1
|
Zhong C, Deng K, Lang X, Shan D, Xie Y, Pan W, Yu J. Therapeutic potential of natural flavonoids in atherosclerosis through endothelium-protective mechanisms: An update. Pharmacol Ther 2025; 271:108864. [PMID: 40274196 DOI: 10.1016/j.pharmthera.2025.108864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/27/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
Atherosclerosis and its associated cardiovascular complications remain significant global public health challenges, underscoring the urgent need for effective therapeutic strategies. Endothelial cells are critical for maintaining vascular health and homeostasis, and their dysfunction is a key contributor to the initiation and progression of atherosclerosis. Targeting endothelial dysfunction has, therefore, emerged as a promising approach for the prevention and management of atherosclerosis. Among natural products, flavonoids, a diverse class of plant-derived phenolic compounds, have garnered significant attention for their anti-atherosclerotic properties. A growing body of evidence demonstrates that flavonoids can mitigate endothelial dysfunction, highlighting their potential as endothelial dysfunction-targeted therapeutics for atherosclerosis. In this review, we summarize current knowledge on the roles of natural flavonoids in modulating various aspects of endothelial dysfunction and their therapeutic effects on atherosclerosis, focusing on the underlying molecular mechanisms. We also discuss the challenges and future prospects of translating natural flavonoids into clinical applications for cardiovascular medicine. This review aims to provide critical insights to advance the development of novel endothelium-protective pharmacotherapies for atherosclerosis.
Collapse
Affiliation(s)
- Chao Zhong
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Keke Deng
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xiaoya Lang
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Dan Shan
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Yanfei Xie
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Wen Pan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi University of Chinese Medicine, Nanchang 330006, China.
| | - Jun Yu
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
2
|
Seneviratne AN, Miller MR. Air pollution and atherosclerosis. Atherosclerosis 2025; 406:119240. [PMID: 40411956 DOI: 10.1016/j.atherosclerosis.2025.119240] [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: 03/16/2025] [Revised: 05/01/2025] [Accepted: 05/02/2025] [Indexed: 05/27/2025]
Abstract
Air pollution is associated with considerable cardiovascular mortality and morbidity. The vascular disease atherosclerosis underlies many cardiovascular conditions, with atherosclerotic plaque rupture being a trigger for stroke and myocardial infarction. The acute and chronic effects of air pollution have the potential to exacerbate many different facets of atherosclerosis. This review provides an overview of how air pollution promotes the development of atherosclerosis. The review summaries the epidemiological evidence between exposure to air pollution and morphological measures of atherosclerosis such as carotid intimal media thickness, coronary artery calcification and aortic artery calcification, before summarising the biological mechanisms by which air pollution promotes atherosclerosis at the different stages of disease progression. We offer our perspective of the weight of evidence between air pollution to atherosclerosis and make recommendations for future research to advance this field. Given the ubiquity of air pollution exposure, we stress the need for urgency in efforts to tackle air pollution and emphasise the potential health gains from minimising the effects of air pollutants on this common and often fatal cardiovascular pathology.
Collapse
Affiliation(s)
| | - Mark R Miller
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
3
|
Lee SK, Xiong T, Qian AS, Yoo JA, Sokeechand BSH, Fuller MT, Gross PL, Austin RC, Igdoura SA, Trigatti BL. Scavenger receptor class B type I knockout mice develop extensive diet-induced coronary artery atherosclerosis in an age-dependent manner. PLoS One 2025; 20:e0318118. [PMID: 40403091 PMCID: PMC12097598 DOI: 10.1371/journal.pone.0318118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 01/10/2025] [Indexed: 05/24/2025] Open
Abstract
OBJECTIVE Homozygous knockout of scavenger receptor class B type I (SR-B1) in mice with atherogenic mutations (such as knockout of the apolipoprotein E or low density lipoprotein receptor genes) results in spontaneous or diet-induced coronary heart disease characterized by atherosclerosis development in the aortic sinus and coronary arteries, platelet accumulation in coronary artery plaques, myocardial fibrosis, and early death. However, the extent of coronary artery atherothrombosis and myocardial fibrosis in mice lacking SR-B1 alone (homozygous SR-B1 knockout mice) has not been examined. Although age is a major risk factor for coronary artery disease, few studies directly examine the effects of age on susceptibility to atherosclerosis or coronary artery atherothrombosis and myocardial fibrosis in mice. Therefore, we set out to examine the effects of age on diet-induced atherosclerosis in female homozygous SR-B1 knockout mice. APPROACH AND RESULTS SR-B1 knockout mice exhibited little-to-no aortic sinus or coronary artery atherosclerosis at 52 weeks of age, when fed a normal diet. However when fed a high-fat, high-cholesterol, cholate-containing (HFCC) diet for 12 weeks from either 14 weeks of age (26-week-old at analysis) or 40 weeks of age (52-week-old at analysis), they developed similar degrees of atherosclerosis in their aortic sinuses. Interestingly, the older aged SR-B1 knockout mice exhibited increased coronary artery atherosclerosis, increased vascular cell adhesion molecule 1 levels and platelet accumulation in coronary arteries, and increased myocardial fibrosis and plasma levels of cardiac troponin I compared to the younger aged mice. Older-aged HFCC diet-fed SR-B1 knockout mice also exhibited reduced survival to humane endpoint. Moreover, older-aged HFCC diet-fed SR-B1 knockout mice exhibited a greater inflammatory state with increased levels of circulating interleukin-6, tumour necrosis factor alpha, and neutrophils, despite plasma lipid levels being unchanged. Consistent with the increased circulating neutrophils, older-aged HFCC diet-fed SR-B1 knockout mice exhibited increased accumulation of the neutrophil marker myeloperoxidase and increased neutrophil extracellular traps in atherosclerotic plaques in the aortic sinus and increased abundance of atherosclerotic coronary arteries containing neutrophil extracellular traps. CONCLUSIONS HFCC diet-fed homozygous SR-B1 knockout mice develop occlusive coronary artery atherothrombosis and myocardial fibrosis in an age-dependent manner, and exhibit an increased inflammatory state with older age. Therefore, aged SR-B1 knockout mice may prove to be an attractive mouse model to analyze age-dependent mechanisms associated with coronary artery disease development, which may facilitate the discovery of more effective therapeutics to treat cardiovascular disease.
Collapse
Affiliation(s)
- Samuel K. Lee
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Ting Xiong
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Alexander S. Qian
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Jeong-Ah Yoo
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - B. Sumayyah H. Sokeechand
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Mark T. Fuller
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Peter L. Gross
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Richard C. Austin
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe’s Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Suleiman A. Igdoura
- Department of Biology and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Bernardo L. Trigatti
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
4
|
Seneviratne AN, Majumdar A, Surendranath K, Miller MR. Environmental modulators of vascular physiology and inflammation. Exp Physiol 2025. [PMID: 40349311 DOI: 10.1113/ep092309] [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: 12/20/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025]
Abstract
Environmental factors play a crucial role in modulating vascular inflammation, contributing significantly to the development of atherosclerosis and cardiovascular disease. This review synthesizes current evidence on how various environmental exposures influence vascular function and inflammation, with a focus on pollutants such as particulate matter and chemical toxins like bisphenols and per- and polyfluoroalkyl substances. These environmental stressors can trigger oxidative stress, chronic inflammation and vascular dysfunction, potentially accelerating the progression of atherosclerosis. We also explore the protective effects of natural compounds and exposure to green spaces in dampening inflammation and reducing cardiovascular risk. By examining the complex interplay between traditional risk factors and environmental exposures, this work highlights the need for comprehensive public health strategies that address both individual lifestyle factors and broader environmental determinants of cardiovascular health. We underscore the importance of further research to elucidate the precise cellular and molecular mechanisms by which environmental factors influence vascular function, with the aim of developing targeted interventions to mitigate their harmful effects and promote cardiovascular well-being.
Collapse
Affiliation(s)
- Anusha N Seneviratne
- Department of Health Studies, Royal Holloway University of London, Egham, Surrey, UK
| | - Anne Majumdar
- Department of Health Studies, Royal Holloway University of London, Egham, Surrey, UK
| | - Kalpana Surendranath
- Genome Engineering Laboratory, School of Life Sciences, University of Westminster, London, UK
| | - Mark R Miller
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
5
|
Gong X, Xu L, Cai P. Friend or foe of tripartite motif-containing protein 21 in cardiovascular disease: A review. Int J Biol Macromol 2025; 308:142682. [PMID: 40164260 DOI: 10.1016/j.ijbiomac.2025.142682] [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: 01/20/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
As an E3 ubiquitin ligase and an Fc receptor, tripartite motif-containing protein 21 (TRIM21) plays a crucial role in immune defense, signal transduction, and cellular regulation. TRIM21 is widely expressed in various tissues, but it is particularly abundant in cardiovascular tissues and is involved in the pathogenesis of various cardiovascular diseases (CVDs). However, although TRIM21 is involved in the regulation of several key molecular pathways in the immune system, its specific role in CVD remains unclear. In this review, we comprehensively summarize the regulatory role of TRIM21 in signaling pathways and discuss the function of TRIM21 in CVD, to provide a systematic understanding of this important protein in CVD and offer insights for further research into the pathogenesis of CVD and its potential applications.
Collapse
Affiliation(s)
- Xiangmei Gong
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Xu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengcheng Cai
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Kazemi A, Deylami N, Shojaei-Zarghani S, Hashempur MH. Effects of Pomegranate on Vascular Endothelial Function: A Systematic Review and Meta-Analysis of Clinical Trials. J Diet Suppl 2025:1-24. [PMID: 40296437 DOI: 10.1080/19390211.2025.2490659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
OBJECTIVE In this systematic review and meta-analysis of clinical trials, we investigated the effect of pomegranate on vascular endothelial function markers. METHODS We searched PubMed, Scopus, Web of Science, and the Cochrane Library until September 2024. Clinical trials that investigated the effect of pomegranate juice, extract, or its components on markers of vascular endothelial function, including flow-mediated dilation (FMD), intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), E-selectin, nitric oxide (NO), interleukin 6 (IL-6), IL-1, and IL-10, in healthy or diseased adults ages ≥ 18 years were eligible to be included. The quality of studies was assessed using the Cochrane tool. Random-effects meta-analysis was used to pool standardized mean differences (SMD) and 95% CI for the outcomes assessed by at least three studies. RESULTS Nineteen studies were included. Analysis of studies indicated a nonsignificant effect on VCAM-1 (SMD: -041, 95%CI: -2.53 to 1.71, I2= 83.6%, n = 3), while a significant reduction in ICAM-1 was observed (SMD: -0.63, 95% CI: -0.85 to -0.40, I2 = 0.0%, n = 3). Pooled analysis of fourteen studies on IL-6 indicated a significant reduction (-0.58, 95% CI: -0.98 to -0.19, I2 = 82.63%). The results of the three studies on E-selectin were inconsistent. The FMD and NO were assessed in only one study. CONCLUSIONS Pomegranate is effective in improving IL-6 and ICAM-1, but not VCAM-1.
Collapse
Affiliation(s)
- Asma Kazemi
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Noushin Deylami
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammad Hashem Hashempur
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Ma M, Liu G, Dai F, Wang X, Qin D, Yin M, Lu L, Wang Z, Wang T, Wang Z, Liu X, Chen Q, Jiao J. A sensing strategy based on novel pyrene-functionalized MOFs for sVCAM-1 detection and prognostic assessment in coronary heart disease. Talanta 2025; 294:128184. [PMID: 40262343 DOI: 10.1016/j.talanta.2025.128184] [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: 03/01/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025]
Abstract
Coronary heart disease (CHD) remains a significant global health concern, with exercise therapy playing a crucial role in rehabilitation. Optimizing exercise intensity is essential, as both insufficient and excessive exercise may lead to suboptimal or adverse outcomes. In response to this challenge, we developed Adaptive Posture-Balance Cardiac Rehabilitation Exercise (APBCRE), which integrates postural balance training with aerobic exercise. To evaluate its therapeutic efficacy, we designed a novel fluorescent biosensor targeting soluble vascular cell adhesion molecule-1 (sVCAM-1), a key biomarker of endothelial inflammation. The biosensor utilizes pyrene-functionalized metal-organic framework (PCA-UiO-66), synthesized via a one-pot approach. Within the MOF, pyrenecarboxylic acid (PCA) exists primarily in the excimer state, exhibiting stable fluorescence emission, a narrow spectral peak, and a wide Stokes shift. For the purpose of identify sVCAM-1, the sVCAM-1-specific aptamer was functionalized to PCA-UiO-66. When sVCAM-1 is present, the aptamer is competitively stripped from the MOF surface by the target. This process induces π-π stacking interactions between the aptamer's phosphate backbone and PCA molecules, facilitating PCA release and transition from the excimer to the monomer state. By quantifying the fluorescence value of monomer state PCA, the fluorescence response allowed precise quantification of sVCAM-1, with a detection limit of 0.69 ng/mL and a range of 2 ng/mL to 50 μg/mL. We analyzed cardiopulmonary performance and serum sVCAM-1 levels in 20 CHD patients who underwent APBCRE. These findings establish APBCRE as an effective rehabilitation strategy that provides both physiological and molecular benefits while also confirming Apt@PCA-UiO-66 as a robust biosensing tool for monitoring therapeutic efficacy and elucidating CHD pathophysiology.
Collapse
Affiliation(s)
- Mei Ma
- Department of Rehabilitation Medicine, Tianjin Chest Hospital, Tianjin, 300192, PR China.
| | - Guangxin Liu
- Department of Rehabilitation Medicine, Tianjin Chest Hospital, Tianjin, 300192, PR China
| | - Fuju Dai
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Xiangrui Wang
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Deyu Qin
- Department of Rehabilitation Medicine, Tianjin Chest Hospital, Tianjin, 300192, PR China
| | - Mengai Yin
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Lina Lu
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Zhijie Wang
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Tong Wang
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Zibo Wang
- Department of Medical Equipment, Tianjin Chest Hospital, Tianjin, 300192, PR China
| | - Xinmeng Liu
- Department of Rehabilitation Medicine, Tianjin Chest Hospital, Tianjin, 300192, PR China
| | - Qiang Chen
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China.
| | - Jun Jiao
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China.
| |
Collapse
|
8
|
Mageswaran N, Zainal SH, Hassan NI, Abd Karim NH, Ismail NAS. Emerging Biomarkers and Electrochemical Biosensors for Early Detection of Premature Coronary Artery Disease. Diagnostics (Basel) 2025; 15:940. [PMID: 40218291 PMCID: PMC11988804 DOI: 10.3390/diagnostics15070940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/04/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025] Open
Abstract
Coronary artery disease (CAD) is one of the primary causes of morbidity and death worldwide. Premature CAD (pCAD) is the term used to describe the 3-10% of CAD occurrences that occur in people under 45 worldwide. Diagnostic difficulties arise from the different risk factor profiles of pCAD and late-onset CAD. Better cardiovascular risk prediction in younger populations has been made possible by the development of biomarker detection tools. This can be applied to a diagnostic tool, including electrochemical biosensors, which have been predicted to be instrumental because of their adaptability for point-of-care applications for quicker diagnoses. These biosensors provide efficient, scalable, and reasonably priced solutions for the quick identification and tracking of CAD. Multiplex biomarker detection has been adopted as a viable approach for early diagnosis and risk assessment due to the constraints of using a single biomarker for pCAD diagnosis. Thus, this study looks at current developments in biosensing technology and discusses established and new cardiac biomarker panels for pCAD identification.
