1
|
Borén J, Packard CJ, Binder CJ. Apolipoprotein B-containing lipoproteins in atherogenesis. Nat Rev Cardiol 2025:10.1038/s41569-024-01111-0. [PMID: 39743565 DOI: 10.1038/s41569-024-01111-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Apolipoprotein B (apoB) is the main structural protein of LDLs, triglyceride-rich lipoproteins and lipoprotein(a), and is crucial for their formation, metabolism and atherogenic properties. In this Review, we present insights into the role of apoB-containing lipoproteins in atherogenesis, with an emphasis on the mechanisms leading to plaque initiation and growth. LDL, the most abundant cholesterol-rich lipoprotein in plasma, is causally linked to atherosclerosis. LDL enters the artery wall by transcytosis and, in vulnerable regions, is retained in the subendothelial space by binding to proteoglycans via specific sites on apoB. A maladaptive response ensues. This response involves modification of LDL particles, which promotes LDL retention and the release of bioactive lipid products that trigger inflammatory responses in vascular cells, as well as adaptive immune responses. Resident and recruited macrophages take up modified LDL, leading to foam cell formation and ultimately cell death due to inadequate cellular lipid handling. Accumulation of dead cells and cholesterol crystallization are hallmarks of the necrotic core of atherosclerotic plaques. Other apoB-containing lipoproteins, although less abundant, have substantially greater atherogenicity per particle than LDL. These lipoproteins probably contribute to atherogenesis in a similar way to LDL but might also induce additional pathogenic mechanisms. Several targets for intervention to reduce the rate of atherosclerotic lesion initiation and progression have now been identified, including lowering plasma lipoprotein levels and modulating the maladaptive responses in the artery wall.
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Zhao Y, Cao H, Wei Y, Zheng T. The impact of different degrees of stenosis on platelet deposition in the left anterior descending branch of the coronary artery. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108445. [PMID: 39369586 DOI: 10.1016/j.cmpb.2024.108445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/15/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND AND OBJECTIVE This study aimed to investigate the impact of different stenotic degrees on platelet deposition in the left anterior descending branch of the coronary artery. METHODS The idealized model of coronary artery stenosis of 30 %, 40 %, 50 %, 60 %, 70 % and four patient-specific models of 22.17 %, 34.88 %, 51.23 % and 62.96 % were established. A discrete phase model was used to calculate the deposition of platelet particles in blood. RESULTS (1) As the stenotic degree increased from 30 % to 70 %, the maximum deposition rates were 4.23e-2 kg/(m2 ·s), 3.47e-2 kg/(m2 ·s), 0.14 kg/(m2 ·s), 0.15 kg/(m2 ·s), and 0.38 kg/(m2 ·s), respectively. (2) The greater the stenotic degree, the more points of platelet deposition. (3) Platelets were mainly deposited at the proximal segment of mild stenosis. When the stenotic degree exceeded 50 %, the deposition position moved to the distal segment of the stenosis. (4) The results in the real coronary artery models were similar to those in the idealized model. CONCLUSION The study suggests that the location and number of platelet deposition are related to the degree of stenosis. Moderate to severe stenosis is more likely to spread downstream.
Collapse
Affiliation(s)
- Yiming Zhao
- Sichuan-tibet Railway Co. LTD, Chengdu 610036, China
| | - Haoyao Cao
- Department of Mechanics & Engineering, College Architecture & Environment, Sichuan University, Chengdu 610065, China; Sichuan University Yibin Park / Yibin Institute of Industrial Technology, Yibin 644000, China
| | - Yongtao Wei
- Department of Mechanics & Engineering, College Architecture & Environment, Sichuan University, Chengdu 610065, China.
| | - Tinghui Zheng
- Department of Mechanics & Engineering, College Architecture & Environment, Sichuan University, Chengdu 610065, China; Med-X Center for Informatics, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Jia X, Bai X, Yin Z, Zheng Q, Zhao Y, Lu Y, Shu Y, Wang Y, Zhang Y, Jin S. Siglec-5 as a novel receptor mediates endothelial cells oxLDL transcytosis to promote atherosclerosis. Transl Res 2024; 274:49-66. [PMID: 39341359 DOI: 10.1016/j.trsl.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Excessive subendothelial retention of oxidized low-density lipoprotein (oxLDL) and subsequent oxLDL engulfment by macrophages leads to the formation of foam cells and the development of atherosclerosis. Our previous study showed that the plasma level of sialic acid-binding immunoglobulin-like lectin 5 (Siglec-5) was a novel biomarker for the prognosis of atherosclerosis in diabetic patients. However, the role and underlying mechanisms of Siglec-5 in atherosclerosis have not been elucidated. METHODS The interaction between oxLDL and Siglec-5 was detected by fluorescence colocalization and coimmunoprecipitation. The effect of oxLDL on Siglec-5 expression was detected in endothelial cells and macrophages, and the effect of Siglec-5 on oxLDL transcytosis and uptake was investigated. Siglec-5 was overexpressed in mice using recombinant adeno-associated virus vector serotype 9 (rAAV9-Siglec-5) to evaluate the effect of Siglec-5 on oxLDL uptake and atherogenesis in vivo. In addition, the effects of Siglec-5 antibodies and soluble Siglec-5 proteins on oxLDL transcytosis and uptake and their role in atherogenesis were investigated in vivo and in vitro. RESULTS We found that oxLDL interacted with Siglec-5 and that oxLDL stimulated the expression of Siglec-5. Siglec-5 promotes the transcytosis and uptake of oxLDL, while both anti-Siglec-5 antibodies and soluble Siglec-5 protein attenuated oxLDL transcytosis and uptake. Interestingly, overexpression of Siglec-5 by recombinant adeno-associated viral vector serotype 9 (rAAV9-Siglec-5) promoted the retention of oxLDL in the aorta of C57BL/6 mice. Moreover, overexpression of Siglec-5 significantly accelerated the formation of atherosclerotic lesions in Apoe-/- mice. Moreover, both anti-Siglec-5 antibodies and soluble Siglec-5 protein significantly alleviated the retention of oxLDL in the aorta of rAAV9-Siglec-5-transfected C57BL/6 mice and the formation of atherosclerotic plaques in rAAV9-Siglec-5-transfected Apoe-/- mice. CONCLUSION Our results suggested that Siglec-5 was a novel receptor that mediated oxLDL transcytosis and promoted the formation of foam cells. Interventions that inhibit the interaction between oxLDL and Siglec-5, including anti-Siglec-5 antibody or soluble Siglec-5 protein treatment, may provide novel therapeutic strategies in treating atherosclerosis.
Collapse
MESH Headings
- Lipoproteins, LDL/metabolism
- Animals
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Transcytosis
- Humans
- Mice
- Endothelial Cells/metabolism
- Lectins/metabolism
- Mice, Inbred C57BL
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Human Umbilical Vein Endothelial Cells/metabolism
- Male
- Macrophages/metabolism
- Antigens, Differentiation, Myelomonocytic
Collapse
Affiliation(s)
- Xiong Jia
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China; Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China
| | - Xiangli Bai
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China; Department of Laboratory Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China
| | - Zhiqiang Yin
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Qijun Zheng
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Yin Zhao
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China
| | - Yajing Lu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China
| | - Yan Shu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China
| | - Yayu Wang
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Yifei Zhang
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China.
| |
Collapse
|
4
|
Izquierdo MC, Cabodevilla AG, Basu D, Nasias D, Kanter JE, Ho W, Gjini J, Fisher EA, Kim J, Lee W, Bornfeldt KE, Goldberg IJ. Hyperchylomicronemia causes endothelial cell inflammation and increases atherosclerosis. RESEARCH SQUARE 2024:rs.3.rs-5451391. [PMID: 39649171 PMCID: PMC11623764 DOI: 10.21203/rs.3.rs-5451391/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
The effect of increased triglycerides (TGs) as an independent factor in atherosclerosis development has been contentious, in part, because severe hypertriglyceridemia associates with low levels of low-density lipoprotein cholesterol (LDL-C). To test whether hyperchylomicronemia, in the absence of markedly reduced LDL-C levels, contributes to atherosclerosis, we created mice with induced whole-body lipoprotein lipase (LpL) deficiency combined with LDL receptor (LDLR) deficiency. On an atherogenic Western-type diet (WD), male and female mice with induced global LpL deficiency (iLpl -/-) and LDLR knockdown (Ldlr kd ) developed hypertriglyceridemia and elevated cholesterol levels; all the increased cholesterol was in chylomicrons or large VLDL. After 12 weeks on a WD, atherosclerotic lesions both in the brachiocephalic artery and the aortic root were more severe in iLpl -/- /Ldlr kd mice compared to the control Ldlr kd mice. One likely mechanism for this is that exposure of the aorta to hyperchylomicronemia led to endothelial cell inflammation. Thus, our data show that intact chylomicrons contribute to atherosclerosis, explain the association of postprandial lipemia and vascular disease, and prove that hyperchylomicronemia is not benign.
Collapse
Affiliation(s)
- Maria Concepcion Izquierdo
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Ainara G. Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Dimitris Nasias
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Jenny E. Kanter
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA 98109
| | - Winnie Ho
- Keenan Centre for Biomedical Research, St. Michael’s Hospital and Division of Critical Care, Department of Medicine, University of Toronto, Canada
| | - Jana Gjini
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Edward A. Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Jeffrey Kim
- Comparative Medicine Research Unit, University of Louisville School of Medicine, Louisville, KY
| | - Warren Lee
- Keenan Centre for Biomedical Research, St. Michael’s Hospital and Division of Critical Care, Department of Medicine, University of Toronto, Canada
| | - Karin E. Bornfeldt
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA 98109
| | - Ira J. Goldberg
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
5
|
Wang Y, Xu F, Li G, Cheng C, Yu B, Zhang Z, Kong D, Chen F, Liu Y, Fang Z, Cao L, Yu Y, Gu Y, He Y. Structure of scavenger receptor SCARF1 and its interaction with lipoproteins. eLife 2024; 13:RP93428. [PMID: 39541158 PMCID: PMC11563577 DOI: 10.7554/elife.93428] [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] [Indexed: 11/16/2024] Open
Abstract
SCARF1 (scavenger receptor class F member 1, SREC-1 or SR-F1) is a type I transmembrane protein that recognizes multiple endogenous and exogenous ligands such as modified low-density lipoproteins (LDLs) and is important for maintaining homeostasis and immunity. But the structural information and the mechanisms of ligand recognition of SCARF1 are largely unavailable. Here, we solve the crystal structures of the N-terminal fragments of human SCARF1, which show that SCARF1 forms homodimers and its epidermal growth factor (EGF)-like domains adopt a long-curved conformation. Then, we examine the interactions of SCARF1 with lipoproteins and are able to identify a region on SCARF1 for recognizing modified LDLs. The mutagenesis data show that the positively charged residues in the region are crucial for the interaction of SCARF1 with modified LDLs, which is confirmed by making chimeric molecules of SCARF1 and SCARF2. In addition, teichoic acids, a cell wall polymer expressed on the surface of gram-positive bacteria, are able to inhibit the interactions of modified LDLs with SCARF1, suggesting the ligand binding sites of SCARF1 might be shared for some of its scavenging targets. Overall, these results provide mechanistic insights into SCARF1 and its interactions with the ligands, which are important for understanding its physiological roles in homeostasis and the related diseases.
Collapse
Affiliation(s)
- Yuanyuan Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Fan Xu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Guangyi Li
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of SciencesShanghaiChina
| | - Chen Cheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bowen Yu
- Department of Immunology, School of Basic Medical Sciences, Weifang Medical UniversityWeifangChina
| | - Ze Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Dandan Kong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fabao Chen
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yali Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhen Fang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Longxing Cao
- School of Life Science, Westlake UniversityHangzhouChina
| | - Yang Yu
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of SciencesShanghaiChina
| | - Yijun Gu
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of SciencesShanghaiChina
| | - Yongning He
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical TranslationShanghaiChina
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
6
|
Wang Q, Guo L, Hao D, Ito M, Mineo C, Shaul PW, Li XA. Elevated free cholesterol levels due to impaired reverse cholesterol transport are a risk factor for polymicrobial sepsis in mice. J Biol Chem 2024; 300:107974. [PMID: 39510181 DOI: 10.1016/j.jbc.2024.107974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
Dysregulated lipid metabolism is commonly observed in septic patients, but how it contributes to sepsis remains largely unknown. Reverse cholesterol transport (RCT) is crucial for regulating cholesterol metabolism in circulation. During RCT, high-density lipoprotein collects cholesterol from peripheral tissues and transports it to the liver's scavenger receptor BI (SR-BI), where SR-BI mediates the uptake of cholesteryl esters (CEs) from high-density lipoprotein for excretion via bile. In this study, we utilized AlbCreSR-BIfl/fl mice, a model with impaired RCT, to investigate the impact of RCT on sepsis. We found that AlbCreSR-BIfl/fl mice were significantly more susceptible to cecal ligation and puncture (CLP)-induced polymicrobial sepsis, with a survival rate of 14.3% compared to 80% in SR-BIfl/fl littermates. Mechanistically, sepsis disrupted cholesterol metabolism, causing a 4.8-fold increase in free cholesterol (FC) levels and a 4-fold increase in the FC/CE ratio in AlbCreSR-BIfl/fl mice compared to SR-BIfl/fl littermates. This disruption led to hemolysis and death. Notably, administering the cholesterol-lowering drug probucol normalized FC levels and the FC/CE ratio, and significantly improved survival in CLP-AlbCreSR-BIfl/fl mice. However, probucol treatment reduced survival in CLP-low-density lipoprotein receptor knockout mice, which had elevated CE levels with a low FC/CE ratio. These results highlight that elevated FC levels with high FC/CE ratio are a risk factor for sepsis. Therefore, selectively targeting elevated FC levels and FC/CE ratio could be a promising therapeutic strategy for managing sepsis.
