1
|
Hara T, Sata M. Pericoronary adipose tissue: potential for pathological diagnosis and therapeutic applications. Cardiovasc Interv Ther 2025:10.1007/s12928-025-01126-5. [PMID: 40185991 DOI: 10.1007/s12928-025-01126-5] [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: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
Excessive accumulation of epicardial adipose tissue (EAT) is known to be a risk factor for coronary artery disease and heart failure. In particular, it is thought that inflammation of pericoronary adipose tissue (PCAT) affects the pathology of various coronary artery diseases (CAD). EAT and PCAT are thought to be new therapeutic targets for preventing cardiovascular disease. Although there are no established drugs that specifically reduce inflammation of EAT or PCAT, the basic approach is to improve lifestyle-related diseases through exercise and diet, and to use metabolic improvement drugs and anti-inflammatory drugs as soft support. Potential candidates include statins, SGLT2 inhibitors, and GLP- 1 receptor agonists. In addition to conventional treatments that target substances within blood vessels, treatments that target EAT and PCAT by directly enveloping the coronary arteries and myocardium from outside the body are expected to further suppress cardiovascular events.
Collapse
Affiliation(s)
- Tomoya Hara
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3 - 18 - 15, Kuramoto-cho, Tokushima, 770 - 8503, Japan.
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3 - 18 - 15, Kuramoto-cho, Tokushima, 770 - 8503, Japan
| |
Collapse
|
2
|
Kanuri B, Maremanda KP, Chattopadhyay D, Essop MF, Lee MKS, Murphy AJ, Nagareddy PR. Redefining Macrophage Heterogeneity in Atherosclerosis: A Focus on Possible Therapeutic Implications. Compr Physiol 2025; 15:e70008. [PMID: 40108774 DOI: 10.1002/cph4.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/20/2025] [Accepted: 03/08/2025] [Indexed: 03/22/2025]
Abstract
Atherosclerosis is a lipid disorder where modified lipids (especially oxidized LDL) induce macrophage foam cell formation in the aorta. Its pathogenesis involves a continuum of persistent inflammation accompanied by dysregulated anti-inflammatory responses. Changes in the immune cell status due to differences in the lesional microenvironment are crucial in terms of plaque development, its progression, and plaque rupture. Ly6Chi monocytes generated through both medullary and extramedullary cascades act as one of the major sources of plaque macrophages and thereby foam cells. Both monocytes and monocyte-derived macrophages also participate in pathological events in atherosclerosis-associated multiple organ systems through inter-organ communications. For years, macrophage phenotypes M1 and M2 have been shown to perpetuate inflammatory and resolution responses; nevertheless, such a dualistic classification is too simplistic and contains severe drawbacks. As the lesion microenvironment is enriched with multiple mediators that possess the ability to activate macrophages to diverse phenotypes, it is obvious that such cells should demonstrate substantial heterogeneity. Considerable research in this regard has indicated the presence of additional macrophage phenotypes that are exclusive to atherosclerotic plaques, namely Mox, M4, Mhem, and M(Hb) type. Furthermore, although the concept of macrophage clusters has come to the fore in recent years with the evolution of high-dimensional techniques, classifications based on such 'OMICS' approaches require extensive functional validation as well as metabolic phenotyping. Bearing this in mind, the current review provides an overview of the status of different macrophage populations and their role during atherosclerosis and also outlines possible therapeutic implications.
Collapse
Affiliation(s)
- Babunageswararao Kanuri
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - Krishna P Maremanda
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - Dipanjan Chattopadhyay
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - M Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Man Kit Sam Lee
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Prabhakara R Nagareddy
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| |
Collapse
|
3
|
Yang X, Hu J, Gao Q, Deng Y, Liu Y, He X, Li C, Yu X, Wan Y, Pi C, Wei Y, Li C. Advances in nano-delivery systems based on diagnosis and theranostics strategy for atherosclerosis. J Drug Target 2025; 33:492-507. [PMID: 39601425 DOI: 10.1080/1061186x.2024.2433560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/21/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
Atherosclerosis (AS) is a chronic systemic inflammatory disease, where early diagnosis and theranostics strategy for AS are crucial for improving outcomes. However, conventional diagnostic techniques are limited in identifying early AS lesions, failing to stop the progression of AS in time. Nano-delivery systems have shown significant potential in AS diagnosis and treatment, offering distinct advantages in plaque identification and enhancing drugs concentration at lesion sites, thereby advancing new-generation theranostics strategy. This review discusses the application of nano-delivery systems based on imaging technology in AS diagnosis, and we further explore recent advancements in combining different imaging technologies with emerging theranostics strategy. In addition, we also discuss the challenges faced by nano-delivery systems for AS diagnosis and theranostics in clinical translation, such as nanoparticle targeting efficiency, cytotoxicity and long-term accumulation, immune clearance and inaccurate disease modelling. Finally, we also provide prospects on nano-delivery systems based on diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jian Hu
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Quanle Gao
- Department of Neurology, Geriatrics, Hejiang County People's Hospital, Luzhou, Sichuan, China
| | - Yiping Deng
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yilin Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xinghui He
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chuang Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Yu
- Chinese Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chao Pi
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yumeng Wei
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
4
|
Guo Y, Akcicek EY, Hippe DS, HashemizadehKolowri S, Wang X, Akcicek H, Canton G, Balu N, Geleri DB, Kim T, Shibata D, Zhang K, Ma X, Ferguson MS, Mossa-Basha M, Hatsukami TS, Yuan C. Long-Term Carotid Plaque Progression and the Role of Intraplaque Hemorrhage: A Deep Learning-Based Analysis of Longitudinal Vessel Wall Imaging. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.12.09.24318661. [PMID: 39711698 PMCID: PMC11661346 DOI: 10.1101/2024.12.09.24318661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Background Carotid atherosclerosis is a major contributor in the etiology of ischemic stroke. Although intraplaque hemorrhage (IPH) is known to increase stroke risk and plaque burden, its long-term effects on plaque dynamics remain unclear. This study aimed to evaluate the long-term impact of IPH on carotid plaque burden progression using deep learning-based segmentation on multi-contrast magnetic resonance vessel wall imaging (VWI). Methods Twenty-eight asymptomatic subjects with carotid atherosclerosis underwent an average of 4.7 ± 0.6 VWI scans over 5.8 ± 1.1 years. Deep learning pipelines were developed and validated to segment the carotid vessel walls and IPH. Bilateral plaque progression was analyzed using generalized estimating equations, and linear mixed-effects models evaluated long-term associations between IPH occurrence, IPH volume, and plaque burden (%WV) progression. Results IPH was detected in 23/50 of arteries. Of arteries without IPH at baseline, 11/39 developed new IPH that persisted, while 5/11 arteries with baseline IPH exhibited it throughout the study. Bilateral plaque growth was significantly correlated (r = 0.54, p < 0.001), but this symmetry was weakened with IPH presence. The progression rate for arteries without IPH was -0.001 %/year (p = 0.90). However, IPH presence or development at any point was associated with a 2.3% absolute increase in %WV on average (p < 0.001). The volume of IPH was also positively associated with increased %WV (p = 0.005). Conclusions Deep learning-based segmentation pipelines were utilized to identify IPH, quantify IPH volume, and measure their effects on carotid plaque burden during long-term follow-up. Findings demonstrated that IPH may persist for extended periods. While arteries without IPH demonstrated minimal progression under contemporary treatment, presence of IPH and greater IPH volume significantly accelerated long-term plaque growth.
Collapse
Affiliation(s)
- Yin Guo
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Ebru Yaman Akcicek
- Department of Radiology and Imaging Science, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Daniel S. Hippe
- Clinical Biostatistics, Clinical Research Division, Fred Hutchison Cancer Center, Seattle, WA, USA
| | | | - Xin Wang
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, USA
| | - Halit Akcicek
- Department of Radiology and Imaging Science, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Gador Canton
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Niranjan Balu
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Duygu Baylam Geleri
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Taewon Kim
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
- Department of Neurology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Dean Shibata
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Kaiyu Zhang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Xiaodong Ma
- Department of Radiology and Imaging Science, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Marina S. Ferguson
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Mahmud Mossa-Basha
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Thomas S. Hatsukami
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, USA
| | - Chun Yuan
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Radiology and Imaging Science, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
5
|
Hara T, Sata M. Roles of perivascular adipose tissue in the pathogenesis of atherosclerosis - an update on recent findings. Front Physiol 2025; 15:1522471. [PMID: 39835204 PMCID: PMC11744021 DOI: 10.3389/fphys.2024.1522471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
Lifestyle-related diseases, such as atherosclerosis and diabetes, are now considered to be a series of diseases caused by chronic inflammation. Adipose tissue is considered to be an endocrine organ that not only plays a role in lipid storage, heat production, and buffering, but also produces physiologically active substances and is involved in chronic inflammation. Perivascular adipose tissue (PVAT) surrounding blood vessels similarly produces inflammatory and anti-inflammatory physiologically active substances that act on blood vessels either directly or via the bloodstream. Epicardial adipose tissue (EAT), which is in direct contact with the coronary arteries inside the pericardium, is thought to have a direct effect on the coronary arteries as well. The presence and inflammatory status of these adipose tissues can be evaluated by imaging tests, and has been shown to be associated with the presence of current cardiovascular disease (CVD) and to be a prognostic factor. It is also expected to become a new diagnostic and therapeutic target for CVD.
Collapse
Affiliation(s)
- Tomoya Hara
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | | |
Collapse
|
6
|
Ma X, Yan P, Ju J, Yang Z, Wang W, Wang Q, Liu X, Xia Z, Sun Q. Vessel wall MRI characteristics associated with intraprocedural stent thrombosis during angioplasty for intracranial atherosclerotic stenosis. J Neurointerv Surg 2024; 16:1348-1352. [PMID: 37989580 PMCID: PMC11672038 DOI: 10.1136/jnis-2023-020941] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/30/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND Few studies have so far explored plaque characteristics on high-resolution magnetic resonance vessel wall imaging (HR-VWI) associated with intraprocedural stent thrombosis (IPST) during angioplasty for intracranial atherosclerotic stenosis (ICAS). We aimed to investigate the plaque features on HR-VWI associated with IPST during stenting for ICAS. METHODS This study recruited 77 patients with ICAS who underwent intracranial stenting using the Gateway-Wingspan system, and were performed with enhanced pre- and post-contrast T1-weighted HR-VWI on a 3.0T MRI scanner before angioplasty. During stenting for ICAS, eight patients (male: 100%, age mean ± standard deviation (SD): 58.7±2.47) developed IPST within 30 minutes after stenting. To ensure comparability, 16 patients who had undergone intracranial stenting but did not develop IPST were matched as controls for this study. Univariable and binary logistic models were used to explore the plaque characteristics on HR-VWI associated with IPST. RESULTS Patients who developed IPST had less plaque diffusion (37.50% vs 81.25%, p=0.036), a more severe degree of area stenosis (median 96.30% vs 81.65%, p<0.01), and a higher plaque enhancement index (median 37.99 vs 13.12, p<0.01) compared with those who did not. After multivariate adjustment, IPST was independently associated with a more severe degree of area stenosis (adjusted odds ratio (OR) 1.20, 95% confidence interval (CI) 1.01-1.43, p=0.044) and a higher plaque enhancement index (adjusted OR 1.17, 95% CI 1.01 to 1.36, p=0.036). CONCLUSION Intraprocedural stent thrombosis during intracranial angioplasty for patients with ICAS may be independently associated with a higher plaque enhancement index and a more severe degree of area stenosis on HR-VWI.
Collapse
Affiliation(s)
- Xiaotong Ma
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Neurology, Liaocheng People’s Hospital, Shandong University, Liaocheng, Shandong, P.R. China
| | - Peng Yan
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Jiachen Ju
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Zhengyu Yang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Wenjuan Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Qiuting Wang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Xiaohui Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Zhangyong Xia
- Department of Neurology, Liaocheng People’s Hospital, Shandong University, Liaocheng, Shandong, P.R. China
- Department of Neurology, Liaocheng People’s Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, P.R. China
- Department of Neurology, the second People’s Hospital of Liaocheng, Liaocheng, Shandong, P.R. China
| | - Qinjian Sun
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
7
|
Papafaklis MI, Koros R, Tsigkas G, Karanasos A, Moulias A, Davlouros P. Reversal of Atherosclerotic Plaque Growth and Vulnerability: Effects of Lipid-Modifying and Anti-Inflammatory Therapeutic Agents. Biomedicines 2024; 12:2435. [PMID: 39595002 PMCID: PMC11591594 DOI: 10.3390/biomedicines12112435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Atherosclerotic plaque development constitutes the primary substrate of coronary artery disease (CAD) and is the outcome of an intricate process involving endothelial damage, inflammation, and lipid retention. The clinical efficacy of many lipid-lowering therapies in patients with CAD has been well established. Over the past few decades, a substantial and significant advance regarding the use of invasive and non-invasive imaging modalities has been observed. Numerous studies have been conducted using these imaging techniques and have investigated the changes in morphology (e.g., atheroma volume) and composition (e.g., lipid burden, fibrous cap thickness, macrophage accumulation) at the plaque level that explain the improved clinical outcomes by various pharmacological interventions. Lipid-lowering agents, such as statins and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, demonstrate direct effects on plaque volume and composition that enhance plaque stabilization and/or regression beyond the reduction of low-density lipoproteins. An increasing amount of clinical research is also focused on the role of inflammation in plaque vulnerability and future adverse cardiac events. Consequently, there is a pressing need to explore therapeutic strategies that are capable of disrupting the inflammatory response as well as reducing atheroma burden and modifying high-risk plaque characteristics. This review provides a comprehensive analysis of the current evidence regarding the effects of traditional and novel therapeutic strategies targeting modification of the lipid profile and inflammatory processes on reversing plaque growth and attenuating vulnerable features, thereby promoting plaque stabilization and passivation.
