Skip main navigation

Sedentary Behavior, Exercise, and Cardiovascular Health

Originally publishedhttps://doi.org/10.1161/CIRCRESAHA.118.312669Circulation Research. 2019;124:799–815

    Abstract

    Sedentary behavior and physical inactivity are among the leading modifiable risk factors worldwide for cardiovascular disease and all-cause mortality. The promotion of physical activity and exercise training (ET) leading to improved levels of cardiorespiratory fitness is needed in all age groups, race, and ethnicities and both sexes to prevent many chronic diseases, especially cardiovascular disease. In this state-of-the-art review, we discuss the negative impact of sedentary behavior and physical inactivity, as well as the beneficial effects of physical activity /ET and cardiorespiratory fitness for the prevention of chronic noncommunicable diseases, including cardiovascular disease. We review the prognostic utility of cardiorespiratory fitness compared with obesity and the metabolic syndrome, as well as the increase of physical activity /ET for patients with heart failure as a therapeutic strategy, and ET dosing. Greater efforts at preventing sedentary behavior and physical inactivity while promoting physical activity, ET, and cardiorespiratory fitness are needed throughout the healthcare system worldwide and particularly in the United States in which the burden of cardiometabolic diseases remains extremely high.

    Although the American Heart Association, the American College of Cardiology, and the American College of Sports Medicine, among other leading organizations, have emphasized that sedentary behavior (SB) and physical inactivity (PI) are major modifiable cardiovascular disease (CVD) risk factors, a sizable percentage of the United States and worldwide population still present with high levels of SB/PI and low levels of physical activity (PA).1–3 Recently, a major emphasis has been directed at making health promotion a priority, including the promotion of PA and exercise training (ET) and improving levels of cardiorespiratory fitness (CRF) in the United States and worldwide in efforts to prevent chronic diseases, especially CVD.2,4

    In this article, we review the adverse consequences of SB and PI and the potential benefits of PA/ET on cardiovascular health. We also review the importance of CRF as perhaps one of the most important CVD risk factors, as well as the prognostic utility of fitness compared with obesity and the metabolic syndrome. The potential for ET and improvements in CRF for patients with heart failure (HF) are also reviewed, including the importance of muscular fitness in addition to aerobic fitness. Finally, we conclude by recommending areas in which greater investigative attention is needed.

    SB and CVD

    In addition to the positive cardiovascular health effects associated with increases in moderate and vigorous PA, there is emerging evidence of several negative health consequences associated with SB, which has been defined as any waking behavior characterized by an energy expenditure ≤1.5 metabolic equivalents of task (METs), while in a seated, reclined or lying posture.5 It is important to emphasize that SB is distinct from PI, where an individual does not perform moderate-to-vigorous PA. Although SB and PA are at opposite ends of the energy expenditure continuum,6 the addition of a postural component as a requirement to be considered sedentary suggests that it is a unique behavior that can be intervened on. One can envision the situation where someone is physically active for the recommended 150 to 300 minutes per week,7 yet they may sit for several hours a day in a sedentary occupation or during their leisure time.

    The American Heart Association recently released a Science Advisory that highlighted the deleterious association between SB and CVD morbidity and mortality.8 However, the American Heart Association report stopped short of making specific quantitative recommendations about target levels of SB and reinforced the need for further research that would inform future quantitative public health guidelines, including the need for interventions using randomized controlled trial designs.8 The American Diabetes Association has incorporated SB into their recent Position Statement on PA/ET and diabetes mellitus, recommending that adults should reduce their overall SB and interrupt prolonged bouts of SB with episodes of light-intensity PA.9 Some countries, such as Australia and the United Kingdom, have also begun to release SB guidelines alongside their PA guidelines,10,11 but they do not make specific quantitative recommendations for adults. Rather, they recommend minimizing time spent sitting and breaking up periods of prolonged sitting. Given the differences in sedentary time between self-reported and objectively measured estimates and the lack of a clear threshold of SB that reduces health risks, it is difficult at the present time to provide a quantitative recommendation. Therefore, future studies are needed to use devices that objectively quantify SB to make strides towards identifying critical thresholds associated with increased risk of CVD.

    The health consequences associated with SB were investigated in a series of preclinical studies conducted in the early 2000s.12,13 Using hindlimb suspension (unloading) in a rat model to mimic human SB, a decrease in lipoprotein lipase activity (the enzyme responsible for hydrolysis of triglyceride-rich lipoproteins), triglyceride uptake into red skeletal muscle, and HDL (high-density lipoprotein) cholesterol concentration occurred within a day’s time.14 Further, a global gene-expression profiling study in rats identified 38 genes that were upregulated by SB (hindlimb unloading) and that 27 of these upregulated genes remained above control levels even after the rats returned to standing and ambulation for 4 hours.15 Furthermore, it is well-accepted that elevated levels of oxidative stress results in pervasive systemic impairments. Mitochondrial dysfunction has been recognized as a significant source of oxidative stress. Within the skeletal muscle cells, PGC-1α (proliferator-activated receptor γ coactivator 1-α), a key regulator of mitochondrial mass/function, and NAD-dependent deacetylase SIRT3 (sirtuin-3), which promotes the expression of PGC-1α, have been found to be lower in sedentary individuals.16 Their inverse relation with levels of reactive oxygen species may partly explain damage and mutations to DNA, which contributes to impaired mitochondrial and subsequently skeletal muscle quality and function. Experimental studies that mimicked SB in a laboratory setting have also provided evidence of greater postprandial glucose and insulin levels during bouts of prolonged sitting (ie, 7 hours) compared with individuals taking frequent standing or walking breaks.17 Compared with prolonged sitting, breaking up sitting time with intermittent, light-intensity activity can increase expression of anti-inflammatory and antioxidative pathway modulators such as nicotinamide N-methyltransferase as well as regulators of glucose transporter type 4 translocation.18 Individuals that chronically sit for long periods of time without intermittent activity likely have reduced expression of key metabolic regulators. Taken together, these results indicate that the gross metabolic disturbances observed with SB result from metabolic alterations at the level of the muscle. While these studies suggest some potential mechanisms involved in SB, substantially more research is required to determine the pathophysiological pathways through which SB impacts risk for CVD, and whether these pathways differ from those associated with PI.

    The preclinical work described above was followed by a large number of epidemiological investigations of the associations between SB, such as daily sitting time or television viewing, and several health outcomes. The evidence is strongest for the associations between SB and mortality from CVD and all-causes and weaker for mortality from cancer.19 However, the weaker association between SB and cancer may be explained by the fact that cancer is a highly heterogeneous disease with several different causes and related treatments. For such reasons epidemiological studies investigating the relationship between SB and cancer mortality should be interpreted with caution.

    The first studies to comprehensively address the association between SB and mortality reported robust and consistent results. A study of 17 013 Canadian adults followed for an average of 12 years, reported a significant dose-response association between daily sitting time and both all-cause and CVD mortality.20 Compared with people who reported sitting almost none of the time, those that reported sitting almost all of the time had a 54% higher risk of dying from all-causes or CVD.20 These results were followed closely by a study that investigated the relationship between television viewing and mortality among 8800 Australian adults followed for a median of 6.6 years.21 When compared with those who reported watching television <2 hours per day, individuals watching ≥4 hours per day experienced a 45% and 80% increased risk of all-cause and CVD mortality, respectively. The results of these early studies have been widely replicated and included in recent meta-analyses investigating the association of SB with television viewing22 and sitting.23 Chau et al23 reported summary hazard ratios (HR) of 1.00 (95% CI, 0.98–1.03), 1.02 (95% CI, 0.99–1.05), and 1.05 (95% CI, 1.02–1.08) for every 1-hour increase in sitting between 0 and 3, >3 to 7, and >7 hours of daily sitting, respectively. Similarly, Sun et al22 reported that television viewing was associated with a significant increased risk for all-cause mortality risk in a curvilinear, direct fashion that increased steadily and more rapidly as television viewing time increased.

    A recent meta-analysis investigated the association between SB and incident CVD events using data from 9 prospective cohort studies including 720 425 participants.24 The authors reported a summary HR of 1.14 (95% CI, 1.09–1.19) comparing the highest (12.5 h/d) versus lowest levels (2.5 h/d) sedentary time. They also observed a significant increased risk at >10 h/d of sedentary time (HR=1.08; 95% CI, 1.00–1.14).24 The reported HR of this meta-analysis seems to confirm the increased CVD risk associated with SB, however, the effects of SB may be less pronounced than what was suggested in prior smaller studies.

    Interactions Between SB and PA

    The effects of SB and PA on health outcomes are currently object of intense scrutiny. Several studies have reported that the relative risks associated with sedentary time are higher among people who are not regularly physically active. For example, a meta-analysis of epidemiological studies reported a summary HR associated with SB of 1.46 (95% CI, 1.22–1.75) in those with low levels of PA versus a summary HR of 1.16 (95% CI, 0.84–1.59) in those with high levels of PA.25 In the largest study to date, Ekelund et al26 pooled data on 1 005 791 participants to examine the combined effects of SB and PA on mortality from CVD, cancer, and all-causes, and they demonstrated that moderate-to-vigorous PA was inversely associated with CVD mortality at every level of sitting (<2, 2–5.9, 6–8, and >8 hours per day). Conversely, sitting time was associated with increased mortality. When studying the effects of SB across different PA levels, while the associations between SB and mortality was significant in individuals with lowest levels of moderate volume PA, the relationship between SB and mortality was no longer significant in individuals who were participating in ≥35.5 MET-hour per week of PA (≈60–75 minutes per day of moderate intensity PA; Figure 1).26 Finally, the results for the joint association of television viewing and PA on CVD mortality were similar to those for sitting and PA.

    Figure 1.

    Figure 1. Hazard ratios for the joint association of sitting time and physical activity with cardiovascular disease (CVD) mortality. Data derived from appendix of Ekelund et al.26

    In summary, in addition to the beneficial effects of PA on risk for CVD (discussed below), there is emerging evidence that excessive SB is also an important CVD risk factor, particularly in those with lowest levels of moderate volume PA. To the contrary, high levels of PA appear to attenuate the negative cardiovascular consequences of SB, but more research is required to better determine the interactions between PA/SB on health outcomes.