Collapse
Affiliation(s)
- Qian Wang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - Ling Guo
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - Dan Hao
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - Misa Ito
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - Chieko Mineo
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Philip W Shaul
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xiang-An Li
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, USA; Lexington VA Healthcare System, Lexington, Kentucky, USA; Department of Physiology, University of Kentucky, Lexington, Kentucky, USA.
| |
Collapse
|
7
|
Potapenko A, Frey K, Schlumpf E, Robert J, Wollscheid B, von Eckardstein A, Rohrer L. The two major splice variants of scavenger receptor BI differ by their interactions with lipoproteins and cellular localization in endothelial cells. J Lipid Res 2024; 65:100665. [PMID: 39393447 PMCID: PMC11585690 DOI: 10.1016/j.jlr.2024.100665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/13/2024] Open
Abstract
The scavenger receptor BI (SR-BI) facilitates the transport of both HDL and LDL through endothelial cells. Its two splice variants, SR-BIvar1 and SR-BIvar2, differ in their carboxy terminal domains. Only SR-BIvar1 contains the putative binding sites for the adapter proteins PDZ domain containing protein 1 (PDZK1) and dedicator of cytokinesis 4 (DOCK4), which limit the cell surface abundance and internalization of the receptor. To investigate the cellular localization of the SR-BI variants and their interaction with lipoproteins in endothelial cells, EA.hy926 cells were stably transfected with vectors encoding untagged, GFP- or mCherry-tagged constructs of the two SR-BI variants. Additionally, the cells were transfected with shRNAs against PDZK1 or DOCK4. Microscopy investigation showed that SR-BIvar1 was predominantly localized on the cell surface together with clathrin whereas SR-BIvar2 was absent from the cell surface but retrieved in endosomes and lysosomes. Accordingly, only SR-BIvar1 increased lipoprotein binding to endothelial while HDL and LDL uptake were enhanced by both variants. Silencing of PDZK1 or DOCK4 only reduced HDL association in SR-BIvar2 overexpressing cells while LDL association was reduced both in WT and SR-BIvar2 overexpressing cells. In conclusion, either SR-BI variant facilitates the uptake of HDL and LDL into endothelial cells, however by different mechanisms and trafficking routes. This dual role may explain why the loss of DOCK4 or PDZK1 differently affects the uptake of HDL and LDL in different endothelial cells.
Collapse
Affiliation(s)
- Anton Potapenko
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Kathrin Frey
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland; Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
| | - Eveline Schlumpf
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Jérôme Robert
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Bernd Wollscheid
- Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland.
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Davidson WS, Vaisar T, Heinecke JW, Bornfeldt KE. Distinct roles of size-defined HDL subpopulations in cardiovascular disease. Curr Opin Lipidol 2024:00041433-990000000-00093. [PMID: 39450930 DOI: 10.1097/mol.0000000000000959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
PURPOSE OF REVIEW Doubts about whether high-density lipoprotein-cholesterol (HDL-C) levels are causally related to atherosclerotic cardiovascular disease (CVD) risk have stimulated research on identifying HDL-related metrics that might better reflect its cardioprotective functions. HDL is made up of different types of particles that vary in size, protein and lipid composition, and function. This review focuses on recent findings on the specific roles of HDL subpopulations defined by size in CVD. RECENT FINDINGS Small HDL particles are more effective than larger particles at promoting cellular cholesterol efflux because apolipoprotein A-I on their surface better engages ABCA1 (ATP binding cassette subfamily A member 1). In contrast, large HDL particles bind more effectively to scavenger receptor class B type 1 on endothelial cells, which helps prevent LDL from moving into the artery wall. The specific role of medium-sized HDL particles, the most abundant subpopulation, is still unclear. SUMMARY HDL is made up of subpopulations of different sizes of particles, with selective functional roles for small and large HDLs. The function of HDL may depend more on the size and composition of its subpopulations than on HDL-C levels. Further research is required to understand how these different HDL subpopulations influence the development of CVD.
Collapse
Affiliation(s)
- W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Tomas Vaisar
- Deaprtment of Medicine, University of Washington School of Medicine
- University of Washington Medicine Diabetes Institute
| | - Jay W Heinecke
- Deaprtment of Medicine, University of Washington School of Medicine
- University of Washington Medicine Diabetes Institute
| | - Karin E Bornfeldt
- Deaprtment of Medicine, University of Washington School of Medicine
- University of Washington Medicine Diabetes Institute
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
9
|
Park SH, Kang MK, Kim DY, Lim SS, Kang YH. Dietary ellagic acid blocks inflammation-associated atherosclerotic plaque formation in cholesterol-fed apoE-deficient mice. Nutr Res Pract 2024; 18:617-632. [PMID: 39398881 PMCID: PMC11464280 DOI: 10.4162/nrp.2024.18.5.617] [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: 03/19/2024] [Revised: 05/09/2024] [Accepted: 07/05/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND/OBJECTIVES Atherosclerosis particularly due to high circulating level of low-density lipoprotein is a major cause of cardiovascular diseases. Ellagic acid is a natural polyphenolic compound rich in pomegranates and berries. Our previous study showed that ellagic acid improved functionality of reverse cholesterol transport in murine model of atherosclerosis. The aim of this study is to investigate whether ellagic acid inhibited inflammation-associated atherosclerotic plaque formation in cholesterol-fed apolipoprotein E (apoE)-knockout (KO) mice. MATERIALS/METHODS Wild type mice and apoE-KO mice were fed a cholesterol-rich Paigen diet for 10 weeks to induce severe atherosclerosis. Concurrently, 10 mg/kg ellagic acid was orally administered to the apoE-KO mice. Plaque lesion formation and lipid deposition were examined by staining with hematoxylin and eosin, Sudan IV and oil red O. RESULTS The plasma leukocyte profile of cholesterol-fed mice was not altered by apoE deficiency. Oral administration of ellagic acid attenuated plaque lesion formation and lipid deposition in the aorta tree of apoE-KO mice. Ellagic acid substantially reduced plasma levels of soluble vascular cell adhesion molecule and interferon-γ in Paigen diet-fed apoE-KO mice. When 10 mg/kg ellagic acid was administered to cholesterol-fed apoE-KO mice, the levels of CD68 and MCP-1 were strongly reduced in aorta vessels. The protein expression level of nitric oxide synthase-2 (NOS2) in the aorta was highly enhanced by supplementation of ellagic acid to apoE-KO mice, but the expression level of heme oxygenase-1 (HO-1) in the aorta was reduced. Furthermore, ellagic acid diminished the increased aorta expression of the inflammatory adhesion molecules in cholesterol-fed apoE-KO mice. The treatment of ellagic acid inhibited the scavenger receptor-B1 expression in the aorta of apoE-KO mice, while the cholesterol efflux-related transporters were not significantly changed. CONCLUSION These results suggest that ellagic acid may be an atheroprotective compound by attenuating apoE deficiency-induced vascular inflammation and reducing atherosclerotic plaque lesion formation.
Collapse
Affiliation(s)
- Sin-Hye Park
- Department of Food Science and Nutrition and Korean Institute of Nutrition, Hallym University, Chuncheon 24252, Korea
| | - Min-Kyung Kang
- Department of Food and Nutrition, Andong National University, Andong 36729, Korea
| | - Dong Yeon Kim
- Department of Food and Nutrition, Andong National University, Andong 36729, Korea
| | - Soon Sung Lim
- Department of Food Science and Nutrition and Korean Institute of Nutrition, Hallym University, Chuncheon 24252, Korea
| | - Young-Hee Kang
- Department of Food Science and Nutrition and Korean Institute of Nutrition, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
10
|
Kim JD, Jain A, Fang L. Mitigating Vascular Inflammation by Mimicking AIBP Mechanisms: A New Therapeutic End for Atherosclerotic Cardiovascular Disease. Int J Mol Sci 2024; 25:10314. [PMID: 39408645 PMCID: PMC11477018 DOI: 10.3390/ijms251910314] [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: 08/14/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Atherosclerosis, characterized by the accumulation of lipoproteins and lipids within the vascular wall, underlies a heart attack, stroke, and peripheral artery disease. Endothelial inflammation is the primary component driving atherosclerosis, promoting leukocyte adhesion molecule expression (e.g., E-selectin), inducing chemokine secretion, reducing the production of nitric oxide (NO), and enhancing the thrombogenic potential. While current therapies, such as statins, colchicine, anti-IL1β, and sodium-glucose cotransporter 2 (SGLT2) inhibitors, target systemic inflammation, none of them addresses endothelial cell (EC) inflammation, a critical contributor to disease progression. Targeting endothelial inflammation is clinically significant because it can mitigate the root cause of atherosclerosis, potentially preventing disease progression, while reducing the side effects associated with broader anti-inflammatory treatments. Recent studies highlight the potential of the APOA1 binding protein (AIBP) to reduce systemic inflammation in mice. Furthermore, its mechanism of action also guides the design of a potential targeted therapy against a particular inflammatory signaling pathway. This review discusses the unique advantages of repressing vascular inflammation or enhancing vascular quiescence and the associated benefits of reducing thrombosis. This approach offers a promising avenue for more effective and targeted interventions to improve patient outcomes.
Collapse
Affiliation(s)
- Jun-Dae Kim
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA;
| | - Longhou Fang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
- Weill Cornell Medical College, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
11
|
Kumar M, Ali W, Yadav K, Kaumri S, Mishra S, Nardi P, Iellamo F, Bernardini S, Pradhan A, Perrone MA. High-Density Lipoprotein-Associated Paraoxonase-1 (PON-1) and Scavenger Receptor Class B Type 1 (SRB-1) in Coronary Artery Disease: Correlation with Disease Severity. J Clin Med 2024; 13:5480. [PMID: 39336967 PMCID: PMC11432482 DOI: 10.3390/jcm13185480] [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: 08/16/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Coronary artery disease (CAD) is the leading cause of death worldwide. High-Density lipoprotein (HDL) is a well-established marker associated with CAD. The current research goes beyond the conventional HDL-C measurement in previous studies and dives into the functional intricacies of HDL. By understanding how HDL works, rather than just how much of it exists, we can better tailor diagnostic and therapeutic strategies for CAD and related conditions. Hence, the current study quantifies the serum levels of two novel HDL-associated markers, Paraoxonase-1 (PON-1) and Scavenger Receptor Class B Type 1 (SRB-1), in CAD cases vs. controls. Methods: A total of 92 subjects, including 69 CAD and 23 healthy controls, were included, based on the prevalence of the disease. Further, based on the severity of the disease, CAD cases were subcategorized as CAD-I, -II, and -III. Serum PON-1 and SRB-1 levels were measured and compared between patient and control groups. Results: The levels of PON-1 and SRB-1 (32.6 ng/mL and 12.49 ng/mL) were significantly lower in CAD patients vs. the healthy control, at 60.36 ng/mL and 15.85 ng/mL, respectively (p < 0.000). A further intergroup comparison showed a statistically significant difference between the CAT-I and -III for PON-1 (p < 0.025), the CAT-I and -III, and CAT-II and -III for SRB-1 (p < 0.000). The receiver operating characteristics (ROC) curve showed cutoff values of 48.20 ng/mL and 14.90 ng/mL for PON-1 and SRB-1. Conclusions: The current study found that serum levels of HDL-associated PON-1 and SRB-1 are significantly lower in CAD cases, and were also inversely related to the increasing severity of coronary artery disease. This inference implies that serum PON-1 and SRB-1 could be used as non-invasive tools for the identification of coronary atherosclerosis and risk assessment in CAD cases.
Collapse
Affiliation(s)
- Manish Kumar
- Department of Pathology, King George Medical University, Lucknow 226003, Uttar Pradesh, India; (M.K.); (S.K.)
| | - Wahid Ali
- Department of Pathology, King George Medical University, Lucknow 226003, Uttar Pradesh, India; (M.K.); (S.K.)
| | - Kusum Yadav
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226003, Uttar Pradesh, India; (K.Y.); (S.M.)
| | - Swati Kaumri
- Department of Pathology, King George Medical University, Lucknow 226003, Uttar Pradesh, India; (M.K.); (S.K.)
| | - Sridhar Mishra
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226003, Uttar Pradesh, India; (K.Y.); (S.M.)
| | - Paolo Nardi
- Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Ferdinando Iellamo
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (F.I.); (M.A.P.)
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Akshyaya Pradhan
- Lari Cardiology Center, King George Medical University, Lucknow 226003, Uttar Pradesh, India;
| | - Marco Alfonso Perrone
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (F.I.); (M.A.P.)
| |
Collapse
|
12
|
Jang E, Ho TWW, Brumell JH, Lefebvre F, Wang C, Lee WL. IL-1β Induces LDL Transcytosis by a Novel Pathway Involving LDLR and Rab27a. Arterioscler Thromb Vasc Biol 2024; 44:2053-2068. [PMID: 38989581 DOI: 10.1161/atvbaha.124.320940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND In early atherosclerosis, circulating LDLs (low-density lipoproteins) traverse individual endothelial cells by an active process termed transcytosis. The CANTOS trial (Canakinumab Antiinflammatory Thrombosis Outcome Study) treated advanced atherosclerosis using a blocking antibody for IL-1β (interleukin-1β); this significantly reduced cardiovascular events. However, whether IL-1β regulates early disease, particularly LDL transcytosis, remains unknown. METHODS We used total internal reflection fluorescence microscopy to quantify transcytosis by human coronary artery endothelial cells exposed to IL-1β. To investigate transcytosis in vivo, we injected wild-type and knockout mice with IL-1β and LDL to visualize acute LDL deposition in the aortic arch. RESULTS Exposure to picomolar concentrations of IL-1β induced transcytosis of LDL but not of albumin by human coronary artery endothelial cells. Surprisingly, expression of the 2 known receptors for LDL transcytosis, ALK-1 (activin receptor-like kinase-1) and SR-BI (scavenger receptor BI), was unchanged or decreased. Instead, IL-1β increased the expression of the LDLR (LDL receptor); this was unexpected because LDLR is not required for LDL transcytosis. Overexpression of LDLR had no effect on basal LDL transcytosis. However, knockdown of LDLR abrogated the effect of IL-1β on transcytosis rates while the depletion of Cav-1 (caveolin-1) did not. Since LDLR was necessary but overexpression had no effect, we reasoned that another player must be involved. Using public RNA sequencing data to curate a list of Rab (Ras-associated binding) GTPases affected by IL-1β, we identified Rab27a. Overexpression of Rab27a alone had no effect on basal transcytosis, but its knockdown prevented induction by IL-1β. This was phenocopied by depletion of the Rab27a effector JFC1 (synaptotagmin-like protein 1). In vivo, IL-1β increased LDL transcytosis in the aortic arch of wild-type but not Ldlr-/- or Rab27a-deficient mice. The JFC1 inhibitor nexinhib20 also blocked IL-1β-induced LDL accumulation in the aorta. CONCLUSIONS IL-1β induces LDL transcytosis by a distinct pathway requiring LDLR and Rab27a; this route differs from basal transcytosis. We speculate that induction of transcytosis by IL-1β may contribute to the acceleration of early disease.
