51
|
Nehl D, Goody PR, Maus K, Pfeifer A, Aikawa E, Bakthiary F, Zimmer S, Nickenig G, Jansen F, Hosen MR. Human and porcine aortic valve endothelial and interstitial cell isolation and characterization. Front Cardiovasc Med 2023; 10:1151028. [PMID: 37408661 PMCID: PMC10318150 DOI: 10.3389/fcvm.2023.1151028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/05/2023] [Indexed: 07/07/2023] Open
Abstract
Background Calcific aortic valve stenosis (AVS) is defined by pathological changes in the aortic valve (AV) and their predominant cell types: valvular interstitial (VICs) and endothelial cells (VECs). Understanding the cellular and molecular mechanisms of this disease is a prerequisite to identify potential pharmacological treatment strategies. In this study, we present a unique aortic valve cell isolation technique to acquire specific human and porcine cell populations and compared VICs and VECs of these species with each other for the first time. Methods AV cells were isolated from tissue obtained from human patients undergoing surgical aortic valve replacement (SAVR) or from porcine hearts. Functional analysis and in vitro experiments revealed that endothelial-to-mesenchymal transition (EndMT) can be induced in hVECs, leading to a significant increase in mesenchymal markers. In vitro calcification experiments of VICs demonstrated pronounced expression of calcification markers and visible calcific deposits in Alizarin Red staining in both species after incubation with pro-calcific media. Results Cells isolated from patient-derived AVs showed mesenchymal and endothelial-specific gene signatures (VIC and VEC, respectively). For instance, von Willebrand factor (vWF) and platelet endothelial adhesion molecule-1 (PECAM1) were upregulated in VECs, while the myofibroblastic markers alpha-smooth muscle actin (α-SMA) and vimentin (VIM) were downregulated in VECs compared to VICs. Analysis of cell function by migration revealed that VECs are more migratory than VICs. Induction of EndMT in vitro in VECs displayed increased expression of EndMT markers and decreased expression of endothelial markers, confirming their mesenchymal transdifferentiation ability. In vitro calcification of VICs revealed upregulation of alkaline phosphatase (ALPL), a hallmark of calcification. In addition, other calcification-related genes such as osteocalcin (BGLAP) and runt-related factor 2 (RUNX2) were upregulated. Alizarin red staining of calcified cells provided a further layer of confirmation that the isolated cells were VICs with osteoblastic differentiation capacity. Conclusion This study aims to take a first step towards standardizing a reproducible isolation technique for specific human and porcine VEC and VIC populations. A comparison of human and porcine aortic valve cells demonstrated that porcine cells may serve as an alternative cellular model system in settings where human tissue is difficult to obtain.
Collapse
Affiliation(s)
- D. Nehl
- Heart Center Bonn, Molecular Cardiology, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - P. R. Goody
- Heart Center Bonn, Molecular Cardiology, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - K. Maus
- Heart Center Bonn, Molecular Cardiology, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - A. Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, Bonn, Germany
| | - E. Aikawa
- Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - F. Bakthiary
- Heart Center Bonn, Department of Cardiac Surgery, University Hospital Bonn, Bonn, Germany
| | - S. Zimmer
- Heart Center Bonn, Molecular Cardiology, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - G. Nickenig
- Heart Center Bonn, Molecular Cardiology, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - F. Jansen
- Heart Center Bonn, Molecular Cardiology, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - M. R. Hosen
- Heart Center Bonn, Molecular Cardiology, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
52
|
Polidori T, De Santis D, Rucci C, Tremamunno G, Piccinni G, Pugliese L, Zerunian M, Guido G, Pucciarelli F, Bracci B, Polici M, Laghi A, Caruso D. Radiomics applications in cardiac imaging: a comprehensive review. LA RADIOLOGIA MEDICA 2023:10.1007/s11547-023-01658-x. [PMID: 37326780 DOI: 10.1007/s11547-023-01658-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 05/26/2023] [Indexed: 06/17/2023]
Abstract
Radiomics is a new emerging field that includes extraction of metrics and quantification of so-called radiomic features from medical images. The growing importance of radiomics applied to oncology in improving diagnosis, cancer staging and grading, and improved personalized treatment, has been well established; yet, this new analysis technique has still few applications in cardiovascular imaging. Several studies have shown promising results describing how radiomics principles could improve the diagnostic accuracy of coronary computed tomography angiography (CCTA) and magnetic resonance imaging (MRI) in diagnosis, risk stratification, and follow-up of patients with coronary heart disease (CAD), ischemic heart disease (IHD), hypertrophic cardiomyopathy (HCM), hypertensive heart disease (HHD), and many other cardiovascular diseases. Such quantitative approach could be useful to overcome the main limitations of CCTA and MRI in the evaluation of cardiovascular diseases, such as readers' subjectiveness and lack of repeatability. Moreover, this new discipline could potentially overcome some technical problems, namely the need of contrast administration or invasive examinations. Despite such advantages, radiomics is still not applied in clinical routine, due to lack of standardized parameters acquisition, inconsistent radiomic methods, lack of external validation, and different knowledge and experience among the readers. The purpose of this manuscript is to provide a recent update on the status of radiomics clinical applications in cardiovascular imaging.
Collapse
Affiliation(s)
- Tiziano Polidori
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy
| | - Domenico De Santis
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy
| | - Carlotta Rucci
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy
| | - Giuseppe Tremamunno
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy
| | - Giulia Piccinni
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy
| | - Luca Pugliese
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy
| | - Marta Zerunian
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy
| | - Gisella Guido
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy
| | - Francesco Pucciarelli
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy
| | - Benedetta Bracci
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy
| | - Michela Polici
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy
| | - Andrea Laghi
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy.
| | - Damiano Caruso
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome - Radiology Unit - Sant'Andrea University Hospital, Via di Grottarossa, 1035-1039, 00189, Rome, Italy
| |
Collapse
|
53
|
Pugliese L, Ricci F, Sica G, Scaglione M, Masala S. Non-Contrast and Contrast-Enhanced Cardiac Computed Tomography Imaging in the Diagnostic and Prognostic Evaluation of Coronary Artery Disease. Diagnostics (Basel) 2023; 13:2074. [PMID: 37370969 DOI: 10.3390/diagnostics13122074] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/07/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023] Open
Abstract
In recent decades, cardiac computed tomography (CT) has emerged as a powerful non-invasive tool for risk stratification, as well as the detection and characterization of coronary artery disease (CAD), which remains the main cause of morbidity and mortality in the world. Advances in technology have favored the increasing use of cardiac CT by allowing better performance with lower radiation doses. Coronary artery calcium, as assessed by non-contrast CT, is considered to be the best marker of subclinical atherosclerosis, and its use is recommended for the refinement of risk assessment in low-to-intermediate risk individuals. In addition, coronary CT angiography (CCTA) has become a gate-keeper to invasive coronary angiography (ICA) and revascularization in patients with acute chest pain by allowing the assessment not only of the extent of lumen stenosis, but also of its hemodynamic significance if combined with the measurement of fractional flow reserve or perfusion imaging. Moreover, CCTA provides a unique incremental value over functional testing and ICA by imaging the vessel wall, thus allowing the assessment of plaque burden, composition, and instability features, in addition to perivascular adipose tissue attenuation, which is a marker of vascular inflammation. There exists the potential to identify the non-obstructive lesions at high risk of progression to plaque rupture by combining all of these measures.
Collapse
Affiliation(s)
- Luca Pugliese
- Radiology Unit, Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Sant'Andrea University Hospital, 00189 Rome, Italy
| | - Francesca Ricci
- Radiology Unit, Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
| | - Giacomo Sica
- Radiology Unit, Monaldi Hospital, 80131 Napoli, Italy
| | - Mariano Scaglione
- Radiology Unit, Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
| | - Salvatore Masala
- Radiology Unit, Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
| |
Collapse
|
54
|
Xie L, Chen J, Wang Y, Jin C, Xie Y, Ma H, Xiang M. Emerging roles of macrophages in heart failure and associated treatment approaches. Ther Adv Chronic Dis 2023; 14:20406223231168755. [PMID: 37152348 PMCID: PMC10155014 DOI: 10.1177/20406223231168755] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/23/2023] [Indexed: 05/09/2023] Open
Abstract
Heart failure is typically caused by different cardiovascular conditions and has a poor prognosis. Despite the advances in treatment in recent decades, heart failure has remained a major cause of morbidity and mortality worldwide. As revealed by in vivo and in vitro experiments, inflammation plays a crucial role in adverse cardiac remodeling, ultimately leading to heart failure. Macrophages are central to the innate immune system, and they are the most indispensable cell type for all cardiac injuries and remodeling stages. The immediate microenvironment regulates their polarization and secretion. In this review, we summarize the phenotypic heterogeneity and governing roles of macrophages in the infarcted, inflamed, and aging heart and assess their significance as potential therapeutic targets in heart failure. We also highlight the current missing links and major challenges in the field that remain to be addressed before macrophages can be exploited for therapeutic applications.
Collapse
Affiliation(s)
- Lan Xie
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinyong Chen
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yidong Wang
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengjiang Jin
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Ma
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou 310009,
China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated
Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou 310009,
China
| |
Collapse
|
55
|
Skenteris NT, Hemme E, Delfos L, Karadimou G, Karlöf E, Lengquist M, Kronqvist M, Zhang X, Maegdefessel L, Schurgers LJ, Arnardottir H, Biessen EAL, Bot I, Matic L. Mast cells participate in smooth muscle cell reprogramming and atherosclerotic plaque calcification. Vascul Pharmacol 2023; 150:107167. [PMID: 36958707 DOI: 10.1016/j.vph.2023.107167] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/14/2023] [Accepted: 03/19/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND Calcification, a key feature of advanced human atherosclerosis, is positively associated with vascular disease burden and adverse events. We showed that macrocalcification can be a stabilizing factor for carotid plaque molecular biology, due to inverse association with immune processes. Mast cells (MCs) are important contributors to plaque instability, but their relationship with macrocalcification is unexplored. With a hypothesis that MC activation negatively associates with carotid plaque macrocalcification, we aimed to investigate the link between MCs and carotid plaque vulnerability, and study MC role in plaque calcification via smooth muscle cells (SMCs). METHODS Pre-operative computed tomography angiographies of patients (n = 40) undergoing surgery for carotid stenosis were used to characterize plaque morphology. Plaque microarrays (n = 40 and n = 126) were used for bioinformatic deconvolution of immune cell populations. Tissue microarrays (n = 103) were used to histologically validate the contribution of activated and resting MCs in plaques. RESULTS Activated MCs and their typical markers were negatively correlated with macrocalcification. The ratio of activated vs. resting MCs was increased in low-calcified plaques from symptomatic patients. There was no modulating effect of medication on MC ratios. In vitro experiments showed that SMC calcification attenuated MC activation, while both active and resting MCs stimulated SMC calcification and induced dedifferentiation towards a pro-inflammatory-, osteochondrocyte-like phenotype, without modulating their migro-proliferative function. CONCLUSIONS Integrative analyses from human plaques showed that MC activation is inversely associated with macrocalcification and positively with parameters of plaque vulnerability. Mechanistically, MCs induce SMC osteogenic reprograming, while matrix calcification in turn attenuates MC activation, offering new therapeutic avenues for exploration.
Collapse
Affiliation(s)
- Nikolaos T Skenteris
- Cardiovascular Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden; Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, the Netherlands
| | - Esmeralda Hemme
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| | - Lucie Delfos
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| | - Glykeria Karadimou
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Eva Karlöf
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Mariette Lengquist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Malin Kronqvist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Xiang Zhang
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Lars Maegdefessel
- Cardiovascular Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden; Technical University Munich, Klinikum rechts der Isar, Department for Vascular and Endovascular Surgery, Germany
| | - Leon J Schurgers
- Department of Biochemistry and CARIM, School for Cardiovascular Diseases, Maastricht University, Netherlands; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Hildur Arnardottir
- Cardiovascular Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, the Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| | - Ljubica Matic
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
56
|
Helicobacter Pylori Virulence Factor Cytotoxin-Associated Gene A (CagA) Induces Vascular Calcification in Coronary Artery Smooth Muscle Cells. Int J Mol Sci 2023; 24:ijms24065392. [PMID: 36982467 PMCID: PMC10049385 DOI: 10.3390/ijms24065392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
Helicobacter pylori (H. pylori) has been associated with cardiovascular diseases. The pro-inflammatory H. pylori virulence factor cytotoxin-associated gene A (CagA) has been detected in serum exosomes of H. pylori-infected subjects and may exert systemic effects throughout the cardiovascular system. The role of H. pylori and CagA in vascular calcification was hitherto unknown. The aim of this study was to determine the vascular effects of CagA through human coronary artery smooth muscle cell (CASMC) osteogenic and pro-inflammatory effector gene expression as well as interleukin 1β secretion and cellular calcification. CagA upregulated bone morphogenic protein 2 (BMP-2) associated with an osteogenic CASMC phenotype switch and induced increased cellular calcification. Furthermore, a pro-inflammatory response was observed. These results support that H. pylori may contribute to vascular calcification through CagA rendering CASMCs osteogenic and inducing calcification.
