1
|
Yang G, Tan X, Zhai Q, Wang Y, Zhang X, Zhao P, Liang F, Lu J, Bao L. Plasma Lipidomics, Gut Microbiota Profile, and Phenotype of Adipose Tissue in an ApoE-/- Mouse Model of Plaque Instability. FRONT BIOSCI-LANDMRK 2025; 30:27236. [PMID: 40152393 DOI: 10.31083/fbl27236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/12/2025] [Accepted: 02/08/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND An appropriate animal model that can simulate the pathological process of atherosclerosis is urgently needed to improve treatment strategies. This study aimed to develop a new atherosclerosis model using ApoE-/- mice and to characterize lipidomics, gut microbiota profiles, and phenotypic alterations in adipose tissue using this model. METHODS After a 14- or 18-week high-fat diet (HFD), male ApoE-/- mice were randomly divided into four groups and treated separately with or without short-term and strong co-stimulation, including ice water bath and intraperitoneal injection of lipopolysaccharide and phenylephrine. As a control group, C57BL/6 mice were fed with conventional chow. The serum lipid levels, aortic arch pathology, adipose tissue phenotypic changes, plasma lipidomics, and 16S rDNA gene sequencing of colon feces were investigated. RESULTS The serum lipid levels were significantly lowered following extended HFD feeding for four weeks. However, co-stimulation increased serum interleukin (IL)-1β levels but did not affect serum lipid profiles. Co-stimulation revealed typical vulnerable atherosclerotic plaque characteristics and defective adipose hypertrophy associated with peroxisome proliferator-activated receptor γ (PPARγ) regulation in adipose tissue and a reduction in mitochondrial uncoupling protein 1 (UCP1) within brown adipose tissue. Plasma lipidomic analysis showed that sphingomyelin (SM), ceramide (Cer), and monohexosylceramide (HexCer) levels in plasma were significantly elevated by HFD feeding, whereas co-stimulation further elevated HexCer levels. Additionally, glycerophosphocholines (16:0/16:0, 18:2/20:4, 18:1/18:1) and HexCer (C12:1, C16:0), Cer (d18:1/16:0), and SM (C16:0) were the most sensitive to co-stimulation. Combined co-stimulation and HFD-fed increased the abundance of Firmicutes, the abundance of f_Erysipelotrichaceae, and the Firmicutes/Bacteroidota ratio but decreased the abundance of microflora promoting bile acid metabolism and short-chain fatty acids (SCFAs) in mouse feces. The results were consistent with the findings of epidemiologic atherosclerotic cardiovascular disease studies. CONCLUSIONS This study established an ApoE-/- mouse atherosclerotic vulnerable plaque model using a multi-index evaluation method. Adipogenic disorders, dysregulation of lipid metabolism at the molecular level, and increasing harmful gut microbiota are significant risk factors for vulnerable plaques, with sphingolipid metabolism receiving the most attention.
Collapse
Affiliation(s)
- Guanlin Yang
- Laboratory of Pharmacology, Zaozhuang Thoracic Hospital, 277500 Zaozhuang, Shandong, China
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Xin Tan
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Qiong Zhai
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Yuewu Wang
- School of Pharmacy, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Xuan Zhang
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Pengwei Zhao
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Fangyuan Liang
- School of Pharmacy, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Jingkun Lu
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - LiLi Bao
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| |
Collapse
|
2
|
Li X, Yang Y, Wang Z, Lin X, Fu X, He X, Liu M, Wang JX, Yu T, Sun P. CircHIPK3 targets DRP1 to mediate hydrogen peroxide-induced necroptosis of vascular smooth muscle cells and atherosclerotic vulnerable plaque formation. J Adv Res 2025; 69:329-341. [PMID: 38621622 PMCID: PMC11954820 DOI: 10.1016/j.jare.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/21/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024] Open
Abstract
INTRODUCTION Necroptosis triggered by H2O2 is hypothesized to be a critical factor in the rupture of atherosclerotic plaques, which may precipitate acute cardiovascular events. Nevertheless, the specific regulatory molecules of this development remain unclear. We aims to elucidate a mechanism from the perspective of circular RNA. OBJECTIVES There are few studies on circRNA in VSMCs necroptosis. The objective of our research is to shed light on the intricate roles that circHIPK3 plays in the process of necroptosis in VSMCs and the development of atherosclerotic plaques that are prone to rupture. Our study elucidates the specific molecular mechanisms by which circHIPK3 regulates necroptosis and atherosclerotic vulnerable plaque formation through targeted proteins. Identifying this mechanism at the cellular level offers a molecular framework for understanding plaque progression and stability regulation, as well as a potential biomarker for the prognosis of susceptible atherosclerotic plaques. METHODS We collected clinical vascular tissue for HE staining and Masson staining to determine the presence and stability of plaques. Then, NCBI database was used to screen out circRNA with elevated expression level in plaque tissue, and the up-regulated circRNA, circHIPK3, was verified by qRT-PCR and FISH. Further, we synthesized circHIPK3's small interference sequence and overexpressed plasmid in vitro, and verified its regulation effect on necroptosis of VSMCs under physiological and pathological conditions by WB, qRT-PCR and PI staining. Through RNA pull down, mass spectrometry and RNA immunoprecipitation, DRP1 was identified as circHIPK3 binding protein and was positively regulated by circHIPK3. Meanwhile, on the basis of silencing of DRP1, the regulation of circHIPK3 on necroptosis is verified to be mediated by DRP1. Finally, we validated the regulation of circHIPK3 on vulnerable plaque formation in ApoE-/- mice. RESULTS We investigated that circHIPK3 was highly expressed in vulnerable plaques, and the increase in expression level promoted H2O2 induced necroptosis of VSMCs. CircHIPK3 targeted the protein DRP1, leading to an elevation in mitochondrial division rate, resulting in increased reactive oxygen species and impaired mitochondrial function, ultimately leading to necroptosis of VSMCs and vulnerable plaque formation. CONCLUSION CircHIPK3 interact with DRP1 involve in H2O2 induced Mitochondrial damage and necroptosis of VSMCs, and Silencing circHIPK3 in vivo can reduce atherosclerotic vulnerable plaque formation. Our research findings may have applications in providing diagnostic biomarkers for vulnerable plaques.
Collapse
MESH Headings
- Plaque, Atherosclerotic/pathology
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/genetics
- Animals
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Necroptosis/drug effects
- Necroptosis/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/cytology
- Humans
- Mice
- Hydrogen Peroxide/pharmacology
- Dynamins/metabolism
- Dynamins/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Male
- Mice, Inbred C57BL
- Cells, Cultured
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Mice, Knockout, ApoE
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Intracellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao 266071, People's Republic of China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China
| | - Xiaotong Lin
- Department of Respiratory Medicine, Qingdao Municipal Hospital, Qingdao 266011, People's Republic of China
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China
| | - Xiangqin He
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China
| | - Meixin Liu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China
| | - Jian-Xun Wang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao 266071, People's Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China; Center for Regenerative Medicine, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China.
| | - Pin Sun
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People's Republic of China.
| |
Collapse
|
3
|
Hao R, Li H, Li X, Liu J, Ji X, Zhang H, Zhang Z, Yang P, Zhai Z. Transcriptomic profiling of lncRNAs and mRNAs in a venous thrombosis mouse model. iScience 2025; 28:111561. [PMID: 39949957 PMCID: PMC11821396 DOI: 10.1016/j.isci.2024.111561] [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: 09/16/2024] [Revised: 10/25/2024] [Accepted: 12/05/2024] [Indexed: 02/16/2025] Open
Abstract
This study explores the role of lncRNAs and mRNAs in venous thromboembolism (VTE) using an inferior vena cava (IVC) mouse model. RNA sequencing identified differentially expressed lncRNAs and mRNAs between model and control groups. Enrichment analyses revealed significant pathways, including HIF-1α signaling, glycolysis/gluconeogenesis, and platelet activation. A lncRNA-miRNA-mRNA network highlighted key regulatory interactions. Validation using qRT-PCR confirmed the RNA-seq findings. These results provide insights into the molecular mechanisms of VTE and suggest potential biomarkers and therapeutic targets for thrombosis.
Collapse
Affiliation(s)
- Risheng Hao
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Capital Medical University, Beijing, China
| | - Haobo Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xincheng Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jixiang Liu
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xiaofan Ji
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hong Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Zhu Zhang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Peiran Yang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Zhenguo Zhai
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
4
|
Deng WQ, Ye ZH, Tang Z, Zhang XL, Lu JJ. Beyond cancer: The potential application of CD47-based therapy in non-cancer diseases. Acta Pharm Sin B 2025; 15:757-791. [PMID: 40177549 PMCID: PMC11959971 DOI: 10.1016/j.apsb.2024.11.018] [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: 08/29/2024] [Revised: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
CD47 is an immune checkpoint widely regarded as a 'don't eat me' signal. CD47-based anti-cancer therapy has received considerable attention, with a significant number of clinical trials conducted. While anti-cancer therapies based on CD47 remain a focal point of interest among researchers, it is noteworthy that an increasing number of studies have found that CD47-based therapy ameliorated the pathological status of non-cancer diseases. This review aims to provide an overview of the recent progress in comprehending the role of CD47-based therapy in non-cancer diseases, including diseases of the circulatory system, nervous system, digestive system, and so on. Furthermore, we sought to delineate the promising mechanisms of CD47-based therapy in treating non-cancer diseases. Our findings suggest that CD47-based agents may exert their effect by regulating phagocytosis, regulating T cells, dendritic cells, and neutrophils, and regulating the secretion of cytokines and chemokines. Additionally, we put forward the orientation of further research to bring to light the potential of CD47 and its binding partners as a target in non-cancer diseases.
Collapse
Affiliation(s)
- Wei-Qing Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zhenghai Tang
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
| | - Xiao-Lei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao 999078, China
| |
Collapse
|
5
|
Mangarova DB, Kaufmann JO, Brangsch J, Kader A, Möckel J, Heyl JL, Verlemann C, Adams LC, Ludwig A, Reimann C, Poller WC, Niehaus P, Karst U, Taupitz M, Hamm B, Weller MG, Makowski MR. ADAMTS4-Specific MR Peptide Probe for the Assessment of Atherosclerotic Plaque Burden in a Mouse Model. Invest Radiol 2025:00004424-990000000-00282. [PMID: 39804796 DOI: 10.1097/rli.0000000000001152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
INTRODUCTION Atherosclerosis is the underlying cause of multiple cardiovascular pathologies. The present-day clinical imaging modalities do not offer sufficient information on plaque composition or rupture risk. A disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) is a strongly upregulated proteoglycan-cleaving enzyme that is specific to cardiovascular diseases, inter alia, atherosclerosis. MATERIALS AND METHODS Male apolipoprotein E-deficient mice received a high-fat diet for 2 (n = 11) or 4 months (n = 11). Additionally, a group (n = 11) receiving pravastatin by drinking water for 4 months alongside the high-fat diet was examined. The control group (n = 10) consisted of C57BL/6J mice on standard chow. Molecular magnetic resonance imaging was performed prior to and after administration of the gadolinium (Gd)-based ADAMTS4-specific probe, followed by ex vivo analyses of the aortic arch, brachiocephalic arteries, and carotid arteries. A P value <0.05 was considered to indicate a statistically significant difference. RESULTS With advancing atherosclerosis, a significant increase in the contrast-to-noise ratio was measured after intravenous application of the probe (mean precontrast = 2.25; mean postcontrast = 11.47, P < 0.001 in the 4-month group). The pravastatin group presented decreased ADAMTS4 expression. A strong correlation between ADAMTS4 content measured via immunofluorescence staining and an increase in the contrast-to-noise ratio was detected ( R2 = 0.69). Microdissection analysis revealed that ADAMTS4 gene expression in the plaque area was significantly greater than that in the arterial wall of a control mouse ( P < 0.001). Laser ablation-inductively coupled plasma-mass spectrometry confirmed strong colocalization of areas positive for ADAMTS4 and Gd. CONCLUSIONS Magnetic resonance imaging using an ADAMTS4-specific agent is a promising method for characterizing atherosclerotic plaques and could improve plaque assessment in the diagnosis and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Dilyana B Mangarova
- From the Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany (D.B.M., J.O.K., J.B., A.K., J.M., J.L.H., C.R., M.T., B.H., M.R.M.); Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany (D.B.M., J.O.K., J.B., A.K., L.C.A., M.R.M.); Department of Chemistry, Humboldt-Universität zu Berlin, Berlin, Germany (J.O.K.); Division 1.5 Protein Analysis, Federal Institute for Materials Research and Testing, Berlin, Germany (J.O.K., M.G.W.); Department of Biology, Chemistry, and Pharmacy, Institute of Biology, Freie Universität Berlin, Berlin, Germany (A.K.); Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Berlin, Germany (J.L.H.); Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany (C.V., P.N., U.K.); Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany (A.L.); DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany (A.L.); and Division of Cardiology, Massachusetts General Hospital, Harvard University, Boston, MA (W.C.P.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Lin A, Miano JM, Fisher EA, Misra A. Chronic inflammation and vascular cell plasticity in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1408-1423. [PMID: 39653823 DOI: 10.1038/s44161-024-00569-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/10/2024] [Indexed: 12/13/2024]
Abstract
Vascular smooth muscle cells, endothelial cells and macrophages undergo phenotypic conversions throughout atherosclerosis progression, both as a consequence of chronic inflammation and as subsequent drivers of it. The inflammatory hypothesis of atherosclerosis has been catapulted to the forefront of cardiovascular research as clinical trials have shown that anti-inflammatory therapy reduces adverse cardiovascular events. However, no current therapies have been specifically designed to target the phenotype of plaque cells. Fate mapping has revealed that plaque cells convert to detrimental and beneficial cell phenotypes during atherosclerosis, with cumulative evidence highlighting that vascular cell plasticity is intimately linked with plaque inflammation, ultimately impacting lesion stability. Here we review vascular cell plasticity during atherosclerosis in the context of the chronic inflammatory plaque microenvironment. We highlight the need to better understand how plaque cells behave during therapeutic intervention. We then propose modulating plaque cell phenotype as an unexplored therapeutic paradigm in the clinical setting.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodelling Group, Heart Research Institute, Sydney, New South Wales, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Joseph M Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Ashish Misra
- Atherosclerosis and Vascular Remodelling Group, Heart Research Institute, Sydney, New South Wales, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
7
|
Hong NE, Chaplin A, Di L, Ravodina A, Bevan GH, Gao H, Asase C, Gangwar RS, Cameron MJ, Mignery M, Cherepanova O, Finn AV, Nayak L, Pieper AA, Maiseyeu A. Nanoparticle-based itaconate treatment recapitulates low-cholesterol/low-fat diet-induced atherosclerotic plaque resolution. Cell Rep 2024; 43:114911. [PMID: 39466775 PMCID: PMC11648168 DOI: 10.1016/j.celrep.2024.114911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/22/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Current pharmacologic treatments for atherosclerosis do not completely protect patients; additional protection can be achieved by dietary modifications, such as a low-cholesterol/low-fat diet (LCLFD), that mediate plaque stabilization and inflammation reduction. However, this lifestyle modification can be challenging for patients. Unfortunately, incomplete understanding of the underlying mechanisms has thwarted efforts to mimic the protective effects of a LCLFD. Here, we report that the tricarboxylic acid cycle intermediate itaconate (ITA), produced by plaque macrophages, is key to diet-induced plaque resolution. ITA is produced by immunoresponsive gene 1 (IRG1), which we observe is highly elevated in myeloid cells of vulnerable plaques and absent from early or stable plaques in mice and humans. We additionally report development of an ITA-conjugated lipid nanoparticle that accumulates in plaque and bone marrow myeloid cells, epigenetically reduces inflammation via H3K27ac deacetylation, and reproduces the therapeutic effects of LCLFD-induced plaque resolution in multiple atherosclerosis models.