Collapse
Affiliation(s)
- Erika Jang
- Keenan Centre for Biomedical Research, St. Michael's Hospital, Toronto, ON, Canada (E.J., T.W.W.H., C.W., W.L.L.)
- Department of Laboratory Medicine and Pathobiology (E.J., T.W.W.H., W.L.L.), University of Toronto, ON, Canada
| | - Tse Wing Winnie Ho
- Keenan Centre for Biomedical Research, St. Michael's Hospital, Toronto, ON, Canada (E.J., T.W.W.H., C.W., W.L.L.)
- Department of Laboratory Medicine and Pathobiology (E.J., T.W.W.H., W.L.L.), University of Toronto, ON, Canada
| | - John H Brumell
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada (J.H.B.)
| | - François Lefebvre
- Canadian Centre for Computational Genomics, McGill University, Montreal, QC, Canada (F.L.)
| | - Changsen Wang
- Keenan Centre for Biomedical Research, St. Michael's Hospital, Toronto, ON, Canada (E.J., T.W.W.H., C.W., W.L.L.)
| | - Warren L Lee
- Keenan Centre for Biomedical Research, St. Michael's Hospital, Toronto, ON, Canada (E.J., T.W.W.H., C.W., W.L.L.)
- Department of Laboratory Medicine and Pathobiology (E.J., T.W.W.H., W.L.L.), University of Toronto, ON, Canada
- Department of Biochemistry (W.L.L.), University of Toronto, ON, Canada
- Department of Medicine and the Interdepartmental Division of Critical Care Medicine (W.L.L.), University of Toronto, ON, Canada
| |
Collapse
|
13
|
Piao J, Su Z, He J, Zhu T, Fan F, Wang X, Yang Z, Zhan H, Luo D. SphK1 deficiency ameliorates the development of atherosclerosis by inhibiting the S1P/S1PR3/Rhoa/ROCK pathway. Cell Signal 2024; 121:111252. [PMID: 38852936 DOI: 10.1016/j.cellsig.2024.111252] [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: 03/15/2024] [Revised: 05/14/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND AND AIMS S1P is an important factor regulating the function of the vascular endothelial barrier. SphK1 is an important limiting enzyme for the synthesis of S1P. However, the role of the SphK1/S1P-mediated vascular endothelial barrier function in atherosclerosis has not been fully revealed. This study explored the roles and mechanisms of SphK1 on atherosclerosis in vivo and in vitro. METHODS In vivo, ApoE-/- and SphK1-/-ApoE-/- mice were fed a high-fat diet to induce atherosclerosis. In vitro, ox-LDL induced HUVECs to establish a cell model. Aortic histological changes were measured by H&E staining, Oil Red O staining, EVG staining, Sirius scarlet staining, immunofluorescence, and Evans Blue Assay. Western blotting was performed to explore the specific mechanism. RESULTS We validated that deficiency of SphK1 resulted in a marked amelioration of atherosclerosis, as indicated by the decreased lipid accumulation, inflammatory factors, oxidative stress, aortic plaque area, inflammatory factor infiltration, VCAM-1 expression, and vascular endothelial permeability. Moreover, deficiency of SphK1 downregulated the expression of aortic S1PR3, Rhoa, ROCK, and F-actin. The results of administration with the SphK1 inhibitor PF-543 and the S1PR3 inhibitor VPC23019 in vitro further confirmed the conclusion that deficiency of SphK1 reduced S1P level and S1PR3 protein expression, inhibited Rhoa/ROCK signaling pathway, regulated protein expression of F-actin, improved vascular endothelial dysfunction and permeability, and exerted anti-atherosclerotic effects. CONCLUSIONS This study revealed that deficiency of SphK1 relieved vascular endothelial barrier function in atherosclerosis mice via SphK1/S1P/S1PR signaling pathway.
Collapse
Affiliation(s)
- Jinyu Piao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Zhuoxuan Su
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Jiqian He
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Tianxin Zhu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Faxin Fan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Xin Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Zhenzhen Yang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Huixia Zhan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Duosheng Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China.
| |
Collapse
|
14
|
Xu J, Wang J, Zhang H, Chen Y, Zhang X, Zhang Y, Xie M, Xiao J, Qiu J, Wang G. Coupled single-cell and bulk RNA-seq analysis reveals the engulfment role of endothelial cells in atherosclerosis. Genes Dis 2024; 11:101250. [PMID: 39022128 PMCID: PMC11252887 DOI: 10.1016/j.gendis.2024.101250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 10/28/2023] [Accepted: 12/05/2023] [Indexed: 07/20/2024] Open
Abstract
The clearance of apoptotic cell debris, containing professional phagocytosis and non-professional phagocytosis, is essential for maintaining the homeostasis of healthy tissues. Here, we discovered that endothelial cells could engulf apoptotic cell debris in atherosclerotic plaque. Single-cell RNA sequencing (RNA-seq) has revealed a unique endothelial cell subpopulation in atherosclerosis, which was strongly associated with vascular injury-related pathways. Moreover, integrated analysis of three vascular injury-related RNA-seq datasets showed that the expression of scavenger receptor class B type 1 (SR-B1) was up-regulated and specifically enriched in the phagocytosis pathway under vascular injury circumstances. Single-cell RNA-seq and bulk RNA-seq indicate that SR-B1 was highly expressed in a unique endothelial cell subpopulation of mouse aorta and strongly associated with the reorganization of cellular adherent junctions and cytoskeleton which were necessary for phagocytosis. Furthermore, SR-B1 was strongly required for endothelial cells to engulf apoptotic cell debris in atherosclerotic plaque of both mouse and human aorta. Overall, this study demonstrated that apoptotic cell debris could be engulfed by endothelial cells through SR-B1 and associated with the reorganization of cellular adherent junctions and cytoskeleton.
Collapse
Affiliation(s)
- Jianxiong Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Jinxuan Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Hongping Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Yidan Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Xiaojuan Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Ying Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
- Chongqing Emergency Medical Center (Chongqing University Central Hospital), Chongqing 400014, China
| | - Ming Xie
- Chongqing Emergency Medical Center (Chongqing University Central Hospital), Chongqing 400014, China
| | - Jun Xiao
- Chongqing Emergency Medical Center (Chongqing University Central Hospital), Chongqing 400014, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
15
|
Mobilia M, Karakashian A, Neupane KR, Hage O, Whitus C, Carter A, Voy C, Johnson LA, Graf GA, Gordon SM. Enhancement of high-density lipoprotein-associated protease inhibitor activity prevents atherosclerosis progression. Atherosclerosis 2024; 396:118544. [PMID: 39126769 PMCID: PMC11404725 DOI: 10.1016/j.atherosclerosis.2024.118544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/28/2024] [Accepted: 07/11/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND AND AIMS Inflammatory cells within atherosclerotic lesions secrete proteolytic enzymes that contribute to lesion progression and destabilization, increasing the risk for an acute cardiovascular event. Elastase is a serine protease, secreted by macrophages and neutrophils, that may contribute to the development of unstable plaque. We previously reported interaction of endogenous protease-inhibitor proteins with high-density lipoprotein (HDL), including alpha-1-antitrypsin, an inhibitor of elastase. These findings support a potential role for HDL as a modulator of protease activity. In this study, we test the hypothesis that enhancement of HDL-associated elastase inhibitor activity is protective against atherosclerotic lesion progression. METHODS We designed an HDL-targeting protease inhibitor (HTPI) that binds to HDL and confers elastase inhibitor activity. Lipoprotein binding and the impact of HTPI on atherosclerosis were examined using mouse models. Histology and immunofluorescence staining of aortic root sections were used to examine the impact of HTPI on lesion morphology and inflammatory features. RESULTS HTPI is a small (1.6 kDa) peptide with an elastase inhibitor domain, a soluble linker, and an HDL-targeting domain. When incubated with human plasma ex vivo, HTPI predominantly binds to HDL. Intravenous administration of HTPI to mice resulted in its binding to plasma HDL and increased elastase inhibitor activity on isolated HDL. Accumulation of HTPI within plaque was observed after administration to Apoe-/- mice. To examine the effect of HTPI treatment on atherosclerosis, prevention and progression studies were performed using Ldlr-/- mice fed Western diet. In both study designs, HTPI-treated mice had reduced lipid deposition in plaque. CONCLUSIONS These data support the hypothesis that HDL-associated anti-elastase activity can improve the atheroprotective potential of HDL and highlight the potential utility of HDL enrichment with anti-protease activity as an approach for stabilization of atherosclerotic lesions.
Collapse
Affiliation(s)
- Maura Mobilia
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | | | - Khaga R Neupane
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Olivia Hage
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Callie Whitus
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Abigail Carter
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Clairity Voy
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Gregory A Graf
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA; Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Scott M Gordon
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA; Department of Physiology, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
16
|
Wang CW, Huang CF, Yeh ML, Chen SC, Hung CH, Kuo CH, Huang JF, Dai CY, Chuang WL, Lung-Yu M. Hepatitis C virus infection associated with coronary and thoracic aortic atherosclerosis. Am J Med Sci 2024; 368:203-213. [PMID: 38368945 DOI: 10.1016/j.amjms.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 01/30/2024] [Accepted: 02/12/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Coronary and thoracic aortic calcification was associated with stroke, coronary heart, and peripheral vascular disease. Hepatitis C virus (HCV) infection is significantly associated with insulin resistance, diabetes mellitus and hepatic steatosis. We aimed to investigate the relationship between HCV infection and coronary, thoracic aortic atherosclerosis. MATERIALS AND METHODS Calcification was detected by chest computed tomography and defined as any Agatston score greater than zero. Metabolic syndrome was based on the modified Adult Treatment Panel III criteria. Fibrosis-4 (FIB-4) and AST-to-platelet ratio (APRI) was calculated. The anti-HCV signal-to-cutoff (S/CO) ratio was determined by the third generation ELISA kit. Atherosclerosis risk was estimated by using multiple logistic regression modeling. RESULTS Being positive for both metabolic syndrome and HCV infection (OR = 2.65, 95% CI: 1.26-5.59, p = 0.007), negative for metabolic syndrome and positive for HCV infection (OR = 2.75, 95% CI: 1.48-5.30, p = 0.001), and positive for metabolic syndrome and negative for HCV infection (OR = 2.42, 95% CI: 1.92-3.07, p < 0.001) were associated with atherosclerosis compared with being negative for both metabolic syndrome and HCV infection (Ptrend< 0.001). HCV infection with liver fibrosis (HCVFIB4>1.4; OR = 2.16, 95% CI: 1.22-3.82, p = 0.008), or (HCVAPRI>0.5; OR = 3.40, 95% CI: 1.28-9.06, p = 0.014) and elevated anti-HCV S/CO ratio (anti-HCVS/CO>10.0; OR = 1.72, 95% CI: 1.01-2.93, p = 0.045) was associated with atherosclerosis. CONCLUSIONS HCV infection with metabolic syndrome, liver fibrosis and elevated anti-HCV S/CO ratio was associated with atherosclerosis.
Collapse
Affiliation(s)
- Chih-Wen Wang
- Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital; School of Medicine and Hepatitis Research Center, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Feng Huang
- Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital; School of Medicine and Hepatitis Research Center, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Lun Yeh
- Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital; School of Medicine and Hepatitis Research Center, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Szu-Chia Chen
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Hsing Hung
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Pediatrics, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chao-Hung Kuo
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jee-Fu Huang
- Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital; School of Medicine and Hepatitis Research Center, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Yen Dai
- Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital; School of Medicine and Hepatitis Research Center, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Long Chuang
- Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital; School of Medicine and Hepatitis Research Center, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming Lung-Yu
- Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital; School of Medicine and Hepatitis Research Center, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan.
| |
Collapse
|
17
|
Su H, Wang X, Wang L, Yuan N. Therapeutic Targeting of Pattern Recognition Receptors to Modulate Inflammation in Atherosclerosis. Cell Biochem Biophys 2024:10.1007/s12013-024-01481-9. [PMID: 39145823 DOI: 10.1007/s12013-024-01481-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
Atherosclerosis (AS), a potentially fatal cardiovascular disease (CVD), is a chronic inflammatory condition. The disease's onset and progression are influenced by inflammatory and immunological mechanisms. The innate immune pathways are essential in the progression of AS, as they are responsible for detecting first danger signals and causing long-term changes in immune cells. The innate immune system possesses distinct receptors known as pattern recognition receptors (PRRs) which can identify both pathogen-associated molecular patterns and danger-associated molecular signals. Activation of PRRs initiates the inflammatory response in various physiological systems, such as the cardiovascular system. This review specifically examines the contribution of the innate immune response and PRRs to the formation and advancement of AS. Studying the role of these particular receptors in AS would enhance our understanding of the development of AS and offer novel approaches for directly improving the inflammatory response associated with it.
Collapse
Affiliation(s)
- Hongyan Su
- Cardiology Department, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130000, China
| | - Xiancheng Wang
- Cardiology Department, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130000, China
| | - Lu Wang
- Cardiology Department, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130000, China
| | - Na Yuan
- Rheumatology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 30000, China.
| |
Collapse
|
18
|
Capellmann S, Kauffmann M, Arock M, Huber M. SR-BI regulates the synergistic mast cell response by modulating the plasma membrane-associated cholesterol pool. Eur J Immunol 2024; 54:e2350788. [PMID: 38708681 DOI: 10.1002/eji.202350788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/07/2024]
Abstract
The high-affinity IgE receptor FcεRI is the mast cell (MC) receptor responsible for the involvement of MCs in IgE-associated allergic disorders. Activation of the FcεRI is achieved via crosslinking by multivalent antigen (Ag) recognized by IgE resulting in degranulation and proinflammatory cytokine production. In comparison to the T- and B-cell receptor complexes, for which several co-receptors orchestrating the initial signaling events have been described, information is scarce about FcεRI-associated proteins. Additionally, it is unclear how FcεRI signaling synergizes with input from other receptors and how regulators affect this synergistic response. We found that the HDL receptor SR-BI (gene name: Scarb1/SCARB1) is expressed in MCs, functionally associates with FcεRI, and regulates the plasma membrane cholesterol content in cholesterol-rich plasma membrane nanodomains. This impacted the activation of MCs upon co-stimulation of the FcεRI with receptors known to synergize with FcεRI signaling. Amongst them, we investigated the co-activation of the FcεRI with the receptor tyrosine kinase KIT, the IL-33 receptor, and GPCRs activated by adenosine or PGE2. Scarb1-deficient bone marrow-derived MCs showed reduced cytokine secretion upon co-stimulation conditions suggesting a role for plasma membrane-associated cholesterol regulating respective MC activation. Mimicking Scarb1 deficiency by cholesterol depletion employing MβCD, we identified PKB and PLCγ1 as cholesterol-sensitive proteins downstream of FcεRI activation in bone marrow-derived MCs. When MCs were co-stimulated with stem cell factor (SCF) and Ag, PLCγ1 activation was boosted, which could be mitigated by cholesterol depletion and SR-BI inhibition. Similarly, SR-BI inhibition attenuated the synergistic response to PGE2 and anti-IgE in the human ROSAKIT WT MC line, suggesting that SR-BI is a crucial regulator of synergistic MC activation.