Collapse
|
57
|
De Azevedo D, Geers J, Gheysens O, Dweck M, Vancraeynest D. 18F-Sodium Fluoride PET/CT in Assessing Valvular Heart and Atherosclerotic Diseases. Semin Nucl Med 2023; 53:241-257. [PMID: 36116988 DOI: 10.1053/j.semnuclmed.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 08/22/2022] [Indexed: 11/11/2022]
Abstract
Aortic valve stenosis is the most common valvular disease in Western countries, while atherosclerotic cardiovascular disease is the foremost cause of death and disability worldwide. Valve degeneration and atherosclerosis are mediated by inflammation and calcification and inevitably progress over time. Computed tomography can visualise the later stages of macroscopic calcification but fails to assess the early stages of microcalcification and cannot differentiate active from burnt out disease states. Molecular imaging has the ability to provide complementary information related to disease activity, which may allow us to detect disease early, to predict disease progression and to monitor preventive or therapeutic strategies for in both aortic stenosis and atherosclerosis. PET/CT is a non-invasive imaging technique that enables visualization of ongoing molecular processes within small structures, such as the coronary arteries or heart valves. 18F-sodium fluoride (18F-NaF) binds hydroxyapatite deposits in the extracellular matrix, with preferential binding to newly developing deposits of microcalcification, which provides an assessment of calcification activity. In recent years, 18F-NaF has attracted the attention of many research groups and has been evaluated in several pathological cardiovascular processes. Histologic validation of the 18F-NaF PET signal in valvular disease and atherosclerosis has been reported in multiple independent studies. The selective high-affinity binding of 18F-NaF to microscopic calcified deposits (beyond the resolution of μCT) has been demonstrated ex vivo, as well as its ability to distinguish between areas of macro- and active microcalcification. In addition, prospective clinical studies have shown that baseline 18F-NaF uptake in patients with aortic stenosis and mitral annular calcification is correlated with subsequent calcium deposition and valvular dysfunction after a follow-up period of 2 years. In patients with surgical bioprosthetic aortic valves but without morphological criteria for prosthetic degeneration, increased 18F-NaF uptake at baseline was associated with subsequent bioprosthetic degeneration over time. Similar data were obtained in a cohort of patients with transcatheter aortic valve implantation. Furthermore, several studies have confirmed the association of coronary 18F-NaF uptake with adverse atherosclerotic plaque features, active disease and future disease progression. 18F-NaF uptake is also associated with future fatal or nonfatal myocardial infarction in patients with established coronary artery disease. The link between 18F-NaF uptake and active atherosclerotic disease has not only been demonstrated in the coronary arteries, but also in peripheral arterial disease, abdominal aortic aneurysms and carotid atherosclerosis. It can be assumed that 18F-NaF PET/CT will strengthen the diagnostic toolbox of practitioners in the coming years. Indeed, there is a strong medical need to diagnose degenerative valvular disease and to detect active atherosclerotic disease states. Finally, the use of 18F-NaF as a biomarker to monitor the efficacy of drug therapies in preventing these pathological processes is attractive. In this review, we consider the role of 18F-NaF PET/CT imaging in cardiac valvular diseases and atherosclerosis.
Collapse
Affiliation(s)
- David De Azevedo
- Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, and IREC/CARD UCLouvain, Brussels, Belgium.
| | - Jolien Geers
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, The Chancellor's Building, Little France Crescent, Midlothian, Edinburgh, UK; Department of Cardiology, CHVZ (Centrum voor Hart en Vaatziekten), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Olivier Gheysens
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc and Institute of Clinical and Experimental Research (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Marc Dweck
- Department of Cardiology, CHVZ (Centrum voor Hart en Vaatziekten), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - David Vancraeynest
- Department of Cardiovascular Diseases, Cliniques Universitaires St. Luc, and IREC/CARD UCLouvain, Brussels, Belgium
| |
Collapse
|
58
|
Yang Z, Wang J, Yuan Y, Cheng T, Ren F, Wang S, Zhang Z. Application of rivaroxaban in patients with non-valvular atrial fibrillation and end-stage kidney disease: A systematic review and meta-analysis. Front Cardiovasc Med 2023; 10:1021959. [PMID: 36844734 PMCID: PMC9951003 DOI: 10.3389/fcvm.2023.1021959] [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: 08/18/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023] Open
Abstract
Background Nowadays, the number of patients with non-valvular atrial fibrillation (NVAF) complicated by end-stage renal disease (ESKD) is increasing. There are significant challenges in anticoagulation with prescription drugs because of the high risk of bleeding and embolism among these patients. However, no randomized controlled trials (RCTs) of warfarin in combination with any non-vitamin K oral anticoagulant (NOACs) have been performed in patients with baseline creatinine clearance (CrCl) <25 ml/min, which makes it difficult to justify the use of anticoagulants in such patients. Then, we aimed to collect and summarize all evidence to enable the anticoagulation of rivaroxaban, which is less cleared by the kidneys, in patients with severe renal insufficiency and to complement and improve the evidence on the use of rivaroxaban for anticoagulation. Methods The present systematic review and meta-analysis searched the databases of PubMed, Embase, the Cochrane Library, CNKI, CBM, and Google Scholar for relevant studies from inception to 1 June 2022, with the restriction of English and Chinese. Eligible cohort studies and RCTs that reported efficacy outcomes [composite of stroke and systemic embolism (SSE), ischemic stroke (ICS), and systemic embolization] or safety outcomes [major bleeding, intracranial hemorrhage (ICH), and gastrointestinal bleeding (GIB)] of rivaroxaban in NVAF patients with ESKD were enrolled. Two authors completed the data extraction and quality assessment work, respectively. The Cochrane Collaboration tool for assessing the risk of bias was used for RCTs, and the NEW-Castle Ottawa scale was used for study quality assessment for cohort studies. Dichotomous variables were calculated as risk factors with 95% confidence intervals (CIs), and meta-analysis was performed to probe the effect of research design, rivaroxaban dose, and controlled drug factors on outcomes. Results In total, three studies were included for meta-analysis, involving 6,071 NVAF patients with ESKD, and two studies were included for qualitative analysis. All included studies were at low risk of bias. A meta-analysis demonstrated that mix-dose rivaroxaban caused no statistical discrepancy in the occurrence of thrombotic and bleeding events when compared to the control group (embolism, LogOR: -0.64, 95% CI: -1.05 to -0.23, P:0.25; bleeding, LogOR: -0.33, 95% CI: -0.63 to -0.03, P:0.15), and low-dose rivaroxaban produced similar results (embolism, LogOR: -1.04, 95% CI: -2.15 to 0.07, P:0.61; bleeding, LogOR: -0.81, 95% CI: -1.19 to -0.44, P:0.93). Conclusion In this study, low-dose rivaroxaban (10 mg, once a day) may benefit more than warfarin in patients with NVAF and ESKD. Systematic review registration https://www.crd.york.ac.uk/prospero/#recordDetails, identifier CRD42022330973.
Collapse
Affiliation(s)
- Zhenzhen Yang
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jieya Wang
- Department of Pharmacy, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China
| | - Ye Yuan
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tian Cheng
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Feifei Ren
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Songsong Wang
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, China,Department of Pharmacology, Hebei Medical University, Shijiazhuang, China, Songsong Wang, ; orcid.org/0000-0003-0528-7920
| | - Zhiqing Zhang
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, China,*Correspondence: Zhiqing Zhang,
| |
Collapse
|
59
|
Wen D, Hu L, Shan J, Zhang H, Hu L, Yuan A, Pu J, Xue S. Mechanical injury accentuates lipid deposition in ApoE -/- mice and advance aortic valve stenosis: A novel modified aortic valve stenosis model. Front Cardiovasc Med 2023; 10:1119746. [PMID: 36818346 PMCID: PMC9932047 DOI: 10.3389/fcvm.2023.1119746] [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: 12/09/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Background Current mouse models still have limitations in studying aortic valve stenosis (AVS). A suitable animal model bearing a close resemblance to the pathophysiological processes of humans needs to be developed. Here, we combined two risk factors to create a mouse model that mimics the pathological features of human AVS. Methods and results We combined WI and hyperlipidemia in ApoE-/- mice to explore the synergistic effect on the stenosis of the aortic valve. Transthoracic echocardiography revealed progressively increased peak velocity with age in ApoE-/- mice to velocities above C57 mice when fed a high-fat diet after wire injury. Moreover, ApoE-/- mice demonstrated lower cusp separation and lower aortic valve area after 8 weeks vs. C57 mice. Gross morphology and MRI showed advanced thickening, sclerosis aortic valve, narrowing of the orifice area, and micro-CT showed obvious calcification in the aortic valves in the hyperlipidemia group after wire injury. Histopathology studies showed thickening and fibrosis of aortic valve leaflets in the hyperlipidemia group after wire injury. Notably, lipid deposition was observed in ApoE-/- mice 8 weeks after wire injury, accompanied by overexpressed apoB and apoA proteins. After wire injury, the hyperlipidemia group exhibited augmented inflammation, ROS production, and apoptosis in the leaflets. Moreover, the combination group exhibited advanced fibro-calcific aortic valves after wire injury. Conclusion Overall, we present the synergistic effect of wire injury and hyperlipidemia on lipoproteins deposition in the development of AVS in ApoE-/- mice, this model bear close resemblance to human AVS pathology.
Collapse
Affiliation(s)
- Dezhong Wen
- Department of Cardiovascular Surgery, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Hu
- Department of Cardiology, Key Laboratory of Coronary Heart Disease, Shanghai Municipal Education Commission, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianggui Shan
- Department of Cardiovascular Surgery, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hengyuan Zhang
- Department of Cardiology, Key Laboratory of Coronary Heart Disease, Shanghai Municipal Education Commission, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liuhua Hu
- Department of Cardiology, Key Laboratory of Coronary Heart Disease, Shanghai Municipal Education Commission, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ancai Yuan
- Department of Cardiology, Key Laboratory of Coronary Heart Disease, Shanghai Municipal Education Commission, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Key Laboratory of Coronary Heart Disease, Shanghai Municipal Education Commission, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Jun Pu,
| | - Song Xue
- Department of Cardiovascular Surgery, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Song Xue,
| |
Collapse
|
60
|
Kwiecinski J. Novel PET Applications and Radiotracers for Imaging Cardiovascular Pathophysiology. Cardiol Clin 2023; 41:129-139. [PMID: 37003671 DOI: 10.1016/j.ccl.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
PET allows the assessment of cardiovascular pathophysiology across a wide range of cardiovascular conditions. By imaging processes directly involved in disease progression and adverse events, such as inflammation and developing calcifications (microcalcifications), PET can not only enhance our understanding of cardiovascular disease, but also, as shown for 18F-sodium fluoride, has the potential to predict hard endpoints. In this review, the recent advances in disease activity assessment with cardiovascular PET, which provide hope that this promising technology could be leveraged in the clinical setting, shall be discussed.
Collapse
Affiliation(s)
- Jacek Kwiecinski
- Department of Interventional Cardiology and Angiology, KKiAI, Institute of Cardiology, Alpejska 42, Warsaw 04-628, Poland.
| |
Collapse
|
61
|
Affiliation(s)
- Xinjiang Cai
- Department of MedicineUniversity of California, Los Angeles (UCLA)Los AngelesCA
| | - Yin Tintut
- Department of MedicineUniversity of California, Los Angeles (UCLA)Los AngelesCA
- Department of PhysiologyUniversity of CaliforniaLos AngelesCA
- Department of Orthopaedic SurgeryUniversity of CaliforniaLos AngelesCA
- VA Greater Los Angeles Healthcare SystemLos AngelesCA
| | - Linda L. Demer
- Department of MedicineUniversity of California, Los Angeles (UCLA)Los AngelesCA
- Department of BioengineeringUniversity of California, Los Angeles (UCLA)Los AngelesCA
- Department of PhysiologyUniversity of CaliforniaLos AngelesCA
| |
Collapse
|
62
|
Bessueille L, Kawtharany L, Quillard T, Goettsch C, Briolay A, Taraconat N, Balayssac S, Gilard V, Mebarek S, Peyruchaud O, Duboeuf F, Bouillot C, Pinkerton A, Mechtouff L, Buchet R, Hamade E, Zibara K, Fonta C, Canet-Soulas E, Millan JL, Magne D. Inhibition of alkaline phosphatase impairs dyslipidemia and protects mice from atherosclerosis. Transl Res 2023; 251:2-13. [PMID: 35724933 DOI: 10.1016/j.trsl.2022.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 11/16/2022]
Abstract
Calcium accumulation in atherosclerotic plaques predicts cardiovascular mortality, but the mechanisms responsible for plaque calcification and how calcification impacts plaque stability remain debated. Tissue-nonspecific alkaline phosphatase (TNAP) recently emerged as a promising therapeutic target to block cardiovascular calcification. In this study, we sought to investigate the effect of the recently developed TNAP inhibitor SBI-425 on atherosclerosis plaque calcification and progression. TNAP levels were investigated in ApoE-deficient mice fed a high-fat diet from 10 weeks of age and in plaques from the human ECLAGEN biocollection (101 calcified and 14 non-calcified carotid plaques). TNAP was inhibited in mice using SBI-425 administered from 10 to 25 weeks of age, and in human vascular smooth muscle cells (VSMCs) with MLS-0038949. Plaque calcification was imaged in vivo with 18F-NaF-PET/CT, ex vivo with osteosense, and in vitro with alizarin red. Bone architecture was determined with µCT. TNAP activation preceded and predicted calcification in human and mouse plaques, and TNAP inhibition prevented calcification in human VSMCs and in ApoE-deficient mice. More unexpectedly, TNAP inhibition reduced the blood levels of cholesterol and triglycerides, and protected mice from atherosclerosis, without impacting the skeletal architecture. Metabolomics analysis of liver extracts identified phosphocholine as a substrate of liver TNAP, who's decreased dephosphorylation upon TNAP inhibition likely reduced the release of cholesterol and triglycerides into the blood. Systemic inhibition of TNAP protects from atherosclerosis, by ameliorating dyslipidemia, and preventing plaque calcification.