Collapse
Affiliation(s)
- Nanthini Mageswaran
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Sarah Husnaini Zainal
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Nurul Izzaty Hassan
- Department of Chemical Sciences, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia; (N.I.H.); (N.H.A.K.)
| | - Nurul Huda Abd Karim
- Department of Chemical Sciences, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia; (N.I.H.); (N.H.A.K.)
| | - Noor Akmal Shareela Ismail
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
9
|
Quagliariello V, Berretta M, Bisceglia I, Giacobbe I, Iovine M, Barbato M, Maurea C, Canale ML, Paccone A, Inno A, Scherillo M, Oliva S, Cadeddu Dessalvi C, Mauriello A, Fonderico C, Maratea AC, Gabrielli D, Maurea N. In the Era of Cardiovascular-Kidney-Metabolic Syndrome in Cardio-Oncology: From Pathogenesis to Prevention and Therapy. Cancers (Basel) 2025; 17:1169. [PMID: 40227756 PMCID: PMC11988012 DOI: 10.3390/cancers17071169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
Cardiovascular-kidney-metabolic (CKM) syndrome represents a complex interplay between cardiovascular disease (CVD), chronic kidney disease (CKD), and metabolic disorders, significantly impacting cancer patients. The presence of CKM syndrome in cancer patients not only worsens their prognosis but also increases the risk of major adverse cardiovascular events (MACE), reduces quality of life (QoL), and affects overall survival (OS). Furthermore, several anticancer therapies, including anthracyclines, tyrosine kinase inhibitors, immune checkpoint inhibitors, and hormonal treatments, can exacerbate CKM syndrome by inducing cardiotoxicity, nephrotoxicity, and metabolic dysregulation. This review explores the pathophysiology of CKM syndrome in cancer patients and highlights emerging therapeutic strategies to mitigate its impact. We discuss the role of novel pharmacological interventions, including sodium-glucose cotransporter-2 inhibitors (SGLT2i), proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), and soluble guanylate cyclase (sGC) activators, as well as dietary and lifestyle interventions. Optimizing the management of CKM syndrome in cancer patients is crucial to improving OS, enhancing QoL, and reducing MACE. By integrating cardiometabolic therapies into oncologic care, we can create a more comprehensive treatment approach that reduces the burden of cardiovascular and renal complications in this vulnerable population. Further research is needed to establish personalized strategies for CKM syndrome prevention and treatment in cancer patients.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Irma Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00148 Rome, Italy
| | - Ilaria Giacobbe
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Matteo Barbato
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Carlo Maurea
- ASL NA1, UOC Neurology and Stroke Unit, Ospedale del Mare, 23807 Naples, Italy
| | | | - Andrea Paccone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Alessandro Inno
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy
| | - Marino Scherillo
- Cardiologia Interventistica e UTIC, A.O. San Pio, Presidio Ospedaliero Gaetano Rummo, 82100 Benevento, Italy
| | - Stefano Oliva
- Cardio-Oncology Unit, IRCCS Istituto Tumori, “Giovanni Paolo II”, 70124 Bari, Italy
| | | | - Alfredo Mauriello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Celeste Fonderico
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Anna Chiara Maratea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Domenico Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00152 Rome, Italy
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| |
Collapse
|
10
|
Čiapienė I, Vėžys J, Lesauskaitė V, Matulevičiūtė I, Meškauskaitė U, Skipskis V, Strazdauskas A, Trumbeckaitė S, Bubulis A, Jūrėnas V, Ostaševičius V, Tamakauskas V, Tatarūnas V. Synergistic Effects of Low-Frequency Ultrasound and Therapeutic Agents on Endothelial and Renal Cells: Emphasis on Cell Functionality, Oxidative Stress, and Inflammatory Markers. Pharmaceuticals (Basel) 2025; 18:404. [PMID: 40143180 PMCID: PMC11945135 DOI: 10.3390/ph18030404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Ischemic heart disease remains the leading cause of death worldwide, with coronary microvascular dysfunction (CMD) as a key complication after ST-elevation myocardial infarction (STEMI). Endothelial dysfunction contributes to CMD, impairing vascular tone and increasing inflammation. While angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs) aid vascular health, their efficacy may improve with therapeutic ultrasound, which enhances drug delivery and endothelial response. This study explores the combined effects of ultrasound and pharmacological treatment on the ACE axis and inflammation in endothelial and renal cells. Methods: Human umbilical vein endothelial cells (HUVECs) and human renal proximal tubular epithelial cell line RPTEC/TERT1 were treated with captopril, losartan, and dexamethasone, alone or combined with low-frequency ultrasound (LFU). Cell viability and wound-healing assays assessed cellular function, while nitric oxide (NO) and reactive oxygen species (ROS) assays were used to evaluate redox signaling. Gene expression related to the ACE axis, inflammation, and vascular and renal cell function was analyzed via qPCR. Results: Captopril and losartan combined with LFU improved endothelial cell viability, wound healing, and NO production at various concentrations, whereas only losartan with LFU enhanced cell viability and wound healing in renal cells. Dexamethasone with LFU increased ROS levels and had variable effects on RPTEC/TERT1 cell survival. Gene expression analysis showed that LFU alone reduced pro-inflammatory markers VCAM-1, ICAM-1, and PTGS2 in captopril-treated HUVECs and similarly affected CYP4F2 in losartan-treated HUVECs. LFU also decreased PTGS2 expression at higher dexamethasone concentrations. In RPTEC/TERT1 cells, LFU alone did not impact SGLT2 or GGT1 expression, but captopril with LFU downregulated GGT1, and dexamethasone with LFU upregulated SGLT2 at higher concentrations. Conclusions: This study demonstrates that LFU enhances the effects of RAS inhibitors by promoting NO synthesis and reducing oxidative stress, while its combination with dexamethasone may have variable, potentially cytotoxic effects on renal cells. Gene expression patterns suggest LFU's anti-inflammatory potential and its role in modulating drug efficacy.
Collapse
Affiliation(s)
- Ieva Čiapienė
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT-50103 Kaunas, Lithuania; (I.Č.); (V.L.); (U.M.); (V.S.); (V.T.)
| | - Joris Vėžys
- Department of Mechanical Engineering, Faculty of Mechanical Engineering and Design, Kaunas University of Technology, Studentu 56, LT-51424 Kaunas, Lithuania;
| | - Vaiva Lesauskaitė
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT-50103 Kaunas, Lithuania; (I.Č.); (V.L.); (U.M.); (V.S.); (V.T.)
| | - Indrė Matulevičiūtė
- Department of Ophthalmology, Lithuanian University of Health Sciences, Eiveniu 2, LT-50161 Kaunas, Lithuania;
| | - Ugnė Meškauskaitė
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT-50103 Kaunas, Lithuania; (I.Č.); (V.L.); (U.M.); (V.S.); (V.T.)
| | - Vilius Skipskis
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT-50103 Kaunas, Lithuania; (I.Č.); (V.L.); (U.M.); (V.S.); (V.T.)
| | - Arvydas Strazdauskas
- Department of Biochemistry, Faculty of Medicine, Lithuanian University of Health Sciences, Eiveniu 4, LT-50161 Kaunas, Lithuania;
| | - Sonata Trumbeckaitė
- Department of Pharmacognosy, Faculty of Pharmacy, Lithuanian University of Health Sciences, Sukileliu 13, LT-50162 Kaunas, Lithuania;
| | - Algimantas Bubulis
- Institute of Mechatronics, Kaunas University of Technology, Studentu 56, LT-51424 Kaunas, Lithuania; (A.B.); (V.J.); (V.O.)
| | - Vytautas Jūrėnas
- Institute of Mechatronics, Kaunas University of Technology, Studentu 56, LT-51424 Kaunas, Lithuania; (A.B.); (V.J.); (V.O.)
| | - Vytautas Ostaševičius
- Institute of Mechatronics, Kaunas University of Technology, Studentu 56, LT-51424 Kaunas, Lithuania; (A.B.); (V.J.); (V.O.)
| | - Vytenis Tamakauskas
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT-50103 Kaunas, Lithuania; (I.Č.); (V.L.); (U.M.); (V.S.); (V.T.)
| | - Vacis Tatarūnas
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT-50103 Kaunas, Lithuania; (I.Č.); (V.L.); (U.M.); (V.S.); (V.T.)
| |
Collapse
|
11
|
Amancherla K, Taravella Oill AM, Bledsoe X, Williams AL, Chow N, Zhao S, Sheng Q, Bearl DW, Hoffman RD, Menachem JN, Siddiqi HK, Brinkley DM, Mee ED, Hadad N, Agrawal V, Schmeckpepper J, Rali AS, Tsai S, Farber-Eger EH, Wells QS, Freedman JE, Tucker NR, Schlendorf KH, Gamazon ER, Shah RV, Banovich N. Dynamic responses to rejection in the transplanted human heart revealed through spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640852. [PMID: 40093136 PMCID: PMC11908199 DOI: 10.1101/2025.02.28.640852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Allograft rejection following solid-organ transplantation is a major cause of graft dysfunction and mortality. Current approaches to diagnosis rely on histology, which exhibits wide diagnostic variability and lacks access to molecular phenotypes that may stratify therapeutic response. Here, we leverage image-based spatial transcriptomics at sub-cellular resolution in longitudinal human cardiac biopsies to characterize transcriptional heterogeneity in 62 adult and pediatric heart transplant (HT) recipients during and following histologically-diagnosed rejection. Across 28 cell types, we identified significant differences in abundance in CD4 + and CD8 + T cells, fibroblasts, and endothelial cells across different biological classes of rejection (cellular, mixed, antibody-mediated). We observed a broad overlap in cellular transcriptional states across histologic rejection severity and biological class and significant heterogeneity within rejection severity grades that would qualify for immunomodulatory treatment. Individuals who had resolved rejection after therapy had a distinct transcriptomic profile relative to those with persistent rejection, including 216 genes across 6 cell types along pathways of inflammation, IL6-JAK-STAT3 signaling, IFNα/IFNγ response, and TNFα signaling. Spatial transcriptomics also identified genes linked to long-term prognostic outcomes post-HT. These results underscore importance of subtyping immunologic states during rejection to stratify immune-cardiac interactions following HT that are therapeutically relevant to short- and long-term rejection-related outcomes.
Collapse
|
12
|
Hullon D, Subeh GK, Volkova Y, Janiec K, Trach A, Mnevets R. The role of glucagon-like peptide-1 receptor (GLP-1R) agonists in enhancing endothelial function: a potential avenue for improving heart failure with preserved ejection fraction (HFpEF). Cardiovasc Diabetol 2025; 24:70. [PMID: 39920668 PMCID: PMC11806760 DOI: 10.1186/s12933-025-02607-w] [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: 09/11/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a prevalent and complex condition with limited effective treatments. Endothelial dysfunction is a significant component of HFpEF pathophysiology, and glucagon-like peptide-1 receptor (GLP-1R) agonists have shown potential benefits in improving endothelial function. This study aims to explore the relationship between endothelial dysfunction in HFpEF and the mechanisms of action of GLP-1R agonists, highlighting their potential therapeutic benefits. A comprehensive review of the literature was conducted to examine the etiology of HFpEF, the role of endothelial dysfunction, and the effects of GLP-1R agonists on endothelial function and heart failure outcomes. The findings indicate that HFpEF is associated with various comorbidities, such as obesity, diabetes mellitus, and hypertension, which contribute to endothelial dysfunction. GLP-1R agonists, including semaglutide and liraglutide, have demonstrated significant cardioprotective effects, such as improving vascular endothelial function, reducing inflammation, and preventing atherosclerosis. Clinical trials, such as the STEP-HFpEF trial, have shown positive results in reducing symptoms and physical restrictions in HFpEF patients. GLP-1R agonists present a promising therapeutic option for HFpEF by targeting endothelial dysfunction and other pathophysiological mechanisms. Further research is needed to elucidate the precise mechanisms through which GLP-1R agonists exert their benefits and to establish their long-term safety and efficacy in diverse HFpEF populations.
Collapse
Affiliation(s)
| | | | | | | | - Adam Trach
- Taras Shevchenko National University of Kyiv, Kiev, Ukraine
| | - Ruslan Mnevets
- Taras Shevchenko National University of Kyiv, Kiev, Ukraine
- Institute of Pediatrics, Obstetrics, and Gynecology named after acad. O.M. Lukyanova of the National Academy of Medical Sciences of Ukraine, Kiev, Ukraine
| |
Collapse
|
13
|
Khuu MP, Paeslack N, Dremova O, Benakis C, Kiouptsi K, Reinhardt C. The gut microbiota in thrombosis. Nat Rev Cardiol 2025; 22:121-137. [PMID: 39289543 DOI: 10.1038/s41569-024-01070-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/01/2024] [Indexed: 09/19/2024]
Abstract
The gut microbiota has emerged as an environmental risk factor that affects thrombotic phenotypes in several cardiovascular diseases. Evidence includes the identification of marker species by sequencing studies of the gut microbiomes of patients with thrombotic disease, the influence of antithrombotic therapies on gut microbial diversity, and preclinical studies in mouse models of thrombosis that have demonstrated the functional effects of the gut microbiota on vascular inflammatory phenotypes and thrombus formation. In addition to impaired gut barrier function promoting low-grade inflammation, gut microbiota-derived metabolites have been shown to act on vascular cell types and promote thrombus formation. Therefore, these meta-organismal pathways that link the metabolic capacities of gut microorganisms with host immune functions have emerged as potential diagnostic markers and novel drug targets. In this Review, we discuss the link between the gut microbiota, its metabolites and thromboembolic diseases.
Collapse
Affiliation(s)
- My Phung Khuu
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Nadja Paeslack
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Olga Dremova
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Corinne Benakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Klytaimnistra Kiouptsi
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| |
Collapse
|
14
|
Shen L, Chen M, Su Y, Bi Y, Shu G, Chen W, Lu C, Zhao Z, Lv L, Zou J, Chen X, Ji J. NIR-II Imaging for Tracking the Spatiotemporal Immune Microenvironment in Atherosclerotic Plaques. ACS NANO 2024; 18:34171-34185. [PMID: 39630481 DOI: 10.1021/acsnano.4c10739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
The inflammatory immune microenvironment is responsible for atherosclerotic plaque erosion and rupture. Near-infrared-II (NIR-II) fluorescence imaging has the potential to continuously monitor the spatiotemporal changes in the plaque immune microenvironment. Herein, we constructed three different NIR-II probes based on benzo[1,2-c;4,5-c']bis[1,2,5]thiadiazole-4,7-bis(9,9-dioctyl-9H-fluoren-2-yl)thiophene (denoted as BBT-2FT): VHPK/BBT-2FT NPs, where VHPK is a specific peptide targeting vascular cell adhesion molecule-1; iNOS/BBT-2FT NPs for modulating the polarization of M1 macrophages by inducible NO synthase (iNOS) antibodies; and Arg-1/BBT-2FT for counterbalancing the inflammatory responses of M1 macrophages. These tracers enable precise tracking of atherosclerotic plaques and M1 and M2 macrophages through NIR-II imaging. VHPK/BBT-2FT NPs can accurately trace atherosclerotic plaques at various stages. Arg-1/BBT-2FT NPs precisely located M2 macrophages in the early plaque microenvironment with upregulation of peroxisome proliferator-activated receptor γ (PPAR-γ), signal transducer and activator of transcription (STAT) 6, and ATP-binding cassette transporter A1 (ABCA1), indicating that M2 macrophage polarization is crucial for early plaque lipid clearance. Meanwhile, iNOS/BBT-2FT NPs accurately tracked M1 macrophages in the advanced plaque microenvironment. The results showed that M1 macrophage polarization induces the formation of an inflammatory microenvironment through anaerobic glycolytic metabolism and pyroptosis in the advanced hypoxic plaque microenvironment, as indicated by the upregulation of hypoxia-inducible factor 1 alpha (HIF-1α), STAT1, NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3), pyruvate dehydrogenase kinase 1 (PDK1), and glucose transporter 1 (GLUT-1). Combining immunological approaches with NIR-II imaging has revealed that hypoxia-induced metabolic reprogramming of macrophages is a key factor in dynamic changes in the immune microenvironment of atherosclerotic plaques. Furthermore, our strategy shows the potential for real-time diagnosis and clinical prevention of unstable plaque rupture in atherosclerosis.
