1
|
Liu Z, Li Y, Bao J, Tian L, Jie Y. Investigating shared diagnostic genes and mechanisms between metabolic syndrome and dry eye disease via integrated bioinformatics analysis and in vivo validation. Exp Eye Res 2025; 256:110374. [PMID: 40280534 DOI: 10.1016/j.exer.2025.110374] [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: 12/31/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025]
Abstract
Recent research has established a bidirectional connection between metabolic syndrome (MetS) and dry eye disease (DED); however, the underlying mechanisms driving their co-occurrence remain poorly understood. This study employed bioinformatics and in vivo validation to investigate the shared diagnostic genes and underlying mechanisms linking MetS and DED. Differential expression analysis using Limma and weighted gene co-expression network analysis (WGCNA) identified 247 shared driver genes from MetS and DED cohorts. Functional enrichment analysis indicated that these genes are associated with immune regulation and inflammatory responses. Key diagnostic genes (Ccl5, Cxcr4, Ccl4, Spp1) were identified via PPI network analysis and validated using a receiver operating characteristic (ROC) curve. The MetS-DED mouse model further demonstrated CXCR4 overexpression in corneal epithelium and liver. These findings elucidate overlapping biomarkers and pathogenic pathways between MetS and DED, providing critical insights for advancing their diagnosis and therapeutic strategies.
Collapse
Affiliation(s)
- Ziyu Liu
- Beijing Tongren Eye Center, Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yaqiong Li
- Beijing Tongren Eye Center, Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jiayu Bao
- Beijing Tongren Eye Center, Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Lei Tian
- Beijing Tongren Eye Center, Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Ying Jie
- Beijing Tongren Eye Center, Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Liu B, Cai Z, Wang Y, Liu X, Zhang B, Zheng Q, Li J, Li C, Cui Y, Lv P, Yang D. Transglutaminase 2 regulates endothelial cell calcification via IL-6-mediated autophagy. Front Pharmacol 2024; 15:1393534. [PMID: 39654623 PMCID: PMC11625581 DOI: 10.3389/fphar.2024.1393534] [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: 02/29/2024] [Accepted: 10/31/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction Endothelial cell (EC) calcification is an important marker of atherosclerotic calcification. ECs play a critical role not only in atherogenesis but also in intimal calcification, as they have been postulated to serve as a source of osteoprogenitor cells that initiate this process. While the role of transglutaminase 2 (TG2) in cellular differentiation, survival, apoptosis, autophagy, and cell adhesion is well established, the mechanism underlying the TG2-mediated regulation of EC calcification is yet to be fully elucidated. Methods The TG2 gene was overexpressed or silenced by using siRNA and recombinant adenovirus. RT-PCR and WB were used to analyze the relative expression of target genes and proteins. 5-BP method analyzed TG2 activity. mCherry-eGFP-LC3 adenovirus and transmission electron microscopy analyzed EC autophagy level. Calcium concentrations were measured by using a calcium colorimetric assay kit. Alizarin red S staining assay analyzed EC calcification level. Elisa analyzed IL-6 level. Establishing EC calcification model by using a calcification medium (CM). Results Our findings demonstrated that CM increased TG2 activity and expression, which activated the NF-κB signaling pathway, and induced IL-6 autocrine signaling in ECs. Furthermore, IL-6 activated the JAK2/STAT3 signaling pathway to suppress cell autophagy and promoted ECs calcification. Discussion ECs are not only critical for atherogenesis but also believed to be a source of osteoprogenitor cells that initiate intimal calcification. Previous research has shown that TG2 plays an important role in the development of VC, but the mechanism by which it exerts this effect is not yet fully understood. Our results demonstrated that TG2 forms complexes with NF-κB components inhibition of autophagy promoted endothelial cell calcification through EndMT. Therefore, our research investigated the molecular mechanism of EC calcification, which can provide new insights into the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Bo Liu
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhiyuan Cai
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Wang
- The First Department of Ocular Fundus Diseases, Zhengzhou Second Hospital, Zhengzhou, Henan, China
| | - Xinye Liu
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Bin Zhang
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Qian Zheng
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Jingye Li
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Cien Li
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuanbo Cui
- Translational Medical Center, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Pengju Lv
- Department of clinical laboratory, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Dongwei Yang
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
3
|
Yu X, Xu L, Su C, Wang C, Wang Z, Wang Y, Lu X, Sun H. Luteolin Protects against Vascular Calcification by Modulating SIRT1/CXCR4 Signaling Pathway and Promoting Autophagy. AAPS J 2024; 26:111. [PMID: 39438407 DOI: 10.1208/s12248-024-00982-y] [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: 08/25/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Vascular calcification (VC) is a common pathological manifestation of atherosclerosis, hypertension, diabetes vascular disease, vascular injury, chronic kidney disease and aging, which is mainly manifested as increased stiffness of the vascular wall. Oxidative stress and autophagy dysfunction are key factors in the pathogenesis of vascular calcification, but the specific mechanisms and the therapeutic strategy of vascular calcification have not been clarified. In the present study, Sirtuin 1 (SIRT1) was screened as the therapeutic targets for vascular calcification by the bioinformatics. SIRT1 is a nicotinamide adenine dinucleotide, which plays an important role in inhibiting oxidative stress and promoting autophagy. Luteolin (LUT), a kind of natural tetrahydroxyl flavonoid, exists in many plants and has many pharmacological effects such as anti-oxidation and anti-apoptosis. We have reported that luteolin has certain anti-osteoporosis effects in the previous study, and it is accepted that the development of vascular calcification is similar to bone formation, indicating that luteolin may also resist vascular calcification. And luteolin is known to activate SIRT1 to some extent. Moreover, the molecular docking analysis predicted that SIRT1 could bind directly to luteolin. Therefore, the purpose of this study was to investigate the potential role of luteolin in inhibiting oxidative stress and promoting autophagy during vascular calcification via modulating SIRT1 expression. The results showed that luteolin significantly improved vascular calcification induced by a high-fat diet (HFD) and vitamin D3 in rats in vivo. In addition, luteolin significantly repressed the formation of mineralized nodules and ALP activity in H2O2-treated A7r5 cells. Luteolin reduced the level of MDA, LDH and ROS generation, inhibited the protein expression of cleaved caspase-3, cleaved caspase-9, β-catenin and BMP-2 in the aortic tissue of the rat and rat smooth muscle cells (A7r5) treated with hydrogen peroxide. At the same time, luteolin could promote the expression of autophagy related proteins. Moreover, luteolin also produced effects to increase the protein expression levels of SIRT1 more than 2 times both in vivo and in vitro. In terms of mechanism, luteolin attenuated vascular calcification by inhibiting oxidative stress and improving autophagy level, via modulating SIRT1 / CXCR4 signaling pathway. In conclusion, this experiment for the first time revealed that LUT protected against VC via modulating SIRT1 / CXCR4 signaling pathway to promote autophagy and inhibit vascular calcification and may be developed as a new therapeutic agent for vascular calcification and atherosclerosis.
Collapse
Affiliation(s)
- Xiaoyu Yu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Lei Xu
- Office of Ethics Committee, the First Affiliated Hospital, Dalian Medical University, No. 222, Zhongshan Road, Xigang District, Dalian, 116011, China
| | - Ce Su
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Zimeng Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Yanna Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Xiaolong Lu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China.
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, 116044, China.
| |
Collapse
|
4
|
Tian Y, Liu YF, Wang YY, Li YZ, Ding WY, Zhang C. Molecular mechanisms of PTEN in atherosclerosis: A comprehensive review. Eur J Pharmacol 2024; 979:176857. [PMID: 39094923 DOI: 10.1016/j.ejphar.2024.176857] [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: 03/09/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall caused by an imbalance of lipid metabolism and a maladaptive inflammatory response. A variety of harmful cellular changes associated with atherosclerosis include endothelial dysfunction, the migration of circulating inflammatory cells to the arterial wall, the production of proinflammatory cytokines, lipid buildup in the intima, local inflammatory responses in blood vessels, atherosclerosis-associated apoptosis, and autophagy. PTEN inhibits the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB/AKT)/mammalian target of rapamycin (mTOR) pathway through its lipid phosphatase activity. Previous studies have shown that PTEN is closely related to atherosclerosis. This article reviews the role of PTEN in atherosclerosis from the perspectives of autophagy, apoptosis, inflammation, proliferation, and angiogenesis.
Collapse
Affiliation(s)
- Yuan Tian
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| | - Yi-Fan Liu
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| | - Yan-Yue Wang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| | - Yong-Zhen Li
- Department of Pathology, The First People's Hospital of Zigong, Zigong, China, 643099, People's Republic of China
| | - Wen-Yan Ding
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China.
| |
Collapse
|
5
|
Gao C, Wan Q, Yan J, Zhu Y, Tian L, Wei J, Feng B, Niu L, Jiao K. Exploring the Link Between Autophagy-Lysosomal Dysfunction and Early Heterotopic Ossification in Tendons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400790. [PMID: 38741381 PMCID: PMC11267276 DOI: 10.1002/advs.202400790] [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: 01/22/2024] [Revised: 03/26/2024] [Indexed: 05/16/2024]
Abstract
Heterotopic ossification (HO), the pathological formation of bone within soft tissues such as tendon and muscle, is a notable complication resulting from severe injury. While soft tissue injury is necessary for HO development, the specific molecular pathology responsible for trauma-induced HO remains a mystery. The previous study detected abnormal autophagy function in the early stages of tendon HO. Nevertheless, it remains to be determined whether autophagy governs the process of HO generation. Here, trauma-induced tendon HO model is used to investigate the relationship between autophagy and tendon calcification. In the early stages of tenotomy, it is observed that autophagic flux is significantly impaired and that blocking autophagic flux promoted the development of more rampant calcification. Moreover, Gt(ROSA)26sor transgenic mouse model experiments disclosed lysosomal acid dysfunction as chief reason behind impaired autophagic flux. Stimulating V-ATPase activity reinstated both lysosomal acid functioning and autophagic flux, thereby reversing tendon HO. This present study demonstrates that autophagy-lysosomal dysfunction triggers HO in the stages of tendon injury, with potential therapeutic targeting implications for HO.
Collapse
Affiliation(s)
- Chang‐He Gao
- Department of StomatologyTangdu HospitalState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- Department of StomatologyThe Third Affiliated Hospital of Xinxiang Medical UniversityXinxiangHenan453000P. R. China
| | - Qian‐Qian Wan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Jan‐Fei Yan
- Department of StomatologyTangdu HospitalState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Yi‐Na Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Lei Tian
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Jian‐Hua Wei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Bin Feng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Li‐Na Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Kai Jiao
- Department of StomatologyTangdu HospitalState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| |
Collapse
|
6
|
Shao Y, Zheng L, Jiang Y. Cadmium toxicity and autophagy: a review. Biometals 2024; 37:609-629. [PMID: 38277035 DOI: 10.1007/s10534-023-00581-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 12/31/2023] [Indexed: 01/27/2024]
Abstract
Cadmium (Cd) is an important environmental pollutant that poses a threat to human health and represents a critical component of air pollutants, food sources, and cigarette smoke. Cd is a known carcinogen and has toxic effects on the environment and various organs in humans. Heavy metals within an organism are difficult to biodegrade, and those that enter the respiratory tract are difficult to remove. Autophagy is a key mechanism for counteracting extracellular (microorganisms and foreign bodies) or intracellular (damaged organelles and proteins that cannot be degraded by the proteasome) stress and represents a self-protective mechanism for eukaryotes against heavy metal toxicity. Autophagy maintains cellular homeostasis by isolating and gathering information about foreign chemicals associated with other molecular events. However, autophagy may trigger cell death under certain pathological conditions, including cancer. Autophagy dysfunction is one of the main mechanisms underlying Cd-induced cytotoxicity. In this review, the toxic effects of Cd-induced autophagy on different human organ systems were evaluated, with a focus on hepatotoxicity, nephrotoxicity, respiratory toxicity, and neurotoxicity. This review also highlighted the classical molecular pathways of Cd-induced autophagy, including the ROS-dependent signaling pathways, endoplasmic reticulum (ER) stress pathway, Mammalian target of rapamycin (mTOR) pathway, Beclin-1 and Bcl-2 family, and recently identified molecules associated with Cd. Moreover, research directions for Cd toxicity regarding autophagic function were proposed. This review presents the latest theories to comprehensively reveal autophagy behavior in response to Cd toxicity and proposes novel potential autophagy-targeted prevention and treatment strategies for Cd toxicity and Cd-associated diseases in humans.
