1
|
Yvan-Charvet L, Barouillet T, Borowczyk C. Haematometabolism rewiring in atherosclerotic cardiovascular disease. Nat Rev Cardiol 2025; 22:414-430. [PMID: 39743562 DOI: 10.1038/s41569-024-01108-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 01/04/2025]
Abstract
Atherosclerotic cardiovascular diseases are the most frequent cause of death worldwide. The clinical complications of atherosclerosis are closely linked to the haematopoietic and immune systems, which maintain homeostatic functions and vital processes in the body. The nodes linking metabolism and inflammation are receiving increasing attention because they are inextricably linked to inflammatory manifestations of non-communicable diseases, including atherosclerosis. Although metabolism and inflammation are essential to survival and involve all tissues, we still know little about how these processes influence each other. In an effort to understand these mechanisms, in this Review we explore whether and how potent cardiovascular risk factors and metabolic modifiers of atherosclerosis influence the molecular and cellular machinery of 'haematometabolism' (metabolic-dependent haematopoietic stem cell skewing) and 'efferotabolism' (metabolic-dependent efferocyte reprogramming). These changes might ultimately propagate a quantitative and qualitative drift of the macrophage supply chain and affect the clinical manifestations of atherosclerosis. Refining our understanding of the different metabolic requirements of these processes could open the possibility of developing therapeutics targeting haematometabolism that, in conjunction with improved dietary habits, help rebalance and promote efficient haematopoiesis and efferocytosis and decrease the risk of atherosclerosis complications.
Collapse
Affiliation(s)
- Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France.
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France.
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| | - Thibault Barouillet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | - Coraline Borowczyk
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France.
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France.
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| |
Collapse
|
2
|
Kraler S, Mueller C, Libby P, Bhatt DL. Acute coronary syndromes: mechanisms, challenges, and new opportunities. Eur Heart J 2025:ehaf289. [PMID: 40358623 DOI: 10.1093/eurheartj/ehaf289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/03/2025] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Despite advances in research and patient management, atherosclerosis and its dreaded acute and chronic sequelae continue to account for one out of three deaths globally. The vast majority of acute coronary syndromes (ACS) arise from either plaque rupture or erosion, but other mechanisms, including calcific nodules, embolism, spontaneous coronary artery dissection, coronary spasm, and microvascular dysfunction, can also cause ACS. This ACS heterogeneity necessitates a paradigm shift in its management that extends beyond the binary interpretation of electrocardiographic and biomarker data. Indeed, given the evolution in the global risk factor profile, the increasing importance of previously underappreciated mechanisms, the evolving appreciation of sex-specific disease characteristics, and the advent of rapidly evolving technologies, a precision medicine approach is warranted. This review provides an update of the mechanisms of ACS, delineates the role of previously underappreciated contributors, discusses sex-specific differences, and explores novel tools for contemporary and personalized management of patients with ACS. Beyond mechanistic insights, it examines evolving imaging techniques, biomarkers, and regression- and machine learning-based approaches for the diagnosis (e.g. CoDE-ACS, MI3) and prognosis (e.g. PRAISE, GRACE, SEX-SHOCK scores) of ACS, along with their implications for future ACS management. A more individualized approach to patients with ACS is advocated, emphasizing the need for innovative studies on emerging technologies, including artificial intelligence, which may collectively facilitate clinical decision-making within a more mechanistic framework, thereby personalizing patient care and potentially improving long-term outcomes.
Collapse
Affiliation(s)
- Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Cardiology and Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland
| | - Christian Mueller
- Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, 1 Gustave Levy Place, Box 1030, New York, NY 10029, USA
| |
Collapse
|
3
|
Wei W, Du Y, Kang X, Liu Y, Liu Y, Guo Q, He Q, Wang J, Zhu S, Zhou JY, Bu W. Nanoscale Rhodium(I) Based Metal-Organic Framework Demonstrating Intense NIR-II Luminescence for Bioimaging. NANO LETTERS 2025; 25:4613-4620. [PMID: 40052801 DOI: 10.1021/acs.nanolett.5c00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Although luminescent metal-organic frameworks (MOFs) have been widely reported, rare examples were found to emit in the second near-infrared (NIR-II, 1000-1700 nm) window. In this work, two nanoscale rhodium(I)-based MOFs (Rh-1@SDS and Rh-1@DSPE-PEG) have been controllably constructed in the aqueous dispersions of sodium dodecyl sulfate (SDS) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-methoxy-poly(ethylene glycol) (DSPE-PEG), wherein micelle- and vesicle-like aggregates form, respectively, with high colloidal stability. The vesicular dispersion of Rh-1@DSPE-PEG exhibits intense NIR-II luminescence at 1125 (1245, shoulder) nm. Consequently, this nanoMOF was used as an NIR-II luminescence probe, indicative of high-resolution systemic and local vascular imaging, where the postoperative recovery process of flap transplantation was clearly visualized. Meanwhile, it also demonstrates superior tumor targeting in the NIR-II window. To the best of our knowledge, this research represents the first example of nanoMOFs having intense NIR-II luminescence and excellent imaging capabilities.
Collapse
Affiliation(s)
- Wenxuan Wei
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
- School of Physical Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Yijing Du
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun 130021, China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| | - Xiaomei Kang
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Yilin Liu
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Yujia Liu
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Qian Guo
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Qun He
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jun Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun 130021, China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| | - Jin Yuan Zhou
- School of Physical Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Weifeng Bu
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| |
Collapse
|
4
|
Odeberg J, Halling A, Ringborn M, Freitag M, Persson ML, Vaara I, Råstam L, Odeberg H, Lindblad U. Markers of inflammation predicts long-term mortality in patients with acute coronary syndrome - a cohort study. BMC Cardiovasc Disord 2025; 25:190. [PMID: 40089663 PMCID: PMC11909928 DOI: 10.1186/s12872-025-04608-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/25/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Chronic low-grade inflammation is a well-known risk factor for coronary heart disease (CHD) and future cardiovascular events. Anti-inflammatory therapy can reduce the risk of ischemic cardiovascular disease (CVD) events following myocardial infarction (MI). However, it remains unknown to what extent inflammation at the time of an acute event predicts long-term outcomes. We explored whether routine blood measurements of inflammatory markers during an acute coronary syndrome (ACS) are predictive of long-term mortality. METHODS In a cohort of 5292 consecutive patients admitted to a coronary intensive care unit with suspected ACS over a four-year period in the Carlscrona Heart Attack Prognosis Study (CHAPS), 908 patients aged 30-74 years (644 men, 264 women) were diagnosed with MI (527) or unstable angina (UA) (381). A 10-year follow-up study was conducted using Swedish national registries, with total mortality and cardiac mortality as primary outcomes. RESULTS Long-term total and cardiac mortality were significantly associated with higher leukocyte counts (e.g., neutrophils, monocytes, p ≤ 0.001), higher levels of inflammatory biomarkers (e.g., C-reactive protein, Serum Amyloid A, fibrinogen, p ≤ 0.001), and elevated neutrophil-lymphocyte ratio (NLR) (p < 0.001) and monocyte-lymphocyte ratio (MLR) (p = 0.002), all measured at ACS admission. These associations were independent of ACS diagnosis. CONCLUSION Our results suggest that level of inflammation at ACS presentation-beyond its established role as a major CHD risk factor-also predicts long-term mortality following ACS. Notably, inflammation at the time of the event was a stronger predictor of long-term mortality than the acute event outcome itself. However, limitations include the observational study design, moderate sample size, and absence of modern high-sensitivity cardiac biomarkers and contemporary ACS management strategies in this cohort. The results should therefore be interpreted in the context of historical clinical practice. While our model-wise complete-case approach ensured consistency, missing data remains a potential source of bias. Future studies in larger, more contemporary cohorts are needed to validate these findings and refine risk stratification strategies.
Collapse
Affiliation(s)
- Jacob Odeberg
- Department of Protein Science, Science for Life Laboratory Stockholm, CBH, KTH Royal Institute of Technology, Stockholm, 100 44, Sweden.
- Department of Clinical Medicine, Faculty of Health Science, Arctic University of Tromsö (UiT), Tromsö, N 9037, Norway.
- Division of Internal Medicine, University Hospital North Norway (UNN), Tromsö, Norway.
- Department of Hematology, Coagulation Unit, Karolinska University Hospital, Stockholm, Sweden.
- Department of Medicine, Solna, Karolinska Institute, Stockholm, SE, 171 77, Sweden.
| | - Anders Halling
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Michael Ringborn
- Thoracic Center, Blekinge County Hospital Karlskrona, Karlskrona, Sweden
| | - Michael Freitag
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Marie Louise Persson
- Department of Laboratory Medicine, Blekinge County Hospital Karlskrona, Karlskrona, Sweden
| | - Ivar Vaara
- Department of Laboratory Medicine, Blekinge County Hospital Karlskrona, Karlskrona, Sweden
| | - Lennart Råstam
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Håkan Odeberg
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Ulf Lindblad
- School of Public Health and Community Medicine/Primary Health Care, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
5
|
Bjarnadóttir S, Aspelund T, Gudmundsson EF, Gudnason V, Andersen K. Why have temporal trends in STEMI and NSTEMI incidence and short-term mortality changed in recent years? A nationwide 35-year cohort study in Iceland. BMJ Open 2025; 15:e087815. [PMID: 40081976 PMCID: PMC11907030 DOI: 10.1136/bmjopen-2024-087815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 02/08/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVES Temporal trends in the incidence of ST-elevation myocardial infarction (STEMI) have been declining in many countries, while the incidence of non-ST elevation myocardial infarction (NSTEMI) has reached a plateau or even increased. The reasons for these changing trends have yet to be explained. We analysed these trends and short-term mortality from acute coronary syndromes in a nationwide cohort study over 35 years in Iceland. DESIGN Retrospective cohort study using a national MI registry. SETTING Iceland. PARTICIPANTS All cases of myocardial infarction in individuals aged 25-74 years in Iceland 1981-2015. METHODS Each case was classified as STEMI, NSTEMI or no ECG taken. ECG recordings were classified according to Minnesota criteria. OUTCOME MEASURES Trends of STEMI and NSTEMI incidence and 1-day and 28-day mortality were obtained from the National Death Registry. RESULTS A total of 10 348 cases were identified (mean age 61 years, 76.4% male). These were categorised as STEMI (32.7%), NSTEMI (45.8%) and no ECG taken (21.5%). We detected a significant 3.7% annual decline in the incidence of first MI. The age-adjusted incidence of STEMI showed an 83.2% decline, most pronounced after 1994, while for NSTEMI the decline was 66.5%, reaching a plateau from the year 1989 onwards. In Iceland, the uptake of highly sensitive biomarkers was initiated in 1997 (cardiac troponin T) and 2012 (high-sensitive troponin T), respectively. CONCLUSIONS The different temporal trends in the incidence of STEMI and NSTEMI cannot be explained only by the uptake of highly sensitive biomarkers in 1997 and 2012. The change in population-level risk factor exposure is likely to have influenced atherosclerotic plaque burden and thrombotic mechanisms. Finally, increasing uptake of cardioprotective pharmacological and interventional therapy may have resulted in a primary preventive effect on plaque rupture and thrombosis and thus on the rates of STEMI and NSTEMI disproportionally.
Collapse
Affiliation(s)
- Sólveig Bjarnadóttir
- Department of Cardiology, Landspitali National University Hospital of Iceland, Reykjavik, Iceland
| | - Thor Aspelund
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland School of Health Sciences, Reykjavík, Iceland
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland School of Health Sciences, Reykjavík, Iceland
| | - Karl Andersen
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland School of Health Sciences, Reykjavík, Iceland
| |
Collapse
|
6
|
Paraskevaidis I, Kourek C, Tsougos E. Chronic Coronary Artery Disease: Wall Disease vs. Lumenopathy. Biomolecules 2025; 15:201. [PMID: 40001504 PMCID: PMC11852618 DOI: 10.3390/biom15020201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Acute and chronic coronary artery disease (CAD) are interconnected, representing two facets of the same condition. Chronic CAD exhibits a dynamic nature, manifesting as stable or acute ischemia, or both. Myocardial ischemia can be transient and reversible. The genesis of CAD involves diverse anatomical and functional mechanisms, including endothelial dysfunction, arteriolar remodeling, capillary rarefaction, and perivascular fibrosis, though no single factor explains its heterogeneity. Chronic CAD is often stable but may present as symptomatic or asymptomatic (e.g., in diabetes) and affect various coronary compartments (epicardial or microcirculation). This complexity necessitates a reappraisal of our approach, as pathophysiological mechanisms vary and often overlap. A comprehensive exploration of these mechanisms using advanced diagnostic techniques can aid in identifying the dynamic processes underlying CAD. The disease may present as obstructive or non-obstructive, stable or unstable, underscoring its diversity. The primary source of CAD lies in the arterial wall, emphasizing the need for research on its components, such as the endothelium and vascular smooth muscle cells, and factors disrupting arterial homeostasis. Shifting focus from arterial luminal status to the arterial wall can provide insights into the genesis of atheromatous plaques, enabling earlier interventions to prevent their development and progression.