    Consequences of PI on Cardiovascular Health

    Cardiovascular health is independently associated with PA, with PI linked with the greatest risk of developing CVD.27,28 The prevalence of PI has increased over recent years, perhaps as the result of a greater adoption of the Western lifestyle, characterized by greater sedentary time, lower participation in active transport, and time spent in leisure or purposeful PA.29–31 Of note, a global examination of PI and noncommunicable disease prevalence estimated that 6% of coronary artery disease, 7% of type 2 diabetes mellitus (T2DM), 10% of breast cancer, and 10% of colon cancer cases were caused by PI. Premature mortality as a result of PI accounted for over 5.3 million global deaths in 2008,28 and in the United States, all-cause and CVD-specific mortality advanced mortality by 4 and 2.4 years, respectively.32 By eliminating PI, it is estimated that life expectancy of the world’s population would increase by 0.68 years.28

    PI is also closely associated with metabolic disorders, such as impaired glucose metabolism, which substantially increases risk of CVD.33 Troubling trends of an increased prevalence of T2DM in children and young adults are in part because of unhealthy lifestyle which promotes PI and the consumption of foods with low nutritional value.34 In a longitudinal examination, 3596 Finnish youth (baseline age, 3–18 years) were followed for 31 years to determine the effects of persistent PI on glucose metabolism in adulthood.35 Compared with participants who were persistently physically inactive, those who increased PA (relative risk, 0.47; 95% CI, 0.29–0.76) or remained persistently active had a lower relative risk for having impaired glucose metabolism at follow-up (0.70; 95% CI, 0.51–0.97). However, individuals who had decreased PA were at similar risk (relative risk, 0.93; 95% CI, 0.66–1.36) to those with persistent PI.

    Although the relation between PI and cardiovascular health is independent and robust, the modulatory effects of PI on cardiovascular health are complex and not completely elucidated. Strong predictors of CVD, such as conduit arterial stiffness and reduced endothelium-dependent dilation (ie, flow-mediated dilation), have been well documented in physically inactive men and women.36 Much of our current understanding of the vascular consequences of becoming inactive has been through extreme models of PI, such as bed rest or limb immobilization.37–39 In contrast, Boyle et al40 sought to implement a real-world model of PI by reducing daily PA levels of highly active volunteers (>10 000 steps per day) to PI levels (<5000 steps per day) over a 5 day period. This short exposure to a physically inactive lifestyle induced a decrease in popliteal artery flow-mediated dilation (baseline, 4.7±0.98%; day 5, 1.72±0.68%, P<0.05) and endothelial cell activation (CD62E+), and an increase of markers of endothelial cell apoptosis (CD31+/CD42b). In line with these findings, both preclinical and clinical investigations have identified oxidative stress as a prominent mediator of endothelial dysfunction.41–44 Imbalances between the production and destruction of reactive oxygen species by antioxidant defense systems associated with inactivity, promote the uncoupling of endothelial nitric oxide synthase. Such abnormalities result in reduced nitric oxide bioavailability and increased production of superoxide.45 Prolonged disruption of endothelial function and associated reduction in vascular compliance because of PI are particularly damaging to cardiovascular health, finally imposing elevated loads on the left ventricle (LV), which may lead to LV stiffening, chamber remodeling and increasing risk of developing HF.46,47

    PA and its Relation to Cardiovascular Health

    The cardioprotective effects of regular PA, whether performed in low or high volumes, are clear and extend across all ages, sex, and race (Table 1).

    Table 1. Physical Activity and Prevention of CVD and CVD Related Events

    Author Population PA Measurement Results
    Lee et al27 40 801 men; 14 336 women PA questionnaire is assessing duration, distance, frequency, and speed of running or jogging. Compared with nonrunners, runners had 30% and 45% lower adjusted risks of all-cause and CV mortality, respectively, with a 3-year life expectancy benefit. During an average 15-year follow-up, persistent runners had a 29% and 50% lower risks of all-cause and CV mortality, respectively, compared with never runners.
    Florido et al48 4881 men; 6470 women Baecke questionnaire Participants maintaining PA recommendations compared with those maintaining poor activity had lower heart failure risk (0.69, 95% CI, 0.60–0.80). Individuals increasing from poor to meeting PA recommendations had reduced heart failure risk (0.77, 95% CI, 0.63–93).
    O’Donovan et al49 27 732 men; 31 273 women Interview is inquiring about housework, walking, sport, and exercise PA performed in previous 4 wk. Risk of CVD mortality in overweight (1.41, 95% CI, 0.94–2.10) and obese (1.41, 95% CI, 0.84–2.38) individuals did not differ compared with normal weight individuals meeting PA guidelines.
    Nes et al50 19 269 men; 20 029 women PAI score Men and women with a PAI score of ≥100 had 17% (95% CI, 7%–27%) and 23% (95% CI, 4%–38%) reduced risk of CVD mortality, respectively, compared with inactive individuals.
    El Saadany et al51 7146 men; 8161 women PA interview Irregular (≤4 days/wk) PA and regular (>4 d/wk) PA were associated with lower risk of CVD mortality (0.66, 95% CI, 0.51–0.85 and 0.58, 95% CI, 0.47–0.72, respectively) compared with no activity. These observations only remained true for women and not men for irregular and regular activity.
    Kubota et al52 34 874 men; 40 038 women Self-administered PA questionnaire regarding leisure-time, commuting, housework PA. Compared with the lowest quartile of daily PA, higher PA levels were associated with reduced risks of total and ischemic stroke. Highest PA level was not associated with reduced risks of hemorrhagic strokes. Second and third quartile had lowest risk of total stroke (0.83, 95% CI, 0.75–0.93 and 0.83, 95% CI, 0.75–0.92, respectively).
    Lear et al53 54 621 men; 76 222 women International Physical Activity Questionnaire Individuals with moderate or high PA levels had a lower risk of major CVD (0.86, 95% CI, 0.78–0.93 and 0.75, 95% CI, 0.69–0.82, respectively) compared with those with low levels of PA.
    Fishman et al54 1412 men; 1617 women Accelerometer Compared with the lowest tertile of activity, those in the second and third highest tertile had significantly lower risk of mortality (0.21, 95% CI, 0.12–0.38 and 0.36, 95% CI, 0.30–0.44, respectively).
    Soares-Miranda et al55 1641 men; 2566 women Minnesota Leisure-Time Activities Questionnaire Walking pace, distance, leisure-time PA, and exercise intensity were associated with lower risk of CHD, stroke, and CVD. Highest leisure-time PA (kcal/wk) compared with lowest quintile had lower risk for CHD (0.57, 95% CI, 0.45–73), stroke (0.56, 95% CI, 0.42–0.75), and CVD (0.59, 95% CI, 0.48–0.72).
    Bell et al56 3707 blacks; 10 018 whites Baecke Questionnaire PA was inversely related to CVD, heart failure, and CHD incidence in both races, and stroke in blacks.
    Shortreed et al57 4729 men and women Self-reported PA Compared with long-term physical inactivity, long-term PA was associated with a CVD rate ratio of 0.95 (95% CI, 0.84–1.07), all-cause mortality rate ratio of 0.81 (95% CI, 0.71–0.93), and CVD attributable mortality rate ratio of 0.83 (95% CI, 0.72–0.97). A greater protective effect of long-term PA on CVD incidence was present for men but not women.
    Gulsvik et al58 5653 men and women Self-reported PA Individuals with a high level of PA compared with low PA levels had lower all-cause (0.63, 95% CI, 0.56–0.71), ischemic heart disease (0.66, 95% CI, 0.52–0.83), and stroke (0.66, 95% CI, 0.47–0.93) mortality risk compared with no activity.
    Wen et al59 199 265 men; 216 910 women Leisure-time PA questionnaire Compared with inactive individuals low-volume activity reduced all-cause mortality and extended life expectancy by 3 y. Exercise 15 min/day =14% reduced risk of all-cause mortality. Every additional 15 min of daily exercise further reduced all-cause mortality by 4% (95% CI, 2.5–7.0). Inactive individuals had a 17% (95% CI, 1.10–1.24) increased risk of mortality compared with low-volume group.
    Tjønna et al60 26 005 men; 27 537 women Self-reported PA Physically active individuals with CVD risk factors had a lower risk (HR, 0.76; 95% CI, 0.61–0.95) compared with the inactive group with CVD risk factors.
    Wisløff et al61 27 143 men; 28 929 women Self-reported PA Compared with those reporting no activity, a single weekly bout of high-intensity exercise lowered risk of CVD death in men (0.61, 95% CI, 0.49–0.75) and women (0.49, 0.27–0.89). No additional benefits when increasing duration or number of sessions per week. Risk reduction increased with increasing age in men but not in women.

    CHD indicates coronary heart disease; CVD, cardiovascular disease; HR, hazards ratio; PA, physical activity; and PAI, physical activity intelligence.

    27,48–61 However, much of the seminal epidemiological work quantifying the volume of PA necessary to curb health risks resulted from subjective assessments of PA, including the use of questionnaires or interviews.62–64 Technological advancements have improved the accuracy and reliability of device-measured PA levels and have become readily available to be applied in large scale epidemiological investigations for objective assessment of PA.65 These advancements coupled with large scale randomized controlled trials have allowed to accurately identifying the specific PA volumes associated with improved markers of cardiovascular health.66–69 These results have made it possible to develop individualized PA recommendations, thus moving away from a one size fits all approach.66

    Despite the known benefits of PA, the adoption of a physically active lifestyle has remained low because of various reasons: personal barriers associated with perceived limitations in self-efficacy, lack of time, and misconceptions of the volume of exercise necessary for cardiovascular health benefits. Despite the evidence supporting the cardiovascular benefits of moderate-to-vigorous PA performed even in bouts of at least 10 minutes, the level of adherence of the general population to the guidelines remains unacceptably low. A recent prospective cohort study assessing PA levels in 1274 older men, over a median follow-up of 5 years investigated the effects of bouts of PA of at least 10 minutes on mortality.70 Accelerometers were used to quantify moderate-to-vigorous PA accumulated in sporadic minutes of PA or in bouts lasting ≥10 minutes. Over the course of a 7-day PA assessment period, only 16% of the older men met the recommended volume of PA when applying the ≥10 minutes criteria, whereas 66% of older men achieved 150 minutes of recommended activity with minutes of accumulated sporadic PA. Despite these stark differences in the proportion meeting PA recommendations, HR for all-cause mortality when PA was accumulated sporadically (HR, 0.59 [95% CI, 0.43–0.81]) did not differ from when PA was performed in ≥10 minutes bouts (HR, 0.58 [95% CI, 0.33–1.00]), suggesting that participation in PA is beneficial irrespective of how it is accumulated. However, this study only included older men, clearly requiring further validation in younger populations and in women, finally allowing to potentially develop even more individualized PA recommendations.