Collapse
Affiliation(s)
- Yueting Shao
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
- School of Public Health, Guangzhou Medical University, Guangzhou, 511436, China
| | - Liting Zheng
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yiguo Jiang
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China.
- School of Public Health, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
7
|
Zhou G, Liu Y, Wu H, Zhang D, Yang Q, Li Y. Research Progress on Histone Deacetylases Regulating Programmed Cell Death in Atherosclerosis. J Cardiovasc Transl Res 2024; 17:308-321. [PMID: 37821683 DOI: 10.1007/s12265-023-10444-z] [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: 05/07/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
Histone deacetylases (HDACs) are epigenetic modifying enzyme that is closely related to chromatin structure and gene transcription, and numerous studies have found that HDACs play an important regulatory role in atherosclerosis disease. Apoptosis, autophagy and programmed necrosis as the three typical programmed cell death modalities that can lead to cell loss and are closely related to the developmental process of atherosclerosis. In recent years, accumulating evidence has shown that the programmed cell death mediated by HDACs is increasingly important in the pathophysiology of atherosclerosis. This paper first gives a brief overview of HDACs, the mechanism of programmed cell death, and their role in atherosclerosis, and then further elaborates on the role and mechanism of HDACs in regulating apoptosis, autophagy, and programmed necrosis in atherosclerosis, respectively, to provide new effective measures and theoretical basis for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Gang Zhou
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| | - Yanfang Liu
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| | - Hui Wu
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China.
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China.
- Department of Cardiology, Yichang Central People's Hospital, Yiling Road 183, Yichang, 443003, Hubei, China.
| | - Dong Zhang
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| | - Qingzhuo Yang
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| | - Yi Li
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| |
Collapse
|
8
|
Zhou G, Liu P, Zhang C, Huang Q, Zhao Z, Wu S, Li D, Liu H. HDAC2 counteracts vascular calcification by activating autophagy in chronic kidney disease. FASEB J 2024; 38:e23470. [PMID: 38354035 DOI: 10.1096/fj.202301429r] [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: 07/14/2023] [Revised: 12/16/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024]
Abstract
Vascular calcification is a major risk factor for cardiovascular disease mortality, with a significant prevalence in chronic kidney disease (CKD). Pharmacological inhibition of histone acetyltransferase has been proven to protect against from vascular calcification. However, the role of Histone Deacetylase 2 (HDAC2) and molecular mechanisms in vascular calcification of CKD remains unknown. An in vivo model of CKD was established using mouse fed with a high adenine and phosphate diet, and an in vitro model was produced using human aortic vascular smooth muscle cells (VSMCs) stimulated with β-glycerophosphate (β-GP). HDAC2 expression was found to be reduced in medial artery of CKD mice and β-GP-induced VSMCs. Overexpression of HDAC2 attenuated OPN and OCN upregulation, α-SMA and SM22α downregulation, and calcium deposition in aortas of CKD. The in vitro results also demonstrated that β-GP-induced osteogenic differentiation was inhibited by HDAC2. Furthermore, we found that HDAC2 overexpression caused an increase in LC3II/I, a decrease in p62, and an induction of autophagic flux. Inhibition of autophagy using its specific inhibitor 3-MA blocked HDAC2's protective effect on osteogenic differentiation in β-GP-treated VSMCs. Taken together, these results suggest that HDAC2 may protect against vascular calcification by the activation of autophagy, laying out a novel insight for the molecular mechanism in vascular calcification of CKD.
Collapse
Affiliation(s)
- Guangyu Zhou
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Pai Liu
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chen Zhang
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qun Huang
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zixia Zhao
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Si Wu
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Detian Li
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hongbo Liu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Hewedy WA, Abdulmalek SA, Ghareeb DA, Habiba ES. AMPK-mediated autophagy is involved in the protective effect of canagliflozin in the vitamin D3 plus nicotine calcification model in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:873-888. [PMID: 37522915 PMCID: PMC10791829 DOI: 10.1007/s00210-023-02627-x] [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: 02/12/2023] [Accepted: 07/12/2023] [Indexed: 08/01/2023]
Abstract
Vascular calcification (VC) is a major risk factor for cardiovascular events. A mutual interplay between inflammation, oxidative stress, apoptosis, and autophagy is implicated in its development. Herein, we aimed to evaluate the potential protective effects of canagliflozin in a vitamin D3 plus nicotine (VDN) model of VC, and to explore potential mechanisms. VC was induced by VDN in adult male Wistar rats on day one. Then, rats were randomly assigned into three groups to receive canagliflozin (10 mg or 20 mg/kg/day) or its vehicle for 4 weeks. Age-matched normal rats served as a control group. After euthanization, aorta and kidneys were harvested for biochemical and histopathological evaluation of calcification. Aortic markers of oxidative stress, alkaline phosphatase (ALP) activity, runt-related transcription factor (Runx2) and bone morphogenic protein-2 (BMP-2) levels were determined. Additionally, the protein expression of autophagic markers, LC3 and p62, and adenosine monophosphate activated protein kinase (AMPK) were also assessed in aortic homogenates. Canagliflozin dose-dependently improved renal function, enhanced the antioxidant capacity of aortic tissues and reduced calcium deposition in rat aortas and kidneys. Both doses of canagliflozin attenuated ALP and osteogenic markers while augmented the expression of autophagic markers and AMPK. Histopathological examination of aortas and kidneys by H&E and Von Kossa stain further support the beneficial effect of canagliflozin. Canagliflozin could alleviate VDN-induced vascular calcification, in a dose dependent manner, via its antioxidant effect and modulation of autophagy. Further studies are needed to verify whether this effect is a member or a class effect.
Collapse
Affiliation(s)
- Wafaa A Hewedy
- Clinical Pharmacology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
- Al-Moassat Medical Campus, Elhadara, Clinical Pharmacology Department, Faculty of Medicine, Alexandria University, 21561, Alexandria, Egypt.
| | - Shaymaa A Abdulmalek
- Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Doaa A Ghareeb
- Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Esraa S Habiba
- Clinical Pharmacology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
10
|
Yaribeygi H, Maleki M, Santos RD, Jamialahmadi T, Sahebkar A. Glp-1 Mimetics and Autophagy in Diabetic Milieu: State-of-the-Art. Curr Diabetes Rev 2024; 20:e250124226181. [PMID: 38299271 DOI: 10.2174/0115733998276570231222105959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 02/02/2024]
Abstract
The diabetic milieu is associated with cascades of pathophysiological pathways that culminate in diabetic complications and tissue injuries. Autophagy is an essential process mandatory for cell survival and tissue homeostasis by degrading damaged organelles and removing injured cells. However, it may turn into a pathological process in an aberrant mode in the diabetic and/or malignant milieu. Moreover, autophagy could serve as a promising therapeutic target for many complications related to tissue injury. Glp-1 mimetics are a class of newer antidiabetic agents that reduce blood glucose through several pathways. However, some evidence suggests that they can provide extra glycemic benefits by modulating autophagy, although there is no complete understanding of this mechanism and its underlying molecular pathways. Hence, in the current review, we aimed to provide new insights on the possible impact of Glp-1 mimetics on autophagy and consequent benefits as well as mediating pathways.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Mina Maleki
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Raul D Santos
- Lipid Clinic Heart Institute (Incor), University of São Paulo, Medical School Hospital, São Paulo, Brazil
| | - Tannaz Jamialahmadi
- Medical Toxicolgy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Medical Toxicolgy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Zhou Z, Li Y, Jiang W, Wang Z. Molecular Mechanism of Calycosin Inhibited Vascular Calcification. Nutrients 2023; 16:99. [PMID: 38201929 PMCID: PMC10781010 DOI: 10.3390/nu16010099] [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: 10/25/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Vascular calcification (VC) is a pathological condition frequently observed in cardiovascular diseases. Primary factors contributing to VC are osteogenic differentiation of vascular smooth muscle and hydroxyapatite deposition. Targeted autophagy (a lysosome-mediated mechanism for degradation/recycling of unnecessary cellular components) is a useful approach for inhibiting VC and promoting vascular cell health. Calycosin has been shown to alleviate atherosclerosis by enhancing macrophage autophagy, but its therapeutic effect on VC has not been demonstrated. Using an in vitro model (rat thoracic aortic smooth muscle cell line A7r5), we demonstrated effective inhibition of VC using calycosin (the primary flavonoid component of astragalus), based on the enhancement of autophagic flux. Calycosin treatment activated AMPK/mTOR signaling to induce initiation of autophagy and restored mTORC1-dependent autophagosome-lysosome fusion in late-stage autophagy by promoting soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex formation, thereby preventing stoppage of autophagy in calcified cells. Calycosin substantially reduced degrees of both osteogenic differentiation and calcium deposition in our VC cell model by enhancing autophagy. The present findings clarify the mechanism whereby calycosin mitigates autophagy stoppage in calcified smooth muscle cells and provide a basis for effective VC treatment via autophagy enhancement.
Collapse
Affiliation(s)
- Zekun Zhou
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Z.Z.); (Y.L.)
| | - Yi Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Z.Z.); (Y.L.)
| | - Wei Jiang
- College of Biological Sciences, China Agricultural University, Beijing 100193, China;
| | - Zengli Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Z.Z.); (Y.L.)
| |
Collapse
|
12
|
Phadwal K, Tan X, Koo E, Zhu D, MacRae VE. Metformin ameliorates valve interstitial cell calcification by promoting autophagic flux. Sci Rep 2023; 13:21435. [PMID: 38052777 PMCID: PMC10698150 DOI: 10.1038/s41598-023-47774-6] [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: 03/15/2023] [Accepted: 11/18/2023] [Indexed: 12/07/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is the most common heart disease of the developed world. It has previously been established that metformin administration reduces arterial calcification via autophagy; however, whether metformin directly regulates CAVD has yet to be elucidated. In the present study we investigated whether metformin alleviates valvular calcification through the autophagy-mediated recycling of Runx2. Calcification was reduced in rat valve interstitial cells (RVICs) by metformin treatment (0.5-1.5 mM) (P < 0.01), with a marked decrease in Runx2 protein expression compared to control cells (P < 0.05). Additionally, upregulated expression of Atg3 and Atg7 (key proteins required for autophagosome formation), was observed following metformin treatment (1 mM). Blocking autophagic flux using Bafilomycin-A1 revealed colocalisation of Runx2 with LC3 puncta in metformin treated RVICs (P < 0.001). Comparable Runx2 accumulation was seen in LC3 positive autolysosomes present within cells that had been treated with both metformin and hydroxychloroquine in combination (P < 0.001). Mechanistic studies employing three-way co-immunoprecipitation with Runx2, p62 and LC3 suggested that Runx2 binds to LC3-II upon metformin treatment in VICs. Together these studies suggest that the utilisation of metformin may represent a novel strategy for the treatment of CAVD.