Collapse
Affiliation(s)
- Laurence Bessueille
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | - Lynn Kawtharany
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | - Thibaut Quillard
- CNRS, INSERM, l'institut du thorax, Nantes Université, Nantes, France
| | - Claudia Goettsch
- Department of Internal Medicine I, Cardiology, Medical Faculty, RWTH Aachen University, Aachen Germany
| | - Anne Briolay
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | - Nirina Taraconat
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III, Paul Sabatier, France
| | - Stéphane Balayssac
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III, Paul Sabatier, France
| | - Véronique Gilard
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III, Paul Sabatier, France
| | - Saida Mebarek
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | | | | | | | | | - Laura Mechtouff
- Stroke Department, Hospices Civils de Lyon, France; CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | - René Buchet
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | - Eva Hamade
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
| | - Caroline Fonta
- Brain and Cognition Research Center CerCo, CNRS UMR5549, Université de Toulouse, France
| | - Emmanuelle Canet-Soulas
- CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | | | - David Magne
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France.
| |
Collapse
|
63
|
Yang K, Shang Y, Yang N, Pan S, Jin J, He Q. Application of nanoparticles in the diagnosis and treatment of chronic kidney disease. Front Med (Lausanne) 2023; 10:1132355. [PMID: 37138743 PMCID: PMC10149997 DOI: 10.3389/fmed.2023.1132355] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/22/2023] [Indexed: 05/05/2023] Open
Abstract
With the development of nanotechnology, nanoparticles have been used in various industries. In medicine, nanoparticles have been used in the diagnosis and treatment of diseases. The kidney is an important organ for waste excretion and maintaining the balance of the internal environment; it filters various metabolic wastes. Kidney dysfunction may result in the accumulation of excess water and various toxins in the body without being discharged, leading to complications and life-threatening conditions. Based on their physical and chemical properties, nanoparticles can enter cells and cross biological barriers to reach the kidneys and therefore, can be used in the diagnosis and treatment of chronic kidney disease (CKD). In the first search, we used the English terms "Renal Insufficiency, Chronic" [Mesh] as the subject word and terms such as "Chronic Renal Insufficiencies," "Chronic Renal Insufficiency," "Chronic Kidney Diseases," "Kidney Disease, Chronic," "Renal Disease, Chronic" as free words. In the second search, we used "Nanoparticles" [Mesh] as the subject word and "Nanocrystalline Materials," "Materials, Nanocrystalline," "Nanocrystals," and others as free words. The relevant literature was searched and read. Moreover, we analyzed and summarized the application and mechanism of nanoparticles in the diagnosis of CKD, application of nanoparticles in the diagnosis and treatment of renal fibrosis and vascular calcification (VC), and their clinical application in patients undergoing dialysis. Specifically, we found that nanoparticles can detect CKD in the early stages in a variety of ways, such as via breath sensors that detect gases and biosensors that detect urine and can be used as a contrast agent to avoid kidney damage. In addition, nanoparticles can be used to treat and reverse renal fibrosis, as well as detect and treat VC in patients with early CKD. Simultaneously, nanoparticles can improve safety and convenience for patients undergoing dialysis. Finally, we summarize the current advantages and limitations of nanoparticles applied to CKD as well as their future prospects.
Collapse
Affiliation(s)
- Kaibi Yang
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yiwei Shang
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Nan Yang
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Shujun Pan
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Juan Jin
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
- *Correspondence: Juan Jin,
| | - Qiang He
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
- Qiang He,
| |
Collapse
|
64
|
Hao N, Zhou Z, Zhang F, Li Y, Hu R, Zou J, Zheng R, Wang L, Xu L, Tan W, Li C, Wang F. Interleukin-29 Accelerates Vascular Calcification via JAK2/STAT3/BMP2 Signaling. J Am Heart Assoc 2022; 12:e027222. [PMID: 36537334 PMCID: PMC9973608 DOI: 10.1161/jaha.122.027222] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Vascular calcification (VC), associated with enhanced cardiovascular morbidity and mortality, is characterized by the osteogenic transdifferentiation of vascular smooth muscle cells. Inflammation promotes VC initiation and progression. Interleukin (IL)-29, a newly discovered member of type III interferon, has recently been implicated in the pathogenesis of autoimmune diseases. Here we evaluated the role of IL-29 in the VC process and underlying inflammatory mechanisms. Methods and Results The mRNA expression of IL-29 was significantly increased and positively associated with an increase in BMP2 (bone morphogenetic protein 2) mRNA level in calcified carotid arteries from patients with coronary artery disease or chronic kidney disease. IL-29 and BMP2 proteins are colocalized in human calcified arteries. IL-29 binding to its specific receptor IL-28Rα (IL-28 receptor α) (IL-29/IL-28Rα) inhibited the proliferation of rat vascular smooth muscle cells without altering cell apoptosis or migration. IL-29 promoted the calcification of rat vascular smooth muscle cells and their osteogenic transdifferentiation in vitro as well as the rat aortic ring calcification ex vivo, induced by the calcification medium or osteogenic medium. The procalcification effect of IL-29 was reduced by pharmacological inhibition of IL-29/IL-28Rα binding as well as suppression of janus kinase 2/signal transducer and activator of transcription pathway activation, accompanied by decreased BMP2 expression in the cultured rat vascular smooth muscle cells. Conclusions These results suggest an important role of IL-29 in VC development, at least partly, via activating the janus kinase 2/signal transducer and activator of transcription 3 signaling. Inhibition of IL-29 or its specific receptor, IL-28Rα, may provide a novel strategy to reduce VC in patients with vascular diseases.
Collapse
Affiliation(s)
- Nannan Hao
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Zihao Zhou
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Feifei Zhang
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Yong Li
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Rui Hu
- Department of Vascular SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Junjie Zou
- Department of Vascular SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Rui Zheng
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Lei Wang
- Department of RheumatologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Lingxiao Xu
- Department of RheumatologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Wenfeng Tan
- Department of RheumatologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Chunjian Li
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Fang Wang
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| |
Collapse
|
65
|
Seguchi M, Aytekin A, Lenz T, Nicol P, Klosterman GR, Beele A, Sabic E, Utsch L, Alyaqoob A, Gorpas D, Ntziachristos V, Jaffer FA, Rauschendorfer P, Joner M. Intravascular molecular imaging: translating pathophysiology of atherosclerosis into human disease conditions. Eur Heart J Cardiovasc Imaging 2022; 24:e1-e16. [PMID: 36002376 DOI: 10.1093/ehjci/jeac163] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/31/2022] [Indexed: 12/25/2022] Open
Abstract
Progression of atherosclerotic plaque in coronary arteries is characterized by complex cellular and non-cellular molecular interactions. Within recent years, atherosclerosis has been recognized as inflammation-driven disease condition, where progressive stages are characterized by morphological changes in plaque composition but also relevant molecular processes resulting in increased plaque vulnerability. While existing intravascular imaging modalities are able to resolve key morphological features during plaque progression, they lack capability to characterize the molecular profile of advanced atherosclerotic plaque. Because hybrid imaging modalities may provide incremental information related to plaque biology, they are expected to provide synergistic effects in detecting high risk patients and lesions. The aim of this article is to review existing literature on intravascular molecular imaging approaches, and to provide clinically oriented proposals of their application. In addition, we assembled an overview of future developments in this field geared towards detection of patients at risk for cardiovascular events.
Collapse
Affiliation(s)
- Masaru Seguchi
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich 80636, Germany
| | - Alp Aytekin
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich 80636, Germany
| | - Tobias Lenz
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich 80636, Germany
| | - Philipp Nicol
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich 80636, Germany
| | - Grace R Klosterman
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich 80636, Germany
| | - Alicia Beele
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich 80636, Germany
| | - Emina Sabic
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich 80636, Germany
| | - Léa Utsch
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich 80636, Germany
| | - Aseel Alyaqoob
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich 80636, Germany
| | - Dimitris Gorpas
- Chair of Biological Imaging and TranslaTUM, Technical University of Munich, Munich 80333, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München GmbH, Neuherberg 85764, Germany
| | - Vasilis Ntziachristos
- Chair of Biological Imaging and TranslaTUM, Technical University of Munich, Munich 80333, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München GmbH, Neuherberg 85764, Germany.,Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK) e.V. (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich 80336, Germany
| | - Farouc A Jaffer
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Philipp Rauschendorfer
- Chair of Biological Imaging and TranslaTUM, Technical University of Munich, Munich 80333, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München GmbH, Neuherberg 85764, Germany.,Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK) e.V. (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich 80336, Germany
| | - Michael Joner
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich 80636, Germany.,Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK) e.V. (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich 80336, Germany
| |
Collapse
|
66
|
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
|
67
|
The role of PI3K/Akt signalling pathway in spinal cord injury. Biomed Pharmacother 2022; 156:113881. [DOI: 10.1016/j.biopha.2022.113881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 11/18/2022] Open
|
68
|
Asadi M, Razi F, Fahimfar N, Shirani S, Behzad G, Salari P. The Association of Coronary Artery Calcium Score and Osteoporosis in Postmenopausal Women: A Cross-Sectional Study. J Bone Metab 2022; 29:245-254. [PMID: 36529867 PMCID: PMC9760776 DOI: 10.11005/jbm.2022.29.4.245] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/06/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The association between osteoporosis, a common metabolic bone disorder, and atherosclerosis has been reported in different studies. In this study, we aimed to investigate the association between the coronary artery calcium score (CACS) and bone mineral density (BMD) at different sites and bone biomarkers in postmenopausal women. METHODS A total of 184 participants were enrolled in this study. The CACS and BMD at different sites, including the spinal, total hip, and femoral neck, were measured using computed tomography angiography and dual energy X-ray absorptiometry, respectively. Serum levels of osteocalcin, β-C-terminal telopeptide (β-CTX), parathyroid hormone, and 25-hydroxy-vitamin D were measured. RESULTS A negative association between CACS and bone biomarker levels (osteocalcin, P=0.021; β-CTX, P=0.013) was noted. The univariable model showed an association between CACS and osteoporosis of the femoral neck (P=0.03). It was found that with an increase of 10 U in CACS, the odds of osteoporosis at the femoral neck escalates by 2% (odds ratio=1.02, 95% confidence interval, 1.002-1.03) using the multivariate logistic regression model, while such an association with osteoporosis could not be found at the spinal site. The best cutoff point of the calcium score was estimated to be 127. CONCLUSIONS The results suggest that in postmenopausal women, coronary atherosclerosis is independently associated with osteoporosis of the femoral neck, but such an association could not be detected with spinal osteoporosis. The importance of screening for osteoporosis in patients with cardiovascular disease and the implications of preventive measures in the primary care setting were highlighted considering the common risk factors.
Collapse
Affiliation(s)
- Mojgan Asadi
- Diabetes Research Center, Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran,
Iran
| | - Farideh Razi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran,
Iran
| | - Noushin Fahimfar
- Osteoporosis Research Center, Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran,
Iran
| | - Shapour Shirani
- Head of Imaging Center, Tehran Heart Center, Tehran University of Medical Sciences, Tehran,
Iran
| | - Ghazal Behzad
- School of Medicine, Tehran University of Medical Sciences, Tehran,
Iran
| | - Pooneh Salari
- Medical Ethics and History of Medicine Research Center, Tehran University of Medical Sciences, Tehran,
Iran
| |
Collapse
|
69
|
Gáll T, Nagy P, Garai D, Potor L, Balla GJ, Balla G, Balla J. Overview on hydrogen sulfide-mediated suppression of vascular calcification and hemoglobin/heme-mediated vascular damage in atherosclerosis. Redox Biol 2022; 57:102504. [PMID: 36240620 PMCID: PMC9576974 DOI: 10.1016/j.redox.2022.102504] [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: 08/18/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 12/04/2022] Open
Abstract
Vulnerable atherosclerotic plaques with hemorrhage considerably contribute to cardiovascular morbidity and mortality. Calcification is the main characteristic of advanced atherosclerotic lesions and calcified aortic valve disease (CAVD). Lyses of red blood cells and hemoglobin (Hb) release occur in human hemorrhagic complicated lesions. During the interaction of cell-free Hb with plaque constituents, Hb is oxidized to ferric and ferryl states accompanied by oxidative changes of the globin moieties and heme release. Accumulation of both ferryl-Hb and metHb has been observed in atherosclerotic plaques. The oxidation hotspots in the globin chain are the cysteine and tyrosine amino acids associated with the generation of Hb dimers, tetramers and polymers. Moreover, fragmentation of Hb occurs leading to the formation of globin-derived peptides. A series of these pro-atherogenic cellular responses can be suppressed by hydrogen sulfide (H2S). Since H2S has been explored to exhibit a wide range of physiologic functions to maintain vascular homeostasis, it is not surprising that H2S may play beneficial effects in the progression of atherosclerosis. In the present review, we summarize the findings about the effects of H2S on atherosclerosis and CAVD with a special emphasis on the oxidation of Hb/heme in atherosclerotic plaque development and vascular calcification.
