1
|
Fujimoto D, Kinoshita D, Suzuki K, Niida T, Yuki H, McNulty I, Lee H, Otake H, Shite J, Ferencik M, Dey D, Kakuta T, Jang IK. Relationship Between Calcified Plaque Burden, Vascular Inflammation, and Plaque Vulnerability in Patients With Coronary Atherosclerosis. JACC Cardiovasc Imaging 2024; 17:1214-1224. [PMID: 39243232 DOI: 10.1016/j.jcmg.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/01/2024] [Accepted: 07/18/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Coronary artery calcification is an integral part of atherosclerosis. It has been suggested that early coronary artery calcification is associated with active inflammation, and advanced calcification forms as inflammation subsides. Inflammation is also an important factor in plaque vulnerability. However, the relationship between coronary artery calcium burden, vascular inflammation, and plaque vulnerability has not been fully investigated. OBJECTIVES This study aimed to correlate calcified plaque burden (CPB) at the culprit lesion with vascular inflammation and plaque vulnerability. METHODS Patients with coronary artery disease who had both computed tomography angiography and optical coherence tomography were included. The authors divided the patients into 4 groups: 1 group without calcification at the culprit lesion; and 3 groups based on the CPB tertiles. CPB was calculated as calcified plaque volume divided by vessel volume in the culprit lesion. The authors compared pericoronary adipose tissue (PCAT) attenuation for vascular inflammation and optical coherence tomography-derived vulnerable features among the 4 groups. RESULTS Among 578 patients, the highest CPB tertile showed significantly lower PCAT attenuation of culprit vessel compared with the other groups. The prevalence of features of plaque vulnerability (including lipid-rich plaque, macrophage, and microvessel) was also lowest in the highest CPB tertile. In the patients with calcification, higher age, statin use, and lower PCAT attenuation were independently associated with CPB. CONCLUSIONS Greater calcium burden is associated with a lower level of vascular inflammation and plaque vulnerability. A greater calcium burden may represent advanced stable plaque without significant inflammatory activity. (Massachusetts General Hospital and Tsuchiura Kyodo General Hospital Coronary Imaging Collaboration; NCT04523194).
Collapse
Affiliation(s)
- Daichi Fujimoto
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daisuke Kinoshita
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Keishi Suzuki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Takayuki Niida
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Haruhito Yuki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Iris McNulty
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hiromasa Otake
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Junya Shite
- Division of Cardiovascular Medicine, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Maros Ferencik
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tsunekazu Kakuta
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan.
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
2
|
Jia K, Luo X, Yi J, Zhang C. Hormonal influence: unraveling the impact of sex hormones on vascular smooth muscle cells. Biol Res 2024; 57:61. [PMID: 39227995 PMCID: PMC11373308 DOI: 10.1186/s40659-024-00542-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
Sex hormones play a pivotal role as endocrine hormones that exert profound effects on the biological characteristics and vascular function of vascular smooth muscle cells (VSMCs). By modulating intracellular signaling pathways, activating nuclear receptors, and regulating gene expression, sex hormones intricately influence the morphology, function, and physiological state of VSMCs, thereby impacting the biological properties of vascular contraction, relaxation, and growth. Increasing evidence suggests that abnormal phenotypic changes in VSMCs contribute to the initiation of vascular diseases, including atherosclerosis. Therefore, understanding the factors governing phenotypic alterations in VSMCs and elucidating the underlying mechanisms can provide crucial insights for refining interventions targeted at vascular diseases. Additionally, the varying levels of different types of sex hormones in the human body, influenced by sex and age, may also affect the phenotypic conversion of VSMCs. This review aims to explore the influence of sex hormones on the phenotypic switching of VSMCs and the development of associated vascular diseases in the human body.
Collapse
Affiliation(s)
- Keran Jia
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xin Luo
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jingyan Yi
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Chunxiang Zhang
- Department of Cardiology, The Affiliated Hospital, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
3
|
Zhang L, Feng Q, Kong W. ECM Microenvironment in Vascular Homeostasis: New Targets for Atherosclerosis. Physiology (Bethesda) 2024; 39:0. [PMID: 38984789 DOI: 10.1152/physiol.00028.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/05/2024] [Accepted: 03/23/2024] [Indexed: 07/11/2024] Open
Abstract
Alterations in vascular extracellular matrix (ECM) components, interactions, and mechanical properties influence both the formation and stability of atherosclerotic plaques. This review discusses the contribution of the ECM microenvironment in vascular homeostasis and remodeling in atherosclerosis, highlighting Cartilage oligomeric matrix protein (COMP) and its degrading enzyme ADAMTS7 as examples, and proposes potential avenues for future research aimed at identifying novel therapeutic targets for atherosclerosis based on the ECM microenvironment.
Collapse
Affiliation(s)
- Lu Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qianqian Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
4
|
Miceli G, Basso MG, Pintus C, Pennacchio AR, Cocciola E, Cuffaro M, Profita M, Rizzo G, Tuttolomondo A. Molecular Pathways of Vulnerable Carotid Plaques at Risk of Ischemic Stroke: A Narrative Review. Int J Mol Sci 2024; 25:4351. [PMID: 38673936 PMCID: PMC11050267 DOI: 10.3390/ijms25084351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/05/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The concept of vulnerable carotid plaques is pivotal in understanding the pathophysiology of ischemic stroke secondary to large-artery atherosclerosis. In macroscopic evaluation, vulnerable plaques are characterized by one or more of the following features: microcalcification; neovascularization; lipid-rich necrotic cores (LRNCs); intraplaque hemorrhage (IPH); thin fibrous caps; plaque surface ulceration; huge dimensions, suggesting stenosis; and plaque rupture. Recognizing these macroscopic characteristics is crucial for estimating the risk of cerebrovascular events, also in the case of non-significant (less than 50%) stenosis. Inflammatory biomarkers, such as cytokines and adhesion molecules, lipid-related markers like oxidized low-density lipoprotein (LDL), and proteolytic enzymes capable of degrading extracellular matrix components are among the key molecules that are scrutinized for their associative roles in plaque instability. Through their quantification and evaluation, these biomarkers reveal intricate molecular cross-talk governing plaque inflammation, rupture potential, and thrombogenicity. The current evidence demonstrates that plaque vulnerability phenotypes are multiple and heterogeneous and are associated with many highly complex molecular pathways that determine the activation of an immune-mediated cascade that culminates in thromboinflammation. This narrative review provides a comprehensive analysis of the current knowledge on molecular biomarkers expressed by symptomatic carotid plaques. It explores the association of these biomarkers with the structural and compositional attributes that characterize vulnerable plaques.
Collapse
Affiliation(s)
- Giuseppe Miceli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Maria Grazia Basso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Chiara Pintus
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Andrea Roberta Pennacchio
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Elena Cocciola
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Mariagiovanna Cuffaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Martina Profita
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Giuliana Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| |
Collapse
|
5
|
Nègre-Salvayre A, Salvayre R. Reactive Carbonyl Species and Protein Lipoxidation in Atherogenesis. Antioxidants (Basel) 2024; 13:232. [PMID: 38397830 PMCID: PMC10886358 DOI: 10.3390/antiox13020232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Atherosclerosis is a multifactorial disease of medium and large arteries, characterized by the presence of lipid-rich plaques lining the intima over time. It is the main cause of cardiovascular diseases and death worldwide. Redox imbalance and lipid peroxidation could play key roles in atherosclerosis by promoting a bundle of responses, including endothelial activation, inflammation, and foam cell formation. The oxidation of polyunsaturated fatty acids generates various lipid oxidation products such as reactive carbonyl species (RCS), including 4-hydroxy alkenals, malondialdehyde, and acrolein. RCS covalently bind to nucleophilic groups of nucleic acids, phospholipids, and proteins, modifying their structure and activity and leading to their progressive dysfunction. Protein lipoxidation is the non-enzymatic post-translational modification of proteins by RCS. Low-density lipoprotein (LDL) oxidation and apolipoprotein B (apoB) modification by RCS play a major role in foam cell formation. Moreover, oxidized LDLs are a source of RCS, which form adducts on a huge number of proteins, depending on oxidative stress intensity, the nature of targets, and the availability of detoxifying systems. Many systems are affected by lipoxidation, including extracellular matrix components, membranes, cytoplasmic and cytoskeletal proteins, transcription factors, and other components. The mechanisms involved in lipoxidation-induced vascular dysfunction are not fully elucidated. In this review, we focus on protein lipoxidation during atherogenesis.
Collapse
Affiliation(s)
- Anne Nègre-Salvayre
- Inserm Unité Mixte de Recherche (UMR), 1297 Toulouse, Centre Hospitalier Universitaire (CHU) Rangueil—BP 84225, 31432 Toulouse CEDEX 4, France;
- Faculty of Medicine, University of Toulouse, 31432 Toulouse, France
| | - Robert Salvayre
- Inserm Unité Mixte de Recherche (UMR), 1297 Toulouse, Centre Hospitalier Universitaire (CHU) Rangueil—BP 84225, 31432 Toulouse CEDEX 4, France;
- Faculty of Medicine, University of Toulouse, 31432 Toulouse, France
| |
Collapse
|
6
|
Mota L, Wang SX, Cronenwett JL, Nolan BW, Malas MB, Schermerhorn ML, Liang P. Association of stroke or death with severity of carotid lesion calcification in patients undergoing carotid artery stenting. J Vasc Surg 2024; 79:305-315.e3. [PMID: 37913944 DOI: 10.1016/j.jvs.2023.10.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023]
Abstract
OBJECTIVE Carotid artery stenting (CAS) for heavily calcified lesions is controversial due to concern for stent failure and increased perioperative stroke risk. However, the degree to which calcification affects outcomes is poorly understood, particularly in transcarotid artery revascularization (TCAR). With the precipitous increase in TCAR use and its expansion to standard surgical-risk patients, we aimed to determine the impact of lesion calcification on CAS outcomes to ensure its safe and appropriate use. METHODS We identified patients in the Vascular Quality Initiative who underwent first-time transfemoral CAS (tfCAS) and TCAR between 2016 and 2021. Patients were stratified into groups based on degree of lesion calcification: no calcification, 1% to 50% calcification, 51% to 99% calcification, and 100% circumferential calcification or intraluminal protrusion. Outcomes included in-hospital and 1-year composite stroke/death, as well as individual stroke, death, and myocardial infarction outcomes. Logistic regression was used to evaluate associations between degree of calcification and these outcomes. RESULTS Among 21,860 patients undergoing CAS, 28% patients had no calcification, 34% had 1% to 50% calcification, 35% had 51% to 99% calcification, and 3% had 100% circumferential calcification/protrusion. Patients with 51% to 99% and circumferential calcification/protrusion had higher odds of in-hospital stroke/death (odds ratio [OR], 1.3; 95% confidence interval [CI], 1.02-1.6; P = .034; OR, 1.9; 95% CI, 1.1-2.9; P = .004, respectively) compared with those with no calcification. Circumferential calcification was also associated with increased risk for in-hospital myocardial infarction (OR, 3.5; 95% CI, 1.5-8.0; P = .003). In tfCAS patients, only circumferential calcification/protrusion was associated with higher in-hospital stroke/death odds (OR, 2.0; 95% CI, 1.2-3.4; P = .013), whereas for TCAR patients, 51% to 99% calcification was associated with increased odds of in-hospital stroke/death (OR, 1.5; 95% CI, 1.1-2.2; P = .025). At 1 year, circumferential calcification/protrusion was associated with higher odds of ipsilateral stroke/death (12.4% vs 6.6%; hazard ratio, 1.64; P = .002). CONCLUSIONS Among patients undergoing CAS, there is an increased risk of in-hospital stroke/death for lesions with >50% calcification or circumferential/protruding plaques. Increasing severity of carotid lesion calcification is a significant risk factor for stroke/death in patients undergoing CAS, regardless of approach.