Collapse
Affiliation(s)
- Ioannis Paraskevaidis
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece;
- Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece;
| | - Christos Kourek
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Elias Tsougos
- Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece;
| |
Collapse
|
7
|
Shen L, Tian Q, Ran Q, Gan Q, Hu Y, Du D, Qin Z, Duan X, Zhu X, Huang W. Z-Ligustilide: A Potential Therapeutic Agent for Atherosclerosis Complicating Cerebrovascular Disease. Biomolecules 2024; 14:1623. [PMID: 39766330 PMCID: PMC11726876 DOI: 10.3390/biom14121623] [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: 11/09/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Atherosclerosis (AS) is one of the major catalysts of ischemic cerebrovascular disease, and the death and disease burden from AS and its cerebrovascular complications are increasing. Z-ligustilide (Z-LIG) is a key active ingredient in Angelica sinensis (Oliv.) Diels and Ligusticum chuanxiong Hort. In this paper, we first introduced LIG's physicochemical properties and pharmacokinetics. Then, we reviewed Z-LIG's intervention and therapeutic mechanisms on AS and its cerebrovascular complications. The mechanisms of Z-LIG intervention in AS include improving lipid metabolism, antioxidant and anti-inflammatory effects, protecting vascular endothelium, and inhibiting vascular endothelial fibrosis, pathological thickening, and plaque calcification. In ischemic cerebrovascular diseases complicated by AS, Z-LIG exerts practical neuroprotective effects in ischemic stroke (IS), transient ischemic attack (TIA), and vascular dementia (VaD) through anti-neuroinflammatory, anti-oxidation, anti-neuronal apoptosis, protection of the blood-brain barrier, promotion of mitochondrial division and angiogenesis, improvement of cholinergic activity, inhibition of astrocyte proliferation, and endoplasmic reticulum stress. This paper aims to provide a basis for subsequent studies of Z-LIG in the prevention and treatment of AS and its cerebrovascular complications and, thus, to promote the development of interventional drugs for AS.
Collapse
Affiliation(s)
- Longyu Shen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Qianqian Tian
- Faculty of Social Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Qiqi Ran
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Qianrong Gan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Yu Hu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Donglian Du
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Zehua Qin
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Xinyi Duan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| | - Xinyun Zhu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
| | - Wei Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (L.S.); (Z.Q.)
| |
Collapse
|
8
|
Schulze-Späte U, Wurschi L, van der Vorst EPC, Hölzle F, Craveiro RB, Wolf M, Noels H. Crosstalk between periodontitis and cardiovascular risk. Front Immunol 2024; 15:1469077. [PMID: 39717783 PMCID: PMC11663742 DOI: 10.3389/fimmu.2024.1469077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/06/2024] [Indexed: 12/25/2024] Open
Abstract
Recent demographic developments resulted in an aged society with a rising disease burden of systemic and non-communicable diseases (NCDs). In cardiovascular disease (CVD), a NCD with high morbidity and mortality, recent preventive strategies include the investigation of comorbidities to reduce its significant economic burden. Periodontal disease, an oral bacterial-induced inflammatory disease of tooth-supporting tissue, is regulated in its prevalence and severity by the individual host response to a dysbiotic oral microbiota. Clinically, both NCDs are highly associated; however, shared risk factors such as smoking, obesity, type II diabetes mellitus and chronic stress represent only an insufficient explanation for the multifaceted interactions of both disease entities. Specifically, the crosstalk between both diseases is not yet fully understood. This review summarizes current knowledge on the clinical association of periodontitis and CVD, and elaborates on how periodontitis-induced pathophysiological mechanisms in patients may contribute to increased cardiovascular risk with focus on atherosclerosis. Clinical implications as well as current and future therapy considerations are discussed. Overall, this review supports novel scientific endeavors aiming at improving the quality of life with a comprehensive and integrated approach to improve well-being of the aging populations worldwide.
Collapse
Affiliation(s)
- Ulrike Schulze-Späte
- Section of Geriodontics, Department of Conservative Dentistry and Periodontics, University Hospital Jena, Jena, Germany
| | - Ludwig Wurschi
- Section of Geriodontics, Department of Conservative Dentistry and Periodontics, University Hospital Jena, Jena, Germany
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), Uniklinik RWTH Aachen, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Cardiorenal Research (AMICARE), Uniklinik RWTH Aachen, RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Frank Hölzle
- Department of Oral and Maxillofacial Surgery, School of Medicine, Uniklinik RWTH Aachen, Aachen, Germany
| | - Rogerio B. Craveiro
- Department of Orthodontics, Dental Clinic, Uniklinik RWTH Aachen, Aachen, Germany
| | - Michael Wolf
- Department of Orthodontics, Dental Clinic, Uniklinik RWTH Aachen, Aachen, Germany
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), Uniklinik RWTH Aachen, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Cardiorenal Research (AMICARE), Uniklinik RWTH Aachen, RWTH Aachen University, Aachen, Germany
- Biochemistry Department, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| |
Collapse
|
9
|
Simony SB, Langsted A, Mortensen MB, Nordestgaard BG, Afzal S. Statin use is associated with less ST-elevation versus non-ST-elevation myocardial infarction in a nationwide study. Atherosclerosis 2024; 399:118625. [PMID: 39437595 DOI: 10.1016/j.atherosclerosis.2024.118625] [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: 06/26/2024] [Revised: 09/05/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND AIMS Statin therapy reduces myocardial infarction rate but whether it is associated with a shift of ST-elevation myocardial infarction (STEMI) towards non-ST-elevation myocardial infarction (non-STEMI) remains unknown. Thus, we tested the hypothesis that statin use is associated with less STEMI relative to non-STEMI in first-time myocardial infarction. METHODS In a nationwide study, including 66,896 patients with first-time myocardial infarction between 2010 and 2021, we obtained multivariable risk estimates for STEMI versus non-STEMI according to any statin use, cumulated statin use, and daily statin dose. Furthermore, we obtained hazard ratios for 60-day mortality (5545 deaths) following myocardial infarction according to type of infarction. RESULTS Odds ratios for STEMI versus non-STEMI were 0.81 (95 % CI:0.77-0.85) and 1.07 (1.01-1.13) in current and previous statin users compared to never statin users. Cumulated statin exposure yielded odds ratios of 0.96 (0.87-1.07) for <2 statin-years, 0.87 (0.79-0.95) for 2-4.9 statin-years, 0.80 (0.74-0.87) for 5-10 statin-years, and 0.75 (0.70-0.80) for >10 statin-years compared to never users. Corresponding odds ratios for statin dose intensity were 0.89 (0.84-0.95) for low-intensity, 0.77 (0.73-0.82) for moderate-intensity, and 0.70 (0.63-0.77) for high-intensity. Results were similar in multiple sensitivity analyses and using a cohort design. The hazard ratio for 60-day mortality after first-time STEMI versus non-STEMI was 2.24 (2.13-2.37). CONCLUSIONS In this nationwide study, prior statin use is associated with less STEMI relative to non-STEMI in a dose dependent manner. This indicates that statin therapy, in addition to reducing myocardial infarction event rates, also result in a less severe presentation of myocardial infarctions.
Collapse
Affiliation(s)
- Sofie B Simony
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 73, Entrance 7 4th floor N5, DK-2730 Herlev, Denmark; Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anne Langsted
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 73, Entrance 7 4th floor N5, DK-2730 Herlev, Denmark; Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Martin B Mortensen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 73, Entrance 7 4th floor N5, DK-2730 Herlev, Denmark; Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Shoaib Afzal
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 73, Entrance 7 4th floor N5, DK-2730 Herlev, Denmark; Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
10
|
Wang X, Mu D, Liang J, Xin R, Zhang Y, Liu R, Yao M, Zhang B. Emerging nanoprobes for the features visualization of vulnerable atherosclerotic plaques. SMART MEDICINE 2024; 3:e20240033. [PMID: 39776593 PMCID: PMC11669784 DOI: 10.1002/smmd.20240033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/28/2024] [Indexed: 01/11/2025]
Abstract
Atherosclerosis (AS) is a major cause of cardiovascular disease. In particular, the unpredictable rupture of vulnerable atherosclerotic plaques (VASPs) can cause serious cardiovascular events such as myocardial infarction, stroke, and even sudden death. Therefore, early evaluation of the vulnerability of atherosclerotic plaques is of great importance. However, clinical imaging techniques are only marginally useful in the presence of severe anatomical structural changes, making it difficult to evaluate plaque vulnerability at an early stage. With the development of molecular imaging and nanotechnology, specific nanoprobes constructed for the pathological features of VASPs have attracted much attention for their ability to visualize VASPs early and noninvasively at the cellular and molecular levels. Here, we outline the pathological features of VASPs, analyze the superiority and limitations of current clinical imaging techniques, introduce the rational design principles of nanoprobes, and systematically summarize the application of nanoprobes to visualize the features of VASPs at the cellular and molecular levels. In addition, we discussed the prospects and urgent challenges in this field, and we believe it will provide new ideas for the early and accurate diagnosis of cardiovascular diseases.
Collapse
Affiliation(s)
- Xin Wang
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Dan Mu
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Jing Liang
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Ruijing Xin
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Yukun Zhang
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Renyuan Liu
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Mei Yao
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Bing Zhang
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
- Medical Imaging CenterAffiliated Drum Tower HospitalMedical School of Nanjing UniversityNanjingChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjingChina
- Department of RadiologyDrum Tower HospitalClinical College of Nanjing Medical UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular MedicineNanjingChina
- Institute of Brain ScienceNanjing UniversityNanjingChina
| |
Collapse
|
11
|
Panduga S, Vasishta S, Subramani R, Vincent S, Mutalik S, Joshi MB. Epidrugs in the clinical management of atherosclerosis: Mechanisms, challenges and promises. Eur J Pharmacol 2024; 980:176827. [PMID: 39038635 DOI: 10.1016/j.ejphar.2024.176827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/03/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Atherosclerosis is a complex and multigenic pathology associated with significant epigenetic reprogramming. Traditional factors (age, sex, obesity, hyperglycaemia, dyslipidaemia, hypertension) and non-traditional factors (foetal indices, microbiome alteration, clonal hematopoiesis, air pollution, sleep disorders) induce endothelial dysfunction, resulting in reduced vascular tone and increased vascular permeability, inflammation and shear stress. These factors induce paracrine and autocrine interactions between several cell types, including vascular smooth muscle cells, endothelial cells, monocytes/macrophages, dendritic cells and T cells. Such cellular interactions lead to tissue-specific epigenetic reprogramming regulated by DNA methylation, histone modifications and microRNAs, which manifests in atherosclerosis. Our review outlines epigenetic signatures during atherosclerosis, which are viewed as potential clinical biomarkers that may be adopted as new therapeutic targets. Additionally, we emphasize epigenetic modifiers referred to as 'epidrugs' as potential therapeutic molecules to correct gene expression patterns and restore vascular homeostasis during atherosclerosis. Further, we suggest nanomedicine-based strategies involving the use of epidrugs, which may selectively target cells in the atherosclerotic microenvironment and reduce off-target effects.
Collapse
Affiliation(s)
- Sushma Panduga
- Department of Biochemistry, Palamur Biosciences Private Limited, Hyderabad, 500026, Telangana, India; PhD Program, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Sampara Vasishta
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Ramamoorthy Subramani
- Department of Biochemistry, Palamur Biosciences Private Limited, Hyderabad, 500026, Telangana, India
| | - Sthevaan Vincent
- Department of Pathology, Palamur Biosciences Private Limited, Hyderabad, 500026, Telangana, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
12
|
Wang Y, Li Y, Lu Y, Li J. Biomimetic Nanoparticles for the Diagnosis and Therapy of Atherosclerosis. CHEM REC 2024; 24:e202400087. [PMID: 39148157 DOI: 10.1002/tcr.202400087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/23/2024] [Indexed: 08/17/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammation of blood vessels, which often has no obvious symptoms in the early stage of the disease, but when atherosclerotic plaques are formed, they often cause lumen blockage, and even plaque rupture leads to thrombosis, that is the essential factor of cardiovascular events, for example myocardial infarction, cerebral infarction, and renal atrophy. Therefore, it is considerably significant for the early recognition and precise therapy of plaque. Biomimetic nanoparticles (BNPs), especially those coated with cell membranes, can retain the biological function of cell membranes or cells, which has led to extensive research and application in the diagnosis and treatment of AS in recent years. In this review, we summarized the roles of various key cells in AS progression, the construction of biomimetic nanoparticles based on these key cells as well as their applications in AS diagnosis and therapy. Furthermore, we give a challenge and prospect of biomimetic nanoparticles in AS, hoping to elevate their application quality and the possibility of clinical translation.