Collapse
Affiliation(s)
- Natalie E Hong
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Alice Chaplin
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Lin Di
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Anastasia Ravodina
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Graham H Bevan
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Huiyun Gao
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Courteney Asase
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Roopesh Singh Gangwar
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Division of Allergy and Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Mark J Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Matthew Mignery
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Olga Cherepanova
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Aloke V Finn
- Department of Internal Medicine, Cardiovascular Division, University of Maryland School of Medicine, Baltimore, MD, USA; CVPath Institute, Inc., Gaithersburg, MD, USA
| | - Lalitha Nayak
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Hematology & Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH, USA; Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Andrei Maiseyeu
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
8
|
Zeller J, Loseff-Silver J, Khoshmanesh K, Baratchi S, Lai A, Nero TL, Roy A, Watson A, Dayawansa N, Sharma P, Barbaro-Wahl A, Chen YC, Moon M, Vidallon MLP, Huang A, Thome J, Cheung Tung Shing KS, Harvie D, Bongiovanni MN, Braig D, Morton CJ, Htun NM, Stub D, Walton A, Horowitz J, Wang X, Pietersz G, Parker MW, Eisenhardt SU, McFadyen JD, Peter K. Shear-Sensing by C-Reactive Protein: Linking Aortic Stenosis and Inflammation. Circ Res 2024; 135:1033-1047. [PMID: 39421928 PMCID: PMC11542976 DOI: 10.1161/circresaha.124.324248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND CRP (C-reactive protein) is a prototypical acute phase reactant. Upon dissociation of the pentameric isoform (pCRP [pentameric CRP]) into its monomeric subunits (mCRP [monomeric CRP]), it exhibits prothrombotic and proinflammatory activity. Pathophysiological shear rates as observed in aortic valve stenosis (AS) can influence protein conformation and function as observed with vWF (von Willebrand factor). Given the proinflammatory function of dissociated CRP and the important role of inflammation in the pathogenesis of AS, we investigated whether shear stress can modify CRP conformation and induce inflammatory effects relevant to AS. METHODS To determine the effects of pathological shear rates on the function of human CRP, pCRP was subjected to pathophysiologically relevant shear rates and analyzed using biophysical and biochemical methods. To investigate the effect of shear on CRP conformation in vivo, we used a mouse model of arterial stenosis. Levels of mCRP and pCRP were measured in patients with severe AS pre- and post-transcatheter aortic valve implantation, and the presence of CRP was investigated on excised valves from patients undergoing aortic valve replacement surgery for severe AS. Microfluidic models of AS were then used to recapitulate the shear rates of patients with AS and to investigate this shear-dependent dissociation of pCRP and its inflammatory function. RESULTS Exposed to high shear rates, pCRP dissociates into its proinflammatory monomers (mCRP) and aggregates into large particles. Our in vitro findings were further confirmed in a mouse carotid artery stenosis model, where the administration of human pCRP led to the deposition of mCRP poststenosis. Patients undergoing transcatheter aortic valve implantation demonstrated significantly higher mCRP bound to circulating microvesicles pre-transcatheter aortic valve implantation compared with post-transcatheter aortic valve implantation. Excised human stenotic aortic valves display mCRP deposition. pCRP dissociated in a microfluidic model of AS and induces endothelial cell activation as measured by increased ICAM-1 (intercellular adhesion molecule 1) and P-selectin expression. mCRP also induces platelet activation and TGF-β (transforming growth factor beta) expression on platelets. CONCLUSIONS We identify a novel mechanism of shear-induced pCRP dissociation, which results in the activation of cells central to the development of AS. This novel mechanosensing mechanism of pCRP dissociation to mCRP is likely also relevant to other pathologies involving increased shear rates, such as in atherosclerotic and injured arteries.
Collapse
Affiliation(s)
- Johannes Zeller
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
- Department of Plastic and Hand Surgery, Medical Center, Faculty of Medicine, University of Freiburg, Breisgau, Germany (J.Z., J.T., D.B., S.U.E.)
| | - Julia Loseff-Silver
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
| | | | - Sara Baratchi
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
- Department of Cardiometabolic Health (S.B., T.L.N., A.R., M.M., M.L.P.V., K.S.C.T.S., C.J.M., X.W., M.W.P., J.D.M., K.P.), Clayton, Victoria, Australia
| | - Austin Lai
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
| | - Tracy L. Nero
- Department of Cardiometabolic Health (S.B., T.L.N., A.R., M.M., M.L.P.V., K.S.C.T.S., C.J.M., X.W., M.W.P., J.D.M., K.P.), Clayton, Victoria, Australia
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute (T.L.N., K.S.C.T.S., C.J.M., M.W.P.), Clayton, Victoria, Australia
| | - Abhishek Roy
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
- Department of Cardiometabolic Health (S.B., T.L.N., A.R., M.M., M.L.P.V., K.S.C.T.S., C.J.M., X.W., M.W.P., J.D.M., K.P.), Clayton, Victoria, Australia
| | - Anna Watson
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
| | - Nalin Dayawansa
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
- Department of Cardiology, Alfred Hospital, Melbourne, Victoria, Australia (N.D., N.M.H., D.S., A. Walton, K.P.)
| | - Prerna Sharma
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
| | - Anastasia Barbaro-Wahl
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
| | - Yung Chih Chen
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
| | - Mitchell Moon
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
- Department of Cardiometabolic Health (S.B., T.L.N., A.R., M.M., M.L.P.V., K.S.C.T.S., C.J.M., X.W., M.W.P., J.D.M., K.P.), Clayton, Victoria, Australia
| | - Mark Louis P. Vidallon
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
- Department of Cardiometabolic Health (S.B., T.L.N., A.R., M.M., M.L.P.V., K.S.C.T.S., C.J.M., X.W., M.W.P., J.D.M., K.P.), Clayton, Victoria, Australia
| | - Angela Huang
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
| | - Julia Thome
- Department of Plastic and Hand Surgery, Medical Center, Faculty of Medicine, University of Freiburg, Breisgau, Germany (J.Z., J.T., D.B., S.U.E.)
| | - Karen S. Cheung Tung Shing
- Department of Cardiometabolic Health (S.B., T.L.N., A.R., M.M., M.L.P.V., K.S.C.T.S., C.J.M., X.W., M.W.P., J.D.M., K.P.), Clayton, Victoria, Australia
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute (T.L.N., K.S.C.T.S., C.J.M., M.W.P.), Clayton, Victoria, Australia
| | - Dalton Harvie
- Department of Chemical Engineering (D.H., M.N.B.), Clayton, Victoria, Australia
| | | | - David Braig
- Department of Plastic and Hand Surgery, Medical Center, Faculty of Medicine, University of Freiburg, Breisgau, Germany (J.Z., J.T., D.B., S.U.E.)
| | - Craig J. Morton
- Department of Cardiometabolic Health (S.B., T.L.N., A.R., M.M., M.L.P.V., K.S.C.T.S., C.J.M., X.W., M.W.P., J.D.M., K.P.), Clayton, Victoria, Australia
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute (T.L.N., K.S.C.T.S., C.J.M., M.W.P.), Clayton, Victoria, Australia
- The University of Melbourne, Parkville, Victoria, Australia; now with CSIRO Biomedical Manufacturing, Clayton, Victoria, Australia (C.J.M.)
| | - Nay M. Htun
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
- Department of Cardiology, Alfred Hospital, Melbourne, Victoria, Australia (N.D., N.M.H., D.S., A. Walton, K.P.)
| | - Dion Stub
- Department of Cardiology, Alfred Hospital, Melbourne, Victoria, Australia (N.D., N.M.H., D.S., A. Walton, K.P.)
- Department of Epidemiology and Preventive Medicine (D.S.), Melbourne, Victoria, Australia
| | - Anthony Walton
- Department of Cardiology, Alfred Hospital, Melbourne, Victoria, Australia (N.D., N.M.H., D.S., A. Walton, K.P.)
| | - John Horowitz
- Department of Cardiology, Queen Elizabeth Hospital, Adelaide, South Australia, Australia (J.H.)
| | - Xiaowei Wang
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
- Department of Cardiometabolic Health (S.B., T.L.N., A.R., M.M., M.L.P.V., K.S.C.T.S., C.J.M., X.W., M.W.P., J.D.M., K.P.), Clayton, Victoria, Australia
| | - Geoffrey Pietersz
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
| | - Michael W. Parker
- Department of Cardiometabolic Health (S.B., T.L.N., A.R., M.M., M.L.P.V., K.S.C.T.S., C.J.M., X.W., M.W.P., J.D.M., K.P.), Clayton, Victoria, Australia
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute (T.L.N., K.S.C.T.S., C.J.M., M.W.P.), Clayton, Victoria, Australia
- ACRF Rational Drug Discovery Centre, St Vincent’s Institute of Medical Research, Fitzroy, Victoria, Australia (M.W.P.)
| | - Steffen U. Eisenhardt
- Department of Plastic and Hand Surgery, Medical Center, Faculty of Medicine, University of Freiburg, Breisgau, Germany (J.Z., J.T., D.B., S.U.E.)
| | - James D. McFadyen
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
- Department of Cardiometabolic Health (S.B., T.L.N., A.R., M.M., M.L.P.V., K.S.C.T.S., C.J.M., X.W., M.W.P., J.D.M., K.P.), Clayton, Victoria, Australia
- Department of Clinical Haematology (J.D.M), School of Translational Medicine Monash University, Melbourne, Victoria, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.Z., J.L.-S., S.B., A.L., A.R., A. Watson, N.D., P.S., A.B.-W., Y.C.C., M.M., M.L.P.V., A.H., N.M.H., X.W., G.P., J.D.M., K.P.)
- Department of Cardiometabolic Health (S.B., T.L.N., A.R., M.M., M.L.P.V., K.S.C.T.S., C.J.M., X.W., M.W.P., J.D.M., K.P.), Clayton, Victoria, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Victoria, Australia (N.D., N.M.H., D.S., A. Walton, K.P.)
| |
Collapse
|
9
|
Cao J, Roth S, Zhang S, Kopczak A, Mami S, Asare Y, Georgakis MK, Messerer D, Horn A, Shemer R, Jacqmarcq C, Picot A, Green JP, Schlegl C, Li X, Tomas L, Dutsch A, Liman TG, Endres M, Wernsdorf SR, Fürle C, Carofiglio O, Zhu J, Brough D, Hornung V, Dichgans M, Vivien D, Schulz C, Dor Y, Tiedt S, Sager HB, Grosse GM, Liesz A. DNA-sensing inflammasomes cause recurrent atherosclerotic stroke. Nature 2024; 633:433-441. [PMID: 39112714 PMCID: PMC11390481 DOI: 10.1038/s41586-024-07803-4] [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: 01/28/2023] [Accepted: 07/09/2024] [Indexed: 08/17/2024]
Abstract
The risk of early recurrent events after stroke remains high despite currently established secondary prevention strategies1. Risk is particularly high in patients with atherosclerosis, with more than 10% of patients experiencing early recurrent events1,2. However, despite the enormous medical burden of this clinical phenomenon, the underlying mechanisms leading to increased vascular risk and recurrent stroke are largely unknown. Here, using a novel mouse model of stroke-induced recurrent ischaemia, we show that stroke leads to activation of the AIM2 inflammasome in vulnerable atherosclerotic plaques via an increase of circulating cell-free DNA. Enhanced plaque inflammation post-stroke results in plaque destabilization and atherothrombosis, finally leading to arterioarterial embolism and recurrent stroke within days after the index stroke. We confirm key steps of plaque destabilization also after experimental myocardial infarction and in carotid artery plaque samples from patients with acute stroke. Rapid neutrophil NETosis was identified as the main source of cell-free DNA after stroke and NET-DNA as the causative agent leading to AIM2 inflammasome activation. Neutralization of cell-free DNA by DNase treatment or inhibition of inflammasome activation reduced the rate of stroke recurrence after experimental stroke. Our findings present an explanation for the high recurrence rate after incident ischaemic events in patients with atherosclerosis. The detailed mechanisms uncovered here provide clinically uncharted therapeutic targets for which we show high efficacy to prevent recurrent events. Targeting DNA-mediated inflammasome activation after remote tissue injury represents a promising avenue for further clinical development in the prevention of early recurrent events.
Collapse
Affiliation(s)
- Jiayu Cao
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Stefan Roth
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany.