Collapse
Affiliation(s)
- Lucas Mota
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA
| | - Sophie X Wang
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA
| | - Jack L Cronenwett
- Section of Vascular Surgery, Dartmouth Institute, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Brian W Nolan
- Division of Vascular and Endovascular Surgery, Maine Medical Center, Portland, ME
| | - Mahmoud B Malas
- Department of Vascular and Endovascular Surgery, University of California San Diego, La Jolla, CA
| | - Marc L Schermerhorn
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA
| | - Patric Liang
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA.
| |
Collapse
|
7
|
Hashmi S, Shah PW, Aherrahrou Z, Aikawa E, Aherrahrou R. Beyond the Basics: Unraveling the Complexity of Coronary Artery Calcification. Cells 2023; 12:2822. [PMID: 38132141 PMCID: PMC10742130 DOI: 10.3390/cells12242822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Coronary artery calcification (CAC) is mainly associated with coronary atherosclerosis, which is an indicator of coronary artery disease (CAD). CAC refers to the accumulation of calcium phosphate deposits, classified as micro- or macrocalcifications, that lead to the hardening and narrowing of the coronary arteries. CAC is a strong predictor of future cardiovascular events, such as myocardial infarction and sudden death. Our narrative review focuses on the pathophysiology of CAC, exploring its link to plaque vulnerability, genetic factors, and how race and sex can affect the condition. We also examined the connection between the gut microbiome and CAC, and the impact of genetic variants on the cellular processes involved in vascular calcification and atherogenesis. We aimed to thoroughly analyze the existing literature to improve our understanding of CAC and its potential clinical and therapeutic implications.
Collapse
Affiliation(s)
- Satwat Hashmi
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi 74800, Pakistan;
| | - Pashmina Wiqar Shah
- Institute for Cardiogenetics, Universität zu Lübeck, 23562 Lübeck, Germany; (P.W.S.); (Z.A.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Heart Centre Lübeck, 23562 Lübeck, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, Universität zu Lübeck, 23562 Lübeck, Germany; (P.W.S.); (Z.A.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Heart Centre Lübeck, 23562 Lübeck, Germany
| | - Elena Aikawa
- Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Rédouane Aherrahrou
- Institute for Cardiogenetics, Universität zu Lübeck, 23562 Lübeck, Germany; (P.W.S.); (Z.A.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Heart Centre Lübeck, 23562 Lübeck, Germany
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| |
Collapse
|
8
|
Kang JH, Asai D, Toita R. Bisphenol A (BPA) and Cardiovascular or Cardiometabolic Diseases. J Xenobiot 2023; 13:775-810. [PMID: 38132710 PMCID: PMC10745077 DOI: 10.3390/jox13040049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Bisphenol A (BPA; 4,4'-isopropylidenediphenol) is a well-known endocrine disruptor. Most human exposure to BPA occurs through the consumption of BPA-contaminated foods. Cardiovascular or cardiometabolic diseases such as diabetes, obesity, hypertension, acute kidney disease, chronic kidney disease, and heart failure are the leading causes of death worldwide. Positive associations have been reported between blood or urinary BPA levels and cardiovascular or cardiometabolic diseases. BPA also induces disorders or dysfunctions in the tissues associated with these diseases through various cell signaling pathways. This review highlights the literature elucidating the relationship between BPA and various cardiovascular or cardiometabolic diseases and the potential mechanisms underlying BPA-mediated disorders or dysfunctions in tissues such as blood vessels, skeletal muscle, adipose tissue, liver, pancreas, kidney, and heart that are associated with these diseases.
Collapse
Affiliation(s)
- Jeong-Hun Kang
- National Cerebral and Cardiovascular Center Research Institute, 6-1 Shinmachi, Kishibe, Osaka 564-8565, Japan
| | - Daisuke Asai
- Laboratory of Microbiology, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Tokyo 194-8543, Japan;
| | - Riki Toita
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Osaka 563-8577, Japan;
- AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology (AIST), 2-1 Yamadaoka, Osaka 565-0871, Japan
| |
Collapse
|
9
|
Qin Z, Yu L, Zhang Y, Xu Q, Li C, Zhao S, Xi X, Tian Y, Wang Z, Tian J, Yu B. Coronary artery calcification and plaque stability: an optical coherence tomography study. Heliyon 2023; 9:e23191. [PMID: 38149191 PMCID: PMC10750051 DOI: 10.1016/j.heliyon.2023.e23191] [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: 01/19/2023] [Revised: 10/12/2023] [Accepted: 11/29/2023] [Indexed: 12/28/2023] Open
Abstract
Background Coronary artery calcification (CAC), a surrogate of atherosclerosis, is related to stent underexpansion and adverse cardiac events. However, the effect of CAC on plaque stability is still controversial and the morphological significance of CAC has yet to be elucidated. Methods A retrospective series of 419 patients with acute coronary syndrome (ACS) who underwent optical coherence tomography (OCT) were enrolled. Patients were classified into three groups based on the calcification size in culprit plaques and the features of the culprit and non-culprit plaques among these groups were compared. Logistic regression was used to analyze independent risk factors for culprit plaque rupture and the nonlinear relationship between calcification parameters and culprit plaque rupture. Furthermore, we compared the detailed calcification parameters of different kinds of plaques. Results A total of 419 culprit plaques and 364 non-culprit plaques were identified. The incidence of calcification was 53.9 % in culprit plaques and 50.3 % in non-culprit plaques. Compared with culprit plaques without calcification, plaque rupture, macrophages and cholesterol crystals were more frequently observed in the spotty calcification group, and the lipid length was longer; the incidence of macrophages and cholesterol crystals was higher in the macrocalcification group. Calcification tended to be smaller in ruptured plaques than in non-ruptured plaques. Moreover, the arc and length of calcification were greater in culprit plaques than in non-culprit plaques. Conclusions Vulnerable features were more frequently observed in culprit plaques with spotty calcification, whereas the presence of macrocalcification calcifications did not significantly increase plaque vulnerability. Calcification tends to be larger in culprit plaques than in non-culprit plaques.
Collapse
Affiliation(s)
- Zhifeng Qin
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Li Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Yanwen Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Qinglu Xu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Chao Li
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Suhong Zhao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Xiangwen Xi
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Yanan Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Zhao Wang
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| |
Collapse
|
10
|
Daghem M, Adamson PD, Wang KL, Doris M, Bing R, van Beek EJR, Forsyth L, Williams MC, Tzolos E, Dey D, Slomka PJ, Dweck MR, Newby DE, Moss AJ. Temporal Changes in Coronary 18F-Fluoride Plaque Uptake in Patients with Coronary Atherosclerosis. J Nucl Med 2023; 64:1478-1486. [PMID: 37591540 PMCID: PMC10478818 DOI: 10.2967/jnumed.122.264331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 12/12/2022] [Indexed: 08/19/2023] Open
Abstract
Coronary 18F-sodium fluoride (18F-fluoride) uptake is a marker of both atherosclerotic disease activity and disease progression. It is currently unknown whether there are rapid temporal changes in coronary 18F-fluoride uptake and whether these are more marked in those with clinically unstable coronary artery disease. This study aimed to determine the natural history of coronary 18F-fluoride uptake over 12 mo in patients with either advanced chronic coronary artery disease or a recent myocardial infarction. Methods: Patients with established multivessel coronary artery disease and either chronic disease or a recent acute myocardial infarction underwent coronary 18F-fluoride PET and CT angiography, which was repeated at 3, 6, or 12 mo. Coronary 18F-fluoride uptake was assessed in each vessel by measuring the coronary microcalcification activity (CMA). Coronary calcification was quantified by measuring calcium score, mass, and volume. Results: Fifty-nine patients had chronic coronary artery disease (median age, 68 y; 93% male), and 52 patients had a recent myocardial infarction (median age, 65 y; 83% male). Reflecting the greater burden of coronary artery disease, baseline CMA values were higher in those with chronic coronary artery disease. Coronary 18F-fluoride uptake (CMA > 0) was associated with higher baseline calcium scores (294 Agatston units [AU] [interquartile range, 116-483 AU] vs. 72 AU [interquartile range, 8-222 AU]; P < 0.001) and more rapid progression of coronary calcification scores (39 AU [interquartile range, 10-82 AU] vs. 12 AU [interquartile range, 1-36 AU]; P < 0.001) than was the absence of uptake (CMA = 0). Coronary 18F-fluoride uptake did not markedly alter over the course of 3, 6, or 12 mo in patients with either chronic coronary artery disease or a recent myocardial infarction. Conclusion: Coronary 18F-fluoride uptake is associated with the severity and progression of coronary artery disease but does not undergo a rapid dynamic change in patients with chronic or unstable coronary artery disease. This finding suggests that coronary 18F-fluoride uptake is a temporally stable marker of established and progressive disease.
Collapse
Affiliation(s)
- Marwa Daghem
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom;
| | - Philip D Adamson
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Kang-Ling Wang
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Mhairi Doris
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Rong Bing
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Edwin J R van Beek
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Laura Forsyth
- Edinburgh Clinical Trials Unit, University of Edinburgh, Edinburgh, United Kingdom
| | - Michelle C Williams
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Evangelos Tzolos
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Centre, Los Angeles, California; and
| | - Piotr J Slomka
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Centre, Los Angeles, California; and
| | - Marc R Dweck
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Alastair J Moss
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Department of Cardiovascular Science, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom
| |
Collapse
|
11
|
Yu F, Duan Y, Liu C, Huang H, Xiao X, He Z. Extracellular vesicles in atherosclerosis and vascular calcification: the versatile non-coding RNAs from endothelial cells and vascular smooth muscle cells. Front Med (Lausanne) 2023; 10:1193660. [PMID: 37469665 PMCID: PMC10352799 DOI: 10.3389/fmed.2023.1193660] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/12/2023] [Indexed: 07/21/2023] Open
Abstract
Atherosclerosis (AS) is characterized by the accumulation of lipids, fibrous elements, and calcification in the innermost layers of arteries. Vascular calcification (VC), the deposition of calcium and phosphate within the arterial wall, is an important characteristic of AS natural history. However, medial arterial calcification (MAC) differs from intimal calcification and cannot simply be explained as the consequence of AS. Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) are directly involved in AS and VC processes. Understanding the communication between ECs and VSMCs is critical in revealing mechanisms underlying AS and VC. Extracellular vesicles (EVs) are found as intercellular messengers in kinds of physiological processes and pathological progression. Non-coding RNAs (ncRNAs) encapsulated in EVs are involved in AS and VC, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). The effects of ncRNAs have not been comprehensively understood, especially encapsulated in EVs. Some ncRNAs have demonstrated significant roles in AS and VC, but it remains unclear the functions of the majority ncRNAs detected in EVs. In this review, we summarize ncRNAs encapsulated in EC-EVs and VSMC-EVs, and the signaling pathways that are involved in AS and VC.
Collapse
Affiliation(s)
- Fengyi Yu
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yingjie Duan
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chongmei Liu
- Department of Pathology, Yueyang People's Hospital, Yueyang, Hunan, China
| | - Hong Huang
- Hengyang Medical School, The First Affiliated Hospital, Institute of Clinical Medicine, University of South China, Hengyang, Hunan, China
| | - Xiangcheng Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhangxiu He
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
12
|
Neels JG, Gollentz C, Chinetti G. Macrophage death in atherosclerosis: potential role in calcification. Front Immunol 2023; 14:1215612. [PMID: 37469518 PMCID: PMC10352763 DOI: 10.3389/fimmu.2023.1215612] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/20/2023] [Indexed: 07/21/2023] Open
Abstract
Cell death is an important aspect of atherosclerotic plaque development. Insufficient efferocytosis of death cells by phagocytic macrophages leads to the buildup of a necrotic core that impacts stability of the plaque. Furthermore, in the presence of calcium and phosphate, apoptotic bodies resulting from death cells can act as nucleation sites for the formation of calcium phosphate crystals, mostly in the form of hydroxyapatite, which leads to calcification of the atherosclerotic plaque, further impacting plaque stability. Excessive uptake of cholesterol-loaded oxidized LDL particles by macrophages present in atherosclerotic plaques leads to foam cell formation, which not only reduces their efferocytosis capacity, but also can induce apoptosis in these cells. The resulting apoptotic bodies can contribute to calcification of the atherosclerotic plaque. Moreover, other forms of macrophage cell death, such as pyroptosis, necroptosis, parthanatos, and ferroptosis can also contribute by similar mechanisms to plaque calcification. This review focuses on macrophage death in atherosclerosis, and its potential role in calcification. Reducing macrophage cell death and/or increasing their efferocytosis capacity could be a novel therapeutic strategy to reduce the formation of a necrotic core and calcification and thereby improving atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Jaap G. Neels
- Université Côte d’Azur, Institut national de la santé et de la recherche médicale (INSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Claire Gollentz
- Université Côte d’Azur, Centre Hospitalier Universitaire (CHU), Institut national de la santé et de la recherche médicale (NSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Giulia Chinetti
- Université Côte d’Azur, Centre Hospitalier Universitaire (CHU), Institut national de la santé et de la recherche médicale (NSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| |
Collapse
|
13
|
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: 3] [Impact Index Per Article: 3.0] [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
|
14
|
Azeez M, Laivuori M, Tolva J, Linder N, Lundin J, Albäck A, Venermo M, Mäyränpää MI, Lokki ML, Lokki AI, Sinisalo J. High relative amount of nodular calcification in femoral plaques is associated with milder lower extremity arterial disease. BMC Cardiovasc Disord 2022; 22:563. [PMID: 36564714 PMCID: PMC9783794 DOI: 10.1186/s12872-022-02945-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/09/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Clinical implications of different types of vascular calcification are poorly understood. The two most abundant forms of calcification, nodular and sheet calcification, have not been quantitatively analyzed in relation to the clinical presentation of lower extremity arterial disease (LEAD). METHODS The study analyzed 51 femoral artery plaques collected during femoral endarterectomy, characterized by the presence of > 90% stenosis. Comprehensive clinical data was obtained from patient records, including magnetic resonance angiography (MRA) images, toe pressure and ankle brachial index measurements and laboratory values. The plaques were longitudinally sectioned, stained with Hematoxylin and Eosin and digitized in a deep learning platform for quantification of the relative area of nodular and sheet calcification to the plaque section area. A deep learning artificial intelligence algorithm was designed and independently validated to reliably quantify nodular calcification and sheet calcification. Vessel measurements and quantity of each calcification category was compared to the risk factors and clinical presentation. RESULTS On average, > 90% stenosed vessels contained 22.4 ± 12.3% of nodular and 14.5 ± 11.8% of sheet calcification. Nodular calcification area proportion in lesions with > 90% stenosis is associated with reduced risk of critically low toe pressure (< 30 mmHg) (OR = 0.910, 95% CI = 0.835-0.992, p < 0.05), severely lowered ankle brachial index (< 0.4) (OR = 0.912, 95% CI = 0.84-0.986, p < 0.05), and semi-urgent operation (OR = 0.882, 95% CI = 0.797-0.976, p < 0.05). Sheet calcification did not show any significant association. CONCLUSIONS Large amount of nodular calcification is associated with less severe LEAD. Patients with nodular calcification may have better flow reserves despite local obstruction.
