1
|
Ahmed B, Rahman AA, Lee S, Malhotra R. The Implications of Aging on Vascular Health. Int J Mol Sci 2024; 25:11188. [PMID: 39456971 PMCID: PMC11508873 DOI: 10.3390/ijms252011188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Vascular aging encompasses structural and functional changes in the vasculature, significantly contributing to cardiovascular diseases, which are the leading cause of death globally. The incidence and prevalence of these diseases increase with age, with most morbidity and mortality attributed to myocardial infarction and stroke. Diagnosing and intervening in vascular aging while understanding the mechanisms behind age-induced vascular phenotypic and pathophysiological alterations offers the potential for delaying and preventing cardiovascular mortality in an aging population. This review delves into various aspects of vascular aging by examining age-related changes in arterial health at the cellular level, including endothelial dysfunction, cellular senescence, and vascular smooth muscle cell transdifferentiation, as well as at the structural level, including arterial stiffness and changes in wall thickness and diameter. We also explore aging-related changes in perivascular adipose tissue deposition, arterial collateralization, and calcification, providing insights into the physiological and pathological implications. Overall, aging induces phenotypic changes that augment the vascular system's susceptibility to disease, even in the absence of traditional risk factors, such as hypertension, diabetes, obesity, and smoking. Overall, age-related modifications in cellular phenotype and molecular homeostasis increase the vulnerability of the arterial vasculature to structural and functional alterations, thereby accelerating cardiovascular risk. Increasing our understanding of these modifications is crucial for success in delaying or preventing cardiovascular diseases. Non-invasive techniques, such as measuring carotid intima-media thickness, pulse wave velocity, and flow-mediated dilation, as well as detecting vascular calcifications, can be used for the early detection of vascular aging. Targeting specific pathological mechanisms, such as cellular senescence and enhancing angiogenesis, holds promise for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Ahmed A. Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sujin Lee
- Division of Vascular Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Rajeev Malhotra
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
2
|
Moghtadaei M, Tagirova S, Ahmet I, Moen J, Lakatta EG, Rose RA. Lifelong longitudinal assessment of the contribution of multi-fractal fluctuations to heart rate and heart rate variability in aging mice: role of the sinoatrial node and autonomic nervous system. GeroScience 2024; 46:5085-5101. [PMID: 38967697 PMCID: PMC11336143 DOI: 10.1007/s11357-024-01267-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024] Open
Abstract
Aging is a major risk factor for sinoatrial node (SAN) dysfunction, which can impair heart rate (HR) control and heart rate variability (HRV). HR and HRV are determined by intrinsic SAN function and its regulation by the autonomic nervous system (ANS). The purpose of this study was to use multi-scale multi-fractal detrended fluctuation analysis (MSMFDFA; a complexity-based approach to analyze multi-fractal dynamics) to longitudinally assess changes in multi-fractal HRV properties and SAN function in ECG time series recorded repeatedly across the full adult lifespan in mice. ECGs were recorded in anesthetized mice in baseline conditions and after autonomic nervous system blockade every three months beginning at 6 months of age until the end of life. MSMFDFA was used to assess HRV and SAN function every three months between 6 and 27 months of age. Intrinsic HR (i.e. HR during ANS blockade) remained relatively stable until 15 months of age, and then progressively declined until study endpoint at 27 months of age. MSMFDFA revealed sudden and rapid changes in multi-fractal properties of the ECG RR interval time series in aging mice. In particular, multi-fractal spectrum width (MFSW, a measure of multi-fractality) was relatively stable between 6 months and 15 months of age and then progressively increased at 27 months of age. These changes in MFSW were evident in baseline conditions and during ANS blockade. Thus, intrinsic SAN function declines progressively during aging and is manifested by age-associated changes in multi-fractal HRV across the lifespan in mice, which can be accurately quantified by MSMFDFA.
Collapse
Affiliation(s)
- Motahareh Moghtadaei
- Department of Cardiac Sciences, Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, GAC66, Health Research Innovation Centre, 3280 Hospital Drive N.W., Calgary, Alberta, T2N 4Z6, Canada
| | - Syevda Tagirova
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ismayil Ahmet
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jack Moen
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| | - Robert A Rose
- Department of Cardiac Sciences, Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, GAC66, Health Research Innovation Centre, 3280 Hospital Drive N.W., Calgary, Alberta, T2N 4Z6, Canada.
| |
Collapse
|
3
|
Schini-Kerth VB, Diouf I, Muzammel H, Said A, Auger C. Natural Products to Promote Vascular Health. Handb Exp Pharmacol 2024. [PMID: 39317849 DOI: 10.1007/164_2024_721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Maintaining good vascular health is a major component in healthy ageing as it reduces the risk of cardiovascular diseases. Endothelial dysfunction, in particular, is a key mechanism in the development of major cardiovascular diseases including hypertension, atherosclerosis and diabetes. Recently, endothelial senescence has emerged as a pivotal early event in age-related endothelial dysfunction. Endothelial function is characterized by an imbalance between the endothelial formation of vasoprotective mechanisms, including the formation of nitric oxide (NO) and endothelium-dependent hyperpolarization responses, and an increased level of oxidative stress involving several pro-oxidant enzymes such as NADPH oxidases and, often also, the appearance of cyclooxygenase-derived vasoconstrictors. Pre-clinical studies have indicated that natural products, in particular several polyphenol-rich foods, can trigger activating pathways in endothelial cells promoting an increased formation of NO and endothelium-dependent hyperpolarization. In addition, some can even exert beneficial effects on endothelial senescence. Moreover, some of these products have been associated with the prevention and/or improvement of established endothelial dysfunction in several experimental models of cardiovascular diseases and in humans with cardiovascular diseases. Therefore, intake of certain natural products, such as dietary and plant-derived polyphenol-rich products, appears to be an attractive approach for a healthy vascular system in ageing.
Collapse
Affiliation(s)
- Valérie B Schini-Kerth
- Translational Cardiovascular Medicine, UR 3074, CRBS, University of Strasbourg, Strasbourg, France.
| | - Ibrahima Diouf
- Translational Cardiovascular Medicine, UR 3074, CRBS, University of Strasbourg, Strasbourg, France
| | - Hira Muzammel
- Translational Cardiovascular Medicine, UR 3074, CRBS, University of Strasbourg, Strasbourg, France
| | - Amissi Said
- Translational Cardiovascular Medicine, UR 3074, CRBS, University of Strasbourg, Strasbourg, France
| | - Cyril Auger
- Regenerative Nanomedicine, INSERM UMR 1260, CRBS, University of Strasbourg, Strasbourg, France
| |
Collapse
|
4
|
Gao D, Zhao B, Yu J, Li X, Yang D, Luo Y, Xia Y, Cai X, Guo Y. Deletion of stimulator of interferons genes aggravated cardiac dysfunction in physiological aged mice. Mech Ageing Dev 2024; 222:111978. [PMID: 39233064 DOI: 10.1016/j.mad.2024.111978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/15/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Stimulator of interferons genes (STING) is crucial for innate immune response. It has been demonstrated that cGAS-STING pathway was the driver of aging-related inflammation. However, whether STING is involved in cardiac dysfunction during the physiological aging process remains unclear. METHODS Gene expression profiles were obtained from the Gene Expression Omnibus database, followed by weighted gene co-expression network analysis, gene ontology analysis and protein network interaction analysis to identify key pathway and genes associated with aging. The effects of STING on cardiac function, glucose homeostasis, inflammation, and autophagy in physiological aging were investigated with STING knockout mice. RESULTS Bioinformatics analysis revealed STING emerged as a hub gene of interest. Subsequent experiments demonstrated the activation of STING pathway in the heart of aged mice. Knockout of STING alleviated the inflammation in aged mice. However, Knockout of STING impaired glucose tolerance, inhibited autophagy, enhanced oxidative stress and aggravated cardiac dysfunction in aged mice. CONCLUSION Although reducing inflammation, long-term STING inhibition by genetic ablation exacerbated cardiac dysfunction in aged mice. Given the multifaceted nature of aging and the diverse cellular functions of STING beyond immune regulation, the negative effects of targeting STING as a strategy to mitigate aging phenotype should be fully considered.
Collapse
Affiliation(s)
- Diansa Gao
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Boying Zhao
- Division of Cardiothoracic Surgery, The Center Hospital of Chongqing University, Chongqing 400016, China
| | - Jiang Yu
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaorong Li
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ding Yang
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuan Luo
- Division of Cardiothoracic Surgery, The Center Hospital of Chongqing University, Chongqing 400016, China
| | - Yong Xia
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiongwei Cai
- Department of Gynecology, Chongqing Health Center for Women and Children, Women and Children's Hospital of Chongqing Medical University, Chongqing 400037, China.
| | - Yongzheng Guo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
5
|
Ma C, Liu Y, Fu Z. Implications of endoplasmic reticulum stress and autophagy in aging and cardiovascular diseases. Front Pharmacol 2024; 15:1413853. [PMID: 39119608 PMCID: PMC11306071 DOI: 10.3389/fphar.2024.1413853] [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: 04/08/2024] [Accepted: 06/24/2024] [Indexed: 08/10/2024] Open
Abstract
The average lifespan of humans has been increasing, resulting in a rapidly rising percentage of older individuals and high morbidity of aging-associated diseases, especially cardiovascular diseases (CVDs). Diverse intracellular and extracellular factors that interrupt homeostatic functions in the endoplasmic reticulum (ER) induce ER stress. Cells employ a dynamic signaling pathway of unfolded protein response (UPR) to buffer ER stress. Recent studies have demonstrated that ER stress triggers various cellular processes associated with aging and many aging-associated diseases, including CVDs. Autophagy is a conserved process involving lysosomal degradation and recycling of cytoplasmic components, proteins, organelles, and pathogens that invade the cytoplasm. Autophagy is vital for combating the adverse influence of aging on the heart. The present report summarizes recent studies on the mechanism of ER stress and autophagy and their overlap in aging and on CVD pathogenesis in the context of aging. It also discusses possible therapeutic interventions targeting ER stress and autophagy that might delay aging and prevent or treat CVDs.