Collapse
Affiliation(s)
- K Phadwal
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK.
| | - X Tan
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
- Guangzhou Institute of Cardiovascular Diseases, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - E Koo
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - D Zhu
- Guangzhou Institute of Cardiovascular Diseases, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - V E MacRae
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| |
Collapse
|
13
|
Xie W, Shan Y, Wu Z, Liu N, Yang J, Zhang H, Sun S, Chi J, Feng W, Lin H, Guo H. Herpud1 deficiency alleviates homocysteine-induced aortic valve calcification. Cell Biol Toxicol 2023; 39:2665-2684. [PMID: 36746840 DOI: 10.1007/s10565-023-09794-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 01/21/2023] [Indexed: 02/08/2023]
Abstract
OBJECTIVES To evaluate the role and therapeutic value of homocysteine (hcy)-inducible endoplasmic reticulum stress (ERS) protein with ubiquitin like domain 1 (Herpud1) in hcy-induced calcific aortic valve disease (CAVD). BACKGROUND The morbidity and mortality rates of calcific aortic valve disease (CAVD) remain high while treatment options are limited. METHODS In vivo, we use the low-density lipoprotein receptor (LDLR) and Herpud1 double knockout (LDLR-/-/Herpud1-/-) mice and used high methionine diet (HMD) to assess of aortic valve calcification lesions, ERS activation, autophagy, and osteogenic differentiation of aortic valve interstitial cells (AVICs). In vitro, the role of Herpud1 in the Hcy-related osteogenic differentiation of AVICs was investigated by manipulating of Herpud1 expression. RESULTS Herpud1 was highly expressed in calcified human and mouse aortic valves as well as primary aortic valve interstitial cells (AVICs). Hcy increased Herpud1 expression through the ERS pathway and promoted CAVD progression. Herpud1 deficiency inhibited hcy-induced CAVD in vitro and in vivo. Herpud1 silencing activated cell autophagy, which subsequently inhibited hcy-induced osteogenic differentiation of AVICs. ERS inhibitor 4-phenyl butyric acid (4-PBA) significantly attenuated aortic valve calcification in HMD-fed low-density lipoprotein receptor-/- (LDLR-/-) mice by suppressing ERS and subsequent Herpud1 biosynthesis. CONCLUSIONS These findings identify a previously unknown mechanism of Herpud1 upregulation in Hcy-related CAVD, suggesting that Herpud1 silencing or inhibition is a viable therapeutic strategy for arresting CAVD progression. HIGHLIGHTS • Herpud1 is upregulated in the leaflets of Hcy-treated mice and patients with CAVD. • In mice, global knockout of Herpud1 alleviates aortic valve calcification and Herpud1 silencing activates cell autophagy, inhibiting osteogenic differentiation of AVICs induced by Hcy. • 4-PBA suppressed Herpud1 expression to alleviate AVIC calcification in Hcy treated AVICs and to mitigate aortic valve calcification in mice.
Collapse
Affiliation(s)
- Wenqing Xie
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, 312000, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China
| | - Yue Shan
- Department of Anesthesiology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, 312000, Zhejiang, China
| | - Zhuonan Wu
- College of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Nan Liu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China
| | - Jinjin Yang
- Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, China
| | - Hanlin Zhang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Shiming Sun
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jufang Chi
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, 312000, Zhejiang, China
- Shaoxing Key Laboratory of Cardio-cerebral Vascular Disease Rehabilitation Technology Innovation and Application, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Weizhong Feng
- Department of Cardiovascular Surgery, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, 312000, Zhejiang, China
| | - Hui Lin
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, 312000, Zhejiang, China.
- Shaoxing Key Laboratory of Cardio-cerebral Vascular Disease Rehabilitation Technology Innovation and Application, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China.
| | - Hangyuan Guo
- College of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, China.
- Shaoxing Key Laboratory of Cardio-cerebral Vascular Disease Rehabilitation Technology Innovation and Application, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China.
| |
Collapse
|
14
|
Lino Cardenas CL, Jiang W, Kajuluri LP, Singh K, Ostrom K, Li R, Cherbonneau F, Boerboom S, Birchenough C, Roh K, Chou EL, Shahrooz Z, Nicholson C, Johnson AL, Lee S, Ichinose F, Bloch DB, Nigwekar S, Ellinor PT, Musolino P, Lindsay ME, Dou Z, Miller CL, Malhotra R. Treatment of calcific arterial disease via enhancement of autophagy using GSK343. iScience 2023; 26:108360. [PMID: 38033629 PMCID: PMC10681952 DOI: 10.1016/j.isci.2023.108360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/31/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
Vascular calcification is a hallmark of atherosclerotic disease and serves as a strong predictor and risk factor for cardiovascular events. Growing evidence suggests that autophagy may play a protective role in early atherosclerosis. The precise effects of autophagy on VSMC-mediated calcification remain unknown. In this study, we utilized multi-omic profiling to investigate impaired autophagy at the transcriptional level as a key driver of VSMC calcification. Our findings revealed that impaired autophagy is an essential determinant of VSMC calcification. We observed that an osteogenic environment affects the open chromatin status of VSMCs, compromising the transcriptional activation of autophagy initiation genes. In vivo experiments involve pharmacological and genetic activation of autophagy using mouse models of spontaneous large (Mgp-/-) and small (Abcc6-/-) artery calcification. Taken together, these data advance our mechanistic understanding of vascular calcification and provide important insights for a broad range of cardiovascular diseases involving VSMC phenotype switch.
Collapse
Affiliation(s)
- Christian L. Lino Cardenas
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Wanlin Jiang
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lova P. Kajuluri
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kuldeep Singh
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Katrina Ostrom
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebecca Li
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Francois Cherbonneau
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sophie Boerboom
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Claire Birchenough
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kangsan Roh
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth L. Chou
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Zarbafian Shahrooz
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Christopher Nicholson
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Adam L. Johnson
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sujin Lee
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fumito Ichinose
- Department of Anesthesia, Critical Care and Pain Medicine at Massachusetts General Hospital, Boston, MA 02114, USA
| | - Donald B. Bloch
- Department of Anesthesia, Critical Care and Pain Medicine at Massachusetts General Hospital, Boston, MA 02114, USA
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sagar Nigwekar
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Patrick T. Ellinor
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Patricia Musolino
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Mark E. Lindsay
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhixun Dou
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Clint L. Miller
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA 22908, USA
| | - Rajeev Malhotra
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Sun X, Zheng Y, Xie L, Zhou Y, Liu R, Ma Y, Zhao M, Liu Y. Autophagy reduces aortic calcification in diabetic mice by reducing matrix vesicle body-mediated IL-1β release. Exp Cell Res 2023; 432:113803. [PMID: 37774764 DOI: 10.1016/j.yexcr.2023.113803] [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: 08/04/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Vascular calcification (VC) is a common pathological process of cardiovascular disease that occurs in patients with type 2 diabetes mellitus (T2DM). However, the molecular basis of VC progression remains unknown. A GEO dataset (GSE146638) was analyzed to show that microbodies and IL-1β may play important roles in the pathophysiology of VC. The release of matrix vesicle bodies (MVBs) and IL-1β and the colocalization of IL-1β with MVBs or autophagosomes were studied by immunofluorescence in an in vivo diabetes mouse model with aortic calcification and an in vitro high glucose cell calcification model. MVB numbers, IL-1β levels and autophagy were increased in calcified mouse aortas and calcified vascular smooth muscle cells (VSMCs). IL-1β colocalized with MVBs and autophagosomes. The MVBs from calcified VSMCs induced the calcification of normal recipient VSMCs, and this effect was alleviated by silencing IL-1β. The autophagy inducer rapamycin reduced IL-1β expression and calcification in VSMCs, while these processes were induced by the autophagy inhibitor chloroquine. In conclusion, our results suggested that MVBs could carry IL-1β out of cells and induce VC in normal VSMCs, and these processes could be counteracted by autophagy. These results suggested that MVB-mediated IL-1β release may be an effective target for treating vascular calcification.
Collapse
Affiliation(s)
- Xiaolei Sun
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Department of Interventional Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Laboratory of Nucleic Acids in Medicine for National High-Level Talents, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China; Cardiovascular and Metabolic Diseases Key Laboratory of Sichuan, Luzhou, 646000, China.
| | - Yang Zheng
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Department of Vascular and Interventional, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Linzhuo Xie
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yuanqun Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China; State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Runyu Liu
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yarong Ma
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Ming Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China.
| | - Yong Liu
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
16
|
Song JH, Liu MY, Ma YX, Wan QQ, Li J, Diao XO, Niu LN. Inflammation-associated ectopic mineralization. FUNDAMENTAL RESEARCH 2023; 3:1025-1038. [PMID: 38933004 PMCID: PMC11197766 DOI: 10.1016/j.fmre.2022.04.020] [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: 01/24/2022] [Revised: 04/06/2022] [Accepted: 04/21/2022] [Indexed: 10/18/2022] Open
Abstract
Ectopic mineralization refers to the deposition of mineralized complexes in the extracellular matrix of soft tissues. Calcific aortic valve disease, vascular calcification, gallstones, kidney stones, and abnormal mineralization in arthritis are common examples of ectopic mineralization. They are debilitating diseases and exhibit excess mortality, disability, and morbidity, which impose on patients with limited social or financial resources. Recent recognition that inflammation plays an important role in ectopic mineralization has attracted the attention of scientists from different research fields. In the present review, we summarize the origin of inflammation in ectopic mineralization and different channels whereby inflammation drives the initiation and progression of ectopic mineralization. The current knowledge of inflammatory milieu in pathological mineralization is reviewed, including how immune cells, pro-inflammatory mediators, and osteogenic signaling pathways induce the osteogenic transition of connective tissue cells, providing nucleating sites and assembly of aberrant minerals. Advances in the understanding of the underlying mechanisms involved in inflammatory-mediated ectopic mineralization enable novel strategies to be developed that may lead to the resolution of these enervating conditions.
Collapse
Affiliation(s)
| | | | | | - Qian-Qian Wan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Centre for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jing Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Centre for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xiao-Ou Diao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Centre for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Li-Na Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Centre for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
17
|
Yang S, Zeng Z, Yuan Q, Chen Q, Wang Z, Xie H, Liu J. Vascular calcification: from the perspective of crosstalk. MOLECULAR BIOMEDICINE 2023; 4:35. [PMID: 37851172 PMCID: PMC10584806 DOI: 10.1186/s43556-023-00146-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Abstract
Vascular calcification (VC) is highly correlated with cardiovascular disease morbidity and mortality, but anti-VC treatment remains an area to be tackled due to the ill-defined molecular mechanisms. Regardless of the type of VC, it does not depend on a single cell but involves multi-cells/organs to form a complex cellular communication network through the vascular microenvironment to participate in the occurrence and development of VC. Therefore, focusing only on the direct effect of pathological factors on vascular smooth muscle cells (VSMCs) tends to overlook the combined effect of other cells and VSMCs, including VSMCs-VSMCs, ECs-VMSCs, Macrophages-VSMCs, etc. Extracellular vesicles (EVs) are a collective term for tiny vesicles with a membrane structure that are actively secreted by cells, and almost all cells secrete EVs. EVs docked on the surface of receptor cells can directly mediate signal transduction or transfer their contents into the cell to elicit a functional response from the receptor cells. They have been proven to participate in the VC process and have also shown attractive therapeutic prospects. Based on the advantages of EVs and the ability to be detected in body fluids, they may become a novel therapeutic agent, drug delivery vehicle, diagnostic and prognostic biomarker, and potential therapeutic target in the future. This review focuses on the new insight into VC molecular mechanisms from the perspective of crosstalk, summarizes how multi-cells/organs interactions communicate via EVs to regulate VC and the emerging potential of EVs as therapeutic methods in VC. We also summarize preclinical experiments on crosstalk-based and the current state of clinical studies on VC-related measures.
Collapse
Affiliation(s)
- Shiqi Yang
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zhaolin Zeng
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qing Yuan
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qian Chen
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zuo Wang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hui Xie
- Department of Orthopaedics, Movement System Injury and Repair Research Centre, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
18
|
Fang J, Qian Y, Chen J, Xu D, Cao N, Zhu G, Hu W, Hu H, Qian N, Yang S, Wang J, Liu X. Human antigen R regulates autophagic flux by stabilizing autophagy-associated mRNA in calcific aortic valve disease. Cardiovasc Res 2023; 119:2117-2129. [PMID: 37183487 DOI: 10.1093/cvr/cvad077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 03/23/2023] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
AIMS The incidence of calcific aortic valve disease (CAVD) has risen over the last decade and is expected to continue rising; however, pharmacological approaches have proven ineffective. In this study, we evaluated the role and underlying mechanisms of human antigen R (HuR)-mediated post-transcriptional regulation in CAVD. METHODS AND RESULTS We found that HuR was significantly upregulated in human calcified aortic valves and primary aortic valvular interstitial cells (VICs) following osteogenic stimulation. Subsequent functional studies revealed that HuR silencing ameliorated calcification both in vitro and in vivo. For the first time, we demonstrated that HuR directly interacted with the transcript of phosphatidylinositol-5-phosphate 4-kinase, type II, alpha (PIP4K2A), which mediates phosphatidylinositol signalling, facilitates autophagy, and acts as an mRNA stabilizer. HuR positively modulated PIP4K2A expression at the post-transcriptional level and consequently influenced the AKT/mTOR/ATG13 pathway to regulate autophagy and CAVD progression. CONCLUSION Our study provides new insights into the post-transcriptional regulatory role of HuR in modulating autophagy-positive factors to regulate the pathogenesis of CAVD. Our findings highlight the potential of HuR as an innovative therapeutic target in CAVD treatment.