Collapse
Affiliation(s)
- Michail I. Papafaklis
- Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Cardiology Division, University Hospital of Patras, 26504 Rio, Greece
| | - Rafail Koros
- Cardiology Division, University Hospital of Patras, 26504 Rio, Greece
| | - Grigorios Tsigkas
- Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Cardiology Division, University Hospital of Patras, 26504 Rio, Greece
| | - Antonios Karanasos
- Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Cardiology Division, University Hospital of Patras, 26504 Rio, Greece
| | | | - Periklis Davlouros
- Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Cardiology Division, University Hospital of Patras, 26504 Rio, Greece
| |
Collapse
|
8
|
Paternò Raddusa MS, Marino A, Celesia BM, Spampinato S, Giarratana C, Venanzi Rullo E, Cacopardo B, Nunnari G. Atherosclerosis and Cardiovascular Complications in People Living with HIV: A Focused Review. Infect Dis Rep 2024; 16:846-863. [PMID: 39311207 PMCID: PMC11417834 DOI: 10.3390/idr16050066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024] Open
Abstract
The intersection of Human Immunodeficiency Virus (HIV) infection and cardiovascular disease (CVD) represents a significant area of concern; advancements in antiretroviral therapy (ART) have notably extended the life expectancy of people living with HIV (PLWH), concurrently elevating the prevalence of chronic conditions such as CVD. This paper explores the multifaceted relationship between HIV infection, ART, and cardiovascular health, focusing on the mechanisms by which HIV and ART contribute to increased cardiovascular risk, including the promotion of endothelial dysfunction, inflammation, immune activation, and metabolic disturbances. We highlight the critical roles of HIV-associated proteins-Tat, Nef, and gp120-in accelerating atherosclerosis through direct and indirect pathways that exacerbate endothelial damage and inflammation. Additionally, we address the persistent challenge of chronic inflammation and immune activation in PLWH, factors that are strongly predictive of non-AIDS-related diseases, including CVD, even in the context of effective viral suppression. The impact of ART on cardiovascular risk is examined, with particular attention to the metabolic implications of specific ART regimens, which can influence lipid profiles and body composition, thereby modifying CVD risk. The therapeutic potential of statins, aspirin, and emerging treatments such as PCSK9 inhibitors in mitigating cardiovascular morbidity and mortality among PLWH is discussed, alongside considerations for their use in conjunction with ART. Our review underscores the necessity for a comprehensive, multidisciplinary approach to cardiovascular care in PLWH, which integrates vigilant cardiovascular risk assessment and management with HIV treatment. As we navigate the evolving landscape of HIV care, the goal remains to optimize treatment outcomes while minimizing cardiovascular risk, ensuring that the gains in longevity afforded by ART translate into improved overall health and quality of life for PLWH.
Collapse
Affiliation(s)
| | - Andrea Marino
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, ARNAS Garibaldi Hospital, University of Catania, 95123 Catania, Italy; (B.M.C.); (B.C.); (G.N.)
| | - Benedetto Maurizio Celesia
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, ARNAS Garibaldi Hospital, University of Catania, 95123 Catania, Italy; (B.M.C.); (B.C.); (G.N.)
| | - Serena Spampinato
- Department of Clinical and Experimental Medicine, University of Messina, 98124 Messina, Italy; (M.S.P.R.); (S.S.); (C.G.); (E.V.R.)
| | - Carmen Giarratana
- Department of Clinical and Experimental Medicine, University of Messina, 98124 Messina, Italy; (M.S.P.R.); (S.S.); (C.G.); (E.V.R.)
| | - Emmanuele Venanzi Rullo
- Department of Clinical and Experimental Medicine, University of Messina, 98124 Messina, Italy; (M.S.P.R.); (S.S.); (C.G.); (E.V.R.)
| | - Bruno Cacopardo
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, ARNAS Garibaldi Hospital, University of Catania, 95123 Catania, Italy; (B.M.C.); (B.C.); (G.N.)
| | - Giuseppe Nunnari
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, ARNAS Garibaldi Hospital, University of Catania, 95123 Catania, Italy; (B.M.C.); (B.C.); (G.N.)
| |
Collapse
|
9
|
Liu Z, Zhang L, Sun B, Ding Y. Association of cardiovascular risk factors and intraplaque neovascularization in symptomatic carotid plaque. Front Neurol 2024; 15:1442656. [PMID: 39253361 PMCID: PMC11381375 DOI: 10.3389/fneur.2024.1442656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/15/2024] [Indexed: 09/11/2024] Open
Abstract
Background and purpose Cardiovascular risk factors are known to contribute to the formation of atherosclerotic plaques, which can result in carotid stenosis. However, the extent to which these factors are associated with intraplaque neovascularization, a key indicator of plaque vulnerability, remains unclear. To investigate this relationship, a study was conducted utilizing contrast-enhanced ultrasound (CEUS) to assess intraplaque neovascularization in symptomatic patients. Methods A cohort of 157 symptomatic patients underwent evaluation using Contrast-Enhanced Ultrasound (CEUS) imaging to assess carotid intraplaque neovascularization, which was quantified based on the degree of plaque enhancement. The collected data encompassed baseline patient characteristics, results from biochemical examinations, cardiovascular risk factors, and medication usage history. Regression analyses were conducted to elucidate the relationship between carotid plaque neovascularization and various cardiovascular risk factors. Results Patients with intraplaque neovascularization were more prone to have diabetes mellitus (OR 3.81, 95% CI 1.94-7.46, p < 0.001), dyslipidemia (OR 2.36, 95% CI 1.22-4.55, p = 0.011) and hypertension (OR 2.92, 95% CI 1.50-5.71, p = 0.002). Smoking increased the risk of having intraplaque neovascularization (OR 2.25, 95% CI 1.12-4.54, p = 0.023). Treatment with statins was significantly lower in patients with intraplaque neovascularization (OR 0.37, 95% CI 0.19-0.72, p = 0.003). In the multivariate analysis, diabetes mellitus (OR 3.27, 95% CI 1.10-9.78, p = 0.034) was independently related to the presence of intraplaque neovascularization. Meanwhile, compared to the patients in the first tertile of serum glucose (< 6.20 mmol/L), the patients in the third tertile (> 13.35 mmol/L) had the most significance of intraplaque neovascularization (OR 5.55, 95% CI 1.85-16.66, p = 0.002). Conclusion The findings indicated that diabetes mellitus is a significant cardiovascular risk factor that is strongly associated with carotid intraplaque neovascularization.
Collapse
Affiliation(s)
- Zehao Liu
- Department of Neurosurgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Lianlian Zhang
- Department of Ultrasonography, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, China
| | - Bing Sun
- Department of Neurosurgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Yasuo Ding
- Department of Neurosurgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| |
Collapse
|
10
|
Xie E, Shen X, Yeo YH, Xing Z, Ebinger JE, Duan Y, Zhang Y, Cheng S, Ji F, Deng J. Exploring the underlying molecular mechanisms of acute myocardial infarction after SARS-CoV-2 infection. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 44:100417. [PMID: 39045234 PMCID: PMC11263507 DOI: 10.1016/j.ahjo.2024.100417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/20/2024] [Indexed: 07/25/2024]
Abstract
An increase in acute myocardial infarction (AMI)-related deaths has been reported during the COVID-19 pandemic. Despite evidence suggesting the association between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and AMI, the underlying mechanisms remain unclear. Here, we integrated mRNA and microRNA expression profiles related to SARS-CoV-2 infection and AMI from public databases. We then performed transcriptomic analysis using bioinformatics and systems biology approaches to explore the potential molecular mechanisms of SARS-CoV-2 infection affects AMI. First, twenty-one common differentially expressed genes (DEGs) were identified from SARS-CoV-2 infection and AMI patients in endothelial cells datasets and then we performed functional analysis to predict the roles of these DEGs. The functional analysis emphasized that the endothelial cell response to cytokine stimulus due to excessive inflammation was essential in these two diseases. Importantly, the tumor necrosis factor and interleukin-17 signaling pathways appeared to be integral factors in this mechanism. Interestingly, most of these common genes were also upregulated in transcriptomic datasets of SARS-CoV-2-infected cardiomyocytes, suggesting that these genes may be shared in cardiac- and vascular-related injuries. We subsequently built a protein-protein interaction network and extracted hub genes and essential modules from this network. At the transcriptional and post-transcriptional levels, regulatory networks with common DEGs were also constructed, and some key regulator signatures were further identified and validated. In summary, our research revealed that a highly activated inflammatory response in patients with COVID-19 might be a crucial factor for susceptibility to AMI and we identified some candidate genes and regulators that could be used as biomarkers or potential therapeutic targets.
Collapse
Affiliation(s)
- Enrui Xie
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Infectious Disease, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaotao Shen
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore
- Department of Genetics, Stanford University, California, USA
| | - Yee Hui Yeo
- Department of Genetics, Stanford University, California, USA
| | - Zixuan Xing
- Department of Infectious Disease, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Joseph E. Ebinger
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Yixuan Duan
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yue Zhang
- Department of Infectious Disease, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- The Eighth Hospital of Xi'an City, Xi'an Jiaotong University, Xi'an, China
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Fanpu Ji
- Department of Infectious Disease, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Medical Research Center of Infectious Diseases, Xi'an, China
| | - Jie Deng
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
11
|
Rademakers T, Manca M, Jin H, Orban T, Perisic LM, Frissen HJM, Rühle F, Hautvast P, van Rijssel J, van Kuijk K, Mees BME, Peutz-Kootstra CJ, Heeneman S, Daemen MJAP, Pasterkamp G, Stoll M, van Zandvoort MAMJ, Hedin U, Dequiedt F, van Buul JD, Sluimer JC, Biessen EAL. Human atherosclerotic plaque transcriptomics reveals endothelial beta-2 spectrin as a potential regulator a leaky plaque microvasculature phenotype. Angiogenesis 2024; 27:461-474. [PMID: 38780883 PMCID: PMC11303431 DOI: 10.1007/s10456-024-09921-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024]
Abstract
The presence of atherosclerotic plaque vessels is a critical factor in plaque destabilization. This may be attributable to the leaky phenotype of these microvessels, although direct proof for this notion is lacking. In this study, we investigated molecular and cellular patterns of stable and hemorrhaged human plaque to identify novel drivers of intraplaque vessel dysfunction. From transcriptome data of a human atherosclerotic lesion cohort, we reconstructed a co-expression network, identifying a gene module strongly and selectively correlated with both plaque microvascular density and inflammation. Spectrin Beta Non-Erythrocytic 1 (sptbn1) was identified as one of the central hubs of this module (along with zeb1 and dock1) and was selected for further study based on its predominant endothelial expression. Silencing of sptbn1 enhanced leukocyte transmigration and vascular permeability in vitro, characterized by an increased number of focal adhesions and reduced junctional VE-cadherin. In vivo, sptbn1 knockdown in zebrafish impaired the development of the caudal vein plexus. Mechanistically, increased substrate stiffness was associated with sptbn1 downregulation in endothelial cells in vitro and in human vessels. Plaque SPTBN1 mRNA and protein expression were found to correlate with an enhanced presence of intraplaque hemorrhage and future cardiovascular disease (CVD) events during follow-up. In conclusion, we identify SPTBN1 as a central hub gene in a gene program correlating with plaque vascularisation. SPTBN1 was regulated by substrate stiffness in vitro while silencing blocked vascular development in vivo, and compromised barrier function in vitro. Together, SPTBN1 is identified as a new potential regulator of the leaky phenotype of atherosclerotic plaque microvessels.
Collapse
Affiliation(s)
- Timo Rademakers
- Department of Pathology, Experimental Vascular Pathology Group, Maastricht University, PO box 5800, 6202 AZ, Maastricht, The Netherlands
- Department of Plasma Proteins, Laboratory for Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Marco Manca
- Department of Pathology, Experimental Vascular Pathology Group, Maastricht University, PO box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Han Jin
- Department of Pathology, Experimental Vascular Pathology Group, Maastricht University, PO box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Tanguy Orban
- Laboratory of Protein Signaling and Interactions, GIGA, Liège Université, Liège, Belgium
| | - Ljubica Matic Perisic
- Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska Hospital, Stockholm, Sweden
| | - Hubertus J M Frissen
- Department of Pathology, Experimental Vascular Pathology Group, Maastricht University, PO box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Frank Rühle
- Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany
| | - Petra Hautvast
- Department of Pathology, Experimental Vascular Pathology Group, Maastricht University, PO box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Jos van Rijssel
- Department of Plasma Proteins, Laboratory for Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Kim van Kuijk
- Department of Pathology, Experimental Vascular Pathology Group, Maastricht University, PO box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Barend M E Mees
- Department of Vascular Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Carine J Peutz-Kootstra
- Department of Pathology, Experimental Vascular Pathology Group, Maastricht University, PO box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Sylvia Heeneman
- Department of Pathology, Experimental Vascular Pathology Group, Maastricht University, PO box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Mat J A P Daemen
- Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - Gerard Pasterkamp
- Laboratory of Clinical Chemistry and Haematology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Monika Stoll
- Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany
- Maastricht Center for Systems Biology (MaCSBio, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
| | - Marc A M J van Zandvoort
- Department of Molecular Cell Biology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Ulf Hedin
- Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska Hospital, Stockholm, Sweden
| | - Franck Dequiedt
- Laboratory of Protein Signaling and Interactions, GIGA, Liège Université, Liège, Belgium
| | - Jaap D van Buul
- Department of Plasma Proteins, Laboratory for Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Judith C Sluimer
- Department of Pathology, Experimental Vascular Pathology Group, Maastricht University, PO box 5800, 6202 AZ, Maastricht, The Netherlands
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Department for Renal and Hypertensive, Rheumatological and Immunological Diseases (Medical Clinic II), RWTH Aachen, Aachen, Germany
| | - Erik A L Biessen
- Department of Pathology, Experimental Vascular Pathology Group, Maastricht University, PO box 5800, 6202 AZ, Maastricht, The Netherlands.
- Institute for Molecular Cardiovascular Research, RWTH Aachen, Aachen, Germany.
| |
Collapse
|
12
|
Niida T, Kinoshita D, Suzuki K, Yuki H, Fujimoto D, Dey D, Lee H, McNulty I, Ferencik M, Yonetsu T, Kakuta T, Jang IK. Layered plaque is associated with high levels of vascular inflammation and vulnerability in patients with stable angina pectoris. J Thromb Thrombolysis 2024; 57:880-887. [PMID: 38649561 DOI: 10.1007/s11239-024-02982-3] [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] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Layered plaque, a signature of previous plaque destabilization and healing, is a known predictor for rapid plaque progression; however, the mechanism of which is unknown. The aim of the current study was to compare the level of vascular inflammation and plaque vulnerability in layered plaques to investigate possible mechanisms of rapid plaque progression. This is a retrospective, observational, single-center cohort study. Patients who underwent both coronary computed tomography angiography (CTA) and optical coherence tomography (OCT) for stable angina pectoris (SAP) were selected. Plaques were defined as any tissue (noncalcified, calcified, or mixed) within or adjacent to the lumen. Perivascular inflammation was measured by pericoronary adipose tissue (PCAT) attenuation at the plaque levels on CTA. Features of plaque vulnerability were assessed by OCT. Layered plaques were defined as plaques presenting one or more layers of different optical densities and a clear demarcation from underlying components on OCT. A total of 475 plaques from 195 patients who presented with SAP were included. Layered plaques (n = 241), compared with non-layered plaques (n = 234), had a higher level of vascular inflammation (-71.47 ± 10.74 HU vs. -73.69 ± 10.91 HU, P = 0.026) as well as a higher prevalence of the OCT features of plaque vulnerability, including lipid-rich plaque (83.8% vs. 66.7%, P < 0.001), thin-cap fibroatheroma (26.1% vs. 17.5%, P = 0.026), microvessels (61.8% vs. 34.6%, P < 0.001), and cholesterol crystals (38.6% vs. 25.6%, P = 0.003). Layered plaque was associated with a higher level of vascular inflammation and a higher prevalence of plaque vulnerability, which might play an important role in rapid plaque progression.Clinical trial registration: https://classic.clinicaltrials.gov/ct2/show/NCT04523194 .
