1
|
Müller L, Di Benedetto S. Inflammaging, immunosenescence, and cardiovascular aging: insights into long COVID implications. Front Cardiovasc Med 2024; 11:1384996. [PMID: 38988667 PMCID: PMC11233824 DOI: 10.3389/fcvm.2024.1384996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024] Open
Abstract
Aging leads to physiological changes, including inflammaging-a chronic low-grade inflammatory state with significant implications for various physiological systems, particularly for cardiovascular health. Concurrently, immunosenescence-the age-related decline in immune function, exacerbates vulnerabilities to cardiovascular pathologies in older individuals. Examining the dynamic connections between immunosenescence, inflammation, and cardiovascular aging, this mini-review aims to disentangle some of these interactions for a better understanding of their complex interplay. In the context of cardiovascular aging, the chronic inflammatory state associated with inflammaging compromises vascular integrity and function, contributing to atherosclerosis, endothelial dysfunction, arterial stiffening, and hypertension. The aging immune system's decline amplifies oxidative stress, fostering an environment conducive to atherosclerotic plaque formation. Noteworthy inflammatory markers, such as the high-sensitivity C-reactive protein, interleukin-6, interleukin-1β, interleukin-18, and tumor necrosis factor-alpha emerge as key players in cardiovascular aging, triggering inflammatory signaling pathways and intensifying inflammaging and immunosenescence. In this review we aim to explore the molecular and cellular mechanisms underlying inflammaging and immunosenescence, shedding light on their nuanced contributions to cardiovascular diseases. Furthermore, we explore the reciprocal relationship between immunosenescence and inflammaging, revealing a self-reinforcing cycle that intensifies cardiovascular risks. This understanding opens avenues for potential therapeutic targets to break this cycle and mitigate cardiovascular dysfunction in aging individuals. Furthermore, we address the implications of Long COVID, introducing an additional layer of complexity to the relationship between aging, immunosenescence, inflammaging, and cardiovascular health. Our review aims to stimulate continued exploration and advance our understanding within the realm of aging and cardiovascular health.
Collapse
Affiliation(s)
- Ludmila Müller
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | | |
Collapse
|
2
|
Abyadeh M, Gupta V, Paulo JA, Mahmoudabad AG, Shadfar S, Mirshahvaladi S, Gupta V, Nguyen CT, Finkelstein DI, You Y, Haynes PA, Salekdeh GH, Graham SL, Mirzaei M. Amyloid-beta and tau protein beyond Alzheimer's disease. Neural Regen Res 2024; 19:1262-1276. [PMID: 37905874 PMCID: PMC11467936 DOI: 10.4103/1673-5374.386406] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT The aggregation of amyloid-beta peptide and tau protein dysregulation are implicated to play key roles in Alzheimer's disease pathogenesis and are considered the main pathological hallmarks of this devastating disease. Physiologically, these two proteins are produced and expressed within the normal human body. However, under pathological conditions, abnormal expression, post-translational modifications, conformational changes, and truncation can make these proteins prone to aggregation, triggering specific disease-related cascades. Recent studies have indicated associations between aberrant behavior of amyloid-beta and tau proteins and various neurological diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, as well as retinal neurodegenerative diseases like Glaucoma and age-related macular degeneration. Additionally, these proteins have been linked to cardiovascular disease, cancer, traumatic brain injury, and diabetes, which are all leading causes of morbidity and mortality. In this comprehensive review, we provide an overview of the connections between amyloid-beta and tau proteins and a spectrum of disorders.
Collapse
Affiliation(s)
| | - Vivek Gupta
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Sina Shadfar
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Shahab Mirshahvaladi
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Christine T.O. Nguyen
- Department of Optometry and Vision Sciences, School of Health Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Yuyi You
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Paul A. Haynes
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Ghasem H. Salekdeh
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Stuart L. Graham
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| |
Collapse
|
3
|
Simistiras A, Georgiopoulos G, Delialis D, Mavraganis G, Oikonomou E, Maneta E, Loutos C, Evangelou E, Stamatelopoulos K. Association of Lipoprotein(a) with arterial stiffness: A Mendelian randomization study. Eur J Clin Invest 2024; 54:e14168. [PMID: 38239089 DOI: 10.1111/eci.14168] [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: 07/19/2023] [Accepted: 01/09/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND In this study we used Mendelian randomization (MR) to investigate the potential causal association of lipoprotein (a) [Lp(a)] levels with pulse wave velocity (PWV). METHODS Genetic variants associated with Lp(a) were retrieved from the UK Biobank GWAS (N = 290,497). A non- overlapping GWAS based on a European cohort (N = 7,000) was used to obtain genetic associations with PWV (outcome) and utilized two different measures for the same trait, brachial-ankle (baPWV) and carotid-femoral (cfPWV) PWV. We applied a two-sample MR using the inverse variance weighting method (IVW) and a series of sensitivity analyses for 170 SNPs that were selected as instrumental variables (IVs). RESULTS Our analyses do not support a causal association between Lp(a) and PWV for neither measurement [βiwv(baPWV) = -.0005, p = .8 and βiwv(cfPWV) = -.006, p = .16]. The above findings were consistent across sensitivity analyses including weighted median, mode-based estimation, MR-Egger regression and MR-PRESSO. CONCLUSION We did not find evidence indicating that Lp(a) is causally associated with PWV, the gold standard marker of arterial stiffness.
Collapse
Affiliation(s)
- Alexandros Simistiras
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
- School of Biomedical Engineering and Imaging Sciences, Kings College London, London, UK
| | - Dimitrios Delialis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Georgios Mavraganis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Ermioni Oikonomou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Eleni Maneta
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Christos Loutos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| |
Collapse
|
4
|
Khan F, Qiu H. Amyloid-β: A potential mediator of aging-related vascular pathologies. Vascul Pharmacol 2023; 152:107213. [PMID: 37625763 DOI: 10.1016/j.vph.2023.107213] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
Aging is one of the most promising risk factors for vascular diseases, however, the precise mechanisms mediating aging-related pathologies are not fully understood. Amyloid beta (Aβ), a peptide produced by the proteolytic processing of amyloid precursor protein (APP), is known as a key mediator of brain damage involved in the pathogenesis of Alzheimer's disease (AD). Recently, it was found that the accumulation of Aβ in the vascular wall is linked to a range of aging-related vascular pathologies, indicating a potential role of Aβ in the pathogenesis of aging-associated vascular diseases. In the present review, we have updated the molecular regulation of Aβ in vascular cells and tissues, summarized the relevance of the Aβ deposition with vascular aging and diseases, and the role of Aβ dysregulation in aging-associated vascular pathologies, including the impaired vascular response, endothelial dysfunction, oxidative stress, and inflammation. This review will provide advanced information in understanding aging-related vascular pathologies and a new avenue to explore therapeutic targets.
Collapse
Affiliation(s)
- Fazlullah Khan
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, The University of Arizona, Phoenix 85004, AZ, USA
| | - Hongyu Qiu
- Translational Cardiovascular Research Center, Department of Internal Medicine, College of Medicine-Phoenix, The University of Arizona, Phoenix 85004, AZ, USA.
| |
Collapse
|
5
|
Świątczak M, Rozwadowska K, Sikorska K, Młodziński K, Świątczak A, Raczak G, Daniłowicz-Szymanowicz L. The potential impact of hereditary hemochromatosis on the heart considering the disease stage and patient age-the role of echocardiography. Front Cardiovasc Med 2023; 10:1202961. [PMID: 37496670 PMCID: PMC10368456 DOI: 10.3389/fcvm.2023.1202961] [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: 04/09/2023] [Accepted: 06/26/2023] [Indexed: 07/28/2023] Open
Abstract
Background Hereditary hemochromatosis (HH) is a genetic disease that leads to increased iron accumulation in several organs. Cardiomyocytes are highly susceptible to this damage owing to their high iron uptake, and cardiovascular complications account for 1/3 of the deaths in the natural course of HH. Additionally, excess iron intake and associated oxidative stress may accelerate the aging of the cardiovascular system, regardless of the age of patients with HH. We aimed to investigate the role of standard and speckle-tracking echocardiography (STE) in revealing heart differences in patients with HH considering the disease stage and the patient age. Methodology Consecutive patients with HH (n = 58) without heart pathologies (except hypertension) and 29 age- and sex-matched healthy individuals underwent echocardiography. Patients were compared according to the time since HH diagnosis (the recently diagnosed HH group [31 patients] with diagnosed HH for less than 6 months and had no more than one venesection; the medium group [11 patients] with diagnosed HH between 6 and 24 months; and the long-lasting group [16 patients] with diagnosed HH for more than 2 years) and the quartile contribution of their age. Results Standard echocardiography revealed differences in diastolic parameters between patients with HH and controls, which were the most prominent between healthy and long-lasting HH patients. Regarding systolic function, left ventricular ejection fraction was lower in HH patients, with the most evident differences between the healthy and recently diagnosed HH patients. STE revealed additional differences in systolic parameters, with LV rotation the worst in recently diagnosed patients and its increase in patients with medium and long-lasting HH. Significantly worse peak systolic longitudinal strain values were observed in all patients with HH. Analyses of the results according to the age quartiles of patients with HH revealed that some changes ocurred earlier than expected according to age. Conclusions Echocardiography can reveal possible heart damage in HH patients at different stages of the disease and highlight potential features of accelerated myocardial aging in these patients.
Collapse
Affiliation(s)
- Michał Świątczak
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Katarzyna Rozwadowska
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Katarzyna Sikorska
- Department of Tropical Medicine and Epidemiology, Faculty of Health Sciences, Medical University of Gdańsk, Gdańsk, Poland
| | - Krzysztof Młodziński
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Agata Świątczak
- Department of Pediatrics, Hematology and Oncology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Grzegorz Raczak
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | | |
Collapse
|
6
|
Wang J, He Y, Zhou D. The role of ubiquitination in microbial infection induced endothelial dysfunction: potential therapeutic targets for sepsis. Expert Opin Ther Targets 2023; 27:827-839. [PMID: 37688775 DOI: 10.1080/14728222.2023.2257888] [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: 05/31/2023] [Revised: 08/11/2023] [Accepted: 09/07/2023] [Indexed: 09/11/2023]
Abstract
INTRODUCTION The ubiquitin system is an evolutionarily conserved and universal means of protein modification that regulates many essential cellular processes. Endothelial dysfunction plays a critical role in the pathophysiology of sepsis and organ failure. However, the mechanisms underlying the ubiquitination-mediated regulation on endothelial dysfunction are not fully understood. AREAS COVERED Here we review the advances in basic and clinical research for relevant papers in PubMed database. We attempt to provide an updated overview of diverse ubiquitination events in endothelial cells, discussing the fundamental role of ubiquitination mediated regulations involving in endothelial dysfunction to provide potential therapeutic targets for sepsis. EXPERT OPINION The central event underlying sepsis syndrome is the overwhelming host inflammatory response to the pathogen infection, leading to endothelial dysfunction. As the key components of the ubiquitin system, E3 ligases are at the center stage of the battle between host and microbial pathogens. Such a variety of ubiquitination regulates a multitude of cellular regulatory processes, including signal transduction, autophagy, inflammasome activation, redox reaction and immune response and so forth. In this review, we discuss the many mechanisms of ubiquitination-mediated regulation with a focus on those that modulate endothelial function to provide potential therapeutic targets for the management of sepsis.
