这是用户在 2025-7-13 17:48 为 https://www.ahajournals.org/doi/full/10.1161/CIRCRESAHA.125.325531?rfr_dat=cr_pub++0pubmed&url_ver=Z... 保存的双语快照页面,由 沉浸式翻译 提供双语支持。了解如何保存?
Skip to main content

Abstract

A demographic shift toward an aging population is occurring worldwide. Commensurate with this trend, age-related diseases are also rising. Although aging is an immutable part of life, biological aging is a highly heterogeneous process influenced by health-related behaviors, genetics, and the environment. The biological effects of aging are evident across all organ systems. Some individuals demonstrate an accelerated aging phenotype, which is associated with substantial morbidity and mortality. Moreover, common cardiovascular conditions, such as heart failure with preserved ejection fraction, can be conceptualized as a consequence of accelerated aging. Physical activity and exercise training modify the biological effects of accelerated aging considerably. They are associated with various salutary benefits and have emerged as potent therapeutics in disease states characterized by the accelerated aging phenotype. In this state-of-the-art review, we discuss the age-related changes observed in cardiovascular and peripheral tissues, the implications of age acceleration, and the effectiveness and mechanisms of physical activity and exercise training to curb the deleterious effects of aging and improve outcomes.
全球人口正朝着老龄化转变。与这一趋势相适应的是,与年龄相关的疾病也在增加。虽然衰老是生命中不可改变的一部分,但生物衰老是一个高度异质性的过程,受健康相关行为、遗传和环境的影响。衰老的生物学效应在所有器官系统中都有体现。一些个体表现出加速衰老的表型,这与大量的发病率和死亡率相关。 此外,常见的心血管疾病,如射血分数保留型心力衰竭,可以被认为是加速衰老的结果。体力活动和运动训练可以显著改变加速衰老的生物学效应。它们与各种有益效果相关,并且已经成为以加速衰老表型为特征的疾病状态下的有效疗法。 在这篇最新的综述中,我们讨论了在心血管和外周组织中观察到的与年龄相关的变化、年龄加速的影响,以及体育活动和运动训练在抑制衰老的不利影响和改善结局方面的有效性和机制。
The population of older adults is expanding across the world. In the United States, people over the age of 65 years is the fastest growing demographic comprising 17% of the total population and has risen from 13.0% in 2010.1 The advanced age of the population is accompanied by increasing exposure to risk factors for cardiometabolic disease resulting in higher prevalence of cardiovascular disease (CVD) in older (vs. younger) adults.2,3 CVD is more complex in older adults, as it is accompanied by multiple comorbidities and a diminished physiological reserve against stressors.4,5
全球老年人口正在不断增长。在美国,65 岁以上的人口是增长最快的群体,占总人口的 17%,高于 2010 年的 13.0%。 1 人口老龄化伴随着心血管代谢疾病风险因素暴露的增加,导致老年人(相对于年轻人)心血管疾病(CVD)的患病率更高。 2,3 在老年人中,心血管疾病更为复杂,因为它伴随着多种合并症和对抗应激源的生理储备的减少。 4,5
Chronological aging occurs in all humans, but biological aging is a highly heterogeneous process that reflects the homeostatic balance between stressors and resilience factors and is potentially modifiable.6,7 Biological aging involves the accumulation of biological stressors and dysfunction at the cellular and subcellular levels. The effects of these biological processes result in the development of clinical geriatric syndromes and cardiometabolic disease and predispose to aging-related CVD.8 Importantly, some of the most common CVDs among older adults, such as heart failure with preserved ejection fraction (HFpEF), mimic an accelerated aging phenotype.9 While these cellular processes develop over time, the rate of progression is partly dependent on health-related behaviors of the individual, such as physical inactivity, nutrition, alcohol, and tobacco use.
所有人类都会经历时间年龄的增长,但生物学年龄是一个高度异质性的过程,它反映了应激因素和恢复力因素之间的稳态平衡,并且具有潜在的可调节性。 6,7 生物学年龄的增长涉及细胞和亚细胞水平上生物应激源和功能障碍的积累。这些生物学过程的影响导致临床老年综合征和心脏代谢疾病的发生,并易于发生与衰老相关的 CVD。 8 重要的是,老年人中最常见的一些心血管疾病,如射血分数保留型心力衰竭 (HFpEF),模拟了一种加速衰老的表型。 9 虽然这些细胞过程随着时间的推移而发展,但进展速度部分取决于个体的健康相关行为,如缺乏运动、营养、酒精和烟草使用。
Physical activity and exercise are important moderators of biological aging and age-related CVD. Higher levels of cardiorespiratory fitness and physical activity have been consistently linked to a lower risk of cardiometabolic disease, including obesity, type 2 diabetes, and coronary heart disease.10–13 Moreover, improving cardiorespiratory fitness with exercise training improves multiple cardiometabolic risk factors and may alter the trajectory of biological aging. In this article, we review the process of cardiovascular aging and the consequences of accelerated aging in the context of CVD development. We also discuss the biological and cellular mechanisms linking biological aging with CVD, the effects of aging on cardiovascular structure and function, and compare normal aging versus accelerated or pathological aging phenotypes. Finally, we explore the role of physical activity, cardiorespiratory fitness, and exercise training on aging and clinical outcomes and review the evidence for and mechanisms through which exercise training may improve cardiorespiratory fitness and curb the effects of cardiovascular aging.
体力活动和锻炼是生物衰老和年龄相关 CVD 的重要调节因素。较高的心肺适能和体力活动水平与较低的心脏代谢疾病风险持续相关,包括肥胖、2 型糖尿病和冠心病。 10–13 此外,通过运动训练提高心肺适能可以改善多种心脏代谢风险因素,并可能改变生物衰老的轨迹。 在本文中,我们将回顾心血管衰老的过程以及在心血管疾病(CVD)发展过程中加速衰老的后果。我们还将讨论生物衰老与心血管疾病相关的生物学和细胞机制,衰老对心血管结构和功能的影响,并比较正常衰老与加速或病理性衰老表型。 最后,我们将探讨体力活动、心肺适能和运动训练在衰老和临床结局中的作用,并回顾运动训练可能改善心肺适能并抑制心血管衰老影响的证据和机制。

Accelerated Aging and the Geroscience Framework for CVD
加速衰老与 CVD 的老年科学框架

While age-related changes to the cardiovascular system are inevitable and can occur without overt CVD, the progression of these changes is influenced by the interplay of health-related behaviors and genetic, molecular, environmental, and social factors.14 The geroscience framework postulates that the development of chronic disease can be viewed as a consequence of biological aging.15 Accelerated aging occurs when biological age outpaces chronological aging, resulting in the premature development of age-related changes at the cellular, tissue, and organ system levels. This culminates in distinct clinical phenotypes characterized by a high burden of frailty, multimorbidity, diminished physical function, low cardiorespiratory fitness, and high risk of adverse events. In contrast to healthy aging, accelerated aging is pathological and results in increased risk of morbidity and mortality.7
虽然与年龄相关的心血管系统变化不可避免,并且可能在没有明显心血管疾病的情况下发生,但这些变化的进展受到健康相关行为以及遗传、分子、环境和社会因素相互作用的影响。 14 衰老科学框架假定,慢性疾病的发生可以被视为生物衰老的结果。 15 当生物学年龄超过实际年龄时,就会发生加速衰老,导致细胞、组织和器官系统层面过早出现与年龄相关的改变。这最终形成独特的临床表型,其特征是虚弱、多病、身体功能下降、心肺适能低以及不良事件风险高。 与健康衰老相反,加速衰老是病理性的,会导致发病率和死亡率风险增加。 7
Rare genetic conditions of premature aging, such as the Hutchinson-Gilford progeria syndrome, have provided key insights into the process of accelerated aging and associated predisposition to CVD.16 The Hutchinson-Gilford progeria syndrome is associated with mutations of the LMNA gene, which results in premature phenotypic manifestations of aging features, including alopecia, skin wrinkling, and osteoporosis. Patients with Hutchinson-Gilford progeria syndrome are also affected by premature CVD, with cardiovascular events such as myocardial infarction and stroke at an early age.16 Similarly, Werner syndrome is caused by mutations in a DNA exonuclease involved in the repair of double-stranded DNA breaks that manifest clinically with premature graying, loss of hair, metabolic dysregulation, subcutaneous fat atrophy, osteoporosis, and premature CVD.17,18 The cardiovascular manifestations of Werner syndrome include atherosclerosis, valvular diseases, and a hypercoagulable state. These syndromes demonstrate that genetic syndromes of accelerated aging can predispose to premature CVD in the absence of traditional cardiovascular risk factors.
罕见的早衰遗传疾病,如早老症,为加速衰老过程以及相关的 CVD 易感性提供了重要的见解。 16 早老症与 LMNA 基因的突变有关,导致过早出现衰老表型特征,包括脱发、皮肤皱纹和骨质疏松症。 患有早衰症的患者也会受到过早发生的心血管疾病 (CVD) 的影响,并在早期发生心肌梗死和卒中等心血管事件。 16 同样,沃纳综合征是由参与双链 DNA 断裂修复的 DNA 外切酶突变引起的,临床表现为过早灰发、脱发、代谢失调、皮下脂肪萎缩、骨质疏松症和过早发生的心血管疾病。 17,18 Werner 综合征的心血管表现包括动脉粥样硬化、瓣膜疾病和高凝状态。这些综合征表明,即使不存在传统的心血管危险因素,加速衰老的遗传综合征也可能导致过早发生 CVD。
While the syndromes of premature aging have distinct underlying genetic causes, they share fundamental cellular mechanisms linking genetic mutation to accelerated biological aging and premature CVD. These include closely inter-linked cellular and molecular processes, commonly referred to as the hallmarks of aging, which are evolutionally conserved across species and provide a framework to understand the role of biological aging in health and disease, including CVD.19 Specifically, 8 hallmarks of aging have been identified as common denominators of aging in the cardiovascular system including genomic instability, epigenetic alterations, loss of proteostasis, disabled macroautophagy, dysregulated nutrition sensing, mitochondrial dysfunction, inflammation, and cellular senescence.19,20 Augmentation of the activity of these hallmarks of aging is implicated in accelerated aging phenotypes and increased risk of CVD.15 Furthermore, amelioration of the activity of specific hallmarks of aging can delay biological aging, highlighting their potential as therapeutic targets to prevent aging-related chronic diseases, including CVD. Table 1 provides a concise overview of the different hallmarks of aging, associated CVD, the key phenotypic manifestations, and potential treatment strategies that may target specific pathways to attenuate accelerated aging and associated CVD.
虽然早衰综合征具有不同的潜在遗传原因,但它们具有将基因突变与加速生物衰老和早发性心血管疾病联系起来的基本细胞机制。这些机制包括密切相关的细胞和分子过程,通常被称为衰老的标志,这些标志在物种进化过程中是保守的,并提供了一个框架来理解生物衰老在健康和疾病(包括心血管疾病)中的作用。 19 具体而言,已确定 8 个衰老标志是心血管系统中衰老的共同因素,包括基因组不稳定性、表观遗传改变、蛋白稳态丧失、巨自噬功能障碍、营养感知失调、线粒体功能障碍、炎症和细胞衰老。 19,20 这些衰老标志活性的增强与加速衰老表型和 CVD 风险增加有关。 15 此外,改善特定衰老标志的活性可以延缓生物衰老,突显了它们作为治疗靶点以预防包括 CVD 在内的与衰老相关的慢性疾病的潜力。表 1 简要概述了不同的衰老标志、相关的 CVD、关键的表型表现以及可能靶向特定通路以减轻加速衰老和相关 CVD 的潜在治疗策略。
Table 1. Hallmarks of Aging, Associated Phenotypic Manifestations and Cardiovascular Diseases, and Potential Treatment Strategies Targeting the Specific Mechanisms
表 1. 衰老的标志、相关的表型表现和心血管疾病,以及针对特定机制的潜在治疗策略
Afib indicates atrial fibrillation; ASCVD, atherosclerotic cardiovascular disease; ATTR, amyloid transthyretin; CAD, coronary artery disease; CV, cardiovascular; CVD, cardiovascular disease; HDAC, histone deacetylase; HF, heart failure; HFpEF, heart failure with preserved ejection fraction; and SGLT2i, sodium-glucose co-transporter 2 inhibitor.
Afib 表示心房颤动;ASCVD,动脉粥样硬化性心血管疾病;ATTR,淀粉样变性转甲状腺素蛋白;CAD,冠状动脉疾病;CV,心血管;CVD,心血管疾病;HDAC,组蛋白去乙酰化酶;HF,心力衰竭;HFpEF,射血分数保留型心力衰竭;SGLT2i,钠-葡萄糖协同转运蛋白 2 抑制剂。

Estimating Biological Age to Identify Individuals at Risk of Accelerated Aging-Related CVD
评估生理年龄以识别具有加速衰老相关心血管疾病风险的个体

Measuring biological age is critical to studying the link between accelerated aging and CVD at a population level. Numerous biological age estimation models incorporating biomarkers, epigenetics, anthropometric, and physiological inputs to measure biological age have been developed and validated. The Phenotypic Age Acceleration model is one such model that utilizes chronologic age and a combination of blood-based chemistry and hematology parameters to estimate biological age.21 A similar score can also be created using the Klemera-Doubal method, which uses all available biomarkers to estimate biological age using mathematical optimization.22 A higher phenotypic age acceleration is associated with multimorbidity and increased mortality risk, and partially mediates the association between unhealthy lifestyles and incident CVD and mortality.21,23 Multiple studies have also demonstrated an association between biological age and abnormal cardiovascular structure and function, including vascular stiffness, adverse cardiac remodeling (lower left ventricular [LV] mass and stroke volume), and increased risk of heart failure (HF).24–26
测量生物学年龄对于在人群水平上研究加速衰老与心血管疾病之间的联系至关重要。已经开发并验证了许多生物学年龄评估模型,这些模型结合了生物标志物、表观遗传学、人体测量学和生理学输入来测量生物学年龄。“表型年龄加速”模型就是其中一种,它利用实际年龄以及基于血液的化学和血液学参数的组合来估计生物学年龄。 21 也可以使用 Klemera-Doubal 方法创建类似的分数,该方法使用所有可用的生物标志物,通过数学优化来估计生物学年龄。 22 表型年龄加速升高与多病共存和死亡风险增加相关,并部分介导不健康的生活方式与新发 CVD 和死亡率之间的关联。 多项研究还表明,生物学年龄与异常心血管结构和功能之间存在关联,包括血管僵硬、不良心脏重塑(左心室[LV]质量和卒中容量降低)以及心力衰竭(HF)风险增加。
Biological aging scores have also been developed to assess epigenetic age using specific DNA methylation patterns. For instance, the GrimAge acceleration model estimates epigenetic age by using 1030 CpG islands, blood-based biomarkers, and clinical risk factors. In the CARDIA (Coronary Artery Disease in Young Adults) cohort, an increase in GrimAge acceleration was associated with an increased risk for incident diabetes27 and risk of CVD and mortality.28 Similarly, in the Atherosclerosis Risk in Communities study, the Horvath and Hannum models (epigenetic scores using DNA methylation patterns) were associated with a higher risk of subclinical atherosclerosis and incident CVD independent of traditional cardiovascular risk factors.29 Similar findings have been noted using other biological age estimation models across different community-dwelling cohorts.28,30,31 Together, these observations support the notion of accelerated aging being a key driver of abnormalities in cardiovascular structure and function, and increased risk of CVD and may be a target for therapeutic interventions.
人们还开发了生物衰老评分,旨在使用特定的 DNA 甲基化模式来评估表观遗传年龄。例如,GrimAge 加速模型通过使用 1030 个 CpG 岛、基于血液的生物标志物和临床危险因素来估计表观遗传年龄。在 CARDIA(年轻人冠状动脉疾病)队列中,GrimAge 加速的增加与新发糖尿病 27 以及 CVD 和死亡风险的增加相关。 28 同样,在社区动脉粥样硬化风险研究中,Horvath 和 Hannum 模型(使用 DNA 甲基化模式的表观遗传评分)与亚临床动脉粥样硬化和独立于传统心血管危险因素的心血管疾病事件的较高风险相关。 29 在不同的社区居住人群队列中使用其他生物年龄估计模型也发现了类似的结果。 28,30,31 总之,这些观察结果支持了加速衰老是心血管结构和功能异常以及心血管疾病风险增加的关键驱动因素的观点,并可能成为治疗干预的靶点。

Frailty: An Accelerated Biological Aging Phenotype Associated With CVD
衰弱:一种与心血管疾病相关的加速生物衰老表型

Accelerated biological aging translates to the accumulation of subclinical disease and deficits across nearly all organ systems. This results in a diminished reserve capacity and increased disease severity that manifests clinically as the frailty syndrome.32 The frailty syndrome is characterized by the loss of reserve across multiple physiological systems, impairing the homeostatic capacity to tolerate external stressors.32 Frailty develops through dysregulation of immune, hormonal, and endocrine systems with upregulation of inflammatory cytokines, which contribute to inflammaging33 and predispose to a catabolic state resulting in sarcopenia. Clinically, these changes appear as impaired functional status and loss of physiological reserve.14,34,35 Genome-wide association studies have implicated multiple genes that are associated with both accelerated aging and CVD in frailty.36 The prevalence of frailty among community-dwelling adults without CVD is ≈15%.37 However, the prevalence of frailty is much higher in older adults with prevalent CVD (reported up to 75%) and differs substantially across CVD subtypes.38
加速的生物衰老转化为亚临床疾病的积累,以及几乎所有器官系统的功能缺陷。这导致储备能力下降和疾病严重程度增加,并在临床上表现为虚弱综合征。 32 虚弱综合征的特征是多个生理系统的储备丧失,从而损害了耐受外部压力的稳态能力。 32 虚弱的发生是由于免疫、激素和内分泌系统失调,以及炎症细胞因子上调,这会导致炎症衰老 33 并易于发生分解代谢状态,从而导致肌少症。临床上,这些变化表现为功能状态受损和生理储备丧失。 14,34,35 全基因组关联研究表明,有多个基因与虚弱相关的加速衰老和心血管疾病有关。 36 在没有 CVD 的社区居住成年人中,虚弱的患病率约为 15%。 37 然而,在患有 CVD 的老年人中,虚弱的患病率要高得多(据报道高达 75%),并且在不同的 CVD 亚型中差异很大。 38
Among age-related CVD, a higher frailty burden has been associated with a greater risk of valvular heart disease, such as aortic stenosis, atrial fibrillation, and HF, particularly HFpEF.39–43 Higher frailty burden is also associated with greater abnormalities in cardiac structure and function among older adults without CVD.43–46 HFpEF is particularly intertwined with frailty and represents a downstream clinical manifestation of accelerated biological aging and the associated decline in physiological reserve.4 HFpEF occurs almost exclusively in older adults, is preceded by an accelerated decline in cardiorespiratory fitness and functional status, and is characterized by severe exercise intolerance.47 Patients with HFpEF also have multiple comorbidities, which contribute to the development of frailty and sarcopenia that potentiates disease severity.4 Indeed, HFpEF is increasingly recognized as a true geriatric syndrome, with accelerated aging, sarcopenia, and frailty as key pathophysiologic determinants (Figure 1).9
在与年龄相关的 CVD 中,较高的衰弱负担与更高的瓣膜性心脏病风险相关,例如主动脉瓣狭窄、房颤和 HF,尤其是 HFpEF。 39–43 较高的衰弱负担也与无 CVD 的老年人的心脏结构和功能异常程度增加有关。 43–46 射血分数保留型心力衰竭(HFpEF)与体弱尤其密切相关,代表着加速生物衰老以及相关的生理储备下降的下游临床表现。 4 射血分数保留型心力衰竭几乎只发生在老年人身上,其发生之前存在心肺适能和功能状态的加速下降,并以严重的运动不耐受为特征。 47 射血分数保留型心力衰竭(HFpEF)患者也常伴有多种合并症,这些合并症会导致虚弱和肌少症,从而加剧疾病的严重程度。 4 事实上,HFpEF 越来越被认为是一种真正的老年综合征,其关键病理生理决定因素是加速衰老、肌少症和虚弱(图 1)。 9
Figure 1. Accelerated aging phenotype and the pathophysiology of heart failure with preserved ejection fraction (HFpEF). The cumulative effects of multimorbidity, obesity, aging, physical inactivity, and systemic inflammation result in an accelerated aging phenotype characterized by sarcopenia, mitochondrial and endothelial dysfunction, and capillary loss, which contribute to diminished physiological reserve (frailty) that leads to multiorgan dysfunction and myopathy of the cardiac and skeletal muscles. These changes have been implicated in the pathophysiology of HFpEF, which is now considered a geriatric syndrome that is characterized by severe impairment in quality of life (QOL), physical function, and exercise capacity and is associated with disability and mortality. Adapted from Pandey et al9 with permission. Illustration credit: Sceyence Studios.
图 1. 加速衰老表型与射血分数保留型心力衰竭(HFpEF)的病理生理学。 多重疾病、肥胖、衰老、缺乏运动和全身性炎症的累积效应会导致加速衰老表型,其特征是肌肉减少症、线粒体和内皮功能障碍以及毛细血管丢失,这些因素会导致生理储备(虚弱)减少,进而导致多器官功能障碍以及心肌和骨骼肌的肌病。 这些变化与射血分数保留型心力衰竭(HFpEF)的病理生理学有关,HFpEF 现在被认为是一种老年综合征,其特征是生活质量(QOL)、身体功能和运动能力的严重受损,并与残疾和死亡率相关。经许可,改自 Pandey 等人 9 。插图来源:Sceyence Studios。

Molecular and Cellular Mechanisms of Cardiovascular Aging
心血管老化的分子和细胞机制

Aging distinctly affects cardiovascular structure and function through cellular and subcellular changes that occur in response to the accumulation of biological, metabolic, and hemodynamic stress over time (Figure 2).48 Aging is associated with an impaired reparative response to cardiac injury and defective activation of reparative fibroblasts in response to growth factors. Cellular renewal and regeneration of cardiomyocytes slow considerably with age and are replaced by cellular senescence and apoptosis.15,49–51 Cardiomyocyte dropout is particularly notable in the cardiac conduction system, with loss of pacemaker cells in the sinoatrial node and the atrioventricular node.52,53 This process initially induces a compensatory remodeling characterized by the alterations of extracellular matrix composition involving the synthesis of myofibroblasts, increased matrix accumulation, and collagen deposition, leading to cardiac fibrosis.54 Cardiac fibrosis process is also potentiated by increased mechanical load and direct senescence–associated fibrogenic actions.55,56 Several molecular pathways are implicated in age-related myocardial fibrosis, including overexpression of chemokines that recruit mononuclear cells and fibroblast progenitors, transforming growth factor-β activation, ET-1 (endothelin-1), and angiotensin-II signaling.57,58 In addition, accumulation of oxidative stress with age results in the deposition of lipofuscin, an intracellular aggregate of oxidized proteins within cells, further contributing to fibrosis.59 Moreover, age-related changes are noted in the cellular function of cardiomyocytes, which affect excitation-contraction coupling, calcium cycling, and cellular metabolism and signaling.60,61 The net effect of progressive fibrosis of the aging heart is an increase in ventricular stiffness and diastolic dysfunction that often progresses to CVD, particularly HFpEF.62–64
衰老通过细胞和亚细胞的变化明显影响心血管结构和功能,这些变化是对生物、代谢和血流动力学应激随时间推移而积累的反应(图 2)。 48 衰老与对心脏损伤的修复反应受损以及修复性成纤维细胞对生长因子激活缺陷有关。 随着年龄的增长,心肌细胞的细胞更新和再生明显减缓,并被细胞衰老和凋亡所取代。 15,49–51 心肌细胞脱落 Cardiac conduction system 中尤为明显,表现为窦房结和房室结起搏细胞的丢失。 52,53 这一过程最初会诱导一种代偿性重塑,其特征是细胞外基质成分的改变,包括肌成纤维细胞的合成、基质积累的增加和胶原蛋白的沉积,从而导致心脏纤维化。 54 心脏纤维化过程也因机械负荷增加和直接的衰老相关纤维化作用而增强。 55,56 多种分子通路与年龄相关的心肌纤维化有关,包括趋化因子过度表达(可募集单核细胞和成纤维细胞祖细胞)、转化生长因子-β激活、ET-1(内皮素-1)和血管紧张素-II 信号传导。 57,58 此外,随着年龄增长,氧化应激的积累导致脂褐素的沉积,脂褐素是细胞内氧化的蛋白质聚集体,进一步促进纤维化。 59 此外,与年龄相关的变化也见于心肌细胞的细胞功能,这些变化会影响兴奋-收缩耦联、钙循环以及细胞代谢和信号传导。 60,61 衰老心脏进行性纤维化的最终结果是心室僵硬度增加和舒张功能障碍,这通常会进展为 CVD,尤其是 HFpEF。 62–64
Figure 2. Multisystem changes related to the aging process. Age-related changes occur in nearly every organ system, and the structural and functional changes of cellular, cardiovascular, skeletomuscular, and adipose tissue result in diminished physiological capacity. LA indicates left atrium; and LV, left ventricle. Illustration credit: Sceyence Studios.
图 2. 与衰老过程相关的多系统变化。与年龄相关的变化几乎发生在每个器官系统中,细胞、心血管、骨骼肌和脂肪组织的结构和功能变化导致生理能力下降。LA 表示左心房;LV 表示左心室。插图来源:Sceyence Studios。

Aging and Cardiovascular Structure and Function
衰老与心血管结构和功能

The cardiac morphology and function evolve considerably with aging, with distinct changes noted among those with normal aging versus accelerated biological aging as highlighted in Table 2 and discussed in more detail in this section. Healthy aging-related changes in cardiac structure and function are often driven by the effects of lifelong exercise activities on the heart and represent a favorable adaptation of the myocardium to the increasing physiological demands.65 In contrast, accelerated age-related changes in cardiac structure and function occur in response to long-standing exposure to pathological stressors (volume or pressure overload) related to cardiovascular risk factors and CVD. Studying effects of healthy aging on the cardiovascular system is challenging because most older adults have cardiovascular risk factors or subclinical CVD (≈95%).66 Excluding those with risk factors and subclinical CVD requires exhaustive workup and is often done variably across studies. The inclusion of participants who may have subclinical CVD may confound the findings of aging-related changes in cardiac structure and function. The participant selection underlies much of the heterogeneity in study findings of cardiac manifestations associated with aging discussed here.
随着年龄的增长,心脏的形态和功能会发生显著变化,正常衰老与加速生物衰老的人群之间存在明显的差异,如表 2 所示,并在本节中进行了更详细的讨论。健康衰老相关的心脏结构和功能变化通常是由终身运动活动对心脏的影响驱动的,代表了心肌对日益增长的生理需求的有利适应。 65 相比之下,长期暴露于与心血管风险因素和 CVD 相关的病理应激(容量或压力超负荷)会导致心脏结构和功能发生加速的年龄相关改变。研究健康衰老对心血管系统的影响具有挑战性,因为大多数老年人都有心血管风险因素或亚临床 CVD(≈95%)。 66 排除那些具有风险因素和亚临床 CVD 的个体需要详尽的检查,并且在不同研究中通常做法不一。纳入可能患有亚临床 CVD 的参与者可能会混淆关于心脏结构和功能中与衰老相关的变化的研究结果。参与者的选择是导致此处讨论的与衰老相关的心脏表现研究结果存在很大异质性的根本原因。
Table 2. Cardiovascular Phenotypes Associated With Normal and Accelerated Biological Aging
表 2. 与正常和加速生物衰老相关的心血管表型

Age-Related Changes to Left Atrial Structure and Function
与年龄相关的左心房结构和功能改变

Age-related increase in left atrial (LA) size has been shown in multiple studies.67–70 However, the dilation of the LA in the absence of a pathological process occurs only after the seventh decade of life.71 The mechanisms of age-related dilation of the LA are not entirely understood but may be in response to chronically elevated LV end-diastolic pressures.72 Aging results in electromechanical changes in the LA, with a decline in LA compliance and function.73–77 LA size is also a significant risk factor for multiple age-related CVDs, such as atrial fibrillation and stroke.78,79 LA strain imaging by echocardiography offers a sensitive assessment of LA function. The reservoir and contraction strain remain unchanged with age, but the conduit function deteriorates, suggesting reduced compliance.76
多项研究表明,左心房 (LA) 大小随年龄增长而增加。 67–70 然而,在没有病理过程的情况下,LA 的扩张仅在七十岁之后发生。 71 与年龄相关的 LA 扩张的机制尚未完全清楚,但可能是对慢性升高的左心室舒张末期压力的反应。 72 衰老会导致 LA 发生机电变化,LA 顺应性和功能下降。 73–77 左心房大小也是多种年龄相关性心血管疾病(如心房颤动和卒中)的重要危险因素。 78,79 通过超声心动图进行左心房应变成像可以灵敏地评估左心房功能。储器和收缩应变随年龄增长保持不变,但导管功能会恶化,表明顺应性降低。 76