Collapse
Affiliation(s)
- Juan Fang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yi Qian
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Department of Cardiology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Jinhua, Zhejiang Province, China
| | - Jinyong Chen
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Dilin Xu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Naifang Cao
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Gangjie Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Wangxing Hu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Haochang Hu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ningjing Qian
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Shuangshuang Yang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jian'an Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Xianbao Liu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
19
|
Zhang J, Pang Q, Wang S, Wu L, Zhang A. Associated factors of cardiac valve calcification and its prognostic effects among patients with chronic kidney disease: a systematic review and meta-analysis. Front Cardiovasc Med 2023; 10:1120634. [PMID: 37180797 PMCID: PMC10169583 DOI: 10.3389/fcvm.2023.1120634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
Background Cardiac valve calcification (CVC) is highly prevalent and a risk factor for adverse outcomes in patients with chronic kidney disease (CKD). This meta-analysis aimed to investigate the risk factors for CVC and association between CVC and mortality in CKD patients. Method Three electronic databases including PubMed, Embase, and Web of Science were searched for relevant studies up to November 2022. Hazard ratios (HR), odds ratios (OR), and 95% confidence intervals (CI) were pooled using random-effect meta-analyses. Results 22 studies were included in the meta-analysis. Pooled analyses showed that CKD patients with CVC were relatively older, had a higher body mass index, left atrial dimension, C-reaction protein level, and a declined ejection fraction. Calcium and phosphate metabolism dysfunction, diabetes, coronary heart disease, and duration of dialysis were all predictors for CVC in CKD patients. The presence of CVC (both aortic valve and mitral valve) increased the risk of all-cause and cardiovascular mortality in CKD patients. However, the prognostic value of CVC for mortality was not significant anymore in patients with peritoneal dialysis. Conclusion CKD patients with CVC had a greater risk of all-cause and cardiovascular mortality. Multiple associated factors for development of CVC in CKD patients should be taken into consideration by healthcare professionals to improve prognosis. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/, identifier [CRD42022364970].
Collapse
Affiliation(s)
- Jialing Zhang
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qi Pang
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shiyuan Wang
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Leiyun Wu
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Aihua Zhang
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
20
|
Ma J, Li Y, Yang X, Liu K, Zhang X, Zuo X, Ye R, Wang Z, Shi R, Meng Q, Chen X. Signaling pathways in vascular function and hypertension: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:168. [PMID: 37080965 PMCID: PMC10119183 DOI: 10.1038/s41392-023-01430-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/22/2023] Open
Abstract
Hypertension is a global public health issue and the leading cause of premature death in humans. Despite more than a century of research, hypertension remains difficult to cure due to its complex mechanisms involving multiple interactive factors and our limited understanding of it. Hypertension is a condition that is named after its clinical features. Vascular function is a factor that affects blood pressure directly, and it is a main strategy for clinically controlling BP to regulate constriction/relaxation function of blood vessels. Vascular elasticity, caliber, and reactivity are all characteristic indicators reflecting vascular function. Blood vessels are composed of three distinct layers, out of which the endothelial cells in intima and the smooth muscle cells in media are the main performers of vascular function. The alterations in signaling pathways in these cells are the key molecular mechanisms underlying vascular dysfunction and hypertension development. In this manuscript, we will comprehensively review the signaling pathways involved in vascular function regulation and hypertension progression, including calcium pathway, NO-NOsGC-cGMP pathway, various vascular remodeling pathways and some important upstream pathways such as renin-angiotensin-aldosterone system, oxidative stress-related signaling pathway, immunity/inflammation pathway, etc. Meanwhile, we will also summarize the treatment methods of hypertension that targets vascular function regulation and discuss the possibility of these signaling pathways being applied to clinical work.
Collapse
Affiliation(s)
- Jun Ma
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yanan Li
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiangyu Yang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Kai Liu
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xin Zhang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xianghao Zuo
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Runyu Ye
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ziqiong Wang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Rufeng Shi
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Qingtao Meng
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
21
|
Feng Y, Yu M, Wang C, Xia J, Huang L, Tang Y, Xiao Q, Pu L, Wang L, Li G, Li Y. BRG1 is involved in vascular calcification in chronic renal disease via autophagy of vascular smooth muscle cells. iScience 2023; 26:106485. [PMID: 37020968 PMCID: PMC10067948 DOI: 10.1016/j.isci.2023.106485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/03/2022] [Accepted: 03/20/2023] [Indexed: 04/04/2023] Open
Abstract
We aimed to investigate the mechanisms of Brahma related gene 1 (BRG1) in promoting vascular calcification in chronic kidney disease (CKD). The expression of BRG1 was examined in high phosphorus stimulated rat aortic smooth muscle cells (RASMCs) and calcified artery tissues from rat models and hemodialysis patients. Autophagosome formation was measured in high phosphorus stimulated RASMCs with and without BRG1 knock-down. We also detected the coexistence of BGR1 and exosomes, and measured the circulatory levels of BRG1 in the hemodialysis patients. BRG1 promoted the osteogenic transdifferentiation of RASMCs. Silencing BRG1 prevented autophagy from being induced by high phosphorus stimulation in RASMCs. Increased expression of BRG1 was observed in calcified blood vessels. Serum BRG1 level increased in the hemodialysis patients. BRG1 was involved in the development of high phosphorus induced osteogenic phenotype in vitro and in vivo, and its underlying mechanism might be facilitating autophagy.
Collapse
|
22
|
Zhang T, Wu J, Yao X, Zhang Y, Wang Y, Han Y, Wu Y, Xu Z, Lan J, Han S, Zou H, Sun Q, Wang D, Zhang J, Wang G. The Aldose Reductase Inhibitor Epalrestat Maintains Blood-Brain Barrier Integrity by Enhancing Endothelial Cell Function during Cerebral Ischemia. Mol Neurobiol 2023; 60:3741-3757. [PMID: 36940077 DOI: 10.1007/s12035-023-03304-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 03/08/2023] [Indexed: 03/21/2023]
Abstract
Excessive activation of aldose reductase (AR) in the brain is a risk factor for aggravating cerebral ischemia injury. Epalrestat is the only AR inhibitor with proven safety and efficacy, which is used in the clinical treatment of diabetic neuropathy. However, the molecular mechanisms underlying the neuroprotection of epalrestat remain unknown in the ischemic brain. Recent studies have found that blood-brain barrier (BBB) damage was mainly caused by increased apoptosis and autophagy of brain microvascular endothelial cells (BMVECs) and decreased expression of tight junction proteins. Thus, we hypothesized that the protective effect of epalrestat is mainly related to regulating the survival of BMVECs and tight junction protein levels after cerebral ischemia. To test this hypothesis, a mouse model of cerebral ischemia was established by permanent middle cerebral artery ligation (pMCAL), and the mice were treated with epalrestat or saline as a control. Epalrestat reduced the ischemic volume, enhanced BBB function, and improved the neurobehavior after cerebral ischemia. In vitro studies revealed that epalrestat increased the expression of tight junction proteins, and reduced the levels of cleaved-caspase3 and LC3 proteins in mouse BMVECs (bEnd.3 cells) exposed to oxygen-glucose deprivation (OGD). In addition, bicalutamide (an AKT inhibitor) and rapamycin (an mTOR inhibitor) increased the epalrestat-induced reduction in apoptosis and autophagy related protein levels in bEnd.3 cells with OGD treatment. Our findings suggest that epalrestat improves BBB function, which may be accomplished by reducing AR activation, promoting tight junction proteins expression, and upregulating AKT/mTOR signaling pathway to inhibit apoptosis and autophagy in BMVECs.
Collapse
Affiliation(s)
- Tongshuai Zhang
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Jinrong Wu
- Department of Anaesthesiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Xinmin Yao
- Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Yao Zhang
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yue Wang
- Department of Anesthesiology, Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Yang Han
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yun Wu
- The Medical Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Zhenyu Xu
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Jing Lan
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Siyu Han
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Haifeng Zou
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Qixu Sun
- Department of Gastroenterology, Penglai People's Hospital, Yantai, 264117, Shandong, China
| | - Dandan Wang
- Wu Lian De Memorial Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| | - Jingyu Zhang
- The Medical Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| | - Guangyou Wang
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
23
|
Vaspin alleviates the lncRNA LEF1-AS1-induced osteogenic differentiation of vascular smooth muscle cells via the Hippo/YAP signaling pathway. Exp Cell Res 2022; 421:113407. [PMID: 36334793 DOI: 10.1016/j.yexcr.2022.113407] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Vascular calcification (VC) is closely related to higher cardiovascular mortality and morbidity, and vascular smooth muscle cell (VSMC) switching to osteogenic-like cells is crucial for VC. LncRNA LEF1-AS1 promotes atherosclerosis and dental pulp stem cells calcification, while its role in VC remains unknown. Visceral adipose tissue-derived serine protease inhibitor (vaspin) is an adipokine regulating bone metabolism. However, the relationship between vaspin and VC is still unclear. We aimed to explore the role of LEF1-AS1 on VSMC osteogenic transition, whether vaspin inhibited LEF1-AS1-mediated osteogenic differentiation of VSMCs, and the responsible mechanism. In this study, quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting analysis indicated that LEF1-AS1 overexpression significantly upregulated osteogenic marker Runt-related transcription factor-2 (RUNX2) level and downregulated VSMC contractile marker α-smooth muscle actin (α-SMA) level. Alizarin red staining, alkaline phosphatase (ALP) staining, ALP activity assay, and calcium content assay also suggested that LEF1-AS1 overexpression promoted calcium deposition in VSMCs. However, vaspin treatment abolished this phenomenon. Mechanistically, LEF1-AS1 markedly decreased phosphorylated YAP level, while vaspin reversed LEF1-AS1-induced phosphorylated YAP decline. Our results revealed that LEF1-AS1 accelerated the osteogenic differentiation of VSMCs by regulating the Hippo/YAP pathway, while vaspin eliminated the LEF1-AS1-meditated VSMCs osteogenic phenotype switch.
Collapse
|
24
|
Li X, Liu XL, Li X, Zhao YC, Wang QQ, Zhong HY, Liu DD, Yuan C, Zheng TF, Zhang M. Dickkopf1 (Dkk1) Alleviates Vascular Calcification by Regulating the Degradation of Phospholipase D1 (PLD1). J Cardiovasc Transl Res 2022; 15:1327-1339. [PMID: 35426038 DOI: 10.1007/s12265-022-10251-y] [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: 12/18/2021] [Accepted: 04/04/2022] [Indexed: 12/16/2022]
Abstract
Vascular calcification (VC) is a significant risk factor for cardiovascular mortality and morbidity in patients with atherosclerosis (AS), chronic kidney disease, and diabetes. Dickkopf1 (Dkk1) is a multifunctional secreted glycoprotein that has been explored as a novel potential antitumor target. Recently, Dkk1 was shown to be closely associated with AS development. However, the role of Dkk1 in VC remains elusive. In this study, we explored the role and molecular mechanisms of Dkk1 in VC based on a smooth muscle-specific Dkk1-knockout (Dkk1SMKO) mouse model. Our data indicated that Dkk1 expression was decreased under calcifying conditions and that Dkk1 overexpression alleviated high phosphate-induced vascular calcification. In vivo, smooth muscle Dkk1-specific knockout aggravated vascular calcification in mice. However, phospholipase D1 (PLD1) overexpression partially weakened the protective effect of Dkk1 against vascular calcification. Mechanistically, Dkk1 slowed vascular calcification by promoting the degradation of PLD1 via the regulating autophagosome formation and maturation. In conclusion, we found that Dkk1 could alleviate vascular calcification by regulating the degradation of PLD1.