Collapse
Affiliation(s)
- Chenguang Ma
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Liu
- 32295 Troops of P.L.A, Liaoyang, China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
6
|
Fang Z, Raza U, Song J, Lu J, Yao S, Liu X, Zhang W, Li S. Systemic aging fuels heart failure: Molecular mechanisms and therapeutic avenues. ESC Heart Fail 2024. [PMID: 39034866 DOI: 10.1002/ehf2.14947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/29/2024] [Accepted: 06/21/2024] [Indexed: 07/23/2024] Open
Abstract
Systemic aging influences various physiological processes and contributes to structural and functional decline in cardiac tissue. These alterations include an increased incidence of left ventricular hypertrophy, a decline in left ventricular diastolic function, left atrial dilation, atrial fibrillation, myocardial fibrosis and cardiac amyloidosis, elevating susceptibility to chronic heart failure (HF) in the elderly. Age-related cardiac dysfunction stems from prolonged exposure to genomic, epigenetic, oxidative, autophagic, inflammatory and regenerative stresses, along with the accumulation of senescent cells. Concurrently, age-related structural and functional changes in the vascular system, attributed to endothelial dysfunction, arterial stiffness, impaired angiogenesis, oxidative stress and inflammation, impose additional strain on the heart. Dysregulated mechanosignalling and impaired nitric oxide signalling play critical roles in the age-related vascular dysfunction associated with HF. Metabolic aging drives intricate shifts in glucose and lipid metabolism, leading to insulin resistance, mitochondrial dysfunction and lipid accumulation within cardiomyocytes. These alterations contribute to cardiac hypertrophy, fibrosis and impaired contractility, ultimately propelling HF. Systemic low-grade chronic inflammation, in conjunction with the senescence-associated secretory phenotype, aggravates cardiac dysfunction with age by promoting immune cell infiltration into the myocardium, fostering HF. This is further exacerbated by age-related comorbidities like coronary artery disease (CAD), atherosclerosis, hypertension, obesity, diabetes and chronic kidney disease (CKD). CAD and atherosclerosis induce myocardial ischaemia and adverse remodelling, while hypertension contributes to cardiac hypertrophy and fibrosis. Obesity-associated insulin resistance, inflammation and dyslipidaemia create a profibrotic cardiac environment, whereas diabetes-related metabolic disturbances further impair cardiac function. CKD-related fluid overload, electrolyte imbalances and uraemic toxins exacerbate HF through systemic inflammation and neurohormonal renin-angiotensin-aldosterone system (RAAS) activation. Recognizing aging as a modifiable process has opened avenues to target systemic aging in HF through both lifestyle interventions and therapeutics. Exercise, known for its antioxidant effects, can partly reverse pathological cardiac remodelling in the elderly by countering processes linked to age-related chronic HF, such as mitochondrial dysfunction, inflammation, senescence and declining cardiomyocyte regeneration. Dietary interventions such as plant-based and ketogenic diets, caloric restriction and macronutrient supplementation are instrumental in maintaining energy balance, reducing adiposity and addressing micronutrient and macronutrient imbalances associated with age-related HF. Therapeutic advancements targeting systemic aging in HF are underway. Key approaches include senomorphics and senolytics to limit senescence, antioxidants targeting mitochondrial stress, anti-inflammatory drugs like interleukin (IL)-1β inhibitors, metabolic rejuvenators such as nicotinamide riboside, resveratrol and sirtuin (SIRT) activators and autophagy enhancers like metformin and sodium-glucose cotransporter 2 (SGLT2) inhibitors, all of which offer potential for preserving cardiac function and alleviating the age-related HF burden.
Collapse
Affiliation(s)
- Zhuyubing Fang
- Cardiovascular Department of Internal Medicine, Karamay Hospital of People's Hospital of Xinjiang Uygur Autonomous Region, Karamay, Xinjiang Uygur Autonomous Region, China
| | - Umar Raza
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Jia Song
- Department of Medicine (Cardiovascular Research), Baylor College of Medicine, Houston, Texas, USA
| | - Junyan Lu
- Department of Cardiology, Zengcheng Branch of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shun Yao
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiaohong Liu
- Cardiovascular Department of Internal Medicine, Karamay Hospital of People's Hospital of Xinjiang Uygur Autonomous Region, Karamay, Xinjiang Uygur Autonomous Region, China
| | - Wei Zhang
- Outpatient Clinic of Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Shujuan Li
- Department of Pediatric Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
7
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
8
|
Cui X, Buonfiglio F, Pfeiffer N, Gericke A. Aging in Ocular Blood Vessels: Molecular Insights and the Role of Oxidative Stress. Biomedicines 2024; 12:817. [PMID: 38672172 PMCID: PMC11048681 DOI: 10.3390/biomedicines12040817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/01/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Acknowledged as a significant pathogenetic driver for numerous diseases, aging has become a focal point in addressing the profound changes associated with increasing human life expectancy, posing a critical concern for global public health. Emerging evidence suggests that factors influencing vascular aging extend their impact to choroidal and retinal blood vessels. The objective of this work is to provide a comprehensive overview of the impact of vascular aging on ocular blood vessels and related diseases. Additionally, this study aims to illuminate molecular insights contributing to vascular cell aging, with a particular emphasis on the choroid and retina. Moreover, innovative molecular targets operating within the domain of ocular vascular aging are presented and discussed.
Collapse
Affiliation(s)
- Xiuting Cui
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (N.P.)
| | | | | | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (N.P.)
| |
Collapse
|
9
|
Fekete M, Major D, Feher A, Fazekas-Pongor V, Lehoczki A. Geroscience and pathology: a new frontier in understanding age-related diseases. Pathol Oncol Res 2024; 30:1611623. [PMID: 38463143 PMCID: PMC10922957 DOI: 10.3389/pore.2024.1611623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/07/2024] [Indexed: 03/12/2024]
Abstract
Geroscience, a burgeoning discipline at the intersection of aging and disease, aims to unravel the intricate relationship between the aging process and pathogenesis of age-related diseases. This paper explores the pivotal role played by geroscience in reshaping our understanding of pathology, with a particular focus on age-related diseases. These diseases, spanning cardiovascular and cerebrovascular disorders, malignancies, and neurodegenerative conditions, significantly contribute to the morbidity and mortality of older individuals. We delve into the fundamental cellular and molecular mechanisms underpinning aging, including mitochondrial dysfunction and cellular senescence, and elucidate their profound implications for the pathogenesis of various age-related diseases. Emphasis is placed on the importance of assessing key biomarkers of aging and biological age within the realm of pathology. We also scrutinize the interplay between cellular senescence and cancer biology as a central area of focus, underscoring its paramount significance in contemporary pathological research. Moreover, we shed light on the integration of anti-aging interventions that target fundamental aging processes, such as senolytics, mitochondria-targeted treatments, and interventions that influence epigenetic regulation within the domain of pathology research. In conclusion, the integration of geroscience concepts into pathological research heralds a transformative paradigm shift in our understanding of disease pathogenesis and promises breakthroughs in disease prevention and treatment.
Collapse
Affiliation(s)
- Monika Fekete
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - David Major
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Agnes Feher
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | | | - Andrea Lehoczki
- Department of Public Health, Semmelweis University, Budapest, Hungary
- Departments of Hematology and Stem Cell Transplantation, South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, Saint Ladislaus Campus, Budapest, Hungary
| |
Collapse
|
10
|
Ananthamohan K, Stelzer JE, Sadayappan S. Hypertrophic cardiomyopathy in MYBPC3 carriers in aging. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:9. [PMID: 38406555 PMCID: PMC10883298 DOI: 10.20517/jca.2023.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is characterized by abnormal thickening of the myocardium, leading to arrhythmias, heart failure, and elevated risk of sudden cardiac death, particularly among the young. This inherited disease is predominantly caused by mutations in sarcomeric genes, among which those in the cardiac myosin binding protein-C3 (MYBPC3) gene are major contributors. HCM associated with MYBPC3 mutations usually presents in the elderly and ranges from asymptomatic to symptomatic forms, affecting numerous cardiac functions and presenting significant health risks with a spectrum of clinical manifestations. Regulation of MYBPC3 expression involves various transcriptional and translational mechanisms, yet the destiny of mutant MYBPC3 mRNA and protein in late-onset HCM remains unclear. Pathogenesis related to MYBPC3 mutations includes nonsense-mediated decay, alternative splicing, and ubiquitin-proteasome system events, leading to allelic imbalance and haploinsufficiency. Aging further exacerbates the severity of HCM in carriers of MYBPC3 mutations. Advancements in high-throughput omics techniques have identified crucial molecular events and regulatory disruptions in cardiomyocytes expressing MYBPC3 variants. This review assesses the pathogenic mechanisms that promote late-onset HCM through the lens of transcriptional, post-transcriptional, and post-translational modulation of MYBPC3, underscoring its significance in HCM across carriers. The review also evaluates the influence of aging on these processes and MYBPC3 levels during HCM pathogenesis in the elderly. While pinpointing targets for novel medical interventions to conserve cardiac function remains challenging, the emergence of personalized omics offers promising avenues for future HCM treatments, particularly for late-onset cases.
Collapse
Affiliation(s)
- Kalyani Ananthamohan
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 45267, USA
| | - Sakthivel Sadayappan
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
11
|
Abdellatif M, Rainer PP, Sedej S, Kroemer G. Hallmarks of cardiovascular ageing. Nat Rev Cardiol 2023; 20:754-777. [PMID: 37193857 DOI: 10.1038/s41569-023-00881-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/18/2023]
Abstract
Normal circulatory function is a key determinant of disease-free life expectancy (healthspan). Indeed, pathologies affecting the cardiovascular system, which are growing in prevalence, are the leading cause of global morbidity, disability and mortality, whereas the maintenance of cardiovascular health is necessary to promote both organismal healthspan and lifespan. Therefore, cardiovascular ageing might precede or even underlie body-wide, age-related health deterioration. In this Review, we posit that eight molecular hallmarks are common denominators in cardiovascular ageing, namely disabled macroautophagy, loss of proteostasis, genomic instability (in particular, clonal haematopoiesis of indeterminate potential), epigenetic alterations, mitochondrial dysfunction, cell senescence, dysregulated neurohormonal signalling and inflammation. We also propose a hierarchical order that distinguishes primary (upstream) from antagonistic and integrative (downstream) hallmarks of cardiovascular ageing. Finally, we discuss how targeting each of the eight hallmarks might be therapeutically exploited to attenuate residual cardiovascular risk in older individuals.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Graz, Austria.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- BioTechMed Graz, Graz, Austria.
| | - Peter P Rainer
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
12
|
Li S, Sun Y. Phytochemicals targeting epidermal growth factor receptor (EGFR) for the prevention and treatment of HNSCC: A review. Medicine (Baltimore) 2023; 102:e34439. [PMID: 37800790 PMCID: PMC10553117 DOI: 10.1097/md.0000000000034439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/30/2023] [Indexed: 10/07/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) develops from the mucosal epithelium of the oral cavity, pharynx, and larynx, and is the most common malignancy of the head and neck, the incidence of which continues to rise. The epidermal growth factor receptor is thought to play a key role in the pathogenesis of HNSCC. Inhibition of epidermal growth factor receptor has been identified as an effective target for the treatment of HNSCC. Many phytochemicals have emerged as potential new drugs for the treatment of HNSCC. A systematic search was conducted for research articles published in PubMed, and Medline on relevant aspects. This review provides an overview of the available literature and reports highlighting the in vitro effects of phytochemicals on epidermal growth factor in various HNSCC cell models and in vivo in animal models and emphasizes the importance of epidermal growth factor as a current therapeutic target for HNSCC. Based on our review, we conclude that phytochemicals targeting the epidermal growth factor receptor are potentially effective candidates for the development of new drugs for the treatment of HNSCC. It provides an idea for further development and application of herbal medicines for cancer treatment.
Collapse
Affiliation(s)
- Shaling Li
- The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Longmatan District, Luzhou City, Sichuan Province, China
| | | |
Collapse
|
13
|
Küçük U, Kırılmaz B, Kaya H, Akşit E, Arslan K. Is elabela/toddler a poor prognostic marker in heart failure patients? Hippokratia 2023; 27:126-131. [PMID: 39372325 PMCID: PMC11451502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Background Elabela/toddler (ELA-32) is a recently identified endogenous apelin receptor ligand. ELA levels are known to rise in heart failure (HF) patients. However, the association between elevated ELA levels and prognosis in these patients remains unknown. We aimed to investigate whether ELA plasma levels are correlated with prognosis in heart failure patients with reduced ejection fraction (HFrEF). Methods This case-control cross-sectional study enrolled 150 patients, including 73 HFrEF patients and 77 age- and gender-matched healthy volunteers. We collected a blood sample at hospital admission to measure ELA-32 levels. The study endpoint was cardiovascular mortality or HF-related hospitalization. We followed up all patients in the study for a mean of 7.48 ± 2.73 months. Results In patients with HFrEF, ELA-32 levels were higher than those in controls. The levels of ELA-32 showed a significant increase at advanced New York Heart Association stages. In the receiver operating characteristics curve analysis, a cut-off value of the serum ELA-32 level of 8.25 ng/mL showed a sensitivity of 76 % and specificity of 82 % for predicting the study endpoint [area under the curve: 0.84; 95 % confidence interval (CI): 0.72-0.98; p <0.001]. Cardiovascular mortality (p =0.042) and HF-related hospitalization (p <0.001) were statistically more significant in patients with ELA-32 levels greater than 8.25. Age [Hazard ratio (HR) =1.023; 95 % CI: 0.964-1.230, p =0.039], N-terminal pro-brain natriuretic peptide (HR =1.300; 95 % CI: 1.017-1.874, p =0.017), left ventricular end-diastolic volume (HR =1.142; 95 % CI 1.022-1.547, p =0.028), and ELA-32 ≥8.25 (HR =2.556; 95 % CI: 1.078-3.941, p <0.001) remained independently associated with the risk of study endpoint. Conclusion For the first time, HF-related hospitalizations and cardiovascular mortality are independently associated with increased ELA-32 levels in patients with HFrEF. HIPPOKRATIA 2023, 27 (4):126-131.