Aging and Changes in Left Ventricular Mass and Size
左心室质量和大小的老化和变化

In the absence of coronary artery disease and hypertension, normal aging does not increase LV mass. Studies of carefully selected normotensive healthy adults using echocardiography and magnetic resonance imaging–based assessment of LV mass have consistently found no increase in LV mass with aging.80–82 In a cross-sectional study of 336 healthy, normotensive adults across a wide age range, Hees et al80 demonstrated distinct sex associations between age and LV morphology. Specifically, among women, they observed an increase in LV wall thickness and a concurrent decrease in LV length with aging such that the mean LV mass remained constant. In contrast, men had a modest decline in LV mass related to decreased LV length without a change in LV wall thickness. In contrast, in an autopsy study of 765 normal hearts from people aged 20 to 99 years, Kitzman et al83 observed an increase in heart weight indexed to body size with aging in women but not men. Specifically, myocardial thickness increases predominantly in the interventricular septum with relatively little LV and right ventricular free wall thickness change. The differences in the methodology of cardiac mass measurements (assessment of LV mass by cardiac magnetic resonance imaging versus crude heart weight using autopsy hearts) may account for the differences noted between these studies. Regarding LV cavity size, evidence from cross-sectional and longitudinal epidemiological cohorts has shown that the LV internal diameter at systole and diastole reduces with normal age, resulting in a smaller LV cavity size.84,85 In addition, there is a decline in LV length, which results in increased sphericity of the heart.71,80 Age-related reduction in LV cavity size and increased wall thickness is reflected in a higher mass/volume ratio (relative wall thickness), corresponding to concentric LV remodeling with normal aging noted in multiple studies.86–89 Accelerated biological aging results in pathological LV hypertrophy characterized by concentric hypertrophy with cardiomyocyte thickening and fibrosis, which results in a marked reduction in the LV cavity size disproportionate to age.90 This is often related to long-term exposure to cardiovascular risk factors such as hypertension and obesity, which accelerates biological aging. In contrast, physical activity and endurance training-related activities, which are lifestyle traits associated with healthy aging, lead to physiological LV hypertrophy with increased LV size and LV mass but not relative wall thickness.91
在没有冠状动脉疾病和高血压的情况下,正常衰老不会增加左心室质量。对精心挑选的血压正常健康成人进行的研究,使用基于超声心动图和磁共振成像的左心室质量评估,一直发现左心室质量不随年龄增长而增加。 80–82 在一项针对 336 名健康、血压正常成人的横断面研究中,Hees 等人 80 证明了年龄与左心室形态之间存在明显的性别关联。 具体而言,在女性中,他们观察到随着年龄的增长,LV 壁厚增加,同时 LV 长度减少,因此平均 LV 质量保持不变。相比之下,男性 LV 质量略有下降,这与 LV 长度的减少有关,而 LV 壁厚度没有变化。相比之下,在 Kitzman 等人 83 对 765 颗年龄在 20 至 99 岁之间的正常心脏进行的尸检研究中,观察到女性的心脏重量指数随年龄增长而增加,而男性则没有。 具体而言,心肌厚度增加主要发生在室间隔,而左心室和右心室游离壁厚度的变化相对较小。心脏质量测量方法(通过心脏磁共振成像评估左心室质量与使用尸检心脏的粗略心脏重量)的差异可能是导致这些研究之间差异的原因。 关于左心室腔大小,横断面和纵向流行病学队列研究的证据表明,随着正常衰老,左心室收缩末期和舒张末期内径减小,导致左心室腔变小。 84,85 此外,左心室长度也会下降,从而导致心脏球形度增加。 71,80 与年龄相关的左心室腔大小缩小和壁厚增加反映在较高的质量/体积比(相对壁厚)中,这与多项研究中发现的正常衰老时发生的心室向心性重塑相对应。 86–89 加速的生物衰老会导致病理性左心室肥厚,其特征为向心性肥厚伴心肌细胞增厚和纤维化,导致左心室腔大小显著缩小,与年龄不成比例。 90 这通常与长期暴露于高血压和肥胖等心血管风险因素有关,这些因素会加速生物衰老。相反,体力活动和耐力训练相关的活动是与健康衰老相关的生活方式特征,可导致生理性左心室肥厚,左心室大小和左心室质量增加,但相对壁厚度没有增加。 91

Aging and Aortic and Mitral Valves
衰老与主动脉瓣和二尖瓣

Age-related changes are also notable in the cardiac valves. Particularly, the aortic and mitral leaflet thickness is noted to increase along the closure margins with aging.92 In the cardiovascular health study, aortic valve sclerosis was present in 26% of participants. Valvular calcification is often regarded as a finding associated with accelerated aging and predisposes to an increased risk of valvular stenosis. In the cardiovascular health study, 2% of the cohort had hemodynamically significant aortic stenosis.93 Mitral annular calcification is also common among older adults and was noted in 10.3% of men and 15.8% of women participants of the Framingham Heart Study.94 While the prevalence of valvular calcification increases with age, the rate of calcification is influenced by genetic, environmental, and immune factors.95 Calcification of the aortic and mitral valves is initially triggered by inflammation, which results in maladaptive ultrastructural changes that result in valvular thickening and apoptosis of valvular interstitial cells and upregulation of disease-related markers such as alkaline phosphatase and osteocalcin.96,97 As such, valvular calcification can occur at an accelerated pace and result in calcific aortic valve disease the most common valvular disease in older adults.98 Furthermore, the presence of mitral annular calcification is associated with a significantly higher risk for adverse cardiovascular outcomes.94
与年龄相关的心脏瓣膜变化也值得关注。特别是,主动脉和二尖瓣叶的厚度随着年龄的增长,在闭合边缘处会明显增加。 92 在一项心血管健康研究中,26%的参与者存在主动脉瓣硬化。瓣膜钙化通常被认为是与加速衰老相关的发现,并增加了瓣膜狭窄的风险。 在心血管健康研究中,2%的队列有血流动力学意义上的主动脉瓣狭窄。 93 二尖瓣环钙化在老年人中也很常见,在 Framingham 心脏研究的参与者中,男性占 10.3%,女性占 15.8%。 94 虽然瓣膜钙化的患病率随着年龄的增长而增加,但钙化的速度受遗传、环境和免疫因素的影响。 95 主动脉和二尖瓣的钙化最初由炎症引发,导致适应不良的超微结构变化,从而导致瓣膜增厚以及瓣膜间质细胞凋亡,并上调疾病相关标志物,如碱性磷酸酶和骨钙素。 96,97 因此,瓣膜钙化可能以更快的速度发生,并导致钙化性主动脉瓣疾病,这是老年人最常见的瓣膜疾病。 98 此外,二尖瓣环钙化的存在与不良心血管事件风险显著升高相关。 94

Age-Related Changes in Cardiac Function
心脏功能的年龄相关变化

Functional changes accompany changes in cardiac structure with aging. A reduction in LV stroke volume with age is noted as the LV cavity size reduces, and the wall thickness increases. In a study of carefully selected volunteers free of CVD, the LV stroke volume index was reduced significantly with normal aging.99 Similarly, the Multiethnic Study of Atherosclerosis and the Framingham Heart Study have demonstrated a cross-sectional association between older age and lower LV stroke volume.84,100 Similarly, LV contractility may also decline with aging. In a study of volunteers free of CVD, LV end-systolic volume, a measure of LV contractility, declined with normal aging.99 Global longitudinal strain is a sensitive marker of myocardial contractility and is also noted to be lower, reflecting diminished contractility of the LV, with age, which is more evident in men than in women.101–105 The extent to which decline in LV contractility is related to healthy aging versus a consequence of underlying subclinical disease has been a matter of debate. In one study of carefully selected volunteers, LV strain was not associated with age alone but also with levels of NT-proBNP (N-terminal pro-B-type natriuretic peptide), implicating neurohormonal stress and subclinical CVD.105 However, increases in NT-ProBNP levels may also be related to the aging process with worsening diastolic function as discussed below. Together, aging results in a decline in stroke volume and reduced contractility but results in normal resting cardiac function, and these changes in LV systolic function are more pronounced and occur earlier with accelerated aging and the presence of underlying subclinical CVD. It is noteworthy that in contrast with SV and contractility measures, LV ejection fraction increases slightly with age and is higher among women than men.101–104 This may be related more to decreases in LV end-diastolic volume than increases in LV stroke volume.
随着年龄增长,心脏结构的变化伴随着功能上的改变。随着左心室腔变小,壁厚增加,左心室射血量随年龄增长而降低。在一项针对精心挑选的、无心血管疾病志愿者的研究中,左心室射血量指数随着正常衰老而显著降低。 99 同样,多种族动脉粥样硬化研究和弗雷明汉心脏研究也证实了老年与较低的左心室射血量之间的横断面关联。 84,100 同样,左心室收缩力也可能随着年龄的增长而下降。在一项针对无心血管疾病 (CVD) 志愿者的研究中,左心室收缩末期容积(一种衡量左心室收缩力的指标)随着正常衰老而下降。 99 全局纵向应变是心肌收缩力的一个敏感标志物,并且也观察到其随着年龄的增长而降低,反映出左心室收缩力减弱,这在男性中比在女性中更为明显。 101–105 左室收缩功能的下降在多大程度上与健康衰老有关,以及在多大程度上是潜在亚临床疾病的后果,一直是争论的问题。在一项精心挑选的志愿者研究中,左室应变与年龄本身无关,而且还与 NT-proBNP(N-末端 B 型脑钠肽前体)水平有关,表明存在神经激素应激和亚临床 CVD。 105 然而,如下文所述,NT-ProBNP 水平的升高也可能与伴随舒张功能恶化的衰老过程有关。总而言之,衰老会导致每搏输出量下降和收缩力降低,但会导致正常的静息心脏功能,并且这些左室收缩功能的变化在加速衰老和存在潜在的亚临床心血管疾病的情况下更为明显且发生更早。 值得注意的是,与 SV 和收缩力指标相比,LV 射血分数随年龄略有增加,并且女性高于男性。 101–104 这可能更多与 LV 舒张末期容积的减少有关,而不是 LV 搏出量的增加。
Alteration in diastolic function with age has been consistently reported across various studies.106–109 Impairment in diastolic filling with a decline in early LA emptying, prolonged late diastolic filling, and longer isovolumic relaxation time are characteristic findings in older adults without overt cardiovascular pathology.107,109 Concurrently, the peak A-wave velocity, representing atrial contraction and late LV filling, increases. The early diastolic peak-filling rate declines by 30% to 50% between the second and eighth decades of life.110 Tissue Doppler imaging–based relaxation velocities measured at the mitral annulus are frequently used to characterize diastolic function and cardiac filling pressures in contemporary clinical practice.111 In the Atherosclerosis Risk in Communities study, older adults without evidence of CVD had lower values for Doppler relaxation velocities without increased risk of adverse cardiovascular events, suggesting that the impairment in relaxation is part of the normal aging process.107 This finding has been recapitulated in multiple cohorts of individuals free of CVD.110,112,113 An analysis from the Baltimore Longitudinal Study of Aging that included participants without CVD who underwent cardiac magnetic resonance imaging demonstrated that the age-related impairment in LV diastolic filling may be driven by changes in left atrial characteristics. Specifically, the investigators found that the association between age and relaxation velocities was entirely attenuated after adjustment for LA pressure.110 This observation suggests that changes in diastolic function with age may be related to changes in LA compliance and contractile properties. Moreover, in a study of volunteers without CVD who underwent hemodynamic assessment, age was associated with slightly higher resting pulmonary capillary wedge pressure (r=0.17; P<0.05).99 In patients with accelerated biological aging, diastolic dysfunction develops at an earlier age and is associated with reduced exercise capacity and future risk of HF.114
在多项研究中,年龄相关的舒张功能改变一直有报道。 106–109 舒张充盈受损,伴随早期左心房排空减少、晚期舒张充盈延长和等容舒张时间延长,是无明显心血管病理的老年人的特征性表现。 107,109 同时,代表心房收缩和晚期左心室充盈的 A 波峰值速度增加。 在 20 岁到 80 岁之间,早期舒张期峰值充盈率下降 30% 到 50%。 110 基于组织多普勒成像的、在二尖瓣环处测量的舒张期松弛速度,常用于描述当代临床实践中的舒张功能和心脏充盈压。 111 在动脉粥样硬化社区风险研究中,无心血管疾病证据的老年人多普勒舒张速度值较低,但不良心血管事件风险并未增加,提示舒张功能受损是正常衰老过程的一部分。 107 这一发现在多个无心血管疾病的个体队列中得到了重复证实。 110,112,113 来自巴尔的摩衰老纵向研究的一项分析,纳入了未患有 CVD 且接受过心脏磁共振成像的参与者,结果表明,与年龄相关的 LV 舒张充盈受损可能由左心房特征的变化驱动。具体而言,研究人员发现,在调整 LA 压力后,年龄与松弛速度之间的关联完全减弱。 110 这一观察结果表明,随着年龄增长,舒张功能的变化可能与左心房顺应性和收缩特性的变化有关。 此外,在一项对未患 CVD 并接受血流动力学评估的志愿者进行的研究中,年龄与略高的静息肺毛细血管楔压相关(r=0.17;P<0.05)。 99 在生物衰老加速的患者中,舒张功能障碍发生年龄较早,并与运动能力下降和未来发生 HF 的风险相关。 114

Age-Related Changes in the Vasculature
血管系统与年龄相关的变化

Aging results in structural and functional changes to the large- and medium-sized arteries, even without atherosclerotic disease.115 The walls of arteries thicken due to hypertrophy of smooth muscle cells and expansion of the extracellular matrix with collagen content, which can irreversibly cross-link with advanced glycation end products.116,117 In addition, calcium deposition occurs in the arteries, and Mönckeberg arteriosclerosis develops.118,119 Together, these changes result in loss of elasticity and increased stiffness of the arterial structures.120 The capacity for endothelium-dependent vasodilation through nitric oxide signaling diminishes at the endothelium level.121–123 Concomitantly, the angiotensin-II production in the arterial wall increases, resulting in constrictive remodeling.117,124 Aging results in an increase in reactive oxidative species and low-grade chronic inflammation that worsens endothelial and microvascular dysfunction.125 Clinically, these changes manifest as mild increases in systolic blood pressure and a decline in diastolic blood pressure due to loss of elasticity and increased vascular stiffness. With age, the coronary arteries become dilated and have increased tortuosity, likely due to increased shear forces.126 In addition, the number of collateral vessels increases in response to the development of atherosclerosis. The development of lipid-rich plaques characteristic of atherosclerosis is not part of normal aging and is a result of accelerated biological aging.
即使没有动脉粥样硬化性疾病,衰老也会导致大中动脉的结构和功能发生变化。 115 由于平滑肌细胞肥大和细胞外基质随胶原含量扩张,动脉壁增厚,这可能会与晚期糖基化终产物发生不可逆的交联。 116,117 此外,钙沉积发生在动脉中,并发生门克贝格动脉硬化。 118,119 共同作用下,这些改变导致动脉结构失去弹性并增加硬度。 120 内皮依赖性血管舒张通过一氧化氮信号传导的能力在内皮水平下降。 121–123 同时,动脉壁中血管紧张素 II 的产生增加,导致血管收缩性重塑。 117,124 衰老导致活性氧增加和低度慢性炎症,从而加剧内皮和微血管功能障碍。 125 临床上,这些变化表现为收缩压轻度升高和舒张压下降,这是由于弹性丧失和血管僵硬度增加所致。随着年龄的增长,冠状动脉扩张并增加弯曲度,这可能是由于剪切力增加所致。 126 此外,侧支血管的数量会随着动脉粥样硬化的发展而增加。富含脂质的斑块是动脉粥样硬化的特征,它的发展并非正常衰老的一部分,而是生物衰老加速的结果。
The influence of lifestyle and environment on aging-related vascular changes is evident from recent studies of individuals from the indigenous Tsimane population in the Bolivian Amazon, who live a preindustrial lifestyle of hunting, gathering, fishing, and farming along the Maniqui River. In a seminal study of Tsimane individuals who underwent coronary artery calcium screening, investigators found a low prevalence of atherosclerosis, with 65% of octogenarians being free of coronary atherosclerosis.127 Similarly, the Yanomami and Ye’kwana peoples from a remote area of the Venezuelan rainforest have low blood pressure. The Yanomami, free entirely of Western influence, did not have an increase in blood pressure with age. In contrast, the Ye’kwena people are exposed to a Western lifestyle, including intermittent exposure to processed foods and salt, and were noted to have an age-related increase in blood pressure in early childhood (age, 1–20 years).128 These findings suggest that age-related changes to the vasculature, which are near universal in the industrialized world, are strongly influenced by lifestyle, diet, behaviors, and the environment and are related to accelerated aging.
生活方式和环境对衰老相关血管变化的影响,在近期对玻利维亚亚马逊地区奇曼人(Tsimane)的研究中可见一斑。奇曼人沿马尼基河(Maniqui River)生活,以狩猎、采集、捕鱼和农耕为生的前工业时代生活方式。 在一项对奇马内人(Tsimane)进行冠状动脉钙化筛查的开创性研究中,研究人员发现动脉粥样硬化的患病率很低,65%的八旬老人没有冠状动脉粥样硬化。 127 同样,来自委内瑞拉热带雨林偏远地区的亚诺玛米人和耶瓜纳人(Yanomami and Ye’kwana)的血压也很低。完全没有西方影响的亚诺玛米人,其血压没有随年龄增长而升高。 相比之下,耶库安纳人接触到西方生活方式,包括偶尔接触加工食品和盐,并注意到他们在儿童早期(1-20 岁)的血压出现与年龄相关的升高。[1] 这些发现表明,血管系统与年龄相关的变化在工业化世界几乎是普遍存在的,它受到生活方式、饮食、行为和环境的强烈影响,并且与加速衰老有关。

Age-Related Changes in Cardiovascular Response to Exercise
运动时心血管反应的年龄相关性变化

Aerobic exercise capacity, the gold-standard measure of cardiovascular performance and reserve, is objectively measured by peak exercise oxygen consumption (VO2peak). Aerobic capacity declines with age.129–131 Studies in healthy volunteers have demonstrated that VO2peak declines by 3% to 8% per decade of life between the ages of 20 and 70 years. However, the rate of decline may vary considerably based on physical activity levels, exercise training, and comorbidities.132 The decline in aerobic capacity accelerates considerably (>20% per 10 years) after the seventh decade of life.133 VO2peak is governed by the Fick principle and is defined as the product of peak exercise cardiac output and peak exercise peripheral arteriovenous oxygen difference (A-VO2diff; Figure 3). Changes in either the cardiac or peripheral reserve or both may account for the decline in VO2peak with aging, as discussed in more detail in the following sections.
有氧运动能力是心血管功能和储备的金标准测量方法,可通过峰值运动耗氧量(VO 2peak )进行客观测量。有氧运动能力随年龄增长而下降。 129–131 对健康志愿者的研究表明,在 20 至 70 岁之间,VO 2peak 每十年下降 3% 至 8%。然而,下降速度可能因体力活动水平、运动训练和合并症而异。 132 在七十岁之后,有氧能力下降明显加速(>10 年内下降 20%)。 133 VO 2peak 受 Fick 原理支配,定义为峰值运动心输出量和峰值运动外周动静脉氧含量差(A-VO 2diff )的乘积(图 3)。心脏或外周储备或两者兼有的变化可能导致 VO 2peak 随年龄增长而下降,这将在以下章节中更详细地讨论。
Figure 3. Mechanisms of age-related decline in exercise capacity. Oxygen delivery can be described by the Fick principle as the product of cardiac output and peripheral oxygen extraction. Aging results in a reduction in peak heart rate, and stroke volume results in reduced exercise cardiac output. Peripheral oxygen extraction at exercise declines or remains the same and is related to reduced capillary density and increased afterload. Together, these mechanisms result in a reduction in aerobic capacity with aging. PCWP indicates pulmonary capillary wedge pressure. Illustration credit: Sceyence Studios.
图 3. 运动能力随年龄增长而下降的机制。氧气输送可用 Fick 原理描述为心输出量和外周氧气提取的乘积。衰老导致峰值心率降低,而每搏输出量减少导致运动心输出量降低。运动时的外周氧气提取下降或保持不变,这与毛细血管密度降低和后负荷增加有关。 总之,这些机制导致有氧能力随年龄增长而下降。PCWP 表示肺毛细血管楔压。插图来源:Sceyence Studios。

Effects of Aging on Cardiac Reserve During Exercise
运动过程中衰老对心脏储备的影响

Underlying structural changes in the myocardium and vasculature result in alterations in the pressure-volume relationships, reducing stroke volume through the Frank-Starling mechanism. The inability to augment stroke volume in response to stress/exercise is characteristic of the aging process. In a cross-sectional study of volunteers free of CVD who underwent resting and exercise hemodynamic assessments, Pandey et al99 demonstrated that age was associated with a decline in VO2peak (40% decline over 5.5 decades) that was primarily driven by reductions in exercise cardiac output due to concurrent reductions in peak exercise stroke volume and heart rate. Aging is associated with a decline in peak heart rate, reduced heart rate responsiveness to autonomic nervous system input, and a decrease in heart rate variability.134–136 The age-related decline in exercise stroke volume is likely driven by a reduced inotropic reserve (reduced contractility with lower ejection fraction and lower end-systolic volume).99 Notably, the decline in peak exercise stroke volume is more prominent in older age (>60 years) than in middle age.99,137,138 In the seminal Dallas Bed Rest Study, no decline in stroke volume at peak exercise was noted on the 30-year follow-up visit (from ages of 20 to 50 years) but a prominent decline was observed at the subsequent 40-year follow-up (by the age of 60 years).138,139 This is particularly relevant considering the accelerated rate of decline in VO2peak that occurs after the sixth decade of life.133 Together, a decline in stroke volume reserve may have an important role in the age-related decline in VO2peak, particularly in the later decades of life.
心肌和血管的潜在结构性改变导致压力-容量关系发生变化,通过 Frank-Starling 机制减少了卒中容量。在应激/运动时无法增加卒中容量是衰老过程的特征。 在一项针对未患心血管疾病的志愿者的横断面研究中,这些志愿者接受了静息和运动血流动力学评估,Pandey 等人 99 证实,年龄与 VO 2peak 的下降有关(在 5.5 个十年中下降了 40%),这主要是由于运动时心输出量的减少所致,而心输出量的减少又是由于峰值运动量时每搏输出量和心率的同步下降所致。 衰老与峰值心率下降、对自主神经系统输入的反应性降低以及心率变异性降低有关 134–136 。与年龄相关的运动性每搏输出量下降可能是由变力储备降低(收缩力降低,射血分数和收缩末期容积降低)驱动的 99 。值得注意的是,峰值运动性每搏输出量的下降在老年(>60 岁)比在中年更明显。 99,137,138 在具有开创性的达拉斯卧床休息研究中,在 30 年随访时(从 20 岁到 50 岁),未观察到峰值运动时的每搏输出量下降,但在随后的 40 年随访时(到 60 岁时)观察到显著下降。 138,139 考虑到 60 岁之后 VO 2peak 的下降速度加快,这一点尤为重要。 133 总之,每搏输出量储备的下降可能在与年龄相关的 VO 2peak 下降中发挥重要作用,尤其是在生命后期。
The effect of aging on LV compliance and relaxation may also contribute to reduced stroke volume reserve.140,141 Some studies using invasive exercise hemodynamic assessments have shown a significant increase in submaximal and near-maximal exercise LV filling pressure and a reduction in the exercise cardiac output to LV filling pressure ratio with aging.141 However, the exaggerated filling pressure response to exercise in older adults is more evident in supine exercise hemodynamic assessment studies140,142 and less so with upright assessment.99 Among individuals with accelerated aging-related CVD, such as HFpEF, the disproportionate rise of filling pressure with exercise is pronounced and often considered pathognomic.143 These observations suggest that progressive decline in LV compliance may be a more consistent feature of accelerated cardiac aging.144
衰老对左心室顺应性和舒张功能的影响也可能导致每搏输出量储备减少。 140,141 一些使用有创运动血流动力学评估的研究表明,随着年龄的增长,次极量和接近极量运动时左心室充盈压显著增加,运动时心输出量与左心室充盈压的比率降低。 141 然而,老年人对运动时充盈压反应的过度增加在卧位运动血流动力学评估研究中更为明显 140,142 ,而在直立评估中则不那么明显 99 。在患有加速衰老相关 CVD 的个体中,如 HFpEF,运动时充盈压不成比例的升高非常明显,通常被认为是病理性的。 143 这些观察结果表明,左心室顺应性的进行性下降可能是加速心脏衰老的一个更一致的特征。 144

Effect of Aging on Peripheral Oxygen Extraction During Peak Exercise
衰老对峰值运动期间外周氧提取的影响

Most studies,139,145–150 though not all,99,137,141 indicate that peak exercise A-VO2diff declines with aging. Several peripheral factors may contribute to reductions in peak exercise A-VO2diff in older adults, including changes in skeletal muscle mass and quality, skeletal muscle blood flow and capillary density, and mitochondrial function. Reductions in skeletal muscle mass and quality are associated with lower VO2peak and physical function performance in older adults.151–153 Reduced muscle mass lowers the amount of metabolically active tissue available to extract and use oxygen during exercise, contributing to reduced peak exercise A-VO2diff observed in older adults. However, the relative contribution of reduced muscle mass to decreased peak exercise A-VO2diff is likely small.154 In contrast, abnormal skeletal muscle qualities are believed to play a larger role. In particular, increased accumulation of intermuscular adipose tissue (termed myosteatosis) is associated with reduced aerobic exercise tolerance,155–158 muscular strength,157,159,160 and physical function.155,156,160,161 Excess adiposity within skeletal muscle likely redirects blood flow away from active skeletal muscle during exercise, reducing muscle blood flow and diminishing functional capacity.162
多数研究( 139,145–150 尽管并非全部 99,137,141 )表明,峰值运动 A-VO 2diff 随年龄增长而下降。若干外周因素可能导致老年人峰值运动 A-VO 2diff 降低,包括骨骼肌质量和质量的变化、骨骼肌血流和毛细血管密度以及线粒体功能。骨骼肌质量和质量的降低与老年人较低的 VO 2peak 和身体功能表现相关。 151–153 肌肉量减少会降低代谢活跃组织的量,而这些组织原本可以在运动过程中提取和利用氧气,从而导致老年人观察到的峰值运动 A-VO 2diff 降低。然而,肌肉量减少对峰值运动 A-VO 2diff 降低的相对贡献可能很小。 154 相比之下,异常的骨骼肌质量被认为起着更大的作用。 特别是,肌间脂肪组织(称为肌脂过多症)的积累增加与有氧运动耐量 155–158 、肌力 157,159,160 和身体功能 155,156,160,161 的降低有关。骨骼肌内过多的脂肪可能在运动期间将血流从活跃的骨骼肌转移,从而减少肌肉血流量并降低功能能力。 162
Skeletal muscle blood flow is lower during submaximal163–166 and maximal exercises167 and is a major contributor to reduced VO2peak in older adults.167,168 While impairments in skeletal muscle blood flow may be due to declines in maximal cardiac output with age, studies have shown that muscle blood flow is reduced during both small muscle mass (single knee extensor and forearm exercise) and large muscle mass (cycling) exercise in older adults. These observations suggest that reduced limb blood flow during exercise may be secondary to peripheral limitations including lower vascular conductance (blood flow/mean arterial pressure).169 The impairment in vascular conductance with aging is the result of lowered nitric oxide and prostaglandin availability,170 coupled with increased endothelium-derived vasoconstrictor (ET-1),171 which together promote endothelial dysfunction and attenuated vasodilatory responses to exercise in older adults. In addition, aging impairs the ability to contract skeletal muscle to blunt sympathetic vasoconstriction (a process known as functional sympatholysis), which is critical for proper regulation of blood flow distribution and oxygen delivery within skeletal muscle during dynamic exercise.169,172,173 Finally, studies have shown that capillary density within skeletal muscle is up to 25% lower in older versus younger adults.174–176 Decreased skeletal muscle capillary density leads to reduced muscle perfusion and red blood cell transit time, which diminishes convective oxygen delivery and extraction during exercise.176,177 As such, skeletal muscle capillary density is an important determinant of peak exercise A-VO2diff.145,178 Together, decreases in skeletal muscle blood flow and capillary density may be major contributors to reduced peak exercise A-VO2diff observed in older adults, as both significantly impact oxygen delivery and uptake within skeletal muscle.155,179
老年人在次极量 163–166 和极量运动 167 期间,骨骼肌血流量较低, 2peak 这是导致 VO 降低的主要原因。 167,168 虽然骨骼肌血流量的下降可能是由于年龄增长导致的最大心输出量下降,但研究表明,老年人在小肌肉群(单膝伸展和前臂运动)和大肌肉群(自行车)运动期间,肌肉血流量都会减少。 这些观察结果表明,运动期间肢体血流量的减少可能是周围限制(包括较低的血管传导性(血流量/平均动脉压))的继发结果。 169 血管传导性随年龄增长而降低是由于一氧化氮和前列腺素的可用性降低, 170 加上内皮衍生血管收缩剂(ET-1)的增加, 171 共同促进内皮功能障碍和老年人运动时血管舒张反应减弱。 此外,衰老会损害收缩骨骼肌以减轻交感神经血管收缩的能力(这一过程被称为功能性交感神经溶解),这对于动态运动期间骨骼肌内血流分布和氧气输送的适当调节至关重要。 169,172,173 最后,研究表明,老年人骨骼肌内的毛细血管密度比年轻人低 25%。 174–176 骨骼肌毛细血管密度降低会导致肌肉灌注减少和红细胞通过时间延长,从而减少运动期间的对流氧气输送和提取。 176,177 因此,骨骼肌毛细血管密度是峰值运动 A-VO 2diff 的一个重要决定因素。 145,178 总之,骨骼肌血流和毛细血管密度的降低可能是导致老年人观察到的峰值运动 A-VO 2diff 降低的主要因素,因为两者都显着影响骨骼肌内的氧气输送和摄取。 155,179