Collapse
Affiliation(s)
- Tamás Gáll
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary; ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology, National Institute of Oncology, Budapest, Hungary; Institute of Oncochemistry, University of Debrecen, Hungary
| | - Dorottya Garai
- Department of Molecular Immunology and Toxicology, National Institute of Oncology, Budapest, Hungary
| | - László Potor
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary; ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | | | - György Balla
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Hungary; ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - József Balla
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary; ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
70
|
Assmann A, Schmidt V, Lepke C, Sugimura Y, Assmann AK, Barth M, Lichtenberg A, Akhyari P. Degeneration of biological heart valve grafts in a rat model of superoxide dismutase-3 deficiency. FASEB J 2022; 36:e22591. [PMID: 36251410 DOI: 10.1096/fj.202200727rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022]
Abstract
While oxidative stress is known as key element in the pathogenesis of atherosclerosis and calcific aortic valve disease, its role in the degeneration of biological cardiovascular grafts has not been clarified yet. Therefore, the present study aimed to examine the impact of oxidative stress on the degeneration of biological cardiovascular allografts in a standardized chronic implantation model realized in rats exhibiting superoxide dismutase 3 deficiency (SOD3(-) ). Rats with SOD3 loss-of-function mutation (n = 24) underwent infrarenal implantation of cryopreserved valved aortic conduits, while SOD3-competent recipients served as controls (n = 28). After a follow-up period of 4 or 12 weeks, comparative analyses addressed degenerative processes, hemodynamics, and evaluation of the oxidative stress model. SOD3(-) rats presented decreased circulating SOD activity (p = .0079). After 12 weeks, 58% of the implant valves in SOD3(-) rats showed regurgitation (vs. 31% in controls, p = .2377). Intima hyperplasia and chondro-osteogenic transformation contributed to progressive graft calcification (p = .0024). At 12 weeks, hydroxyapatite deposition (p = .0198) and the gene expression of runt-related transcription factor-2 (Runx2) (p = .0093) were significantly enhanced in group SOD3(-) . This study provides the first in vivo evidence that impaired systemic antioxidant activity contributes to biological cardiovascular graft degeneration.
Collapse
Affiliation(s)
- Alexander Assmann
- Department of Cardiac Surgery and Research Group for Experimental Surgery, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Vera Schmidt
- Department of Cardiac Surgery and Research Group for Experimental Surgery, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Caroline Lepke
- Department of Cardiac Surgery and Research Group for Experimental Surgery, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Yukiharu Sugimura
- Department of Cardiac Surgery and Research Group for Experimental Surgery, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Anna Kathrin Assmann
- Department of Cardiac Surgery and Research Group for Experimental Surgery, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Mareike Barth
- Department of Cardiac Surgery and Research Group for Experimental Surgery, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Artur Lichtenberg
- Department of Cardiac Surgery and Research Group for Experimental Surgery, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany.,CARID-Cardiovascular Research Institute Düsseldorf, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Payam Akhyari
- Department of Cardiac Surgery and Research Group for Experimental Surgery, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| |
Collapse
|
71
|
Molnár AÁ, Pásztor D, Merkely B. Cellular Senescence, Aging and Non-Aging Processes in Calcified Aortic Valve Stenosis: From Bench-Side to Bedside. Cells 2022; 11:cells11213389. [PMID: 36359785 PMCID: PMC9659237 DOI: 10.3390/cells11213389] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 11/30/2022] Open
Abstract
Aortic valve stenosis (AS) is the most common valvular heart disease. The incidence of AS increases with age, however, a significant proportion of elderly people have no significant AS, indicating that both aging and nonaging pathways are involved in the pathomechanism of AS. Age-related and stress-induced cellular senescence accompanied by further active processes represent the key elements of AS pathomechanism. The early stage of aortic valve degeneration involves dysfunction and disruption of the valvular endothelium due to cellular senescence and mechanical stress on blood flow. These cells are replaced by circulating progenitor cells, but in an age-dependent decelerating manner. When endothelial denudation is no longer replaced by progenitor cells, the path opens for focal lipid deposition, initiating subsequent oxidation, inflammation and micromineralisation. Later stages of AS feature a complex active process with extracellular matrix remodeling, fibrosis and calcification. Echocardiography is the gold standard method for diagnosing aortic valve disease, although computed tomography and cardiac magnetic resonance are useful additional imaging methods. To date, no medical treatment has been proven to halt the progression of AS. Elucidation of differences and similarities between vascular and valvular calcification pathomechanisms may help to find effective medical therapy and reduce the increasing health burden of the disease.
Collapse
|
72
|
Van der Heiden K, Barrett HE, Meester EJ, van Gaalen K, Krenning BJ, Beekman FJ, de Blois E, de Swart J, Verhagen HJM, van der Lugt A, Norenberg JP, de Jong M, Bernsen MR, Gijsen FJH. SPECT/CT imaging of inflammation and calcification in human carotid atherosclerosis to identify the plaque at risk of rupture. J Nucl Cardiol 2022; 29:2487-2496. [PMID: 34318395 PMCID: PMC9553768 DOI: 10.1007/s12350-021-02745-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/28/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Calcification and inflammation are atherosclerotic plaque compositional biomarkers that have both been linked to stroke risk. The aim of this study was to evaluate their co-existing prevalence in human carotid plaques with respect to plaque phenotype to determine the value of hybrid imaging for the detection of these biomarkers. METHODS Human carotid plaque segments, obtained from endarterectomy, were incubated in [111In]In-DOTA-butylamino-NorBIRT ([111In]In-Danbirt), targeting Leukocyte Function-associated Antigen-1 (LFA-1) on leukocytes. By performing SPECT/CT, both inflammation from DANBIRT uptake and calcification from CT imaging were assessed. Plaque phenotype was classified using histology. RESULTS On a total plaque level, comparable levels of calcification volume existed with different degrees of inflammation and vice versa. On a segment level, an inverse relationship between calcification volume and inflammation was evident in highly calcified segments, which classify as fibrocalcific, stable plaque segments. In contrast, segments with little or no calcification presented with a moderate to high degree of inflammation, often coinciding with the more dangerous fibrous cap atheroma phenotype. CONCLUSION Calcification imaging alone can only accurately identify highly calcified, stable, fibrocalcific plaques. To identify high-risk plaques, with little or no calcification, hybrid imaging of calcification and inflammation could provide diagnostic benefit.
Collapse
Affiliation(s)
- K Van der Heiden
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands.
| | - H E Barrett
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - E J Meester
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - K van Gaalen
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | - B J Krenning
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - F J Beekman
- MiLabs, B.V, Utrecht, The Netherlands
- Section Biomedical Imaging, Department Radiation Science & Technology, Delft University of Technology, Delft, The Netherlands
- Department of Translational Neuroscience, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - E de Blois
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - J de Swart
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - H J M Verhagen
- Department of Vascular Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - A van der Lugt
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - J P Norenberg
- Radiopharmaceutical Sciences, University of New Mexico, Albuquerque, NM, USA
| | - M de Jong
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - M R Bernsen
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Applied Molecular Imaging Erasmus Core Facility, Erasmus MC Rotterdam, Rotterdam, The Netherlands
| | - F J H Gijsen
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
73
|
Greco A, Herrmann J, Babic M, Gummi MR, van der Giet M, Tölle M, Schuchardt M. Molecular Imaging and Quantification of Smooth Muscle Cell and Aortic Tissue Calcification In Vitro and Ex Vivo with a Fluorescent Hydroxyapatite-Specific Probe. Biomedicines 2022; 10:biomedicines10092271. [PMID: 36140372 PMCID: PMC9496085 DOI: 10.3390/biomedicines10092271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/22/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Vessel calcification is characterized by the precipitation of hydroxyapatite (HAP) in the vasculature. Currently, no causal therapy exists to reduce or prevent vessel calcification. Studying the underlying pathways within vascular smooth muscle cells and testing pharmacological intervention is a major challenge in the vascular research field. This study aims to establish a rapid and efficient working protocol for specific HAP detection in cells and tissue using the synthetic bisphosphonate fluorescence dye OsteoSense™. This protocol facilitates especially early quantification of the fluorescence signal and permits co-staining with other markers of interest, enabling smaller experimental set-ups with lesser primary cells consumption and fast workflows. The fluorescence-based detection of vascular calcification with OsteoSense™ combines a high specificity with improved sensitivity. Therefore, this methodology can improve research of the pathogenesis of vascular calcification, especially for testing the therapeutic benefit of inhibitors in the case of in vitro and ex vivo settings.
Collapse
|
74
|
Turner ME, Bartoli‐Leonard F, Aikawa E. Small particles with large impact: Insights into the unresolved roles of innate immunity in extracellular vesicle‐mediated cardiovascular calcification. Immunol Rev 2022; 312:20-37. [DOI: 10.1111/imr.13134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Mandy E Turner
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Francesca Bartoli‐Leonard
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Elena Aikawa
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
- Division of Cardiovascular Medicine Department of Medicine Center for Excellence in Vascular Biology Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| |
Collapse
|
75
|
Kitagawa T, Nakano Y. Innovative atherosclerosis imaging using 18F-NaF PET/CT: Its clinical potential. J Nucl Cardiol 2022; 29:1724-1728. [PMID: 33686582 DOI: 10.1007/s12350-021-02576-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Toshiro Kitagawa
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
76
|
Kwiecinski J. 18F-fluorodeoxyglucose and 18F-sodium fluoride for imaging atherosclerotic plaque activity. J Nucl Cardiol 2022; 29:1710-1712. [PMID: 35277833 DOI: 10.1007/s12350-022-02947-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 10/18/2022]
Affiliation(s)
- Jacek Kwiecinski
- Department of Interventional Cardiology and Angiology, Institute of Cardiology, Warsaw, Poland.
| |
Collapse
|
77
|
Atkins SK, Sonawane AR, Brouwhuis R, Barrientos J, Ha A, Rogers M, Tanaka T, Okui T, Kuraoka S, Singh SA, Aikawa M, Aikawa E. Induced pluripotent stem cell-derived smooth muscle cells to study cardiovascular calcification. Front Cardiovasc Med 2022; 9:925777. [PMID: 35958427 PMCID: PMC9357895 DOI: 10.3389/fcvm.2022.925777] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular calcification is the lead predictor of cardiovascular events and the top cause of morbidity and mortality worldwide. To date, only invasive surgical options are available to treat cardiovascular calcification despite the growing understanding of underlying pathological mechanisms. Key players in vascular calcification are vascular smooth muscle cells (SMCs), which transform into calcifying SMCs and secrete mineralizing extracellular vesicles that form microcalcifications, subsequently increasing plaque instability and consequential plaque rupture. There is an increasing, practical need for a large scale and inexhaustible source of functional SMCs. Here we describe an induced pluripotent stem cell (iPSC)-derived model of SMCs by differentiating iPSCs toward SMCs to study the pathogenesis of vascular calcification. Specifically, we characterize the proteome during iPSC differentiation to better understand the cellular dynamics during this process. First, we differentiated human iPSCs toward an induced-SMC (iSMC) phenotype in a 10-day protocol. The success of iSMC differentiation was demonstrated through morphological analysis, immunofluorescent staining, flow cytometry, and proteomics characterization. Proteomics was performed throughout the entire differentiation time course to provide a robust, well-defined starting and ending cell population. Proteomics data verified iPSC differentiation to iSMCs, and functional enrichment of proteins on different days showed the key pathways changing during iSMC development. Proteomics comparison with primary human SMCs showed a high correlation with iSMCs. After iSMC differentiation, we initiated calcification in the iSMCs by culturing the cells in osteogenic media for 17 days. Calcification was verified using Alizarin Red S staining and proteomics data analysis. This study presents an inexhaustible source of functional vascular SMCs and calcifying vascular SMCs to create an in vitro model of vascular calcification in osteogenic conditions, with high potential for future applications in cardiovascular calcification research.