Collapse
Affiliation(s)
- Junshuai Wang
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Yang He
- Department of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Daixing Zhou
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| |
Collapse
|
7
|
Zgutka K, Tkacz M, Tomasiak P, Tarnowski M. A Role for Advanced Glycation End Products in Molecular Ageing. Int J Mol Sci 2023; 24:9881. [PMID: 37373042 PMCID: PMC10298716 DOI: 10.3390/ijms24129881] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Ageing is a composite process that involves numerous changes at the cellular, tissue, organ and whole-body levels. These changes result in decreased functioning of the organism and the development of certain conditions, which ultimately lead to an increased risk of death. Advanced glycation end products (AGEs) are a family of compounds with a diverse chemical nature. They are the products of non-enzymatic reactions between reducing sugars and proteins, lipids or nucleic acids and are synthesised in high amounts in both physiological and pathological conditions. Accumulation of these molecules increases the level of damage to tissue/organs structures (immune elements, connective tissue, brain, pancreatic beta cells, nephrons, and muscles), which consequently triggers the development of age-related diseases, such as diabetes mellitus, neurodegeneration, and cardiovascular and kidney disorders. Irrespective of the role of AGEs in the initiation or progression of chronic disorders, a reduction in their levels would certainly provide health benefits. In this review, we provide an overview of the role of AGEs in these areas. Moreover, we provide examples of lifestyle interventions, such as caloric restriction or physical activities, that may modulate AGE formation and accumulation and help to promote healthy ageing.
Collapse
Affiliation(s)
- Katarzyna Zgutka
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, Żołnierska 54, 70-210 Szczecin, Poland
| | - Marta Tkacz
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, Żołnierska 54, 70-210 Szczecin, Poland
| | - Patrycja Tomasiak
- Institute of Physical Culture Sciences, University of Szczecin, 70-453 Szczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, Żołnierska 54, 70-210 Szczecin, Poland
| |
Collapse
|
8
|
Zhao X, Zhou S, Liu Y, Gong C, Xiang L, Li S, Wang P, Wang Y, Sun L, Zhang Q, Yang Y. Parishin alleviates vascular ageing in mice by upregulation of Klotho. J Cell Mol Med 2023; 27:1398-1409. [PMID: 37032511 PMCID: PMC10183705 DOI: 10.1111/jcmm.17740] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/11/2023] Open
Abstract
Senescence of vascular endothelial cells is the major risk of vascular dysfunction and disease among elderly people. Parishin, which is a phenolic glucoside derived from Gastrodia elata, significantly prolonged yeast lifespan. However, the action of parishin in vascular ageing remains poorly understood. Here, we treated human coronary artery endothelial cells (HCAEC) and naturally aged mice by parishin. Parishin alleviated HCAEC senescence and general age-related features in vascular tissue in naturally aged mice. Network pharmacology approach was applied to determine the compound-target networks of parishin. Our analysis indicated that parishin had a strong binding affinity for Klotho. Expression of Klotho, a protein of age-related declines, was upregulated by parishin in serum and vascular tissue in naturally aged mice. Furthermore, FoxO1, on Klotho/FoxO1 signalling pathway, was increased in the parishin-intervened group, accompanied by the downregulated phosphorylated FoxO1. Taken together, parishin can increase Klotho expression to alleviate vascular endothelial cell senescence and vascular ageing.
Collapse
Affiliation(s)
- Xinxiu Zhao
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Shixian Zhou
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Yang Liu
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Caixia Gong
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Lan Xiang
- College of Pharmaceutical SciencesZhejiang University866 Yu Hang Tang RoadHangzhouChina
| | - Shumin Li
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Peixia Wang
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Yuejun Wang
- Zhejiang Aged Care HospitalHangzhou Normal UniversityHangzhouZhejiangChina
| | - Linlin Sun
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Qin Zhang
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Yunmei Yang
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| |
Collapse
|
9
|
Ajoolabady A, Pratico D, Vinciguerra M, Lip GYH, Franceschi C, Ren J. Inflammaging: mechanisms and role in the cardiac and vasculature. Trends Endocrinol Metab 2023; 34:373-387. [PMID: 37076375 DOI: 10.1016/j.tem.2023.03.005] [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] [Received: 09/28/2022] [Revised: 03/04/2023] [Accepted: 03/16/2023] [Indexed: 04/21/2023]
Abstract
Aging triggers a wide range of cellular and molecular aberrations in the body, giving rise to inflammation and associated diseases. In particular, aging is associated with persistent low-grade inflammation even in absence of inflammatory stimuli, a phenomenon commonly referred to as 'inflammaging'. Accumulating evidence has revealed that inflammaging in vascular and cardiac tissues is associated with the emergence of pathological states such as atherosclerosis and hypertension. In this review we survey molecular and pathological mechanisms of inflammaging in vascular and cardiac aging to identify potential targets, natural therapeutic compounds, and other strategies to suppress inflammaging in the heart and vasculature, as well as in associated diseases such as atherosclerosis and hypertension.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Manlio Vinciguerra
- Liverpool Centre for Cardiovascular Science, Liverpool Johns Moore University, Liverpool, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, UK; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| | - Claudio Franceschi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Institute of Neurological Sciences of Bologna, Bologna, Italy; Department of Applied Mathematics and Laboratory of Systems Biology of Aging, Lobachevsky University, Nizhny Novgorod, Russia.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
10
|
Tan WW, Adler M, Rochow S, Myint PK. An unusual case of neurocardiogenic syncope: A case report and systematic review. J R Coll Physicians Edinb 2023; 53:19-22. [PMID: 36642954 DOI: 10.1177/14782715221147969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
We present a case of syncopal episode in emergency department (ED) and subsequent admission to the geriatric assessment unit. The patient presented with self-limiting central abdominal pain. Given a history of previous aortic aneurysm repair, a contrast CT angiogram was performed. With no evidence of leaking aneurysm, the patient was discharged from the ED. The syncopal episode happened while waiting for a taxi. A review of the earlier CT scan showed the presence of air in the venous circulatory system. In hindsight, it was thought the syncopal episode occurred due to air embolism introduced during or shortly after venous cannulation. We discuss the aetiology of venous air embolism and highlight the lack of evidence regarding tolerable amounts of air in the circulatory system. Physiological changes associated with age may suggest that elderly patients are uniquely maladapted to overcome sudden insults to their cardiovascular status.
Collapse
Affiliation(s)
- Wei Wen Tan
- Department of Medicine for the Elderly, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Maciej Adler
- Department of Medicine for the Elderly, Aberdeen Royal Infirmary, Aberdeen, UK
- Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Stuart Rochow
- Department of Medicine for the Elderly, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Phyo Kyaw Myint
- Department of Medicine for the Elderly, Aberdeen Royal Infirmary, Aberdeen, UK
- Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
11
|
Vijakumaran U, Shanmugam J, Heng JW, Azman SS, Yazid MD, Haizum Abdullah NA, Sulaiman N. Effects of Hydroxytyrosol in Endothelial Functioning: A Comprehensive Review. Molecules 2023; 28:molecules28041861. [PMID: 36838850 PMCID: PMC9966213 DOI: 10.3390/molecules28041861] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Pharmacologists have been emphasizing and applying plant and herbal-based treatments in vascular diseases for decades now. Olives, for example, are a traditional symbol of the Mediterranean diet. Hydroxytyrosol is an olive-derived compound known for its antioxidant and cardioprotective effects. Acknowledging the merit of antioxidants in maintaining endothelial function warrants the application of hydroxytyrosol in endothelial dysfunction salvage and recovery. Endothelial dysfunction (ED) is an impairment of endothelial cells that adversely affects vascular homeostasis. Disturbance in endothelial functioning is a known precursor for atherosclerosis and, subsequently, coronary and peripheral artery disease. However, the effects of hydroxytyrosol on endothelial functioning were not extensively studied, limiting its value either as a nutraceutical supplement or in clinical trials. The action of hydroxytyrosol in endothelial functioning at a cellular and molecular level is gathered and summarized in this review. The favorable effects of hydroxytyrosol in the improvement of endothelial functioning from in vitro and in vivo studies were scrutinized. We conclude that hydroxytyrosol is capable to counteract oxidative stress, inflammation, vascular aging, and arterial stiffness; thus, it is beneficial to preserve endothelial function both in vitro and in vivo. Although not specifically for endothelial dysfunction, hydroxytyrosol safety and efficacy had been demonstrated via in vivo and clinical trials for cardiovascular-related studies.
Collapse
|
12
|
Serna E, Mauricio MD, San-Miguel T, Guerra-Ojeda S, Verdú D, Valls A, Arc-Chagnaud C, De la Rosa A, Viña J. Glucose 6-P Dehydrogenase Overexpression Improves Aging-Induced Endothelial Dysfunction in Aorta from Mice: Role of Arginase II. Int J Mol Sci 2023; 24:ijms24043622. [PMID: 36835034 PMCID: PMC9961129 DOI: 10.3390/ijms24043622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
The increase of vascular arginase activity during aging causes endothelial dysfunction. This enzyme competes with the endothelial nitric oxide synthase (eNOS) for L-arginine substrate. Our hypothesis is that glucose 6-P dehydrogenase (G6PD) overexpression could improve the endothelial function modulating the arginase pathway in aorta from mice. For this study, three groups of male mice were used: young wild type (WT) (6-9 months), old WT (21-22 months) and old G6PD-Tg (21-22 months) mice. Vascular reactivity results showed a reduced acetylcholine-dependent relaxation in the old WT but not old G6PD-Tg group. Endothelial dysfunction was reverted by nor-NOHA, an arginase inhibitor. Mice overexpressing G6PD underexpressed arginase II and also displayed a lower activity of this enzyme. Moreover, histological analyses demonstrated that age causes a thickness of aortic walls, but this did not occur in G6PD-Tg mice. We conclude that the overexpressing G6PD mouse is a model to improve vascular health via the arginase pathway.
Collapse
Affiliation(s)
- Eva Serna
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
- Correspondence:
| | - Maria D Mauricio
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Teresa San-Miguel
- Department of Pathology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Sol Guerra-Ojeda
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - David Verdú
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Alicia Valls
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Coralie Arc-Chagnaud
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Adrián De la Rosa
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - José Viña
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
13
|
Kassis A, Fichot MC, Horcajada MN, Horstman AMH, Duncan P, Bergonzelli G, Preitner N, Zimmermann D, Bosco N, Vidal K, Donato-Capel L. Nutritional and lifestyle management of the aging journey: A narrative review. Front Nutr 2023; 9:1087505. [PMID: 36761987 PMCID: PMC9903079 DOI: 10.3389/fnut.2022.1087505] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/28/2022] [Indexed: 01/25/2023] Open
Abstract
With age, the physiological responses to occasional or regular stressors from a broad range of functions tend to change and adjust at a different pace and restoring these functions in the normal healthy range becomes increasingly challenging. Even if this natural decline is somehow unavoidable, opportunities exist to slow down and attenuate the impact of advancing age on major physiological processes which, when weakened, constitute the hallmarks of aging. This narrative review revisits the current knowledge related to the aging process and its impact on key metabolic functions including immune, digestive, nervous, musculoskeletal, and cardiovascular functions; and revisits insights into the important biological targets that could inspire effective strategies to promote healthy aging.
Collapse
Affiliation(s)
- Amira Kassis
- Whiteboard Nutrition Science, Beaconsfield, QC, Canada,Amira Kassis,
| | | | | | | | - Peter Duncan
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | | | - Nicolas Preitner
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Diane Zimmermann
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Nabil Bosco
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Karine Vidal
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Laurence Donato-Capel
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland,*Correspondence: Laurence Donato-Capel,
| |
Collapse
|
14
|
Zolotaryova NA, Vastyanov RS, Gunenko II. Portable Device use for Arterial Stiffness Determination as a Control Method at the Recovery Stage of Rehabilitation. ACTA BALNEOLOGICA 2022. [DOI: 10.36740/abal202206112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Aim: To create a device for PWPV evaluation which would be cheap and could be easily used not only at the inpatient but also at the restorative outpatient and health-resort stages of rehabilitation.