    The benefits of PA on cardiovascular health and to combat the aging process are multifaceted (Figure 2).71–97 Aging is associated with a decline in LV as well as vascular function, finally altering the interaction between the LV and arterial system (ventricular-arterial coupling). The impairment in ventricular-arterial coupling is related, at least in part, to an increase in arterial stiffening, which increases the afterload on the heart and consequently increases LV stiffening. Prolonged exposure to these conditions as it occurs with aging increases the risk of developing HF. However, lifelong exercise, 4 to 5 sessions per week can prevent age-related decrements in compliance and distensibility,68 while maintaining youthful arterial compliance and function.69 Although the exact mechanisms responsible for the above described ET-induced cardiovascular benefits are not clear, several hypotheses have been proposed. Rodent models have provided evidence that ET enhances calcium handling through the sarcoendoplasmic reticulum calcium transport ATPase as well as an increase in its mRNA expression.70,98 Furthermore, ET reduces circulating markers of systemic inflammation, such as C-reactive protein,99 which may protect against inflammation-mediated myocardial fibrosis and dysfunction. In regard to the peripheral vasculature, regular PA can reduce mitochondrial reactive oxygen species production, enhance cellular antioxidant defense proteins, and reduce mitochondrial fission (a sign of mitochondrial dysfunction).100 Collectively, these beneficial effects of ET may contribute to improving compliance, reducing stiffness, and afterload, finally reducing the risk of future cardiac dysfunction.

    Figure 2.

    Figure 2. The multidimensional mechanisms associated with the deleterious effects of sedentary behavior and the beneficial effects of physical activity that occur within the mitochondria, skeletal muscle, myocardium, and conduit arteries. β-HAD indicates β-hydroxyacyl CoA dehydrogenase; AGE, advanced glycation end products; Akt, protein kinase B; CRP, C-reactive protein; FOXO3a, forkhead box O3;IL-6, interleukin-6; MHC, myosin heavy chain; MMP, matrix metalloproteinase; mTOR, mammalian target of rapamycin; MuRF-1, muscle RING-finger protein-1; NADPH, nicotinamide adenine dinucleotide phosphate; NO, nitric oxide; NOS, nitric oxide synthase; PGC-1α, peroxisome proliferator-activated receptor γ coactivator 1-α; PI3K, phosphoinositide 3-kinase; SERCA2a, sarcoplasmic reticulum calcium adenosine triphosphatase; and SIRT3; nicotinamide adenine dinucleotide-dependent deacetylase sirtuin-3 SOD.

    Importance of Cardiorespiratory Fitness

    PA and ET are associated with improvements in cardiovascular health and longevity, however, much of these benefits may result from the improvements in CRF following increased PA, which is a stronger predictor of prognosis compared with PA/ET alone.1–3,101,102 While the explanatory factors for the different prognostic ability of CRF versus PA/ET are complex, a potential reason may be related to the well-documented observations of interindividual fitness changes to the same volume of PA/ET.103,104 The gold standard for CRF remains the measurement of peak oxygen consumption (VO2) by cardiopulmonary exercise testing using gas exchange analysis. Other assessments of exercise capacity, such as estimated METs, determined by speed and incline on the treadmill using standard algorithms, or even 6-minute walk test, particularly in patients with coronary heart disease (CHD) and HF, have been potent predictors of prognosis.105–108 The potential benefits of improved PA, ET, and CRF are numerous and are summarized in Table 2.

    Table 2. Potential Benefits of Physical Activity, Exercise Training, and Cardiorespiratory Fitness on Prognosis

    Physiological Benefits
    Reduced blood pressure109 Reduced systemic inflammation110
    Improved heart rate variability111 Decreased myocardial oxygen demands112
    Improved endothelial function113 Maintain lean mass114
    Improved insulin sensitivity115 Reduced visceral adiposity116
    Reduced myocardial infarction117 Increased capillary density118
    Reduced blood and plasma viscosity119 Improved mood and psychological stress120
    Increased mitochondrial density79 Improved sleep121
    Reduced risk of developing
     Hypertension122 Osteoporosis123
     Depression124 Osteoarthritis125
     Metabolic syndrome126 Dementia and Alzheimer Disease127
     Diabetes mellitus128 Breast, colon, and other cancers129

    Similar to high levels of PA, high levels of CRF are associated with reduced prevalence of many CVD and CHD risk factors, including hypertension, obesity, metabolic syndrome, and T2DM.101,102,130,131 Clearly, many studies have demonstrated the powerful impact of CRF on prognosis, which has been noted in large population-based studies, clinical cohorts, and in those at high CVD risk and in CVD populations, such as CHD and HF.1–3,101,102,131

    Nearly a decade ago, a very high-profile meta-analysis by Kodama et al132 of 33 studies in over 100 000 individuals observed that every 1 estimated MET increase in CRF was associated with 13% and 15% reductions in all-cause and CVD/CHD mortality, respectively. This large meta-analysis also defined age- and sex-specific levels of CRF that were associated with lowest event rates in women (40 years: 7 METs; 50 years: 6 METs; and 60 years: 5METs) and men (40 years: 9 METs; 50 years: 8 METs; and 60 years: 7 METs).

    Additionally, CRF is associated with prognosis in those individuals with high-risks of CVD, including those with metabolic syndrome, pre-T2DM or T2DM.1–3,101,102,131,133 In such high-risk individuals, often those with high levels of CRF have a better prognosis that do unfit individuals without these disorders. High levels of CRF have also been protective against lifetime CVD risks. In fact, subjects with a high burden of CHD risk factors but increased level of CRF have lifetime CVD risks similar to or even lower than those with lower risks factors,134 further supporting the powerful role of CRF even in those with otherwise high CVD risk.

    Many studies have also investigated the effects of the changes in CRF over time on CVD risk factors and on CVD morbidity and mortality.1–3,101,102,131,133 Particularly, Sui et al131 have recently reviewed the impact of changes in CRF on improvements in various CVD and CHD risk factors. Using the Aerobics Center Longitudinal Study (N=9777), Blair et al135 reported that men classified in the lowest 20th percentile of CRF based on age at their first examination but fit at the time of their second examination several years later had a 52% reduction in CVD mortality compared with men who remained unfit. Similarly, Lee et al136 evaluated the effects of the changes in CRF on CVD mortality over a mean 11.4 years follow-up in 14 345 subjects using the same Aerobics Center Longitudinal Study data set. They demonstrated that those individuals who presented with a preserved CRF or an increased CRF after 6.3 years from the initial CRF assessment had significant reductions in CVD mortality by 27% and 42%, respectively. Importantly, for every 1 estimated MET increase, all-cause and CVD mortality were reduced by 15% and 19%, respectively. Erikssen et al137 and others have noted similar findings about improvements in CRF over time.

    The assessment of CRF represents the synergistic functioning of multiple organ systems to effectively transport oxygen from the air to the mitochondria of the working skeletal muscle, which must produce the necessary energy to meet the demands of activity as well as effectively remove the resultant metabolic byproducts that impair the ability of the muscle to sustain activity when accumulated in excess. Considering the highly prognostic nature of CRF and its representation of the whole-body physiological function, its assessment has been used as the primary end point also in non-ET interventions (ie, pharmacological) in HF patients. Pharmacological interventions, such as angiotensin-converting enzyme inhibitors and sildenafil therapy in patients with HF with reduced ejection fraction (HFrEF) have been effective in significantly increasing CRF.138,139 Conversely, these pharmacological interventions have not been as effective in patients with HF with preserved ejection fraction (HFpEF), highlighting the need to develop nonpharmacological therapeutics, as also described in the next paragraphs. For example, HFpEF patients randomized to 24 weeks of phosphodiesterase-5 inhibitor did not experience significant increases in CRF or clinical status when compared with placebo.140 Similarly, a 12-month intervention of daily spironolactone did not result in CRF improvement.141 More recently, novel interventions aimed at enhancing the nitric oxide signaling pathway to increase its bioavailability have been tested in clinical trials. However, despite promising results in small pilot studies, a 4-week intervention of inhaled inorganic nitrite (a precursor to nitric oxide) did not improve CRF in patients with HFpEF.142 Although an effective pharmacological intervention to manage HFpEF has not been found, future studies are encouraged to continue to use CRF as a primary outcome of interest.

    Among the traditional risk factors for CVD, CRF has consistently shown to be one of the strongest prognosticators. A greater CRF in men with the metabolic syndrome protects against all-cause and CVD mortality to similar to what is seen in healthy men.143 In addition to investigating the effects of CRF in patients with metabolic syndrome, the relation between CRF and obesity has also received much attention. As outlined by Kennedy et al,144 the independent effects of fitness versus fatness have been debated. Several of the authors of this review, as well as others, have evaluated the independent effects of excess adiposity (ie, obesity) and CRF on subsequent CVD and all-cause mortality.1–3,133,144–148 In fact, considerable evidence indicates the high levels of CRF significantly attenuate or even eliminates the elevated risk of CVD- and all-cause mortality in overweight and obese individuals. This has been reported in patients with dyslipidemia and T2DM as well as in the general population. Indeed, CRF markedly alters the relationship of fatness and subsequent prognosis. Recently, Barry et al146 performed a meta-analysis on 8 studies and 9 independent groups to assess the joint impact of body mass index (BMI), a surrogate for increased adiposity, and CRF on CVD mortality. Unfit individuals had 2× to 3× higher mortality risk across all levels of BMI. Both overweight fit and obese fit had 25% and 42% increased mortality risk, respectively, compared with normal weight fit, which is considerably <2-fold increased risk reported in overweight unfit individuals.

    We have recently reviewed the impact of CRF on prognosis in the obesity paradox, especially in CHD, HF, and atrial fibrillation.2,145,147–149 The obesity paradox describes the improved prognosis typically reported in epidemiological studies in patients with class I and II obesity compared with normal weight and underweight individuals in the setting of established CVD, particularly CHD, HF, and more recently atrial fibrillation. In a study of 9563 patients with CHD followed for an average of over 13 years, those in the bottom tertile of CRF based on age and sex showed an obesity paradox. In fact, in this group of unfit individuals, measures of adiposity, such as higher BMI, % body fat, and waist circumference, were associated with improved prognosis compared with the thinner but similarly unfit individuals.150 However, the relatively fit CHD patients (not in the bottom tertile for age- and sex-based CRF) had an excellent prognosis that was similar in all groups of adiposity, suggesting that increased BMI, % body fat, and waist circumference were no longer protective in the setting of preserved or increased CRF. Similarly, in 2066 patients with HFrEF,151 those individuals with reduced CRF defined as peak VO2 <14 mL/kg per minute and with concomitant obesity presented a more favorable prognosis compared with normal weight individuals. However, an obesity paradox was not reported in those patients with a relatively preserved CRF (peak VO2 ≥14 mL/kg per minute), suggesting that in patients with HF, obesity may only be protective in the setting of reduced CRF and that perhaps therapeutics aiming at improving CRF in patients with HF may result in greater benefits as compared to those targeting body weight alone. In addition to CHD and HF, CRF levels and related improvements over time have been associated with markedly improved prognosis also in patients with atrial fibrillation.149,152

    A recent study from Norway (HUNT study [The Nord-Trøndelag Health Study]) also demonstrated that PA levels were stronger predictors of survival compared with BMI.153 In fact, while changes in PA markedly impacted mortality, with increased PA levels associated with an improved prognosis, changes in BMI, including weight loss, did not affect mortality rate.154 Taken together, these data support the importance of increased CRF and PA to reduce CVD- and all-cause mortality risks, independent of obesity.