Collapse
Affiliation(s)
- Takayuki Niida
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Daisuke Kinoshita
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Keishi Suzuki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Haruhito Yuki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Daichi Fujimoto
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Iris McNulty
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Maros Ferencik
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Taishi Yonetsu
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tsunekazu Kakuta
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
13
|
Li Y, Zhang L, Yang W, Lin L, Pan J, Lu M, Zhang Z, Li Y, Li C. Notoginsenoside R 1 decreases intraplaque neovascularization by governing pericyte-endothelial cell communication via Ang1/Tie2 axis in atherosclerosis. Phytother Res 2024; 38:4036-4052. [PMID: 38886264 DOI: 10.1002/ptr.8257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
Atherosclerosis represents the major cause of mortality worldwide and triggers higher risk of acute cardiovascular events. Pericytes-endothelial cells (ECs) communication is orchestrated by ligand-receptor interaction generating a microenvironment which results in intraplaque neovascularization, that is closely associated with atherosclerotic plaque instability. Notoginsenoside R1 (R1) exhibits anti-atherosclerotic bioactivity, but its effect on angiogenesis in atherosclerotic plaque remains elusive. The aim of our study is to explore the therapeutic effect of R1 on vulnerable plaque and investigate its potential mechanism against intraplaque neovascularization. The impacts of R1 on plaque stability and intraplaque neovascularization were assessed in ApoE-/- mice induced by high-fat diet. Pericytes-ECs direct or non-direct contact co-cultured with VEGF-A stimulation were used as the in vitro angiogenesis models. Overexpressing Ang1 in pericytes was performed to investigate the underlying mechanism. In vivo experiments, R1 treatment reversed atherosclerotic plaque vulnerability and decreased the presence of neovessels in ApoE-/- mice. Additionally, R1 reduced the expression of Ang1 in pericytes. In vitro experiments demonstrated that R1 suppressed pro-angiogenic behavior of ECs induced by pericytes cultured with VEGF-A. Mechanistic studies revealed that the anti-angiogenic effect of R1 was dependent on the inhibition of Ang1 and Tie2 expression, as the effects were partially reversed after Ang1 overexpressing in pericytes. Our study demonstrated that R1 treatment inhibited intraplaque neovascularization by governing pericyte-EC association via suppressing Ang1-Tie2/PI3K-AKT paracrine signaling pathway. R1 represents a novel therapeutic strategy for atherosclerotic vulnerable plaques in clinical application.
Collapse
Affiliation(s)
- Yuan Li
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenqing Yang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinyuan Pan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunlun Li
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
14
|
Konishi T, Kawakami R, Vozenilek AE, Ghosh SKB, Xu W, Grogan A, Shah P, Tanaka T, Sekimoto T, Shiraki T, Kawai K, Sato Y, Mori M, Sakamoto A, Hisadome H, Ashida K, Bellissard A, Williams D, Dryanovski D, Kutys R, Cheng Q, Romero M, Chahal D, Virmani R, Finn AV. Mechanisms of Medial Wall Thinning in Chronic Total Occlusion. JACC Cardiovasc Interv 2024; 17:1719-1728. [PMID: 38970581 DOI: 10.1016/j.jcin.2024.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/25/2024] [Accepted: 05/07/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND The success rate of percutaneous coronary intervention (PCI) for chronic total occlusion (CTO) is lower and the risk for complications higher compared with other non-CTO PCI. Although interventionalists focus on intimal plaque characteristics, the coronary media is an important (especially for techniques involving antegrade dissection and re-entry) but poorly understood structure in CTO PCI. OBJECTIVES The aim of the present study was to investigate coronary medial wall thinning in CTO lesions and determine how this thinning might affect CTO PCI. METHODS A total of 2,586 sections were investigated, from arteries with evidence of CTO from 54 subjects (1,383 sections) and arteries without evidence of CTO from 54 subjects with non-coronary-related deaths (1,203 sections) after matching for age, gender, body weight, and body height. RESULTS The medial thickness in subjects with CTO was lower than that in those with non-coronary-related death (P < 0.001). In subjects with CTO, CTO lesions had thinner medial walls compared with those with lower luminal narrowing (P < 0.001). At the CTO distal segments, the 6- to 12-mm distal segment from the distal end of the CTO had significantly less luminal narrowing (P < 0.001), and similar medial thickness, compared with the distal end of the CTO. Immunohistochemical analysis revealed that short-duration CTO had more cleaved caspase-3-positive cells in media and had significantly more CD3+, CD4+, CD8+, and CD4+CD28null T cells compared with long-duration CTO. CONCLUSIONS CTO lesions demonstrated coronary medial thinning compared with non-CTO lesions. Further investigation of the cause-and-effect relationship among inflammation, apoptosis, and coronary medial wall thinning is warranted in future mechanistic studies.
Collapse
Affiliation(s)
- Takao Konishi
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA; Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Aimee E Vozenilek
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Saikat Kumar B Ghosh
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Weili Xu
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Alyssa Grogan
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Palak Shah
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Takamasa Tanaka
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Teruo Sekimoto
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Tatsuya Shiraki
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Kenji Kawai
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Yu Sato
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Masayuki Mori
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Atsushi Sakamoto
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | | | - Kazuhiro Ashida
- Cardiovascular Center, Seirei Yokohama Hospital, Yokohama, Japan
| | - Arielle Bellissard
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Desiree Williams
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Dilyan Dryanovski
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Robert Kutys
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Qi Cheng
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Maria Romero
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Diljon Chahal
- School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Aloke V Finn
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA; School of Medicine, University of Maryland, Baltimore, Maryland, USA.
| |
Collapse
|
15
|
Cen K, Huang Y, Xie Y, Liu Y. The guardian of intracranial vessels: Why the pericyte? Biomed Pharmacother 2024; 176:116870. [PMID: 38850658 DOI: 10.1016/j.biopha.2024.116870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Intracranial atherosclerotic stenosis (ICAS) is a pathological condition characterized by progressive narrowing or complete blockage of intracranial blood vessels caused by plaque formation. This condition leads to reduced blood flow to the brain, resulting in cerebral ischemia and hypoxia. Ischemic stroke (IS) resulting from ICAS poses a significant global public health challenge, especially among East Asian populations. However, the underlying causes of the notable variations in prevalence among diverse populations, as well as the most effective strategies for preventing and treating the rupture and blockage of intracranial plaques, remain incompletely comprehended. Rupture of plaques, bleeding, and thrombosis serve as precipitating factors in the pathogenesis of luminal obstruction in intracranial arteries. Pericytes play a crucial role in the structure and function of blood vessels and face significant challenges in regulating the Vasa Vasorum (VV)and preventing intraplaque hemorrhage (IPH). This review aims to explore innovative therapeutic strategies that target the pathophysiological mechanisms of vulnerable plaques by modulating pericyte biological function. It also discusses the potential applications of pericytes in central nervous system (CNS) diseases and their prospects as a therapeutic intervention in the field of biological tissue engineering regeneration.
Collapse
Affiliation(s)
- Kuan Cen
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China
| | - YinFei Huang
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China
| | - Yu Xie
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China
| | - YuMin Liu
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China.
| |
Collapse
|
16
|
Yamamoto K, Mizutani K, Akiyama T, Nogawa H, Toda M. Vasa vasorum: The role in intracranial physiology and pathophysiology. Surg Neurol Int 2024; 15:188. [PMID: 38974550 PMCID: PMC11225505 DOI: 10.25259/sni_214_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/04/2024] [Indexed: 07/09/2024] Open
Abstract
Background Vasa vasorum (VVs) is a Latin word representing vessels of vessels. VVs are usually found on the adventitia of the parent vessel and infrequently reach the media and intima, depending on the size and type of the parent vessels and physiological and pathological conditions. The VVs include arteries, capillaries, veins, and lymphatic vessels, involving the oxygenation and nourishment of the vessel's wall to sustain its healthy state. Accumulated studies have revealed that VVs are involved in various intracranial lesions, including atherosclerotic diseases, aneurysms, and shunt diseases. The current review aims to review and integrate past and recent findings and knowledge on VVs and to facilitate our understanding of VVs and intracranial pathology involving VVs. Methods A literature review was carried out with a focus on the role of VVs by searching the Pubmed database. Results We identified 71 articles that discuss the role of VVs. We discussed the anatomical structure, physiological significance, and pathological significance of the VV. Conclusion VV is not only involved in the nutrition and metabolism of the vascular wall but is also deeply involved in the pathogenesis of inflammation, ischemia, and thrombosis of the vascular wall. In addition, in the central nervous system, intracranial vascular wall nutrient particularities and VVs are closely related to the pathogenesis of cerebral aneurysms, subarachnoid hemorrhage, arteriovenous shunt disease, atherosclerotic lesions, and other conditions.
Collapse
Affiliation(s)
| | - Katsuhiro Mizutani
- Department of Neurosurgery, Keio University, School of Medicine, Shinjuku, Japan
| | | | | | | |
Collapse
|
17
|
Delanghe JR, Delrue C, Speeckaert R, Speeckaert MM. Unlocking the link between haptoglobin polymorphism and noninfectious human diseases: insights and implications. Crit Rev Clin Lab Sci 2024; 61:275-297. [PMID: 38013410 DOI: 10.1080/10408363.2023.2285929] [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: 07/21/2023] [Accepted: 11/16/2023] [Indexed: 11/29/2023]
Abstract
Haptoglobin (Hp) is a polymorphic protein that was initially described as a hemoglobin (Hb)-binding protein. The major functions of Hp are to scavenge Hb, prevent iron loss, and prevent heme-based oxidation. Hp regulates angiogenesis, nitric oxide homeostasis, immune responses, and prostaglandin synthesis. Genetic polymorphisms in the Hp gene give rise to different phenotypes, including Hp 1-1, Hp 2-1, and Hp 2-2. Extensive research has been conducted to investigate the association between Hp polymorphisms and several medical conditions including cardiovascular disease, inflammatory bowel disease, cancer, transplantation, and hemoglobinopathies. Generally, the Hp 2-2 phenotype is associated with increased disease risk and poor outcomes. Over the years, the Hp 2 allele has spread under genetic pressures. Individuals with the Hp 2-2 phenotype generally exhibit lower levels of CD163 expression in macrophages. The decreased expression of CD163 may be associated with the poor antioxidant capacity in the serum of subjects carrying the Hp 2-2 phenotype. However, the Hp 1-1 phenotype may confer protection in some cases. The Hp1 allele has strong antioxidant, anti-inflammatory, and immunomodulatory properties. It is important to note that the benefits of the Hp1 allele may vary depending on genetic and environmental factors as well as the specific disease or condition under consideration. Therefore, the Hp1 allele may not necessarily confer advantages in all situations, and its effects may be context-dependent. This review highlights the current understanding of the role of Hp polymorphisms in cardiovascular disease, inflammatory bowel disease, cancer, transplantation, hemoglobinopathies, and polyuria.
Collapse
Affiliation(s)
- Joris R Delanghe
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium
| | | | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium
- Research Foundation-Flanders (FWO), Brussels, Belgium
| |
Collapse
|
18
|
Lv Y, Ma X, Zhao W, Ju J, Yan P, Li S, Xue Y, Sui Y, Shao S, Sun Q, Qiu C. Association of plaque characteristics with long-term stroke recurrence in patients with intracranial atherosclerotic disease: a 3D high-resolution MRI-based cohort study. Eur Radiol 2024; 34:3022-3031. [PMID: 37870623 PMCID: PMC11126465 DOI: 10.1007/s00330-023-10278-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 07/31/2023] [Accepted: 08/12/2023] [Indexed: 10/24/2023]
Abstract
OBJECTIVES To evaluate the predictive ability of plaque characteristics for long-term stroke recurrence among patients with symptomatic intracranial atherosclerotic disease (ICAD). METHODS This cohort study included 132 patients with acute ischemic stroke (AIS) attributed to ICAD who were recruited between July 2017 and December 2020 and followed until stroke recurrence or December 2021. Plaque surface irregularity, degree of stenosis, plaque burden, remodeling ratio, enhancement ratio, and intraplaque hemorrhage were assessed with 3-dimensional high-resolution magnetic resonance vessel wall imaging (3D HR-MRI). Data were analyzed using Cox models, receiver operating characteristic (ROC) curves, and Kaplan-Meier survival analysis. RESULTS Of the 132 patients, during a median follow-up of 2.8 years, stroke recurrence occurred in 35 patients. The multivariable-adjusted hazard ratio (95% confidence interval) of stroke recurrence was 3.15 (1.34-7.42) per 10% increase in plaque burden and 2.17 (1.27-3.70) for enhancement ratio. The area under the curve (AUC) to predict stroke recurrence was 0.725 (95% CI 0.629-0.822) for plaque burden, 0.692 (95% CI 0.593-0.792) for enhancement ratio, and only 0.595 (95% CI 0.492-0.699) for the Essen stroke risk score. The Kaplan-Meier survival analysis further demonstrated significant differences in survival of free recurrent stroke between patients with plaque burden or enhancement ratio below and above the optimum cut-offs (both p < 0.001). CONCLUSION Higher plaque burden and enhancement ratio are independent risk factors for long-term stroke recurrence among patients with symptomatic ICAD, and valuable imaging markers for predicting and stratifying risk of stroke recurrence. CLINICAL RELEVANCE STATEMENT In patients with symptomatic ICAD, the results of this high-resolution magnetic resonance vessel wall imaging study have potential implications for optimal management of intracranial plaques and secondary prevention of stroke recurrence based on plaque burden and enhancement ratio. KEY POINTS • Identification of intracranial plaque characteristics responsible for stroke recurrence is essential to preventing stroke recurrence in patients with symptomatic intracranial atherosclerotic disease. • Higher plaque burden and enhancement ratio are independent risk factors for stroke recurrence. • Plaque burden and enhancement ratio are valuable imaging markers in the prediction and stratification of the risk of stroke recurrence.
Collapse
Affiliation(s)
- Yaodong Lv
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Xiaotong Ma
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Weihua Zhao
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Jiachen Ju
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Peng Yan
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Shan Li
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yuan Xue
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - YanLing Sui
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Sai Shao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Qinjian Sun
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Chengxuan Qiu
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet-Stockholm University, Stockholm, Sweden
| |
Collapse
|
19
|
Miceli G, Basso MG, Pintus C, Pennacchio AR, Cocciola E, Cuffaro M, Profita M, Rizzo G, Tuttolomondo A. Molecular Pathways of Vulnerable Carotid Plaques at Risk of Ischemic Stroke: A Narrative Review. Int J Mol Sci 2024; 25:4351. [PMID: 38673936 PMCID: PMC11050267 DOI: 10.3390/ijms25084351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/05/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The concept of vulnerable carotid plaques is pivotal in understanding the pathophysiology of ischemic stroke secondary to large-artery atherosclerosis. In macroscopic evaluation, vulnerable plaques are characterized by one or more of the following features: microcalcification; neovascularization; lipid-rich necrotic cores (LRNCs); intraplaque hemorrhage (IPH); thin fibrous caps; plaque surface ulceration; huge dimensions, suggesting stenosis; and plaque rupture. Recognizing these macroscopic characteristics is crucial for estimating the risk of cerebrovascular events, also in the case of non-significant (less than 50%) stenosis. Inflammatory biomarkers, such as cytokines and adhesion molecules, lipid-related markers like oxidized low-density lipoprotein (LDL), and proteolytic enzymes capable of degrading extracellular matrix components are among the key molecules that are scrutinized for their associative roles in plaque instability. Through their quantification and evaluation, these biomarkers reveal intricate molecular cross-talk governing plaque inflammation, rupture potential, and thrombogenicity. The current evidence demonstrates that plaque vulnerability phenotypes are multiple and heterogeneous and are associated with many highly complex molecular pathways that determine the activation of an immune-mediated cascade that culminates in thromboinflammation. This narrative review provides a comprehensive analysis of the current knowledge on molecular biomarkers expressed by symptomatic carotid plaques. It explores the association of these biomarkers with the structural and compositional attributes that characterize vulnerable plaques.