Collapse
Affiliation(s)
- Lin Shen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui 323000, China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui 323000, China
| | - Yanping Su
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui 323000, China
| | - Yanran Bi
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui 323000, China
| | - Gaofeng Shu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui 323000, China
| | - Weiqian Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui 323000, China
| | - Chenying Lu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui 323000, China
| | - Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui 323000, China
| | - Lingchun Lv
- Department of Cardiovascular Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Jianhua Zou
- Departments of Diagnostic Radiology Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios 138667, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios 138667, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Lower Kent Ridge Road, 4 Science Drive 2, Singapore 117544, Singapore
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, No 289, Kuocang Road, Lishui 323000, China
| |
Collapse
|
15
|
Sanip Z, Rasool AHG, Pahimi N, Bokti NA, Yusof Z, Mohamed MS, Isa WYHW. Elevated Inflammation and Adhesion Molecule hsCRP, GDF-15 and VCAM-1 in Angina Patients with Non-obstructive Coronary Artery Disease. Malays J Med Sci 2024; 31:148-159. [PMID: 39830114 PMCID: PMC11740812 DOI: 10.21315/mjms2024.31.6.12] [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: 07/27/2024] [Accepted: 11/07/2024] [Indexed: 01/22/2025] Open
Abstract
Background Non-obstructive coronary artery disease (NOCAD) is a condition in stable patients that experience angina despite not having significant coronary obstructive lesion. Knowledge on the role of certain biomarkers in patients with NOCAD is still limited. This study aimed to evaluate the roles of inflammation and adhesion molecules in the development of NOCAD. The correlations between the peripheral and coronary levels of the inflammatory biomarkers and adhesion molecules were also investigated. Methods In this cross-sectional study, symptomatic angina patients scheduled for coronary angiograms were recruited and separated into obstructive coronary artery disease (OCAD) and NOCAD groups based on those angiograms. Peripheral and coronary blood samples were taken to measure inflammation biomarkers [high sensitivity C-reactive protein (hsCRP) and growth differentiation factor 15 (GDF-15)], and adhesion molecules [vascular cell adhesion molecule-1 (VCAM-1)]. Subjects without angina symptoms were recruited for the control group. Results The hsCRP, GDF-15, and VCAM-1 levels were higher in the OCAD and NOCAD groups than in the control group. VCAM-1 levels successfully predicted the incidence of NOCAD [p = 0.010, area under the curve (AUC) = 0.716]. All biomarkers' levels in the peripheral and coronary blood were correlated in OCAD and NOCAD patients (p < 0.001). Conclusion Elevated levels of the hsCRP, GDF-15, and VCAM-1 were found with NOCAD, despite the absent of significant coronary obstruction. VCAM-1 successfully predicted NOCAD and may thus serve as an early NOCAD biomarker. Significant correlations of hsCRP, GDF-15, and VCAM-1 level in peripheral and coronary blood indicate that the peripheral levels of these biomarkers reflect the levels and changes that occur at the coronary level.
Collapse
Affiliation(s)
- Zulkefli Sanip
- Central Research Laboratory, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Nurnajwa Pahimi
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
- Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Nur Adilah Bokti
- Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
- Cardiology Unit, Hospital Universiti Sains Malaysia, Kelantan, Malaysia
| | - Zurkurnai Yusof
- Cardiology Unit, Hospital Universiti Sains Malaysia, Kelantan, Malaysia
| | | | - W Yus Haniff W Isa
- Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
- Cardiology Unit, Hospital Universiti Sains Malaysia, Kelantan, Malaysia
| |
Collapse
|
16
|
Barbhuyan T, Patel RB, Budnik I, Chauhan AK. Genetic ablation of myeloid integrin α9 attenuates early atherosclerosis. J Leukoc Biol 2024; 116:1208-1214. [PMID: 39036986 PMCID: PMC11531806 DOI: 10.1093/jleuko/qiae161] [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: 03/07/2024] [Revised: 06/04/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024] Open
Abstract
Integrin α9β1 is known to stabilize leukocyte adhesion to the activated endothelium. We determined the role of myeloid cell α9β1 in early atherosclerosis in two models: α9Mye-KOApoe-/- or the Ldlr-/- mice transplanted with bone marrow (BM) from α9Mye-KO mice fed a high-fat "Western" diet for 4 wk. α9Mye-KOApoe-/- mice exhibited reduced early lesions in the aortae and aortic sinuses (P < 0.05 vs α9WT Apoe-/- mice). Similar results were obtained in α9Mye-KO BM→Ldlr-/- mice (P < 0.05 vs α9WT BM→Ldlr-/- mice). Reduced early atherosclerosis in α9Mye-KOApoe-/- mice was associated with decreased neutrophil and neutrophil extracellular traps (NETs) content in the aortic lesions (P < 0.05 vs α9WTApoe-/-). Vascular cell adhesion molecule-1-stimulated neutrophils from α9Mye-KO mice exhibited reduced adhesion, transmigration, and NETs formation (NETosis) (P < 0.05 vs α9WT neutrophils). Reduced NETosis was associated with decreased extracellular signal-regulated kinase phosphorylation, peptidyl arginine deiminase 4, and citrullinated histone H3 expression. In summary, genetic ablation of myeloid cell-specific α9 reduces early atherosclerosis, most likely by reducing neutrophil adhesion, transmigration, and NETosis.
Collapse
Affiliation(s)
- Tarun Barbhuyan
- Division of Hematology/Oncology, Department of Internal Medicine, University of Iowa, 3160 Medical Labs, Iowa City, IA 52242, United States
| | - Rakesh B Patel
- Division of Hematology/Oncology, Department of Internal Medicine, University of Iowa, 3160 Medical Labs, Iowa City, IA 52242, United States
| | - Ivan Budnik
- Division of Hematology/Oncology, Department of Internal Medicine, University of Iowa, 3160 Medical Labs, Iowa City, IA 52242, United States
| | - Anil K Chauhan
- Division of Hematology/Oncology, Department of Internal Medicine, University of Iowa, 3160 Medical Labs, Iowa City, IA 52242, United States
| |
Collapse
|
17
|
Luo H, Zhao S, Zi J, Hu Y, Yao Y, Xiong J. Benzo[b]fluoranthene damages coronary artery and affects atherosclerosis markers in mice and umbilical vein endothelial cells. Toxicol Lett 2024; 401:150-157. [PMID: 39395681 DOI: 10.1016/j.toxlet.2024.10.007] [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: 04/22/2024] [Revised: 09/03/2024] [Accepted: 10/08/2024] [Indexed: 10/14/2024]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) exposure is associated with cardiovascular diseases. Toxic effects of PAHs are diverse, while cardiovascular consequences of benzo[b]fluoranthene (B[b]F) are unclear. Here, we reported the impacts of B[b]F on coronary artery and atherosclerosis markers both in mice and umbilical vein endothelial EAhy.926 cells. In mice, we found that B[b]F decreases heart-to-body weight ratio, affects aortic physiology, elevates serum low-density lipoprotein and total cholesterol, increases aortic levels of collagen fiber and atherosclerotic marker vascular cell adhesion molecule-1 (VCAM-1), and downregulates oxidative stress related nuclear factor erythroid 2-related factor 2 (Nrf2). In EAhy.926 cells, we showed that B[b]F inhibits cell proliferation and migration in a dose-dependent manner, induces cell cycle arrest and apoptosis, increases reactive oxygen species, upregulates VCAM-1 level, and suppresses expression of Nrf2. Taken together, our findings reveal that B[b]F exposure may contribute to coronary artery damage and potentially induce atherosclerosis, possibly via the Nrf2-related signaling pathways.
Collapse
Affiliation(s)
- Hang Luo
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Shanshan Zhao
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; Fucheng Center for Disease Control and Prevention, Mianyang 621000, China
| | - Jing Zi
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yifan Hu
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yuqin Yao
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China.
| | - Jingyuan Xiong
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu 610041, China.
| |
Collapse
|
18
|
Kennelly JP, Xiao X, Gao Y, Kim S, Hong SG, Villanueva M, Ferrari A, Vanharanta L, Nguyen A, Nagari RT, Burton NR, Tol MJ, Becker AP, Lee MJ, Ikonen E, Backus KM, Mack JJ, Tontonoz P. Cholesterol binding to VCAM-1 promotes vascular inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613543. [PMID: 39345495 PMCID: PMC11429921 DOI: 10.1101/2024.09.17.613543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Hypercholesterolemia has long been implicated in endothelial cell (EC) dysfunction, but the mechanisms by which excess cholesterol causes vascular pathology are incompletely understood. Here we used a cholesterol-mimetic probe to map cholesterol-protein interactions in primary human ECs and discovered that cholesterol binds to and stabilizes the adhesion molecule VCAM-1. We show that accessible plasma membrane (PM) cholesterol in ECs is acutely responsive to inflammatory stimuli and that the nonvesicular cholesterol transporter Aster-A regulates VCAM-1 stability in activated ECs by controlling the size of this pool. Deletion of Aster-A in ECs increases VCAM-1 protein, promotes immune cell recruitment to vessels, and impairs pulmonary immune homeostasis. Conversely, depleting cholesterol from the endothelium in vivo dampens VCAM-1 induction in response to inflammatory stimuli. These findings identify cholesterol binding to VCAM-1 as a key step during EC activation and provide a biochemical explanation for the ability of excess membrane cholesterol to promote immune cell recruitment to the endothelium.
Collapse
Affiliation(s)
- John P. Kennelly
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA); Los Angeles, CA 90095, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
- These authors contributed equally
| | - Xu Xiao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA); Los Angeles, CA 90095, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
- These authors contributed equally
| | - Yajing Gao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA); Los Angeles, CA 90095, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
| | - Sumin Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Soon-Gook Hong
- Molecular Biology Institute, UCLA, Los Angeles, CA 90095, USA
- Department of Medicine, Division of Cardiology, UCLA, Los Angeles, CA, USA
| | | | - Alessandra Ferrari
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA); Los Angeles, CA 90095, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
| | - Lauri Vanharanta
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
| | - Alexander Nguyen
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA); Los Angeles, CA 90095, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Rohith T. Nagari
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA); Los Angeles, CA 90095, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
| | - Nikolas R. Burton
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095, United States
| | - Marcus J. Tol
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA); Los Angeles, CA 90095, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
| | - Andrew P. Becker
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
| | - Min Jae Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Elina Ikonen
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
| | - Keriann M. Backus
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095, United States
- DOE Institute for Genomics and Proteomics, UCLA, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California 90095, United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California 90095, United States
| | - Julia J. Mack
- Molecular Biology Institute, UCLA, Los Angeles, CA 90095, USA
- Department of Medicine, Division of Cardiology, UCLA, Los Angeles, CA, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA); Los Angeles, CA 90095, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
19
|
Piao J, Su Z, He J, Zhu T, Fan F, Wang X, Yang Z, Zhan H, Luo D. SphK1 deficiency ameliorates the development of atherosclerosis by inhibiting the S1P/S1PR3/Rhoa/ROCK pathway. Cell Signal 2024; 121:111252. [PMID: 38852936 DOI: 10.1016/j.cellsig.2024.111252] [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: 03/15/2024] [Revised: 05/14/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND AND AIMS S1P is an important factor regulating the function of the vascular endothelial barrier. SphK1 is an important limiting enzyme for the synthesis of S1P. However, the role of the SphK1/S1P-mediated vascular endothelial barrier function in atherosclerosis has not been fully revealed. This study explored the roles and mechanisms of SphK1 on atherosclerosis in vivo and in vitro. METHODS In vivo, ApoE-/- and SphK1-/-ApoE-/- mice were fed a high-fat diet to induce atherosclerosis. In vitro, ox-LDL induced HUVECs to establish a cell model. Aortic histological changes were measured by H&E staining, Oil Red O staining, EVG staining, Sirius scarlet staining, immunofluorescence, and Evans Blue Assay. Western blotting was performed to explore the specific mechanism. RESULTS We validated that deficiency of SphK1 resulted in a marked amelioration of atherosclerosis, as indicated by the decreased lipid accumulation, inflammatory factors, oxidative stress, aortic plaque area, inflammatory factor infiltration, VCAM-1 expression, and vascular endothelial permeability. Moreover, deficiency of SphK1 downregulated the expression of aortic S1PR3, Rhoa, ROCK, and F-actin. The results of administration with the SphK1 inhibitor PF-543 and the S1PR3 inhibitor VPC23019 in vitro further confirmed the conclusion that deficiency of SphK1 reduced S1P level and S1PR3 protein expression, inhibited Rhoa/ROCK signaling pathway, regulated protein expression of F-actin, improved vascular endothelial dysfunction and permeability, and exerted anti-atherosclerotic effects. CONCLUSIONS This study revealed that deficiency of SphK1 relieved vascular endothelial barrier function in atherosclerosis mice via SphK1/S1P/S1PR signaling pathway.
Collapse
Affiliation(s)
- Jinyu Piao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Zhuoxuan Su
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Jiqian He
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Tianxin Zhu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Faxin Fan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Xin Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Zhenzhen Yang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Huixia Zhan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China
| | - Duosheng Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China.
| |
Collapse
|
20
|
Barreca MM, Raimondo S, Conigliaro A, Siragusa S, Napolitano M, Alessandro R, Corrado C. The Combination of Natural Compounds Escin-Bromelain-Ginkgo Biloba-Sage Miltiorrhiza (EBGS) Reduces Platelet Adhesion to TNFα-Activated Vascular Endothelium through FAK Signaling. Int J Mol Sci 2024; 25:9252. [PMID: 39273200 PMCID: PMC11395133 DOI: 10.3390/ijms25179252] [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: 07/25/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
Thrombosis is a key process that determines acute coronary syndrome and ischemic stroke and is the leading cause of morbidity and mortality in the world, together with cancer. Platelet adhesion and subsequent activation and aggregation are critical processes that cause thrombus formation after endothelial damage. To date, high hopes are associated with compounds of natural origin, which show anticoagulant action without undesirable effects and can be proposed as supportive therapies. We investigated the effect of the new combination of four natural compounds, escin-bromelain-ginkgo biloba-sage miltiorrhiza (EBGS), on the initial process of the coagulation cascade, which is the adhesion of platelets to activated vascular endothelium. Our results demonstrated that EBGS pretreatment of endothelial cells reduces platelet adhesion even in the presence of the monocyte-lymphocyte population. Our data indicate that EBGS exerts its effects by inhibiting the transcription of adhesion molecules, including P-selectin, platelet membrane glycoprotein GP1b, integrins αV and β3, and reducing the secretion of the pro-inflammatory cytokines interleukin 6, interleukin 8, and the metalloproteinases MMP-2 and MMP-9. Furthermore, we demonstrated that EBGS inhibited the expression of focal adhesion kinase (FAK), strictly involved in platelet adhesion, and whose activity is correlated with that of integrin β3. The results shown in this manuscript suggest a possible inhibitory role of the new combination EBGS in the reduction in platelet adhesion to activated endothelium, thus possibly preventing coagulation cascade initiation.
Collapse
Affiliation(s)
- Maria Magdalena Barreca
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Biology and Genetics Section, University of Palermo, 90133 Palermo, Italy; (M.M.B.); (S.R.); (A.C.); (R.A.)
| | - Stefania Raimondo
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Biology and Genetics Section, University of Palermo, 90133 Palermo, Italy; (M.M.B.); (S.R.); (A.C.); (R.A.)
| | - Alice Conigliaro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Biology and Genetics Section, University of Palermo, 90133 Palermo, Italy; (M.M.B.); (S.R.); (A.C.); (R.A.)
| | - Sergio Siragusa
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Haematology Section, University of Palermo, 90127 Palermo, Italy; (S.S.); (M.N.)
| | - Mariasanta Napolitano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Haematology Section, University of Palermo, 90127 Palermo, Italy; (S.S.); (M.N.)
| | - Riccardo Alessandro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Biology and Genetics Section, University of Palermo, 90133 Palermo, Italy; (M.M.B.); (S.R.); (A.C.); (R.A.)
| | - Chiara Corrado
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Biology and Genetics Section, University of Palermo, 90133 Palermo, Italy; (M.M.B.); (S.R.); (A.C.); (R.A.)
| |
Collapse
|
21
|
Natarajan N, Florentin J, Johny E, Xiao H, O'Neil SP, Lei L, Shen J, Ohayon L, Johnson AR, Rao K, Li X, Zhao Y, Zhang Y, Tavakoli S, Shiva S, Das J, Dutta P. Aberrant mitochondrial DNA synthesis in macrophages exacerbates inflammation and atherosclerosis. Nat Commun 2024; 15:7337. [PMID: 39187565 PMCID: PMC11347661 DOI: 10.1038/s41467-024-51780-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 08/16/2024] [Indexed: 08/28/2024] Open
Abstract
There is a large body of evidence that cellular metabolism governs inflammation, and that inflammation contributes to the progression of atherosclerosis. However, whether mitochondrial DNA synthesis affects macrophage function and atherosclerosis pathology is not fully understood. Here we show, by transcriptomic analyzes of plaque macrophages, spatial single cell transcriptomics of atherosclerotic plaques, and functional experiments, that mitochondrial DNA (mtDNA) synthesis in atherosclerotic plaque macrophages are triggered by vascular cell adhesion molecule 1 (VCAM-1) under inflammatory conditions in both humans and mice. Mechanistically, VCAM-1 activates C/EBPα, which binds to the promoters of key mitochondrial biogenesis genes - Cmpk2 and Pgc1a. Increased CMPK2 and PGC-1α expression triggers mtDNA synthesis, which activates STING-mediated inflammation. Consistently, atherosclerosis and inflammation are less severe in Apoe-/- mice lacking Vcam1 in macrophages. Downregulation of macrophage-specific VCAM-1 in vivo leads to decreased expression of LYZ1 and FCOR, involved in STING signalling. Finally, VCAM-1 expression in human carotid plaque macrophages correlates with necrotic core area, mitochondrial volume, and oxidative damage to DNA. Collectively, our study highlights the importance of macrophage VCAM-1 in inflammation and atherogenesis pathology and proposes a self-acerbating pathway involving increased mtDNA synthesis.