Collapse
Affiliation(s)
- Xuan Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107, Wen Hua Xi Road, Jinan, 250012, Shandong, China
| | - Xiao-Lin Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107, Wen Hua Xi Road, Jinan, 250012, Shandong, China
| | - Xiao Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107, Wen Hua Xi Road, Jinan, 250012, Shandong, China
| | - Ya-Chao Zhao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107, Wen Hua Xi Road, Jinan, 250012, Shandong, China
| | - Qian-Qian Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107, Wen Hua Xi Road, Jinan, 250012, Shandong, China
| | - Hong-Yu Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107, Wen Hua Xi Road, Jinan, 250012, Shandong, China
| | - Dong-Dong Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107, Wen Hua Xi Road, Jinan, 250012, Shandong, China
| | - Chong Yuan
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107, Wen Hua Xi Road, Jinan, 250012, Shandong, China
| | - Teng-Fei Zheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107, Wen Hua Xi Road, Jinan, 250012, Shandong, China.
| | - Mei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107, Wen Hua Xi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
25
|
Guo L, Wang Y, Li S, Zhou L, Li D. GALNT3 protects against phosphate-induced calcification in vascular smooth muscle cells by enhancing active FGF23 and inhibiting the wnt/β-catenin signaling pathway. Cell Signal 2022; 100:110477. [PMID: 36162588 DOI: 10.1016/j.cellsig.2022.110477] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/12/2022] [Accepted: 09/20/2022] [Indexed: 11/03/2022]
Abstract
Vascular calcification (VC) acts as a notable risk factor in the cardiovascular system. Disorder of phosphorus (Pi) metabolism promotes VC. Recent findings show that polypeptide N-acetylgalactosaminyltransferase 3(GALNT3) is Pi responsive and with potent effects on Pi homeostasis. However, whether GALNT3 is involved in high Pi-induced VC remains unclear. The present study investigated the potential role of GALNT3 as a novel regulator of VC. In vitro, human aortic smooth muscle cells (HASMCs) calcification was induced by inorganic Pi, while in vivo, C57BL/6 J mice were used to determine the effects of GALNT3 on Vitamin D3-induced medial arterial calcification. Alizarin red staining, Von Kossa staining, calcium and alkaline phosphatase (ALP) activity were performed to test VC. We showed that expression of GALNT3 was increased in the calcified HASMCs and aortas of the calcified mice.In vitro, overexpression of GALNT3 increased the levels of active full-length FGF23, accompanied by suppression of the osteoblast-related factors (Runx2 and BMP2), and further inhibited the formation of calcified nodules. Moreover, the protein levels of Wnt3a and active β-catenin were determined and it was found that GALNT3 significantly inhibited their expression. LiCl, a Wnt/β-catenin signaling activator, was observed to reverse the protective effect of GALNT3 overexpression. The opposite results were observed in the GALNT3 knockdown cells. In vivo, overexpression of GALNT3 by adeno-associated virus decreased the serum Pi and slowed the formation of aortic calcification in the calcified mice. In conclusion, our results indicate that GALNT3 counteracts high Pi-induced osteoblastic differentiation of VSMCs and protects against the initiation and progression of VC by inhibiting the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Liwei Guo
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang, Henan, China.
| | - Yikai Wang
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang, Henan, China
| | - Shijie Li
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang, Henan, China
| | - Lulu Zhou
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang, Henan, China
| | - Duan Li
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
26
|
Repression of the antiporter SLC7A11/glutathione/glutathione peroxidase 4 axis drives ferroptosis of vascular smooth muscle cells to facilitate vascular calcification. Kidney Int 2022; 102:1259-1275. [PMID: 36063875 DOI: 10.1016/j.kint.2022.07.034] [Citation(s) in RCA: 184] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 06/13/2022] [Accepted: 07/27/2022] [Indexed: 01/12/2023]
Abstract
Vascular calcification is a common pathologic condition in patients with chronic kidney disease (CKD). Cell death such as apoptosis plays a critical role in vascular calcification. Ferroptosis is a type of iron-catalyzed and regulated cell death resulting from excessive iron-dependent reactive oxygen species and lipid peroxidation. However, it is unclear whether ferroptosis of vascular smooth muscle cells (VSMCs) regulates vascular calcification in CKD. Our results showed that high calcium and phosphate concentrations induced ferroptosis in rat VSMCs in vitro. Inhibition of ferroptosis by ferrostatin-1 dose-dependently reduced mineral deposition in rat VSMCs under pro-osteogenic conditions, as indicated by alizarin red staining and quantification of calcium content. In addition, gene expression analysis revealed that ferrostatin-1 inhibited osteogenic differentiation of rat VSMCs. Similarly, ferrostatin-1 remarkably attenuated calcification of rat and human arterial rings ex vivo and aortic calcification in vitamin D3-overloaded mice in vivo. Moreover, inhibition of ferroptosis by either ferrostatin-1 or deferoxamine attenuated aortic calcification in rats with CKD. Mechanistically, high calcium and phosphate downregulated expression of SLC7A11 (a cystine-glutamate antiporter), and reduced glutathione (GSH) content in VSMCs. Additionally, GSH depletion induced by erastin (a small molecule initiating ferroptotic cell death) significantly promoted calcification of VSMCs under pro-osteogenic conditions, whereas GSH supplement by N-acetylcysteine reduced calcification of VSMCs. Consistently, knockdown of SLC7A11 by siRNA markedly promoted VSMC calcification. Furthermore, high calcium and phosphate downregulated glutathione peroxidase 4 (GPX4) expression, and reduced glutathione peroxidase activity. Inhibition of GPX4 by RSL3 promoted VSMC calcification. Thus, repression of the SLC7A11/GSH/GPX4 axis triggers ferroptosis of VSMCs to promote vascular calcification under CKD conditions, providing a novel targeting strategy for vascular calcification.
Collapse
|
27
|
Tang Y, Xie J, Chen X, Sun L, Xu L, Chen X. A novel link between silent information regulator 1 and autophagy in cerebral ischemia-reperfusion. Front Neurosci 2022; 16:1040182. [PMID: 36507335 PMCID: PMC9726917 DOI: 10.3389/fnins.2022.1040182] [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: 09/09/2022] [Accepted: 11/03/2022] [Indexed: 11/24/2022] Open
Abstract
Cerebral ischemia is one of the leading causes of death and disability worldwide. Although revascularization via reperfusion combined with advanced anticoagulant therapy is currently a gold standard treatment for patients, the reperfusion itself also results in a serious dysfunction termed cerebral ischemia-reperfusion (I/R) injury. Silent information regulator 1 (sirtuin 1, SIRT1), is a classic NAD+-dependent deacetylase, which has been proposed as an important mediator in the alleviation of cerebral ischemia through modulating multiple physiological processes, including apoptosis, inflammation, DNA repair, oxidative stress, and autophagy. Recent growing evidence suggests that SIRT1-mediated autophagy plays a key role in the pathophysiological process of cerebral I/R injury. SIRT1 could both activate and inhibit the autophagy process by mediating different autophagy pathways, such as the SIRT1-FOXOs pathway, SIRT1-AMPK pathway, and SIRT1-p53 pathway. However, the autophagic roles of SIRT1 in cerebral I/R injury have not been systematically summarized. Here, in this review, we will first introduce the molecular mechanisms and effects of SIRT1 in cerebral ischemia and I/R injury. Next, we will discuss the involvement of autophagy in the pathogenesis of cerebral I/R injury. Finally, we will summarize the latest advances in the interaction between SIRT1 and autophagy in cerebral I/R injury. A good understanding of these relationships would serve to consolidate a framework of mechanisms underlying SIRT1's neuroprotective effects and provides evidence for the development of drugs targeting SIRT1.
Collapse
|
28
|
Phadwal K, Koo E, Jones RA, Forsythe RO, Tang K, Tang Q, Corcoran BM, Caporali A, MacRae VE. Metformin protects against vascular calcification through the selective degradation of Runx2 by the p62 autophagy receptor. J Cell Physiol 2022; 237:4303-4316. [PMID: 36166694 DOI: 10.1002/jcp.30887] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 09/04/2022] [Accepted: 09/06/2022] [Indexed: 11/09/2022]
Abstract
Vascular calcification is associated with aging, type 2 diabetes, and atherosclerosis, and increases the risk of cardiovascular morbidity and mortality. It is an active, highly regulated process that resembles physiological bone formation. It has previously been established that pharmacological doses of metformin alleviate arterial calcification through adenosine monophosphate-activated protein kinase (AMPK)-activated autophagy, however the specific pathway remains elusive. In the present study we hypothesized that metformin protects against arterial calcification through the direct autophagic degradation of runt-related transcription factor 2 (Runx2). Calcification was blunted in vascular smooth muscle cells (VSMCs) by metformin in a dose-dependent manner (0.5-1.5 mM) compared to control cells (p < 0.01). VSMCs cultured under high-phosphate (Pi) conditions in the presence of metformin (1 mM) showed a significant increase in LC3 puncta following bafilomycin-A1 (Baf-A; 5 nM) treatment compared to control cells (p < 0.001). Furthermore, reduced expression of Runx2 was observed in the nuclei of metformin-treated calcifying VSMCs (p < 0.0001). Evaluation of the functional role of autophagy through Atg3 knockdown in VSMCs showed aggravated Pi-induced calcification (p < 0.0001), failure to induce autophagy (punctate LC3) (p < 0.001) and increased nuclear Runx2 expression (p < 0.0001) in VSMCs cultured under high Pi conditions in the presence of metformin (1 mM). Mechanistic studies employing three-way coimmunoprecipitation with Runx2, p62, and LC3 revealed that p62 binds to both LC3 and Runx2 upon metformin treatment in VSMCs. Furthermore, immunoblotting with LC3 revealed that Runx2 specifically binds with p62 and LC3-II in metformin-treated calcified VSMCs. Lastly, we investigated the importance of the autophagy pathway in vascular calcification in a clinical setting. Ex vivo clinical analyses of calcified diabetic lower limb artery tissues highlighted a negative association between Runx2 and LC3 in the vascular calcification process. These studies suggest that exploitation of metformin and its analogues may represent a novel therapeutic strategy for clinical intervention through the induction of AMPK/Autophagy Related 3 (Atg3)-dependent autophagy and the subsequent p62-mediated autophagic degradation of Runx2.
Collapse
Affiliation(s)
- Kanchan Phadwal
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Eve Koo
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Ross A Jones
- Edinburgh Medical School, The University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neurone Disease Research, The University of Edinburgh, Edinburgh, UK
| | - Rachael O Forsythe
- Centre for Cardiovascular Science, The Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Vascular Surgery, Edinburgh Royal Infirmary, Edinburgh, UK
| | - Keyi Tang
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Qiyu Tang
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Brendan M Corcoran
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Andrea Caporali
- Centre for Cardiovascular Science, The Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Vicky E MacRae
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
29
|
Zhao H, Liu H, Liu Y, Jin J, He Q, Lin B. The role of extracellular vesicles in vascular calcification in chronic kidney disease. Front Med (Lausanne) 2022; 9:997554. [PMID: 36388921 PMCID: PMC9651939 DOI: 10.3389/fmed.2022.997554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/10/2022] [Indexed: 09/08/2024] Open
Abstract
Widespread vascular calcification (VC) in patients with chronic kidney disease (CKD) is the pathological basis for the development of cardiovascular disease, and VC has been identified as an independent risk factor for increased cardiovascular mortality in cases of CKD. While VC was earlier thought to be a passive deposition process following calcium and phosphorus supersaturation, recent studies have suggested that it is an active, modifiable, biological process similar to bone development. The involvement of extracellular vesicles (EVs) in the process of VC has been reported as an important transporter of material transport and intercellular communication. This paper reviews the mechanism of the role of EVs, especially exosomes, in VC and the regulation of VC by stem cell-derived EVs, and discusses the possible and promising application of related therapeutic targets in the clinical setting.
Collapse
Affiliation(s)
- Huan Zhao
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejian, China
| | - Haojie Liu
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejian, China
| | - Yueming Liu
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejian, China
| | - Juan Jin
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejian, China
| | - Qiang He
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Bo Lin
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejian, China
| |
Collapse
|
30
|
Klotho Ameliorates Vascular Calcification via Promoting Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7192507. [PMID: 36338347 PMCID: PMC9629936 DOI: 10.1155/2022/7192507] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 02/05/2023]
Abstract
Vascular calcification (VC) is regarded as a common feature of vascular aging. Klotho deficiency reportedly contributes to VC, which can be ameliorated by restoration of Klotho expression. However, the specific mechanisms involved remain unclear. Here, we investigated the role of autophagy in the process of Klotho-inhibiting VC. The clinical study results indicated that, based on Agatston score, serum Klotho level was negatively associated with aortic calcification. Then, Klotho-deficient mice exhibited aortic VC, which could be alleviated with the supplementation of Klotho protein. Moreover, autophagy increased in the aorta of Klotho-deficient mice and protected against VC. Finally, we found that Klotho ameliorated calcification by promoting autophagy both in the aorta of Klotho-deficient mice and in mouse vascular smooth muscle cells (MOVAS) under calcifying conditions. These findings indicate that Klotho deficiency induces increased autophagy to protect against VC and that Klotho expression further enhances autophagy to ameliorate calcification. This study is beneficial to exploring the underlying mechanisms of Klotho regulating VC, which has important guiding significance for future clinical studies in the treatment of VC.