Collapse
Affiliation(s)
- Giuseppe Miceli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Maria Grazia Basso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Chiara Pintus
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Andrea Roberta Pennacchio
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Elena Cocciola
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Mariagiovanna Cuffaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Martina Profita
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Giuliana Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| |
Collapse
|
20
|
Shen Y, Wang X, Gu N, Liu Z, Rong J, Shen C, Zhang W, Chen P, Deng Y, Deng C, Ma S, Zhao Y, Zhao R, Shi B. Association between periprocedural myocardial injury and neointimal characteristics in patients with in-stent restenosis: an optical coherence tomography study. Cardiovasc Diagn Ther 2024; 14:5-17. [PMID: 38434558 PMCID: PMC10904304 DOI: 10.21037/cdt-23-390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/05/2024] [Indexed: 03/05/2024]
Abstract
Background The relationship between neointimal characteristics of in-stent restenosis (ISR) and periprocedural myocardial injury (PMI) remains unclear. Therefore, this study aimed to investigate the relationship between PMI and neointimal characteristics of ISR by using optical coherence tomography (OCT). Methods This was a retrospective study. We enrolled 140 patients diagnosed with ISR with normal baseline high-sensitivity troponin T (hs-cTnT) levels who underwent OCT and subsequent revascularization by means of drug-coated balloon (DCB) or drug-eluting stent (DES) between October 2018 and October 2022 in the Affiliated Hospital of Zunyi Medical University. Based on the 4th universal definition of myocardial infarction, patients whose hs-cTnT were increased five times above the upper reference limit (URL) after percutaneous coronary interventions (PCI) were deemed to PMI. The patients were subdivided into PMI (n=53) and non-PMI (n=87) groups. In the univariable analysis, variables in the baselines, angiography characteristics and OCT findings were analyzed with binary logistic regression. A P value of <0.2 was included in the multivariable model. Multivariable logistic regression analysis was used to identify the independent predictors of PMI. Results The prevalence of intra-intimal microvessels in patients with PMI was higher than in those without PMI (58.5% vs. 32.2%, P=0.003). The ratio of intra-stent plaque rupture (PR) was also higher in patients with PMI (60.4% vs. 40.2%, P=0.021). Multivariable logistic regression analysis showed that intra-intimal microvessels [odds ratio (OR): 3.193, 95% confidence interval (CI): 1.280-7.966; P=0.013] and intra-stent PR (OR: 2.124, 95% CI: 1.153-4.732; P=0.035) were independently associated with PMI. Conclusions Intra-intimal microvessels and intra-stent PR were independently associated with PMI. Accurate identification and recognition of intra-intimal microvessels and intra-stent PR may be helpful in preventing PMI.
Collapse
Affiliation(s)
- Youcheng Shen
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xi Wang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ning Gu
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhijiang Liu
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jidong Rong
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Changyin Shen
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wei Zhang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Panke Chen
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yi Deng
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chancui Deng
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shuai Ma
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
21
|
Ma Y, Wang Q, Du S, Luo J, Sun X, Jia B, Ge J, Dong J, Jiang S, Li Z. Multipathway Regulation for Targeted Atherosclerosis Therapy Using Anti-miR-33-Loaded DNA Origami. ACS NANO 2024. [PMID: 38321605 DOI: 10.1021/acsnano.3c10213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Given the multifactorial pathogenesis of atherosclerosis (AS), a chronic inflammatory disease, combination therapy arises as a compelling approach to effectively address the complex interplay of pathogenic mechanisms for a more desired treatment outcome. Here, we present cRGD/ASOtDON, a nanoformulation based on a self-assembled DNA origami nanostructure for the targeted combination therapy of AS. cRGD/ASOtDON targets αvβ3 integrin receptors overexpressed on pro-inflammatory macrophages and activated endothelial cells in atherosclerotic lesions, alleviates the oxidative stress induced by extracellular and endogenous reactive oxygen species, facilitates the polarization of pro-inflammatory macrophages toward the anti-inflammatory M2 phenotype, and inhibits foam cell formation by promoting cholesterol efflux from macrophages by downregulating miR-33. The antiatherosclerotic efficacy and safety profile of cRGD/ASOtDON, as well as its mechanism of action, were validated in an AS mouse model. cRGD/ASOtDON treatment reversed AS progression and restored normal morphology and tissue homeostasis of the diseased artery. Compared to probucol, a clinical antiatherosclerotic drug with a similar mechanism of action, cRGD/ASOtDON enabled the desired therapeutic outcome at a notably lower dosage. This study demonstrates the benefits of targeted combination therapy in AS management and the potential of self-assembled DNA nanoformulations in addressing multifactorial inflammatory conditions.
Collapse
Affiliation(s)
- Yuxuan Ma
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Qi Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Shiyu Du
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Jingwei Luo
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Xiaolei Sun
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Bin Jia
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Jingru Ge
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Jun Dong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P. R. China
| | - Shuoxing Jiang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| | - Zhe Li
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu 210023, P. R. China
| |
Collapse
|
22
|
Los J, Mensink FB, Mohammadnia N, Opstal TSJ, Damman P, Volleberg RHJA, Peeters DAM, van Royen N, Garcia-Garcia HM, Cornel JH, El Messaoudi S, van Geuns RJM. Invasive coronary imaging of inflammation to further characterize high-risk lesions: what options do we have? Front Cardiovasc Med 2024; 11:1352025. [PMID: 38370159 PMCID: PMC10871865 DOI: 10.3389/fcvm.2024.1352025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/15/2024] [Indexed: 02/20/2024] Open
Abstract
Coronary atherosclerosis remains a leading cause of morbidity and mortality worldwide. The underlying pathophysiology includes a complex interplay of endothelial dysfunction, lipid accumulation and inflammatory pathways. Multiple structural and inflammatory features of the atherosclerotic lesions have become targets to identify high-risk lesions. Various intracoronary imaging devices have been developed to assess the morphological, biocompositional and molecular profile of the intracoronary atheromata. These techniques guide interventional and therapeutical management and allow the identification and stratification of atherosclerotic lesions. We sought to provide an overview of the inflammatory pathobiology of atherosclerosis, distinct high-risk plaque features and the ability to visualize this process with contemporary intracoronary imaging techniques.
Collapse
Affiliation(s)
- Jonathan Los
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Frans B. Mensink
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Tjerk S. J. Opstal
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Cardiology, Northwest Clinics, Alkmaar, Netherlands
| | - Peter Damman
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Denise A. M. Peeters
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Niels van Royen
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Jan H. Cornel
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Cardiology, Northwest Clinics, Alkmaar, Netherlands
- Dutch Network for Cardiovascular Research (WCN), Utrecht, Netherlands
| | - Saloua El Messaoudi
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | |
Collapse
|
23
|
Zhang L, Li Y, Yang W, Lin L, Li J, Liu D, Li C, Wu J, Li Y. Protocatechuic aldehyde increases pericyte coverage and mitigates pericyte damage to enhance the atherosclerotic plaque stability. Biomed Pharmacother 2023; 168:115742. [PMID: 37871558 DOI: 10.1016/j.biopha.2023.115742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/15/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023] Open
Abstract
Pericyte dysfunction and loss contribute substantially to the destabilization and rupture of atherosclerotic plaques. Protocatechuic aldehyde (PCAD), a natural polyphenol, exerts anti-atherosclerotic effects. However, the effects and mechanisms of this polyphenol on pericyte recruitment, coverage, and pericyte function remain unknown. We here treated apolipoprotein E-deficient mice having high-fat diet-induced atherosclerosis with PCAD. PCAD achieved therapeutic effects similar to rosuvastatin in lowering lipid levels and thus preventing atherosclerosis progression. With PCAD administration, plaque phenotype exhibited higher stability with markedly reduced lesion vulnerability, which is characterized by reduced lipid content and macrophage accumulation, and a consequent increase in collagen deposition. PCAD therapy increased pericyte coverage in the plaques, reduced VEGF-A production, and inhibited intraplaque neovascularization. PCAD promoted pericyte proliferation, adhesion, and migration to mitigate ox-LDL-induced pericyte dysfunction, which thus maintained the capillary network structure and stability. Furthermore, TGFBR1 silencing partially reversed the protective effect exerted by PCAD on human microvascular pericytes. PCAD increased pericyte coverage and impeded ox-LDL-induced damages through TGF-β1/TGFBR1/Smad2/3 signaling. All these novel findings indicated that PCAD increases pericyte coverage and alleviates pericyte damage to improve the stability of atherosclerotic plaques, which is accomplished by regulating TGF-β1/TGFBR1/Smad2/3 signaling in pericytes.
Collapse
Affiliation(s)
- Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yuan Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wenqing Yang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jie Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Dekun Liu
- Shool of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Yunlun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Shandong Provincial Engineering Laboratory of Traditional Chinese Medicine Precision Therapy for Cardiovascular Diseases, Jinan 250355, China.
| |
Collapse
|
24
|
Yan A, Gotlieb AI. The microenvironment of the atheroma expresses phenotypes of plaque instability. Cardiovasc Pathol 2023; 67:107572. [PMID: 37595697 DOI: 10.1016/j.carpath.2023.107572] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/06/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
Data from histopathology studies of human atherosclerotic tissue specimens and from vascular imaging studies support the concept that the local arterial microenvironment of a stable atheroma promotes destabilizing conditions that result in the transition to an unstable atheroma. Destabilization is characterized by several different plaque phenotypes that cause major clinical events such as acute coronary syndrome and cerebrovascular strokes. There are several rupture-associated phenotypes causing thrombotic vascular occlusion including simple fibrous cap rupture of an atheroma, fibrous cap rupture at site of previous rupture-and-repair of an atheroma, and nodular calcification with rupture. Endothelial erosion without rupture has more recently been shown to be a common phenotype to promote thrombosis as well. Microenvironment features that are linked to these phenotypes of plaque instability are neovascularization arising from the vasa vasorum network leading to necrotic core expansion, intraplaque hemorrhage, and cap rupture; activation of adventitial and perivascular adipose tissue cells leading to secretion of cytokines, growth factors, adipokines in the outer artery wall that destabilize plaque structure; and vascular smooth muscle cell phenotypic switching through transdifferentiation and stem/progenitor cell activation resulting in the promotion of inflammation, calcification, and secretion of extracellular matrix, altering fibrous cap structure, and necrotic core growth. As the technology evolves, studies using noninvasive vascular imaging will be able to investigate the transition of stable to unstable atheromas in real time. A limitation in the field, however, is that reliable and predictable experimental models of spontaneous plaque rupture and/or erosion are not currently available to study the cell and molecular mechanisms that regulate the conversion of the stable atheroma to an unstable plaque.
Collapse
Affiliation(s)
- Angela Yan
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Avrum I Gotlieb
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Mekke JM, Sakkers TR, Verwer MC, van den Dungen NAM, Song Y, Miller CL, Finn AV, Pasterkamp G, Mokry M, den Ruijter HM, Vink A, de Kleijn DPV, de Borst GJ, Haitjema S, van der Laan SW. The accumulation of erythrocytes quantified and visualized by Glycophorin C in carotid atherosclerotic plaque reflects intraplaque hemorrhage and pre-procedural neurological symptoms. Sci Rep 2023; 13:17104. [PMID: 37816779 PMCID: PMC10564864 DOI: 10.1038/s41598-023-43369-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023] Open
Abstract
The accumulation of erythrocyte membranes within an atherosclerotic plaque may contribute to the deposition of free cholesterol and thereby the enlargement of the necrotic core. Erythrocyte membranes can be visualized and quantified in the plaque by immunostaining for the erythrocyte marker glycophorin C. Hence, we theorized that the accumulation of erythrocytes quantified by glycophorin C could function as a marker for plaque vulnerability, possibly reflecting intraplaque hemorrhage (IPH), and offering predictive value for pre-procedural neurological symptoms. We employed the CellProfiler-integrated slideToolKit workflow to visualize and quantify glycophorin C, defined as the total plaque area that is positive for glycophorin C, in single slides of culprit lesions obtained from the Athero-Express Biobank of 1819 consecutive asymptomatic and symptomatic patients who underwent carotid endarterectomy. Our assessment included the evaluation of various parameters such as lipid core, calcifications, collagen content, SMC content, and macrophage burden. These parameters were evaluated using a semi-quantitative scoring method, and the resulting data was dichotomized as predefined criteria into categories of no/minor or moderate/heavy staining. In addition, the presence or absence of IPH was also scored. The prevalence of IPH and pre-procedural neurological symptoms were 62.4% and 87.1%, respectively. The amount of glycophorin staining was significantly higher in samples from men compared to samples of women (median 7.15 (IQR:3.37, 13.41) versus median 4.06 (IQR:1.98, 8.32), p < 0.001). Glycophorin C was associated with IPH adjusted for clinical confounders (OR 1.90; 95% CI 1.63, 2.21; p = < 0.001). Glycophorin C was significantly associated with ipsilateral pre-procedural neurological symptoms (OR:1.27, 95%CI:1.06-1.41, p = 0.005). Sex-stratified analysis, showed that this was also the case for men (OR 1.37; 95%CI 1.12, 1.69; p = 0.003), but not for women (OR 1.15; 95%CI 0.77, 1.73; p = 0.27). Glycophorin C was associated with classical features of a vulnerable plaque, such as a larger lipid core, a higher macrophage burden, less calcifications, a lower collagen and SMC content. There were marked sex differences, in men, glycophorin C was associated with calcifications and collagen while these associations were not found in women. To conclude, the accumulation of erythrocytes in atherosclerotic plaque quantified and visualized by glycophorin C was independently associated with the presence of IPH, preprocedural symptoms in men, and with a more vulnerable plaque composition in both men and women. These results strengthen the notion that the accumulation of erythrocytes quantified by glycophorin C can be used as a marker for plaque vulnerability.