Collapse
Affiliation(s)
- Mae Azeez
- grid.7737.40000 0004 0410 2071Transplantation Laboratory, Department of Pathology, University of Helsinki, Haartmaninkatu 3, FIN-00014 Helsinki, Finland
| | - Mirjami Laivuori
- grid.15485.3d0000 0000 9950 5666Department of Vascular Surgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, PL 340, FIN-00029 HUS, Helsinki, Finland
| | - Johanna Tolva
- grid.7737.40000 0004 0410 2071Transplantation Laboratory, Department of Pathology, University of Helsinki, Haartmaninkatu 3, FIN-00014 Helsinki, Finland
| | - Nina Linder
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland, HILIFE, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Johan Lundin
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland, HILIFE, University of Helsinki, FIN-00014 Helsinki, Finland ,grid.4714.60000 0004 1937 0626Present Address: Department of Public Health Sciences, Global Health/IHCAR, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Anders Albäck
- grid.15485.3d0000 0000 9950 5666Department of Vascular Surgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, PL 340, FIN-00029 HUS, Helsinki, Finland
| | - Maarit Venermo
- grid.15485.3d0000 0000 9950 5666Department of Vascular Surgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, PL 340, FIN-00029 HUS, Helsinki, Finland
| | - Mikko I. Mäyränpää
- grid.7737.40000 0004 0410 2071Department of Pathology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 3, FIN-00014 Helsinki, Finland
| | - Marja-Liisa Lokki
- grid.7737.40000 0004 0410 2071Transplantation Laboratory, Department of Pathology, University of Helsinki, Haartmaninkatu 3, FIN-00014 Helsinki, Finland
| | - A. Inkeri Lokki
- grid.7737.40000 0004 0410 2071Transplantation Laboratory, Department of Pathology, University of Helsinki, Haartmaninkatu 3, FIN-00014 Helsinki, Finland ,grid.7737.40000 0004 0410 2071Translational Immunology Research Program, Research Programs Unit, University of Helsinki, Haartmaninkatu 8, FIN-00014 Helsinki, Finland ,grid.15485.3d0000 0000 9950 5666Department of Cardiology, Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, PL 340, FIN-00029 HUS Helsinki, Finland
| | - Juha Sinisalo
- grid.15485.3d0000 0000 9950 5666Department of Cardiology, Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, PL 340, FIN-00029 HUS Helsinki, Finland
| |
Collapse
|
15
|
Zhao Y, Malik S, Criqui MH, Allison MA, Budoff MJ, Sandfort V, Wong ND. Coronary calcium density in relation to coronary heart disease and cardiovascular disease in adults with diabetes or metabolic syndrome: the Multi-ethnic Study of Atherosclerosis (MESA). BMC Cardiovasc Disord 2022; 22:536. [PMID: 36494811 PMCID: PMC9733236 DOI: 10.1186/s12872-022-02956-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 11/14/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Coronary artery calcium (CAC) density is inversely associated with coronary heart disease (CHD) and cardiovascular disease (CVD) risk. We examined this relation in those with diabetes mellitus (DM) or metabolic syndrome (MetS). METHODS We studied 3,818 participants with non-zero CAC scores from the Multiethnic Study of Atherosclerosis and classified them as DM, MetS (without DM) or neither DM/MetS. Risk factor-adjusted CAC density was calculated and examined in relation to incident CHD and CVD events over a median follow-up of 15 years among these three disease groups. RESULTS Adjusted CAC density was 2.54, 2.61 and 2.69 among those with DM, MetS or neither DM/MetS. Hazard ratios (HRs) for CHD per 1 SD increase of CAC density was 0.91 (95% CI: 0.72-1.16), 0.70 (95% CI: 0.56-0.87) and 0.79 (95% CI: 0.66-0.95) for those with DM, MetS or neither DM/MetS groups and were 0.77 (95% CI: 0.64-0.94), 0.83 (95% CI: 0.70-0.99) and 0.82 (95% CI: 0.71-0.95) for CVD, respectively. Adjustment for CAC density increased the HRs of CAC volume for CHD/CVD events. Compared to prediction models with or without single CAC measures, c-statistics of models with CAC volume and density were the highest ranging 0.67-0.72. CONCLUSION CAC density is lower among patients with DM or MetS than those with neither DM/MetS and is inversely associated with future CHD/CVD risk among them. Including CAC density in risk assessment among those with MetS may improve prediction of CHD and CVD.
Collapse
Affiliation(s)
- Yanglu Zhao
- grid.19006.3e0000 0000 9632 6718Department of Epidemiology, University of California Los Angeles, Los Angeles, USA ,grid.266093.80000 0001 0668 7243Heart Disease Prevention Program, University of California Irvine, Irvine, USA
| | - Shaista Malik
- grid.266093.80000 0001 0668 7243Heart Disease Prevention Program, University of California Irvine, Irvine, USA
| | - Michael H. Criqui
- grid.266100.30000 0001 2107 4242Division of Preventive Medicine, University of California San Diego, San Diego, USA
| | - Matthew A. Allison
- grid.266100.30000 0001 2107 4242Division of Preventive Medicine, University of California San Diego, San Diego, USA
| | - Matthew J. Budoff
- grid.239844.00000 0001 0157 6501Lindquist Institute, Harbor-UCLA Medical Center, Los Angeles, USA
| | - Veit Sandfort
- grid.94365.3d0000 0001 2297 5165Radiology & Imaging Sciences, National Institutes of Health, Bethesda, USA
| | - Nathan D. Wong
- grid.19006.3e0000 0000 9632 6718Department of Epidemiology, University of California Los Angeles, Los Angeles, USA ,grid.266093.80000 0001 0668 7243Heart Disease Prevention Program, University of California Irvine, Irvine, USA
| |
Collapse
|
16
|
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: 1.0] [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
|
17
|
Fote GM, Raefsky S, Mock K, Chaudhari A, Shafie M, Yu W. Intracranial Arterial Calcifications: Potential Biomarkers of Stroke Risk and Outcome. Front Neurol 2022; 13:900579. [PMID: 36119671 PMCID: PMC9475140 DOI: 10.3389/fneur.2022.900579] [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: 03/20/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022] Open
Abstract
Intracranial artery calcifications (IAC), a common and easily identifiable finding on computed tomorgraphy angiography (CTA), has gained recognition as a possible risk factor for ischemic stroke. While atherosclerosis of intracranial arteries is believed to be a mechanism that commonly contributes to ischemic stroke, and coronary artery calcification is well-established as a predictor of both myocardial infarction (MI) and ischemic stroke risk, IAC is not currently used as a prognostic tool for stroke risk or recurrence. This review examines the pathophysiology and prevalence of IAC, and current evidence suggesting that IAC may be a useful tool for prediction of stroke incidence, recurrence, and response to acute ischemic stroke therapy.
Collapse
Affiliation(s)
- Gianna M. Fote
- School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Sophia Raefsky
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Kelton Mock
- School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Amit Chaudhari
- Department of Neurology, University of California, Irvine, Irvine, CA, United States
- *Correspondence: Amit Chaudhari
| | - Mohammad Shafie
- Department of Neurology, University of California, Irvine, Irvine, CA, United States
| | - Wengui Yu
- Department of Neurology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
18
|
Aftermath of AGE-RAGE Cascade in the pathophysiology of cardiovascular ailments. Life Sci 2022; 307:120860. [DOI: 10.1016/j.lfs.2022.120860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/21/2022]
|
19
|
Tian L, Wang Y, Zhang R. Galectin-3 induces vascular smooth muscle cells calcification via AMPK/TXNIP pathway. Aging (Albany NY) 2022; 14:5086-5096. [PMID: 35771146 PMCID: PMC9271303 DOI: 10.18632/aging.204130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 05/23/2022] [Indexed: 12/15/2022]
Abstract
Galectin-3 plays an important role in atherosclerosis. Upregulation of VSMCs calcification is involved in the progression and development of vulnerable plaques. Thioredoxin-interacting protein (TXNIP) has been regarded as an important determinant in regulating inflammation and oxidative stress. In this study, we evaluated the role of TXNIP in galectin-3-induced vascular calcification. A primary culture of mouse VSMCs was established by enzymatic digestion of aorta. Small interfering (si) RNA was used to knock down the expression of target gene. VSMCs were treated with 3-methyladenine (3-MA) or compound C respectively. Western blot was performed to detect the protein level in VSMCs, Alkaline phosphatase (ALP) and Alizarin red staining was used to observe calcium deposition. Dihydroethidium (DHE) staining was used to observe the reactive oxygen species (ROS) production. Here we showed that galectin-3 increased aorta and VSMCs calcification, which was associated with AMPK/TXNIP upregulation and autophagy activation. TXNIP inhibition decreased galectin-3-induced aorta and VSMCs calcification and autophagy activation. 3-MA or Atg5 siRNA decreased galectin-3-induced upregulation of Runx2, BMP2 and OPN. AMPK mediated galectin-3-induced VSMCs osteogenic differentiation. These findings illustrated that TXNIP mediated galectin-3-induced vascular calcification, AMPK and autophagy activation were also associated with this process.
Collapse
Affiliation(s)
- Lei Tian
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Bayarsaikhan G, Bayarsaikhan D, Lee J, Lee B. Targeting Scavenger Receptors in Inflammatory Disorders and Oxidative Stress. Antioxidants (Basel) 2022; 11:936. [PMID: 35624800 PMCID: PMC9137717 DOI: 10.3390/antiox11050936] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/04/2022] [Accepted: 05/07/2022] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress and inflammation cannot be considered as diseases themselves; however, they are major risk factors for the development and progression of the pathogenesis underlying many illnesses, such as cancer, neurological disorders (including Alzheimer's disease and Parkinson's disease), autoimmune and metabolic disorders, etc. According to the results obtained from extensive studies, oxidative stress-induced biomolecules, such as advanced oxidation protein products, advanced glycation end products, and advanced lipoxidation end products, are critical for an accelerated level of inflammation and oxidative stress-induced cellular damage, as reflected in their strong affinity to a wide range of scavenger receptors. Based on the limitations of antioxidative and anti-inflammatory molecules in practical applications, targeting such interactions between harmful molecules and their cellular receptors/signaling with advances in gene engineering technology, such as CRISPR or TALEN, may prove to be a safe and effective alternative. In this review, we summarize the findings of recent studies focused on the deletion of scavenger receptors under oxidative stress as a development in the therapeutic approaches against the diseases linked to inflammation and the contribution of advanced glycation end products (AGEs), advanced lipid peroxidation products (ALEs), and advanced oxidation protein products (AOPPs).