Collapse
Affiliation(s)
- Niranjana Natarajan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Jonathan Florentin
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Ebin Johny
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Hanxi Xiao
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Joint CMU-Pitt PhD program in Computational Biology, Pittsburgh, PA, USA
| | - Scott Patrick O'Neil
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Liqun Lei
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Jixing Shen
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Lee Ohayon
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Aaron R Johnson
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Krithika Rao
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Xiaoyun Li
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Yanwu Zhao
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Yingze Zhang
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Sina Tavakoli
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
- University of Pittsburgh School of Medicine Department of Pharmacology & Chemical Biology, Pittsburgh, PA, USA
| | - Jishnu Das
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
22
|
Shapira M, Roguin A, Fayad I, Medlij L, khateeb A, Egbaria D, Amsalem N, Abu Fanne R. Predictive value of baseline alpha defensin level in patients with stable coronary artery disease: A retrospective single center study. IJC HEART & VASCULATURE 2024; 53:101465. [PMID: 39091435 PMCID: PMC11292519 DOI: 10.1016/j.ijcha.2024.101465] [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: 03/04/2024] [Revised: 06/24/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024]
Abstract
Background Inflammation plays a central role in atherogenesis. The major neutrophilic peptide alpha-defensin is a promising evolving risk factor for atherosclerosis. The aim of the present study was to examine the role of alpha-defensin in predicting future major adverse cardiovascular events (MACE) occurrence in fully revascularized patients with stable CAD under optimal medical therapy. Methods and results We retrospectively examined the prognostic value of baseline plasma alpha-defensin levels in predicting MACE occurrence in 174 fully revascularized patients for stable CAD between March 2016 and January 2017. Alpha-defensin levels were found 20 % higher among demised patients (10,859 pg/ml, IQR [6,920 to 23,320] vs. 9,020 pg/ml, IQR [5,540 to 16,180] pg/ml, P = 0.15). The absolute increase in mortality risk in patients with alpha-defensin levels greater than the median values was 72.5 % (P = 0.33). Log-rank analysis proved both recurrent PCI for de novo lesions (14.9 % and 2.3 %) and the composite of mortality and recurrent PCI for de novo lesions (27.6 % vs. 9.2 %) were significantly related to alpha-defensin values greater than the median (>9200 pg/ml). Conclusion Baseline plasma alpha-defensin is an independent predictor of mortality and recurrent PCI among patients with stable CAD. Alpha-defensin may evolve as a promising factor in cardiovascular risk assessment beyond traditional risk factors. Targeting alpha-defensin to ameliorate MACE occurrence should be addressed in future studies.
Collapse
Affiliation(s)
- Maanit Shapira
- Division of laboratories, Hillel Yaffe Medical Center, Hadera, Israel
- Faculty of Medicine - Technion Israel Institute of Technology, Israel
| | - Ariel Roguin
- Faculty of Medicine - Technion Israel Institute of Technology, Israel
- Department of Cardiology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Ibraheem Fayad
- Faculty of Medicine - Technion Israel Institute of Technology, Israel
- Department of Cardiology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Lina Medlij
- Faculty of Medicine - Technion Israel Institute of Technology, Israel
- Department of Cardiology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Aysha khateeb
- Faculty of Medicine - Technion Israel Institute of Technology, Israel
- Department of Cardiology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Dema Egbaria
- Faculty of Medicine - Technion Israel Institute of Technology, Israel
- Department of Cardiology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Naama Amsalem
- Faculty of Medicine - Technion Israel Institute of Technology, Israel
- Department of Cardiology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Rami Abu Fanne
- Faculty of Medicine - Technion Israel Institute of Technology, Israel
- Department of Cardiology, Hillel Yaffe Medical Center, Hadera, Israel
| |
Collapse
|
23
|
Shi H, Song J, Gao L, Shan X, Panicker SR, Yao L, McDaniel M, Zhou M, McGee S, Zhong H, Griffin CT, Xia L, Shao B. Deletion of Talin1 in Myeloid Cells Facilitates Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 2024; 44:1799-1812. [PMID: 38899470 DOI: 10.1161/atvbaha.123.319677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 04/23/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Integrin-regulated monocyte recruitment and cellular responses of monocyte-derived macrophages are critical for the pathogenesis of atherosclerosis. In the canonical model, talin1 controls ligand binding to integrins, a prerequisite for integrins to mediate leukocyte recruitment and induce immune responses. However, the role of talin1 in the development of atherosclerosis has not been studied. Our study investigated how talin1 in myeloid cells regulates the progression of atherosclerosis. METHODS On an Apoe-/- background, myeloid talin1-deficient mice and the control mice were fed with a high-fat diet for 8 or 12 weeks to induce atherosclerosis. The atherosclerosis development in the aorta and monocyte recruitment into atherosclerotic lesions were analyzed. RESULTS Myeloid talin1 deletion facilitated the formation of atherosclerotic lesions and macrophage deposition in lesions. Talin1 deletion abolished integrin β2-mediated adhesion of monocytes but did not impair integrin α4β1-dependent cell adhesion in a flow adhesion assay. Strikingly, talin1 deletion did not prevent Mn2+- or chemokine-induced activation of integrin α4β1 to the high-affinity state for ligands. In an in vivo competitive homing assay, monocyte infiltration into inflamed tissues was prohibited by antibodies to integrin α4β1 but was not affected by talin1 deletion or antibodies to integrin β2. Furthermore, quantitative polymerase chain reaction and ELISA (enzyme-linked immunosorbent assay) analysis showed that macrophages produced cytokines to promote inflammation and the proliferation of smooth muscle cells. Ligand binding to integrin β3 inhibited cytokine generation in macrophages, although talin1 deletion abolished the negative effects of integrin β3. CONCLUSIONS Integrin α4β1 controls monocyte recruitment during atherosclerosis. Talin1 is dispensable for integrin α4β1 activation to the high-affinity state and integrin α4β1-mediated monocyte recruitment. Yet, talin1 is required for integrin β3 to inhibit the production of inflammatory cytokines in macrophages. Thus, intact monocyte recruitment and elevated inflammatory responses cause enhanced atherosclerosis in talin1-deficient mice. Our study provides novel insights into the roles of myeloid talin1 and integrins in the progression of atherosclerosis.
Collapse
Affiliation(s)
- Huiping Shi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center (H.S., L.X.)
| | - Jianhua Song
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Liang Gao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Xindi Shan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Sumith R Panicker
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Longbiao Yao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Michael McDaniel
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Meixiang Zhou
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Samuel McGee
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Hui Zhong
- Lindsley F. Kimball Research Institute, New York Blood Center (H.Z., B.S.)
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center (H.S., L.X.)
| | - Bojing Shao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (H.S., J.S., L.G., X.S., S.R.P., L.Y., M.M., M.Z., S.M., C.T.G., L.X., B.S.)
- Lindsley F. Kimball Research Institute, New York Blood Center (H.Z., B.S.)
| |
Collapse
|
24
|
Short SA, Wilkinson K, Long DL, Crews DC, Gutierrez OM, Irvin MR, Wheeler M, Cushman M, Cheung KL. Endothelial Dysfunction Biomarkers and CKD Incidence in the REGARDS Cohort. Kidney Int Rep 2024; 9:2016-2027. [PMID: 39081743 PMCID: PMC11284378 DOI: 10.1016/j.ekir.2024.04.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction Chronic kidney disease (CKD) is only partly caused by traditional risk factors. Endothelial dysfunction is common in CKD and may contribute to CKD incidence. We studied the association of circulating biomarkers reflecting endothelial dysfunction with incident CKD. Methods The Reasons for Geographical and Racial Differences in Stroke (REGARDS) study is a prospective cohort of 30,239 Black or White adults aged ≥45 years. Baseline levels of intercellular cellular adhesion molecule 1 (ICAM-1), vascular cellular adhesion molecule 1 (VCAM-1), factor VIII (FVIII), and E-selectin were measured in 3300 participants without baseline CKD or albuminuria who attended a second visit 9.4 years later. Kidney outcomes were incident CKD (estimated glomerular filtration rate [eGFR] <60 ml/min per 1.73 m2 and ≥40% decline or onset of new end-stage kidney disease), incident ≥30% eGFR decline, and incident albuminuria (albumin-to-creatinine ratio [ACR] ≥30 mg/g). Sequentially adjusted logistic regression models assessed the association of biomarkers with kidney outcomes. Results Median age of participants was 62 years, 49% were women, and 46% identified as Black. Of the participants, 228 (6.9%) developed CKD, 613 (18.9%) experienced ≥30% decline in eGFR, and 356 (11.4%) developed albuminuria. The adjusted odds ratios (ORs) for incident CKD per 1 SD increment biomarker was 1.12 for ICAM-1 (95% confidence interval [CI]: 1.02-1.22), 1.10 for VCAM-1 (95% CI: 1.01-1.20), 1.15 for FVIII (95% CI: 1.06-1.24), and 1.10 for E-selectin (95% CI: 1.01-1.20). Results were similar for incident ≥30% eGFR decline but not albuminuria, where only higher FVIII was positively associated. Conclusion Higher concentration of ICAM-1, VCAM-1, FVIII, and E-selectin were associated with incident CKD and ≥30% eGFR decline in a large cohort study. Higher FVIII was also associated with incident albuminuria.
Collapse
Affiliation(s)
- Samuel A.P. Short
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Katherine Wilkinson
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - D. Leann Long
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Deidra C. Crews
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Orlando M. Gutierrez
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Marguerite R. Irvin
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Marsha Wheeler
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Mary Cushman
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT
| | - Katharine L. Cheung
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT
| |
Collapse
|
25
|
Tan JN, Husain K, Jubri Z, Chan KM, Ugusman A, Jantan I, Fauzi NM. Anti-atherogenic mechanism of ethanol extract of Christia vespertilionis (L.f.) Bakh. F. Leaves in vitro. Int Immunopharmacol 2024; 134:112148. [PMID: 38718657 DOI: 10.1016/j.intimp.2024.112148] [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: 01/11/2024] [Revised: 04/08/2024] [Accepted: 04/20/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Vascular inflammation is the key event in early atherogenesis. Pro-inflammatory endothelial cells induce monocyte recruitment into the sub-endothelial layer of the artery. This requires endothelial expression of adhesion molecules namely intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1), alongside chemokines production. Christia vespertilionis (L.f.) Bakh.f. (CV) possesses anti-inflammatory property. However, its potential anti-atherogenic effect in the context of vascular inflammation has yet to be explored. PURPOSE To evaluate the anti-atherogenic mechanism of 80% ethanol extract of CV leaves on tumor necrosis factor-α (TNF-α)-activated human umbilical vein endothelial cells (HUVECs). METHODS Qualitative analysis of the CV extract was carried out by using liquid chromatography with tandem mass spectrometry (LC-MS/MS). The cell viability of HUVECs treated with CV extract was determined by MTT assay. The effect of CV extract on monocyte adhesion was determined by monocyte-endothelial adhesion assay. Protein expressions of ICAM-1, VCAM-1 and nuclear factor-kappa B (NF-κB) signaling pathway were determined by western blot while production of monocyte chemoattractant protein-1 (MCP-1) was determined by ELISA. RESULTS LC-MS/MS analysis showed that CV extract composed of five main compounds, including schaftoside, orientin, isovitexin, 6-caffeoyl-D-glucose, and 3,3'-di-O-methyl ellagic acid. Treatment of CV extract at a concentration range from 5 to 60 µg/mL for 24 h maintained HUVECs viability above 90 %, therefore concentrations of 20, 40 and 60 μg/mL were selected for the subsequent experiments. All concentrations of CV extract showed a significant inhibitory effect on monocyte adhesion to TNF-α-activated HUVECs (p < 0.05). In addition, the protein expressions of ICAM-1 and VCAM-1 were significantly attenuated by CV in a concentration dependent manner (p < 0.001). At all tested concentrations, CV extract also exhibited significant inhibition on the production of MCP-1 (p < 0.05). Moreover, CV extract significantly inhibited TNF-α-induced phosphorylation of inhibitor of nuclear factor-κB kinase alpha/beta (IKKα/β), inhibitor kappa B-alpha (IκBα), NF-κB and nuclear translocation of NF-κB (p < 0.05). CONCLUSION CV extract inhibited monocyte adhesion to endothelial cells by suppressing protein expressions of cell adhesion molecules and production of chemokines through downregulation of NF-κB signaling pathway. Thus, CV has the potential to be developed as an anti-atherogenic agent for early treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jiah Ning Tan
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Khairana Husain
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Zakiah Jubri
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Kok Meng Chan
- Centre for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia; Product Stewardship and Toxicology, Group Health, Safety and Environment (GHSE), Petroliam Nasional Berhad (PETRONAS), Kuala Lumpur, Malaysia
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ibrahim Jantan
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Malaysia; Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Norsyahida Mohd Fauzi
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| |
Collapse
|
26
|
Lee J, Park J, Song KM, Lee YG, Choi HK. Actinidia arguta Extract Containing Myo-Inositol Suppresses TNF- α-Induced VCAM-1 Expression and Monocyte Adhesion to Endothelial Cells via Inhibition of the PTEN/Akt/GSK-3 β and NF- κB Signaling Pathways. J Med Food 2024; 27:419-427. [PMID: 38656897 DOI: 10.1089/jmf.2023.k.0326] [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] [Indexed: 04/26/2024] Open
Abstract
The primary inflammatory process in atherosclerosis, a major contributor to cardiovascular disease, begins with monocyte adhering to vascular endothelial cells. Actinidia arguta (kiwiberry) is an edible fruit that contains various bioactive components. While A. arguta extract (AAE) has been recognized for its anti-inflammatory characteristics, its specific inhibitory effect on early atherogenic events has not been clarified. We used tumor necrosis factor-α (TNF-α)-stimulated human umbilical vein endothelial cells (HUVECs) for an in vitro model. AAE effectively hindered the attachment of THP-1 monocytes and reduced the expression of vascular cell adhesion molecule-1 (VCAM-1) in HUVECs. Transcriptome analysis revealed that AAE treatment upregulated phosphatase and tensin homolog (PTEN), subsequently inhibiting phosphorylation of AKT and glycogen synthase kinase 3β (GSK3β) in HUVECs. AAE further hindered phosphorylation of AKT downstream of the nuclear factor kappa B (NF-κB) signaling pathway, leading to suppression of target gene expression. Oral administration of AAE suppressed TNF-α-stimulated VCAM-1 expression, monocyte-derived macrophage infiltration, and proinflammatory cytokine expression in C57BL/6 mouse aortas. Myo-inositol, identified as the major compound in AAE, played a key role in suppressing THP-1 monocyte adhesion in HUVECs. These findings suggest that AAE could serve as a nutraceutical for preventing atherosclerosis by inhibiting its initial pathogenesis.