Collapse
Affiliation(s)
- U Küçük
- Department of Cardiology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - B Kırılmaz
- Department of Cardiology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - H Kaya
- Department of Cardiology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - E Akşit
- Department of Cardiology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - K Arslan
- Department of Cardiology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
14
|
Biwer LA, Lu Q, Ibarrola J, Stepanian A, Man JJ, Carvajal BV, Camarda ND, Zsengeller Z, Skurnik G, Seely EW, Karumanchi SA, Jaffe IZ. Smooth Muscle Mineralocorticoid Receptor Promotes Hypertension After Preeclampsia. Circ Res 2023; 132:674-689. [PMID: 36815487 PMCID: PMC10119809 DOI: 10.1161/circresaha.122.321228] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 02/16/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND Preeclampsia is a syndrome of high blood pressure (BP) with end organ damage in late pregnancy that is associated with high circulating soluble VEGF receptor (sFlt1 [soluble Fms-like tyrosine kinase 1]). Women exposed to preeclampsia have a substantially increased risk of hypertension after pregnancy, but the mechanism remains unknown, leaving a missed interventional opportunity. After preeclampsia, women have enhanced sensitivity to hypertensive stress. Since smooth muscle cell mineralocorticoid receptors (SMC-MR) are activated by hypertensive stimuli, we hypothesized that high sFlt1 exposure in pregnancy induces a postpartum state of enhanced SMC-MR responsiveness. METHODS Postpartum BP response to high salt intake was studied in women with prior preeclampsia. MR transcriptional activity was assessed in vitro in sFlt1-treated SMC by reporter assays and PCR. Preeclampsia was modeled by transient sFlt1 expression in pregnant mice. Two months post-partum, mice were exposed to high salt and then to AngII (angiotensin II) and BP and vasoconstriction were measured. RESULTS Women exposed to preeclampsia had significantly enhanced salt sensitivity of BP verses those with a normotensive pregnancy. sFlt1 overexpression during pregnancy in mice induced elevated BP and glomerular endotheliosis, which resolved post-partum. The sFlt1 exposed post-partum mice had significantly increased BP response to 4% salt diet and to AngII infusion. In vitro, SMC-MR transcriptional activity in response to aldosterone or AngII was significantly increased after transient exposure to sFlt1 as was aldosterone-induced expression of AngII type 1 receptor. Post-partum, SMC-MR-KO mice were protected from the enhanced response to hypertensive stimuli after preeclampsia. Mechanistically, preeclampsia mice exposed to postpartum hypertensive stimuli develop enhanced aortic stiffness, microvascular myogenic tone, AngII constriction, and AngII type 1 receptor expression, all of which were prevented in SMC-MR-KO littermates. CONCLUSIONS These data support that sFlt1-induced vascular injury during preeclampsia produces a persistent state of enhanced sensitivity of SMC-MR to activation. This contributes to postpartum hypertension in response to common stresses and supports testing of MR antagonism to mitigate the increased cardiovascular risk in women after PE.
Collapse
Affiliation(s)
- Lauren A. Biwer
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
| | - Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
| | - Alec Stepanian
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA
| | - Joshua J. Man
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA
| | - Brigett V. Carvajal
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA
| | - Nicholas D. Camarda
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA
| | | | | | - Ellen W. Seely
- Division of Endocrinology, Brigham and Women’s Hospital, Boston MA
| | - S. Ananth Karumanchi
- Department of Medicine, Beth Israel Deaconess Hospital, Boston MA
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles CA
| | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA
| |
Collapse
|
15
|
Peng Y, Liu Z, Fu G, Zhao B, Gong M, Lu Z, Zhou Y, Chen L, Su H, Lou W, Chen G, He X, Gu J, Kong J. Identification microenvironment immune features and key genes in elderly stroke patients. Medicine (Baltimore) 2023; 102:e33108. [PMID: 36862915 PMCID: PMC9981407 DOI: 10.1097/md.0000000000033108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND The purpose of this study was to identify the signaling pathways and immune microenvironments related to elderly stroke patients. METHODS We downloaded the public transcriptome data (GSE37587) from the gene expression omnibus and divided the patients into young and old groups and identified differentially expressed genes (DEGs). Gene ontology function analysis, Kyoto encyclopedia of genes and genomes pathway analysis, and gene set enrichment analysis (GSEA) were performed. A protein-protein interaction network was constructed and hub genes were identified. Gene-miRNA, gene-TF, and gene-drug networks were constructed using the network analyst database. The immune infiltration score was evaluated using single-sample gene set enrichment analysis GSEA, its correlation with age was computed and visualized using R software. RESULTS We identified 240 DEGs, including 222 upregulated and 18 downregulated DEGs. Gene ontology enrichment was significantly enriched in response to the virus, type I interferon signaling pathway, cytological component, focal adhesion, cell-substrate adherents junction, and the cytosolic ribosome. GSEA identified the following mechanisms: heme metabolism, interferon gamma response, and interferon alpha response. Ten hub genes included interferon alpha-inducible protein 27, human leucocyte antigen-G, interferon-induced protein with tetratricopeptide repeats 2, 2'-5'-oligoadenylate synthetase 2, interferon alpha-inducible protein 6, interferon alpha-inducible protein 44-like, interferon-induced protein with tetratricopeptide repeats 3, interferon regulatory factor 5, myxovirus resistant 1, and interferon-induced protein with tetratricopeptide repeats 1. Quantitative analysis of immune infiltration showed that increased age was significantly positively correlated with myeloid-derived suppressor cells and natural killer T cells, and negatively correlated with immature dendritic cells. CONCLUSION The present research could help us better understand the molecular mechanisms and immune microenvironment of elderly patients with stroke.
Collapse
Affiliation(s)
- Yisheng Peng
- Department of Radiological Intervention, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, P.R. China
| | - Zhengli Liu
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Guanqi Fu
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Boxiang Zhao
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Maofeng Gong
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Zhaoxuan Lu
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Yangyi Zhou
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Liang Chen
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Haobo Su
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Wensheng Lou
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Guoping Chen
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Xu He
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Jianping Gu
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Jie Kong
- Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
- * Correspondence: Jie Kong, Department of Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China (e-mail: )
| |
Collapse
|
16
|
Li T, Meng Y, Ding P, Wang H, Liu J, Xia C, Chen Y, Li J. Pathological implication of CaMKII in NF-κB pathway and SASP during cardiomyocytes senescence. Mech Ageing Dev 2023; 209:111758. [PMID: 36462537 DOI: 10.1016/j.mad.2022.111758] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 11/06/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022]
Abstract
Senescence-associated secretory phenotype (SASP) could be developed during heart ageing. But the role of SASP in cardiomyocytes senescence and its molecular mechanism remains undetermined. In this study, we observed elevated Ca2+/calmodulin -dependent protein kinase II (CaMKII) activation in both physiological aged heart and premature senescent cardiomyocytes. Notably, we confirmed the gradual SASP development induced by NF-κB activation in long-term cultured cardiomyocytes. Transgenic inhibition of CaMKII in mice (AC3-I mice) alleviated the NF-κB activation, chronic sterile inflammation and ageing-associated cardiomyopathy. Correspondingly, pharmacological inhibition of CaMKII with KN93 mitigated SASP and hindered cardiomyocytes senescence. Meanwhile, increased NF-κB activation and exacerbated cardiomyocytes senescence were observed with transgenic CaMKII activation. Collectively, our results indicated that the increased CaMKII activation accompanying ageing could aggravate NF-κB activation and SASP development and facilitate cardiomyocytes senescence and heart ageing.
Collapse
Affiliation(s)
- Ting Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yidi Meng
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peiwu Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongfei Wang
- Department of Cardiac Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaorui Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingdong Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
17
|
Forte M, Rodolico D, Ameri P, Catalucci D, Chimenti C, Crotti L, Schirone L, Pingitore A, Torella D, Iacovone G, Valenti V, Schiattarella GG, Perrino C, Sciarretta S. Molecular mechanisms underlying the beneficial effects of exercise and dietary interventions in the prevention of cardiometabolic diseases. J Cardiovasc Med (Hagerstown) 2022; 24:e3-e14. [PMID: 36729582 DOI: 10.2459/jcm.0000000000001397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cardiometabolic diseases still represent a major cause of mortality worldwide. In addition to pharmacological approaches, lifestyle interventions can also be adopted for the prevention of these morbid conditions. Lifestyle changes include exercise and dietary restriction protocols, such as calorie restriction and intermittent fasting, which were shown to delay cardiovascular ageing and elicit health-promoting effects in preclinical models of cardiometabolic diseases. Beneficial effects are mediated by the restoration of multiple molecular mechanisms in heart and vessels that are compromised by metabolic stress. Exercise and dietary restriction rescue mitochondrial dysfunction, oxidative stress and inflammation. They also improve autophagy. The result of these effects is a marked improvement of vascular and heart function. In this review, we provide a comprehensive overview of the molecular mechanisms involved in the beneficial effects of exercise and dietary restriction in models of diabetes and obesity. We also discuss clinical studies and gap in animal-to-human translation.
Collapse
Affiliation(s)
- Maurizio Forte
- Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli
| | - Daniele Rodolico
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico.,Department of Internal Medicine, University of Genova, Genova
| | - Daniele Catalucci
- Humanitas Research Hospital, IRCCS, Rozzano.,National Research Council, Institute of Genetic and Biomedical Research - UOS, Milan
| | - Cristina Chimenti
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome
| | - Lia Crotti
- Istituto Auxologico Italiano, IRCCS, Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital.,Department of Medicine and Surgery, Università Milano-Bicocca, Milan
| | - Leonardo Schirone
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina
| | - Annachiara Pingitore
- Department of General and Specialistic Surgery 'Paride Stefanini' Sapienza University of Rome
| | - Daniele Torella
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro
| | | | | | - Gabriele G Schiattarella
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Cinzia Perrino
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Sebastiano Sciarretta
- Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli.,Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina
| | | |
Collapse
|
18
|
Yu H, Liao K, Hu Y, Lv D, Luo M, Liu Q, Huang L, Luo S. Role of the cGAS-STING Pathway in Aging-related Endothelial Dysfunction. Aging Dis 2022; 13:1901-1918. [PMID: 36465181 PMCID: PMC9662267 DOI: 10.14336/ad.2022.0316] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/16/2022] [Indexed: 07/30/2023] Open
Abstract
Endothelial dysfunction develops gradually with age, and is the foundation of many age-related diseases in the elderly. The purpose of this study was to investigate the role of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway in aging-related endothelial dysfunction. Endothelial functional parameters and biochemical indices of vascular function were examined in 2-, 6-, 12- and 24-month-old mice. Then, 6-month-old mice were administered RU.521, a specific inhibitor of cGAS, for 6 months, and endothelial functional parameters and biochemical indices of vascular function were re-examined. An in vitro model of cell senescence was established by treating human aortic endothelial cells (HAECs) with D-Galactose (D-GAL). Using inhibitors or siRNA interference, cGAS and STING were suppressed or silenced in senescent HAECs, and changes in the expression of eNOS, the senescence markers, p53, p21 and p16, components of the cGAS-STING pathway and Senescence-Associated β-galactosidase (SA-β-gal) staining were examined. Finally, cGAS, STING and p-IRF3 levels were measured in aorta tissue sections from eight patients. A decline in endothelial function, up-regulation of p53, p21 and p16 expression, and activation of the cGAS-STING pathway were observed in aging mice. Inhibition of cGAS was found to improve endothelial function and reverse the increased expression of aging markers. Our in vitro data demonstrated that D-GAL induced a decrease in eNOS expression and cell senescence, which could be partly reversed by cGAS inhibitor, STING inhibitor, siRNA-cGAS and siRNA-STING treatment. Higher expression levels of cGAS, STING and p-IRF3 were observed in aged human aortic intima tissue compared to young aortic intima tissue. Our study demonstrated that activation of the cGAS-STING pathway played a vital role in aging-related endothelial dysfunction. Thus, the cGAS-STING pathway may be a potential target for the prevention of cardiovascular diseases in the elderly.