Collapse
Affiliation(s)
- Sandro Capellmann
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Marlies Kauffmann
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Michel Arock
- Department of Hematological Biology, Pitié-Salpêtrière Charles-Foix Hospital, AP-HP Sorbonne University, Paris, France
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
19
|
Adolph TE, Tilg H. Western diets and chronic diseases. Nat Med 2024; 30:2133-2147. [PMID: 39085420 DOI: 10.1038/s41591-024-03165-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024]
Abstract
'Westernization', which incorporates industrial, cultural and dietary trends, has paralleled the rise of noncommunicable diseases across the globe. Today, the Western-style diet emerges as a key stimulus for gut microbial vulnerability, chronic inflammation and chronic diseases, affecting mainly the cardiovascular system, systemic metabolism and the gut. Here we review the diet of modern times and evaluate the threat it poses for human health by summarizing recent epidemiological, translational and clinical studies. We discuss the links between diet and disease in the context of obesity and type 2 diabetes, cardiovascular diseases, gut and liver diseases and solid malignancies. We collectively interpret the evidence and its limitations and discuss future challenges and strategies to overcome these. We argue that healthcare professionals and societies must react today to the detrimental effects of the Western diet to bring about sustainable change and improved outcomes in the future.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
20
|
Mullis DM, Padilla-Lopez A, Wang H, Zhu Y, Elde S, Bonham SA, Yajima S, Kocher ON, Krieger M, Woo YJ. Stromal cell-derived factor-1 alpha improves cardiac function in a novel diet-induced coronary atherosclerosis model, the SR-B1ΔCT/LDLR KO mouse. Atherosclerosis 2024; 395:117518. [PMID: 38627162 PMCID: PMC11254567 DOI: 10.1016/j.atherosclerosis.2024.117518] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 02/28/2024] [Accepted: 03/12/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND AND AIMS There are a limited number of pharmacologic therapies for coronary artery disease, and few rodent models of occlusive coronary atherosclerosis and consequent myocardial infarction with which one can rapidly test new therapeutic approaches. Here, we characterize a novel, fertile, and easy-to-use HDL receptor (SR-B1)-based model of atherogenic diet-inducible, fatal coronary atherosclerosis, the SR-B1ΔCT/LDLR KO mouse. Additionally, we test intramyocardial injection of Stromal Cell-Derived Factor-1 alpha (SDF-1α), a potent angiogenic cytokine, as a possible therapy to rescue cardiac function in this mouse. METHODS SR-B1ΔCT/LDLR KO mice were fed the Paigen diet or standard chow diet, and we determined the effects of the diets on cardiac function, histology, and survival. After two weeks of feeding either the Paigen diet (n = 24) or standard chow diet (n = 20), the mice received an intramyocardial injection of either SDF-1α or phosphate buffered saline (PBS). Cardiac function and angiogenesis were assessed two weeks later. RESULTS When six-week-old mice were fed the Paigen diet, they began to die as early as 19 days later and 50% had died by 38 days. None of the mice maintained on the standard chow diet died by day 72. Hearts from mice on the Paigen diet showed evidence of cardiomegaly, myocardial infarction, and occlusive coronary artery disease. For the five mice that survived until day 28 that underwent an intramyocardial injection of PBS on day 15, the average ejection fraction (EF) decreased significantly from day 14 (the day before injection, 52.1 ± 4.3%) to day 28 (13 days after the injection, 30.6 ± 6.8%) (paired t-test, n = 5, p = 0.0008). Of the 11 mice fed the Paigen diet and injected with SDF-1α on day 15, 8 (72.7%) survived to day 28. The average EF for these 8 mice increased significantly from 48.2 ± 7.2% on day 14 to63.6 ± 6.9% on day 28 (Paired t-test, n = 8, p = 0.003). CONCLUSIONS This new mouse model and treatment with the promising angiogenic cytokine SDF-1α may lead to new therapeutic approaches for ischemic heart disease.
Collapse
MESH Headings
- Animals
- Chemokine CXCL12/metabolism
- Chemokine CXCL12/genetics
- Disease Models, Animal
- Mice, Knockout
- Coronary Artery Disease
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Scavenger Receptors, Class B/genetics
- Male
- Neovascularization, Physiologic/drug effects
- Mice, Inbred C57BL
- Diet, Atherogenic
- Mice
- Ventricular Function, Left
- Myocardium/pathology
- Myocardium/metabolism
- Diet, High-Fat
Collapse
Affiliation(s)
- Danielle M Mullis
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | | | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Yuanjia Zhu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Stefan Elde
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Spencer A Bonham
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Shin Yajima
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Olivier N Kocher
- Department of Pathology, Beth Israel Hospital, Harvard Medical School, Boston, MA, USA
| | - Monty Krieger
- Department of Biology, Massachusetts Institute of Technology, MA, USA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
21
|
Kothari V, Ho TW, Cabodevilla AG, He Y, Kramer F, Shimizu-Albergine M, Kanter JE, Snell-Bergeon J, Fisher EA, Shao B, Heinecke JW, Wobbrock JO, Lee WL, Goldberg IJ, Vaisar T, Bornfeldt KE. Imbalance of APOB Lipoproteins and Large HDL in Type 1 Diabetes Drives Atherosclerosis. Circ Res 2024; 135:335-349. [PMID: 38828596 PMCID: PMC11223987 DOI: 10.1161/circresaha.123.323100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 04/25/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Individuals with type 1 diabetes (T1D) generally have normal or even higher HDL (high-density lipoprotein)-cholesterol levels than people without diabetes yet are at increased risk for atherosclerotic cardiovascular disease (CVD). Human HDL is a complex mixture of particles that can vary in cholesterol content by >2-fold. To investigate if specific HDL subspecies contribute to the increased atherosclerosis associated with T1D, we created mouse models of T1D that exhibit human-like HDL subspecies. We also measured HDL subspecies and their association with incident CVD in a cohort of people with T1D. METHODS We generated LDL receptor-deficient (Ldlr-/-) mouse models of T1D expressing human APOA1 (apolipoprotein A1). Ldlr-/-APOA1Tg mice exhibited the main human HDL subspecies. We also generated Ldlr-/-APOA1Tg T1D mice expressing CETP (cholesteryl ester transfer protein), which had lower concentrations of large HDL subspecies versus mice not expressing CETP. HDL particle concentrations and sizes and proteins involved in lipoprotein metabolism were measured by calibrated differential ion mobility analysis and targeted mass spectrometry in the mouse models of T1D and in a cohort of individuals with T1D. Endothelial transcytosis was analyzed by total internal reflection fluorescence microscopy. RESULTS Diabetic Ldlr-/-APOA1Tg mice were severely hyperglycemic and hyperlipidemic and had markedly elevated plasma APOB levels versus nondiabetic littermates but were protected from the proatherogenic effects of diabetes. Diabetic Ldlr-/-APOA1Tg mice expressing CETP lost the atheroprotective effect and had increased lesion necrotic core areas and APOB accumulation, despite having lower plasma APOB levels. The detrimental effects of low concentrations of larger HDL particles in diabetic mice expressing CETP were not explained by reduced cholesterol efflux. Instead, large HDL was more effective than small HDL in preventing endothelial transcytosis of LDL mediated by scavenger receptor class B type 1. Finally, in humans with T1D, increased concentrations of larger HDL particles relative to APOB100 negatively predicted incident CVD independently of HDL-cholesterol levels. CONCLUSIONS Our results suggest that the balance between APOB lipoproteins and the larger HDL subspecies contributes to atherosclerosis progression and incident CVD in the setting of T1D and that larger HDLs exert atheroprotective effects on endothelial cells rather than by promoting macrophage cholesterol efflux.
Collapse
MESH Headings
- Adult
- Animals
- Female
- Humans
- Male
- Mice
- Middle Aged
- Apolipoprotein A-I/blood
- Apolipoprotein A-I/metabolism
- Apolipoprotein B-100/metabolism
- Apolipoprotein B-100/genetics
- Apolipoprotein B-100/blood
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/blood
- Atherosclerosis/pathology
- Cholesterol Ester Transfer Proteins/genetics
- Cholesterol Ester Transfer Proteins/metabolism
- Cholesterol Ester Transfer Proteins/blood
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/blood
- Disease Models, Animal
- Lipoproteins, HDL/blood
- Lipoproteins, HDL/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Receptors, LDL/metabolism
Collapse
Affiliation(s)
- Vishal Kothari
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Tse W.W. Ho
- Keenan Centre for Biomedical Research, St. Michael’s Hospital, Toronto, Canada (T.W.W.H., W.L.L.)
- Department of Laboratory Medicine and Pathobiology (T.W.W.H., W.L.L.)
| | | | - Yi He
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Farah Kramer
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Masami Shimizu-Albergine
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Jenny E. Kanter
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Janet Snell-Bergeon
- Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora (J.S.-B.)
| | - Edward A. Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (E.A.F.)
| | - Baohai Shao
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Jay W. Heinecke
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | | | - Warren L. Lee
- Keenan Centre for Biomedical Research, St. Michael’s Hospital, Toronto, Canada (T.W.W.H., W.L.L.)
- Department of Laboratory Medicine and Pathobiology (T.W.W.H., W.L.L.)
- Interdepartmental Division of Critical Care and the Department of Biochemistry, University of Toronto, Canada (W.L.L.)
| | - Ira J. Goldberg
- Division of Endocrinology, Diabetes and Metabolism (A.G.C., I.J.G.)
| | - Tomas Vaisar
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
| | - Karin E. Bornfeldt
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute (V.K., Y.H., F.K., M.S.-A., J.E.K., B.S., J.W.H., T.V., K.E.B.)
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle (K.E.B.)
| |
Collapse
|
22
|
Terao J. Caveolae and caveolin-1 as targets of dietary polyphenols for protection against vascular endothelial dysfunction. J Clin Biochem Nutr 2024; 75:7-16. [PMID: 39070533 PMCID: PMC11273273 DOI: 10.3164/jcbn.24-30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 07/30/2024] Open
Abstract
Caveolae, consisting of caveolin-1 proteins, are ubiquitously present in endothelial cells and contribute to normal cardiovascular functions by acting as a platform for cellular signaling pathways as well as transcytosis and endocytosis. However, caveolin-1 is thought to have a proatherogenic role by inhibiting endothelial nitric oxide synthase activity and Nrf2 activation, or by promoting inflammation through NF-κB activation. Dietary polyphenols were suggested to exert anti-atherosclerotic effects by a mechanism involving the inhibition of endothelial dysfunction, by which they can regulate redox-sensitive signaling pathways in relation to NF-κB and Nrf2 activation. Some monomeric polyphenols and microbiota-derived catabolites from monomeric polyphenols or polymeric tannins might be responsible for the inhibition, because they can be transferred into the circulation from the digestive tract. Several polyphenols were reported to modulate caveolin-1 expression or its localization in caveolae. Therefore, we hypothesized that circulating polyphenols affect caveolae functions by altering its structure leading to the release of caveolin-1 from caveolae, and attenuating redox-sensitive signaling pathway-dependent caveolin-1 overexpression. Further studies using circulating polyphenols at a physiologically relevant level are necessary to clarify the mechanism of action of dietary polyphenols targeting caveolae and caveolin-1.
Collapse
Affiliation(s)
- Junji Terao
- Faculty of Medicine, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
23
|
Shu LX, Cao LL, Guo X, Wang ZB, Wang SZ. Mechanism of efferocytosis in atherosclerosis. J Mol Med (Berl) 2024; 102:831-840. [PMID: 38727748 DOI: 10.1007/s00109-024-02439-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 06/29/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease that occurs in the intima of large and medium-sized arteries with the immune system's involvement. It is a common pathological basis for high morbidity and mortality of cardiovascular diseases. Abnormal proliferation of apoptotic cells and necrotic cells leads to AS plaque expansion, necrotic core formation, and rupture. In the early stage of AS, macrophages exert an efferocytosis effect to engulf and degrade apoptotic, dead, damaged, or senescent cells by efferocytosis, thus enabling the regulation of the organism. In the early stage of AS, macrophages rely on this effect to slow down the process of AS. However, in the advanced stage of AS, the efferocytosis of macrophages within the plaque is impaired, which leads to the inability of macrophages to promptly remove the apoptotic cells (ACs) from the organism promptly, causing exacerbation of AS. Moreover, upregulation of CD47 expression in AS plaques also protects ACs from phagocytosis by macrophages, resulting in a large amount of residual ACs in the plaque, further expanding the necrotic core. In this review, we discussed the molecular mechanisms involved in the process of efferocytosis and how efferocytosis is impaired and regulated during AS, hoping to provide new insights for treating AS.