Collapse
Affiliation(s)
- Govigerel Bayarsaikhan
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Korea; (G.B.); (D.B.); (J.L.)
| | - Delger Bayarsaikhan
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Korea; (G.B.); (D.B.); (J.L.)
| | - Jaewon Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Korea; (G.B.); (D.B.); (J.L.)
| | - Bonghee Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Korea; (G.B.); (D.B.); (J.L.)
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Gachon University, Incheon 405-760, Korea
| |
Collapse
|
21
|
Cansby E, Kumari S, Caputo M, Xia Y, Porosk R, Robinson J, Wang H, Olsson BM, Vallin J, Grantham J, Soomets U, Svensson LT, Sihlbom C, Marschall HU, Edsfeldt A, Goncalves I, Mahlapuu M. Silencing of STE20-type kinase STK25 in human aortic endothelial and smooth muscle cells is atheroprotective. Commun Biol 2022; 5:379. [PMID: 35440683 PMCID: PMC9018782 DOI: 10.1038/s42003-022-03309-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 03/22/2022] [Indexed: 02/07/2023] Open
Abstract
Recent studies highlight the importance of lipotoxic damage in aortic cells as the major pathogenetic contributor to atherosclerotic disease. Since the STE20-type kinase STK25 has been shown to exacerbate ectopic lipid storage and associated cell injury in several metabolic organs, we here investigate its role in the main cell types of vasculature. We depleted STK25 by small interfering RNA in human aortic endothelial and smooth muscle cells exposed to oleic acid and oxidized LDL. In both cell types, the silencing of STK25 reduces lipid accumulation and suppresses activation of inflammatory and fibrotic pathways as well as lowering oxidative and endoplasmic reticulum stress. Notably, in smooth muscle cells, STK25 inactivation hinders the shift from a contractile to a synthetic phenotype. Together, we provide several lines of evidence that antagonizing STK25 signaling in human aortic endothelial and smooth muscle cells is atheroprotective, highlighting this kinase as a new potential therapeutic target for atherosclerotic disease. Silencing of STK25, an STE20-type kinase, in human aortic endothelial and smooth muscle cells reduces lipid accumulation and suppresses inflammation and fibrotic pathways, ultimately exerting atheroprotective effects.
Collapse
Affiliation(s)
- Emmelie Cansby
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sima Kumari
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mara Caputo
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ying Xia
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Rando Porosk
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Jonathan Robinson
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Gothenburg, Sweden
| | - Hao Wang
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Gothenburg, Sweden
| | | | - Josefine Vallin
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Julie Grantham
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ursel Soomets
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - L Thomas Svensson
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Gothenburg, Sweden
| | - Carina Sihlbom
- Proteomics Core Facility, University of Gothenburg, Gothenburg, Sweden
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Andreas Edsfeldt
- Department of Clinical Sciences Malmö, Clinical Research Center, Lund University, Malmö, Sweden.,Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Malmö, Sweden
| | - Isabel Goncalves
- Department of Clinical Sciences Malmö, Clinical Research Center, Lund University, Malmö, Sweden.,Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden
| | - Margit Mahlapuu
- Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
22
|
Dai X, Liu S, Cheng L, Huang T, Guo H, Wang D, Xia M, Ling W, Xiao Y. Epigenetic Upregulation of H19 and AMPK Inhibition Concurrently Contribute to S-Adenosylhomocysteine Hydrolase Deficiency-Promoted Atherosclerotic Calcification. Circ Res 2022; 130:1565-1582. [PMID: 35410483 DOI: 10.1161/circresaha.121.320251] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND S-adenosylhomocysteine (SAH) is a risk factor of cardiovascular disease; inhibition of SAH hydrolase (SAHH) results in SAH accumulation and induces endothelial dysfunction and atherosclerosis. However, the effect and mechanism of SAHH in atherosclerotic calcification is still unclear. We aimed to explore the role and mechanism of SAHH in atherosclerotic calcification. METHODS The relationship between SAHH and atherosclerotic calcification was investigated in patients with coronary atherosclerotic calcification. Different in vivo genetic models were used to examine the effect of SAHH deficiency on atherosclerotic calcification. Human aortic and murine vascular smooth muscle cells (VSMCs) were cultured to explore the underlying mechanism of SAHH on osteoblastic differentiation of VSMCs. RESULTS The expression and activity of SAHH were decreased in calcified human coronary arteries and inversely associated with coronary atherosclerotic calcification severity, whereas plasma SAH and total homocysteine levels were positively associated with coronary atherosclerotic calcification severity. Heterozygote knockout of SAHH promoted atherosclerotic calcification. Specifically, VSMC-deficient but not endothelial cell-deficient or macrophage-deficient SAHH promoted atherosclerotic calcification. Mechanistically, SAHH deficiency accumulated SAH levels and induced H19-mediated Runx2 (runt-related transcription factor 2)-dependent osteoblastic differentiation of VSMCs by inhibiting DNMT3b (DNA methyltransferase 3 beta) and leading to hypomethylation of the H19 promoter. On the other hand, SAHH deficiency resulted in lower intracellular levels of adenosine and reduced AMPK (AMP-activated protein kinase) activation. Adenosine supplementation activated AMPK and abolished SAHH deficiency-induced expression of H19 and Runx2 and osteoblastic differentiation of VSMCs. Finally, AMPK activation by adenosine inhibited H19 expression by inducing Sirt1-mediated histone H3 hypoacetylation and DNMT3b-mediated hypermethylation of the H19 promoter in SAHH deficiency VSMCs. CONCLUSIONS We have confirmed a novel correlation between SAHH deficiency and atherosclerotic calcification and clarified a new mechanism that epigenetic upregulation of H19 and AMPK inhibition concurrently contribute to SAHH deficiency-promoted Runx2-dependent atherosclerotic calcification.
Collapse
Affiliation(s)
- Xin Dai
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China (X.D., S.L., L.C., T.H., Y.X.)
| | - Si Liu
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China (X.D., S.L., L.C., T.H., Y.X.)
| | - Lokyu Cheng
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China (X.D., S.L., L.C., T.H., Y.X.)
| | - Ting Huang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China (X.D., S.L., L.C., T.H., Y.X.)
| | - Honghui Guo
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, China (H.G.)
| | - Dongliang Wang
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China (D.W., M.X., W.L.)
| | - Min Xia
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China (D.W., M.X., W.L.)
| | - Wenhua Ling
- Department of Nutrition, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China (D.W., M.X., W.L.)
| | - Yunjun Xiao
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China (X.D., S.L., L.C., T.H., Y.X.)
| |
Collapse
|
23
|
Jebari-Benslaiman S, Galicia-García U, Larrea-Sebal A, Olaetxea JR, Alloza I, Vandenbroeck K, Benito-Vicente A, Martín C. Pathophysiology of Atherosclerosis. Int J Mol Sci 2022; 23:ijms23063346. [PMID: 35328769 PMCID: PMC8954705 DOI: 10.3390/ijms23063346] [Citation(s) in RCA: 260] [Impact Index Per Article: 130.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/12/2022] [Accepted: 03/18/2022] [Indexed: 11/26/2022] Open
Abstract
Atherosclerosis is the main risk factor for cardiovascular disease (CVD), which is the leading cause of mortality worldwide. Atherosclerosis is initiated by endothelium activation and, followed by a cascade of events (accumulation of lipids, fibrous elements, and calcification), triggers the vessel narrowing and activation of inflammatory pathways. The resultant atheroma plaque, along with these processes, results in cardiovascular complications. This review focuses on the different stages of atherosclerosis development, ranging from endothelial dysfunction to plaque rupture. In addition, the post-transcriptional regulation and modulation of atheroma plaque by microRNAs and lncRNAs, the role of microbiota, and the importance of sex as a crucial risk factor in atherosclerosis are covered here in order to provide a global view of the disease.
Collapse
Affiliation(s)
- Shifa Jebari-Benslaiman
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
| | - Unai Galicia-García
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Fundación Biofisika Bizkaia, Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain
| | - Asier Larrea-Sebal
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Fundación Biofisika Bizkaia, Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain
| | | | - Iraide Alloza
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - Koen Vandenbroeck
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Bizkaia, Spain
| | - Asier Benito-Vicente
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Correspondence: (A.B.-V.); (C.M.); Tel.: +34-946-01-2741 (C.M.)
| | - César Martín
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Correspondence: (A.B.-V.); (C.M.); Tel.: +34-946-01-2741 (C.M.)
| |
Collapse
|
24
|
Molecular imaging in atherosclerosis. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00483-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Purpose
As atherosclerosis is a prominent cause of morbidity and mortality, early detection of atherosclerotic plaques is vital to prevent complications. Imaging plays a significant role in this goal. Molecular imaging and structural imaging detect different phases of atherosclerotic progression. In this review, we explain the relation between these types of imaging with the physiopathology of plaques, along with their advantages and disadvantages. We also discuss in detail the most commonly used positron emission tomography (PET) radiotracers for atherosclerosis imaging.
Method
A comprehensive search was conducted to extract articles related to imaging of atherosclerosis in PubMed, Google Scholar, and Web of Science. The obtained papers were reviewed regarding precise relation with our topic. Among the search keywords utilized were "atherosclerosis imaging", "atherosclerosis structural imaging", "atherosclerosis CT scan" "positron emission tomography", "PET imaging", "18F-NaF", "18F-FDG", and "atherosclerosis calcification."
Result
Although structural imaging such as computed tomography (CT) offers essential information regarding plaque structure and morphologic features, these modalities can only detect macroscopic alterations that occur later in the disease’s progression, when the changes are frequently irreversible. Molecular imaging modalities like PET, on the other hand, have the advantage of detecting microscopic changes and allow us to treat these plaques before irreversible changes occur. The two most commonly used tracers in PET imaging of atherosclerosis are 18F-sodium fluoride (18F-NaF) and 18F-fluorodeoxyglucose (18F-FDG). While there are limitations in the use of 18F-FDG for the detection of atherosclerosis in coronary arteries due to physiological uptake in myocardium and high luminal blood pool activity of 18F-FDG, 18F-NaF PET is less affected and can be utilized to analyze the coronary arteries in addition to the peripheral vasculature.
Conclusion
Molecular imaging with PET/CT has become a useful tool in the early detection of atherosclerosis. 18F-NaF PET/CT shows promise in the early global assessment of atherosclerosis, but further prospective studies are needed to confirm its role in this area.
Collapse
|
25
|
Gonçalves I, Oduor L, Matthes F, Rakem N, Meryn J, Skenteris NT, Aspberg A, Orho-Melander M, Nilsson J, Matic L, Edsfeldt A, Sun J, Bengtsson E. Osteomodulin Gene Expression Is Associated With Plaque Calcification, Stability, and Fewer Cardiovascular Events in the CPIP Cohort. Stroke 2022; 53:e79-e84. [PMID: 35135320 DOI: 10.1161/strokeaha.121.037223] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Stable atherosclerotic plaques are characterized by thick fibrous caps of smooth muscle cells, collagen, and macrocalcifications. Identifying factors of plaque stability is necessary to design drugs to prevent plaque rupture and symptoms. Osteomodulin, originally identified in bones, is expressed by bone synthesizing osteoblasts and involved in mineralization. In the present study, we analyzed osteomodulin expression in human carotid plaques, its link with plaque phenotype, calcification, and future cardiovascular events. METHODS Osteomodulin gene expression (OMD; n=82) was determined by RNA sequencing and osteomodulin protein levels by immunohistochemistry (n=45) in carotid plaques obtained by endarterectomy from patients with or without cerebrovascular symptoms from the CPIP (Carotid Plaque Imaging Project) cohort, Skåne University Hospital, Sweden. Plaque components were assessed by immunohistochemistry, RNA sequencing, and multiplex analysis. Patients were followed for cardiovascular events or cardiovascular death during a median of 57 or 70 months, respectively, using national registers. RESULTS OMD levels were increased in plaques from asymptomatic patients compared to symptomatics. High OMD levels were associated with fewer cardiovascular events during follow-up. OMD correlated positively with smooth muscle α-actin (ACTA2; r=0.73, P=10-13) and collagen (COL1A2; r=0.4, P=0.0002), but inversely with CD68 gene expression (r=-0.67, P=10-11), lipids (r=-0.37, P=0.001), intraplaque hemorrhage (r=-0.32, P=0.010), inflammatory cytokine, and matrix metalloproteinase plaque contents. OMD was positively associated with MSX2 (Msh Homeobox 2) (r=0.32, P=0.003), a marker of preosteoblast differentiation, BMP4 (bone morphogenetic protein) (r=0.50, P=0.000002) and BMP6 (r=0.47, P=0.000007), plaque calcification (r=0.35, P=0.016), and was strongly upregulated in osteogenically stimulated smooth muscle cells, which was further increased upon BMP stimulation. Osteomodulin protein was present in calcified regions. Osteomodulin protein levels were associated with plaque calcification (r=0.41, P=0.006) and increased in macrocalcified plaques. CONCLUSIONS These data show that osteomodulin mRNA and protein levels are associated with plaque calcification in human atherosclerosis. Furthermore, osteomodulin mRNA, but not protein levels, is associated with plaque stability.