Collapse
Affiliation(s)
- Huilin Yu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Ke Liao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Yu Hu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Dingyi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Qian Liu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Longxiang Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Suxin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Sahinturk S, Demirel S, Ozyener F, Isbil N. Vascular Functional Effect Mechanisms of Elabela in Rat Thoracic Aorta. Ann Vasc Surg 2022; 84:381-397. [PMID: 35472496 DOI: 10.1016/j.avsg.2022.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Elabela is a recently discovered peptide hormone. The present study aims to investigate the vasorelaxant effect mechanisms of elabela in the rat thoracic aorta. METHODS The vascular rings obtained from the thoracic aortas of the male Wistar albino rats were placed in the isolated tissue bath system. Resting tension was set to 1 gram. After the equilibration period, the vessel rings were contracted with phenylephrine or potassium chloride. Once a stable contraction was achieved, elabela-32 was applied cumulatively (10-9-10-6 molar) to the vascular rings. The experimental protocol was repeated in the presence of specific signaling pathway inhibitors or potassium channel blockers to determine the effect mechanisms of elabela. RESULTS Elabela showed a significant vasorelaxant effect in a concentration-dependent manner (P < 0.001). The vasorelaxant effect level of elabela was significantly reduced by the apelin receptor antagonist F13A, cyclooxygenase inhibitor indomethacin, adenosine monophosphate-activated protein kinase inhibitor dorsomorphin, protein kinase C inhibitor bisindolmaleimide, large-conductance calcium-activated potassium channel blocker iberiotoxin, and intermediate-conductance calcium-activated potassium channel blocker TRAM-34 (P < 0.001). However, the vasorelaxant effect level of elabela was not significantly affected by the endothelial nitric oxide synthase inhibitor nitro-L-arginine methyl ester and mitogen-activated protein kinase inhibitor U0126. CONCLUSIONS Elabela exhibits a prominent vasodilator effect in rat thoracic aorta. Apelin receptor, prostanoids, adenosine monophosphate-activated protein kinase, protein kinase C, and calcium-activated potassium channels are involved in the vasorelaxant effect mechanisms of elabela.
Collapse
Affiliation(s)
- Serdar Sahinturk
- Physiology Department, Bursa Uludag University Medicine School, Bursa, Turkey.
| | - Sadettin Demirel
- Physiology Department, Bursa Uludag University Medicine School, Bursa, Turkey
| | - Fadil Ozyener
- Physiology Department, Bursa Uludag University Medicine School, Bursa, Turkey
| | - Naciye Isbil
- Physiology Department, Bursa Uludag University Medicine School, Bursa, Turkey
| |
Collapse
|
20
|
Versaci F, Valenti V, Forte M, Cammisotto V, Nocella C, Bartimoccia S, Schirone L, Schiavon S, Vecchio D, D’Ambrosio L, Spinosa G, D’Amico A, Chimenti I, Violi F, Frati G, Pignatelli P, Sciarretta S, Pastori D, Carnevale R. Aging-Related Decline of Autophagy in Patients with Atrial Fibrillation-A Post Hoc Analysis of the ATHERO-AF Study. Antioxidants (Basel) 2022; 11:antiox11040698. [PMID: 35453383 PMCID: PMC9030744 DOI: 10.3390/antiox11040698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/26/2022] [Accepted: 03/31/2022] [Indexed: 01/02/2023] Open
Abstract
Background: Aging is an independent risk factor for cardiovascular diseases. The autophagy process may play a role in delaying aging and improving cardiovascular function in aging. Data regarding autophagy in atrial fibrillation (AF) patients are lacking. Methods: A post hoc analysis of the prospective ATHERO-AF cohort study, including 150 AF patients and 150 sex- and age-matched control subjects (CS), was performed. For the analysis, the population was divided into three age groups: <50−60, 61−70, and >70 years. Oxidative stress (Nox2 activity and hydrogen peroxide, H2O2), platelet activation (PA) by sP-selectin and CD40L, endothelial dysfunction (nitric oxide, NO), and autophagy parameters (P62 and ATG5 levels) were assessed. Results: Nox2 activity and H2O2 production were higher in the AF patients than in the CS; conversely, antioxidant capacity was decreased in the AF patients compared to the CS, as was NO production. Moreover, sP-selectin and CD40L were higher in the AF patients than in the CS. The autophagy process was also significantly impaired in the AF patients. We found a significant difference in oxidative stress, PA, NO production, and autophagy across the age groups. Autophagy markers correlated with oxidative stress, PA, and endothelial dysfunction in both groups. Conclusions: This study provides evidence that the autophagy process may represent a mechanism for increased cardiovascular risk in the AF population.
Collapse
Affiliation(s)
- Francesco Versaci
- Department of Cardiology, Santa Maria Goretti Hospital, 04100 Latina, Italy; (F.V.); (V.V.)
| | - Valentina Valenti
- Department of Cardiology, Santa Maria Goretti Hospital, 04100 Latina, Italy; (F.V.); (V.V.)
| | - Maurizio Forte
- IRCCS Neuromed, 86077 Pozzilli, Italy; (M.F.); (G.F.); (S.S.)
| | - Vittoria Cammisotto
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (C.N.); (P.P.)
| | - Cristina Nocella
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (C.N.); (P.P.)
| | - Simona Bartimoccia
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Leonardo Schirone
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Sonia Schiavon
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Daniele Vecchio
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Luca D’Ambrosio
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Giulia Spinosa
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Alessandra D’Amico
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy;
| | - Isotta Chimenti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | | | - Giacomo Frati
- IRCCS Neuromed, 86077 Pozzilli, Italy; (M.F.); (G.F.); (S.S.)
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (C.N.); (P.P.)
- Mediterranea Cardiocentro, 80122 Naples, Italy;
| | - Sebastiano Sciarretta
- IRCCS Neuromed, 86077 Pozzilli, Italy; (M.F.); (G.F.); (S.S.)
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
| | - Daniele Pastori
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (C.N.); (P.P.)
- Correspondence: (D.P.); (R.C.); Tel.: +39-0649970941 (D.P.); +39-07731757245 (R.C.); Fax: +39-0649972309 (D.P.); +39-07731757245 (R.C.)
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (L.S.); (S.S.); (D.V.); (L.D.); (G.S.); (I.C.)
- Mediterranea Cardiocentro, 80122 Naples, Italy;
- Correspondence: (D.P.); (R.C.); Tel.: +39-0649970941 (D.P.); +39-07731757245 (R.C.); Fax: +39-0649972309 (D.P.); +39-07731757245 (R.C.)
| |
Collapse
|
21
|
Li HM, Liu X, Meng ZY, Wang L, Zhao LM, Chen H, Wang ZX, Cui H, Tang XQ, Li XH, Han WN, Bai X, Lin Y, Liu H, Zhang Y, Yang BF. Kanglexin delays heart aging by promoting mitophagy. Acta Pharmacol Sin 2022; 43:613-623. [PMID: 34035486 PMCID: PMC8888756 DOI: 10.1038/s41401-021-00686-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Heart aging is characterized by structural and diastolic dysfunction of the heart. However, there is still no effective drug to prevent and treat the abnormal changes in cardiac function caused by aging. Here, we present the preventive effects of emodin and its derivative Kanglexin (KLX) against heart aging. We found that the diastolic dysfunction and cardiac remodeling in mice with D-galactose (D-gal)-induced aging were markedly mitigated by KLX and emodin. In addition, the senescence of neonatal mouse cardiomyocytes induced by D-gal was also reversed by KLX and emodin treatment. However, KLX exhibited better anti-heart aging effects than emodin at the same dose. Dysregulated mitophagy was observed in aging hearts and in senescent neonatal mouse cardiomyocytes, and KLX produced a greater increase in mitophagy than emodin. The mitophagy-promoting effects of KLX and emodin were ascribed to their abilities to enhance the protein stability of Parkin, a key modulator in mitophagy, with different potencies. Molecular docking and SPR analysis demonstrated that KLX has a higher affinity for the ubiquitin-like (UBL) domain of Parkin than emodin. The UBL domain might contribute to the stabilizing effects of KLX on Parkin. In conclusion, this study identifies KLX and emodin as effective anti-heart aging drugs that activate Parkin-mediated mitophagy and outlines their putative therapeutic importance.
Collapse
Affiliation(s)
- Hui-min Li
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Xin Liu
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Zi-yu Meng
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Lei Wang
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Li-min Zhao
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Hui Chen
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Zhi-xia Wang
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Hao Cui
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Xue-qing Tang
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Xiao-han Li
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Wei-na Han
- grid.410736.70000 0001 2204 9268Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Xue Bai
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Yuan Lin
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Heng Liu
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Yong Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China. .,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, 150086, China. .,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, 150086, China.
| | - Bao-feng Yang
- grid.410736.70000 0001 2204 9268Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China ,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, 150086 China ,grid.1008.90000 0001 2179 088XDepartment of Pharmacology and Therapeutics, Melbourne School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences University of Melbourne, Melbourne, Australia
| |
Collapse
|
22
|
Jansen HJ, Moghtadaei M, Rafferty SA, Rose RA. Loss of natriuretic peptide receptor C enhances sinoatrial node dysfunction in aging and frail mice. J Gerontol A Biol Sci Med Sci 2021; 77:902-908. [PMID: 34865023 DOI: 10.1093/gerona/glab357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Indexed: 11/14/2022] Open
Abstract
Heart rate is controlled by the sinoatrial node (SAN). SAN dysfunction is highly prevalent in aging; however, not all individuals age at the same rate. Rather, health status during aging is affected by frailty. Natriuretic peptides regulate SAN function in part by activating natriuretic peptide receptor C (NPR-C). The impacts of NPR-C on HR and SAN function in aging and as a function of frailty are unknown. Frailty was measured in aging wildtype (WT) and NPR-C knockout (NPR-C -/-) mice using a mouse clinical frailty index (FI). HR and SAN structure and function were investigated using intracardiac electrophysiology in anesthetized mice, high-resolution optical mapping in intact atrial preparations, histology and molecular biology. NPR-C -/- mice rapidly became frail leading to shortened lifespan. HR and SAN recovery time were increased in older vs. younger mice and this was exacerbated in NPR-C -/- mice; however, there was substantial variability among age groups and genotypes. HR and SAN recovery time were correlated with FI score and fell along a continuum regardless of age or genotype. Optical mapping demonstrates impairments in SAN function that were also strongly correlated with FI score. SAN fibrosis was increased in aged and NPR-C -/- mice and was graded by FI score. Loss of NPR-C results in accelerated aging due to a rapid decline in health status in association with impairments in HR and SAN function. Frailty assessment was effective and often better able to distinguish aging-dependent changes in SAN function in the setting of shorted lifespan due to loss of NPR-C.