Collapse
Affiliation(s)
- Li-Xia Shu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Liu-Li Cao
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Xin Guo
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Zong-Bao Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China.
| |
Collapse
|
24
|
Zhang N, Nao J, Zhang S, Dong X. Novel insights into the activating transcription factor 4 in Alzheimer's disease and associated aging-related diseases: Mechanisms and therapeutic implications. Front Neuroendocrinol 2024; 74:101144. [PMID: 38797197 DOI: 10.1016/j.yfrne.2024.101144] [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: 02/05/2024] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
Ageing is inherent to all human beings, most mechanistic explanations of ageing results from the combined effects of various physiological and pathological processes. Additionally, aging pivotally contributes to several chronic diseases. Activating transcription factor 4 (ATF4), a member of the ATF/cAMP response element-binding protein family, has recently emerged as a pivotal player owing to its indispensable role in the pathophysiological processes of Alzheimer's disease and aging-related diseases. Moreover, ATF4 is integral to numerous biological processes. Therefore, this article aims to comprehensively review relevant research on the role of ATF4 in the onset and progression of aging-related diseases, elucidating its potential mechanisms and therapeutic approaches. Our objective is to furnish scientific evidence for the early identification of risk factors in aging-related diseases and pave the way for new research directions for their treatment. By elucidating the signaling pathway network of ATF4 in aging-related diseases, we aspire to gain a profound understanding of the molecular and cellular mechanisms, offering novel strategies for addressing aging and developing related therapeutics.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Neurology, the Seventh Clinical College of China Medical University, No. 24 Central Street, Xinfu District, Fushun 113000, Liaoning, China.
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110000, Liaoning, China.
| | - Shun Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110000, Liaoning, China.
| | - Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110000, Liaoning, China.
| |
Collapse
|
25
|
Getz GS, Reardon CA. Insights from Murine Studies on the Site Specificity of Atherosclerosis. Int J Mol Sci 2024; 25:6375. [PMID: 38928086 PMCID: PMC11204064 DOI: 10.3390/ijms25126375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Atherosclerosis is an inflammatory reaction that develops at specific regions within the artery wall and at specific sites of the arterial tree over a varying time frame in response to a variety of risk factors. The mechanisms that account for the interaction of systemic factors and atherosclerosis-susceptible regions of the arterial tree to mediate this site-specific development of atherosclerosis are not clear. The dynamics of blood flow has a major influence on where in the arterial tree atherosclerosis develops, priming the site for interactions with atherosclerotic risk factors and inducing cellular and molecular participants in atherogenesis. But how this accounts for lesion development at various locations along the vascular tree across differing time frames still requires additional study. Currently, murine models are favored for the experimental study of atherogenesis and provide the most insight into the mechanisms that may contribute to the development of atherosclerosis. Based largely on these studies, in this review, we discuss the role of hemodynamic shear stress, SR-B1, and other factors that may contribute to the site-specific development of atherosclerosis.
Collapse
Affiliation(s)
- Godfrey S. Getz
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA;
| | | |
Collapse
|
26
|
Zhao S, Guo H, Qiu L, Zhong C, Xue J, Qin M, Zhang Y, Xu C, Xie Y, Yu J. Saponins from Allii Macrostemonis Bulbus attenuate atherosclerosis by inhibiting macrophage foam cell formation and inflammation. Sci Rep 2024; 14:12917. [PMID: 38839811 PMCID: PMC11153636 DOI: 10.1038/s41598-024-61209-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/02/2024] [Indexed: 06/07/2024] Open
Abstract
Allii Macrostemonis Bulbus (AMB) is a traditional Chinese medicine with medicinal and food homology. AMB has various biological activities, including anti-coagulation, lipid-lowering, anti-tumor, and antioxidant effects. Saponins from Allium macrostemonis Bulbus (SAMB), the predominant beneficial compounds, also exhibited lipid-lowering and anti-inflammatory properties. However, the effect of SAMB on atherosclerosis and the underlying mechanisms are still unclear. This study aimed to elucidate the pharmacological impact of SAMB on atherosclerosis. In apolipoprotein E deficiency (ApoE-/-) mice with high-fat diet feeding, oral SAMB administration significantly attenuated inflammation and atherosclerosis plaque formation. The in vitro experiments demonstrated that SAMB effectively suppressed oxidized-LDL-induced foam cell formation by down-regulating CD36 expression, thereby inhibiting lipid endocytosis in bone marrow-derived macrophages. Additionally, SAMB effectively blocked LPS-induced inflammatory response in bone marrow-derived macrophages potentially through modulating the NF-κB/NLRP3 pathway. In conclusion, SAMB exhibits a potential anti-atherosclerotic effect by inhibiting macrophage foam cell formation and inflammation. These findings provide novel insights into potential preventive and therapeutic strategies for the clinical management of atherosclerosis.
Collapse
Affiliation(s)
- Shutian Zhao
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Huijun Guo
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Liang Qiu
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Chao Zhong
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jing Xue
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Manman Qin
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Yifeng Zhang
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Chuanming Xu
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
| | - Yanfei Xie
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
| | - Jun Yu
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
27
|
Goldberg IJ, Cabodevilla AG, Younis W. In the Beginning, Lipoproteins Cross the Endothelial Barrier. J Atheroscler Thromb 2024; 31:854-860. [PMID: 38616110 PMCID: PMC11150724 DOI: 10.5551/jat.rv22017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 04/16/2024] Open
Abstract
Atherosclerosis begins with the infiltration of cholesterol-containing lipoproteins into the arterial wall. White blood cell (WBC)-associated inflammation follows. Despite decades of research using genetic and pharmacologic methods to alter WBC function, in humans, the most effective method to prevent the initiation and progression of disease remains low-density lipoprotein (LDL) reduction. However, additional approaches to reducing cardiovascular disease would be useful as residual risk of events continues even with currently effective LDL-reducing treatments. Some of this residual risk may be due to vascular toxicity of triglyceride-rich lipoproteins (TRLs). Another option is that LDL transcytosis continues, albeit at reduced rates due to lower circulating levels of this lipoprotein. This review will address these two topics. The evidence that TRLs promote atherosclerosis and the processes that allow LDL and TRLs to be taken up by endothelial cells leading to their accumulation with the subendothelial space.
Collapse
Affiliation(s)
- Ira J Goldberg
- Division of Endocrinology, New York University Grossman School of Medicine
| | | | - Waqas Younis
- Division of Endocrinology, New York University Grossman School of Medicine
| |
Collapse
|
28
|
Cheng X, Shihabudeen Haider Ali MS, Baki VB, Moran M, Su H, Sun X. Multifaceted roles of Meg3 in cellular senescence and atherosclerosis. Atherosclerosis 2024; 392:117506. [PMID: 38518516 PMCID: PMC11088985 DOI: 10.1016/j.atherosclerosis.2024.117506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 02/11/2024] [Accepted: 03/05/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND AND AIMS Long noncoding RNAs are involved in the pathogenesis of atherosclerosis. As long noncoding RNAs maternally expressed gene 3 (Meg3) prevents cellular senescence of hepatic vascular endothelium and obesity-induced insulin resistance, we decided to examine its role in cellular senescence and atherosclerosis. METHODS AND RESULTS By analyzing our data and human and mouse data from the Gene Expression Omnibus database, we found that Meg3 expression was reduced in humans and mice with cardiovascular disease, indicating its potential role in atherosclerosis. In Ldlr-/- mice fed a Western diet for 12 weeks, Meg3 silencing by chemically modified antisense oligonucleotides attenuated the formation of atherosclerotic lesions by 34.9% and 20.1% in male and female mice, respectively, revealed by en-face Oil Red O staining, which did not correlate with changes in plasma lipid profiles. Real-time quantitative PCR analysis of cellular senescence markers p21 and p16 revealed that Meg3 deficiency aggravates hepatic cellular senescence but not cellular senescence at aortic roots. Human Meg3 transgenic mice were generated to examine the role of Meg3 gain-of-function in the development of atherosclerosis induced by PCSK9 overexpression. Meg3 overexpression promotes atherosclerotic lesion formation by 29.2% in Meg3 knock-in mice independent of its effects on lipid profiles. Meg3 overexpression inhibits hepatic cellular senescence, while it promotes aortic cellular senescence likely by impairing mitochondrial function and delaying cell cycle progression. CONCLUSIONS Our data demonstrate that Meg3 promotes the formation of atherosclerotic lesions independent of its effects on plasma lipid profiles. In addition, Meg3 regulates cellular senescence in a tissue-specific manner during atherosclerosis. Thus, we demonstrated that Meg3 has multifaceted roles in cellular senescence and atherosclerosis.
Collapse
Affiliation(s)
- Xiao Cheng
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE, 68588, USA
| | | | - Vijaya Bhaskar Baki
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE, 68588, USA
| | - Matthew Moran
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE, 68588, USA
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA; Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Xinghui Sun
- Department of Biochemistry, University of Nebraska - Lincoln, Beadle Center, 1901 Vine St, Lincoln, NE, 68588, USA; Nebraska Center for the Prevention of Obesity Diseases Through Dietary Molecules, University of Nebraska - Lincoln, USA.
| |
Collapse
|
29
|
Younis W, Goldberg IJ. Can another lipid, sphingosine-1-phosphate, treat atherosclerosis? Cardiovasc Res 2024; 120:435-436. [PMID: 38563326 DOI: 10.1093/cvr/cvae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Affiliation(s)
- Waqas Younis
- Division of Endocrinology, New York University Grossman School of Medicine, 435 East 30th Street, SB 617, New York, NY 10016, USA
| | - Ira J Goldberg
- Division of Endocrinology, New York University Grossman School of Medicine, 435 East 30th Street, SB 617, New York, NY 10016, USA
| |
Collapse
|
30
|
Velagapudi S, Wang D, Poti F, Feuerborn R, Robert J, Schlumpf E, Yalcinkaya M, Panteloglou G, Potapenko A, Simoni M, Rohrer L, Nofer JR, von Eckardstein A. Sphingosine-1-phosphate receptor 3 regulates the transendothelial transport of high-density lipoproteins and low-density lipoproteins in opposite ways. Cardiovasc Res 2024; 120:476-489. [PMID: 38109696 PMCID: PMC11060483 DOI: 10.1093/cvr/cvad183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 10/08/2023] [Accepted: 10/20/2023] [Indexed: 12/20/2023] Open
Abstract
AIMS The entry of lipoproteins from blood into the arterial wall is a rate-limiting step in atherosclerosis. It is controversial whether this happens by filtration or regulated transendothelial transport.Because sphingosine-1-phosphate (S1P) preserves the endothelial barrier, we investigated in vivo and in vitro, whether S1P and its cognate S1P-receptor 3 (S1P3) regulate the transendothelial transport of lipoproteins. METHODS AND RESULTS Compared to apoE-haploinsufficient mice (CTRL), apoE-haploinsufficient mice with additional endothelium-specific knock-in of S1P3 (S1P3-iECKI) showed decreased transport of LDL and Evan's Blue but increased transport of HDL from blood into the peritoneal cave. After 30 weeks of high-fat diet feeding, S1P3-iECKI mice had lower levels of non-HDL-cholesterol and less atherosclerosis than CTRL mice. In vitro stimulation with an S1P3 agonist increased the transport of 125I-HDL but decreased the transport of 125I-LDL through human aortic endothelial cells (HAECs). Conversely, inhibition or knock-down of S1P3 decreased the transport of 125I-HDL but increased the transport of 125I-LDL. Silencing of SCARB1 encoding scavenger receptor B1 (SR-BI) abrogated the stimulation of 125I-HDL transport by the S1P3 agonist. The transendothelial transport of 125I-LDL was decreased by silencing of SCARB1 or ACVLR1 encoding activin-like kinase 1 but not by interference with LDLR. None of the three knock-downs prevented the stimulatory effect of S1P3 inhibition on transendothelial 125I-LDL transport. CONCLUSION S1P3 regulates the transendothelial transport of HDL and LDL oppositely by SR-BI-dependent and SR-BI-independent mechanisms, respectively. This divergence supports a contention that lipoproteins pass the endothelial barrier by specifically regulated mechanisms rather than passive filtration.
Collapse
Affiliation(s)
- Srividya Velagapudi
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Dongdong Wang
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Francesco Poti
- Department of Medicine and Surgery—Unit of Neurosciences, University of Parma, Parma, Italy
- Department of Biomedical, Metabolic and Neural Sciences—Unit of Endocrinology, University of Modena and Reggio Emilia, Modena, Italy
| | - Renata Feuerborn
- Central Laboratory Facility, University Hospital of Münster, Münster, Germany
| | - Jerome Robert
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Eveline Schlumpf
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Mustafa Yalcinkaya
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Grigorios Panteloglou
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Anton Potapenko
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences—Unit of Endocrinology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Jerzy-Roch Nofer
- Central Laboratory Facility, University Hospital of Münster, Münster, Germany
- Institute of Laboratory Medicine, Marien-Hospital Osnabrück, Niels-Stensen-Kliniken, Osnabrück, Germany
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| |
Collapse
|
31
|
Hao D, Guo L, Wang Q, Ito M, Huang B, Mineo C, Shaul PW, Li XA. Relative Adrenal Insufficiency Is a Risk Factor for Pediatric Sepsis: A Proof-of-Concept Study. J Infect Dis 2024; 229:1166-1177. [PMID: 37633660 PMCID: PMC11011189 DOI: 10.1093/infdis/jiad369] [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: 03/29/2023] [Revised: 08/16/2023] [Accepted: 08/24/2023] [Indexed: 08/28/2023] Open
Abstract
Glucocorticoid (GC) therapy had been strongly recommended for pediatric sepsis (grade 1A). However, the recommendation was changed to grade 2C in 2020 due to weak evidence. About 32.8% of patients with pediatric septic develop relative adrenal insufficiency (RAI). But whether GC therapy should be determined by RAI status is controversial. This study utilized 21-day-old SF1CreSRBIfl/fl mice as the first pediatric RAI mouse model to assess the pathogenesis of RAI and evaluate GC therapy. RAI mice exhibited a substantially higher mortality rate in cecal ligation and puncture and cecal slurry-induced sepsis. These mice featured persistent inflammatory responses and were effectively rescued by GC therapy. RNA sequencing analysis revealed persistent inflammatory responses in RAI mice, caused by transcriptional dysregulation of AP-1 and NF-κB, and cytokine-induced secondary inflammatory response. Our findings support a precision medicine approach to guide GC therapy for pediatric patients based on the status of RAI.