Collapse
Affiliation(s)
- Samantha K. Atkins
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Abhijeet R. Sonawane
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- *Correspondence: Abhijeet R. Sonawane,
| | - Romi Brouwhuis
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Johana Barrientos
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Anna Ha
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Maximillian Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Takeshi Tanaka
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Takehito Okui
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Shiori Kuraoka
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Sasha A. Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Elena Aikawa,
| |
Collapse
|
78
|
Patel NR, Setya K, Pradhan S, Lu M, Demer LL, Tintut Y. Microarchitectural Changes of Cardiovascular Calcification in Response to In Vivo Interventions Using Deep-Learning Segmentation and Computed Tomography Radiomics. Arterioscler Thromb Vasc Biol 2022; 42:e228-e241. [PMID: 35708025 PMCID: PMC9339530 DOI: 10.1161/atvbaha.122.317761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Coronary calcification associates closely with cardiovascular risk, but its progress is accelerated in response to some interventions widely used to reduce risk. This paradox suggests that qualitative, not just quantitative, changes in calcification may affect plaque stability. To determine if the microarchitecture of calcification varies with aging, Western diet, statin therapy, and high intensity, progressive exercise, we assessed changes in a priori selected computed tomography radiomic features (intensity, size, shape, and texture). METHODS Longitudinal computed tomography scans of mice (Apoe-/-) exposed to each of these conditions were autosegmented by deep learning segmentation, and radiomic features of the largest deposits were analyzed. RESULTS Over 20 weeks of aging, intensity and most size parameters increased, but surface-area-to-volume ratio (a measure of porosity) decreased, suggesting stabilization. However, texture features (coarseness, cluster tendency, and nonuniformity) increased, suggesting heterogeneity and likely destabilization. Shape parameters showed no significant changes, except sphericity, which showed a decrease. The Western diet had significant effects on radiomic features related to size and texture, but not intensity or shape. In mice undergoing either pravastatin treatment or exercise, the selected radiomic features of their computed tomography scans were not significantly different from those of their respective controls. Interestingly, the total number of calcific deposits increased significantly less in the 2 intervention groups compared with the respective controls, suggesting more coalescence and/or fewer de novo deposits. CONCLUSIONS Thus, aging and standard interventions alter the microarchitectural features of vascular calcium deposits in ways that may alter plaque biomechanical stability.
Collapse
Affiliation(s)
- Nikhil Rajesh Patel
- Department of Medicine, University of California, Los Angeles. (N.R.P., K.S., S.P., M.L., L.L.D., Y.T.)
| | - Kulveer Setya
- Department of Medicine, University of California, Los Angeles. (N.R.P., K.S., S.P., M.L., L.L.D., Y.T.)
| | - Stuti Pradhan
- Department of Medicine, University of California, Los Angeles. (N.R.P., K.S., S.P., M.L., L.L.D., Y.T.)
| | - Mimi Lu
- Department of Medicine, University of California, Los Angeles. (N.R.P., K.S., S.P., M.L., L.L.D., Y.T.)
| | - Linda L Demer
- Department of Medicine, University of California, Los Angeles. (N.R.P., K.S., S.P., M.L., L.L.D., Y.T.).,Department of Bioengineering, University of California, Los Angeles. (L.L.D.).,Department of Physiology, University of California, Los Angeles. (L.L.D., Y.T.).,VA Greater Los Angeles Healthcare System, CA (L.L.D., Y.T.)
| | - Yin Tintut
- Department of Medicine, University of California, Los Angeles. (N.R.P., K.S., S.P., M.L., L.L.D., Y.T.).,Department of Physiology, University of California, Los Angeles. (L.L.D., Y.T.).,Department of Orthopaedic Surgery, University of California, Los Angeles. (Y.T.).,VA Greater Los Angeles Healthcare System, CA (L.L.D., Y.T.)
| |
Collapse
|
79
|
Ying KE, Feng W, Ying WZ, Li X, Xing D, Sun Y, Chen Y, Sanders PW. Dietary salt initiates redox signaling between endothelium and vascular smooth muscle through NADPH oxidase 4. Redox Biol 2022; 52:102296. [PMID: 35378363 PMCID: PMC8980891 DOI: 10.1016/j.redox.2022.102296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/16/2022] [Indexed: 11/19/2022] Open
Abstract
Prevention of phenotype switching of vascular smooth muscle cells is an important determinant of normal vascular physiology. Hydrogen peroxide (H2O2) promotes osteogenic differentiation of vascular smooth muscle cells through expression of Runt related transcription factor 2 (Runx2). In this study, an increase in dietary NaCl increased endothelial H2O2 generation through NOX4, a NAD(P)H oxidase. The production of H2O2 was sufficient to increase Runx2, osteopontin and osteocalcin in adjacent vascular smooth muscle cells from control littermate mice but was inhibited in mice lacking endothelial Nox4. A vascular smooth muscle cell culture model confirmed the direct involvement of the activation of protein kinase B (Akt) with inactivation of FoxO1 and FoxO3a observed in the control mice on the high NaCl diet. The present study also showed a reduction of catalase activity in aortas during high NaCl intake. The findings demonstrated an interesting cell-cell communication in the vascular wall that was initiated with H2O2 production by endothelium and was regulated by dietary NaCl intake. A better understanding of how dietary salt intake alters vascular biology may improve treatment of vascular disease that involves activation of Runx2.
Collapse
Affiliation(s)
- Kai Er Ying
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA
| | - Wenguang Feng
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA
| | - Wei-Zhong Ying
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA
| | - Xingsheng Li
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA
| | - Dongqi Xing
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA
| | - Yong Sun
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA; Birmingham Department of Veterans Affairs Health Care System, Birmingham, AL, 35233, USA
| | - Paul W Sanders
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA; Birmingham Department of Veterans Affairs Health Care System, Birmingham, AL, 35233, USA.
| |
Collapse
|
80
|
Bakhshian Nik A, Ng HH, Garcia Russo M, Iacoviello F, Shearing PR, Bertazzo S, Hutcheson JD. The Time-Dependent Role of Bisphosphonates on Atherosclerotic Plaque Calcification. J Cardiovasc Dev Dis 2022; 9:jcdd9060168. [PMID: 35735797 PMCID: PMC9225625 DOI: 10.3390/jcdd9060168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerotic plaque calcification directly contributes to the leading cause of morbidity and mortality by affecting plaque vulnerability and rupture risk. Small microcalcifications can increase plaque stress and promote rupture, whereas large calcifications can stabilize plaques. Drugs that target bone mineralization may lead to unintended consequences on ectopic plaque calcification and cardiovascular outcomes. Bisphosphonates, common anti-osteoporotic agents, have elicited unexpected cardiovascular events in clinical trials. Here, we investigated the role of bisphosphonate treatment and timing on the disruption or promotion of vascular calcification and bone minerals in a mouse model of atherosclerosis. We started the bisphosphonate treatment either before plaque formation, at early plaque formation times associated with the onset of calcification, or at late stages of plaque development. Our data indicated that long-term bisphosphonate treatment (beginning prior to plaque development) leads to higher levels of plaque calcification, with a narrower mineral size distribution. When given later in plaque development, we measured a wider distribution of mineral size. These morphological alterations might be associated with a higher risk of plaque rupture by creating stress foci. Yet, bone mineral density positively correlated with the duration of the bisphosphonate treatment.
Collapse
Affiliation(s)
- Amirala Bakhshian Nik
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (A.B.N.); (H.H.N.); (M.G.R.)
| | - Hooi Hooi Ng
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (A.B.N.); (H.H.N.); (M.G.R.)
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Manuel Garcia Russo
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (A.B.N.); (H.H.N.); (M.G.R.)
| | - Francesco Iacoviello
- Department of Chemical Engineering, University College London, London WC1E 7JE, UK; (F.I.); (P.R.S.)
| | - Paul R. Shearing
- Department of Chemical Engineering, University College London, London WC1E 7JE, UK; (F.I.); (P.R.S.)
| | - Sergio Bertazzo
- Department of Medical Physics and Biomedical Engineering, University College London, London WC1E 6BT, UK;
| | - Joshua D. Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (A.B.N.); (H.H.N.); (M.G.R.)
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA
- Correspondence: ; Tel.: +1-305-348-0157
| |
Collapse
|
81
|
Wang X, Chen X, Chen Z, Zhang M. Arterial Calcification and Its Association With Stroke: Implication of Risk, Prognosis, Treatment Response, and Prevention. Front Cell Neurosci 2022; 16:845215. [PMID: 35634461 PMCID: PMC9130460 DOI: 10.3389/fncel.2022.845215] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/28/2022] [Indexed: 01/07/2023] Open
Abstract
Stroke is a leading cause of death worldwide. Vascular calcification (VC), defined as deposition of calcium-phosphate complexes in the vessels, is considered as the characteristic of vascular aging. Calcifications at different vessel layers have different implications. Intimal calcification is closely related to atherosclerosis and affects plaque stability, while medial calcification can cause arterial stiffening and reduce compliance. Accumulating evidence suggested that arterial calcifications, including calcifications in the intracranial artery, coronary artery, and carotid artery, are associated with the risk, prognosis, and treatment response of stroke. VC can not only serve as a marker of atherosclerosis, but cause cerebral hemodynamic impairment. In addition, calcifications in large arteries are associated with cerebral small vessel disease. In this review, we summarize the findings of recently published studies focusing on the relationship between large artery calcification and the risk, prognosis, treatment response, and prevention of stroke and also discuss possible mechanisms behind those associations.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xinghang Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhuohui Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Mengqi Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Mengqi Zhang,
| |
Collapse
|
82
|
Greenberg HZE, Zhao G, Shah AM, Zhang M. Role of oxidative stress in calcific aortic valve disease and its therapeutic implications. Cardiovasc Res 2022; 118:1433-1451. [PMID: 33881501 PMCID: PMC9074995 DOI: 10.1093/cvr/cvab142] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the end result of active cellular processes that lead to the progressive fibrosis and calcification of aortic valve leaflets. In western populations, CAVD is a significant cause of cardiovascular morbidity and mortality, and in the absence of effective drugs, it will likely represent an increasing disease burden as populations age. As there are currently no pharmacological therapies available for preventing, treating, or slowing the development of CAVD, understanding the mechanisms underlying the initiation and progression of the disease is important for identifying novel therapeutic targets. Recent evidence has emerged of an important causative role for reactive oxygen species (ROS)-mediated oxidative stress in the pathophysiology of CAVD, inducing the differentiation of valve interstitial cells into myofibroblasts and then osteoblasts. In this review, we focus on the roles and sources of ROS driving CAVD and consider their potential as novel therapeutic targets for this debilitating condition.
Collapse
Affiliation(s)
- Harry Z E Greenberg
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Guoan Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Henan, China
| | - Ajay M Shah
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Min Zhang
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
83
|
Zeng ZL, Yuan Q, Zu X, Liu J. Insights Into the Role of Mitochondria in Vascular Calcification. Front Cardiovasc Med 2022; 9:879752. [PMID: 35571215 PMCID: PMC9099050 DOI: 10.3389/fcvm.2022.879752] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/14/2022] [Indexed: 12/22/2022] Open
Abstract
Vascular calcification (VC) is a growing burden in aging societies worldwide, and with a significant increase in all-cause mortality and atherosclerotic plaque rupture, it is frequently found in patients with aging, diabetes, atherosclerosis, or chronic kidney disease. However, the mechanism of VC is still not yet fully understood, and there are still no effective therapies for VC. Regarding energy metabolism factories, mitochondria play a crucial role in maintaining vascular physiology. Discoveries in past decades signifying the role of mitochondrial homeostasis in normal physiology and pathological conditions led to tremendous advances in the field of VC. Therapies targeting basic mitochondrial processes, such as energy metabolism, damage in mitochondrial DNA, or free-radical generation, hold great promise. The remarkably unexplored field of the mitochondrial process has the potential to shed light on several VC-related diseases. This review focuses on current knowledge of mitochondrial dysfunction, dynamics anomalies, oxidative stress, and how it may relate to VC onset and progression and discusses the main challenges and prerequisites for their therapeutic applications.
Collapse
Affiliation(s)
- ZL Zeng
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Key Laboratory for Arteriosclerology of Hunan Province, Department of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Qing Yuan
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xuyu Zu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- *Correspondence: Xuyu Zu
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Jianghua Liu
| |
Collapse
|
84
|
Bartlett CL, Cave EM, Crowther NJ, Ferris WF. A new perspective on the function of Tissue Non-Specific Alkaline Phosphatase: from bone mineralization to intra-cellular lipid accumulation. Mol Cell Biochem 2022; 477:2093-2106. [PMID: 35471716 DOI: 10.1007/s11010-022-04429-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/31/2022] [Indexed: 11/29/2022]
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) is one of four isozymes, which include germ cell, placental and intestinal alkaline phosphatases. The TNAP isozyme has 3 isoforms (liver, bone and kidney) which differ by tissue expression and glycosylation pattern. Despite a long history of investigation, the exact function of TNAP in many tissues is largely unknown. Only the bone isoform has been well characterised during mineralization where the enzyme hydrolyses pyrophosphate to inorganic phosphate, which combines with calcium to form hydroxyapatite crystals deposited as new bone. The inorganic phosphate also increases gene expression of proteins that support tissue mineralization. Recent studies have shown that TNAP is expressed in preadipocytes from several species, and that inhibition of TNAP activity causes attenuation of intracellular lipid accumulation in these and other lipid-storing cells. The mechanism by which TNAP stimulates lipid accumulation is not known; however, proteins that are important for controlling phosphate levels in bone are also expressed in adipocytes. This review examines the evidence that inorganic phosphate generated by TNAP promotes transcription that enhances the expression of the regulators of lipid storage and consequently, that TNAP has a major function of lipid metabolism.