Collapse
Affiliation(s)
- Isabel Gonçalves
- Department of Clinical Sciences Malmö, Lund University, Sweden. (I.G., L.O., F.M., N.R., J.M., M.O.-M., J.N., A.E., J.S., E.B.).,Cardiology, Skåne University Hospital, Lund University, Sweden. (I.G., A.E.)
| | - Loureen Oduor
- Department of Clinical Sciences Malmö, Lund University, Sweden. (I.G., L.O., F.M., N.R., J.M., M.O.-M., J.N., A.E., J.S., E.B.)
| | - Frank Matthes
- Department of Clinical Sciences Malmö, Lund University, Sweden. (I.G., L.O., F.M., N.R., J.M., M.O.-M., J.N., A.E., J.S., E.B.)
| | - Narjess Rakem
- Department of Clinical Sciences Malmö, Lund University, Sweden. (I.G., L.O., F.M., N.R., J.M., M.O.-M., J.N., A.E., J.S., E.B.)
| | - Jakob Meryn
- Department of Clinical Sciences Malmö, Lund University, Sweden. (I.G., L.O., F.M., N.R., J.M., M.O.-M., J.N., A.E., J.S., E.B.)
| | - Nikolaos-Taxiarchis Skenteris
- Department of Clinical Sciences Malmö, Lund University, Sweden. (I.G., L.O., F.M., N.R., J.M., M.O.-M., J.N., A.E., J.S., E.B.).,Department of Molecular Medicine and Surgery, Karolinska Institutet, Sweden (N.-T.S., L.M.)
| | - Anders Aspberg
- Department of Clinical Sciences, Lund University, Sweden. (A.A.)
| | | | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Sweden. (I.G., L.O., F.M., N.R., J.M., M.O.-M., J.N., A.E., J.S., E.B.)
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Sweden (N.-T.S., L.M.)
| | - Andreas Edsfeldt
- Department of Clinical Sciences Malmö, Lund University, Sweden. (I.G., L.O., F.M., N.R., J.M., M.O.-M., J.N., A.E., J.S., E.B.).,Cardiology, Skåne University Hospital, Lund University, Sweden. (I.G., A.E.).,Wallenberg Centre for Molecular Medicine, Lund University, Sweden. (A.E.)
| | - Jiangming Sun
- Department of Clinical Sciences Malmö, Lund University, Sweden. (I.G., L.O., F.M., N.R., J.M., M.O.-M., J.N., A.E., J.S., E.B.)
| | - Eva Bengtsson
- Department of Clinical Sciences Malmö, Lund University, Sweden. (I.G., L.O., F.M., N.R., J.M., M.O.-M., J.N., A.E., J.S., E.B.)
| |
Collapse
|
26
|
Liu H, Pietersz G, Peter K, Wang X. Nanobiotechnology approaches for cardiovascular diseases: site-specific targeting of drugs and nanoparticles for atherothrombosis. J Nanobiotechnology 2022; 20:75. [PMID: 35135581 PMCID: PMC8822797 DOI: 10.1186/s12951-022-01279-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/21/2022] [Indexed: 02/18/2023] Open
Abstract
Atherosclerosis and atherothrombosis, the major contributors to cardiovascular diseases (CVDs), represent the leading cause of death worldwide. Current pharmacological therapies have been associated with side effects or are insufficient at halting atherosclerotic progression effectively. Pioneering work harnessing the passive diffusion or endocytosis properties of nanoparticles and advanced biotechnologies in creating recombinant proteins for site-specific delivery have been utilized to overcome these limitations. Since CVDs are complex diseases, the most challenging aspect of developing site-specific therapies is the identification of an individual and unique antigenic epitope that is only expressed in lesions or diseased areas. This review focuses on the pathological mechanism of atherothrombosis and discusses the unique targets that are important during disease progression. We review recent advances in site-specific therapy using novel targeted drug-delivery and nanoparticle-carrier systems. Furthermore, we explore the limitations and future perspectives of site-specific therapy for CVDs.
Collapse
Affiliation(s)
- Haikun Liu
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Geoffrey Pietersz
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Burnet Institute, Melbourne, VIC, Australia.,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia.,Department of Medicine, Monash University, Melbourne, VIC, Australia.,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia. .,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia. .,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia. .,Department of Medicine, Monash University, Melbourne, VIC, Australia. .,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia.
| |
Collapse
|
27
|
Gimnich OA, Zil-E-Ali A, Brunner G. Imaging Approaches to the Diagnosis of Vascular Diseases. Curr Atheroscler Rep 2022; 24:85-96. [PMID: 35080717 DOI: 10.1007/s11883-022-00988-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Vascular imaging is a complex field including numerous modalities and imaging markers. This review is focused on important and recent findings in atherosclerotic carotid artery plaque imaging with an emphasis on developments in magnetic resonance imaging (MRI) and computed tomography (CT). RECENT FINDINGS Recent evidence shows that carotid plaque characteristics and not only established measures of carotid plaque burden and stenosis are associated independently with cardiovascular outcomes. On carotid MRI, the presence of a lipid-rich necrotic core (LRNC) has been associated with incident cardiovascular disease (CVD) events independent of wall thickness, a traditional measure of plaque burden. On carotid MRI, intraplaque hemorrhage (IPH) presence has been identified as an independent predictor of stroke. The presence of a fissured carotid fibrous cap has been associated with contrast enhancement on CT angiography imaging. Carotid artery plaque characteristics have been associated with incident CVD events, and advanced plaque imaging techniques may gain additional prominence in the clinical treatment decision process.
Collapse
Affiliation(s)
- Olga A Gimnich
- Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Ahsan Zil-E-Ali
- Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Gerd Brunner
- Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA.
| |
Collapse
|
28
|
Kawtharany L, Bessueille L, Issa H, Hamade E, Zibara K, Magne D. Inflammation and Microcalcification: A Never-Ending Vicious Cycle in Atherosclerosis? J Vasc Res 2022; 59:137-150. [PMID: 35038712 DOI: 10.1159/000521161] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/17/2021] [Indexed: 11/19/2022] Open
Abstract
Inflammatory cells and cytokines are known for long to worsen the development of atherosclerotic plaques in mice, and intense efforts are today devoted to develop anti-inflammatory therapeutic strategies to slow down plaque development. Increasing data indicate that plaque inflammation is intimately associated with microcalcifications, which exert harmful effects eventually culminating with plaque rupture. In this review article, we will first introduce microcalcification location, detection, and effects in atherosclerotic plaques. Then, we will present the numerous data suggesting that inflammatory cells and molecules are responsible for the formation of microcalcifications and the articles showing that microcalcifications stimulate macrophages and smooth muscle cells to produce more pro-inflammatory cytokines. Finally, we will discuss the possibility that microcalcifications might stimulate smooth muscle cells to produce larger and more stable calcifications to stabilize plaques, to exit the vicious cycle associating inflammation and microcalcification in atherosclerotic plaques.
Collapse
Affiliation(s)
- Lynn Kawtharany
- ICBMS UMR CNRS 5246, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Hawraa Issa
- College of Public Health, Phoenicia University, Zahrani, Lebanon
| | - Eva Hamade
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - David Magne
- ICBMS UMR CNRS 5246, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
29
|
Tian L, Huang CK, Ding F, Zhang R. Galectin-3 Mediates Thrombin-Induced Vascular Smooth Muscle Cell Migration. Front Cardiovasc Med 2021; 8:686200. [PMID: 34746246 PMCID: PMC8563778 DOI: 10.3389/fcvm.2021.686200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 09/09/2021] [Indexed: 11/25/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) migration is an important step in the progression and development of vulnerable plaques. Thrombin is involved in both physiological and pathological processes of atherosclerosis. Therefore, the elucidation of the mechanisms underlying thrombin-induced VSMC migration is essential for devising effective treatments aimed at the prevention of plaque instability. In this study, we found that thrombin activated MAPK signaling pathways and increased the expression of galectin-3, which was also a well-known factor in atherosclerosis. Knockdown of galectin-3 by specific small interfering RNA (siRNA) blocked thrombin-induced activation of ERK1/2 and p38 MAPK, but not JNK MAPK. Src/FAK phosphorylation was also shown to be activated by thrombin. FAK autophosphorylation at Y397 was most significantly inhibited by galectin-3 siRNA. Galectin-3 siRNA or specific inhibitor (P38 MAPK inhibitor and ERK1/2 inhibitor) effectively prevented thrombin-induced VSMC migration via reducing paxillin expression. These findings demonstrate, for the first time, that thrombin stimulation of VSMC migration and paxillin expression are regulated by galectin-3, and ERK1/2, p38 MAPK, and Src/FAK signaling pathways are involved in this process. These results are beneficial to clarify the role of galectin-3 in thrombin-induced advanced lesions in atherosclerosis and shed new insights into the regulatory mechanism of VSMC migration in combating plaque rupture.
Collapse
Affiliation(s)
- Lei Tian
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun-Kai Huang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fenghua Ding
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Worssam MD, Jørgensen HF. Mechanisms of vascular smooth muscle cell investment and phenotypic diversification in vascular diseases. Biochem Soc Trans 2021; 49:2101-2111. [PMID: 34495326 PMCID: PMC8589433 DOI: 10.1042/bst20210138] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 12/31/2022]
Abstract
In contrast with the heart, the adult mammalian vasculature retains significant remodelling capacity, dysregulation of which is implicated in disease development. In particular, vascular smooth muscle cells (VSMCs) play major roles in the pathological vascular remodelling characteristic of atherosclerosis, restenosis, aneurysm and pulmonary arterial hypertension. Clonal lineage tracing revealed that the VSMC-contribution to disease results from the hyperproliferation of few pre-existing medial cells and suggested that VSMC-derived cells from the same clone can adopt diverse phenotypes. Studies harnessing the powerful combination of lineage tracing and single-cell transcriptomics have delineated the substantial diversity of VSMC-derived cells in vascular lesions, which are proposed to have both beneficial and detrimental effects on disease severity. Computational analyses further suggest that the pathway from contractile VSMCs in healthy arteries to phenotypically distinct lesional cells consists of multiple, potentially regulatable, steps. A better understanding of how individual steps are controlled could reveal effective therapeutic strategies to minimise VSMC functions that drive pathology whilst maintaining or enhancing their beneficial roles. Here we review current knowledge of VSMC plasticity and highlight important questions that should be addressed to understand how specific stages of VSMC investment and phenotypic diversification are controlled. Implications for developing therapeutic strategies in pathological vascular remodelling are discussed and we explore how cutting-edge approaches could be used to elucidate the molecular mechanisms underlying VSMC regulation.
Collapse
Affiliation(s)
- Matthew D. Worssam
- Cardiovascular Medicine Division, University of Cambridge, Cambridge, U.K
| | - Helle F. Jørgensen
- Cardiovascular Medicine Division, University of Cambridge, Cambridge, U.K
| |
Collapse
|
31
|
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
|
32
|
Nakahara T, Narula J, Fox JJ, Jinzaki M, Strauss HW. Temporal relationship between 18F-sodium fluoride uptake in the abdominal aorta and evolution of CT-verified vascular calcification. J Nucl Cardiol 2021; 28:1936-1945. [PMID: 31741329 DOI: 10.1007/s12350-019-01934-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 09/26/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND Fluoride-18 sodium fluoride (18F-NaF) localizes in microcalcifications in atheroma. The microcalcifications may aggregate, passing the resolution threshold to visualize on computed tomography (CT). We evaluated serial NaF positron emission tomography (PET)-CT scans to determine the temporal relationship between vascular NaF uptake and CT evident calcification in the abdominal aorta. METHODS Prostate cancer patients who had at least 3 NaF PET-CT scans over at least 1.5 years were retrospectively enrolled. Regions of interest were traced in the abdominal aorta on both PET and CT images, excluding skeletal NaF activity. The maximum standardized uptake value (SUVmax) of NaF and the density and volume of calcium (exceeding 130 HU) were summed and divided by the number of slices to produce the SUVmax/slice and the mm3·slice-1 of calcium. RESULTS Of 437 patients, 45 patients met criteria. NaF uptake waxed and waned between scans, while the calcium volume plateaued or increased over time. NaF uptake correlated with calcium volume on the baseline scan (P = .60, < .0001†) and calcium volume increment, especially from 1.0 to 1.5 years (r = .79, P < .0001†). Patients with persistently high NaF uptake showed a higher calcium volume increment (0-1.5 years) than patients with low or transiently high NaF uptake. CONCLUSIONS Abdominal aortic NaF uptake varied over time. NaF uptake on the baseline scans and high NaF uptake on the serial scans preceded an increase in calcium volume, especially by 1.0-1.5 years. Persistently high NaF uptake was associated with a greater increment in calcium volume than patients with transiently elevated or persistently low fluoride uptake.