Collapse
Affiliation(s)
- Dan Hao
- Department of Pharmacology and Nutritional Sciences
| | - Ling Guo
- Saha Cardiovascular Research Center
| | | | - Misa Ito
- Department of Pharmacology and Nutritional Sciences
| | - Bin Huang
- Division of Cancer Biostatistics, College of Medicine, University of Kentucky, Lexington
| | - Chieko Mineo
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas
| | - Philip W Shaul
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas
| | - Xiang-An Li
- Department of Pharmacology and Nutritional Sciences
- Saha Cardiovascular Research Center
- Lexington VA Healthcare System
- Department of Physiology, College of Medicine, University of Kentucky, Lexington
| |
Collapse
|
32
|
Wang J, Xu J, Liu T, Yu C, Xu F, Wang G, Li S, Dai X. Biomechanics-mediated endocytosis in atherosclerosis. Front Cardiovasc Med 2024; 11:1337679. [PMID: 38638885 PMCID: PMC11024446 DOI: 10.3389/fcvm.2024.1337679] [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: 11/13/2023] [Accepted: 03/04/2024] [Indexed: 04/20/2024] Open
Abstract
Biomechanical forces, including vascular shear stress, cyclic stretching, and extracellular matrix stiffness, which influence mechanosensitive channels in the plasma membrane, determine cell function in atherosclerosis. Being highly associated with the formation of atherosclerotic plaques, endocytosis is the key point in molecule and macromolecule trafficking, which plays an important role in lipid transportation. The process of endocytosis relies on the mobility and tension of the plasma membrane, which is sensitive to biomechanical forces. Several studies have advanced the signal transduction between endocytosis and biomechanics to elaborate the developmental role of atherosclerosis. Meanwhile, increased plaque growth also results in changes in the structure, composition and morphology of the coronary artery that contribute to the alteration of arterial biomechanics. These cross-links of biomechanics and endocytosis in atherosclerotic plaques play an important role in cell function, such as cell phenotype switching, foam cell formation, and lipoprotein transportation. We propose that biomechanical force activates the endocytosis of vascular cells and plays an important role in the development of atherosclerosis.
Collapse
Affiliation(s)
- Jinxuan Wang
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
- Department of Cardiology, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jianxiong Xu
- School of Health Management, Xihua University, Chengdu, China
| | - Tianhu Liu
- Department of Cardiology, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Cardiology and Vascular Health Research Center, Chengdu Medical College, Chengdu, China
| | - Chaoping Yu
- Department of Cardiology, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Cardiology and Vascular Health Research Center, Chengdu Medical College, Chengdu, China
| | - Fengcheng Xu
- Department of Cardiology, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Cardiology and Vascular Health Research Center, Chengdu Medical College, Chengdu, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Shun Li
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Xiaozhen Dai
- Department of Cardiology, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Cardiology and Vascular Health Research Center, Chengdu Medical College, Chengdu, China
- School of Biosciences and Technology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
33
|
Stroope C, Nettersheim FS, Coon B, Finney AC, Schwartz MA, Ley K, Rom O, Yurdagul A. Dysregulated cellular metabolism in atherosclerosis: mediators and therapeutic opportunities. Nat Metab 2024; 6:617-638. [PMID: 38532071 PMCID: PMC11055680 DOI: 10.1038/s42255-024-01015-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
Accumulating evidence over the past decades has revealed an intricate relationship between dysregulation of cellular metabolism and the progression of atherosclerotic cardiovascular disease. However, an integrated understanding of dysregulated cellular metabolism in atherosclerotic cardiovascular disease and its potential value as a therapeutic target is missing. In this Review, we (1) summarize recent advances concerning the role of metabolic dysregulation during atherosclerosis progression in lesional cells, including endothelial cells, vascular smooth muscle cells, macrophages and T cells; (2) explore the complexity of metabolic cross-talk between these lesional cells; (3) highlight emerging technologies that promise to illuminate unknown aspects of metabolism in atherosclerosis; and (4) suggest strategies for targeting these underexplored metabolic alterations to mitigate atherosclerosis progression and stabilize rupture-prone atheromas with a potential new generation of cardiovascular therapeutics.
Collapse
Affiliation(s)
- Chad Stroope
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Felix Sebastian Nettersheim
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Brian Coon
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Cardiovascular Biology Research Program, OMRF, Oklahoma City, OK, USA
- Department of Cell Biology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
- Immunology Center of Georgia (IMMCG), Augusta University Immunology Center of Georgia, Augusta, GA, USA
| | - Oren Rom
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
34
|
Fung KYY, Ho TWW, Xu Z, Neculai D, Beauchemin CAA, Lee WL, Fairn GD. Apolipoprotein A1 and high-density lipoprotein limit low-density lipoprotein transcytosis by binding SR-B1. J Lipid Res 2024; 65:100530. [PMID: 38479648 PMCID: PMC11004410 DOI: 10.1016/j.jlr.2024.100530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 04/09/2024] Open
Abstract
Atherosclerosis results from the deposition and oxidation of LDL and immune cell infiltration in the sub-arterial space leading to arterial occlusion. Studies have shown that transcytosis transports circulating LDL across endothelial cells lining blood vessels. LDL transcytosis is initiated by binding to either scavenger receptor B1 (SR-B1) or activin A receptor-like kinase 1 on the apical side of endothelial cells leading to its transit and release on the basolateral side. HDL is thought to partly protect individuals from atherosclerosis due to its ability to remove excess cholesterol and act as an antioxidant. Apolipoprotein A1 (APOA1), an HDL constituent, can bind to SR-B1, raising the possibility that APOA1/HDL can compete with LDL for SR-B1 binding, thereby limiting LDL deposition in the sub-arterial space. To examine this possibility, we used in vitro approaches to quantify the internalization and transcytosis of fluorescent LDL in coronary endothelial cells. Using microscale thermophoresis and affinity capture, we find that SR-B1 and APOA1 interact and that binding is enhanced when using the cardioprotective variant of APOA1 termed Milano (APOA1-Milano). In male mice, transiently increasing the levels of HDL reduced the acute deposition of fluorescently labeled LDL in the atheroprone inner curvature of the aorta. Reduced LDL deposition was also observed when increasing circulating wild-type APOA1 or the APOA1-Milano variant, with a more robust inhibition from the APOA1-Milano. The results suggest that HDL may limit SR-B1-mediated LDL transcytosis and deposition, adding to the mechanisms by which it can act as an atheroprotective particle.
Collapse
Affiliation(s)
- Karen Y Y Fung
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Tse Wing Winnie Ho
- Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Zizhen Xu
- Department of Cell Biology, and Department of Pathology Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dante Neculai
- Department of Cell Biology, and Department of Pathology Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Catherine A A Beauchemin
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Interdisciplinary Theoretical and Mathematical Sciences (iTHEMS) program, RIKEN, Wako, Saitama, Japan
| | - Warren L Lee
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Gregory D Fairn
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
35
|
Vittum Z, Cocchiaro S, Mensah SA. Basal endothelial glycocalyx's response to shear stress: a review of structure, function, and clinical implications. Front Cell Dev Biol 2024; 12:1371769. [PMID: 38562144 PMCID: PMC10982814 DOI: 10.3389/fcell.2024.1371769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
The endothelial glycocalyx encompasses the entire endothelial cell, transducing extracellular signals and regulating vascular permeability and barrier functions. The apical glycocalyx, which forms the lumen of the vessel, and the basal glycocalyx, at the smooth muscle cell interface, are often investigated separately as they are exposed to vastly different stimuli. The apical glycocalyx directly senses fluid shear forces transmitting them intracellularly through connection to the cytoskeleton of the endothelial cell. The basal glycocalyx has demonstrated sensitivity to shear due to blood flow transmitted through the cytoskeleton, promoting alternate signaling processes. In this review, we discuss current literature on the basal glycocalyx's response to shear stress in the context of mechanotransduction and remodeling. The possible implications of basal glycocalyx degradation in pathologies are also explored. Finally, this review seeks to highlight how addressing the gaps discussed would improve our wholistic understanding of the endothelial glycocalyx and its role in maintaining vascular homeostasis.
Collapse
Affiliation(s)
- Zoe Vittum
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
| | - Samantha Cocchiaro
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
| | - Solomon A. Mensah
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
- Mechanical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
| |
Collapse
|
36
|
Xia Y, Gao D, Wang X, Liu B, Shan X, Sun Y, Ma D. Role of Treg cell subsets in cardiovascular disease pathogenesis and potential therapeutic targets. Front Immunol 2024; 15:1331609. [PMID: 38558816 PMCID: PMC10978666 DOI: 10.3389/fimmu.2024.1331609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
In the genesis and progression of cardiovascular diseases involving both innate and adaptive immune responses, inflammation plays a pivotal and dual role. Studies in experimental animals indicate that certain immune responses are protective, while others exacerbate the disease. T-helper (Th) 1 cell immune responses are recognized as key drivers of inflammatory progression in cardiovascular diseases. Consequently, the CD4+CD25+FOXP3+ regulatory T cells (Tregs) are gaining increasing attention for their roles in inflammation and immune regulation. Given the critical role of Tregs in maintaining immune-inflammatory balance and homeostasis, abnormalities in their generation or function might lead to aberrant immune responses, thereby initiating pathological changes. Numerous preclinical studies and clinical trials have unveiled the central role of Tregs in cardiovascular diseases, such as atherosclerosis. Here, we review the roles and mechanisms of Treg subsets in cardiovascular conditions like atherosclerosis, hypertension, myocardial infarction and remodeling, myocarditis, dilated cardiomyopathy, and heart failure. While the precise molecular mechanisms of Tregs in cardiac protection remain elusive, therapeutic strategies targeting Tregs present a promising new direction for the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Yunpeng Sun
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, China
| | - Dashi Ma
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
37
|
Su JH, Hong Y, Han CC, Yu J, Guan X, Zhu YM, Wang C, Ma MM, Pang RP, Ou JS, Zhou JG, Zhang ZY, Ban T, Liang SJ. Dual action of macrophage miR-204 confines cyclosporine A-induced atherosclerosis. Br J Pharmacol 2024; 181:640-658. [PMID: 37702564 DOI: 10.1111/bph.16240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 07/15/2023] [Accepted: 08/31/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Atherosclerosis induced by cyclosporine A (CsA), an inhibitor of the calcineurin/nuclear factor of activated T cells (NFAT) pathway, is a major concern after organ transplantation. However, the atherosclerotic mechanisms of CsA remain obscure. We previously demonstrated that calcineurin/NFAT signalling inhibition contributes to atherogenesis via suppressing microRNA-204 (miR-204) transcription. We therefore hypothesised that miR-204 is involved in the development of CsA-induced atherosclerosis. EXPERIMENTAL APPROACH ApoE-/- mice with macrophage-miR-204 overexpression were generated to determine the effects of miR-204 on CsA-induced atherosclerosis. Luciferase reporter assays and chromatin immunoprecipitation sequencing were performed to explore the targets mediating miR-204 effects. KEY RESULTS CsA alone did not significantly affect atherosclerotic lesions or serum lipid levels. However, it exacerbated high-fat diet-induced atherosclerosis and hyperlipidemia in C57BL/6J and ApoE-/- mice, respectively. miR-204 levels decreased in circulating monocytes and plaque lesions during CsA-induced atherosclerosis. The upregulation of miR-204 in macrophages inhibited CsA-induced atherosclerotic plaque formation but did not affect serum lipid levels. miR-204 limited the CsA-induced foam cell formation by reducing the expression of the scavenger receptors SR-BII and CD36. SR-BII was post-transcriptionally regulated by mature miR-204-5p via 3'-UTR targeting. Additionally, nuclear-localised miR-204-3p prevented the CsA-induced binding of Ago2 to the CD36 promoter, suppressing CD36 transcription. SR-BII or CD36 expression restoration dampened the beneficial effects of miR-204 on CsA-induced atherosclerosis. CONCLUSION AND IMPLICATIONS Macrophage miR-204 ameliorates CsA-induced atherosclerosis, suggesting that miR-204 may be a potential target for the prevention and treatment of CsA-related atherosclerotic side effects.
Collapse
Affiliation(s)
- Jia-Hui Su
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yu Hong
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Department of Pharmacy, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Cong-Cong Han
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jie Yu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Guan
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Ya-Mei Zhu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Cheng Wang
- Program of Kidney and Cardiovascular Diseases, the Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Ming-Ming Ma
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Rui-Ping Pang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jing-Song Ou
- Division of Cardiac Surgery, The Key Laboratory of Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jia-Guo Zhou
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Program of Kidney and Cardiovascular Diseases, the Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Zi-Yi Zhang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Tao Ban
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Si-Jia Liang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Program of Kidney and Cardiovascular Diseases, the Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
38
|
Nakamura Y, Kulkarni NN, Takahashi T, Alimohamadi H, Dokoshi T, Liu E, Shia M, Numata T, Luo EW, Gombart AF, Yang X, Secrest P, Gordts PL, Tsimikas S, Wong GC, Gallo RL. Increased LL37 in psoriasis and other inflammatory disorders promotes LDL uptake and atherosclerosis. J Clin Invest 2024; 134:e172578. [PMID: 38194294 PMCID: PMC10904043 DOI: 10.1172/jci172578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
Patients with chronic inflammatory disorders such as psoriasis have an increased risk of cardiovascular disease and elevated levels of LL37, a cathelicidin host defense peptide that has both antimicrobial and proinflammatory properties. To explore whether LL37 could contribute to the risk of heart disease, we examined its effects on lipoprotein metabolism and show that LL37 enhanced LDL uptake in macrophages through the LDL receptor (LDLR), scavenger receptor class B member 1 (SR-B1), and CD36. This interaction led to increased cytosolic cholesterol in macrophages and changes in expression of lipid metabolism genes consistent with increased cholesterol uptake. Structure-function analysis and synchrotron small-angle x-ray scattering showed structural determinants of the LL37-LDL complex that underlie its ability to bind its receptors and promote uptake. This function of LDL uptake is unique to cathelicidins from humans and some primates and was not observed with cathelicidins from mice or rabbits. Notably, Apoe-/- mice expressing LL37 developed larger atheroma plaques than did control mice, and a positive correlation between plasma LL37 and oxidized phospholipid on apolipoprotein B (OxPL-apoB) levels was observed in individuals with cardiovascular disease. These findings provide evidence that LDL uptake can be increased via interaction with LL37 and may explain the increased risk of cardiovascular disease associated with chronic inflammatory disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Adrian F. Gombart
- Linus Pauling Institute, Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, USA
| | | | - Patrick Secrest
- Department of Medicine, Division of Endocrinology and Metabolism, and
| | - Philip L.S.M. Gordts
- Department of Medicine, Division of Endocrinology and Metabolism, and
- Glycobiology Research and Training Center, UCSD, La Jolla, California, USA
| | | | - Gerard C.L. Wong
- Department of Bioengineering, UCLA, Los Angeles, California, USA
| | | |
Collapse
|
39
|
Nasr M, Fay A, Lupieri A, Malet N, Darmon A, Zahreddine R, Swiader A, Wahart A, Viaud J, Nègre-Salvayre A, Hirsch E, Monteyne D, Perez-Morgà D, Dupont N, Codogno P, Ramel D, Morel E, Laffargue M, Gayral S. PI3KCIIα-Dependent Autophagy Program Protects From Endothelial Dysfunction and Atherosclerosis in Response to Low Shear Stress in Mice. Arterioscler Thromb Vasc Biol 2024; 44:620-634. [PMID: 38152888 DOI: 10.1161/atvbaha.123.319978] [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: 08/08/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND The ability to respond to mechanical forces is a basic requirement for maintaining endothelial cell (ECs) homeostasis, which is continuously subjected to low shear stress (LSS) and high shear stress (HSS). In arteries, LSS and HSS have a differential impact on EC autophagy processes. However, it is still unclear whether LSS and HSS differently tune unique autophagic machinery or trigger specific autophagic responses in ECs. METHODS Using fluid flow system to generate forces on EC and multiscale imaging analyses on ApoE-/- mice whole arteries, we studied the cellular and molecular mechanism involved in autophagic response to LSS or HSS on the endothelium. RESULTS We found that LSS and HSS trigger autophagy activation by mobilizing specific autophagic signaling modules. Indeed, LSS-induced autophagy in endothelium was independent of the class III PI3K (phosphoinositide 3-kinase) VPS34 (vacuolar sorting protein 34) but controlled by the α isoform of class II PI3K (phosphoinositide 3-kinase class II α [PI3KCIIα]). Accordingly, reduced PI3KCIIα expression in ApoE-/- mice (ApoE-/-PI3KCIIα+/-) led to EC dysfunctions associated with increased plaque deposition in the LSS regions. Mechanistically, we revealed that PI3KCIIα inhibits mTORC1 (mammalian target of rapamycin complex 1) activation and that rapamycin treatment in ApoE-/-PI3KCIIα+/- mice specifically rescue autophagy in arterial LSS regions. Finally, we demonstrated that absence of PI3KCIIα led to decreased endothelial primary cilium biogenesis in response to LSS and that ablation of primary cilium mimics PI3KCIIα-decreased expression in EC dysfunction, suggesting that this organelle could be the mechanosensor linking PI3KCIIα and EC homeostasis. CONCLUSIONS Our data reveal that mechanical forces variability within the arterial system determines EC autophagic response and supports a central role of PI3KCIIα/mTORC1 axis to prevent EC dysfunction in LSS regions.