Materials and Methods: The clinical examination included 90 people who were randomized into the group of observation (n=75) with patients of AH various degrees. 15 practically healthy persons were involved into the control group. PWPV was measured using a specially designed device according to the generally accepted carotid-femoral technique.
Results: Our results showed that the PWPV of healthy persons measured using original device failed to differ statistically from the literature reference values and indicated the correspondence of the measurements we obtained with the data received using traditional apparatus. Out data of original PWPV measurement obtained from examined and treated patients do not differ from the results obtained in wide range of patients aged from 40 to 70 years with the 1st -2nd stages of AH.
Conclusions: We described an efficacy of PWPV determination using an original device. The use of the proposed device does not contradict with the generally accepted method of vascular stiffness measuring and studying. The results obtained on it are comparable with the data of the most used stationary device SphygmoCor for these purposes. The prospects of the device using are without the limitations in patients with vascular diseases at the stage of rehabilitation.
Collapse
|
15
|
Vascular Aging and Damage in Patients with Iron Metabolism Disorders. Diagnostics (Basel) 2022; 12:diagnostics12112817. [PMID: 36428877 PMCID: PMC9689457 DOI: 10.3390/diagnostics12112817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/06/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Vascular aging is a physiological, multifactorial process that involves every type of vessel, from large arteries to microcirculation. This manifests itself as impaired vasomotor function, altered secretory phenotype, deteriorated intercellular transport function, structural remodeling, and aggravated barrier function between the blood and the vascular smooth muscle layer. Iron disorders, particularly iron overload, may lead to oxidative stress and, among other effects, vascular aging. The elevated transferrin saturation and serum iron levels observed in iron overload lead to the formation of a non-transferrin-bound iron (NTBI) fraction with high pro-oxidant activity. NTBI can induce the production of reactive oxygen species (ROS), which induce lipid peroxidation and mediate iron-related damage as the elements of oxidative stress in many tissues, including heart and vessels' mitochondria. However, the available data make it difficult to precisely determine the impact of iron metabolism disorders on vascular aging; therefore, the relationship requires further investigation. Our study aims to present the current state of knowledge on vascular aging in patients with deteriorated iron metabolism.
Collapse
|
16
|
Downregulation of P300/CBP-Associated Factor Protects from Vascular Aging via Nrf2 Signal Pathway Activation. Int J Mol Sci 2022; 23:ijms232012574. [PMID: 36293441 PMCID: PMC9603891 DOI: 10.3390/ijms232012574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 12/14/2022] Open
Abstract
Increasing evidence has shown that vascular aging has a key role in the pathogenesis of vascular diseases. P300/CBP-associated factor (PCAF) is involved in many vascular pathological processes, but the role of PCAF in vascular aging is unknown. This study aims to explore the role and underlying mechanism of PCAF in vascular aging. The results demonstrated that the expression of PCAF was associated with age and aging, and remarkably increased expression of PCAF was present in human atherosclerotic coronary artery. Downregulation of PCAF could reduce angiotensin II (AngII)-induced senescence of rat aortic endothelial cells (ECs) in vitro. In addition, inhibition of PCAF with garcinol alleviated AngII-induced vascular senescence phenotype in mice. Downregulation of PCAF could alleviate AngII-induced oxidative stress injury in ECs and vascular tissue. Moreover, PCAF and nuclear factor erythroid-2-related factor 2 (Nrf2) could interact directly, and downregulation of PCAF alleviated vascular aging by promoting the activation of Nrf2 and enhancing the expression of its downstream anti-aging factors. The silencing of Nrf2 with small interfering RNA attenuated the protective effect of PCAF downregulation from vascular aging. These findings indicate that downregulation of PCAF alleviates oxidative stress by activating the Nrf2 signaling pathway and ultimately inhibits vascular aging. Thus, PCAF may be a promising target for aging-related cardiovascular disease.
Collapse
|
17
|
RNA modifications in aging-associated cardiovascular diseases. Aging (Albany NY) 2022; 14:8110-8136. [PMID: 36178367 PMCID: PMC9596201 DOI: 10.18632/aging.204311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 09/17/2022] [Indexed: 11/25/2022]
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide that bears an enormous healthcare burden and aging is a major contributing factor to CVDs. Functional gene expression network during aging is regulated by mRNAs transcriptionally and by non-coding RNAs epi-transcriptionally. RNA modifications alter the stability and function of both mRNAs and non-coding RNAs and are involved in differentiation, development, and diseases. Here we review major chemical RNA modifications on mRNAs and non-coding RNAs, including N6-adenosine methylation, N1-adenosine methylation, 5-methylcytidine, pseudouridylation, 2′ -O-ribose-methylation, and N7-methylguanosine, in the aging process with an emphasis on cardiovascular aging. We also summarize the currently available methods to detect RNA modifications and the bioinformatic tools to study RNA modifications. More importantly, we discussed the specific implication of the RNA modifications on mRNAs and non-coding RNAs in the pathogenesis of aging-associated CVDs, including atherosclerosis, hypertension, coronary heart diseases, congestive heart failure, atrial fibrillation, peripheral artery disease, venous insufficiency, and stroke.
Collapse
|
18
|
Gkaliagkousi E, Lazaridis A, Dogan S, Fraenkel E, Tuna BG, Mozos I, Vukicevic M, Yalcin O, Gopcevic K. Theories and Molecular Basis of Vascular Aging: A Review of the Literature from VascAgeNet Group on Pathophysiological Mechanisms of Vascular Aging. Int J Mol Sci 2022; 23:ijms23158672. [PMID: 35955804 PMCID: PMC9368987 DOI: 10.3390/ijms23158672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Vascular aging, characterized by structural and functional alterations of the vascular wall, is a hallmark of aging and is tightly related to the development of cardiovascular mortality and age-associated vascular pathologies. Over the last years, extensive and ongoing research has highlighted several sophisticated molecular mechanisms that are involved in the pathophysiology of vascular aging. A more thorough understanding of these mechanisms could help to provide a new insight into the complex biology of this non-reversible vascular process and direct future interventions to improve longevity. In this review, we discuss the role of the most important molecular pathways involved in vascular ageing including oxidative stress, vascular inflammation, extracellular matrix metalloproteinases activity, epigenetic regulation, telomere shortening, senescence and autophagy.
Collapse
Affiliation(s)
- Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Faculty of Medicine, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece
- Correspondence: (E.G.); (K.G.)
| | - Antonios Lazaridis
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Faculty of Medicine, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, 34755 Istanbul, Turkey
| | - Emil Fraenkel
- 1st Department of Internal Medicine, University Hospital, Pavol Jozef Šafárik University of Košice, Trieda SNP 1, 04066 Košice, Slovakia
| | - Bilge Guvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, 34755 Istanbul, Turkey
| | - Ioana Mozos
- Department of Functional Sciences-Pathophysiology, Center for Translational Research and Systems Medicine, “Victor Babes” University of Medicine and Pharmacy, 300173 Timisoara, Romania
| | - Milica Vukicevic
- Cardiac Surgery Clinic, Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Ozlem Yalcin
- Department of Physiology, School of Medicine, Koc University, 34450 Istanbul, Turkey
| | - Kristina Gopcevic
- Laboratory for Analytics of Biomolecules, Department of Chemistry in Medicine, Faculty of Medicine, 11000 Belgrade, Serbia
- Correspondence: (E.G.); (K.G.)
| |
Collapse
|
19
|
Xiao XT, He SQ, Wu NN, Lin XC, Zhao J, Tian C. Green Tea Polyphenols Prevent Early Vascular Aging Induced by High-Fat Diet via Promoting Autophagy in Young Adult Rats. Curr Med Sci 2022; 42:981-990. [PMID: 35896932 DOI: 10.1007/s11596-022-2604-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/03/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Epidemiology studies indicate that green tea polyphenols (GTP) perform a protective effect on cardiovascular diseases, but the underlying mechanisms are complex. The present study aimed to investigate the effect of GTP on high-fat diets (HFD) induced-early vascular aging. METHODS Six-week young adult Wistar rats were fed with standard chow or HFD in the presence and absence of GTP (200 mg/kg body weight) for 18 weeks. In vitro experiment, human umbilical vascular endothelial cells (HUVECs) were treated with palmitic acid (PA) and GTP. RESULTS The results showed that GTP alleviated the disorganized arterial wall and the increased intima-media thickness induced by HFD. In addition, the vascular oxidative injury was suppressed following GTP treatment. Furthermore, GTP elevated the ratio of LC3-II/LC3-I and suppressed expression of p62/SQSTM1, and restored SIRT3 expression in the aorta of HFD rats. Consistently, in cultured HUVECs, GTP inhibited cell senescence indicated by SA-β-gal and promoted endothelial autophagy compared with the PA treatment group. The activity of SIRT3 was specifically inhibited by 3-TYP, and the protective effect of GTP was consequently abolished. CONCLUSION The findings indicated that GTP protected against early vascular senescence in young HFD rats via ameliorating oxidative injury and promoting autophagy which was partially regulated by the SIRT3 pathway.
Collapse
Affiliation(s)
- Xiang-Tian Xiao
- Medical College of Wuhan University of Science and Technology, Wuhan, 430081, China
| | - Shui-Qing He
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Nan-Nan Wu
- School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, 400016, China
| | - Xue-Chun Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Zhao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chong Tian
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
20
|
Dela Justina V, Miguez JSG, Priviero F, Sullivan JC, Giachini FR, Webb RC. Sex Differences in Molecular Mechanisms of Cardiovascular Aging. FRONTIERS IN AGING 2022; 2:725884. [PMID: 35822017 PMCID: PMC9261391 DOI: 10.3389/fragi.2021.725884] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD) is still the leading cause of illness and death in the Western world. Cardiovascular aging is a progressive modification occurring in cardiac and vascular morphology and physiology where increased endothelial dysfunction and arterial stiffness are observed, generally accompanied by increased systolic blood pressure and augmented pulse pressure. The effects of biological sex on cardiovascular pathophysiology have long been known. The incidence of hypertension is higher in men, and it increases in postmenopausal women. Premenopausal women are protected from CVD compared with age-matched men and this protective effect is lost with menopause, suggesting that sex-hormones influence blood pressure regulation. In parallel, the heart progressively remodels over the course of life and the pattern of cardiac remodeling also differs between the sexes. Lower autonomic tone, reduced baroreceptor response, and greater vascular function are observed in premenopausal women than men of similar age. However, postmenopausal women have stiffer arteries than their male counterparts. The biological mechanisms responsible for sex-related differences observed in cardiovascular aging are being unraveled over the last several decades. This review focuses on molecular mechanisms underlying the sex-differences of CVD in aging.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | - Fernanda Priviero
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Fernanda R Giachini
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil.,Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - R Clinton Webb
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
21
|
Image-free ultrasound for local and regional vascular stiffness assessment: the ARTSENS Plus. J Hypertens 2022; 40:1537-1544. [PMID: 35730407 DOI: 10.1097/hjh.0000000000003181] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The combined assessment of vascular health markers is crucial for identifying the cumulative burden of vascular risk factors early on, as well as the extent of vascular aging for effective prediction of future cardiovascular events. This work addresses the need for a currently nonexistent device or system that facilitates such combined assessment in clinical practice and large-scale screening settings. We report an image-free ultrasound device - ARTSENS Plus - developed for the measurement of local and regional arterial stiffness, central and peripheral blood pressure (BP), and vessel dimensions, all in one examination. METHODS A preclinical study on 90 asymptomatic individuals verified the device's functionality under ARTERY Society guidelines. The device's accuracy of stiffness measures was validated against the reference measures. RESULTS The interoperator and intraoperator variability was less than 7%. Carotid artery's lumen diameter and local stiffness indices and carotid-femoral regional pulse wave velocity showed excellent agreement with the references (absolute errors were less than 4.1, 9, and 4.1%, respectively). The carotid SBP was 10.02% lower than that of the brachial artery, as expected. CONCLUSION The study demonstrated the device's ability to perform an effortless and reliable evaluation of the local and regional vascular stiffness and central BP with an accuracy that meets clinical standards.