Collapse
Affiliation(s)
- Cara-Lesley Bartlett
- Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Eleanor Margaret Cave
- Department of Chemical Pathology, University of the Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa
| | - Nigel John Crowther
- Department of Chemical Pathology, University of the Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa.,Department of Chemical Pathology, National Health Laboratory Service, Johannesburg, South Africa
| | - William Frank Ferris
- Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
85
|
Yaker L, Tebani A, Lesueur C, Dias C, Jung V, Bekri S, Guerrera IC, Kamel S, Ausseil J, Boullier A. Extracellular Vesicles From LPS-Treated Macrophages Aggravate Smooth Muscle Cell Calcification by Propagating Inflammation and Oxidative Stress. Front Cell Dev Biol 2022; 10:823450. [PMID: 35356285 PMCID: PMC8959646 DOI: 10.3389/fcell.2022.823450] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/09/2022] [Indexed: 12/29/2022] Open
Abstract
Background: Vascular calcification (VC) is a cardiovascular complication associated with a high mortality rate among patients with diseases such as atherosclerosis and chronic kidney disease. During VC, vascular smooth muscle cells (VSMCs) undergo an osteogenic switch and secrete a heterogeneous population of extracellular vesicles (EVs). Recent studies have shown involvement of EVs in the inflammation and oxidative stress observed in VC. We aimed to decipher the role and mechanism of action of macrophage-derived EVs in the propagation of inflammation and oxidative stress on VSMCs during VC. Methods: The macrophage murine cell line RAW 264.7 treated with lipopolysaccharide (LPS-EK) was used as a cellular model for inflammatory and oxidative stress. EVs secreted by these macrophages were collected by ultracentrifugation and characterized by transmission electron microscopy, cryo-electron microscopy, nanoparticle tracking analysis, and the analysis of acetylcholinesterase activity, as well as that of CD9 and CD81 protein expression by western blotting. These EVs were added to a murine VSMC cell line (MOVAS-1) under calcifying conditions (4 mM Pi—7 or 14 days) and calcification assessed by the o-cresolphthalein calcium assay. EV protein content was analyzed in a proteomic study and EV cytokine content assessed using an MSD multiplex immunoassay. Results: LPS-EK significantly decreased macrophage EV biogenesis. A 24-h treatment of VSMCs with these EVs induced both inflammatory and oxidative responses. LPS-EK-treated macrophage-derived EVs were enriched for pro-inflammatory cytokines and CAD, PAI-1, and Saa3 proteins, three molecules involved in inflammation, oxidative stress, and VC. Under calcifying conditions, these EVs significantly increase the calcification of VSMCs by increasing osteogenic markers and decreasing contractile marker expression. Conclusion: Our results show that EVs derived from LPS-EK–treated-macrophages are able to induce pro-inflammatory and pro-oxidative responses in surrounding cells, such as VSMCs, thus aggravating the VC process.
Collapse
Affiliation(s)
- Linda Yaker
- MP3CV-UR7517, CURS-University of Picardie Jules Verne, Amiens, France
| | - Abdellah Tebani
- INSERM U1245, CHU Rouen, Normandie University, UNIROUEN, Rouen, France
- Department of Metabolic Biochemistry, Rouen University Hospital, Rouen, France
| | - Céline Lesueur
- INSERM U1245, CHU Rouen, Normandie University, UNIROUEN, Rouen, France
| | - Chloé Dias
- Infinity, INSERM UMR1291, CNRS UMR5051, University of Toulouse III, Toulouse, France
| | - Vincent Jung
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, University of Paris—Federative Research Structure Necker, Paris, France
| | - Soumeya Bekri
- INSERM U1245, CHU Rouen, Normandie University, UNIROUEN, Rouen, France
- Department of Metabolic Biochemistry, Rouen University Hospital, Rouen, France
| | - Ida Chiara Guerrera
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, University of Paris—Federative Research Structure Necker, Paris, France
| | - Saïd Kamel
- MP3CV-UR7517, CURS-University of Picardie Jules Verne, Amiens, France
- Laboratory of Biochemistry, CHU Amiens-Picardie, Amiens, France
| | - Jérôme Ausseil
- Infinity, INSERM UMR1291, CNRS UMR5051, University of Toulouse III, Toulouse, France
- Service de Biochimie, Institut Fédératif de Biologie, CHU Toulouse, Toulouse, France
| | - Agnès Boullier
- MP3CV-UR7517, CURS-University of Picardie Jules Verne, Amiens, France
- Laboratory of Biochemistry, CHU Amiens-Picardie, Amiens, France
- *Correspondence: Agnès Boullier,
| |
Collapse
|
86
|
Zhang M, Xie Z, Long H, Ren K, Hou L, Wang Y, Xu X, Lei W, Yang Z, Ahmed S, Zhang H, Zhao G. Current advances in the imaging of atherosclerotic vulnerable plaque using nanoparticles. Mater Today Bio 2022; 14:100236. [PMID: 35341094 PMCID: PMC8943324 DOI: 10.1016/j.mtbio.2022.100236] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/13/2022] [Accepted: 03/05/2022] [Indexed: 01/29/2023]
Abstract
Vulnerable atherosclerotic plaques of the artery wall that pose a significant risk of cardio-cerebral vascular accidents remain the global leading cause of morbidity and mortality. Thus, early delineation of vulnerable atherosclerotic plaques is of clinical importance for prevention and treatment. The currently available imaging technologies mainly focus on the structural assessment of the vascular wall. Unfortunately, several disadvantages in these strategies limit the improvement in imaging effect. Nanoparticle technology is a novel diagnostic strategy for targeting and imaging pathological biomarkers. New functionalized nanoparticles that detect hallmarks of vulnerable plaques are promising for advance further control of this critical illness. The review aims to address the current opportunities and challenges for the use of nanoparticle technology in imagining vulnerable plaques.
Collapse
|
87
|
Brandt KJ, Burger F, Baptista D, Roth A, Fernandes da Silva R, Montecucco F, Mach F, Miteva K. Single-Cell Analysis Uncovers Osteoblast Factor Growth Differentiation Factor 10 as Mediator of Vascular Smooth Muscle Cell Phenotypic Modulation Associated with Plaque Rupture in Human Carotid Artery Disease. Int J Mol Sci 2022; 23:1796. [PMID: 35163719 PMCID: PMC8836240 DOI: 10.3390/ijms23031796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 12/18/2022] Open
Abstract
(1) Background: Vascular smooth muscle cells (VSMCs) undergo a complex phenotypic switch in response to atherosclerosis environmental triggers, contributing to atherosclerosis disease progression. However, the complex heterogeneity of VSMCs and how VSMC dedifferentiation affects human carotid artery disease (CAD) risk has not been clearly established. (2) Method: A single-cell RNA sequencing analysis of CD45- cells derived from the atherosclerotic aorta of Apolipoprotein E-deficient (Apoe-/-) mice on a normal cholesterol diet (NCD) or a high cholesterol diet (HCD), respecting the site-specific predisposition to atherosclerosis was performed. Growth Differentiation Factor 10 (GDF10) role in VSMCs phenotypic switch was investigated via flow cytometry, immunofluorescence in human atherosclerotic plaques. (3) Results: scRNAseq analysis revealed the transcriptomic profile of seven clusters, five of which showed disease-relevant gene signature of VSMC macrophagic calcific phenotype, VSMC mesenchymal chondrogenic phenotype, VSMC inflammatory and fibro-phenotype and VSMC inflammatory phenotype. Osteoblast factor GDF10 involved in ossification and osteoblast differentiation emerged as a hallmark of VSMCs undergoing phenotypic switch. Under hypercholesteremia, GDF10 triggered VSMC osteogenic switch in vitro. The abundance of GDF10 expressing osteogenic-like VSMCs cells was linked to the occurrence of carotid artery disease (CAD) events. (4) Conclusions: Taken together, these results provide evidence about GDF10-mediated VSMC osteogenic switch, with a likely detrimental role in atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Karim J. Brandt
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Daniela Baptista
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Aline Roth
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Rafaela Fernandes da Silva
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 6627, Brazil
- Swiss Institute for Translational and Entrepreneurial Medicine, Freiburgstrasse 3, 3010 Bern, Switzerland
| | - Fabrizio Montecucco
- Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy;
- First Clinic of Internal Medicine, Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - Francois Mach
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Kapka Miteva
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| |
Collapse
|
88
|
Skenteris NT, Seime T, Witasp A, Karlöf E, Wasilewski GB, Heuschkel MA, Jaminon AM, Oduor L, Dzhanaev R, Kronqvist M, Lengquist M, Peeters FE, Söderberg M, Hultgren R, Roy J, Maegdefessel L, Arnardottir H, Bengtsson E, Goncalves I, Quertermous T, Goettsch C, Stenvinkel P, Schurgers LJ, Matic L. Osteomodulin attenuates smooth muscle cell osteogenic transition in vascular calcification. Clin Transl Med 2022; 12:e682. [PMID: 35184400 PMCID: PMC8858609 DOI: 10.1002/ctm2.682] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 11/28/2021] [Accepted: 12/02/2021] [Indexed: 12/29/2022] Open
Abstract
RATIONALE Vascular calcification is a prominent feature of late-stage diabetes, renal and cardiovascular disease (CVD), and has been linked to adverse events. Recent studies in patients reported that plasma levels of osteomodulin (OMD), a proteoglycan involved in bone mineralisation, associate with diabetes and CVD. We hypothesised that OMD could be implicated in these diseases via vascular calcification as a common underlying factor and aimed to investigate its role in this context. METHODS AND RESULTS In patients with chronic kidney disease, plasma OMD levels correlated with markers of inflammation and bone turnover, with the protein present in calcified arterial media. Plasma OMD also associated with cardiac calcification and the protein was detected in calcified valve leaflets by immunohistochemistry. In patients with carotid atherosclerosis, circulating OMD was increased in association with plaque calcification as assessed by computed tomography. Transcriptomic and proteomic data showed that OMD was upregulated in atherosclerotic compared to control arteries, particularly in calcified plaques, where OMD expression correlated positively with markers of smooth muscle cells (SMCs), osteoblasts and glycoproteins. Immunostaining confirmed that OMD was abundantly present in calcified plaques, localised to extracellular matrix and regions rich in α-SMA+ cells. In vivo, OMD was enriched in SMCs around calcified nodules in aortic media of nephrectomised rats and in plaques from ApoE-/- mice on warfarin. In vitro experiments revealed that OMD mRNA was upregulated in SMCs stimulated with IFNγ, BMP2, TGFβ1, phosphate and β-glycerophosphate, and by administration of recombinant human OMD protein (rhOMD). Mechanistically, addition of rhOMD repressed the calcification process of SMCs treated with phosphate by maintaining their contractile phenotype along with enriched matrix organisation, thereby attenuating SMC osteoblastic transformation. Mechanistically, the role of OMD is exerted likely through its link with SMAD3 and TGFB1 signalling, and interplay with BMP2 in vascular tissues. CONCLUSION We report a consistent association of both circulating and tissue OMD levels with cardiovascular calcification, highlighting the potential of OMD as a clinical biomarker. OMD was localised in medial and intimal α-SMA+ regions of calcified cardiovascular tissues, induced by pro-inflammatory and pro-osteogenic stimuli, while the presence of OMD in extracellular environment attenuated SMC calcification.