Collapse
Affiliation(s)
- Mouin Nasr
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Alexis Fay
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Adrien Lupieri
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Nicole Malet
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Anne Darmon
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Rana Zahreddine
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Audrey Swiader
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Amandine Wahart
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Julien Viaud
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Anne Nègre-Salvayre
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Emilio Hirsch
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy (E.H.)
| | - Daniel Monteyne
- IBMM-DBM, Department of Molecular Parasitology, University of Brussels, Gosselies, Belgium (D.M., D.P.-M.)
| | - David Perez-Morgà
- IBMM-DBM, Department of Molecular Parasitology, University of Brussels, Gosselies, Belgium (D.M., D.P.-M.)
| | - Nicolas Dupont
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, France (N.D., P.C., E.M.)
| | - Patrice Codogno
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, France (N.D., P.C., E.M.)
| | - Damien Ramel
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Etienne Morel
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, France (N.D., P.C., E.M.)
| | - Muriel Laffargue
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| | - Stephanie Gayral
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut national de la Santé et de la Recherche (INSERM) 1297, University of Toulouse 3, France (M.N., A.F., A.L., N.M., A.D., R.Z., A.S., A.W., J.V., A.N.-S., D.R., M.L., S.G.)
| |
Collapse
|
40
|
Li Z, Zou X, Lu R, Wan X, Sun S, Wang S, Qu Y, Zhang Y, Li Z, Yang L, Fang S. Arsenic trioxide alleviates atherosclerosis by inhibiting CD36-induced endocytosis and TLR4/NF-κB-induced inflammation in macrophage and ApoE -/- mice. Int Immunopharmacol 2024; 128:111452. [PMID: 38237221 DOI: 10.1016/j.intimp.2023.111452] [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: 08/10/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND Inflammation and lipid accumulation are key events in atherosclerosis progression. Despite arsenic trioxide's (ATO) toxicity, at appropriate doses, it is a useful treatment for various diseases treatment. ATO prevents vascular restenosis; however, its effects on atherosclerotic plaque development and instability remain unclear. METHODS ApoE-/- mice were fed high-fat diet for 4 months, and starting at the third month, ATO was intravenously administered every other day. Atherosclerotic lesion size, histological characteristics, and related protein and lipid profiles were assessed using samples from the aorta, carotid artery, and serum. The anti-inflammatory and anti-pyroptosis effects of ATO were investigated by stimulating RAW264.7 and THP-1 cell lines with oxidized low-density lipoprotein (ox-LDL) or lipopolysaccharide (LPS). RESULTS ATO reduced atherosclerotic lesion formation and plasma lipid levels in ApoE-/- mice. In the serum and aortic plaques, ATO reduced the levels of pro-inflammatory factors, including interleukin (IL) 6 and tumor necrosis factor α, but increased IL-10 levels. Mechanistically, ATO promoted the CD36-mediated internalization of ox-LDL in a peroxisome proliferator-activated receptor γ-dependent manner. Furthermore, ATO downregulated Toll-like receptor 4 (TLR4) expression in plaques and macrophages and inhibited p65 nuclear translocation and IκBα degradation. ATO reduced macrophage pyroptosis by downregulating NLR family pyrin domain-containing 3 (NLRP3) expression and caspase 1 activation. CONCLUSION ATO has potential atheroprotective effects, especially in macrophages. The mechanisms were inhibition of CD36-mediated foam cell formation and suppression of inflammatory responses and pyroptosis mediated by TLR4/nuclear factor κB and NLRP3 activation. Our findings provide evidence supporting the potential atheroprotective value of ATO.
Collapse
Affiliation(s)
- Zhaoying Li
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiaoyi Zou
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Rongzhe Lu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xin Wan
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China; Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Song Sun
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China; Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Shanjie Wang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yinan Qu
- Department of Cardiac Function, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Yun Zhang
- Univ Texas MD Anderson Canc Ctr, Dept Clin Canc Prevent, Houston, TX 77030 USA
| | - Zhangyi Li
- Department of biochemistry and life sciences, Faculty of Arts and Sciences, Queen's University, Kingston, Ontario, Canada
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang Province, China.
| | - Shaohong Fang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
41
|
Li Y, Xiong Z, Jiang WL, Tian D, Zhou H, Hou Q, Xiao L, Zhang M, Huang L, Zhong L, Zhou L, Zeng GG. An innovative viewpoint on the existing and prospectiveness of SR-B1. Curr Probl Cardiol 2024; 49:102226. [PMID: 38040207 DOI: 10.1016/j.cpcardiol.2023.102226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
Scavenger Receptor Class B Type 1 (SR-B1), a receptor protein expressed on the cell membrane, plays a crucial role in the metabolism and transport of cholesterol and other lipids, contributing significantly to the homeostasis of lipid levels within the body. Bibliometric analysis involves the application of mathematical and statistical methods to quantitatively analyze different types of documents. It involves the analysis of structural and temporal trends in scholarly articles, coupled with the identification of subject emphasis and variations. Through a bibliometric analysis, this study examines the historical background, current research trends, and future directions in the exploration of SR-B1. By offering insights into the research status and development of SR-B1, this paper aims to assist researchers in identifying novel pathways and areas of investigation in this field of study. Following the screening process, it can be concluded that research on SR-B1 has consistently remained a topic of significant interest over the past 17 years. Interestingly, SR-B1 has recently garnered attention in areas beyond its traditional research focus, including the field of cancer. The primary objective of this review is to provide a concise and accessible overview of the development process of SR-B1 that can help readers who are not well-versed in SR-B1 research quickly grasp its key aspects. Furthermore, this review aims to offer insights and suggestions to researchers regarding potential future research directions and areas of emphasis relating to SR-B1.
Collapse
Affiliation(s)
- Yonggui Li
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhijie Xiong
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wan-Li Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Dandan Tian
- School of Nursing, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Haiyou Zhou
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Qin Hou
- Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Liang Xiao
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Mengjie Zhang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Liubin Huang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Lianping Zhong
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Gastroenterology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Li Zhou
- Department of Pathology, Chongqing Public Health Medical Center, Southwest University Public Health Hospital, Chongqing, China
| | - Guang-Gui Zeng
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
42
|
Su Z, Wang J, Xiao C, Zhong W, Liu J, Liu X, Zhu YZ. Functional role of Ash2l in oxLDL induced endothelial dysfunction and atherosclerosis. Cell Mol Life Sci 2024; 81:62. [PMID: 38280036 PMCID: PMC10821849 DOI: 10.1007/s00018-024-05130-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/29/2024]
Abstract
Endothelial injury and dysfunction in the artery wall fuel the process of atherosclerosis. As a key epigenetic regulator, Ash2l (Absent, small, or homeotic-Like 2) is involved in regulating vascular injury and its complications. However, the role of Ash2l in atherosclerosis has not yet been fully elucidated. Here, we found increased Ash2l expression in high-cholesterol diet-fed ApoE-/- mice and oxidized LDL (oxLDL) treated endothelial cells (ECs). Furthermore, Ash2l promoted the scavenger receptors transcription by catalyzing histone H3 lysine 4 (H3K4) trimethylation at the promoter region of transcription factor peroxisome proliferator-activated receptor-γ (PPARγ) and triggered the activation of the pro-inflammatory nuclear factor-kappa B (NF-κB) by enhancing interaction between CD36 and toll-like receptor 4 (TLR4). Meanwhile, enhanced expression of scavenger receptors drove more oxLDL uptake by ECs. In vivo studies revealed that ECs-specific Ash2l knockdown reduced atherosclerotic lesion formation and promoted fibrous cap stability in the aorta of ApoE-/- mice, which was partly associated with a reduced endothelial activation by suppressing scavenger receptors and the uptake of lipids by ECs. Collectively, our findings identify Ash2l as a novel regulator that mediates endothelial injury and atherosclerosis. Targeting Ash2l may provide valuable insights for developing novel therapeutic candidates for atherosclerosis.
Collapse
Affiliation(s)
- Zhenghua Su
- School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Jinghuan Wang
- School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Chenxi Xiao
- School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Wen Zhong
- School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Jiayao Liu
- School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Xinhua Liu
- School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, China.
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, 825, Zhangheng Road, Pudong New District, Shanghai, China.
| | - Yi Zhun Zhu
- School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, China.
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy and 1st affiliate hospital, Macau University of Science and Technology, Macau, China.
- School of Pharmacy, Macau University of Science and Technology Taipa, Macau, China.
| |
Collapse
|
43
|
Bornfeldt KE. Apolipoprotein C3: form begets function. J Lipid Res 2024; 65:100475. [PMID: 37972731 PMCID: PMC10805671 DOI: 10.1016/j.jlr.2023.100475] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023] Open
Abstract
Increased circulating levels of apolipoprotein C3 (APOC3) predict cardiovascular disease (CVD) risk in humans, and APOC3 promotes atherosclerosis in mouse models. APOC3's mechanism of action is due in large part to its ability to slow the clearance of triglyceride-rich lipoproteins (TRLs) and their remnants when APOC3 is carried by these lipoproteins. However, different pools and forms of APOC3 exert distinct biological effects or associations with atherogenic processes. Thus, lipid-free APOC3 induces inflammasome activation in monocytes whereas lipid particle-bound APOC3 does not. APOC3-enriched LDL binds better to the vascular glycosaminoglycan biglycan than does LDL depleted of APOC3. Patterns of APOC3 glycoforms predict CVD risk differently. The function of APOC3 bound to HDL is largely unknown. There is still much to learn about the mechanisms of action of different forms and pools of APOC3 in atherosclerosis and CVD, and whether APOC3 inhibition would prevent CVD risk in patients on LDL-cholesterol lowering medications.
Collapse
Affiliation(s)
- Karin E Bornfeldt
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, UW Medicine Diabetes Institute and Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
44
|
Peng J, Yu L, Huang L, Paschoal VA, Chu H, de Souza CO, Varre JV, Oh DY, Kohler JJ, Xiao X, Xu L, Holland WL, Shaul PW, Mineo C. Hepatic sialic acid synthesis modulates glucose homeostasis in both liver and skeletal muscle. Mol Metab 2023; 78:101812. [PMID: 37777009 PMCID: PMC10583174 DOI: 10.1016/j.molmet.2023.101812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
OBJECTIVE Sialic acid is a terminal monosaccharide of glycans in glycoproteins and glycolipids, and its derivation from glucose is regulated by the rate-limiting enzyme UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE). Although the glycans on key endogenous hepatic proteins governing glucose metabolism are sialylated, how sialic acid synthesis and sialylation in the liver influence glucose homeostasis is unknown. Studies were designed to fill this knowledge gap. METHODS To decrease the production of sialic acid and sialylation in hepatocytes, a hepatocyte-specific GNE knockdown mouse model was generated, and systemic glucose metabolism, hepatic insulin signaling and glucagon signaling were evaluated in vivo or in primary hepatocytes. Peripheral insulin sensitivity was also assessed. Furthermore, the mechanisms by which sialylation in the liver influences hepatic insulin signaling and glucagon signaling and peripheral insulin sensitivity were identified. RESULTS Liver GNE deletion in mice caused an impairment of insulin suppression of hepatic glucose production. This was due to a decrease in the sialylation of hepatic insulin receptors (IR) and a decline in IR abundance due to exaggerated degradation through the Eph receptor B4. Hepatic GNE deficiency also caused a blunting of hepatic glucagon receptor (GCGR) function which was related to a decline in its sialylation and affinity for glucagon. An accompanying upregulation of hepatic FGF21 production caused an enhancement of skeletal muscle glucose disposal that led to an overall increase in glucose tolerance and insulin sensitivity. CONCLUSION These collective observations reveal that hepatic sialic acid synthesis and sialylation modulate glucose homeostasis in both the liver and skeletal muscle. By interrogating how hepatic sialic acid synthesis influences glucose control mechanisms in the liver, a new metabolic cycle has been identified in which a key constituent of glycans generated from glucose modulates the systemic control of its precursor.