| | - Sijia Zhang
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Anna Kopczak
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Samira Mami
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Yaw Asare
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Programme in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Denise Messerer
- Medizinische Klinik und Poliklinik I, LMU University Hospital, LMU Munich, Munich, Germany
| | - Amit Horn
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel
| | - Ruth Shemer
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel
| | - Charlene Jacqmarcq
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Audrey Picot
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Jack P Green
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK
| | - Christina Schlegl
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Xinghai Li
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Lukas Tomas
- Medizinische Klinik und Poliklinik I, LMU University Hospital, LMU Munich, Munich, Germany
| | - Alexander Dutsch
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Thomas G Liman
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Endres
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Saskia R Wernsdorf
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Christina Fürle
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Olga Carofiglio
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Jie Zhu
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - David Brough
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK
| | - Veit Hornung
- Gene Center and Department of Biochemistry, LMU Munich, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Denis Vivien
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Caen, France
- Research Clinical Department, Caen Normandie University Hospital, Caen, France
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Immunopharmacology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Hendrik B Sager
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Gerrit M Grosse
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
10
|
Shamsuzzaman S, Deaton RA, Salamon A, Doviak H, Serbulea V, Milosek VM, Evans MA, Karnewar S, Saibaba S, Alencar GF, Shankman LS, Walsh K, Bekiranov S, Kocher O, Krieger M, Kull B, Persson M, Michaëlsson E, Bergenhem N, Heydarkhan-Hagvall S, Owens GK. Novel Mouse Model of Myocardial Infarction, Plaque Rupture, and Stroke Shows Improved Survival With Myeloperoxidase Inhibition. Circulation 2024; 150:687-705. [PMID: 38881440 PMCID: PMC11347105 DOI: 10.1161/circulationaha.123.067931] [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: 11/10/2023] [Accepted: 05/22/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Thromboembolic events, including myocardial infarction (MI) or stroke, caused by the rupture or erosion of unstable atherosclerotic plaques are the leading cause of death worldwide. Although most mouse models of atherosclerosis develop lesions in the aorta and carotid arteries, they do not develop advanced coronary artery lesions. Moreover, they do not undergo spontaneous plaque rupture with MI and stroke or do so at such a low frequency that they are not viable experimental models to study late-stage thrombotic events or to identify novel therapeutic approaches for treating atherosclerotic disease. This has stymied the development of more effective therapeutic approaches for reducing these events beyond what has been achieved with aggressive lipid lowering. Here, we describe a diet-inducible mouse model that develops widespread advanced atherosclerosis in coronary, brachiocephalic, and carotid arteries with plaque rupture, MI, and stroke. METHODS We characterized a novel mouse model with a C-terminal mutation in the scavenger receptor class B, type 1 (SR-BI), combined with Ldlr knockout (designated SR-BI∆CT/∆CT/Ldlr-/-). Mice were fed Western diet (WD) for 26 weeks and analyzed for MI and stroke. Coronary, brachiocephalic, and carotid arteries were analyzed for atherosclerotic lesions and indices of plaque stability. To validate the utility of this model, SR-BI∆CT/∆CT/Ldlr-/- mice were treated with the drug candidate AZM198, which inhibits myeloperoxidase, an enzyme produced by activated neutrophils that predicts rupture of human atherosclerotic lesions. RESULTS SR-BI∆CT/∆CT/Ldlr-/- mice show high (>80%) mortality rates after 26 weeks of WD feeding because of major adverse cardiovascular events, including spontaneous plaque rupture with MI and stroke. Moreover, WD-fed SR-BI∆CT/∆CT/Ldlr-/- mice displayed elevated circulating high-sensitivity cardiac troponin I and increased neutrophil extracellular trap formation within lesions compared with control mice. Treatment of WD-fed SR-BI∆CT/∆CT/Ldlr-/- mice with AZM198 showed remarkable benefits, including >90% improvement in survival and >60% decrease in the incidence of plaque rupture, MI, and stroke, in conjunction with decreased circulating high-sensitivity cardiac troponin I and reduced neutrophil extracellular trap formation within lesions. CONCLUSIONS WD-fed SR-BI∆CT/∆CT/Ldlr-/- mice more closely replicate late-stage clinical events of advanced human atherosclerotic disease than previous models and can be used to identify and test potential new therapeutic agents to prevent major adverse cardiac events.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Diet, Western/adverse effects
- Disease Models, Animal
- Enzyme Inhibitors/therapeutic use
- Enzyme Inhibitors/pharmacology
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardial Infarction/pathology
- Myocardial Infarction/drug therapy
- Peroxidase/metabolism
- Plaque, Atherosclerotic/drug therapy
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Rupture, Spontaneous
- Scavenger Receptors, Class B/genetics
- Scavenger Receptors, Class B/metabolism
- Stroke/drug therapy
- Stroke/prevention & control
Collapse
Affiliation(s)
- Sohel Shamsuzzaman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Rebecca A. Deaton
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Anita Salamon
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Heather Doviak
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Vlad Serbulea
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Victoria M. Milosek
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Molecular Physiology and Biophysics, University of Virginia, Charlottesville, VA, USA
| | - Megan A. Evans
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Santosh Karnewar
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Subhi Saibaba
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Gabriel F. Alencar
- Beirne B Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Laura S. Shankman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kenneth Walsh
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Stefan Bekiranov
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Olivier Kocher
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Medical Deaconess Medical Center Harvard Medical School, Boston, MA, USA
| | - Monty Krieger
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bengt Kull
- BioPharmaceuticals R&D Early Cardiovascular Renal and Metabolism (CVRM) Bioscience Cardiovascular, AstraZeneca, Mölndal 43183, Sweden
| | - Marie Persson
- BioPharmaceuticals R&D Early Cardiovascular Renal and Metabolism (CVRM) DMPK, AstraZeneca, Mölndal 43183, Sweden
| | - Erik Michaëlsson
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal, and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Nils Bergenhem
- Alliance Management, Business Development & Licensing, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA
| | - Sepideh Heydarkhan-Hagvall
- AstraZeneca R&D, Chief Medical Office, Global Patient Safety, Pepparedsleden 1, Mölndal, SE43183, Sweden
| | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Molecular Physiology and Biophysics, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
11
|
Liu Y, Wu Y, Wang C, Hu W, Zou S, Ren H, Zuo Y, Qu L. MiR-127-3p enhances macrophagic proliferation via disturbing fatty acid profiles and oxidative phosphorylation in atherosclerosis. J Mol Cell Cardiol 2024; 193:36-52. [PMID: 38795767 DOI: 10.1016/j.yjmcc.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/05/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Atherosclerosis is a chronic pathology, leading to acute coronary heart disease or stroke. MiR-127 has been found significantly upregulated in advanced atherosclerosis. But its function in atherosclerosis remains unexplored. We explored the role of miR-127-3p in regulating atherosclerosis development and its downstream mechanisms. METHODS The expression profile of miR-127 in carotid atherosclerotic plaques of 23 patients with severe carotid stenosis was detected by RT-qPCR and in situ hybridization. Primary bone marrow-derived macrophages (BMDM) stimulated with oxidized low-density lipoprotein were used as an in vitro model. CCK-8, EdU, RT-qPCR, and flow cytometry were used to detect the proliferative capacity and polarization of BMDM, which were infected by lentivirus-carrying plasmid to upregulate or downregulate miR-127-3p expression, respectively. RNA sequencing combined with bioinformatic analysis and targeted fatty acid metabolomics approach were used to detect the transcriptome and lipid metabolites. The association between miR-127-3p and its target was verified by dual-luciferase activity reporting and Western blotting. Oxygen consumption rate of BMDM were detected using seahorse analysis. High-cholesterol-diet-fed low density lipoprotein deficient (LDLR-/-) mice, with-or-without carotid tandem-stenosis surgery, were treated with miR-127-3p agomir or antagomir to examine its effect on plaque development and stability. RESULTS miR-127-3p, not -5p, is elevated in human advanced carotid atheroma and its expression is positively associated with macrophage accummulation in plaques. In vitro, miR-127-3p-overexpressed macrophage exhibites increased proliferation capacity and facilitates M1 polariztion whereas the contrary trend is present in miR-127-3p-inhibited macrophage. Stearoyl-CoA desaturase-1 (SCD1) is one potential target of miR-127-3p. miR-127-3p mimics decreases the activity of 3' untranslated regions of SCD-1. Furthermore, miR-127-3p downregulates SCD1 expression, and reversing the expression of SCD1 attenuates the increased proliferation induced by miR-127-3p overexpression in macrophage. miR-127-3p overexpression could also lead to decreased content of unsaturated fatty acids (UFAs), increased content of acetyl CoA and increased level of oxidative phosphorylation. In vivo, miR-127-3p agomir significantly increases atherosclerosis progression, macrophage proliferation and decreases SCD1 expression and the content of UFAs in aortic plaques of LDLR-/- mice. Conversely, miR-127-3p antagomir attenuated atherosclerosis, macrophage proliferation in LDLR-/- mice, and enhanced carotid plaque stability in mice with vulnerable plaque induced. CONCLUSION MiR-127-3p enhances proliferation in macrophages through downregulating SCD-1 expression and decreasing the content of unsaturated fatty acid, thereby promoting atherosclerosis development and decreasing plaque stability. miR-127-3p/SCD1/UFAs might provide potential therapeutic target for anti-inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Yandong Liu
- Department of Geriatrics, 905th Hospital of PLA NAVY, Shanghai, China; Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Yicheng Wu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Chao Wang
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Weilin Hu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Sili Zou
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Huiqiong Ren
- Department of Geriatrics, 905th Hospital of PLA NAVY, Shanghai, China.
| | - Yong Zuo
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lefeng Qu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
12
|
Li J, Chen R, Zhou J, Wang Y, Zhao X, Liu C, Zhou P, Chen Y, Song L, Li N, Yan H, Zhao H. Atherosclerotic Autoantigen ALDH4A1 as a Novel Immunological Indicator for Plaque Erosion in Patients with ST Segment Elevated Myocardial Infarction. Thromb Haemost 2024; 124:584-594. [PMID: 38109905 DOI: 10.1055/s-0043-1777265] [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] [Indexed: 12/20/2023]
Abstract
OBJECTIVE Aldehyde dehydrogenase 4A1 (ALDH4A1) was recently reported to be a novel autoantigen of atherosclerosis. However, its role in different phenotypes of acute coronary syndrome remains unclear. Herein, we planned to explore the circulating and regional expression of ALDH4A1 in patients with plaque rupture (PR) and plaque erosion (PE) determined by optical coherence tomography (OCT). METHODS AND RESULTS After applying the inclusion and exclusion criteria, a prospective series of 312 patients with ST segment elevated myocardial infarction (STEMI), including 161 patients with PR and 151 patients with PE determined by OCT, were enrolled for plasma ALDH4A1 testing. In addition, ALDH4A1 was quantified using immunofluorescence in aspirated coronary thrombus samples obtained from 31 patients with PR and 25 patients with PE. In addition, we established an atherosclerosis mouse model and analyzed the distribution of ALDH4A1 expression in different mouse organs. Furthermore, we compared the level of ALDH4A1 in the spleen and carotid artery between Apoe-/- and C57 mice. The results showed that the plasma level of ALDH4A1 was significantly higher in STEMI patients with PE than in those with PR (4.6 ng/mL [2.2-8.7] vs. 3.5 ng/mL [1.6-5.6] p = 0.005). The expression of ALDH4A1 in aspirated coronary thrombi was also significantly higher in patients with PE than in those with PR (mean gray value: 32.0 [23.6-40.6] vs. 16.8 [14.0-24.5], p < 0.001). In animal models, the expression of ALDH4A1 is much higher in the spleen than in other organs, and the level of ALDH4A1 is significantly elevated in the spleen and carotid artery of Apoe-/- mice compared with C57 mice. CONCLUSION The high levels of ALDH4A1 in the plasma and aspirated coronary thrombi independently correlated with PE in patients with STEMI. These results suggested that ALDH4A1 is involved in the mechanism of PE and serves as a promising biomarker and treatment target for patients with PE.
Collapse
Affiliation(s)
- Jiannan Li
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China
| | - Runzhen Chen
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jinying Zhou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Wang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoxiao Zhao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Chen Liu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China
| | - Peng Zhou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yi Chen
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Li Song
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Coronary Heart Disease Center, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Nan Li
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hongbing Yan
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China
- Coronary Heart Disease Center, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Hanjun Zhao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Coronary Heart Disease Center, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Li Y, Wang Y, Xia Z, Xie Y, Ke D, Song B, Mu D, Yu R, Xie J. Noninvasive platelet membrane-coated Fe 3O 4 nanoparticles identify vulnerable atherosclerotic plaques. SMART MEDICINE 2024; 3:e20240006. [PMID: 39188703 PMCID: PMC11235982 DOI: 10.1002/smmd.20240006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/27/2024] [Indexed: 08/28/2024]
Abstract
Vulnerable atherosclerotic plaques serve as the primary pathological basis for fatal cardiovascular and cerebrovascular diseases. The precise identification and treatment of these vulnerable plaques hold paramount clinical importance in mitigating the incidence of myocardial infarction and stroke. Nevertheless, the identification of vulnerable plaques within the diffuse atherosclerotic plaques dispersed throughout the systemic circulation continues to pose a substantial challenge in clinical practice. Double emulsion solvent evaporation method, specifically the water-in-oil-in-water (W/O/W) technique, was employed to fabricate Fe3O4-based poly (lactic-co-glycolic acid) (PLGA) nanoparticles (Fe3O4@PLGA). Platelet membranes (PM) were extracted through hypotonic lysis, followed by ultrasound-assisted encapsulation onto the surface of Fe3O4@PLGA, resulting in the formation of PM-coated Fe3O4 nanoparticles (PM/Fe3O4@PLGA). Characterization of PM/Fe3O4@PLGA involved the use of dynamic light scattering, transmission electron microscopy, western blotting, and magnetic resonance imaging (MRI). A model of atherosclerotic vulnerable plaques was constructed by carotid artery coarctation and a high-fat diet fed to ApoE-/- (Apolipoprotein E knockout) mice. Immunofluorescence and MRI techniques were employed to verify the functionality of PM/Fe3O4@PLGA. In this study, we initially synthesized Fe3O4@PLGA as the core material. Subsequently, a platelet membrane was employed as a coating for the Fe3O4@PLGA, aiming to enable the detection of vulnerable atherosclerotic plaques through MRI. In vitro, PM/Fe3O4@PLGA not only exhibited excellent biosafety but also showed targeted collagen characteristics and MR imaging performance. In vivo, the adhesion of PM/Fe3O4@PLGA to atherosclerotic lesions was confirmed in a mouse model of vulnerable atherosclerotic plaques. Simultaneously, PM/Fe3O4@PLGA as a novel contrast agent for MRI has shown effective identification of vulnerable atherosclerotic plaques. In terms of safety profile in vivo, PM/Fe3O4@PLGA has not demonstrated significant organ toxicity or inflammatory response in the bloodstream. In this study, we successfully developed a platelet-membrane-coated nanoparticle system for the targeted delivery of Fe3O4@PLGA to vulnerable atherosclerotic plaques. This innovative system allows for the visualization of vulnerable plaques using MRI, thereby demonstrating its potential for enhancing the clinical diagnosis of vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Yuyu Li
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Beijing Institute of Heart, Lung, and Blood Vessel DiseasesBeijing Anzhen Hospital Affiliated to Capital Medical UniversityBeijingChina
| | - Yujie Wang
- Department of RadiologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Zequn Xia
- Department of CardiologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Yangjing Xie
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Department of MedicineCardiovascular CenterMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Daozheng Ke
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Bing Song
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Dan Mu
- Department of RadiologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of RadiologyNanjing Drum Tower Hospital Clinical College of Jiangsu UniversityNanjingChina
| | - Ronghui Yu
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Jun Xie
- Department of CardiologyNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Department of CardiologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| |
Collapse
|
14
|
Ruan Y, Meng S, Jia R, Cao X, Jin Z. MicroRNA-322-5p protects against myocardial infarction through targeting BTG2. Am J Med Sci 2024; 367:397-405. [PMID: 38437946 DOI: 10.1016/j.amjms.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 01/21/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024]
Abstract
BACKGROUND Numerous studies have explored the therapeutic potential of microRNA (miR) in myocardial infarction (MI) treatment. This study focuses on the role of miR-322-5p in MI, particularly in its regulatory interaction with B-cell translocation gene 2 (BTG2). MATERIALS AND METHODS Expression levels of miR-322-5p and BTG2 were assessed in a rat MI model. Adenovirus altering miR-322-5p or BTG2 expression were administered to MI rats. Evaluation included cardiac function, inflammation, myocardial injury, pathological changes, apoptosis, and NF-κB pathway-related genes in MI rats post-targeted treatment. The miR-322-5p and BTG2 targeting relationship was investigated. RESULTS MI rats exhibited low miR-322-5p and high BTG2 expression in the myocardial tissues. Restoration of miR-322-5p enhanced cardiac function, alleviated inflammation and myocardial injury, mitigated pathological changes and apoptosis, and deactivated the NF-κB pathway in MI rats. BTG2 expression was negatively-regulated by miR-322-5p. Overexpressed BTG2 counteracted miR-322-5p-induced cardioprotection on MI rats. CONCLUSION This study provides evidence that miR-322-5p protects against MI by suppressing BTG2 expression.
Collapse
Affiliation(s)
- Yang Ruan
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuai Meng
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ruofei Jia
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaojing Cao
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zening Jin
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| |
Collapse
|
15
|
Li B, Lu M, Wang H, Sheng S, Guo S, Li J, Tian Y. Macrophage Ferroptosis Promotes MMP2/9 Overexpression Induced by Hemin in Hemorrhagic Plaque. Thromb Haemost 2024; 124:568-580. [PMID: 37696298 DOI: 10.1055/a-2173-3602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
BACKGROUND Intra-plaque hemorrhage (IPH) leads to rapid plaque progression and instability through upregulation of matrix metalloproteinases (MMPs) and collagen degradation. Hemoglobin-derived hemin during IPH promotes plaque instability. We investigated whether hemin affects MMP overexpression in macrophages and explored the underlying mechanisms. MATERIAL AND METHODS In vivo, hemorrhagic plaque models were established in rabbits and ApoE-/- mice. Ferrostatin-1 was used to inhibit ferroptosis. Plaque size, collagen, and MMP2/9 levels were evaluated using immunohistochemistry, H&E, Sirius Red, and Masson staining. In vitro, mouse peritoneal macrophages were extracted. Western blot and ELISA were used to measure MMP2/9 levels. Bioinformatics analysis investigated the association between MMPs and ferroptosis pathway genes. Macrophage ferroptosis was assessed by evaluating cell viability, lipid reactive oxygen species, mitochondrial ultrastructure, iron content, and COX2 levels after pretreatment with cell death inhibitors. Hemin's impact on ferroptosis and MMP expression was studied using Ferrostatin-1 and SB202190. RESULTS In the rabbit hemorrhagic plaques, hemin deposition and overexpression of MMP2/9 were observed, particularly in macrophage-enriched regions. In vitro, hemin induced ferroptosis and MMP2/9 expression in macrophages. Ferrostatin-1 and SB202190 inhibited hemin-induced MMP2/9 overexpression. Ferrostatin-1 inhibited p38 phosphorylation in macrophages. Ferostatin-1 inhibits macrophage ferroptosis, reduces MMP2/9 levels in plaques, and stabilizes the hemorrhagic plaques. CONCLUSION Our results suggested that hemin-induced macrophage ferroptosis promotes p38 pathway activation and MMP2/9 overexpression, which may play a crucial role in increasing hemorrhagic plaque vulnerability. These findings provide insights into the pathogenesis of hemorrhagic plaques and suggest that targeting macrophage ferroptosis may be a promising strategy for stabilizing vulnerable plaque.