Physical Activity, Fitness, and Cardiovascular Health
体力活动、身体素质与心血管健康

Physical Activity and Accelerated Aging
体力活动与加速衰老

Physical activity levels over the life course are an important prognostic indicator of adverse outcomes.180 Physical activity is defined as energy utilized beyond basal metabolic rate, can include all activities of daily life, and is measured as kilocalories or metabolic equivalents. Historically, it was assessed by self-report but recently has been measured objectively using wearable accelerometers. Higher levels of physical activity have been associated with reduced biological age, as assessed by the Phenotypic Age Acceleration model in the National Health and Nutrition Examination Survey cohort.181 Across multiple cohort studies, higher physical activity levels are a strong predictor of healthy aging, as defined by different metrics including functional independence and less decline in functional status,182,183 lower frailty burden,184 and the composite of absence of chronic disease, functional, and cognitive impairments.185 The association of higher physical activity levels with healthy aging is consistent in middle age and older age185 and independent of baseline functional status.183
生命历程中的体力活动水平是预测不良结局的重要指标。 180 体力活动被定义为超出基础代谢率所消耗的能量,可以包括所有的日常生活活动,并以千卡或代谢当量来衡量。过去,体力活动通常通过自我报告进行评估,但最近开始使用可穿戴加速度计进行客观测量。 在全国健康和营养调查队列中,通过表型年龄加速模型评估,较高水平的体力活动与生物学年龄的降低有关。 181 在多项队列研究中,较高的体力活动水平是健康老龄化的有力预测指标,健康老龄化由不同的指标定义,包括功能独立性和功能状态下降较少, 182,183 较低的虚弱负担, 184 以及没有慢性疾病、功能和认知障碍的综合指标。 185 较高的体力活动水平与健康老龄化之间的关联在中年和老年均一致, 185 并且与基线功能状态无关。 183

Physical Activity and Risk of Mortality and Incident CVDs
体力活动与死亡和心血管疾病发病风险

Higher self-reported physical activity levels have been consistently associated with reduced risk of mortality and CVD in several epidemiological cohort studies.186–188 These observations are consistent with self-reported and objectively measured physical activity levels (Table 3). A large individual-level meta-analysis of 8 cohort studies that included 36 383 individuals with objectively measured physical activity levels demonstrated that higher overall physical activity levels were associated with lower mortality risk.189 However, a nonlinear dose-response relationship was noted between total physical activity volume and all-cause mortality, with the most significant risk reduction noted in moving from the lowest quartile to the next quartile of physical activity (52% risk reduction) and modest marginal improvements with subsequent increments in physical activity levels. These observations are consistent with the current guidelines recommend 150 to 300 min/wk of moderate to vigorous aerobic physical activity190
在多项流行病学队列研究中,较高的自我报告体力活动水平与死亡率和心血管疾病风险降低持续相关。 186–188 这些观察结果与自我报告和客观测量的体力活动水平相符(表 3)。 一项对 8 个队列研究进行的大型个体水平荟萃分析(纳入了 36 383 名具有客观测量的体力活动水平的个体)表明,较高的总体体力活动水平与较低的死亡风险相关。 189 然而,总的体力活动量与全因死亡率之间存在非线性剂量-反应关系,其中最显著的风险降低出现在从最低四分位数到下一个四分位数的体力活动(风险降低 52%),而随后的体力活动水平增加带来的边际改善较为适度。 这些观察结果与目前的指南相符,指南建议每周进行 150 至 300 分钟的中等至高强度有氧体力活动 190
Table 3. Summary Findings From Key Studies Evaluating the Association Between Objectively Measured Physical Activity Levels With Adverse Cardiovascular Outcomes
表 3. 评估客观测量的体力活动水平与不良心血管结局之间关系的关键研究的总结性发现
Afib indicates atrial fibrillation; CHD, coronary heart disease; CV, cardiovascular; CVD, cardiovascular disease; HF, heart failure; HFpEF, heart failure with preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction; and MI, myocardial infarction.
Afib 指房颤;CHD,冠心病;CV,心血管;CVD,心血管疾病;HF,心力衰竭;HFpEF,射血分数保留型心力衰竭;HFrEF,射血分数降低型心力衰竭;MI,心肌梗死。
In contrast, recent studies have observed a more graded and linear dose-response relationship for objectively measured physical activity levels and risk of nonfatal incident CVD. A contemporary analysis from the UK Biobank cohort, which included 103 687 participants with objective measures of physical activity by accelerometers, demonstrated a 27% to 53% risk reduction in incident CVD in the second and fourth (most active) quartiles, respectively, compared with the referent (least active) group.191 These findings suggest that the cardiovascular benefit of physical activity may be more consistent and graded across the spectrum of the dose-response curve. The cardiovascular benefits of light-intensity physical activity in reducing the risk of adverse CV events at moderate doses seem to be more exaggerated among older adults. A recent analysis of 8 prospective studies, which included 20 152 individuals with objectively measured physical activity in the form of daily step counts, found that 6000 to 9000 steps per day were associated with a 40% to 50% lower risk of incident CVD among older adults over the age of 60 years.192 In contrast, no association was noted among younger adults. Similar findings were reported in the Women’s Health Initiative cohort, where higher levels of light physical activity were associated with a graded reduction in incident CVD independent of cardiovascular risk factors among older women (mean age, 78.5 years).193
相比之下,近期的研究观察到,客观测量的体力活动水平与非致命性突发心血管疾病风险之间存在更均衡和线性的剂量-反应关系。一项来自英国生物样本库队列的当代分析,纳入了 103687 名通过加速计客观测量体力活动的参与者,结果表明,与参考组(活动量最少组)相比,第二和第四(活动量最大)四分位数中,突发心血管疾病的风险分别降低了 27%至 53%。 191 这些发现表明,体力活动对心血管的益处可能在剂量-反应曲线的整个范围内更加一致和分级。对于老年人而言,适度剂量的轻度体力活动在降低不良心血管事件风险方面的心血管益处似乎更为显著。 最近一项对 8 项前瞻性研究的分析,纳入了 20152 名通过每日步数客观测量体力活动的人,发现每天 6000 至 9000 步与 60 岁以上老年人新发 CVD 风险降低 40%至 50%相关 192 。相比之下,在年轻人中未观察到这种关联。 在“妇女健康倡议”队列中也报告了类似的结果,其中较高的轻度体力活动水平与老年妇女(平均年龄 78.5 岁)心血管疾病事件的逐步减少相关,且与心血管危险因素无关。 193

Physical Activity and Risk of Incident HF and Its Subtypes
体力活动与新发心力衰竭及其亚型的风险

A linear dose-dependent relationship has been reported between physical activity levels and the risk of HF.194–198 This is consistent with self-reported and objectively measured physical activity levels. In a pooled analysis of 12 prospective cohort studies with over 370 460 participants, participants engaging in guideline-recommended levels of physical activity (500 metabolic equivalents-min/wk) had modest reductions in HF risk (10% lower risk). In contrast, large reductions in HF risk were noted among those reporting physical activity at twice (19% lower risk) and 4× (35% lower risk) the guideline-recommended levels.196 Similar observations were also reported in the UK Biobank cohort using objectively measured physical activity, where higher levels of moderate physical activity were associated with a lower risk of incident HF in a graded fashion up to high levels much beyond the current guideline-recommended levels of 300 min/wk (66% lower risk).199 It is noteworthy that the association between physical activity and HF risk differs for HF subtypes, HFpEF and HF with reduced ejection fraction (HFrEF), such that higher physical activity levels are more strongly associated with a lower risk of HFpEF but not HFrEF. In a pooled analysis, leisure-time physical activity level above 1000 metabolic equivalents-min/wk was associated with a 19% risk reduction for HFpEF. In contrast, physical activity levels were not associated with HFrEF risk.200 Similar findings of a stronger association between physical activity levels and risk of HFpEF but not HFrEF were confirmed in the Objective Physical Activity and Cardiovascular Health in Older Women study using objective measures of physical activity levels.197
一项研究报告指出,体力活动水平与心力衰竭风险之间存在线性剂量依赖关系。 194–198 这与自我报告和客观测量的体力活动水平相符。在一项对超过 370460 名参与者的 12 项前瞻性队列研究的汇总分析中,参与符合指南推荐水平体力活动(500 代谢当量-分钟/周)的参与者,其心力衰竭风险略有降低(风险降低 10%)。 相比之下,报告身体活动量达到指南推荐量 2 倍(风险降低 19%)和 4 倍(风险降低 35%)的人群,心力衰竭风险显著降低。 196 英国生物样本库队列也报告了类似的观察结果,该研究使用了客观测量的体力活动,结果显示,较高水平的适度体力活动与较低的心力衰竭发生风险相关,并且呈梯度关系,直至远高于当前指南推荐的每周 300 分钟的水平(风险降低 66%)。 199 值得注意的是,体力活动与心力衰竭(HF)风险之间的关联因心力衰竭亚型而异,表现为射血分数保留型心力衰竭(HFpEF)和射血分数降低型心力衰竭(HFrEF),较高的体力活动水平与较低的 HFpEF 风险更密切相关,但与 HFrEF 则不然。在一项汇总分析中,高于 1000 代谢当量-分钟/周的休闲时间体力活动水平与 HFpEF 风险降低 19%相关。相比之下,体力活动水平与 HFrEF 风险无关。 200 在老年妇女客观体力活动与心血管健康研究中,使用体力活动水平的客观测量也证实了类似的发现,即体力活动水平与 HFpEF 风险之间存在更强的关联,但与 HFrEF 的关联则不然。 197
In addition to physical activity levels, higher levels of cardiorespiratory fitness, a gold-standard integrative measure of exercise capacity affected by aging as discussed previously, have also been associated with lower risk of adverse CV outcomes, particularly HFpEF. There is a monotonic decline in VO2peak with aging in middle age, with an accelerated decline noted in older adults (>60 years).133 Seminal studies from the Cooper Center for Longitudinal Study have demonstrated that lower levels of cardiorespiratory fitness in mid-life and greater decline in cardiorespiratory fitness with aging are strongly associated with higher risk of HF in older age.194,201 Furthermore, the association of baseline and decline in cardiorespiratory fitness with the risk of HF is stronger for HFpEF than HFrEF.47 The stronger association of physical activity and cardiorespiratory fitness levels with the risk of HFpEF but not HFrEF is particularly relevant in the context of HFpEF representing an accelerated cardiac aging phenotype. It highlights the potential role of physical activity and exercise interventions in mitigating accelerated aging-related CVD.
除了体力活动水平,较高的心肺适能水平(这是衡量运动能力的金标准综合指标,并会受到衰老的影响,如前所述)也与较低的不良心血管事件风险相关,尤其是射血分数保留型心力衰竭 (HFpEF)。随着年龄增长,中年人的 VO 2peak 呈单调下降趋势,老年人(>60 岁)则出现加速下降。 133 库珀纵向研究中心的重要研究表明,中年时期较低的心肺适能水平以及随着年龄增长心肺适能的更大程度下降与老年时期较高的心力衰竭风险密切相关。 194,201 此外,基线心肺适能和心肺适能下降与心力衰竭风险的关联对于射血分数保留型心力衰竭 (HFpEF) 比射血分数降低型心力衰竭 (HFrEF) 更强。 47 身体活动和心肺适能水平与 HFpEF 风险的关联更强,但与 HFrEF 的关联不强,这在 HFpEF 代表加速心脏衰老表型的情况下尤为重要。这突出了身体活动和运动干预在减轻与加速衰老相关的 CVD 方面的潜在作用。

Physical Activity, Cardiorespiratory Fitness, and Cardiac Structure and Function
体力活动、心肺健康与心脏结构和功能

Several observational cohort studies have demonstrated a strong, graded association between higher physical activity and cardiorespiratory fitness, and lower prevalence of CVD risk factors, including diabetes, hypertension, hyperlipidemia, and obesity.202–206 However, differences in antecedent development of CVD risk factors do not fully mediate the high physical activity and cardiorespiratory fitness–associated reduction in the risk of HF, implicating a more direct effect on the cardiovascular structure and function.201 Development of HF with aging is characterized by intermediate development of subclinical abnormalities in cardiac structure and function. Numerous observational studies have demonstrated a predictable and consistent association between physical activity, cardiorespiratory fitness, and subclinical HF phenotypes. In the CARDIA study, lower cardiorespiratory fitness in young adulthood was associated with measures of abnormal LV systolic and diastolic function in middle age, and the association between low cardiorespiratory fitness and adverse diastolic function was independent of traditional cardiovascular risk factor burden.207 Moreover, in the Dallas Heart Study, low cardiorespiratory fitness and objectively measured physical activity levels among middle-aged adults were associated with higher peak systolic circumferential strain (worse cardiac contractility), irrespective of risk factor burden, demonstrating an independent association between low fitness levels and adverse cardiac remodeling.208,209 Notably, higher levels of cardiorespiratory fitness among participants of the Cooper Center for Longitudinal Study were associated with better diastolic function and less concentric remodeling.210 Together, these observations suggest that higher levels of physical activity and cardiorespiratory fitness may contribute to better cardiometabolic health and attenuate the development of accelerated cardiac aging phenotype.
多项观察性队列研究表明,较高的体力活动和心肺功能与较低的 CVD 风险因素(包括糖尿病、高血压、高脂血症和肥胖)患病率之间存在显著且分级的关联。 202–206 然而,心血管疾病风险因素先前发展的差异并不能完全解释高体力活动和心肺适能相关的心力衰竭风险降低,这意味着对心血管结构和功能有更直接的影响。 201 随着年龄增长而发生的心力衰竭的特征是心脏结构和功能亚临床异常的中间发展阶段。 大量观察性研究已证实,体力活动、心肺适能和亚临床 HF 表型之间存在可预测且一致的关联。在 CARDIA 研究中,青年时期较低的心肺适能与中年时期异常的左心室收缩和舒张功能相关,并且低心肺适能与不良舒张功能之间的关联独立于传统心血管风险因素负担。 207 此外,在达拉斯心脏研究中,中年人的低心肺适能和客观测量的体力活动水平与较高的峰值收缩环向应变(较差的心脏收缩力)相关,而与风险因素负担无关,这表明低适能水平与不良心脏重塑之间存在独立的关联。 208,209 值得注意的是,在库珀纵向研究中心参与者中,较高的心肺适能水平与更好的舒张功能和更少的心脏向心性重构相关联。 210 总之,这些观察结果表明,较高水平的体力活动和心肺适能可能有助于更好的心血管代谢健康,并减轻加速心脏衰老表型的发生。

Exercise Training, Accelerated Aging, and Cardiovascular Health
运动训练、加速衰老和心血管健康

Effects of Exercise Training on Accelerated Aging Phenotype
运动训练对加速衰老表型的影响

Small randomized controlled trials (RCTs) have evaluated the effects of different exercise training interventions on biological aging. In a single-center RCT of 30 middle-aged participants, a 4-week high-intensity exercise training significantly reduced biological age as assessed by a transcriptomic clock model.211 Benefits of exercise on reversing accelerated biological aging are augmented by add-on dietary and nutritional interventions. To this end, in a secondary analysis of an RCT of 107 participants who underwent different diet and exercise interventions, diet and combination of diet with exercise significantly reduced biological age as assessed by different models (the Klemera-Doubal method biological age, the homeostatic dysregulation score, and the health aging index).212 In a larger Vitamin D3 - Omega-3 - Home Exercise - Healthy Aging and Longevity Trial that enrolled 777 participants, a combination of exercise with nutritional supplementation of omega-III fatty acids significantly reduced biological age as assessed by DNA methylation-based models (Phenotypic Age Acceleration, GrimAge, and GrimAge2).213 Together, these observations highlight the effectiveness of exercise, alone or combined with dietary interventions, in reversing accelerated aging phenotypes.
一些小型随机对照试验 (RCT) 已经评估了不同运动训练干预对生物衰老的影响。在一项针对 30 名中年参与者的单中心 RCT 中,为期 4 周的高强度运动训练显著降低了生物年龄,这是通过转录组时钟模型评估的。 211 运动在逆转加速生物衰老方面的益处可以通过附加的饮食和营养干预来增强。 为此,在一项针对 107 名受试者进行不同饮食和运动干预的 RCT 的二次分析中,饮食以及饮食与运动的结合显著降低了通过不同模型评估的生物学年龄(Klemera-Doubal 法生物学年龄、稳态失调评分和健康老龄化指数)。 212 在一项纳入了 777 名参与者的大型维生素 D3 - Omega-3 - 居家运动 - 健康衰老与长寿试验中,运动与 omega-III 脂肪酸营养补充剂的结合显著降低了通过基于 DNA 甲基化的模型(表型年龄加速、GrimAge 和 GrimAge2)评估的生物学年龄。 213 总之,这些观察结果突出了运动(单独或与饮食干预相结合)在逆转加速衰老表型方面的有效性。

Effects of Exercise Training on Cardiorespiratory Fitness and Cardiometabolic Outcomes
运动训练对心肺适能和心脏代谢结果的影响

Several trials and associated meta-analyses have demonstrated improvements in cardiometabolic parameters with exercise training. A significant increase in VO2peak with exercise training has been shown in older individuals without CVD (including sedentary214,215 and overweight/obese216 cohorts) and among those with CAD,217,218 HFrEF219 (Heart Failure: A Controlled Trial Investigating Outcomes of Exercise Training,220 Study of Myocardial Recovery After Exercise Training in Heart Failure221 and the Exercise Rehabilitation Trial222), and HFpEF.223–228 Direct comparisons of outcomes between studies of different populations should be interpreted with caution, but, on average, exercise training appears to provide higher increases in VO2peak in older sedentary or overweight/obese populations (>3.5 mL/kg per min)214–216 compared with patients with CAD (>2.5 mL/kg per min)218 and those with HF (0.4–2.7 mL/kg per min).220,223,229–233
多项试验和相关的荟萃分析表明,运动训练可以改善心脏代谢参数。 在没有 CVD 的老年人(包括久坐 214,215 和超重/肥胖 216 人群)以及 CAD 217,218 、HFrEF 219 患者(心力衰竭:一项调查运动训练结果的对照试验, 220 心力衰竭运动训练后心肌恢复研究 221 和运动康复试验 222 )和 HFpEF 患者中,运动训练可显著提高 VO 2peak223–228 不同人群研究结果的直接比较应谨慎解读,但平均而言,运动训练似乎能使老年久坐或超重/肥胖人群的 VO2max增加幅度更高(>3.5 mL/kg/min), 214–216 而冠心病(CAD)患者(>2.5 mL/kg/min) 218 和心力衰竭(HF)患者(0.4–2.7 mL/kg/min)的增加幅度相对较低。 220,223,229–233
In addition to cardiorespiratory fitness, exercise training studies have demonstrated benefits for various cardiovascular and metabolic health parameters.234,235 Across various primary and secondary prevention populations, exercise training has improved blood pressure,236 hemoglobin A1c in diabetes237,238 and prediabetes,239 lipid profile,240,241 and combined CVD risk profiles.242 Exercise training also typically reduces body weight/waist circumference (in many studies of different populations),243 but may even more reliably reduce pathogenic metrics such as adipose tissue or visceral fat with associated stable or decreased body weight.238,244,245 Individual studies and associated populations/outcomes are too numerous and diverse to highlight each individually. Among this array of studies, select notable trials contributing to the findings above include the Comparison of the Cardiovascular Benefits of Resistance, Aerobic, and Combined Exercise trial242 (n=406 adults with overweight/obesity), the Sapporo Fitness Club Trial246 (n=561 adults with overweight/obesity and 2 additional CVD risk factors), and Studies of Targeted Risk Reduction Interventions through Defined Exercise trials (n=111 and n=137, respectively; sedentary, overweight/obese adults with dyslipidemia) programs.240,247
除了心肺适能外,运动训练研究也证实了其对多种心血管和代谢健康参数的益处。 234,235 在各种一级和二级预防人群中,运动训练改善了血压, 236 糖尿病 237,238 和糖尿病前期患者的糖化血红蛋白(hemoglobin A1c), 239 血脂谱, 240,241 以及综合心血管疾病(CVD)风险状况。 242 运动训练通常也能减轻体重/腰围(在许多不同人群的研究中), 243 但可能更可靠地减少致病指标,如脂肪组织或内脏脂肪,同时体重保持稳定或下降。 238,244,245 由于各个研究及其相关人群/结局数量众多且各不相同,因此无法在此逐一强调。 在这些研究中,对上述发现做出贡献的著名试验包括:阻力、有氧和联合运动对心血管益处的比较试验 242 (n=406,超重/肥胖成人)、札幌健身俱乐部试验 246 (n=561,超重/肥胖且具有 2 项额外 CVD 风险因素的成人)以及通过定义的运动试验进行靶向风险降低干预研究(分别为 n=111 和 n=137;久坐、超重/肥胖且患有血脂异常的成人)项目。 240,247

Central Versus Peripheral Mechanisms of Exercise Training Benefits
运动训练获益的中心机制与外周机制

Several mechanisms underlie the beneficial effects of exercise training (Figure 4). From the standpoint of improvement in exercise capacity, as assessed by VO2peak, the mechanisms can be broadly characterized as effects on the 2 key components of the Fick equation: exercise cardiac output (central effect) and peripheral oxygen extraction at peak exercise (peripheral effects).
运动训练产生有益效果有几个潜在机制(图 4)。从提高运动能力的角度来看,如通过 VO 2peak 评估,这些机制可以大致分为对 Fick 方程的 2 个关键组成部分的影响:运动心输出量(中枢效应)和峰值运动时的外周氧提取(外周效应)。
Figure 4. Multisystem mechanisms of benefits of exercise training. Exercise training results in beneficial adaptations in multiple tissues, which attenuate the changes associated with aging. Exercise training results in increased cardiac compliance, improvement in stroke volume, and cardiac output. At the level of the peripheral vasculature, improvement in peak exercise oxygen extraction occurs as a result of improved endothelial function and muscle blood flow. The skeletal muscle size increases with a resultant increase in contractile function. Reduction in visceral and ectopic adiposity contributes to improved insulin sensitivity and reduction in inflammation. At the subcellular level, mitochondrial content and energetics improve with exercise. Together, these adaptations result in improved aerobic capacity, increased strength that results in better physical function, reduced burden of frailty, and greater independence. Illustration credit: Sceyence Studios.
图 4. 运动训练获益的多系统机制。运动训练可在多个组织中产生有益的适应性改变,从而减轻与衰老相关的变化。运动训练可提高心脏顺应性,改善每搏输出量和心输出量。在外周血管层面,由于内皮功能和肌肉血流的改善,峰值运动氧提取率得以提高。 骨骼肌体积增大,导致收缩功能增强。内脏和异位脂肪减少有助于提高胰岛素敏感性并减轻炎症。在亚细胞水平上,线粒体含量和能量学随着运动而改善。总之,这些适应导致有氧能力提高,力量增强,从而改善身体功能,减轻虚弱负担,并提高独立性。 插图鸣谢:Sceyence Studios。

Favorable Effects of Exercise Training on Cardiac Exercise Reserve
运动训练对心脏储备功能的有利影响

Mechanistic studies from athletes and those individuals who participate in a high volume of lifelong exercise provide mechanistic and phenomenological insight into how exercise alters cardiac structure and function to improve exercise cardiac performance favorably. The cardiac hypertrophy in athletes, athlete’s heart, has been well described in the literature.248–250 This adaptive hypertrophy of the heart occurs in response to the exercise training stimuli, resulting in larger cardiac size, stroke volume, higher VO2peak, and cardiac output.251 A major contribution to our understanding of the physiological effects of exercise training on cardiac performance across the age spectrum has been made by a series of studies done by Levine et al65 over the last 2 decades.139,146,252–255 In a seminal study from his group, Arbab-Zadeh et al65 demonstrated that athletes with prolonged and sustained endurance training over the life course have higher stroke volume and better LV compliance than sedentary but healthy older adults. Moreover, LV compliance in older endurance athletes did not differ from that of younger controls. These findings suggest that exercise training may prevent age-related LV stiffness and diastolic impairment. A subsequent study by the group characterized the relationship between lifelong exercise dose and cardiac compliance in older age.252 Older lifelong competitive exercisers (>3 days of exercise per week) had better LV compliance than their sedentary counterparts. In addition, LV compliance and VO2peak increased with higher doses of lifelong exercise, suggesting that exercise volume over the lifetime directly results in positive adaptations on diastolic function. Furthermore, prolonged, high-intensity exercise training can reverse some adverse cardiac remodeling noted in sedentary middle-aged adults. Specifically, Howden et al255 demonstrated that 2 years of high-intensity training improved cardiorespiratory fitness and LV stiffness among healthy, middle-aged, sedentary adults. Heida et al observed similar improvements in LV stiffness with 1-year of high-intensity training among those with LV hypertrophy and elevated cardiac biomarkers (stage B HF).256 It is noteworthy that the favorable effects of high intensity, prolonged exercise training on LV stiffness have been noted in middle-aged adults but not older adults or those with established HFpEF. Specifically, 1 year of vigorous exercise training in older adults did not reverse LV stiffness or improve exercise cardiac output or stroke volume reserve.253 Still, it did increase arterial elastance and improved cardiorespiratory fitness. Similarly, among patients with established HFpEF, a 1-year progressive endurance exercise training program did not improve LV compliance, arterial stiffness, or exercise capacity.254 These observations suggest that among older sedentary adults or those with established HFpEF, the myocardial stiffness may be less amenable to improvement with exercise, and the favorable effects of exercise on VO2peak may be more related to its effects on peripheral oxygen extraction as discussed in the following.
来自运动员和终身参与大量运动的个体的机制研究,为我们提供了关于运动如何改变心脏结构和功能,从而有利地改善运动时心脏功能的机制性和现象学方面的深入见解。运动员的心脏肥大,即“运动员心脏”,在文献中已有充分描述。 248–250 这种心脏的适应性肥大是对运动训练刺激的反应,导致心脏尺寸增大、卒中量增加、VO 2peak 值升高以及心输出量增加。 251 Levine 等人 65 在过去 20 年中所做的一系列研究,对我们理解运动训练对不同年龄段人群心脏功能的生理影响做出了重要贡献。 139,146,252–255 在他所在研究小组的一项具有开创性的研究中,Arbab-Zadeh 等人 65 证明,与久坐但健康的年长者相比,一生中接受长期和持续耐力训练的运动员具有更高的卒中量和更好的左心室顺应性。此外,年长耐力运动员的左心室顺应性与年轻对照组没有差异。这些发现表明,运动训练可能预防与年龄相关的左心室僵硬和舒张功能障碍。 该小组随后的研究描述了终生运动剂量与老年人心脏顺应性之间的关系。 252 年长的终生竞技运动者(每周运动 >3 天)比久坐的同龄人具有更好的左心室顺应性。此外,左心室顺应性和 VO 2peak 随着终生运动剂量的增加而增加,表明终生运动量直接导致舒张功能的积极适应。 此外,长时间、高强度的运动训练可以逆转久坐不动的中年人出现的一些不良心脏重塑。具体而言,Howden 等人 255 证实,在健康、中年、久坐不动的成年人中,为期 2 年的高强度训练改善了心肺功能和左心室(LV)僵硬度。Heida 等人观察到,在患有 LV 肥厚和心脏生物标志物升高(B 期心力衰竭,HF)的患者中,为期 1 年的高强度训练也观察到类似的 LV 僵硬度改善。 值得注意的是,高强度、长时间的运动训练对左心室僵硬度的有利影响已在中年人中得到证实,但在老年人或已确诊的射血分数保留型心力衰竭(HFpEF)患者中未见报道。具体而言,老年人进行为期 1 年的剧烈运动训练并未逆转左心室僵硬度,也未改善运动时的心输出量或每搏输出量储备 253 。尽管如此,它确实增加了动脉弹性,并改善了心肺功能。 同样,在已确诊为 HFpEF 的患者中,为期 1 年的渐进式耐力运动训练计划并未改善 LV 顺应性、动脉僵硬度或运动能力。 254 这些观察结果表明,在老年久坐的成年人或已确诊为 HFpEF 的患者中,心肌僵硬度可能不太容易通过运动得到改善,而运动对 VO 2peak 的有利影响可能更多地与其对外周氧气提取的影响有关,如下文所述。