Collapse
Affiliation(s)
- Jangho Lee
- Divisions of Functional Food Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Joon Park
- Divisions of Strategic Food Technology Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Kyung-Mo Song
- Divisions of Strategic Food Technology Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Yu Geon Lee
- Divisions of Functional Food Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Hyo-Kyoung Choi
- Divisions of Functional Food Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| |
Collapse
|
27
|
Guo W, Yang C, Zou J, Yu T, Li M, He R, Chen K, Hell RCR, Gross ER, Zou X, Lu Y. Interleukin-1β polarization in M1 macrophage mediates myocardial fibrosis in diabetes. Int Immunopharmacol 2024; 131:111858. [PMID: 38492336 PMCID: PMC11330059 DOI: 10.1016/j.intimp.2024.111858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Diabetes is a global health problem whose common complication is diabetic cardiomyopathy, characterized by chronic inflammation of the heart muscle. Macrophages are the main white blood cells found in the resting heart. Therefore, we investigated the underling mechanism of macrophage on myocardial fibrosis in diabetes. METHODS Here, echocardiography was utilized to evaluate cardiac function, and the degree of myocardial fibrosis was assessed using Masson's trichrome staining, followed by single-cell RNA sequencing (scRNA-seq) to analyze the phenotype, function, developmental trajectory, and interactions between immune cells, endothelial cells (ECs), and fibroblasts (FBs) in the hearts of db/db mice at different stages of diabetes. Macrophages and cardiac fibroblasts were also co-cultured in order to study the signaling between macrophages and fibroblasts. RESULTS We found that with the development of diabetes mellitus, myocardial hypertrophy and fibrosis occurred that was accompanied by cardiac dysfunction. A significant proportion of immune cells, endothelial cells, and fibroblasts were identified by RNA sequencing. The most significant changes observed were in macrophages, which undergo M1 polarization and are critical for oxidative stress and extracellular matrix (ECM) formation. We further found that M1 macrophages secreted interleukin-1β (IL-1β), which interacted with the receptor on the surface of fibroblasts, to cause myocardial fibrosis. In addition, crosstalk between M1 macrophages and endothelial cells also plays a key role in fibrosis and immune response regulation through IL-1β and corresponding receptors. CONCLUSIONS M1 macrophages mediate diabetic myocardial fibrosis through interleukin-1β interaction with fibroblasts.
Collapse
Affiliation(s)
- Wenli Guo
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Chen Yang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Jiawei Zou
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tingting Yu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Mingde Li
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Ruilin He
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Keyang Chen
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Rafaela C R Hell
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, 94305 CA, United States
| | - Eric R Gross
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, 94305 CA, United States
| | - Xin Zou
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai 201508, China.
| | - Yao Lu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Ambulatory Surgery Center, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
28
|
Manzo OL, Nour J, Sasset L, Marino A, Rubinelli L, Palikhe S, Smimmo M, Hu Y, Bucci MR, Borczuk A, Elemento O, Freed JK, Norata GD, Di Lorenzo A. Rewiring Endothelial Sphingolipid Metabolism to Favor S1P Over Ceramide Protects From Coronary Atherosclerosis. Circ Res 2024; 134:990-1005. [PMID: 38456287 PMCID: PMC11009055 DOI: 10.1161/circresaha.123.323826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Growing evidence correlated changes in bioactive sphingolipids, particularly S1P (sphingosine-1-phosphate) and ceramides, with coronary artery diseases. Furthermore, specific plasma ceramide species can predict major cardiovascular events. Dysfunction of the endothelium lining lesion-prone areas plays a pivotal role in atherosclerosis. Yet, how sphingolipid metabolism and signaling change and contribute to endothelial dysfunction and atherosclerosis remain poorly understood. METHODS We used an established model of coronary atherosclerosis in mice, combined with sphingolipidomics, RNA-sequencing, flow cytometry, and immunostaining to investigate the contribution of sphingolipid metabolism and signaling to endothelial cell (EC) activation and dysfunction. RESULTS We demonstrated that hemodynamic stress induced an early metabolic rewiring towards endothelial sphingolipid de novo biosynthesis, favoring S1P signaling over ceramides as a protective response. This finding is a paradigm shift from the current belief that ceramide accrual contributes to endothelial dysfunction. The enzyme SPT (serine palmitoyltransferase) commences de novo biosynthesis of sphingolipids and is inhibited by NOGO-B (reticulon-4B), an ER membrane protein. Here, we showed that NOGO-B is upregulated by hemodynamic stress in myocardial EC of ApoE-/- mice and is expressed in the endothelium lining coronary lesions in mice and humans. We demonstrated that mice lacking NOGO-B specifically in EC (Nogo-A/BECKOApoE-/-) were resistant to coronary atherosclerosis development and progression, and mortality. Fibrous cap thickness was significantly increased in Nogo-A/BECKOApoE-/- mice and correlated with reduced necrotic core and macrophage infiltration. Mechanistically, the deletion of NOGO-B in EC sustained the rewiring of sphingolipid metabolism towards S1P, imparting an atheroprotective endothelial transcriptional signature. CONCLUSIONS These data demonstrated that hemodynamic stress induced a protective rewiring of sphingolipid metabolism, favoring S1P over ceramide. NOGO-B deletion sustained the rewiring of sphingolipid metabolism toward S1P protecting EC from activation under hemodynamic stress and refraining coronary atherosclerosis. These findings also set forth the foundation for sphingolipid-based therapeutics to limit atheroprogression.
Collapse
Affiliation(s)
- Onorina Laura Manzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Jasmine Nour
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Department of Excellence of Pharmacological and Biomolecular Sciences, University of Milan, Via G. Balzaretti, 9 – 20133, Milano, Italy
| | - Linda Sasset
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Alice Marino
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Luisa Rubinelli
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Sailesh Palikhe
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Martina Smimmo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Yang Hu
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA
| | - Maria Rosaria Bucci
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano 49, Naples 80131, Italy
| | - Alain Borczuk
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Olivier Elemento
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA
| | - Julie K. Freed
- Department of Anesthesiology, Medical College of Wisconsin Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Rd. Milwaukee, WI 53226, USA
| | - Giuseppe Danilo Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences, University of Milan, Via G. Balzaretti, 9 – 20133, Milano, Italy
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| |
Collapse
|
29
|
Abstract
BACKGROUND Approximately one in four stroke patients suffer from recurrent vascular events, underlying the necessity to improve secondary stroke prevention strategies. Immune mechanisms are causally associated with coronary atherosclerosis. However, stroke is a heterogeneous disease and the relative contribution of inflammation across stroke mechanisms is not well understood. The optimal design of future randomized control trials (RCTs) of anti-inflammatory therapies to prevent recurrence after stroke must be informed by a clear understanding of the prognostic role of inflammation according to stroke subtype and individual patient factors. AIM In this narrative review, we discuss (1) inflammatory pathways in the etiology of ischemic stroke subtypes; (2) the evidence on inflammatory markers and vascular recurrence after stroke; and (3) review RCT evidence of anti-inflammatory agents for vascular prevention. SUMMARY OF REVIEW Experimental work, genetic epidemiological data, and plaque-imaging studies all implicate inflammation in atherosclerotic stroke. However, emerging evidence also suggests that inflammatory mechanisms are also important in other stroke mechanisms. Advanced neuroimaging techniques support the role of neuroinflammation in blood-brain barrier dysfunction in cerebral small vessel disease (cSVD). Systemic inflammatory processes also promote atrial cardiopathy, incident and recurrent atrial fibrillation (AF). Although several inflammatory markers have been associated with recurrence after stroke, interleukin-6 (IL-6) and high-sensitivity C-reactive protein (hsCRP) are presently the most promising markers to identify patients at increased vascular risk. Several RCTs have shown that anti-inflammatory therapies reduce vascular risk, including stroke, in coronary artery disease (CAD). Some, but not all of these trials, selected patients on the basis of elevated hsCRP. Although unproven after stroke, targeting inflammation to reduce recurrence is a compelling strategy and several RCTs are ongoing. CONCLUSION Evidence points toward the importance of inflammation across multiple stroke etiologies and potential benefit of anti-inflammatory targets in secondary stroke prevention. Taking the heterogeneous stroke etiologies into account, the use of serum biomarkers could be useful to identify patients with residual inflammatory risk and perform biomarker-led patient selection for future RCTs.
Collapse
Affiliation(s)
- Annaelle Zietz
- Department of Neurology and Stroke Center, University Hospital Basel and University of Basel, Basel, Switzerland
- Neurology and Neurorehabilitation, University Department of Geriatric Medicine Felix Platter, University of Basel, Basel, Switzerland
| | - Sarah Gorey
- Health Research Board (HRB) Stroke Clinical Trials Network Ireland (SCTNI), Dublin, Ireland
- School of Medicine, University College Dublin (UCD), Dublin, Ireland
- Department of Geriatric Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Peter J Kelly
- Health Research Board (HRB) Stroke Clinical Trials Network Ireland (SCTNI), Dublin, Ireland
- School of Medicine, University College Dublin (UCD), Dublin, Ireland
- Department of Neurology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Mira Katan
- Department of Neurology and Stroke Center, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - John J McCabe
- Health Research Board (HRB) Stroke Clinical Trials Network Ireland (SCTNI), Dublin, Ireland
- School of Medicine, University College Dublin (UCD), Dublin, Ireland
- Department of Geriatric Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| |
Collapse
|
30
|
Castro R, Adair JH, Mastro AM, Neuberger T, Matters GL. VCAM-1-targeted nanoparticles to diagnose, monitor and treat atherosclerosis. Nanomedicine (Lond) 2024; 19:723-735. [PMID: 38420919 DOI: 10.2217/nnm-2023-0282] [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] [Indexed: 03/02/2024] Open
Abstract
Vascular cell adhesion molecule-1 (VCAM-1) was identified over 2 decades ago as an endothelial adhesion receptor involved in leukocyte recruitment and cell-based immune responses. In atherosclerosis, a chronic inflammatory disease of the blood vessels that is the leading cause of death in the USA, endothelial VCAM-1 is robustly expressed beginning in the early stages of the disease. The interactions of circulating immune cells with VCAM-1 on the activated endothelial cell surface promote the uptake of monocytes and the progression of atherosclerotic lesions in susceptible vessels. Herein, we review the role of VCAM-1 in atherosclerosis and the use of VCAM-1 binding peptides, antibodies and aptamers as targeting agents for nanoplatforms for early detection and treatment of atherosclerotic disease.
Collapse
Affiliation(s)
- Rita Castro
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Pharmaceutical Sciences & Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisboa, Portugal
| | - James H Adair
- Department of Materials Science, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Pharmacology, The Pennsylvania State University, University Park, PA 16802, USA
| | | | - Thomas Neuberger
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of The Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Gail L Matters
- Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
31
|
Fishbein I, Inamdar VV, Alferiev IS, Bratinov G, Zviman MM, Yekhilevsky A, Nagaswami C, Gardiner KL, Levy RJ, Stachelek SJ. Hypercholesterolemia exacerbates in-stent restenosis in rabbits: Studies of the mitigating effect of stent surface modification with a CD47-derived peptide. Atherosclerosis 2024; 390:117432. [PMID: 38241977 PMCID: PMC10939830 DOI: 10.1016/j.atherosclerosis.2023.117432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/07/2023] [Accepted: 12/20/2023] [Indexed: 01/21/2024]
Abstract
BACKGROUND AND AIMS Hypercholesterolemia (HC) has previously been shown to augment the restenotic response in animal models and humans. However, the mechanistic aspects of in-stent restenosis (ISR) on a hypercholesterolemic background, including potential augmentation of systemic and local inflammation precipitated by HC, are not completely understood. CD47 is a transmembrane protein known to abort crucial inflammatory pathways. Our studies have examined the interrelation between HC, inflammation, and ISR and investigated the therapeutic potential of stents coated with a CD47-derived peptide (pepCD47) in the hypercholesterolemic rabbit model. METHODS PepCD47 was immobilized on metal foils and stents using polybisphosphonate coordination chemistry and pyridyldithio/thiol conjugation. Cytokine expression in buffy coat-derived cells cultured over bare metal (BM) and pepCD47-derivatized foils demonstrated an M2/M1 macrophage shift with pepCD47 coating. HC and normocholesterolemic (NC) rabbit cohorts underwent bilateral implantation of BM and pepCD47 stents (HC) or BM stents only (NC) in the iliac location. RESULTS A 40 % inhibition of cell attachment to pepCD47-modified compared to BM surfaces was observed. HC increased neointimal growth at 4 weeks post BM stenting. These untoward outcomes were mitigated in hypercholesterolemic rabbits treated with pepCD47-derivatized stents. Compared to NC animals, inflammatory cytokine immunopositivity and macrophage infiltration of peri-strut areas increased in HC animals and were attenuated in HC rabbits treated with pepCD47 stents. CONCLUSIONS Augmented inflammatory responses underlie severe ISR morphology in hypercholesterolemic rabbits. Blockage of initial platelet and leukocyte attachment to stent struts through CD47 functionalization of stents mitigates the pro-restenotic effects of hypercholesterolemia.
Collapse
Affiliation(s)
- Ilia Fishbein
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| | - Vaishali V Inamdar
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ivan S Alferiev
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - George Bratinov
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Menekhem M Zviman
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | | | | | - Kristin L Gardiner
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Robert J Levy
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Stanley J Stachelek
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
32
|
Bajpai D, Rajasekar A. Genetic Association of ICAM-1 (rs5498) Gene Polymorphism With Susceptibility to Stage II Grade B Periodontitis: A Case-Control Study in South Indian Population. Cureus 2024; 16:e56629. [PMID: 38650775 PMCID: PMC11034711 DOI: 10.7759/cureus.56629] [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: 02/19/2024] [Accepted: 03/21/2024] [Indexed: 04/25/2024] Open
Abstract
INTRODUCTION In the contemporary perspective, periodontitis is considered a complex issue triggered and perpetuated by bacteria but strongly influenced by the way the body reacts to bacterial plaque. Recent research has indicated that variations in genes might have an impact on the development of periodontitis. This study was conducted to explore a probable link between the genetic variations in intercellular adhesion molecule-1 (ICAM-1) represented by rs5498 and the occurrence of periodontitis. Methods: A total of 100 participants, 50 with periodontitis and 50 with periodontally healthy or mild gingivitis, were recruited for this study. Whole blood drawn from the participants was used to obtain genomic DNA. The ICAM-1 gene polymorphism (rs5498) was determined using polymerase chain reaction (PCR) amplification and digestion. The ICAM-1 gene's flanking primers were used to amp up the DNA. For statistical analysis, the genotype that was analyzed using the pattern of restriction fragment length polymorphism was recorded. The Chi-square test compared genotype and allele frequency distributions between both groups. The odds ratio with 95% confidence intervals with each individual allele or genotype was used to compute the risk. Statistical significance was established in all tests when the p-value was less than 0.05. RESULTS There was no discernible difference between the genotype frequencies of patients and controls χ2df (P = 0.6065). The findings demonstrated that no significant difference was present between the two groups for homozygous or heterozygous mutant genotypes (AA vs. AG+GG; P = 0.6854). There was no discernible difference in the detected frequencies of the A allele (58% vs. 61%), G allele (42% vs. 39%), TT (16% vs. 24%), AG (40% vs. 36%), and TT genotypes in the studied groups. CONCLUSION According to the results of the current investigation, the ICAM-1 (rs5498) gene polymorphism is not associated with periodontitis in the population investigated.