    Importance of Exercise in HF

    Currently, over 6 million adults have been diagnosed with HF and more sobering is the projected increase to over 8 million by the year 2030.155 Among elderly individuals, HF-related exacerbations is the most common cause for hospitalization, placing a significant burden on individuals as well as the health care system. Exercise intolerance, typically defined as reduced CRF, is the major symptom in patients with HF.156,157 As described above, increasing PA and ET remain the most effective therapeutic strategies to improve CRF.1,158 ET induces improvements in CRF typically objectively assessed with peak VO2 during a maximal cardiopulmonary exercise test159,160 in a wide spectrum of HF phenotypes,161 including HFrEF162 and HFpEF.163 The effects of ET on CRF have been tested in several small randomized controlled trials and supported by meta-analyses suggesting beneficial effects of ET on clinical outcomes.164,165 However, the majority of these studies were performed in a single-center and limited by the small sample size, making them likely unpowered to detect meaningful improvements in strong clinical outcomes (eg, all-cause mortality and HF hospitalizations).

    The largest randomized controlled trial testing the efficacy and safety of ET on clinical outcomes is the multicenter HF-ACTION (Heart Failure: A Controlled Trial Investigating Outcomes of Exercise Training), which randomized >2300 stable patients with HFrEF (LVEF ≤35%) with New York Heart Association class II to IV to 36 supervised sessions of aerobic ET and home-based ET in addition to standard of care or to standard of care alone.162 After a median follow-up of about 30 months, patients randomized to the ET group experienced a modest 4% improvement in peak VO2162 which was lower than the anticipated 10% to 15% suggested by the smaller studies.166 One of the reasons for the small improvement in CRF was perhaps the low adherence to the prescribed ET, with only 30% of patients achieving the targeted level of ET in terms of recommended minutes/wk.162 Nevertheless, the ET training failed to reduce the risk for the primary composite end point of all-cause mortality and all-cause hospitalizations. However, after prespecified statistical adjustments for key prognostic factors of morbidity, ET was associated with a significant 13% relative risk reduction for the primary composite end point as well as a 15% relative risk reduction for the composite secondary end point of CVD mortality and HF hospitalizations.162 A secondary analysis of the trial also suggested a greater reduction for the primary and secondary composite end points in those patients who achieved the targeted goal of weekly ET which was also consistent with a greater improvement in CRF reported in this subgroup.167 Importantly, ET was safe as the number of adverse events did not differ between the intervention and control groups.162 Clearly, the HF-ACTION has added important information on the beneficial effects of ET, however, those were limited to HF patients with an LVEF ≤35%, and to date similar large multicenter studies in HFpEF, or even in HFrEF but with LVEF ≥35%, are lacking and, in fact, highly encouraged, perhaps this time with the use of additional tools to improve adherence to ET during the course of the study.168 Nevertheless, ET seems to exert similar, if not even greater benefit, at least on CRF, in patients HFpEF.163

    The improvements in CRF following ET in the different forms of HF seems to result from a variety of mechanisms.160,169 Peak VO2, typically reported in a milliliter of oxygen consumption per kilogram of body weight per minute (mL/kg per minute), following the Fick principle results from the product of cardiac output and arteriovenous oxygen difference [C(a−v)O2], which is clearly also affected by hemoglobin concentrations:

    Peak VO2=(stroke volume×heart rate)max×[C(a−v)O2]max

    ET can, therefore, improve CRF (ie, peak VO2) by affecting one or more of these variables.

    In patients with HFrEF, in which the effects of ET have been investigated the most compared with HFpEF, the ET-induced changes in CRF have been associated with a combination of improvements in cardiac factors and peripheral noncardiac factors,170 measured using both invasive and noninvasive assessments. In patients with HFrEF, ET can improve systolic function (ie, LVEF) and cardiac remodeling, by reducing LV end-diastolic volume and LV end-systolic volume,171 finally resulting in improved peak cardiac output.172 Importantly, the improvement in CRF induced by ET also result from improvements in peripheral factors, particularly an increase in systemic arterial-venous oxygen difference, leg blood flow, and oxygen delivery.172 Such effects are typically independent of changes in body weight, which highlights the importance of targeting CRF in HFrEF, independent of changes in body mass.

    In addition to ET, several pharmacological strategies have also shown improvements in CRF in HFrEF173 leading to larger CVD outcomes trials investigating the effects on clinical outcomes of such therapies. However, as described in the prior sections of this review, several failures in HFpEF have been reported in the last years as well as many efficacy, design, and ethical issues that have been associated with exploring advanced therapies that involve stem cell interventions.174 Such disappointing results have increased the attention on the effects of ET in this population. Indeed, ET alone is perhaps the most powerful tool to improve CRF in HFpEF, particularly when combined with weight loss strategies (ie, caloric restriction) in patients with concomitant obesity.175 The mechanisms of improvements in CRF mediated by ET, however, differ significantly from what described previously in HFrEF. A meta-analysis of 6 randomized controlled trials investigating the effects of ET in HFpEF found that ET improves CRF but without significantly affecting cardiac systolic and diastolic function.163 Such results suggest that noncardiac limitations may be major contributors for exercise intolerance in HFpEF. Few studies have confirmed that ET training improves CRF in older patients with HFpEF176 but without inducing significant changes in resting and peak cardiac output and cardiac index, proposing improvements in peak C(a−v)O2 as major contributors for improved CRF in this population.171 Prior small studies, however, have shown some degree of improvements in cardiac diastolic function in patients with HFpEF.177 Clearly, the lack of a universal definition of HFpEF plays a major role in determining potential improvements in cardiac versus noncardiac factor, or perhaps a combination of both. The presence of cardiac diastolic dysfunction, particularly when assessed invasively and during exercise,178–181 may identify those patients in which ET could affect cardiac function to a greater degree compared with those who have been diagnosed with HFpEF, but without meeting major diastolic dysfunction criteria for the initial HFpEF diagnosis.182 HFpEF is a highly heterogeneous population, to the extent that different phenotypes of HFpEF have been proposed in the literature.183 Individuals that gain weight over time have been shown to have increased diastolic stiffening,184,185 which may partly explain the high prevalence of obesity found in patients with HFpEF (Figure 3). Obese HFpEF patients have an increased plasma volume, greater degree of concentric LV remodeling and right ventricular dilation, more right ventricular dysfunction,186 higher biventricular fillings pressures during exercise, and lower CRF compared with nonobese HFpEF and control individuals.187 ET has been proven to be effective in most phenotypes and subgroups, more recently also the in patients with HFpEF who also have class II and III obesity,175 which represent one of the most common comorbid condition in this population and in which adiposity188,189 and peripheral noncardiac factors176 have been recognized as major determinants of reduced CRF. Initiating ET before the development of HFpEF would certainly be an effective way to reverse the deleterious effects of a sedentary lifestyle.190

    Figure 3.

    Figure 3. Proposed mechanisms by which obesity can contribute towards the development and progression of heart failure with preserved ejection fraction (HFpEF). BNP indicates B-type natriuretic peptide; CV, cardiovascular; LV, left ventricular; and RV, right ventricular. Adapted from Pandey et al184 with permission. Copyright ©2018, Elsevier.

    Patients with HF are also characterized by reduced subjective assessment of quality of life, typically measured using validated questionnaires.191 ET is an effective strategy to improve quality of life in both HFrEF192 and HFpEF,193 further supporting the importance of its implementation in the care of patients with HF.193

    Importance of Exercise in Muscular Fitness

    Body composition compartments play a central role in determining CRF.189,194–196 Particularly, the levels of lean mass (LM) of the extremities (ie, appendicular LM) are considered the best surrogate for appendicular skeletal muscle mass,197 major determinant of CRF.198 In addition to the amount of LM, its composition and functionality are also important. Recently, the ratio between intermuscular fat and skeletal muscle mass area assessed with magnetic resonance imaging was found to be the strongest predictor for exercise intolerance in patients with HFpEF.194 Furthermore, when a reduced amount of LM is associated with reduced functionality, patients typically present with sarcopenia,197 which has been associated with worse CRF and outcomes in several chronic diseases, recently also in HF.199 When sarcopenia is coupled with excess adiposity (ie, obesity), it can be defined as sarcopenic obesity, which is associated with an even worse CRF compared with sarcopenia alone.189,200,201 For such reasons, preserving or perhaps even increasing LM with resistance training in association with aerobic exercise may represent the most effective therapeutic strategy to improve muscular fitness in the setting of HF.202,203 This may be particularly true in older adults, in which LM loss occurs physiologically, therefore, increasing the risk of sarcopenia and sarcopenic obesity, but also in the more advanced stages of HF, which are characterized by the presence of a systemic catabolic state responsible for the loss of LM, often associated with concomitant loss of FM, which is, when unintentional, a critical negative prognostic factor in HF.203,204 Randomized trials testing these hypotheses and also investigating the intensity of the resistance ET in association with more established protocols involving aerobic ET are clearly needed.

    Exercise Dosing

    There continues to be considerable controversy about the optimal dose of PA/ET for CVD and general prevention; however, substantial evidence suggests that any level of PA/ET is better than none.1–3,133,205 Physical Activity Federal Guidelines call for a minimum of 150 minutes per week of moderate aerobic PA or 75 minutes per week of vigorous PA, while the Institute of Medicine suggests that 60 minutes daily of total PA is ideal.1,2,7,19,160 The majority of the general population does not meet these guidelines, with only 10% meeting these minimum recommend level of PA using objective assessments, such as accelerometers.1,2,206,207 However, recent evidence indicates substantial benefits even with ET doses much lower than what recommended in these guidelines.