Collapse
Affiliation(s)
- Bicheng Li
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, P. R. China
| | - Minqiao Lu
- Department of Pathophysiology and Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, P. R. China
| | - Hui Wang
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, P. R. China
| | - Siqi Sheng
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, P. R. China
| | - Shuyuan Guo
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, P. R. China
| | - Jia Li
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, P. R. China
| | - Ye Tian
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, P. R. China
- Department of Pathophysiology and Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, P. R. China
| |
Collapse
|
16
|
Jiang Y, Tong W, Li Y, Ma Q, Chen Y. Melatonin inhibits the formation of intraplaque neovessels in ApoE-/- mice via PPARγ- RhoA-ROCK pathway. Biochem Biophys Res Commun 2024; 696:149391. [PMID: 38184922 DOI: 10.1016/j.bbrc.2023.149391] [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: 09/28/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND According to former research, the atherosclerotic plaque is thought to be aggravated by intraplaque neovessels (IPN) and intraplaque hemorrhage (IPH). Intriguingly, a lower incidence of IPH was found in plaque treated with melatonin. In this study, we attempted to investigate the impact and underlying mechanism regarding the influences of melatonin upon IPN. METHODS A mouse model was established by subjecting the high fat diet (HFD)-fed ApoE-/- mice to tandem stenosis (TS) surgery with melatonin and GW9662, a PPARγ antagonist, being given by gavage. In vitro experiment was conducted with HUVECs exposing to according treatments of VEGF, melatonin, GW9662, or Y27632. RESULTS Plaque and IPN were attenuated by treatment with melatonin, which was then reversed by blocking PPARγ. Western blotting results showed that melatonin increased PPARγ and decreased RhoA/ROCK signaling in carotid artery. Elevated RhoA/ROCK signaling was observed in melatonin-treated mice when PPARγ was blocked. In accordance with it, experiments using protein and mRNA from HUVECs revealed that melatonin inhibited the RhoA/ROCK signaling by enhancing PPARγ. According to in vitro study, melatonin was able to inhibit cell migration and angiogenesis, which was aborted by GW9662. Blockage of ROCK using Y27632 was able to cease the effect of GW9662 and restored the suppression on cell migration and angiogenesis by melatonin. CONCLUSIONS Our study demonstrates that melatonin is able to curb development of plaque and IPN formation by inhibiting the migration of endothelial cells via PPARγ- RhoA-ROCK pathway. That provides a therapeutic potential for both melatonin and PPARγ agonist targeting IPN, IPH, and atherosclerotic plaque.
Collapse
Affiliation(s)
- YuFan Jiang
- School of Medicine, Nankai University, Tianjin, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei Tong
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yueyang Li
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qiang Ma
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - YunDai Chen
- School of Medicine, Nankai University, Tianjin, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
17
|
Ping L, Zhi-Ming L, Bi-Shan Z, Lei Z, Bo Y, Yi-Chun Z, Ming-Jie W. S-propargyl-cysteine promotes the stability of atherosclerotic plaque via maintaining vascular muscle contractile phenotype. Front Cell Dev Biol 2024; 11:1291170. [PMID: 38328305 PMCID: PMC10847265 DOI: 10.3389/fcell.2023.1291170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/22/2023] [Indexed: 02/09/2024] Open
Abstract
Introduction: Plaque rupture in atherosclerosis contributes to various acute cardiovascular events. As a new sulfide-containing donor, S-propargyl-cysteine (SPRC) has been reported to play a beneficial role in cardioprotection, potentially through its anti-inflammatory, anti-oxidative and anti-atherogenic activities. Our previous study observed an increase in eNOS phosphorylation in endothelial cells. However, it remains unclear whether SPRC influences vascular smooth muscle cells (VSMCs) within the plaque and if this effect contributes to plaque stabilization. Methods: An atherosclerotic unstable plaque mouse model was established by subjecting ApoE-/- mice to tandem stenosis of the right carotid artery along with a Western diet. Daily SPRC administration was conducted for 13 weeks. Plaque morphology and stability were assessed using MRI scanning and histopathological staining. In our in vitro studies, we stimulated human artery vascular smooth muscle cells (HAVSMCs) with platelet-derived growth factor-BB (PDGF-BB), both with and without 100 μM SPRC treatment. Cell phenotype was assessed using both Western blot and Real-time PCR. Cell proliferation was assessed using the BrdU cell proliferation kit and immunofluorescence of Ki-67, while cell migration was measured using scratch wound healing and transwell assay. MiR-143-3p overexpression and knockdown experiments were used to investigate whether it mediates the effect of SPRC on VSMC phenotype. Results and Discussion: SPRC treatment reduced plasma lipid levels, increased collagen content and decreased cell apoptosis in atherosclerotic plaques, indicating improved plaque stability. Both in vivo and in vitro studies elucidated the role of SPRC in preserving the contractile phenotype of VSMCs through up-regulation of miR-143-3p expression. Furthermore, SPRC suppressed the pro-proliferation and pro-migration effects of PDGF-BB on HAVSMCs. Overall, these findings suggest that the inhibitory effect of SPRC on phenotype switch from contractile to synthetic VSMCs may contribute to its beneficial role in enhancing plaque stability.
Collapse
Affiliation(s)
- Li Ping
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, The Innovative Research Team of High-level Local Universities in Shanghai, Fudan University, Shanghai, China
| | - Li Zhi-Ming
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, The Innovative Research Team of High-level Local Universities in Shanghai, Fudan University, Shanghai, China
| | - Zhang Bi-Shan
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, The Innovative Research Team of High-level Local Universities in Shanghai, Fudan University, Shanghai, China
| | - Zhu Lei
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Bo
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhu Yi-Chun
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, The Innovative Research Team of High-level Local Universities in Shanghai, Fudan University, Shanghai, China
| | - Wang Ming-Jie
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, The Innovative Research Team of High-level Local Universities in Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Liu J, Zhang D, Brahmandam A, Matsubara Y, Gao M, Tian J, Liu B, Shu C, Dardik A. Bioinformatics identifies predictors of arteriovenous fistula maturation. J Vasc Access 2024; 25:172-186. [PMID: 35686495 PMCID: PMC9734286 DOI: 10.1177/11297298221102298] [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] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Arteriovenous fistulae (AVF) are the preferred access for hemodialysis but still have poor rates of maturation and patency limiting their clinical use. The underlying mechanisms of venous remodeling remain poorly understood, and only limited numbers of unbiased approaches have been reported. METHODS Biological Gene Ontology (GO) term enrichment analysis and differentially expressed genes (DEG) analysis were performed for three AVF datasets. A microRNA enrichment analysis and L1000CDS2 query were performed to identify factors predicting AVF patency. RESULTS The inflammatory and immune responses were activated during both early and late phases of AVF maturation, with upregulation of neutrophil and leukocyte regulation, cytokine production, and cytokine-mediated signaling. In men with failed AVF, negative regulation of myeloid-leukocyte differentiation and regulation of macrophage activation were significantly upregulated. Compared to non-diabetic patients, diabetic patients had significantly reduced immune response-related enrichment such as cell activation in immune response, regulation of immune-effector process, and positive regulation of defense response; in addition, diabetic patients showed no enrichment of the immune response-regulating signaling pathway. CONCLUSIONS These data show coordinated, and differential regulation of genes associated with AVF maturation, and different patterns of several pathways are associated with sex differences in AVF failure. Inflammatory and immune responses are activated during AVF maturation and diabetes may impair AVF maturation by altering these responses. These findings suggest several novel molecular targets to improve sex specific AVF maturation.
Collapse
Affiliation(s)
- Jia Liu
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Dingyao Zhang
- The Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
| | - Anand Brahmandam
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Yutaka Matsubara
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- The Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan
| | - Mingjie Gao
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Jingru Tian
- The Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
| | - Bing Liu
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| | - Chang Shu
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Alan Dardik
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
19
|
Yan A, Gotlieb AI. The microenvironment of the atheroma expresses phenotypes of plaque instability. Cardiovasc Pathol 2023; 67:107572. [PMID: 37595697 DOI: 10.1016/j.carpath.2023.107572] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/06/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
Data from histopathology studies of human atherosclerotic tissue specimens and from vascular imaging studies support the concept that the local arterial microenvironment of a stable atheroma promotes destabilizing conditions that result in the transition to an unstable atheroma. Destabilization is characterized by several different plaque phenotypes that cause major clinical events such as acute coronary syndrome and cerebrovascular strokes. There are several rupture-associated phenotypes causing thrombotic vascular occlusion including simple fibrous cap rupture of an atheroma, fibrous cap rupture at site of previous rupture-and-repair of an atheroma, and nodular calcification with rupture. Endothelial erosion without rupture has more recently been shown to be a common phenotype to promote thrombosis as well. Microenvironment features that are linked to these phenotypes of plaque instability are neovascularization arising from the vasa vasorum network leading to necrotic core expansion, intraplaque hemorrhage, and cap rupture; activation of adventitial and perivascular adipose tissue cells leading to secretion of cytokines, growth factors, adipokines in the outer artery wall that destabilize plaque structure; and vascular smooth muscle cell phenotypic switching through transdifferentiation and stem/progenitor cell activation resulting in the promotion of inflammation, calcification, and secretion of extracellular matrix, altering fibrous cap structure, and necrotic core growth. As the technology evolves, studies using noninvasive vascular imaging will be able to investigate the transition of stable to unstable atheromas in real time. A limitation in the field, however, is that reliable and predictable experimental models of spontaneous plaque rupture and/or erosion are not currently available to study the cell and molecular mechanisms that regulate the conversion of the stable atheroma to an unstable plaque.
Collapse
Affiliation(s)
- Angela Yan
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Avrum I Gotlieb
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Tao Y, Lan X, Zhang Y, Fu C, Liu L, Cao F, Guo W. Biomimetic nanomedicines for precise atherosclerosis theranostics. Acta Pharm Sin B 2023; 13:4442-4460. [PMID: 37969739 PMCID: PMC10638499 DOI: 10.1016/j.apsb.2022.11.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/13/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis (AS) is a leading cause of the life-threatening cardiovascular disease (CVD), creating an urgent need for efficient, biocompatible therapeutics for diagnosis and treatment. Biomimetic nanomedicines (bNMs) are moving closer to fulfilling this need, pushing back the frontier of nano-based drug delivery systems design. This review seeks to outline how these nanomedicines (NMs) might work to diagnose and treat atherosclerosis, to trace the trajectory of their development to date and in the coming years, and to provide a foundation for further discussion about atherosclerotic theranostics.
Collapse
Affiliation(s)
- Ying Tao
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Biomedical Engineering & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Xinmiao Lan
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Yang Zhang
- Department of Cardiology, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Chenxing Fu
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Lu Liu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR 999077, China
| | - Feng Cao
- Department of Cardiology, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Weisheng Guo
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Biomedical Engineering & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| |
Collapse
|
21
|
Chen W, Nadel J, Tumanov S, Stocker R. Near-Infrared Autofluorescence (NIRAF) in Atherosclerotic Plaque Dissociates from Intraplaque Hemorrhage and Bilirubin. Int J Mol Sci 2023; 24:10727. [PMID: 37445903 DOI: 10.3390/ijms241310727] [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: 05/23/2023] [Revised: 06/17/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Near-infrared autofluorescence (NIRAF) in unstable atherosclerotic plaque has been suggested as a novel imaging technology for high-risk atherosclerosis. Intraplaque hemorrhage (IPH) and bilirubin, derived from the subsequent degradation of heme, have been proposed as the source of NIRAF, although their roles and the underlying mechanism responsible for NIRAF remain unclear. To test the proposed role of bilirubin as the source of NIRAF in high-risk atherosclerosis, Biliverdin reductase a gene and apolipoprotein E gene double-knockout (Bvra-/-Apoe-/-) mice were subjected to the Western diet and tandem stenosis (TS) surgery, as a model of both bilirubin deficiency and plaque instability. Human coronary arteries containing atherosclerotic plaques were obtained from heart transplant recipients. The NIRAF was determined by in vivo fluorescence emission computed tomography, and ex vivo infrared imaging. The cholesterol content was quantified by HPLC with UV detection. In Bvra+/+Apoe-/- TS mice, the NIRAF intensity was significantly higher in unstable plaque than in stable plaque, yet the NIRAF in unstable plaque was undistinguishable in Bvra+/+Apoe-/- and littermate Bvra-/-Apoe-/- TS mice. Moreover, the unstable plaque in TS mice exhibited a lower NIRAF compared with highly cellular plaque that lacked most of the features of unstable plaque. In human coronary arteries, the NIRAF associated with cholesterol-rich, calcified lesions, rather than just cholesterol-rich lesions. The NIRAF in atherosclerotic plaque can be dissociated from IPH and bilirubin, such that the compositional meaning of an elevated NIRAF remains obscure.
Collapse
Affiliation(s)
- Weiyu Chen
- Heart Research Institute, Sydney, NSW 2042, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - James Nadel
- Heart Research Institute, Sydney, NSW 2042, Australia
- St Vincent's Hospital, Sydney, NSW 2010, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Sergey Tumanov
- Heart Research Institute, Sydney, NSW 2042, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Roland Stocker
- Heart Research Institute, Sydney, NSW 2042, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
22
|
Cheng XW, Narisawa M, Wang H, Piao L. Overview of multifunctional cysteinyl cathepsins in atherosclerosis-based cardiovascular disease: from insights into molecular functions to clinical implications. Cell Biosci 2023; 13:91. [PMID: 37202785 DOI: 10.1186/s13578-023-01040-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 05/01/2023] [Indexed: 05/20/2023] Open
Abstract
Cysteinyl cathepsins (CTSs) are widely known to have a proteolysis function that mediates recycling of unwanted proteins in endosomes and lysosomes, and investigation of CTSs has greatly improved with advances in live-imaging techniques both in vivo and in vitro, leading to three key findings. (1) CTSs are relocated from the lysosomes to other cellular spaces (i.e., cytosol, nucleus, nuclear membrane, plasma membrane, and extracellular milieu). (2) In addition to acidic cellular compartments, CTSs also exert biological activity in neutral environments. (3) CTSs also exert multiple nontraditional functions in, for example, extracellular matrix metabolism, cell signaling transduction, protein processing/trafficking, and cellular events. Various stimuli regulate the expression and activities of CTSs in vivo and vitro-e.g., inflammatory cytokines, oxidative stress, neurohormones, and growth factors. Accumulating evidence has confirmed the participation of CTSs in vascular diseases characterized by atherosclerosis, plaque rupture, thrombosis, calcification, aneurysm, restenosis/in-stent-restenosis, and neovasel formation. Circulating and tissue CTSs are promising as biomarkers and as a diagnostic imaging tool in patients with atherosclerosis-based cardiovascular disease (ACVD), and pharmacological interventions with their specific and non-specific inhibitors, and cardiovascular drugs might have potential for the therapeutic targeting of CTSs in animals. This review focuses on the update findings on CTS biology and the involvement of CTSs in the initiation and progression of ACVD and discusses the potential use of CTSs as biomarkers and small-molecule targets to prevent deleterious nontraditional functions in ACVD.
Collapse
Affiliation(s)
- Xian Wu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital, 1327 Juzijie, Yanjin, Jilin, 133000, People's Republic of China.
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanjin, 133000, Jilin, People's Republic of China.
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, Jilin PR. 133000, China.