Collapse
Affiliation(s)
- Devika Bajpai
- Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Arvina Rajasekar
- Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
33
|
Ailuno G, Baldassari S, Balboni A, Pastorino S, Zuccari G, Cortese K, Barbieri F, Drava G, Florio T, Caviglioli G. Development of Biotinylated Liposomes Encapsulating Metformin for Therapeutic Targeting of Inflammation-Based Diseases. Pharmaceutics 2024; 16:235. [PMID: 38399288 PMCID: PMC10893420 DOI: 10.3390/pharmaceutics16020235] [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: 12/28/2023] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Inflammation is a physiological response to a damaging stimulus but sometimes can be the cause of the onset of neurodegenerative diseases, atherosclerosis, and cancer. These pathologies are characterized by the overexpression of inflammatory markers like endothelial adhesion molecules, such as Vascular Cell Adhesion Molecule-1 (VCAM-1). In the present work, the development of liposomes for therapeutic targeted delivery to inflamed endothelia is described. The idea is to exploit a three-step pretargeting system based on the biotin-avidin high-affinity interaction: the first step involves a previously described biotin derivative bearing a VCAM-1 binding peptide; in the second step, the avidin derivative NeutrAvidinTM, which strongly binds to the biotin moiety, is injected; the final step is the administration of biotinylated liposomes that would bind to NeutravidinTM immobilized onto VCAM-1 overexpressing endothelium. Stealth biotinylated liposomes, prepared via the thin film hydration method followed by extrusion and purification via size exclusion chromatography, have been thoroughly characterized for their chemico-physical and morphological features and loaded with metformin hydrochloride, a potential anti-inflammatory agent. The three-step system, tested in vitro on different cell lines via confocal microscopy, FACS analysis and metformin uptake, has proved its suitability for therapeutic applications.
Collapse
Affiliation(s)
- Giorgia Ailuno
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (S.B.); (A.B.); (G.Z.); (G.D.); (G.C.)
| | - Sara Baldassari
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (S.B.); (A.B.); (G.Z.); (G.D.); (G.C.)
| | - Alice Balboni
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (S.B.); (A.B.); (G.Z.); (G.D.); (G.C.)
| | - Sara Pastorino
- Territorial Pharmacy of Azienda Sociosanitaria Ligure 2, Via Carlo Collodi 13, 17100 Savona, Italy;
| | - Guendalina Zuccari
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (S.B.); (A.B.); (G.Z.); (G.D.); (G.C.)
| | - Katia Cortese
- Department of Experimental Medicine, University of Genoa, Via Antonio de Toni 14, 16132 Genova, Italy;
| | - Federica Barbieri
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV 2, 16132 Genova, Italy; (F.B.); (T.F.)
| | - Giuliana Drava
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (S.B.); (A.B.); (G.Z.); (G.D.); (G.C.)
| | - Tullio Florio
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV 2, 16132 Genova, Italy; (F.B.); (T.F.)
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Gabriele Caviglioli
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (S.B.); (A.B.); (G.Z.); (G.D.); (G.C.)
| |
Collapse
|
34
|
Ferrell M, Wang Z, Anderson JT, Li XS, Witkowski M, DiDonato JA, Hilser JR, Hartiala JA, Haghikia A, Cajka T, Fiehn O, Sangwan N, Demuth I, König M, Steinhagen-Thiessen E, Landmesser U, Tang WHW, Allayee H, Hazen SL. A terminal metabolite of niacin promotes vascular inflammation and contributes to cardiovascular disease risk. Nat Med 2024; 30:424-434. [PMID: 38374343 PMCID: PMC11841810 DOI: 10.1038/s41591-023-02793-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 12/22/2023] [Indexed: 02/21/2024]
Abstract
Despite intensive preventive cardiovascular disease (CVD) efforts, substantial residual CVD risk remains even for individuals receiving all guideline-recommended interventions. Niacin is an essential micronutrient fortified in food staples, but its role in CVD is not well understood. In this study, untargeted metabolomics analysis of fasting plasma from stable cardiac patients in a prospective discovery cohort (n = 1,162 total, n = 422 females) suggested that niacin metabolism was associated with incident major adverse cardiovascular events (MACE). Serum levels of the terminal metabolites of excess niacin, N1-methyl-2-pyridone-5-carboxamide (2PY) and N1-methyl-4-pyridone-3-carboxamide (4PY), were associated with increased 3-year MACE risk in two validation cohorts (US n = 2,331 total, n = 774 females; European n = 832 total, n = 249 females) (adjusted hazard ratio (HR) (95% confidence interval) for 2PY: 1.64 (1.10-2.42) and 2.02 (1.29-3.18), respectively; for 4PY: 1.89 (1.26-2.84) and 1.99 (1.26-3.14), respectively). Phenome-wide association analysis of the genetic variant rs10496731, which was significantly associated with both 2PY and 4PY levels, revealed an association of this variant with levels of soluble vascular adhesion molecule 1 (sVCAM-1). Further meta-analysis confirmed association of rs10496731 with sVCAM-1 (n = 106,000 total, n = 53,075 females, P = 3.6 × 10-18). Moreover, sVCAM-1 levels were significantly correlated with both 2PY and 4PY in a validation cohort (n = 974 total, n = 333 females) (2PY: rho = 0.13, P = 7.7 × 10-5; 4PY: rho = 0.18, P = 1.1 × 10-8). Lastly, treatment with physiological levels of 4PY, but not its structural isomer 2PY, induced expression of VCAM-1 and leukocyte adherence to vascular endothelium in mice. Collectively, these results indicate that the terminal breakdown products of excess niacin, 2PY and 4PY, are both associated with residual CVD risk. They also suggest an inflammation-dependent mechanism underlying the clinical association between 4PY and MACE.
Collapse
Affiliation(s)
- Marc Ferrell
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Systems Biology and Bioinformatics Program, Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA
| | - Zeneng Wang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James T Anderson
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Xinmin S Li
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Marco Witkowski
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Joseph A DiDonato
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James R Hilser
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jaana A Hartiala
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Arash Haghikia
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tomas Cajka
- West Coast Metabolomics Center, University of California, Davis, Davis, CA, USA
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, Davis, CA, USA
| | - Naseer Sangwan
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ilja Demuth
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Maximilian König
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - W H Wilson Tang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Hooman Allayee
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stanley L Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
35
|
Boutagy NE, Gamez-Mendez A, Fowler JW, Zhang H, Chaube BK, Esplugues E, Kuo A, Lee S, Horikami D, Zhang J, Citrin KM, Singh AK, Coon BG, Lee MY, Suarez Y, Fernandez-Hernando C, Sessa WC. Dynamic metabolism of endothelial triglycerides protects against atherosclerosis in mice. J Clin Invest 2024; 134:e170453. [PMID: 38175710 PMCID: PMC10866653 DOI: 10.1172/jci170453] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Blood vessels are continually exposed to circulating lipids, and elevation of ApoB-containing lipoproteins causes atherosclerosis. Lipoprotein metabolism is highly regulated by lipolysis, largely at the level of the capillary endothelium lining metabolically active tissues. How large blood vessels, the site of atherosclerotic vascular disease, regulate the flux of fatty acids (FAs) into triglyceride-rich (TG-rich) lipid droplets (LDs) is not known. In this study, we showed that deletion of the enzyme adipose TG lipase (ATGL) in the endothelium led to neutral lipid accumulation in vessels and impaired endothelial-dependent vascular tone and nitric oxide synthesis to promote endothelial dysfunction. Mechanistically, the loss of ATGL led to endoplasmic reticulum stress-induced inflammation in the endothelium. Consistent with this mechanism, deletion of endothelial ATGL markedly increased lesion size in a model of atherosclerosis. Together, these data demonstrate that the dynamics of FA flux through LD affects endothelial cell homeostasis and consequently large vessel function during normal physiology and in a chronic disease state.
Collapse
Affiliation(s)
- Nabil E. Boutagy
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
| | - Ana Gamez-Mendez
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
| | - Joseph W.M. Fowler
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
| | - Hanming Zhang
- Vascular Biology and Therapeutics Program, and
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Bal K. Chaube
- Vascular Biology and Therapeutics Program, and
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Enric Esplugues
- Vascular Biology and Therapeutics Program, and
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Andrew Kuo
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Sungwoon Lee
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
| | - Daiki Horikami
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
| | - Jiasheng Zhang
- Department of Cardiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kathryn M. Citrin
- Vascular Biology and Therapeutics Program, and
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Abhishek K. Singh
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
| | - Brian G. Coon
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Monica Y. Lee
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago School of Medicine, Chicago, Illinois, USA
| | - Yajaira Suarez
- Vascular Biology and Therapeutics Program, and
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Carlos Fernandez-Hernando
- Vascular Biology and Therapeutics Program, and
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - William C. Sessa
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
- Department of Cardiology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
36
|
Ma XF, Zhou YR, Zhou ZX, Liu HT, Zhou BB, Deng NH, Zhou K, Tian Z, Wu ZF, Liu XY, Fu MG, Jiang ZS. TRIM65 Suppresses oxLDL-induced Endothelial Inflammation by Interaction with VCAM-1 in Atherogenesis. Curr Med Chem 2024; 31:4898-4911. [PMID: 37608612 DOI: 10.2174/0929867331666230822152350] [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: 12/27/2022] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND AND OBJECTIVE Endothelial cell activation, characterized by increased levels of vascular cell adhesion molecule 1 (VCAM-1), plays a crucial role in the development of atherosclerosis (AS). Therefore, inhibition of VCAM-1-mediated inflammatory response is of great significance in the prevention and treatment of AS. The tripartite motif (TRIM) protein-TRIM65 is involved in the regulation of cancer development, antivirals and inflammation. We aimed to study the functions of TRIM65 in regulating endothelial inflammation by interacting with VCAM-1 in atherogenesis. METHODS AND RESULTS In vitro, we report that human umbilical vein endothelial cells (HUVECs) treated with oxidized low-density lipoprotein (oxLDL) significantly upregulate the expression of TRIM65 in a time- and dose-dependent manner. Overexpression of TRIM65 reduces oxLDL-triggered VCAM-1 protein expression, decreases monocyte adhesion to HUVECs and inhibits the production of the inflammatory cytokines IL-1β, IL-6, IL-8, and TNF-α as well as endothelial oxLDL transcytosis. In contrast, siRNA-mediated knockdown of TRIM65 promotes the expression of VCAM-1, resulting in increased adhesion of monocytes and the release of the inflammatory cytokines IL-1β, IL-6, IL-8, and TNF-α and enhances endothelial oxLDL transcytosis. In vivo, we measured the high expression of TRIM65 in ApoE-/- mouse aortic plaques compared to C57BL/6J mouse aortic plaques. Then, we examined whether the blood levels of VCAM-1 were higher in TRIM65 knockout ApoE-/- mice than in control mice induced by a Western diet. Furthermore, Western blot results showed that the protein expression of VCAM-1 was markedly enhanced in TRIM65 knockout ApoE-/- mouse aortic tissues compared to that of the controls. Immunofluorescence staining revealed that the expression of VCAM-1 was significantly increased in atherosclerotic plaques of TRIM65-/-/ApoE-/- aortic vessels compared to ApoE-/- controls. Mechanistically, TRIM65 specifically interacts with VCAM-1 and targets it for K48-linked ubiquitination. CONCLUSION Our studies indicate that TRIM65 attenuates the endothelial inflammatory response by targeting VCAM-1 for ubiquitination and provides a potential therapeutic target for the inhibition of endothelial inflammation in AS.
Collapse
Affiliation(s)
- Xiao-Feng Ma
- Department of Cardiology, Affiliated Nanhua Hospital, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Yi-Ren Zhou
- Department of Cardiology, Affiliated Nanhua Hospital, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Zhi-Xiang Zhou
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Hui-Ting Liu
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Bo-Bin Zhou
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Nian-Hua Deng
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Kun Zhou
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Zhen Tian
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Ze-Fan Wu
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Xi-Yan Liu
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Ming-Gui Fu
- Department of Basic Medical Science, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Zhi-Sheng Jiang
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| |
Collapse
|
37
|
Otunla AA, Shanmugarajah K, Davies AH, Lucia Madariaga M, Shalhoub J. The Biological Parallels Between Atherosclerosis and Cardiac Allograft Vasculopathy: Implications for Solid Organ Chronic Rejection. Cardiol Rev 2024; 32:2-11. [PMID: 38051983 DOI: 10.1097/crd.0000000000000437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Atherosclerosis and solid organ chronic rejection are pervasive chronic disease states that account for significant morbidity and mortality in developed countries. Recently, a series of shared molecular pathways have emerged, revealing biological parallels from early stages of development up to the advanced forms of pathology. These shared mechanistic processes are inflammatory in nature, reflecting the importance of inflammation in both disorders. Vascular inflammation triggers endothelial dysfunction and disease initiation through aberrant vasomotor control and shared patterns of endothelial activation. Endothelial dysfunction leads to the recruitment of immune cells and the perpetuation of the inflammatory response. This drives lesion formation through the release of key cytokines such as IFN-y, TNF-alpha, and IL-2. Continued interplay between the adaptive and innate immune response (represented by T lymphocytes and macrophages, respectively) promotes lesion instability and thrombotic complications; hallmarks of advanced disease in both atherosclerosis and solid organ chronic rejection. The aim of this study is to identify areas of overlap between atherosclerosis and chronic rejection. We then discuss new approaches to improve current understanding of the pathophysiology of both disorders, and eventually design novel therapeutics.
Collapse
Affiliation(s)
- Afolarin A Otunla
- From the Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | | | - Alun H Davies
- Section of Vascular Surgery, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
- Imperial Vascular Unit, Imperial College Healthcare NHS Trust, London, United Kingdom
| | | | - Joseph Shalhoub
- Section of Vascular Surgery, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
- Imperial Vascular Unit, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
38
|
Chetan IM, Vesa ȘC, Domokos Gergely B, Beyer RS, Tomoaia R, Cabau G, Vulturar DM, Pop D, Todea D. Increased Levels of VCAM-1 in Patients with High Cardiovascular Risk and Obstructive Sleep Apnea Syndrome. Biomedicines 2023; 12:48. [PMID: 38255155 PMCID: PMC10813101 DOI: 10.3390/biomedicines12010048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
(1) Background: Although obstructive sleep apnea (OSA) is associated with increased cardiovascular morbidity, the link between OSA and cardiovascular disease (CVD) is not completely elucidated. Thus, we aim to assess cardiovascular risk (CVR) using SCORE 2 and SCORE 2 for older persons (SCORE 2OP), and to evaluate the association between the endothelial biomarkers VCAM-1, ICAM-1, epicardial fat, and sleep study parameters in order to improve current clinical practices and better understand the short-and long-term CVRs in OSA patients. (2) Methods: 80 OSA patients and 37 healthy volunteers were enrolled in the study. SCORE2 and SCORE 2 OP regional risk charts (validated algorithms to predict the 10-year risk of first-onset CVD) were used for the analysis of CVR. Two-dimensional echocardiography was performed on all patients and epicardial fat thickness was measured. VCAM-1 and ICAM-1 serum levels were assessed in all patients. (3) Results: OSA patients were classified as being at high CVR, regardless of the type of score achieved. Increased EFT was observed in the OSA group. VCAM-1 was associated with a high CVR in OSA patients, but no significant correlation was observed between adhesion molecules and epicardial fat thickness. (4) Conclusions: OSA patients have a high CVR according to the SCORE 2 and SCORE 2OP risk scores. VCAM-1 may be associated with a high CVR in OSA patients. Extending conventional risk stratification scores by adding other potential biomarkers improves the risk stratification and guide treatment eligibility for CVD prevention in the OSA population.
