1
|
Shi D, Ning Z, Zhang Y, Guo X, Wei Y, Liu M. Research Trends in Vascular Aging in the Last Decade: A Comprehensive Bibliometric Analysis. Vasc Health Risk Manag 2025; 21:411-423. [PMID: 40417310 PMCID: PMC12103870 DOI: 10.2147/vhrm.s517302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/08/2025] [Indexed: 05/27/2025] Open
Abstract
Background In recent years, vascular aging has emerged as a hot topic in become an important direction of aging research, but a comprehensive bibliometric analysis has not been conducted. Methods The Web of Science database was searched for articles and reviews on vascular aging from January 1, 2014, to August 20, 2024, and the literature was analyzed and knowledge maps were constructed using CiteSpace, VOSviewer, pajek and Scimago Graphica software for econometric analysis and knowledge graph construction of the literature. Results A total of 38,910 authors from 7622 institutions in 111 countries published 7277 papers in 1344 academic journals, with a significant increase in publication volume. The United States is the country with the highest productivity and citation rates, and Mayo Clinic is the most active institution. Tarantini S published the most papers, while Csiszar A received the most citations. Retina-The Journal of Retinal and Vitreous Diseases journal published the most papers, and Circulation journal received the most citations. The main research aspects include age-related macular degeneration, arteriosclerosis, and oxidative stress, which are the main keywords in this field. In the last decade, the term c reactive protein has attracted great attention with its strongest citation explosion. Conclusion In the past decade, the research focus on vascular aging has been increasing year by year. Age-related macular degeneration, arteriosclerosis, oxidative stress and vascular endothelial cells are the emerging research directions in this field.
Collapse
Affiliation(s)
- Dandan Shi
- Xiyuan Hospital, China Academy of Chinese Medicine Science, Beijing, People’s Republic of China
| | - Ziqi Ning
- Xiyuan Hospital, China Academy of Chinese Medicine Science, Beijing, People’s Republic of China
| | - Yaoyao Zhang
- Xiyuan Hospital, China Academy of Chinese Medicine Science, Beijing, People’s Republic of China
| | - Xiaochen Guo
- Xiyuan Hospital, China Academy of Chinese Medicine Science, Beijing, People’s Republic of China
| | - Yun Wei
- Xiyuan Hospital, China Academy of Chinese Medicine Science, Beijing, People’s Republic of China
| | - Meixia Liu
- Xiyuan Hospital, China Academy of Chinese Medicine Science, Beijing, People’s Republic of China
| |
Collapse
|
2
|
Peck M, Connelly P, Lucas-Herald AK. Cardiometabolic outcomes of early onset hypogonadism in males. Best Pract Res Clin Endocrinol Metab 2025:102004. [PMID: 40399185 DOI: 10.1016/j.beem.2025.102004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Testosterone is an important vascular hormone, with multiple effects reported on the vasculature. As such, boys and men with early onset hypogonadism may have altered cardiovascular function, with the potential to result in adverse cardiometabolic outcomes in adulthood. Given the fact that cardiovascular changes in the young can affect future cardiovascular health, there is a need to better understand the influence of androgens on the vasculature in those with conditions such as 46, XY Disorders of Sex Development and Klinefelter Syndrome. This review summarises what is known about hypogonadism and the effects of testosterone supplementation in adults with hypogonadism, as well as what is currently understood in those with early onset hypogonadism specifically. A number of research gaps persist in this area and there is a need for international collaborative studies to address these for future generations of affected individuals.
Collapse
Affiliation(s)
- Mariska Peck
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, 1345 Govan Road, Glasgow G51 4TF, UK.
| | - Paul Connelly
- Department of Endocrinology, Queen Elizabeth University Hospital, Govan Road, 1345 Govan Road, Glasgow G51 4TF, UK.
| | - Angela K Lucas-Herald
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, 1345 Govan Road, Glasgow G51 4TF, UK.
| |
Collapse
|
3
|
Chen FY, Lee CW, Chen YJ, Lin YH, Yeh CF, Lin CC, Cheng HM. Pathophysiology and blood pressure measurements of hypertension in the elderly. J Formos Med Assoc 2025:S0929-6646(25)00145-7. [PMID: 40328594 DOI: 10.1016/j.jfma.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 12/20/2024] [Accepted: 03/26/2025] [Indexed: 05/08/2025] Open
Abstract
Hypertension is a serious concern among the elderly, posing health risks and treatment challenges. In the first part of this review, we explore the pathophysiological mechanisms linking hypertension and aging, focusing on physiological changes such as arterial stiffening, in autonomic nervous system dysfunction, disruption of the renin-angiotensin-aldosterone system, aging-induced alterations in renal function, and the impact of salt-sensitive hypertension. The mnemonic "King/A+++/Hermès" aids in recalling the pathophysiology of aging-related hypertension. Accurate blood pressure measurements are important to determine the prognosis and treatment in the elderly; however, obtaining reliable measurement data in this population is not always easy, as changes in cardiovascular structure and hemodynamics with aging may lead to increased blood pressure variability. In the second part of this review, we evaluate the effectiveness of blood pressure monitoring in older adults, emphasizing the importance of precise measurements. We also assess the efficacy of home and ambulatory monitoring, discuss orthostatic hypotension and orthostatic hypertension, outline challenges in measuring blood pressure in individuals with atrial fibrillation, and explore the impact of aging on measurement errors. We conclude that understanding the interplay between hypertension and aging, coupled with precise blood pressure monitoring strategies tailored for older adults, is essential for effective management and to improve overall cardiovascular health in the elderly population.
Collapse
Affiliation(s)
- Fan-Yu Chen
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Wei Lee
- Institute of Public Health and Community Medicine Research Center, National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan; Cardiovascular Division, Department of Internal Medicine, MacKay Memorial Hospital, MacKay Medical College, New Taipei City, Taiwan; MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Yu-Jen Chen
- Department of Cardiology, Taipei Municipal Wanfang Hospital, Taipei, Taiwan
| | - Yen-Hung Lin
- Primary Aldosteronism Center at National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Fan Yeh
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Ching Lin
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Hao-Min Cheng
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan; Center for Evidence-Based Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; PhD Program of Interdisciplinary Medicine (PIM), National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
4
|
Mancusi C, Basile C, Fucile I, Palombo C, Lembo M, Buso G, Agabiti-Rosei C, Visco V, Gigante A, Tocci G, Maloberti A, Tognola C, Pucci G, Curcio R, Cicco S, Piani F, Marozzi MS, Milan A, Leone D, Cogliati C, Schiavon R, Salvetti M, Ciccarelli M, De Luca N, Volpe M, Muiesan ML. Aortic Remodeling in Patients with Arterial Hypertension: Pathophysiological Mechanisms, Therapeutic Interventions and Preventive Strategies-A Position Paper from the Heart and Hypertension Working Group of the Italian Society of Hypertension. High Blood Press Cardiovasc Prev 2025; 32:255-273. [PMID: 40082374 PMCID: PMC12098454 DOI: 10.1007/s40292-025-00710-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/18/2025] [Indexed: 03/16/2025] Open
Abstract
In patient with arterial hypertension the whole aorta is exposed to increased wall stress due to pressure overload. Different blood pressure (BP) components have been reported as main determinant of aortic remodelling. In particular increased diastolic BP has been associated with aortic dilatation across all its segments with smaller increase in aortic root and ascending aorta related to increased systolic BP and pulse pressure. Optimal BP control is crucial to prevent development of aortic aneurysm and acute aortic disease. Many studies have evaluated the role of different antihypertensive drug classes for prevention of adverse aortic remodelling including beneficial effects of ACEIs, ARBs, dihydropyridinic calcium channel blockers and Beta-blockers. The present review discusses pathophysiological mechanisms, therapeutic interventions and preventive strategies for development of aortic remodeling in patients with arterial hypertension.
Collapse
Affiliation(s)
- Costantino Mancusi
- Department of Advanced Biomedical Science, Hypertension Research Center, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy.
| | - Christian Basile
- Department of Advanced Biomedical Science, Hypertension Research Center, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Ilaria Fucile
- Department of Advanced Biomedical Science, Hypertension Research Center, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Carlo Palombo
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Maria Lembo
- Department of Advanced Biomedical Science, Hypertension Research Center, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Giacomo Buso
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Division of Internal Medicine, ASST Spedali Civili Brescia, Brescia, Italy
- Centro per lo Studio dell'Ipertensione Arteriosa e Fattori di Rischio Cardiovascolari, Brescia, Italy
| | - Claudia Agabiti-Rosei
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Division of Internal Medicine, ASST Spedali Civili Brescia, Brescia, Italy
- Centro per lo Studio dell'Ipertensione Arteriosa e Fattori di Rischio Cardiovascolari, Brescia, Italy
| | - Valeria Visco
- Cardiovascular Research Unit, Department of Medicine and Surgery, University of Salerno, Via Salvador Allende, 84081, Baronissi, Italy
| | - Antonietta Gigante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuliano Tocci
- Hypertension Unit, Division of Cardiology, Department of Clinical and Molecular Medicine, University of Rome Sapienza, Sant'Andrea Hospital, Rome, Italy
| | - Alessandro Maloberti
- School of Medicine and surgery, University of Milano-Bicocca, Milan, Italy
- Cardiology 4, "A.De Gasperis" Cardio Center, ASST GOM Niguarda Ca' Granda, Milan, Italy
| | - Chiara Tognola
- School of Medicine and surgery, University of Milano-Bicocca, Milan, Italy
- Cardiology 4, "A.De Gasperis" Cardio Center, ASST GOM Niguarda Ca' Granda, Milan, Italy
| | - Giacomo Pucci
- Unit of Internal and Traslational Medicine, Terni University Hospital, Terni, Italy
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rosa Curcio
- Unit of Internal Medicine, Terni University Hospital, Terni, Italy
| | - Sebastiano Cicco
- Unit of Internal Medicine "Guido Baccelli" and Unit of Hypertension "Anna Maria Pirrelli", Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), University of Bari Aldo Moro, AUOC Policlinico di Bari, Bari, Italy
| | - Federica Piani
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138, Bologna, Italy
| | - Marialuisa Sveva Marozzi
- Unit of Internal Medicine "Guido Baccelli" and Unit of Hypertension "Anna Maria Pirrelli", Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), University of Bari Aldo Moro, AUOC Policlinico di Bari, Bari, Italy
| | - Alberto Milan
- Division of Internal Medicine, Candiolo Cancer Institute FPO- IRCCS, Candiolo, TO, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Dario Leone
- Division of Internal Medicine, Candiolo Cancer Institute FPO- IRCCS, Candiolo, TO, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Chiara Cogliati
- Department of Biomedical and Clinical Sciences, University of Milan and Internal Medicine, L.Sacco Hospital, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Riccardo Schiavon
- Internal Medicine, L.Sacco Hospital, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Massimo Salvetti
- Department of Clinical and Experimental Sciences, University of Brescia & ASST Spedali Civili di Brescia, Brescia, Italy
| | - Michele Ciccarelli
- Cardiovascular Research Unit, Department of Medicine and Surgery, University of Salerno, Via Salvador Allende, 84081, Baronissi, Italy
| | - Nicola De Luca
- Department of Advanced Biomedical Science, Hypertension Research Center, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | | | - Maria Lorenza Muiesan
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Division of Internal Medicine, ASST Spedali Civili Brescia, Brescia, Italy
- Centro per lo Studio dell'Ipertensione Arteriosa e Fattori di Rischio Cardiovascolari, Brescia, Italy
- Cardiovascular Research Unit, Department of Medicine and Surgery, University of Salerno, Via Salvador Allende, 84081, Baronissi, Italy
| |
Collapse
|
5
|
Sirivarasai J, Shantavasinkul PC, Thitiwiwatkul M, Monsuwan W, Panpunuan P, Sritara P. Association Between Plasma Homocysteine, Folate, Vitamin B12 Levels, and Metabolic Dysfunction Indices in Elderly with Arterial Stiffness. J Clin Med 2025; 14:2998. [PMID: 40364032 PMCID: PMC12072721 DOI: 10.3390/jcm14092998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/07/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Arterial stiffness is a prevalent age-related condition that can significantly increase the risk of cardiovascular disease and mortality in older adults. Understanding the factors that contribute to vascular health, including metabolic dysfunction and hyperhomocysteinemia, alongside vitamin B status, is essential for developing effective interventions. This study aimed to explore the relationship between plasma levels of homocysteine, folate, and vitamin B12, as well as various indices of metabolic dysfunction, in elderly individuals with arterial stiffness. Methods: We conducted a cross-sectional analysis involving 884 participants aged 65 and older, assessing arterial stiffness using the cardio/ankle vascular index method. Additionally, we collected fasting blood samples to evaluate plasma homocysteine, folate, vitamin B12 levels, and other relevant biochemical markers. Results: Higher plasma homocysteine levels are significantly correlated with elevated CAVI scores and increased indices of metabolic dysfunction (p < 0.05). Furthermore, a multivariate logistic regression analysis demonstrated that elevated plasma homocysteine levels, along with higher levels of lipid accumulation product (LAP), triglyceride/glucose index (TyG), and visceral adiposity index (VAI), are associated with increased arterial stiffness. Conclusions: These findings suggest that monitoring and optimizing homocysteine, folate, and vitamin B12 levels may be beneficial for preventing or managing arterial stiffness and related metabolic disorders in the elderly population.
Collapse
Affiliation(s)
- Jintana Sirivarasai
- Nutrition Unit, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | | | - Manasid Thitiwiwatkul
- Faculty of Medicine Ramathibodi Hospital, Mahidol University and HRH Princess Chulabhorn College of Medical Science, Bangkok 10400, Thailand;
| | - Wutarak Monsuwan
- Nutrition Unit, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Pachara Panpunuan
- Division of Cardiology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (P.P.); (P.S.)
| | - Piyamitr Sritara
- Division of Cardiology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (P.P.); (P.S.)
| |
Collapse
|
6
|
Lambert GW, Patel M, Lambert EA. The Influence of the Sympathetic Nervous System on Cardiometabolic Health in Response to Weight Gain or Weight Loss. Metabolites 2025; 15:286. [PMID: 40422864 DOI: 10.3390/metabo15050286] [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: 03/19/2025] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/28/2025] Open
Abstract
Alterations in sympathetic nervous activity are evident in response to changes in body weight. Sympathetic nervous activity and sympathetic responses to weight change are regionalized, with alterations in end organ function dependent on the changes occurring in the brain regulatory pathways invoked and in the effector organs engaged. The obesity-induced activation of the sympathetic nervous system likely contributes to the initiation and worsening of cardiometabolic risk factors, including elevated blood pressure, cardiac dysfunction, dyslipidaemia, increased fasting blood glucose, insulin resistance, and non-alcoholic steatohepatitis. Unintended weight loss, as occurs in cachexia, is driven, at least in part, by the activation of sympathetic nervous-stimulated thermogenesis. The complexity of sympathetic nervous regulation renders the use of global measures of sympathetic activity problematic and the development of targeted therapies difficult, but these are not without promise or precedent. Knowledge of the central and peripheral pathways involved in sympathetic nervous regulation has opened up opportunities for pharmacological, surgical, and device-based approaches to mitigating the burden of disease development and progression. In this narrative review, we elaborate on sympathetic activity in response to changes in body weight, the brain pathways involved, and the cardiovascular and metabolic risks associated with perturbations in regional sympathetic activity.
Collapse
Affiliation(s)
- Gavin W Lambert
- School of Health Sciences and Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
| | - Mariya Patel
- School of Health Sciences and Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
| | - Elisabeth A Lambert
- School of Health Sciences and Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
- Medical Technology Victoria (MedTechVic) Research Hub, Hawthorn, VIC 3122, Australia
| |
Collapse
|
7
|
Tai GJ, Ma YJ, Feng JL, Li JP, Qiu S, Yu QQ, Liu RH, Wankumbu SC, Wang X, Li XX, Xu M. NLRP3 inflammasome-mediated premature immunosenescence drives diabetic vascular aging dependent on the induction of perivascular adipose tissue dysfunction. Cardiovasc Res 2025; 121:77-96. [PMID: 38643484 DOI: 10.1093/cvr/cvae079] [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: 05/16/2023] [Revised: 11/29/2023] [Accepted: 02/06/2024] [Indexed: 04/23/2024] Open
Abstract
AIMS The vascular aging process accelerated by type 2 diabetes mellitus (T2DM) is responsible for the elevated risk of associated cardiovascular diseases. Metabolic disorder-induced immune senescence has been implicated in multi-organ/tissue damage. Herein, we sought to determine the role of immunosenescence in diabetic vascular aging and to investigate the underlying mechanisms. METHODS AND RESULTS Aging hallmarks of the immune system appear prior to the vasculature in streptozotocin (STZ)/high-fat diet (HFD)-induced T2DM mice or db/db mice. Transplantation of aged splenocytes or diabetic splenocytes into young mice triggered vascular senescence and injury compared with normal control splenocyte transfer. RNA sequencing profile and validation in immune tissues revealed that the toll-like receptor 4-nuclear factor-kappa B-NLRP3 axis might be the mediator of diabetic premature immunosenescence. The absence of Nlrp3 attenuated immune senescence and vascular aging during T2DM. Importantly, senescent immune cells, particularly T cells, provoked perivascular adipose tissue (PVAT) dysfunction and alternations in its secretome, which in turn impair vascular biology. In addition, senescent immune cells may uniquely affect vasoconstriction via influencing PVAT. Lastly, rapamycin alleviated diabetic immune senescence and vascular aging, which may be partly due to NLRP3 signalling inhibition. CONCLUSION These results indicated that NLRP3 inflammasome-mediated immunosenescence precedes and drives diabetic vascular aging. The contribution of senescent immune cells to vascular aging is a combined effect of their direct effects and induction of PVAT dysfunction, the latter of which can uniquely affect vasoconstriction. We further demonstrated that infiltration of senescent T cells in PVAT was increased and associated with PVAT secretome alterations. Our findings suggest that blocking the NLRP3 pathway may prevent early immunosenescence and thus mitigate diabetic vascular aging and damage, and targeting senescent T cells or PVAT might also be the potential therapeutic approach.