Collapse
|
31
|
Liu YZ, Li ZX, Zhang LL, Wang D, Liu YP. Phenotypic plasticity of vascular smooth muscle cells in vascular calcification: Role of mitochondria. Front Cardiovasc Med 2022; 9:972836. [PMID: 36312244 PMCID: PMC9597684 DOI: 10.3389/fcvm.2022.972836] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 09/20/2022] [Indexed: 12/02/2022] Open
Abstract
Vascular calcification (VC) is an important hallmark of cardiovascular disease, the osteo-/chondrocyte phenotype differentiation of vascular smooth muscle cells (VSMCs) is the main cause of vascular calcification. Accumulating evidence shows that mitochondrial dysfunction may ultimately be more detrimental in the VSMCs calcification. Mitochondrial participate in essential cellular functions, including energy production, metabolism, redox homeostasis regulation, intracellular calcium homeostasis, apoptosis, and signal transduction. Mitochondrial dysfunction under pathological conditions results in mitochondrial reactive oxygen species (ROS) generation and metabolic disorders, which further lead to abnormal phenotypic differentiation of VSMCs. In this review, we summarize existing studies targeting mitochondria as a treatment for VC, and focus on VSMCs, highlighting recent progress in determining the roles of mitochondrial processes in regulating the phenotype transition of VSMCs, including mitochondrial biogenesis, mitochondrial dynamics, mitophagy, mitochondrial energy metabolism, and mitochondria/ER interactions. Along these lines, the impact of mitochondrial homeostasis on VC is discussed.
Collapse
|
32
|
Ceccherini E, Cecchettini A, Gisone I, Persiani E, Morales MA, Vozzi F. Vascular Calcification: In Vitro Models under the Magnifying Glass. Biomedicines 2022; 10:biomedicines10102491. [PMID: 36289753 DOI: 10.3390/biomedicines10102491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular calcification is a systemic disease contributing to cardiovascular morbidity and mortality. The pathophysiology of vascular calcification involves calcium salt deposition by vascular smooth muscle cells that exhibit an osteoblast-like phenotype. Multiple conditions drive the phenotypic switch and calcium deposition in the vascular wall; however, the exact molecular mechanisms and the connection between vascular smooth muscle cells and other cell types are not fully elucidated. In this hazy landscape, effective treatment options are lacking. Due to the pathophysiological complexity, several research models are available to evaluate different aspects of the calcification process. This review gives an overview of the in vitro cell models used so far to study the molecular processes underlying vascular calcification. In addition, relevant natural and synthetic compounds that exerted anticalcifying properties in in vitro systems are discussed.
Collapse
Affiliation(s)
- Elisa Ceccherini
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Antonella Cecchettini
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Ilaria Gisone
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Elisa Persiani
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Maria Aurora Morales
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Federico Vozzi
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| |
Collapse
|
33
|
Jiang H, Li L, Zhang L, Zang G, Sun Z, Wang Z. Role of endothelial cells in vascular calcification. Front Cardiovasc Med 2022; 9:895005. [PMID: 35928939 PMCID: PMC9343736 DOI: 10.3389/fcvm.2022.895005] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular calcification (VC) is active and regulates extraosseous ossification progress, which is an independent predictor of cardiovascular disease (CVD) morbidity and mortality. Endothelial cells (ECs) line the innermost layer of blood vessels and directly respond to changes in flow shear stress and blood composition. Together with vascular smooth muscle cells, ECs maintain vascular homeostasis. Increased evidence shows that ECs have irreplaceable roles in VC due to their high plasticity. Endothelial progenitor cells, oxidative stress, inflammation, autocrine and paracrine functions, mechanotransduction, endothelial-to-mesenchymal transition (EndMT), and other factors prompt ECs to participate in VC. EndMT is a dedifferentiation process by which ECs lose their cell lineage and acquire other cell lineages; this progress coexists in both embryonic development and CVD. EndMT is regulated by several signaling molecules and transcription factors and ultimately mediates VC via osteogenic differentiation. The specific molecular mechanism of EndMT remains unclear. Can EndMT be reversed to treat VC? To address this and other questions, this study reviews the pathogenesis and research progress of VC, expounds the role of ECs in VC, and focuses on the regulatory factors underlying EndMT, with a view to providing new concepts for VC prevention and treatment.
Collapse
Affiliation(s)
- Han Jiang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Guangyao Zang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- *Correspondence: Zhongqun Wang,
| |
Collapse
|
34
|
Zhou H, Wang L, Liu S, Wang W. The role of phosphoinositide 3-kinases in immune-inflammatory responses: potential therapeutic targets for abdominal aortic aneurysm. Cell Cycle 2022; 21:2339-2364. [PMID: 35792922 DOI: 10.1080/15384101.2022.2094577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The pathogenesis of abdominal aortic aneurysm (AAA) includes inflammatory responses, matrix metalloproteinases (MMPs) degradation, VSMC apoptosis, oxidative stress, and angiogenesis, among which the inflammatory response plays a key role. At present, surgery is the only curing treatment, and no effective drug can delay AAA progression in clinical practice. Therefore, searching for a signaling pathway related to the immune-inflammatory response is an essential direction for developing drugs targeting AAA. Recent studies have confirmed that the PI3K family plays an important role in many inflammatory diseases and is involved in regulating various cellular functions, especially in the immune-inflammatory response. This review focuses on the role of each isoform of PI3K in each stage of AAA immune-inflammatory response, making available explorations for a deeper understanding of the mechanism of inflammation and immune response during the formation and development of AAA.
Collapse
Affiliation(s)
- Haiyang Zhou
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shuai Liu
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
35
|
Rao Z, Zheng Y, Xu L, Wang Z, Zhou Y, Chen M, Dong N, Cai Z, Li F. Endoplasmic Reticulum Stress and Pathogenesis of Vascular Calcification. Front Cardiovasc Med 2022; 9:918056. [PMID: 35783850 PMCID: PMC9243238 DOI: 10.3389/fcvm.2022.918056] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/30/2022] [Indexed: 12/05/2022] Open
Abstract
Vascular calcification (VC) is characterized by calcium phosphate deposition in blood vessel walls and is associated with many diseases, as well as increased cardiovascular morbidity and mortality. However, the molecular mechanisms underlying of VC development and pathogenesis are not fully understood, thus impeding the design of molecular-targeted therapy for VC. Recently, several studies have shown that endoplasmic reticulum (ER) stress can exacerbate VC. The ER is an intracellular membranous organelle involved in the synthesis, folding, maturation, and post-translational modification of secretory and transmembrane proteins. ER stress (ERS) occurs when unfolded/misfolded proteins accumulate after a disturbance in the ER environment. Therefore, downregulation of pathological ERS may attenuate VC. This review summarizes the relationship between ERS and VC, focusing on how ERS regulates the development of VC by promoting osteogenic transformation, inflammation, autophagy, and apoptosis, with particular interest in the molecular mechanisms occurring in various vascular cells. We also discuss, the therapeutic effects of ERS inhibition on the progress of diseases associated with VC are detailed.
Collapse
Affiliation(s)
- Zhenqi Rao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yidan Zheng
- Basic Medical School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihao Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhejun Cai
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
36
|
Ren SC, Mao N, Yi S, Ma X, Zou JQ, Tang X, Fan JM. Vascular Calcification in Chronic Kidney Disease: An Update and Perspective. Aging Dis 2022; 13:673-697. [PMID: 35656113 PMCID: PMC9116919 DOI: 10.14336/ad.2021.1024] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/24/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease is a devastating condition resulting from irreversible loss of nephron numbers and function and leading to end-stage renal disease and mineral disorders. Vascular calcification, an ectopic deposition of calcium-phosphate salts in blood vessel walls and heart valves, is an independent risk factor of cardiovascular morbidity and mortality in chronic kidney disease. Moreover, aging and related metabolic disorders are essential risk factors for chronic kidney disease and vascular calcification. Marked progress has been recently made in understanding and treating vascular calcification in chronic kidney disease. However, there is a paucity of systematic reviews summarizing this progress, and investigating unresolved issues is warranted. In this systematic review, we aimed to overview the underlying mechanisms of vascular calcification in chronic kidney diseases and discuss the impact of chronic kidney disease on the pathophysiology of vascular calcification. Additionally, we summarized potential clinical diagnostic biomarkers and therapeutic applications for vascular calcification with chronic kidney disease. This review may offer new insights into the pathogenesis, diagnosis, and therapeutic intervention of vascular calcification.
Collapse
Affiliation(s)
- Si-Chong Ren
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
- Center for Translational Medicine, Sichuan Academy of Traditional Chinese Medicine, Chengdu, China.
| | - Nan Mao
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Si Yi
- Chengdu Medical College, Chengdu, China.
- Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, China.
| | - Xin Ma
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Jia-Qiong Zou
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jun-Ming Fan
- Chengdu Medical College, Chengdu, China.
- Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, China.
| |
Collapse
|
37
|
Burtenshaw D, Regan B, Owen K, Collins D, McEneaney D, Megson IL, Redmond EM, Cahill PA. Exosomal Composition, Biogenesis and Profiling Using Point-of-Care Diagnostics—Implications for Cardiovascular Disease. Front Cell Dev Biol 2022; 10:853451. [PMID: 35721503 PMCID: PMC9198276 DOI: 10.3389/fcell.2022.853451] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
Arteriosclerosis is an important age-dependent disease that encompasses atherosclerosis, in-stent restenosis (ISR), pulmonary hypertension, autologous bypass grafting and transplant arteriosclerosis. Endothelial dysfunction and the proliferation of vascular smooth muscle cell (vSMC)-like cells is a critical event in the pathology of arteriosclerotic disease leading to intimal-medial thickening (IMT), lipid retention and vessel remodelling. An important aspect in guiding clinical decision-making is the detection of biomarkers of subclinical arteriosclerosis and early cardiovascular risk. Crucially, relevant biomarkers need to be good indicators of injury which change in their circulating concentrations or structure, signalling functional disturbances. Extracellular vesicles (EVs) are nanosized membraneous vesicles secreted by cells that contain numerous bioactive molecules and act as a means of intercellular communication between different cell populations to maintain tissue homeostasis, gene regulation in recipient cells and the adaptive response to stress. This review will focus on the emerging field of EV research in cardiovascular disease (CVD) and discuss how key EV signatures in liquid biopsies may act as early pathological indicators of adaptive lesion formation and arteriosclerotic disease progression. EV profiling has the potential to provide important clinical information to complement current cardiovascular diagnostic platforms that indicate or predict myocardial injury. Finally, the development of fitting devices to enable rapid and/or high-throughput exosomal analysis that require adapted processing procedures will be evaluated.
Collapse
Affiliation(s)
- Denise Burtenshaw
- Vascular Biology and Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Brian Regan
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Kathryn Owen
- Southern Health and Social Care Trust, Craigavon Area Hospital, Craigavon, United Kingdom
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), Ulster University, Belfast, United Kingdom
| | - David Collins
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - David McEneaney
- Southern Health and Social Care Trust, Craigavon Area Hospital, Craigavon, United Kingdom
| | - Ian L. Megson
- Division of Biomedical Sciences, Centre for Health Science, UHI Institute of Health Research and Innovation, Inverness, United Kingdom
| | - Eileen M. Redmond
- Department of Surgery, University of Rochester, Rochester, NY, United States
| | - Paul Aidan Cahill
- Vascular Biology and Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
- *Correspondence: Paul Aidan Cahill,
| |
Collapse
|
38
|
Li M, Wang ZW, Fang LJ, Cheng SQ, Wang X, Liu NF. Programmed cell death in atherosclerosis and vascular calcification. Cell Death Dis 2022; 13:467. [PMID: 35585052 PMCID: PMC9117271 DOI: 10.1038/s41419-022-04923-5] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 04/30/2022] [Accepted: 05/06/2022] [Indexed: 12/14/2022]
Abstract
The concept of cell death has been expanded beyond apoptosis and necrosis to additional forms, including necroptosis, pyroptosis, autophagy, and ferroptosis. These cell death modalities play a critical role in all aspects of life, which are noteworthy for their diverse roles in diseases. Atherosclerosis (AS) and vascular calcification (VC) are major causes for the high morbidity and mortality of cardiovascular disease. Despite considerable advances in understanding the signaling pathways associated with AS and VC, the exact molecular basis remains obscure. In the article, we review the molecular mechanisms that mediate cell death and its implications for AS and VC. A better understanding of the mechanisms underlying cell death in AS and VC may drive the development of promising therapeutic strategies.