Favorable Effects of Exercise Training on Peripheral Oxygen Extraction in Skeletal Muscles
运动训练对骨骼肌外周氧气提取的有利影响

Skeletal muscle is remarkably adaptive in its structure and function to physiological stimuli such as exercise, even in older sedentary adults.257 Indeed, 3 months of aerobic exercise training in sedentary older adults have been shown to improve VO2peak by ≈18% secondary to increases in peak exercise A-VO2diff, with no changes in peak cardiac output.145 Contrary to effects on LV stiffness, which vary by age, exercise training elicits similar skeletal muscle adaptations in older and younger sedentary individuals.145,258 These include modulation of functional sympatholysis,259,260 increase in skeletal muscle mitochondrial content261–264 and function,263,264 capillarizations,262,265 and capillary-to-fiber ratio.265,266 Increases in capillary density increase red blood cell mean transit time and the area for diffusion while decreasing the diffusion distance from the red blood cell to the muscle fiber.177 Furthermore, increases in skeletal muscle mitochondrial content and function improve oxygen utilization and ATP production. These exercise training–induced favorable changes in skeletal muscle are similar to those observed in athletes and contribute to increased peak exercise A-VO2dif.169,258,259,261
即使在老年久坐的成年人中,骨骼肌在结构和功能上对运动等生理刺激也具有显著的适应性。 257 事实上,研究表明,久坐的老年人经过 3 个月的有氧运动训练后,由于峰值运动 A-VO 2diff 增加,VO 2peak 改善了约 18%,而峰值心输出量没有变化。 145 与对左心室僵硬度的影响(随年龄而异)相反,运动训练在老年和年轻的久坐个体中引起类似的骨骼肌适应。 145,258 这些包括功能性交感神经溶解的调节, 259,260 骨骼肌线粒体含量 261–264 和功能的增加, 263,264 毛细血管化, 262,265 以及毛细血管与纤维比率。 265,266 毛细血管密度增加会延长红细胞的平均通过时间和扩散面积,同时缩短红细胞到肌纤维的扩散距离。 177 此外,骨骼肌线粒体含量和功能的增加可改善氧气利用率和 ATP 的产生。这些运动训练引起的骨骼肌的有利变化与在运动员中观察到的相似,并有助于增加峰值运动 A-VO 2dif169,258,259,261
Among older patients with HFpEF, supervised exercise training improves exercise capacity mainly through favorable effects on peripheral mechanisms.267,268 Haykowsky et al267 demonstrated that among older adults with HFpEF, the favorable effect of exercise training on cardiac output was only responsible for 16% of the improvement in VO2peak, and the increase in peripheral oxygen extraction was responsible for most of the aerobic capacity improvement. A subanalysis from the Optimising Exercise Training in Prevention and Treatment of Diastolic Heart Failure trial (OptimEX-Clin) with patients with chronic stable HFpEF demonstrated that aerobic exercise training improved skeletal muscle energy metabolism and reduced markers of muscle atrophy after 3 months of exercise training.269 Together, these findings suggest a more pronounced role of exercise training on skeletal muscle function in exerting its beneficial effects in patients with HFpEF.
在患有 HFpEF 的老年患者中,监督下的运动训练主要通过对周围机制的有利影响来提高运动能力。 267,268 Haykowsky 等人 267 证明,在患有 HFpEF 的老年人中,运动训练对心输出量的有利影响仅占 VO 2peak 改善的 16%,而周围氧气提取的增加是大部分有氧能力改善的原因。 一项对慢性稳定型射血分数保留型心力衰竭(HFpEF)患者进行的“优化运动训练预防和治疗舒张性心力衰竭试验(OptimEX-Clin)”的子分析表明,有氧运动训练改善了骨骼肌能量代谢,并在运动训练 3 个月后减少了肌肉萎缩的标志物。 269 总之,这些发现表明,运动训练在骨骼肌功能方面发挥了更显著的作用,从而在 HFpEF 患者中发挥其有益作用。
As such, the response to exercise training in different organ systems remains heterogeneous and an area of extensive investigation.270 The ongoing multicenter Molecular Transducers of Physical Activity Consortium study has identified thousands of exercise training–induced changes across a wide variety of target tissues, highlighting that exercise leads to a pleiotropic, whole-organism-wide stimulus that interacts with genetic, cellular, and subcellular functions.271–273 In this way, the biological effects of exercise seem to mirror the aging process, where aging occurs across multiple organ systems and results in cellular and subcellular changes, the cumulative effects of which emerge as biological aging.
因此,不同器官系统对运动训练的反应仍然是异质性的,并且是一个广泛的研究领域。 270 正在进行的多中心“体力活动分子转导器联盟”研究已经确定了运动训练在多种目标组织中引起的数千种变化,强调运动会导致多效性的、全身性的刺激,并与遗传、细胞和亚细胞功能相互作用。 271–273 这样看来,运动的生物学效应似乎与衰老过程相似,衰老发生在多个器官系统中,并导致细胞和亚细胞的变化,这些变化的累积效应表现为生物学衰老。

Exercise Interventions for Prevention and Management of HF
用于预防和控制心力衰竭的运动干预措施

Exercise and Physical Activity Interventions for HF Prevention in At-Risk Older Adults
针对有风险老年人预防心力衰竭的运动和体力活动干预措施

Adequately powering and executing RCTs evaluating the effects of structured exercise training and physical activity interventions on the risk of HF in otherwise healthy, community-dwelling adults remain challenging. As a result, few individual trials have been successfully performed to assess the effects of exercise training on HF incidence as a secondary or exploratory outcome. The Generation-100 RCT274 remains one of the largest and longest exercise training studies in healthy participants (n=1567), assessing 5 years of exercise training (moderate-intensity, continuous exercise training [MICT] versus high-intensity interval training [HIIT] versus activity recommendation) in a specific population: healthy and fit older patients (aged 70–77 years) in Norway. In this study, exercise training resulted in an improvement in cardiovascular risk factors but did not accrue enough HF events to assess its effect on the same.275 In the LIFE study (n=1635),276 structured physical activity intervention reduced major mobility disability but did show a significant effect on the risk of HF, again limited by fewer events and lack of power.277 In the Action for Health in Diabetes trial (n=5145) that enrolled individuals with type 2 diabetes and overweight or obesity, an intensive lifestyle intervention targeting improved physical activity and weight loss did not lead to significant reductions in risk of HF despite relatively longer follow-up and a higher number of HF hospitalization events than prior studies.278 However, in exploratory analyses, greater improvement in cardiorespiratory fitness on follow-up was associated with a lower risk of HF, particularly HFpEF.47 Together, while RCT data for the prevention of HF with exercise and physical activity intervention are lacking owing to the lack of adequately powered RCTs with long-term follow-up, the convergence of epidemiological, mechanistic, and guideline-based evidence provides a compelling rationale for exercise and physical activity intervention as a key strategy in the primary prevention of HF.
充分支持并执行 RCT,以评估结构化运动训练和体力活动干预对原本健康的社区居住成年人发生心力衰竭风险的影响,仍然具有挑战性。因此,很少有单独的试验能够成功评估运动训练对心力衰竭发生率的影响,作为次要或探索性结局。 Generation-100 RCT 274 仍然是针对健康参与者(n=1567)进行的最大型、历时最长的运动训练研究之一,该研究在特定人群中评估了 5 年的运动训练效果(中等强度持续运动训练 [MICT] 与高强度间歇训练 [HIIT] 与活动推荐):挪威健康且身体健康的年长患者(年龄 70-77 岁)。 在本研究中,运动训练改善了心血管危险因素,但没有积累足够的心力衰竭(HF)事件来评估其对心力衰竭的影响。 275 在 LIFE 研究(n=1635) 276 中,有组织的体育活动干预减少了主要的行动障碍,但没有显示出对 HF 风险的显著影响,同样也受到事件较少和缺乏统计功效的限制。 277 在糖尿病健康行动试验(Action for Health in Diabetes trial, n=5145)中,该试验纳入了 2 型糖尿病和超重或肥胖的个体,一项旨在改善身体活动和减轻体重的强化生活方式干预措施,尽管随访时间相对较长,且心力衰竭(HF)住院事件的数量多于之前的研究,但并未显著降低 HF 风险。 278 然而,在探索性分析中,随访时心肺适能的更大改善与较低的 HF 风险相关,尤其是 HFpEF。 47 总而言之,虽然由于缺乏具有长期随访的、充分赋能的 RCT,因此缺乏通过运动和体育活动干预预防 HF 的 RCT 数据,但流行病学、机制和基于指南的证据的融合为将运动和体育活动干预作为 HF 一级预防的关键策略提供了令人信服的理由。

Exercise Training for Management of Individuals With Established HF
针对已确诊 HF 患者的运动训练管理

Several studies have examined the utility of exercise training in patients with prevalent HFrEF. Specifically, Heart Failure: A Controlled Trial Investigating Outcomes of Exercise Training (HF-ACTION) was one of the seminal exercise training trials with a sufficient sample size (n=2331) to evaluate mortality and hospitalization outcomes in patients with chronic stable HFrEF.220 Although the protocol-specified primary analysis demonstrated no reduction in hospitalization/mortality with the exercise training intervention, the prespecified analysis adjusting for risk factors found a significant reduction in all-cause mortality or hospitalization (Hazard Ratio, 0.89 [95% CI, 0.81–0.99]; P=0.03).279 Similarly, a small (n=123) study subsequently evaluated supervised exercise training for 10 years (2x/wk) and found a significant reduction in hospitalization and cardiac mortality.280 Comprehensive study-level pooled analyses have demonstrated up to 30% reduction in the risk of all-cause hospitalization with exercise-based cardiac rehabilitation (versus control) in adults with HF with no difference in the risk of mortality.281 Among HF subtypes, multiple meta-analyses of RCTs of exercise training in participants with HFpEF have demonstrated consistent improvement in exercise capacity and quality of life with exercise training.233,268 Furthermore, there have been signals that patients with HFpEF may benefit more in exercise capacity gains with exercise training than patients with HFrEF. In the HF-ACTION trial, participants gained only 0.4 to 0.6 mL/kg per min in VO2peak although this may have been impacted by intervention adherence.220 Multiple trials in HFpEF, on the other hand, have demonstrated a median effect of ≈2 mL/kg per min in VO2peak.233 Furthermore, the only direct, head-to-head comparison of exercise training across HF subtypes found significantly greater improvement in VO2peak in patients with HFpEF compared with those with HFrEF.282 More recently, in the Rehabilitation Therapy in Older Acute Heart Failure Patients (REHAB-HF) trial, a multidomain rehabilitation intervention was associated with greater improvement in physical function, quality of life, and exercise capacity among participants hospitalized with acute decompensated HFpEF versus HFrEF.283 For clinical outcomes, study-level pooled analysis has demonstrated greater risk reduction in risk of hospitalization with exercise training (versus control) in patients with HFpEF versus HFrEF (HFpEF: odds ratio, 0.43 [95% CI, 0.30–0.62]; P<0.001 versus HFrEF: odds ratio, 0.71 [95% CI, 0.54–0.94]; P=0.02).284 The ongoing Physical Rehabilitation for Older Patients with Acute HFpEF (Unique identifier: NCT05525663) trial is an adequately powered RCT that is evaluating the effects of a multidomain rehabilitation intervention on clinical outcomes of mortality of HF hospitalization in older adults with acute decompensated HFpEF.
多项研究检验了运动训练在患有 HFrEF 患者中的效用。具体来说,Heart Failure: A Controlled Trial Investigating Outcomes of Exercise Training (HF-ACTION) 是一项具有足够样本量 (n=2331) 的重要运动训练试验,旨在评估慢性稳定 HFrEF 患者的死亡率和住院结局。 220 尽管方案规定的主要分析表明,运动训练干预并未降低住院/死亡率,但对风险因素进行调整的预先指定分析发现,全因死亡率或住院率显著降低(风险比,0.89 [95% CI,0.81–0.99];P=0.03)。 279 同样,一项小型研究(n=123)随后评估了为期 10 年(2 次/周)的监督运动训练,发现住院率和心脏死亡率显著降低。 280 综合性研究层面的汇总分析表明,在心力衰竭成人患者中,以运动为基础的心脏康复(相对于对照组)可降低高达 30%的全因住院风险,而死亡风险无差异。 281 在心力衰竭亚型中,针对射血分数保留型心力衰竭(HFpEF)患者进行运动训练的多个 RCTs 的荟萃分析表明,运动训练可始终如一地提高运动能力和生活质量。 233,268 此外,有迹象表明,与 HFrEF 患者相比,HFpEF 患者可能从运动训练中获得更大的运动能力提升。在 HF-ACTION 试验中,参与者的 VO2峰值仅增加了 0.4 至 0.6 mL/kg/min 2peak ,但这可能受到了干预依从性的影响。 220 另一方面,多项 HFpEF 试验表明,VO2峰值的中位数效应约为 2 mL/kg/min 2peak 233 此外,唯一一项针对不同 HF 亚型进行运动训练的直接、头对头比较研究发现,HFpEF 患者的 VO 2peak 改善程度明显高于 HFrEF 患者。 282 最近,在老年急性心力衰竭患者康复治疗(REHAB-HF)试验中,多领域康复干预与急性失代偿性射血分数保留型心力衰竭(HFpEF)患者相比,射血分数降低型心力衰竭(HFrEF)住院患者在身体功能、生活质量和运动能力方面的改善更大。 283 对于临床结局,研究层面的汇总分析表明,在 HFpEF 患者中,运动训练(相对于对照组)在降低住院风险方面比 HFrEF 患者效果更佳(HFpEF:优势比,0.43 [95% CI,0.30–0.62];P<0.001,而 HFrEF:优势比,0.71 [95% CI,0.54–0.94];P=0.02)。 284 正在进行的针对急性 HFpEF 老年患者的身体康复(Unique identifier:NCT05525663)试验是一项充分赋能的 RCT,正在评估多领域康复干预对急性失代偿性 HFpEF 老年患者死亡率或 HF 住院等临床结局的影响。

Treatment Benefits Across Exercise Regimens of Different Intensities, Durations, and Modalities
不同强度、持续时间和方式的运动方案对治疗的益处

The design of early exercise training and cardiac rehabilitation programs primarily relied on aerobic, MICT regimens; these included a graduated increase in effort/exercise time over the training period and were typically enacted through walking or biking exertion. In the past decade, alternative and adjunct modalities, including HIIT and resistance training, have been studied in primary and secondary prevention populations. In general, results have either been neutral between MICT and HIIT221,285,286 or slightly favored HIIT in a subset of studies in healthy or overweight/obese populations.216,275,276 Several meta-analyses have suggested that HIIT may be superior to MICT in HFpEF,233,287,288 but these should be interpreted with extreme caution, given that (1) the largest direct comparison study (OptimEx-Clin; n=180) was neutral; 2) other contributing studies were small (n=24 and n=15)289,290; (3) 1 of the 3 included studies demonstrated no improvement in VO2peak with MICT versus control,290 in contrast to many other studies that have found significant improvement with MICT in HFpEF; and (4) several of these meta-analyses233,288 utilized a secondary outcome point estimate from OptimEx-Clin (12-month change in VO2peak, trending to favor HIIT) compared with the primary outcome of 3-month change in VO2peak (which trended to favor MICT without statistical significance), which would have been more consistent for comparison to the other included studies. It should be noted that the evidence base for MICT is much larger (ie, the number of trials and total participants studied) and may be implementable in a broader range of patients (ie, frail and musculoskeletal limitations). However, this remains an area of active research. From existing data, it appears that the clinical focus should be on implementing proven exercise training regimens (MICT or HIIT) based on patients’ preferences and logistical feasibility and increasing the frequency/duration of training, as tolerated.240,286 Furthermore, resistance training can improve body composition/lean mass, strength, and cardiometabolic metrics and, therefore, has increasingly become of interest for exercise training regimens. In aggregate, resistance training in isolation has demonstrated less reliable cardiometabolic results,234,239,242,245 but it improves VO2peak216,247,291 and has been shown to have supportive/additive effects alongside aerobic training for leg strength and muscle quality.227
早期运动训练和心脏康复计划的设计主要依赖于有氧、中等强度持续训练(MICT)方案;这些方案包括在训练期间逐步增加运动强度/运动时间,并且通常通过步行或自行车运动来实现。在过去的十年中,包括高强度间歇训练(HIIT)和阻力训练在内的替代和辅助方式已在 primary 和 secondary 预防人群中进行了研究。 一般来说,在健康或超重/肥胖人群的部分研究中,MICT 和 HIIT 的结果要么是中性的 221,285,286 ,要么略微偏向 HIIT。 216,275,276 几项荟萃分析表明,在高射血分数心力衰竭 (HFpEF) 患者中,高强度间歇训练 (HIIT) 可能优于中等强度持续训练 (MICT), 233,287,288 但鉴于以下原因,应极其谨慎地解读这些分析:(1) 最大的直接比较研究(OptimEx-Clin;n=180)结果为中性;2) 其他有贡献的研究规模较小(n=24 和 n=15) 289,290 ;(3) 在纳入的 3 项研究中,有 1 项研究显示,与对照组相比,MICT 对 VO 2peak 没有改善, 290 这与其他许多研究发现 MICT 可显著改善 HFpEF 的结果相反;(4) 这些荟萃分析中,有几项 233,288 使用了 OptimEx-Clin 的次要终点估计值(12 个月 VO 2peak 变化,趋势偏向 HIIT),而不是 3 个月 VO 2peak 变化的主要终点(趋势偏向 MICT,但无统计学意义),而与纳入的其他研究相比,主要终点更具一致性。 应该指出的是,关于中等强度持续训练(MICT)的证据基础要大得多(即,试验次数和研究的总参与者人数),并且可能适用于更广泛的患者(即,体弱者和肌肉骨骼受限者)。然而,这仍然是一个活跃的研究领域。 根据现有数据,临床重点似乎应该放在根据患者的偏好和后勤可行性实施已证实有效的运动训练方案(MICT 或 HIIT),并在可耐受的情况下增加训练的频率/持续时间。 240,286 此外,阻力训练可以改善身体成分/瘦体重、力量和心脏代谢指标,因此,越来越受到运动训练方案的关注。 总的来说,孤立的阻力训练显示出不太可靠的心脏代谢结果, 234,239,242,245 但它改善了 VO 2peak 216,247,291 ,并且已被证明在腿部力量和肌肉质量方面与有氧训练一起具有支持/累加效果。 227

Durable and Legacy Effects of Exercise Training
运动训练的持久和长期影响

In contrast to data for immediate response outcomes (assessed at intervention completion), the evidence base for the intermediate and long-term functional effects of exercise training is limited. Definitions for the timeline of longer-term effects are not standardized, but durable effects may be generally defined as between 6 and 24 months after the completion of the intervention. In contrast, legacy effects are typically described as years after completion of the intervention, long after cessation of the treatment.292–294 In the few existing studies, legacy effects have often been studied ≈10 years after the intervention293–295 but may be reasonably defined as early as 2 years after intervention completion and up to 20 years.296,297
与即时反应结果(在干预结束时评估)的数据相比,运动训练对中期和长期功能影响的证据基础有限。长期影响的时间线定义尚未标准化,但持久影响通常可定义为干预完成后 6 至 24 个月之间。 相比之下,后效通常被描述为干预完成后数年,即治疗停止很久之后出现的效应。 292–294 在为数不多的现有研究中,后效通常在干预后约 10 年进行研究 293–295 ,但可以合理地定义为干预完成后最早 2 年到最长 20 年的时间范围内。 296,297
Regarding the durable effects of exercise training, several studies have evaluated effects in the months to 1 year after the initial, supervised regimen was completed. Often, a home prescription or recommendation for continued exercise training, with variable adherence monitoring, is utilized between immediate evaluation (ie, 3–6 months) and durable evaluation (ie, 6–24 months after completion of supervised regimen). The largest studies to evaluate the question of durability include HF-ACTION (HFrEF)220 and OptimEx-Clin (HFpEF).285 In addition to the primary mortality/hospitalization outcomes, HF-ACTION demonstrated some degree of maintenance of benefits for exercise capacity (VO2peak and exercise time, not six minute walk distance, which attenuated to nonsignificance), depression, and quality-of-life metrics at 12 months220,298,299 and up to 26 months for quality of life.299 In contrast, the OptimEx-Clin evaluation at 12 months suggested that exercise benefits (primary outcome and VO2peak) regressed markedly in the months following exercise training. However, the intensity of monitoring and guidance in the 3- to 12-month home phase may have differed between the 2 programs.
关于运动训练的持久效果,有多项研究评估了在最初的监督方案完成后几个月到 1 年内的效果。通常,在即时评估(即 3-6 个月)和持久评估(即完成监督方案后 6-24 个月)之间,会采用家庭处方或持续运动训练的建议,并进行不同程度的依从性监测。 关于持久性的最大型研究包括 HF-ACTION(HFrEF) 220 和 OptimEx-Clin(HFpEF)。 285 除了主要死亡率/住院结局外,HF-ACTION 还证明了运动能力(VO 2peak 和运动时间,而非六分钟步行距离,后者减弱至无统计学意义)、抑郁症和生活质量指标在 12 个月时 220,298,299 以及长达 26 个月的生活质量方面,均在一定程度上保持了益处。 299 相比之下,OptimEx-Clin 研究在 12 个月时的评估表明,运动带来的益处(主要结局和 VO 2peak )在运动训练后的几个月里明显消退。然而,在这两个项目中,3 至 12 个月家庭阶段的监测和指导强度可能有所不同。
Data regarding the legacy effects of exercise training are even more limited. The STRRIDE program remains one of the few trials to re-evaluate participants in the legacy phase, through a 10-year Reunion study.293 Participants from the exercise training groups were found to have an attenuated decline in VO2peak and smaller increases in waist circumference. Furthermore, legacy exercise training participants were found to have improved fasting insulin and mean arterial blood pressure compared with controls. These encouraging findings have limitations, including a tightly defined trial cohort, the opportunity at randomization for control arm participants to pursue the exercise training arm (including potential bias), and the lack of inclusion of dropout participants in the Reunion study. Future investigation of both the durable and legacy effects of exercise training across population cohorts is needed to define current results and to support the development of strategies to improve long-term effects.
关于运动训练的遗留效应的数据甚至更加有限。STRRIDE 计划仍然是为数不多的在遗留阶段重新评估参与者的试验之一,通过一项为期 10 年的重聚研究。 293 结果发现,运动训练组的参与者 VO 2peak 的下降幅度有所减缓,腰围的增加幅度也有所减小。 此外,与对照组相比,既往运动训练参与者的空腹胰岛素和平均动脉血压均有所改善。这些令人鼓舞的发现也存在局限性,包括定义严格的试验队列、随机分组时对照组参与者有机会参加运动训练组(包括潜在的偏倚),以及 Reunion 研究中未纳入退出研究的参与者。 未来需要对不同人群队列中运动训练的持久效应和长期效应进行进一步研究,以明确目前的结果,并支持制定改善长期效果的策略。
Figure 5. Current challenges and potential solutions to the implementation of exercise training interventions. Despite the notable benefits of exercise training, implementation remains challenging with notable gaps that result in reduced access and utilization among vulnerable patients who have the greatest need and may benefit the most from exercise training interventions and cardiac rehabilitation. Innovative approaches that implement exercise training at home, tailoring training to specific deficits, and early implementation for vulnerable populations along with expanded coverage to improve access may enhance the effectiveness of the intervention and address the implementation gap. HFpEF indicates heart failure with preserved ejection fraction. Illustration credit: Sceyence Studios.
图 5. 实施运动训练干预措施的当前挑战和潜在解决方案。尽管运动训练具有显著益处,但实施仍然具有挑战性,存在显著差距,导致弱势患者获得和利用的机会减少,而这些患者最需要运动训练干预措施和心脏康复,并且可能从中获益最多。 实施居家运动训练、针对特定缺陷定制训练、及早对弱势群体实施,以及扩大覆盖面以提高可及性的创新方法,可能会增强干预的有效性,并解决实施差距。HFpEF 表示射血分数保留型心力衰竭。图示版权:Sceyence Studios。

Current State and Future of Exercise Interventions in HF
心力衰竭运动干预的现状与未来

Despite the growing evidence supporting exercise as a therapeutic strategy to reduce the burden and severity of aging-related diseases, including CVD, implementing exercise training interventions in the real world remains challenging (Figure 5). Cardiac rehabilitation, which encompasses a holistic approach to lifestyle intervention incorporating nutritional counseling, risk factor management, and psychosocial support with exercise training, improves the quality of life and functional status across a broad range of cardiovascular conditions, including HF. However, only <5% of patients with HF attend center-based cardiac rehabilitation sessions.300 Moreover, insurance reimbursement for cardiac rehabilitation is unavailable for patients with HFpEF because these patients were not included in the large outcome studies. Furthermore, while the benefits of exercise training appear to be most evident in patients with a higher burden of frailty and physical dysfunction, older, frail patients with multiple comorbidities are not commonly referred to cardiac rehabiliation (CR).301–303 Home-based rehabilitation programs have emerged as a potential solution to improve access to CR for vulnerable patients who may not be able to attend center-based programs.304–307 These programs have demonstrated good adherence and have been shown to improve the quality of life in patients with HFpEF.304
尽管越来越多的证据表明运动是一种治疗策略,可以减轻与衰老相关的疾病(包括 CVD)的负担和严重程度,但在现实世界中实施运动训练干预仍然具有挑战性(图 5)。 心脏康复包括一种针对生活方式干预的整体方法,其中结合了营养咨询、危险因素管理以及伴有运动训练的心理社会支持,能够改善包括心力衰竭(HF)在内的多种心血管疾病患者的生活质量和功能状态。然而,仅有不到 5%的心力衰竭患者参加以中心为基础的心脏康复课程。 300 此外,由于 HFpEF 患者未纳入大型结局研究,因此他们无法获得心脏康复的保险报销。再者,虽然运动训练的益处在虚弱和身体功能障碍负担较重的患者中最为明显,但患有多种合并症的老年虚弱患者通常不会被转诊至心脏康复(CR)。 301–303 以家庭为基础的康复计划已成为一种潜在的解决方案,可以改善弱势患者获得心脏康复(CR)的机会,这些患者可能无法参加以中心为基础的计划。 304–307 这些计划已显示出良好的依从性,并已被证明可以提高射血分数保留型心力衰竭(HFpEF)患者的生活质量。 304
Patients hospitalized with HF are more prone to develop worsening sarcopenia and frailty as a result of prolonged bed rest, increased catabolic state, and acute HF.4 However, these patients are not eligible for CR until 4 weeks after hospital discharge. These observations highlight the mismatch between those individuals at the highest risk and those most likely to benefit from CR and access and utilization of CR. As a result, novel approaches to implementing exercise training and CR are warranted. Older adults with HF are more likely to have HFpEF, a higher burden of frailty, and physical dysfunction. Rehabilitation programs that are aimed at restoring physical function with a progressive focus on balance, strength, and endurance may directly address the underlying physical deficits of these patients and may be a more prudent strategy than aerobic endurance training. As discussed above, the REHAB-HF trial demonstrated the feasibility and efficacy of such a program in improving physical function and quality of life among older adults with recent HF hospitalization.308 Moreover, the REHAB-HF trial implemented an early, tailored progressive intervention initiated in the hospital and continued in the outpatient setting. Such a program proactively addresses the deficits in physical function that accrue due to HF hospitalization, particularly in older adults.
因长期卧床、分解代谢状态增加和急性心力衰竭,住院的 HF 患者更容易发生恶化的肌肉减少症和体弱。 4 然而,这些患者在出院后 4 周才有资格参加 CR。这些观察结果突显了最高风险人群与最有可能从 CR 中获益、获得和利用 CR 的人群之间的不匹配。 因此,有必要采取新的方法来实施运动训练和 CR。患有 HF 的老年人更有可能患有 HFpEF,更高的虚弱负担和身体功能障碍。旨在通过逐步关注平衡、力量和耐力来恢复身体功能的康复计划可以直接解决这些患者的潜在身体缺陷,并且可能比有氧耐力训练更谨慎。 如上所述,REHAB-HF 试验表明了该项目在改善近期因心衰住院的老年人的身体机能和生活质量方面的可行性和有效性。 308 此外,REHAB-HF 试验实施了一项早期、定制的渐进式干预,该干预在医院启动并在门诊环境中继续进行。该项目主动解决因心衰住院导致的身体机能缺陷,尤其是在老年人中。
Physical activity and exercise training are important tools for clinicians and public health officials to mitigate the deleterious effects of accelerated aging and forestall the onset of chronic disease, multimorbidity, and frailty. The evidence supporting the benefits of higher physical activity levels is incontrovertible. Moreover, dedicated exercise training interventions are effective across a broad range of geriatric and cardiovascular conditions in improving quality of life, physical function, and exercise capacity.309–312 Interestingly, these interventions appear to be most effective for participants with a higher burden of frailty and multimorbidity, that is, accelerated aging phenotype.302,303 This observation is consistent with the pleiotropic mechanisms of exercise training, which are well suited to address the multisystem changes that occur due to aging and are more pronounced in adults with multimorbidity and frailty. However, implementing exercise training for patients with frailty and multimorbidity has been challenging.300,301 Recognizing its importance, novel approaches using multidomain interventions and home-based physical rehabilitation programs are being developed and tested for older patients with a high burden of frailty.
体力活动和运动训练是临床医生和公共卫生官员的重要工具,可以减轻加速衰老带来的有害影响,并延缓慢性病、多病共病和虚弱的发生。支持较高体力活动水平益处的证据是无可辩驳的。 此外,专门的运动训练干预措施在改善老年人和心血管疾病患者的生活质量、身体机能和运动能力方面非常有效。 309–312 有趣的是,这些干预措施对于那些体弱和多病负担较重的参与者,即加速衰老表型,似乎最为有效。 302,303 这一观察结果与运动训练的多效性机制相符,该机制非常适合解决因衰老而发生的多系统变化,并且在患有多种疾病和体弱的成年人中更为明显。然而,对体弱和患有多种疾病的患者实施运动训练一直具有挑战性。 300,301 认识到其重要性,目前正在开发和测试使用多领域干预和居家物理康复计划的新方法,用于治疗高衰弱负担的老年患者。