Collapse
Affiliation(s)
- Takehiro Nakahara
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Jagat Narula
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Josef J Fox
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Masahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - H William Strauss
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
33
|
den Harder AM, Wolterink JM, Bartstra JW, Spiering W, Zwakenberg SR, Beulens JW, Slart RHJA, Luurtsema G, Mali WP, de Jong PA. Vascular uptake on 18F-sodium fluoride positron emission tomography: precursor of vascular calcification? J Nucl Cardiol 2021; 28:2244-2254. [PMID: 31975332 PMCID: PMC8648691 DOI: 10.1007/s12350-020-02031-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/14/2019] [Accepted: 12/31/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND Microcalcifications cannot be identified with the present resolution of CT; however, 18F-sodium fluoride (18F-NaF) positron emission tomography (PET) imaging has been proposed for non-invasive identification of microcalcification. The primary objective of this study was to assess whether 18F-NaF activity can assess the presence and predict the progression of CT detectable vascular calcification. METHODS AND RESULTS The data of two longitudinal studies in which patients received a 18F-NaF PET-CT at baseline and after 6 months or 1-year follow-up were used. The target to background ratio (TBR) was measured on PET at baseline and CT calcification was quantified in the femoral arteries at baseline and follow-up. 128 patients were included. A higher TBR at baseline was associated with higher calcification mass at baseline and calcification progression (β = 1.006 [1.005-1.007] and β = 1.002 [1.002-1.003] in the studies with 6 months and 1-year follow-up, respectively). In areas without calcification at baseline and where calcification developed at follow-up, the TBR was .11-.13 (P < .001) higher compared to areas where no calcification developed. CONCLUSION The activity of 18F-NaF is related to the amount of calcification and calcification progression. In areas where calcification formation occurred, the TBR was slightly but significantly higher.
Collapse
Affiliation(s)
- Annemarie M den Harder
- Department of Radiology, Utrecht University Medical Center, P.O. Box 85500, E01.132, 3508 GA, Utrecht, The Netherlands.
| | - Jelmer M Wolterink
- Image Sciences Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jonas W Bartstra
- Department of Radiology, Utrecht University Medical Center, P.O. Box 85500, E01.132, 3508 GA, Utrecht, The Netherlands
| | - Wilko Spiering
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sabine R Zwakenberg
- Julius Centre for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Joline W Beulens
- Julius Centre for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Riemer H J A Slart
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gert Luurtsema
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Willem P Mali
- Department of Radiology, Utrecht University Medical Center, P.O. Box 85500, E01.132, 3508 GA, Utrecht, The Netherlands
| | - Pim A de Jong
- Department of Radiology, Utrecht University Medical Center, P.O. Box 85500, E01.132, 3508 GA, Utrecht, The Netherlands
| |
Collapse
|
34
|
Sun C, He B, Sun M, Lv X, Wang F, Chen J, Zhang J, Ye Z, Wen J, Liu P. Yes-Associated Protein in Atherosclerosis and Related Complications: A Potential Therapeutic Target That Requires Further Exploration. Front Cardiovasc Med 2021; 8:704208. [PMID: 34513949 PMCID: PMC8430249 DOI: 10.3389/fcvm.2021.704208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis and its complications diseases remain leading causes of cardiovascular morbidity and mortality, bringing a massive burden on public health worldwide. Atherosclerosis is recognized as chronic inflammation, and involves several highly correlated processes, including lipid metabolism dysfunction, endothelial cell dysfunction, inflammation, oxidative stress, vascular smooth muscle cell activation, platelet activation, thrombosis, altered matrix metabolism, and vascular remodeling. Within the past few decades, accumulating evidence has shown that the Yes-associated protein (YAP), the major effector of the Hippo pathway, can play a crucial role in pathogenesis and development of atherosclerosis. Activation of YAP-related pathways, which are induced by alerting flow pattern and matrix stiffness among others, can regulate processes including vascular endothelial cell dysfunction, monocyte infiltration, and smooth muscle cell migration, which contribute to atherosclerotic lesion formation. Further, YAP potentially modulates atherosclerotic complications such as vascular calcification and intraplaque hemorrhage, which require further investigation. Here, we summarized the relevant literature to outline current findings detailing the relationship between of YAP and atherosclerosis and highlight areas for future research.
Collapse
Affiliation(s)
- Congrui Sun
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Bin He
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Mingsheng Sun
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xiaoshuo Lv
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Feng Wang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jie Chen
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jianbin Zhang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Zhidong Ye
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jianyan Wen
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.,Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Peng Liu
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.,Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
35
|
Vulnerable atherosclerotic plaque features: findings from coronary imaging. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2021; 18:577-584. [PMID: 34404993 PMCID: PMC8352771 DOI: 10.11909/j.issn.1671-5411.2021.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Pathological studies have suggested that features of vulnerable atherosclerotic plaques likely to progress and lead to acute cardiovascular events have specific characteristics. Given the progress of intravascular coronary imaging technology, some large prospective studies have detected features of vulnerable atherosclerotic plaques using these imaging modalities. However, the rate of cardiovascular events, such as acute coronary syndrome, has been found to be considerably reduced in the limited follow-up period available in the statin era. Additionally, not all disrupted plaques lead to thrombus formation with clinical presentation. If sub-occlusive or occlusive thrombus formation does not occur, a thrombus on a disrupted plaque will organize without any symptoms, forming a “healed plaque”. Although vulnerable plaque detection using intracoronary imaging is focused on “thin-cap fibroatheroma” leading to plaque rupture, superficial plaque erosion is increasingly recognized; however, the underlying mechanism of thrombus formation on eroded plaques is not well understood. One of intravascular imaging, optical coherence tomography (OCT) has the highest image resolution and has enabled detailed characterization of the plaque in vivo. Here, we reviewed the status and limitations of intravascular imaging in terms of detecting vulnerable plaque through mainly OCT studies. We suggested that vulnerable plaque should be reconsidered in terms of eroded plaque and healed plaque and that both plaque and circulating blood should be assessed in greater detail accordingly.
Collapse
|
36
|
Nakamizo T, Cologne J, Cordova K, Yamada M, Takahashi T, Misumi M, Fujiwara S, Matsumoto M, Kihara Y, Hida A, Ohishi W. Radiation effects on atherosclerosis in atomic bomb survivors: a cross-sectional study using structural equation modeling. Eur J Epidemiol 2021; 36:401-414. [PMID: 33742296 PMCID: PMC8076141 DOI: 10.1007/s10654-021-00731-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/10/2021] [Indexed: 12/24/2022]
Abstract
Past reports indicated that total-body irradiation at low to moderate doses could be responsible for cardiovascular disease risks, but the mechanism remains unclear. The purpose of this study was to investigate the association between radiation exposure and atherosclerosis, an underlying pathology of cardiovascular diseases, in the Japanese atomic bomb survivors. We performed a cross-sectional study measuring 14 clinical-physiological atherosclerosis indicators during clinical exams from 2010 to 2014 in 3274 participants of the Adult Health Study cohort. Multivariable analyses were performed by using a structural equation model with latent factors representing underlying atherosclerotic pathologies: (1) arterial stiffness, (2) calcification, and (3) plaque as measured with indicators chosen a priori on the basis of clinical-physiological knowledge. Radiation was linearly associated with calcification (standardized coefficient per Gy 0.15, 95 % confidence interval: CI [0.070, 0.23]) and plaque (0.11, 95 % CI [0.029, 0.20]), small associations that were comparable to about 2 years of aging per Gy of radiation exposure, but not with arterial stiffness (0.036, 95 % CI [− 0.025, 0.095]). The model fitted better and had narrower confidence intervals than separate ordinary regression models explaining individual indicators independently. The associations were less evident when the dose range was restricted to a maximum of 2 or 1 Gy. By combining individual clinical-physiological indicators that are correlated because of common, underlying atherosclerotic pathologies, we found a small, but significant association of radiation with atherosclerosis.
Collapse
Affiliation(s)
- Tomoki Nakamizo
- Department of Clinical Studies, Radiation Effects Research Foundation (RERF), Nagasaki, Japan.
| | - John Cologne
- Department of Statistics, RERF, Hiroshima, Japan
| | | | | | - Tetsuya Takahashi
- Faculty of Rehabilitation, Hiroshima International University, Hiroshima, Japan
| | | | - Saeko Fujiwara
- Department of Clinical Studies, RERF, Hiroshima, Japan.,Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan
| | - Masayasu Matsumoto
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Hiroshima, Japan.,Department of Neurology, Sakai City Medical Center, Osaka, Japan
| | - Yasuki Kihara
- Department of Clinical Studies, RERF, Hiroshima, Japan.,Department of Cardiovascular Medicine, Hiroshima University, Hiroshima, Japan.,Kobe City Medical Center General Hospital, Hyogo, Japan
| | - Ayumi Hida
- Department of Clinical Studies, Radiation Effects Research Foundation (RERF), Nagasaki, Japan
| | - Waka Ohishi
- Department of Clinical Studies, RERF, Hiroshima, Japan
| |
Collapse
|
37
|
The Napkin-Ring Sign – the Story Behind Invasive Coronary Angiography. JOURNAL OF INTERDISCIPLINARY MEDICINE 2021. [DOI: 10.2478/jim-2021-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Coronary artery disease (CAD) represents one of the leading causes of morbidity and mortality across Europe. Most of the patients do not experience any warning sign before the coronary event develops, therefore screening this group of patients is essential to prevent major cardiac events. Coronary computed tomography angiography (CCTA) offers a noninvasive approach of the coronary arteries, providing information not only on the presence and severity of the coronary stenosis, but is also able to characterize the structure of the coronary wall. CCTA allows complex evaluation of the extension of CAD, and by assessing the structure of the atherosclerotic plaque, it can identify its degree of vulnerability. The napkin-ring sign (NRS) represents a ring-like attenuation of the non-calcified portion of the coronary lesion and has a high specificity (96–100%) for the identification of thin cap fibroatheroma (TCFA) or culprit lesion in acute coronary syndromes (ACS). It is also an independent predictor for ACS events and the strongest predictor for future ACS. Modern CCTA can provide submillimeter isotropic spatial resolution. Thus, CT attenuation-based tissue interpretation enables the assessment of total coronary plaque burden and individual plaque components, with a similar accuracy as intravascular ultrasoud-based investigations. This review aims to present the important role of CCTA as a potent screening tool for patients with CAD, and the current evidences in the detection and quantification of vulnerable plaques.
Collapse
|
38
|
Song Y, Wu H, Wen D, Zhu B, Graner P, Ciancibello L, Rajeswaran H, Salem K, Hajmomenian M, Gilkeson RC, Wilson DL. Detection of coronary calcifications with dual energy chest X-rays: clinical evaluation. Int J Cardiovasc Imaging 2021; 37:767-774. [PMID: 33113069 PMCID: PMC7979464 DOI: 10.1007/s10554-020-02072-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/13/2020] [Indexed: 10/23/2022]
Abstract
Our goal is to assess the ability of physicians to detect coronary calcifications in dual energy chest X-rays processed by a previously developed advanced algorithm. Because the chest X-ray is the most common imaging procedure, because the presence of coronary calcium provides proof of coronary artery disease, and because adherence to therapy can improve health, successful detection could positively impact healthcare for a large number of patients. Both dual energy chest and corroborative CT calcium score images were acquired. Dual energy images were processed with the advanced techniques, including sliding organ registration, so as to enhance coronary calcifications in two-shot dual energy acquisitions. We performed ROC to determine physicians' ability to detect coronary calcifications. Since detection might be easier with heavier calcifications, we used various Agatston score cut-points for determining cases actually positive with calcification in the ROC analysis. In many cases, coronary calcifications were made more visible with the advanced processing as compared to conventional processing. At an Agatston cut-point of 300, coronary calcifications were detected with AUC = 0.85. There were marginal effects on detection performance found with increased X-ray exposure, nearby Agatston cut-point values, and coronary artery territory. Coronary calcifications can be detected in dual energy chest X-rays. The ability to detect disease compares very favorably to other accepted screening methods (e.g., X-ray mammography). As the chest X-ray is an already ordered procedure, there is an opportunity to detect a very large number of persons with coronary artery disease at zero or low cost.