Collapse
|
22
|
Stamatelopoulos K, Delialis D, Georgiopoulos G, Tselegkidi MI, Theodorakakou F, Dialoupi I, Bambatsias D, Petropoulos I, Vergaro G, Ikonomidis I, Tzortzis S, Briasoulis A, Kanakakis J, Trougakos I, Dimopoulos MA, Kastritis E. Determining patterns of vascular function and structure in wild-type transthyretin cardiac amyloidosis. A comparative study. Int J Cardiol 2022; 363:102-110. [PMID: 35716935 DOI: 10.1016/j.ijcard.2022.06.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/26/2022] [Accepted: 06/10/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND The impact of wild-type transthyretin-related cardiac amyloidosis (ATTRwt) on functional and structural peripheral vascular measures is unknown. In the present study, we explored patterns of vascular dysfunction in patients with ATTRwt in comparison to diseases with similar cardiac phenotype. METHODS Treatment-naïve patients with ATTRwt (n = 32) were compared to: 1. Age-and sex-matched reference population without amyloidosis (n = 32), 2. Age-and sex-matched patients with systemic AL amyloidosis (n = 32), and 3. patients with cardiac AL amyloidosis (AL-HF, n = 23) or elderly patients with heart failure with preserved ejection fraction (HFpEF) (n = 16). All subjects underwent peripheral vascular assessment using carotid artery ultrasonography, brachial artery flow-mediated dilation (FMD), measurement of arterial stiffness and aortic hemodynamics including heart rate-adjusted time of return of reflected waves (Tr/HR). RESULTS After adjustment for traditional cardiovascular risk factors and coronary artery disease (core model), peripheral and aortic blood pressures (BP) were lower in patients with ATTRwt (p < 0.05) whereas other vascular markers were preserved compared to the reference non-amyloidosis group. ATTRwt was independently associated with lower BP and longer Tr/HR compared to AL. Compared to AL-HF, FMD was lower in ATTRwt (p = 0.033). ATTRwt patients had lower BP and higher Tr/HR than HFpEF (p < 0.05). By ROC analysis, Tr/HR discriminated ATTRwt vs. AL-HF (sensitivity 93%, specificity 75%) and HFpEF (sensitivity 100%, specificity 94%) and lower FMD increased the likelihood for ATTRwt at low Tr/HR values. CONCLUSION ATTRwt patients present a distinct peripheral vascular fingerprint which is different from AL-HF or HFpEF, consisting of lower peripheral and aortic BP, prolonged Tr/HR and FMD at reference-population range.
Collapse
Affiliation(s)
- Kimon Stamatelopoulos
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece..
| | - Dimitrios Delialis
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece.; Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Maria-Irini Tselegkidi
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Foteini Theodorakakou
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Ioanna Dialoupi
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Dimitrios Bambatsias
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Ioannis Petropoulos
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Giuseppe Vergaro
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Ignatios Ikonomidis
- 2(nd) Department of Cardiology, School of Medicine of the National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Stavros Tzortzis
- 2(nd) Department of Cardiology, School of Medicine of the National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Alexandros Briasoulis
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - John Kanakakis
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Ioannis Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, University of Athens Medical School, Athens, Greece.; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy..
| |
Collapse
|
23
|
Milusev A, Rieben R, Sorvillo N. The Endothelial Glycocalyx: A Possible Therapeutic Target in Cardiovascular Disorders. Front Cardiovasc Med 2022; 9:897087. [PMID: 35647072 PMCID: PMC9136230 DOI: 10.3389/fcvm.2022.897087] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/21/2022] [Indexed: 12/15/2022] Open
Abstract
The physiological, anti-inflammatory, and anti-coagulant properties of endothelial cells (ECs) rely on a complex carbohydrate-rich layer covering the luminal surface of ECs, called the glycocalyx. In a range of cardiovascular disorders, glycocalyx shedding causes endothelial dysfunction and inflammation, underscoring the importance of glycocalyx preservation to avoid disease initiation and progression. In this review we discuss the physiological functions of the glycocalyx with particular focus on how loss of endothelial glycocalyx integrity is linked to cardiovascular risk factors, like hypertension, aging, diabetes and obesity, and contributes to the development of thrombo-inflammatory conditions. Finally, we consider the role of glycocalyx components in regulating inflammatory responses and discuss possible therapeutic interventions aiming at preserving or restoring the endothelial glycocalyx and therefore protecting against cardiovascular disease.
Collapse
Affiliation(s)
- Anastasia Milusev
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Robert Rieben
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Nicoletta Sorvillo
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- *Correspondence: Nicoletta Sorvillo
| |
Collapse
|
24
|
Barcena ML, Aslam M, Pozdniakova S, Norman K, Ladilov Y. Cardiovascular Inflammaging: Mechanisms and Translational Aspects. Cells 2022; 11:cells11061010. [PMID: 35326461 PMCID: PMC8946971 DOI: 10.3390/cells11061010] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/07/2022] [Accepted: 03/15/2022] [Indexed: 12/14/2022] Open
Abstract
Aging is one of the major non-reversible risk factors for several chronic diseases, including cancer, type 2 diabetes, dementia, and cardiovascular diseases (CVD), and it is a key cause of multimorbidity, disability, and frailty (decreased physical activity, fatigue, and weight loss). The underlying cellular mechanisms are complex and consist of multifactorial processes, such as telomere shortening, chronic low-grade inflammation, oxidative stress, mitochondrial dysfunction, accumulation of senescent cells, and reduced autophagy. In this review, we focused on the molecular mechanisms and translational aspects of cardiovascular aging-related inflammation, i.e., inflammaging.
Collapse
Affiliation(s)
- Maria Luisa Barcena
- Department of Geriatrics and Medical Gerontology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (S.P.); (K.N.); (Y.L.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-525-359
| | - Muhammad Aslam
- Experimental Cardiology, Department of Internal Medicine I, Justus Liebig University, Aulweg 129, 35392 Giessen, Germany;
- Department of Cardiology, Kerckhoff Clinic GmbH, 61231 Bad Nauheim, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Rhein-Main, 61231 Bad Nauheim, Germany
| | - Sofya Pozdniakova
- Department of Geriatrics and Medical Gerontology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (S.P.); (K.N.); (Y.L.)
- Barcelona Biomedical Research Park (PRBB), Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003 Barcelona, Spain
| | - Kristina Norman
- Department of Geriatrics and Medical Gerontology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (S.P.); (K.N.); (Y.L.)
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- Department of Nutrition & Gerontology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Yury Ladilov
- Department of Geriatrics and Medical Gerontology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (S.P.); (K.N.); (Y.L.)
- Department of Cardiovascular Surgery, Heart Center Brandenburg, Brandenburg Medical School Theodor Fontane, University Hospital, Ladeburger Str. 17, 16321 Bernau, Germany
| |
Collapse
|
25
|
Evsevyeva MY, Eremin MV, Rostovtseva MV, Sergeeva OV, Rusidi AV, Kudryavtseva VD, Shchetinin EV. Preventive Screening of Young People from the Perspective of Vascular Aging Phenotypes: the Role of Body Weight. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2022. [DOI: 10.20996/1819-6446-2022-02-14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aim. To study the cardiovascular stiffness index CAVI in relation to main risk factors (RF) in young people from the point of their vascular aging phenotype and gender.Material and methods. 264 young people (93 boys and 171 girls) at age of 18 to 25 years were examined. Vascular screening was performed with evaluating of CAVI. All subjects were divided into tertile groups for this indicator, taking into account gender. Such interval analysis of gender- and age- homogeneous sample allows to identify carriers of the phenotypes of premature (upper CAVI-tercile), normal (middle CAVI-tercile), and favorable or healthy (low CAVI-tercile) vascular aging. The first of them is also called EVA (early vascular aging) syndrome. In these groups, the incidence of carriers of insufficient, normal body mass (BM) and overweight were estimated. In three groups formed by BM indicator, the parameters of CAVI, hemodynamic and metabolic status in response to changes in BM were determined.Results. Among of the presented RF, the correlation was significant for CAVI with a weight -0.428 (p<0.001) for R-CAVI, -0.453 (p<0.001) for L- CAVI, and even more significant with the BM index - at the level of -0.410 (p<0.001) for R-CAVI and -0.462 (p<0.001) for L-CAVI. This connection is found in boys. Among boys and girls with excessive BM, the most favorable vascular aging phenotype is observed 2.7 and 2.2 times more often than the EVA syndrome. In contrast, among girls with insufficient BM, a favorable vascular phenotype is detected almost twice rarely as compared with EVA syndrome. In similar group of boys, a healthy vascular phenotype is not observed at all. Among normal-weight boys and girls, the occurrence of the two extreme vascular phenotypes is almost identical. The arterial rigidity decrease on the background of BM increase occurs despite the сhanges of hemo- dynamic and metabolic indicators. This changes are especially pronounced in girls. The difference between the extreme weight categories in terms of R- CAVI and L-CAVI for boys was 1.4, while for girls it was only 0.3 for R-CAVI and 0.4 for L-CAVI.Conclusion. When carrying out preventive measures among young people, one should not limit oneself to evaluating only traditional RF. Based on the concept of vascular aging, angiological screening should be more widely implemented. It will allow for more individualized preventive interventions among young people in the future.
Collapse
|
26
|
Physical Performance, Cardiovascular Health and Psychosocial Wellbeing in Older Adults Compared to Oldest-Old Residential Seniors. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19031451. [PMID: 35162467 PMCID: PMC8835371 DOI: 10.3390/ijerph19031451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023]
Abstract
Background: This study analyzed physical, cardiovascular, and psychosocial health in different age groups at the far end of the lifespan. Methods: Sixty-two residential seniors participated in this cross-sectional study and were assigned according to age to either the older adults (n = 27; age: 74.8 (3.6); f: 23) or the oldest-old group (n = 35; age: 87.2 (5.0); f: 28). Gait speed, functional mobility, handgrip strength, and pulse wave velocity (PWV) were measured. Additionally, questionnaires to assess quality of life were applied. Mean between-group differences (Δ) and Hedge’s g with 95 % confidence intervals were calculated. Results: Oldest-old had moderately lower handgrip strength (Δ = −31.3 N, 95% CI [−66.30; −1.65], Hedge’s g = 0.49 [−0.97; 0.03]) and relevant lower gait speed than the older adults (Δ = −0.11 m/s [−0.28; 0.05], g = 0.34 [−0.89; 0.20]). All other physical parameters showed trivial differences. Very large effects were found in PWV in favor of the older adults (Δ = −2.65 m/s [−3.26; −2.04], g = −2.14 [−2.81; −1.36]). The questionnaires showed trivial to small differences. Conclusion: We found small differences in physical as well as psychosocial health between age groups with large inter-individual variance. Large differences were found in arterial stiffness, which increases with age. Exercise programs in nursing homes should consider physical, psychosocial, and cardiovascular variables more than age.