Footnote  脚注

Nonstandard Abbreviations and Acronyms
非标准缩写和首字母缩略词

A-VO2diff
arteriovenous oxygen difference
动静脉氧含量差
CARDIA
Coronary Artery Disease in Young Adults
青年人群冠状动脉疾病
CVD
cardiovascular disease  心血管疾病
ET-1
endothelin-1  内皮素-1
HF  心衰
heart failure  心力衰竭
HFpEF  射血分数保留型心力衰竭
heart failure with preserved ejection fraction
射血分数保留型心力衰竭
HFrEF  HFrEF(射血分数降低型心力衰竭)
heart failure with reduced ejection fraction
射血分数降低型心力衰竭
HIIT  高强度间歇训练
high-intensity interval training
高强度间歇训练
LA  左心房
left atrium  左心房
LV  左室
left ventricle  左心室
MICT
moderate-intensity, continuous exercise training
中等强度持续运动训练
NT-proBNP
N-terminal pro-B-type natriuretic peptide
N 端脑钠肽前体
RCT
randomized controlled trial
随机对照试验
VO2peak
peak exercise oxygen consumption
峰值运动氧耗量

References  参考文献

1.
Zoe Caplan MR. The Older Population: 2020. Bureau TUSC; 2023.
Zoe Caplan MR. 2020 年老年人口统计. 美国人口普查局; 2023.
2.
Domanski MJ, Tian X, Wu CO, Reis JP, Dey AK, Gu Y, Zhao L, Bae S, Liu K, Hasan AA, et al. Time course of LDL cholesterol exposure and cardiovascular disease event risk. J Am Coll Cardiol. 2020;76:1507–1516. doi: 10.1016/j.jacc.2020.07.059
Domanski MJ, Tian X, Wu CO, Reis JP, Dey AK, Gu Y, Zhao L, Bae S, Liu K, Hasan AA, et al. LDL 胆固醇暴露的时间过程与心血管疾病事件风险。《美国心脏病学会杂志》。2020;76:1507–1516. doi: 10.1016/j.jacc.2020.07.059
3.
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Barone Gibbs B, Beaton AZ, Boehme AK, et al; American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee. 2024 Heart disease and stroke statistics: a report of US and global data from the American Heart Association. Circulation. 2024;149:e347–e913. doi: 10.1161/CIR.0000000000001209
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Barone Gibbs B, Beaton AZ, Boehme AK, et al; American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee. 2024 年心脏病和中风统计数据:美国心脏协会关于美国和全球数据的报告。Circulation. 2024;149:e347–e913. doi: 10.1161/CIR.0000000000001209
4.
Pandey A, Kitzman D, Reeves G. Frailty is intertwined with heart failure: mechanisms, prevalence, prognosis, assessment, and management. JACC Heart Fail. 2019;7:1001–1011. doi: 10.1016/j.jchf.2019.10.005
Pandey A, Kitzman D, Reeves G. 衰弱与心力衰竭密切相关:机制、患病率、预后、评估和管理。JACC Heart Fail. 2019;7:1001–1011. doi: 10.1016/j.jchf.2019.10.005

eLetters  电子来信

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.
eLetter 应与近期发表在该期刊上的一篇文章相关,而不是提供未发表数据的论坛。评论会审查语气和语言的适当性。评论不经过同行评审。可接受的评论仅发布在期刊网站上。评论不会在期刊上发表,也不会在 PubMed 中收录。评论不应超过 500 字,并且仅在线发布。参考文献限制为 10 个。 我们将邀请评论中引用的文章的作者酌情回复。

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.
有关 AHA/ASA 科学声明和指南的评论和反馈应通过其通信页面发送给 AHA/ASA 稿件监督委员会。

Information & Authors

Information

Published In

Go to Circulation Research
Circulation Research
Pages: 205 - 230
PubMed: 40608862

History

Received: 20 February 2025
Revision received: 19 May 2025
Accepted: 3 June 2025
Published online: 3 July 2025
Published in print: 7 July 2025

Permissions

Request permissions for this article.

Keywords

  1. aging
  2. cardiovascular diseases
  3. exercise
  4. frailty
  5. heart failure

Subjects

Authors

Affiliations

Notes

For Sources of Funding and Disclosures, see page 221.
Correspondence to: Dalane W. Kitzman, MD, Department of Cardiovascular Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, Email dkitzman@wakehealth.edu
Ambarish Pandey, MD, MSCS, Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, Email ambarish.pandey@utsouthwestern.edu

Disclosures

D.W. Kitzman has been a consultant for AstraZeneca, Pfizer, Corvia Medical, Bayer, Boehringer Ingelheim, Novo Nordisk, Rivus, and St. Luke’s Medical Center; received grant support from Novartis, AstraZeneca, Bayer, Pfizer, Novo Nordisk, Rivus, and St. Luke’s Medical Center; and owns stocks in Gilead Sciences. A. Pandey received research support from the National Institutes of Health, the American Heart Association, Applied Therapeutics, Roche, Ultromics, Gilead Sciences, and AstraZeneca; honoraria outside of this study as an advisor/consultant for Tricog Health, Inc, Lilly USA, Rivus, Cytokinetics, Roche Diagnostics, Axon therapies, Medtronic, Edward Lifesciences, Science37, Novo Nordisk, Bayer, Merck, Sarfez Pharmaceuticals, Emmi Solutions, Semler Scientific, Ultromics, Merck, kardigan, Kieele Health, Anumana, and Acorai; and nonfinancial support from Pfizer and Merck. The other authors report no conflicts.

Sources of Funding

Dr. Kitzman is supported by NIH grants U01AG076928, R01AG078153, R01AG045551, P30AG021332, U24AG059624, and U01HL160272 and the Kermit Glenn Phillips II Chair in Cardiovascular Medicine.

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB

Figures

Figure 1. Accelerated aging phenotype and the pathophysiology of heart failure with preserved ejection fraction (HFpEF). The cumulative effects of multimorbidity, obesity, aging, physical inactivity, and systemic inflammation result in an accelerated aging phenotype characterized by sarcopenia, mitochondrial and endothelial dysfunction, and capillary loss, which contribute to diminished physiological reserve (frailty) that leads to multiorgan dysfunction and myopathy of the cardiac and skeletal muscles. These changes have been implicated in the pathophysiology of HFpEF, which is now considered a geriatric syndrome that is characterized by severe impairment in quality of life (QOL), physical function, and exercise capacity and is associated with disability and mortality. Adapted from Pandey et al9 with permission. Illustration credit: Sceyence Studios.
Figure 2. Multisystem changes related to the aging process. Age-related changes occur in nearly every organ system, and the structural and functional changes of cellular, cardiovascular, skeletomuscular, and adipose tissue result in diminished physiological capacity. LA indicates left atrium; and LV, left ventricle. Illustration credit: Sceyence Studios.
Figure 3. Mechanisms of age-related decline in exercise capacity. Oxygen delivery can be described by the Fick principle as the product of cardiac output and peripheral oxygen extraction. Aging results in a reduction in peak heart rate, and stroke volume results in reduced exercise cardiac output. Peripheral oxygen extraction at exercise declines or remains the same and is related to reduced capillary density and increased afterload. Together, these mechanisms result in a reduction in aerobic capacity with aging. PCWP indicates pulmonary capillary wedge pressure. Illustration credit: Sceyence Studios.
Figure 4. Multisystem mechanisms of benefits of exercise training. Exercise training results in beneficial adaptations in multiple tissues, which attenuate the changes associated with aging. Exercise training results in increased cardiac compliance, improvement in stroke volume, and cardiac output. At the level of the peripheral vasculature, improvement in peak exercise oxygen extraction occurs as a result of improved endothelial function and muscle blood flow. The skeletal muscle size increases with a resultant increase in contractile function. Reduction in visceral and ectopic adiposity contributes to improved insulin sensitivity and reduction in inflammation. At the subcellular level, mitochondrial content and energetics improve with exercise. Together, these adaptations result in improved aerobic capacity, increased strength that results in better physical function, reduced burden of frailty, and greater independence. Illustration credit: Sceyence Studios.
Figure 5. Current challenges and potential solutions to the implementation of exercise training interventions. Despite the notable benefits of exercise training, implementation remains challenging with notable gaps that result in reduced access and utilization among vulnerable patients who have the greatest need and may benefit the most from exercise training interventions and cardiac rehabilitation. Innovative approaches that implement exercise training at home, tailoring training to specific deficits, and early implementation for vulnerable populations along with expanded coverage to improve access may enhance the effectiveness of the intervention and address the implementation gap. HFpEF indicates heart failure with preserved ejection fraction. Illustration credit: Sceyence Studios.

Tables

Table 1. Hallmarks of Aging, Associated Phenotypic Manifestations and Cardiovascular Diseases, and Potential Treatment Strategies Targeting the Specific Mechanisms
Table 2. Cardiovascular Phenotypes Associated With Normal and Accelerated Biological Aging
Table 3. Summary Findings From Key Studies Evaluating the Association Between Objectively Measured Physical Activity Levels With Adverse Cardiovascular Outcomes