Collapse
Affiliation(s)
- Hailey J Jansen
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, Calgary, Alberta, Canada
| | - Motahareh Moghtadaei
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, Calgary, Alberta, Canada
| | - Sara A Rafferty
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Robert A Rose
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
23
|
Voglhuber J, Ljubojevic-Holzer S, Abdellatif M, Sedej S. Targeting Cardiovascular Risk Factors Through Dietary Adaptations and Caloric Restriction Mimetics. Front Nutr 2021; 8:758058. [PMID: 34660673 PMCID: PMC8514725 DOI: 10.3389/fnut.2021.758058] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022] Open
Abstract
The average human life expectancy continues to rise globally and so does the prevalence and absolute burden of cardiovascular disease. Dietary restriction promotes longevity and improves various cardiovascular risk factors, including hypertension, obesity, diabetes mellitus, and metabolic syndrome. However, low adherence to caloric restriction renders this stringent dietary intervention challenging to adopt as a standard practice for cardiovascular disease prevention. Hence, alternative eating patterns and strategies that recapitulate the salutary benefits of caloric restriction are under intense investigation. Here, we first provide an overview of alternative interventions, including intermittent fasting, alternate-day fasting and the Mediterranean diet, along with their cardiometabolic effects in animal models and humans. We then present emerging pharmacological alternatives, including spermidine, NAD+ precursors, resveratrol, and metformin, as promising caloric restriction mimetics, and briefly touch on the mechanisms underpinning their cardiometabolic and health-promoting effects. We conclude that implementation of feasible dietary approaches holds the promise to attenuate the burden of cardiovascular disease and facilitate healthy aging in humans.
Collapse
Affiliation(s)
- Julia Voglhuber
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Senka Ljubojevic-Holzer
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Graz, Austria
- Centre de Recherche des Cordeliers, Equipe labellisée par La Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institute Universitaire de France, Paris, France
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
| |
Collapse
|
24
|
Dynamic Crosstalk between Vascular Smooth Muscle Cells and the Aged Extracellular Matrix. Int J Mol Sci 2021; 22:ijms221810175. [PMID: 34576337 PMCID: PMC8468233 DOI: 10.3390/ijms221810175] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 01/15/2023] Open
Abstract
Vascular aging is accompanied by the fragmentation of elastic fibers and collagen deposition, leading to reduced distensibility and increased vascular stiffness. A rigid artery facilitates elastin to degradation by MMPs, exposing vascular cells to greater mechanical stress and triggering signaling mechanisms that only exacerbate aging, creating a self-sustaining inflammatory environment that also promotes vascular calcification. In this review, we highlight the role of crosstalk between smooth muscle cells and the vascular extracellular matrix (ECM) and how aging promotes smooth muscle cell phenotypes that ultimately lead to mechanical impairment of aging arteries. Understanding the underlying mechanisms and the role of associated changes in ECM during aging may contribute to new approaches to prevent or delay arterial aging and the onset of cardiovascular diseases.
Collapse
|
25
|
Abstract
Heart failure (HF) is a growing epidemic with high morbidity and mortality at an international scale. The apelin-APJ receptor pathway has been implicated in HF, making it a promising therapeutic target. APJ has been shown to be activated by a novel endogenous peptide ligand known as Elabela (ELA, also called Toddler or Apela), with a critical role in cardiac development and function. Activation of the ELA-APJ receptor axis exerts a wide range of physiological effects, including depressor response, positive inotropic action, diuresis, anti-inflammatory, anti-fibrotic, and anti-remodeling, leading to its cardiovascular protection. The ELA-APJ axis is essential for diverse biological processes and has been shown to regulate fluid homeostasis, myocardial contractility, vasodilation, angiogenesis, cellular differentiation, apoptosis, oxidative stress, cardiorenal fibrosis, and dysfunction. The beneficial effects of the ELA-APJ receptor system are well-established by treating hypertension, myocardial infarction, and HF. Additionally, administration of ELA protects human embryonic stem cells against apoptosis and stress-induced cell death and promotes survival and self-renewal in an APJ-independent manner (X receptor) via the phosphatidylinositol 3-kinase/Akt pathway, which may provide a new therapeutic approach for HF. Thus, targeting the ELA-APJ axis has emerged as a pre-warning biomarker and a novel therapeutic approach against progression of HF. An increased understanding of cardiovascular actions of ELA will help to develop effective interventions. This article gives an overview of the characteristics of the ELA-apelin-APJ axis and summarizes the current knowledge on its cardioprotective roles, potential mechanisms, and prospective application for acute and chronic HF.
Collapse
Affiliation(s)
- Zheng Ma
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Juan-Juan Song
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Sara Martin
- Santa Rosa Family Medicine Residency, Santa Rosa, CA, 95403, USA
| | - Xin-Chun Yang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Jiu-Chang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
26
|
Czibik G, Mezdari Z, Murat Altintas D, Bréhat J, Pini M, d'Humières T, Delmont T, Radu C, Breau M, Liang H, Martel C, Abatan A, Sarwar R, Marion O, Naushad S, Zhang Y, Halfaoui M, Suffee N, Morin D, Adnot S, Hatem S, Yavari A, Sawaki D, Derumeaux G. Dysregulated Phenylalanine Catabolism Plays a Key Role in the Trajectory of Cardiac Aging. Circulation 2021; 144:559-574. [PMID: 34162223 DOI: 10.1161/circulationaha.121.054204] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Aging myocardium undergoes progressive cardiac hypertrophy and interstitial fibrosis with diastolic and systolic dysfunction. Recent metabolomics studies shed light on amino acids in aging. The present study aimed to dissect how aging leads to elevated plasma levels of the essential amino acid phenylalanine and how it may promote age-related cardiac dysfunction. METHODS We studied cardiac structure and function, together with phenylalanine catabolism in wild-type (WT) and p21-/- mice (male; 2-24 months), with the latter known to be protected from cellular senescence. To explore phenylalanine's effects on cellular senescence and ectopic phenylalanine catabolism, we treated cardiomyocytes (primary adult rat or human AC-16) with phenylalanine. To establish a role for phenylalanine in driving cardiac aging, WT male mice were treated twice a day with phenylalanine (200 mg/kg) for a month. We also treated aged WT mice with tetrahydrobiopterin (10 mg/kg), the essential cofactor for the phenylalanine-degrading enzyme PAH (phenylalanine hydroxylase), or restricted dietary phenylalanine intake. The impact of senescence on hepatic phenylalanine catabolism was explored in vitro in AML12 hepatocytes treated with Nutlin3a (a p53 activator), with or without p21-targeting small interfering RNA or tetrahydrobiopterin, with quantification of PAH and tyrosine levels. RESULTS Natural aging is associated with a progressive increase in plasma phenylalanine levels concomitant with cardiac dysfunction, whereas p21 deletion delayed these changes. Phenylalanine treatment induced premature cardiac deterioration in young WT mice, strikingly akin to that occurring with aging, while triggering cellular senescence, redox, and epigenetic changes. Pharmacological restoration of phenylalanine catabolism with tetrahydrobiopterin administration or dietary phenylalanine restriction abrogated the rise in plasma phenylalanine and reversed cardiac senescent alterations in aged WT mice. Observations from aged mice and human samples implicated age-related decline in hepatic phenylalanine catabolism as a key driver of elevated plasma phenylalanine levels and showed increased myocardial PAH-mediated phenylalanine catabolism, a novel signature of cardiac aging. CONCLUSIONS Our findings establish a pathogenic role for increased phenylalanine levels in cardiac aging, linking plasma phenylalanine levels to cardiac senescence via dysregulated phenylalanine catabolism along a hepatic-cardiac axis. They highlight phenylalanine/PAH modulation as a potential therapeutic strategy for age-associated cardiac impairment.
Collapse
Affiliation(s)
- Gabor Czibik
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
- Department of Physiology (G.C., T.d'H., S.A., G.D.), AP-HP, Henri Mondor Hospital, FHU-SENEC, Créteil, France
| | - Zaineb Mezdari
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Dogus Murat Altintas
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Juliette Bréhat
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Maria Pini
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Thomas d'Humières
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
- Department of Physiology (G.C., T.d'H., S.A., G.D.), AP-HP, Henri Mondor Hospital, FHU-SENEC, Créteil, France
| | - Thaïs Delmont
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Costin Radu
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
- Department of Cardiac Surgery (C.R.), AP-HP, Henri Mondor Hospital, FHU-SENEC, Créteil, France
| | - Marielle Breau
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Hao Liang
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Cecile Martel
- Mitologics SAS (C.M.), Université Paris-Est Créteil, France
| | - Azania Abatan
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Rizwan Sarwar
- Experimental Therapeutics, Radcliffe Department of Medicine (R.S., A.Y.), University of Oxford, United Kingdom
| | - Ophélie Marion
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Suzain Naushad
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Yanyan Zhang
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Maissa Halfaoui
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Nadine Suffee
- Sorbonne Universités, INSERM UMR_S1166, Faculté de Médecine UPMC, Paris, France (N.S., S.H.)
- Institute of Cardiometabolism and Nutrition, ICAN, Paris, France (N.S., S.H.)
| | - Didier Morin
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Serge Adnot
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
- Department of Physiology (G.C., T.d'H., S.A., G.D.), AP-HP, Henri Mondor Hospital, FHU-SENEC, Créteil, France
| | - Stéphane Hatem
- Sorbonne Universités, INSERM UMR_S1166, Faculté de Médecine UPMC, Paris, France (N.S., S.H.)
- Institute of Cardiometabolism and Nutrition, ICAN, Paris, France (N.S., S.H.)
| | - Arash Yavari
- Experimental Therapeutics, Radcliffe Department of Medicine (R.S., A.Y.), University of Oxford, United Kingdom
- Wellcome Centre for Human Genetics (A.Y.), University of Oxford, United Kingdom
| | - Daigo Sawaki
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
| | - Geneviève Derumeaux
- INSERM (L'Institut National de la Santé et de la Recherche Médicale) U955 (G.C., Z.M., D.M.A., J.B., M.P., T.d'H., T.D., C.R., M.B., H.L., A.A., O.M., S.N., Y.Z., M.H., D.M., S.A., D.S., G.D.), Université Paris-Est Créteil, France
- Department of Physiology (G.C., T.d'H., S.A., G.D.), AP-HP, Henri Mondor Hospital, FHU-SENEC, Créteil, France
| |
Collapse
|
27
|
Dorey TW, Jansen HJ, Moghtadaei M, Jamieson KL, Rose RA. Impacts of frailty on heart rate variability in aging mice: Roles of the autonomic nervous system and sinoatrial node. Heart Rhythm 2021; 18:1999-2008. [PMID: 34371195 DOI: 10.1016/j.hrthm.2021.07.069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Heart rate variability (HRV) is determined by intrinsic sinoatrial node (SAN) activity and the autonomic nervous system (ANS). HRV is reduced in aging; however, aging is heterogeneous. Frailty, which can be measured using a frailty index (FI), can quantify health status in aging separately from chronological age. OBJECTIVE The purpose of this study was to investigate the impacts of age and frailty on HRV in mice. METHODS Frailty was measured in aging mice between 10 and 130 weeks of age. HRV was assessed using time domain, frequency domain, and Poincaré plot analyses in anesthetized mice at baseline and after ANS blockade, as well as in isolated atrial preparations. RESULTS HRV was reduced in aged mice (90-130 weeks and 50-80 weeks old) compared to younger mice (10-30 weeks old); however, there was substantial variability within age groups. In contrast, HRV was strongly correlated with FI score regardless of chronological age. ANS blockade resulted in reductions in heart rate that were largest in 90- to 130-week-old mice and were correlated with FI score. HRV after ANS blockade or in isolated atrial preparations was increased in aged mice but again showed high variability among age groups. HRV was correlated with FI score after ANS blockade and in isolated atrial preparations. CONCLUSION HRV is reduced in aging mice in association with a shift in sympathovagal balance and increased intrinsic SAN beating variability; however, HRV is highly variable within age groups. HRV was strongly correlated with frailty, which was able to detect differences in HRV separately from chronological age.