Collapse
Affiliation(s)
- Nikolaos T. Skenteris
- Cardiovascular Medicine UnitDepartment of MedicineKarolinska InstituteStockholmSweden
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
- Department of Biochemistry and CARIMSchool for Cardiovascular DiseasesMaastricht UniversityMaastrichtNetherlands
| | - Till Seime
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| | - Anna Witasp
- Division of Renal MedicineDepartment of Clinical SciencesIntervention and TechnologyKarolinska InstituteStockholmSweden
| | - Eva Karlöf
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| | - Grzegorz B. Wasilewski
- Department of Biochemistry and CARIMSchool for Cardiovascular DiseasesMaastricht UniversityMaastrichtNetherlands
- Nattopharma ASA, OsloNorway
| | - Marina A. Heuschkel
- Department of Biochemistry and CARIMSchool for Cardiovascular DiseasesMaastricht UniversityMaastrichtNetherlands
- Department of Internal Medicine I‐CardiologyMedical FacultyRWTH Aachen University, Aachen, Germany
| | - Armand M.G. Jaminon
- Department of Biochemistry and CARIMSchool for Cardiovascular DiseasesMaastricht UniversityMaastrichtNetherlands
| | - Loureen Oduor
- Department of Clinical Sciences Malmö and CardiologySkåne University HospitalLund UniversityLundSweden
| | - Robert Dzhanaev
- Department of Biochemistry and CARIMSchool for Cardiovascular DiseasesMaastricht UniversityMaastrichtNetherlands
- Biointerface GroupHelmholtz Institute for Biomedical EngineeringRWTH Aachen UniversityAachenGermany
| | - Malin Kronqvist
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| | - Mariette Lengquist
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| | - Frederique E.C.M. Peeters
- Department of Cardiology and CARIMSchool for Cardiovascular DiseasesMaastricht University Medical CenterMaastrichtNetherlands
| | - Magnus Söderberg
- CardiovascularRenal and Metabolism SafetyClinical Pharmacology and Safety SciencesR&D, AstraZenecaGothenburgSweden
| | - Rebecka Hultgren
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| | - Joy Roy
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| | - Lars Maegdefessel
- Cardiovascular Medicine UnitDepartment of MedicineKarolinska InstituteStockholmSweden
- Klinikum rechts der IsarDepartment for Vascular and Endovascular SurgeryTechnical University MunichMunichGermany
| | - Hildur Arnardottir
- Cardiovascular Medicine UnitDepartment of MedicineKarolinska InstituteStockholmSweden
| | - Eva Bengtsson
- Department of Clinical Sciences Malmö and CardiologySkåne University HospitalLund UniversityLundSweden
| | - Isabel Goncalves
- Department of Clinical Sciences Malmö and CardiologySkåne University HospitalLund UniversityLundSweden
| | - Thomas Quertermous
- Department of Cardiovascular Medicine, University of StanfordStanfordCaliforniaUSA
| | - Claudia Goettsch
- Department of Internal Medicine I‐CardiologyMedical FacultyRWTH Aachen University, Aachen, Germany
| | - Peter Stenvinkel
- Division of Renal MedicineDepartment of Clinical SciencesIntervention and TechnologyKarolinska InstituteStockholmSweden
| | - Leon J. Schurgers
- Department of Biochemistry and CARIMSchool for Cardiovascular DiseasesMaastricht UniversityMaastrichtNetherlands
- Institute of Experimental Medicine and Systems BiologyRWTH Aachen UniversityAachenGermany
| | - Ljubica Matic
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| |
Collapse
|
89
|
Cote B, Elbarbry F, Bui F, Su JW, Seo K, Nguyen A, Lee M, Rao DA. Mechanistic Basis for the Role of Phytochemicals in Inflammation-Associated Chronic Diseases. Molecules 2022; 27:molecules27030781. [PMID: 35164043 PMCID: PMC8838908 DOI: 10.3390/molecules27030781] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 12/15/2022] Open
Abstract
Chronic inflammatory diseases occur in a large portion of the population and are associated with a poor diet. Key natural products found in fruits and vegetables may assist in lowering inflammation associated with chronic diseases such as obesity, diabetes, cardiovascular diseases, and cancer. This review seeks to examine the roles of several natural products, resveratrol (RES), quercetin (QUE), curcumin (CUR), piperine (PIP), epigallocatechin gallate (EGCG), and gingerol (GIN), in their ability to attenuate inflammatory markers in specific diseases states. Additionally, we will discuss findings in past and ongoing clinical trials, detail possible phytochemical–drug interactions, and provide a brief resource for researchers and healthcare professionals on natural product and supplement regulation as well as names of databases with information on efficacy, indications, and natural product–drug interactions. As diet and over-the-counter supplement use are modifiable factors and patients are interested in using complementary and alternative therapies, understanding the mechanisms by which natural products have demonstrated efficacy and the types of drugs they interact with and knowing where to find information on herbs and supplements is important for practicing healthcare providers and researchers interested in this field.
Collapse
Affiliation(s)
- Brianna Cote
- College of Pharmacy, Oregon State University, Portland, OR 97201, USA;
| | - Fawzy Elbarbry
- School of Pharmacy, Pacific University, Hillsboro, OR 97123, USA; (F.E.); (F.B.); (K.S.); (A.N.); (M.L.)
| | - Fiona Bui
- School of Pharmacy, Pacific University, Hillsboro, OR 97123, USA; (F.E.); (F.B.); (K.S.); (A.N.); (M.L.)
| | - Joe W. Su
- School of Pharmacy, West Coast University, Los Angeles, CA 90004, USA;
| | - Karen Seo
- School of Pharmacy, Pacific University, Hillsboro, OR 97123, USA; (F.E.); (F.B.); (K.S.); (A.N.); (M.L.)
| | - Arthur Nguyen
- School of Pharmacy, Pacific University, Hillsboro, OR 97123, USA; (F.E.); (F.B.); (K.S.); (A.N.); (M.L.)
| | - Max Lee
- School of Pharmacy, Pacific University, Hillsboro, OR 97123, USA; (F.E.); (F.B.); (K.S.); (A.N.); (M.L.)
| | - Deepa A. Rao
- School of Pharmacy, Pacific University, Hillsboro, OR 97123, USA; (F.E.); (F.B.); (K.S.); (A.N.); (M.L.)
- Correspondence:
| |
Collapse
|
90
|
Burger F, Baptista D, Roth A, Brandt KJ, da Silva RF, Montecucco F, Mach F, Miteva K. Single-Cell RNA-Seq Reveals a Crosstalk between Hyaluronan Receptor LYVE-1-Expressing Macrophages and Vascular Smooth Muscle Cells. Cells 2022; 11:411. [PMID: 35159221 PMCID: PMC8834524 DOI: 10.3390/cells11030411] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/11/2022] Open
Abstract
Background: Atherosclerosis is a chronic inflammatory disease where macrophages participate in the progression of the disease. However, the role of resident-like macrophages (res-like) in the atherosclerotic aorta is not completely understood. Methods: A single-cell RNA sequencing analysis of CD45+ leukocytes in the atherosclerotic aorta of apolipoprotein E-deficient (Apoe-/-) mice on a normal cholesterol diet (NCD) or a high cholesterol diet (HCD), respecting the side-to-specific predisposition to atherosclerosis, was performed. A population of res-like macrophages expressing hyaluronan receptor LYVE-1 was investigated via flow cytometry, co-culture experiments, and immunofluorescence in human atherosclerotic plaques from carotid artery disease patients (CAD). Results: We identified 12 principal leukocyte clusters with distinct atherosclerosis disease-relevant gene expression signatures. LYVE-1+ res-like macrophages, expressing a high level of CC motif chemokine ligand 24 (CCL24, eotaxin-2), expanded under hypercholesteremia in Apoe-/- mice and promoted VSMC phenotypic modulation to osteoblast/chondrocyte-like cells, ex vivo, in a CCL24-dependent manner. Moreover, the abundance of LYVE-1+CCL24+ macrophages and elevated systemic levels of CCL24 were associated with vascular calcification and CAD events. Conclusions: LYVE-1 res-like macrophages, via the secretion of CCL24, promote the transdifferentiation of VSMC to osteogenic-like cells with a possible role in vascular calcification and likely a detrimental role in atherosclerotic plaque destabilization.
Collapse
Affiliation(s)
- Fabienne Burger
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1206 Geneva, Switzerland; (F.B.); (D.B.); (A.R.); (K.J.B.); (R.F.d.S.); (F.M.)
| | - Daniela Baptista
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1206 Geneva, Switzerland; (F.B.); (D.B.); (A.R.); (K.J.B.); (R.F.d.S.); (F.M.)
| | - Aline Roth
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1206 Geneva, Switzerland; (F.B.); (D.B.); (A.R.); (K.J.B.); (R.F.d.S.); (F.M.)
| | - Karim J. Brandt
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1206 Geneva, Switzerland; (F.B.); (D.B.); (A.R.); (K.J.B.); (R.F.d.S.); (F.M.)
| | - Rafaela Fernandes da Silva
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1206 Geneva, Switzerland; (F.B.); (D.B.); (A.R.); (K.J.B.); (R.F.d.S.); (F.M.)
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 6627, Brazil
- Swiss Institute for Translational and Entrepreneurial Medicine, Freiburgstrasse 3, 3010 Bern, Switzerland
| | - Fabrizio Montecucco
- Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy;
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - François Mach
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1206 Geneva, Switzerland; (F.B.); (D.B.); (A.R.); (K.J.B.); (R.F.d.S.); (F.M.)
| | - Kapka Miteva
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1206 Geneva, Switzerland; (F.B.); (D.B.); (A.R.); (K.J.B.); (R.F.d.S.); (F.M.)
| |
Collapse
|
91
|
Goettsch C, Strzelecka-Kiliszek A, Bessueille L, Quillard T, Mechtouff L, Pikula S, Canet-Soulas E, Luis MJ, Fonta C, Magne D. TNAP as a therapeutic target for cardiovascular calcification: a discussion of its pleiotropic functions in the body. Cardiovasc Res 2022; 118:84-96. [PMID: 33070177 PMCID: PMC8752354 DOI: 10.1093/cvr/cvaa299] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/11/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular calcification (CVC) is associated with increased morbidity and mortality. It develops in several diseases and locations, such as in the tunica intima in atherosclerosis plaques, in the tunica media in type 2 diabetes and chronic kidney disease, and in aortic valves. In spite of the wide occurrence of CVC and its detrimental effects on cardiovascular diseases (CVD), no treatment is yet available. Most of CVC involve mechanisms similar to those occurring during endochondral and/or intramembranous ossification. Logically, since tissue-nonspecific alkaline phosphatase (TNAP) is the key-enzyme responsible for skeletal/dental mineralization, it is a promising target to limit CVC. Tools have recently been developed to inhibit its activity and preclinical studies conducted in animal models of vascular calcification already provided promising results. Nevertheless, as its name indicates, TNAP is ubiquitous and recent data indicate that it dephosphorylates different substrates in vivo to participate in other important physiological functions besides mineralization. For instance, TNAP is involved in the metabolism of pyridoxal phosphate and the production of neurotransmitters. TNAP has also been described as an anti-inflammatory enzyme able to dephosphorylate adenosine nucleotides and lipopolysaccharide. A better understanding of the full spectrum of TNAP's functions is needed to better characterize the effects of TNAP inhibition in diseases associated with CVC. In this review, after a brief description of the different types of CVC, we describe the newly uncovered additional functions of TNAP and discuss the expected consequences of its systemic inhibition in vivo.
Collapse
Affiliation(s)
- Claudia Goettsch
- Department of Internal Medicine I, Cardiology, Medical Faculty, RWTH Aachen
University, Aachen, Germany
| | - Agnieszka Strzelecka-Kiliszek
- Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental
Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Laurence Bessueille
- Institute of Molecular and Supramolecular Chemistry and Biochemistry
(ICBMS), UMR CNRS 5246, Université Claude Bernard Lyon 1, Bâtiment
Raulin, 43 Bd du 11 novembre 1918, Lyon 69622 Villeurbanne Cedex, France
| | - Thibaut Quillard
- PHY-OS Laboratory, UMR 1238 INSERM, Université de Nantes, CHU
de Nantes, France
| | - Laura Mechtouff
- Stroke Department, Hospices Civils de Lyon, France
- CREATIS Laboratory, CNRS UMR 5220, Inserm U1044, Université Claude Bernard
Lyon 1, Lyon, France
| | - Slawomir Pikula
- Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental
Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Emmanuelle Canet-Soulas
- CarMeN Laboratory, Univ Lyon, INSERM, INRA, INSA Lyon, Université Claude
Bernard Lyon 1, Lyon, France
| | - Millan Jose Luis
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery
Institute, La Jolla, CA 92037, USA
| | - Caroline Fonta
- Brain and Cognition Research Center CerCo, CNRS UMR5549, Université de
Toulouse, France
| | - David Magne
- Institute of Molecular and Supramolecular Chemistry and Biochemistry
(ICBMS), UMR CNRS 5246, Université Claude Bernard Lyon 1, Bâtiment
Raulin, 43 Bd du 11 novembre 1918, Lyon 69622 Villeurbanne Cedex, France
| |
Collapse
|
92
|
Berman DS, Kwiecinski J. Imaging Coronary Inflammatory Risk. JACC Cardiovasc Imaging 2021; 15:472-475. [PMID: 34922869 DOI: 10.1016/j.jcmg.2021.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 11/26/2022]
Affiliation(s)
- Daniel S Berman
- Department of Imaging and Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, California, USA.
| | - Jacek Kwiecinski
- Department of Interventional Cardiology and Angiology, Institute of Cardiology, Warsaw, Poland
| |
Collapse
|
93
|
Wang D, Wang X, Huang L, Pan Z, Liu K, Du B, Xue Y, Li B, Zhang Y, Wang H, Li D, Sun H. Unraveling an Innate Mechanism of Pathological Mineralization-Regulated Inflammation by a Nanobiomimetic System. Adv Healthc Mater 2021; 10:e2101586. [PMID: 34601825 DOI: 10.1002/adhm.202101586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/15/2021] [Indexed: 12/18/2022]
Abstract
Pathological mineralization (PTM) often occurs under inflammation and affects the prognosis of diseases, such as atherosclerosis and cancers. However, how the PTM impacts inflammation has not been well explored. Herein, poly lactic-co-glycolic acid (PLGA)/gelatin/hydroxyapatite (HA) electrospun nanofibers are rationally designed as an ideal PTM microenvironment biomimetic system for unraveling the role of PTM on inflammation. The results demonstrate that the inflammatory response decreases continuously during the process of mineralization. When mature macromineralization forms, the inflammation almost completely disappears. Mechanistically, the PTM formation is mediated by matrix proteins, local high calcium, and cell debris (nuclei), or actively regulated by the lysosomal/plasma membrane components secreted by macrophages. These inflammatory inducible factors (calcium, cell debris, etc.) can be "buried" through PTM process, resulting in reduced immune responses. Overall, the present study demonstrates that PTM is an innate mechanism of inflammation subsidence, providing valuable insight into understanding the action of mineralization on inflammation.