Collapse
Affiliation(s)
- Min Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Zhen-Wei Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Li-Juan Fang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Shou-Quan Cheng
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Xin Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China.
| |
Collapse
|
39
|
Qiao Y. Reactive Oxygen Species in Cardiovascular Calcification: Role of Medicinal Plants. Front Pharmacol 2022; 13:858160. [PMID: 35370681 PMCID: PMC8964595 DOI: 10.3389/fphar.2022.858160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/25/2022] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular calcification, including vascular calcification and calcific aortic valve disease (CAVD), is a serious worldwide health problem, especially in older adults. The mechanisms underlying cardiovascular calcifications are complex and multifactorial. An increase in reactive oxygen species (ROS) and oxidative stress play important roles in the initiation and development of cardiovascular calcification. This mini-review summarizes the recent evidence that supports the association of ROS with vascular calcification and CAVD and discusses the role of medicinal plants for the prevention and treatment of cardiovascular calcification.
Collapse
Affiliation(s)
- Yu Qiao
- King's College London, London, United Kingdom
| |
Collapse
|
40
|
Irisin protects against vascular calcification by activating autophagy and inhibiting NLRP3-mediated vascular smooth muscle cell pyroptosis in chronic kidney disease. Cell Death Dis 2022; 13:283. [PMID: 35354793 PMCID: PMC8967887 DOI: 10.1038/s41419-022-04735-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 02/24/2022] [Accepted: 03/15/2022] [Indexed: 01/18/2023]
Abstract
Irisin protects the cardiovascular system against vascular diseases. However, its role in chronic kidney disease (CKD) -associated vascular calcification (VC) and the underlying mechanisms remain unclear. In the present study, we investigated the potential link among Irisin, pyroptosis, and VC under CKD conditions. During mouse vascular smooth muscle cell (VSMC) calcification induced by β-glycerophosphate (β-GP), the pyroptosis level was increased, as evidenced by the upregulated expression of pyroptosis-related proteins (cleaved CASP1, GSDMD-N, and IL1B) and pyroptotic cell death (increased numbers of PI-positive cells and LDH release). Reducing the pyroptosis levels by a CASP1 inhibitor remarkably decreased calcium deposition in β-GP-treated VSMCs. Further experiments revealed that the pyroptosis pathway was activated by excessive reactive oxygen species (ROS) production and subsequent NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in calcified VSMCs. Importantly, Irisin effectively inhibited β-GP-induced calcium deposition in VSMCs in vitro and in mice aortic rings ex vivo. Overexpression of Nlrp3 attenuated the suppressive effect of Irisin on VSMC calcification. In addition, Irisin could induce autophagy and restore autophagic flux in calcified VSMCs. Adding the autophagy inhibitor 3-methyladenine or chloroquine attenuated the inhibitory effect of Irisin on β-GP-induced ROS production, NLRP3 inflammasome activation, pyroptosis, and calcification in VSMCs. Finally, our in vivo study showed that Irisin treatment promoted autophagy, downregulated ROS level and thereby suppressed pyroptosis and medial calcification in aortic tissues of adenine-induced CKD mice. Together, our findings for the first time demonstrated that Irisin protected against VC via inducing autophagy and inhibiting VSMC pyroptosis in CKD, and Irisin might serve as an effective therapeutic agent for CKD-associated VC.
Collapse
|
41
|
Li T, Yu H, Zhang D, Feng T, Miao M, Li J, Liu X. Matrix Vesicles as a Therapeutic Target for Vascular Calcification. Front Cell Dev Biol 2022; 10:825622. [PMID: 35127686 PMCID: PMC8814528 DOI: 10.3389/fcell.2022.825622] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/04/2022] [Indexed: 01/01/2023] Open
Abstract
Vascular calcification (VC) is linked to an increased risk of heart disease, stroke, and atherosclerotic plaque rupture. It is a cell-active process regulated by vascular cells rather than pure passive calcium (Ca) deposition. In recent years, extracellular vesicles (EVs) have attracted extensive attention because of their essential role in the process of VC. Matrix vesicles (MVs), one type of EVs, are especially critical in extracellular matrix mineralization and the early stages of the development of VC. Vascular smooth muscle cells (VSMCs) have the potential to undergo phenotypic transformation and to serve as a nucleation site for hydroxyapatite crystals upon extracellular stimulation. However, it is not clear what underlying mechanism that MVs drive the VSMCs phenotype switching and to result in calcification. This article aims to review the detailed role of MVs in the progression of VC and compare the difference with other major drivers of calcification, including aging, uremia, mechanical stress, oxidative stress, and inflammation. We will also bring attention to the novel findings in the isolation and characterization of MVs, and the therapeutic application of MVs in VC.
Collapse
Affiliation(s)
- Tiantian Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Hongchi Yu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Demao Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Tang Feng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Michael Miao
- Division of Oral & Craniofacial Health Sciences, University of North Carolina Adams School of Dentistry, Chapel Hill, NC, United States
| | - Jianwei Li
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Jianwei Li, ; Xiaoheng Liu,
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Jianwei Li, ; Xiaoheng Liu,
| |
Collapse
|
42
|
Lu H, Du W, Ren L, Hamblin MH, Becker RC, Chen YE, Fan Y. Vascular Smooth Muscle Cells in Aortic Aneurysm: From Genetics to Mechanisms. J Am Heart Assoc 2021; 10:e023601. [PMID: 34796717 PMCID: PMC9075263 DOI: 10.1161/jaha.121.023601] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Aortic aneurysm, including thoracic aortic aneurysm and abdominal aortic aneurysm, is the second most prevalent aortic disease following atherosclerosis, representing the ninth-leading cause of death globally. Open surgery and endovascular procedures are the major treatments for aortic aneurysm. Typically, thoracic aortic aneurysm has a more robust genetic background than abdominal aortic aneurysm. Abdominal aortic aneurysm shares many features with thoracic aortic aneurysm, including loss of vascular smooth muscle cells (VSMCs), extracellular matrix degradation and inflammation. Although there are limitations to perfectly recapitulating all features of human aortic aneurysm, experimental models provide valuable tools to understand the molecular mechanisms and test novel therapies before human clinical trials. Among the cell types involved in aortic aneurysm development, VSMC dysfunction correlates with loss of aortic wall structural integrity. Here, we discuss the role of VSMCs in aortic aneurysm development. The loss of VSMCs, VSMC phenotypic switching, secretion of inflammatory cytokines, increased matrix metalloproteinase activity, elevated reactive oxygen species, defective autophagy, and increased senescence contribute to aortic aneurysm development. Further studies on aortic aneurysm pathogenesis and elucidation of the underlying signaling pathways are necessary to identify more novel targets for treating this prevalent and clinical impactful disease.
Collapse
Affiliation(s)
- Haocheng Lu
- Department of Internal MedicineCardiovascular CenterUniversity of Michigan Medical CenterAnn ArborMI
| | - Wa Du
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Lu Ren
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Milton H. Hamblin
- Department of PharmacologyTulane University School of MedicineNew OrleansLA
| | - Richard C. Becker
- Division of Cardiovascular Health and DiseaseDepartment of Internal MedicineUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Y. Eugene Chen
- Department of Internal MedicineCardiovascular CenterUniversity of Michigan Medical CenterAnn ArborMI
| | - Yanbo Fan
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOH
- Division of Cardiovascular Health and DiseaseDepartment of Internal MedicineUniversity of Cincinnati College of MedicineCincinnatiOH
| |
Collapse
|
43
|
Hyperphosphatemia-induced degradation of transcription factor EB exacerbates vascular calcification. Biochim Biophys Acta Mol Basis Dis 2021; 1868:166323. [PMID: 34921974 DOI: 10.1016/j.bbadis.2021.166323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/16/2021] [Accepted: 12/08/2021] [Indexed: 11/23/2022]
Abstract
AIMS Chronic kidney disease (CKD) and subsequent hyperphosphatemia causes vascular calcification (VC), a strong predictor of mortality. Dysregulation of the autophagy-lysosomal pathway in vascular smooth muscle cells (VSMCs) mediates hyperphosphatemia-dependent VC. However, the process through which lysosomes become dysfunctional remains unknown. Transcription factor EB (TFEB) is a master regulator of lysosome biogenesis. The present study examined the hypothesis that TFEB dysfunction causes VC progression. METHODS AND RESULTS Inorganic phosphate (Pi) dose-dependently promoted VC in mouse aorta ex vivo, in rat VSMCs in vitro, and in human aortic smooth muscle cells in vitro, all accompanied by a decrease in TFEB protein. Lysosomal inhibitors or TFEB knockdown using small interfering RNA exacerbated Pi-induced VC in VSMCs. Conversely, TFEB downregulation was not observed in the hypercalcemia-sensitive VC model induced by excessive vitamin D dosages. Feeding rats an adenine-containing diet caused CKD and hyperphosphatemia. VC occurred in the adenine-fed rat aorta and regressed after adenine cessation. In this CKD model, aortic TFEB expression decreased at VC onset but recovered to average levels during recovery from VC after adenine cessation. The calcified area of the CKD rat aorta exhibited lysosomal damage and enhanced TFEB ubiquitination. Hyperphosphatemia in vitro increased insoluble TFEB and decreased soluble TFEB in VSMCs, both of which were abrogated by the proteasome inhibitor, MG-132. CONCLUSION Hyperphosphatemia caused VC via TFEB downregulation in VSMCs. Under hyperphosphatemia, TFEB was insolubilized and degraded via the ubiquitin-proteasome system. Our results suggest a new mechanism for the pathogenesis of VC under CKD and hyperphosphatemia.
Collapse
|
44
|
Kan S, Duan M, Liu Y, Wang C, Xie J. Role of Mitochondria in Physiology of Chondrocytes and Diseases of Osteoarthritis and Rheumatoid Arthritis. Cartilage 2021; 13:1102S-1121S. [PMID: 34894777 PMCID: PMC8804744 DOI: 10.1177/19476035211063858] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PURPOSE OF REVIEW Mitochondria are recognized to be one of the most important organelles in chondrocytes for their role in triphosphate (ATP) generation through aerobic phosphorylation. Mitochondria also participate in many intracellular processes involving modulating reactive oxygen species (ROS), responding to instantaneous hypoxia stress, regulating cytoplasmic transport of calcium ion, and directing mitophagy to maintain the homeostasis of individual chondrocytes. DESIGNS To summarize the specific role of mitochondria in chondrocytes, we screened related papers in PubMed database and the search strategy is ((mitochondria) AND (chondrocyte)) AND (English [Language]). The articles published in the past 5 years were included and 130 papers were studied. RESULTS In recent years, the integrity of mitochondrial structure has been regarded as a prerequisite for normal chondrocyte survival and defect in mitochondrial function has been found in cartilage-related diseases, such as osteoarthritis (OA) and rheumatoid arthritis (RA). However, the understanding of mitochondria in cartilage is still largely limited. The mechanism on how the changes in mitochondrial structure and function directly lead to the occurrence and development of cartilage-related diseases remains to be elusive. CONCLUSION This review aims to summarize the role of mitochondria in chondrocytes under the physiological and pathological changes from ATP generation, calcium homeostasis, redox regulation, mitophagy modulation, mitochondria biogenesis to immune response activation. The enhanced understanding of molecular mechanisms in mitochondria might offer some new cues for cartilage remodeling and pathological intervention.