Collapse
Affiliation(s)
- Jun Peng
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| | - Liming Yu
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Linzhang Huang
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Vivian A Paschoal
- Dept. of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Haiyan Chu
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Camila O de Souza
- Dept. of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Joseph V Varre
- Dept. of Nutrition & Integrative Physiology, University of Utah College of Health, 250 1850 E, Salt Lake City, UT, 84112, USA
| | - Da Young Oh
- Dept. of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Jennifer J Kohler
- Dept. of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Xue Xiao
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Lin Xu
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - William L Holland
- Dept. of Nutrition & Integrative Physiology, University of Utah College of Health, 250 1850 E, Salt Lake City, UT, 84112, USA
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA; Dept. of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| |
Collapse
|
45
|
Henry A, Lee WL. Unexpected Antiatherogenic Effect: Myeloid-Derived Growth Factor Inhibits LDL Transcytosis. Arterioscler Thromb Vasc Biol 2023; 43:2115-2118. [PMID: 37823266 DOI: 10.1161/atvbaha.123.320163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Affiliation(s)
- Andria Henry
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Canada (A.H., L.W.W.)
- Department of Biochemistry, University of Toronto, Canada (A.H., L.W.W.)
| | - Warren L Lee
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada (W.L.L.)
- Department of Medicine and the Interdepartmental Division of Critical Care Medicine, University of Toronto, Canada (W.L.L.)
- St. Michael's Hospital, Unity Health Toronto, Canada (W.L.L.)
| |
Collapse
|
46
|
Xu J, Ma H, Shi L, Zhou H, Cheng Y, Tong J, Meng B, Xu X, He K, Ding S, Zhang J, Yue L, Xiang G. Inflammatory Cell-Derived MYDGF Attenuates Endothelial LDL Transcytosis to Protect Against Atherogenesis. Arterioscler Thromb Vasc Biol 2023; 43:e443-e467. [PMID: 37767706 DOI: 10.1161/atvbaha.123.319905] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Inflammation contributes to the pathogenesis of atherosclerosis. But little is known about the potential benefits of inflammatory cells to atherosclerosis. The aim of this study was to investigate the function of inflammatory cells/endothelium axis and determine whether and how inflammatory cell-derived MYDGF (myeloid-derived growth factor) inhibited endothelial LDL (low-density lipoprotein) transcytosis. METHODS In in vivo experiments, both loss- and gain-of-function strategies were used to evaluate the effect of inflammatory cell-derived MYDGF on LDL transcytosis. We generated monocyte/macrophage-targeted MYDGF-null mice on an Ldlr (LDL receptor)-/- background in the loss-of-function strategy and restored the inflammatory cell-derived MYDGF by bone marrow transplantation and inflammatory cell-specific overexpression of MYDGF mice model in the gain-of-function strategy. In in vitro experiments, coculture experiments between primary mouse aortic endothelial cells and macrophages and mouse aortic endothelial cells supplemented with or without recombinant MYDGF were conducted. RESULTS Inflammatory cell-derived MYDGF deficiency aggravated endothelial LDL transcytosis, drove LDL uptake by artery wall, and thus exacerbated atherosclerosis in vivo. Inflammatory cell-derived MYDGF restoration by bone marrow transplantation and inflammatory cell MYDGF overexpression alleviated LDL transport across the endothelium, prevented LDL accumulation in the subendothelial space, and subsequently ameliorated atherosclerosis in vivo. Furthermore, in the in vitro study, macrophages isolated from MYDGF+/+ mice and recombinant MYDGF attenuated LDL transcytosis and uptake in mouse aortic endothelial cells. Mechanistically, MYDGF inhibited MAP4K4 (mitogen-activated protein kinase kinase kinase kinase isoform 4) phosphorylation, enhanced activation of Akt (protein kinase B)-1, and diminished the FoxO (forkhead box O) 3a signaling cascade to exert protective effects of MYDGF on LDL transcytosis and atherosclerosis. CONCLUSIONS The findings support a role for inflammatory cell-derived MYDGF served as a cross talk factor between inflammatory cells and endothelial cells that inhibits LDL transcytosis across endothelium. MYDGF may become a novel therapeutic drug for atherosclerosis, and the beneficial effects of inflammatory cell in atherosclerosis deserve further attention.
Collapse
Affiliation(s)
- Jinling Xu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China (J.X., L.S., Y.C., J.T., K.H., S.D., G.X.)
| | - Huaxing Ma
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, China (H.M.)
| | - Lingfeng Shi
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China (J.X., L.S., Y.C., J.T., K.H., S.D., G.X.)
| | - Hui Zhou
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Hunan, China (H.Z.)
| | - Yangyang Cheng
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, China (H.M.)
| | - Jiayue Tong
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China (J.X., L.S., Y.C., J.T., K.H., S.D., G.X.)
| | - Biying Meng
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
| | - Xiaoli Xu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
| | - Kaiyue He
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China (J.X., L.S., Y.C., J.T., K.H., S.D., G.X.)
| | - Sheng Ding
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China (J.X., L.S., Y.C., J.T., K.H., S.D., G.X.)
| | - Jiajia Zhang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
| | - Ling Yue
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
| | - Guangda Xiang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, China (J.X., L.S., Y.C., J.T., B.M., X.X., J.Z., L.Y., G.X.)
- The First School of Clinical Medicine, Southern Medical University, Guangdong, China (J.X., L.S., Y.C., J.T., K.H., S.D., G.X.)
| |
Collapse
|
47
|
Erlandsson L, Ohlsson L, Masoumi Z, Rehnström M, Cronqvist T, Edvinsson L, Hansson SR. Preliminary evidence that blocking the uptake of placenta-derived preeclamptic extracellular vesicles protects the vascular endothelium and prevents vasoconstriction. Sci Rep 2023; 13:18425. [PMID: 37891193 PMCID: PMC10611745 DOI: 10.1038/s41598-023-45830-9] [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: 07/18/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023] Open
Abstract
Preeclampsia (PE) is a pregnancy syndrome characterized by hypertension and organ damage manifesting after 20 gestational weeks. The etiology is of multifactorial origin, where placental stress causes increased levels of placenta-derived extracellular vesicles (STBEVs) in the maternal circulation, shown to cause inflammation, endothelial activation, vasoconstriction, and anti-angiogenic activity. General endothelial dysfunction is believed to be initiated by endothelial insult during pregnancy that alters vascular function resulting in increased arterial stiffness, cardiac dysfunction, and increased risk of cardiovascular disease later in life. We compared the effect of normal and PE derived STBEVs in vitro on vascular contractility of human subcutaneous arteries using wire myography. Cellular structures of exposed vessels were investigated by transmission electron microscopy. We explored strategies to pharmacologically block the effects of the STBEVs on human vessels. The PE STBEVs caused significantly stronger angiotensin II-mediated contractions and extended structural damage to human subcutaneous arteries compared to normal STBEVs. These negative effects could be reduced by blocking vesicle uptake by endothelial cells, using chlorpromazine or specific antibodies towards the LOX-1 receptor. The therapeutic potential of blocking vesicle uptake should be further explored, to reduce the permanent damage caused on the vasculature during PE pregnancy to prevent future cardiovascular risk.
Collapse
Affiliation(s)
- Lena Erlandsson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, BMC C14, Klinikgatan 28, 221 85, Lund, Sweden.
| | - Lena Ohlsson
- Experimental Vascular Research, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Zahra Masoumi
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, BMC C14, Klinikgatan 28, 221 85, Lund, Sweden
| | - Mimmi Rehnström
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, BMC C14, Klinikgatan 28, 221 85, Lund, Sweden
| | - Tina Cronqvist
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, BMC C14, Klinikgatan 28, 221 85, Lund, Sweden
| | - Lars Edvinsson
- Experimental Vascular Research, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Skåne University Hospital, Lund, Sweden
| | - Stefan R Hansson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, BMC C14, Klinikgatan 28, 221 85, Lund, Sweden
- Skåne University Hospital, Lund, Sweden
| |
Collapse
|
48
|
Oladosu O, Esobi IC, Powell RR, Bruce T, Stamatikos A. Dissecting the Impact of Vascular Smooth Muscle Cell ABCA1 versus ABCG1 Expression on Cholesterol Efflux and Macrophage-like Cell Transdifferentiation: The Role of SR-BI. J Cardiovasc Dev Dis 2023; 10:416. [PMID: 37887863 PMCID: PMC10607678 DOI: 10.3390/jcdd10100416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
Cholesterol-laden macrophages are recognized as a major contributor to atherosclerosis. However, recent evidence indicates that vascular smooth muscle cells (VSMC) that accumulate cholesterol and transdifferentiate into a macrophage-like cell (MLC) phenotype also play a role in atherosclerosis. Therefore, removing cholesterol from MLC may be a potential atheroprotective strategy. The two transporters which remove cholesterol from cells are ABCA1 and ABCG1, as they efflux cholesterol to apoAI and HDL, respectively. In this study, the well-characterized immortalized VSMC line MOVAS cells were edited to generate ABCA1- and ABCG1-knockout (KO) MOVAS cell lines. We cholesterol-loaded ABCA1-KO MOVAS cells, ABCG1-KO MOVAS cells, and wild-type MOVAS cells to convert cells into a MLC phenotype. When we measured apoAI- and HDL-mediated cholesterol efflux in these cells, we observed a drastic decrease in apoAI-mediated cholesterol efflux within ABCA1-KO MOVAS MLC, but HDL-mediated cholesterol efflux was only partially reduced in ABCG1-KO MOVAS cells. Since SR-BI also participates in HDL-mediated cholesterol efflux, we assessed SR-BI protein expression in ABCG1-KO MOVAS MLC and observed SR-BI upregulation, which offered a possible mechanism explaining why HDL-mediated cholesterol efflux remains maintained in ABCG1-KO MOVAS MLC. When we used lentivirus for shRNA-mediated knockdown of SR-BI in ABCG1-KO MOVAS MLC, this decreased HDL-mediated cholesterol efflux when compared to ABCG1-KO MOVAS MLC with unmanipulated SR-BI expression. Taken together, these major findings suggest that SR-BI expression in MLC of a VSMC origin plays a compensatory role in HDL-mediated cholesterol efflux when ABCG1 expression becomes impaired and provides insight on SR-BI demonstrating anti-atherogenic properties within VSMC/MLC.
Collapse
Affiliation(s)
- Olanrewaju Oladosu
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (O.O.); (I.C.E.)
| | - Ikechukwu C. Esobi
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (O.O.); (I.C.E.)
| | - Rhonda R. Powell
- Clemson Light Imaging Facility, Clemson University, Clemson, SC 29634, USA; (R.R.P.); (T.B.)
| | - Terri Bruce
- Clemson Light Imaging Facility, Clemson University, Clemson, SC 29634, USA; (R.R.P.); (T.B.)
| | - Alexis Stamatikos
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (O.O.); (I.C.E.)
| |
Collapse
|
49
|
Francis GA. The Greatly Under-Represented Role of Smooth Muscle Cells in Atherosclerosis. Curr Atheroscler Rep 2023; 25:741-749. [PMID: 37665492 PMCID: PMC10564813 DOI: 10.1007/s11883-023-01145-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 09/05/2023]
Abstract
PURPOSE OF REVIEW This article summarizes previous and recent research on the fundamental role of arterial smooth muscle cells (SMCs) as drivers of initial and, along with macrophages, later stages of human atherosclerosis. RECENT FINDINGS Studies using human tissues and SMC lineage-tracing mice have reinforced earlier observations that SMCs drive initial atherogenesis in humans and contribute a multitude of phenotypes including foam cell formation hitherto attributed primarily to macrophages in atherosclerosis. Arterial smooth muscle cells (SMCs) are the primary cell type in human pre-atherosclerotic intima and are responsible for the retention of lipoproteins that drive the development of atherosclerosis. Despite this, images of atherogenesis still depict the process as initially devoid of SMCs, primarily macrophage driven, and indicate only relatively minor roles such as fibrous cap formation to intimal SMCs. This review summarizes historical and recent observations regarding the importance of SMCs in the formation of a pre-atherosclerotic intima, initial and later foam cell formation, and the phenotypic changes that give rise to multiple different roles for SMCs in human and mouse lesions. Potential SMC-specific therapies in atherosclerosis are presented.
Collapse
Affiliation(s)
- Gordon A Francis
- Centre for Heart Lung Innovation, Providence Research, St. Paul's Hospital, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
50
|
Aronova A, Tosato F, Naser N, Asare Y. Innate Immune Pathways in Atherosclerosis-From Signaling to Long-Term Epigenetic Reprogramming. Cells 2023; 12:2359. [PMID: 37830572 PMCID: PMC10571887 DOI: 10.3390/cells12192359] [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: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
Innate immune pathways play a crucial role in the development of atherosclerosis, from sensing initial danger signals to the long-term reprogramming of immune cells. Despite the success of lipid-lowering therapy, anti-hypertensive medications, and other measures in reducing complications associated with atherosclerosis, cardiovascular disease (CVD) remains the leading cause of death worldwide. Consequently, there is an urgent need to devise novel preventive and therapeutic strategies to alleviate the global burden of CVD. Extensive experimental research and epidemiological studies have demonstrated the dominant role of innate immune mechanisms in the progression of atherosclerosis. Recently, landmark trials including CANTOS, COLCOT, and LoDoCo2 have provided solid evidence demonstrating that targeting innate immune pathways can effectively reduce the risk of CVD. These groundbreaking trials mark a significant paradigm shift in the field and open new avenues for atheroprotective treatments. It is therefore crucial to comprehend the intricate interplay between innate immune pathways and atherosclerosis for the development of targeted therapeutic interventions. Additionally, unraveling the mechanisms underlying long-term reprogramming may offer novel strategies to reverse the pro-inflammatory phenotype of immune cells and restore immune homeostasis in atherosclerosis. In this review, we present an overview of the innate immune pathways implicated in atherosclerosis, with a specific focus on the signaling pathways driving chronic inflammation in atherosclerosis and the long-term reprogramming of immune cells within atherosclerotic plaque. Elucidating the molecular mechanisms governing these processes presents exciting opportunities for the development of a new class of immunotherapeutic approaches aimed at reducing inflammation and promoting plaque stability. By addressing these aspects, we can potentially revolutionize the management of atherosclerosis and its associated cardiovascular complications.
Collapse
Affiliation(s)
| | | | | | - Yaw Asare
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), 80539 Munich, Germany
| |
Collapse
|