Collapse
Affiliation(s)
- Tristan W Dorey
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Hailey J Jansen
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Motahareh Moghtadaei
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - K Lockhart Jamieson
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robert A Rose
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
28
|
Wang L, Ren J. Aging as a risk factor for cardiac surgery: Blunted ischemic-reperfusion stress response? J Card Surg 2021; 36:3641-3642. [PMID: 34250658 DOI: 10.1111/jocs.15806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/21/2022]
Abstract
Biological aging is commonly associated with compromised cardiovascular function. In particular, cardiac aging is featured by unfavorable left ventricular remodeling, loss of compliance, and poor contractile reserve. Among possible contributing factors for cardiovascular aging, dampened autophagy response has received much attention. Recent evidence also noted a decline in ischemia-reperfusion (I/R) response and mitochondrial content in aging. I/R stress is commonly seen in the setting of cardiac surgery representing a clinical dilemma accompanied by possible myocardial injury if not handled correctly. Although overproduction of reactive oxygen species has been indicated to play a role in aging-associated more pronounced I/R injury, little is known for the mechanism of action (in particular the role of autophagy and mitophagy) and possible sequelae of poor I/R stress response in aging.
Collapse
Affiliation(s)
- Lin Wang
- Department of Geriatrics, Xijing Hospital, The Air Force Military Medical University, Xi'an, China
| | - Jun Ren
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA.,Department of Cardiology, Shanghai Institute for Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
29
|
Vaccinium Species (Ericaceae): From Chemical Composition to Bio-Functional Activities. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11125655] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The genus Vaccinium L. (Ericaceae) includes more than 450 species, which mainly grow in cooler areas of the northern hemisphere. Vaccinium species have been used in traditional medicine of different cultures and the berries are widely consumed as food. Indeed, Vaccinium supplement-based herbal medicine and functional food, mainly from V. myrtillus and V. macrocarpon, are used in Europe and North America. Biological studies support traditional uses since, for many Vaccinium components, important biological functions have been described, including antioxidant, antitumor, anti-inflammatory, antidiabetic and endothelium protective activities. Vaccinium components, such as polyphenols, anthocyanins and flavonoids, are widely recognized as modulators of cellular pathways involved in pathological conditions, thus indicating that Vaccinium may be an important source of bioactive molecules. This review aims to better describe the bioactivity of Vaccinium species, focusing on anti-inflammatory and endothelial protective cellular pathways, modulated by their components, to better understand their importance for public health.
Collapse
|
30
|
Kim SH, Monticone RE, McGraw KR, Wang M. Age-associated proinflammatory elastic fiber remodeling in large arteries. Mech Ageing Dev 2021; 196:111490. [PMID: 33839189 PMCID: PMC8154723 DOI: 10.1016/j.mad.2021.111490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/24/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
Elastic fibers are the main components of the extracellular matrix of the large arterial wall. Elastic fiber remodeling is an intricate process of synthesis and degradation of the core elastin protein and microfibrils accompanied by the assembly and disassembly of accessory proteins. Age-related morphological, structural, and functional proinflammatory remodeling within the elastic fiber has a profound effect upon the integrity, elasticity, calcification, amyloidosis, and stiffness of the large arterial wall. An age-associated increase in arterial stiffness is a major risk factor for the pathogenesis of diseases of the large arteries such as hypertensive and atherosclerotic vasculopathy. This mini review is an update on the key molecular, cellular, functional, and structural mechanisms of elastic fiber proinflammatory remodeling in large arteries with aging. Targeting structural and functional integrity of the elastic fiber may be an effective approach to impede proinflammatory arterial remodeling with advancing age.
Collapse
Affiliation(s)
- Soo Hyuk Kim
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Robert E Monticone
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Kimberly R McGraw
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA.
| |
Collapse
|
31
|
Dagher O, Mury P, Thorin-Trescases N, Noly PE, Thorin E, Carrier M. Therapeutic Potential of Quercetin to Alleviate Endothelial Dysfunction in Age-Related Cardiovascular Diseases. Front Cardiovasc Med 2021; 8:658400. [PMID: 33860002 PMCID: PMC8042157 DOI: 10.3389/fcvm.2021.658400] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
The vascular endothelium occupies a catalog of functions that contribute to the homeostasis of the cardiovascular system. It is a physically active barrier between circulating blood and tissue, a regulator of the vascular tone, a biochemical processor and a modulator of coagulation, inflammation, and immunity. Given these essential roles, it comes to no surprise that endothelial dysfunction is prodromal to chronic age-related diseases of the heart and arteries, globally termed cardiovascular diseases (CVD). An example would be ischemic heart disease (IHD), which is the main cause of death from CVD. We have made phenomenal advances in treating CVD, but the aging endothelium, as it senesces, always seems to out-run the benefits of medical and surgical therapies. Remarkably, many epidemiological studies have detected a correlation between a flavonoid-rich diet and a lower incidence of mortality from CVD. Quercetin, a member of the flavonoid class, is a natural compound ubiquitously found in various food sources such as fruits, vegetables, seeds, nuts, and wine. It has been reported to have a wide range of health promoting effects and has gained significant attention over the years. A growing body of evidence suggests quercetin could lower the risk of IHD by mitigating endothelial dysfunction and its risk factors, such as hypertension, atherosclerosis, accumulation of senescent endothelial cells, and endothelial-mesenchymal transition (EndoMT). In this review, we will explore these pathophysiological cascades and their interrelation with endothelial dysfunction. We will then present the scientific evidence to quercetin's anti-atherosclerotic, anti-hypertensive, senolytic, and anti-EndoMT effects. Finally, we will discuss the prospect for its clinical use in alleviating myocardial ischemic injuries in IHD.
Collapse
Affiliation(s)
- Olina Dagher
- Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | - Pauline Mury
- Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | | | - Pierre Emmanuel Noly
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | - Eric Thorin
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | - Michel Carrier
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| |
Collapse
|
32
|
Soares ADS, Trevisol DJ, Schuelter-Trevisol F. Medication discrepancies in a hospital in Southern Brazil: the importance of medication reconciliation for patient safety. BRAZ J PHARM SCI 2021. [DOI: 10.1590/s2175-979020200004181064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Alessandra de Sá Soares
- Universidade do Sul de Santa Catarina, Brasil; Universidade do Sul de Santa Catarina, Brasil
| | - Daisson José Trevisol
- Universidade do Sul de Santa Catarina, Brasil; Universidade do Sul de Santa Catarina, Brasil
| | | |
Collapse
|
33
|
Cicalese S, Okuno K, Eguchi S. Detection of Protein Aggregation and Proteotoxicity Induced by Angiotensin II in Vascular Smooth Muscle Cells. J Cardiovasc Pharmacol 2021; 77:43-48. [PMID: 33079831 DOI: 10.1097/fjc.0000000000000934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/06/2020] [Indexed: 12/26/2022]
Abstract
ABSTRACT Disruption of protein quality control occurs with aging and cardiovascular pathologies including arterial stiffness and hypertension. Angiotensin II (Ang II) is believed to induce endoplasmic reticulum stress in vascular smooth muscle cells (VSMCs), thus contributing to vascular remodeling and dysfunction. However, whether Ang II increases formation of protein aggregates and mediates proteotoxicity in VSMCs remain obscure. Accordingly, this study aimed to establish a quantitative method of protein aggregate detection induced by Ang II and to investigate their potential involvement in inflammatory and senescence responses. Proteostat staining showed increased aggregate numbers per cell on Ang II exposure. Immunoblot analysis further showed an increase in preamyloid oligomer presence in a detergent insoluble protein fraction purified from VSMCs stimulated with Ang II. Moreover, these responses were attenuated by treatment with chemical chaperone, 4-phenylbutyrate. 4-phenylbutyrate further blocked Ang II-induced senescence associated β-galactosidase activity and THP-1 monocyte adhesion in VSMCs. These data suggest that Ang II induces proteotoxicity in VSMCs which likely contributes to aging and inflammation in the vasculature.
Collapse
MESH Headings
- Angiotensin II/toxicity
- Animals
- Cell Adhesion/drug effects
- Cellular Senescence/drug effects
- Endoplasmic Reticulum Stress/drug effects
- Humans
- Male
- Monocytes/drug effects
- Monocytes/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Protein Aggregates
- Protein Aggregation, Pathological
- Rats, Sprague-Dawley
- THP-1 Cells
- Rats
Collapse
Affiliation(s)
- Stephanie Cicalese
- Department of Physiology, Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | | | | |
Collapse
|
34
|
Li X, Lu L, Chen J, Zhang C, Chen H, Huang H. New Insight into the Mechanisms of Ginkgo Biloba Extract in Vascular Aging Prevention. Curr Vasc Pharmacol 2020; 18:334-345. [PMID: 31223090 DOI: 10.2174/1570161117666190621150725] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/07/2019] [Accepted: 05/14/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Aging-associated vascular dysfunction promotes cardiovascular diseases. Recently, Ginkgo biloba extract (GBE) has attracted considerable attention in the prevention of aged vasculature. METHODS This review discusses the pathophysiological alterations in aged vasculature and the underlying mechanisms of GBE in vascular aging suppression. RESULTS Both arterial stiffening and endothelial dysfunction are critical aging-related vascular phenotypes that result in the progression of cardiovascular diseases in the general population. Consistent oxidative stress and inflammatory reaction lead to vascular dysfunction. GBE ameliorates aging-related vascular dysfunction, due to its antioxidant and anti-inflammatory properties. The main effects of GBE in aged vasculature might be associated with the longevity signaling pathways. GBE also attenuates the progression of vascular aging in diabetes mellitus via regulation of glucose and lipid metabolism. CONCLUSION GBE plays an important role in the prevention of vascular aging process. It is a promising therapeutic approach to ameliorate aging-related vascular dysfunction and cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaoxue Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Liuyi Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jie Chen
- Department of Radiation Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chao Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong Chen
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Hui Huang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
35
|
Mechanisms of Vascular Aging, A Geroscience Perspective: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:931-941. [PMID: 32130929 DOI: 10.1016/j.jacc.2019.11.061] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/15/2019] [Accepted: 11/19/2019] [Indexed: 12/25/2022]
Abstract
Age-related pathological alterations of the vasculature have a critical role in morbidity and mortality of older adults. In epidemiological studies, age is the single most important cardiovascular risk factor that dwarfs the impact of traditional risk factors. To develop novel therapeutic interventions for prevention of age-related vascular pathologies, it is crucial to understand the cellular and molecular mechanisms of vascular aging. In this review, shared molecular mechanisms of aging are considered in terms of their contribution to the pathogenesis of macrovascular and microvascular diseases associated with old age. The role of cellular senescence in development of vascular aging phenotypes is highlighted, and potential interventions to prevent senescence and to eliminate senescent cells for prevention of vascular pathologies are presented. The evidence supporting a role for interorgan communication and circulating progeronic and antigeronic factors in vascular aging is discussed.