Collapse
Affiliation(s)
- Shiyi Kan
- State Key Laboratory of Oral Diseases,
West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mengmeng Duan
- State Key Laboratory of Oral Diseases,
West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yang Liu
- State Key Laboratory of Oral Diseases,
West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chunli Wang
- “111” Project Laboratory of
Biomechanics and Tissue Repair, Bioengineering College, Chongqing University,
Chongqing, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases,
West China Hospital of Stomatology, Sichuan University, Chengdu, China,“111” Project Laboratory of
Biomechanics and Tissue Repair, Bioengineering College, Chongqing University,
Chongqing, China,Lab of Bone & Joint Disease, State
Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan
University, Chengdu, China,Jing Xie, Lab of Bone & Joint Disease,
State Key Laboratory of Oral Diseases, West China Hospital of Stomatology,
Sichuan University, Chengdu 610064, Sichuan, China.
| |
Collapse
|
45
|
Shen J, Zhao M, Zhang C, Sun X. IL-1β in atherosclerotic vascular calcification: From bench to bedside. Int J Biol Sci 2021; 17:4353-4364. [PMID: 34803503 PMCID: PMC8579452 DOI: 10.7150/ijbs.66537] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/11/2021] [Indexed: 01/19/2023] Open
Abstract
Atherosclerotic vascular calcification contributes to increased risk of death in patients with cardiovascular diseases. Assessing the type and severity of inflammation is crucial in the treatment of numerous cardiovascular conditions. IL-1β, a potent proinflammatory cytokine, plays diverse roles in the pathogenesis of atherosclerotic vascular calcification. Several large-scale, population cohort trials have shown that the incidence of cardiovascular events is clinically reduced by the administration of anti-IL-1β therapy. Anti-IL-1β therapy might reduce the incidence of cardiovascular events by affecting atherosclerotic vascular calcification, but the mechanism underlying this effect remains unclear. In this review, we summarize current knowledge on the role of IL-1β in atherosclerotic vascular calcification, and describe the latest results reported in clinical trials evaluating anti-IL-1β therapies for the treatment of cardiovascular diseases. This review will aid in improving current understanding of the pathophysiological roles of IL-1β and mechanisms underlying its activity.
Collapse
Affiliation(s)
- Jialing Shen
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Ming Zhao
- Department of Interventional Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Chunxiang Zhang
- Laboratory of Nucleic Acids in Medicine for National high-level talents, Southwest Medical University, Luzhou 646000, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Xiaolei Sun
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Department of Interventional Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Laboratory of Nucleic Acids in Medicine for National high-level talents, Southwest Medical University, Luzhou 646000, China.,School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Research Excellence, Faculty of Life Science and Medicine, King's College London, London SE5 9NU, United Kingdom.,Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, China
| |
Collapse
|
46
|
Tian L, Huang CK, Ding F, Zhang R. Galectin-3 Mediates Thrombin-Induced Vascular Smooth Muscle Cell Migration. Front Cardiovasc Med 2021; 8:686200. [PMID: 34746246 PMCID: PMC8563778 DOI: 10.3389/fcvm.2021.686200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 09/09/2021] [Indexed: 11/25/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) migration is an important step in the progression and development of vulnerable plaques. Thrombin is involved in both physiological and pathological processes of atherosclerosis. Therefore, the elucidation of the mechanisms underlying thrombin-induced VSMC migration is essential for devising effective treatments aimed at the prevention of plaque instability. In this study, we found that thrombin activated MAPK signaling pathways and increased the expression of galectin-3, which was also a well-known factor in atherosclerosis. Knockdown of galectin-3 by specific small interfering RNA (siRNA) blocked thrombin-induced activation of ERK1/2 and p38 MAPK, but not JNK MAPK. Src/FAK phosphorylation was also shown to be activated by thrombin. FAK autophosphorylation at Y397 was most significantly inhibited by galectin-3 siRNA. Galectin-3 siRNA or specific inhibitor (P38 MAPK inhibitor and ERK1/2 inhibitor) effectively prevented thrombin-induced VSMC migration via reducing paxillin expression. These findings demonstrate, for the first time, that thrombin stimulation of VSMC migration and paxillin expression are regulated by galectin-3, and ERK1/2, p38 MAPK, and Src/FAK signaling pathways are involved in this process. These results are beneficial to clarify the role of galectin-3 in thrombin-induced advanced lesions in atherosclerosis and shed new insights into the regulatory mechanism of VSMC migration in combating plaque rupture.
Collapse
Affiliation(s)
- Lei Tian
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun-Kai Huang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fenghua Ding
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
47
|
Liu Q, Luo Y, Zhao Y, Xiang P, Zhu J, Jing W, Jin W, Chen M, Tang R, Yu H. Nano-hydroxyapatite accelerates vascular calcification via lysosome impairment and autophagy dysfunction in smooth muscle cells. Bioact Mater 2021; 8:478-493. [PMID: 34541414 PMCID: PMC8429627 DOI: 10.1016/j.bioactmat.2021.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/20/2021] [Accepted: 06/02/2021] [Indexed: 12/22/2022] Open
Abstract
Vascular calcification (VC) is a common characteristic of aging, diabetes, chronic renal failure, and atherosclerosis. The basic component of VC is hydroxyapatite (HAp). Nano-sized HAp (nHAp) has been identified to play an essential role in the development of pathological calcification of vasculature. However, whether nHAp can induce calcification in vivo and the mechanism of nHAp in the progression of VC remains unclear. We discovered that nHAp existed both in vascular smooth muscle cells (VSMCs) and their extracellular matrix (ECM) in the calcified arteries from patients. Synthetic nHAp had similar morphological and chemical properties as natural nHAp recovered from calcified artery. nHAp stimulated osteogenic differentiation and accelerated mineralization of VSMCs in vitro. Synthetic nHAp could also directly induce VC in vivo. Mechanistically, nHAp was internalized into lysosome, which impaired lysosome vacuolar H+-ATPase for its acidification, therefore blocked autophagic flux in VSMCs. Lysosomal re-acidification by cyclic-3′,5′-adenosine monophosphate (cAMP) significantly enhanced autophagic degradation and attenuated nHAp-induced calcification. The accumulated autophagosomes and autolysosomes were converted into calcium-containing exosomes which were secreted into ECM and accelerated vascular calcium deposit. Inhibition of exosome release in VSMCs decreased calcium deposition. Altogether, our results demonstrated a repressive effect of nHAp on lysosomal acidification, which inhibited autophagic degradation and promoted a conversion of the accumulated autophagic vacuoles into exosomes that were loaded with undissolved nHAp, Ca2+, Pi and ALP. These exosomes bud off the plasma membrane, deposit within ECM, and form calcium nodules. Vascular calcification was thus accelerated by nHAP through blockage of autophagic flux in VSMCs. We first demonstrated that nHAp was internalized into the vascular cell in human calcified aorta. Nano-HAp impairs lysosomal acidification and degradation, and causesblockage of autophagy flux in VSMCs. The accumulated autophagosomes and autolysosomes induced by nHAp in VSMCs are converted into exosomes which promote calcification development.
Collapse
Affiliation(s)
- Qi Liu
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, China
| | - Yi Luo
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, China
| | - Yun Zhao
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, Shandong Province, 266071, China.,Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, China
| | - Pingping Xiang
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, China
| | - Jinyun Zhu
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, China
| | - Wangwei Jing
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, China
| | - Wenjing Jin
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
| | - Mingyao Chen
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
| | - Hong Yu
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310009, China
| |
Collapse
|
48
|
Lin F, Yao X, Kong C, Liu X, Zhao Z, Rao S, Wang L, Li S, Wang J, Dai Q. 25-Hydroxycholesterol protecting from cerebral ischemia-reperfusion injury through the inhibition of STING activity. Aging (Albany NY) 2021; 13:20149-20163. [PMID: 34406977 PMCID: PMC8436919 DOI: 10.18632/aging.203337] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/01/2021] [Indexed: 11/25/2022]
Abstract
Middle cerebral artery occlusion (MCAO) injury refers to impaired blood supply to the brain that is caused by a cerebrovascular disease, resulting in local brain tissue ischemia, hypoxic necrosis, and rapid neurological impairment. Nevertheless, the mechanisms involved are unclear, and pharmacological interventions are lacking. 25-Hydroxycholesterol (25-HC) was reported to be involved in cholesterol and lipid metabolism as an oxysterol molecule. This study aimed to determine whether 25-HC exerts a cerebral protective effect on MCAO injury and investigate its potential mechanism. 25-HC was administered prior to reperfusion in a mouse model of MCAO injury. 25-HC evidently decreased infarct size induced by MCAO and enhanced brain function. It reduced stimulator of interferon gene (STING) activity and regulated mTOR to inhibit autophagy and induce cerebral ischemia tolerance. Thus, 25-HC improved MCAO injury through the STING channel. As indicated in this preliminary study, 25-HC improved MCAO injury by inhibiting STING activity and autophagy as well as by reducing brain nerve cell apoptosis. Thus, it is a potential treatment drug for brain injury.
Collapse
Affiliation(s)
- Feihong Lin
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinyu Yao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chang Kong
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xia Liu
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhangfan Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Suhuan Rao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shan Li
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junlu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qinxue Dai
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
49
|
Different types of cell death in vascular diseases. Mol Biol Rep 2021; 48:4687-4702. [PMID: 34013393 DOI: 10.1007/s11033-021-06402-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/08/2021] [Indexed: 10/21/2022]
Abstract
In a mature organism, tissue homeostasis is regulated by cell division and cell demise as the two major physiological procedures. There is increasing evidence that deregulation of these processes is important in the pathogenicity of main diseases, including myocardial infarction, stroke, atherosclerosis, and inflammatory diseases. Therefore, there are ongoing efforts to discover modulating factors of the cell cycle and cell demise planners aiming at shaping innovative therapeutically modalities to the therapy of such diseases. Although the life of a cell is terminated by several modes of action, a few cell deaths exist-some of which resemble apoptosis and/or necrosis, and most of them are different from one another-that contribute to a wide range of functions to either support or disrupt the homoeostasis. Even in normal physiological conditions, cell life is severe within the cardiovascular system. Cells are persistently undergoing stretch, contraction, injurious metabolic byproducts, and hemodynamic forces, and a few of cells sustain decade-long lifetimes. The duration of vascular disease causes further exposure of vascular cells to a novel range of offences, most of which induce cell death. There is growing evidence on consequences of direct damage to a cell, as well as on responses of adjacent and infiltrating cells, which also have an effect on the pathology. In this study, by focusing on different pathways of cell death in different vascular diseases, an attempt is made to open a new perspective on the therapeutic goals associated with cell death in these diseases.
Collapse
|
50
|
Luo Z, Zeng A, Chen Y, He S, He S, Jin X, Li C, Mei W, Lu Q. Ligustilide inhibited Angiotensin II induced A7r5 cell autophagy via Akt/mTOR signaling pathway. Eur J Pharmacol 2021; 905:174184. [PMID: 34004211 DOI: 10.1016/j.ejphar.2021.174184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022]
Abstract
Autophagy is essential to vessel homeostasis and function in the cardiovascular system. Ligustilide (LIG) is one of the main active ingredients extracted from traditional Chinese medicines, such as Ligusticum chuanxiong, Angelica, and other umbelliferous plants, and reported to have cardiovascular protective effects. In this study, we explore the effects and the potential mechanism of ligustilide on the Ang II-induced autophagy in A7r5 cells. Our results showed that ligustilide inhibited the Ang II-induced autophagy in A7r5 cells and down regulated the expression of autophagy-related proteins LC3, ULK1, and Beclin-1. Ligustilide exerted a protective effect on the reduction of the concentrations of reactive oxygen species and Ca2+ and upregulated the nitric oxide concentration in A7r5 cells with Ang II-induced autophagy. Additionally, the analyses of network pharmacological targets and potential signal pathways indicated that the target of ligustilide to regulate autophagy was related to the Akt/mTOR signaling pathway. Furthermore, ligustilide could upregulate the expression of p-Akt and p-mTOR and inhibit the expression of LC3II in A7r5 cells with Ang II-induced autophagy. These findings showed that ligustilide inhibited the autophagic flux in A7r5 cells induced by Ang II via the activation of the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Zhenhui Luo
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ao Zeng
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuankun Chen
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shumiao He
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Siqing He
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaobao Jin
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chunmei Li
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenjie Mei
- Guangdong Province Engineering and Technology Center for Molecular Probe and Bio-medicine Imaging, Guangzhou, China
| | - Qun Lu
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Province Engineering and Technology Center for Molecular Probe and Bio-medicine Imaging, Guangzhou, China.
| |
Collapse
|