Collapse
MESH Headings
- Animals
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
- Inflammasomes/metabolism
- Inflammasomes/genetics
- Inflammasomes/immunology
- Signal Transduction
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/pathology
- Male
- Adipose Tissue/metabolism
- Adipose Tissue/immunology
- Adipose Tissue/physiopathology
- Adipose Tissue/pathology
- Mice, Inbred C57BL
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes Mellitus, Type 2/pathology
- Immunosenescence
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/immunology
- Diabetic Angiopathies/physiopathology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/prevention & control
- Cellular Senescence
- Mice, Knockout
- Vasoconstriction
- T-Lymphocytes/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- T-Lymphocytes/pathology
- NF-kappa B/metabolism
- Mice
- Spleen/metabolism
- Spleen/transplantation
- Toll-Like Receptor 4
Collapse
Affiliation(s)
- Guang-Jie Tai
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Yan-Jie Ma
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Jun-Lin Feng
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Jia-Peng Li
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Shu Qiu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Qing-Qing Yu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Ren-Hua Liu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Silumbwe Ceaser Wankumbu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| | - Xin Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Xiao-Xue Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87 Dingjiaqiao, Nanjing 210009, China
| | - Ming Xu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tong jia Lane, Nanjing 210009, China
| |
Collapse
|
8
|
Teixido-Tura G, Dux-Santoy L, Badia C, Limeres J, Guala A, Evangelista Masip A, Ferreira-González I, Rodríguez-Palomares J. Present and future of aortic risk assessment in patients with heritable thoracic aortic diseases. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2025; 78:358-367. [PMID: 39536939 DOI: 10.1016/j.rec.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Heritable thoracic aortic diseases (HTAD) are a group of diverse genetic conditions characterized by an increased risk of aortic complications. The standard surveillance of these patients involves monitoring aortic diameters until a defined threshold is reached, at which point preventive aortic surgery is recommended. However, assessing aortic risk in these patients is far more complex and, in many aspects, remains incompletely understood. Several factors contribute to this complexity, including the diversity and low prevalence of the conditions within HTAD and the limited understanding of the factors influencing the progression of aortic dilation and the advent of acute aortic events. This article reviews current knowledge on clinical, genetic, and imaging factors related to aortic risk in HTAD and explores their potential future roles in improving risk assessment. By advancing our understanding of these factors, we aim to enhance the precision of risk stratification and develop more effective, personalized management strategies for HTAD patients, with the final goal of improving clinical outcomes and quality of life in individuals affected by these genetic disorders.
Collapse
Affiliation(s)
- Gisela Teixido-Tura
- Departamento de Cardiología, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain.
| | | | - Clara Badia
- Departamento de Cardiología, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Javier Limeres
- Departamento de Cardiología, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Andrea Guala
- Vall d'Hebron Institut de Recerca, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | | | - Ignacio Ferreira-González
- Departamento de Cardiología, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain; Departamento de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - José Rodríguez-Palomares
- Departamento de Cardiología, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Vall d'Hebron Institut de Recerca, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Departamento de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| |
Collapse
|
9
|
Toffoli B, Comar C, Grillo A, Barbato V, Vincis E, Baldi V, Berti S, Volpato T, Zorat F, Crocè SL, Emmi G, Fabris B, Puato M, Bernardi S. PNPLA3 Polymorphism Is Inversely Correlated with Aortic Stiffness in Patients with Metabolic Dysfunction-Associated Steatotic Liver Disease Without Fibrosis. Int J Mol Sci 2025; 26:3256. [PMID: 40244110 PMCID: PMC11989603 DOI: 10.3390/ijms26073256] [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: 01/16/2025] [Revised: 03/25/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) corresponds to the condition of increased hepatic fat levels, which is the leading cause of hepatic failure and carcinoma. It is also an independent risk factor for cardiovascular disease (CVD) and mortality. MASLD can be due to obesity with insulin resistance and/or genetic predisposition, i.e., polymorphism in the patatin-like phospholipase domain-containing 3 (PNPLA3) gene. PNPLA3 polymorphism has been associated with increased hepatic fat levels, fibrosis, cirrhosis, and hepatocellular carcinoma, while its association with CVD remains to be fully understood. The aim of the current study was to examine whether the vascular phenotype of patients with MASLD differed between carriers and noncarriers of the PNPLA3 polymorphism. Adult patients with MASLD underwent clinical assessment, PNPLA3 genotyping, arterial tonometry for aortic stiffness measurement, and ultrasound examination of carotid arteries. In total, 117 patients with MASLD and no fibrosis (median hepatic stiffness was 4.71 kPa) were recruited. Carriers of the PNPLA3 polymorphism were younger and exhibited higher levels of ALT and APRI, as compared to wild-type subjects. On the other hand, carriers of the PNPLA3 polymorphism had not only a better metabolic profile (i.e., lower glucose and glycated hemoglobin) but also lower blood pressure, carotid intima-media thickness (IMT), and cardiovascular risk. In addition, PNPLA3 polymorphism was negatively correlated with aortic stiffness, which is a marker of arteriolosclerosis and vascular ageing. Our data are consistent with previous observations that in case of genetically-driven MASLD, there is an inverse association with common predictors of CVD. Our data support the view that the main contributors to CVD risk in patients with MASLD remain conventional cardiometabolic risk factors (i.e., age, glucose) that are more likely to be found in metabolic syndrome-related MASLD rather than genetically-driven MASLD, at least in the first stages of the disease.
Collapse
Affiliation(s)
- Barbara Toffoli
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital Strada di Fiume, 34100 Trieste, Italy; (A.G.); (V.B.); (E.V.); (V.B.); (S.B.); (S.L.C.); (G.E.); (B.F.); (M.P.); (S.B.)
| | - Consuelo Comar
- UCO Medicina Clinica ASUGI, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy; (C.C.); (F.Z.)
| | - Andrea Grillo
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital Strada di Fiume, 34100 Trieste, Italy; (A.G.); (V.B.); (E.V.); (V.B.); (S.B.); (S.L.C.); (G.E.); (B.F.); (M.P.); (S.B.)
- UCO Medicina Clinica ASUGI, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy; (C.C.); (F.Z.)
| | - Vincenzo Barbato
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital Strada di Fiume, 34100 Trieste, Italy; (A.G.); (V.B.); (E.V.); (V.B.); (S.B.); (S.L.C.); (G.E.); (B.F.); (M.P.); (S.B.)
| | - Emanuele Vincis
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital Strada di Fiume, 34100 Trieste, Italy; (A.G.); (V.B.); (E.V.); (V.B.); (S.B.); (S.L.C.); (G.E.); (B.F.); (M.P.); (S.B.)
| | - Veronica Baldi
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital Strada di Fiume, 34100 Trieste, Italy; (A.G.); (V.B.); (E.V.); (V.B.); (S.B.); (S.L.C.); (G.E.); (B.F.); (M.P.); (S.B.)
| | - Silvia Berti
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital Strada di Fiume, 34100 Trieste, Italy; (A.G.); (V.B.); (E.V.); (V.B.); (S.B.); (S.L.C.); (G.E.); (B.F.); (M.P.); (S.B.)
| | - Teresa Volpato
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital Strada di Fiume, 34100 Trieste, Italy; (A.G.); (V.B.); (E.V.); (V.B.); (S.B.); (S.L.C.); (G.E.); (B.F.); (M.P.); (S.B.)
| | - Francesca Zorat
- UCO Medicina Clinica ASUGI, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy; (C.C.); (F.Z.)
| | - Saveria Lory Crocè
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital Strada di Fiume, 34100 Trieste, Italy; (A.G.); (V.B.); (E.V.); (V.B.); (S.B.); (S.L.C.); (G.E.); (B.F.); (M.P.); (S.B.)
- Centro Clinico Studi Fegato ASUGI, Maggiore Teaching Hospital, Piazza dell’Ospitale, 34100 Trieste, Italy
| | - Giacomo Emmi
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital Strada di Fiume, 34100 Trieste, Italy; (A.G.); (V.B.); (E.V.); (V.B.); (S.B.); (S.L.C.); (G.E.); (B.F.); (M.P.); (S.B.)
- UCO Medicina Clinica ASUGI, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy; (C.C.); (F.Z.)
| | - Bruno Fabris
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital Strada di Fiume, 34100 Trieste, Italy; (A.G.); (V.B.); (E.V.); (V.B.); (S.B.); (S.L.C.); (G.E.); (B.F.); (M.P.); (S.B.)
| | - Massimo Puato
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital Strada di Fiume, 34100 Trieste, Italy; (A.G.); (V.B.); (E.V.); (V.B.); (S.B.); (S.L.C.); (G.E.); (B.F.); (M.P.); (S.B.)
- SSD Angiologia ASUGI, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
| | - Stella Bernardi
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital Strada di Fiume, 34100 Trieste, Italy; (A.G.); (V.B.); (E.V.); (V.B.); (S.B.); (S.L.C.); (G.E.); (B.F.); (M.P.); (S.B.)
- SS Endocrinologia ASUGI, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
| |
Collapse
|
10
|
Alanis GA, Boutouyrie P, Abouqateb M, Bruno RM, Andrieu M, Vedie B, Geromin D, Danchin N, Laurent S, Jouven X, Empana JP. Accelerated Vascular Aging as a Possible Mechanism of Troponin I Release in the Absence of Clinically Manifested Myocardial Injury. J Am Heart Assoc 2025; 14:e037718. [PMID: 40145294 DOI: 10.1161/jaha.124.037718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 02/28/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND We examined the association between clusters of vascular aging manifestations and ultrasensitivity cardiac troponin I in individuals without cardiovascular disease. METHODS AND RESULTS A cross-sectional analysis was conducted using baseline data from PPS-3 (Paris Prospective Study III), a French cohort of 10 157 participants. Cardiac troponin I was measured with an ultrasensitive immunoassay with a limit of detection of 0.013 pg/mL. Vascular aging manifestations were assessed via echotracking of the right common carotid artery to measure structural and functional parameters. Hierarchical clustering was used to identify clusters of vascular aging. Multinomial regression assessed the association between vascular aging clusters and cardiac troponin I quintiles. The study included 8722 cardiovascular disease-free participants (mean±SD age, 59.5±6.3 years; 39% women). Three vascular aging clusters were identified. Cluster 1 (n=4158; 47.4%) was characterized as healthy vascular aging with the lowest arteriosclerosis and atherosclerosis indices; cluster 2 (n=2237; 25.5%) was characterized by the highest arteriosclerosis indices, including increased pulse wave velocity, β index, and Young elastic modulus and lowest distensibility coefficient; and cluster 3 (n=2377; 27.0%) was characterized by the highest atherosclerosis indices, including more frequent plaque prevalence and greater intima-media thickness. Compared with healthy vascular aging, arteriosclerosis and atherosclerosis clusters showed a graded positive association with cardiac troponin I quintiles, independent of traditional risk factors. The adjusted odds ratio for belonging to the highest quintile (quintile 5 versus quintile 1) was 1.55 (95% CI, 1.31-1.92) for arteriosclerosis and 2.66 (95% CI, 2.18-3.23) for atherosclerosis clusters. CONCLUSIONS Vascular aging manifestations of arteriosclerosis and atherosclerosis may partly explain the release of troponin I into the bloodstream in adults without clinical cardiovascular disease. REGISTRATION URL: https://clinicaltrials.gov; Unique identifier: NCT00741728.
Collapse
Affiliation(s)
- G A Alanis
- Integrative Epidemiology of Cardiovascular Disease Université Paris Cité, INSERM U970 Paris France
| | - P Boutouyrie
- Paris Cardiovascular Research Centre, Team Arterial Diseases in Women Université Paris Cité, INSERM U970 Paris France
| | - M Abouqateb
- Integrative Epidemiology of Cardiovascular Disease Université Paris Cité, INSERM U970 Paris France
| | - R M Bruno
- Paris Cardiovascular Research Centre, Team Arterial Diseases in Women Université Paris Cité, INSERM U970 Paris France
| | - M Andrieu
- Platform CYBIO Université Paris Cité, INSERM U1016, Cochin Institute Paris France
| | - B Vedie
- AP-HP, Department of Biochemistry, Tissue and Blood Samples Biobank Georges Pompidou European Hospital Paris France
| | - D Geromin
- AP-HP, Department of Biochemistry, Tissue and Blood Samples Biobank Georges Pompidou European Hospital Paris France
| | - N Danchin
- Preventive and Clinical Investigation Center (IPC) Paris France
| | - S Laurent
- Paris Cardiovascular Research Centre, Team Arterial Diseases in Women Université Paris Cité, INSERM U970 Paris France
| | - X Jouven
- Integrative Epidemiology of Cardiovascular Disease Université Paris Cité, INSERM U970 Paris France
| | - J P Empana
- Integrative Epidemiology of Cardiovascular Disease Université Paris Cité, INSERM U970 Paris France
| |
Collapse
|
11
|
Xu T, Zhang Y, Zhou Y, Yin L, Min X, Wu S, Zhang C, Ruan L. Association of Vascular Aging Phenotypes with Adverse Clinical Outcomes in the Chinese Population: A Multicentre Study. Clin Interv Aging 2025; 20:403-414. [PMID: 40177341 PMCID: PMC11963814 DOI: 10.2147/cia.s485597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/08/2025] [Indexed: 04/05/2025] Open
Abstract
Purpose This study aimed to investigate the clinical implications of vascular aging (VAg) phenotypes based on the difference between chronological age (CA) and vascular age (VA). Patients and Methods We defined VA as the predicted age in a multivariable linear regression model including structural and functional parameters of arteries and conventional risk factors, in a multicentric, cross-sectional cohort (n=15580). According to the 10th and 90th percentiles of Δ-age (CA minus VA), we then classified the status of VAg into 3 phenotypes: the early VAg (EVA), the Normal VAg and the supernormal VAg (SUPERNOVA). We used Cox survival analysis to investigate the association between VAg phenotypes and the risk for adverse clinical outcomes (including all-cause death and cardiovascular disease) in an independent, prospective cohort (n=5316). Results In the prospective cohort (11.07 years, 927 events), when compared to the Normal VAg phenotype, EVA had an increased risk (HR: 2.43; 95% CI: 1.80-3.27) and SUPERNOVA had a decrease risk (HR: 0.75; 95% CI: 0.64-0.90) of adverse clinical outcomes, in particular stroke events. EVA also showed a higher risk of myocardial infarction (HR: 3.21, 95% CI: 1.56-6.62) and all-cause death (HR: 1.79, 95% CI: 1.12-2.85). The associations were independent of the atherosclerotic cardiovascular disease risk score. Further, the C-statistics increased 0.010 (P < 0.001), 0.013 (P < 0.001) and 0.016 (P < 0.001) separately when adding baPWV, adding the combination of baPWV and CIMT, and adding the VAg phenotypes to a model of conventional risk factors in predicting cardiovascular events. Conclusion This is the first study to evaluate the clinical implications of VAg phenotypes using multicentric data and undergone external validation in China. Our results emphasized that the classification of VAg phenotypes may be a potential tool to identify individuals who were susceptible to or resilient to VAg.
Collapse
Affiliation(s)
- Ting Xu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Yucong Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Yi Zhou
- Department of Cardiovascular Ultrasound and Non-Invasive Cardiology, Sichuan Provincial People’s Hospital, Sichuan Academy of Medical Sciences, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Lixue Yin
- Department of Cardiovascular Ultrasound and Non-Invasive Cardiology, Sichuan Provincial People’s Hospital, Sichuan Academy of Medical Sciences, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Xinwen Min
- Department of Cardiology, Sinopharm Dongfeng Central Hospital, Hubei University of Medicine, Shiyan, 442008, People’s Republic of China
| | - Shouling Wu
- Department of Cardiology, Kailuan Hospital, Hebei United University, Tangshan, 063000, People’s Republic of China
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Lei Ruan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
12
|
Loboda D, Golba KS, Gurowiec P, Bredelytė A, Razbadauskas A, Sarecka-Hujar B. Variability in Arterial Stiffness and Vascular Endothelial Function After COVID-19 During 1.5 Years of Follow-Up-Systematic Review and Meta-Analysis. Life (Basel) 2025; 15:520. [PMID: 40283075 PMCID: PMC12028431 DOI: 10.3390/life15040520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/29/2025] Open
Abstract
Increasing long-term observations suggest that coronavirus disease 2019 (COVID-19) vasculopathy may persist even 1.5 years after the acute phase, potentially accelerating the development of atherosclerotic cardiovascular diseases. This study systematically reviewed the variability of brachial flow-mediated dilation (FMD) and carotid-femoral pulse wave velocity (cfPWV) from the acute phase of COVID-19 through 16 months of follow-up (F/U). Databases including PubMed, Web of Science, MEDLINE, and Embase were screened for a meta-analysis without language or date restrictions (PROSPERO reference CRD42025642888, last search conducted on 1 February 2025). The quality of the included studies was assessed using the Newcastle-Ottawa Quality Scale. We considered all studies (interventional pre-post studies, prospective observational studies, prospective randomized, and non-randomized trials) that assessed FMD or cfPWV in adults (aged ≥ 18 years) with or after laboratory-confirmed COVID-19 compared with non-COVID-19 controls or that assessed changes in these parameters during the F/U. Twenty-one studies reported differences in FMD, and 18 studies examined cfPWV between COVID-19 patients and control groups during various stages: acute/subacute COVID-19 (≤30 days from disease onset), early (>30-90 days), mid-term (>90-180 days), late (>180-270 days), and very late (>270 days) post-COVID-19 recovery. Six studies assessed variability in FMD, while nine did so for cfPWV during the F/U. Data from 14 FMD studies (627 cases and 694 controls) and 15 cfPWV studies (578 cases and 703 controls) were included in our meta-analysis. FMD showed a significant decrease compared to controls during the acute/subacute phase (standardized mean difference [SMD]= -2.02, p < 0.001), with partial improvements noted from the acute/subacute phase to early recovery (SMD = 0.95, p < 0.001) and from early to mid-term recovery (SMD = 0.92, p = 0.006). Normalization compared to controls was observed in late recovery (SMD = 0.12, p = 0.69). In contrast, cfPWV values, which were higher than controls in the acute/subacute phase (SMD = 1.27, p < 0.001), remained elevated throughout the F/U, with no significant changes except for a decrease from mid-term to very late recovery (SMD= -0.39, p < 0.001). In the very late recovery, cfPWV values remained higher than those of controls (SMD = 0.45, p = 0.010). In the manuscript, we discuss how various factors, including the severity of acute COVID-19, the persistence of long-term COVID-19 syndrome, and the patient's initial vascular age, depending on metrics age and cardiovascular risk factors, influenced the time and degree of FMD and cfPWV improvement.