    In a large study of 416 175 individuals from Taiwan, Wen et al59 noted a dose-response relationship between aerobic PA and subsequent mortality, with some mortality reductions noted with just 15 minutes per day of moderate PA (Figure 4). In fact, progressive reductions in mortality were noted up to ≤90 daily minutes of moderate PA and ≤30 to 40 minutes of vigorous PA, which was defined as only 6.5 to 8.5 METs. In a recent large running study from 55 000 people from the Aerobics Center Longitudinal Study, including 13 000 runners and 42 000 nonrunners, who were followed on average for nearly 15 years, runners had impressive reductions in mortality and CVD mortality by 30% and 45%, respectively, compared with nonrunners, with an average increase in life expectantly and CVD life expectancy of 3 and 4.1 years, respectively.27 Persistent runners had the full benefits, while those who had stopped running or started running during the study had nearly half the benefits compared with never runners. These results are not unexpected, and many would believe that there are benefits of running but also there may be selection bias, in that those able to run may be healthier than nonrunners.

    Figure 4.

    Figure 4. Daily physical activity duration and all-cause mortality reduction. Reprinted from Wen et al59 with permission. Copyright ©2011, Elsevier.

    However, interesting findings emerge when assessing running dosing, by dividing runners into quintiles (Q) of exercise volumes, such as miles per week, times per week and minutes per week. In fact, Q1 runners (<6 miles per week, 1–2 times per week, and <51 minutes per week) had similar all-cause and CVD-mortality risks compared with Q2–Q4 runners, with a slight trend to lower mortality to Q5 runners (Figure 5).27 These results suggest that weekly running, which is often considered to be a relatively high intensity form of ET that is common and convenient, the maximal benefits on all-cause and CVD-mortality occurred at low doses, including ET doses well below the current International PA guidelines. In a subsequent analysis of the Q5 runners who were divided into tertiles, the top 8% of runners with regards to dosing appeared to lose the benefits, at least compared with the lower dose runners (Figure 6),208 suggesting that more may not be better but also raising the possibility that more could be worse with regard to ET dosing.

    Figure 5.

    Figure 5. Hazard ratios (HRs) of all-cause and cardiovascular mortality by running characteristic (weekly running time, distance, frequency, total amount, and speed). Participants were classified into 6 groups: nonrunners (reference group) and 5 quintiles of each running characteristic. All HRs were adjusted for baseline age (y), sex, examination year, smoking status (never, former, or current), alcohol consumption (heavy drinker or not), other physical activities except running (0, 1–499, or ≥500 metabolic equivalent [MET] minutes/wk), and parental history of cardiovascular disease (yes or no). All P values for HRs across running characteristics were <0.05 for all-cause and cardiovascular mortality except for running frequency of ≥6 times/wk (P=0.11) and speed of <6.0 miles per hour (P=0.10) for cardiovascular mortality. Reprinted from Lee et al27 with permission. Copyright ©2014, Elsevier.

    Figure 6.

    Figure 6. Hazard ratios (HRs) of all-cause and cardiovascular disease (CVD) mortality by weekly running time, distance, frequency, and total amount. Participants were classified into 8 groups: nonrunners and 5 quintiles of each running dose (Q1–Q5) with the last quintile (Q5) additionally categorized into 3 tertiles (Q5-T1, Q5-T2, and Q5-T3) using larger markers (7 groups for running frequency because of limited numbers in ≥7 times/wk). All HRs were adjusted for baseline age (y), sex, examination year, smoking status (never, former, or current), alcohol consumption (heavy drinker or not), other physical activities except running (0, 1–499, or ≥500 metabolic equivalent task minutes per week [MET min/wk]), and parental CVD (yes or no). The number of participants (number of all-cause deaths) were 42 121 (2857), 2710 (110), 2584 (116), 2505 (103), 2647 (112), 850 (33), 822 (30), and 898 (52) in the corresponding 8 running time groups from nonrunners to Q5-T3; 42 121 (2857), 2626 (105), 2473 (120), 2961 (123), 2218 (92), 885 (36), 1027 (40), and 826 (40) in running distance; 42 121 (2857), 2757 (62), 3076 (105), 2817 (131), 2500 (143), 1215 (66), and 651 (49) in running frequency; and 42 121 (2857), 2609 (109), 2598 (122), 2558 (116), 2626 (105), 863 (31), 886 (30), and 876 (43) in total running amount. The number of participants (number of CVD deaths) were 40 319 (1055), 2628 (28), 2501 (33), 2435 (33), 2567 (32), 827 (10), 801 (9), and 863 (17) in the corresponding 8 running time groups from nonrunners to Q5-T3; 40 319 (1055), 2550 (29), 2386 (33), 2874 (36), 2156 (30), 858 (9), 1001 (14), and 797 (11) in running distance; 40 319 (1055), 2714 (19), 2993 (22), 2725 (39), 2396 (39), 1174 (25), and 620 (18) in running frequency; and 40 319 (1055), 2531 (31), 2508 (32), 2477 (35), 2553 (32), 842 (10), 864 (8), and 847 (14) in total running amount. The bars indicate 95% CIs, and HRs appear next to the bars. Reprinted from Lee et al208 with permission. Copyright ©2016, Elsevier.

    Future Considerations

    Although the benefits of PA/ET and deleterious effects of SB/PI are well established, further study on potential benefits of ET on major clinical events in HFpEF, T2DM, and other chronic diseases is needed. Similarly, the relative values of high-intensity interval training and resistance training on major clinical events in these populations may also need further investigation.209 Additionally, much of our limited understanding of the mechanistic consequences of SB derives from preclinical models and cross-sectional examinations of sedentary individuals. Efforts have recently been made to characterize the effects of prolonged acute (ie, hours) sitting on metabolic parameters such as postprandial glucose and insulin responses, however, no study has examined the cellular and molecular responses in various tissues across different populations and PA status. Another area that warrants attention is elucidating the high degree of interindividual variation in CRF responses to exercise interventions. Ross et al103 have made great strides in addressing the influence of amount and intensity on CRF outcomes, however, there still remains a large gap in identifying molecular characteristics that may provide insight into who responds or does not respond to exercise. Finally, we should recognize that the PA/ET fields of medicine have not done an excellent job of promoting PA/ET throughout the world and in many diseases, including the patients with CVD.However, we highly encourage research investigating novel strategies to improve adherence to the recommendations described above, finally resulting in increased PA/ET and reduced SB/PI across the globe.2,210

    Conclusions

    In this State-of-the-Art review, we discussed the potential benefits of PA/ET and the adverse effects of SB/PI in the primary and secondary prevention of chronic diseases, especially CVD. The constellation of data reviewed in this article marked the benefits of increased PA/ET, particularly as they lead to increased CRF, finally resulting in improved prognosis in a large spectrum of metabolic diseases and CVD. Greater implementation of this therapy, therefore, is desperately needed worldwide.

    Nonstandard Abbreviations and Acronyms

    BMI

    body mass index

    CHD

    coronary heart disease

    CRF

    cardiorespiratory fitness

    CVD

    cardiovascular disease

    ET

    exercise training

    HDL

    high-density lipoprotein

    HF

    heart failure

    HF-ACTION

    Heart Failure: A Controlled Trial Investigating Outcomes of Exercise Training

    HFpEF

    heart failure with preserved ejection fraction

    HFrEF

    heart failure with reduced ejection fraction

    HR

    hazards ratio

    LM

    lean mass

    LV

    left ventricle

    METs

    metabolic equivalents of task

    PA

    physical activity

    PGC-1α

    proliferator-activated receptor γ coactivator 1-α

    PI

    physical inactivity

    SB

    sedentary behavior

    SIRT

    sirtuin

    T2DM

    type 2 diabetes mellitus

    VO2

    oxygen consumption

    Footnotes

    Correspondence to Carl J. Lavie, MD, John Ochsner Heart and Vascular Institute, Ochsner Clinical School, The University of Queensland School of Medicine, 1514 Jefferson Hwy, New Orleans, LA 70121. Email