Collapse
Affiliation(s)
- Yingnan Song
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hao Wu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Di Wen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Bo Zhu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Philipp Graner
- Department of Radiology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
- Department of Radiology, University Hospital Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Leslie Ciancibello
- Department of Radiology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
- Department of Radiology, University Hospital Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Haran Rajeswaran
- Department of Radiology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
- Department of Radiology, University Hospital Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Karma Salem
- Department of Radiology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
- Department of Radiology, University Hospital Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Mehrdad Hajmomenian
- Department of Radiology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
- Department of Radiology, University Hospital Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Robert C Gilkeson
- Department of Radiology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
- Department of Radiology, University Hospital Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - David L Wilson
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Department of Radiology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
| |
Collapse
|
39
|
Hu Y, Hu P, Hu B, Chen W, Cheng D, Shi H. Dynamic monitoring of active calcification in atherosclerosis by 18F-NaF PET imaging. Int J Cardiovasc Imaging 2021; 37:731-739. [PMID: 32926308 DOI: 10.1007/s10554-020-02019-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 04/20/2020] [Indexed: 11/26/2022]
Abstract
The objective was to dynamically monitor the progression of atherosclerotic plaques in ApoE-/- mice with 18F-NaF PET imaging. The ApoE-/- mice were used to develop atherosclerosis models, and the C57BL/6 J mice were used as control. 18F-NaF PET was performed when the mice were 12, 20, and 30 weeks of age. Serum lipids and lipoproteins profiles, inflammatory cytokines, and calcification factors were tested by ELISA. The lipid distribution, morphology, and calcification of plaque were evaluated by Oil Red O, HE, and alizarin red staining. The correlation between imaging and the extent of calcification was analyzed by Pearson correlation analysis. The uptake of 18F-NaF in the aorta was gradually increased with each weekly extension. Compared with the ApoE-/- mice at the age of 12 weeks and 20 weeks, the levels of lipoprotein, inflammatory cytokines, and calcification factors were higher at 30 weeks. In Oil Red O, HE, and alizarin red staining, the extent of the lipid area and calcification increased with time. The correlation analysis showed that the uptake of 18F-NaF in the aorta was related to the extent of calcification. 18F-NaF may dynamically monitor the progression of atherosclerotic plaques and ongoing microcalcification formation.
Collapse
Affiliation(s)
- Yan Hu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Pengcheng Hu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Bingxin Hu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Weijia Chen
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Dengfeng Cheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China.
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China.
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China.
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China.
| |
Collapse
|
40
|
Bartstra JW, van den Beukel TC, Van Hecke W, Mali WPTM, Spiering W, Koek HL, Hendrikse J, de Jong PA, den Harder AM. Intracranial Arterial Calcification: Prevalence, Risk Factors, and Consequences: JACC Review Topic of the Week. J Am Coll Cardiol 2021; 76:1595-1604. [PMID: 32972537 DOI: 10.1016/j.jacc.2020.07.056] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/09/2020] [Accepted: 07/20/2020] [Indexed: 02/07/2023]
Abstract
Intracranial large and small arterial calcifications are a common incidental finding on computed tomography imaging in the general population. Here we provide an overview of the published reports on prevalence of intracranial arterial calcifications on computed tomography imaging and histopathology in relation to risk factors and clinical outcomes. We performed a systematic search in Medline, with a search filter using synonyms for computed tomography scanning, (histo)pathology, different intracranial arterial beds, and calcification. We found that intracranial calcifications are a frequent finding in all arterial beds with the highest prevalence in the intracranial internal carotid artery. In general, prevalence increases with age. Longitudinal studies on calcification progression and intervention studies are warranted to investigate the possible causal role of calcification on clinical outcomes. This might open up new therapeutic directions in stroke and dementia prevention and the maintenance of the healthy brain.
Collapse
Affiliation(s)
- Jonas W Bartstra
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| | - Tim C van den Beukel
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Wim Van Hecke
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Willem P T M Mali
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Wilko Spiering
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Huiberdina L Koek
- Department of Geriatrics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jeroen Hendrikse
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Pim A de Jong
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Annemarie M den Harder
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
41
|
Montanaro M, Scimeca M, Anemona L, Servadei F, Giacobbi E, Bonfiglio R, Bonanno E, Urbano N, Ippoliti A, Santeusanio G, Schillaci O, Mauriello A. The Paradox Effect of Calcification in Carotid Atherosclerosis: Microcalcification is Correlated with Plaque Instability. Int J Mol Sci 2021; 22:ijms22010395. [PMID: 33401449 PMCID: PMC7796057 DOI: 10.3390/ijms22010395] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 12/14/2022] Open
Abstract
Background: this study aims to investigate the possible association among the histopathologic features of carotid plaque instability, the presence of micro- or macrocalcifications, the expression of in situ inflammatory biomarkers, and the occurrence of the major risk factors in this process in a large series of carotid plaques. Methods: a total of 687 carotid plaques from symptomatic and asymptomatic patients were collected. Histological evaluation was performed to classify the calcium deposits in micro or macrocalcifications according to their morphological features (location and size). Immunohistochemistry was performed to study the expression of the main inflammatory biomarkers. Results: results here reported demonstrated that calcifications are very frequent in carotid plaques, with a significant difference between the presence of micro- and macrocalcifications. Specifically, microcalcifications were significantly associated to high inflamed unstable plaques. Paradoxically, macrocalcifications seem to stabilize the plaque and are associated to a M2 macrophage polarization instead. Discussion: the characterization of mechanisms involved in the formation of carotid calcifications can lay the foundation for developing new strategies for the management of patients affected by carotid atherosclerosis. Data of this study could provide key elements for an exhaustive evaluation of carotid plaque calcifications allowing to establish the risk of associated clinical events.
Collapse
Affiliation(s)
- Manuela Montanaro
- Department of Experimental Medicine, University “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (M.S.); (L.A.); (F.S.); (E.G.); (R.B.); (E.B.); (G.S.)
| | - Manuel Scimeca
- Department of Experimental Medicine, University “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (M.S.); (L.A.); (F.S.); (E.G.); (R.B.); (E.B.); (G.S.)
- Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
| | - Lucia Anemona
- Department of Experimental Medicine, University “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (M.S.); (L.A.); (F.S.); (E.G.); (R.B.); (E.B.); (G.S.)
| | - Francesca Servadei
- Department of Experimental Medicine, University “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (M.S.); (L.A.); (F.S.); (E.G.); (R.B.); (E.B.); (G.S.)
| | - Erica Giacobbi
- Department of Experimental Medicine, University “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (M.S.); (L.A.); (F.S.); (E.G.); (R.B.); (E.B.); (G.S.)
| | - Rita Bonfiglio
- Department of Experimental Medicine, University “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (M.S.); (L.A.); (F.S.); (E.G.); (R.B.); (E.B.); (G.S.)
- Fondazione Umberto Veronesi (FUV), Piazza Velasca 5, 20122 Milano, Italy
| | - Elena Bonanno
- Department of Experimental Medicine, University “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (M.S.); (L.A.); (F.S.); (E.G.); (R.B.); (E.B.); (G.S.)
| | - Nicoletta Urbano
- Nuclear Medicine Unit, Department of Oncohaematology, Policlinico “Tor Vergata”, viale oxford 81, 00133 Rome, Italy;
| | - Arnaldo Ippoliti
- Vascular Surgery, Department of Biomedicine and Prevention, Policlinico “Tor Vergata”, viale oxford 81, 00133 Rome, Italy;
| | - Giuseppe Santeusanio
- Department of Experimental Medicine, University “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (M.S.); (L.A.); (F.S.); (E.G.); (R.B.); (E.B.); (G.S.)
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, University “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (M.S.); (L.A.); (F.S.); (E.G.); (R.B.); (E.B.); (G.S.)
- Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
- Correspondence: ; Tel.: +39-0620903908
| |
Collapse
|
42
|
Molecular Aspects and Prognostic Significance of Microcalcifications in Human Pathology: A Narrative Review. Int J Mol Sci 2020; 22:ijms22010120. [PMID: 33374380 PMCID: PMC7795544 DOI: 10.3390/ijms22010120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022] Open
Abstract
The presence of calcium deposits in human lesions is largely used as imaging biomarkers of human diseases such as breast cancer. Indeed, the presence of micro- or macrocalcifications is frequently associated with the development of both benign and malignant lesions. Nevertheless, the molecular mechanisms involved in the formation of these calcium deposits, as well as the prognostic significance of their presence in human tissues, have not been completely elucidated. Therefore, a better characterization of the biological process related to the formation of calcifications in different tissues and organs, as well as the understanding of the prognostic significance of the presence of these calcium deposits into human tissues could significantly improve the management of patients characterized by microcalcifications associated lesions. Starting from these considerations, this narrative review highlights the most recent histopathological and molecular data concerning the formation of calcifications in breast, thyroid, lung, and ovarian diseases. Evidence reported here could deeply change the current point of view concerning the role of ectopic calcifications in the progression of human diseases and also in the patients’ management. In fact, the presence of calcifications can suggest an unfavorable prognosis due to dysregulation of normal tissues homeostasis.
Collapse
|
43
|
Blanda V, Bracale UM, Di Taranto MD, Fortunato G. Galectin-3 in Cardiovascular Diseases. Int J Mol Sci 2020; 21:ijms21239232. [PMID: 33287402 PMCID: PMC7731136 DOI: 10.3390/ijms21239232] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
Galectin-3 (Gal-3) is a β-galactoside-binding protein belonging to the lectin family with pleiotropic regulatory activities and several physiological cellular functions, such as cellular growth, proliferation, apoptosis, differentiation, cellular adhesion, and tissue repair. Inflammation, tissue fibrosis and angiogenesis are the main processes in which Gal-3 is involved. It is implicated in the pathogenesis of several diseases, including organ fibrosis, chronic inflammation, cancer, atherosclerosis and other cardiovascular diseases (CVDs). This review aims to explore the connections of Gal-3 with cardiovascular diseases since they represent a major cause of morbidity and mortality. We herein discuss the evidence on the pro-inflammatory role of Gal-3 in the atherogenic process as well as the association with plaque features linked to lesion stability. We report the biological role and molecular mechanisms of Gal-3 in other CVDs, highlighting its involvement in the development of cardiac fibrosis and impaired myocardium remodelling, resulting in heart failure and atrial fibrillation. The role of Gal-3 as a prognostic marker of heart failure is described together with possible diagnostic applications to other CVDs. Finally, we report the tentative use of Gal-3 inhibition as a therapeutic approach to prevent cardiac inflammation and fibrosis.
Collapse
Affiliation(s)
- Valeria Blanda
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (V.B.); (G.F.)
- Istituto Zooprofilattico Sperimentale della Sicilia, via Gino Marinuzzi 3, 90129 Palermo, Italy
| | - Umberto Marcello Bracale
- Dipartimento di Sanità Pubblica, Università degli Studi di Napoli Federico II, 80131 Naples, Italy;
| | - Maria Donata Di Taranto
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (V.B.); (G.F.)
- CEINGE S.C.a r.l. Biotecnologie Avanzate, 80131 Naples, Italy
- Correspondence: ; Tel.: +39-081-7463530
| | - Giuliana Fortunato
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (V.B.); (G.F.)