Collapse
Affiliation(s)
- Danuta Loboda
- Department of Electrocardiology and Heart Failure, Medical University of Silesia in Katowice, 40-635 Katowice, Poland; (K.S.G.); (P.G.)
| | - Krzysztof S. Golba
- Department of Electrocardiology and Heart Failure, Medical University of Silesia in Katowice, 40-635 Katowice, Poland; (K.S.G.); (P.G.)
| | - Piotr Gurowiec
- Department of Electrocardiology and Heart Failure, Medical University of Silesia in Katowice, 40-635 Katowice, Poland; (K.S.G.); (P.G.)
| | - Aelita Bredelytė
- Faculty of Health Sciences, Klaipėda University, LT-92294 Klaipeda, Lithuania; (A.B.); (A.R.)
| | - Artūras Razbadauskas
- Faculty of Health Sciences, Klaipėda University, LT-92294 Klaipeda, Lithuania; (A.B.); (A.R.)
- Chemotherapy Unit, Department of Oncology, Klaipeda University Hospital, LT-92288 Klaipeda, Lithuania
| | - Beata Sarecka-Hujar
- Department of Basic Biomedical Science, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland;
| |
Collapse
|
13
|
Pucci G, Alcidi R, Grassi G. Pulse Wave Velocity Assessment: A Useful Tool to Screen Asymptomatic Individuals at High Cardiovascular Risk? J Clin Hypertens (Greenwich) 2025; 27:e70040. [PMID: 40127413 PMCID: PMC11932551 DOI: 10.1111/jch.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/26/2025]
Affiliation(s)
- Giacomo Pucci
- Unit of Internal and Translational MedicineSanta Maria Terni HospitalTerniItaly
| | - Riccardo Alcidi
- Unit of Internal and Translational MedicineSanta Maria Terni HospitalTerniItaly
| | - Guido Grassi
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMilanItaly
| |
Collapse
|
14
|
Alanis GA, Boutouyrie P, Abouqateb M, Bruno RM, Climie RE, van Sloten T, Danchin N, Pannier B, Laurent S, Jouven X, Empana JP. Vascular ageing manifestations and hypertension in the community. Am J Prev Cardiol 2025; 21:100918. [PMID: 39807445 PMCID: PMC11728902 DOI: 10.1016/j.ajpc.2024.100918] [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: 08/17/2024] [Revised: 10/31/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
Objective To investigate the association between joint manifestations of vascular ageing (VA) and hypertension. Methods We used baseline (2008-2012) and follow-up data (up to 2024) from the Paris Prospective Study III, a French cohort of 10,157 participants. Prevalent and incident hypertension were determined at baseline (blood pressure ≥140/90 mmHg or on medication) and at 2, 4, 6, 8 and 10 years of follow-up (self-reported antihypertensive treatment). VA manifestations were assessed at baseline via echo-tracking in the right common carotid artery. Clustering analysis identified patterns of VA and their association with hypertension was assessed with logistic regression. Results The cross-sectional analysis included 9,096 participants (mean age: 59±6 years, 39 % female). Hypertension prevalence was 36 % (n = 3,276). Three clusters of VA manifestations were identified. Cluster 1 (n = 4,326;47.6 %) was characterized by healthy vascular ageing (HVA), Cluster 2 by increased arteriosclerosis (ART) (n = 2,274;25.0 %) and Cluster 3 by greater atherosclerosis prevalence (ATH) (n = 2,496;27.4 %). Compared to the HVA cluster, ART (aOR 3.94; 95 % CI 3.50;4.45) and ATH clusters (aOR 2.69; 95 % CI 2.38;3.04) were associated with prevalent hypertension. The prospective analysis included 5,310 normotensives with 754 (14.1 %) cases of incident hypertension (median follow-up of 10.05 years [range: 10.00;10.15]). Both ART (aOR 1.34; 95 % CI 1.08;1.65) and ATH (aOR 1.70; 95 % CI 1.40;2.07) clusters were associated with incident hypertension. Conclusion Vascular ageing manifestations reflecting increased carotid arteriosclerosis and atherosclerosis are related to prevalent and incident hypertension.
Collapse
Affiliation(s)
- Guillermo A. Alanis
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Centre, Team Integrative epidemiology of cardiovascular diseases, Paris, France
| | - Pierre Boutouyrie
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Centre, Team Arterial diseases in women, Paris, France
- APHP, Georges Pompidou European Hospital, DMU CARTE, Department of Pharmacology, Paris, France
| | - Mouad Abouqateb
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Centre, Team Integrative epidemiology of cardiovascular diseases, Paris, France
| | - Rosa Maria Bruno
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Centre, Team Arterial diseases in women, Paris, France
- APHP, Georges Pompidou European Hospital, DMU CARTE, Department of Pharmacology, Paris, France
| | - Rachel E. Climie
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Centre, Team Integrative epidemiology of cardiovascular diseases, Paris, France
- Menzies Institute for Medical Research, University of Tasmanian, Hobart, Australia
| | - Thomas van Sloten
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Bruno Pannier
- Investigations Précliniques de Paris (IPC), Paris, France
| | - Stéphane Laurent
- APHP, Georges Pompidou European Hospital, DMU CARTE, Department of Pharmacology, Paris, France
| | - Xavier Jouven
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Centre, Team Integrative epidemiology of cardiovascular diseases, Paris, France
| | - Jean-Philippe Empana
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Centre, Team Integrative epidemiology of cardiovascular diseases, Paris, France
- Menzies Institute for Medical Research, University of Tasmanian, Hobart, Australia
| |
Collapse
|
15
|
Virsolvy A, Benmira AM, Allal S, Demattei C, Sutra T, Cristol JP, Jouy N, Richard S, Perez-Martin A. Benefits of Dietary Supplementation with Specific Silicon-Enriched Spirulina on Arterial Function in Healthy Elderly Individuals: A Randomized, Placebo-Controlled Trial. Nutrients 2025; 17:864. [PMID: 40077730 PMCID: PMC11901655 DOI: 10.3390/nu17050864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Vascular aging is associated with increased arterial stiffness and changes in the wall structure, leading to a loss of elasticity. Silicon is abundant in arteries and plays a key role in the synthesis and stabilization of elastin fibers. In animal models of accelerated cardiovascular aging, a specific nutritional supplement based on silicon-enriched spirulina (SpSi) has been shown to have beneficial effects on vascular function. The present study, designed as a randomized, double-blind, placebo-controlled trial, aimed to evaluate the effectiveness of this SpSi supplement on aging-related changes in vascular function among healthy older adults. Methods: Here, 120 healthy volunteers aged 60-75 years were enrolled and randomly assigned to either the SpSi group (n = 60) or placebo group (n = 60). Over 6 months, the participants received either 3.5 g of specific 1% silicon-enriched spirulina (SpSi group) or placebo tablets daily. The primary outcome was the assessment of arterial wall pressure waveforms, which included blood pressure (BP) readings and the determination of the aortic pulse wave velocity (aPWV). Secondary outcomes included the vasomotor endothelial function through post-ischemic vasorelaxation, measured using the reactive hyperemia index (RHI), and carotid intima-media thickness. Results: When considering the entire sample, none of the studied parameters differed between the placebo and SpSi groups. However, when focusing on individuals with high-normal blood pressure (i.e., systolic BP between 130 and 150 mmHg) and aPWV levels above cutoff values (>10 m/s), the BP decreased by 8% (p < 0.001) and aPWV decreased by 13.5% (p < 0.0001) in subjects receiving SpSi. In individuals with BP and aPWV levels below the cutoff values, no effect was observed. Conclusions: In healthy elderly individuals, SpSi supplementation improved high-normal blood pressure and aortic pulse wave velocity, suggesting an enhanced vascular function.
Collapse
Affiliation(s)
- Anne Virsolvy
- PhyMedExp, Université de Montpellier, INSERM U1046, CNRS UMR 9214, 34295 Montpellier, France; (J.-P.C.); (S.R.)
| | - Amir Mokhfi Benmira
- Department of Vascular Medicine, Nîmes University Hospital, Université de Montpellier, 30900 Nîmes, France; (A.M.B.); (S.A.); (A.P.-M.)
| | - Salim Allal
- Department of Vascular Medicine, Nîmes University Hospital, Université de Montpellier, 30900 Nîmes, France; (A.M.B.); (S.A.); (A.P.-M.)
| | - Christophe Demattei
- BESPIM–Laboratoire de Biostatistique, Epidémiologie Clinique, Santé Publique Innovation et Méthodologie, Nïmes University Hospital, Université de Montpellier, 30900 Nîmes, France;
| | - Thibault Sutra
- Department of Biochemistry, Montpellier University Hospital, 34295 Montpellier, France;
| | - Jean-Paul Cristol
- PhyMedExp, Université de Montpellier, INSERM U1046, CNRS UMR 9214, 34295 Montpellier, France; (J.-P.C.); (S.R.)
- Department of Biochemistry, Montpellier University Hospital, 34295 Montpellier, France;
| | | | - Sylvain Richard
- PhyMedExp, Université de Montpellier, INSERM U1046, CNRS UMR 9214, 34295 Montpellier, France; (J.-P.C.); (S.R.)
| | - Antonia Perez-Martin
- Department of Vascular Medicine, Nîmes University Hospital, Université de Montpellier, 30900 Nîmes, France; (A.M.B.); (S.A.); (A.P.-M.)
- IDESP-Institut Desbrest D’Epidémiologie et de Santé Publique, Université de Montpellier, INSERM U1318, 34090 Montpellier, France
| |
Collapse
|
16
|
Vieira-da-Silva MA, Bauab Filho AB, Imanichi F, Lessa Silva RC, Marchiori Vieira L, Roma Uyemura J, Humsi MJ, Yugar-Toledo JC, Cosenso-Martin LN, Vilela-Martin JF. The correlation between age, blood pressure variability and estimated pulse wave velocity. Sci Rep 2025; 15:6990. [PMID: 40011569 PMCID: PMC11865459 DOI: 10.1038/s41598-025-91023-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
Studies have separately compared the association of age with blood pressure variability (BPV) or pulse wave velocity (PWV). We aimed to establish the association between age and metrics of short-term BPV and PWV in the same sample. 508 under 60 years (< 60) and 141 in their sixties (≥ 60) measured blood pressure (OBP), and PWV using the oscillometric technique (br-PWV). They recorded an ambulatory BP monitoring (ABPM) to obtain variables for systolic (SBPV) and diastolic BPV (DBPV). We also estimated PWV using formulas. We calculated the Pearson correlation coefficient (r) and determination coefficient (R2) of all parameters with age. The correlation between age and PWV was very strong (br-PWV; r = 0.901; p < 0.001). It was poor for SBPV, 24-h weighted SD r = 0.492; p < 0.001, and not significant for DBPV, 24-h weighted SD r = 0.220; p < 0.001. The correlation and determination values were generally better in the group comprising ≥ 60-years, with R2 values robust for PWV (br-PWV = 0.812) weaker for SBPV (24-h weighted SD = 0.243) and deemed irrelevant for DBPV (24-h weighted SD = 0.048). Our study shows that PWV metrics are firmly and significantly more influenced by age than short-term BPV.
Collapse
Affiliation(s)
- Marco A Vieira-da-Silva
- Internal Medicine Department, Federal University of the Triangulo Mineiro, Uberaba, Brazil.
- Cardiac Diagnostic Center (CDC), Dr. Thomaz Ulhoa 544 Square, Uberaba, 38025-050, Brazil.
| | - André B Bauab Filho
- Internal Medicine Department, Federal University of the Triangulo Mineiro, Uberaba, Brazil
| | - Felipe Imanichi
- Medical Sciences School of the Santa Casa of Sao Paulo, Sao Paulo, Brazil
| | | | | | - Jessica Roma Uyemura
- Internal Medicine Department, State Medical School in Sao Jose Do Rio Preto (FAMERP), Sao Jose Do Rio Preto, Brazil
| | - Marcelo J Humsi
- Internal Medicine Department, State Medical School in Sao Jose Do Rio Preto (FAMERP), Sao Jose Do Rio Preto, Brazil
| | - Juan C Yugar-Toledo
- Internal Medicine Department, State Medical School in Sao Jose Do Rio Preto (FAMERP), Sao Jose Do Rio Preto, Brazil
| | - Luciana N Cosenso-Martin
- Internal Medicine Department, State Medical School in Sao Jose Do Rio Preto (FAMERP), Sao Jose Do Rio Preto, Brazil
| | - Jose F Vilela-Martin
- Internal Medicine Department, State Medical School in Sao Jose Do Rio Preto (FAMERP), Sao Jose Do Rio Preto, Brazil
| |
Collapse
|
17
|
Pescari D, Mihuta MS, Bena A, Stoian D. Independent Predictors of Circulating Trimethylamine N-Oxide (TMAO) and Resistin Levels in Subjects with Obesity: Associations with Carotid Intima-Media Thickness and Metabolic Parameters. Nutrients 2025; 17:798. [PMID: 40077669 PMCID: PMC11902032 DOI: 10.3390/nu17050798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Obesity contributes to cardiometabolic risk, including subclinical atherosclerosis and insulin resistance. This study examines the predictive roles of trimethylamine N-oxide (TMAO) and resistin in relation to carotid intima-media thickness and metabolic parameters; Methods: Sixty adults (18-71 years) with varying body weights were assessed for body composition, subclinical atherosclerosis, and blood biomarkers, including TMAO and resistin; Results: TMAO correlated strongly with CIMT (r = 0.674, p < 0.001), indicating its role in subclinical atherosclerosis. Logistic regression identified TMAO (threshold 380; AUC = 0.880, accuracy = 91.7%) as a predictor of cardiometabolic risk. Resistin was associated with CIMT, WHR, and total cholesterol, inversely linked to LDL cholesterol (p = 0.003). Less active participants exhibited higher TMAO (p = 0.001) and resistin (p = 0.02). Family histories of obesity and diabetes correlated with elevated TMAO, while resistin linked to shorter sleep duration and diabetes history, highlighting their importance in obesity-related cardiometabolic risks; Conclusions: TMAO is strongly linked to abdominal fat, insulin resistance, and subclinical atherosclerosis, while resistin is associated with lipid metabolism and aging. Their combined assessment enhances the prediction of obesity-related cardiometabolic risk, supporting their role in risk stratification and targeted interventions.
Collapse
Affiliation(s)
- Denisa Pescari
- Department of Doctoral Studies, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Monica Simina Mihuta
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Andreea Bena
- Discipline of Endocrinology, Second Department of Internal Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Dana Stoian
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| |
Collapse
|
18
|
Kakaletsis N, Kotsis V, Hosomi N, Nezu T, Michel P, Guillaume T, Strambo D, Kim YS, Sung W, Vemmos K, Korompoki E, Acampa M, Putaala J, Tulkki L, Hermann M, Rejmer P, Bath PM, Woodhouse LJ, Protogerou AD, Athanasopoulou E, Milionis H, Ntaios G, Savopoulos C. Early vascular aging ambulatory score in acute ischemic stroke. NPJ AGING 2025; 11:13. [PMID: 39984473 PMCID: PMC11845580 DOI: 10.1038/s41514-025-00202-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 01/25/2025] [Indexed: 02/23/2025]
Abstract
Understanding the impact of early vascular aging (EVA) on acute ischemic stroke (AIS) outcomes may provide new insights for improving prognostic assessments and developing targeted therapeutic strategies. This study aimed to validate the EVA ambulatory score (EVAAs) in AIS patients, assessing its association with stroke type, severity, and prognosis. Among the 2,730 AIS patients with a mean age of 72.0 ± 14.4 years, 83.4% exhibited EVA. EVA was identified as an independent predictor of poor outcome at both discharges (aOR:1.72, 95%CI:1.25-2.36, p < 0.001) and at 90 days (aOR:2.22, 95%CI:1.49-3.31, p < 0.001). In subgroup analyses, EVAAs showed improved predictive value in AIS patients with a lower cardiovascular disease burden and a non-atherogenic lipid profile. The EVAAs, as an indicator of EVA that could be easily integrated into daily clinical practice, are a significant predictor of adverse outcomes in AIS patients.