Collapse
|
27
|
Bogatyreva FM, Kaplunova VRY, Kozhevnikova MV, Shakaryants GA, Khabarova NV, Privalova EV, Belenkov YN. [Assessment of the structural and functional state of blood vessels in patients with hypertrophic cardiomyopathy]. KARDIOLOGIIA 2021; 61:16-21. [PMID: 35057717 DOI: 10.18087/cardio.2021.12.n1718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/07/2021] [Indexed: 06/14/2023]
Abstract
Aim To evaluate the structural and functional condition of the vasculature using fingertip photoplethysmography and computerized videocapillaroscopy in patients with hypertrophic cardiomyopathy (HCMP).Material and methods The study included patients with HCMP (n=48; 28 (57 %) men; age, 54.3±13.6 years) and healthy volunteers (control group, n=33, 15 (45 %) men; age, 58.2±8.8 years). Standard laboratory and instrumental examination (blood count and biochemistry, electrocardiography, echocardiography, Holter electrocardiogram monitoring) were performed for all HCMP patients. The condition of vascular wall at various levels of the vasculature was evaluated by fingertip photoplethysmography (apparatus Angioscan-01) and computerized nail-fold videocapillaroscopy (apparatus Capillaroscan-01). The photoplethysmography study analyzed structural parameters, including the arterial wall stiffness index (aSI) of large blood vessels and the resistance index (RI) of small muscular arteries. Endothelial dysfunction was evaluated by the occlusion index (OI) and phase shift (PS). The capillaroscopy study assessed structural parameters, including the resting capillary density (rCD) and the capillary density following venous occlusion (voCD), and functional parameters, including the percentage of perfused capillaries (PPC), the percentage of restored capillaries (PRC), and the capillary density after the reactive hyperemia test (rhCD).Results The study showed increases in aSI (8.8 [6.8; 12.2] and RI (32.5 [17.4; 47.9] in the HCMP group. The OI was significantly lower in the HCMP group (1.3 [1.1; 1.5]) than in the control group (1.8 [1.5; 2.7], р<0.001). Also, PS values were significantly decreased in the HCMP group (4.4 [2.3; 8.6]) compared to the control group (8.4 [5.1; 12.1]. p=0.018). Disorders of structural and functional capillary indexes were observed in HCMP patients compared to the control group; rCD and voCD were decreased in the HCMP group (60 [52.6; 68] and 88 [75; 90], respectively) compared to the control group (75.8 [60; 87] and 90 [73; 101]), however, no intergroup difference reached a statistical significance. The rhCD, PPC, and PRC values were decreased in the HCMP group (66.3 [55; 72], 86.7 [70.9; 104.2] and 1.7 [-6.95; 20.3], respectively) compared to the control group (86 [68.6; 100], 103 [96; 114] and 18.4 [8.1; 27.4], respectively); PPC and PRC values were significantly different (р<0.005 and p<0.004, respectively).Conclusion In patients with HCMP, fingertip photoplethysmography and computerized videocapillaroscopy showed increased wall stiffness in both large blood vessels and microvasculature, pronounced endothelial dysfunction, and decreases in capillary density and percentage of restored capillaries following respective tests.
Collapse
Affiliation(s)
- F M Bogatyreva
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - V Ra Yu Kaplunova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - M V Kozhevnikova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - G A Shakaryants
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - N V Khabarova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - E V Privalova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - Yu N Belenkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| |
Collapse
|
28
|
Ministrini S, Puspitasari YM, Beer G, Liberale L, Montecucco F, Camici GG. Sirtuin 1 in Endothelial Dysfunction and Cardiovascular Aging. Front Physiol 2021; 12:733696. [PMID: 34690807 PMCID: PMC8527036 DOI: 10.3389/fphys.2021.733696] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/30/2021] [Indexed: 01/10/2023] Open
Abstract
Sirtuin 1 (SIRT1) is a histone deacetylase belonging to the family of Sirtuins, a class of nicotinamide adenine dinucleotide (NAD+)-dependent enzymes with multiple metabolic functions. SIRT1 localizes in the nucleus and cytoplasm, and is implicated in the regulation of cell survival in response to several stimuli, including metabolic ones. The expression of SIRT1 is associated with lifespan and is reduced with aging both in animal models and in humans, where the lack of SIRT1 is regarded as a potential mediator of age-related cardiovascular diseases. In this review, we will summarize the extensive evidence linking SIRT1 functional and quantitative defects to cellular senescence and aging, with particular regard to their role in determining endothelial dysfunction and consequent cardiovascular diseases. Ultimately, we outline the translational perspectives for this topic, in order to highlight the missing evidence and the future research steps.
Collapse
Affiliation(s)
- Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland.,Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Georgia Beer
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland.,First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,Istituto di Ricerca e Cura a Carattere Scientifico Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
29
|
Koutsaliaris IK, Moschonas IC, Pechlivani LM, Tsouka AN, Tselepis AD. Inflammation, Oxidative Stress, Vascular Aging And Atherosclerotic Ischemic Stroke. Curr Med Chem 2021; 29:5496-5509. [PMID: 34547993 DOI: 10.2174/0929867328666210921161711] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 11/22/2022]
Abstract
Vascular aging is a crucial risk factor for atherosclerotic ischemic stroke. Vascular aging is characterized by oxidative stress, endothelial dysfunction, inflammation, intimal and media thickening, as well as the gradual development of arterial stiffness, among other pathophysiological features. Regarding oxidative stress, increased concentration of reactive oxygen and nitrogen species is linked to atherosclerotic ischemic stroke in vascular aging. Additionally, oxidative stress is associated with an inflammatory response. Inflammation is related to aging through the "inflammaging" theory, which is characterized by decreased ability to cope with a variety of stressors, in combination with an increased pro-inflammatory state. Vascular aging is correlated with changes in cerebral arteries that are considered predictors of the risk for atherosclerotic ischemic stroke. The aim of the present review is to present the role of oxidative stress and inflammation in vascular aging, as well as their involvement in atherosclerotic ischemic stroke.
Collapse
Affiliation(s)
- Ioannis K Koutsaliaris
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 45110, Ioannina. Greece
| | - Iraklis C Moschonas
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 45110, Ioannina. Greece
| | - Louisa M Pechlivani
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 45110, Ioannina. Greece
| | - Aikaterini N Tsouka
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 45110, Ioannina. Greece
| | - Alexandros D Tselepis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 45110, Ioannina. Greece
| |
Collapse
|
30
|
Dutka M, Bobiński R, Wojakowski W, Francuz T, Pająk C, Zimmer K. Osteoprotegerin and RANKL-RANK-OPG-TRAIL signalling axis in heart failure and other cardiovascular diseases. Heart Fail Rev 2021; 27:1395-1411. [PMID: 34313900 PMCID: PMC9197867 DOI: 10.1007/s10741-021-10153-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/16/2021] [Indexed: 01/29/2023]
Abstract
Osteoprotegerin (OPG) is a glycoprotein involved in the regulation of bone remodelling. OPG regulates osteoclast activity by blocking the interaction between the receptor activator of nuclear factor kappa B (RANK) and its ligand (RANKL). More and more studies confirm the relationship between OPG and cardiovascular diseases. Numerous studies have confirmed that a high plasma concentration of OPG and a low concentration of tumour necrosis factor–related apoptosis inducing ligand (TRAIL) together with a high OPG/TRAIL ratio are predictors of poor prognosis in patients with myocardial infarction. A high plasma OPG concentration and a high ratio of OPG/TRAIL in the acute myocardial infarction are a prognostic indicator of adverse left ventricular remodelling and of the development of heart failure. Ever more data indicates the participation of OPG in the regulation of the function of vascular endothelial cells and the initiation of the atherosclerotic process in the arteries. Additionally, it has been shown that TRAIL has a protective effect on blood vessels and exerts an anti-atherosclerotic effect. The mechanisms of action of both OPG and TRAIL within the cells of the vascular wall are complex and remain largely unclear. However, these mechanisms of action as well as their interaction in the local vascular environment are of great interest to researchers. This article presents the current state of knowledge on the mechanisms of action of OPG and TRAIL in the circulatory system and their role in cardiovascular diseases. Understanding these mechanisms may allow their use as a therapeutic target in cardiovascular diseases in the future.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland.
| | - Rafał Bobiński
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Wojciech Wojakowski
- Department of Cardiology and Structural Heart Disease, Medical University of Silesia, Katowice, Poland
| | - Tomasz Francuz
- Department of Biochemistry, Medical University of Silesia, Katowice, Poland
| | - Celina Pająk
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Karolina Zimmer
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| |
Collapse
|
31
|
Piotrowicz K, Gryglewska B, Grodzicki T, Gąsowski J. Arterial stiffness and frailty - A systematic review and metaanalysis. Exp Gerontol 2021; 153:111480. [PMID: 34265411 DOI: 10.1016/j.exger.2021.111480] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022]
Abstract
Frailty and cardiovascular disease share some of the pathophysiologic features. Our objective was to review and metaanalyse the available published evidence on the topic. We performed a comprehensive literature search for studies where pulse wave velocity (PWV) or carotid-ankle vascular index (CAVI) has been linked with frailty in older persons. Of the initial 362 abstracts, after the application of the PRISMA approach, 5 were analysed in detail. We calculated within-study and pooled standardised mean differences of aortic stiffness measures between frail and non-frail (0.62 [0.31-0.92], p < 0.0001, I2 = 88%), and pre-frail and non-frail (0.32 [0.14-0.51], p = 0.0006, I2 = 72%) groups. In two studies it was possible to extract directly or calculate based on published data the odds ratios for the concomitant frailty, associated in one case with CAVI greater by 1 m/s and in another with cfPWV >13 m/s, indicating greater probability of concomitant frailty given greater aortic stiffness. Across the studies, the prevalence of hypertension, diabetes mellitus, hyperlipidaemia, and smoking tended to increase from non-frail, to pre-frail, and frail groups, presenting a possibility of important confounding, but also a common pathophysiology. In conclusion, the pooled analysis of the published cross-sectional study results indicates a relation between aortic stiffness and frailty in older subjects. However, well designed prospective studies are needed to answer the questions of causality.
Collapse
Affiliation(s)
- Karolina Piotrowicz
- Jagiellonian University Medical College, Department of Internal Medicine and Gerontology, Kraków, Poland
| | - Barbara Gryglewska
- Jagiellonian University Medical College, Department of Internal Medicine and Gerontology, Kraków, Poland
| | - Tomasz Grodzicki
- Jagiellonian University Medical College, Department of Internal Medicine and Gerontology, Kraków, Poland
| | - Jerzy Gąsowski
- Jagiellonian University Medical College, Department of Internal Medicine and Gerontology, Kraków, Poland.
| |
Collapse
|
32
|
Xu H, Li S, Liu YS. Roles and Mechanisms of DNA Methylation in Vascular Aging and Related Diseases. Front Cell Dev Biol 2021; 9:699374. [PMID: 34262910 PMCID: PMC8273304 DOI: 10.3389/fcell.2021.699374] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/07/2021] [Indexed: 12/20/2022] Open
Abstract
Vascular aging is a pivotal risk factor promoting vascular dysfunction, the development and progression of vascular aging-related diseases. The structure and function of endothelial cells (ECs), vascular smooth muscle cells (VSMCs), fibroblasts, and macrophages are disrupted during the aging process, causing vascular cell senescence as well as vascular dysfunction. DNA methylation, an epigenetic mechanism, involves the alteration of gene transcription without changing the DNA sequence. It is a dynamically reversible process modulated by methyltransferases and demethyltransferases. Emerging evidence reveals that DNA methylation is implicated in the vascular aging process and plays a central role in regulating vascular aging-related diseases. In this review, we seek to clarify the mechanisms of DNA methylation in modulating ECs, VSMCs, fibroblasts, and macrophages functions and primarily focus on the connection between DNA methylation and vascular aging-related diseases. Therefore, we represent many vascular aging-related genes which are modulated by DNA methylation. Besides, we concentrate on the potential clinical application of DNA methylation to serve as a reliable diagnostic tool and DNA methylation-based therapeutic drugs for vascular aging-related diseases.