Media

Share

Share

Share article link

Share

References

References

1.
Zoe Caplan MR. The Older Population: 2020. Bureau TUSC; 2023.
2.
Domanski MJ, Tian X, Wu CO, Reis JP, Dey AK, Gu Y, Zhao L, Bae S, Liu K, Hasan AA, et al. Time course of LDL cholesterol exposure and cardiovascular disease event risk. J Am Coll Cardiol. 2020;76:1507–1516. doi: 10.1016/j.jacc.2020.07.059
3.
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Barone Gibbs B, Beaton AZ, Boehme AK, et al; American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee. 2024 Heart disease and stroke statistics: a report of US and global data from the American Heart Association. Circulation. 2024;149:e347–e913. doi: 10.1161/CIR.0000000000001209
4.
Pandey A, Kitzman D, Reeves G. Frailty is intertwined with heart failure: mechanisms, prevalence, prognosis, assessment, and management. JACC Heart Fail. 2019;7:1001–1011. doi: 10.1016/j.jchf.2019.10.005
5.
Bell SP, Orr NM, Dodson JA, Rich MW, Wenger NK, Blum K, Harold JG, Tinetti ME, Maurer MS, Forman DE. What to expect from the evolving field of geriatric cardiology. J Am Coll Cardiol. 2015;66:1286–1299. doi: 10.1016/j.jacc.2015.07.048
6.
Kuo PL, Schrack JA, Shardell MD, Levine M, Moore AZ, An Y, Elango P, Karikkineth A, Tanaka T, de Cabo R, et al. A roadmap to build a phenotypic metric of ageing: insights from the Baltimore Longitudinal Study of Aging. J Intern Med. 2020;287:373–394. doi: 10.1111/joim.13024
7.
Ferrucci L, Levine ME, Kuo PL, Simonsick EM. Time and the metrics of aging. Circ Res. 2018;123:740–744. doi: 10.1161/CIRCRESAHA.118.312816
8.
Hu C, Zhang X, Teng T, Ma ZG, Tang QZ. Cellular senescence in cardiovascular diseases: a systematic review. Aging Dis. 2022;13:103–128. doi: 10.14336/AD.2021.0927
9.
Pandey A, Shah SJ, Butler J, Kellogg DL, Lewis GD, Forman DE, Mentz RJ, Borlaug BA, Simon MA, Chirinos JA, et al. Exercise intolerance in older adults with heart failure with preserved ejection fraction: JACC state-of-the-art review. J Am Coll Cardiol. 2021;78:1166–1187. doi: 10.1016/j.jacc.2021.07.014
10.
Gander JC, Sui X, Hebert JR, Hazlett LJ, Cai B, Lavie CJ, Blair SN. Association of cardiorespiratory fitness with coronary heart disease in asymptomatic men. Mayo Clin Proc. 2015;90:1372–1379. doi: 10.1016/j.mayocp.2015.07.017
11.
Wei M, Kampert JB, Barlow CE, Nichaman MZ, Gibbons LW, Paffenbarger RS, Blair SN. Relationship between low cardiorespiratory fitness and mortality in normal-weight, overweight, and obese men. JAMA. 1999;282:1547–1553. doi: 10.1001/jama.282.16.1547
12.
Earnest CP, Artero EG, Sui X, Lee DC, Church TS, Blair SN. Maximal estimated cardiorespiratory fitness, cardiometabolic risk factors, and metabolic syndrome in the aerobics center longitudinal study. Mayo Clin Proc. 2013;88:259–270. doi: 10.1016/j.mayocp.2012.11.006
13.
Blair SN, Jackson AS. Physical fitness and activity as separate heart disease risk factors: a meta-analysis. Med Sci Sports Exerc. 2001;33:762–764. doi: 10.1097/00005768-200105000-00013
14.
Lakatta EG. So! What’s aging? Is cardiovascular aging a disease? J Mol Cell Cardiol. 2015;83:1–13. doi: 10.1016/j.yjmcc.2015.04.005
15.
Paneni F, Diaz Canestro C, Libby P, Luscher TF, Camici GG. The aging cardiovascular system: understanding it at the cellular and clinical levels. J Am Coll Cardiol. 2017;69:1952–1967. doi: 10.1016/j.jacc.2017.01.064
16.
Capell BC, Collins FS, Nabel EG. Mechanisms of cardiovascular disease in accelerated aging syndromes. Circ Res. 2007;101:13–26. doi: 10.1161/CIRCRESAHA.107.153692
17.
Yu CE, Oshima J, Fu YH, Wijsman EM, Hisama F, Alisch R, Matthews S, Nakura J, Miki T, Ouais S, et al. Positional cloning of the Werner’s syndrome gene. Science. 1996;272:258–262. doi: 10.1126/science.272.5259.258
18.
Kato H, Maezawa Y. Atherosclerosis and cardiovascular diseases in progeroid syndromes. J Atheroscler Thromb. 2022;29:439–447. doi: 10.5551/jat.RV17061
19.
Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023;186:243–278. doi: 10.1016/j.cell.2022.11.001
20.
Abdellatif M, Rainer PP, Sedej S, Kroemer G. Hallmarks of cardiovascular ageing. Nat Rev Cardiol. 2023;20:754–777. doi: 10.1038/s41569-023-00881-3
21.
Liu Z, Kuo PL, Horvath S, Crimmins E, Ferrucci L, Levine M. A new aging measure captures morbidity and mortality risk across diverse subpopulations from NHANES IV: a cohort study. PLoS Med. 2018;15:e1002718. doi: 10.1371/journal.pmed.1002718
22.
Klemera P, Doubal S. A new approach to the concept and computation of biological age. Mech Ageing Dev. 2006;127:240–248. doi: 10.1016/j.mad.2005.10.004
23.
Li X, Cao X, Zhang J, Fu J, Mohedaner M, Danzengzhuoga, Sun X, Yang G, Yang Z, Kuo CL, et al. Accelerated aging mediates the associations of unhealthy lifestyles with cardiovascular disease, cancer, and mortality. J Am Geriatr Soc. 2024;72:181–193. doi: 10.1111/jgs.18611
24.
Spikes TA, Alonso A, Thorpe RJ, Pelkmans J, Higgins M, Sullivan S, Dunbar SB, Michopoulos V, Searles C, Lewis TT, et al. Biological age as a predictor of arterial stiffness in young and early midlife black and white women. Am J Hypertens. 2025. doi: 10.1093/ajh/hpaf059
25.
Mao R, Wang F, Zhong Y, Meng X, Zhang T, Li J. Association of biological age acceleration with cardiac morphology, function, and incident heart failure: insights from UK Biobank participants. Eur Heart J Cardiovasc Imaging. 2024;25:1315–1323. doi: 10.1093/ehjci/jeae126
26.
Sanchez-Cabo F, Fuster V, Silla-Castro JC, Gonzalez G, Lorenzo-Vivas E, Alvarez R, Callejas S, Benguria A, Gil E, Nunez E, et al. Subclinical atherosclerosis and accelerated epigenetic age mediated by inflammation: a multi-omics study. Eur Heart J. 2023;44:2698–2709. doi: 10.1093/eurheartj/ehad361
27.
Kim K, Joyce BT, Zheng Y, Schreiner PJ, Jacobs DR, Catov JM, Shikany JM, Carnethon MR, Greenland P, Van Horn LV, et al. DNA methylation grimage and incident diabetes: the Coronary Artery Risk Development in Young Adults (CARDIA) study. Diabetes. 2021;70:1404–1413. doi: 10.2337/db20-1167
28.
Lu AT, Quach A, Wilson JG, Reiner AP, Aviv A, Raj K, Hou L, Baccarelli AA, Li Y, Stewart JD, et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging (Albany NY). 2019;11:303–327. doi: 10.18632/aging.101684
29.
Roetker NS, Pankow JS, Bressler J, Morrison AC, Boerwinkle E. Prospective study of epigenetic age acceleration and incidence of cardiovascular disease outcomes in the ARIC study (Atherosclerosis Risk in Communities). Circ Genom Precis Med. 2018;11:e001937. doi: 10.1161/CIRCGEN.117.001937
30.
Levine ME, Lu AT, Quach A, Chen BH, Assimes TL, Bandinelli S, Hou L, Baccarelli AA, Stewart JD, Li Y, et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging (Albany NY). 2018;10:573–591. doi: 10.18632/aging.101414
31.
Jia L, Zhang W, Chen X. Common methods of biological age estimation. Clin Interv Aging. 2017;12:759–772. doi: 10.2147/CIA.S134921
32.
Afilalo J, Alexander KP, Mack MJ, Maurer MS, Green P, Allen LA, Popma JJ, Ferrucci L, Forman DE. Frailty assessment in the cardiovascular care of older adults. J Am Coll Cardiol. 2014;63:747–762. doi: 10.1016/j.jacc.2013.09.070
33.
Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci. 2014;69:S4–S9. doi: 10.1093/gerona/glu057
34.
Forman DE, Kuchel GA, Newman JC, Kirkland JL, Volpi E, Taffet GE, Barzilai N, Pandey A, Kitzman DW, Libby P, et al. Impact of geroscience on therapeutic strategies for older adults with cardiovascular disease: JACC scientific statement. J Am Coll Cardiol. 2023;82:631–647. doi: 10.1016/j.jacc.2023.05.038
35.
Noren Hooten N, Pacheco NL, Smith JT, Evans MK. The accelerated aging phenotype: the role of race and social determinants of health on aging. Ageing Res Rev. 2022;73:101536. doi: 10.1016/j.arr.2021.101536
36.
Melzer D, Pilling LC, Ferrucci L. The genetics of human ageing. Nat Rev Genet. 2020;21:88–101. doi: 10.1038/s41576-019-0183-6
37.
Bandeen-Roche K, Seplaki CL, Huang J, Buta B, Kalyani RR, Varadhan R, Xue QL, Walston JD, Kasper JD. Frailty in older adults: a nationally representative profile in the United States. J Gerontol A Biol Sci Med Sci. 2015;70:1427–1434. doi: 10.1093/gerona/glv133
38.
Marinus N, Vigorito C, Giallauria F, Haenen L, Jansegers T, Dendale P, Feys P, Meesen R, Timmermans A, Spildooren J, et al. Frailty is highly prevalent in specific cardiovascular diseases and females, but significantly worsens prognosis in all affected patients: a systematic review. Ageing Res Rev. 2021;66:101233. doi: 10.1016/j.arr.2020.101233
39.
Pandey A, Khan MS, Garcia K, Simpson F, Bahnson J, Patel KV, Singh S, Vaduganathan M, Bertoni A, Kitzman D, et al. Association of baseline and longitudinal changes in frailty burden and risk of heart failure in type 2 diabetes-findings from the look AHEAD trial. J Gerontol A Biol Sci Med Sci. 2022;77:2489–2497. doi: 10.1093/gerona/glac094
40.
Nguyen DD, Arnold SV. Impact of frailty on disease-specific health status in cardiovascular disease. Heart. 2023;109:977–983. doi: 10.1136/heartjnl-2022-321631
41.
Wong CWY, Li PWC, Yu DSF, Ho BMH, Chan BS. Estimated prevalence of frailty and prefrailty in patients undergoing coronary artery or valvular surgeries/procedures: a systematic review and proportional meta-analysis. Ageing Res Rev. 2024;96:102266. doi: 10.1016/j.arr.2024.102266
42.
Proietti M, Romiti GF, Raparelli V, Diemberger I, Boriani G, Dalla Vecchia LA, Bellelli G, Marzetti E, Lip GY, Cesari M. Frailty prevalence and impact on outcomes in patients with atrial fibrillation: a systematic review and meta-analysis of 1,187,000 patients. Ageing Res Rev. 2022;79:101652. doi: 10.1016/j.arr.2022.101652
43.
Segar MW, Singh S, Goyal P, Hummel SL, Maurer MS, Forman DE, Butler J, Pandey A. Prefrailty, impairment in physical function, and risk of incident heart failure among older adults. J Am Geriatr Soc. 2021;69:2486–2497. doi: 10.1111/jgs.17218
44.
Newman AB, Gottdiener JS, McBurnie MA, Hirsch CH, Kop WJ, Tracy R, Walston JD, Fried LP, Cardiovascular Health Study Research G. Associations of subclinical cardiovascular disease with frailty. J Gerontol A Biol Sci Med Sci. 2001;56:M158–M166. doi: 10.1093/gerona/56.3.m158
45.
Ramonfaur D, Skali H, Claggett B, Windham BG, Palta P, Kitzman D, Ndumele C, Konety S, Shah AM. Bidirectional association between frailty and cardiac structure and function: the Atherosclerosis Risk in Communities Study. J Am Heart Assoc. 2023;12:e029458. doi: 10.1161/JAHA.122.029458
46.
Nadruz W, Kitzman D, Windham BG, Kucharska-Newton A, Butler K, Palta P, Griswold ME, Wagenknecht LE, Heiss G, Solomon SD, et al. Cardiovascular dysfunction and frailty among older adults in the community: the ARIC study. J Gerontol A Biol Sci Med Sci. 2017;72:958–964. doi: 10.1093/gerona/glw199
47.
Pandey A, Patel KV, Bahnson JL, Gaussoin SA, Martin CK, Balasubramanyam A, Johnson KC, McGuire DK, Bertoni AG, Kitzman D, et al; Look AHEAD Research Group. Association of intensive lifestyle intervention, fitness, and body mass index with risk of heart failure in overweight or obese adults with type 2 diabetes mellitus: an analysis from the look AHEAD trial. Circulation. 2020;141:1295–1306. doi: 10.1161/CIRCULATIONAHA.119.044865
48.
North BJ, Sinclair DA. The intersection between aging and cardiovascular disease. Circ Res. 2012;110:1097–1108. doi: 10.1161/CIRCRESAHA.111.246876
49.
Hastings MH, Zhou Q, Wu C, Shabani P, Huang S, Yu X, Singh AP, Guseh JS, Li H, Lerchenmuller C, et al. Cardiac aging: from hallmarks to therapeutic opportunities. Cardiovasc Res. 2024. doi: 10.1093/cvr/cvae124
50.
Tuttle CSL, Waaijer MEC, Slee-Valentijn MS, Stijnen T, Westendorp R, Maier AB. Cellular senescence and chronological age in various human tissues: a systematic review and meta-analysis. Aging Cell. 2020;19:e13083. doi: 10.1111/acel.13083
51.
Camici GG, Savarese G, Akhmedov A, Luscher TF. Molecular mechanism of endothelial and vascular aging: implications for cardiovascular disease. Eur Heart J. 2015;36:3392–3403. doi: 10.1093/eurheartj/ehv587
52.
Thery C, Gosselin B, Lekieffre J, Warembourg H. Pathology of sinoatrial node. Correlations with electrocardiographic findings in 111 patients. Am Heart J. 1977;93:735–740. doi: 10.1016/s0002-8703(77)80070-7
53.
Shiraishi I, Takamatsu T, Minamikawa T, Onouchi Z, Fujita S. Quantitative histological analysis of the human sinoatrial node during growth and aging. Circulation. 1992;85:2176–2184. doi: 10.1161/01.cir.85.6.2176
54.
Boyle AJ, Shih H, Hwang J, Ye J, Lee B, Zhang Y, Kwon D, Jun K, Zheng D, Sievers R, et al. Cardiomyopathy of aging in the mammalian heart is characterized by myocardial hypertrophy, fibrosis and a predisposition towards cardiomyocyte apoptosis and autophagy. Exp Gerontol. 2011;46:549–559. doi: 10.1016/j.exger.2011.02.010
55.
Biernacka A, Frangogiannis NG. Aging and cardiac fibrosis. Aging Dis. 2011;2:158–173.
56.
Meyer K, Hodwin B, Ramanujam D, Engelhardt S, Sarikas A. Essential role for premature senescence of myofibroblasts in myocardial fibrosis. J Am Coll Cardiol. 2016;67:2018–2028. doi: 10.1016/j.jacc.2016.02.047
57.
Brooks WW, Conrad CH. Myocardial fibrosis in transforming growth factor beta(1)heterozygous mice. J Mol Cell Cardiol. 2000;32:187–195. doi: 10.1006/jmcc.1999.1065
58.
Lu L, Guo J, Hua Y, Huang K, Magaye R, Cornell J, Kelly DJ, Reid C, Liew D, Zhou Y, et al. Cardiac fibrosis in the ageing heart: contributors and mechanisms. Clin Exp Pharmacol Physiol. 2017;44:55–63. doi: 10.1111/1440-1681.12753
59.
Gray DA, Woulfe J. Lipofuscin and aging: a matter of toxic waste. Sci Aging Knowledge Environ. 2005;2005:re1. doi: 10.1126/sageke.2005.5.re1
60.
Xie S, Xu SC, Deng W, Tang Q. Metabolic landscape in cardiac aging: insights into molecular biology and therapeutic implications. Signal Transduct Target Ther. 2023;8:114. doi: 10.1038/s41392-023-01378-8
61.
Feridooni HA, Dibb KM, Howlett SE. How cardiomyocyte excitation, calcium release and contraction become altered with age. J Mol Cell Cardiol. 2015;83:62–72. doi: 10.1016/j.yjmcc.2014.12.004
62.
Schelbert EB, Fridman Y, Wong TC, Abu Daya H, Piehler KM, Kadakkal A, Miller CA, Ugander M, Maanja M, Kellman P, et al. Temporal relation between myocardial fibrosis and heart failure with preserved ejection fraction: association with baseline disease severity and subsequent outcome. JAMA Cardiol. 2017;2:995–1006. doi: 10.1001/jamacardio.2017.2511
63.
Yi CJ, Wu CO, Tee M, Liu CY, Volpe GJ, Prince MR, Hundley GW, Gomes AS, van der Geest RJ, Heckbert S, et al. The association between cardiovascular risk and cardiovascular magnetic resonance measures of fibrosis: the Multi-Ethnic Study of Atherosclerosis (MESA). J Cardiovasc Magn Reson. 2015;17:15. doi: 10.1186/s12968-015-0121-5
64.
Duprez DA, Gross MD, Kizer JR, Ix JH, Hundley WG, Jacobs DR. Predictive value of collagen biomarkers for heart failure with and without preserved ejection fraction: MESA (Multi-Ethnic Study of Atherosclerosis). J Am Heart Assoc. 2018;7:e007885. doi: 10.1161/JAHA.117.007885
65.
Arbab-Zadeh A, Dijk E, Prasad A, Fu Q, Torres P, Zhang R, Thomas JD, Palmer D, Levine BD. Effect of aging and physical activity on left ventricular compliance. Circulation. 2004;110:1799–1805. doi: 10.1161/01.CIR.0000142863.71285.74
66.
Shah AM, Claggett B, Loehr LR, Chang PP, Matsushita K, Kitzman D, Konety S, Kucharska-Newton A, Sueta CA, Mosley TH, et al. Heart failure stages among older adults in the community: the Atherosclerosis Risk in Communities Study. Circulation. 2017;135:224–240. doi: 10.1161/CIRCULATIONAHA.116.023361
67.
Boyd AC, Schiller NB, Leung D, Ross DL, Thomas L. Atrial dilation and altered function are mediated by age and diastolic function but not before the eighth decade. JACC Cardiovasc Imaging. 2011;4:234–242. doi: 10.1016/j.jcmg.2010.11.018
68.
Nikitin NP, Witte KK, Thackray SD, Goodge LJ, Clark AL, Cleland JG. Effect of age and sex on left atrial morphology and function. Eur J Echocardiogr. 2003;4:36–42. doi: 10.1053/euje.2002.0611
69.
Triposkiadis F, Tentolouris K, Androulakis A, Trikas A, Toutouzas K, Kyriakidis M, Gialafos J, Toutouzas P. Left atrial mechanical function in the healthy elderly: new insights from a combined assessment of changes in atrial volume and transmitral flow velocity. J Am Soc Echocardiogr. 1995;8:801–809. doi: 10.1016/s0894-7317(05)80004-5
70.
Gardin JM, Henry WL, Savage DD, Ware JH, Burn C, Borer JS. Echocardiographic measurements in normal subjects: evaluation of an adult population without clinically apparent heart disease. J Clin Ultrasound. 1979;7:439–447. doi: 10.1002/jcu.1870070606
71.
Fleg JL, Strait J. Age-associated changes in cardiovascular structure and function: a fertile milieu for future disease. Heart Fail Rev. 2012;17:545–554. doi: 10.1007/s10741-011-9270-2
72.
Abhayaratna WP, Seward JB, Appleton CP, Douglas PS, Oh JK, Tajik AJ, Tsang TS. Left atrial size: physiologic determinants and clinical applications. J Am Coll Cardiol. 2006;47:2357–2363. doi: 10.1016/j.jacc.2006.02.048
73.
Sun JP, Yang Y, Guo R, Wang D, Lee AP, Wang XY, Lam YY, Fang F, Yang XS, Yu CM. Left atrial regional phasic strain, strain rate and velocity by speckle-tracking echocardiography: normal values and effects of aging in a large group of normal subjects. Int J Cardiol. 2013;168:3473–3479. doi: 10.1016/j.ijcard.2013.04.167
74.
Evin M, Redheuil A, Soulat G, Perdrix L, Ashrafpoor G, Giron A, Lamy J, Defrance C, Roux C, Hatem SN, et al. Left atrial aging: a cardiac magnetic resonance feature-tracking study. Am J Physiol Heart Circ Physiol. 2016;310:H542–H549. doi: 10.1152/ajpheart.00504.2015
75.
Boyd AC, Richards DA, Marwick T, Thomas L. Atrial strain rate is a sensitive measure of alterations in atrial phasic function in healthy ageing. Heart. 2011;97:1513–1519. doi: 10.1136/heartjnl-2011-300134
76.
Okamatsu K, Takeuchi M, Nakai H, Nishikage T, Salgo IS, Husson S, Otsuji Y, Lang RM. Effects of aging on left atrial function assessed by two-dimensional speckle tracking echocardiography. J Am Soc Echocardiogr. 2009;22:70–75. doi: 10.1016/j.echo.2008.11.006
77.
Abou R, Leung M, Tonsbeek AM, Podlesnikar T, Maan AC, Schalij MJ, Ajmone Marsan N, Delgado V, Bax JJ. Effect of aging on left atrial compliance and electromechanical properties in subjects without structural heart disease. Am J Cardiol. 2017;120:140–147. doi: 10.1016/j.amjcard.2017.03.243
78.
Benjamin EJ, D’Agostino RB, Belanger AJ, Wolf PA, Levy D. Left atrial size and the risk of stroke and death. The Framingham Heart Study. Circulation. 1995;92:835–841. doi: 10.1161/01.cir.92.4.835
79.
Habibi M, Zareian M, Ambale Venkatesh B, Samiei S, Imai M, Wu C, Launer LJ, Shea S, Gottesman RF, Heckbert SR, et al. Left atrial mechanical function and incident ischemic cerebrovascular events independent of AF: insights from the MESA study. JACC Cardiovasc Imaging. 2019;12:2417–2427. doi: 10.1016/j.jcmg.2019.02.021
80.
Hees PS, Fleg JL, Lakatta EG, Shapiro EP. Left ventricular remodeling with age in normal men versus women: novel insights using three-dimensional magnetic resonance imaging. Am J Cardiol. 2002;90:1231–1236. doi: 10.1016/s0002-9149(02)02840-0
81.
Gardin JM, Siscovick D, Anton-Culver H, Lynch JC, Smith VE, Klopfenstein HS, Bommer WJ, Fried L, O’Leary D, Manolio TA. Sex, age, and disease affect echocardiographic left ventricular mass and systolic function in the free-living elderly. The Cardiovascular Health Study. Circulation. 1995;91:1739–1748. doi: 10.1161/01.cir.91.6.1739
82.
Dannenberg AL, Levy D, Garrison RJ. Impact of age on echocardiographic left ventricular mass in a healthy population (the Framingham Study). Am J Cardiol. 1989;64:1066–1068. doi: 10.1016/0002-9149(89)90816-3
83.
Kitzman DW, Scholz DG, Hagen PT, Ilstrup DM, Edwards WD. Age-related changes in normal human hearts during the first 10 decades of life. Part II (maturity): a quantitative anatomic study of 765 specimens from subjects 20 to 99 years old. Mayo Clin Proc. 1988;63:137–146. doi: 10.1016/s0025-6196(12)64946-5
84.
Cheng S, Fernandes VR, Bluemke DA, McClelland RL, Kronmal RA, Lima JA. Age-related left ventricular remodeling and associated risk for cardiovascular outcomes: the Multi-Ethnic Study of Atherosclerosis. Circ Cardiovasc Imaging. 2009;2:191–198. doi: 10.1161/CIRCIMAGING.108.819938
85.
Cheng S, Xanthakis V, Sullivan LM, Lieb W, Massaro J, Aragam J, Benjamin EJ, Vasan RS. Correlates of echocardiographic indices of cardiac remodeling over the adult life course: longitudinal observations from the Framingham Heart Study. Circulation. 2010;122:570–578. doi: 10.1161/CIRCULATIONAHA.110.937821
86.
Ganau A, Saba PS, Roman MJ, de Simone G, Realdi G, Devereux RB. Ageing induces left ventricular concentric remodelling in normotensive subjects. J Hypertens. 1995;13:1818–1822.
87.
Slotwiner DJ, Devereux RB, Schwartz JE, Pickering TG, de Simone G, Ganau A, Saba PS, Roman MJ. Relation of age to left ventricular function in clinically normal adults. Am J Cardiol. 1998;82:621–626. doi: 10.1016/s0002-9149(98)00390-7
88.
Peverill RE. Changes in left ventricular size, geometry, pump function and left heart pressures during healthy aging. Rev Cardiovasc Med. 2021;22:717–729. doi: 10.31083/j.rcm2203079
89.
Vriz O, Pirisi M, Habib E, Galzerano D, Fadel B, Antonini-Canterin F, Veldtman G, Bossone E. Age related structural and functional changes in left ventricular performance in healthy subjects: a 2D echocardiographic study. Int J Cardiovasc Imaging. 2019;35:2037–2047. doi: 10.1007/s10554-019-01665-y
90.
Tracy E, Rowe G, LeBlanc AJ. Cardiac tissue remodeling in healthy aging: the road to pathology. Am J Physiol Cell Physiol. 2020;319:C166–C182. doi: 10.1152/ajpcell.00021.2020
91.
Kusy K, Blazejewski J, Gilewski W, Karasek D, Banach J, Bujak R, Zielinski J, Sinkiewicz W, Grzesk G. Aging athlete’s heart: an echocardiographic evaluation of competitive sprint- versus endurance-trained master athletes. J Am Soc Echocardiogr. 2021;34:1160–1169. doi: 10.1016/j.echo.2021.06.009
92.
Sahasakul Y, Edwards WD, Naessens JM, Tajik AJ. Age-related changes in aortic and mitral valve thickness: implications for two-dimensional echocardiography based on an autopsy study of 200 normal human hearts. Am J Cardiol. 1988;62:424–430. doi: 10.1016/0002-9149(88)90971-x
93.
Stewart BF, Siscovick D, Lind BK, Gardin JM, Gottdiener JS, Smith VE, Kitzman DW, Otto CM. Clinical factors associated with calcific aortic valve disease. Cardiovascular Health Study. J Am Coll Cardiol. 1997;29:630–634. doi: 10.1016/s0735-1097(96)00563-3
94.
Fox CS, Vasan RS, Parise H, Levy D, O’Donnell CJ, D’Agostino RB, Benjamin EJ, Framingham Heart S. Mitral annular calcification predicts cardiovascular morbidity and mortality: the Framingham Heart Study. Circulation. 2003;107:1492–1496. doi: 10.1161/01.cir.0000058168.26163.bc
95.
Moncla LM, Briend M, Bosse Y, Mathieu P. Calcific aortic valve disease: mechanisms, prevention and treatment. Nat Rev Cardiol. 2023;20:546–559. doi: 10.1038/s41569-023-00845-7
96.
Otto CM, Kuusisto J, Reichenbach DD, Gown AM, O’Brien KD. Characterization of the early lesion of ‘degenerative’ valvular aortic stenosis. Histological and immunohistochemical studies. Circulation. 1994;90:844–853. doi: 10.1161/01.cir.90.2.844
97.
Rodriguez KJ, Piechura LM, Porras AM, Masters KS. Manipulation of valve composition to elucidate the role of collagen in aortic valve calcification. BMC Cardiovasc Disord. 2014;14:29. doi: 10.1186/1471-2261-14-29
98.
Freeman RV, Otto CM. Spectrum of calcific aortic valve disease: pathogenesis, disease progression, and treatment strategies. Circulation. 2005;111:3316–3326. doi: 10.1161/CIRCULATIONAHA.104.486738
100.
Chuang ML, Gona P, Hautvast GL, Salton CJ, Breeuwer M, O’Donnell CJ, Manning WJ. CMR reference values for left ventricular volumes, mass, and ejection fraction using computer-aided analysis: the Framingham Heart Study. J Magn Reson Imaging. 2014;39:895–900. doi: 10.1002/jmri.24239
101.
Cheng S, Larson MG, McCabe EL, Osypiuk E, Lehman BT, Stanchev P, Aragam J, Benjamin EJ, Solomon SD, Vasan RS. Age- and sex-based reference limits and clinical correlates of myocardial strain and synchrony: the Framingham Heart Study. Circ Cardiovasc Imaging. 2013;6:692–699. doi: 10.1161/CIRCIMAGING.112.000627
102.
Lee KY, Kim HL, Kim KJ. Sex difference in the age-related decline of global longitudinal strain of left ventricle. Sci Rep. 2023;13:18441. doi: 10.1038/s41598-023-42286-9
103.
Sugimoto T, Dulgheru R, Bernard A, Ilardi F, Contu L, Addetia K, Caballero L, Akhaladze N, Athanassopoulos GD, Barone D, et al. Echocardiographic reference ranges for normal left ventricular 2D strain: results from the EACVI NORRE study. Eur Heart J Cardiovasc Imaging. 2017;18:833–840. doi: 10.1093/ehjci/jex140
104.
Subramanian V, Keshvani N, Segar MW, Kondamudi NJ, Chandra A, Maddineni B, Matulevicius SA, Michos ED, Lima JAC, Berry JD, et al. Association of global longitudinal strain by feature tracking cardiac magnetic resonance imaging with adverse outcomes among community-dwelling adults without cardiovascular disease: The Dallas Heart Study. Eur J Heart Fail. 2024;26:208–215. doi: 10.1002/ejhf.3158
105.
Potter EL, Wright L, Yang H, Marwick TH. Normal range of global longitudinal strain in the elderly: the impact of subclinical disease. JACC Cardiovasc Imaging. 2021;14:300–302. doi: 10.1016/j.jcmg.2020.07.014
106.
Gardin JM, Arnold AM, Bild DE, Smith VE, Lima JA, Klopfenstein HS, Kitzman DW. Left ventricular diastolic filling in the elderly: the cardiovascular health study. Am J Cardiol. 1998;82:345–351. doi: 10.1016/s0002-9149(98)00339-7
107.
Shah AM, Claggett B, Kitzman D, Biering-Sorensen T, Jensen JS, Cheng S, Matsushita K, Konety S, Folsom AR, Mosley TH, et al. Contemporary assessment of left ventricular diastolic function in older adults: the Atherosclerosis Risk in Communities Study. Circulation. 2017;135:426–439. doi: 10.1161/CIRCULATIONAHA.116.024825
108.
Sagie A, Benjamin EJ, Galderisi M, Larson MG, Evans JC, Fuller DL, Lehman B, Levy D. Echocardiographic assessment of left ventricular structure and diastolic filling in elderly subjects with borderline isolated systolic hypertension (the Framingham Heart Study). Am J Cardiol. 1993;72:662–665. doi: 10.1016/0002-9149(93)90881-c
109.
Downes TR, Nomeir AM, Smith KM, Stewart KP, Little WC. Mechanism of altered pattern of left ventricular filling with aging in subjects without cardiac disease. Am J Cardiol. 1989;64:523–527. doi: 10.1016/0002-9149(89)90433-5
110.
Hees PS, Fleg JL, Dong SJ, Shapiro EP. MRI and echocardiographic assessment of the diastolic dysfunction of normal aging: altered LV pressure decline or load? Am J Physiol Heart Circ Physiol. 2004;286:H782–H788. doi: 10.1152/ajpheart.01092.2002
111.
Nagueh SF, Smiseth OA, Appleton CP, Byrd BF, Dokainish H, Edvardsen T, Flachskampf FA, Gillebert TC, Klein AL, Lancellotti P, et al. Recommendations for the evaluation of left ventricular diastolic function by echocardiography: an update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. J Am Soc Echocardiogr. 2016;29:277–314. doi: 10.1016/j.echo.2016.01.011
112.
Henein M, Lindqvist P, Francis D, Morner S, Waldenstrom A, Kazzam E. Tissue Doppler analysis of age-dependency in diastolic ventricular behaviour and filling: a cross-sectional study of healthy hearts (the Umea General Population Heart Study). Eur Heart J. 2002;23:162–171. doi: 10.1053/euhj.2001.3032
113.
Sohn DW, Chai IH, Lee DJ, Kim HC, Kim HS, Oh BH, Lee MM, Park YB, Choi YS, Seo JD, et al. Assessment of mitral annulus velocity by Doppler tissue imaging in the evaluation of left ventricular diastolic function. J Am Coll Cardiol. 1997;30:474–480. doi: 10.1016/s0735-1097(97)88335-0
114.
Nayor M, Cooper LL, Enserro DM, Xanthakis V, Larson MG, Benjamin EJ, Aragam J, Mitchell GF, Vasan RS. Left ventricular diastolic dysfunction in the community: impact of diagnostic criteria on the burden, correlates, and prognosis. J Am Heart Assoc. 2018;7:e008291. doi: 10.1161/JAHA.117.008291
115.
Ungvari Z, Tarantini S, Donato AJ, Galvan V, Csiszar A. Mechanisms of vascular aging. Circ Res. 2018;123:849–867. doi: 10.1161/CIRCRESAHA.118.311378
116.
AlGhatrif M, Lakatta EG. The conundrum of arterial stiffness, elevated blood pressure, and aging. Curr Hypertens Rep. 2015;17:12. doi: 10.1007/s11906-014-0523-z
117.
Lacolley P, Regnault V, Avolio AP. Smooth muscle cell and arterial aging: basic and clinical aspects. Cardiovasc Res. 2018;114:513–528. doi: 10.1093/cvr/cvy009
118.
Lakatta EG, Levy D. Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: part I: aging arteries: a “set up” for vascular disease. Circulation. 2003;107:139–146. doi: 10.1161/01.cir.0000048892.83521.58
119.
Leopold JA. Vascular calcification: an age-old problem of old age. Circulation. 2013;127:2380–2382. doi: 10.1161/CIRCULATIONAHA.113.003341
120.
Ungvari Z, Kaley G, de Cabo R, Sonntag WE, Csiszar A. Mechanisms of vascular aging: new perspectives. J Gerontol A Biol Sci Med Sci. 2010;65:1028–1041. doi: 10.1093/gerona/glq113
121.
Donato AJ, Eskurza I, Silver AE, Levy AS, Pierce GL, Gates PE, Seals DR. Direct evidence of endothelial oxidative stress with aging in humans: relation to impaired endothelium-dependent dilation and upregulation of nuclear factor-kappaB. Circ Res. 2007;100:1659–1666. doi: 10.1161/01.RES.0000269183.13937.e8
122.