Collapse
Affiliation(s)
- Nikolaos Kakaletsis
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece.
| | - Vasilios Kotsis
- Third Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Naohisa Hosomi
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Tomohisa Nezu
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Patrik Michel
- Stroke Center and Neurology Service, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Thevoz Guillaume
- Stroke Center and Neurology Service, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Davide Strambo
- Stroke Center and Neurology Service, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Young Seo Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Wonjae Sung
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Konstantinos Vemmos
- Department of Clinical Therapeutics of Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni Korompoki
- Department of Clinical Therapeutics of Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maurizio Acampa
- Stroke Unit, Department of Medical Sciences, Surgery and Neurosciences, "Santa Maria Alle Scotte" General Hospital, University of Siena, Siena, Italy
| | - Jukka Putaala
- Department of Neurology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Lauri Tulkki
- Department of Neurology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Matthias Hermann
- University Heart Center Zurich, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Protazy Rejmer
- University Heart Center Zurich, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Philip M Bath
- Stroke Trials Unit, Mental Health & Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Lisa J Woodhouse
- Stroke Trials Unit, Mental Health & Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Athanase D Protogerou
- Cardiovascular Prevention & Research Unit, Clinic & Laboratory of Pathophysiology, Department of Medicine, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Elpida Athanasopoulou
- Cardiovascular Prevention & Research Unit, Clinic & Laboratory of Pathophysiology, Department of Medicine, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Haralampos Milionis
- Department of Internal Medicine, Medical School, University of Ioannina, University Hospital of Ioannina, Ioannina, Greece
| | - George Ntaios
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Christos Savopoulos
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
19
|
Kasal DA, Sena V, Huguenin GVB, De Lorenzo A, Tibirica E. Microvascular endothelial dysfunction in vascular senescence and disease. Front Cardiovasc Med 2025; 12:1505516. [PMID: 40041173 PMCID: PMC11878104 DOI: 10.3389/fcvm.2025.1505516] [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: 10/03/2024] [Accepted: 01/21/2025] [Indexed: 03/06/2025] Open
Abstract
Cardiovascular disease (CVD) is the main cause of morbidity and mortality in the adult and the elderly, with increasing prevalence worldwide. A growing body of research has focused on the earliest stage of vascular decline-endothelial dysfunction (ED)-which at the microvascular level can anticipate in decades the diagnosis of CVD. This review aims to provide a prospect of the literature regarding the development of ED as an indissociable feature of the aging of the cardiovascular system, highlighting the role of inflammation in the process. Vascular aging consists of a lifelong continuum, which starts with cell respiration and its inherent production of reactive oxygen species. Molecular imbalance is followed by cellular epigenetic changes, which modulate immune cells, such as macrophage and lymphocyte subtypes. These mechanisms are influenced by lifestyle habits, which affect inflammation hotspots in organism, such as visceral fat and gut microbiota. The process can ultimately lead to an environment committed to the loss of the physiological functions of endothelial cells. In addition, we discuss lifestyle changes targeting the connection between age-related inflammation and vascular dysfunction. Addressing microvascular ED represents a critical endeavor in order to prevent or delay vascular aging and associated diseases.
Collapse
Affiliation(s)
- Daniel A. Kasal
- Research and Teaching Department, National Institute of Cardiology, Rio de Janeiro, Brazil
- Internal Medicine Department, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Viviane Sena
- Research and Teaching Department, National Institute of Cardiology, Rio de Janeiro, Brazil
| | - Grazielle Vilas Bôas Huguenin
- Research and Teaching Department, National Institute of Cardiology, Rio de Janeiro, Brazil
- Nutrition and Dietetics Department, Fluminense Federal University, Rio de Janeiro, Brazil
| | - Andrea De Lorenzo
- Research and Teaching Department, National Institute of Cardiology, Rio de Janeiro, Brazil
| | - Eduardo Tibirica
- Research and Teaching Department, National Institute of Cardiology, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Phua TJ. Hallmarks of aging: middle-aging hypovascularity, tissue perfusion and nitric oxide perspective on healthspan. FRONTIERS IN AGING 2025; 5:1526230. [PMID: 39839443 PMCID: PMC11747043 DOI: 10.3389/fragi.2024.1526230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025]
Abstract
Aging is a complex process marked by various changes at both cellular and systemic levels, impacting the functioning and lifespan of organisms. Over time, researchers have pinpointed several significant hallmarks of aging that lead to the gradual deterioration of tissue function, regulation, and homeostasis associated with aging in humans. Despite this, the intricate interactions and cumulative effects of these hallmarks are still mostly uncharted territory. Understanding this complex web is a major challenge in Geroscience, yet it is crucial for developing effective strategies that promote healthy aging, reduce medical costs, and ensure the sustainability of health systems. Gaining insights in this area is essential for creating interventions that can slow the aging process, enhance healthspan, and decrease the likelihood of age-related diseases. The integration of knowledge from various fields concerning the middle-aging nitric oxide (NO)-mediated hypovascularity hypoxia hemodynamic hypothesis points to a systems-based approach to the biological hallmarks of aging. Key evidence suggests a systemic connection between the endocrine system (specifically sex hormones), endogenous NO deficiency, and the vascular system, which serves as a network of microvascular structures crucial for tissue perfusion functions at cellular level. These processes also involve oxidative stress and inflammation triggered by hypoxia.
Collapse
Affiliation(s)
- Teow J. Phua
- Molecular Medicine, NSW Health Pathology, John Hunter Hospital, Newcastle, NSW, Australia
| |
Collapse
|
21
|
Puzantian H, Townsend RR. Three Ankle-Brachial Index Ranges and Incident CKD in Diabetes: A Goldilocks Perspective on the "Just Right" Range. Am J Kidney Dis 2025; 85:11-13. [PMID: 39570274 DOI: 10.1053/j.ajkd.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 11/22/2024]
Affiliation(s)
- Houry Puzantian
- Hariri School of Nursing, American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Raymond R Townsend
- Renal, Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
22
|
Zanelli S, Agnoletti D, Alastruey J, Allen J, Bianchini E, Bikia V, Boutouyrie P, Bruno RM, Climie R, Djeldjli D, Gkaliagkousi E, Giudici A, Gopcevic K, Grillo A, Guala A, Hametner B, Joseph J, Karimpour P, Kodithuwakku V, Kyriacou PA, Lazaridis A, Lønnebakken MT, Martina MR, Mayer CC, Nabeel PM, Navickas P, Nemcsik J, Orter S, Park C, Pereira T, Pucci G, Rey ABA, Salvi P, Seabra ACG, Seeland U, van Sloten T, Spronck B, Stansby G, Steens I, Stieglitz T, Tan I, Veerasingham D, Wassertheurer S, Weber T, Westerhof BE, Charlton PH. Developing technologies to assess vascular ageing: a roadmap from VascAgeNet. Physiol Meas 2024; 45:121001. [PMID: 38838703 PMCID: PMC11697036 DOI: 10.1088/1361-6579/ad548e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 03/15/2024] [Accepted: 06/05/2024] [Indexed: 06/07/2024]
Abstract
Vascular ageing (vascular ageing) is the deterioration of arterial structure and function which occurs naturally with age, and which can be accelerated with disease. Measurements of vascular ageing are emerging as markers of cardiovascular risk, with potential applications in disease diagnosis and prognosis, and for guiding treatments. However, vascular ageing is not yet routinely assessed in clinical practice. A key step towards this is the development of technologies to assess vascular ageing. In this Roadmap, experts discuss several aspects of this process, including: measurement technologies; the development pipeline; clinical applications; and future research directions. The Roadmap summarises the state of the art, outlines the major challenges to overcome, and identifies potential future research directions to address these challenges.
Collapse
Affiliation(s)
- Serena Zanelli
- Laboratoire Analyse, Géométrie et Applications, Université Sorbonne Paris Nord, Paris, France
- Axelife, Paris, France
| | - Davide Agnoletti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna Policlinico Sant’Orsola, Bologna, Italy
- Cardiovascular Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Jordi Alastruey
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King’s College London, London SE1 7EU, United Kingdom
| | - John Allen
- Research Centre for Intelligent Healthcare, Coventry University, Coventry CV1 5RW, United Kingdom
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Elisabetta Bianchini
- Institute of Clinical Physiology, Italian National Research Council (CNR), Pisa, Italy
| | - Vasiliki Bikia
- Stanford University, Stanford, California, United States
- Swiss Federal Institute of Technology of Lausanne, Lausanne, Switzerland
| | - Pierre Boutouyrie
- INSERM U970 Team 7, Paris Cardiovascular Research Centre
- PARCC, University Paris Descartes, AP-HP, Pharmacology Unit, Hôpital Européen Georges Pompidou, 56
Rue Leblanc, Paris 75015, France
| | - Rosa Maria Bruno
- INSERM U970 Team 7, Paris Cardiovascular Research Centre
- PARCC, University Paris Descartes, AP-HP, Pharmacology Unit, Hôpital Européen Georges Pompidou, 56
Rue Leblanc, Paris 75015, France
| | - Rachel Climie
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | | | | | - Alessandro Giudici
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| | | | - Andrea Grillo
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Andrea Guala
- Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain
- CIBER-CV, Instituto de Salud Carlos III, Madrid, Spain
| | - Bernhard Hametner
- Center for Health & Bioresources, Medical Signal Analysis, AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | - Jayaraj Joseph
- Department of Electrical Engineering, Indian Institute of Technology Madras, Chennai 600 036, India
| | - Parmis Karimpour
- Research Centre for Biomedical Engineering, City, University of London, London EC1V 0HB, United Kingdom
| | | | - Panicos A Kyriacou
- Research Centre for Biomedical Engineering, City, University of London, London EC1V 0HB, United Kingdom
| | - Antonios Lazaridis
- Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Mai Tone Lønnebakken
- Department of Heart Disease, Haukeland University Hospital and Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Christopher Clemens Mayer
- Center for Health & Bioresources, Medical Signal Analysis, AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | - P M Nabeel
- Healthcare Technology Innovation Centre, IIT Madras, Chennai 600 113, India
| | - Petras Navickas
- Clinic of Cardiac and Vascular Diseases, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - János Nemcsik
- Department of Family Medicine, Semmelweis University, Budapest, Hungary
| | - Stefan Orter
- Center for Health & Bioresources, Medical Signal Analysis, AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | - Chloe Park
- MRC Unit for Lifelong Health and Ageing at UCL, 1–19 Torrington Place, London WC1E 7HB, UK
| | - Telmo Pereira
- Polytechnic University of Coimbra, Coimbra Health School, Rua 5 de Outubro—S. Martinho do Bispo, Apartado 7006, 3046-854 Coimbra, Portugal
| | - Giacomo Pucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Unit of Internal Medicine, ‘Santa Maria’ Terni Hospital, Terni, Italy
| | - Ana Belen Amado Rey
- Laboratory for Biomedical Microtechnology, Department of Microsystems Engineering—IMTEK, IMBIT—NeuroProbes, BrainLinks-BrainTools Center, University of Freiburg, Freiburg, Germany
| | - Paolo Salvi
- Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Ana Carolina Gonçalves Seabra
- Laboratory for Biomedical Microtechnology, Department of Microsystems Engineering—IMTEK, IMBIT—NeuroProbes, BrainLinks-BrainTools Center, University of Freiburg, Freiburg, Germany
| | - Ute Seeland
- Institute of Social Medicine, Epidemiology and Health Economics, Charitè—Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thomas van Sloten
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bart Spronck
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University,
Sydney, Australia
| | - Gerard Stansby
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
- Northern Vascular Centre, Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom
| | - Indra Steens
- Department of Internal Medicine, Maastricht University, Maastricht, The Netherlands
| | - Thomas Stieglitz
- Laboratory for Biomedical Microtechnology, Department of Microsystems Engineering—IMTEK, IMBIT—NeuroProbes, BrainLinks-BrainTools Center, University of Freiburg, Freiburg, Germany
- Bernstein Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Isabella Tan
- Macquarie University, Sydney, Australia
- The George Institute for Global Health, Sydney, Australia
| | | | - Siegfried Wassertheurer
- Center for Health & Bioresources, Medical Signal Analysis, AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | - Thomas Weber
- Cardiology Department, Klinikum Wels-Grieskirchen, Wels, Austria
| | - Berend E Westerhof
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Neonatology, Radboud University Medical Center, Radboud Institute for Health Sciences, Amalia Children’s Hospital, Nijmegen, The Netherlands
| | - Peter H Charlton
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, United Kingdom
- Research Centre for Biomedical Engineering, City, University of London, London EC1V 0HB, United Kingdom
| |
Collapse
|
23
|
Jae SY, Lee KH, Kim HJ, Kunutsor SK, Pierce GL, Hui SSC, Kang M. Association Between Cardiorespiratory Fitness and Trend of Age-Related Rise in Arterial Stiffness in Individuals With and Without Hypertension or Diabetes. Am J Hypertens 2024; 38:46-54. [PMID: 39306668 DOI: 10.1093/ajh/hpae124] [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: 06/16/2024] [Revised: 08/19/2024] [Accepted: 09/17/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND This study aimed to investigate whether higher cardiorespiratory fitness (CRF) can modify the trend of age-related rise in arterial stiffness in individuals with and without hypertension (HTN) or diabetes. METHODS The study included 4,935 participants who underwent maximal cardiopulmonary exercise testing with respiratory gas analysis in a health screening program. CRF was directly measured using peak oxygen uptake during the cardiopulmonary exercise test, while arterial stiffness was evaluated using brachial-ankle pulse wave velocity (baPWV). RESULTS Participants with high CRF levels had significantly lower baPWV compared with those with low CRF levels, regardless of HTN or diabetes status (P < 0.05). The trend of baPWV increased with age, but the rate of age-related increase in baPWV was lower in individuals with moderate-to-high CRF levels compared with those with low CRF levels, regardless of HTN or diabetes status. Joint association analysis indicated that the trend of age-related increase in baPWV was the lowest in fit individuals without HTN or diabetes compared with unfit individuals with HTN or diabetes (P < 0.01). However, the trend of age-related increase in baPWV was not attenuated in fit with HTN or diabetes compared with unfit with HTN or diabetes. CONCLUSIONS These findings suggest that higher CRF levels may mitigate the trend of age-related rise in arterial stiffness in individuals with and without HTN or diabetes. However, this attenuating trend appears more pronounced in individuals without HTN or diabetes.
Collapse
Affiliation(s)
- Sae Young Jae
- Department of Sport Science, University of Seoul, Seoul, Republic of Korea
| | - Kyung Hyun Lee
- Department of Digital Health, SAIHST, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun Jeong Kim
- Department of Sport Science, University of Seoul, Seoul, Republic of Korea
| | - Setor K Kunutsor
- Diabetes Research Centre, Leicester General Hospital, University of Leicester, Leicester, UK
- Department of Internal Medicine, Section of Cardiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gary L Pierce
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa, USA
| | - Stanley Sai-Chuen Hui
- Department of Sports Science and Physical Education, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Mira Kang
- Department of Digital Health, SAIHST, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Health Promotion Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
24
|
Söderström M, Grönlund C, Liv P, Nyman E, Näslund U, Wester P. Aortic arterial stiffness associates with carotid intima-media thickness and carotid plaques in younger middle-aged healthy people. Blood Press 2024; 33:2405161. [PMID: 39291635 DOI: 10.1080/08037051.2024.2405161] [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: 06/20/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE Aortic stiffness, assessed as estimated aortic pulse wave velocity (aPWV), and carotid intima-media thickness (cIMT) are markers of vascular age, and carotid plaques are a marker of early atherosclerosis. In this cross-sectional study we aimed to investigate the association between aPWV, cIMT and plaques across different age groups and in women and men, in a middle-aged healthy population. MATERIALS AND METHODS Participants in the 6.5-year follow-up of the VIPVIZA trial who were aged 47, 57 and 67 underwent an oscillometric measurement which estimates aPWV between 2020 and 2023. Carotid ultrasound examinations were also performed. Linear and ordinal regression models were used to investigate how aPWV associates with cIMT and with carotid plaques, for the overall study group and stratified for age groups and sex. RESULTS A total of 1046 subjects were included in the analyses. Linear associations between aPWV and cIMT (β = 0.018, 95% CI: 0.006-0.030, p = 0.003), and between aPWV and plaques (OR: 1.19, 95% CI: 1.03-1.38, p = 0.018), were seen in the 57-year-olds. In the 47-year-olds a significant association was seen between aPWV and plaques (OR: 2.98 95% CI: 1.44-6.14, p = 0.003). No significant associations were seen in the 67-year-olds. For women, a significant association between aPWV and cIMT (β = 0.011, 95% CI: 0.004-0.017, p = 0.002) was shown. CONCLUSION Estimated aPWV was positively associated with increasing cIMT and the presence of carotid plaques in younger middle-aged individuals, and with cIMT in women, suggesting that measurement of estimated aPWV may improve cardiovascular risk assessment in younger middle-aged individuals and women.Clinical Trial Registration date 8 May 2013: URL: www.clinicaltrials.gov. Unique identifier: NCT01849575.
Collapse
Affiliation(s)
- Martina Söderström
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Christer Grönlund
- Department of Diagnostics and Intervention, Radiation Physics and Biomedical Engineering, Umeå University, Umeå, Sweden
| | - Per Liv
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Emma Nyman
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Ulf Näslund
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Per Wester
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
25
|
Luo X, Pang Z, Li J, Anh M, Kim BS, Gao G. Bioengineered human arterial equivalent and its applications from vascular graft to in vitro disease modeling. iScience 2024; 27:111215. [PMID: 39555400 PMCID: PMC11565542 DOI: 10.1016/j.isci.2024.111215] [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] [Indexed: 11/19/2024] Open
Abstract
Arterial disorders such as atherosclerosis, thrombosis, and aneurysm pose significant health risks, necessitating advanced interventions. Despite progress in artificial blood vessels and animal models aimed at understanding pathogenesis and developing therapies, limitations in graft functionality and species discrepancies restrict their clinical and research utility. Addressing these issues, bioengineered arterial equivalents (AEs) with enhanced vascular functions have been developed, incorporating innovative technologies that improve clinical outcomes and enhance disease progression modeling. This review offers a comprehensive overview of recent advancements in bioengineered AEs, systematically summarizing the bioengineered technologies used to construct these AEs, and discussing their implications for clinical application and pathogenesis understanding. Highlighting current breakthroughs and future perspectives, this review aims to inform and inspire ongoing research in the field, potentially transforming vascular medicine and offering new avenues for preclinical and clinical advances.