Collapse
Affiliation(s)
- Ioana-Maria Chetan
- Department of Pneumology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.-M.C.); (B.D.G.); (D.M.V.); (D.T.)
| | - Ștefan Cristian Vesa
- Department of Pharmacology, Toxicology and Clinical Pharmacology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Bianca Domokos Gergely
- Department of Pneumology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.-M.C.); (B.D.G.); (D.M.V.); (D.T.)
| | | | - Raluca Tomoaia
- Department of Cardiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (R.T.)
| | - Georgiana Cabau
- Department of Medical Genetics, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Damiana Maria Vulturar
- Department of Pneumology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.-M.C.); (B.D.G.); (D.M.V.); (D.T.)
| | - Dana Pop
- Department of Cardiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (R.T.)
| | - Doina Todea
- Department of Pneumology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.-M.C.); (B.D.G.); (D.M.V.); (D.T.)
| |
Collapse
|
39
|
Chan A, Torelli S, Cheng E, Batchelder R, Waliany S, Neal J, Witteles R, Nguyen P, Cheng P, Zhu H. Immunotherapy-Associated Atherosclerosis: A Comprehensive Review of Recent Findings and Implications for Future Research. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2023; 25:715-735. [PMID: 38213548 PMCID: PMC10776491 DOI: 10.1007/s11936-023-01024-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 01/13/2024]
Abstract
Purpose of the Review Even as immune checkpoint inhibitors (ICIs) have transformed the lifespan of many patients, they may also trigger acceleration of long-term cardiovascular disease. Our review aims to examine the current landscape of research on ICI-mediated atherosclerosis and address key questions regarding its pathogenesis and impact on patient management. Recent Findings Preclinical mouse models suggest that T cell dysregulation and proatherogenic cytokine production are key contributors to plaque development after checkpoint inhibition. Clinical data also highlight the significant burden of atherosclerotic cardiovascular disease (ASCVD) in patients on immunotherapy, although the value of proactively preventing and treating ASCVD in this population remains an open area of inquiry. Current treatment options include dietary/lifestyle modification and traditional medications to manage hypertension, hyperlipidemia, and diabetes risk factors; no current targeted therapies exist. Summary Early identification of high-risk patients is crucial for effective preventive strategies and timely intervention. Future research should focus on refining screening tools, elucidating targetable mechanisms driving ICI atherosclerosis, and evaluating long-term cardiovascular outcomes in cancer survivors who received immunotherapy. Moreover, close collaboration between oncologists and cardiologists is essential to optimize patient outcomes.
Collapse
Affiliation(s)
- Antonia Chan
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Stefan Torelli
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Evaline Cheng
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Ryan Batchelder
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Sarah Waliany
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Joel Neal
- Department of Medicine, Division of Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA USA
| | - Ronald Witteles
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Patricia Nguyen
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| | - Paul Cheng
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| | - Han Zhu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| |
Collapse
|
40
|
Terasawa M, Zang L, Hiramoto K, Shimada Y, Mitsunaka M, Uchida R, Nishiura K, Matsuda K, Nishimura N, Suzuki K. Oral Administration of Rhamnan Sulfate from Monostroma nitidum Suppresses Atherosclerosis in ApoE-Deficient Mice Fed a High-Fat Diet. Cells 2023; 12:2666. [PMID: 37998401 PMCID: PMC10670814 DOI: 10.3390/cells12222666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023] Open
Abstract
Oral administration of rhamnan sulfate (RS), derived from the seaweed Monostroma nitidum, markedly suppresses inflammatory damage in the vascular endothelium and organs of lipopolysaccharide-treated mice. This study aimed to analyze whether orally administered RS inhibits the development of atherosclerosis, a chronic inflammation of the arteries. ApoE-deficient female mice were fed a normal or high-fat diet (HFD) with or without RS for 12 weeks. Immunohistochemical and mRNA analyses of atherosclerosis-related genes were performed. The effect of RS on the migration of RAW264.7 cells was also examined in vitro. RS administration suppressed the increase in blood total cholesterol and triglyceride levels. In the aorta of HFD-fed mice, RS reduced vascular smooth muscle cell proliferation, macrophage accumulation, and elevation of VCAM-1 and inhibited the reduction of Robo4. Increased mRNA levels of Vcam1, Mmp9, and Srebp1 in atherosclerotic areas of HFD-fed mice were also suppressed with RS. Moreover, RS directly inhibited the migration of RAW264.7 cells in vitro. Thus, in HFD-fed ApoE-deficient mice, oral administration of RS ameliorated abnormal lipid metabolism and reduced vascular endothelial inflammation and hyperpermeability, macrophage infiltration and accumulation, and smooth muscle cell proliferation in the arteries leading to atherosclerosis. These results suggest that RS is an effective functional food for the prevention of atherosclerosis.
Collapse
Affiliation(s)
- Masahiro Terasawa
- Konan Chemical Manufacturing Co., Ltd., Kitagomizuka, Kusu-cho, Yokkaichi 510-0103, Japan; (M.T.); (R.U.); (K.N.); (K.M.)
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Minamitamagaki-cho, Suzuka 513-8670, Japan;
| | - Liqing Zang
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan; (L.Z.); (N.N.)
| | - Keiichi Hiramoto
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Minamitamagaki-cho, Suzuka 513-8670, Japan;
| | - Yasuhito Shimada
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu 514-8507, Japan; (Y.S.); (M.M.)
| | - Mari Mitsunaka
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu 514-8507, Japan; (Y.S.); (M.M.)
| | - Ryota Uchida
- Konan Chemical Manufacturing Co., Ltd., Kitagomizuka, Kusu-cho, Yokkaichi 510-0103, Japan; (M.T.); (R.U.); (K.N.); (K.M.)
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Minamitamagaki-cho, Suzuka 513-8670, Japan;
| | - Kaoru Nishiura
- Konan Chemical Manufacturing Co., Ltd., Kitagomizuka, Kusu-cho, Yokkaichi 510-0103, Japan; (M.T.); (R.U.); (K.N.); (K.M.)
| | - Koichi Matsuda
- Konan Chemical Manufacturing Co., Ltd., Kitagomizuka, Kusu-cho, Yokkaichi 510-0103, Japan; (M.T.); (R.U.); (K.N.); (K.M.)
| | - Norihiro Nishimura
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan; (L.Z.); (N.N.)
| | - Koji Suzuki
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Minamitamagaki-cho, Suzuka 513-8670, Japan;
| |
Collapse
|
41
|
Lee SK, Malik RA, Zhou J, Wang W, Gross PL, Weitz JI, Ramachandran R, Trigatti BL. PAR4 Inhibition Reduces Coronary Artery Atherosclerosis and Myocardial Fibrosis in SR-B1/LDLR Double Knockout Mice. Arterioscler Thromb Vasc Biol 2023; 43:2165-2178. [PMID: 37675637 PMCID: PMC10597419 DOI: 10.1161/atvbaha.123.319767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND SR-B1 (scavenger receptor class B type 1)/LDLR (low-density lipoprotein receptor) double knockout mice fed a high-fat, high-cholesterol diet containing cholate exhibit coronary artery disease characterized by occlusive coronary artery atherosclerosis, platelet accumulation in coronary arteries, and myocardial fibrosis. Platelets are involved in atherosclerosis development, and PAR (protease-activated receptor) 4 has a prominent role in platelet function in mice. However, the role of PAR4 on coronary artery disease in mice has not been tested. METHODS We tested the effects of a PAR4 inhibitory pepducin (RAG8) on diet-induced aortic sinus and coronary artery atherosclerosis, platelet accumulation in atherosclerotic coronary arteries, and myocardial fibrosis in SR-B1/LDLR double knockout mice. SR-B1/LDLR double knockout mice were fed a high-fat, high-cholesterol diet containing cholate and injected daily with 20 mg/kg of either the RAG8 pepducin or a control reverse-sequence pepducin (SRQ8) for 20 days. RESULTS Platelets from the RAG8-treated mice exhibited reduced thrombin and PAR4 agonist peptide-mediated activation compared with those from control SRQ8-treated mice when tested ex vivo. Although aortic sinus atherosclerosis levels did not differ, RAG8-treated mice exhibited reduced coronary artery atherosclerosis, reduced platelet accumulation in atherosclerotic coronary arteries, and reduced myocardial fibrosis. These protective effects were not accompanied by changes in circulating lipids, inflammatory cytokines, or immune cells. However, RAG8-treated mice exhibited reduced VCAM-1 (vascular cell adhesion molecule 1) protein levels in nonatherosclerotic coronary artery cross sections and reduced leukocyte accumulation in atherosclerotic coronary artery cross sections compared with those from SRQ8-treated mice. CONCLUSIONS The PAR4 inhibitory RAG8 pepducin reduced coronary artery atherosclerosis and myocardial fibrosis in SR-B1/LDLR double knockout mice fed a high-fat, high-cholesterol diet containing cholate. Furthermore, RAG8 reduced VCAM-1 in nonatherosclerotic coronary arteries and reduced leukocyte and platelet accumulation in atherosclerotic coronary arteries. These findings identify PAR4 as an attractive target in reducing coronary artery disease development, and the use of RAG8 may potentially be beneficial in cardiovascular disease.
Collapse
Affiliation(s)
- Samuel K. Lee
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Ontario, Canada (S.K.L., W.W., J.I.W., B.L.T.)
| | - Rida A. Malik
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Department of Medicine (R.A.M., J.Z., P.L.G., J.I.W.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
| | - Ji Zhou
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Department of Medicine (R.A.M., J.Z., P.L.G., J.I.W.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
| | - Wei Wang
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Ontario, Canada (S.K.L., W.W., J.I.W., B.L.T.)
| | - Peter L. Gross
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Department of Medicine (R.A.M., J.Z., P.L.G., J.I.W.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
| | - Jeffrey I. Weitz
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Department of Medicine (R.A.M., J.Z., P.L.G., J.I.W.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Ontario, Canada (S.K.L., W.W., J.I.W., B.L.T.)
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.R.)
| | - Bernardo L. Trigatti
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Ontario, Canada (S.K.L., W.W., J.I.W., B.L.T.)
| |
Collapse
|
42
|
Chang HW, Hsu MJ, Chien LN, Chi NF, Yu MC, Chen HC, Lin YF, Hu CJ. Role of the Autism Risk Gene Shank3 in the Development of Atherosclerosis: Insights from Big Data and Mechanistic Analyses. Cells 2023; 12:2546. [PMID: 37947623 PMCID: PMC10647789 DOI: 10.3390/cells12212546] [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: 09/07/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Increased medical attention is needed as the prevalence of autism spectrum disorder (ASD) rises. Both cardiovascular disorder (CVD) and hyperlipidemia are closely associated with adult ASD. Shank3 plays a key genetic role in ASD. We hypothesized that Shank3 contributes to CVD development in young adults with ASD. In this study, we investigated whether Shank3 facilitates the development of atherosclerosis. Using Gene Set Enrichment Analysis software (Version No.: GSEA-4.0.3), we analyzed the data obtained from Shank3 knockout mice (Gene Expression Omnibus database), a human population-based study cohort (from Taiwan's National Health Insurance Research Database), and a Shank3 knockdown cellular model. Shank3 knockout upregulated the expression of genes of cholesterol homeostasis and fatty acid metabolism but downregulated the expression of genes associated with inflammatory responses. Individuals with autism had higher risks of hyperlipidemia (adjusted hazard ratio (aHR): 1.39; p < 0.001), major adverse cardiac events (aHR: 2.67; p < 0.001), and stroke (aHR: 3.55; p < 0.001) than age- and sex-matched individuals without autism did. Shank3 downregulation suppressed tumor necrosis factor-α-induced fatty acid synthase expression; vascular cell adhesion molecule 1 expression; and downstream signaling pathways involving p38, Jun N-terminal kinase, and nuclear factor-κB. Thus, Shank3 may influence the development of early-onset atherosclerosis and CVD in ASD. Furthermore, regulating Shank3 expression may reduce inflammation-related disorders, such as atherosclerosis, by inhibiting tumor necrosis factor-alpha-mediated inflammatory cascades.
Collapse
Affiliation(s)
- Hsiu-Wen Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Neurology, Sijhih Cathay General Hospital, New Taipei City 22174, Taiwan
| | - Ming-Jen Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan (H.-C.C.)
| | - Li-Nien Chien
- Institute of Health and Welfare Policy, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
| | - Nai-Fang Chi
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei 11267, Taiwan;
| | - Meng-Chieh Yu
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan (H.-C.C.)
| | - Hsiu-Chen Chen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan (H.-C.C.)
| | - Yuan-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
43
|
Pickett JR, Wu Y, Zacchi LF, Ta HT. Targeting endothelial vascular cell adhesion molecule-1 in atherosclerosis: drug discovery and development of vascular cell adhesion molecule-1-directed novel therapeutics. Cardiovasc Res 2023; 119:2278-2293. [PMID: 37595265 PMCID: PMC10597632 DOI: 10.1093/cvr/cvad130] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/14/2023] [Accepted: 07/04/2023] [Indexed: 08/20/2023] Open
Abstract
Vascular cell adhesion molecule-1 (VCAM-1) has been well established as a critical contributor to atherosclerosis and consequently as an attractive therapeutic target for anti-atherosclerotic drug candidates. Many publications have demonstrated that disrupting the VCAM-1 function blocks monocyte infiltration into the sub-endothelial space, which effectively prevents macrophage maturation and foam cell transformation necessary for atherosclerotic lesion formation. Currently, most VCAM-1-inhibiting drug candidates in pre-clinical and clinical testing do not directly target VCAM-1 itself but rather down-regulate its expression by inhibiting upstream cytokines and transcriptional regulators. However, the pleiotropic nature of these regulators within innate immunity means that optimizing dosage to a level that suppresses pathological activity while preserving normal physiological function is extremely challenging and oftentimes infeasible. In recent years, highly specific pharmacological strategies that selectively inhibit VCAM-1 function have emerged, particularly peptide- and antibody-based novel therapeutics. Studies in such VCAM-1-directed therapies so far remain scarce and are limited by the constraints of current experimental atherosclerosis models in accurately representing the complex pathophysiology of the disease. This has prompted the need for a comprehensive review that recounts the evolution of VCAM-1-directed pharmaceuticals and addresses the current challenges in novel anti-atherosclerotic drug development.
Collapse
Affiliation(s)
- Jessica R Pickett
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, West Creek Road, Nathan, QLD 4111, Australia
- School of Environment and Science, Griffith University, Kessels Road, Nathan, QLD 4111, Australia
| | - Yuao Wu
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, West Creek Road, Nathan, QLD 4111, Australia
| | - Lucia F Zacchi
- Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, St. Lucia, QLD 4072, Australia
- School of Chemistry and Molecular Biosciences, the University of Queensland, St. Lucia, QLD 4072, Australia
| | - Hang T Ta
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, West Creek Road, Nathan, QLD 4111, Australia
- School of Environment and Science, Griffith University, Kessels Road, Nathan, QLD 4111, Australia
| |
Collapse
|
44
|
Vu T, Smith JA. The pathophysiology and management of depression in cardiac surgery patients. Front Psychiatry 2023; 14:1195028. [PMID: 37928924 PMCID: PMC10623009 DOI: 10.3389/fpsyt.2023.1195028] [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: 03/27/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Background Depression is common in the cardiac surgery population. This contemporary narrative review aims to explore the main pathophysiological disturbances underpinning depression specifically within the cardiac surgery population. The common non-pharmacological and pharmacological management strategies used to manage depression within the cardiac surgery patient population are also explored. Methods A total of 1291 articles were identified through Ovid Medline and Embase. The findings from 39 studies were included for qualitative analysis in this narrative review. Results Depression is associated with several pathophysiological and behavioral factors which increase the likelihood of developing coronary heart disease which may ultimately require surgical intervention. The main pathophysiological factors contributing to depression are well characterized and include autonomic nervous system dysregulation, excessive inflammation and disruption of the hypothalamic-pituitary-adrenal axis. There are also several behavioral factors in depressed patients associated with the development of coronary heart disease including poor diet, insufficient exercise, poor compliance with medications and reduced adherence to cardiac rehabilitation. The common preventative and management modalities used for depression following cardiac surgery include preoperative and peri-operative education, cardiac rehabilitation, cognitive behavioral therapy, religion/prayer/spirituality, biobehavioral feedback, anti-depressant medications, and statins. Conclusion This contemporary review explores the pathophysiological mechanisms leading to depression following cardiac surgery and the current management modalities. Further studies on the preventative and management strategies for postoperative depression in the cardiac surgery patient population are warranted.