Collapse
Affiliation(s)
- Hui Xu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-Related Disease Research, Central South University, Changsha, China
| | - Shuang Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-Related Disease Research, Central South University, Changsha, China
| | - You-Shuo Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-Related Disease Research, Central South University, Changsha, China
| |
Collapse
|
33
|
Blöchl M, Schaare HL, Kunzmann U, Nestler S. The Age-Dependent Association Between Vascular Risk Factors and Depressed Mood. J Gerontol B Psychol Sci Soc Sci 2021; 77:284-294. [PMID: 34080633 DOI: 10.1093/geronb/gbab063] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Cumulative burden of vascular risk factors (VRFs) has been linked to an increased risk of depressed mood. However, the role of age in this association is still unclear. Here, we investigated whether VRF burden is associated with levels and changes in depressed mood and whether these associations become stronger or weaker from mid- to later life. METHOD We used longitudinal data from 5,689 participants (52-89 years) of the English Longitudinal Study of Ageing. A composite score incorporated the presence of 5 VRFs: hypertension, diabetes, smoking, obesity, and hypercholesterolemia. Second-order latent growth models were used to test whether levels and changes of depressed mood differed as a function of baseline VRF burden, and whether these associations were moderated by age. RESULTS Baseline VRF burden showed a small association with higher levels of depressed mood (estimate = 0.081; 95% CI: 0.024, 0.138, p = .005). This association varied with age, such that it was stronger in midlife compared to later life (estimate = -0.007; 95% CI: -0.013, -0.002, p = .017). There was no evidence that VRF burden was associated with changes in depressed mood. DISCUSSION Our findings suggest that VRF burden in midlife, but less so in later life, predicts individual differences in depressed mood. These findings are consistent with reports on the importance of midlife VRFs and support the idea that promotion of vascular health in this age group or earlier in life may be critical to maintain mental health across adulthood.
Collapse
Affiliation(s)
- Maria Blöchl
- Department of Psychology, University of Münster, Germany
- Department for Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - H Lina Schaare
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour), Research Centre Jülich, Germany
| | - Ute Kunzmann
- Department of Psychology, Leipzig University, Germany
| | | |
Collapse
|
34
|
Khosravipour M, Rajati F. Sensorineural hearing loss and risk of stroke: a systematic review and meta-analysis. Sci Rep 2021; 11:11021. [PMID: 34040004 PMCID: PMC8155183 DOI: 10.1038/s41598-021-89695-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 04/29/2021] [Indexed: 02/04/2023] Open
Abstract
The aim of this systematic review and meta-analysis study was to clarify the effects of sensorineural hearing loss (SNHL) on the incidence of stroke. In line with this, PubMed, Scopus, Web of Science, and ScienceDirect databases were searched using related keywords and MeSH terms from inception to March 1, 2020. Out of the 1961 initial records, eight cohort studies comprising 4,564,202 participants were included, and their qualities were assessed using the Newcastle-Ottawa Scale (NOS). Then, the random-effects model was used to pool HR (95% CI) for risk of stroke; and heterogeneity was presented with I2 index. Subgroup analysis and publication bias tests were performed, and the pooled HR (95% CI) of stroke in SNHL was estimated as 1.31 (1.08, 1.53) for the unadjusted model and 1.33 (1.18, 1.49) for the adjusted model. Subgroup analysis indicates a significantly higher risk of stroke in patients with sudden SNHL (SSNHL) in comparison to age-related HL (ARHL) both in the unadjusted model, [HR = 1.46; 95% CI (1.08, 1.63)] versus [HR = 1.14; 95% CI (0.64, 1.65)], and in the adjusted model, [HR = 1.44; 95% CI (1.15, 1.74)] versus [HR = 1.29; 95% CI (1.24, 1.34)]. Our study showed that patients with SNHL face a higher risk of stroke than those without SNHL. It is necessary to perform hematologic and neurological examinations to help clinicians detect patients who are potentially at risk for stroke.
Collapse
Affiliation(s)
- Masoud Khosravipour
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Rajati
- Research Center for Environmental Determinants of Health, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
35
|
Abstract
The endothelium is a crucial regulator of vascular homeostasis by controlling barrier integrity as well acting as an important signal transducer, thereby illustrating that endothelial cells are not inert cells. In the context of atherosclerosis, this barrier function is impaired and endothelial cells become activated, resulting in the upregulation of adhesion molecules, secretion of cytokines and chemokines and internalization of integrins. Finally, this leads to increased vessel permeability, thereby facilitating leukocyte extravasation as well as fostering a pro-inflammatory environment. Additionally, activated endothelial cells can form migrating tip cells and proliferative stalk cells, resulting in the formation of new blood vessels. Emerging evidence has accumulated indicating that cellular metabolism is crucial in fueling these pro-atherosclerotic processes, including neovascularization and inflammation, thereby contributing to plaque progression and altering plaque stability. Therefore, further research is necessary to unravel the complex mechanisms underlying endothelial cell metabolic changes, and exploit this knowledge for finding and developing potential future therapeutic strategies. In this review we discuss the metabolic alterations endothelial cells undergo in the context of inflammation and atherosclerosis and how this relates to changes in endothelial functioning. Finally, we will describe several metabolic targets that are currently being used for therapeutic interventions.
Collapse
|
36
|
Additive contribution of microRNA-34a/b/c to human arterial ageing and atherosclerosis. Atherosclerosis 2021; 327:49-58. [PMID: 34038763 DOI: 10.1016/j.atherosclerosis.2021.05.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 05/05/2021] [Accepted: 05/12/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Preclinical data suggest that the ageing-induced miR-34a regulates vascular senescence. Herein we sought to assess whether the miR-34 family members miR-34a, miR-34b and miR-34c are involved in human arterial disease. METHODS Expression levels of miR-34a/b/c were quantified by TaqMan assay in peripheral blood mononuclear cells (PBMCs) derived from a consecutive cohort of 221 subjects who underwent cardiovascular risk assessment and thorough vascular examination for aortic stiffness and extent of arterial atherosclerosis. RESULTS High miR-34a was independently associated with the presence of CAD [OR (95%C.I.): 3.87 (1.56-9.56); p = 0.003] and high miR-34c with the number of diseased arterial beds [OR (95%C.I.): 1.88 (1.034-3.41); p = 0.038], while concurrent high expression of miR-34-a/c or all three miR-34a/b/c was associated with aortic stiffening (miR-34a/c: p = 0.022; miR-34a/b/c: p = 0.041) and with the extent of atherosclerosis [OR (95%C.I.) for number of coronary arteries [miR-34a/c: 3.29 (1.085-9.95); miR-34a/b/c: 6.06 (1.74-21.2)] and number of diseased arterial beds [miR-34a/c: 3.51 (1.45-8.52); miR-34a/b/c: 2.89 (1.05-7.92)] after controlling for possible confounders (p < 0.05 for all). Mechanistically, the increased levels of miR-34a or miR-34c were inversely associated with expression of SIRT1 or JAG1, NOTCH2, CTNNB1 and ATF1, respectively. The association of miR-34a/c or miR-34a/b/c with CAD was mainly mediated through SIRT1 and to a lesser extent through JAG1 as revealed by generalized structural equation modeling. Leukocyte-specific ablation of miR-34a/b/c ameliorates atherosclerotic plaque development and increases Sirt1 and Jag1 expression in an atherosclerosis mouse model confirming the human findings. CONCLUSIONS The present study reveals the clinical significance of the additive role of miR-34a/b/c in vascular ageing and atherosclerotic vascular disease.
Collapse
|
37
|
Rabanal-Ruiz Y, Llanos-González E, Alcain FJ. The Use of Coenzyme Q10 in Cardiovascular Diseases. Antioxidants (Basel) 2021; 10:antiox10050755. [PMID: 34068578 PMCID: PMC8151454 DOI: 10.3390/antiox10050755] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023] Open
Abstract
CoQ10 is an endogenous antioxidant produced in all cells that plays an essential role in energy metabolism and antioxidant protection. CoQ10 distribution is not uniform among different organs, and the highest concentration is observed in the heart, though its levels decrease with age. Advanced age is the major risk factor for cardiovascular disease and endothelial dysfunction triggered by oxidative stress that impairs mitochondrial bioenergetic and reduces NO bioavailability, thus affecting vasodilatation. The rationale of the use of CoQ10 in cardiovascular diseases is that the loss of contractile function due to an energy depletion status in the mitochondria and reduced levels of NO for vasodilatation has been associated with low endogenous CoQ10 levels. Clinical evidence shows that CoQ10 supplementation for prolonged periods is safe, well-tolerated and significantly increases the concentration of CoQ10 in plasma up to 3–5 µg/mL. CoQ10 supplementation reduces oxidative stress and mortality from cardiovascular causes and improves clinical outcome in patients undergoing coronary artery bypass graft surgery, prevents the accumulation of oxLDL in arteries, decreases vascular stiffness and hypertension, improves endothelial dysfunction by reducing the source of ROS in the vascular system and increases the NO levels for vasodilation.
Collapse
Affiliation(s)
- Yoana Rabanal-Ruiz
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071 Ciudad Real, Spain; (Y.R.-R.); (E.L.-G.)
- Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research CRIB, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Emilio Llanos-González
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071 Ciudad Real, Spain; (Y.R.-R.); (E.L.-G.)
- Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research CRIB, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Francisco Javier Alcain
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-La Mancha, 13071 Ciudad Real, Spain; (Y.R.-R.); (E.L.-G.)
- Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research CRIB, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
- Correspondence:
| |
Collapse
|
38
|
Minghetti A, Donath L, Zahner L, Hanssen H, Faude O. Beneficial effects of an intergenerational exercise intervention on health-related physical and psychosocial outcomes in Swiss preschool children and residential seniors: a clinical trial. PeerJ 2021; 9:e11292. [PMID: 33987002 PMCID: PMC8086583 DOI: 10.7717/peerj.11292] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/26/2021] [Indexed: 01/22/2023] Open
Abstract
Background Intergenerational exercise possesses the potential to becoming an innovative strategy for promoting physical activity in seniors and children. Although this approach has gained attraction within the last decade, controlled trials on physical and psychosocial effects have not been performed yet. Methods Sixty-eight healthy preschool children (age: 4.9 y (SD 0.7)) and 47 residential seniors (age: 81.7 y (7.1)) participated in this five-armed intervention study. All participants were assigned to either an intergenerational (IG), peer (PG) or a control group (CON). Children were tested on gross motor skills (TGMD-2), jump performance and handgrip strength. Social-emotional skills questionnaires (KOMPIK) were assessed by kindergarten teachers. Seniors performed the Short Physical Performance Battery (SPPB), including gait speed. Arterial stiffness parameters were also examined. Questionnaires assessing psychosocial wellbeing were filled in with staff. IG and PG received one comparable exercise session a week lasting 45 minutes for 25-weeks. CON received no intervention. Measurements were performed before and after the intervention. Results In children: IG improved all measured physical parameters. When adjusted for baseline values, large effects were observed in favor of IG compared to CON in TGMD-2 (Cohen's d=0.78 [0.33;1.24]) and in handgrip strength (d = 1.07 [0.63;1.51]). No relevant differences were found in KOMPIK between groups (-0.38<d≤0.14). In seniors: IG showed moderate to very large improvements in all main physical performance (0.61<d≤2.53) and psychosocial parameters (0.89<d≤1.20) compared to CON. Conclusion IG children showed large benefits in motor skills compared to CON while IG seniors benefit especially in psychosocial wellbeing and functional mobility necessary for everyday life. Intergenerational exercise is comparable and in certain dimensions superior to peer group exercise and a promising strategy to integratively improve mental health as well as physical fitness in preschool children and residential seniors.