Collapse
Affiliation(s)
- Xi Luo
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Zherui Pang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology, Zhuhai 519088, China
| | - Minjun Anh
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Soo Kim
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
26
|
Ahmed B, Rahman AA, Lee S, Malhotra R. The Implications of Aging on Vascular Health. Int J Mol Sci 2024; 25:11188. [PMID: 39456971 PMCID: PMC11508873 DOI: 10.3390/ijms252011188] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Vascular aging encompasses structural and functional changes in the vasculature, significantly contributing to cardiovascular diseases, which are the leading cause of death globally. The incidence and prevalence of these diseases increase with age, with most morbidity and mortality attributed to myocardial infarction and stroke. Diagnosing and intervening in vascular aging while understanding the mechanisms behind age-induced vascular phenotypic and pathophysiological alterations offers the potential for delaying and preventing cardiovascular mortality in an aging population. This review delves into various aspects of vascular aging by examining age-related changes in arterial health at the cellular level, including endothelial dysfunction, cellular senescence, and vascular smooth muscle cell transdifferentiation, as well as at the structural level, including arterial stiffness and changes in wall thickness and diameter. We also explore aging-related changes in perivascular adipose tissue deposition, arterial collateralization, and calcification, providing insights into the physiological and pathological implications. Overall, aging induces phenotypic changes that augment the vascular system's susceptibility to disease, even in the absence of traditional risk factors, such as hypertension, diabetes, obesity, and smoking. Overall, age-related modifications in cellular phenotype and molecular homeostasis increase the vulnerability of the arterial vasculature to structural and functional alterations, thereby accelerating cardiovascular risk. Increasing our understanding of these modifications is crucial for success in delaying or preventing cardiovascular diseases. Non-invasive techniques, such as measuring carotid intima-media thickness, pulse wave velocity, and flow-mediated dilation, as well as detecting vascular calcifications, can be used for the early detection of vascular aging. Targeting specific pathological mechanisms, such as cellular senescence and enhancing angiogenesis, holds promise for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Ahmed A. Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sujin Lee
- Division of Vascular Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Rajeev Malhotra
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
27
|
Artola Arita V, Beigrezaei S, Franco OH. Risk factors for cardiovascular disease: the known unknown. Eur J Prev Cardiol 2024; 31:e106-e107. [PMID: 38099566 DOI: 10.1093/eurjpc/zwad392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 10/11/2024]
Affiliation(s)
- Vicente Artola Arita
- Department of Global Public Health and Bioethics, Julius Center, University Medical Center (UMC) Utrecht, Universiteitsweg 100, 3584 CX Utrecht, The Netherlands
| | - Sara Beigrezaei
- Department of Global Public Health and Bioethics, Julius Center, University Medical Center (UMC) Utrecht, Universiteitsweg 100, 3584 CX Utrecht, The Netherlands
| | - Oscar H Franco
- Department of Global Public Health and Bioethics, Julius Center, University Medical Center (UMC) Utrecht, Universiteitsweg 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
28
|
Azizzadeh M, Karimi A, Breyer-Kohansal R, Hartl S, Breyer MK, Gross C, Boutouyrie P, Bruno RM, Hametner B, Wassertheurer S, Burghuber OC, Weber T. Reference equations for pulse wave velocity, augmentation index, amplitude of forward and backward wave in a European general adult population. Sci Rep 2024; 14:23151. [PMID: 39367200 PMCID: PMC11452679 DOI: 10.1038/s41598-024-74162-5] [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: 06/27/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
Pulsatile hemodynamics have been shown to be independent predictors of cardiovascular events. The aim of the current study was to describe four pulsatile hemodynamic markers in a large, well-established, population-based cohort and to provide reference equations for sex- and age-based standardization of these measurements. 6828 adult participants from the Austrian LEAD (Lung, hEart, sociAl, boDy) cohort study, who were free from overt cardiovascular disease, non-diabetic based on blood test results, and had no history of pharmacological treatment for hypertension, dyslipidemia, and diabetes, comprised the "reference population". Carotid-femoral pulse wave velocity (cfPWV), augmentation index (AIx), amplitude of forward wave (Pf), and backward wave (Pb) were described in different age categories for both sexes. Sex-specific reference equations for cfPWV, AIx, Pf, and Pb with age as the predictive variable were created using the Lambda-Mu-Sigma (LMS) method. All four parameters increased with age. CfPWV and Pf were higher in males than females, especially in young and middle-age groups (P < 0.001). AIx was higher in females than males in all age categories (P < 0.001). Pb was also higher in females than males in age groups older than 40 years (P < 0.01). Reference equations for the skewness (Lambda), median (Mu), and coefficient of variation (Sigma) values were determined, enabling the calculation of sex- and age-standardized values (z-scores) for each individual's pulsatile hemodynamic measurement, and an online application was developed. Reference equations derived from a large population-based dataset constitute a suitable tool for the standardization of pulsatile hemodynamics and for the accurate interpretation of vascular aging.
Collapse
Affiliation(s)
- Mohammad Azizzadeh
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria.
- Faculty of Medicine, Sigmund Freud Private University, Vienna, Austria.
| | - Ahmad Karimi
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Faculty of Medicine, Sigmund Freud Private University, Vienna, Austria
| | - Robab Breyer-Kohansal
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Department of Respiratory and Pulmonary Diseases, Clinic Hietzing, Vienna Healthcare Group, Vienna, Austria
| | - Sylvia Hartl
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Faculty of Medicine, Sigmund Freud Private University, Vienna, Austria
| | - Marie-Kathrin Breyer
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Department of Respiratory and Pulmonary Diseases, Clinic Penzing, Vienna Healthcare Group, Vienna, Austria
| | - Christoph Gross
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Faculty of Medicine, Sigmund Freud Private University, Vienna, Austria
| | - Pierre Boutouyrie
- Inserm, PARCC, Université Paris Cité, 75015, Paris, France
- Service de Pharmacologie et Hypertension, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - Rosa Maria Bruno
- Inserm, PARCC, Université Paris Cité, 75015, Paris, France
- Service de Pharmacologie et Hypertension, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | - Bernhard Hametner
- AIT Austrian Institute of Technology, Center for Health and Bioresources, Vienna, Austria
| | | | - Otto Chris Burghuber
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Faculty of Medicine, Sigmund Freud Private University, Vienna, Austria
| | - Thomas Weber
- Department of Cardiology, Klinikum Wels-Grieskirchen, Wels, Austria
| |
Collapse
|
29
|
Kakaletsis N, Protogerou AD, Kotsis V, Vemmos K, Korompoki E, Kollias A, Milionis H, Ntaios G, Savopoulos C. Advanced vascular aging and outcomes after acute ischemic stroke: a systematic review and meta-analysis. J Hum Hypertens 2024; 38:676-686. [PMID: 39317753 DOI: 10.1038/s41371-024-00961-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Pulse wave velocity (PWV) is as a reliable marker of arterial stiffness and vascular aging, surpassing traditional risk factors in predicting detrimental cardiovascular events. The present meta-analysis aims to investigate PWV thresholds and assess its prognostic value in outcomes of acute ischemic stroke (AIS). A search was conducted in PubMed, Cochrane, Web of Science, and Scopus for studies published up to January 2024, focusing on patients admitted with AIS, wherein arterial stiffness was assessed through PWV measurements during hospitalization. Identified studies reported PWV values in individuals with both favorable and unfavorable outcomes at the end of follow-up. Initially, 35 eligible studies provided data for weighted mean baPWV (11,953 AIS patients) and cfPWV (2,197 AIS patients) calculations. The average age was 67 years, with approximately 60% male, 67% hypertensive, 30% diabetic and 30% smoker participants. The weighted mean systolic blood pressure was approximately 150 mmHg. In AIS patients, the mean PWV was 10 m/s for standard cfPWV and 20 m/s for baPWV. Nine cohort studies (6,006 AIS patients) were included in the quantitative analysis of clinical outcomes. Higher PWV levels were associated with poorer functional outcomes (2.3 m/s higher, 95%CI:1.2-3.4, p < 0.001; I2 = 87.4%). AIS patients with arterial stiffness/vascular aging (higher PWV) had approximately 46.2% increased risk of poor functional outcome, 12.7% higher risk of mortality, 13.9% greater risk of major adverse cardiovascular events, and 13.9% greater risk of stroke recurrence over the long term compared to those without arterial stiffness. Advanced vascular aging, as indicated by PWV, significantly predicts adverse outcomes in AIS patients. Integrating the assessment of vascular aging into clinical practice can improve risk perception in these patients.
Collapse
Affiliation(s)
- Nikolaos Kakaletsis
- Second Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece.
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece.
| | - Athanase D Protogerou
- Cardiovascular Prevention & Research Unit, Clinic & Laboratory of Pathophysiology, Department of Medicine, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasilios Kotsis
- Third Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Konstantinos Vemmos
- Department of Clinical Therapeutics of Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni Korompoki
- Department of Clinical Therapeutics of Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Kollias
- Third Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, Athens, Greece
| | - Haralampos Milionis
- Department of Internal Medicine, Medical School, University of Ioannina, University Hospital of Ioannina, Ioannina, Greece
| | - George Ntaios
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Christos Savopoulos
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
30
|
Dorogovtsev V, Yankevich D, Martyushev-Poklad A, Borisov I, Grechko AV. The Importance of Orthostatic Increase in Pulse Wave Velocity in the Diagnosis of Early Vascular Aging. J Clin Med 2024; 13:5713. [PMID: 39407773 PMCID: PMC11476871 DOI: 10.3390/jcm13195713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/08/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Vascular aging can be assessed by arterial stiffness measured through pulse wave velocity (PWV). Increased PWV predicts arterial hypertension, cardiovascular events and all-cause mortality. Detection of early signs of vascular aging remains an unmet problem. To search for the most sensitive markers for the early increase in vascular stiffness in a healthy population. Methods: One-hundred and twenty healthy subjects were divided in three equal age groups: <30 years, 30-45 years and >45 years. Head-up tilt test (HUTT) protocol was applied, providing a standardized hydrostatic column height. PWV at the brachial-ankle artery site (baPWV) was measured using a multichannel sphygmomanometer ABI System 100 PWV in three positions: in the baseline horizontal (supine) position-baPWVb; during the head tilt-up with an individual angle of inclination-baPWVt; and when returning to supine. Results: The most sensitive marker of early stiffness increase in a healthy population is the relative orthostatic increase in baPWV, ΔbaPWV/baPWVb, where ΔbaPWV = baPWVt - baPWVb. The significance of differences in this parameter between the young and elderly groups reached p = 0.000075 and p = 0.000006, respectively. Conclusions: The proposed index ΔbaPWV/baPWVb can be considered as a promising sensitive early biomarker of vascular aging and as a potential effective indicator in cardiovascular prevention. A longitudinal cohort study is needed to confirm this assumption.
Collapse
Affiliation(s)
- Victor Dorogovtsev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia; (D.Y.); (A.M.-P.); (I.B.); (A.V.G.)
| | | | | | | | | |
Collapse
|
31
|
Ciolac EG, Babjakova J, de Abreu RM, Mao SJ, Qian G, Teixeira do Amaral V, Wrzesinski B, Ferron AJT, Ossowski Z, Francisqueti-Ferron FV, Yeniğün SC, Fernandes B, Rodrigues LM, Gabulova R. Role of sex and training characteristics on exercise effects on cardiovascular aging: protocol for a systematic review with meta-analysis of randomized trials. Syst Rev 2024; 13:234. [PMID: 39277764 PMCID: PMC11401294 DOI: 10.1186/s13643-024-02644-8] [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: 08/24/2023] [Accepted: 08/22/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Cardiovascular diseases remain a leading global cause of mortality worldwide especially in older adults. Although it is known that regular exercise reduces cardiovascular diseases incidence, its effects on specific cardiovascular aging parameters considering the influence of sex and different exercise designs are still not fully understood. Therefore, this systematic review and meta-analysis aims to evaluate the effects of different physical exercise protocols on age-related cardiovascular outcomes in older adults. METHODS This systematic review and meta-analysis will be reported in agreement with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. Articles will be eligible if they are randomized controlled trials with a primary objective of evaluating the chronic effects of exercise interventions on cardiovascular aging parameters. Search strategy will be performed from the inception to September 30th, 2023, in the following electronic databases: MEDLINE (Ovid), SCOPUS (Elsevier), Embase, Sport Discus (EBSCO), Cochrane Central Register of Controlled Trials (CENTRAL), and Web of Science Core Collection (Clarivate Analytics). Data will be extracted and managed through Research Electronic Data Capture (REDCap) software. The Tool for the assEssment of Study qualiTy and reporting in EXercise (TESTEX) will be used to assess the methodological quality of included studies. Additionally, the quality of the findings will be evaluated using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) profiler. Meta-analysis based on the random-effects model will be performed (if deemed suitable, considering the methodological and clinical heterogeneity of the studies) to estimate the effects of exercise training on cardiovascular aging variables (i.e., cardiac output; arterial stiffness; stroke volume; endothelial function; and carotid intima-media thickness). Heterogeneity will be assessed with the I2 statistics, while the publication bias will be assessed based on Egger's test. DISCUSSION To the best of our knowledge, this will be the first systematic review and meta-analysis to investigate the impact of sex and training protocols on the cardiovascular aging parameters. Moreover, the findings of this systematic review and meta-analysis will provide evidence for health professionals in the management of elderly patients in order to optimize the exercise prescription to face the cardiovascular alterations related to the aging process, considering the effects of different protocols according to sex. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42023441015 .
Collapse
Affiliation(s)
- Emmanuel Gomes Ciolac
- Exercise and Chronic Disease Research Laboratory, Department of Physical Education, School of Sciences, São Paulo State University (Unesp), Av. Eng. Luiz Edmundo Carrijo Coube 14-01, Bauru, Sao Paulo, 17033-360, Brazil.
| | - Jana Babjakova
- Faculty of Medicine, Comenius University in Bratislava, Institute of Hygiene, Bratislava, Slovakia
| | - Raphael Martins de Abreu
- Department of Physiotherapy, LUNEX University, International University of Health, Exercise & Sports S.A, Differdange, Luxembourg
- LUNEX ASBL Luxembourg Health & Sport Sciences Research Institute, Differdange, Luxembourg
| | - Su-Jie Mao
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Guoping Qian
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Vanessa Teixeira do Amaral
- Exercise and Chronic Disease Research Laboratory, Department of Physical Education, School of Sciences, São Paulo State University (Unesp), Av. Eng. Luiz Edmundo Carrijo Coube 14-01, Bauru, Sao Paulo, 17033-360, Brazil
| | - Bartlomiej Wrzesinski
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Artur Junio Togneri Ferron
- Exercise and Chronic Disease Research Laboratory, Department of Physical Education, School of Sciences, São Paulo State University (Unesp), Av. Eng. Luiz Edmundo Carrijo Coube 14-01, Bauru, Sao Paulo, 17033-360, Brazil
| | - Zbigniew Ossowski
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Fabiane Valentini Francisqueti-Ferron
- Exercise and Chronic Disease Research Laboratory, Department of Physical Education, School of Sciences, São Paulo State University (Unesp), Av. Eng. Luiz Edmundo Carrijo Coube 14-01, Bauru, Sao Paulo, 17033-360, Brazil
- Medical School, São Paulo State University (Unesp), Botucatu, Brazil
| | - Seda Cansu Yeniğün
- Faculty of Health Sciences, Akdeniz University, Kumluca , Antalya, Turkey
| | - Bianca Fernandes
- Exercise and Chronic Disease Research Laboratory, Department of Physical Education, School of Sciences, São Paulo State University (Unesp), Av. Eng. Luiz Edmundo Carrijo Coube 14-01, Bauru, Sao Paulo, 17033-360, Brazil
| | - Luis Monteiro Rodrigues
- Research Center for Biosciences & Health Technologies (CBIOS), Universidade Lusófona, Lisbon, Portugal
| | - Rahima Gabulova
- Department of Family Medicine, Azerbaijan Medical University, Baku, Azerbaijan
| |
Collapse
|
32
|
Badhwar S, Marais L, Khettab H, Poli F, Li Y, Segers P, Aasmul S, de Melis M, Baets R, Greenwald S, Bruno RM, Boutouyrie P. Clinical Validation of Carotid-Femoral Pulse Wave Velocity Measurement Using a Multi-Beam Laser Vibrometer: The CARDIS Study. Hypertension 2024; 81:1986-1995. [PMID: 38934112 PMCID: PMC11319084 DOI: 10.1161/hypertensionaha.124.22729] [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: 01/10/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Carotid-femoral pulse wave velocity (cfPWV) is the gold standard for noninvasive arterial stiffness assessment, an independent predictor of cardiovascular disease, and a potential parameter to guide therapy. However, cfPWV is not routinely measured in clinical practice due to the unavailability of a low-cost, operator-friendly, and independent device. The current study validated a novel laser Doppler vibrometry (LDV)-based measurement of cfPWV against the reference technique. METHODS In 100 (50 men) hypertensive patients, cfPWV was measured using applanation tonometry (Sphygmocor) and the novel LDV device. This device has 2 handpieces with 6 laser beams each that simultaneously measure vibrations from the skin surface at carotid and femoral sites. Pulse wave velocity is calculated using ECG for the identification of cardiac cycles. An ECG-independent method was also devised. Cardiovascular risk score was calculated for patients between 40 and 75 years old using the WHO risk scoring chart. RESULTS LDV-based cfPWV correlated significantly with tonometry (r=0.86, P<0.0001 ECG-dependent [cfPWVLDV_ECG] and r=0.80, P<0.001 ECG-independent [cfPWVLDV_w/oECG] methods). Bland-Altman analysis showed nonsignificant bias (0.65 m/s) and acceptable SD (1.27 m/s) between methods. Intraobserver coefficient of variance for LDV was 4.7% (95% CI, 3.0%-5.5%), and interobserver coefficient of variance was 5.87%. CfPWV correlated significantly with CVD risk (r=0.64, P<0.001; r=0.41, P=0.003; and r=0.37, P=0.006 for tonometry, LDV-with, and LDV-without ECG, respectively). CONCLUSIONS The study demonstrates clinical validity of the LDV device. The LDV provides a simple, noninvasive, operator-independent method to measure cfPWV for assessing arterial stiffness, comparable to the standard existing techniques. REGISTRATION URL: https://clinicaltrials.gov/study/NCT03446430; Unique identifier: NCT03446430.
Collapse
Affiliation(s)
- Smriti Badhwar
- Paris Cardiovascular Research Center (PARCC) Institut National de la Santé Et de la Researche Médicale (INSERM), Paris, France (S.B., L.M., F.P., R.M.B., P.B.)
| | - Louise Marais
- Paris Cardiovascular Research Center (PARCC) Institut National de la Santé Et de la Researche Médicale (INSERM), Paris, France (S.B., L.M., F.P., R.M.B., P.B.)
| | - Hakim Khettab
- Hôpital européen Georges-Pompidou (HEGP), Assistance publique-Hôpitaux de Paris (APHP), Paris, France (H.K., R.M.B, P.B.)
| | - Federica Poli
- Paris Cardiovascular Research Center (PARCC) Institut National de la Santé Et de la Researche Médicale (INSERM), Paris, France (S.B., L.M., F.P., R.M.B., P.B.)
| | - Yanlu Li
- Photonics Research Group, Ghent University-imec, Belgium (Y.L., R.B.)