Collapse
Affiliation(s)
- Tony Vu
- Department of Cardiothoracic Surgery, The Alfred Hospital, Melbourne, VIC, Australia
- Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| | - Julian A. Smith
- Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
- Department of Cardiothoracic Surgery, Monash Health, Melbourne, VIC, Australia
| |
Collapse
|
45
|
Teichmann E, Blessing E, Hinz B. Non-Psychoactive Phytocannabinoids Inhibit Inflammation-Related Changes of Human Coronary Artery Smooth Muscle and Endothelial Cells. Cells 2023; 12:2389. [PMID: 37830604 PMCID: PMC10571842 DOI: 10.3390/cells12192389] [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: 08/18/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
Atherosclerosis is associated with vascular smooth muscle cell proliferation, chronic vascular inflammation, and leukocyte adhesion. In view of the cardioprotective effects of cannabinoids described in recent years, the present study investigated the impact of the non-psychoactive phytocannabinoids cannabidiol (CBD) and tetrahydrocannabivarin (THCV) on proliferation and migration of human coronary artery smooth muscle cells (HCASMC) and on inflammatory markers in human coronary artery endothelial cells (HCAEC). In HCASMC, CBD and THCV at nontoxic concentrations exhibited inhibitory effects on platelet-derived growth factor-triggered proliferation (CBD) and migration (CBD, THCV). When interleukin (IL)-1β- and lipopolysaccharide (LPS)-stimulated HCAEC were examined, both cannabinoids showed a concentration-dependent decrease in the expression of vascular cell adhesion molecule-1 (VCAM-1), which was mediated independently of classical cannabinoid receptors and was not accompanied by a comparable inhibition of intercellular adhesion molecule-1. Further inhibitor experiments demonstrated that reactive oxygen species, p38 mitogen-activated protein kinase activation, histone deacetylase, and nuclear factor κB (NF-κB) underlie IL-1β- and LPS-induced expression of VCAM-1. In this context, CBD and THCV were shown to inhibit phosphorylation of NF-κB regulators in LPS- but not IL-1β-stimulated HCAEC. Stimulation of HCAEC with IL-1β and LPS was associated with increased adhesion of monocytes, which, however, could not be significantly abolished by CBD and THCV. In summary, the results highlight the potential of the non-psychoactive cannabinoids CBD and THCV to regulate inflammation-related changes in HCASMC and HCAEC. Considering their effect on both cell types studied, further preclinical studies could address the use of CBD and THCV in drug-eluting stents for coronary interventions.
Collapse
Affiliation(s)
| | | | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany; (E.T.); (E.B.)
| |
Collapse
|
46
|
Bourrie BCT, Forgie AJ, Makarowski A, Cotter PD, Richard C, Willing BP. Consumption of kefir made with traditional microorganisms resulted in greater improvements in LDL cholesterol and plasma markers of inflammation in males when compared to a commercial kefir: a randomized pilot study. Appl Physiol Nutr Metab 2023; 48:668-677. [PMID: 37224566 DOI: 10.1139/apnm-2022-0463] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Kefir has long been associated with health benefits; however, recent evidence suggests that these benefits are dependent on the specific microbial composition of the kefir consumed. This study aimed to compare how consumption of a commercial kefir without traditional kefir organisms and a pitched kefir containing traditional organisms affected plasma lipid levels, glucose homeostasis, and markers of endothelial function and inflammation in males with elevated LDL cholesterol. We utilized a crossover design in n = 21 participants consisting of two treatments of 4 weeks each in random order separated by a 4-week washout. Participants received either commercial kefir or pitched kefir containing traditional kefir organisms for each treatment period. Participants consumed 2 servings of kefir (350 g) per day. Plasma lipid profile, glucose, insulin, markers of endothelial function, and inflammation were measured in the fasting state before and after each treatment period. Differences within each treatment period and comparison of treatment delta values were performed using paired t tests and Wilcoxon signed-rank test, respectively. When compared to baseline, pitched kefir consumption reduced LDL-C, ICAM-1, and VCAM-1, while commercial kefir consumption increased TNF-α. Pitched kefir consumption resulted in greater reductions in IL-8, CRP, VCAM-1, and TNF-α when compared to commercial kefir consumption. These findings provide strong evidence that microbial composition is an important factor in the metabolic health benefits associated with kefir consumption. They also provide support for larger studies examining these to assess whether traditional kefir organisms are necessary to confer health benefits to individuals at risk of developing cardiovascular disease.
Collapse
Affiliation(s)
- Benjamin C T Bourrie
- Agricultural Food and Nutritional Science, Agriculture/Forestry Center, University of Alberta, Edmonton, AB, Canada
| | - Andrew J Forgie
- Agricultural Food and Nutritional Science, Agriculture/Forestry Center, University of Alberta, Edmonton, AB, Canada
| | - Alexander Makarowski
- Agricultural Food and Nutritional Science, Agriculture/Forestry Center, University of Alberta, Edmonton, AB, Canada
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
- VistaMilk, Fermoy, Co. Cork, Ireland
| | - Caroline Richard
- Agricultural Food and Nutritional Science, Agriculture/Forestry Center, University of Alberta, Edmonton, AB, Canada
| | - Benjamin P Willing
- Agricultural Food and Nutritional Science, Agriculture/Forestry Center, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
47
|
McCabe JJ, Evans NR, Gorey S, Bhakta S, Rudd JHF, Kelly PJ. Imaging Carotid Plaque Inflammation Using Positron Emission Tomography: Emerging Role in Clinical Stroke Care, Research Applications, and Future Directions. Cells 2023; 12:2073. [PMID: 37626883 PMCID: PMC10453446 DOI: 10.3390/cells12162073] [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: 07/11/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
Atherosclerosis is a chronic systemic inflammatory condition of the vasculature and a leading cause of stroke. Luminal stenosis severity is an important factor in determining vascular risk. Conventional imaging modalities, such as angiography or duplex ultrasonography, are used to quantify stenosis severity and inform clinical care but provide limited information on plaque biology. Inflammatory processes are central to atherosclerotic plaque progression and destabilization. 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) is a validated technique for quantifying plaque inflammation. In this review, we discuss the evolution of FDG-PET as an imaging modality to quantify plaque vulnerability, challenges in standardization of image acquisition and analysis, its potential application to routine clinical care after stroke, and the possible role it will play in future drug discovery.
Collapse
Affiliation(s)
- John J. McCabe
- Health Research Board Stroke Clinical Trials Network Ireland, Catherine McAuley Centre, Nelson Street, D07 KX5K Dublin, Ireland; (S.G.); (P.J.K.)
- Neurovascular Unit for Applied Translational and Therapeutics Research, Catherine McAuley Centre, Nelson Street, D07 KX5K Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Stroke Service, Department of Medicine for the Elderly, Mater Misericordiae University Hospital, Eccles Street, D07 R2WY Dublin, Ireland
| | - Nicholas R. Evans
- Department of Clinical Neurosciences, Box 83, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK; (N.R.E.); (S.B.)
| | - Sarah Gorey
- Health Research Board Stroke Clinical Trials Network Ireland, Catherine McAuley Centre, Nelson Street, D07 KX5K Dublin, Ireland; (S.G.); (P.J.K.)
- Neurovascular Unit for Applied Translational and Therapeutics Research, Catherine McAuley Centre, Nelson Street, D07 KX5K Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Stroke Service, Department of Medicine for the Elderly, Mater Misericordiae University Hospital, Eccles Street, D07 R2WY Dublin, Ireland
| | - Shiv Bhakta
- Department of Clinical Neurosciences, Box 83, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK; (N.R.E.); (S.B.)
| | - James H. F. Rudd
- Division of Cardiovascular Medicine, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK;
| | - Peter J. Kelly
- Health Research Board Stroke Clinical Trials Network Ireland, Catherine McAuley Centre, Nelson Street, D07 KX5K Dublin, Ireland; (S.G.); (P.J.K.)
- Neurovascular Unit for Applied Translational and Therapeutics Research, Catherine McAuley Centre, Nelson Street, D07 KX5K Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Stroke Service, Department of Medicine for the Elderly, Mater Misericordiae University Hospital, Eccles Street, D07 R2WY Dublin, Ireland
| |
Collapse
|
48
|
Simeunovic A, Brunborg C, Heier M, Seljeflot I, Dahl-Jørgensen K, Margeirsdottir HD. Sustained low-grade inflammation in young participants with childhood onset type 1 diabetes: The Norwegian atherosclerosis and childhood diabetes (ACD) study. Atherosclerosis 2023; 379:117151. [PMID: 37349194 DOI: 10.1016/j.atherosclerosis.2023.05.020] [Citation(s) in RCA: 2] [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: 11/15/2022] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND AND AIMS Persons with type 1 diabetes (T1D) have increased mortality from cardiovascular disease. Early inflammation is important in the development of atherosclerosis. We aimed to evaluate the extent of inflammation and difference in mean over a five-year period in young persons with T1D compared to healthy controls. METHODS The Norwegian Atherosclerosis and Childhood Diabetes (ACD) study is a prospective population-based cohort study on atherosclerosis development in childhood-onset T1D compared to healthy controls, with follow-ups every fifth year. The original study cohort consisted of 314 children with T1D on intensive insulin treatment and 120 healthy controls of similar age. Circulating levels of VCAM-1, TNA-α, P-selectin, E-selectin, CRP, IL-6, IL-18, MCP-1, MMP-9 and TIMP-1 were measured by ELISAs at baseline and at the five-year follow-up. RESULTS The group with T1D had mean age 13.7 (SD = 2.8) years, disease duration 5.6 (SD = 3.4) years and HbA1c 68 (SD = 13.1) mmol/mol at baseline. Levels of almost all inflammatory markers were significantly increased in the group with T1D compared to controls, and significant mean-difference between the two groups over the five-year period was observed in four markers: IL-18, P-selectin, E-selectin and TIMP-1. CONCLUSIONS The early low-grade inflammation present in young individuals with T1D five years after diagnosis is sustained at ten-year disease duration, with moderate changes for most markers of inflammation over time. The evolving inflammatory profile indicates an accelerated chain of events in the progression of early atheromatosis in T1D.
Collapse
Affiliation(s)
- Aida Simeunovic
- Department of Paediatric and Adolescent Medicine, Akershus University Hospital, Lørenskog, Norway; Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; University of Oslo, Institute of Clinical Medicine, Faculty of Medicine, Oslo, Norway; Oslo Diabetes Research Centre, Oslo, Norway.
| | - Cathrine Brunborg
- Centre for Biostatistics and Epidemiology, Research Support Services, Oslo University Hospital, Oslo, Norway
| | - Martin Heier
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; Oslo Diabetes Research Centre, Oslo, Norway
| | - Ingebjørg Seljeflot
- University of Oslo, Institute of Clinical Medicine, Faculty of Medicine, Oslo, Norway; Center for Clinical Heart Research and Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Knut Dahl-Jørgensen
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; University of Oslo, Institute of Clinical Medicine, Faculty of Medicine, Oslo, Norway; Oslo Diabetes Research Centre, Oslo, Norway
| | - Hanna Dis Margeirsdottir
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; Oslo Diabetes Research Centre, Oslo, Norway
| |
Collapse
|
49
|
Festa J, Hussain A, Hackney A, Desai U, Sahota TS, Singh H, Da Boit M. Elderberry extract improves molecular markers of endothelial dysfunction linked to atherosclerosis. Food Sci Nutr 2023; 11:4047-4059. [PMID: 37457144 PMCID: PMC10345675 DOI: 10.1002/fsn3.3393] [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: 09/29/2022] [Revised: 02/28/2023] [Accepted: 04/12/2023] [Indexed: 07/18/2023] Open
Abstract
Endothelial dysfunction (ED), secondary to diminished nitric oxide (NO) production and oxidative stress, is an early subclinical marker of atherosclerosis. Reduced NO bioavailability enhances the adhesion of monocytes to endothelial cells and promotes atherosclerosis. Elderberry extract (EB) is known to contain high levels of anthocyanins which could exert vascular protective effects. Specifically, we investigated the functional capacity of EB on various markers of ED. Human umbilical vein endothelial cells (HUVEC) were pretreated with EB 50 μg/mL and stimulated with TNF-α 10 ng/mL. Cell viability, apoptosis, oxidative stress; eNOS, Akt, Nrf2, NOX-4, and NF-κB at the protein level were measured. A co-culture model was used to determine whether EB could prevent the adhesion of monocytes (THP-1) to HUVECs. Moreover, the expression of adhesion molecules and pro-inflammatory cytokines were also measured. It was demonstrated that EB prevented TNF-α induced apoptosis and reactive oxygen species production in HUVECs. Additionally, EB upregulated Akt and eNOS activity, and Nrf2 expression in response to TNF-α, whereas it decreased NOX-4 expression and NF-κB activity. EB prevented the adhesion of monocytes to HUVECs, as well as reduced IL-6 and MCP-1 levels, which was associated with inhibition of VCAM-1 expression. Our results demonstrate that EB upregulates key cellular markers of endothelial function and ameliorates markers of ED. EB could be used as a potential nutritional aid for preventing atherosclerosis progression.
Collapse
Affiliation(s)
- Joseph Festa
- Leicester School of Allied Health SciencesDe Montfort UniversityLeicesterUK
| | - Aamir Hussain
- Leicester School of Allied Health SciencesDe Montfort UniversityLeicesterUK
| | - Amon Hackney
- Leicester School of Allied Health SciencesDe Montfort UniversityLeicesterUK
| | - Unmesh Desai
- Leicester School of PharmacyFaculty of Health and Life SciencesDe Montfort UniversityLeicesterUK
| | - Tarsem S. Sahota
- Leicester School of PharmacyFaculty of Health and Life SciencesDe Montfort UniversityLeicesterUK
| | - Harprit Singh
- Leicester School of Allied Health SciencesDe Montfort UniversityLeicesterUK
| | - Mariasole Da Boit
- Leicester School of Allied Health SciencesDe Montfort UniversityLeicesterUK
| |
Collapse
|
50
|
Dri E, Lampas E, Lazaros G, Lazarou E, Theofilis P, Tsioufis C, Tousoulis D. Inflammatory Mediators of Endothelial Dysfunction. Life (Basel) 2023; 13:1420. [PMID: 37374202 PMCID: PMC10305352 DOI: 10.3390/life13061420] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Endothelial dysfunction (ED) is characterized by imbalanced vasodilation and vasoconstriction, elevated reactive oxygen species (ROS), and inflammatory factors, as well as deficiency of nitric oxide (NO) bioavailability. It has been reported that the maintenance of endothelial cell integrity serves a significant role in human health and disease due to the involvement of the endothelium in several processes, such as regulation of vascular tone, regulation of hemostasis and thrombosis, cell adhesion, smooth muscle cell proliferation, and vascular inflammation. Inflammatory modulators/biomarkers, such as IL-1α, IL-1β, IL-6, IL-12, IL-15, IL-18, and tumor necrosis factor α, or alternative anti-inflammatory cytokine IL-10, and adhesion molecules (ICAM-1, VCAM-1), involved in atherosclerosis progression have been shown to predict cardiovascular diseases. Furthermore, several signaling pathways, such as NLRP3 inflammasome, that are associated with the inflammatory response and the disrupted H2S bioavailability are postulated to be new indicators for endothelial cell inflammation and its associated endothelial dysfunction. In this review, we summarize the knowledge of a plethora of reviews, research articles, and clinical trials concerning the key inflammatory modulators and signaling pathways in atherosclerosis due to endothelial dysfunction.
Collapse
Affiliation(s)
- Eirini Dri
- 1st Department of Cardiology, Hippokration General Hospital, Kapodistrian University of Athens Medical School, Vas. Sofias 114, 11528 Athens, Greece
| | - Evangelos Lampas
- Department of Cardiology, Konstantopouleio General Hospital, 14233 Athens, Greece
| | - George Lazaros
- 1st Department of Cardiology, Hippokration General Hospital, Kapodistrian University of Athens Medical School, Vas. Sofias 114, 11528 Athens, Greece
| | - Emilia Lazarou
- 1st Department of Cardiology, Hippokration General Hospital, Kapodistrian University of Athens Medical School, Vas. Sofias 114, 11528 Athens, Greece
| | - Panagiotis Theofilis
- 1st Department of Cardiology, Hippokration General Hospital, Kapodistrian University of Athens Medical School, Vas. Sofias 114, 11528 Athens, Greece
| | - Costas Tsioufis
- 1st Department of Cardiology, Hippokration General Hospital, Kapodistrian University of Athens Medical School, Vas. Sofias 114, 11528 Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, Hippokration General Hospital, Kapodistrian University of Athens Medical School, Vas. Sofias 114, 11528 Athens, Greece
| |
Collapse
|