Collapse
Affiliation(s)
- Joost M Mekke
- Division of Surgical Specialties, Department of Vascular Surgery, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Tim R Sakkers
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Maarten C Verwer
- Division of Surgical Specialties, Department of Vascular Surgery, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Noortje A M van den Dungen
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Yipei Song
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Clint L Miller
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22908, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, 22908, USA
| | | | - Gerard Pasterkamp
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Michal Mokry
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Dominique P V de Kleijn
- Division of Surgical Specialties, Department of Vascular Surgery, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
- Netherlands Heart Institute, Moreelsepark 1, 3511 EP, Utrecht, The Netherlands
| | - Gert J de Borst
- Division of Surgical Specialties, Department of Vascular Surgery, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Saskia Haitjema
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Sander W van der Laan
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
26
|
Chen D, Zhao Z, Liu P, Liu X, Wang X, Ren Q, Chang B. Adventitial Vasa Vasorum Neovascularization in Femoral Artery of Type 2 Diabetic Patients with Macroangiopathy Is Associated with Macrophages and Lymphocytes as well as the Occurrence of Cardiovascular Events. Thromb Haemost 2023; 123:989-998. [PMID: 37037199 DOI: 10.1055/s-0043-1768162] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
OBJECTIVES This study was conducted to assess the relationship between adventitial vasa vasorum neovascularization (VVn) in femoral artery of type 2 diabetic patients with macroangiopathy and the recruitment of macrophages and lymphocytes, and to relate the density of VVn to the occurrence of cardiovascular events. MATERIALS Femoral artery samples were obtained from amputation cases. A total of 55 type 2 diabetic patients with macroangiopathy, 15 autopsy cases with type 2 diabetes without atherosclerosis. METHODS Hematoxylin and eosin (H&E) staining to observe the histopathological features; Victoria blue staining to analyze the histological features; immunohistochemistry (CD34, CD68, CD20, and CD3) to determine the VVn density and the expression of macrophages, B lymphocytes, and T lymphocytes. RESULTS Type 2 diabetic patients with macroangiopathy showed a higher mean adventitial VVn density in femoral artery (48.40 ± 9.39 no./mm2) than patients with type 2 diabetes without atherosclerosis (19.75 ± 6.28 no./mm2) (p < 0.01). In addition, the VVn density was positively associated with the expression of CD68 macrophages (r = 0.62, p < 0.01) and CD20 B lymphocytes (r = 0.59, p < 0.01). Type 2 diabetic patients with high VVn density showed more adverse cardiovascular events (27/35 vs. 8/20 events, p = 0.006). In multivariable analysis adjusted for main risk factors for cardiovascular disease, VVn was still independently associated with adverse cardiovascular events (p = 0.01). CONCLUSION VVn density in type 2 diabetic patients with macroangiopathy is positively correlated with the adventitial immune-inflammatory cell numbers and the development of atherosclerotic lesions. Furthermore, VVn density is associated with adverse cardiovascular events.
Collapse
Affiliation(s)
- Dong Chen
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Zixi Zhao
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Peng Liu
- Department of Surgery, Binhai New Area Hospital of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Xinbang Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Xin Wang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Qiuyue Ren
- Department of Endocrinology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People's Republic of China
| | - Bai Chang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
27
|
Burke-Kleinman J, Gotlieb AI. Progression of Arterial Vasa Vasorum from Regulator of Arterial Homeostasis to Promoter of Atherogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1468-1484. [PMID: 37356574 DOI: 10.1016/j.ajpath.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 06/27/2023]
Abstract
The vasa vasorum (vessels of vessels) are a dynamic microvascular system uniquely distributed to maintain physiological homeostasis of the artery wall by supplying nutrients and oxygen to the outer layers of the artery wall, adventitia, and perivascular adipose tissue, and in large arteries, to the outer portion of the medial layer. Vasa vasorum endothelium and contractile mural cells regulate direct access of bioactive cells and factors present in both the systemic circulation and the arterial perivascular adipose tissue and adventitia to the artery wall. Experimental and human data show that proatherogenic factors and cells gain direct access to the artery wall via the vasa vasorum and may initiate, promote, and destabilize the plaque. Activation and growth of vasa vasorum occur in all blood vessel layers primarily by angiogenesis, producing fragile and permeable new microvessels that may cause plaque hemorrhage and fibrous cap rupture. Ironically, invasive therapies, such as angioplasty and coronary artery bypass grafting, injure the vasa vasorum, leading to treatment failures. The vasa vasorum function both as a master integrator of arterial homeostasis and, once perturbed or injured, as a promotor of atherogenesis. Future studies need to be directed at establishing reliable in vivo and in vitro models to investigate the cellular and molecular regulation of the function and dysfunction of the arterial vasa vasorum.
Collapse
Affiliation(s)
- Jonah Burke-Kleinman
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Avrum I Gotlieb
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Avagimyan A, Pogosova N, Kakturskiy L, Sheibani M, Urazova O, Trofimenko A, Navarsdyan G, Jndoyan Z, Abgaryan K, Fogacci F, Galli M, Agati L, Kobalava Z, Shafie D, Marzilli M, Gogiashvili L, Sarrafzadegan N. HIV-Related Atherosclerosis: State-of-the-Art-Review. Curr Probl Cardiol 2023; 48:101783. [PMID: 37172874 DOI: 10.1016/j.cpcardiol.2023.101783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
The infection caused by the Human Immunodeficiency Virus (HIV) has spread rapidly across the globe, assuming the characteristics of an epidemic in some regions. Thanks to the introduction of antiretroviral therapy into routine clinical practice, there was a considerable breakthrough in the treatment of HIV, that is now HIV is potentially well-controlled even in low-income countries. To date, HIV infection has moved from the group of life-threatening conditions to the group of chronic and well controlled ones and the quality of life and life expectancy of HIV+ people, with an undetectable viral load is closer to that of an HIV- people. However, unsolved issues still persist. For example: people living with HIV are more prone to the age-related diseases, especially atherosclerosis. For this reason, a better understanding of the mechanisms of HIV-associated destabilization of vascular homeostasis seems to be an urgent duty, that may lead to the development of new protocols, bringing the possibilities of pathogenetic therapies to a new level. The purpose of the article was to evaluate the pathological aspects of HIV-induced atherosclerosis.
Collapse
Affiliation(s)
- Ashot Avagimyan
- Assistant Professor, Anatomical Pathology and Clinical Morphology Department, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia.
| | - Nana Pogosova
- Professor, Deputy of General Director for Science and Preventive Cardiology, National Medical Research Centre of Cardiology after E. Chazov, Moscow, Russia
| | - Lev Kakturskiy
- Professor, Scientific Director, Research Institute of Human Morphology FSBI «Petrovskiy NRCS, Moscow, Russia
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Razi Drug Research Centre, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Olga Urazova
- Professor, Head of Pathophysiology Department, Siberian State Medical University, Tomsk, Russia
| | - Artem Trofimenko
- Associate Professor, Pathophysiology Department, Kuban State Medical University, Krasnodar, Russia
| | - Grizelda Navarsdyan
- Professor, Pathophysiology Department, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
| | - Zinaida Jndoyan
- Professor, Head of Internal Diseases Propedeutics Department, Yerevan State Medical University after M. Heratsi, Armenia
| | - Kristina Abgaryan
- Associate Professor, Medical Microbiology Department, Yerevan State Medical University after M.Heratsi, Armenia
| | - Federica Fogacci
- Research Fellow, Atherosclerosis and Metabolic Disorders Research Unit, University of Bologna, Bologna, Italy
| | - Mattia Galli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Luciano Agati
- Professor of Cardiology Department, Head of Cardiology Unit Azienda Policlinico Umberto II, Sapienza University, Rome, Italy
| | - Zhanna Kobalava
- Professor, Head of Internal Disease, Cardiology and Clinical Pharmacology Department, Peoples' Friendship University of Russia (RUDN), Moscow, Russia
| | - Davood Shafie
- Isfahan Cardiovascular Research Institute, Isfahan, Iran
| | - Mario Marzilli
- Professor, Head of Cardiovascular Medicine Division, University of Pisa, Pisa, Italy
| | - Liana Gogiashvili
- Professor, Head of Experimental and Clinical Pathology Department, Al. Natishvili Institute of Experimental Morphology, I. Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Nizal Sarrafzadegan
- Professor, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
29
|
Lin PK, Davis GE. Extracellular Matrix Remodeling in Vascular Disease: Defining Its Regulators and Pathological Influence. Arterioscler Thromb Vasc Biol 2023; 43:1599-1616. [PMID: 37409533 PMCID: PMC10527588 DOI: 10.1161/atvbaha.123.318237] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/23/2023] [Indexed: 07/07/2023]
Abstract
Because of structural and cellular differences (ie, degrees of matrix abundance and cross-linking, mural cell density, and adventitia), large and medium-sized vessels, in comparison to capillaries, react in a unique manner to stimuli that induce vascular disease. A stereotypical vascular injury response is ECM (extracellular matrix) remodeling that occurs particularly in larger vessels in response to injurious stimuli, such as elevated angiotensin II, hyperlipidemia, hyperglycemia, genetic deficiencies, inflammatory cell infiltration, or exposure to proinflammatory mediators. Even with substantial and prolonged vascular damage, large- and medium-sized arteries, persist, but become modified by (1) changes in vascular wall cellularity; (2) modifications in the differentiation status of endothelial cells, vascular smooth muscle cells, or adventitial stem cells (each can become activated); (3) infiltration of the vascular wall by various leukocyte types; (4) increased exposure to critical growth factors and proinflammatory mediators; and (5) marked changes in the vascular ECM, that remodels from a homeostatic, prodifferentiation ECM environment to matrices that instead promote tissue reparative responses. This latter ECM presents previously hidden matricryptic sites that bind integrins to signal vascular cells and infiltrating leukocytes (in coordination with other mediators) to proliferate, invade, secrete ECM-degrading proteinases, and deposit injury-induced matrices (predisposing to vessel wall fibrosis). In contrast, in response to similar stimuli, capillaries can undergo regression responses (rarefaction). In summary, we have described the molecular events controlling ECM remodeling in major vascular diseases as well as the differential responses of arteries versus capillaries to key mediators inducing vascular injury.
Collapse
Affiliation(s)
- Prisca K. Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - George E. Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| |
Collapse
|
30
|
Sakamoto A, Suwa K, Kawakami R, Finn AV, Maekawa Y, Virmani R, Finn AV. Significance of Intra-plaque Hemorrhage for the Development of High-Risk Vulnerable Plaque: Current Understanding from Basic to Clinical Points of View. Int J Mol Sci 2023; 24:13298. [PMID: 37686106 PMCID: PMC10487895 DOI: 10.3390/ijms241713298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Acute coronary syndromes due to atherosclerotic coronary artery disease are a leading cause of morbidity and mortality worldwide. Intra-plaque hemorrhage (IPH), caused by disruption of intra-plaque leaky microvessels, is one of the major contributors of plaque progression, causing a sudden increase in plaque volume and eventually plaque destabilization. IPH and its healing processes are highly complex biological events that involve interactions between multiple types of cells in the plaque, including erythrocyte, macrophages, vascular endothelial cells and vascular smooth muscle cells. Recent investigations have unveiled detailed molecular mechanisms by which IPH leads the development of high-risk "vulnerable" plaque. Current advances in clinical diagnostic imaging modalities, such as magnetic resonance image and intra-coronary optical coherence tomography, increasingly allow us to identify IPH in vivo. To date, retrospective and prospective clinical trials have revealed the significance of IPH as detected by various imaging modalities as a reliable prognostic indicator of high-risk plaque. In this review article, we discuss recent advances in our understanding for the significance of IPH on the development of high-risk plaque from basic to clinical points of view.
Collapse
Affiliation(s)
- Atsushi Sakamoto
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu 431-3125, Japan; (K.S.); (Y.M.)
| | - Kenichiro Suwa
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu 431-3125, Japan; (K.S.); (Y.M.)
| | - Rika Kawakami
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| | - Alexandra V. Finn
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| | - Yuichiro Maekawa
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu 431-3125, Japan; (K.S.); (Y.M.)
| | - Renu Virmani
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| | - Aloke V. Finn
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| |
Collapse
|
31
|
Shen Y, Xu M, Ren L, Li X, Han X, Cao X, Yao J, Yan B. A novel retinoic acid drug, EYE-502, inhibits choroidal neovascularization by targeting endothelial cells and pericytes. Sci Rep 2023; 13:10439. [PMID: 37369771 DOI: 10.1038/s41598-023-37619-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 06/24/2023] [Indexed: 06/29/2023] Open
Abstract
Choroidal neovascularization (CNV) occurs in neovascular age-related macular degeneration (AMD) and often leads to permanent visual impairment. Intravitreal injection of anti-vascular endothelial growth factor (VEGF) agents is the gold standard for the treatment of CNV. However, anti-VEGF treatment did not always cause vision improvement and sometimes had detrimental effects on normal retinal tissues. Herein, we identified a novel retinoic acid drug, EYE-502, which had great therapeutic effects on CNV. Administration of EYE-502 could inhibit VEGF-induced dysfunction of endothelial cells (ECs) and reduce platelet-derived growth factor (PDGF)-induced recruitment of pericytes to ECs in vitro. Administration of EYE-502 could reduce the area of choroidal sprouting and laser-induced CNV, exhibiting similar anti-angiogenic effects as aflibercept. Moreover, administration of EYE-502 could reduce pericyte coverage in the sprouting vessels and choroidal neovascularization. Mechanistically, EYE-502 primarily bound to retinoic acid receptors (RARs) and exerted the anti-angiogenic effects by targeting ECs and pericytes via affecting the activation of Wnt/β-catenin and PDGF/PDGFR/PI3K/Akt signaling. Taken together, this study reports a novel retinoic acid drug, EYE-502, which can exert the anti-angiogenic effects by simultaneous targeting of ECs and pericytes.
Collapse
Affiliation(s)
- Yaming Shen
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Miao Xu
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Ling Ren
- Eye Institute, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiumiao Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaoyan Han
- Eye Institute, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xin Cao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jin Yao
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China.
| | - Biao Yan
- Eye Institute, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
32
|
Brown PA, Brown PD. Extracellular vesicles and atherosclerotic peripheral arterial disease. Cardiovasc Pathol 2023; 63:107510. [PMID: 36460259 DOI: 10.1016/j.carpath.2022.107510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/24/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
Atherogenesis involves a complex multifactorial process including chronic inflammation that requires the participation of several cell types and molecules. In addition to their role in vascular homeostasis, extracellular vesicles also appear to play an important role in atherogenesis, including monocyte transmigration and foam cell formation, SMC proliferation and migration, leukocyte transmigration, and thrombosis. Peripheral arterial disease, a major form of peripheral vascular disease, is characterized by structural or functional impairment of peripheral arterial supply, often secondary to atherosclerosis. Elevated levels of extracellular vesicles have been demonstrated in patients with peripheral arterial disease and implicated in the development of atherosclerosis within peripheral vascular beds. However, extracellular vesicles also appear capable of delivering cargo with atheroprotective effects. This capability has been exploited in vesicles engineered to carry content capable of neovascularization, suggesting potential for therapeutic angiogenesis. This dual capacity holds substantial promise for diagnosis and therapy, including possibly limb- and life-saving options for peripheral arterial disease management.