van der Loo B, Labugger R, Skepper JN, Bachschmid M, Kilo J, Powell JM, Palacios-Callender M, Erusalimsky JD, Quaschning T, Malinski T, et al. Enhanced peroxynitrite formation is associated with vascular aging. J Exp Med. 2000;192:1731–1744. doi: 10.1084/jem.192.12.1731
123.
Sun D, Huang A, Yan EH, Wu Z, Yan C, Kaminski PM, Oury TD, Wolin MS, Kaley G. Reduced release of nitric oxide to shear stress in mesenteric arteries of aged rats. Am J Physiol Heart Circ Physiol. 2004;286:H2249–H2256. doi: 10.1152/ajpheart.00854.2003
124.
Lakatta EG, Levy D. Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: part II: the aging heart in health: links to heart disease. Circulation. 2003;107:346–354. doi: 10.1161/01.cir.0000048893.62841.f7
125.
Jablonski KL, Seals DR, Eskurza I, Monahan KD, Donato AJ. High-dose ascorbic acid infusion abolishes chronic vasoconstriction and restores resting leg blood flow in healthy older men. J Appl Physiol (1985). 2007;103:1715–1721. doi: 10.1152/japplphysiol.00533.2007
126.
Kahe F, Sharfaei S, Pitliya A, Jafarizade M, Seifirad S, Habibi S, Chi G. Coronary artery tortuosity: a narrative review. Coron Artery Dis. 2020;31:187–192. doi: 10.1097/MCA.0000000000000769
127.
Kaplan H, Thompson RC, Trumble BC, Wann LS, Allam AH, Beheim B, Frohlich B, Sutherland ML, Sutherland JD, Stieglitz J, et al. Coronary atherosclerosis in indigenous South American Tsimane: a cross-sectional cohort study. Lancet. 2017;389:1730–1739. doi: 10.1016/S0140-6736(17)30752-3
128.
Mueller NT, Noya-Alarcon O, Contreras M, Appel LJ, Dominguez-Bello MG. Association of age with blood pressure across the lifespan in isolated Yanomami and Yekwana villages. JAMA Cardiol. 2018;3:1247–1249. doi: 10.1001/jamacardio.2018.3676
129.
Rogers MA, Hagberg JM, Martin WH, Ehsani AA, Holloszy JO. Decline in VO2max with aging in master athletes and sedentary men. J Appl Physiol (1985). 1990;68:2195–2199. doi: 10.1152/jappl.1990.68.5.2195
130.
Tanaka H, Desouza CA, Jones PP, Stevenson ET, Davy KP, Seals DR. Greater rate of decline in maximal aerobic capacity with age in physically active vs. sedentary healthy women. J Appl Physiol (1985). 1997;83:1947–1953. doi: 10.1152/jappl.1997.83.6.1947
131.
Toth MJ, Gardner AW, Ades PA, Poehlman ET. Contribution of body composition and physical activity to age-related decline in peak VO2 in men and women. J Appl Physiol (1985). 1994;77:647–652. doi: 10.1152/jappl.1994.77.2.647
132.
Talbot LA, Metter EJ, Fleg JL. Leisure-time physical activities and their relationship to cardiorespiratory fitness in healthy men and women 18-95 years old. Med Sci Sports Exerc. 2000;32:417–425. doi: 10.1097/00005768-200002000-00024
133.
Fleg JL, Morrell CH, Bos AG, Brant LJ, Talbot LA, Wright JG, Lakatta EG. Accelerated longitudinal decline of aerobic capacity in healthy older adults. Circulation. 2005;112:674–682. doi: 10.1161/CIRCULATIONAHA.105.545459
134.
Tanaka H, Monahan KD, Seals DR. Age-predicted maximal heart rate revisited. J Am Coll Cardiol. 2001;37:153–156. doi: 10.1016/s0735-1097(00)01054-8
135.
Yaniv Y, Ahmet I, Tsutsui K, Behar J, Moen JM, Okamoto Y, Guiriba TR, Liu J, Bychkov R, Lakatta EG. Deterioration of autonomic neuronal receptor signaling and mechanisms intrinsic to heart pacemaker cells contribute to age-associated alterations in heart rate variability in vivo. Aging Cell. 2016;15:716–724. doi: 10.1111/acel.12483
136.
Christou DD, Seals DR. Decreased maximal heart rate with aging is related to reduced beta-adrenergic responsiveness but is largely explained by a reduction in intrinsic heart rate. J Appl Physiol (1985). 2008;105:24–29. doi: 10.1152/japplphysiol.90401.2008
137.
Higginbotham MB, Morris KG, Williams RS, Coleman RE, Cobb FR. Physiologic basis for the age-related decline in aerobic work capacity. Am J Cardiol. 1986;57:1374–1379. doi: 10.1016/0002-9149(86)90221-3
138.
McGavock JM, Hastings JL, Snell PG, McGuire DK, Pacini EL, Levine BD, Mitchell JH. A forty-year follow-up of the Dallas Bed Rest and Training study: the effect of age on the cardiovascular response to exercise in men. J Gerontol A Biol Sci Med Sci. 2009;64:293–299. doi: 10.1093/gerona/gln025
139.
McGuire DK, Levine BD, Williamson JW, Snell PG, Blomqvist CG, Saltin B, Mitchell JH. A 30-year follow-up of the Dallas bedrest and training study: I. Effect of age on the cardiovascular response to exercise. Circulation. 2001;104:1350–1357.
140.
van Empel VP, Kaye DM, Borlaug BA. Effects of healthy aging on the cardiopulmonary hemodynamic response to exercise. Am J Cardiol. 2014;114:131–135. doi: 10.1016/j.amjcard.2014.04.011
141.
Wolsk E, Bakkestrøm R, Thomsen JH, Balling L, Andersen MJ, Dahl JS, Hassager C, Møller JE, Gustafsson F. The influence of age on hemodynamic parameters during rest and exercise in healthy individuals. JACC Heart Fail. 2017;5:337–346. doi: 10.1016/j.jchf.2016.10.012
142.
Zeder K, Avian A, Mak S, Giannakoulas G, Kawut SM, Maron BA, Humbert M, Olschewski H, Kovacs G. Pulmonary arterial wedge pressure in healthy subjects: a meta-analysis. Eur Respir J. 2024;64:2400967. doi: 10.1183/13993003.00967-2024
143.
Zile MR, Baicu CF, Gaasch WH. Diastolic heart failure--abnormalities in active relaxation and passive stiffness of the left ventricle. N Engl J Med. 2004;350:1953–1959. doi: 10.1056/NEJMoa032566
144.
Borlaug BA, Nishimura RA, Sorajja P, Lam CS, Redfield MM. Exercise hemodynamics enhance diagnosis of early heart failure with preserved ejection fraction. Circ Heart Fail. 2010;3:588–595. doi: 10.1161/CIRCHEARTFAILURE.109.930701
145.
Beere PA, Russell SD, Morey MC, Kitzman DW, Higginbotham MB. Aerobic exercise training can reverse age-related peripheral circulatory changes in healthy older men. Circulation. 1999;100:1085–1094. doi: 10.1161/01.cir.100.10.1085
146.
Carrick-Ranson G, Hastings JL, Bhella PS, Shibata S, Fujimoto N, Palmer D, Boyd K, Levine BD. The effect of age-related differences in body size and composition on cardiovascular determinants of VO2max. J Gerontol A Biol Sci Med Sci. 2012;68:608–616. doi: 10.1093/gerona/gls220
147.
Fuller A, Okwose N, Scragg J, Eggett C, Luke P, Bandali A, Velicki R, Greaves L, MacGowan GA, Jakovljevic DG. The effect of age on mechanisms of exercise tolerance: reduced arteriovenous oxygen difference causes lower oxygen consumption in older people. Exp Gerontol. 2021;149:111340. doi: 10.1016/j.exger.2021.111340
148.
Houghton D, Jones TW, Cassidy S, Siervo M, MacGowan GA, Trenell MI, Jakovljevic DG. The effect of age on the relationship between cardiac and vascular function. Mech Ageing Dev. 2016;153:1–6. doi: 10.1016/j.mad.2015.11.001
149.
Ogawa T, Spina RJ, Martin WH, Kohrt WM, Schechtman KB, Holloszy JO, Ehsani AA. Effects of aging, sex, and physical training on cardiovascular responses to exercise. Circulation. 1992;86:494–503. doi: 10.1161/01.cir.86.2.494
150.
Rodeheffer RJ, Gerstenblith G, Becker LC, Fleg JL, Weisfeldt ML, Lakatta EG. Exercise cardiac output is maintained with advancing age in healthy human subjects: cardiac dilatation and increased stroke volume compensate for a diminished heart rate. Circulation. 1984;69:203–213. doi: 10.1161/01.cir.69.2.203
151.
Batsis JA, Villareal DT. Sarcopenic obesity in older adults: aetiology, epidemiology and treatment strategies. Nat Rev Endocrinol. 2018;14:513–537. doi: 10.1038/s41574-018-0062-9
152.
Distefano G, Goodpaster BH. Effects of exercise and aging on skeletal muscle. Cold Spring Harb Perspect Med. 2018;8:a029785. doi: 10.1101/cshperspect.a029785
153.
Fleg JL, Lakatta EG. Role of muscle loss in the age-associated reduction in VO2 max. J Appl Physiol (1985). 1988;65:1147–1151. doi: 10.1152/jappl.1988.65.3.1147
154.
Proctor DN, Joyner MJ. Skeletal muscle mass and the reduction of VO2max in trained older subjects. J Appl Physiol (1985). 1997;82:1411–1415. doi: 10.1152/jappl.1997.82.5.1411
155.
Haykowsky MJ, Kouba EJ, Brubaker PH, Nicklas BJ, Eggebeen J, Kitzman DW. Skeletal muscle composition and its relation to exercise intolerance in older patients with heart failure and preserved ejection fraction. Am J Cardiol. 2014;113:1211–1216. doi: 10.1016/j.amjcard.2013.12.031
156.
Marcus RL, Addison O, Dibble LE, Foreman KB, Morrell G, Lastayo P. Intramuscular adipose tissue, sarcopenia, and mobility function in older individuals. J Aging Res. 2012;2012:629637. doi: 10.1155/2012/629637
157.
Tuttle LJ, Sinacore DR, Mueller MJ. Intermuscular adipose tissue is muscle specific and associated with poor functional performance. J Aging Res. 2012;2012:172957. doi: 10.1155/2012/172957
158.
Haykowsky MJ, Brubaker PH, Morgan TM, Kritchevsky S, Eggebeen J, Kitzman DW. Impaired aerobic capacity and physical functional performance in older heart failure patients with preserved ejection fraction: role of lean body mass. J Gerontol A Biol Sci Med Sci. 2013;68:968–975. doi: 10.1093/gerona/glt011
159.
Goodpaster BH, Park SW, Harris TB, Kritchevsky SB, Nevitt M, Schwartz AV, Simonsick EM, Tylavsky FA, Visser M, Newman AB. The loss of skeletal muscle strength, mass, and quality in older adults: the health, aging and body composition study. J Gerontol A Biol Sci Med Sci. 2006;61:1059–1064. doi: 10.1093/gerona/61.10.1059
160.
Hilton TN, Tuttle LJ, Bohnert KL, Mueller MJ, Sinacore DR. Excessive adipose tissue infiltration in skeletal muscle in individuals with obesity, diabetes mellitus, and peripheral neuropathy: association with performance and function. Phys Ther. 2008;88:1336–1344. doi: 10.2522/ptj.20080079
161.
Visser M, Kritchevsky SB, Goodpaster BH, Newman AB, Nevitt M, Stamm E, Harris TB. Leg muscle mass and composition in relation to lower extremity performance in men and women aged 70 to 79: the health, aging and body composition study. J Am Geriatr Soc. 2002;50:897–904. doi: 10.1046/j.1532-5415.2002.50217.x
162.
Heinonen I, Bucci M, Kemppainen J, Knuuti J, Nuutila P, Boushel R, Kalliokoski KK. Regulation of subcutaneous adipose tissue blood flow during exercise in humans. J Appl Physiol (1985). 2012;112:1059–1063. doi: 10.1152/japplphysiol.00732.2011
163.
Kirby BS, Voyles WF, Simpson CB, Carlson RE, Schrage WG, Dinenno FA. Endothelium-dependent vasodilatation and exercise hyperaemia in ageing humans: impact of acute ascorbic acid administration. J Physiol. 2009;587:1989–2003. doi: 10.1113/jphysiol.2008.167320
164.
Poole JG, Lawrenson L, Kim J, Brown C, Richardson RS. Vascular and metabolic response to cycle exercise in sedentary humans: effect of age. Am J Physiol Heart Circ Physiol. 2003;284:H1251–H1259. doi: 10.1152/ajpheart.00790.2002
165.
Proctor DN, Koch DW, Newcomer SC, Le KU, Leuenberger UA. Impaired leg vasodilation during dynamic exercise in healthy older women. J Appl Physiol (1985). 2003;95:1963–1970. doi: 10.1152/japplphysiol.00472.2003
166.
Proctor DN, Shen PH, Dietz NM, Eickhoff TJ, Lawler LA, Ebersold EJ, Loeffler DL, Joyner MJ. Reduced leg blood flow during dynamic exercise in older endurance-trained men. J Appl Physiol (1985). 1998;85:68–75. doi: 10.1152/jappl.1998.85.1.68
167.
Proctor DN, Koch DW, Newcomer SC, Le KU, Smithmyer SL, Leuenberger UA. Leg blood flow and VO2 during peak cycle exercise in younger and older women. Med Sci Sports Exerc. 2004;36:623–631. doi: 10.1249/01.mss.0000121951.10417.b5
168.
Dinenno FA, Seals DR, DeSouza CA, Tanaka H. Age-related decreases in basal limb blood flow in humans: time course, determinants and habitual exercise effects. J Physiol. 2001;531:573–579. doi: 10.1111/j.1469-7793.2001.0573i.x
169.
Hearon CM, Dinenno FA. Regulation of skeletal muscle blood flow during exercise in ageing humans. J Physiol. 2016;594:2261–2273. doi: 10.1113/JP270593
170.
Schrage WG, Eisenach JH, Joyner MJ. Ageing reduces nitric-oxide- and prostaglandin-mediated vasodilatation in exercising humans. J Physiol. 2007;579:227–236. doi: 10.1113/jphysiol.2006.124313
171.
Barrett-O’Keefe Z, Ives SJ, Trinity JD, Morgan G, Rossman MJ, Donato AJ, Runnels S, Morgan DE, Gmelch BS, Bledsoe AD, et al. Endothelin-A-mediated vasoconstriction during exercise with advancing age. J Gerontol A Biol Sci Med Sci. 2015;70:554–565. doi: 10.1093/gerona/glu065
172.
Fadel PJ, Wang Z, Watanabe H, Arbique D, Vongpatanasin W, Thomas GD. Augmented sympathetic vasoconstriction in exercising forearms of postmenopausal women is reversed by oestrogen therapy. J Physiol. 2004;561:893–901. doi: 10.1113/jphysiol.2004.073619
173.
Koch DW, Leuenberger UA, Proctor DN. Augmented leg vasoconstriction in dynamically exercising older men during acute sympathetic stimulation. J Physiol. 2003;551:337–344. doi: 10.1113/jphysiol.2003.042747
174.
Coggan AR, Spina RJ, King DS, Rogers MA, Brown M, Nemeth PM, Holloszy JO. Histochemical and enzymatic comparison of the gastrocnemius muscle of young and elderly men and women. J Gerontol. 1992;47:B71–B76. doi: 10.1093/geronj/47.3.b71
175.
Groen BB, Hamer HM, Snijders T, van Kranenburg J, Frijns D, Vink H, van Loon LJ. Skeletal muscle capillary density and microvascular function are compromised with aging and type 2 diabetes. J Appl Physiol (1985). 2014;116:998–1005. doi: 10.1152/japplphysiol.00919.2013
176.
Landers-Ramos RQ, Prior SJ. The microvasculature and skeletal muscle health in aging. Exerc Sport Sci Rev. 2018;46:172–179. doi: 10.1249/JES.0000000000000151
177.
Hellsten Y, Gliemann L. Peripheral limitations for performance: Muscle capillarization. Scand J Med Sci Sports. 2024;34:e14442. doi: 10.1111/sms.14442
178.
Haykowsky MJ, Brubaker PH, John JM, Stewart KP, Morgan TM, Kitzman DW. Determinants of exercise intolerance in elderly heart failure patients with preserved ejection fraction. J Am Coll Cardiol. 2011;58:265–274. doi: 10.1016/j.jacc.2011.02.055
179.
Kitzman DW, Nicklas B, Kraus WE, Lyles MF, Eggebeen J, Morgan TM, Haykowsky M. Skeletal muscle abnormalities and exercise intolerance in older patients with heart failure and preserved ejection fraction. Am J Physiol Heart Circ Physiol. 2014;306:H1364–H1370. doi: 10.1152/ajpheart.00004.2014
180.
Singh MA. Exercise to prevent and treat functional disability. Clin Geriatr Med. 2002;18:431–462, vi. doi: 10.1016/s0749-0690(02)00016-2
181.
Wu D, Jia Y, Liu Y, Pan X, Li P, Shang M. Dose response of leisure time physical activity and biological aging in type 2 diabetes: a cross sectional study. Sci Rep. 2024;14:26253. doi: 10.1038/s41598-024-77359-w
182.
Mor V, Murphy J, Masterson-Allen S, Willey C, Razmpour A, Jackson ME, Greer D, Katz S. Risk of functional decline among well elders. J Clin Epidemiol. 1989;42:895–904. doi: 10.1016/0895-4356(89)90103-0
183.
Miller ME, Rejeski WJ, Reboussin BA, Ten Have TR, Ettinger WH. Physical activity, functional limitations, and disability in older adults. J Am Geriatr Soc. 2000;48:1264–1272. doi: 10.1111/j.1532-5415.2000.tb02600.x
184.
Peterson MJ, Giuliani C, Morey MC, Pieper CF, Evenson KR, Mercer V, Cohen HJ, Visser M, Brach JS, Kritchevsky SB, et al; Health, Aging and Body Composition Study Research Group. Physical activity as a preventative factor for frailty: the health, aging, and body composition study. J Gerontol A Biol Sci Med Sci. 2009;64:61–68. doi: 10.1093/gerona/gln001
185.
Hamer M, Lavoie KL, Bacon SL. Taking up physical activity in later life and healthy ageing: the English longitudinal study of ageing. Br J Sports Med. 2014;48:239–243. doi: 10.1136/bjsports-2013-092993
186.
Paffenbarger RS, Wolf PA, Notkin J, Thorne MC. Chronic disease in former college students. I. Early precursors of fatal coronary heart disease. Am J Epidemiol. 1966;83:314–328. doi: 10.1093/oxfordjournals.aje.a120587
187.
Paffenbarger RS, Wing AL, Hyde RT. Physical activity as an index of heart attack risk in college alumni. Am J Epidemiol. 1978;108:161–175. doi: 10.1093/oxfordjournals.aje.a112608
188.
Lee IM, Paffenbarger RS. Associations of light, moderate, and vigorous intensity physical activity with longevity. The Harvard Alumni Health Study. Am J Epidemiol. 2000;151:293–299. doi: 10.1093/oxfordjournals.aje.a010205
189.
Ekelund U, Tarp J, Steene-Johannessen J, Hansen BH, Jefferis B, Fagerland MW, Whincup P, Diaz KM, Hooker SP, Chernofsky A, et al. Dose-response associations between accelerometry measured physical activity and sedentary time and all cause mortality: systematic review and harmonised meta-analysis. BMJ. 2019;366:l4570. doi: 10.1136/bmj.l4570
190.
Bull FC, Al-Ansari SS, Biddle S, Borodulin K, Buman MP, Cardon G, Carty C, Chaput JP, Chastin S, Chou R, et al. World Health Organization 2020 guidelines on physical activity and sedentary behaviour. Br J Sports Med. 2020;54:1451–1462. doi: 10.1136/bjsports-2020-102955.
191.
Ramakrishnan R, Doherty A, Smith-Byrne K, Rahimi K, Bennett D, Woodward M, Walmsley R, Dwyer T. Accelerometer measured physical activity and the incidence of cardiovascular disease: evidence from the UK Biobank cohort study. PLoS Med. 2021;18:e1003487. doi: 10.1371/journal.pmed.1003487
192.
Paluch AE, Bajpai S, Ballin M, Bassett DR, Buford TW, Carnethon MR, Chernofsky A, Dooley EE, Ekelund U, Evenson KR, et al; Steps for Health Collaborative. Prospective association of daily steps with cardiovascular disease: a harmonized meta-analysis. Circulation. 2023;147:122–131. doi: 10.1161/CIRCULATIONAHA.122.061288
193.
LaCroix AZ, Bellettiere J, Rillamas-Sun E, Di C, Evenson KR, Lewis CE, Buchner DM, Stefanick ML, Lee IM, Rosenberg DE, et al; Women’s Health Initiative (WHI). Association of light physical activity measured by accelerometry and incidence of coronary heart disease and cardiovascular disease in older women. JAMA Netw Open. 2019;2:e190419. doi: 10.1001/jamanetworkopen.2019.0419
194.
Berry JD, Pandey A, Gao A, Leonard D, Farzaneh-Far R, Ayers C, DeFina L, Willis B. Physical fitness and risk for heart failure and coronary artery disease. Circ Heart Fail. 2013;6:627–634. doi: 10.1161/CIRCHEARTFAILURE.112.000054
195.
Andersen K, Mariosa D, Adami HO, Held C, Ingelsson E, Lagerros YT, Nyren O, Ye W, Bellocco R, Sundstrom J. Dose-response relationship of total and leisure time physical activity to risk of heart failure: a prospective cohort study. Circ Heart Fail. 2014;7:701–708. doi: 10.1161/CIRCHEARTFAILURE.113.001010
196.
Pandey A, Garg S, Khunger M, Darden D, Ayers C, Kumbhani DJ, Mayo HG, de Lemos JA, Berry JD. Dose-response relationship between physical activity and risk of heart failure: a meta-analysis. Circulation. 2015;132:1786–1794. doi: 10.1161/CIRCULATIONAHA.115.015853
197.
LaMonte MJ, LaCroix AZ, Nguyen S, Evenson KR, Di C, Stefanick ML, Hyde ET, Anuskiewicz B, Eaton CB. Accelerometer-measured physical activity, sedentary time, and heart failure risk in women aged 63 to 99 years. JAMA Cardiol. 2024;9:336–345. doi: 10.1001/jamacardio.2023.5692
198.
LaMonte MJ, Buchner DM, Rillamas-Sun E, Di C, Evenson KR, Bellettiere J, Lewis CE, Lee IM, Tinker LF, Seguin R, et al. Accelerometer-measured physical activity and mortality in women aged 63 to 99. J Am Geriatr Soc. 2018;66:886–894. doi: 10.1111/jgs.15201
199.
Ho FK, Zhou Z, Petermann-Rocha F, Para-Soto S, Boonpor J, Welsh P, Gill JMR, Gray SR, Sattar N, Pell JP, et al. Association between device-measured physical activity and incident heart failure: a prospective cohort study of 94 739 UK biobank participants. Circulation. 2022;146:883–891. doi: 10.1161/CIRCULATIONAHA.122.059663
200.
Pandey A, LaMonte M, Klein L, Ayers C, Psaty BM, Eaton CB, Allen NB, de Lemos JA, Carnethon M, Greenland P, et al. Relationship between physical activity, body mass index, and risk of heart failure. J Am Coll Cardiol. 2017;69:1129–1142. doi: 10.1016/j.jacc.2016.11.081
201.
Pandey A, Patel M, Gao A, Willis BL, Das SR, Leonard D, Drazner MH, de Lemos JA, DeFina L, Berry JD. Changes in mid-life fitness predicts heart failure risk at a later age independent of interval development of cardiac and noncardiac risk factors: the Cooper Center Longitudinal Study. Am Heart J. 2015;169:290–297.e1. doi: 10.1016/j.ahj.2014.10.017
202.
Blodgett JM, Ahmadi MN, Atkin AJ, Chastin S, Chan HW, Suorsa K, Bakker EA, Hettiarcachchi P, Johansson PJ, Sherar LB, et al; ProPASS Collaboration. Device-measured physical activity and cardiometabolic health: the Prospective Physical Activity, Sitting, and Sleep (ProPASS) consortium. Eur Heart J. 2024;45:458–471. doi: 10.1093/eurheartj/ehad717
203.
Khurshid S, Weng LC, Nauffal V, Pirruccello JP, Venn RA, Al-Alusi MA, Benjamin EJ, Ellinor PT, Lubitz SA. Wearable accelerometer-derived physical activity and incident disease. NPJ Digit Med. 2022;5:131. doi: 10.1038/s41746-022-00676-9
204.
Carnethon MR, Evans NS, Church TS, Lewis CE, Schreiner PJ, Jacobs DR, Sternfeld B, Sidney S. Joint associations of physical activity and aerobic fitness on the development of incident hypertension: coronary artery risk development in young adults. Hypertension. 2010;56:49–55. doi: 10.1161/HYPERTENSIONAHA.109.147603
205.
Forman JP, Stampfer MJ, Curhan GC. Diet and lifestyle risk factors associated with incident hypertension in women. JAMA. 2009;302:401–411. doi: 10.1001/jama.2009.1060
206.
Asferg C, Mogelvang R, Flyvbjerg A, Frystyk J, Jensen JS, Marott JL, Appleyard M, Schnohr P, Jensen GB, Jeppesen JR. Interaction between leptin and leisure-time physical activity and development of hypertension. Blood Press. 2011;20:362–369. doi: 10.3109/00365599.2011.586248
207.
Pandey A, Allen NB, Ayers C, Reis JP, Moreira HT, Sidney S, Rana JS, Jacobs DR, Chow LS, de Lemos JA, et al. Fitness in young adulthood and long-term cardiac structure and function: the CARDIA study. JACC Heart Fail. 2017;5:347–355. doi: 10.1016/j.jchf.2016.11.014
208.
Kondamudi N, Thangada N, Patel KV, Ayers C, Chandra A, Berry JD, Neeland IJ, Pandey A. Regional adiposity, cardiorespiratory fitness, and left ventricular strain: an analysis from the Dallas Heart Study. J Cardiovasc Magn Reson. 2021;23:78. doi: 10.1186/s12968-021-00757-w
209.
Thangada ND, Patel KV, Peden B, Agusala V, Kozlitina J, Garg S, Drazner MH, Ayers C, Berry JD, Pandey A. Cross-sectional associations of objectively measured sedentary time, physical activity, and fitness with cardiac structure and function: findings from the Dallas Heart Study. J Am Heart Assoc. 2021;10:e015601. doi: 10.1161/JAHA.119.015601
210.
Brinker SK, Pandey A, Ayers CR, Barlow CE, DeFina LF, Willis BL, Radford NB, Farzaneh-Far R, de Lemos JA, Drazner MH, et al. Association of cardiorespiratory fitness with left ventricular remodeling and diastolic function: the Cooper Center Longitudinal Study. JACC Heart Fail. 2014;2:238–246. doi: 10.1016/j.jchf.2014.01.004
211.
Lohman T, Bains G, Cole S, Gharibvand L, Berk L, Lohman E. High-Intensity interval training reduces transcriptomic age: a randomized controlled trial. Aging Cell. 2023;22:e13841. doi: 10.1111/acel.13841
212.
Ho E, Qualls C, Villareal DT. Effect of diet, exercise, or both on biological age and healthy aging in older adults with obesity: secondary analysis of a randomized controlled trial. J Nutr Health Aging. 2022;26:552–557. doi: 10.1007/s12603-022-1812-x
213.
Bischoff-Ferrari HA, Gangler S, Wieczorek M, Belsky DW, Ryan J, Kressig RW, Stahelin HB, Theiler R, Dawson-Hughes B, Rizzoli R, et al. Individual and additive effects of vitamin D, omega-3 and exercise on DNA methylation clocks of biological aging in older adults from the DO-HEALTH trial. Nat Aging. 2025;5:376–385. doi: 10.1038/s43587-024-00793-y
214.
Huang G, Gibson CA, Tran ZV, Osness WH. Controlled endurance exercise training and VO2max changes in older adults: a meta-analysis. Prev Cardiol. 2005;8:217–225. doi: 10.1111/j.0197-3118.2005.04324.x
215.
Howden EJ, Levine BD. Reversing the cardiac effects of sedentary. Circulation. 2018;138:1759–1760. doi: 10.1161/circulationaha.118.036249
216.
van Baak MA, Pramono A, Battista F, Beaulieu K, Blundell JE, Busetto L, Carraça EV, Dicker D, Encantado J, Ermolao A, et al. Effect of different types of regular exercise on physical fitness in adults with overweight or obesity: systematic review and meta-analyses. Obes Rev. 2021;22:1–11. doi: 10.1111/obr.13239
217.
Conraads VM, Pattyn N, De Maeyer C, Beckers PJ, Coeckelberghs E, Cornelissen VA, Denollet J, Frederix G, Goetschalckx K, Hoymans VY, et al. Aerobic interval training and continuous training equally improve aerobic exercise capacity in patients with coronary artery disease: the SAINTEX-CAD study. Int J Cardiol. 2015;179:203–210. doi: 10.1016/j.ijcard.2014.10.155
218.
Kristiansen J, Sjúrðarson T, Grove EL, Rasmussen J, Kristensen SD, Hvas AM, Mohr M. Feasibility and impact of whole-body high-intensity interval training in patients with stable coronary artery disease: a randomised controlled trial. Sci Rep. 2022;12:1–12. doi: 10.1038/s41598-022-21655-w
219.
Ismail H, McFarlane JR, Nojoumian AH, Dieberg G, Smart NA. Clinical outcomes and cardiovascular responses to different exercise training intensities in patients with heart failure. a systematic review and meta-analysis. JACC Heart Fail. 2013;1:514–522. doi: 10.1016/j.jchf.2013.08.006
220.
O’Connor CM, Whellan DJ, Lee KL, Keteyian SJ, Cooper LS, Ellis SJ, Leifer ES, Kraus WE, Kitzman DW, Blumenthal JA, et al; HF-ACTION Investigators. Efficacy and safety of exercise training in patients with chronic heart failure HF-ACTION randomized controlled trial. JAMA. 2009;301:1439–1450. doi: 10.1001/jama.2009.454
221.
Ellingsen O, Halle M, Conraads V, Støylen A, Dalen H, Delagardelle C, Larsen AI, Hole T, Mezzani A, Van Craenenbroeck EM, et al; SMARTEX Heart Failure Study (Study of Myocardial Recovery After Exercise Training in Heart Failure) Group. High-intensity interval training in patients with heart failure with reduced ejection fraction. Circulation. 2017;135:839–849. doi: 10.1161/CIRCULATIONAHA.116.022924
222.
McKelvie RS, Teo KK, Roberts R, McCartney N, Humen D, Montague T, Hendrican K, Yusuf S. Effects of exercise training in patients with heart failure: the Exercise Rehabilitation Trial (EXERT). Am Heart J. 2002;144:23–30. doi: 10.1067/mhj.2002.123310
223.
Pandey A, Parashar A, Kumbhani DJ, Agarwal S, Garg J, Kitzman D, Levine BD, Drazner M, Berry JD. Exercise training in patients with heart failure and preserved ejection fraction meta-analysis of randomized control trials. Circ Heart Fail. 2015;8:33–40. doi: 10.1161/CIRCHEARTFAILURE.114.001615
224.
Kitzman DW, Brubaker P, Morgan T, Haykowsky M, Hundley G, Kraus WE, Eggebeen J, Nicklas BJ. Effect of caloric restriction or aerobic exercise training on peak oxygen consumption and quality of life in obese older patients with heart failure with preserved ejection fraction: a randomized clinical trial. JAMA. 2016;315:36–46. doi: 10.1001/jama.2015.17346
225.
Edelmann F, Gelbrich G, Dngen HD, Fröhling S, Wachter R, Stahrenberg R, Binder L, Töpper A, Lashki DJ, Schwarz S, et al. Exercise training improves exercise capacity and diastolic function in patients with heart failure with preserved ejection fraction: results of the Ex-DHF (exercise training in diastolic heart failure) pilot study. J Am Coll Cardiol. 2011;58:1780–1791. doi: 10.1016/j.jacc.2011.06.054
226.
Kitzman DW, Brubaker PH, Morgan TM, Stewart KP, Little WC. Exercise training in older patients with Heart failure and preserved ejection fraction: a randomized, controlled, single-blind trial. Circ Heart Fail. 2010;3:659–667. doi: 10.1161/CIRCHEARTFAILURE.110.958785
227.
Brubaker PH, Nicklas BJ, Houston DK, Hundley WG, Chen H, Molina AJA, Lyles WM, Nelson B, Upadhya B, Newland R, et al. A randomized, controlled trial of resistance training added to caloric restriction plus aerobic exercise training in obese heart failure with preserved ejection fraction. Circ Heart Fail. 2023;16:e010161–E010161. doi: 10.1161/CIRCHEARTFAILURE.122.010161
228.
Sachdev V, Sharma K, Keteyian SJ, Alcain CF, Desvigne-Nickens P, Fleg JL, Florea VG, Franklin BA, Guglin M, Halle M, et al; American Heart Association Heart Failure and Transplantation Committee of the Council on Clinical Cardiology. Supervised exercise training for chronic heart failure with preserved ejection fraction: a scientific statement from the American Heart Association and American College of Cardiology. J Am Coll Cardiol. 2023;81:1524–1542. doi: 10.1016/j.jacc.2023.02.012
229.
van Tol BAF, Huijsmans RJ, Kroon DW, Schothorst M, Kwakkel G. Effects of exercise training on cardiac performance, exercise capacity and quality of life in patients with heart failure: a meta-analysis. Eur J Heart Fail. 2006;8:841–850. doi: 10.1016/j.ejheart.2006.02.013
230.
Rees K, Taylor RRS, Singh S, Coats AJS, Ebrahim S. Exercise based rehabilitation for heart failure. Cochrane Database Syst Rev. 2004;(3):CD003331. doi: 10.1002/14651858.CD003331.pub2
231.
Taylor RS, Walker S, Ciani O, Warren F, Smart NA, Piepoli M, Davos CH. Exercise-based cardiac rehabilitation for chronic heart failure: the EXTRAMATCH II individual participant data meta-analysis. Health Technol Assess. 2019;23:1–98. doi: 10.3310/hta23250
232.
Smart N, Marwick TH. Exercise training for patients with heart failure: a systematic review of factors that improve mortality and morbidity. Am J Med. 2004;116:693–706. doi: 10.1016/j.amjmed.2003.11.033
233.
Baral R, Ho JSY, Soroya AN, Hanger M, Clarke RE, Memon SF, Glatzel H, Ahmad M, Providencia R, Bray JJH, et al. Exercise training improves exercise capacity and quality of life in heart failure with preserved ejection fraction: a systematic review and meta-analysis of randomized controlled trials. European Heart Journal Open. 2024;4:1–9. doi: 10.1093/ehjopen/oeae033
234.