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, 4668550, Japan
| | - Hailong Wang
- Department of Cardiology and Hypertension, Yanbian University Hospital, 1327 Juzijie, Yanjin, Jilin, 133000, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanjin, 133000, Jilin, People's Republic of China
| | - Limei Piao
- Department of Cardiology and Hypertension, Yanbian University Hospital, 1327 Juzijie, Yanjin, Jilin, 133000, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanjin, 133000, Jilin, People's Republic of China
| |
Collapse
|
23
|
Schwarz N, Fernando S, Chen YC, Salagaras T, Rao SR, Liyanage S, Williamson AE, Toledo-Flores D, Dimasi C, Sargeant TJ, Manavis J, Eddy E, Kanellakis P, Thompson PL, Tan JTM, Snel MF, Bursill CA, Nicholls SJ, Peter K, Psaltis PJ. Colchicine exerts anti-atherosclerotic and -plaque-stabilizing effects targeting foam cell formation. FASEB J 2023; 37:e22846. [PMID: 36856983 DOI: 10.1096/fj.202201469r] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 02/02/2023] [Accepted: 02/15/2023] [Indexed: 03/02/2023]
Abstract
Colchicine is a broad-acting anti-inflammatory agent that has attracted interest for repurposing in atherosclerotic cardiovascular disease. Here, we studied its ability at a human equivalent dose of 0.5 mg/day to modify plaque formation and composition in murine atherosclerosis and investigated its actions on macrophage responses to atherogenic stimuli in vitro. In atherosclerosis induced by high-cholesterol diet, Apoe-/- mice treated with colchicine had 50% reduction in aortic oil Red O+ plaque area compared to saline control (p = .001) and lower oil Red O+ staining of aortic sinus lesions (p = .03). In vitro, addition of 10 nM colchicine inhibited foam cell formation from murine and human macrophages after treatment with oxidized LDL (ox-LDL). Mechanistically, colchicine downregulated glycosylation and surface expression of the ox-LDL uptake receptor, CD36, and reduced CD36+ staining in aortic sinus plaques. It also decreased macrophage uptake of cholesterol crystals, resulting in lower intracellular lysosomal activity, inhibition of the NLRP3 inflammasome, and reduced secretion of IL-1β and IL-18. Colchicine's anti-atherosclerotic actions were accentuated in a mouse model of unstable plaque induced by carotid artery tandem stenosis surgery, where it decreased lesion size by 48% (p = .01), reduced lipid (p = .006) and necrotic core area (p = .007), increased collagen content and cap-to-necrotic core ratio (p = .05), and attenuated plaque neutrophil extracellular traps (p < .001). At low dose, colchicine's effects were not accompanied by the evidence of microtubule depolymerization. Together, these results show that colchicine exerts anti-atherosclerotic and plaque-stabilizing effects at low dose by inhibiting foam cell formation and cholesterol crystal-induced inflammation. This provides a new framework to support its repurposing for atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Nisha Schwarz
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Sanuja Fernando
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Yung-Chih Chen
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Thalia Salagaras
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Sushma R Rao
- Proteomics, Metabolomics and MS-Imaging Facility, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Sanuri Liyanage
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Anna E Williamson
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Deborah Toledo-Flores
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Catherine Dimasi
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Timothy J Sargeant
- Lifelong Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Jim Manavis
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Eleanor Eddy
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter Kanellakis
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter L Thompson
- Faculty of Health and Medical Sciences, Internal Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Marten F Snel
- Proteomics, Metabolomics and MS-Imaging Facility, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Christina A Bursill
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Monash University, Melbourne, Victoria, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
24
|
Currie GM. The emerging role of artificial intelligence and digital twins in pre-clinical molecular imaging. Nucl Med Biol 2023; 120-121:108337. [PMID: 37030076 DOI: 10.1016/j.nucmedbio.2023.108337] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/27/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023]
Abstract
INTRODUCTION Pre-clinical molecular imaging, particularly with mice, is an essential part of drug and radiopharmaceutical development. There remain ethical challenges to reduce, refine and replace animal imaging where possible. METHOD A number of approaches have been adopted to reduce the use of mice including using algorithmic approaches to animal modelling. Digital twins have been used to create a virtual model of mice, however, exploring the potential of deep learning approaches to digital twin development may enhance capabilities and application in research. RESULTS Generative adversarial networks produce generated images that sufficiently resemble reality that they could be adapted to create digital twins. Specific genetic mouse models have greater homogeneity making them more receptive to modelling and suitable specifically for digital twin simulation. CONCLUSION There are numerous benefits of digital twins in pre-clinical imaging including improved outcomes, fewer animal studies, shorter development timelines and lower costs.
Collapse
|
25
|
Nadel J, Jabbour A, Stocker R. Arterial myeloperoxidase in the detection and treatment of vulnerable atherosclerotic plaque: a new dawn for an old light. Cardiovasc Res 2023; 119:112-120. [PMID: 35587708 DOI: 10.1093/cvr/cvac081] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/01/2022] [Accepted: 04/20/2022] [Indexed: 11/14/2022] Open
Abstract
Intracellular myeloperoxidase (MPO) plays a specific role in the innate immune response; however, upon release into the extracellular space in the setting of inflammation, drives oxidative tissue injury. Extracellular MPO has recently been shown to be abundant in unstable atheroma and causally linked to plaque destabilization, erosion, and rupture, identifying it as a potential target for the surveillance and treatment of vulnerable atherosclerosis. Through the compartmentalization of MPO's protective and deleterious effects, extracellular MPO can be selectively detected using non-invasive molecular imaging and targeted by burgeoning pharmacotherapies. Given its causal relationship to plaque destabilization coupled with an ability to preserve its beneficial properties, MPO is potentially a superior translational inflammatory target compared with other immunomodulatory therapies and imaging biomarkers utilized to date. This review explores the role of MPO in plaque destabilization and provides insights into how it can be harnessed in the management of patients with vulnerable atherosclerotic plaque.
Collapse
Affiliation(s)
- James Nadel
- Heart Research Institute, The University of Sydney, 7 Eliza St, Newtown, 2042 Sydney, NSW, Australia
- Cardiology Department, St Vincent's Hospital, Sydney, Australia
- School of Medicine, University of New South Wales, Sydney, Australia
| | - Andrew Jabbour
- Cardiology Department, St Vincent's Hospital, Sydney, Australia
- School of Medicine, University of New South Wales, Sydney, Australia
| | - Roland Stocker
- Heart Research Institute, The University of Sydney, 7 Eliza St, Newtown, 2042 Sydney, NSW, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| |
Collapse
|
26
|
Chen YC, Smith M, Ying YL, Makridakis M, Noonan J, Kanellakis P, Rai A, Salim A, Murphy A, Bobik A, Vlahou A, Greening DW, Peter K. Quantitative proteomic landscape of unstable atherosclerosis identifies molecular signatures and therapeutic targets for plaque stabilization. Commun Biol 2023; 6:265. [PMID: 36914713 PMCID: PMC10011552 DOI: 10.1038/s42003-023-04641-4] [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: 07/18/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Atherosclerotic plaque rupture leading to myocardial infarction is a major global health burden. Applying the tandem stenosis (TS) mouse model, which distinctively exhibits the characteristics of human plaque instability/rupture, we use quantitative proteomics to understand and directly compare unstable and stable atherosclerosis. Our data highlight the disparate natures and define unique protein signatures of unstable and stable atherosclerosis. Key proteins and pathway networks are identified such as the innate immune system, and neutrophil degranulation. The latter includes calprotectin S100A8/A9, which we validate in mouse and human unstable plaques, and we demonstrate the plaque-stabilizing effects of its inhibition. Overall, we provide critical insights into the unique proteomic landscape of unstable atherosclerosis (as distinct from stable atherosclerosis and vascular tissue). We further establish the TS model as a reliable preclinical tool for the discovery and testing of plaque-stabilizing drugs. Finally, we provide a knowledge resource defining unstable atherosclerosis that will facilitate the identification and validation of long-sought-after therapeutic targets and drugs for plaque stabilization.
Collapse
Affiliation(s)
- Yung-Chih Chen
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Meaghan Smith
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Ya-Lan Ying
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Manousos Makridakis
- Proteomics Research Unit, Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Jonathan Noonan
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Peter Kanellakis
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Alin Rai
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
- Molecular Proteomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
| | - Agus Salim
- Department of Bioinformatics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Andrew Murphy
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
- Haematopoiseis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Alex Bobik
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Immunology, Monash University, Centre for Inflammatory Disease, School of Clinical Sciences, Monash Health, Melbourne, VIC, Australia
| | - Antonia Vlahou
- Proteomics Research Unit, Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - David W Greening
- Central Clinical School, Monash University, Melbourne, VIC, Australia.
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia.
- Molecular Proteomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia.
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Central Clinical School, Monash University, Melbourne, VIC, Australia.
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia.
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia.
| |
Collapse
|
27
|
Tong W, Zhang Y, Hui H, Feng X, Ning B, Yu T, Wang W, Shang Y, Zhang G, Zhang S, Tian F, He W, Chen Y, Tian J. Sensitive magnetic particle imaging of haemoglobin degradation for the detection and monitoring of intraplaque haemorrhage in atherosclerosis. EBioMedicine 2023; 90:104509. [PMID: 36905783 PMCID: PMC10023936 DOI: 10.1016/j.ebiom.2023.104509] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND Intraplaque haemorrhage (IPH) drives atherosclerosis progression and is a key imaging biomarker of unstable plaques. Non-invasive and sensitive monitoring of IPH is challenging due to the compositional complexity and dynamic nature of atherosclerotic plaques. Magnetic particle imaging (MPI) is a highly sensitive, radiation-free, and no-tissue-background tomographic technique that detects superparamagnetic nanoparticles. Thus, we aimed to investigate whether MPI can in vivo detect and monitor IPH. METHODS Thirty human carotid endarterectomy samples were collected and scanned with MPI. The tandem stenosis (TS) model was employed to establish unstable plaques with IPH in ApoE-/- mice. MPI and 7 T T1-weighted magnetic resonance imaging (MRI) were performed on TS ApoE-/- mice. Plaque specimens were analyzed histologically. FINDINGS Human carotid endarterectomy samples exhibited endogenous MPI signals, which histologically colocalized with IPH. In vitro experiments identified haemosiderin, a haemoglobin degradation product, as a potential source of MPI signals. Longitudinal MPI of TS ApoE-/- mice detected IPH at unstable plaques, of which MPI signal-to-noise ratio values increased from 6.43 ± 1.74 (four weeks) to 10.55 ± 2.30 (seven weeks) and reduced to 7.23 ± 1.44 (eleven weeks). In contrast, 7 T T1-weighted MRI did not detect the small-size IPH (329.91 ± 226.82 μm2) at four weeks post-TS. The time-course changes in IPH were shown to correlate with neovessel permeability providing a possible mechanism for signal changes over time. INTERPRETATION MPI is a highly sensitive imaging technology that allows the identification of atherosclerotic plaques with IPH and may help detect and monitor unstable plaques in patients. FUNDING This work was supported in part by the Beijing Natural Science Foundation under Grant JQ22023; the National Key Research and Development Program of China under Grant 2017YFA0700401; the National Natural Science Foundation of China under Grant 62027901, 81827808, 81730050, 81870178, 81800221, 81527805, and 81671851; the CAS Youth Innovation Promotion Association under Grant Y2022055 and CAS Key Technology Talent Program; and the Project of High-Level Talents Team Introduction in Zhuhai City (Zhuhai HLHPTP201703).
Collapse
Affiliation(s)
- Wei Tong
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, China; CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yingqian Zhang
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Hui Hui
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Beijing Key Laboratory of Molecular Imaging, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Xin Feng
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Beijing Key Laboratory of Molecular Imaging, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Bin Ning
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Tengfei Yu
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Wei Wang
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Yaxin Shang
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing, 100069, China
| | - Guanghao Zhang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Suhui Zhang
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Feng Tian
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Wen He
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| | - Yundai Chen
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Beijing Key Laboratory of Molecular Imaging, Beijing, 100190, China; Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, 100191, China; Zhuhai Precision Medical Center, Zhuhai People's Hospital, Affiliated with Jinan University, Zhuhai, 519000, China.
| |
Collapse
|
28
|
Zhang X, Centurion F, Misra A, Patel S, Gu Z. Molecularly targeted nanomedicine enabled by inorganic nanoparticles for atherosclerosis diagnosis and treatment. Adv Drug Deliv Rev 2023; 194:114709. [PMID: 36690300 DOI: 10.1016/j.addr.2023.114709] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/20/2022] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Atherosclerosis, a chronic cardiovascular disease caused by plaque development in arteries, remains a leading cause of morbidity and mortality. Atherosclerotic plaques are characterized by the expression and regulation of key molecules such as cell surface receptors, cytokines, and signaling pathway proteins, potentially facilitating precise diagnosis and treatment on a molecular level by specifically targeting the characteristic molecules. In this review, we highlight the recent progress in the past five years on developing molecularly targeted nanomedicine for imaging detection and treatment of atherosclerosis with the use of inorganic nanoparticles. Through targeted delivery of imaging contrast nanoparticles to specific molecules in atherogenesis, atherosclerotic plaque development at different stages could be identified and monitored via various molecular imaging modalities. We also review molecularly targeted therapeutic approaches that target and regulate molecules associated with lipid regulation, inflammation, and apoptosis. The review is concluded with discussion on current challenges and future development of nanomedicine for atherosclerotic diagnosis and treatment.
Collapse
Affiliation(s)
- Xiuwen Zhang
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Franco Centurion
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ashish Misra
- Heart Research Institute, Sydney, NSW 2042, Australia; Faculty of Medicine and Health, The University of Sydney, NSW 2006, Australia
| | - Sanjay Patel
- Heart Research Institute, Sydney, NSW 2042, Australia; Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia; Sydney Medical School, The University of Sydney, NSW 2006, Australia
| | - Zi Gu
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; Australian Centre for NanoMedicine (ACN), University of New South Wales, Sydney, NSW 2052, Australia; UNSW RNA Institute, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
29
|
Bartlett B, Lee S, Ludewick HP, Siew T, Verma S, Waterer G, Corrales-Medina VF, Dwivedi G. A multiple comorbidities mouse lung infection model in ApoE‑deficient mice. Biomed Rep 2023; 18:21. [PMID: 36846615 PMCID: PMC9944256 DOI: 10.3892/br.2023.1603] [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: 08/04/2022] [Accepted: 12/09/2022] [Indexed: 02/09/2023] Open
Abstract
Acute pneumonia is characterised by a period of intense inflammation. Inflammation is now considered to be a key step in atherosclerosis progression. In addition, pre-existing atherosclerotic inflammation is considered to play a role in pneumonia progression and risk. In the present study, a multiple comorbidities murine model was used to study respiratory and systemic inflammation that results from pneumonia in the setting of atherosclerosis. Firstly, a minimal infectious dose of Streptococcus pneumoniae (TIGR4 strain) to produce clinical pneumonia with a low mortality rate (20%) was established. C57Bl/6 ApoE -/- mice were fed a high-fat diet prior to administering intranasally 105 colony forming units of TIGR4 or phosphate-buffered saline (PBS). At days 2, 7 and 28 post inoculation (PI), the lungs of mice were imaged by magnetic resonance imaging (MRI) and positron emission tomography (PET). Mice were euthanised and investigated for changes in lung morphology and changes in systemic inflammation using ELISA, Luminex assay and real-time PCR. TIGR4-inoculated mice presented with varying degrees of lung infiltrate, pleural effusion and consolidation on MRI at all time points up to 28 days PI. Moreover, PET scans identified significantly higher FDG uptake in the lungs of TIGR4-inoculated mice up to 28 days PI. The majority (90%) TIGR4-inoculated mice developed pneumococcal-specific IgG antibody response at 28 days PI. Consistent with these observations, TIGR4-inoculated mice displayed significantly increased inflammatory gene expression [interleukin (IL)-1β and IL-6] in the lungs and significantly increased levels of circulating inflammatory protein (CCL3) at 7 and 28 days PI respectively. The mouse model developed by the authors presents a discovery tool to understand the link between inflammation related to acute infection such as pneumonia and increased risk of cardiovascular disease observed in humans.