Collapse
Affiliation(s)
- Paul A Brown
- Department of Basic Medical Sciences, University of the West Indies, Mona, Jamaica.
| | - Paul D Brown
- Department of Basic Medical Sciences, University of the West Indies, Mona, Jamaica
| |
Collapse
|
33
|
Tissue oxygenation stabilizes neovessels and mitigates hemorrhages in human atherosclerosis-induced angiogenesis. Angiogenesis 2023; 26:63-76. [PMID: 35947328 DOI: 10.1007/s10456-022-09851-8] [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: 04/01/2022] [Accepted: 07/21/2022] [Indexed: 11/01/2022]
Abstract
Progression of atherosclerosis is associated with a maladaptive form of angiogenesis which contributes to intraplaque hemorrhage and plaque disruption. Hypoxia has been implicated in mechanisms of angiogenic neovessel fragility and atherosclerotic plaque destabilization. We used ex vivo and in vivo models to characterize the effect of oxygen (O2) on the formation, stability and tendency to bleed of human plaque-induced neovessels. Plaque explants potently stimulated the ex vivo angiogenic response of rat aortic rings at atmospheric O2 levels. Severe hypoxia (1% O2) inhibited plaque-induced angiogenesis and pericyte recruitment causing neovessel breakdown, whereas increasing O2 levels dose dependently enhanced pericyte numbers and neovessel stability. Plaque fragments implanted subcutaneously with or without aortic rings in SCID mice stimulated the host angiogenic response with plaques causing minimal or no hemorrhages and plaques co-implanted with aortic rings causing marked hemorrhages. Plaque/aortic ring-induced hemorrhages were reduced in mice exposed to moderate hyperoxia (50% O2). Hyperoxia downregulated expression of the hypoxia-sensitive genes Ca9, Ca12 and VegfA and increased influx into implants of mesenchymal cells reactive for the pericyte marker NG2. In both ex vivo and in vivo models, O2 promoted expression of vasostabilizing genes required for pericyte recruitment (Angpt1, Pdgfb), basement membrane assembly (Col4A1), and tight junction formation (Cldn5 and/or Ocln). Our results suggest that formation of neovessels that are stable, pericyte-coated, and resistant to bleeding requires adequate tissue oxygenation. Understanding the mechanisms by which O2 stabilizes neovessels and mitigates neovessel bleeding may lead to new therapies for the prevention of atherosclerosis complications.
Collapse
|
34
|
Wang JY, Liu K, Wang YB, Deng YB, Sun J. Effects of norepinephrine on plaque hypoxia in atherosclerotic rabbits. Front Cardiovasc Med 2023; 10:1121413. [PMID: 36873394 PMCID: PMC9974659 DOI: 10.3389/fcvm.2023.1121413] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Background Hypoxia plays a vital role throughout the whole process of atherosclerotic vulnerable plaque formation, which may be induced by a reduced oxygen supply. The vasa vasorum can be affected by norepinephrine (NE) and cause a reduced oxygen supply, ultimately leading to plaque hypoxia. This study aimed to investigate the effects of norepinephrine, which can increase the tension of the vasa vasorum, on plaque hypoxia, evaluated by contrast-enhanced ultrasound imaging. Methods Atherosclerosis (AS) was induced in New Zealand white rabbits by a combination of a cholesterol-rich diet and aortic balloon dilation. After the atherosclerotic model was well established, NE was intravenously administered three times per day for 2 weeks. Contrast-enhanced ultrasound (CEUS) and immunohistochemistry staining were performed to evaluate the expression of hypoxia-inducible factor alpha (HIF-α) and vascular endothelial growth factor (VEGF) in atherosclerotic plaques. Results The plaque blood flow decreased after long-term norepinephrine administration. The expression of HIF-α and VEGF in atherosclerotic plaques concentrated in the outer medial layers increased, which indicated that NE might cause plaque hypoxia by contraction of the vasa vasorum. Conclusion Apparent hypoxia of atherosclerotic plaques after long-term NE administration was mainly caused by decreased plaque blood flow due to the contraction of the vasa vasorum and high blood pressure.
Collapse
Affiliation(s)
- Jia-Yu Wang
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Liu
- Department of Medical Ultrasound, The Third People's Hospital of Hubei, Wuhan, China
| | - Yu-Bo Wang
- Department of Medical Ultrasound, Maternal and Child Healthcare Hospital of Hubei, Wuhan, China
| | - You-Bin Deng
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Sun
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Current Concepts and Future Applications of Non-Invasive Functional and Anatomical Evaluation of Coronary Artery Disease. Life (Basel) 2022; 12:life12111803. [PMID: 36362957 PMCID: PMC9696378 DOI: 10.3390/life12111803] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/31/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Over the last decades, significant advances have been achieved in the treatment of coronary artery disease (CAD). Proper non-invasive diagnosis and appropriate management based on functional information and the extension of ischemia or viability remain the cornerstone in the fight against adverse CAD events. Stress echocardiography and single photon emission computed tomography are often used for the evaluation of ischemia. Advancements in non-invasive imaging modalities such as computed tomography (CT) coronary angiography and cardiac magnetic resonance imaging (MRI) have not only allowed non-invasive imaging of coronary artery lumen but also provide additional functional information. Other characteristics regarding the plaque morphology can be further evaluated with the latest modalities achieving a morpho-functional evaluation of CAD. Advances in the utilization of positron emission tomography (PET), as well as software advancements especially regarding cardiac CT, may provide additional prognostic information to a more evidence-based treatment decision. Since the armamentarium on non-invasive imaging modalities has evolved, the knowledge of the capabilities and limitations of each imaging modality should be evaluated in a case-by-case basis to achieve the best diagnosis and treatment decision. In this review article, we present the most recent advances in the noninvasive anatomical and functional evaluation of CAD.
Collapse
|
36
|
Lee J, Kim JN, Gomez-Perez L, Gharaibeh Y, Motairek I, Pereira GTR, Zimin VN, Dallan LAP, Hoori A, Al-Kindi S, Guagliumi G, Bezerra HG, Wilson DL. Automated Segmentation of Microvessels in Intravascular OCT Images Using Deep Learning. Bioengineering (Basel) 2022; 9:648. [PMID: 36354559 PMCID: PMC9687448 DOI: 10.3390/bioengineering9110648] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 09/03/2024] Open
Abstract
Microvessels in vascular plaque are associated with plaque progression and are found in plaque rupture and intra-plaque hemorrhage. To analyze this characteristic of vulnerability, we developed an automated deep learning method for detecting microvessels in intravascular optical coherence tomography (IVOCT) images. A total of 8403 IVOCT image frames from 85 lesions and 37 normal segments were analyzed. Manual annotation was performed using a dedicated software (OCTOPUS) previously developed by our group. Data augmentation in the polar (r,θ) domain was applied to raw IVOCT images to ensure that microvessels appear at all possible angles. Pre-processing methods included guidewire/shadow detection, lumen segmentation, pixel shifting, and noise reduction. DeepLab v3+ was used to segment microvessel candidates. A bounding box on each candidate was classified as either microvessel or non-microvessel using a shallow convolutional neural network. For better classification, we used data augmentation (i.e., angle rotation) on bounding boxes with a microvessel during network training. Data augmentation and pre-processing steps improved microvessel segmentation performance significantly, yielding a method with Dice of 0.71 ± 0.10 and pixel-wise sensitivity/specificity of 87.7 ± 6.6%/99.8 ± 0.1%. The network for classifying microvessels from candidates performed exceptionally well, with sensitivity of 99.5 ± 0.3%, specificity of 98.8 ± 1.0%, and accuracy of 99.1 ± 0.5%. The classification step eliminated the majority of residual false positives and the Dice coefficient increased from 0.71 to 0.73. In addition, our method produced 698 image frames with microvessels present, compared with 730 from manual analysis, representing a 4.4% difference. When compared with the manual method, the automated method improved microvessel continuity, implying improved segmentation performance. The method will be useful for research purposes as well as potential future treatment planning.
Collapse
Affiliation(s)
- Juhwan Lee
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Justin N Kim
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Lia Gomez-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Yazan Gharaibeh
- Department of Biomedical Engineering, Faculty of Engineering, The Hashemite University, Zarqa, 13133, Jordan
| | - Issam Motairek
- Cardiovascular Imaging Core Laboratory, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Gabriel T R Pereira
- Cardiovascular Imaging Core Laboratory, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Vladislav N Zimin
- Cardiovascular Imaging Core Laboratory, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Luis A P Dallan
- Cardiovascular Imaging Core Laboratory, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Ammar Hoori
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sadeer Al-Kindi
- Cardiovascular Imaging Core Laboratory, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Giulio Guagliumi
- Cardiovascular Department, Galeazzi San'Ambrogio Hospital, Innovation District Milan, 20157 Milan, Italy
| | - Hiram G Bezerra
- Interventional Cardiology Center, Heart and Vascular Institute, University of South Florida, Tampa, FL 33606, USA
| | - David L Wilson
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Radiology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
37
|
Zhao XQ, Sun J, Hippe DS, Isquith DA, Canton G, Yamada K, Balu N, Crouse JR, Anderson TJ, Huston J, O’Brien KD, Hatsukami TS, Yuan C. Magnetic Resonance Imaging of Intraplaque Hemorrhage and Plaque Lipid Content With Continued Lipid-Lowering Therapy: Results of a Magnetic Resonance Imaging Substudy in AIM-HIGH. Circ Cardiovasc Imaging 2022; 15:e014229. [PMID: 36378778 PMCID: PMC9773914 DOI: 10.1161/circimaging.122.014229] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Intraplaque hemorrhage (IPH) is associated with plaque progression and ischemic events, and plaque lipid content (% lipid core) predicts the residual atherosclerotic cardiovascular disease risk. This study examined the impact of IPH on lipid content change in the setting of intensive lipid-lowering therapy. METHODS In total, 214 AIM-HIGH (Atherothrombosis Intervention in Metabolic Syndrome with Low High-Density Lipoprotein/High Triglycerides: Impact on Global Health Outcomes) participants with clinically established ASCVD and low high-density lipoprotein cholesterol received cartoid MRI at baseline and 2 years to assess changes in carotid morphology and composition. Patients were randomized to extended-release niacin or placebo, and all received simvastatin with optional ezetimibe as necessary to lower low-density lipoprotein cholesterol to 40 to 80 mg/dL. Changes in lipid content and carotid morphology were tested using the Wilcoxon signed-rank test. Differences between subjects with and without IPH and between subjects assigned extended-release niacin or placebo were tested using the Wilcoxon rank-sum test. Linear regression was used to test the association of IPH and lipid content changes after adjusting for clinical risk factors. RESULTS Among 156 patients (61±9 years; 81% men) with complete MRI, prior statin use: <1 year, 26%; 1 to 5 years, 37%; >5 years, 37%. Triglycerides and ApoB decreased significantly, whereas high-density lipoprotein cholesterol and ApoA1 increased significantly over time. Plaque lipid content was significantly reduced (-0.5±2.4 %/year, P = 0.017) without a significant difference between the 2 treatment groups. However, the lipid content increased in plaques with IPH but regressed in plaques without IPH (1.2±2.5 %/year versus -1.0±2.2, P = 0.006). Additionally, IPH was associated with a decrease in lumen area (-0.4±0.9 mm2/year versus 0.3±1.4, P = 0.033). IPH remained significantly associated with increase in lipid content in multivariable analysis (54.4%, 95% CI: 26.8, 88.0, P < 0.001). CONCLUSIONS Carotid plaques under continued intensive lipid-lowering therapy moved toward stabilization. However, plaques with IPH showed greater increases in lipid content and greater decreases in lumen area than plaques without IPH. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT01178320.
Collapse
Affiliation(s)
- Xue-Qiao Zhao
- Department of Medicine (Division of Cardiology), University of Washington, Seattle, Washington
| | - Jie Sun
- Department of Radiology, University of Washington, Seattle, Washington
| | - Daniel S. Hippe
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Daniel A. Isquith
- Department of Medicine (Division of Cardiology), University of Washington, Seattle, Washington
| | - Gador Canton
- Department of Radiology, University of Washington, Seattle, Washington
| | - Kiyofumi Yamada
- Department of Radiology, University of Washington, Seattle, Washington
| | - Niranjan Balu
- Department of Radiology, University of Washington, Seattle, Washington
| | - John R. Crouse
- Department of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Todd J. Anderson
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - John Huston
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Kevin D. O’Brien
- Department of Medicine (Division of Cardiology), University of Washington, Seattle, Washington
| | - Thomas S. Hatsukami
- Department of Surgery (Division of Vascular Surgery), University of Washington, Seattle, Washington
| | - Chun Yuan
- Department of Radiology, University of Washington, Seattle, Washington
| | | |
Collapse
|
38
|
Bogdanov L, Shishkova D, Mukhamadiyarov R, Velikanova E, Tsepokina A, Terekhov A, Koshelev V, Kanonykina A, Shabaev A, Frolov A, Zagorodnikov N, Kutikhin A. Excessive Adventitial and Perivascular Vascularisation Correlates with Vascular Inflammation and Intimal Hyperplasia. Int J Mol Sci 2022; 23:ijms232012156. [PMID: 36293013 PMCID: PMC9603343 DOI: 10.3390/ijms232012156] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 12/24/2022] Open
Abstract
Albeit multiple studies demonstrated that vasa vasorum (VV) have a crucial importance in vascular pathology, the informative markers and metrics of vascular inflammation defining the development of intimal hyperplasia (IH) have been vaguely studied. Here, we employed two rat models (balloon injury of the abdominal aorta and the same intervention optionally complemented with intravenous injections of calciprotein particles) and a clinical scenario (arterial and venous conduits for coronary artery bypass graft (CABG) surgery) to investigate the pathophysiological interconnections among VV, myeloperoxidase-positive (MPO+) clusters, and IH. We found that the amounts of VV and MPO+ clusters were strongly correlated; further, MPO+ clusters density was significantly associated with balloon-induced IH and increased at calciprotein particle-provoked endothelial dysfunction. Likewise, number and density of VV correlated with IH in bypass grafts for CABG surgery at the pre-intervention stage and were higher in venous conduits which more frequently suffered from IH as compared with arterial grafts. Collectively, our results underline the pathophysiological importance of excessive VV upon the vascular injury or at the exposure to cardiovascular risk factors, highlight MPO+ clusters as an informative marker of adventitial and perivascular inflammation, and propose another mechanistic explanation of a higher long-term patency of arterial grafts upon the CABG surgery.