Collapse
|
36
|
Wang L, Liu S, Pan B, Cai H, Zhou H, Yang P, Wang W. The role of autophagy in abdominal aortic aneurysm: protective but dysfunctional. Cell Cycle 2020; 19:2749-2759. [PMID: 32960711 PMCID: PMC7714418 DOI: 10.1080/15384101.2020.1823731] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/17/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Autophagy, an evolutionarily conserved mechanism that promotes cell survival by recycling nutrients and degrading long-lived proteins and dysfunctional organelles, is an important defense mechanism, and its attenuation has been well documented in senescence and aging-related diseases. Abdominal aortic aneurysm (AAA), a well-known aging-related disease, has been defined as a chronic degenerative process in the abdominal aortic wall; however, the complete mechanism is unknown, and a clinical treatment is lacking. Accumulating evidence has recently revealed that numerous drugs that can induce autophagy are effective in the treatment of AAA. The purpose of this systematic review was to focus on the cross-talk between autophagy and high-risk factors and the potential pathogenesis of AAA to understand not only the host defense and pathogenesis but also potential treatments.
Collapse
Affiliation(s)
- Lei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuai Liu
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Baihong Pan
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huoying Cai
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haiyang Zhou
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pu Yang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
37
|
Sciarretta S, Forte M, Castoldi F, Frati G, Versaci F, Sadoshima J, Kroemer G, Maiuri MC. Caloric restriction mimetics for the treatment of cardiovascular diseases. Cardiovasc Res 2020; 117:1434-1449. [PMID: 33098415 DOI: 10.1093/cvr/cvaa297] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/09/2020] [Indexed: 12/25/2022] Open
Abstract
Caloric restriction mimetics (CRMs) are emerging as potential therapeutic agents for the treatment of cardiovascular diseases. CRMs include natural and synthetic compounds able to inhibit protein acetyltransferases, to interfere with acetyl coenzyme A biosynthesis, or to activate (de)acetyltransferase proteins. These modifications mimic the effects of caloric restriction, which is associated with the activation of autophagy. Previous evidence demonstrated the ability of CRMs to ameliorate cardiac function and reduce cardiac hypertrophy and maladaptive remodelling in animal models of ageing, mechanical overload, chronic myocardial ischaemia, and in genetic and metabolic cardiomyopathies. In addition, CRMs were found to reduce acute ischaemia-reperfusion injury. In many cases, these beneficial effects of CRMs appeared to be mediated by autophagy activation. In the present review, we discuss the relevant literature about the role of different CRMs in animal models of cardiac diseases, emphasizing the molecular mechanisms underlying the beneficial effects of these compounds and their potential future clinical application.
Collapse
Affiliation(s)
- Sebastiano Sciarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 40100 Latina, Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Maurizio Forte
- Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Francesca Castoldi
- Centre de Recherche des Cordeliers, Team "Metabolism, Cancer & Immunity", INSERM UMRS1138, Université de Paris, Sorbonne Université, 75006 Paris, France.,Cell Biology and Metabolomics platforms, Gustave Roussy Cancer Campus, 94805 Villejuif, France
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 40100 Latina, Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Francesco Versaci
- Division of Cardiology, S. Maria Goretti Hospital, 04100 Latina, Italy
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Avenue, G-609, Newark, NJ 07103, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Team "Metabolism, Cancer & Immunity", INSERM UMRS1138, Université de Paris, Sorbonne Université, 75006 Paris, France.,Cell Biology and Metabolomics platforms, Gustave Roussy Cancer Campus, 94805 Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou Jiangsu 215163, China.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, Team "Metabolism, Cancer & Immunity", INSERM UMRS1138, Université de Paris, Sorbonne Université, 75006 Paris, France.,Cell Biology and Metabolomics platforms, Gustave Roussy Cancer Campus, 94805 Villejuif, France
| |
Collapse
|
38
|
The carotid plaque as paradigmatic case of site-specific acceleration of aging process: The microRNAs and the inflammaging contribution. Ageing Res Rev 2020; 61:101090. [PMID: 32474155 DOI: 10.1016/j.arr.2020.101090] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/31/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is considered a chronic inflammatory disease of arteries associated with the aging process. Many risk factors have been identified and they are mainly related to life-styles, gene-environment interactions and socioeconomic status. Carotid and coronary artery diseases are the two major atherosclerotic conditions, being the primary cause of stroke and heart attack, respectively. Nevertheless, carotid plaque assumes particular aspects not only for the specific molecular mechanisms, but also for the types of atheroma which may be associated with a better or a worst prognosis. The identification of circulating blood biomarkers able to distinguish carotid plaque types (stable or vulnerable) is a crucial step for the improvement of adequate therapeutic approaches avoiding or delaying endarterectomy in the oldest old individuals (> 80 years), a population predicted to growth in the next years. The review highlights the most recent knowledge on carotid plaque molecular mechanisms, focusing on microRNAs (miRs), as a site-specific accelerated aging within the conceptual framework of Geroscience for new affordable therapies.
Collapse
|
39
|
Associations of cardiovascular biomarkers and plasma albumin with exceptional survival to the highest ages. Nat Commun 2020; 11:3820. [PMID: 32732919 PMCID: PMC7393489 DOI: 10.1038/s41467-020-17636-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 07/07/2020] [Indexed: 12/22/2022] Open
Abstract
Supercentenarians (those aged ≥110 years) are approaching the current human longevity limit by preventing or surviving major illness. Identifying specific biomarkers conducive to exceptional survival might provide insights into counter-regulatory mechanisms against aging-related disease. Here, we report associations between cardiovascular disease-related biomarkers and survival to the highest ages using a unique dataset of 1,427 oldest individuals from three longitudinal cohort studies, including 36 supercentenarians, 572 semi-supercentenarians (105–109 years), 288 centenarians (100–104 years), and 531 very old people (85–99 years). During follow-up, 1,000 participants (70.1%) died. Overall, N-terminal pro-B-type natriuretic peptide (NT-proBNP), interleukin-6, cystatin C and cholinesterase are associated with all-cause mortality independent of traditional cardiovascular risk factors and plasma albumin. Of these, low NT-proBNP levels are statistically associated with a survival advantage to supercentenarian age. Only low albumin is associated with high mortality across age groups. These findings expand our knowledge on the biology of human longevity. Supercentenarians are approaching the current longevity limit by avoiding or surviving major illness, thus identifying biomarkers for exceptional survival might provide insights into the protection against disease of aging. Here, the authors show low NT-proBNP and high albumin in plasma are the biological correlates of survival to the highest ages.
Collapse
|
40
|
Li T, Mu N, Yin Y, Yu L, Ma H. Targeting AMP-Activated Protein Kinase in Aging-Related Cardiovascular Diseases. Aging Dis 2020; 11:967-977. [PMID: 32765957 PMCID: PMC7390518 DOI: 10.14336/ad.2019.0901] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 09/01/2019] [Indexed: 12/11/2022] Open
Abstract
Aging is a pivotal risk factor for developing cardiovascular diseases (CVD) due to the lifelong exposure to various risk factors that may affect the heart and vasculature during aging. AMP-activated protein kinase (AMPK), a serine/threonine protein kinase, is a pivotal endogenous energy regulator that protects against various pathological alterations. In this report, we first introduced the protective mechanisms of AMPK signaling in myocardium, such as oxidative stress, apoptosis, inflammation, autophagy and inflammatory response. Next, we introduced the potential correlation between AMPK and cardiac aging. Then, we highlighted the roles of AMPK signaling in cardiovascular diseases, including myocardial ischemia, cardiomyopathy, and heart failure. Lastly, some potential directions and further perspectives were expanded. The information extends our understanding on the protective roles of AMPK in myocardial aging, which may contribute to the design of drug targets and sheds light on potential treatments of AMPK for aging-related CVD.
Collapse
Affiliation(s)
- Tian Li
- 1Department of physiology and pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Nan Mu
- 1Department of physiology and pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yue Yin
- 1Department of physiology and pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Lu Yu
- 2Department of pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Heng Ma
- 1Department of physiology and pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
41
|
Forte M, Stanzione R, Cotugno M, Bianchi F, Marchitti S, Rubattu S. Vascular ageing in hypertension: Focus on mitochondria. Mech Ageing Dev 2020; 189:111267. [PMID: 32473170 DOI: 10.1016/j.mad.2020.111267] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/25/2022]
Abstract
Hypertension is a common age-related disease, along with vascular and neurodegenerative diseases. Vascular ageing increases during hypertension, but hypertension itself accelerates vascular ageing, thus creating a vicious circle. Vascular stiffening, endothelial dysfunction, impaired contractility and vasorelaxation are the main alterations related to vascular ageing, as a consequence of vascular smooth muscle and endothelial cells senescence. Several molecular mechanisms have been involved into the functional and morphological changes of the aged vessels. Among them, oxidative stress, inflammation, extracellular matrix deregulation and mitochondrial dysfunction are the best characterized. In the present review, we discuss relevant literature about the biology of vascular and cerebrovascular ageing with a particular focus on mitochondria signalling. We underline the therapeutic strategies, able to improve mitochondrial health, which may represent a promising tool to decrease vascular dysfunction associated with ageing and hypertension-related complications.
Collapse
Affiliation(s)
- Maurizio Forte
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy
| | | | - Maria Cotugno
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy
| | - Franca Bianchi
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy
| | | | - Speranza Rubattu
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy.
| |
Collapse
|
42
|
Wang M, Monticone RE, McGraw KR. Proinflammation, profibrosis, and arterial aging. Aging Med (Milton) 2020; 3:159-168. [PMID: 33103036 PMCID: PMC7574637 DOI: 10.1002/agm2.12099] [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: 01/13/2020] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/18/2022] Open
Abstract
Aging is a major risk factor for quintessential cardiovascular diseases, which are closely related to arterial proinflammation. The age-related alterations of the amount, distribution, and properties of the collagen fibers, such as cross-links and degradation in the arterial wall, are the major sequelae of proinflammation. In the aging arterial wall, collagen types I, II, and III are predominant, and are mainly produced by stiffened vascular smooth muscle cells (VSMCs) governed by proinflammatory signaling, leading to profibrosis. Profibrosis is regulated by an increase in the proinflammatory molecules angiotensin II, milk fat globule-EGF-VIII, and transforming growth factor-beta 1 (TGF-β1) signaling and a decrease in the vasorin signaling cascade. The release of these proinflammatory factors triggers the activation of matrix metalloproteinase type II (MMP-2) and activates profibrogenic TGF-β1 signaling, contributing to profibrosis. The age-associated increase in activated MMP-2 cleaves latent TGF-β and subsequently increases TGF-β1 activity leading to collagen deposition in the arterial wall. Furthermore, a blockade of the proinflammatory signaling pathway alleviates the fibrogenic signaling, reduces profibrosis, and prevents arterial stiffening with aging. Thus, age-associated proinflammatory-profibrosis coupling is the underlying molecular mechanism of arterial stiffening with advancing age.
Collapse
Affiliation(s)
- Mingyi Wang
- Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Baltimore Maryland
| | - Robert E Monticone
- Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Baltimore Maryland
| | - Kimberly R McGraw
- Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Baltimore Maryland
| |
Collapse
|
43
|
Nalobin D, Alipkina S, Gaidamaka A, Glukhov A, Khuchua Z. Telomeres and Telomerase in Heart Ontogenesis, Aging and Regeneration. Cells 2020; 9:cells9020503. [PMID: 32098394 PMCID: PMC7072777 DOI: 10.3390/cells9020503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 12/14/2022] Open
Abstract
The main purpose of the review article is to assess the contributions of telomere length and telomerase activity to the cardiac function at different stages of development and clarify their role in cardiac disorders. It has been shown that the telomerase complex and telomeres are of great importance in many periods of ontogenesis due to the regulation of the proliferative capacity of heart cells. The review article also discusses the problems of heart regeneration and the identification of possible causes of dysfunction of telomeres and telomerase.