Collapse
Affiliation(s)
- Yan Wang
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yize Li
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yuqing Lu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Jingjing Li
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China
| |
Collapse
|
13
|
Molinaro R, Sellar RS, Vromman A, Sausen G, Folco E, Sukhova GK, McConke ME, Corbo C, Ebert BL, Libby P. The clonal hematopoiesis mutation Jak2 V617F aggravates endothelial injury and thrombosis in arteries with erosion-like intimas. Int J Cardiol 2024; 409:132184. [PMID: 38759798 PMCID: PMC11753411 DOI: 10.1016/j.ijcard.2024.132184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/03/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Superficial plaque erosion causes many acute coronary syndromes. However, mechanisms of plaque erosion remain poorly understood, and we lack directed therapeutics for thrombotic complication. Human eroded plaques can harbor neutrophil extracellular traps (NETs) that propagate endothelial damage at experimental arterial lesions that recapitulate superficial erosion. Clonal Hematopoiesis of Indeterminate Potential (CHIP) denotes age-related clonal expansion of bone marrow-derived cells harboring somatic mutations in the absence of overt hematological disease. CHIP heightens the risk of cardiovascular disease, with the greatest increase seen in individuals with JAK2V617F. Neutrophils from mice and humans with JAK2V617F undergo NETosis more readily than Jak2WT (wild-type) cells. We hypothesized that JAK2V617F, by increasing propensity to NETosis, exacerbates aspects of superficial erosion. METHODS AND RESULTS We generated Jak2V617F and Jak2WT mice with heterozygous Jak2V617F in myeloid cells. We induced areas of denuded endothelium that recapitulate features of superficial erosion and assessed endothelial integrity, cellular composition of the erosion, thrombosis rates, and response to ruxolitinib, a clinically available JAK1/2 inhibitor, in relation to genotype. Following experimental erosion, Jak2V617F mice have greater impairment of endothelial barrier function and increased rates of arterial thrombosis. Neointimas in Jak2V617F mice exhibit increased apoptosis, NETosis, and platelet recruitment. Jak2V617F mice treated with ruxolitinib show increased endothelial continuity and reduced apoptosis in the neointima comparable to levels in Jak2WT. CONCLUSIONS These observations provide new mechanistic insight into the pathophysiology of superficial erosion, the heightened risk for myocardial infarction in JAK2V617F CHIP, and point the way to personalized therapeutics based on CHIP status.
Collapse
Affiliation(s)
- Roberto Molinaro
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Rob S Sellar
- Department of Haematology, UCL Cancer Institute, London, UK; Dana-Farber Cancer Institute, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Amélie Vromman
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Grasiele Sausen
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Eduardo Folco
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Galina K Sukhova
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Marie E McConke
- Dana-Farber Cancer Institute, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Claudia Corbo
- University of Milano-Bicocca, Department of Medicine and Surgery, NANOMIB Center, Monza 20900, Italy; IRCCS Istituto Ortopedico Galeazzi, Milan 20161, Italy
| | - Benjamin L Ebert
- Dana-Farber Cancer Institute, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA; Howard Hughes Medical Institute, Boston, MA, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
14
|
Bruoha S, Galli M, Sabouret P, Yosefy C, Taha L, Gragnano F, Savage MP, Shuvy M, Biondi-Zoccai G, Glikson M, Asher E. Atherosclerotic Plaque Erosion: Mechanisms, Clinical Implications, and Potential Therapeutic Strategies-A Review. J Cardiovasc Pharmacol 2024; 83:547-556. [PMID: 38421206 DOI: 10.1097/fjc.0000000000001554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
Atherosclerosis is an insidious and progressive inflammatory disease characterized by the formation of lipid-laden plaques within the intima of arterial walls with potentially devastating consequences. While rupture of vulnerable plaques has been extensively studied, a distinct mechanism known as plaque erosion (PE) has gained recognition and attention in recent years. PE, characterized by the loss of endothelial cell lining in the presence of intact fibrous cap, contributes to a significant and growing proportion of acute coronary events. However, despite a heterogeneous substrate underlying coronary thrombosis, treatment remains identical. This article provides an overview of atherosclerotic PE characteristics and its underlying mechanisms, highlights its clinical implications, and discusses potential therapeutic strategies.
Collapse
Affiliation(s)
- Sharon Bruoha
- Department of Cardiology, Barzilai Medical Center, the Ben-Gurion University of the Negev, Israel
| | - Mattia Galli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Pierre Sabouret
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
- National College of French Cardiologists, 13 rue Niepce, 75014 Paris, France
| | - Chaim Yosefy
- Department of Cardiology, Barzilai Medical Center, the Ben-Gurion University of the Negev, Israel
| | - Louay Taha
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. "Sant'Anna e San Sebastiano", Caserta, Italy
| | - Michael P Savage
- Division of Cardiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mony Shuvy
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy ; and
- Mediterranea Cardiocentro, Naples, Italy
| | - Michael Glikson
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Elad Asher
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| |
Collapse
|
15
|
Libby P. Inflammation and the pathogenesis of atherosclerosis. Vascul Pharmacol 2024; 154:107255. [PMID: 38157682 DOI: 10.1016/j.vph.2023.107255] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/02/2023] [Indexed: 01/03/2024]
Abstract
The notion that inflammation contributes to atherosclerosis has now gained considerable currency. Inflammation participates in atherosclerosis inception, progression, and thrombotic complications. Induced expression of endothelial leukocyte adhesion molecules and chemoattractant cytokines recruit blood cells to the arterial intima. Lesions can contain virtually every type of leukocyte. Monocytes mature into macrophages and imbibe lipids becoming foam cells, a hallmark of the atherosclerotic lesion. T lymphocytes can instruct the more numerous macrophages to express genes involved in the progression of the atheroma and its eventual destabilization. Inflammation is becoming clinically actionable to refine risk prediction, allocate treatments, and as a therapeutic target.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
16
|
Heusch G. Myocardial ischemia/reperfusion: Translational pathophysiology of ischemic heart disease. MED 2024; 5:10-31. [PMID: 38218174 DOI: 10.1016/j.medj.2023.12.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/27/2023] [Accepted: 12/12/2023] [Indexed: 01/15/2024]
Abstract
Ischemic heart disease is the greatest health burden and most frequent cause of death worldwide. Myocardial ischemia/reperfusion is the pathophysiological substrate of ischemic heart disease. Improvements in prevention and treatment of ischemic heart disease have reduced mortality in developed countries over the last decades, but further progress is now stagnant, and morbidity and mortality from ischemic heart disease in developing countries are increasing. Significant problems remain to be resolved and require a better pathophysiological understanding. The present review attempts to briefly summarize the state of the art in myocardial ischemia/reperfusion research, with a view on both its coronary vascular and myocardial aspects, and to define the cutting edges where further mechanistic knowledge is needed to facilitate translation to clinical practice.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
17
|
Park S, Yuki H, Niida T, Suzuki K, Kinoshita D, McNulty I, Broersen A, Dijkstra J, Lee H, Kakuta T, Ye JC, Jang IK. A novel deep learning model for a computed tomography diagnosis of coronary plaque erosion. Sci Rep 2023; 13:22992. [PMID: 38151502 PMCID: PMC10752868 DOI: 10.1038/s41598-023-50483-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023] Open
Abstract
Patients with acute coronary syndromes caused by plaque erosion might be managed conservatively without stenting. Currently, the diagnosis of plaque erosion requires an invasive imaging procedure. We sought to develop a deep learning (DL) model that enables an accurate diagnosis of plaque erosion using coronary computed tomography angiography (CTA). A total of 532 CTA scans from 395 patients were used to develop a DL model: 426 CTA scans from 316 patients for training and internal validation, and 106 separate scans from 79 patients for validation. Momentum Distillation-enhanced Composite Transformer Attention (MD-CTA), a novel DL model that can effectively process the entire set of CTA scans to diagnose plaque erosion, was developed. The novel DL model, compared to the convolution neural network, showed significantly improved AUC (0.899 [0.841-0.957] vs. 0.724 [0.622-0.826]), sensitivity (87.1 [70.2-96.4] vs. 71.0 [52.0-85.8]), and specificity (85.3 [75.3-92.4] vs. 68.0 [56.2-78.3]), respectively, for the patient-level prediction. Similar results were obtained at the slice-level prediction AUC (0.897 [0.890-0.904] vs. 0.757 [0.744-0.770]), sensitivity (82.2 [79.8-84.3] vs. 68.9 [66.2-71.6]), and specificity (80.1 [79.1-81.0] vs. 67.3 [66.3-68.4]), respectively. This newly developed DL model enables an accurate CT diagnosis of plaque erosion, which might enable cardiologists to provide tailored therapy without invasive procedures.Clinical Trial Registration: http://www.clinicaltrials.gov , NCT04523194.
Collapse
Affiliation(s)
- Sangjoon Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Haruhito Yuki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB 800, Boston, MA, 02114, USA
| | - Takayuki Niida
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB 800, Boston, MA, 02114, USA
| | - Keishi Suzuki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB 800, Boston, MA, 02114, USA
| | - Daisuke Kinoshita
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB 800, Boston, MA, 02114, USA
| | - Iris McNulty
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB 800, Boston, MA, 02114, USA
| | - Alexander Broersen
- Department of Radiology, Division of Image Processing, Leiden University Medical Center, Leiden, the Netherlands
| | - Jouke Dijkstra
- Department of Radiology, Division of Image Processing, Leiden University Medical Center, Leiden, the Netherlands
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA n, USA
| | - Tsunekazu Kakuta
- Department of Cardiology, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Jong Chul Ye
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
- Kim Jaechul Graduate School of Artificial Intelligence, Department of Mathematical Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-Ro, Daejeon, 34141, South Korea.
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB 800, Boston, MA, 02114, USA.
| |
Collapse
|
18
|
Noothi SK, Ahmed MR, Agrawal DK. Residual risks and evolving atherosclerotic plaques. Mol Cell Biochem 2023; 478:2629-2643. [PMID: 36897542 PMCID: PMC10627922 DOI: 10.1007/s11010-023-04689-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023]
Abstract
Atherosclerotic disease of the coronary and carotid arteries is the primary global cause of significant mortality and morbidity. The chronic occlusive diseases have changed the epidemiological landscape of health problems both in developed and the developing countries. Despite the enormous benefit of advanced revascularization techniques, use of statins, and successful attempts of targeting modifiable risk factors, like smoking and exercise in the last four decades, there is still a definite "residual risk" in the population, as evidenced by many prevalent and new cases every year. Here, we highlight the burden of the atherosclerotic diseases and provide substantial clinical evidence of the residual risks in these diseases despite advanced management settings, with emphasis on strokes and cardiovascular risks. We critically discussed the concepts and potential underlying mechanisms of the evolving atherosclerotic plaques in the coronary and carotid arteries. This has changed our understanding of the plaque biology, the progression of unstable vs stable plaques, and the evolution of plaque prior to the occurrence of a major adverse atherothrombotic event. This has been facilitated using intravascular ultrasound, optical coherence tomography, and near-infrared spectroscopy in the clinical settings to achieve surrogate end points. These techniques are now providing exquisite information on plaque size, composition, lipid volume, fibrous cap thickness and other features that were previously not possible with conventional angiography.
Collapse
Affiliation(s)
- Sunil K Noothi
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, USA
| | - Mohamed Radwan Ahmed
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, USA.
| |
Collapse
|
19
|
Zhou T, Zha M, Tang H, Li K, Jiang X. Controlling NIR-II emitting gold organic/inorganic nanohybrids with tunable morphology and surface PEG density for dynamic visualization of vascular dysfunction. Chem Sci 2023; 14:8842-8849. [PMID: 37621431 PMCID: PMC10445439 DOI: 10.1039/d3sc02290k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/16/2023] [Indexed: 08/26/2023] Open
Abstract
Luminescent Au nanoparticles (AuNPs) and their organic/inorganic nanohybrids are of interest due to their favorable properties and promising biomedical applications. However, most existing AuNP-based hybrid nanostructures cannot satisfy high efficiency in synthesis, deep tissue penetration, and long blood circulation simultaneously, thus cannot be employed in dynamic monitoring of biomedical applications. In this paper, using Pluronic F127 as a template, we report a robust approach for one-pot synthesis of AuNP-based organic/inorganic nanohybrids (AuNHs) with bright luminescence in the second near-infrared (NIR-II) window, tunable shape, and controllable surface polyethylene glycol (PEG) density. The nanohybrids could be controlled from a necklace-like shape with a dense brush PEG configuration to a spherical structure with a brush PEG coating, which greatly impacts the in vivo biological behavior. Compared to spherical AuNHs, the necklace-shaped AuNHs present a higher quantum yield and longer blood circulation, which are superior to most of the individual AuNPs. With these outstanding features, the necklace-shaped AuNHs could achieve real-time, dynamic visualization of vascular dysfunction, capable of directing the precise administration of thrombolytics (a medicine for the breakdown of blood clots). These findings could provide a powerful guide for designing novel NIR-II nanoprobes toward in vivo dynamic information visualization.