Wewege MA, Thom JM, Rye K-A, Parmenter BJ. Aerobic, resistance or combined training: A systematic review and meta-analysis of exercise to reduce cardiovascular risk in adults with metabolic syndrome. Atherosclerosis. 2018;274:162–171. doi: 10.1016/j.atherosclerosis.2018.05.002
235.
Pedersen BK, Saltin B. Exercise as medicine - evidence for prescribing exercise as therapy in 26 different chronic diseases. Scand J Med Sci Sports. 2015;25:1–72. doi: 10.1111/sms.12581
236.
Cornelissen VA, Smart NA, Survey NE. Exercise training for blood pressure: a systematic review and meta- analysis. J Am Heart Assoc. 2013;2:e004473.
237.
Sigal RJ, Kenny GP, Boulé NG, Wells GA, Prud’homme D, Fortier M, Reid RD, Tulloch H, Coyle D, Phillips P, et al. Effects of aerobic training, resistance training, or both on glycemic control in type 2 diabetes: a randomized trial. Ann Intern Med. 2007;147:357–369. doi: 10.7326/0003-4819-147-6-200709180-00005.
238.
Boule NG, Haddad E, Kenny GP, Wells GA, Sigal RJ. Effects of exercise on glycemic control and body mass in type 2 diabetes mellitus a meta-analysis of controlled clinical trials. JAMA. 2001;286:11–15. doi: 10.1001/jama.286.10.1218
239.
Huang L, Fang Y, Tang L. Comparisons of different exercise interventions on glycemic control and insulin resistance in prediabetes: a network. BMC Endocr Disord. 2021;21:181. doi: 10.1186/s12902-021-00846-y
240.
Kraus WE, Houmard JA, Duscha BD, Knetzger KJ, Wharton MB, McCartney JS, Bales CW, Henes S, Samsa GP, Otvos JD, et al. Effects of the amount and intensity of exercise on plasma lipoproteins. N Engl J Med. 2002;347:1483–1492. doi: 10.1056/nejmoa020194
241.
Kodama S, Tanaka S, Saito K, Shu M, Sone Y, Onitake F, Suzuki E, Shimano H, Yamamoto S, Kondo K, et al. Effect of aerobic exercise training on serum levels of high-density lipoprotein cholesterol. Arch Intern Med. 2007;167:999–1008. doi: 10.1001/archinte.167.10.999
242.
Lee DC, Brellenthin AG, Lanningham-Foster LM, Kohut ML, Li Y. Aerobic, resistance, or combined exercise training and cardiovascular risk profile in overweight or obese adults: the CardioRACE trial. Eur Heart J. 2024;45:1127–1142. doi: 10.1093/eurheartj/ehad827
243.
Thorogood A, Mottillo S, Shimony A, Filion KB, Joseph L, Genest J, Pilote L, Poirier P, Schiffrin EL, Eisenberg MJ. Isolated aerobic exercise and weight loss: a systematic review and meta-analysis of randomized controlled trials. Am J Med. 2011;124:747–755. doi: 10.1016/j.amjmed.2011.02.037
244.
Thomas DE, Elliott EJ, Naughton GA. Exercise for type 2 diabetes mellitus (review). Cochrane Database Syst Rev. 2006;2006:CD002968. doi: 10.1002/14651858.CD002968.pub2
245.
Ismail I, Keating SE, Baker MK, Johnson NA. A systematic review and meta-analysis of the effect of aerobic vs. resistance exercise training on visceral fat. Obes Rev. 2011;13:68–91. doi: 10.1111/j.1467-789x.2011.00931.x
246.
Nishijima H, Satake K, Igarashi K, Morita N, Kanazawa N, Okita K. Effects of exercise in overweight Japanese with multiple cardiovascular risk factors. Med Sci Sports Exerc. 2007;39:926–933. doi: 10.1249/mss.0b013e3180383d84
247.
Collins KA, Fos LB, Ross LM, Slentz CA, Davis PG, Willis LH, Piner LW, Bateman LA, Houmard JA, Kraus WE. Aerobic, resistance, and combination training on health-related quality of life: the STRRIDE-AT/RT randomized trial. Front Sports Act Living. 2021;2:1–10. doi: 10.3389/fspor.2020.620300
248.
La Gerche A, Burns AT, Taylor AJ, Macisaac AI, Heidbuchel H, Prior DL. Maximal oxygen consumption is best predicted by measures of cardiac size rather than function in healthy adults. Eur J Appl Physiol. 2012;112:2139–2147. doi: 10.1007/s00421-011-2184-9
249.
Steding K, Engblom H, Buhre T, Carlsson M, Mosen H, Wohlfart B, Arheden H. Relation between cardiac dimensions and peak oxygen uptake. J Cardiovasc Magn Reson. 2010;12:8. doi: 10.1186/1532-429X-12-8
250.
Scharhag J, Schneider G, Urhausen A, Rochette V, Kramann B, Kindermann W. Athlete’s heart: right and left ventricular mass and function in male endurance athletes and untrained individuals determined by magnetic resonance imaging. J Am Coll Cardiol. 2002;40:1856–1863. doi: 10.1016/s0735-1097(02)02478-6
251.
Levine BD. VO2max: What do we know, and what do we still need to know? J Physiol. 2008;586:25–34. doi: 10.1113/jphysiol.2007.147629
252.
Bhella PS, Hastings JL, Fujimoto N, Shibata S, Carrick-Ranson G, Palmer MD, Boyd KN, Adams-Huet B, Levine BD. Impact of lifelong exercise “dose” on left ventricular compliance and distensibility. J Am Coll Cardiol. 2014;64:1257–1266. doi: 10.1016/j.jacc.2014.03.062
253.
Fujimoto N, Prasad A, Hastings JL, Arbab-Zadeh A, Bhella PS, Shibata S, Palmer D, Levine BD. Cardiovascular effects of 1 year of progressive and vigorous exercise training in previously sedentary individuals older than 65 years of age. Circulation. 2010;122:1797–1805. doi: 10.1161/CIRCULATIONAHA.110.973784
254.
Fujimoto N, Prasad A, Hastings JL, Bhella PS, Shibata S, Palmer D, Levine BD. Cardiovascular effects of 1 year of progressive endurance exercise training in patients with heart failure with preserved ejection fraction. Am Heart J. 2012;164:869–877. doi: 10.1016/j.ahj.2012.06.028
255.
Howden EJ, Sarma S, Lawley JS, Opondo M, Cornwell W, Stoller D, Urey MA, Adams-Huet B, Levine BD. Reversing the cardiac effects of sedentary aging in middle age-A randomized controlled trial: implications for heart failure prevention. Circulation. 2018;137:1549–1560. doi: 10.1161/CIRCULATIONAHA.117.030617
256.
Hieda M, Sarma S, Hearon CM, MacNamara JP, Dias KA, Samels M, Palmer D, Livingston S, Morris M, Levine BD. One-year committed exercise training reverses abnormal left ventricular myocardial stiffness in patients with stage B heart failure with preserved ejection fraction. Circulation. 2021;144:934–946. doi: 10.1161/CIRCULATIONAHA.121.054117
257.
Melov S, Tarnopolsky MA, Beckman K, Felkey K, Hubbard A. Resistance exercise reverses aging in human skeletal muscle. PLoS One. 2007;2:e465. doi: 10.1371/journal.pone.0000465
258.
Laurin JL, Reid JJ, Lawrence MM, Miller BF. Long-term aerobic exercise preserves muscle mass and function with age. Curr Opin Physiol. 2019;10:70–74. doi: 10.1016/j.cophys.2019.04.019
259.
Mortensen SP, Nyberg M, Winding K, Saltin B. Lifelong physical activity preserves functional sympatholysis and purinergic signalling in the ageing human leg. J Physiol. 2012;590:6227–6236. doi: 10.1113/jphysiol.2012.240093
260.
Mizuno M, Iwamoto GA, Vongpatanasin W, Mitchell JH, Smith SA. Exercise training improves functional sympatholysis in spontaneously hypertensive rats through a nitric oxide-dependent mechanism. Am J Physiol Heart Circ Physiol. 2014;307:H242–H251. doi: 10.1152/ajpheart.00103.2014
261.
Distefano G, Standley RA, Zhang X, Carnero EA, Yi F, Cornnell HH, Coen PM. Physical activity unveils the relationship between mitochondrial energetics, muscle quality, and physical function in older adults. J Cachexia Sarcopenia Muscle. 2018;9:279–294. doi: 10.1002/jcsm.12272
262.
Coggan AR, Spina RJ, King DS, Rogers MA, Brown M, Nemeth PM, Holloszy JO. Skeletal muscle adaptations to endurance training in 60- to 70-yr-old men and women. J Appl Physiol (1985). 1992;72:1780–1786. doi: 10.1152/jappl.1992.72.5.1780
263.
Robinson MM, Dasari S, Konopka AR, Johnson ML, Manjunatha S, Esponda RR, Carter RE, Lanza IR, Nair KS. Enhanced protein translation underlies improved metabolic and physical adaptations to different exercise training modes in young and old humans. Cell Metab. 2017;25:581–592. doi: 10.1016/j.cmet.2017.02.009
264.
Wyckelsma VL, Levinger I, McKenna MJ, Formosa LE, Ryan MT, Petersen AC, Anderson MJ, Murphy RM. Preservation of skeletal muscle mitochondrial content in older adults: relationship between mitochondria, fibre type and high-intensity exercise training. J Physiol. 2017;595:3345–3359. doi: 10.1113/JP273950
265.
Charles M, Charifi N, Verney J, Pichot V, Feasson L, Costes F, Denis C. Effect of endurance training on muscle microvascular filtration capacity and vascular bed morphometry in the elderly. Acta Physiol (Oxf). 2006;187:399–406. doi: 10.1111/j.1748-1716.2006.01585.x
266.
Hepple RT, Mackinnon SL, Thomas SG, Goodman JM, Plyley MJ. Quantitating the capillary supply and the response to resistance training in older men. Pflugers Arch. 1997;433:238–244. doi: 10.1007/s004240050273
267.
Haykowsky MJ, Brubaker PH, Stewart KP, Morgan TM, Eggebeen J, Kitzman DW. Effect of endurance training on the determinants of peak exercise oxygen consumption in elderly patients with stable compensated heart failure and preserved ejection fraction. J Am Coll Cardiol. 2012;60:120–128. doi: 10.1016/j.jacc.2012.02.055
268.
Pandey A, Parashar A, Kumbhani D, Agarwal S, Garg J, Kitzman D, Levine B, Drazner M, Berry J. Exercise training in patients with heart failure and preserved ejection fraction: meta-analysis of randomized control trials. Circ Heart Fail. 2015;8:33–40. doi: 10.1161/CIRCHEARTFAILURE.114.001615
269.
Winzer EB, Augstein A, Schauer A, Mueller S, Fischer-Schaepmann T, Goto K, Hommel J, van Craenenbroeck EM, Wisloff U, Pieske B, et al. Impact of different training modalities on molecular alterations in skeletal muscle of patients with heart failure with preserved ejection fraction: a substudy of the OptimEx trial. Circ Heart Fail. 2022;15:e009124. doi: 10.1161/CIRCHEARTFAILURE.121.009124
270.
Chow LS, Gerszten RE, Taylor JM, Pedersen BK, van Praag H, Trappe S, Febbraio MA, Galis ZS, Gao Y, Haus JM, et al. Exerkines in health, resilience and disease. Nat Rev Endocrinol. 2022;18:273–289. doi: 10.1038/s41574-022-00641-2
271.
Amar D, Gay NR, Jimenez-Morales D, Jean Beltran PM, Ramaker ME, Raja AN, Zhao B, Sun Y, Marwaha S, Gaul DA, et al; MoTrPAC Study Group. The mitochondrial multi-omic response to exercise training across rat tissues. Cell Metab. 2024;36:1411–1429.e10. doi: 10.1016/j.cmet.2023.12.021
272.
Vetr NG, Gay NR, MoTr PACSG, Montgomery SB. The impact of exercise on gene regulation in association with complex trait genetics. Nat Commun. 2024;15:3346. doi: 10.1038/s41467-024-45966-w
273.
MoTr PACSG, Lead Analyst, MoTr PACSG. Temporal dynamics of the multi-omic response to endurance exercise training. Nature. 2024;629:174–183. doi: 10.1038/s41586-023-06877-w
274.
Stensvold D, Viken H, Steinshamn SL, Dalen H, Støylen A, Loennechen JP, Reitlo LS, Zisko N, Bækkerud FH, Tari AR, et al. Effect of exercise training for five years on all cause mortality in older adults-the generation 100 study: randomised controlled trial. BMJ. 2020;371:m3485. doi: 10.1136/bmj.m3485
275.
Letnes JM, Berglund I, Johnson KE, Dalen H, Nes BM, Lydersen S, Viken H, Hassel E, Steinshamn S, Vesterbekkmo EK, et al. Effect of 5 years of exercise training on the cardiovascular risk profile of older adults: the generation 100 randomized trial. Eur Heart J. 2022;43:2065–2075. doi: 10.1093/eurheartj/ehab721
276.
Pahor M, Guralnik JM, Ambrosius WT, Blair S, Bonds DE, Church TS, Espeland MA, Fielding RA, Gill TM, Groessl EJ, et al; LIFE study investigators. Effect of structured physical activity on prevention of major mobility disability in older adults: the LIFE study randomized clinical trial. JAMA. 2014;311:2387–2396. doi: 10.1001/jama.2014.5616
277.
Newman AB, Dodson JA, Church TS, Buford TW, Fielding RA, Kritchevsky S, Beavers D, Pahor M, Stafford RS, Szady AD, et al; LIFE Study Group. Cardiovascular events in a physical activity intervention compared with a successful aging intervention: the LIFE study randomized trial. JAMA Cardiol. 2016;1:568–574. doi: 10.1001/jamacardio.2016.1324
278.
Look ARG, Wing RR, Bolin P, Brancati FL, Bray GA, Clark JM, Coday M, Crow RS, Curtis JM, Egan CM, et al. Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. N Engl J Med. 2013;369:145–154. doi: 10.1056/NEJMoa1212914
279.
Keteyian SJ, Kraus WE, Brawner CA, Connor CMO, Whellan DJ, Cooper LS, Fleg JL, Kitzman DW, Cohen-solal A, Blumenthal JA, et al. Relation between volume of exercise and clinical outcomes in patients with heart failure. JACC. 2012;60:1899–1905. doi: 10.1016/j.jacc.2012.08.958
280.
Belardinelli R, Georgiou D, Cianci G, Purcaro A. 10-year exercise training in chronic heart failure: a randomized controlled trial. J Am Coll Cardiol. 2012;60:1521–1528. doi: 10.1016/j.jacc.2012.06.036
281.
Molloy CD, Long L, Mordi IR, Bridges C, Sagar VA, Davies EJ, Coats AJS, Dalal H, Rees K, Singh SJ, et al. Exercise-based cardiac rehabilitation for adults with heart failure – 2023 Cochrane systematic review and meta-analysis. Eur J Heart Fail. 2023;25:2263–2273. doi: 10.1002/ejhf.3046
282.
Pandey A, Kitzman DW, Brubaker P, Haykowsky MJ, Morgan T, Becton JT, Berry JD. Response to endurance exercise training in older adults with heart failure with preserved or reduced ejection fraction. J Am Geriatr Soc. 2017;65:1698–1704. doi: 10.1111/jgs.14867
283.
Mentz RJ, Whellan DJ, Reeves GR, Pastva AM, Duncan P, Upadhya B, Nelson MB, Chen H, Reed SD, Rosenberg PB, et al. Rehabilitation intervention in older patients with acute heart failure with preserved versus reduced ejection fraction. JACC Heart Fail. 2021;9:747–757. doi: 10.1016/j.jchf.2021.05.007
284.
Al Hennawi H, Bedi A, Khan MK, Zohaib M, Khan IA, Mazzoni JA. Impact of exercise training on clinical outcomes and quality of life in chronic congestive heart failure: a systematic review and meta-analysis: impact of exercise-based cardiac rehabilitation on heart failure. Curr Probl Cardiol. 2024;49:102756–102756. doi: 10.1016/j.cpcardiol.2024.102756
285.
Mueller S, Winzer EB, Duvinage A, Gevaert AB, Edelmann F, Haller B, Pieske-Kraigher E, Beckers P, Bobenko A, Hommel J, et al; OptimEx-Clin Study Group. Effect of high-intensity interval training, moderate continuous training, or guideline-based physical activity advice on peak oxygen consumption in patients with heart failure with preserved ejection fraction: a randomized clinical trial. JAMA. 2021;325:542–551. doi: 10.1001/jama.2020.26812
286.
Mueller S, Kabelac M, Fegers-Wustrow I, Winzer EB, Gevaert AB, Beckers P, Haller B, Edelmann F, Christle JW, Haykowsky MJ, et al. Comparison of exercise training modalities and change in peak oxygen consumption in heart failure with preserved ejection fraction: a secondary analysis of the OptimEx-Clin trial. Eur J Prev Cardiol. 2024. doi: 10.1093/eurjpc/zwae332
287.
Walters GWM, Yeo JL, Bilak JM, Pepper C, Gulsin GS, Freeman SC, Gray LJ, McCann GP, Brady EM. The effectiveness of lifestyle interventions in heart failure with preserved ejection fraction: a systematic review and network meta-analysis. J Card Fail. 2024;30:994–1009. doi: 10.1016/j.cardfail.2024.01.015
288.
Siddiqi TJ, Rashid AM, Javaid SS, Siddiqi AK, Usman MS, Hervir O, Kamimura D, Lavie CJ, Mentz RJ, Butler J, et al. High-intensity interval training versus moderate continuous training in patients with heart failure with preserved ejection fraction: a systematic review and meta-analysis. Curr Probl Cardiol. 2023;48:101720–101720. doi: 10.1016/j.cpcardiol.2023.101720
289.
Donelli da Silveira A, Beust de Lima J, da Silva Piardi D, dos Santos Macedo D, Zanini M, Nery R, Laukkanen JA, Stein R. High-intensity interval training is effective and superior to moderate continuous training in patients with heart failure with preserved ejection fraction: a randomized clinical trial. Eur J Prev Cardiol. 2020;27:1733–1743. doi: 10.1177/2047487319901206
290.
Angadi SS, Mookadam F, Lee CD, Tucker WJ, Haykowsky MJ, Gaesser GA. High-intensity interval training vs. moderate-intensity continuous exercise training in heart failure with preserved ejection fraction: a pilot study. J Appl Physiol (1985). 2015;119:753–758. doi: 10.1152/japplphysiol.00518.2014
291.
Sharif SM, Smith JR, Bruhn EJ, Shultz AM, Olson TP. Impact of structured resistance training on locomotor muscle composition in patients with HFpEF. J Card Fail. 2024;30:410–414. doi: 10.1016/j.cardfail.2023.10.475
292.
Coppo R. Is a legacy effect possible in IgA nephropathy? Nephrol Dial Transplant. 2013;28:1657–1662. doi: 10.1093/ndt/gft016
293.
Johnson JL, Slentz CA, Ross LM, Huffman KM, Kraus WE. Ten-year legacy effects of three eight-month exercise training programs on cardiometabolic health parameters. Front Physiol. 2019;10:1–9. doi: 10.3389/fphys.2019.00452
294.
Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HAW. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med. 2008;359:1577–1589. doi: 10.1056/NEJMoa0806470
295.
Sever PS, Chang CL, Gupta AK, Whitehouse A, Poulter NR; ASCOT Investigators. The Anglo-Scandinavian cardiac outcomes trial: 11-year mortality follow-up of the lipid-lowering arm in the UK. Eur Heart J. 2011;32:2525–2532. doi: 10.1093/eurheartj/ehr333
296.
Li G, Zhang P, Wang J, Gregg EW, Yang W, Gong Q, Li H, Li H, Jiang Y, An Y, et al. The long-term effect of lifestyle interventions to prevent diabetes in the China Da Qing Diabetes Prevention Study: a 20-year follow-up study. Lancet. 2008;371:1783–1789. doi: 10.1016/S0140-6736(08)60766-7
297.
Kashef MA, Giugliano G. Legacy effect of statins: 20-year follow up of the West of Scotland Coronary Prevention Study (WOSCOPS). Glob Cardiol Sci Pract. 2016;2016:e201635. doi: 10.21542/gcsp.2016.35
298.
Blumenthal JA, Babyak MA, O’Connor C, Keteyian S, Landzberg J, Howlett J, Kraus W, Gottlieb S, Blackburn G, Swank A, et al. Effects of exercise training on depressive symptoms in patients with chronic heart failure: the HF-ACTION randomized trial. JAMA. 2012;308:465–474. doi: 10.1001/jama.2012.8720
299.
Flynn KE, Piña IL, Whellan DJ, Lin L, Blumenthal JA, Ellis SJ, Fine LJ, Howlett G, Keteyian SJ, Kitzman DW, et al. Effects of exercise training on health status in patients with chronic heart failure: HF-ACTION randomized controlled trial. JAMA. 2009;301:1451–1459. doi: 10.1001/jama.2009.457
300.
Keshvani N, Subramanian V, Wrobel CA, Solomon N, Alhanti B, Greene SJ, DeVore AD, Yancy CW, Allen LA, Fonarow GC, et al. Patterns of referral and postdischarge utilization of cardiac rehabilitation among patients hospitalized with heart failure: an analysis from the GWTG-HF registry. Circ Heart Fail. 2023;16:e010144. doi: 10.1161/CIRCHEARTFAILURE.122.010144
301.
Golwala H, Pandey A, Ju C, Butler J, Yancy C, Bhatt DL, Hernandez AF, Fonarow GC. Temporal trends and factors associated with cardiac rehabilitation referral among patients hospitalized with heart failure: findings from get with the guidelines-heart failure registry. J Am Coll Cardiol. 2015;66:917–926. doi: 10.1016/j.jacc.2015.06.1089
302.
Pandey A, Kitzman DW, Nelson MB, Pastva AM, Duncan P, Whellan DJ, Mentz RJ, Chen H, Upadhya B, Reeves GR. Frailty and effects of a multidomain physical rehabilitation intervention among older patients hospitalized for acute heart failure: a secondary analysis of a randomized clinical trial. JAMA Cardiol. 2023;8:167–176. doi: 10.1001/jamacardio.2022.4903
303.
Pandey A, Segar MW, Singh S, Reeves GR, O’Connor C, Pina I, Whellan D, Kraus WE, Mentz RJ, Kitzman DW. Frailty status modifies the efficacy of exercise training among patients with chronic heart failure and reduced ejection fraction: an analysis from the HF-ACTION trial. Circulation. 2022;146:80–90. doi: 10.1161/CIRCULATIONAHA.122.059983
304.
Subramanian V, Betts T, Segar MW, Patel L, Keshvani N, Chandra A, Pandey A. m-health based cardiac rehabilitation program for management of older patients with HFpEF: a pilot RCT. JACC Heart Fail. 2025;13:653–656. doi: 10.1016/j.jchf.2024.10.024
305.
Schmidt C, Magalhaes S, Gois Basilio P, Santos C, Oliveira MI, Ferreira JP, Ribeiro F, Santos M. Center- vs home-based cardiac rehabilitation in patients with heart failure: EXIT-HF randomized controlled trial. JACC Heart Fail. 2025;13:695. doi: 10.1016/j.jchf.2024.09.024
306.
Dalal HM, Taylor RS, Jolly K, Davis RC, Doherty P, Miles J, van Lingen R, Warren FC, Green C, Wingham J, et al. The effects and costs of home-based rehabilitation for heart failure with reduced ejection fraction: the REACH-HF multicentre randomized controlled trial. Eur J Prev Cardiol. 2019;26:262–272. doi: 10.1177/2047487318806358
307.
Maddison R, Rawstorn JC, Stewart RAH, Benatar J, Whittaker R, Rolleston A, Jiang Y, Gao L, Moodie M, Warren I, et al. Effects and costs of real-time cardiac telerehabilitation: randomised controlled non-inferiority trial. Heart. 2019;105:122–129. doi: 10.1136/heartjnl-2018-313189
308.
Kitzman DW, Whellan DJ, Duncan P, Pastva AM, Mentz RJ, Reeves GR, Nelson MB, Chen H, Upadhya B, Reed SD, et al. Physical rehabilitation for older patients hospitalized for heart failure. N Engl J Med. 2021;385:203–216. doi: 10.1056/NEJMoa2026141
309.
Khodadad Kashi S, Mirzazadeh ZS, Saatchian V. A systematic review and meta-analysis of resistance training on quality of life, depression, muscle strength, and functional exercise capacity in older adults aged 60 years or more. Biol Res Nurs. 2023;25:88–106. doi: 10.1177/10998004221120945
310.
Rocha JNS, Pereira-Monteiro MR, Vasconcelos ABS, Pantoja-Cardoso A, Aragao-Santos JC, Da Silva-Grigoletto ME. Different resistance training volumes on strength, functional fitness, and body composition of older people: a systematic review with meta-analysis. Arch Gerontol Geriatr. 2024;119:105303. doi: 10.1016/j.archger.2023.105303
311.
Sachdev V, Sharma K, Keteyian SJ, Alcain CF, Desvigne-Nickens P, Fleg JL, Florea VG, Franklin BA, Guglin M, Halle M, et al; American Heart Association Heart Failure and Transplantation Committee of the Council on Clinical Cardiology; Council on Arteriosclerosis, Thrombosis and Vascular Biology; and American College of Cardiology. Supervised exercise training for chronic heart failure with preserved ejection fraction: a scientific statement from the American Heart Association and American College of Cardiology. Circulation. 2023;147:e699–e715. doi: 10.1161/CIR.0000000000001122
312.
Lavie CJ, Arena R, Swift DL, Johannsen NM, Sui X, Lee DC, Earnest CP, Church TS, O’Keefe JH, Milani RV, et al. Exercise and the cardiovascular system: clinical science and cardiovascular outcomes. Circ Res. 2015;117:207–219. doi: 10.1161/CIRCRESAHA.117.305205
313.
Sano S, Horitani K, Ogawa H, Halvardson J, Chavkin NW, Wang Y, Sano M, Mattisson J, Hata A, Danielsson M, et al. Hematopoietic loss of Y chromosome leads to cardiac fibrosis and heart failure mortality. Science. 2022;377:292–297. doi: 10.1126/science.abn3100
314.
Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG, Shvartz E, McConkey M, Gupta N, Gabriel S, Ardissino D, et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N Engl J Med. 2017;377:111–121. doi: 10.1056/NEJMoa1701719
315.
Yu B, Roberts MB, Raffield LM, Zekavat SM, Nguyen NQH, Biggs ML, Brown MR, Griffin G, Desai P, Correa A, et al; National Heart, Lung, and Blood Institute TOPMed Consortium. Supplemental association of clonal hematopoiesis with incident heart failure. J Am Coll Cardiol. 2021;78:42–52. doi: 10.1016/j.jacc.2021.04.085
316.
Schuermans A, Honigberg MC, Raffield LM, Yu B, Roberts MB, Kooperberg C, Desai P, Carson AP, Shah AM, Ballantyne CM, et al. Clonal hematopoiesis and incident heart failure with preserved ejection fraction. JAMA Netw Open. 2024;7:e2353244. doi: 10.1001/jamanetworkopen.2023.53244
317.
Park HW, Ahn Y, Jeong MH, Cho JG, Park JC, Kang JC, Shin MG, Shin JH, Suh SP, Ryang DW, et al. Chronic atrial fibrillation associated with somatic mitochondrial DNA mutations in human atrial tissue. J Clin Pathol. 2007;60:948–950. doi: 10.1136/jcp.2007.047134
318.
Attia SM, Helal GK, Alhaider AA. Assessment of genomic instability in normal and diabetic rats treated with metformin. Chem Biol Interact. 2009;180:296–304. doi: 10.1016/j.cbi.2009.03.001
319.
Piskovatska V, Stefanyshyn N, Storey KB, Vaiserman AM, Lushchak O. Metformin as a geroprotector: experimental and clinical evidence. Biogerontology. 2019;20:33–48. doi: 10.1007/s10522-018-9773-5
320.
Pathak GA, Wendt FR, De Lillo A, Nunez YZ, Goswami A, De Angelis F, Fuciarelli M, Kranzler HR, Gelernter J, Polimanti R. Epigenomic profiles of African-American transthyretin Val122Ile carriers reveals putatively dysregulated amyloid mechanisms. Circ Genom Precis Med. 2021;14:e003011. doi: 10.1161/CIRCGEN.120.003011
321.
De Lillo A, Pathak GA, De Angelis F, Di Girolamo M, Luigetti M, Sabatelli M, Perfetto F, Frusconi S, Manfellotto D, Fuciarelli M, et al. Epigenetic profiling of Italian patients identified methylation sites associated with hereditary transthyretin amyloidosis. Clin Epigenetics. 2020;12:176. doi: 10.1186/s13148-020-00967-6
322.
Yang M, Chao K, Wang Z, Xue R, Zhang X, Wang D. Accelerated biological aging and risk of atrial fibrillation: a cohort study. Heart Rhythm. 2024. doi: 10.1016/j.hrthm.2024.11.017
323.
Wallner M, Eaton DM, Berretta RM, Liesinger L, Schittmayer M, Gindlhuber J, Wu J, Jeong MY, Lin YH, Borghetti G, et al. HDAC inhibition improves cardiopulmonary function in a feline model of diastolic dysfunction. Sci Transl Med. 2020;12:eaay7205. doi: 10.1126/scitranslmed.aay7205
324.
Travers JG, Wennersten SA, Pena B, Bagchi RA, Smith HE, Hirsch RA, Vanderlinden LA, Lin YH, Dobrinskikh E, Demos-Davies KM, et al. HDAC inhibition reverses preexisting diastolic dysfunction and blocks covert extracellular matrix remodeling. Circulation. 2021;143:1874–1890. doi: 10.1161/CIRCULATIONAHA.120.046462
325.
Mesgarzadeh JS, Romine IC, Smith-Cohen EM, Grandjean JMD, Kelly JW, Genereux JC, Wiseman RL. ATF6 activation reduces amyloidogenic transthyretin secretion through increased interactions with endoplasmic reticulum proteostasis factors. Cells. 2022;11:1661. doi: 10.3390/cells11101661
326.
Kassiotis C, Ballal K, Wellnitz K, Vela D, Gong M, Salazar R, Frazier OH, Taegtmeyer H. Markers of autophagy are downregulated in failing human heart after mechanical unloading. Circulation. 2009;120:S191–S197. doi: 10.1161/CIRCULATIONAHA.108.842252
327.
Matai L, Sarkar GC, Chamoli M, Malik Y, Kumar SS, Rautela U, Jana NR, Chakraborty K, Mukhopadhyay A. Dietary restriction improves proteostasis and increases life span through endoplasmic reticulum hormesis. Proc Natl Acad Sci U S A. 2019;116:17383–17392. doi: 10.1073/pnas.1900055116
328.
Bulawa CE, Connelly S, Devit M, Wang L, Weigel C, Fleming JA, Packman J, Powers ET, Wiseman RL, Foss TR, et al. Tafamidis, a potent and selective transthyretin kinetic stabilizer that inhibits the amyloid cascade. Proc Natl Acad Sci U S A. 2012;109:9629–9634. doi: 10.1073/pnas.1121005109
329.
Wolff CA, Reid JJ, Musci RV, Linden MA, Konopka AR, Peelor FF, Miller BF, Hamilton KL, Bruns DR. Differential effects of rapamycin and metformin in combination with rapamycin on mechanisms of proteostasis in cultured skeletal myotubes. J Gerontol A Biol Sci Med Sci. 2020;75:32–39. doi: 10.1093/gerona/glz058
330.
Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, Taniike M, Omiya S, Mizote I, Matsumura Y, Asahi M, et al. The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress. Nat Med. 2007;13:619–624. doi: 10.1038/nm1574
331.
Zhu H, Tannous P, Johnstone JL, Kong Y, Shelton JM, Richardson JA, Le V, Levine B, Rothermel BA, Hill JA. Cardiac autophagy is a maladaptive response to hemodynamic stress. J Clin Invest. 2007;117:1782–1793. doi: 10.1172/jci27523
332.
Castoldi F, Humeau J, Martins I, Lachkar S, Loew D, Dingli F, Durand S, Enot D, Bossut N, Chery A, et al. Autophagy-mediated metabolic effects of aspirin. Cell Death Discov. 2020;6:129. doi: 10.1038/s41420-020-00365-0
333.
Elmorsy EA, Saber S, Hamad RS, Abdel-Reheim MA, El-Kott AF, AlShehri MA, Morsy K, Negm S, Youssef ME. Mechanistic insights into carvedilol’s potential protection against doxorubicin-induced cardiotoxicity. Eur J Pharm Sci. 2024;200:106849. doi: 10.1016/j.ejps.2024.106849
334.
Madeo F, Bauer MA, Carmona-Gutierrez D, Kroemer GS. Spermidine: a physiological autophagy inducer acting as an anti-aging vitamin in humans? Autophagy. 2019;15:165–168. doi: 10.1080/15548627.2018.1530929
335.
Schiattarella GG, Hill JA. Therapeutic targeting of autophagy in cardiovascular disease. J Mol Cell Cardiol. 2016;95:86–93. doi: 10.1016/j.yjmcc.2015.11.019
336.
Kulkarni AS, Gubbi S, Barzilai N. Benefits of metformin in attenuating the hallmarks of aging. Cell Metab. 2020;32:15–30. doi: 10.1016/j.cmet.2020.04.001
337.
Sabbah HN. Targeting the mitochondria in heart failure: a translational perspective. JACC Basic Transl Sci. 2020;5:88–106. doi: 10.1016/j.jacbts.2019.07.009
338.
Goh KY, He L, Song J, Jinno M, Rogers AJ, Sethu P, Halade GV, Rajasekaran NS, Liu X, Prabhu SD, et al. Mitoquinone ameliorates pressure overload-induced cardiac fibrosis and left ventricular dysfunction in mice. Redox Biol. 2019;21:101100. doi: 10.1016/j.redox.2019.101100
339.
Suda M, Katsuumi G, Tchkonia T, Kirkland JL, Minamino T. Potential clinical implications of senotherapies for cardiovascular disease. Circ J. 2024;88:277–284. doi: 10.1253/circj.CJ-23-0657
340.
Chaib S, Tchkonia T, Kirkland JL. Cellular senescence and senolytics: the path to the clinic. Nat Med. 2022;28:1556–1568. doi: 10.1038/s41591-022-01923-y
341.
Ajufo E, Kany S, Ramo JT, Churchill TW, Guseh JS, Aragam KG, Ellinor PT, Khurshid S. Accelerometer-measured sedentary behavior and risk of future cardiovascular disease. J Am Coll Cardiol. 2024;85:473–486. doi: 10.1016/j.jacc.2024.10.065
View full text|Download PDF
查看全文下载 PDF

Comment Response