Collapse
|
39
|
Li M, Yun W, Wang G, Li A, Gao J, He Q. Roles and mechanisms of garlic and its extracts on atherosclerosis: A review. Front Pharmacol 2022; 13:954938. [PMID: 36263122 PMCID: PMC9574545 DOI: 10.3389/fphar.2022.954938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
The prevention and treatment of cardiovascular diseases (CVDs) have achieved initial results, but the number of CVDs patients will increase rapidly in the next 10 years. Atherosclerosis (AS) is a significant risk factor for CVDs. The impact of lifestyle and daily diet varies considerably between different countries and continents and has been shown to affect the development of various diseases such as diabetes and CVDs. Primary and secondary prevention using alternative supplements and methods to avoid or reduce the use of traditional pharmacological drugs have also become popular. One of the reasons for this is that pharmacological drugs with lipid-lowering, and blood pressure-lowering effects cause many side effects that may negatively impact the quality of life. Patients are now emphasizing reliance on lifestyle changes to reduce cardiovascular risks. Garlic is a medicinal and edible plant that has been used for a long time. In order to reveal garlic application in the prevention and treatment of AS, reviewing the latest domestic and international studies through searching databases. The result shows that the antiatherogenic role of garlic is eximious. And the mechanisms are mainly related to hypolipidemic, antioxidant, antithrombotic, inhibiting angiogenesis, protecting endothelial cells, anti-inflammatory, anti-apoptotic, inhibiting vascular smooth muscle proliferation, and regulating gut microbiota. The main signaling pathways involve AMPK/TLRs, Keap1/Nrf2, PI3K/AKT, PPARγ/LXRα, GEF-H1/RhoA/Rac, etc. The antiatherogenic actions and molecular mechanism of garlic were reviewed in this study to obtain a robust evidence basis for the clinical application and mechanistic study and provide a theoretical basis for further utilization of garlic.
Collapse
Affiliation(s)
- Min Li
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wingyan Yun
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Clinical Department of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Guibin Wang
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Anqi Li
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Clinical Department of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Gao
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Clinical Department of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingyong He
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Qingyong He,
| |
Collapse
|
40
|
Kawai K, Vozenilek AE, Kawakami R, Sato Y, Ghosh SKB, Virmani R, Finn AV. Understanding the role of alternative macrophage phenotypes in human atherosclerosis. Expert Rev Cardiovasc Ther 2022; 20:689-705. [PMID: 35942866 DOI: 10.1080/14779072.2022.2111301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
INTRODUCTION Atherosclerosis-based ischemic heart disease is still the primary cause of death throughout the world. Over the past decades there has been no significant changes in the therapeutic approaches to atherosclerosis, which are mainly based on lipid lowering therapies and management of comorbid conditions such as diabetes and hypertension. The involvement of macrophages in atherosclerosis has been recognized for decades. More recently, a more detailed and sophisticated understanding of their various phenotypes and roles in the atherosclerotic process has been recognized. This new data is revealing how specific subtypes of macrophage-induced inflammation may have distinct effects on atherosclerosis progression and may provide new approaches for treatment, based upon targeting of specific macrophage subtypes. AREAS COVERED We will comprehensively review the spectrum of macrophage phenotypes and how they contribute to atherosclerotic plaque development and progression. EXPERT OPINION Various signals derived from atherosclerotic lesions drive macrophages into complex subsets with different gene expression profiles, phenotypes, and functions, not all of which are understood. Macrophage phenotypes include those that enhance, heal, and regress the atherosclerotic lesions though various mechanisms. Targeting of specific macrophage phenotypes may provide a promising and novel approach to prevent atherosclerosis progression.
Collapse
Affiliation(s)
- Kenji Kawai
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aimee E Vozenilek
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Yu Sato
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | | | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aloke V Finn
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA.,University of Maryland, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
41
|
Gong F, Wang Z, Mo R, Wang Y, Su J, Li X, Omonova CTQ, Khamis AM, Zhang Q, Dong M, Su Z. Nano-sponge-like liposomes remove cholesterol crystals for antiatherosclerosis. J Control Release 2022; 349:940-953. [PMID: 35870569 DOI: 10.1016/j.jconrel.2022.07.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/25/2022] [Accepted: 07/18/2022] [Indexed: 10/15/2022]
Abstract
Atherosclerotic cardiovascular diseases remain the leading causes of morbidity and mortality worldwide. Cholesterol crystals in atherosclerotic plaques play an essential role in atherosclerosis progression. However, no clinical drugs have been used for removing cholesterol crystals from plaque to counter atherosclerosis. Previous studies identified the hydrophobic domain of lipid bilayer in liposomes acted as sinks for solubilizing hydrophobic cholesterol. Moreover, adjusting the composition of the lipid bilayer in liposomes can enhance its hydrophobic molecule loading capacity. Therefore, in this study, ginsenosides Rb1 (Rb1), one of main active components of ginseng which has a similar structure to cholesterol, is anchored into soy phospholipids bilayer with its hydrophobic region to prepare nano-sponge-like liposomes (Rb1-LPs), aiming to amplify the solubilization of cholesterol in lipid bilayer. For targeting delivery to atherosclerotic plaques, Annexin V (AnxV), a protein that can specifically recognize phosphatidylserine upregulated in atherosclerotic plaques, is applied to decorate the surface of Rb1-LPs by click reaction to obtain the final preparation of AnxV-Rb1-LPs. The in vitro studies showed that incorporating Rb1 into lipid bilayer remarkably increased the affinity of the lipid bilayer to free cholesterol and the solubilization of cholesterol crystals. Additionally, nano-sponge-like liposomes could efficiently reduce the accumulation of cholesterol crystals and improve cholesterol efflux, finally inhibiting inflammation and apoptosis in cholesterol-laden cells. Furthermore, AnxV-Rb1-LPs could efficiently accumulate in atherosclerotic plaques after intravenous injection, exert nano-sponge-like functions to remove intra- and extracellular cholesterol crystals, ultimately alleviating inflammation and apoptosis in atherosclerotic plaques for antiatherosclerosis. Therefore, AnxV-Rb1-LPs provide a potential strategy for removing cholesterol crystals in atherosclerotic plaques and can be further utilized in other diseases with excessive cholesterol accumulation.
Collapse
Affiliation(s)
- Fanglin Gong
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Zibin Wang
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Rui Mo
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yutong Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Jin Su
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xianglong Li
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Charos Tuychi Qizi Omonova
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Amari Mohamed Khamis
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Qing Zhang
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, PR China.
| | - Mei Dong
- Jiangsu Provincial Enginerring Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Zhigui Su
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
42
|
Fukushima D, Kondo K, Harada N, Terazono S, Uchino K, Shibuya K, Sugo N. Quantitative comparison between carotid plaque hardness and histopathological findings: an observational study. Diagn Pathol 2022; 17:58. [PMID: 35818059 PMCID: PMC9275256 DOI: 10.1186/s13000-022-01239-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 06/12/2022] [Indexed: 12/02/2022] Open
Abstract
Background Plaque hardness in carotid artery stenosis correlates with cerebral infarction. This study aimed to quantitatively compare plaque hardness with histopathological findings and identify the pathological factors involved in plaque hardness. Methods This study included 84 patients (89 lesions) undergoing carotid endarterectomy (CEA) at our institution. Plaque hardness was quantitatively measured immediately after excision using a hardness meter. Collagen and calcification were evaluated as the pathological factors. Collagen was stained with Elastica van Gieson stain, converted to a gray-scale image, and displayed in a 256-step histogram. The median gray-scale median (GSM) was used as the collagen content. The degree of calcification was defined by the hematoxylin–eosin stain as follows: "0:" no calcification, "1:" scattered microcalcification, or "2:" calcification greater than 1 mm or more than 2% of the total calcification. Carotid echocardiographic findings, specifically echoluminance or the brightness of the narrowest lesion of the plaque, classified as hypo-, iso-, or hyper-echoic by comparison with the intima-media complex surrounding the plaque, and clinical data were reviewed. Results Plaque hardness was significantly negatively correlated with GSM [Spearman's correlation coefficient: -0.7137 (p < 0.0001)]: the harder the plaque, the higher the collagen content. There were significant differences between plaque hardness and degree of calcification between "0" and "2" (p = 0.0206). For plaque hardness and echoluminance (hypo-iso-hyper), significant differences were found between hypo-iso (p = 0.0220), hypo-hyper (p = 0.0006), and iso-hyper (p = 0.0015): the harder the plaque, the higher the luminance. In single regression analysis, GSM, sex, and diabetes mellitus were significant variables, and in multiple regression analysis, only GSM was extracted as a significant variable. Conclusions Plaque hardness was associated with a higher amount of collagen, which is the main component of the fibrous cap. Greater plaque hardness was associated with increased plaque stability. The degree of calcification may also be associated with plaque hardness.
Collapse
Affiliation(s)
- Daisuke Fukushima
- Department of Neurosurgery (Omori), School of Medicine, Faculty of Medicine, Toho University, Tokyo, 143-8541, Japan.
| | - Kosuke Kondo
- Department of Neurosurgery (Omori), School of Medicine, Faculty of Medicine, Toho University, Tokyo, 143-8541, Japan
| | - Naoyuki Harada
- Department of Neurosurgery (Omori), School of Medicine, Faculty of Medicine, Toho University, Tokyo, 143-8541, Japan
| | - Sayaka Terazono
- Department of Neurosurgery (Omori), School of Medicine, Faculty of Medicine, Toho University, Tokyo, 143-8541, Japan
| | - Kei Uchino
- Department of Neurosurgery (Omori), School of Medicine, Faculty of Medicine, Toho University, Tokyo, 143-8541, Japan
| | - Kazutoshi Shibuya
- Department of Pathology, Toho University Omori Medical Center, Tokyo, Japan
| | - Nobuo Sugo
- Department of Neurosurgery (Omori), School of Medicine, Faculty of Medicine, Toho University, Tokyo, 143-8541, Japan
| |
Collapse
|
43
|
Li F, Wang Y, Hu T, Wu Y. Application and interpretation of vessel wall magnetic resonance imaging for intracranial atherosclerosis: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:714. [PMID: 35845481 PMCID: PMC9279807 DOI: 10.21037/atm-22-2364] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/01/2022] [Indexed: 11/06/2022]
Abstract
Background and Objective Atherosclerosis is a systemic disease that occurs in the arteries, and it is the most important causative factor of ischemic stroke. Vessel wall magnetic resonance imaging (VWMRI) is one of the best non-invasive methods for displaying the vascular features of intracranial atherosclerosis. The main clinical applications of this technique include the exploration of the pathogenesis of intracranial atherosclerotic lesions, follow-up monitoring, and treatment prognosis judgment. As the demand for intracranial VWMRI increases in clinical practice, radiologists should be aware of the selection of imaging parameters and how they affect image quality, clinical indications, evaluation methods, and limitations in interpreting these images. Therefore, this review focused on describing how to perform and interpret VWMRI of intracranial atherosclerotic lesions. Methods We searched the studies on the application of VWMRI in the PubMed database from January 1, 2000 to March 31, 2022, and focused on the analysis of related studies on VWMRI in atherosclerotic lesions, including technical application, expert consensus, imaging characteristics, and the clinical significance of intracranial atherosclerotic lesions. Key Content and Findings We reviewed and summarized recent advances in the clinical application of VWMRI in atherosclerotic diseases. Currently accepted principles and expert consensus recommendations for intracranial VWMRI include high spatial resolution, multiplanar two and three-dimensional imaging, multiple tissue-weighted sequences, and blood and cerebrospinal fluid suppression. Understanding the characteristics of VWMRI of normal intracranial arteries is the basis for interpreting VWMRI of atherosclerotic lesions. Evaluating VWMRI imaging features of intracranial atherosclerotic lesions includes plaque morphological and enhancement characteristics. The evaluation of atherosclerotic plaque stability is the highlight of VWMRI. Conclusions VWMRI has a wide range of clinical applications and can address important clinical questions and provide critical information for treatment decisions. VWMRI plays a key role in the comprehensive evaluation and prevention of intracranial atherosclerosis. However, intracranial VWMRI is still unable to obtain in vivo plaque pathological specimens for imaging—pathological comparison is the most significant limitation of this technique. Further technical improvements are expected to reduce acquisition time and may ultimately contribute to a better understanding of the underlying pathology of lesions on VWMRI.
Collapse
Affiliation(s)
- Fangbing Li
- Department of Radiology, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yilin Wang
- Department of Radiology, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Tianxiang Hu
- Department of Radiology, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yejun Wu
- Department of Radiology, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
44
|
Chen Q, Liang L, Zhang Z, Du J, Tang L, Cao W, Kong X, Xu D. Pathological analysis of vascularization of the arterialized veins in failed arteriovenous fistulas among uremic patients. J Vasc Interv Radiol 2022; 33:904-912.e1. [PMID: 35605817 DOI: 10.1016/j.jvir.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/29/2022] [Accepted: 05/08/2022] [Indexed: 10/18/2022] Open
Abstract
PURPOSE To assess venous wall vascularization and its correlation with neointimal hyperplasia (NIH) in failed arteriovenous fistulas (AVFs). METHODS A total of 43 uremic patients who received a first AVF creation and 39 patients who received reconstruction of failed fistulas were enrolled in the study. A 5-10 mm vein segment adjacent to future fistula creation or reconstruction site was surgically removed and assessed using histopathological analyses and stained by immunohistochemistry to quantify vasa vasorum density (VVD). RESULTS Both the intimal thickness (70.68 [28.81-99.54] vs. 4.53 [2.69-7.30] μm, P < 0.001) and the intimal thickness/medial thickness ratio (2.20 [0.77-4.36] vs. 0.15 [0.10-0.30], P < 0.001) were higher in failed AVFs than in pre-access veins. CD31 and factor VIII marked VVDs both in the intima (6.31 [1.62-12.53] vs. 0.0 [0.0-0.0], P < 0.001; 7.82 [3.33-11.61] vs. 0.0 [0.0-0.0], P < 0.001) and media (10.0 [7.59-12.95] vs. 3.71 [2.44-4.87], P < 0.001; 8.33 [5.55-13.0] vs. 3.57 [2.53-4.82], P < 0.001), and the intimal VVD/medial VVD ratio (0.67 [0.19-1.08] vs. 0.0 [0.0-0.0], P < 0.001; 0.71 [0.39-1.14] vs. 0.0 [0.0-0.0], P < 0.001) were significantly higher in failed AVFs than in pre-access veins. There was also a positive relationship between the intimal VVD/medial VVD ratio and the intimal thickness/medial thickness ratio (P < 0.001). In addition, compared to pre-access veins, vascular endothelial cell growth factor-A (VEGF-A) expression was higher in failed AVFs. CONCLUSIONS Vascularization of the vessel wall was noticeably more developed in the arterialized veins, especially among the NIH regions in failed AVFs.