Collapse
Affiliation(s)
- Tingyao Zhou
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology No. 1088 Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
- Institute for Advanced Study, Shenzhen University No. 3688 Nanhai Avenue, Nanshan District Shenzhen Guangdong 518060 P. R. China
| | - Menglei Zha
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology No. 1088 Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| | - Hao Tang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology No. 1088 Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| | - Kai Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology No. 1088 Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| | - Xingyu Jiang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology No. 1088 Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| |
Collapse
|
20
|
Dabravolski SA, Sukhorukov VN, Melnichenko AA, Khotina VA, Orekhov AN. Potential Application of the Plant-Derived Essential Oils for Atherosclerosis Treatment: Molecular Mechanisms and Therapeutic Potential. Molecules 2023; 28:5673. [PMID: 37570643 PMCID: PMC10420188 DOI: 10.3390/molecules28155673] [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: 06/21/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Essential oils (EOs) are complex secondary metabolites identified in many plant species. Plant-derived EOs have been widely used in traditional medicine for centuries for their health-beneficial effects. Some EOs and their active ingredients have been reported to improve the cardiovascular system, in particular to provide an anti-atherosclerotic effect. The objective of this review is to highlight the recent research investigating the anti-inflammatory, anti-oxidative and lipid-lowering properties of plant-derived EOs and discuss their mechanisms of action. Also, recent clinical trials exploring anti-inflammatory and anti-oxidative activities of EOs are discussed. Future research on EOs has the potential to identify new bioactive compounds and invent new effective agents for treatment of atherosclerosis and related diseases such as diabetes, metabolic syndrome and obesity.
Collapse
Affiliation(s)
- Siarhei A. Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Snunit 51, P.O. Box 78, Karmiel 2161002, Israel
| | - Vasily N. Sukhorukov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia; (V.N.S.); (A.A.M.); (V.A.K.); (A.N.O.)
| | - Alexandra A. Melnichenko
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia; (V.N.S.); (A.A.M.); (V.A.K.); (A.N.O.)
| | - Victoria A. Khotina
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia; (V.N.S.); (A.A.M.); (V.A.K.); (A.N.O.)
| | - Alexander N. Orekhov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia; (V.N.S.); (A.A.M.); (V.A.K.); (A.N.O.)
| |
Collapse
|
21
|
Petkovic A, Erceg S, Munjas J, Ninic A, Vladimirov S, Davidovic A, Vukmirovic L, Milanov M, Cvijanovic D, Mitic T, Sopic M. LncRNAs as Regulators of Atherosclerotic Plaque Stability. Cells 2023; 12:1832. [PMID: 37508497 PMCID: PMC10378138 DOI: 10.3390/cells12141832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Current clinical data show that, despite constant efforts to develop novel therapies and clinical approaches, atherosclerotic cardiovascular diseases (ASCVD) are still one of the leading causes of death worldwide. Advanced and unstable atherosclerotic plaques most often trigger acute coronary events that can lead to fatal outcomes. However, despite the fact that different plaque phenotypes may require different treatments, current approaches to prognosis, diagnosis, and classification of acute coronary syndrome do not consider the diversity of plaque phenotypes. Long non-coding RNAs (lncRNAs) represent an important class of molecules that are implicated in epigenetic control of numerous cellular processes. Here we review the latest knowledge about lncRNAs' influence on plaque development and stability through regulation of immune response, lipid metabolism, extracellular matrix remodelling, endothelial cell function, and vascular smooth muscle function, with special emphasis on pro-atherogenic and anti-atherogenic lncRNA functions. In addition, we present current challenges in the research of lncRNAs' role in atherosclerosis and translation of the findings from animal models to humans. Finally, we present the directions for future lncRNA-oriented research, which may ultimately result in patient-oriented therapeutic strategies for ASCVD.
Collapse
Affiliation(s)
- Aleksa Petkovic
- Clinical-Hospital Centre "Dr Dragiša Mišović-Dedinje", 11000 Belgrade, Serbia
| | - Sanja Erceg
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| | - Jelena Munjas
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| | - Ana Ninic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| | - Sandra Vladimirov
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandar Davidovic
- Intern Clinic, Clinical Ward for Cardiovascular Diseases, Clinical-Hospital Centre Zvezdara, 11000 Belgrade, Serbia
- Department for Internal Medicine, Faculty of Dentistry, University of Belgrade, 11000 Belgrade, Serbia
| | - Luka Vukmirovic
- Intern Clinic, Clinical Ward for Cardiovascular Diseases, Clinical-Hospital Centre Zvezdara, 11000 Belgrade, Serbia
| | - Marko Milanov
- Intern Clinic, Clinical Ward for Cardiovascular Diseases, Clinical-Hospital Centre Zvezdara, 11000 Belgrade, Serbia
| | - Dane Cvijanovic
- Intern Clinic, Clinical Ward for Cardiovascular Diseases, Clinical-Hospital Centre Zvezdara, 11000 Belgrade, Serbia
| | - Tijana Mitic
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Miron Sopic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
22
|
DeFilippis AP, Lidani KCF, Nam Y, Trainor PJ, Johnson WC, Heckbert SR, McClelland RL, Blaha MJ, Nasir K. Risk factor associations with individual myocardial infarction subtypes and acute non-ischemic myocardial injury in the Multi-Ethnic Study of Atherosclerosis (MESA): Design and rationale. Am Heart J 2023; 260:151-173. [PMID: 36868395 PMCID: PMC10227529 DOI: 10.1016/j.ahj.2023.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/21/2023] [Accepted: 02/18/2023] [Indexed: 05/07/2023]
Abstract
BACKGROUND Despite different prevalence, pathobiology, and prognosis between etiologically distinct myocardial infarction (MI) subtypes, prospective study of risk factor for MI in large NHLBI-sponsored cardiovascular cohorts is limited to acute MI as a singular entity. Therefore, we sought to utilize the Multi-Ethnic Study of Atherosclerosis (MESA), a large prospective primary prevention cardiovascular study, to define the incidence and risk factor profile of individual myocardial injury subtypes. METHODS We describe the rationale and design of re-adjudicating 4,080 events that occurred over the first 14 years of follow-up in MESA for the presence and subtype of myocardial injury as defined by the Fourth Universal Definition of MI: MI type 1 to 5, acute non-ischemic myocardial injury, and chronic myocardial injury. The project utilizes a 2-physician adjudication process via examination of medical records, abstracted data collection forms, cardiac biomarker results, and electrocardiograms of all relevant clinical events. Comparison of the magnitude and direction of associations between baseline traditional and novel cardiovascular risk factors with incident and recurrent acute MI subtypes and acute non-ischemic myocardial injury events will be made. CONCLUSIONS This project will result in one of the first large prospective cardiovascular cohort with modern classification of acute MI subtypes, as well as a full accounting of non-ischemic myocardial injury events, with implications for numerous ongoing and future studies in MESA. By creating precise MI phenotypes, and defining their epidemiology, this project will allow for discovery of novel pathobiology-specific risk factors, allow for development of more accurate risk prediction, and suggest more targeted preventive strategies.
Collapse
Affiliation(s)
- Andrew P DeFilippis
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN.
| | - Karita C F Lidani
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Yunbi Nam
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA
| | - Patrick J Trainor
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM
| | - W Craig Johnson
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA
| | - Susan R Heckbert
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA
| | - Robyn L McClelland
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA
| | - Michael J Blaha
- Ciccarone Center for the Prevention of Heart Disease, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Khurram Nasir
- Division of Cardiovascular Prevention and Wellness, Department of Cardiology, Houston Methodist DeBakey Heart & Vascular Center, Houston, TX
| |
Collapse
|
23
|
Chen Y, Chen S, Han Y, Xu Q, Zhao X. Neutrophil-to-Lymphocyte Ratio and Platelet-to-Lymphocyte Ratio are Important Indicators for Predicting in-Hospital Death in Elderly AMI Patients. J Inflamm Res 2023; 16:2051-2061. [PMID: 37215380 PMCID: PMC10198281 DOI: 10.2147/jir.s411086] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/06/2023] [Indexed: 05/24/2023] Open
Abstract
Aim To investigate the role of neutrophil-to-lymphocyte ratio(NLR) and platelet-to-lymphocyte(PLR) in predicting the risk of in-hospital mortality in elderly acute myocardial infarction(AMI) patients. Methods This study was a single-center, retrospective and observational study. From December 2015 to December 2021, a total of 1550 elderly patients (age ≥ 60 years) with AMI with complete clinical history data were enrolled in the Second Hospital of Dalian Medical University. Routine blood tests were performed on admission, and NLR and PLR were calculated based on neutrophil, platelet, and lymphocyte counts. Outcome was defined as all-cause mortality during hospitalization. Cox regression and restricted spline cubic(RCS) models were used to evaluate the association of NLR and in-hospital mortality risk and the association of PLR with in-hospital mortality risk, respectively. Results (1) A total of 132 (8.5%) patients died during hospitalization. From the results of blood routine, the white blood cell, neutrophil, NLR and PLR in the death group were higher than those in the non-death group, while the lymphocyte was lower than that in the non-death group, and the difference was statistically significant (P < 0.05). (2) The results of receiver operating characteristic(ROC) curves analysis showed that the predictive ability of NLR (AUC = 0.790) for in-hospital death was better than that of PLR (AUC = 0.637). (3) Multivariate Cox proportional regression hazard models showed that high NLR was associated with the risk of in-hospital mortality in elderly AMI patients (HR = 3.091, 95% CI 2.097-4.557, P < 0.001), while high PLR was not. (4) RCS models showed a nonlinear dose-response relationship between NLR and in-hospital death (P for nonlinear = 0.0007). Conclusion High NLR (> 6.69) is associated with the risk of in-hospital mortality in elderly patients with AMI and can be an independent predictor of poor short-term prognosis in elderly patients with AMI.
Collapse
Affiliation(s)
- Yan Chen
- Department of Cardiology, the Second Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Shengyue Chen
- Department of Cardiology, the Second Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Yuanyuan Han
- Department of Cardiology, the Second Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Qing Xu
- Department of Cardiology, the Second Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Xin Zhao
- Department of Cardiology, the Second Hospital of Dalian Medical University, Dalian, People’s Republic of China
| |
Collapse
|
24
|
Lorenzatti D, Piña P, Csecs I, Schenone AL, Gongora CA, Garcia MJ, Blaha MJ, Budoff MJ, Williams MC, Dey D, Berman DS, Virani SS, Slipczuk L. Does Coronary Plaque Morphology Matter Beyond Plaque Burden? Curr Atheroscler Rep 2023; 25:167-180. [PMID: 36808390 DOI: 10.1007/s11883-023-01088-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2023] [Indexed: 02/23/2023]
Abstract
PURPOSE OF REVIEW Imaging of adverse coronary plaque features by coronary computed tomography angiography (CCTA) has advanced greatly and at a fast pace. We aim to describe the evolution, present and future in plaque analysis, and its value in comparison to plaque burden. RECENT FINDINGS Recently, it has been demonstrated that in addition to plaque burden, quantitative and qualitative assessment of coronary plaque by CCTA can improve the prediction of future major adverse cardiovascular events in diverse coronary artery disease scenarios. The detection of high-risk non-obstructive coronary plaque can lead to higher use of preventive medical therapies such as statins and aspirin, help identify culprit plaque, and differentiate between myocardial infarction types. Even more, over traditional plaque burden, plaque analysis including pericoronary inflammation can potentially be useful tools for tracking disease progression and response to medical therapy. The identification of the higher risk phenotypes with plaque burden, plaque characteristics, or ideally both can allow the allocation of targeted therapies and potentially monitor response. Further observational data are now required to investigate these key issues in diverse populations, followed by rigorous randomized controlled trials.
Collapse
Affiliation(s)
- Daniel Lorenzatti
- Cardiology Division, Montefiore Healthcare Network/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Pamela Piña
- Cardiology Division, Montefiore Healthcare Network/Albert Einstein College of Medicine, Bronx, NY, USA
- Cardiology Division, CEDIMAT Cardiovascular Center, Santo Domingo, Dominican Republic
| | - Ibolya Csecs
- Cardiology Division, Montefiore Healthcare Network/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aldo L Schenone
- Cardiology Division, Montefiore Healthcare Network/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Carlos A Gongora
- Cardiology Division, Montefiore Healthcare Network/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mario J Garcia
- Cardiology Division, Montefiore Healthcare Network/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael J Blaha
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Matthew J Budoff
- Department of Medicine, Lundquist Institute at Harbor UCLA Medical Center, Torrance, CA, USA
| | - Michelle C Williams
- BHF Centre of Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, Queen's Medical Research Institute University of Edinburgh, Edinburgh, UK
| | - Damini Dey
- Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daniel S Berman
- Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Salim S Virani
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Section of Cardiology, Department of Medicine, The Aga Khan University, Karachi, Pakistan
| | - Leandro Slipczuk
- Cardiology Division, Montefiore Healthcare Network/Albert Einstein College of Medicine, Bronx, NY, USA.