- Center for Nano- and Biophotonics, Ghent University, Belgium (Y.L., R.B.)
| | | | - Soren Aasmul
- Medtronic Bakken Research Center, Maastricht, The Netherlands (S.A., M.d.M.)
| | - Mirko de Melis
- Medtronic Bakken Research Center, Maastricht, The Netherlands (S.A., M.d.M.)
| | - Roel Baets
- Photonics Research Group, Ghent University-imec, Belgium (Y.L., R.B.)
- Center for Nano- and Biophotonics, Ghent University, Belgium (Y.L., R.B.)
| | - Steve Greenwald
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (S.G.)
| | - Rosa Maria Bruno
- Paris Cardiovascular Research Center (PARCC) Institut National de la Santé Et de la Researche Médicale (INSERM), Paris, France (S.B., L.M., F.P., R.M.B., P.B.)
- Hôpital européen Georges-Pompidou (HEGP), Assistance publique-Hôpitaux de Paris (APHP), Paris, France (H.K., R.M.B, P.B.)
- Université Paris Cité, Faculté de Médecine, France (R.M.B, P.B.)
| | - Pierre Boutouyrie
- Paris Cardiovascular Research Center (PARCC) Institut National de la Santé Et de la Researche Médicale (INSERM), Paris, France (S.B., L.M., F.P., R.M.B., P.B.)
- Hôpital européen Georges-Pompidou (HEGP), Assistance publique-Hôpitaux de Paris (APHP), Paris, France (H.K., R.M.B, P.B.)
- Université Paris Cité, Faculté de Médecine, France (R.M.B, P.B.)
| |
Collapse
|
33
|
Kaufmann CC, Breyer MK, Hartl S, Gross C, Schiffers C, Wouters EFM, Breyer-Kohansal R, Weber T, Huber K, Agusti A, Burghuber OC. Association of Preserved Ratio Impaired Spirometry with Arterial Stiffness. Ann Am Thorac Soc 2024; 21:1289-1298. [PMID: 38820245 DOI: 10.1513/annalsats.202310-859oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 05/23/2024] [Indexed: 06/02/2024] Open
Abstract
Rationale: Preserved ratio impaired spirometry (PRISm) is a recently recognized spirometric pattern defined by a ratio of forced expiratory volume in 1 second to forced vital capacity of at least 0.70 and a forced expiratory volume in 1 second <80% of reference. For unclear reasons, PRISm is associated with increased cardiovascular (CV) morbidity and mortality. Arterial stiffness is a major mechanism of CV disease, which can be measured by carotid-femoral pulse-wave velocity (cfPWV). Objectives: We explored the hypothesis that cfPWV would be increased in individuals with PRISm and airflow limitation (AL). Methods: We measured forced spirometry, lung volumes by body plethysmography, and cfPWV in 9,466 subjects recruited from the general population in the Austrian cross-sectional LEAD (Lung, Heart, Social, Body) study and tested the association of arterial stiffness with PRISm and AL by multivariable linear regression analysis. Individuals younger than 18 years were excluded from the study. Results: Individuals with PRISm (n = 431; 4.6%) were of similar age to those with normal spirometry (n = 8,136; 85.9%) and significantly younger than those with AL (n = 899; 9.5%). Arterial hypertension, diabetes mellitus, coronary artery disease, heart failure, and peripheral arterial occlusive disease were significantly more common in individuals with PRISm than in those with normal lung function and similar to those with AL. There was a significant association between PRISm and arterial stiffness on bivariate linear regression analysis (crude model, β = 0.038; 95% confidence interval [CI], 0.016-0.058), which persisted after robust adjustment for clinical confounders upon multivariable analysis (final model, β = 0.017; 95% CI, 0.001-0.032). cfPWV was significantly higher in individuals with PRISm irrespective of the presence of established CV disease or pulmonary restriction. AL also showed a significant association with arterial stiffness on multivariable linear regression analysis (final model, β = 0.025; 95% CI, 0.009-0.042). Conclusions: Arterial stiffness measured by cfPWV is increased in individuals with PRISm independent of CV disease and risk factors. The pathobiological mechanisms underlying this association deserve further research.
Collapse
Affiliation(s)
- Christoph C Kaufmann
- 3rd Medical Department, Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), Vienna, Austria
| | - Marie-Kathrin Breyer
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Department of Respiratory and Pulmonary Diseases, Clinic Penzing, and
| | - Sylvia Hartl
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Faculty of Medicine, Sigmund Freud University, Vienna, Austria
| | | | | | - Emiel F M Wouters
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
- Faculty of Medicine, Sigmund Freud University, Vienna, Austria
| | - Robab Breyer-Kohansal
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Clinic Hietzing, Vienna Healthcare Group, Vienna, Austria
| | - Thomas Weber
- Department of Cardiology, Klinikum Wels-Grieskirchen, Wels, Austria; and
| | - Kurt Huber
- 3rd Medical Department, Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), Vienna, Austria
- Faculty of Medicine, Sigmund Freud University, Vienna, Austria
| | - Alvar Agusti
- Catedra Salud Respiratoria, Universidad de Barcelona, Clinic Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer, Centro de Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
| | - Otto C Burghuber
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Faculty of Medicine, Sigmund Freud University, Vienna, Austria
| |
Collapse
|
34
|
Pescari D, Borlea A, Mihuta S, Stoian D. Development of a comprehensive risk prediction model for arterial stiffness assessment in individuals with obesity. Front Med (Lausanne) 2024; 11:1430437. [PMID: 39224613 PMCID: PMC11368134 DOI: 10.3389/fmed.2024.1430437] [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: 05/09/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Obesity in adults is a known risk factor for cardiovascular events and is associated with a decline in arterial elasticity. This study aims to evaluate the utility of pulse wave analysis (PWA) parameters in routine clinical practice for the primary prevention of cardiovascular events by developing a prediction model for arterial stiffness among obese and overweight individuals. Methods The study enrolled 84 adult patients, aged 18 to 85 years, with varying degrees of weight status, including optimal weight, overweight, and obesity. The lifestyle habits, the personal and family history of cardiometabolic diseases, as well the clinical evaluation that included BMI (body mass index), WHR (waist-to-hip ratio), WC (waist circumferance) were performed. PWA evaluation was conducted using the Mobil-O-Graph device, assessing the following parameters: pulse wave velocity (PWV), augmentation index (AIx), heart rate (HR), central pulse pressure (cPP), peripheral and central blood pressure (SBP, DBP, cSBP, cDBP). Body composition analysis was performed using the TANITA BC-418 body analyzer. Laboratory results from the past 3 months were also collected during initial nutritional consultations for each patient. Results Family history of cardiovascular events showed positive correlations with all PWA parameters, while diabetes history only with PWV and family history of obesity with PWV, DBP, and cSBP. Insufficient sleep duration showed positive associations with all arterial stiffness parameters except cDBP. Smoking status correlated with significantly elevated PWV and Aix values, while insufficient physical activity was associated solely with PWV. Positive correlations were showed between current weight and PWV, while WC demonstrated positive associations with PWV, SBP, and cSBP. Body composition analysis revealed significant associations between trunk adipose tissue mass (%) and PWV, SBP, and cSBP. Hydration status (%) emerged as an independent predictor for PWV, exhibiting an inverse relationship. HOMA-IR (Homeostatic Model Assessment for Insulin Resistance) showed a strong positive correlation with PWV. Negative associations were observed with HDL-c and vitamin D. Threshold values for age, cDBP and Cardiac Index providing positive diagnostic for vascular impairment. Conclusion The assessment of arterial stiffness can be considered a reliable approach to prevent obesity-related cardiovascular events and facilitate the comprehensive management of such pathologies.
Collapse
Affiliation(s)
- Denisa Pescari
- Department of Doctoral Studies, “Victor Babeş” University of Medicine and Pharmacy, Timişoara, Romania
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeş” University of Medicine and Pharmacy, Timişoara, Romania
| | - Andreea Borlea
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeş” University of Medicine and Pharmacy, Timişoara, Romania
- Discipline of Endocrinology, Second Department of Internal Medicine, “Victor Babeş” University of Medicine and Pharmacy, Timişoara, Romania
| | - Simina Mihuta
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeş” University of Medicine and Pharmacy, Timişoara, Romania
| | - Dana Stoian
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeş” University of Medicine and Pharmacy, Timişoara, Romania
- Discipline of Endocrinology, Second Department of Internal Medicine, “Victor Babeş” University of Medicine and Pharmacy, Timişoara, Romania
| |
Collapse
|
35
|
Kakaletsis N, Kotsis V, Protogerou AD, Vemmos K, Korompoki E, Kollias A, Karagiannis T, Milionis H, Ntaios G, Savopoulos C. Early vascular aging in acute ischemic stroke: A systematic review and meta-analysis. J Stroke Cerebrovasc Dis 2024; 33:107800. [PMID: 38797457 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107800] [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: 03/26/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND While arterial stiffening is a known risk factor for cardiovascular diseases, it remains unclear whether there is an early vascular aging (EVA) in patients who have experienced acute ischemic stroke (AIS). This systematic review and meta-analysis aims to investigate whether patients with AIS exhibit EVA through pulse wave velocity (PWV) measurements shortly after the stroke onset, shedding light on the relationship between arterial stiffness, hypertension, and stroke. METHODS Thirteen case-control studies were included, comparing PWV measurements between AIS patients and non-AIS individuals. A meta-analysis was performed to compare PWV levels, age, blood pressure, and the prevalence of different cardiovascular risk factors among 1711 AIS patients and 1551 controls. RESULTS Despite AIS patients showing higher PWV compared to controls (mean difference: 1.72 m/s, 95 % CI: 1.05-2.38, p < 0.001; I2 = 88.3 %), their age did not significantly differ (95 % CI: -0.47-0.94, p = 0.519; I2 = 0 %), suggesting EVA in AIS patients. Moreover, AIS patients exhibited elevated systolic and diastolic blood pressure and had higher odds of smoking, hypertension, diabetes, and male gender compared to controls. CONCLUSIONS This study's findings underscore the presence of EVA in AIS patients, evident through increased PWV measurements shortly after stroke onset. Notably, smoking, hypertension, and diabetes mellitus emerge as substantial factors contributing to accelerated arterial stiffness within this population.
Collapse
Affiliation(s)
- N Kakaletsis
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece; Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - V Kotsis
- Third Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - A D Protogerou
- Cardiovascular Prevention & Research Unit, Clinic & Laboratory of Pathophysiology, Department of Medicine, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - K Vemmos
- Department of Clinical Therapeutics of Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - E Korompoki
- Department of Clinical Therapeutics of Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - A Kollias
- Third Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, Athens, Greece
| | - T Karagiannis
- Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - H Milionis
- Department of Internal Medicine, Medical School, University of Ioannina, University Hospital of Ioannina, Ioannina, Greece
| | - G Ntaios
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - C Savopoulos
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
36
|
Savulescu-Fiedler I, Mihalcea R, Dragosloveanu S, Scheau C, Baz RO, Caruntu A, Scheau AE, Caruntu C, Benea SN. The Interplay between Obesity and Inflammation. Life (Basel) 2024; 14:856. [PMID: 39063610 PMCID: PMC11277997 DOI: 10.3390/life14070856] [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: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is an important condition affecting the quality of life of numerous patients and increasing their associated risk for multiple diseases, including tumors and immune-mediated disorders. Inflammation appears to play a major role in the development of obesity and represents a central point for the activity of cellular and humoral components in the adipose tissue. Macrophages play a key role as the main cellular component of the adipose tissue regulating the chronic inflammation and modulating the secretion and differentiation of various pro- and anti-inflammatory cytokines. Inflammation also involves a series of signaling pathways that might represent the focus for new therapies and interventions. Weight loss is essential in decreasing cardiometabolic risks and the degree of associated inflammation; however, the latter can persist for long after the excess weight is lost, and can involve changes in macrophage phenotypes that can ensure the metabolic adjustment. A clear understanding of the pathophysiological processes in the adipose tissue and the interplay between obesity and chronic inflammation can lead to a better understanding of the development of comorbidities and may ensure future targets for the treatment of obesity.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Razvan Mihalcea
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 030167 Bucharest, Romania
| | - Radu Octavian Baz
- Clinical Laboratory of Radiology and Medical Imaging, “Sf. Apostol Andrei” County Emergency Hospital, 900591 Constanta, Romania
- Department of Radiology and Medical Imaging, Faculty of Medicine, “Ovidius” University, 900527 Constanta, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- “Prof. Dr. Matei Balș” National Institute for Infectious Diseases, 021105 Bucharest, Romania
| |
Collapse
|
37
|
Chandra Sekar N, Khoshmanesh K, Baratchi S. Bioengineered models of cardiovascular diseases. Atherosclerosis 2024; 393:117565. [PMID: 38714426 DOI: 10.1016/j.atherosclerosis.2024.117565] [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: 11/25/2023] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 05/09/2024]
Abstract
Age-associated cardiovascular diseases (CVDs), predominantly resulting from artery-related disorders such as atherosclerosis, stand as a leading cause of morbidity and mortality among the elderly population. Consequently, there is a growing interest in the development of clinically relevant bioengineered models of CVDs. Recent developments in bioengineering and material sciences have paved the way for the creation of intricate models that closely mimic the structure and surroundings of native cardiac tissues and blood vessels. These models can be utilized for basic research purposes and for identifying pharmaceutical interventions and facilitating drug discovery. The advancement of vessel-on-a-chip technologies and the development of bioengineered and humanized in vitro models of the cardiovascular system have the potential to revolutionize CVD disease modelling. These technologies offer pathophysiologically relevant models at a fraction of the cost and time required for traditional experimentation required in vivo. This progress signifies a significant advancement in the field, transitioning from conventional 2D cell culture models to advanced 3D organoid and vessel-on-a-chip models. These innovative models are specifically designed to explore the complexities of vascular aging and stiffening, crucial factors in the development of cardiovascular diseases. This review summarizes the recent progress of various bioengineered in vitro platforms developed for investigating the pathophysiology of human cardiovascular system with more focus on advanced 3D vascular platforms.
Collapse
Affiliation(s)
- Nadia Chandra Sekar
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia
| | - Khashayar Khoshmanesh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; School of Engineering, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Sara Baratchi
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
38
|
Günes-Altan M, Bosch A, Striepe K, Bramlage P, Schiffer M, Schmieder RE, Kannenkeril D. Is GFR decline induced by SGLT2 inhibitor of clinical importance? Cardiovasc Diabetol 2024; 23:184. [PMID: 38811998 PMCID: PMC11138027 DOI: 10.1186/s12933-024-02223-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/01/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Use of sodium-glucose-cotransporter-2 (SGLT2) inhibitors often causes an initial decline in glomerular filtration rate (GFR). This study addresses the question whether the initial decline of renal function with SGLT2 inhibitor treatment is related to vascular changes in the systemic circulation. METHODS We measured GFR (mGFR) and estimated GFR (eGFR) in 65 patients with type 2 diabetes (T2D) at baseline and after 12 weeks of treatment randomized either to a combination of empagliflozin and linagliptin (SGLT2 inhibitor based treatment group) (n = 34) or metformin and insulin (non-SGLT2 inhibitor based treatment group) (n = 31). mGFR was measured using the gold standard clearance technique by constant infusion of inulin. In addition to blood pressure (BP), we measured pulse wave velocity (PWV) under standardized conditions reflecting vascular compliance of large arteries, as PWV is considered to be one of the most reliable vascular parameter of cardiovascular (CV) prognosis. RESULTS Both mGFR and eGFR decreased significantly after initiating treatment, but no correlation was found between change in mGFR and change in eGFR in either treatment group (SGLT2 inhibitor based treatment group: r=-0.148, p = 0.404; non-SGLT2 inhibitor based treatment group: r = 0.138, p = 0.460). Noticeably, change in mGFR correlated with change in PWV (r = 0.476, p = 0.005) in the SGLT2 inhibitor based treatment group only and remained significant after adjustment for the change in systolic BP and the change in heart rate (r = 0.422, p = 0.018). No such correlation was observed between the change in eGFR and the change in PWV in either treatment group. CONCLUSIONS Our main finding is that after initiating a SGLT2 inhibitor based therapy an exaggerated decline in mGFR was related with improved vascular compliance of large arteries reflecting the pharmacologic effects of SGLT2 inhibitor in the renal and systemic vascular bed. Second, in a single patient with T2D, eGFR may not be an appropriate parameter to assess the true change of renal function after receiving SGLT2 inhibitor based therapy. TRIAL REGISTRATION clinicaltrials.gov (NCT02752113).