    References

    • 1. Lavie CJ, Arena R, Swift DL, Johannsen NM, Sui X, Lee DC, Earnest CP, Church TS, O’Keefe JH, Milani RV, Blair SN . Exercise and the cardiovascular system: clinical science and cardiovascular outcomes.Circ Res. 2015; 117:207–219. doi: 10.1161/CIRCRESAHA.117.305205LinkGoogle Scholar
    • 2. Fletcher GF, Landolfo C, Niebauer J, Ozemek C, Arena R, Lavie CJ . Promoting physical activity and exercise: JACC health promotion series.J Am Coll Cardiol. 2018; 72:1622–1639. doi: 10.1016/j.jacc.2018.08.2141CrossrefMedlineGoogle Scholar
    • 3. Wisloff U, Lavie CJ . Taking physical activity, exercise, and fitness to a higher level.Prog Cardiovasc Dis. 2017; 60:1–2. doi: 10.1016/j.pcad.2017.06.002CrossrefMedlineGoogle Scholar
    • 4. Turco JV, Inal-Veith A, Fuster V . Cardiovascular health promotion: an issue that can no longer wait.J Am Coll Cardiol. 2018; 72:908–913. doi: 10.1016/j.jacc.2018.07.007CrossrefMedlineGoogle Scholar
    • 5. Tremblay MS, Aubert S, Barnes JD, Saunders TJ, Carson V, Latimer-Cheung AE, Chastin SFM, Altenburg TM, Chinapaw MJM ; SBRN Terminology Consensus Project Participants. Sedentary behavior research network (SBRN) - terminology consensus project process and outcome.Int J Behav Nutr Phys Act. 2017; 14:75. doi: 10.1186/s12966-017-0525-8CrossrefMedlineGoogle Scholar
    • 6. Gibbs BB, Hergenroeder AL, Katzmarzyk PT, Lee IM, Jakicic JM . Definition, measurement, and health risks associated with sedentary behavior.Med Sci Sports Exerc. 2015; 47:1295–1300. doi: 10.1249/MSS.0000000000000517CrossrefMedlineGoogle Scholar
    • 7. US Department of Health and Human Services. 2008 Physical Activity Guidelines for Americans: US Department of Health and Human Services, Centers for Disease Control and Prevention. 2008. https://health.gov/paguidelines/2008/. Accessed November 1, 2018.Google Scholar
    • 8. Young DR, Hivert MF, Alhassan S, Camhi SM, Ferguson JF, Katzmarzyk PT, Lewis CE, Owen N, Perry CK, Siddique J, Yong CM ; Physical Activity Committee of the Council on Lifestyle and Cardiometabolic Health; Council on Clinical Cardiology; Council on Epidemiology and Prevention; Council on Functional Genomics and Translational Biology; and Stroke Council. Sedentary behavior and cardiovascular morbidity and mortality: a science advisory from the American Heart Association.Circulation. 2016; 134:e262–e279. doi: 10.1161/CIR.0000000000000440LinkGoogle Scholar
    • 9. Colberg SR, Sigal RJ, Yardley JE, Riddell MC, Dunstan DW, Dempsey PC, Horton ES, Castorino K, Tate DF . Physical activity/exercise and diabetes: a position statement of the American Diabetes Association.Diabetes Care. 2016; 39:2065–2079. doi: 10.2337/dc16-1728CrossrefMedlineGoogle Scholar
    • 10. UK Department of Health. Start Active, Stay Active: A Report on Physical Activity for Health From the Four Home Country’s Chief Medical Officers. London, United Kingdom: Crown Copyright; 2011.Google Scholar
    • 11. Commonwealth of Australia. Australia’s Physical Activity and Sedentary Behaviour Guidelines for Adults (18–64Years). Canberra, Australia: Australian Government Department of Health; 2014.Google Scholar
    • 12. Hamilton MT, Hamilton DG, Zderic TW . Exercise physiology versus inactivity physiology: an essential concept for understanding lipoprotein lipase regulation.Exerc Sport Sci Rev. 2004; 32:161–166.CrossrefMedlineGoogle Scholar
    • 13. Hamilton MT, Hamilton DG, Zderic TW . Role of low energy expenditure and sitting in obesity, metabolic syndrome, type 2 diabetes, and cardiovascular disease.Diabetes. 2007; 56:2655–2667. doi: 10.2337/db07-0882CrossrefMedlineGoogle Scholar
    • 14. Bey L, Hamilton MT . Suppression of skeletal muscle lipoprotein lipase activity during physical inactivity: a molecular reason to maintain daily low-intensity activity.J Physiol. 2003; 551:673–682. doi: 10.1113/jphysiol.2003.045591CrossrefMedlineGoogle Scholar
    • 15. Bey L, Akunuri N, Zhao P, Hoffman EP, Hamilton DG, Hamilton MT . Patterns of global gene expression in rat skeletal muscle during unloading and low-intensity ambulatory activity.Physiol Genomics. 2003; 13:157–167. doi: 10.1152/physiolgenomics.00001.2002CrossrefMedlineGoogle Scholar
    • 16. Joseph AM, Adhihetty PJ, Buford TW, Wohlgemuth SE, Lees HA, Nguyen LM, Aranda JM, Sandesara BD, Pahor M, Manini TM, Marzetti E, Leeuwenburgh C . The impact of aging on mitochondrial function and biogenesis pathways in skeletal muscle of sedentary high- and low-functioning elderly individuals.Aging Cell. 2012; 11:801–809. doi: 10.1111/j.1474-9726.2012.00844.xCrossrefMedlineGoogle Scholar
    • 17. Pulsford RM, Blackwell J, Hillsdon M, Kos K . Intermittent walking, but not standing, improves postprandial insulin and glucose relative to sustained sitting: a randomised cross-over study in inactive middle-aged men.J Sci Med Sport. 2017; 20:278–283. doi: 10.1016/j.jsams.2016.08.012CrossrefMedlineGoogle Scholar
    • 18. Latouche C, Jowett JB, Carey AL, Bertovic DA, Owen N, Dunstan DW, Kingwell BA . Effects of breaking up prolonged sitting on skeletal muscle gene expression.J Appl Physiol (1985). 2013; 114:453–460. doi: 10.1152/japplphysiol.00978.2012CrossrefMedlineGoogle Scholar
    • 19. 2018 Physical Activity Guidelines Advisory Committee. Physical Activity Guidelines Advisory Committee Scientific Report. Washington, DC: U.S. Department of Health and Human Services; 2018.Google Scholar
    • 20. Katzmarzyk PT, Church TS, Craig CL, Bouchard C . Sitting time and mortality from all causes, cardiovascular disease, and cancer.Med Sci Sports Exerc. 2009; 41:998–1005. doi: 10.1249/MSS.0b013e3181930355CrossrefMedlineGoogle Scholar
    • 21. Dunstan DW, Barr EL, Healy GN, Salmon J, Shaw JE, Balkau B, Magliano DJ, Cameron AJ, Zimmet PZ, Owen N . Television viewing time and mortality: the Australian Diabetes, Obesity and Lifestyle Study (AusDiab).Circulation. 2010; 121:384–391. doi: 10.1161/CIRCULATIONAHA.109.894824LinkGoogle Scholar
    • 22. Sun JW, Zhao LG, Yang Y, Ma X, Wang YY, Xiang YB . Association between television viewing time and all-cause mortality: a meta-analysis of cohort studies.Am J Epidemiol. 2015; 182:908–916. doi: 10.1093/aje/kwv164CrossrefMedlineGoogle Scholar
    • 23. Chau JY, Grunseit AC, Chey T, Stamatakis E, Brown WJ, Matthews CE, Bauman AE, van der Ploeg HP . Daily sitting time and all-cause mortality: a meta-analysis.PLoS One. 2013; 8:e80000. doi: 10.1371/journal.pone.0080000CrossrefMedlineGoogle Scholar
    • 24. Pandey A, Salahuddin U, Garg S, Ayers C, Kulinski J, Anand V, Mayo H, Kumbhani DJ, de Lemos J, Berry JD . Continuous dose-response association between sedentary time and risk for cardiovascular disease: a meta-analysis.JAMA Cardiol. 2016; 1:575–583. doi: 10.1001/jamacardio.2016.1567CrossrefMedlineGoogle Scholar
    • 25. Biswas A, Oh PI, Faulkner GE, Bajaj RR, Silver MA, Mitchell MS, Alter DA . Sedentary time and its association with risk for disease incidence, mortality, and hospitalization in adults: a systematic review and meta-analysis.Ann Intern Med. 2015; 162:123–132. doi: 10.7326/M14-1651CrossrefMedlineGoogle Scholar
    • 26. Ekelund U, Steene-Johannessen J, Brown WJ, Fagerland MW, Owen N, Powell KE, Bauman A, Lee IM ; Lancet Physical Activity Series 2 Executive Committee; Lancet Sedentary Behaviour Working Group. Does physical activity attenuate, or even eliminate, the detrimental association of sitting time with mortality? A harmonised meta-analysis of data from more than 1 million men and women.Lancet. 2016; 388:1302–1310. doi: 10.1016/S0140-6736(16)30370-1CrossrefMedlineGoogle Scholar
    • 27. Lee DC, Pate RR, Lavie CJ, Sui X, Church TS, Blair SN . Leisure-time running reduces all-cause and cardiovascular mortality risk.J Am Coll Cardiol. 2014; 64:472–481. doi: 10.1016/j.jacc.2014.04.058CrossrefMedlineGoogle Scholar
    • 28. Lee IM, Shiroma EJ, Lobelo F, Puska P, Blair SN, Katzmarzyk PT ; Lancet Physical Activity Series Working Group. Effect of physical inactivity on major non-communicable diseases worldwide: an analysis of burden of disease and life expectancy.Lancet. 2012; 380:219–229. doi: 10.1016/S0140-6736(12)61031-9CrossrefMedlineGoogle Scholar
    • 29. World Health Organization.GlobalRecommendations on Physical Activity for Health. Geneva;2010.Google Scholar
    • 30. Dumith SC, Hallal PC, Reis RS, Kohl HW . Worldwide prevalence of physical inactivity and its association with human development index in 76 countries.Prev Med. 2011; 53:24–28. doi: 10.1016/j.ypmed.2011.02.017CrossrefMedlineGoogle Scholar
    • 31. Oggioni C, Lara J, Wells JC, Soroka K, Siervo M . Shifts in population dietary patterns and physical inactivity as determinants of global trends in the prevalence of diabetes: an ecological analysis.Nutr Metab Cardiovasc Dis. 2014; 24:1105–1111. doi: 10.1016/j.numecd.2014.05.005CrossrefMedlineGoogle Scholar
    • 32. Borrell LN . The effects of smoking and physical inactivity on advancing mortality in U.S. adults.Ann Epidemiol. 2014; 24:484–487. doi: 10.1016/j.annepidem.2014.02.016CrossrefMedlineGoogle Scholar
    • 33. Mayer-Davis EJ, Dabelea D, Lawrence JM . Incidence trends of type 1 and type 2 diabetes among youths, 2002-2012.N Engl J Med. 2017; 377:301. doi: 10.1056/NEJMc1706291CrossrefMedlineGoogle Scholar