Collapse
Affiliation(s)
- Benjamin Bartlett
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia 6150, Australia
- School of Medicine, The University of Western Australia, Perth, Western Australia 6009, Australia
| | - Silvia Lee
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia 6150, Australia
- Department of Microbiology, Pathwest Laboratory Medicine, Perth, Western Australia 6000, Australia
| | - Herbert P. Ludewick
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia 6150, Australia
- Heart and Lung Research Institute, Harry Perkins Institute of Medical Research, Perth, Western Australia 6150, Australia
| | - Teck Siew
- Department of Nuclear Medicine, Fiona Stanley Hospital, Perth, Western Australia 6150, Australia
- Royal Perth Hospital, Perth, Western Australia 6000, Australia
| | - Shipra Verma
- Department of Nuclear Medicine, Fiona Stanley Hospital, Perth, Western Australia 6150, Australia
- Department of Geriatric Medicine, Fiona Stanley Hospital, Perth, Western Australia 6150, Australia
| | - Grant Waterer
- School of Medicine, The University of Western Australia, Perth, Western Australia 6009, Australia
- Royal Perth Hospital, Perth, Western Australia 6000, Australia
| | - Vicente F. Corrales-Medina
- Department of Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Girish Dwivedi
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia 6150, Australia
- School of Medicine, The University of Western Australia, Perth, Western Australia 6009, Australia
- Department of Cardiology, Fiona Stanley Hospital, Perth, Western Australia 6150, Australia
| |
Collapse
|
30
|
Wu Y, Deng C, Xu J, Wang W, Chen Y, Qin X, Lv Q, Xie M. Enhanced Local Delivery of microRNA-145a-5P into Mouse Aorta via Ultrasound-Targeted Microbubble Destruction Inhibits Atherosclerotic Plaque Formation. Mol Pharm 2023; 20:1086-1095. [PMID: 36656656 DOI: 10.1021/acs.molpharmaceut.2c00799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) play a key role in the formation and rupture of atherosclerotic plaques. Previous studies have confirmed that microRNA-145 (miR-145) is involved in the phenotypic regulation of VSMCs and reduction of atherosclerosis. At present, seeking safe and effective gene delivery remains a key problem restricting the development of gene therapy. In recent years, ultrasound-targeted microbubble destruction (UTMD) has become a safe and effective transfection method that is widely used in the basic research of gene therapy for heart and tumor diseases. Here, we synthesized cationic microbubbles to encapsulate miR-145 and targeted their release into VSMCs in vitro and in vivo using ultrasound. The feasibility of this gene therapy was verified by fluorescence microscopy and an in vivo imaging system. The results showed that treatment with miR-145 delivered via UTMD considerably improved the gene transfection efficiency and promoted the contraction phenotype of VSMCs in vitro. In vivo, this treatment reduced the atherosclerotic plaque area by 48.04% compared with treatment with free miR-145. Therefore, UTMD-mediated miRNA therapy may provide a new targeted therapeutic approach for atherosclerotic plaques.
Collapse
Affiliation(s)
- Yu Wu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Cheng Deng
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Jia Xu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Wei Wang
- Department of Ultrasound, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaojuan Qin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Qing Lv
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| |
Collapse
|
31
|
Vigder N, Suarna C, Corcilius L, Nadel J, Chen W, Payne R, Tumanov S, Stocker R. An improved method for the detection of myeloperoxidase chlorinating activity in biological systems using the redox probe hydroethidine. Free Radic Biol Med 2023; 195:23-35. [PMID: 36565892 DOI: 10.1016/j.freeradbiomed.2022.12.014] [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: 10/16/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Conversion of the redox probe hydroethidine (HE) to 2-chloroethidium (2-Cl-E+) by myeloperoxidase (MPO)-derived hypochlorous acid (HOCl) provides comparable specificity and superior sensitivity to measurement of 3-chlorotyrosine (3-Cl-Tyr), the gold standard biomarker for MPO chlorinating activity in biological systems. However, a limitation of the former method is the complex mixture of products formed by the reaction of HE with reagent HOCl, coupled with the difficult purification of 2-Cl-E+ from this mixture for analytical purposes. This limitation prompted us to test whether 2-Cl-E+ could be formed by reaction of HE with the strong and widely used chlorinating agent, N-chlorosuccinimide (NCS). Unexpectedly, such reaction yielded 2-chlorohydroethidine (2-Cl-HE) as the major product in addition to 2-Cl-E+, as assessed by high performance liquid chromatography (HPLC), mass spectrometry (MS), and nuclear magnetic resonance (NMR). 2-Cl-HE was also observed to be the major chlorination product formed from HE with both reagent and enzymatically generated HOCl, just as it was formed ex vivo in different healthy and diseased mouse and human tissues upon incubation with glucose/glucose oxidase to generate a flux of hydrogen peroxide (H2O2). Quantification of 2-Cl-HE plus 2-Cl-E+ improved the sensitivity of the HE-based method compared with measurement of only 2-Cl-E+. Moreover, 2-chlorodimidium (2-Cl-D+) was developed as a practical internal standard instead of the previously used internal standard, deuterated 2-Cl-E+ (d5-2-Cl-E+). Overall, the present study describes an improved method for the detection of MPO/chlorinating activity in biological systems of health and disease.
Collapse
Affiliation(s)
- Niv Vigder
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Cacang Suarna
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Leo Corcilius
- School of Chemistry, The University of Sydney, Sydney, New South Wales, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, New South Wales, Australia
| | - James Nadel
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; St Vincent's Hospital, Sydney, Australia; University of New South Wales, Sydney, Australia
| | - Weiyu Chen
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Richard Payne
- School of Chemistry, The University of Sydney, Sydney, New South Wales, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, New South Wales, Australia
| | - Sergey Tumanov
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Roland Stocker
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
32
|
van der Vorst EPC, Maas SL, Theodorou K, Peters LJF, Jin H, Rademakers T, Gijbels MJ, Rousch M, Jansen Y, Weber C, Lehrke M, Lebherz C, Yildiz D, Ludwig A, Bentzon JF, Biessen EAL, Donners MMPC. Endothelial ADAM10 controls cellular response to oxLDL and its deficiency exacerbates atherosclerosis with intraplaque hemorrhage and neovascularization in mice. Front Cardiovasc Med 2023; 10:974918. [PMID: 36776254 PMCID: PMC9911417 DOI: 10.3389/fcvm.2023.974918] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
Introduction The transmembrane protease A Disintegrin And Metalloproteinase 10 (ADAM10) displays a "pattern regulatory function," by cleaving a range of membrane-bound proteins. In endothelium, it regulates barrier function, leukocyte recruitment and angiogenesis. Previously, we showed that ADAM10 is expressed in human atherosclerotic plaques and associated with neovascularization. In this study, we aimed to determine the causal relevance of endothelial ADAM10 in murine atherosclerosis development in vivo. Methods and results Endothelial Adam10 deficiency (Adam10 ecko ) in Western-type diet (WTD) fed mice rendered atherogenic by adeno-associated virus-mediated PCSK9 overexpression showed markedly increased atherosclerotic lesion formation. Additionally, Adam10 deficiency was associated with an increased necrotic core and concomitant reduction in plaque macrophage content. Strikingly, while intraplaque hemorrhage and neovascularization are rarely observed in aortic roots of atherosclerotic mice after 12 weeks of WTD feeding, a majority of plaques in both brachiocephalic artery and aortic root of Adam10ecko mice contained these features, suggestive of major plaque destabilization. In vitro, ADAM10 knockdown in human coronary artery endothelial cells (HCAECs) blunted the shedding of lectin-like oxidized LDL (oxLDL) receptor-1 (LOX-1) and increased endothelial inflammatory responses to oxLDL as witnessed by upregulated ICAM-1, VCAM-1, CCL5, and CXCL1 expression (which was diminished when LOX-1 was silenced) as well as activation of pro-inflammatory signaling pathways. LOX-1 shedding appeared also reduced in vivo, as soluble LOX-1 levels in plasma of Adam10ecko mice was significantly reduced compared to wildtypes. Discussion Collectively, these results demonstrate that endothelial ADAM10 is atheroprotective, most likely by limiting oxLDL-induced inflammation besides its known role in pathological neovascularization. Our findings create novel opportunities to develop therapeutics targeting atherosclerotic plaque progression and stability, but at the same time warrant caution when considering to use ADAM10 inhibitors for therapy in other diseases.
Collapse
Affiliation(s)
- Emiel P. C. van der Vorst
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands,Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, Aachen, Germany,Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University Hospital, Aachen, Germany,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Munich, Germany,German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Sanne L. Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, Aachen, Germany,Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University Hospital, Aachen, Germany
| | - Kosta Theodorou
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Linsey J. F. Peters
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands,Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, Aachen, Germany,Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University Hospital, Aachen, Germany
| | - Han Jin
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Timo Rademakers
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Marion J. Gijbels
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands,Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands,Department of Medical Biochemistry, Amsterdam UMC, Locatie AMC, Amsterdam, Netherlands
| | - Mat Rousch
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Yvonne Jansen
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Munich, Germany,German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Michael Lehrke
- Department of Internal Medicine I, RWTH Aachen University Hospital, Aachen, Germany
| | - Corinna Lebherz
- Department of Internal Medicine I, RWTH Aachen University Hospital, Aachen, Germany
| | - Daniela Yildiz
- Institute of Molecular Pharmacology, RWTH Aachen University Hospital, Aachen, Germany,Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jacob F. Bentzon
- Experimental Pathology of Atherosclerosis Laboratory, Spanish National Center for Cardiovascular Research (CNIC), Madrid, Spain,Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Erik A. L. Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands,Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, Aachen, Germany
| | - Marjo M. P. C. Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands,*Correspondence: Marjo M. P. C. Donners,
| |
Collapse
|
33
|
Li J, Zhao N, Zhang W, Li P, Yin X, Zhang W, Wang H, Tang B. Assessing the Progression of Early Atherosclerosis Mice Using a Fluorescence Nanosensor for the Simultaneous Detection and Imaging of pH and Phosphorylation. Angew Chem Int Ed Engl 2023; 62:e202215178. [PMID: 36357335 DOI: 10.1002/anie.202215178] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 10/30/2022] [Accepted: 11/09/2022] [Indexed: 11/12/2022]
Abstract
The inflammatory microenvironment involves changes in pH and protein phosphorylation state and is closely related to the occurrence and development of atherosclerosis (AS). Herein, we constructed a dual-detection fluorescence nanosensor PCN-NP-HPZ based on post modification of MOFs, which realized the simultaneous detection and imaging of pH and phosphorylation through the pH-sensitive group piperazine and the ZrIV node of the MOFs. The sensors were used to monitor changes in blood pH and phosphate levels at different time stages during atherosclerotic plaque formation. Two-photon fluorescence imaging was also performed in the vascular endothelium. Blood tests combined with two-photon fluorescence images indicated that in the early stage of AS, blood and tissue pH levels were lower than that of the normal mice, while phosphate and tissue phosphorylation levels were higher than that of the normal mice. The present study provides a new analysis method for the assessment of early atherosclerotic disease.
Collapse
Affiliation(s)
- Jin Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Na Zhao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Xia Yin
- Molecular Science and Biomedicine Laboratory(MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Wen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| |
Collapse
|
34
|
May L, Bartolo B, Harrison D, Guzik T, Drummond G, Figtree G, Ritchie R, Rye KA, de Haan J. Translating atherosclerosis research from bench to bedside: navigating the barriers for effective preclinical drug discovery. Clin Sci (Lond) 2022; 136:1731-1758. [PMID: 36459456 PMCID: PMC9727216 DOI: 10.1042/cs20210862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/21/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide. An ongoing challenge remains the development of novel pharmacotherapies to treat CVD, particularly atherosclerosis. Effective mechanism-informed development and translation of new drugs requires a deep understanding of the known and currently unknown biological mechanisms underpinning atherosclerosis, accompanied by optimization of traditional drug discovery approaches. Current animal models do not precisely recapitulate the pathobiology underpinning human CVD. Accordingly, a fundamental limitation in early-stage drug discovery has been the lack of consensus regarding an appropriate experimental in vivo model that can mimic human atherosclerosis. However, when coupled with a clear understanding of the specific advantages and limitations of the model employed, preclinical animal models remain a crucial component for evaluating pharmacological interventions. Within this perspective, we will provide an overview of the mechanisms and modalities of atherosclerotic drugs, including those in the preclinical and early clinical development stage. Additionally, we highlight recent preclinical models that have improved our understanding of atherosclerosis and associated clinical consequences and propose model adaptations to facilitate the development of new and effective treatments.
Collapse
Affiliation(s)
- Lauren T. May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | | | - David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN, U.S.A
| | - Tomasz Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
| | - Gemma A. Figtree
- Kolling Research Institute, University of Sydney, Sydney, Australia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Rebecca H. Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney 2052, Australia
| | - Judy B. de Haan
- Cardiovascular Inflammation and Redox Biology Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- Department Cardiometabolic Health, University of Melbourne, Parkville, Victoria 3010, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria 3086, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| |
Collapse
|
35
|
Gogiraju R, Renner L, Bochenek ML, Zifkos K, Molitor M, Danckwardt S, Wenzel P, Münzel T, Konstantinides S, Schäfer K. Arginase-1 Deletion in Erythrocytes Promotes Vascular Calcification via Enhanced GSNOR (S-Nitrosoglutathione Reductase) Expression and NO Signaling in Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2022; 42:e291-e310. [PMID: 36252109 DOI: 10.1161/atvbaha.122.318338] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Erythrocytes (red blood cells) participate in the control of vascular NO bioavailability. The purpose of this study was to determine whether and how genetic deletion of ARG1 (arginase-1) affects vascular smooth muscle cell NO signaling, osteoblastic differentiation, and atherosclerotic lesion calcification. METHODS Atherosclerosis-prone mice with conditional, erythrocyte-restricted deletion of ARG1 (apoE-/- red blood cell.ARG1 knockout) were generated and vascular calcification studied using molecular imaging of the osteogenic activity agent OsteoSense, Alizarin staining or immunohistochemistry, qPCR of osteogenic markers and ex vivo assays. RESULTS Atherosclerotic lesion size at the aortic root did not differ, but calcification was significantly more pronounced in apoE-/- mice lacking erythrocyte ARG1. Incubation of murine and human VSMCs with lysed erythrocyte membranes from apoE-/- red blood cell. ARG1-knockout mice accelerated their osteogenic differentiation, and mRNA transcripts of osteogenic markers decreased following NO scavenging. In addition to NO signaling via sGC (soluble guanylyl cyclase), overexpression of GSNOR (S-nitrosoglutathione reductase) enhanced degradation of S-nitrosoglutathione to glutathione and reduced protein S-nitrosation of HSP (heat shock protein)-70 were identified as potential mechanisms of vascular smooth muscle cell calcification in mice lacking ARG1 in erythrocytes, and calcium phosphate deposition was enhanced by heat shock and prevented by GSNOR inhibition. Messenger RNA levels of enzymes metabolizing the arginase products L-ornithine and L-proline also were elevated in VSMCs, paralleled by increased proliferation, myofibroblast marker and collagen type 1 expression. CONCLUSIONS Our findings support an important role of erythrocyte ARG1 for NO bioavailability and L-arginine metabolism in VSMCs, which controls atherosclerotic lesion composition and calcification.