Collapse
Affiliation(s)
- Alice Minghetti
- Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Lars Donath
- Department of Intervention Research in Exercise Training, German Sport University, Cologne, Germany
| | - Lukas Zahner
- Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Henner Hanssen
- Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Oliver Faude
- Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| |
Collapse
|
39
|
Fan W, Guo Y, Cao S, Cao S, Xie Y, Liu X, Jin B. Tetrahydroxystilbene glucoside alleviates angiotensin II induced HUVEC senescence via SIRT1. Can J Physiol Pharmacol 2021; 99:389-394. [PMID: 32898442 DOI: 10.1139/cjpp-2020-0202] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tetrahydroxystilbene glucoside (TSG), an active ingredient of Polygonum multiflorum, has been known for certain anti-aging effects. In this study, the possible protective mechanism of TSG on human umbilical vein endothelial cell (HUVEC) senescence induced by angiotensin II (Ang II) was investigated. The results revealed that TSG pretreatment could reduce the percentage of senescence-associated β-galactosidase (SA-β-gal) positive cells and decrease the expression levels of the cellular senescence biomarker proteins p53 and PAI-1. At the same time, the expression of SIRT1 in senescent cells showed an upward trend due to TSG treatment. When inhibiting the expression of SIRT1 by EX527, our results showed that TSG reversed the effect of EX527 by promoting the expression level of SIRT1, reducing the expression of SA-β-gal positive cells and the expression level of p53 and PAI-1 proteins. The present study demonstrated that TSG could protect against HUVEC senescence induced by Ang II, potentially through modulation of SIRT1 activity.
Collapse
Affiliation(s)
- Wenxue Fan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yan Guo
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shidong Cao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shuyu Cao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yuefeng Xie
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Xiangdong Liu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Bo Jin
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| |
Collapse
|
40
|
Izzo C, Vitillo P, Di Pietro P, Visco V, Strianese A, Virtuoso N, Ciccarelli M, Galasso G, Carrizzo A, Vecchione C. The Role of Oxidative Stress in Cardiovascular Aging and Cardiovascular Diseases. Life (Basel) 2021; 11:60. [PMID: 33467601 PMCID: PMC7829951 DOI: 10.3390/life11010060] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Aging can be seen as process characterized by accumulation of oxidative stress induced damage. Oxidative stress derives from different endogenous and exogenous processes, all of which ultimately lead to progressive loss in tissue and organ structure and functions. The oxidative stress theory of aging expresses itself in age-related diseases. Aging is in fact a primary risk factor for many diseases and in particular for cardiovascular diseases and its derived morbidity and mortality. Here we highlight the role of oxidative stress in age-related cardiovascular aging and diseases. We take into consideration the molecular mechanisms, the structural and functional alterations, and the diseases accompanied to the cardiovascular aging process.
Collapse
Affiliation(s)
- Carmine Izzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Paolo Vitillo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Paola Di Pietro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Valeria Visco
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Andrea Strianese
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Nicola Virtuoso
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Gennaro Galasso
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
- Department of Angio-Cardio-Neurology, Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Isernia, Italy
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
- Department of Angio-Cardio-Neurology, Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Isernia, Italy
| |
Collapse
|
41
|
Chen Z, Xiong ZF, Liu X. Research progress on the interaction between circadian clock and early vascular aging. Exp Gerontol 2021; 146:111241. [PMID: 33453324 DOI: 10.1016/j.exger.2021.111241] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/15/2022]
Abstract
Considerable researches implicate that the circadian clock regulates the responsive rhythms of organs and sets the orderly aging process of cells indirectly. It influences an array of diverse biological process including intestinal flora, peripheral inflammatory responses, and redox homeostasis. People with sleep disoders and other kinds of circadian disruptions are prone to have vascular aging earlier. Meanwhile, those people are always faced with chronic vascular inflammation. It has not been elucidated that the specific mechanism of the interaction between the circadian system and early vascular aging. To explore the biphasic relationship between vascular aging and the circadian system, we summarize what is linking circadian clock with early vascular aging through four major prospect: inflammatory process, oxidative stress response, intestinal flora, and cellular senescence. Meanwhile, we discuss the hypothesis that the deterioration of circadian rhythms may exacerbate the process of early vascular aging, leading to the cardiovascular diseases. It will help us to provide new ideas for understanding the process of vascular aging and exploring the possible ways to design personalized chronotherapies.
Collapse
Affiliation(s)
- Zhuoying Chen
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Zhi-Fan Xiong
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Xiangjie Liu
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China.
| |
Collapse
|
42
|
Jin W, Chowienczyk P, Alastruey J. Estimating pulse wave velocity from the radial pressure wave using machine learning algorithms. PLoS One 2021; 16:e0245026. [PMID: 34181640 PMCID: PMC8238176 DOI: 10.1371/journal.pone.0245026] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 06/02/2021] [Indexed: 01/04/2023] Open
Abstract
One of the European gold standard measurement of vascular ageing, a risk factor for cardiovascular disease, is the carotid-femoral pulse wave velocity (cfPWV), which requires an experienced operator to measure pulse waves at two sites. In this work, two machine learning pipelines were proposed to estimate cfPWV from the peripheral pulse wave measured at a single site, the radial pressure wave measured by applanation tonometry. The study populations were the Twins UK cohort containing 3,082 subjects aged from 18 to 110 years, and a database containing 4,374 virtual subjects aged from 25 to 75 years. The first pipeline uses Gaussian process regression to estimate cfPWV from features extracted from the radial pressure wave using pulse wave analysis. The mean difference and upper and lower limits of agreement (LOA) of the estimation on the 924 hold-out test subjects from the Twins UK cohort were 0.2 m/s, and 3.75 m/s & -3.34 m/s, respectively. The second pipeline uses a recurrent neural network (RNN) to estimate cfPWV from the entire radial pressure wave. The mean difference and upper and lower LOA of the estimation on the 924 hold-out test subjects from the Twins UK cohort were 0.05 m/s, and 3.21 m/s & -3.11m/s, respectively. The percentage error of the RNN estimates on the virtual subjects increased by less than 2% when adding 20% of random noise to the pressure waveform. These results show the possibility of assessing the vascular ageing using a single peripheral pulse wave (e.g. the radial pressure wave), instead of cfPWV. The proposed code for the machine learning pipelines is available from the following online depository (https://github.com/WeiweiJin/Estimate-Cardiovascular-Risk-from-Pulse-Wave-Signal).
Collapse
Affiliation(s)
- Weiwei Jin
- Department of Biomedical Engineering, King’s College London, London, United Kingdom
- * E-mail: ,
| | - Philip Chowienczyk
- Department of Clinical Pharmacology, St. Thomas’ Hospital, King’s College London, London, United Kingdom
| | - Jordi Alastruey
- Department of Biomedical Engineering, King’s College London, London, United Kingdom
- World-Class Research Centre, Digital Biodesign and Personalized Healthcare, Sechenov University, Moscow, Russia
| |
Collapse
|
43
|
Stakos DA, Stamatelopoulos K, Bampatsias D, Sachse M, Zormpas E, Vlachogiannis NI, Tual-Chalot S, Stellos K. The Alzheimer's Disease Amyloid-Beta Hypothesis in Cardiovascular Aging and Disease: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:952-967. [PMID: 32130931 PMCID: PMC7042886 DOI: 10.1016/j.jacc.2019.12.033] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
Aging-related cellular and molecular processes including low-grade inflammation are major players in the pathogenesis of cardiovascular disease (CVD) and Alzheimer's disease (AD). Epidemiological studies report an independent interaction between the development of dementia and the incidence of CVD in several populations, suggesting the presence of overlapping molecular mechanisms. Accumulating experimental and clinical evidence suggests that amyloid-beta (Aβ) peptides may function as a link among aging, CVD, and AD. Aging-related vascular and cardiac deposition of Αβ induces tissue inflammation and organ dysfunction, both important components of the Alzheimer's disease amyloid hypothesis. In this review, the authors describe the determinants of Aβ metabolism, summarize the effects of Aβ on atherothrombosis and cardiac dysfunction, discuss the clinical value of Αβ1-40 in CVD prognosis and patient risk stratification, and present the therapeutic interventions that may alter Aβ metabolism in humans.
Collapse
Affiliation(s)
- Dimitrios A Stakos
- Cardiology Department, Democritus University of Thrace, Alexandroupolis, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece; Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Dimitrios Bampatsias
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Marco Sachse
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Medical School, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Eleftherios Zormpas
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nikolaos I Vlachogiannis
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Simon Tual-Chalot
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Konstantinos Stellos
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Department of Cardiology, Freeman Hospital, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom; NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
44
|
Lambrinoudaki I, Delialis D, Georgiopoulos G, Tual-Chalot S, Vlachogiannis NI, Patras R, Aivalioti E, Armeni E, Augoulea A, Tsoltos N, Soureti A, Stellos K, Stamatelopoulos K. Circulating Amyloid Beta 1-40 Is Associated with Increased Rate of Progression of Atherosclerosis in Menopause: A Prospective Cohort Study. Thromb Haemost 2020; 121:650-658. [PMID: 33202443 DOI: 10.1055/s-0040-1721144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Accumulating evidence suggests that circulating amyloidβ 1-40 (Αβ1-40), a proatherogenic aging peptide, may serve as a novel biomarker in cardiovascular disease (CVD). We aimed to explore the role of plasma Αβ1-40 and its patterns of change over time in atherosclerosis progression in postmenopausal women, a population with substantial unrecognized CVD risk beyond traditional risk factors (TRFs). METHODS In this prospective study, Αβ1-40 was measured in plasma by enzyme-linked immunosorbent assay and atherosclerosis was assessed using carotid high-resolution ultrasonography at baseline and after a median follow-up of 28.2 months in 152 postmenopausal women without history or symptoms of CVD. RESULTS At baseline, high Αβ1-40 was independently associated with higher carotid bulb intima-media thickness (cbIMT) and the sum of maximal wall thickness in all carotid sites (sumWT) (p < 0.05). Αβ1-40 levels increased over time and were associated with decreasing renal function (p < 0.05 for both). Women with a pattern of increasing or persistently high Αβ1-40 levels presented accelerated progression of cbIMT and maximum carotid wall thickness and sumWT (p < 0.05 for all) after adjustment for baseline Αβ1-40 levels, TRFs, and renal function. CONCLUSION In postmenopausal women, a pattern of increasing or persistently high Αβ1-40 was associated with the rate of progression of subclinical atherosclerosis irrespective of its baseline levels. These findings provide novel insights into a link between Αβ1-40 and atherosclerosis progression in menopause and warrant further research to clarify the clinical value of monitoring its circulating levels as an atherosclerosis biomarker in women without clinically overt CVD.