    • 34. Saunders RP, Dowda M, Mciver K, McDonald SM, Pate RR . Physical and social contexts of physical activity behaviors of fifth and seventh grade youth.J Sch Health. 2018; 88:122–131. doi: 10.1111/josh.12587CrossrefMedlineGoogle Scholar
    • 35. Kallio P, Pahkala K, Heinonen OJ, Tammelin T, Hirvensalo M, Telama R, Juonala M, Magnussen CG, Rovio S, Helajärvi H, Hutri-Kähönen N, Viikari J, Raitakari OT . Physical inactivity from youth to adulthood and risk of impaired glucose metabolism.Med Sci Sports Exerc. 2018; 50:1192–1198. doi: 10.1249/MSS.0000000000001555CrossrefMedlineGoogle Scholar
    • 36. Santos-Parker JR, LaRocca TJ, Seals DR . Aerobic exercise and other healthy lifestyle factors that influence vascular aging.Adv Physiol Educ. 2014; 38:296–307. doi: 10.1152/advan.00088.2014CrossrefMedlineGoogle Scholar
    • 37. Bleeker MW, De Groot PC, Poelkens F, Rongen GA, Smits P, Hopman MT . Vascular adaptation to 4 wk of deconditioning by unilateral lower limb suspension.Am J Physiol Heart Circ Physiol. 2005; 288:H1747–H1755. doi: 10.1152/ajpheart.00966.2004CrossrefMedlineGoogle Scholar
    • 38. Bleeker MW, De Groot PC, Rongen GA, Rittweger J, Felsenberg D, Smits P, Hopman MT . Vascular adaptation to deconditioning and the effect of an exercise countermeasure: results of the Berlin Bed Rest study.J Appl Physiol (1985). 2005; 99:1293–1300. doi: 10.1152/japplphysiol.00118.2005CrossrefMedlineGoogle Scholar
    • 39. Birk GK, Dawson EA, Timothy Cable N, Green DJ, Thijssen DH . Effect of unilateral forearm inactivity on endothelium-dependent vasodilator function in humans.Eur J Appl Physiol. 2013; 113:933–940. doi: 10.1007/s00421-012-2505-7CrossrefMedlineGoogle Scholar
    • 40. Boyle LJ, Credeur DP, Jenkins NT, Padilla J, Leidy HJ, Thyfault JP, Fadel PJ . Impact of reduced daily physical activity on conduit artery flow-mediated dilation and circulating endothelial microparticles.J Appl Physiol (1985). 2013; 115:1519–1525. doi: 10.1152/japplphysiol.00837.2013CrossrefMedlineGoogle Scholar
    • 41. Eskurza I, Monahan KD, Robinson JA, Seals DR . Effect of acute and chronic ascorbic acid on flow-mediated dilatation with sedentary and physically active human ageing.J Physiol. 2004; 556:315–324. doi: 10.1113/jphysiol.2003.057042CrossrefMedlineGoogle Scholar
    • 42. Moreau KL, Gavin KM, Plum AE, Seals DR . Oxidative stress explains differences in large elastic artery compliance between sedentary and habitually exercising postmenopausal women.Menopause. 2006; 13:951–958. doi: 10.1097/01.gme.0000243575.09065.48CrossrefMedlineGoogle Scholar
    • 43. de Picciotto NE, Gano LB, Johnson LC, Martens CR, Sindler AL, Mills KF, Imai S, Seals DR . Nicotinamide mononucleotide supplementation reverses vascular dysfunction and oxidative stress with aging in mice.Aging Cell. 2016; 15:522–530. doi: 10.1111/acel.12461CrossrefMedlineGoogle Scholar
    • 44. Durrant JR, Seals DR, Connell ML, Russell MJ, Lawson BR, Folian BJ, Donato AJ, Lesniewski LA . Voluntary wheel running restores endothelial function in conduit arteries of old mice: direct evidence for reduced oxidative stress, increased superoxide dismutase activity and down-regulation of NADPH oxidase.J Physiol. 2009; 587:3271–3285. doi: 10.1113/jphysiol.2009.169771CrossrefMedlineGoogle Scholar
    • 45. Moreau KL, Ozemek C . Vascular adaptations to habitual exercise in older adults: time for the sex talk.Exerc Sport Sci Rev. 2017; 45:116–123. doi: 10.1249/JES.0000000000000104CrossrefMedlineGoogle Scholar
    • 46. Borlaug BA, Kass DA . Ventricular-vascular interaction in heart failure.Cardiol Clin. 2011; 29:447–459. doi: 10.1016/j.ccl.2011.06.004CrossrefMedlineGoogle Scholar
    • 47. Borlaug BA, Redfield MM, Melenovsky V, Kane GC, Karon BL, Jacobsen SJ, Rodeheffer RJ . Longitudinal changes in left ventricular stiffness: a community-based study.Circ Heart Fail. 2013; 6:944–952. doi: 10.1161/CIRCHEARTFAILURE.113.000383LinkGoogle Scholar
    • 48. Florido R, Kwak L, Lazo M, Nambi V, Ahmed HM, Hegde SM, Gerstenblith G, Blumenthal RS, Ballantyne CM, Selvin E, Folsom AR, Coresh J, Ndumele CE . Six-year changes in physical activity and the risk of incident heart failure: ARIC study.Circulation. 2018; 137:2142–2151. doi: 10.1161/CIRCULATIONAHA.117.030226LinkGoogle Scholar
    • 49. O’Donovan G, Stamatakis E, Stensel DJ, Hamer M . The importance of vigorous-intensity leisure-time physical activity in reducing cardiovascular disease mortality risk in the obese.Mayo Clin Proc. 2018; 93:1096–1103. doi: 10.1016/j.mayocp.2018.01.016CrossrefMedlineGoogle Scholar
    • 50. Nes BM, Gutvik CR, Lavie CJ, Nauman J, Wisløff U . Personalized activity intelligence (PAI) for prevention of cardiovascular disease and promotion of physical activity.Am J Med. 2017; 130:328–336. doi: 10.1016/j.amjmed.2016.09.031CrossrefMedlineGoogle Scholar
    • 51. El Saadany T, Richard A, Wanner M, Rohrmann S . Sex-specific effects of leisure-time physical activity on cause-specific mortality in NHANES III.Prev Med. 2017; 101:53–59. doi: 10.1016/j.ypmed.2017.05.029CrossrefMedlineGoogle Scholar
    • 52. Kubota Y, Iso H, Yamagishi K, Sawada N, Tsugane S ; JPHC Study Group. Daily total physical activity and incident stroke: the Japan public health center-based prospective study.Stroke. 2017; 48:1730–1736. doi: 10.1161/STROKEAHA.117.017560LinkGoogle Scholar
    • 53. Lear SA, Hu W, Rangarajan S, et al . The effect of physical activity on mortality and cardiovascular disease in 130 000 people from 17 high-income, middle-income, and low-income countries: the PURE study.Lancet. 2017; 390:2643–2654. doi: 10.1016/S0140-6736(17)31634-3CrossrefMedlineGoogle Scholar
    • 54. Fishman EI, Steeves JA, Zipunnikov V, Koster A, Berrigan D, Harris TA, Murphy R . Association between objectively measured physical activity and mortality in NHANES.Med Sci Sports Exerc. 2016; 48:1303–1311. doi: 10.1249/MSS.0000000000000885CrossrefMedlineGoogle Scholar
    • 55. Soares-Miranda L, Siscovick DS, Psaty BM, Longstreth WT, Mozaffarian D . Physical activity and risk of coronary heart disease and stroke in older adults: the cardiovascular health study.Circulation. 2016; 133:147–155. doi: 10.1161/CIRCULATIONAHA.115.018323LinkGoogle Scholar
    • 56. Bell EJ, Lutsey PL, Windham BG, Folsom AR . Physical activity and cardiovascular disease in African Americans in Atherosclerosis Risk in Communities.Med Sci Sports Exerc. 2013; 45:901–907. doi: 10.1249/MSS.0b013e31827d87ecCrossrefMedlineGoogle Scholar
    • 57. Shortreed SM, Peeters A, Forbes AB . Estimating the effect of long-term physical activity on cardiovascular disease and mortality: evidence from the Framingham Heart Study.Heart. 2013; 99:649–654. doi: 10.1136/heartjnl-2012-303461CrossrefMedlineGoogle Scholar
    • 58. Gulsvik AK, Thelle DS, Samuelsen SO, Myrstad M, Mowé M, Wyller TB . Ageing, physical activity and mortality–a 42-year follow-up study.Int J Epidemiol. 2012; 41:521–530. doi: 10.1093/ije/dyr205CrossrefMedlineGoogle Scholar
    • 59. Wen CP, Wai JP, Tsai MK, Yang YC, Cheng TY, Lee MC, Chan HT, Tsao CK, Tsai SP, Wu X . Minimum amount of physical activity for reduced mortality and extended life expectancy: a prospective cohort study.Lancet. 2011; 378:1244–1253. doi: 10.1016/S0140-6736(11)60749-6CrossrefMedlineGoogle Scholar
    • 60. Tjønna AE, Lund Nilsen TI, Slørdahl SA, Vatten L, Wisløff U . The association of metabolic clustering and physical activity with cardiovascular mortality: the HUNT study in Norway.J Epidemiol Community Health. 2010; 64:690–695. doi: 10.1136/jech.2008.084467CrossrefMedlineGoogle Scholar
    • 61. Wisløff U, Nilsen TI, Drøyvold WB, Mørkved S, Slørdahl SA, Vatten LJ . A single weekly bout of exercise may reduce cardiovascular mortality: how little pain for cardiac gain? ‘The HUNT study, Norway’.Eur J Cardiovasc Prev Rehabil. 2006; 13:798–804. doi: 10.1097/01.hjr.0000216548.84560.acCrossrefMedlineGoogle Scholar
    • 62. 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.CrossrefMedlineGoogle Scholar
    • 63. 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.CrossrefMedlineGoogle Scholar
    • 64. 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.CrossrefMedlineGoogle Scholar
    • 65. Loprinzi PD, Lee H, Cardinal BJ, Crespo CJ, Andersen RE, Smit E . The relationship of actigraph accelerometer cut-points for estimating physical activity with selected health outcomes: results from NHANES 2003-06.Res Q Exerc Sport. 2012; 83:422–430. doi: 10.1080/02701367.2012.10599877MedlineGoogle Scholar
    • 66. Arena R, Ozemek C, Laddu D, Campbell T, Rouleau CR, Standley R, Bond S, Abril EP, Hills AP, Lavie CJ . Applying precision medicine to healthy living for the prevention and treatment of cardiovascular disease.Curr Probl Cardiol. 2018; 43:448–483. doi: 10.1016/j.cpcardiol.2018.06.001CrossrefMedlineGoogle Scholar
    • 67. Ross R, Hudson R, Day AG, Lam M . Dose-response effects of exercise on abdominal obesity and risk factors for cardiovascular disease in adults: study rationale, design and methods.Contemp Clin Trials. 2013; 34:155–160. doi: 10.1016/j.cct.2012.10.010CrossrefMedlineGoogle Scholar
    • 68. 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.2014.03.062CrossrefMedlineGoogle Scholar
    • 69. Seals DR, Desouza CA, Donato AJ, Tanaka H . Habitual exercise and arterial aging.J Appl Physiol (1985). 2008; 105:1323–1332. doi: 10.1152/japplphysiol.90553.2008CrossrefMedlineGoogle Scholar