Collapse
Affiliation(s)
- Rajinikanth Gogiraju
- Department of Cardiology, Cardiology I (R.G., L.R., M.L.B., M.M., P.W., T.M., K.S.), University Medical Center Mainz, Germany
| | - Luisa Renner
- Department of Cardiology, Cardiology I (R.G., L.R., M.L.B., M.M., P.W., T.M., K.S.), University Medical Center Mainz, Germany
| | - Magdalena L Bochenek
- Department of Cardiology, Cardiology I (R.G., L.R., M.L.B., M.M., P.W., T.M., K.S.), University Medical Center Mainz, Germany.,Center for Thrombosis and Hemostasis (M.L.B., K.Z., M.M., S.D., P.W., S.K.), University Medical Center Mainz, Germany
| | - Konstantinos Zifkos
- Center for Thrombosis and Hemostasis (M.L.B., K.Z., M.M., S.D., P.W., S.K.), University Medical Center Mainz, Germany
| | - Michael Molitor
- Department of Cardiology, Cardiology I (R.G., L.R., M.L.B., M.M., P.W., T.M., K.S.), University Medical Center Mainz, Germany.,Center for Thrombosis and Hemostasis (M.L.B., K.Z., M.M., S.D., P.W., S.K.), University Medical Center Mainz, Germany
| | - Sven Danckwardt
- Center for Thrombosis and Hemostasis (M.L.B., K.Z., M.M., S.D., P.W., S.K.), University Medical Center Mainz, Germany.,Institute for Clinical Chemistry (S.D.), University Medical Center Mainz, Germany
| | - Philip Wenzel
- Department of Cardiology, Cardiology I (R.G., L.R., M.L.B., M.M., P.W., T.M., K.S.), University Medical Center Mainz, Germany.,Center for Thrombosis and Hemostasis (M.L.B., K.Z., M.M., S.D., P.W., S.K.), University Medical Center Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Cardiology I (R.G., L.R., M.L.B., M.M., P.W., T.M., K.S.), University Medical Center Mainz, Germany
| | - Stavros Konstantinides
- Center for Thrombosis and Hemostasis (M.L.B., K.Z., M.M., S.D., P.W., S.K.), University Medical Center Mainz, Germany
| | - Katrin Schäfer
- Department of Cardiology, Cardiology I (R.G., L.R., M.L.B., M.M., P.W., T.M., K.S.), University Medical Center Mainz, Germany
| |
Collapse
|
36
|
Singh D, Rai V, Agrawal DK. Non-Coding RNAs in Regulating Plaque Progression and Remodeling of Extracellular Matrix in Atherosclerosis. Int J Mol Sci 2022; 23:13731. [PMID: 36430208 PMCID: PMC9692922 DOI: 10.3390/ijms232213731] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/31/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022] Open
Abstract
Non-coding RNAs (ncRNAs) regulate cell proliferation, migration, differentiation, inflammation, metabolism of clinically important biomolecules, and other cellular processes. They do not encode proteins but are involved in the regulatory network of various proteins that are directly related to the pathogenesis of diseases. Little is known about the ncRNA-associated mechanisms of atherosclerosis and related cardiovascular disorders. Remodeling of the extracellular matrix (ECM) is critical in the pathogenesis of atherosclerosis and related disorders; however, its regulatory proteins are the potential subjects to explore with special emphasis on epigenetic regulatory components. The activity of regulatory proteins involved in ECM remodeling is regulated by various ncRNA molecules, as evident from recent research. Thus, it is important to critically evaluate the existing literature to enhance the understanding of nc-RNAs-regulated molecular mechanisms regulating ECM components, remodeling, and progression of atherosclerosis. This is crucial since deregulated ECM remodeling contributes to atherosclerosis. Thus, an in-depth understanding of ncRNA-associated ECM remodeling may identify novel targets for the treatment of atherosclerosis and other cardiovascular diseases.
Collapse
Affiliation(s)
| | | | - Devendra K. Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
37
|
Therapeutic inhibition of MPO stabilizes pre-existing high risk atherosclerotic plaque. Redox Biol 2022; 58:102532. [PMID: 36375379 PMCID: PMC9663534 DOI: 10.1016/j.redox.2022.102532] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/29/2022] [Accepted: 11/03/2022] [Indexed: 11/06/2022] Open
Abstract
Currently there are no established therapies to treat high-risk patients with unstable atherosclerotic lesions that are prone to rupture and can result in thrombosis, abrupt arterial occlusion, and a precipitous infarction. Rather than being stenotic, rupture-prone non-occlusive plaques are commonly enriched with inflammatory cells and have a thin fibrous cap. We reported previously that inhibition of the pro-inflammatory enzyme myeloperoxidase (MPO) with the suicide inhibitor AZM198 prevents formation of unstable plaque in the Tandem Stenosis (TS) mouse model of plaque instability. However, in our previous study AZM198 was administered to animals before unstable plaque was present and hence it did not test the significant unmet clinical need present in high-risk patients with vulnerable atherosclerosis. In the present study we therefore asked whether pharmacological inhibition of MPO with AZM198 can stabilize pre-existing unstable lesions in an interventional setting using the mouse model of plaque instability. In vivo molecular magnetic resonance imaging of arterial MPO activity using bis-5-hydroxytryptamide-DTPA-Gd and histological analyses revealed that arterial MPO activity was elevated one week after TS surgery, prior to the presence of unstable lesions observed two weeks after TS surgery. Animals with pre-existing unstable plaque were treated with AZM198 for one or five weeks. Both short- and long-term intervention effectively inhibited arterial MPO activity and increased fibrous cap thickness, indicative of a more stable plaque phenotype. Plaque stabilization was observed without AZM198 affecting the arterial content of Ly6B.2+- and CD68+-cells and MPO protein. These findings demonstrate that inhibition of arterial MPO activity converts unstable into stable atherosclerotic lesions in a preclinical model of plaque instability and highlight the potential therapeutic potency of MPO inhibition for the management of high-risk patients and the development of novel protective strategies against cardiovascular diseases.
Collapse
|
38
|
He F, Wang X, Hua L, Guo T. Numerical analysis of hemodynamics in pulmonary artery stenosis. Biomed Mater Eng 2022; 34:235-246. [DOI: 10.3233/bme-221418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Pulmonary artery stenosis is a serious threat to people’s life and health. OBJECTIVE: The hydrodynamic mechanism of pulmonary artery stenosis is investigated. METHODS: Numerical analysis of hemodynamics in pulmonary artery stenosis using computational fluid dynamics techniques is a well-established method. An idealized model of pulmonary stenosis is established, and the model is divided into main pulmonary artery, right and left pulmonary arteries, and their branches. The sections at different positions are intercepted to study the distribution trend of maximum velocity, pressure and wall shear stress. RESULTS: The numerical simulation results show that the pressure drop at both ends of the narrow is large. High velocity and wall shear stress exist in the center of stenosis, and the wall shear stress at the distal end of stenosis gradually decreases, resulting in endothelial dysfunction. CONCLUSIONS: To some extent, this study helps clinicians make diagnosis and treatment plans in advance and improve prognosis. This method could be used in the numerical simulation of practical models.
Collapse
Affiliation(s)
- Fan He
- School of Science, , , China
| | | | - Lu Hua
- , , State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, , , China
| | - Tingting Guo
- , , State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, , , China
| |
Collapse
|
39
|
Shen Y, Xu LR, Yan D, Zhou M, Han TL, Lu C, Tang X, Lin CP, Qian RZ, Guo DQ. BMAL1 modulates smooth muscle cells phenotypic switch towards fibroblast-like cells and stabilizes atherosclerotic plaques by upregulating YAP1. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166450. [PMID: 35598770 DOI: 10.1016/j.bbadis.2022.166450] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/03/2022] [Accepted: 05/13/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Ischemic heart diseases and ischemic stroke are closely related to circadian clock and unstable atherosclerotic plaques. Vascular smooth muscle cells (VSMCs) can stabilize or destabilize an atherosclerotic lesion through phenotypic switch. BMAL1 is not only an indispensable core component in circadian clock but also an important regulator in atherosclerosis and VSMCs proliferation. However, little is known about the modulation mechanisms of BMAL1 in VSMCs phenotypic switch and atherosclerotic plaque stability. METHODS We integrated histological analysis of human plaques, in vivo experiments of VSMC-specific Bmal1-/- mice, in vitro experiments, and gene set enrichment analysis (GSEA) of public datasets of human plaques to explore the function of BMAL1 in VSMCs phonotypic switch and plaque stability. FINDINGS Comparing to human unstable plaques, BMAL1 was higher in stable plaques, accompanied by elevated YAP1 and fibroblast maker FSP1 which were positively correlated with BMAL1. In response to Methyl-β-cyclodextrin-cholesterol, oxidized-low-density-lipoprotein and platelet-derived-growth-factor-BB, VSMCs embarked on phenotypic switch and upregulated BMAL, YAP1 and FSP1. Besides, BMAL1 overexpression promoted VSMCs phonotypic switch towards fibroblast-like cells by transcriptionally upregulating the expression of YAP1. BMAL1 or YAP1 knock-down inhibited VSMCs phonotypic switch and downregulated FSP1. Furthermore, VSMC-specific Bmal1-/- mice exhibited VSMCs with lower YAP1 and FSP1 levels, and more vulnerable plaques with less collagen content. In addition, BMAL1 suppressed the migration of VSMCs. The GSEA results of public datasets were consistent with our laboratory findings. INTERPRETATION Our results highlight the importance of BMAL1 as a major regulator in VSMCs phenotypic switch towards fibroblast-like cells which stabilize an atherosclerotic plaque.
Collapse
Affiliation(s)
- Yang Shen
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Li-Rong Xu
- Department of Pathology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dong Yan
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Min Zhou
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Tong-Lei Han
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Chao Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Rd., Shanghai 200032, China
| | - Xiao Tang
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Chang-Po Lin
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China.
| | - Rui-Zhe Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Rd., Shanghai 200032, China.
| | - Da-Qiao Guo
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China.
| |
Collapse
|
40
|
El Hage R, Knippschild U, Arnold T, Hinterseher I. Stem Cell-Based Therapy: A Promising Treatment for Diabetic Foot Ulcer. Biomedicines 2022; 10:1507. [PMID: 35884812 PMCID: PMC9312797 DOI: 10.3390/biomedicines10071507] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Diabetic foot ulcer (DFU) is a severe complication of diabetes and a challenging medical condition. Conventional treatments for DFU have not been effective enough to reduce the amputation rates, which urges the need for additional treatment. Stem cell-based therapy for DFU has been investigated over the past years. Its therapeutic effect is through promoting angiogenesis, secreting paracrine factors, stimulating vascular differentiation, suppressing inflammation, improving collagen deposition, and immunomodulation. It is controversial which type and origin of stem cells, and which administration route would be the most optimal for therapy. We reviewed the different types and origins of stem cells and routes of administration used for the treatment of DFU in clinical and preclinical studies. Diabetes leads to the impairment of the stem cells in the diseased patients, which makes it less ideal to use autologous stem cells, and requires looking for a matching donor. Moreover, angioplasty could be complementary to stem cell therapy, and scaffolds have a positive impact on the healing process of DFU by stem cell-based therapy. In short, stem cell-based therapy is promising in the field of regenerative medicine, but more studies are still needed to determine the ideal type of stem cells required in therapy, their safety, proper dosing, and optimal administration route.
Collapse
Affiliation(s)
- Racha El Hage
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, Fehrbelliner Str. 38, 16816 Neuruppin, Germany;
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Center, Ulm University, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (U.K.); (T.A.)
| | - Tobias Arnold
- Department of General and Visceral Surgery, Surgery Center, Ulm University, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (U.K.); (T.A.)
| | - Irene Hinterseher
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, Fehrbelliner Str. 38, 16816 Neuruppin, Germany;
- Berlin Institute of Health, Vascular Surgery Clinic, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane und der Brandenburgischen Technischen Universität Cottbus—Senftenberg, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany
| |
Collapse
|
41
|
Genetic Variants of MIR27A, MIR196A2 May Impact the Risk for the Onset of Coronary Artery Disease in the Pakistani Population. Genes (Basel) 2022; 13:genes13050747. [PMID: 35627132 PMCID: PMC9141586 DOI: 10.3390/genes13050747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 02/04/2023] Open
Abstract
Genetic variants in microRNA genes have a detrimental effect on miRNA-mediated regulation of gene expression and may contribute to coronary artery disease (CAD). CAD is the primary cause of mortality worldwide. Several environmental, genetic, and epigenetic factors are responsible for CAD susceptibility. The contribution of protein-coding genes is extensively studied. However, the role of microRNA genes in CAD is at infancy. The study is aimed to investigate the impact of rs895819, rs11614913, and rs2168518 variants in MIR27A, MIR196A2, and MIR4513, respectively, in CAD using allele-specific PCR. Results: For variant rs11614913, significant distribution of the genotypes among the cases and controls was determined by co-dominant [χ2 = 54.4; p value ≤ 0.0001], dominant (C/C vs. C/T + T/T) [OR = 0.257 (0.133-0.496); p value ≤ 0.0001], recessive (T/T vs. C/T + C/C) [OR = 1.56 (0.677-0.632); p value = 0.398], and additive models [OR = 0.421 (0.262-0.675); p value = 0.0004]. Similarly, a significant association of rs895819 was determined by co-dominant [χ2 = 9.669; p value ≤ 0.008], dominant (A/A vs. A/G + G/G) [OR = 0.285 (0.1242-0.6575); p value ≤ 0.0034], recessive (G/G vs. A/G + A/A) [OR = 0.900 (0.3202-3.519); p value = 1.000], and additive models [OR = 0.604 (0.3640-1.002); p value = 0.05] while no significant association of rs2168518 with CAD was found. Conclusion: The variants rs895819 and rs11614913 are the susceptibility factors for CAD.
Collapse
|
42
|
Li J, Thiele S, Kirk RW, Quirk BC, Hoogendoorn A, Chen YC, Peter K, Nicholls SJ, Verjans JW, Psaltis PJ, Bursill C, Herkommer AM, Giessen H, McLaughlin RA. 3D-Printed Micro Lens-in-Lens for In Vivo Multimodal Microendoscopy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107032. [PMID: 35229467 DOI: 10.1002/smll.202107032] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/19/2022] [Indexed: 06/14/2023]
Abstract
Multimodal microendoscopes enable co-located structural and molecular measurements in vivo, thus providing useful insights into the pathological changes associated with disease. However, different optical imaging modalities often have conflicting optical requirements for optimal lens design. For example, a high numerical aperture (NA) lens is needed to realize high-sensitivity fluorescence measurements. In contrast, optical coherence tomography (OCT) demands a low NA to achieve a large depth of focus. These competing requirements present a significant challenge in the design and fabrication of miniaturized imaging probes that are capable of supporting high-quality multiple modalities simultaneously. An optical design is demonstrated which uses two-photon 3D printing to create a miniaturized lens that is simultaneously optimized for these conflicting imaging modalities. The lens-in-lens design contains distinct but connected optical surfaces that separately address the needs of both fluorescence and OCT imaging within a lens of 330 µm diameter. This design shows an improvement in fluorescence sensitivity of >10x in contrast to more conventional fiber-optic design approaches. This lens-in-lens is then integrated into an intravascular catheter probe with a diameter of 520 µm. The first simultaneous intravascular OCT and fluorescence imaging of a mouse artery in vivo is reported.
Collapse
Affiliation(s)
- Jiawen Li
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, SA, 5005, Australia
- School of Electrical and Electronic Engineering, University of Adelaide, Adelaide, SA, 5005, Australia
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Simon Thiele
- Institute of Applied Optics (ITO) and Research Center SCoPE, University of Stuttgart, 70569, Stuttgart, Germany
| | - Rodney W Kirk
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, SA, 5005, Australia
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, SA, 5005, Australia
- School of Biomedicine, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Bryden C Quirk
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, SA, 5005, Australia
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, SA, 5005, Australia
- School of Biomedicine, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Ayla Hoogendoorn
- Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Yung Chih Chen
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Department of Cardiometabolic Health, Bio21 Institute, Melbourne Medical School, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Department of Cardiometabolic Health, Bio21 Institute, Melbourne Medical School, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Stephen J Nicholls
- Victorian Heart Institute, Monash University, Melbourne, VIC, 3168, Australia
| | - Johan W Verjans
- Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
- Department of Cardiology, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Peter J Psaltis
- Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
- Department of Cardiology, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Christina Bursill
- Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Alois M Herkommer
- Institute of Applied Optics (ITO) and Research Center SCoPE, University of Stuttgart, 70569, Stuttgart, Germany
| | - Harald Giessen
- 4th Physics Institute and Research Center SCoPE, University of Stuttgart, 70569, Stuttgart, Germany
| | - Robert A McLaughlin
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, SA, 5005, Australia
- Institute for Photonics and Advanced Sensing, The University of Adelaide, Adelaide, SA, 5005, Australia
- School of Biomedicine, University of Adelaide, Adelaide, SA, 5005, Australia
| |
Collapse
|
43
|
Jebari-Benslaiman S, Galicia-García U, Larrea-Sebal A, Olaetxea JR, Alloza I, Vandenbroeck K, Benito-Vicente A, Martín C. Pathophysiology of Atherosclerosis. Int J Mol Sci 2022; 23:ijms23063346. [PMID: 35328769 PMCID: PMC8954705 DOI: 10.3390/ijms23063346] [Citation(s) in RCA: 376] [Impact Index Per Article: 125.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/12/2022] [Accepted: 03/18/2022] [Indexed: 11/26/2022] Open
Abstract
Atherosclerosis is the main risk factor for cardiovascular disease (CVD), which is the leading cause of mortality worldwide. Atherosclerosis is initiated by endothelium activation and, followed by a cascade of events (accumulation of lipids, fibrous elements, and calcification), triggers the vessel narrowing and activation of inflammatory pathways. The resultant atheroma plaque, along with these processes, results in cardiovascular complications. This review focuses on the different stages of atherosclerosis development, ranging from endothelial dysfunction to plaque rupture. In addition, the post-transcriptional regulation and modulation of atheroma plaque by microRNAs and lncRNAs, the role of microbiota, and the importance of sex as a crucial risk factor in atherosclerosis are covered here in order to provide a global view of the disease.