- Clinical Cardiology, Advanced Cardiac Imaging, CV Atherosclerosis and Lipid Disorder Center, Montefiore Health System, NewYork, USA.
| |
Collapse
|
25
|
Senders ML, Calcagno C, Tawakol A, Nahrendorf M, Mulder WJM, Fayad ZA. PET/MR imaging of inflammation in atherosclerosis. Nat Biomed Eng 2023; 7:202-220. [PMID: 36522465 DOI: 10.1038/s41551-022-00970-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 10/25/2022] [Indexed: 12/23/2022]
Abstract
Myocardial infarction, stroke, mental disorders, neurodegenerative processes, autoimmune diseases, cancer and the human immunodeficiency virus impact the haematopoietic system, which through immunity and inflammation may aggravate pre-existing atherosclerosis. The interplay between the haematopoietic system and its modulation of atherosclerosis has been studied by imaging the cardiovascular system and the activation of haematopoietic organs via scanners integrating positron emission tomography and resonance imaging (PET/MRI). In this Perspective, we review the applicability of integrated whole-body PET/MRI for the study of immune-mediated phenomena associated with haematopoietic activity and cardiovascular disease, and discuss the translational opportunities and challenges of the technology.
Collapse
Affiliation(s)
- Max L Senders
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Claudia Calcagno
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ahmed Tawakol
- Cardiology Division and Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Willem J M Mulder
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands.
- Laboratory of Chemical Biology, Department of Biochemical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
26
|
Fröbert O, Götberg M, Erlinge D, Akhtar Z, Christiansen EH, MacIntyre CR, Oldroyd KG, Motovska Z, Erglis A, Moer R, Hlinomaz O, Jakobsen L, Engstrøm T, Jensen LO, Fallesen CO, Jensen SE, Angerås O, Calais F, Kåregren A, Lauermann J, Mokhtari A, Nilsson J, Persson J, Stalby P, Islam AKMM, Rahman A, Malik F, Choudhury S, Collier T, Pocock SJ, Pernow J. Clinical impact of influenza vaccination after ST- and non-ST-segment elevation myocardial infarction - insights from the IAMI trial. Am Heart J 2023; 255:82-89. [PMID: 36279930 DOI: 10.1016/j.ahj.2022.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/23/2022] [Accepted: 10/01/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Influenza vaccination early after myocardial infarction (MI) improves prognosis but vaccine effectiveness may differ dependent on type of MI. METHODS A total of 2,571 participants were prospectively enrolled in the Influenza vaccination after myocardial infarction (IAMI) trial and randomly assigned to receive in-hospital inactivated influenza vaccine or saline placebo. The trial was conducted at 30 centers in eight countries from October 1, 2016 to March 1, 2020. Here we report vaccine effectiveness in the 2,467 participants with ST-segment elevation MI (STEMI, n = 1,348) or non-ST-segment elevation MI (NSTEMI, n = 1,119). The primary endpoint was the composite of all-cause death, MI, or stent thrombosis at 12 months. Cumulative incidence of the primary and key secondary endpoints by randomized treatment and NSTEMI/STEMI was estimated using the Kaplan-Meier method. Treatment effects were evaluated with formal interaction testing to assess for effect modification. RESULTS Baseline risk was higher in participants with NSTEMI. In the NSTEMI group the primary endpoint occurred in 6.5% of participants assigned to influenza vaccine and 10.5% assigned to placebo (hazard ratio [HR], 0.60; 95% CI, 0.39-0.91), compared to 4.1% assigned to influenza vaccine and 4.5% assigned to placebo in the STEMI group (HR, 0.90; 95% CI, 0.54-1.50, P = .237 for interaction). Similar findings were seen for the key secondary endpoints of all-cause death and cardiovascular death. The Kaplan-Meier risk difference in all-cause death at one year was more pronounced in participants with NSTEMI (NSTEMI: HR, 0.47; 95% CI 0.28-0.80, STEMI: HR, 0.86; 95% CI, 0.43-1.70, interaction P = .028). CONCLUSIONS The beneficial effect of influenza vaccination on adverse cardiovascular events may be enhanced in patients with NSTEMI compared to those with STEMI.
Collapse
Affiliation(s)
- Ole Fröbert
- Örebro University, Faculty of Health, Department of Cardiology, Örebro, Örebro, Sweden.
| | - Matthias Götberg
- Department of Cardiology, Skane University Hospital, Clinical Sciences, Lund University, Lund, Scania, Sweden
| | - David Erlinge
- Department of Cardiology, Skane University Hospital, Clinical Sciences, Lund University, Lund, Scania, Sweden
| | - Zubair Akhtar
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Dhaka, Bangladesh; The Kirby Institute, UNSW Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Chandini R MacIntyre
- The Kirby Institute, UNSW Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Keith G Oldroyd
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom and West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Glasgow, Scotland, United Kingdom
| | - Zuzana Motovska
- Cardiocenter, Third Faculty of Medicine, Charles University, Prague, Czech Republic and University Hospital Kralovske Vinohrady, Prague, Bohemia, Czech Republic
| | - Andrejs Erglis
- Pauls Stradins Clinical University Hospital, University of Latvia, Riga, Riga, Latvia
| | - Rasmus Moer
- LHL-sykehuset Gardermoen, Oslo, Ostiandet, Norway
| | - Ota Hlinomaz
- nternational clinical research center, St. Anne University Hospital and Masaryk University, Brno, South Moravian, Czech Republic
| | - Lars Jakobsen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Aarhus , Denmark
| | - Thomas Engstrøm
- Rigshospitalet, University of Copenhagen, Copenhagen, Copenhagen, Denmark
| | - Lisette O Jensen
- Department of Cardiology, Odense University Hospital, Odense, Odense, Denmark
| | | | - Svend E Jensen
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark and Department of Clinical Medicine, Aalborg University, Aalborg, Kommune, Denmark
| | - Oskar Angerås
- Sahlgrenska University Hospital, Gothenburg, Sweden and Institute of Medicine, Department of molecular and clinical medicine, Gothenburg University, Gothenburg, Västergötland , Sweden
| | - Fredrik Calais
- Örebro University, Faculty of Health, Department of Cardiology, Örebro, Örebro, Sweden
| | - Amra Kåregren
- Västmanlands sjukhus Västerås, Västerås, Västmanland, Sweden
| | - Jörg Lauermann
- Department of Cardiology, Jönköping, Region Jönköping County, and Department of Health, Medicine and Caring, Linköping University, Linköping, Östergötland, Sweden
| | - Arash Mokhtari
- Department of Cardiology, Skane University Hospital, Clinical Sciences, Lund University, Lund, Scania, Sweden
| | - Johan Nilsson
- Cardiology, Heart Centre, Department of Public Health and Clinical Medicine, Umeå University, Umea, Västerbotten län, Sweden
| | - Jonas Persson
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Karolinska Institutet, Danderyd University Hospital, Stockholm, Södermanland and Uppland, Sweden
| | - Per Stalby
- Department of Cardiology, Karlstad Central Hospital, Karlstad, Värmland, Sweden
| | - Abu K M M Islam
- National Institute of Cardiovascular Diseases, Sher-e-Bangla Nagar, Dhaka, Dhaka, Bangladesh
| | - Afzalur Rahman
- National Institute of Cardiovascular Diseases, Sher-e-Bangla Nagar, Dhaka, Dhaka, Bangladesh
| | - Fazila Malik
- National Heart Foundation Hospital & Research Institute, Dhaka, Dhaka, Bangladesh
| | - Sohel Choudhury
- National Heart Foundation Hospital & Research Institute, Dhaka, Dhaka, Bangladesh
| | - Timothy Collier
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, London, United Kingdom
| | - Stuart J Pocock
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, London, United Kingdom
| | - John Pernow
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Stockholm, Sweden
| |
Collapse
|
27
|
Wang KL, Meah MN, Bularga A, Singh T, Williams MC, Newby DE. Computed tomography coronary angiography in non-ST-segment elevation myocardial infarction. Br J Radiol 2022; 95:20220346. [PMID: 36017975 PMCID: PMC9733606 DOI: 10.1259/bjr.20220346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/15/2022] [Accepted: 08/04/2022] [Indexed: 11/05/2022] Open
Abstract
Electrocardiography and high-sensitivity cardiac troponin testing are routinely applied as the initial step for clinical evaluation of patients with suspected non-ST-segment elevation myocardial infarction. Once diagnosed, patients with non-ST-segment elevation myocardial infarction are commenced on antithrombotic and secondary preventative therapies before undergoing invasive coronary angiography to determine the strategy of coronary revascularisation. However, this clinical pathway is imperfect and can lead to challenges in the diagnosis, management, and clinical outcomes of these patients. Computed tomography coronary angiography (CTCA) has increasingly been utilised in the setting of patients with suspected non-ST-segment elevation myocardial infarction, where it has an important role in avoiding unnecessary invasive coronary angiography and reducing downstream non-invasive functional testing for myocardial ischaemia. CTCA is an excellent gatekeeper for the cardiac catheterisation laboratory. In addition, CTCA provides complementary information for patients with myocardial infarction in the absence of obstructive coronary artery disease and highlights alternative or incidental diagnoses for those with cardiac troponin elevation. However, the routine application of CTCA has yet to demonstrate an impact on subsequent major adverse cardiovascular events. There are several ongoing studies evaluating CTCA and its associated technologies that will define and potentially expand its application in patients with suspected or diagnosed non-ST-segment elevation myocardial infarction. We here review the current evidence relating to the clinical application of CTCA in patients with non-ST-segment elevation myocardial infarction and highlight the areas where CTCA is likely to have an increasing important role and impact for our patients.
Collapse
Affiliation(s)
| | - Mohammed N Meah
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Anda Bularga
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Trisha Singh
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Michelle C Williams
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - David E Newby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
28
|
Henein MY, Vancheri S, Longo G, Vancheri F. The Role of Inflammation in Cardiovascular Disease. Int J Mol Sci 2022; 23:12906. [PMID: 36361701 PMCID: PMC9658900 DOI: 10.3390/ijms232112906] [Citation(s) in RCA: 275] [Impact Index Per Article: 91.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/15/2022] [Accepted: 10/24/2022] [Indexed: 07/21/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease, in which the immune system has a prominent role in its development and progression. Inflammation-induced endothelial dysfunction results in an increased permeability to lipoproteins and their subendothelial accumulation, leukocyte recruitment, and platelets activation. Recruited monocytes differentiate into macrophages which develop pro- or anti-inflammatory properties according to their microenvironment. Atheroma progression or healing is determined by the balance between these functional phenotypes. Macrophages and smooth muscle cells secrete inflammatory cytokines including interleukins IL-1β, IL-12, and IL-6. Within the arterial wall, low-density lipoprotein cholesterol undergoes an oxidation. Additionally, triglyceride-rich lipoproteins and remnant lipoproteins exert pro-inflammatory effects. Macrophages catabolize the oxidized lipoproteins and coalesce into a lipid-rich necrotic core, encapsulated by a collagen fibrous cap, leading to the formation of fibro-atheroma. In the conditions of chronic inflammation, macrophages exert a catabolic effect on the fibrous cap, resulting in a thin-cap fibro-atheroma which makes the plaque vulnerable. However, their morphology may change over time, shifting from high-risk lesions to more stable calcified plaques. In addition to conventional cardiovascular risk factors, an exposure to acute and chronic psychological stress may increase the risk of cardiovascular disease through inflammation mediated by an increased sympathetic output which results in the release of inflammatory cytokines. Inflammation is also the link between ageing and cardiovascular disease through increased clones of leukocytes in peripheral blood. Anti-inflammatory interventions specifically blocking the cytokine pathways reduce the risk of myocardial infarction and stroke, although they increase the risk of infections.