Collapse
Affiliation(s)
- Irene Lambrinoudaki
- Menopause Clinic, 2nd Department of Obstetrics and Gynecology, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Delialis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.,School of Biomedical Engineering & Imaging Sciences, Rayne Institute, St. Thomas' Hospital, London, United Kingdom
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nikolaos I Vlachogiannis
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Raphael Patras
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Evmorfia Aivalioti
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Eleni Armeni
- Menopause Clinic, 2nd Department of Obstetrics and Gynecology, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Areti Augoulea
- Menopause Clinic, 2nd Department of Obstetrics and Gynecology, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Tsoltos
- Menopause Clinic, 2nd Department of Obstetrics and Gynecology, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Soureti
- Menopause Clinic, 2nd Department of Obstetrics and Gynecology, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom.,Department of Cardiology, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom
| | - Kimon Stamatelopoulos
- Menopause Clinic, 2nd Department of Obstetrics and Gynecology, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece.,Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
45
|
Dall'Olio L, Curti N, Remondini D, Safi Harb Y, Asselbergs FW, Castellani G, Uh HW. Prediction of vascular aging based on smartphone acquired PPG signals. Sci Rep 2020; 10:19756. [PMID: 33184391 PMCID: PMC7661535 DOI: 10.1038/s41598-020-76816-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/23/2020] [Indexed: 02/04/2023] Open
Abstract
Photoplethysmography (PPG) measured by smartphone has the potential for a large scale, non-invasive, and easy-to-use screening tool. Vascular aging is linked to increased arterial stiffness, which can be measured by PPG. We investigate the feasibility of using PPG to predict healthy vascular aging (HVA) based on two approaches: machine learning (ML) and deep learning (DL). We performed data preprocessing, including detrending, demodulating, and denoising on the raw PPG signals. For ML, ridge penalized regression has been applied to 38 features extracted from PPG, whereas for DL several convolutional neural networks (CNNs) have been applied to the whole PPG signals as input. The analysis has been conducted using the crowd-sourced Heart for Heart data. The prediction performance of ML using two features (AUC of 94.7%) – the a wave of the second derivative PPG and tpr, including four covariates, sex, height, weight, and smoking – was similar to that of the best performing CNN, 12-layer ResNet (AUC of 95.3%). Without having the heavy computational cost of DL, ML might be advantageous in finding potential biomarkers for HVA prediction. The whole workflow of the procedure is clearly described, and open software has been made available to facilitate replication of the results.
Collapse
Affiliation(s)
- Lorenzo Dall'Olio
- Department of Physics and Astronomy, University of Bologna, 40126, Bologna, BO, Italy
| | - Nico Curti
- Department of Specialised, Diagnostic and Experimental Medicine, University of Bologna, 40126, Bologna, BO, Italy
| | - Daniel Remondini
- Department of Physics and Astronomy, University of Bologna, 40126, Bologna, BO, Italy
| | | | - Folkert W Asselbergs
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands.,Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, WC1E 6BT, UK.,Health Data Research UK and Institute of Health Informatics, University College London, London, NW1 2BE, UK
| | - Gastone Castellani
- Department of Specialised, Diagnostic and Experimental Medicine, University of Bologna, 40126, Bologna, BO, Italy
| | - Hae-Won Uh
- Department of Biostatistics and Research Support, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
46
|
Shi HZ, Zeng JC, Shi SH, Giannakopoulos H, Zhang QZ, Le AD. Extracellular Vesicles of GMSCs Alleviate Aging-Related Cell Senescence. J Dent Res 2020; 100:283-292. [PMID: 33073684 DOI: 10.1177/0022034520962463] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Healthy aging is a complex biological process with progressive accumulation of senescent cells characterized by stable cell cycle arrest, resulting in impaired homeostasis, regenerative potential, and gradual functional decline in multiple tissues and organs, whereby the aberrant activation of mammalian target of rapamycin (mTOR) signaling networks plays a central role. Herein, we explored the effects of extracellular vesicles (EVs) released by gingiva-derived mesenchymal stem cells (GMSC-EVs) on oxidative stress-induced cellular senescence in human endothelial cells and skin fibroblasts and their antiaging potentials. Our results showed that GMSC-EVs robustly abrogated oxidative stress-induced upregulation in the expression of cellular senescence-related genes, such as β-galactosidase, p21, p53, and γH2AX, and mTOR/pS6 signaling pathway, in human umbilical vein endothelial cells (HUVECs) and skin fibroblasts. Meanwhile, GMSC-EVs restored oxidative stress-induced impairment in proliferation and tube formation by HUVECs. Systemic administration of GMSC-EVs attenuated aging-associated elevation in the expression levels of p21, mTOR/pS6, interleukin 6, and tumor necrosis factor α in skin and heart tissues of aged mice. These findings suggest that GMSC-EVs could be a potential alternative source of cell-free product for attenuation of aging-related skin and vascular dysfunctions due to their potent inhibitory effects on oxidative stress-induced cellular senescence in endothelial cells and skin fibroblasts.
Collapse
Affiliation(s)
- H Z Shi
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
- Health Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - J C Zeng
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - S H Shi
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - H Giannakopoulos
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
- Department of Oral & Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, Perelman Center for Advanced Medicine, Philadelphia, PA, USA
| | - Q Z Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - A D Le
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
- Department of Oral & Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, Perelman Center for Advanced Medicine, Philadelphia, PA, USA
| |
Collapse
|
47
|
Gocmez SS, Yazir Y, Gacar G, Demirtaş Şahin T, Arkan S, Karson A, Utkan T. Etanercept improves aging-induced cognitive deficits by reducing inflammation and vascular dysfunction in rats. Physiol Behav 2020; 224:113019. [DOI: 10.1016/j.physbeh.2020.113019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 11/15/2022]
|
48
|
Machado-Oliveira G, Ramos C, Marques ARA, Vieira OV. Cell Senescence, Multiple Organelle Dysfunction and Atherosclerosis. Cells 2020; 9:E2146. [PMID: 32977446 PMCID: PMC7598292 DOI: 10.3390/cells9102146] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/19/2020] [Accepted: 09/20/2020] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is an age-related disorder associated with long-term exposure to cardiovascular risk factors. The asymptomatic progression of atherosclerotic plaques leads to major cardiovascular diseases (CVD), including acute myocardial infarctions or cerebral ischemic strokes in some cases. Senescence, a biological process associated with progressive structural and functional deterioration of cells, tissues and organs, is intricately linked to age-related diseases. Cell senescence involves coordinated modifications in cellular compartments and has been demonstrated to contribute to different stages of atheroma development. Senescence-based therapeutic strategies are currently being pursued to treat and prevent CVD in humans in the near-future. In addition, distinct experimental settings allowed researchers to unravel potential approaches to regulate anti-apoptotic pathways, facilitate excessive senescent cell clearance and eventually reverse atherogenesis to improve cardiovascular function. However, a deeper knowledge is required to fully understand cellular senescence, to clarify senescence and atherogenesis intertwining, allowing researchers to establish more effective treatments and to reduce the cardiovascular disorders' burden. Here, we present an objective review of the key senescence-related alterations of the major intracellular organelles and analyze the role of relevant cell types for senescence and atherogenesis. In this context, we provide an updated analysis of therapeutic approaches, including clinically relevant experiments using senolytic drugs to counteract atherosclerosis.
Collapse
Affiliation(s)
- Gisela Machado-Oliveira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (C.R.); (A.R.A.M.)
| | | | | | - Otília V. Vieira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (C.R.); (A.R.A.M.)
| |
Collapse
|
49
|
Pecher SJ, Potthast AB, von Versen-Höynck F, Das AM. Impact of Short-Term Hypoxia on Sirtuins as Regulatory Elements in HUVECs. J Clin Med 2020; 9:jcm9082604. [PMID: 32796661 PMCID: PMC7464651 DOI: 10.3390/jcm9082604] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/07/2020] [Accepted: 08/09/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Sirtuins (SIRT) are NAD+-dependent deacetylases that are involved in stress response, antioxidative defense, and longevity via posttranslational modifications. SIRT1 directly activates nitric oxide synthase (NOS). Aging is associated with a reduced sirtuin function and reduction of the cofactor NAD+. Age-related atherosclerosis and vascular diseases are linked to a compromised sirtuin function. Vascular events like stroke and cardiac infarction result in acute hypoxia, which can additionally impact sirtuins and thus the vascular function. This prompted us to study sirtuins in intact HUVECs, under acute, short-term hypoxic conditions. Methods: We measured intracellular sirtuin and NAD+ levels in HUVECs exposed to hypoxia (2% O₂) for 10–120 min, compared to normoxic controls. SIRT1, SIRT3, and SIRT4 were measured at the protein (Western Blot) and the transcript level (qRT-PCR), SIRT1 and SIRT3 at the enzyme level (fluorometrically), and NAD+ levels were measured spectrophotometrically. Results: We observed a reduction of SIRT1 and SIRT4 at the protein level, a downregulation of SIRT1 at the transcript level and increased NAD+ levels under hypoxia. SIRT3 was not affected by hypoxia. Conclusions: Downregulation of SIRT1 under hypoxia might reduce production of the reactive oxygen species (ROS) via the respiratory chain and inhibit the mitochondrial ATP-synthase, resulting in energy conservation. NOS might be impaired if SIRT1 is decreased. Increased NAD+ levels might compensate these effects. Hypoxic downregulation of SIRT4 might lead to mitochondrial uncoupling, hence endothelial dysfunction, and ADP/ATP-translocase 2 (ANT2)-inhibition. NAD+ upregulation might partly compensate this effect.
Collapse
Affiliation(s)
- Simone Johanna Pecher
- Clinic for Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School (MHH), Carl-Neuberg-Straße 1, 30625 Hannover, Germany; (S.J.P.); (A.B.P.)
| | - Arne Björn Potthast
- Clinic for Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School (MHH), Carl-Neuberg-Straße 1, 30625 Hannover, Germany; (S.J.P.); (A.B.P.)
| | - Frauke von Versen-Höynck
- Department of Obstetrics and Gynecology, Hannover Medical School (MHH), Carl-Neuberg-Straße 1, 30625 Hannover, Germany;
| | - Anibh Martin Das
- Clinic for Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School (MHH), Carl-Neuberg-Straße 1, 30625 Hannover, Germany; (S.J.P.); (A.B.P.)
- Correspondence: ; Tel.: +49-511-532-3220
| |
Collapse
|
50
|
Dominic A, Banerjee P, Hamilton DJ, Le NT, Abe JI. Time-dependent replicative senescence vs. disturbed flow-induced pre-mature aging in atherosclerosis. Redox Biol 2020; 37:101614. [PMID: 32863187 PMCID: PMC7767754 DOI: 10.1016/j.redox.2020.101614] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/07/2020] [Accepted: 06/18/2020] [Indexed: 12/19/2022] Open
Abstract
Accumulation of senescent cells has a causative role in the pathology of age-related disorders including atherosclerosis (AS) and cardiovascular diseases (CVDs). However, the concept of senescence is now drastically changing, and the new concept of senescence-associated reprogramming/stemness has emerged, suggesting that senescence is not merely related to “cell cycle arrest” or halting various cellular functions. It is well known that disturbed flow (D-flow) accelerates pre-mature aging and plays a significant role in the development of AS. We will discuss in this review that pre-mature aging induced by D-flow is not comparable to time-dependent aging, particularly with a focus on the possible involvement of senescence-associated secretory phenotype (SASP) in senescence-associated reprogramming/stemness, or increasing cell numbers. We will also present our outlook of nicotinamide adenine dinucleotides (NAD)+ deficiency-induced mitochondrial reactive oxygen species (mtROS) in evoking SASP by activating DNA damage response (DDR). MtROS plays a key role in developing cross-talk between nuclear-mitochondria, SASP, and ultimately atherosclerosis formation. Although senescence induced by time and various stress factors is a classical concept, we wish that the readers will see the undergoing Copernican-like change in this concept, as well as to recognize the significant contrast between pre-mature aging induced by D-flow and time-dependent aging.
Collapse
Affiliation(s)
- Abishai Dominic
- Department of Molecular and Cellular Biology Texas A&M Health Science Center, USA; Department of Cardio-Vascular Regeneration, Houston Methodist Research Institute, Texas, USA
| | - Priyanka Banerjee
- Department of Cardio-Vascular Regeneration, Houston Methodist Research Institute, Texas, USA
| | - Dale J Hamilton
- Department of Medicine, Center for Bioenergetics Houston Methodist Research Institute, Texas, USA
| | - Nhat-Tu Le
- Department of Cardio-Vascular Regeneration, Houston Methodist Research Institute, Texas, USA.
| | - Jun-Ichi Abe
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|