Collapse
Affiliation(s)
- Denis Nalobin
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russian
- Correspondence: ; Tel.: +7-916-939-0990
| | - Svetlana Alipkina
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russian
| | - Anna Gaidamaka
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russian
| | - Alexander Glukhov
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russian
- Department of Biochemistry, Sechenov First Moscow State Medical University, 119991 Moscow, Russian
| | - Zaza Khuchua
- Department of Biochemistry, Sechenov First Moscow State Medical University, 119991 Moscow, Russian
- Institute of Chemical Biology Ilia State University, 0162 Tbilisi, Georgia
- Division of Molecular and Cardiovascular Biology, Cincinnati Children’s Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
44
|
Calabrese EJ, Bhatia TN, Calabrese V, Dhawan G, Giordano J, Hanekamp YN, Kapoor R, Kozumbo WJ, Leak RK. Cytotoxicity models of Huntington’s disease and relevance of hormetic mechanisms: A critical assessment of experimental approaches and strategies. Pharmacol Res 2019; 150:104371. [DOI: 10.1016/j.phrs.2019.104371] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/17/2022]
|
45
|
Lushchak O, Strilbytska O, Koliada A, Zayachkivska A, Burdyliuk N, Yurkevych I, Storey KB, Vaiserman A. Nanodelivery of phytobioactive compounds for treating aging-associated disorders. GeroScience 2019; 42:117-139. [PMID: 31686375 DOI: 10.1007/s11357-019-00116-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 10/04/2019] [Indexed: 12/15/2022] Open
Abstract
Aging population presents a major challenge for many countries in the world and has made the development of efficient means for healthspan extension a priority task for researchers and clinicians worldwide. Anti-aging properties including antioxidant, anti-inflammatory, anti-tumor, and cardioprotective activities have been reported for various phytobioactive compounds (PBCs) including resveratrol, quercetin, curcumin, catechin, etc. However, the therapeutic potential of orally administered PBCs is limited by their poor stability, bioavailability, and solubility in the gastrointestinal tract. Recently, innovative nanotechnology-based approaches have been developed to improve the bioactivity of PBCs and enhance their potential in preventing and/or treating age-associated disorders, primarily those caused by aging-related chronic inflammation. PBC-loaded nanoparticles designed for oral administration provide many benefits over conventional formulations, including enhanced stability and solubility, prolonged half-life, improved epithelium permeability and bioavailability, enhanced tissue targeting, and minimized side effects. The present review summarizes recent advances in this rapidly developing research area.
Collapse
Affiliation(s)
- Oleh Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str., Ivano-Frankivsk, 76018, Ukraine.
| | - Olha Strilbytska
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str., Ivano-Frankivsk, 76018, Ukraine
| | - Alexander Koliada
- Laboratory of Epigenetics, D.F. Chebotarev Institute of Gerontology, NAMS, 67 Vyshgorodska str., Kyiv, 04114, Ukraine
| | - Alina Zayachkivska
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str., Ivano-Frankivsk, 76018, Ukraine
| | - Nadia Burdyliuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str., Ivano-Frankivsk, 76018, Ukraine
| | - Ihor Yurkevych
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str., Ivano-Frankivsk, 76018, Ukraine
| | - Kenneth B Storey
- Department of Biology, Carleton University, 1125 Colonel by Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Alexander Vaiserman
- Laboratory of Epigenetics, D.F. Chebotarev Institute of Gerontology, NAMS, 67 Vyshgorodska str., Kyiv, 04114, Ukraine.
| |
Collapse
|
46
|
Mitophagy, Mitochondrial Dynamics, and Homeostasis in Cardiovascular Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9825061. [PMID: 31781358 PMCID: PMC6875274 DOI: 10.1155/2019/9825061] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022]
Abstract
Biological aging is an inevitable and independent risk factor for a wide array of chronic diseases including cardiovascular and metabolic diseases. Ample evidence has established a pivotal role for interrupted mitochondrial homeostasis in the onset and development of aging-related cardiovascular anomalies. A number of culprit factors have been suggested in aging-associated mitochondrial anomalies including oxidative stress, lipid toxicity, telomere shortening, metabolic disturbance, and DNA damage, with recent findings revealing a likely role for compromised mitochondrial dynamics and mitochondrial quality control machinery such as autophagy. Mitochondria undergo consistent fusion and fission, which are crucial for mitochondrial homeostasis and energy adaptation. Autophagy, in particular, mitochondria-selective autophagy, namely, mitophagy, refers to a highly conservative cellular process to degrade and clear long-lived or damaged cellular organelles including mitochondria, the function of which gradually deteriorates with increased age. Mitochondrial homeostasis could be achieved through a cascade of independent but closely related processes including fusion, fission, mitophagy, and mitochondrial biogenesis. With improved health care and increased human longevity, the ever-rising aging society has imposed a high cardiovascular disease prevalence. It is thus imperative to understand the role of mitochondrial homeostasis in the regulation of lifespan and healthspan. Targeting mitochondrial homeostasis should offer promising novel therapeutic strategies against aging-related complications, particularly cardiovascular diseases.
Collapse
|
47
|
Abstract
Aging of the vasculature plays a central role in morbidity and mortality of older people. To develop novel treatments for amelioration of unsuccessful vascular aging and prevention of age-related vascular pathologies, it is essential to understand the cellular and functional changes that occur in the vasculature during aging. In this review, the pathophysiological roles of fundamental cellular and molecular mechanisms of aging, including oxidative stress, mitochondrial dysfunction, impaired resistance to molecular stressors, chronic low-grade inflammation, genomic instability, cellular senescence, epigenetic alterations, loss of protein homeostasis, deregulated nutrient sensing, and stem cell dysfunction in the vascular system are considered in terms of their contribution to the pathogenesis of both microvascular and macrovascular diseases associated with old age. The importance of progeronic and antigeronic circulating factors in relation to development of vascular aging phenotypes are discussed. Finally, future directions and opportunities to develop novel interventions to prevent/delay age-related vascular pathologies by targeting fundamental cellular and molecular aging processes are presented.
Collapse
Affiliation(s)
- Zoltan Ungvari
- From the Vascular Cognitive Impairment Laboratory, Reynolds Oklahoma Center on Aging (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City
- Department of Geriatric Medicine, Translational Geroscience Laboratory (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City
- Department of Medical Physics and Informatics, University of Szeged, Hungary (Z.U., A.C.)
- Department of Pulmonology, Semmelweis University of Medicine, Budapest, Hungary (Z.U.)
| | - Stefano Tarantini
- From the Vascular Cognitive Impairment Laboratory, Reynolds Oklahoma Center on Aging (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City
- Department of Geriatric Medicine, Translational Geroscience Laboratory (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City
| | - Anthony J Donato
- Division of Geriatrics, Department of Internal Medicine, University of Utah, Salt Lake City (A.J.D.)
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, UT (A.J.D.)
| | - Veronica Galvan
- Barshop Institute for Longevity and Aging Studies (V.G.), University of Texas Health Science Center at San Antonio
- Department of Physiology (V.G.), University of Texas Health Science Center at San Antonio
| | - Anna Csiszar
- From the Vascular Cognitive Impairment Laboratory, Reynolds Oklahoma Center on Aging (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City
- Department of Geriatric Medicine, Translational Geroscience Laboratory (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City
- Department of Medical Physics and Informatics, University of Szeged, Hungary (Z.U., A.C.)
| |
Collapse
|
48
|
Kiss T, Giles CB, Tarantini S, Yabluchanskiy A, Balasubramanian P, Gautam T, Csipo T, Nyúl-Tóth Á, Lipecz A, Szabo C, Farkas E, Wren JD, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) supplementation promotes anti-aging miRNA expression profile in the aorta of aged mice, predicting epigenetic rejuvenation and anti-atherogenic effects. GeroScience 2019; 41:419-439. [PMID: 31463647 PMCID: PMC6815288 DOI: 10.1007/s11357-019-00095-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 08/21/2019] [Indexed: 12/11/2022] Open
Abstract
Understanding molecular mechanisms involved in vascular aging is essential to develop novel interventional strategies for treatment and prevention of age-related vascular pathologies. Recent studies provide critical evidence that vascular aging is characterized by NAD+ depletion. Importantly, in aged mice, restoration of cellular NAD+ levels by treatment with the NAD+ booster nicotinamide mononucleotide (NMN) exerts significant vasoprotective effects, improving endothelium-dependent vasodilation, attenuating oxidative stress, and rescuing age-related changes in gene expression. Strong experimental evidence shows that dysregulation of microRNAs (miRNAs) has a role in vascular aging. The present study was designed to test the hypothesis that age-related NAD+ depletion is causally linked to dysregulation of vascular miRNA expression. A corollary hypothesis is that functional vascular rejuvenation in NMN-treated aged mice is also associated with restoration of a youthful vascular miRNA expression profile. To test these hypotheses, aged (24-month-old) mice were treated with NMN for 2 weeks and miRNA signatures in the aortas were compared to those in aortas obtained from untreated young and aged control mice. We found that protective effects of NMN treatment on vascular function are associated with anti-aging changes in the miRNA expression profile in the aged mouse aorta. The predicted regulatory effects of NMN-induced differentially expressed miRNAs in aged vessels include anti-atherogenic effects and epigenetic rejuvenation. Future studies will uncover the mechanistic role of miRNA gene expression regulatory networks in the anti-aging effects of NAD+ booster treatments and determine the links between miRNAs regulated by NMN and sirtuin activators and miRNAs known to act in the conserved pathways of aging and major aging-related vascular diseases.
Collapse
Affiliation(s)
- Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics / Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Cory B Giles
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Oklahoma Medical Research Foundation, Genes & Human Disease Research Program, Oklahoma City, OK and Department of Biochemistry and Molecular Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Tripti Gautam
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics / Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
- Department of Public Health / Doctoral School of Basic and Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Institute of Biophysics, Biological Research Centre / Theoretical Medicine Doctoral School, Hungarian Academy of Sciences, Szeged, Hungary
| | - Agnes Lipecz
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics / Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
- Department of Public Health / Doctoral School of Basic and Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Csaba Szabo
- Chair of Pharmacology, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Eszter Farkas
- Department of Medical Physics and Informatics / Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Jonathan D Wren
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Oklahoma Medical Research Foundation, Genes & Human Disease Research Program, Oklahoma City, OK and Department of Biochemistry and Molecular Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics / Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA.
- Department of Medical Physics and Informatics / Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary.
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- Department of Public Health / Doctoral School of Basic and Translational Medicine, Semmelweis University, Budapest, Hungary.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
49
|
Laurent S, Boutouyrie P, Cunha PG, Lacolley P, Nilsson PM. Concept of Extremes in Vascular Aging. Hypertension 2019; 74:218-228. [DOI: 10.1161/hypertensionaha.119.12655] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Stephane Laurent
- From the Department of Pharmacology, INSERM U970, Assistance Publique Hôpitaux de Paris, Université Paris Descartes, France (S.L., P.B.)
| | - Pierre Boutouyrie
- From the Department of Pharmacology, INSERM U970, Assistance Publique Hôpitaux de Paris, Université Paris Descartes, France (S.L., P.B.)
| | - Pedro Guimarães Cunha
- Center for the Research and Treatment of Arterial Hypertension and Cardiovascular Risk, Serviço de Medicina Interna do Hospital da Senhora da Oliveira, Guimarães, Portugal (P.G.C.)
- Life and Health Science Research Institute, School of Medicine, University of Minho, Guimarães, Portugal (P.G.C.)
| | | | - Peter M. Nilsson
- Department of Clinical Sciences, Lund University, Skane University Hospital, Malmo, Sweden (P.M.N.)
| |
Collapse
|
50
|
Yoon SK, Okyere BA, Strasser D. Polypharmacy and Rational Prescribing: Changing the Culture of Medicine One Patient at a Time. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2019. [DOI: 10.1007/s40141-019-00220-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|