Collapse
Affiliation(s)
- Michael Y. Henein
- Institute of Public Health and Clinical Medicine, Umea University, 90187 Umea, Sweden
- Institute of Environment & Health and Societies, Brunel University, Middlesex SW17 0RE, UK
- Molecular and Clinical Sciences Research Institute, St. George’s University, London UB8 3PH, UK
| | - Sergio Vancheri
- Interventional Neuroradiology Department, Besançon University Hospital, 25000 Besançon, France
| | - Giovanni Longo
- Cardiovascular and Interventional Department, S.Elia Hospital, 93100 Caltanissetta, Italy
| | - Federico Vancheri
- Department of Internal Medicine, S.Elia Hospital, 93100 Caltanissetta, Italy
| |
Collapse
|
29
|
Tateishi K, Saito Y, Yamazaki T, Kitahara H, Kobayashi Y. Impact of in-hospital versus ambulatory glucose variability on coronary plaque vulnerability in patients with coronary artery disease. DIABETES EPIDEMIOLOGY AND MANAGEMENT 2022; 8:100104. [DOI: 10.1016/j.deman.2022.100104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
30
|
Caselli C, Di Giorgi N, Ragusa R, Lorenzoni V, Smit J, El Mahdiui M, Buechel RR, Teresinska A, Pizzi MN, Roque A, Poddighe R, Knuuti J, Schütte M, Parodi O, Pelosi G, Scholte A, Rocchiccioli S, Neglia D. Association of MMP9 with adverse features of plaque progression and residual inflammatory risk in patients with chronic coronary syndrome (CCS). Vascul Pharmacol 2022; 146:107098. [PMID: 36100166 DOI: 10.1016/j.vph.2022.107098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/18/2022] [Accepted: 08/26/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND AIMS MMP-9 is a predictor of atherosclerotic plaque instability and adverse cardiovascular events, but longitudinal data on the association between MMP9 and coronary disease progression are lacking. This study is aimed at investigating whether MMP9 is associated with atherosclerotic plaque progression and the related molecular basis in stable patients with chronic coronary syndrome (CCS). METHODS MMP9 serum levels were measured in 157 CCS patients (58 ± 8 years of age; 66% male) undergoing coronary computed tomography angiography at baseline and after a follow up period of 6.5 ± 1.1 years to assess progression of Total, Fibrous, Fibro-fatty, Necrotic Core, and Dense Calcium plaque volumes (PV). Gene expression analysis was evaluated in whole blood using a transcriptomic approach by RNA-seq. RESULTS At multivariate analysis, serum MMP9 was associated with annual change of Total and Necrotic Core PV (Coefficient 3.205, SE 1.321, P = 0.017; 1.449, SE 0.690, P = 0.038, respectively), while MMP9 gene expression with Necrotic Core PV (Coefficient 70.559, SE 32.629, P = 0.034), independently from traditional cardiovascular risk factors, medications, and presence of obstructive CAD. After transcriptomic analysis, MMP9 expression was linked to expression of genes involved in the innate immunity. CONCLUSIONS Among CCS patients, MMP9 is an independent predictive marker of progression of adverse coronary plaques, possibly reflecting the activity of inflammatory pathways conditioning adverse plaque phenotypes. Thus, blood MMP9 might be used for the identification of patients with residual risk even with optimal management of classical cardiovascular risk factors who may derive the greatest benefit from targeted anti-inflammatory drugs.
Collapse
Affiliation(s)
- Chiara Caselli
- Institute of Clinical Physiology CNR, Via G. Moruzzi 1, Pisa, Italy.
| | | | - Rosetta Ragusa
- Institute of Clinical Physiology CNR, Via G. Moruzzi 1, Pisa, Italy.
| | - Valentina Lorenzoni
- Sant'Anna School of Advanced Studies, Piazza Martiri della Libertà, 33, Pisa, Italy.
| | - Jeff Smit
- Department of Cardiology, Heart Lung Center, Leiden University Medical Centre, Albinusdreef 2, RC, Leiden, the Netherlands.
| | - Mohammed El Mahdiui
- Department of Cardiology, Heart Lung Center, Leiden University Medical Centre, Albinusdreef 2, RC, Leiden, the Netherlands.
| | - Ronny R Buechel
- Department of Nuclear Medicine, Cardiac Imaging, University Hospital and University of Zurich, Switzerland.
| | | | - Maria N Pizzi
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Albert Roque
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | | | - Juhani Knuuti
- PET Center, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, Turku, Finland.
| | - Moritz Schütte
- Alacris Theranostics GmbH, Max-Planck-Straße 3, 12489 Berlin, Germany.
| | - Oberdan Parodi
- Fondazione Toscana G. Monasterio, Via G. Moruzzi 1, Pisa, Italy
| | - Gualtiero Pelosi
- Institute of Clinical Physiology CNR, Via G. Moruzzi 1, Pisa, Italy.
| | - Arthur Scholte
- Department of Cardiology, Heart Lung Center, Leiden University Medical Centre, Albinusdreef 2, RC, Leiden, the Netherlands.
| | | | - Danilo Neglia
- Fondazione Toscana G. Monasterio, Via G. Moruzzi 1, Pisa, Italy.
| | | |
Collapse
|
31
|
Tian W, Zhang T, Wang X, Zhang J, Ju J, Xu H. Global research trends in atherosclerosis: A bibliometric and visualized study. Front Cardiovasc Med 2022; 9:956482. [PMID: 36082127 PMCID: PMC9445883 DOI: 10.3389/fcvm.2022.956482] [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: 05/30/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIncreasing evidence has spurred a considerable evolution of concepts related to atherosclerosis, prompting the need to provide a comprehensive view of the growing literature. By retrieving publications in the Web of Science Core Collection (WoSCC) of Clarivate Analytics, we conducted a bibliometric analysis of the scientific literature on atherosclerosis to describe the research landscape.MethodsA search was conducted of the WoSCC for articles and reviews serving exclusively as a source of information on atherosclerosis published between 2012 and 2022. Microsoft Excel 2019 was used to chart the annual productivity of research relevant to atherosclerosis. Through CiteSpace and VOSviewer, the most prolific countries or regions, authors, journals, and resource-, intellectual-, and knowledge-sharing in atherosclerosis research, as well as co-citation analysis of references and keywords, were analyzed.ResultsA total of 20,014 publications were retrieved. In terms of publications, the United States remains the most productive country (6,390, 31,93%). The most publications have been contributed by Johns Hopkins Univ (730, 3.65%). ALVARO ALONSO produced the most published works (171, 0.85%). With a betweenness centrality of 0.17, ERIN D MICHOS was the most influential author. The most prolific journal was identified as Atherosclerosis (893, 4.46%). Circulation received the most co-citations (14,939, 2.79%). Keywords with the ongoing strong citation bursts were “nucleotide-binding oligomerization (NOD), Leucine-rich repeat (LRR)-containing protein (NLRP3) inflammasome,” “short-chain fatty acids (SCFAs),” “exosome,” and “homeostasis,” etc.ConclusionThe research on atherosclerosis is driven mostly by North America and Europe. Intensive research has focused on the link between inflammation and atherosclerosis, as well as its complications. Specifically, the NLRP3 inflammasome, interleukin-1β, gut microbiota and SCFAs, exosome, long non-coding RNAs, autophagy, and cellular senescence were described to be hot issues in the field.
Collapse
Affiliation(s)
- Wende Tian
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tai Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Gastroenterology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyi Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jianqing Ju
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jianqing Ju,
| | - Hao Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Hao Xu,
| |
Collapse
|
32
|
Cao Z, Yuan G, Zeng L, Bai L, Liu X, Wu M, Sun R, Chen Z, Jiang Y, Gao Q, Chen Y, Zhang Y, Pan Y, Wang J. Macrophage-Targeted Sonodynamic/Photothermal Synergistic Therapy for Preventing Atherosclerotic Plaque Progression Using CuS/TiO 2 Heterostructured Nanosheets. ACS NANO 2022; 16:10608-10622. [PMID: 35759554 DOI: 10.1021/acsnano.2c02177] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Sonodynamic therapy (SDT) and photothermal therapy (PTT) are two effective strategies for the treatment of atherosclerotic plaques. However, the low yield of reactive oxygen species (ROS) of conventional organic sonosensitizers and the low biosafety of hyperthermia limit the therapeutic efficacy of SDT and PTT. Herein, we report copper sulfide/titanium oxide heterostructure nanosheets modified with hyaluronic acid (HA) and PEG (HA-HNSs) for low-intensity sonodynamic and mild-photothermal synergistic therapy for early atherosclerotic plaques. CuS/TiO2 heterostructure nanosheets (HNSs) show high electron-hole separation efficiency and superior sonodynamic performance, because it has high surface energy crystal facets as well as a narrow band. Moreover, HNSs exhibit intense absorbance in the NIR-II region, which endows the nanosheets with excellent photothermal performance. With a further modification of HA, HA-HNSs can selectively target intraplaque proinflammatory macrophages through CD44-HA interaction. Because SDT reduces the expression of heat shock protein 90 and PTT facilitates the sonocatalytic process, the combination of SDT and PTT based on HA-HNSs could synergistically induce proinflammatory macrophage apoptosis. More importantly, the synergistic therapy prevents the progression of early atherosclerotic plaque by removing lesional macrophages and mitigating inflammation. Taken together, this work provides a macrophage-targeting sonodynamic/photothermal synergistic therapy, which is an effective translational clinical intervention for early atherosclerotic plaques.
Collapse
|
33
|
Semaglutide treatment attenuates vessel remodelling in ApoE-/- mice following vascular injury and blood flow perturbation. ATHEROSCLEROSIS PLUS 2022; 49:32-41. [PMID: 36644202 PMCID: PMC9833261 DOI: 10.1016/j.athplu.2022.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 01/18/2023]
Abstract
Background and aims Randomized clinical studies have shown a reduction in cardiovascular outcomes with glucagon-like peptide 1 receptor agonist (GLP-1RA) treatment with the hypothesized mechanisms being an underlying effect on atherosclerosis. Here, we aimed to assess the pharmacological effects of semaglutide in an atheroprone murine model that recapitulates central mechanisms related to vascular smooth muscle cell (VSMC) phenotypic switching and endothelial dysfunction known to operate within the atherosclerotic plaque. Methods In study A, we employed an electrical current to the carotid artery in ApoE-/- mice to induce severe VSMC injury and death, after which the arteries were allowed to heal for 4 weeks. In study B, a constrictive cuff was added for 6 h at the site of the healed segment to induce a disturbance in blood flow. Results Compared to vehicle, semaglutide treatment reduced the intimal and medial area by ∼66% (p = 0.007) and ∼11% (p = 0.0002), respectively. Following cuff placement, expression of the pro-inflammatory marker osteopontin and macrophage marker Mac-2 was reduced (p < 0.05) in the semaglutide-treated group compared to vehicle. GLP-1R were not expressed in murine carotid artery and human coronary vessels with and without atherosclerotic plaques, and semaglutide treatment did not affect proliferation of cultured primary human VSMCs. Conclusions Semaglutide treatment reduced vessel remodelling following electrical injury and blood flow perturbation in an atheroprone mouse model. This effect appears to be driven by anti-inflammatory and -proliferative mechanisms independent of GLP-1 receptor-mediated signalling in the resident vascular cells. This mechanism of action may be important for cardiovascular protection.
Collapse
|
34
|
Shishikura D, Octavia Y, Hayat U, Thondapu V, Barlis P. Atherogenesis and Inflammation. Interv Cardiol 2022. [DOI: 10.1002/9781119697367.ch1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
35
|
Endothelial Dysfunction Induced by Extracellular Neutrophil Traps Plays Important Role in the Occurrence and Treatment of Extracellular Neutrophil Traps-Related Disease. Int J Mol Sci 2022; 23:ijms23105626. [PMID: 35628437 PMCID: PMC9147606 DOI: 10.3390/ijms23105626] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 01/27/2023] Open
Abstract
Many articles have demonstrated that extracellular neutrophil traps (NETs) are often described as part of the antibacterial function. However, since the components of NETs are non-specific, excessive NETs usually cause inflammation and tissue damage. Endothelial dysfunction (ED) caused by NETs is the major focus of tissue damage, which is highly related to many inflammatory diseases. Therefore, this review summarizes the latest advances in the primary and secondary mechanisms between NETs and ED regarding inflammation as a mediator. Moreover, the detailed molecular mechanisms with emphasis on the disadvantages from NETs are elaborated: NETs can use its own enzymes, release particles as damage-associated molecular patterns (DAMPs) and activate the complement system to interact with endothelial cells (ECs), drive ECs damage and eventually aggravate inflammation. In view of the role of NETs-induced ED in different diseases, we also discussed possible molecular mechanisms and the treatments of NETs-related diseases.
Collapse
|
36
|
Li Q, Ma X, Shao Q, Yang Z, Wang Y, Gao F, Zhou Y, Yang L, Wang Z. Prognostic Impact of Multiple Lymphocyte-Based Inflammatory Indices in Acute Coronary Syndrome Patients. Front Cardiovasc Med 2022; 9:811790. [PMID: 35592392 PMCID: PMC9110784 DOI: 10.3389/fcvm.2022.811790] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/18/2022] [Indexed: 12/14/2022] Open
Abstract
Background The aim of this study was to evaluate the prognostic values of five lymphocyte-based inflammatory indices (platelet-lymphocyte ratio [PLR], neutrophil-lymphocyte ratio [NLR], monocyte-lymphocyte ratio [MLR], systemic immune inflammation index [SII], and system inflammation response index [SIRI]) in patients with acute coronary syndrome (ACS). Methods A total of 1,701 ACS patients who underwent percutaneous coronary intervention (PCI) were included in this study and followed up for major adverse cardiovascular events (MACE) including all-cause death, non-fatal ischemic stroke, and non-fatal myocardial infarction. The five indices were stratified by the optimal cutoff value for comparison. The association between each of the lymphocyte-based inflammatory indices and MACE was assessed by the Cox proportional hazards regression analysis. Results During the median follow-up of 30 months, 107 (6.3%) MACE were identified. The multivariate COX analysis showed that all five indices were independent predictors of MACE, and SIRI seemingly performed best (Hazard ratio [HR]: 3.847; 95% confidence interval [CI]: [2.623–5.641]; p < 0.001; C-statistic: 0.794 [0.731–0.856]). The addition of NLR, MLR, SII, or SIRI to the Global Registry of Acute Coronary Events (GRACE) risk score, especially SIRI (C-statistic: 0.699 [0.646–0.753], p < 0.001; net reclassification improvement [NRI]: 0.311 [0.209–0.407], p < 0.001; integrated discrimination improvement [IDI]: 0.024 [0.010–0.046], p < 0.001), outperformed the GRACE risk score alone in the risk predictive performance. Conclusion Lymphocyte-based inflammatory indices were significantly and independently associated with MACE in ACS patients who underwent PCI. SIRI seemed to be better than the other four indices in predicting MACE, and the combination of SIRI with the GRACE risk score could predict MACE more accurately.