Collapse
Affiliation(s)
- Dongyang Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun, Jilin, 130021, P. R. China
| | - Xiaomeng Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun, Jilin, 130021, P. R. China
| | - Lei Huang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun, Jilin, 130021, P. R. China
| | - Ziyi Pan
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun, Jilin, 130021, P. R. China
| | - Kexuan Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun, Jilin, 130021, P. R. China
| | - Beibei Du
- Department of Cardiology, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Avenue, Changchun, Jilin, 130033, P. R. China
| | - Ying Xue
- HOOKE Instruments Ltd., 77 Yingkou Road, Changchun, Jilin, 130033, P. R. China
| | - Bei Li
- HOOKE Instruments Ltd., 77 Yingkou Road, Changchun, Jilin, 130033, P. R. China
- The State Key Lab of Applied Optics, Changchun Institute of Optics, Fine Mechanics and Physics, CAS, 3888 East Nanhu Road, Changchun, Jilin, 130033, P. R. China
| | - Yuan Zhang
- Department of Anesthesiology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, 130021, P. R. China
| | - Huan Wang
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, Jilin, 130022, P. R. China
| | - Daowei Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun, Jilin, 130021, P. R. China
| | - Hongchen Sun
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, 763 Heguang Road, Changchun, Jilin, 130021, P. R. China
| |
Collapse
|
94
|
Libby P. Inflammation during the life cycle of the atherosclerotic plaque. Cardiovasc Res 2021; 117:2525-2536. [PMID: 34550337 PMCID: PMC8783385 DOI: 10.1093/cvr/cvab303] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation orchestrates each stage of the life cycle of atherosclerotic plaques. Indeed, inflammatory mediators likely link many traditional and emerging risk factors with atherogenesis. Atheroma initiation involves endothelial activation with recruitment of leucocytes to the arterial intima, where they interact with lipoproteins or their derivatives that have accumulated in this layer. The prolonged and usually clinically silent progression of atherosclerosis involves periods of smouldering inflammation, punctuated by episodes of acute activation that may arise from inflammatory mediators released from sites of extravascular injury or infection or from subclinical disruptions of the plaque. Smooth muscle cells and infiltrating leucocytes can proliferate but also undergo various forms of cell death that typically lead to formation of a lipid-rich 'necrotic' core within the evolving intimal lesion. Extracellular matrix synthesized by smooth muscle cells can form a fibrous cap that overlies the lesion's core. Thus, during progression of atheroma, cells not only procreate but perish. Inflammatory mediators participate in both processes. The ultimate clinical complication of atherosclerotic plaques involves disruption that provokes thrombosis, either by fracture of the plaque's fibrous cap or superficial erosion. The consequent clots can cause acute ischaemic syndromes if they embarrass perfusion. Incorporation of the thrombi can promote plaque healing and progressive intimal thickening that can aggravate stenosis and further limit downstream blood flow. Inflammatory mediators regulate many aspects of both plaque disruption and healing process. Thus, inflammatory processes contribute to all phases of the life cycle of atherosclerotic plaques, and represent ripe targets for mitigating the disease.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston, MA, USA
| |
Collapse
|
95
|
Yuan X, Yuan W, Ding L, Shi M, Luo L, Wan Y, Oh J, Zhou Y, Bian L, Deng DYB. Cell-adaptable dynamic hydrogel reinforced with stem cells improves the functional repair of spinal cord injury by alleviating neuroinflammation. Biomaterials 2021; 279:121190. [PMID: 34736145 DOI: 10.1016/j.biomaterials.2021.121190] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022]
Abstract
Spinal cord injury (SCI) is one of the most challenging clinical issues. It is characterized by the disruption of neural circuitry and connectivity, resulting in neurological disability. Adipose-derived stem cells (ADSCs) serve as a promising source of therapeutic cells for SCI treatment. However, the therapeutic outcomes of direct ADSCs transplantation are limited in the presence of an inflammatory microenvironment. Herein, a cell-adaptable neurogenic (CaNeu) hydrogel was developed as a delivery vehicle for ADSCs to promote neuronal regeneration after SCI. The dynamic network of CaNeu hydrogel loaded with ADSCs provides a cell-infiltratable matrix that enhances axonal growth and eventually leads to improved motor evoked potential, hindlimb strength, and coordination of complete spinal cord transection in rats. Furthermore, the CaNeu hydrogel also establishes an anti-inflammatory microenvironment by inducing a shift in the polarization of the recruited macrophages toward the pro-regeneration (M2) phenotype. Our study showed that the CaNeu-hydrogel‒mediated ADSCs delivery resulted in significantly suppressed neuroinflammation and apoptosis, and that this phenomenon involved the PI3K/Akt signaling pathway. Our findings indicate that the CaNeu hydrogel is a valuable delivery vehicle to assist stem cell therapy for SCI, providing a promising strategy for central nervous system diseases.
Collapse
Affiliation(s)
- Xin Yuan
- Department of Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Weihao Yuan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, 999077, Hong Kong, China; Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lu Ding
- Department of Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Ming Shi
- Department of Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Liang Luo
- Department of Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Yong Wan
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiwon Oh
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, 999077, Hong Kong, China
| | - Yanfang Zhou
- Department of Pathophysiology, Guangdong Medical University, Dongguan, 523808, China.
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China.
| | - David Y B Deng
- Department of Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
96
|
Zebhi B, Lazkani M, Bark D. Calcific Aortic Stenosis-A Review on Acquired Mechanisms of the Disease and Treatments. Front Cardiovasc Med 2021; 8:734175. [PMID: 34604358 PMCID: PMC8486019 DOI: 10.3389/fcvm.2021.734175] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/18/2021] [Indexed: 11/13/2022] Open
Abstract
Calcific aortic stenosis is a progressive disease that has become more prevalent in recent decades. Despite advances in research to uncover underlying biomechanisms, and development of new generations of prosthetic valves and replacement techniques, management of calcific aortic stenosis still comes with unresolved complications. In this review, we highlight underlying molecular mechanisms of acquired aortic stenosis calcification in relation to hemodynamics, complications related to the disease, diagnostic methods, and evolving treatment practices for calcific aortic stenosis.
Collapse
Affiliation(s)
- Banafsheh Zebhi
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Mohamad Lazkani
- Medical Center of the Rockies, University of Colorado Health, Loveland, CO, United States
| | - David Bark
- Department of Pediatrics, Washington University in Saint Louis, Saint Louis, MO, United States.,Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, United States
| |
Collapse
|
97
|
Atorvastatin Promotes Macrocalcification, But Not Microcalcification in Atherosclerotic Rabbits: An 18F-NaF PET/CT Study. J Cardiovasc Pharmacol 2021; 78:544-550. [PMID: 34651601 DOI: 10.1097/fjc.0000000000001088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/29/2021] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Our study aimed to investigate the effect of atorvastatin on plaque calcification by matching the results obtained by 18F-sodium fluoride (18F-NaF) positron emission tomography (PET)/computed tomography (CT) with data from histologic sections. METHODS AND RESULTS The rabbits were divided into 2 groups as follows: an atherosclerosis group (n = 10) and an atorvastatin group (n = 10). All rabbits underwent an abdominal aortic operation and were fed a high-fat diet to induce atherosclerosis. Plasma samples were used to analyze serum inflammation markers and blood lipid levels. 18F-NaF PET/CT scans were performed twice. The plaque area, macrophage number and calcification were measured, and the data from the pathological sections were matched with the 18F-NaF PET/CT scan results. The mean standardized uptake value (0.725 ± 0.126 vs. 0.603 ± 0.071, P < 0.001) and maximum standardized uptake value (1.024 ± 0.116 vs. 0.854 ± 0.091, P < 0.001) significantly increased in the atherosclerosis group, but only slightly increased in the atorvastatin group (0.616 ± 0.103 vs. 0.613 ± 0.094, P = 0.384; 0.853 ± 0.099 vs.0.837 ± 0.089, P < 0.001, respectively). The total calcium density was significantly increased in rabbits treated with atorvastatin compared with rabbits not treated with atorvastatin (1.64 ± 0.90 vs. 0.49 ± 0.35, P < 0.001), but the microcalcification level was significantly lower. There were more microcalcification deposits in the areas with increased radioactive uptake of 18F-NaF. CONCLUSIONS Our study suggests that the anti-inflammatory activity of atorvastatin may promote macrocalcification but not microcalcification within atherosclerotic plaques. 18F-NaF PET/CT can detect plaque microcalcifications.
Collapse
|
98
|
Gaudieri V, Zampella E, D'Antonio A, Cuocolo A. 18F-sodium fluoride and vascular calcification: Some like it hot. J Nucl Cardiol 2021; 28:2255-2257. [PMID: 32356181 DOI: 10.1007/s12350-020-02125-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 10/24/2022]
Affiliation(s)
- Valeria Gaudieri
- Department of Advanced Biomedical Sciences, University Federico II, Naples, Italy
| | - Emilia Zampella
- Department of Advanced Biomedical Sciences, University Federico II, Naples, Italy
| | - Adriana D'Antonio
- Department of Advanced Biomedical Sciences, University Federico II, Naples, Italy
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, University Federico II, Naples, Italy.
| |
Collapse
|
99
|
Imaging Inflammation in Patients and Animals: Focus on PET Imaging the Vulnerable Plaque. Cells 2021; 10:cells10102573. [PMID: 34685553 PMCID: PMC8533866 DOI: 10.3390/cells10102573] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/18/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023] Open
Abstract
Acute coronary syndrome (ACS) describes a range of conditions associated with the rupture of high-risk or vulnerable plaque. Vulnerable atherosclerotic plaque is associated with many changes in its microenvironment which could potentially cause rapid plaque progression. Present-day PET imaging presents a plethora of radiopharmaceuticals designed to image different characteristics throughout plaque progression. Improved knowledge of atherosclerotic disease pathways has facilitated a growing number of pathophysiological targets for more innovative radiotracer design aimed at identifying at-risk vulnerable plaque and earlier intervention opportunity. This paper reviews the efficacy of PET imaging radiotracers 18F-FDG, 18F-NaF, 68Ga-DOTATATE, 64Cu-DOTATATE and 68Ga-pentixafor in plaque characterisation and risk assessment, as well as the translational potential of novel radiotracers in animal studies. Finally, we discuss our murine PET imaging experience and the challenges encountered.
Collapse
|
100
|
Waring OJ, Skenteris NT, Biessen EAL, Donners MMPC. Two-faced Janus: The dual role of macrophages in atherosclerotic calcification. Cardiovasc Res 2021; 118:2768-2777. [PMID: 34550346 PMCID: PMC9586561 DOI: 10.1093/cvr/cvab301] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/20/2021] [Indexed: 12/19/2022] Open
Abstract
Calcification is an independent predictor of atherosclerosis-related cardiovascular events. Microcalcification is linked to inflamed, unstable lesions, in comparison to the fibrotic stable plaque phenotype generally associated with advanced calcification. This paradox relates to recognition that calcification presents in a wide spectrum of manifestations that differentially impact plaque’s fate. Macrophages, the main inflammatory cells in atherosclerotic plaque, have a multifaceted role in disease progression. They crucially control the mineralization process, from microcalcification to the osteoid metaplasia of bone-like tissue. It is a bilateral interaction that weighs heavily on the overall plaque fate but remains rather unexplored. This review highlights current knowledge about macrophage phenotypic changes in relation to and interaction with the calcifying environment. On the one hand, macrophage-led inflammation kickstarts microcalcification through a multitude of interlinked mechanisms, which in turn stimulates phenotypic changes in vascular cell types to drive microcalcification. Macrophages may also modulate the expression/activity of calcification inhibitors and inducers, or eliminate hydroxyapatite nucleation points. Contrarily, direct exposure of macrophages to an early calcifying milieu impacts macrophage phenotype, with repercussions for plaque progression and/or stability. Macrophages surrounding macrocalcification deposits show a more reparative phenotype, modulating extracellular matrix, and expressing osteoclast genes. This phenotypic shift favours gradual displacement of the pro-inflammatory hubs; the lipid necrotic core, by macrocalcification. Parallels to bone metabolism may explain many of these changes to macrophage phenotype, with advanced calcification able to show homeostatic osteoid metaplasia. As the targeted treatment of vascular calcification developing in atherosclerosis is thus far severely lacking, it is crucial to better understand its mechanisms of development.
Collapse
Affiliation(s)
- O J Waring
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - N T Skenteris
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands.,Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Solna, Sweden
| | - E A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands.,Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, German
| | - M M P C Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|