- CEINGE S.C.a r.l. Biotecnologie Avanzate, 80131 Naples, Italy
| |
Collapse
|
44
|
Gijsen FJH, Vis B, Barrett HE, Zadpoor AA, Verhagen HJ, Bos D, van der Steen AFW, Akyildiz AC. Morphometric and Mechanical Analyses of Calcifications and Fibrous Plaque Tissue in Carotid Arteries for Plaque Rupture Risk Assessment. IEEE Trans Biomed Eng 2020; 68:1429-1438. [PMID: 33186100 DOI: 10.1109/tbme.2020.3038038] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Atherosclerotic plaque rupture in carotid arteries is a major source of cerebrovascular events. Calcifications are highly prevalent in carotid plaques, but their role in plaque rupture remains poorly understood. This work studied the morphometric features of calcifications in carotid plaques and their effect on the stress distribution in the fibrous plaque tissue at the calcification interface, as a potential source of plaque rupture and clinical events. METHODS A comprehensive morphometric analysis of 65 histology cross-sections from 16 carotid plaques was performed to identify the morphology (size and shape) and location of plaque calcifications, and the fibrous tissue fiber organization around them. Calcification-specific finite element models were constructed to examine the fibrous plaque tissue stresses at the calcification interface. Statistical correlation analysis was performed to elucidate the impact of calcification morphology and fibrous tissue organization on interface stresses. RESULTS Hundred-seventy-one calcifications were identified on the histology cross-sections, which showed great variation in morphology. Four distinct patterns of fiber organization in the plaque tissue were observed around the calcification. They were termed as attached, pushed-aside, encircling and random patterns. The stress analyses showed that calcifications are correlated with high interface stresses, which might be comparable to or even above the plaque strength. The stress levels depended on the calcification morphology and fiber organization. Thicker calcification with a circumferential slender shape, located close to the lumen were correlated most prominently to high interface stresses. CONCLUSION Depending on its morphology and the fiber organization around it, a calcification in an atherosclerotic plaque can act as a stress riser and cause high interface stresses. SIGNIFICANCE This study demonstrated the potential of calcifications in atherosclerotic plaques to cause elevated stresses in plaque tissue and provided a biomechanical explanation for the histopathological findings of calcification-associated plaque rupture.
Collapse
|
45
|
Kurihara O, Kim HO, Russo M, Araki M, Nakajima A, Lee H, Takano M, Mizuno K, Jang IK. Relation of Low-Density Lipoprotein Cholesterol Level to Plaque Rupture. Am J Cardiol 2020; 134:48-54. [PMID: 32892992 DOI: 10.1016/j.amjcard.2020.08.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 01/28/2023]
Abstract
Statin therapy reduces low-density lipoprotein cholesterol (LDL-C), inflammation, and atherosclerotic cardiovascular disease. We investigated the association between LDL-C and statin therapy on the prevalence of plaque rupture (PR). Patients with acute coronary syndromes who underwent optical coherence tomography imaging of the culprit lesion were divided into 4 groups based on LDL-C level and statin use (Group 1: LDL-C ≤ 100 without statin; Group 2; LDL-C ≤ 100 with statin; Group 3: LDL-C > 100 with statin; Group 4: LDL-C > 100 without statin), and the prevalence of PR was compared between the groups. Among 896 patients, PR was diagnosed in 444 (49.6%) patients. The prevalence of PR was significantly different among the 4 groups (p = 0.007): it was highest in the high LDL-C without statin group and lowest in the low LDL-C without statin group (53.9% and 39.2%, respectively). Compared with the high LDL-C without statin group, the low LDL-C without statin and low LDL-C with statin groups had a significantly lower prevalence of PR (p = 0.001, p = 0.040, respectively), and the low LDL-C with statin group had a significantly higher prevalence of calcification (p = 0.037). The patients with naturally low LDL-C have the lowest risk of PR. The patients with low LDL-C achieved by statin therapy had a higher prevalence of calcification. When LDL-C level is elevated, early and aggressive treatment with statin may help to prevent PR by stabilizing plaques through calcification.
Collapse
|
46
|
Borrás T, Cowley DO, Asokan P, Pandya K. Generation of a Matrix Gla (Mgp) floxed mouse, followed by conditional knockout, uncovers a new Mgp function in the eye. Sci Rep 2020; 10:18583. [PMID: 33122788 PMCID: PMC7596545 DOI: 10.1038/s41598-020-75031-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 10/09/2020] [Indexed: 01/07/2023] Open
Abstract
The ability to ablate a gene in a given tissue by generating a conditional knockout (cKO) is crucial for determining its function in the targeted tissue. Such tissue-specific ablation is even more critical when the gene's conventional knockout (KO) is lethal, which precludes studying the consequences of its deletion in other tissues. Therefore, here we describe a successful strategy that generated a Matrix Gla floxed mouse (Mgp.floxed) by the CRISPR/Cas9 system, that subsequently allowed the generation of cKOs by local viral delivery of the Cre-recombinase enzyme. MGP is a well-established inhibitor of calcification gene, highly expressed in arteries' smooth muscle cells and chondrocytes. MGP is also one of the most abundant genes in the trabecular meshwork, the eye tissue responsible for maintenance of intraocular pressure (IOP) and development of Glaucoma. Our strategy entailed one-step injection of two gRNAs, Cas9 protein and a long-single-stranded-circular DNA donor vector (lsscDNA, 6.7 kb) containing two loxP sites in cis and 900-700 bp 5'/3' homology arms. Ocular intracameral injection of Mgp.floxed mice with a Cre-adenovirus, led to an Mgp.TMcKO mouse which developed elevated IOP. Our study discovered a new role for the Mgp gene as a keeper of physiological IOP in the eye.
Collapse
Affiliation(s)
- Teresa Borrás
- Department of Ophthalmology, University of North Carolina School of Medicine, 4109C Neuroscience Research Building CB 7041, 115 Mason Farm Road, Chapel Hill, NC, 27599-7041, USA.
| | - Dale O Cowley
- Animal Models Core, University of North Carolina, Chapel Hill, NC, USA
| | - Priyadarsini Asokan
- Department of Ophthalmology, University of North Carolina School of Medicine, 4109C Neuroscience Research Building CB 7041, 115 Mason Farm Road, Chapel Hill, NC, 27599-7041, USA
| | - Kumar Pandya
- Animal Models Core, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
47
|
Tsai CH, Lin LY, Lin YH, Tsai IJ, Huang JW. Abdominal aorta calcification predicts cardiovascular but not non-cardiovascular outcome in patients receiving peritoneal dialysis: A prospective cohort study. Medicine (Baltimore) 2020; 99:e21730. [PMID: 32925715 PMCID: PMC7489593 DOI: 10.1097/md.0000000000021730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Abdominal aorta calcification (AAC) is associated with worse clinical outcomes in dialysis patients. However, the long-term prognostic values of AAC to cardiovascular (CV) and non-CV mortality in patients starting peritoneal dialysis (PD) remain unknown. This study is aimed to the analyze the predictive power of AAC to CV and non-CV mortality in PD patients. We prospectively enrolled 123 patients undergoing PD. All patients received quantitative analysis of AAC via abdominal computer tomography at enrollment. The AAC ratio was measured by the area of the whole aorta affected by aortic calcification above the iliac bifurcation. The CV mortality and non-CV mortality during the follow-up period were investigated using the Cox proportional hazard model and time-dependent receiver operating characteristic (ROC) analysis. After median 6.8 (interquartile range, 3.6-9.2) years of follow-up, there were 18 CV mortality, 24 non-CV mortality and 42 total mortality. The age and AAC ratio were significantly higher in CV mortality group compared with others without CV mortality. In time-dependent ROC analysis, AAC had excellent predictive power of CV mortality (AUC:0.787) but not non-CV mortality (AUC:0.537). The best cutoff value of AAC ratio to predict CV mortality was 39%. In addition, AAC was not associated with non-CV mortality but remained to be a significantly predictor of CV mortality after adjusted with clinical covariates in different Cox proportional hazard models. AAC has excellent prognostic value of CV mortality but is unable to predict non-CV morality in patients undergoing PD.
Collapse
Affiliation(s)
- Cheng-Hsuan Tsai
- Department of Internal Medicine, National Taiwan University Hospital, JinShan Branch, New Taipei
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine
| | - Lian-Yu Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine
| | - Yen-Hung Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine
| | - I-Jung Tsai
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine
| | - Jenq-Wen Huang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
48
|
Zheng Z, Zhao Q, Wei J, Wang B, Wang H, Meng L, Xin Y, Jiang X. Medical prevention and treatment of radiation-induced carotid injury. Biomed Pharmacother 2020; 131:110664. [PMID: 32861067 DOI: 10.1016/j.biopha.2020.110664] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/06/2020] [Accepted: 08/20/2020] [Indexed: 11/30/2022] Open
Abstract
Radiotherapy has significantly improved the survival of cancer patients but is also associated with several adversities, including radiation-induced carotid injury (RICI). The RICI mechanisms are complex, including vessel inflammatory injury, carotid atherosclerosis, intimal proliferation, media necrosis, and peri-adventitial fibrosis. The main manifestation and adverse consequence of RICI is carotid artery stenosis (CAS), which can lead to stroke and transient ischemic attack. Currently, carotid artery injury is primarily diagnosed via color-coded duplex sonography. Early detection of traumatic changes in the carotid artery depends on measurements of carotid intima-media thickness; serum biomarker testing also shows great potential. CAS is mainly treated with carotid endarterectomy or carotid angioplasty and stent implantation. Notably, bone marrow mesenchymal stem cells are advantageous in RICI treatment and reduce carotid inflammation, oxidative stress, and delaying atherosclerosis. This review summarizes the mechanisms, examination methods, and latest treatments for RICI to provide data for its clinical prevention and treatment.
Collapse
Affiliation(s)
- Zhuangzhuang Zheng
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| | - Qin Zhao
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| | - Jinlong Wei
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| | - Bin Wang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| | - Huanhuan Wang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL, 33612, USA.
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| |
Collapse
|
49
|
Abstract
Matrix mineralization can be divided into physiological mineralization and pathological mineralization. There is a consensus among existing studies that matrix vesicles (MVs) are the starting sites of bone mineralization, and each component of MVs serves a certain function in mineralization. In addition, ectopic MVs pathologically promote undesired calcification, the primary focus of which is the promotion of vascular calcification. However, the specific mechanisms of the actions of MVs in bone-vascular axis cross-talk have not been fully elucidated. This review summarizes the latest research in this field and explores the roles of MVs in the bone-vascular axis with the aim of generating new ideas for the prevention and treatment of vascular calcification and bone metabolic disease.
Collapse
|
50
|
Pugliese L, Spiritigliozzi L, Di Tosto F, Ricci F, Cavallo AU, Di Donna C, De Stasio V, Presicce M, Benelli L, D'Errico F, Pasqualetto M, Floris R, Chiocchi M. Association of plaque calcification pattern and attenuation with instability features and coronary stenosis and calcification grade. Atherosclerosis 2020; 311:150-157. [PMID: 32771265 DOI: 10.1016/j.atherosclerosis.2020.06.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 06/10/2020] [Accepted: 06/24/2020] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND AIMS Coronary computed tomography (CT) allows calculating coronary artery calcium score (CACS). However, other CT features might be more strongly related to plaque vulnerability and risk of future coronary events. This study investigated the association of plaque calcification pattern and attenuation with plaque instability features, coronary artery disease (CAD) grade and CACS. METHODS One-hundred patients with coronary stenosis associated with calcified plaques were considered for this analysis. CACS, CAD grade, calcification pattern and attenuation, features of plaque instability, and epicardial adipose tissue (EAT) thickness and attenuation were assessed with non-contrast and contrast-enhanced CT angiography. RESULTS Of 373 calcified plaques, 131 were responsible for the highest degree of coronary stenosis (1.31 ± 0.53 per patient). Participants were stratified according to the features of the highest-grade lesion(s) into patients with large (35%), spotty (52%) or mixed (13%) calcification pattern and tertiles of plaque calcification attenuation (using the mean value for multiple lesions). Patients with large calcification pattern or higher plaque calcification attenuation had higher stenosis and CACS grade (and EAT attenuation), but lower plaque instability score, whereas those with spotty calcification pattern or lower plaque calcification attenuation had lower stenosis and CACS grade (and EAT attenuation), but higher plaque instability score. Among the instability features, low attenuation and napkin-ring sign, but not positive remodeling, were associated with a spotty pattern and a lower calcification attenuation. CONCLUSIONS Both the pattern and attenuation of calcification should be considered, in addition to CACS, for risk stratification of heavily calcified high-risk patients with non-critical coronary stenosis.
Collapse
Affiliation(s)
- Luca Pugliese
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy.
| | - Luigi Spiritigliozzi
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Federica Di Tosto
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Francesca Ricci
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Armando U Cavallo
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Carlo Di Donna
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Vincenzo De Stasio
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Matteo Presicce
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Leonardo Benelli
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Francesca D'Errico
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Monia Pasqualetto
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Roberto Floris
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Marcello Chiocchi
- Department of Biomedicine and Prevention, Division of Diagnostic Imaging, Tor Vergata University of Rome and Unit of Diagnostic Imaging, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| |
Collapse
|