Collapse
|
37
|
Toczek J, Riou L. Considerations on PET/MR imaging of carotid plaque inflammation with 68Ga-Pentixafor. J Nucl Cardiol 2022; 29:503-505. [PMID: 32914318 DOI: 10.1007/s12350-020-02354-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Jakub Toczek
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Laurent Riou
- Laboratoire Radiopharmaceutiques Biocliniques, Faculté de Médecine de Grenoble, UMR UGA - INSERM U1039, Grenoble, France.
| |
Collapse
|
38
|
Pasterkamp G, den Ruijter HM, Giannarelli C. False Utopia of One Unifying Description of the Vulnerable Atherosclerotic Plaque: A Call for Recalibration That Appreciates the Diversity of Mechanisms Leading to Atherosclerotic Disease. Arterioscler Thromb Vasc Biol 2022; 42:e86-e95. [PMID: 35139657 DOI: 10.1161/atvbaha.121.316693] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Atherosclerosis is a complex disease characterized by the formation of arterial plaques with a broad diversity of morphological phenotypic presentations. Researchers often apply one description of the vulnerable plaque as a gold standard in preclinical and clinical research that could be applied as a surrogate measure of a successful therapeutic intervention, despite the variability in lesion characteristics that may underly a thrombotic occlusion. The complex mechanistic interplay underlying progression of atherosclerotic disease is a consequence of the broad range of determinants such as sex, risk factors, hemodynamics, medications, and the genetic landscape. Currently, we are facing an overwhelming amount of data based on genetic, transcriptomic, proteomic, and metabolomic studies that all point to heterogeneous molecular profiles of atherosclerotic lesions that lead to a myocardial infarction or stroke. The observed molecular diversity implies that one unifying model cannot fully recapitulate the natural history of atherosclerosis. Despite emerging data obtained from -omics studies, a description of a natural history of atherosclerotic disease in which cell-specific expression of proteins or genes are included is still lacking. This also applies to the insights provided by genome-wide association studies. This review will critically discuss the dogma that the progression of atherosclerotic disease can be captured in one unifying natural history model of atherosclerosis.
Collapse
Affiliation(s)
- Gerard Pasterkamp
- Circulatory Health Laboratories (G.P., H.M.d.R.), University Medical Center Utrecht, the Netherlands.,Central Diagnostics Laboratories (G.P.), University Medical Center Utrecht, the Netherlands
| | - Hester M den Ruijter
- Circulatory Health Laboratories (G.P., H.M.d.R.), University Medical Center Utrecht, the Netherlands.,Laboratory of Experimental Cardiology (H.M.d.R.), University Medical Center Utrecht, the Netherlands
| | - Chiara Giannarelli
- NYU Cardiovascular Research Center (C.G.), New York University Grossman School of Medicine.,Department of Pathology (C.G.), New York University Grossman School of Medicine
| |
Collapse
|
39
|
Tateishi K, Saito Y, Kitahara H, Kobayashi Y. Relation of glucose variability to vulnerable plaque formation in patients with coronary artery disease. Heart Vessels 2022; 37:1516-1525. [PMID: 35353201 DOI: 10.1007/s00380-022-02063-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/18/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Although glucose variability (GV) is reportedly associated with coronary plaque vulnerability, namely lipid-rich plaque, details are not fully understood. The aim of this study was to evaluate relations of GV after discharge to vulnerable plaque formation assessed by near-infrared spectroscopy intravascular ultrasound (NIRS-IVUS) in patients with and without diabetes. METHODS A total of 40 patients undergoing percutaneous coronary intervention under NIRS-IVUS guidance were included, among whom 13 (33%) had diabetes and 20 (50%) presented with acute myocardial infarction (MI). GV was evaluated by a flush glucose monitoring system, primarily with mean amplitude of glycemic excursion (MAGE). Lipid-rich plaque was assessed by maximum lipid core burden index in 4 mm (maxLCBI4mm) in the target lesion using NIRS-IVUS. RESULTS Mean MAGE and maxLCBI4mm were 69.7 ± 25.6 mg/dl and 508.0 ± 294.9. Intra-day GV was not significantly associated with maxLCBI4mm in the entire study population, while MAGE was correlated with maxLCBI4mm in non-diabetic patients (r = 0.46, p = 0.02). In patients with and without acute MI presentation, no significant relations were found between MAGE and maxLCBI4mm. CONCLUSION GV was associated with lipid core plaque formation, especially in non-diabetic patients.
Collapse
Affiliation(s)
- Kazuya Tateishi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677, Japan.
| | - Yuichi Saito
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677, Japan
| | - Hideki Kitahara
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677, Japan
| |
Collapse
|
40
|
Ren F, Yuan Q, Han M, Jiang Z, Zhu H, Yun B, Li Z. In vivo static and dynamic angiography of thrombosis by using multi-functional lanthanide nanoprobes. Sci Bull (Beijing) 2022; 67:461-465. [PMID: 36546164 DOI: 10.1016/j.scib.2021.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/13/2021] [Accepted: 10/20/2021] [Indexed: 01/06/2023]
Affiliation(s)
- Feng Ren
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Qiang Yuan
- The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Mengxiao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Zhilin Jiang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Hongqin Zhu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Baofeng Yun
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China.
| |
Collapse
|
41
|
Jiang Z, Xia B, Ren F, Bao B, Xing W, He T, Li Z. Boosting Vascular Imaging-Performance and Systemic Biosafety of Ultra-Small NaGdF 4 Nanoparticles via Surface Engineering with Rationally Designed Novel Hydrophilic Block Co-Polymer. SMALL METHODS 2022; 6:e2101145. [PMID: 35107219 DOI: 10.1002/smtd.202101145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/03/2021] [Indexed: 06/14/2023]
Abstract
Revealing the anatomical structures, functions, and distribution of vasculature via contrast agent (CA) enhanced magnetic resonance imaging (MRI) is crucial for precise medical diagnosis and therapy. The clinically used MRI CAs strongly rely on Gd-chelates, which exhibit low T1 relaxivities and high risks of nephrogenic systemic fibrosis (NSF) for patients with renal dysfunction. It is extremely important to develop high-performance and safe CAs for MRI. Herein, it is reported that ultra-small NaGdF4 nanoparticles (UGNs) can serve as an excellent safe MRI CA via surface engineering with rationally designed novel hydrophilic block co-polymer (BPn ). By optimizing the polymer molecular weights, the polymer-functionalized UGNs (i.e., UGNs-BP14 ) are obtained to exhibit remarkably higher relaxivity (11.8 mm-1 s-1 at 3.0 T) than Gd-DTPA (3.6 mm-1 s-1 ) due to their ultracompact and abundant hydrophilic surface coating. The high performance of UGNs-BP14 enables us to sensitively visualize microvasculature with a small diameter of ≈0.17 mm for up to 2 h, which is the thinnest blood vessel and the longest time window for low field (1.0 T) MR angiography ever reported, and cannot be achieved by using the clinically used Gd-DTPA under the same conditions. More importantly, renal clearable UGNs-BP14 show lower risks of inducing NSF in comparison with Gd-DTPA due to their negligible release of Gd3+ ions after modification with the novel hydrophilic block copolymer. The study presents a novel avenue for boosting imaging-performance and systemic biosafety of UGNs as a robust MRI CA with great potential in precise diagnosis of vasculature-related diseases.
Collapse
Affiliation(s)
- Zhilin Jiang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Bin Xia
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Hefei University of Technology, Hefei, Anhui, 230009, P. R. China
| | - Feng Ren
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| | - Bolin Bao
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
- Department of Radiology, Affiliated Hospital 3, Soochow University, Changzhou, 213003, P. R. China
| | - Wei Xing
- Department of Radiology, Affiliated Hospital 3, Soochow University, Changzhou, 213003, P. R. China
| | - Tao He
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Hefei University of Technology, Hefei, Anhui, 230009, P. R. China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
42
|
Canu M, Broisat A, Riou L, Vanzetto G, Fagret D, Ghezzi C, Djaileb L, Barone-Rochette G. Non-invasive Multimodality Imaging of Coronary Vulnerable Patient. Front Cardiovasc Med 2022; 9:836473. [PMID: 35282382 PMCID: PMC8907666 DOI: 10.3389/fcvm.2022.836473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/01/2022] [Indexed: 01/07/2023] Open
Abstract
Atherosclerotic plaque rupture or erosion remain the primary mechanism responsible for myocardial infarction and the major challenge of cardiovascular researchers is to develop non-invasive methods of accurate risk prediction to identify vulnerable plaques before the event occurs. Multimodal imaging, by CT-TEP or CT-SPECT, provides both morphological and activity information about the plaque and cumulates the advantages of anatomic and molecular imaging to identify vulnerability features among coronary plaques. However, the rate of acute coronary syndromes remains low and the mechanisms leading to adverse events are clearly more complex than initially assumed. Indeed, recent studies suggest that the detection of a state of vulnerability in a patient is more important than the detection of individual sites of vulnerability as a target of focal treatment. Despite this evolution of concepts, multimodal imaging offers a strong potential to assess patient's vulnerability. Here we review the current state of multimodal imaging to identify vulnerable patients, and then focus on emerging imaging techniques and precision medicine.
Collapse
Affiliation(s)
- Marjorie Canu
- Department of Cardiology, University Hospital, Grenoble Alpes, Grenoble, France
| | - Alexis Broisat
- Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, LRB, Grenoble, France
| | - Laurent Riou
- Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, LRB, Grenoble, France
| | - Gerald Vanzetto
- Department of Cardiology, University Hospital, Grenoble Alpes, Grenoble, France
- Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, LRB, Grenoble, France
- French Alliance Clinical Trial, French Clinical Research Infrastructure Network, Paris, France
| | - Daniel Fagret
- Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, LRB, Grenoble, France
- Department of Nuclear Medicine, University Hospital, Grenoble Alpes, Grenoble, France
| | - Catherine Ghezzi
- Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, LRB, Grenoble, France
| | - Loic Djaileb
- Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, LRB, Grenoble, France
- Department of Nuclear Medicine, University Hospital, Grenoble Alpes, Grenoble, France
| | - Gilles Barone-Rochette
- Department of Cardiology, University Hospital, Grenoble Alpes, Grenoble, France
- Univ. Grenoble Alpes, INSERM, CHU Grenoble Alpes, LRB, Grenoble, France
- French Alliance Clinical Trial, French Clinical Research Infrastructure Network, Paris, France
- *Correspondence: Gilles Barone-Rochette
| |
Collapse
|
43
|
Vinci R, Pedicino D, Bonanni A, d'Aiello A, Pisano E, Ponzo M, Severino A, Ciampi P, Canonico F, Russo G, Di Sario M, Vergallo R, Filomia S, Montone RA, Flego D, Stefanini L, Piacentini R, Conte C, Cribari F, Massetti M, Crea F, Liuzzo G. Monocyte-Platelet Aggregates Triggered by CD31 Molecule in Non-ST Elevation Myocardial Infarction: Clinical Implications in Plaque Rupture. Front Cardiovasc Med 2022; 8:741221. [PMID: 35146002 PMCID: PMC8821091 DOI: 10.3389/fcvm.2021.741221] [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: 07/14/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Despite the recent innovations in cardiovascular care, atherothrombosis is still a major complication of acute coronary syndromes (ACS). We evaluated the involvement of the CD31 molecule in thrombotic risk through the formation of monocyte-platelet (Mo-Plt) aggregates in patients with ACS with no-ST-segment elevation myocardial infarction (NSTEMI) on top of dual anti-platelet therapy (DAPT). We enrolled 19 control (CTRL) subjects, 46 stable angina (SA), and 86 patients with NSTEMI, of which, 16 with Intact Fibrous Cap (IFC) and 19 with Ruptured Fibrous Cap (RFC) as assessed by the Optical Coherence Tomography (OCT). The expression of CD31 on monocytes and platelets was measured. Following the coronary angiography, 52 NSTEMIs were further stratified according to thrombus grade (TG) evaluation. Finally, a series of ex vivo experiments verified whether the CD31 participates in Mo-Plt aggregate formation. In patients with NSTEMI, CD31 was reduced on monocytes and was increased on platelets, especially in NSTEMI presented with RFC plaques compared to those with IFC lesions, and in patients with high TG compared to those with zero/low TG. Ex vivo experiments documented an increase in Mo-Plt aggregates among NSTEMI, which significantly decreased after the CD31 ligation, particularly in patients with RFC plaques. In NSTEMI, CD31 participates in Mo-Plt aggregate formation in spite of optimal therapy and DAPT, suggesting the existence of alternative thrombotic pathways, as predominantly displayed in patients with RFC.