Collapse
Affiliation(s)
- Shifa Jebari-Benslaiman
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
| | - Unai Galicia-García
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Fundación Biofisika Bizkaia, Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain
| | - Asier Larrea-Sebal
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Fundación Biofisika Bizkaia, Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain
| | | | - Iraide Alloza
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - Koen Vandenbroeck
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Bizkaia, Spain
| | - Asier Benito-Vicente
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Correspondence: (A.B.-V.); (C.M.); Tel.: +34-946-01-2741 (C.M.)
| | - César Martín
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Correspondence: (A.B.-V.); (C.M.); Tel.: +34-946-01-2741 (C.M.)
| |
Collapse
|
44
|
Functional Phenotypes of Intraplaque Macrophages and Their Distinct Roles in Atherosclerosis Development and Atheroinflammation. Biomedicines 2022; 10:biomedicines10020452. [PMID: 35203661 PMCID: PMC8962399 DOI: 10.3390/biomedicines10020452] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/04/2022] [Accepted: 02/11/2022] [Indexed: 12/01/2022] Open
Abstract
Macrophages are the key inflammatory cell type involved in all stages of atherosclerosis development and progression, as demonstrated by numerous studies. Correspondingly, macrophages are currently regarded as a promising therapeutic target for the development of new treatment approaches. The macrophage population is heterogeneous and dynamic, as these cells can switch between a number of distinct functional states with pro- and anti-atherogenic activity in response to various stimuli. An atherosclerotic plaque microenvironment defined by cytokine levels, cell-to-cell interactions, lipid accumulation, hypoxia, neoangiogenesis, and intraplaque haemorrhage may guide local macrophage polarization processes within the lesion. In this review, we discuss known functional phenotypes of intraplaque macrophages and their distinct contribution to ahteroinflammation.
Collapse
|
45
|
Izquierdo-Garcia D, Diyabalanage H, Ramsay IA, Rotile NJ, Mauskapf A, Choi JK, Witzel T, Humblet V, Jaffer FA, Brownell AL, Tawakol A, Catana C, Conrad MF, Caravan P, Ay I. Imaging High-Risk Atherothrombosis Using a Novel Fibrin-Binding Positron Emission Tomography Probe. Stroke 2022; 53:595-604. [PMID: 34965737 PMCID: PMC8792326 DOI: 10.1161/strokeaha.121.035638] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND PURPOSE High-risk atherosclerosis is an underlying cause of cardiovascular events, yet identifying the specific patient population at immediate risk is still challenging. Here, we used a rabbit model of atherosclerotic plaque rupture and human carotid endarterectomy specimens to describe the potential of molecular fibrin imaging as a tool to identify thrombotic plaques. METHODS Atherosclerotic plaques in rabbits were induced using a high-cholesterol diet and aortic balloon injury (N=13). Pharmacological triggering was used in a group of rabbits (n=9) to induce plaque disruption. Animals were grouped into thrombotic and nonthrombotic plaque groups based on gross pathology (gold standard). All animals were injected with a novel fibrin-specific probe 68Ga-CM246 followed by positron emission tomography (PET)/magnetic resonance imaging 90 minutes later. 68Ga-CM246 was quantified on the PET images using tissue-to-background (back muscle) ratios and standardized uptake value. RESULTS Both tissue-to-background (back muscle) ratios and standardized uptake value were significantly higher in the thrombotic versus nonthrombotic group (P<0.05). Ex vivo PET and autoradiography of the abdominal aorta correlated positively with in vivo PET measurements. Plaque disruption identified by 68Ga-CM246 PET agreed with gross pathology assessment (85%). In ex vivo surgical specimens obtained from patients undergoing elective carotid endarterectomy (N=12), 68Ga-CM246 showed significantly higher binding to carotid plaques compared to a D-cysteine nonbinding control probe. CONCLUSIONS We demonstrated that molecular fibrin PET imaging using 68Ga-CM246 could be a useful tool to diagnose experimental and clinical atherothrombosis. Based on our initial results using human carotid plaque specimens, in vivo molecular imaging studies are warranted to test 68Ga-CM246 PET as a tool to stratify risk in atherosclerotic patients.
Collapse
Affiliation(s)
- David Izquierdo-Garcia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA,Harvard-MIT Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA
| | | | - Ian A. Ramsay
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA,Collagen Medical, LLC, Belmont, MA,The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA
| | - Nicholas J. Rotile
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA,The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA
| | - Adam Mauskapf
- Cardiovascular Research Center, Division of Cardiology, Department of Medicine Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ji-Kyung Choi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | - Thomas Witzel
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | | | - Farouc A. Jaffer
- Cardiovascular Research Center, Division of Cardiology, Department of Medicine Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Anna-Liisa Brownell
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | - Ahmed Tawakol
- Nuclear Cardiology, Division of Cardiology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ciprian Catana
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA,The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA
| | - Mark F. Conrad
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA,The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA
| | - Ilknur Ay
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| |
Collapse
|
46
|
Gerhardt T, Haghikia A, Stapmanns P, Leistner DM. Immune Mechanisms of Plaque Instability. Front Cardiovasc Med 2022; 8:797046. [PMID: 35087883 PMCID: PMC8787133 DOI: 10.3389/fcvm.2021.797046] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/15/2021] [Indexed: 01/08/2023] Open
Abstract
Inflammation crucially drives atherosclerosis from disease initiation to the emergence of clinical complications. Targeting pivotal inflammatory pathways without compromising the host defense could compliment therapy with lipid-lowering agents, anti-hypertensive treatment, and lifestyle interventions to address the substantial residual cardiovascular risk that remains beyond classical risk factor control. Detailed understanding of the intricate immune mechanisms that propel plaque instability and disruption is indispensable for the development of novel therapeutic concepts. In this review, we provide an overview on the role of key immune cells in plaque inception and progression, and discuss recently identified maladaptive immune phenomena that contribute to plaque destabilization, including epigenetically programmed trained immunity in myeloid cells, pathogenic conversion of autoreactive regulatory T-cells and expansion of altered leukocytes due to clonal hematopoiesis. From a more global perspective, the article discusses how systemic crises such as acute mental stress or infection abruptly raise plaque vulnerability and summarizes recent advances in understanding the increased cardiovascular risk associated with COVID-19 disease. Stepping outside the box, we highlight the role of gut dysbiosis in atherosclerosis progression and plaque vulnerability. The emerging differential role of the immune system in plaque rupture and plaque erosion as well as the limitations of animal models in studying plaque disruption are reviewed.
Collapse
Affiliation(s)
- Teresa Gerhardt
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Arash Haghikia
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Philip Stapmanns
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
| | - David Manuel Leistner
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: David Manuel Leistner
| |
Collapse
|
47
|
ADAM and ADAMTS disintegrin and metalloproteinases as major factors and molecular targets in vascular malfunction and disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:255-363. [PMID: 35659374 PMCID: PMC9231755 DOI: 10.1016/bs.apha.2021.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A Disintegrin and Metalloproteinase (ADAM) and A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) are two closely related families of proteolytic enzymes. ADAMs are largely membrane-bound enzymes that act as molecular scissors or sheddases of membrane-bound proteins, growth factors, cytokines, receptors and ligands, whereas ADAMTS are mainly secreted enzymes. ADAMs have a pro-domain, and a metalloproteinase, disintegrin, cysteine-rich and transmembrane domain. Similarly, ADAMTS family members have a pro-domain, and a metalloproteinase, disintegrin, and cysteine-rich domain, but instead of a transmembrane domain they have thrombospondin motifs. Most ADAMs and ADAMTS are activated by pro-protein convertases, and can be regulated by G-protein coupled receptor agonists, Ca2+ ionophores and protein kinase C. Activated ADAMs and ADAMTS participate in numerous vascular processes including angiogenesis, vascular smooth muscle cell proliferation and migration, vascular cell apoptosis, cell survival, tissue repair, and wound healing. ADAMs and ADAMTS also play a role in vascular malfunction and cardiovascular diseases such as hypertension, atherosclerosis, coronary artery disease, myocardial infarction, heart failure, peripheral artery disease, and vascular aneurysm. Decreased ADAMTS13 is involved in thrombotic thrombocytopenic purpura and microangiopathies. The activity of ADAMs and ADAMTS can be regulated by endogenous tissue inhibitors of metalloproteinases and other synthetic small molecule inhibitors. ADAMs and ADAMTS can be used as diagnostic biomarkers and molecular targets in cardiovascular disease, and modulators of ADAMs and ADAMTS activity may provide potential new approaches for the management of cardiovascular disorders.
Collapse
|
48
|
Chen YC, Jandeleit-Dahm K, Peter K. Sodium-Glucose Co-Transporter 2 (SGLT2) Inhibitor Dapagliflozin Stabilizes Diabetes-Induced Atherosclerotic Plaque Instability. J Am Heart Assoc 2021; 11:e022761. [PMID: 34970931 PMCID: PMC9075188 DOI: 10.1161/jaha.121.022761] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Background Diabetes is known to accelerate atherosclerosis and increase plaque instability. However, there has been a lack of suitable animal models to study the effect of diabetes on plaque instability. We hypothesized that the tandem stenosis mouse model, which reflects plaque instability/rupture as seen in patients, can be applied to study the effects of diabetes and respective therapeutics on plaque instability/rupture. Methods and Results ApoE−/− mice at 7 weeks of age were rendered diabetic with streptozotocin and 5 weeks later were surgically subjected to tandem stenosis in the right carotid artery and fed with a high‐fat diet for 7 weeks. As a promising new antidiabetic drug class, a sodium glucose co‐transporter 2 inhibitor was tested in this new model. Diabetic mice showed an increase in the size of unstable atherosclerotic plaques and in the plaque instability markers MCP‐1, CD68, and necrotic core size. Mice treated with dapagliflozin demonstrated attenuated glucose and triglyceride levels. Importantly, these mice demonstrated plaque stabilization with enhanced collagen accumulation, increased fibrosis, increased cap‐to‐lesion height ratios, and significant upregulation of the vasculoprotective NADPH oxidase 4 expression. Conclusions The tandem stenosis mouse model in combination with the application of streptozotocin represents a highly suitable and unique mouse model for studying plaque destabilization under diabetic conditions. Furthermore, for the first time, we provide evidence of plaque‐stabilizing effects of sodium‐glucose co‐transporter 2 inhibitor. Our data also suggest that this newly developed mouse model is an attractive preclinical tool for testing antidiabetic drugs for the highly sought‐after potential to stabilize atherosclerotic plaques.
Collapse
Affiliation(s)
- Yung-Chih Chen
- Baker Heart & Diabetes Institute Melbourne Australia.,Central Clinical School Monash University Melbourne Australia.,Department of Cardiometabolic Health University of Melbourne Australia
| | - Karin Jandeleit-Dahm
- Department of Diabetes Monash University Melbourne Australia.,German Diabetes Centre Leibniz Centre for Diabetes Research at Heinrich Heine University Dusseldorf Germany
| | - Karlheinz Peter
- Baker Heart & Diabetes Institute Melbourne Australia.,Central Clinical School Monash University Melbourne Australia.,Department of Cardiometabolic Health University of Melbourne Australia.,Department of Cardiology The Alfred Hospital Melbourne Australia
| |
Collapse
|
49
|
Hagemeyer CE. Mopping up crystals to keep the blood flowing. EBioMedicine 2021; 75:103786. [PMID: 34963098 PMCID: PMC8717595 DOI: 10.1016/j.ebiom.2021.103786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 11/29/2022] Open
Affiliation(s)
- Christoph E Hagemeyer
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia.
| |
Collapse
|
50
|
Luo Y, Guo Y, Wang H, Yu M, Hong K, Li D, Li R, Wen B, Hu D, Chang L, Zhang J, Yang B, Sun D, Schwendeman AS, Eugene Chen Y. Phospholipid nanoparticles: Therapeutic potentials against atherosclerosis via reducing cholesterol crystals and inhibiting inflammation. EBioMedicine 2021; 74:103725. [PMID: 34879325 PMCID: PMC8654800 DOI: 10.1016/j.ebiom.2021.103725] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 01/11/2023] Open
Abstract
Background Atherosclerosis-related cardiovascular diseases (CVDs) are the leading cause of mortality worldwide. Cholesterol crystals (CCs) induce inflammation in atherosclerosis and are associated with unstable plaques and poor prognosis, but no drug can remove CCs in the clinic currently. Methods We generated a phospholipid-based and high-density lipoprotein (HDL)-like nanoparticle, miNano, and determined CC-dissolving capacity, cholesterol efflux property, and anti-inflammation effects of miNano in vitro. Both normal C57BL/6J and Apoe-deficient mice were used to explore the accumulation of miNano in atherosclerotic plaques. The efficacy and safety of miNano administration to treat atherosclerosis were evaluated in the Ldlr-deficient atherosclerosis model. The CC-dissolving capacity of miNano was also detected using human atherosclerotic plaques ex vivo. Findings We found that miNano bound to and dissolved CCs efficiently in vitro, and miNano accumulated in atherosclerotic plaques, co-localized with CCs and macrophages in vivo. Administration of miNano inhibited atherosclerosis and improved plaque stability by reducing CCs and macrophages in Ldlr-deficient mice with favorable safety profiles. In macrophages, miNano prevented foam cell formation by enhancing cholesterol efflux and suppressed inflammatory responses via inhibiting TLR4-NF-κB pathway. Finally, in an ex vivo experiment, miNano effectively dissolved CCs in human aortic atherosclerotic plaques. Interpretation Together, our work finds that phospholipid-based and HDL-like nanoparticle, miNano, has the potential to treat atherosclerosis by targeting CCs and stabilizing plaques. Funding This work was supported by the National Institutes of Health HL134569, HL109916, HL136231, and HL137214 to Y.E.C, HL138139 to J.Z., R21NS111191 to A.S., by the American Heart Association 15SDG24470155, Grant Awards (U068144 from Bio-interfaces and G024404 from M-BRISC) at the University of Michigan to Y.G., by the American Heart Association 19PRE34400017 and Rackham Helen Wu award to M.Y., NIH T32 GM07767 to K. H., Barbour Fellowship to D.L.
Collapse
Affiliation(s)
- Yonghong Luo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Second Xiangya Hospital, Central South University, Hunan Province, China
| | - Yanhong Guo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Huilun Wang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Minzhi Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kristen Hong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dan Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ruiting Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Die Hu
- Second Xiangya Hospital, Central South University, Hunan Province, China
| | - Lin Chang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jifeng Zhang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Bo Yang
- Department of Cardiac Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anna S Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Y Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Cardiac Surgery, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|