Collapse
Affiliation(s)
- Qinlan Chen
- Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, No.44, Wenhua West Road, Jinan, China
| | - Liming Liang
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, PR China; Nephrology Research Institute of Shandong Province, No.16766, Jingshi Road, Jinan, 250014, PR China
| | - Ziheng Zhang
- Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, No.44, Wenhua West Road, Jinan, China
| | - Jing Du
- Department of Blood Purification Center, Weifang People's Hospital, No.151, Guangwen Street, Kuiwen District, Weifang, China
| | - Lijun Tang
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, PR China; Nephrology Research Institute of Shandong Province, No.16766, Jingshi Road, Jinan, 250014, PR China
| | - Wei Cao
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, PR China; Nephrology Research Institute of Shandong Province, No.16766, Jingshi Road, Jinan, 250014, PR China
| | - Xianglei Kong
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, PR China; Nephrology Research Institute of Shandong Province, No.16766, Jingshi Road, Jinan, 250014, PR China
| | - Dongmei Xu
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, No.16766, Jingshi Road, Jinan, 250014, PR China; Nephrology Research Institute of Shandong Province, No.16766, Jingshi Road, Jinan, 250014, PR China
| |
Collapse
|
45
|
Dose-dependent impact of statin therapy intensity on circulating progenitor cells in patients undergoing percutaneous coronary intervention for the treatment of acute versus chronic coronary syndrome. PLoS One 2022; 17:e0267433. [PMID: 35587929 PMCID: PMC9119492 DOI: 10.1371/journal.pone.0267433] [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: 04/26/2021] [Accepted: 04/09/2022] [Indexed: 11/30/2022] Open
Abstract
Background By low-density lipoprotein (LDL) reduction, statins play an important role in cardiovascular risk modification. Incompletely understood pleiotropic statin effects include vasoprotection that might originate from mobilisation and differentiation of vascular progenitor cells. Data on the potentially differential impact of statin treatment intensity on circulating progenitor cells in patients undergoing percutaneous coronary intervention (PCI) are scarce. This study examines the potential association of different permanent statin treatment regimens on circulating progenitor cells in patients with coronary syndrome. Methods and results In a monocentric prospective all-comers study, 105 consecutive cases scheduled for coronary angiography due to either (A) non-invasive proof of ischemia and chronic coronary syndrome (CCS) or (B) troponin-positive acute coronary syndrome (ACS) were included. According to the 2018 American College of Cardiology Guidelines on Blood Cholesterol, patients were clustered depending on their respective permanent statin treatment regimen in either a high- to moderate-intensity statin treatment (HIST) or a low-intensity statin treatment (LIST) group. Baseline characteristics including LDL levels were comparable. From blood drawn at the time of PCI, peripheral blood mononuclear cells were isolated, cultivated and counted and, by density gradient centrifugation, levels of circulating progenitor cells were determined using fluorescence-activated cell sorting (FACS) analysis. In ACS patients both absolute and relative numbers of circulating early-outgrowth endothelial progenitor cells (EPCs) concurrently were significantly lower in the HIST group as compared to the LIST group. This effect was more pronounced in ACS patients than in CCS patients. Both in ACS and CCS patients, HIST caused a significant reduction of the number of circulating SMPCs. Conclusions In patients undergoing PCI, a dose intensity-dependent and LDL level-independent pro-differentiating vasoprotective pleiotropic capacity of statins for EPC and SMPC is demonstrated.
Collapse
|
46
|
Lv X, Wang F, Sun M, Sun C, Fan X, Ma B, Yang Y, Ye Z, Liu P, Wen J. Differential Gene Expression and Immune Cell Infiltration in Carotid Intraplaque Hemorrhage Identified Using Integrated Bioinformatics Analysis. Front Cardiovasc Med 2022; 9:818585. [PMID: 35656397 PMCID: PMC9152291 DOI: 10.3389/fcvm.2022.818585] [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: 11/19/2021] [Accepted: 03/11/2022] [Indexed: 11/24/2022] Open
Abstract
Background Intraplaque hemorrhage (IPH) is an important feature of unstable plaques and an independent risk factor for cardiovascular events. However, the molecular mechanisms contributing to IPH are incompletely characterized. We aimed to identify novel biomarkers and interventional targets for IPH and to characterize the role of immune cells in IPH pathogenesis. Methods The microarray dataset GSE163154 which contain IPH and non-IPH plaque samples was obtained from the Gene Expression Omnibus (GEO). R software was adopted for identifying differentially expressed genes (DEGs) and conducting functional investigation. The hub genes were carried by protein-protein interaction (PPI) network and were validated by the GSE120521 dataset. CIBERSORT deconvolution was used to determine differential immune cell infiltration and the relationship of immune cells and hub genes. We confirmed expression of proteins encoded by the hub genes by immunohistochemistry and western blotting in 8 human carotid endarterectomy samples with IPH and 8 samples without IPH (non-IPH). Results We detected a total of 438 differentially expressed genes (DEGs), of which 248 were upregulated and 190 were downregulated. DEGs were mainly involved in inflammatory related pathways, including neutrophil activation, neutrophil degranulation, neutrophil-mediated immunity, leukocyte chemotaxis, and lysosomes. The hub genes found through the method of degree in the PPI network showed that ITGB2 and ITGAM might play an important role in IPH. Receiver operating characteristic (ROC) results also showed a good performance of these two genes in the test and validation dataset. We found that the proportions of infiltrating immune cells in IPH and non-IPH samples differed, especially in terms of M0 and M2 macrophages. Immunohistochemistry and western blotting analysis showed that expression levels of ITGB2 and ITGAM increased significantly in carotid atherosclerotic plaques with IPH. Conclusion ITGB2 and ITGAM are key hub genes of IPH and may play an important role in the biological process of IPH. Our findings advance our understanding of the underlying mechanisms of IPH pathogenesis and provide valuable information and directions for future research into novel targets for IPH diagnosis and immunotherapy.
Collapse
Affiliation(s)
- Xiaoshuo Lv
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Feng Wang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Mingsheng Sun
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Congrui Sun
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xueqiang Fan
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Bo Ma
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Yuguang Yang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Zhidong Ye
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Peng Liu
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
- Peng Liu
| | - Jianyan Wen
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
- *Correspondence: Jianyan Wen
| |
Collapse
|
47
|
Increased frequency of proangiogenic tunica intima endothelial kinase 2 (Tie2) expressing monocytes in individuals with type 2 diabetes mellitus. Cardiovasc Diabetol 2022; 21:72. [PMID: 35549955 PMCID: PMC9102255 DOI: 10.1186/s12933-022-01497-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/18/2022] [Indexed: 12/02/2022] Open
Abstract
Background Individuals with type 2 diabetes mellitus (T2DM) have an increased risk for developing macrovascular disease (MVD) manifested by atherosclerosis. Phenotypically and functionally different monocyte subsets (classical; CD14++CD16−, non-classical; CD14+CD16++, and intermediate; CD14++CD16+) including pro-angiogenic monocytes expressing Tie2 (TEMs) can be identified. Here we investigated monocyte heterogeneity and its association with T2DM and MVD. Methods Individuals with (N = 51) and without (N = 56) T2DM were recruited and allocated to "non-MVD" or "with MVD" (i.e., peripheral or coronary artery disease) subgroups. Blood monocyte subsets were quantified based on CD14, CD16 and Tie2 expression levels. Plasma levels of Tie2-ligands angiopoietin-1 and angiopoietin-2 were determined using ELISA. Carotid endarterectomy samples from individuals with (N = 24) and without (N = 22) T2DM were stained for intraplaque CD68+ macrophages (inflammation) and CD34+ (angiogenesis), as plaque vulnerability markers. Results Monocyte counts were similar between individuals with T2DM and healthy controls (non-diabetic, non-MVD). Non-classical monocytes were reduced (p < 0.05) in T2DM, whereas the percentage of TEMs within the intermediate subset was increased (p < 0.05). T2DM was associated with increased angiopoietin-1 (p < 0.05) and angiopoietin-2 (p = 0.0001) levels. Angiopoietin-2 levels were higher in T2DM individuals with MVD compared with non-MVD (p < 0.01). Endarterectomized plaques showed no differences in macrophage influx and microvessel number between individuals with and without T2DM. Conclusions Monocyte subset distribution is altered in T2DM with reduced non-classical monocytes and increased TEM percentage in the intermediate monocyte subset. Increased angiopoietin-2 levels together with increased frequency of TEMs might promote plaque vulnerability in T2DM which could however not be confirmed at tissue level in advanced atherosclerotic lesions.
Collapse
|
48
|
Dietz M, Kamani CH, Dunet V, Fournier S, Rubimbura V, Testart Dardel N, Schaefer A, Jreige M, Boughdad S, Nicod Lalonde M, Schaefer N, Mewton N, Prior JO, Treglia G. Overview of the RGD-Based PET Agents Use in Patients With Cardiovascular Diseases: A Systematic Review. Front Med (Lausanne) 2022; 9:887508. [PMID: 35602497 PMCID: PMC9120643 DOI: 10.3389/fmed.2022.887508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/19/2022] [Indexed: 01/02/2023] Open
Abstract
Studies using arginine–glycine–aspartate (RGD)-PET agents in cardiovascular diseases have been recently published. The aim of this systematic review was to perform an updated, evidence-based summary about the role of RGD-based PET agents in patients with cardiovascular diseases to better address future research in this setting. Original articles within the field of interest reporting the role of RGD-based PET agents in patients with cardiovascular diseases were eligible for inclusion in this systematic review. A systematic literature search of PubMed/MEDLINE and Cochrane library databases was performed until October 26, 2021. Literature shows an increasing role of RGD-based PET agents in patients with cardiovascular diseases. Overall, two main topics emerged: the infarcted myocardium and atherosclerosis. The existing studies support that αvβ3 integrin expression in the infarcted myocardium is well evident in RGD PET/CT scans. RGD-based PET radiotracers accumulate at the site of infarction as early as 3 days and seem to be peaking at 1–3 weeks post myocardial infarction before decreasing, but only 1 study assessed serial changes of myocardial RGD-based PET uptake after ischemic events. RGD-based PET uptake in large vessels showed correlation with CT plaque burden, and increased signal was found in patients with prior cardiovascular events. In human atherosclerotic carotid plaques, increased PET signal was observed in stenotic compared with non-stenotic areas based on MR or CT angiography data. Histopathological analysis found a co-localization between tracer accumulation and areas of αvβ3 expression. Promising applications using RGD-based PET agents are emerging, such as prediction of remodeling processes in the infarcted myocardium or detection of active atherosclerosis, with potentially significant clinical impact.
Collapse
Affiliation(s)
- Matthieu Dietz
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital, Lausanne, Switzerland
- INSERM U1060, CarMeN Laboratory, University of Lyon, Lyon, France
| | - Christel H. Kamani
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital, Lausanne, Switzerland
- Cardiology Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Vincent Dunet
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
| | - Stephane Fournier
- Cardiology Department, Lausanne University Hospital, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
| | - Vladimir Rubimbura
- Cardiology Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Nathalie Testart Dardel
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Ana Schaefer
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Mario Jreige
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Sarah Boughdad
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Marie Nicod Lalonde
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
| | - Niklaus Schaefer
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
| | - Nathan Mewton
- INSERM U1060, CarMeN Laboratory, University of Lyon, Lyon, France
- Cardiovascular Hospital Louis Pradel, Department of Heart Failure, Hospices Civils de Lyon, Lyon, France
- Clinical Investigation Center Inserm 1407, Lyon, France
| | - John O. Prior
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
- *Correspondence: John O. Prior
| | - Giorgio Treglia
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Università della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
49
|
Holmes DR, Alkhouli MA, Klaas JP, Brinjikji W, Savastano LE, Lanzino G, Benson JC. Change of Heart: The Underexplored Role of Plaque Hemorrhage in the Evaluation of Stroke of Undetermined Etiology. J Am Heart Assoc 2022; 11:e025323. [PMID: 35475334 PMCID: PMC9238607 DOI: 10.1161/jaha.122.025323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the evaluation of embolic strokes of undetermined source, great emphasis is often placed on cardiovascular disease, namely on atrial fibrillation. Other pathophysiologic mechanisms, however, may also be involved. Carotid artery intraplaque hemorrhage (IPH)—the presence of blood components within an atheromatous plaque—has become increasingly recognized as a possible etiologic mechanism in some cryptogenic strokes. IPH is a marker of plaque instability and is associated with ipsilateral neurologic ischemic events, even in nonstenotic carotid plaques. As recognition of carotid IPH as an etiology of embolic strokes has grown, so too has the complexity with which such patients are evaluated and treated, particularly because overlaps exist in the risk factors for atrial fibrillation and IPH. In this article, we review what is currently known about carotid IPH and how this clinical entity should be approached in the context of the evaluation of embolic strokes of undetermined source.
Collapse
Affiliation(s)
- David R Holmes
- Department of Cardiovascular Medicine Mayo Clinic Rochester MN
| | | | | | | | | | | | | |
Collapse
|
50
|
Chamaria S, Ueyama H, Yasumura K, Johnson KW, Vengrenyuk Y, Okamoto N, Barman N, Bhatheja S, Kapur V, Hasan C, Sweeney J, Baber U, Sharma SK, Narula J, Kini AS. Coronary plaque vulnerability in statin-treated patients with elevated LDL-C and hs-CRP: optical coherence tomography study. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2022; 38:1157-1167. [PMID: 38819727 DOI: 10.1007/s10554-021-02238-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/03/2021] [Indexed: 11/30/2022]
Abstract
There have been no previous attempts to assess coronary plaque morphology in statin-treated patients with combined residual cholesterol and inflammatory risk. The aim of this study was to characterize the morphology using optical coherence tomography (OCT) and to investigate the underlying molecular mechanisms. Two hundred seventy statin-treated patients with stable coronary artery disease who underwent OCT imaging prior to elective percutaneous coronary intervention were included in this single-center retrospective analysis. Subjects were stratified into four groups based on low-density lipoprotein cholesterol (LDL-C) and high-sensitivity C-reactive protein (hs-CRP) levels using 70 mg/dl and 2 mg/L as cut-offs, respectively. OCT images of the target lesions were assessed. For a subset of patients, peripheral blood mononuclear cells (PBMC) were isolated, and gene expression was characterized using microarray analysis. Patients with high LDL-C and high hs-CRP demonstrated a higher frequency of lipid-rich plaques (LRP) (91%, P = 0.03) by OCT. LRPs in these patients had a greater maximal lipid arc (186.6 ± 92.5°, P = 0.047). In addition, plaques from patients who did not achieve dual-target were less frequently calcified (P = 0.003). If calcification was present, it was characterized by a lower maximal arc (P = 0.016) and shorter length (P = 0.025). PBMC gene expression analysis demonstrated functional enrichment of toll-like receptors (TLRs) 1-9 to be associated with high LDL-C and hs-CRP. Obstructive coronary lesions in patients on statin therapy with combined residual cholesterol and inflammatory risk demonstrated a higher prevalence of LRP with greater maximal lipid arcs and more frequent spotty calcifications. PBMC from these patients revealed functional enrichment of TLR 1-9.
Collapse
Affiliation(s)
- Surbhi Chamaria
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Hiroki Ueyama
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
- Department of Medicine, Icahn School of Medicine At Mount Sinai, Mount Sinai Beth Israel, New York, NY, USA
| | - Keisuke Yasumura
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Kipp W Johnson
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Yuliya Vengrenyuk
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Naotaka Okamoto
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Nitin Barman
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Samit Bhatheja
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Vishal Kapur
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Choudhury Hasan
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Joseph Sweeney
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Usman Baber
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Samin K Sharma
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Jagat Narula
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Annapoorna S Kini
- Division of Cardiology, Mount Sinai Hospital and Icahn School of Medicine At Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA.
| |
Collapse
|