Collapse
Affiliation(s)
- Ramona Vinci
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Daniela Pedicino
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- *Correspondence: Daniela Pedicino
| | - Alice Bonanni
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alessia d'Aiello
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Eugenia Pisano
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Myriana Ponzo
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Severino
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Pellegrino Ciampi
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Canonico
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giulio Russo
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marianna Di Sario
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rocco Vergallo
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Simone Filomia
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rocco Antonio Montone
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Davide Flego
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Lucia Stefanini
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Roberto Piacentini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cristina Conte
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Cribari
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Massimo Massetti
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Filippo Crea
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanna Liuzzo
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
44
|
Zhao X, Zhang F, Lei Z. The pursuit of polymethine fluorophores with NIR-II emission and high brightness for in vivo applications. Chem Sci 2022; 13:11280-11293. [PMID: 36320587 PMCID: PMC9533410 DOI: 10.1039/d2sc03136a] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/25/2022] [Indexed: 11/21/2022] Open
Abstract
Polymethine cyanine dyes, as the most important class of organic near-infrared-II (NIR-II) fluorophores, recently received increasing attention due to their high molar extinction coefficients, intensive fluorescence brightness, and flexible wavelength tunability for fluorescent bioimaging applications. Very recently, remarkable advances have been made in the development of NIR-II polymethine fluorophores with improved optical performance, mainly including tunable fluorescence, improved brightness, improved water solubility and stability. In this review, we summarize the recent research advances in molecular tailoring design strategies of NIR-II polymethine fluorophores, and then emphasize the representative bioimaging and biosensing applications. The potential challenges and perspectives of NIR-II polymethine fluorophores in this emerging field are also discussed. This review may provide guidance and reference for further development of high-performance NIR-II polymethine fluorophores to boost their clinical translation in the future. Overview of historical development for polymethine fluorophores with NIR-II emission and high brightness for in vivo applications.![]()
Collapse
Affiliation(s)
- Xuan Zhao
- Minhang Hospital and Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai 200433, China
| | - Fan Zhang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai 200433, China
| | - Zuhai Lei
- Minhang Hospital and Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
45
|
Mao L, Yin R, Yang L, Zhao D. Role of advanced glycation end products on vascular smooth muscle cells under diabetic atherosclerosis. Front Endocrinol (Lausanne) 2022; 13:983723. [PMID: 36120471 PMCID: PMC9470882 DOI: 10.3389/fendo.2022.983723] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease and leading cause of cardiovascular diseases. The progression of AS is a multi-step process leading to high morbidity and mortality. Hyperglycemia, dyslipidemia, advanced glycation end products (AGEs), inflammation and insulin resistance which strictly involved in diabetes are closely related to the pathogenesis of AS. A growing number of studies have linked AGEs to AS. As one of the risk factors of cardiac metabolic diseases, dysfunction of VSMCs plays an important role in AS pathogenesis. AGEs are increased in diabetes, participate in the occurrence and progression of AS through multiple molecular mechanisms of vascular cell injury. As the main functional cells of vascular, vascular smooth muscle cells (VSMCs) play different roles in each stage of atherosclerotic lesions. The interaction between AGEs and receptor for AGEs (RAGE) accelerates AS by affecting the proliferation and migration of VSMCs. In addition, increasing researches have reported that AGEs promote osteogenic transformation and macrophage-like transformation of VSMCs, and affect the progression of AS through other aspects such as autophagy and cell cycle. In this review, we summarize the effect of AGEs on VSMCs in atherosclerotic plaque development and progression. We also discuss the AGEs that link AS and diabetes mellitus, including oxidative stress, inflammation, RAGE ligands, small noncoding RNAs.
Collapse
Affiliation(s)
| | | | | | - Dong Zhao
- *Correspondence: Longyan Yang, ; Dong Zhao,
| |
Collapse
|
46
|
Kränkel N. The "real world" is relative-and biased. Eur J Prev Cardiol 2021; 29:1331-1333. [PMID: 34940857 DOI: 10.1093/eurjpc/zwab227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Nicolle Kränkel
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
47
|
Fukumoto Y. Need more attention to peripheral arterial diseases, especially in women. Eur J Prev Cardiol 2021; 28:e7-e8. [PMID: 33611385 DOI: 10.1177/2047487319898308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Yoshihiro Fukumoto
- Department of Internal Medicine, Kurume University School of Medicine, Japan
| |
Collapse
|
48
|
Zhang X, Cheng M, Gao N, Li Y, Yan C, Tian X, Liu D, Qiu M, Wang X, Luan B, Deng J, Wang S, Tian H, Wang G, Ma X, Stone GW, Han Y. Utility of S100A12 as an Early Biomarker in Patients With ST-Segment Elevation Myocardial Infarction. Front Cardiovasc Med 2021; 8:747511. [PMID: 34977174 PMCID: PMC8718434 DOI: 10.3389/fcvm.2021.747511] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/22/2021] [Indexed: 01/16/2023] Open
Abstract
Importance: S100A12 is a calcium binding protein which is involved in inflammation and progression of atherosclerosis.Objective: We sought to investigate the utility of S100A12 as a biomarker for the early diagnosis and prognostication of patients presenting with ST-segment elevation myocardial infarction (STEMI).Design, Setting, and Participants: S100A12 was measured in 1023 patients presenting to the emergency department with acute chest pain between June 2012 and November 2015. An independent cohort of 398 patients enrolled at 3 different hospitals served as a validation cohort.Main Outcomes and Measures: The primary clinical endpoint of interest was major adverse cardiac and cerebral events (MACCE) defined as a composite of all-cause death, MI, stroke, or hospitalization for heart failure.Results: A total of 438/1023 patients (42.8%) in the diagnosis cohort were adjudicated as STEMI, among whom plasma S100A12 levels increased within 30 min and peaked 1–2 h after symptom onset. Compared with high-sensitivity cardiac troponin T and creatine kinase-MB isoenzyme, S100A12 more accurately identified STEMI, especially within the first 2 h after symptom onset (area under the curve 0.963 compared with 0.860 for hscTnT and 0.711 for CK-MB, both P < 0.05). These results were consistent in the 243-patient validation cohort. The 1-year rate of MACCE was greatest in patients in the highest peak S100A12 tertile, intermediate in the middle tertile and least in the lowest tertile (9.3 vs. 5.7 vs. 3.0% respectively, Ptrend = 0.0006). By multivariable analysis the peak plasma concentration of S100A12 was an independent predictor of MACCE within 1 year after STEMI (HR, 1.001, 95%CI, 1.000–1.002; P = 0.0104).Conclusions and Relevance: S100A12 rapidly identified patients with STEMI, more accurately than other cardiac biomarkers, especially within the first 2 h after symptom onset. The peak plasma S100A12 level was a strong predictor of 1-year prognosis after STEMI.
Collapse
Affiliation(s)
- Xiaolin Zhang
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Minghui Cheng
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Naijing Gao
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Yi Li
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaoxiang Tian
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Dan Liu
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Miaohan Qiu
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaozeng Wang
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Bo Luan
- Department of Cardiology, Liaoning Provincial People's Hospital, Shenyang, China
| | - Jie Deng
- Department of Cardiology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shouli Wang
- Department of Cardiology, General Hospital of the Strategic Support Force of the Chinese People's Liberation Army, Beijing, China
| | - Hongyan Tian
- Department of Cardiology First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Geng Wang
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Gregg W. Stone
- Icahn School of Medicine at Mount Sinai, Mount Sinai Heart and the Cardiovascular Research Foundation, New York, NY, United States
| | - Yaling Han
- Cardiovascular Research Institute and Department of Cardiology, The General Hospital of Northern Theater Command, Shenyang, China
- *Correspondence: Yaling Han
| |
Collapse
|
49
|
Ren F, Jiang Z, Han M, Zhang H, Yun B, Zhu H, Li Z. NIR‐II Fluorescence imaging for cerebrovascular diseases. VIEW 2021. [DOI: 10.1002/viw.20200128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Feng Ren
- Center for Molecular Imaging and Nuclear Medicine State Key Laboratory of Radiation Medicine and Protection School for Radiological and Interdisciplinary Sciences (RAD‐X) Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions Suzhou 215123 P. R. China
| | - Zhilin Jiang
- Center for Molecular Imaging and Nuclear Medicine State Key Laboratory of Radiation Medicine and Protection School for Radiological and Interdisciplinary Sciences (RAD‐X) Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions Suzhou 215123 P. R. China
| | - Mengxiao Han
- Center for Molecular Imaging and Nuclear Medicine State Key Laboratory of Radiation Medicine and Protection School for Radiological and Interdisciplinary Sciences (RAD‐X) Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions Suzhou 215123 P. R. China
| | - Hao Zhang
- Center for Molecular Imaging and Nuclear Medicine State Key Laboratory of Radiation Medicine and Protection School for Radiological and Interdisciplinary Sciences (RAD‐X) Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions Suzhou 215123 P. R. China
| | - Baofeng Yun
- Center for Molecular Imaging and Nuclear Medicine State Key Laboratory of Radiation Medicine and Protection School for Radiological and Interdisciplinary Sciences (RAD‐X) Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions Suzhou 215123 P. R. China
| | - Hongqin Zhu
- Center for Molecular Imaging and Nuclear Medicine State Key Laboratory of Radiation Medicine and Protection School for Radiological and Interdisciplinary Sciences (RAD‐X) Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions Suzhou 215123 P. R. China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine State Key Laboratory of Radiation Medicine and Protection School for Radiological and Interdisciplinary Sciences (RAD‐X) Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions Suzhou 215123 P. R. China
| |
Collapse
|
50
|
Molinaro R, Yu M, Sausen G, Bichsel CA, Corbo C, Folco EJ, Lee GY, Liu Y, Tesmenitsky Y, Shvartz E, Sukhova GK, Kloss F, Croce KJ, Farokhzad OC, Shi J, Libby P. Targeted delivery of protein arginine deiminase-4 inhibitors to limit arterial intimal NETosis and preserve endothelial integrity. Cardiovasc Res 2021; 117:2652-2663. [PMID: 33751034 PMCID: PMC8783386 DOI: 10.1093/cvr/cvab074] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
AIMS Recent evidence suggests that 'vulnerable plaques', which have received intense attention as underlying mechanism of acute coronary syndromes over the decades, actually rarely rupture and cause clinical events. Superficial plaque erosion has emerged as a growing cause of residual thrombotic complications of atherosclerosis in an era of increased preventive measures including lipid lowering, antihypertensive therapy, and smoking cessation. The mechanisms of plaque erosion remain poorly understood, and we currently lack validated effective diagnostics or therapeutics for superficial erosion. Eroded plaques have a rich extracellular matrix, an intact fibrous cap, sparse lipid, and few mononuclear cells, but do harbour neutrophil extracellular traps (NETs). We recently reported that NETs amplify and propagate the endothelial damage at the site of arterial lesions that recapitulate superficial erosion in mice. We showed that genetic loss of protein arginine deiminase (PAD)-4 function inhibited NETosis and preserved endothelial integrity. The current study used systemic administration of targeted nanoparticles to deliver an agent that limits NETs formation to probe mechanisms of and demonstrate a novel therapeutic approach to plaque erosion that limits endothelial damage. METHODS AND RESULTS We developed Collagen IV-targeted nanoparticles (Col IV NP) to deliver PAD4 inhibitors selectively to regions of endothelial cell sloughing and collagen IV-rich basement membrane exposure. We assessed the binding capability of the targeting ligand in vitro and evaluated Col IV NP targeting to areas of denuded endothelium in vivo in a mouse preparation that recapitulates features of superficial erosion. Delivery of the PAD4 inhibitor GSK484 reduced NET accumulation at sites of intimal injury and preserved endothelial continuity. CONCLUSIONS NPs directed to Col IV show selective uptake and delivery of their payload to experimentally eroded regions, illustrating their translational potential. Our results further support the role of PAD4 and NETs in superficial erosion.
Collapse
Affiliation(s)
- Roberto Molinaro
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Business Development of Research, IRCCS San Raffaele Hospital, Milan, Italy
| | - Mikyung Yu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Grasiele Sausen
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Colette A Bichsel
- Department of Surgery, Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Claudia Corbo
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine and Surgery, Nanomedicine Center NANOMIB, University of Milano-Bicocca, Milano, Italy
| | - Eduardo J Folco
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Gha Young Lee
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yuan Liu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yevgenia Tesmenitsky
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Eugenia Shvartz
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Galina K Sukhova
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Frederik Kloss
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kevin J Croce
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|