Collapse
Affiliation(s)
- Merve Günes-Altan
- Department of Nephrology and Hypertension, University Hospital Erlangen Friedrich-Alexander University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany
- Department of Cardiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnnberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany
| | - Agnes Bosch
- Department of Nephrology and Hypertension, University Hospital Erlangen Friedrich-Alexander University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany
| | - Kristina Striepe
- Department of Nephrology and Hypertension, University Hospital Erlangen Friedrich-Alexander University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany
| | - Peter Bramlage
- Institute for Pharmacology and Preventive Medicine, Bahnhofstraße 20, 49661, Cloppenburg, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, University Hospital Erlangen Friedrich-Alexander University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany
| | - Roland E Schmieder
- Department of Nephrology and Hypertension, University Hospital Erlangen Friedrich-Alexander University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany.
| | - Dennis Kannenkeril
- Department of Nephrology and Hypertension, University Hospital Erlangen Friedrich-Alexander University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054, Erlangen, Germany
| |
Collapse
|
39
|
Li X, Chen Y, Liu B, Ye M, Liu B, Lu L, Guo R. Associations Between Estimated Pulse Wave Velocity and Five-Year All-Cause Mortality in Patients with Atherosclerotic Cardiovascular Disease with and without Standard Modifiable Risk Factors: Evidence From NHANES 1999-2016. Clin Epidemiol 2024; 16:367-377. [PMID: 38827433 PMCID: PMC11144008 DOI: 10.2147/clep.s457054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/16/2024] [Indexed: 06/04/2024] Open
Abstract
Aim The study aimed to analyze the associations between estimated pulse wave velocity (ePWV) and 5-year mortality in atherosclerotic cardiovascular disease (ASCVD) patients with and without standard modifiable risk factors (SMuRFs), which included smoking status, hypertension, diabetes, and hypercholesterolemia. Methods The present retrospective cohort study utilized data from the National Health and Nutrition Examination Survey (NHANES) between 1999 and 2016. Patients with ASCVD who completed both the questionnaire survey and serum testing were included. Patients were categorized into the ≥1 SMuRF group if they had at least one SMuRF, while those without any SMuRFs were classified into the SMuRF-less group. The ePWV, which was calculated using the age and mean blood pressure, was evenly divided into three categories: low (Q1), medium (Q2), and high (Q3). Multivariable weighted Cox proportional-hazard regression analyses were utilized to explore the risk factors associated with 5-year mortality in patients with and without SMuRFs. And restricted cubic spline curve (RCS) was used to assess their nonlinear correlation. Results A total of 1901 patients with ASCVD were included in the study. For the patients in ≥1 SMuRF group, the Q3 group included patients who were older, with a higher proportion of males, more comorbidities, and a lower body mass index than the Q1 group (P<0.05). The Cox proportional-hazard regression model results revealed, the Q3 group had a higher risk of 5-year mortality than the Q1 group [hazard ratio (HR) 4.30, 95% confidence interval (CI) (2.66, 6.95), P<0.001]. RCS demonstrated a linear trend between high level of ePWV and decreased risks of mortality. Similar results were observed in the SMuRF-less group [HR 10.62, 95% CI (1.22, 92.06), P=0.032]. Conclusion A high level of ePWV signified a higher risk of 5-year mortality in ASCVD patients with and without SMuRFs.
Collapse
Affiliation(s)
- Xicong Li
- Department of Cardiovascular, Kunming Medical University, Kunming, Yunnan, People’s Republic of China
- Department of Cardiovascular, 920th Hospital of Joint Logistics Support Force, Chinese People’s Liberation Army (PLA), Kunming, Yunnan, People’s Republic of China
| | - Yubiao Chen
- Department of Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Baiyun Liu
- Department of Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Mingyuan Ye
- Department of Otolaryngology, Zhuhai Maternity and Child Health Hospital, Zhuhai, Guangdong, People’s Republic of China
| | - Bei Liu
- Department of Cardiovascular, 920th Hospital of Joint Logistics Support Force, Chinese People’s Liberation Army (PLA), Kunming, Yunnan, People’s Republic of China
| | - Lifei Lu
- Department of Respiratory Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Ruiwei Guo
- Department of Cardiovascular, 920th Hospital of Joint Logistics Support Force, Chinese People’s Liberation Army (PLA), Kunming, Yunnan, People’s Republic of China
| |
Collapse
|
40
|
Martina MR, Park C, Alastruey J, Bruno RM, Climie R, Dogan S, Tuna BG, Jerončić A, Manouchehri M, Panayiotou AG, Tamarri S, Terentes-Printzios D, Testa M, Triantafyllou A, Mayer CC, Bianchini E. Medical device regulation in vascular ageing assessment: a VascAgeNet survey exploring knowledge and perception. Expert Rev Med Devices 2024; 21:335-347. [PMID: 38557297 DOI: 10.1080/17434440.2024.2334931] [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: 12/25/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Regulation has a key role for medical devices throughout their lifecycle aiming to guarantee effectiveness and safety for users. Requirements of Regulation (EU) 2017/745 (MDR) have an impact on novel and previously approved systems. Identification of key stakeholders' needs can support effective implementation of MDR improving the translation to clinical practice of vascular ageing assessment. The aim of this work is to explore knowledge and perception of medical device regulatory framework in vascular ageing field. RESEARCH DESIGN AND METHODS A survey was developed within VascAgeNet and distributed in the community by means of the EUSurvey platform. RESULTS Results were derived from 94 participants (27% clinicians, 62% researchers, 11% companies) and evidenced mostly a fair knowledge of MDR (despite self-judged as poor by 51%). Safety (83%), validation (56%), risk management (50%) were considered relevant and associated with the regulatory process. Structured support and regulatory procedures connected with medical devices in daily practice at the institutional level are lacking (only 33% report availability of a regulatory department). CONCLUSIONS Regulation was recognized relevant by the VascAgeNet community and specific support and training in medical device regulatory science was considered important. A direct link with the regulatory sector is not yet easily available.
Collapse
Affiliation(s)
| | - Chloe Park
- University College London (UCL), London, UK
| | - Jordi Alastruey
- Department of Biomedical Engineering, King's College London, London, UK
| | - Rosa Maria Bruno
- PARCC (Paris Cardiovascular Research Center, Inserm U970), Université Paris Cité, Inserm, Paris, France
| | - Rachel Climie
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Soner Dogan
- Department of Medical Biology, Yeditepe University, School of Medicine, Istanbul, Turkiye
| | - Bilge Guvenc Tuna
- Department of Biophysics, Yeditepe University, School of Medicine, Istanbul, Turkiye
| | - Ana Jerončić
- Department of Research in Biomedicine and Health & Laboratory of Vascular Aging and Cardiovascular Prevention, University of Split School of Medicine, Split, Croatia
| | | | - Andrie G Panayiotou
- Department of Rehabilitation Sciences, Cyprus University of Technology, Limassol, Cyprus
| | | | - Dimitrios Terentes-Printzios
- First Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration Hospital, Athens, Greece
| | | | - Areti Triantafyllou
- 3rd Clinic of Internal Medicine, Papageorgiou GH, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christopher C Mayer
- AIT Austrian Institute of Technology GmbH, Center for Health & Bioresources, Medical Signal Analysis, Vienna, Austria
| | - Elisabetta Bianchini
- Institute of Clinical Physiology - Italian National Research Council (CNR-IFC), Pisa, Italy
| |
Collapse
|
41
|
Hellqvist H, Karlsson M, Hoffman J, Kahan T, Spaak J. Estimation of aortic stiffness by finger photoplethysmography using enhanced pulse wave analysis and machine learning. Front Cardiovasc Med 2024; 11:1350726. [PMID: 38529332 PMCID: PMC10961400 DOI: 10.3389/fcvm.2024.1350726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/16/2024] [Indexed: 03/27/2024] Open
Abstract
Introduction Aortic stiffness plays a critical role in the evolution of cardiovascular diseases, but the assessment requires specialized equipment. Photoplethysmography (PPG) and single-lead electrocardiogram (ECG) are readily available in healthcare and wearable devices. We studied whether a brief PPG registration, alone or in combination with single-lead ECG, could be used to reliably estimate aortic stiffness. Methods A proof-of-concept study with simultaneous high-resolution index finger recordings of infrared PPG, single-lead ECG, and finger blood pressure (Finapres) was performed in 33 participants [median age 44 (range 21-66) years, 19 men] and repeated within 2 weeks. Carotid-femoral pulse wave velocity (cfPWV; two-site tonometry with SphygmoCor) was used as a reference. A brachial single-cuff oscillometric device assessed aortic pulse wave velocity (aoPWV; Arteriograph) for further comparisons. We extracted 136 established PPG waveform features and engineered 13 new with improved coupling to the finger blood pressure curve. Height-normalized pulse arrival time (NPAT) was derived using ECG. Machine learning methods were used to develop prediction models. Results The best PPG-based models predicted cfPWV and aoPWV well (root-mean-square errors of 0.70 and 0.52 m/s, respectively), with minor improvements by adding NPAT. Repeatability and agreement were on par with the reference equipment. A new PPG feature, an amplitude ratio from the early phase of the waveform, was most important in modelling, showing strong correlations with cfPWV and aoPWV (r = -0.81 and -0.75, respectively, both P < 0.001). Conclusion Using new features and machine learning methods, a brief finger PPG registration can estimate aortic stiffness without requiring additional information on age, anthropometry, or blood pressure. Repeatability and agreement were comparable to those obtained using non-invasive reference equipment. Provided further validation, this readily available simple method could improve cardiovascular risk evaluation, treatment, and prognosis.
Collapse
Affiliation(s)
- Henrik Hellqvist
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Karlsson
- Marcus Wallenberg Laboratory for Sound and Vibration Research, Department of Engineering Mechanics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Johan Hoffman
- Division of Computational Science and Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Thomas Kahan
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Spaak
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
42
|
Tang Q, Liu S, Tao C, Wang J, Zhao H, Wang G, Zhao X, Ren Q, Zhang L, Su B, Xu J, An H. A new method for vascular age estimation based on relative risk difference in vascular aging. Comput Biol Med 2024; 171:108155. [PMID: 38430740 DOI: 10.1016/j.compbiomed.2024.108155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/26/2024] [Accepted: 02/12/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE The current models of estimating vascular age (VA) primarily rely on the regression label expressed with chronological age (CA), which does not account individual differences in vascular aging (IDVA) that are difficult to describe by CA. This may lead to inaccuracies in assessing the risk of cardiovascular disease based on VA. To address this limitation, this work aims to develop a new method for estimating VA by considering IDVA. This method will provide a more accurate assessment of cardiovascular disease risk. METHODS Relative risk difference in vascular aging (RRDVA) is proposed to replace IDVA, which is represented as the numerical difference between individual predicted age (PA) and the corresponding mean PA of healthy population. RRDVA and CA are regard as the influence factors to acquire VA. In order to acquire PA of all samples, this work takes CA as the dependent variable, and mines the two most representative indicators from arteriosclerosis data as the independent variables, to establish a regression model for obtaining PA. RESULTS The proposed VA based on RRDVA is significantly correlated with 27 indirect indicators for vascular aging evaluation. Moreover, VA is better than CA by comparing the correlation coefficients between VA, CA and 27 indirect indicators, and RRDVA greater than zero presents a higher risk of disease. CONCLUSION The proposed VA overcomes the limitation of CA in characterizing IDVA, which may help young groups with high disease risk to promote healthy behaviors.
Collapse
Affiliation(s)
- Qingfeng Tang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The University Key Laboratory of Intelligent Perception and Computing of Anhui Province, Anqing Normal University, Anqing 246133, China; Anhui Engineering Research Center of Intelligent Perception and Elderly Care, Chuzhou University, Chuzhou 239000, China.
| | - Shiping Liu
- The University Key Laboratory of Intelligent Perception and Computing of Anhui Province, Anqing Normal University, Anqing 246133, China.
| | - Chao Tao
- The University Key Laboratory of Intelligent Perception and Computing of Anhui Province, Anqing Normal University, Anqing 246133, China.
| | - Jue Wang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Huanhuan Zhao
- Anhui Engineering Research Center of Intelligent Perception and Elderly Care, Chuzhou University, Chuzhou 239000, China; School of Computer and Information Engineering, Chuzhou University, Chuzhou 239000, China.
| | - Guangjun Wang
- The University Key Laboratory of Intelligent Perception and Computing of Anhui Province, Anqing Normal University, Anqing 246133, China.
| | - Xu Zhao
- Health Management & Physical Examination Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China.
| | - Qun Ren
- Health Management & Physical Examination Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China.
| | - Liangliang Zhang
- The University Key Laboratory of Intelligent Perception and Computing of Anhui Province, Anqing Normal University, Anqing 246133, China.
| | - Benyue Su
- The University Key Laboratory of Intelligent Perception and Computing of Anhui Province, Anqing Normal University, Anqing 246133, China; School of Mathematics and Computer Science, Tongling University, Tongling 244061, China.
| | - Jiatuo Xu
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hui An
- Health Management & Physical Examination Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China.
| |
Collapse
|
43
|
Fraile-Martinez O, De Leon-Oliva D, Boaru DL, De Castro-Martinez P, Garcia-Montero C, Barrena-Blázquez S, García-García J, García-Honduvilla N, Alvarez-Mon M, Lopez-Gonzalez L, Diaz-Pedrero R, Guijarro LG, Ortega MA. Connecting epigenetics and inflammation in vascular senescence: state of the art, biomarkers and senotherapeutics. Front Genet 2024; 15:1345459. [PMID: 38469117 PMCID: PMC10925776 DOI: 10.3389/fgene.2024.1345459] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/15/2024] [Indexed: 03/13/2024] Open
Abstract
Vascular diseases pose major health challenges, and understanding their underlying molecular mechanisms is essential to advance therapeutic interventions. Cellular senescence, a hallmark of aging, is a cellular state characterized by cell-cycle arrest, a senescence-associated secretory phenotype macromolecular damage, and metabolic dysregulation. Vascular senescence has been demonstrated to play a key role in different vascular diseases, such as atherosclerosis, peripheral arterial disease, hypertension, stroke, diabetes, chronic venous disease, and venous ulcers. Even though cellular senescence was first described in 1961, significant gaps persist in comprehending the epigenetic mechanisms driving vascular senescence and its subsequent inflammatory response. Through a comprehensive analysis, we aim to elucidate these knowledge gaps by exploring the network of epigenetic alterations that contribute to vascular senescence. In addition, we describe the consequent inflammatory cascades triggered by these epigenetic modifications. Finally, we explore translational applications involving biomarkers of vascular senescence and the emerging field of senotherapy targeting this biological process.
Collapse
Affiliation(s)
- Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Joaquin García-García
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, Alcala deHenares, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala deHenares, Spain
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala deHenares, Spain
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, Alcala deHenares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, Alcala deHenares, Spain
| |
Collapse
|
44
|
Pucci G, Grillo A, Dalakleidi KV, Fraenkel E, Gkaliagkousi E, Golemati S, Guala A, Hametner B, Lazaridis A, Mayer CC, Mozos I, Pereira T, Veerasingam D, Terentes-Printzios D, Agnoletti D. Atrial Fibrillation and Early Vascular Aging: Clinical Implications, Methodology Issues and Open Questions-A Review from the VascAgeNet COST Action. J Clin Med 2024; 13:1207. [PMID: 38592046 PMCID: PMC10931681 DOI: 10.3390/jcm13051207] [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: 01/08/2024] [Revised: 02/02/2024] [Accepted: 02/18/2024] [Indexed: 04/10/2024] Open
Abstract
Atrial fibrillation (AF), the most common cardiac arrhythmia, is associated with adverse CV outcomes. Vascular aging (VA), which is defined as the progressive deterioration of arterial function and structure over a lifetime, is an independent predictor of both AF development and CV events. A timing identification and treatment of early VA has therefore the potential to reduce the risk of AF incidence and related CV events. A network of scientists and clinicians from the COST Action VascAgeNet identified five clinically and methodologically relevant questions regarding the relationship between AF and VA and conducted a narrative review of the literature to find potential answers. These are: (1) Are VA biomarkers associated with AF? (2) Does early VA predict AF occurrence better than chronological aging? (3) Is early VA a risk enhancer for the occurrence of CV events in AF patients? (4) Are devices measuring VA suitable to perform subclinical AF detection? (5) Does atrial-fibrillation-related rhythm irregularity have a negative impact on the measurement of vascular age? Results showed that VA is a powerful and independent predictor of AF incidence, however, its role as risk modifier for the occurrence of CV events in patients with AF is debatable. Limited and inconclusive data exist regarding the reliability of VA measurement in the presence of rhythm irregularities associated with AF. To date, no device is equipped with tools capable of detecting AF during VA measurements. This represents a missed opportunity to effectively perform CV prevention in people at high risk. Further advances are needed to fill knowledge gaps in this field.
Collapse
Affiliation(s)
- Giacomo Pucci
- Unit of Internal Medicine, Santa Maria University Hospital, 05100 Terni, Italy
- Department of Medicine and Surgery, University of Perugia, 06125 Perugia, Italy
| | - Andrea Grillo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Kalliopi V Dalakleidi
- Biomedical Simulations and Imaging (BIOSIM) Laboratory, School of Electrical and Computer Engineering, National Technical University of Athens, 15780 Athens, Greece
| | - Emil Fraenkel
- 1st Department of Internal Medicine, Faculty of General Medicine, Pavol Jozef Šafárik University, 04011 Košice, Slovakia
| | - Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, 54124 Thessaloniki, Greece
| | - Spyretta Golemati
- Medical School, National and Kapodistrian University of Athens, 10675 Athens, Greece
| | - Andrea Guala
- Vall d'Hebrón Research Institute (VHIR), 08035 Barcelona, Spain
- CIBER CV, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Bernhard Hametner
- AIT Austrian Institute of Technology, Center for Health & Bioresources, Medical Signal Analysis, 1210 Vienna, Austria
| | - Antonios Lazaridis
- 3rd Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, 54124 Thessaloniki, Greece
| | - Christopher C Mayer
- AIT Austrian Institute of Technology, Center for Health & Bioresources, Medical Signal Analysis, 1210 Vienna, Austria
| | - Ioana Mozos
- Department of Functional Sciences-Pathophysiology, Center for Translational Research and Systems Medicine, "Victor Babes" University of Medicine and Pharmacy, 300173 Timisoara, Romania
| | - Telmo Pereira
- H&TRC-Health & Technology Research Center, Coimbra Health School, Polytechnic University of Coimbra, 3000-331 Coimbra, Portugal
- Laboratory for Applied Research in Health (Labinsaúde), Polytechnic University of Coimbra, 3000-331 Coimbra, Portugal
| | - Dave Veerasingam
- Department of Cardiothoracic Surgery, Galway University Hospitals, H91 YR71 Galway, Ireland
| | - Dimitrios Terentes-Printzios
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Davide Agnoletti
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Cardiovascular Internal Medicine, Medical and Surgical Sciences Department, University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
45
|
Mulligan LJ, Ungerleider J, Friedman A, Sanders B, Thrash J, Ewert D, Mitrev L, Hill JC. Evaluation of ventricular-vascular coupling with critical care metrics: An in silico approach. Physiol Rep 2024; 12:e15920. [PMID: 38296348 PMCID: PMC10830386 DOI: 10.14814/phy2.15920] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 02/05/2024] Open
Abstract
Mean arterial pressure and cardiac output provide insufficient guidance for the management of intraoperative hypotension (IOH). In silico models offer additional insights into acute changes in hemodynamic parameters that may be encountered during IOH. A computational model (CM) generated parameters quantifying ventricular-vascular coupling, and pressure-volume construct across levels of aortic compliance (CA ). We studied how a loss from normal-to-stiff CA impacts critical care metrics of hemodynamics during vascular occlusion. Pulse pressure (PP), end-systolic pressure (Pes ), arterial compliance (Art-ca), arterial elastance (Art-ea), and dynamic arterial elastance (Eadyn), along mechanical efficiency (ME) were measured at five levels of CA . A loss in CA impacted all variables. During steady-state conditions, PP, Pes , and stroke work increased significantly as CA decreased. Art-ca decreased and Art-ea increased similarly; Eadyn increased and ME decreased. During a decrease in preload across all CA levels, arterial dynamics measures remained linear. The CM demonstrated that a loss in CA impacts measures of arterial dynamics during steady-state and transient conditions and the model demonstrates that critical care metrics are sensitive to changes in CA . While Art-ca and Art-ea were sensitive to changes in preload, Eadyn did not change.
Collapse
Affiliation(s)
- Lawrence J. Mulligan
- Department of AnesthesiologyCooper University HospitalCamdenNew JerseyUSA
- Cooper Medical School of Rowan UniversityCamdenNew JerseyUSA
| | | | - Adam Friedman
- Cooper Medical School of Rowan UniversityCamdenNew JerseyUSA
| | | | | | - Daniel Ewert
- University of North DakotaGrand ForksNorth DakotaUSA
| | - Ludmil Mitrev
- Department of AnesthesiologyCooper University HospitalCamdenNew JerseyUSA
- Cooper Medical School of Rowan UniversityCamdenNew JerseyUSA
| | - Jeffrey C. Hill
- School of Medical Imaging and TherapeuticsMassachusetts College of Pharmacy and Health Sciences UniversityWorcesterMassachusettsUSA
| |
Collapse
|
46
|
Bianchini E, Climie RE, Mayer CC, Martina MR, Nandi M, Schmidt-Trucksäss A, Segers P, Park C, Pucci G, Terentes-Printzios D, Charlton PH, for VascAgeNet. Unified Language for Knowledge Dissemination: The Vascular Ageing Glossary, an Initiative by VascAgeNet. Artery Res 2024; 30:1-7. [PMID: 39086596 PMCID: PMC11286687 DOI: 10.1007/s44200-023-00041-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/07/2023] [Indexed: 08/02/2024] Open
Abstract
Objectives In general, a terminology shared and agreed by different stakeholders is important to facilitate communication and cooperation. This holds true in the field of vascular ageing for the benefit of global cardiovascular health. The need to promote a common language and understanding across this area was recognised by VascAgeNet, a collaborative network with relevant and diverse expertise in the vascular ageing field, supported by the European Cooperation in Science and Technology. To contribute to the spread of unified terms in the vascular ageing field, a glossary was created by VascAgeNet and this paper describes the systematic process used for its development. Methods An initial list of terms and preliminary definitions were collated from the network. A dedicated team was created to design the glossary development process, to facilitate its implementation and to maximise outreach and dissemination. The key steps of the process were to determine: (1) the target audience; (2) a list of priority terms; (3) a template structure for definitions; (4) methods for collecting feedback and (5) the dissemination plan. Results An implementation strategy was provided for each key step and shared within the network; main decisions were agreed by all members of the glossary team. Small groups of definitions were released on a regular basis within a pilot phase including 19 terms (status: 05.09.2023) that were published openly at https://vascagenet.eu/official-glossary. Conclusions The strategy for creating the first Vascular Ageing Glossary has been successfully designed and developed within VascAgeNet. A pilot phase covering the first publicly available terms was completed. The glossary is a living document, available to the scientific community, which aims to unify the vascular ageing language. Supplementary Information The online version contains supplementary material available at 10.1007/s44200-023-00041-5.
Collapse
Affiliation(s)
- Elisabetta Bianchini
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy
| | - Rachel E. Climie
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool St, Hobart, 7000 Australia
| | - Christopher Clemens Mayer
- Center for Health and Bioresources, Medical Signal Analysis, AIT Austrian Institute of Technology GmbH, Giefinggasse 4, 1210 Vienna, Austria
| | | | - Manasi Nandi
- School of Cancer and Pharmaceutical Science, Faculty of Life Sciences and Medicine, King’s College London, 150 Stamford Street, London, SE19NH UK
| | - Arno Schmidt-Trucksäss
- Division of Sport and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052 Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Patrick Segers
- Institute of Biomedical Engineering and Technology, Ghent University, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - Chloe Park
- MRC Unit for Lifelong Health and Ageing at UCL, UCL, London, UK
| | - Giacomo Pucci
- Unit of Internal Medicine, Department of Medicine and Surgery, Santa Maria Terni Hospital, University of Perugia, Terni, Italy
| | - Dimitrios Terentes-Printzios
- First Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration Hospital, Athens, Greece
| | - Peter H. Charlton
- Strangeways Research Laboratory, Department of Public Health and Primary Care, University of Cambridge, 2 Worts Causeway, Cambridge, CB1 8RN UK
| | - for VascAgeNet
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool St, Hobart, 7000 Australia
- Center for Health and Bioresources, Medical Signal Analysis, AIT Austrian Institute of Technology GmbH, Giefinggasse 4, 1210 Vienna, Austria
- School of Cancer and Pharmaceutical Science, Faculty of Life Sciences and Medicine, King’s College London, 150 Stamford Street, London, SE19NH UK
- Division of Sport and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Grosse Allee 6, 4052 Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
- Institute of Biomedical Engineering and Technology, Ghent University, C. Heymanslaan 10, 9000 Ghent, Belgium
- MRC Unit for Lifelong Health and Ageing at UCL, UCL, London, UK
- Unit of Internal Medicine, Department of Medicine and Surgery, Santa Maria Terni Hospital, University of Perugia, Terni, Italy
- First Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration Hospital, Athens, Greece
- Strangeways Research Laboratory, Department of Public Health and Primary Care, University of Cambridge, 2 Worts Causeway, Cambridge, CB1 8RN UK
| |
Collapse
|
47
|
Cavero-Redondo I, Saz-Lara A, Martínez-García I, Otero-Luis I, Martínez-Rodrigo A. Validation of an early vascular aging construct model for comprehensive cardiovascular risk assessment using external risk indicators for improved clinical utility: data from the EVasCu study. Cardiovasc Diabetol 2024; 23:33. [PMID: 38218806 PMCID: PMC10787504 DOI: 10.1186/s12933-023-02104-y] [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: 10/31/2023] [Accepted: 12/27/2023] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Cardiovascular diseases (CVDs) remain a major global health concern, necessitating advanced risk assessment beyond traditional factors. Early vascular aging (EVA), characterized by accelerated vascular changes, has gained importance in cardiovascular risk assessment. METHODS The EVasCu study in Spain examined 390 healthy participants using noninvasive measurements. A construct of four variables (Pulse Pressure, Pulse Wave Velocity, Glycated Hemoglobin, Advanced Glycation End Products) was used for clustering. K-means clustering with principal component analysis revealed two clusters, healthy vascular aging (HVA) and early vascular aging (EVA). External validation variables included sociodemographic, adiposity, glycemic, inflammatory, lipid profile, vascular, and blood pressure factors. RESULTS EVA cluster participants were older and exhibited higher adiposity, poorer glycemic control, dyslipidemia, altered vascular properties, and higher blood pressure. Significant differences were observed for age, smoking status, body mass index, waist circumference, fat percentage, glucose, insulin, C-reactive protein, diabetes prevalence, lipid profiles, arterial stiffness, and blood pressure levels. These findings demonstrate the association between traditional cardiovascular risk factors and EVA. CONCLUSIONS This study validates a clustering model for EVA and highlights its association with established risk factors. EVA assessment can be integrated into clinical practice, allowing early intervention and personalized cardiovascular risk management.
Collapse
Affiliation(s)
- Iván Cavero-Redondo
- Health and Social Research Center, University of Castilla-La Mancha, Cuenca, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Alicia Saz-Lara
- Health and Social Research Center, University of Castilla-La Mancha, Cuenca, Spain.
| | | | - Iris Otero-Luis
- Health and Social Research Center, University of Castilla-La Mancha, Cuenca, Spain
| | - Arturo Martínez-Rodrigo
- Research Group in Electronic, Biomedical, and Telecommunication Engineering, University of Castilla-La Mancha, Cuenca, Spain
| |
Collapse
|
48
|
Mathieu AJW, Pascual MS, Charlton PH, Volovaya M, Venton J, Aston PJ, Nandi M, Alastruey J. Advanced waveform analysis of the photoplethysmogram signal using complementary signal processing techniques for the extraction of biomarkers of cardiovascular function. JRSM Cardiovasc Dis 2024; 13:20480040231225384. [PMID: 38314325 PMCID: PMC10838030 DOI: 10.1177/20480040231225384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 02/06/2024] Open
Abstract
Introduction Photoplethysmogram signals from wearable devices typically measure heart rate and blood oxygen saturation, but contain a wealth of additional information about the cardiovascular system. In this study, we compared two signal-processing techniques: fiducial point analysis and Symmetric Projection Attractor Reconstruction, on their ability to extract new cardiovascular information from a photoplethysmogram signal. The aim was to identify fiducial point analysis and Symmetric Projection Attractor Reconstruction indices that could classify photoplethysmogram signals, according to age, sex and physical activity. Methods Three datasets were used: an in-silico dataset of simulated photoplethysmogram waves for healthy male participants (25-75 years old); an in-vivo dataset containing 10-min photoplethysmogram recordings from 57 healthy subjects at rest (18-39 or > 70 years old; 53% female); and an in-vivo dataset containing photoplethysmogram recordings collected for 4 weeks from a single subject, in daily life. The best-performing indices from the in-silico study (5/48 fiducial point analysis and 6/49 Symmetric Projection Attractor Reconstruction) were applied to the in-vivo datasets. Results Key fiducial point analysis and Symmetric Projection Attractor Reconstruction indices, which showed the greatest differences between groups, were found to be consistent across datasets. These indices were related to systolic augmentation, diastolic peak positioning and prominence, and waveform variability. Both fiducial point analysis and Symmetric Projection Attractor Reconstruction techniques provided indices that supported the classification of age and physical activity, but not sex. Conclusions Both fiducial point analysis and Symmetric Projection Attractor Reconstruction techniques demonstrated utility in identifying cardiovascular differences between individuals and within an individual over time. Future research should investigate the potential utility of these techniques for extracting information on fitness and disease, to support healthcare-decision making.
Collapse
Affiliation(s)
- Aristide Jun Wen Mathieu
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King's College London, St Thomas' Hospital, London, UK
| | - Miquel Serna Pascual
- School of Cancer and Pharmaceutical Science, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Peter H Charlton
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Maria Volovaya
- School of Cancer and Pharmaceutical Science, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Jenny Venton
- School of Cancer and Pharmaceutical Science, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Philip J Aston
- Department of Mathematics, University of Surrey, Guildford, UK
| | - Manasi Nandi
- School of Cancer and Pharmaceutical Science, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Jordi Alastruey
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King's College London, St Thomas' Hospital, London, UK
| |
Collapse
|
49
|
Gyöngyösi H, Szőllősi GJ, Csenteri O, Jancsó Z, Móczár C, Torzsa P, Andréka P, Vajer P, Nemcsik J. Differences between SCORE, Framingham Risk Score, and Estimated Pulse Wave Velocity-Based Vascular Age Calculation Methods Based on Data from the Three Generations Health Program in Hungary. J Clin Med 2023; 13:205. [PMID: 38202212 PMCID: PMC10779891 DOI: 10.3390/jcm13010205] [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: 11/17/2023] [Revised: 12/28/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Early vascular ageing contributes to cardiovascular (CV) morbidity and mortality. There are different possibilities to calculate vascular age including methods based on CV risk scores, but different methods might identify different subjects with early vascular ageing. We aimed to compare SCORE and Framingham Risk Score (FRS)-based vascular age calculation methods on subjects that were involved in a national screening program in Hungary. We also aimed to compare the distribution of subjects identified with early vascular ageing based on estimated pulse wave velocity (ePWV). The Three Generations for Health program focuses on the development of primary health care in Hungary. One of the key elements of the program is the identification of risk factors of CV diseases. Vascular ages based on the SCORE and FRS were calculated based on previous publications and were compared with chronological age and with each other in the total population and in patients with hypertension or diabetes. ePWV was calculated based on a method published previously. Supernormal, normal, and early vascular ageing were defined as <10%, 10-90%, and >90% ePWV values for the participants. In total, 99,231 subjects were involved in the study, and among them, 49,191 patients had hypertension (HT) and 15,921 patients had diabetes (DM). The chronological age of the total population was 54.0 (48.0-60.0) years, while the SCORE and FRS vascular ages were 59.0 (51.0-66.0) and 64.0 (51-80) years, respectively. In the HT patients, the chronological, SCORE, and FRS vascular ages were 57.0 (51.0-62.0), 63.0 (56.0-68.0), and 79.0 (64.0-80.0) years, respectively. In the DM patients, the chronological, SCORE, and FRS vascular ages were 58.0 (52.0-62.0), 63.0 (56.0-68.0), and 80.0 (76.0-80.0) years, respectively. Based on ePWV, the FRS identified patients with an elevated vascular age with high sensitivity (97.3%), while in the case of the SCORE, the sensitivity was much lower (13.3%). In conclusion, different vascular age calculation methods can provide different vascular age results in a population-based cohort. The importance of this finding for the implementation in CV preventive strategies requires further studies.
Collapse
Affiliation(s)
- Helga Gyöngyösi
- Department of Family Medicine, Semmelweis University, 1085 Budapest, Hungary; (H.G.); (C.M.); (P.T.)
| | - Gergő József Szőllősi
- Gottsegen National Cardiovascular Center, 1096 Budapest, Hungary; (G.J.S.); (O.C.); (Z.J.); (P.A.)
- Coordination Center for Research in Social Sciences, Faculty of Economics and Business, University of Debrecen, 4032 Debrecen, Hungary
| | - Orsolya Csenteri
- Gottsegen National Cardiovascular Center, 1096 Budapest, Hungary; (G.J.S.); (O.C.); (Z.J.); (P.A.)
| | - Zoltán Jancsó
- Gottsegen National Cardiovascular Center, 1096 Budapest, Hungary; (G.J.S.); (O.C.); (Z.J.); (P.A.)
| | - Csaba Móczár
- Department of Family Medicine, Semmelweis University, 1085 Budapest, Hungary; (H.G.); (C.M.); (P.T.)
| | - Péter Torzsa
- Department of Family Medicine, Semmelweis University, 1085 Budapest, Hungary; (H.G.); (C.M.); (P.T.)
| | - Péter Andréka
- Gottsegen National Cardiovascular Center, 1096 Budapest, Hungary; (G.J.S.); (O.C.); (Z.J.); (P.A.)
| | - Péter Vajer
- Gottsegen National Cardiovascular Center, 1096 Budapest, Hungary; (G.J.S.); (O.C.); (Z.J.); (P.A.)
| | - János Nemcsik
- Department of Family Medicine, Semmelweis University, 1085 Budapest, Hungary; (H.G.); (C.M.); (P.T.)
| |
Collapse
|
50
|
Hong J, Nandi M, Charlton PH, Alastruey J. Noninvasive hemodynamic indices of vascular aging: an in silico assessment. Am J Physiol Heart Circ Physiol 2023; 325:H1290-H1303. [PMID: 37737734 PMCID: PMC10908403 DOI: 10.1152/ajpheart.00454.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023]
Abstract
Vascular aging (VA) involves structural and functional changes in blood vessels that contribute to cardiovascular disease. Several noninvasive pulse wave (PW) indices have been proposed to assess the arterial stiffness component of VA in the clinic and daily life. This study investigated 19 of these indices, identified in recent review articles on VA, by using a database comprising 3,837 virtual healthy subjects aged 25-75 yr, each with unique PW signals simulated under various levels of artificial noise to mimic real measurement errors. For each subject, VA indices were calculated from filtered PW signals and compared with the precise theoretical value of aortic Young's modulus (EAo). In silico PW indices showed age-related changes that align with in vivo population studies. The cardio-ankle vascular index (CAVI) and all pulse wave velocity (PWV) indices showed strong linear correlations with EAo (Pearson's rp > 0.95). Carotid distensibility showed a strong negative nonlinear correlation (Spearman's rs < -0.99). CAVI and distensibility exhibited greater resilience to noise compared with PWV indices. Blood pressure-related indices and photoplethysmography (PPG)-based indices showed weaker correlations with EAo (rp and rs < 0.89, |rp| and |rs| < 0.84, respectively). Overall, blood pressure-related indices were confounded by more cardiovascular properties (heart rate, stroke volume, duration of systole, large artery diameter, and/or peripheral vascular resistance) compared with other studied indices, and PPG-based indices were most affected by noise. In conclusion, carotid-femoral PWV, CAVI and carotid distensibility emerged as the superior clinical VA indicators, with a strong EAo correlation and noise resilience. PPG-based indices showed potential for daily VA monitoring under minimized noise disturbances.NEW & NOTEWORTHY For the first time, 19 noninvasive pulse wave indices for assessing vascular aging were examined together in a single database of nearly 4,000 subjects aged 25-75 yr. The dataset contained precise values of the aortic Young's modulus and other hemodynamic measures for each subject, which enabled us to test each index's ability to measure changes in aortic stiffness while accounting for confounding factors and measurement errors. The study provides freely available tools for analyzing these and additional indices.
Collapse
Affiliation(s)
- Jingyuan Hong
- Division of Imaging Sciences and Biomedical Engineering, King's College London, St. Thomas' Hospital, London, United Kingdom
| | - Manasi Nandi
- School of Cancer and Pharmaceutical Science, King's College London, London, United Kingdom
| | - Peter H Charlton
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Jordi Alastruey
- Division of Imaging Sciences and Biomedical Engineering, King's College London, St. Thomas' Hospital, London, United Kingdom
| |
Collapse
|