    • 70. Wisløff U, Ellingsen Ø, Kemi OJ . High-intensity interval training to maximize cardiac benefits of exercise training?Exerc Sport Sci Rev. 2009; 37:139–146. doi: 10.1097/JES.0b013e3181aa65fcCrossrefMedlineGoogle Scholar
    • 71. 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.0000465CrossrefMedlineGoogle Scholar
    • 72. Barrientos A, Casademont J, Rötig A, Miró O, Urbano-Márquez A, Rustin P, Cardellach F . Absence of relationship between the level of electron transport chain activities and aging in human skeletal muscle.Biochem Biophys Res Commun. 1996; 229:536–539. doi: 10.1006/bbrc.1996.1839CrossrefMedlineGoogle Scholar
    • 73. Kang C, Yeo D, Ji LL . Muscle immobilization activates mitophagy and disrupts mitochondrial dynamics in mice.Acta Physiol (Oxf). 2016; 218:188–197. doi: 10.1111/apha.12690CrossrefMedlineGoogle Scholar
    • 74. Conley KE, Amara CE, Bajpeyi S, Costford SR, Murray K, Jubrias SA, Arakaki L, Marcinek DJ, Smith SR . Higher mitochondrial respiration and uncoupling with reduced electron transport chain content in vivo in muscle of sedentary versus active subjects.J Clin Endocrinol Metab. 2013; 98:129–136. doi: 10.1210/jc.2012-2967CrossrefMedlineGoogle Scholar
    • 75. Konopka AR, Sreekumaran Nair K . Mitochondrial and skeletal muscle health with advancing age.Mol Cell Endocrinol. 2013; 379:19–29. doi: 10.1016/j.mce.2013.05.008CrossrefMedlineGoogle Scholar
    • 76. Short KR, Vittone JL, Bigelow ML, Proctor DN, Rizza RA, Coenen-Schimke JM, Nair KS . Impact of aerobic exercise training on age-related changes in insulin sensitivity and muscle oxidative capacity.Diabetes. 2003; 52:1888–1896.CrossrefMedlineGoogle Scholar
    • 77. Cobley JN, Bartlett JD, Kayani A, Murray SW, Louhelainen J, Donovan T, Waldron S, Gregson W, Burniston JG, Morton JP, Close GL . PGC-1α transcriptional response and mitochondrial adaptation to acute exercise is maintained in skeletal muscle of sedentary elderly males.Biogerontology. 2012; 13:621–631. doi: 10.1007/s10522-012-9408-1CrossrefMedlineGoogle Scholar
    • 78. Lanza IR, Short DK, Short KR, Raghavakaimal S, Basu R, Joyner MJ, McConnell JP, Nair KS . Endurance exercise as a countermeasure for aging.Diabetes. 2008; 57:2933–2942. doi: 10.2337/db08-0349CrossrefMedlineGoogle Scholar
    • 79. Meinild Lundby AK, Jacobs RA, Gehrig S, de Leur J, Hauser M, Bonne TC, Fluck D, Dandanell S, Kirk N, Kaech A, Ziegler U, Larsen S, Lundby C . Exercise training increases skeletal muscle mitochondrial volume density by enlargement of existing mitochondria and not de novo biogenesis.Acta Physiol (Oxf). 2018; 222:e12905.CrossrefGoogle Scholar
    • 80. Safdar A, Hamadeh MJ, Kaczor JJ, Raha S, Debeer J, Tarnopolsky MA . Aberrant mitochondrial homeostasis in the skeletal muscle of sedentary older adults.PLoS One. 2010; 5:e10778. doi: 10.1371/journal.pone.0010778CrossrefMedlineGoogle Scholar
    • 81. Powers SK, Wiggs MP, Duarte JA, Zergeroglu AM, Demirel HA . Mitochondrial signaling contributes to disuse muscle atrophy.Am J Physiol Endocrinol Metab. 2012; 303:E31–E39. doi: 10.1152/ajpendo.00609.2011CrossrefMedlineGoogle Scholar
    • 82. Haddad F, Adams GR, Bodell PW, Baldwin KM . Isometric resistance exercise fails to counteract skeletal muscle atrophy processes during the initial stages of unloading.J Appl Physiol (1985). 2006; 100:433–441. doi: 10.1152/japplphysiol.01203.2005CrossrefMedlineGoogle Scholar
    • 83. Reid N, Healy GN, Gianoudis J, Formica M, Gardiner PA, Eakin EE, Nowson CA, Daly RM . Association of sitting time and breaks in sitting with muscle mass, strength, function, and inflammation in community-dwelling older adults.Osteoporos Int. 2018; 29:1341–1350. doi: 10.1007/s00198-018-4428-6CrossrefMedlineGoogle Scholar
    • 84. Andersen JL . Muscle fibre type adaptation in the elderly human muscle.Scand J Med Sci Sports. 2003; 13:40–47.CrossrefMedlineGoogle Scholar
    • 85. Gries KJ, Raue U, Perkins RK, Lavin KM, Overstreet BS, D’Acquisto LJ, Graham B, Finch WH, Kaminsky LA, Trappe TA, Trappe SW . Cardiovascular and skeletal muscle health with lifelong exercise.J Appl Physiol (1985). 2018; 125:1636–1645.CrossrefMedlineGoogle Scholar
    • 86. Konopka AR, Douglass MD, Kaminsky LA, Jemiolo B, Trappe TA, Trappe S, Harber MP . Molecular adaptations to aerobic exercise training in skeletal muscle of older women.J Gerontol A Biol Sci Med Sci. 2010; 65:1201–1207. doi: 10.1093/gerona/glq109CrossrefMedlineGoogle Scholar
    • 87. Wright KJ, Thomas MM, Betik AC, Belke D, Hepple RT . Exercise training initiated in late middle age attenuates cardiac fibrosis and advanced glycation end-product accumulation in senescent rats.Exp Gerontol. 2014; 50:9–18. doi: 10.1016/j.exger.2013.11.006CrossrefMedlineGoogle Scholar
    • 88. Hieda M, Howden E, Shibata S, Fujimoto N, Bhella PS, Hastings JL, Tarumi T, Sarma S, Fu Q, Zhang R, Levine BD . Impact of lifelong exercise training dose on ventricular-arterial coupling.Circulation. 2018; 138:2638–2647. doi: 10.1161/CIRCULATIONAHA.118.035116LinkGoogle Scholar
    • 89. Xiao RP, Spurgeon HA, O’Connor F, Lakatta EG . Age-associated changes in beta-adrenergic modulation on rat cardiac excitation-contraction coupling.J Clin Invest. 1994; 94:2051–2059. doi: 10.1172/JCI117559CrossrefMedlineGoogle Scholar
    • 90. Davies CH, Ferrara N, Harding SE . Beta-adrenoceptor function changes with age of subject in myocytes from non-failing human ventricle.Cardiovasc Res. 1996; 31:152–156.MedlineGoogle Scholar
    • 91. Janczewski AM, Lakatta EG . Modulation of sarcoplasmic reticulum Ca(2+) cycling in systolic and diastolic heart failure associated with aging.Heart Fail Rev. 2010; 15:431–445. doi: 10.1007/s10741-010-9167-5CrossrefMedlineGoogle Scholar
    • 92. Kemi OJ, Ellingsen O, Ceci M, Grimaldi S, Smith GL, Condorelli G, Wisløff U . Aerobic interval training enhances cardiomyocyte contractility and Ca2+ cycling by phosphorylation of CaMKII and Thr-17 of phospholamban.J Mol Cell Cardiol. 2007; 43:354–361. doi: 10.1016/j.yjmcc.2007.06.013CrossrefMedlineGoogle Scholar
    • 93. Kemi OJ, Wisløff U . Mechanisms of exercise-induced improvements in the contractile apparatus of the mammalian myocardium.Acta Physiol (Oxf). 2010; 199:425–439. doi: 10.1111/j.1748-1716.2010.02132.xCrossrefMedlineGoogle Scholar
    • 94. Taddei S, Virdis A, Ghiadoni L, Salvetti G, Bernini G, Magagna A, Salvetti A . Age-related reduction of NO availability and oxidative stress in humans.Hypertension. 2001; 38:274–279.LinkGoogle Scholar
    • 95. Ryan MJ . An update on immune system activation in the pathogenesis of hypertension.Hypertension. 2013; 62:226–230. doi: 10.1161/HYPERTENSIONAHA.113.00603LinkGoogle Scholar
    • 96. Pierce GL, Donato AJ, LaRocca TJ, Eskurza I, Silver AE, Seals DR . Habitually exercising older men do not demonstrate age-associated vascular endothelial oxidative stress.Aging Cell. 2011; 10:1032–1037. doi: 10.1111/j.1474-9726.2011.00748.xCrossrefMedlineGoogle Scholar
    • 97. Rossman MJ, Kaplon RE, Hill SD, McNamara MN, Santos-Parker JR, Pierce GL, Seals DR, Donato AJ . Endothelial cell senescence with aging in healthy humans: prevention by habitual exercise and relation to vascular endothelial function.Am J Physiol Heart Circ Physiol. 2017; 313:H890–H895. doi: 10.1152/ajpheart.00416.2017CrossrefMedlineGoogle Scholar
    • 98. Buttrick PM, Kaplan M, Leinwand LA, Scheuer J . Alterations in gene expression in the rat heart after chronic pathological and physiological loads.J Mol Cell Cardiol. 1994; 26:61–67. doi: 10.1006/jmcc.1994.1008CrossrefMedlineGoogle Scholar
    • 99. Stewart LK, Flynn MG, Campbell WW, Craig BA, Robinson JP, Timmerman KL, McFarlin BK, Coen PM, Talbert E . The influence of exercise training on inflammatory cytokines and C-reactive protein.Med Sci Sports Exerc. 2007; 39:1714–1719. doi: 10.1249/mss.0b013e31811ece1cCrossrefMedlineGoogle Scholar
    • 100. Gioscia-Ryan RA, Battson ML, Cuevas LM, Zigler MC, Sindler AL, Seals DR . Voluntary aerobic exercise increases arterial resilience and mitochondrial health with aging in mice.Aging (Albany NY). 2016; 8:2897–2914. doi: 10.18632/aging.101099CrossrefMedlineGoogle Scholar
    • 101. Harber MP, Kaminsky LA, Arena R, Blair SN, Franklin BA, Myers J, Ross R . Impact of cardiorespiratory fitness on all-cause and disease-specific mortality: advances since 2009.Prog Cardiovasc Dis. 2017; 60:11–20. doi: 10.1016/j.pcad.2017.03.001CrossrefMedlineGoogle Scholar
    • 102. Nauman J, Tauschek LC, Kaminsky LA, Nes BM, Wisløff U . Global fitness levels: findings from a web-based surveillance report.Prog Cardiovasc Dis. 2017; 60:78–88. doi: 10.1016/j.pcad.2017.01.009CrossrefMedlineGoogle Scholar
    • 103. Ross R, de Lannoy L, Stotz PJ . Separate effects of intensity and amount of exercise on interindividual cardiorespiratory fitness response.Mayo Clin Proc. 2015; 90:1506–1514. doi: 10.1016/j.mayocp.2015.07.024CrossrefMedlineGoogle Scholar
    • 104. Bouchard C, Sarzynski MA, Rice TK, Kraus WE, Church TS, Sung YJ, Rao DC, Rankinen T . Genomic predictors of the maximal O2 uptake response to standardized exercise training programs.J Appl Physiol (1985). 2011; 110:1160–1170. doi: 10.1152/japplphysiol.00973.2010CrossrefMedlineGoogle Scholar
    • 105. Balady GJ, Arena R, Sietsema K, Myers J, Coke L, Fletcher GF, Forman D, Franklin B, Guazzi M, Gulati M, Keteyian SJ, Lavie CJ, Macko R, Mancini D, Milani RV ; American Heart Association Exercise, Cardiac Rehabilitation, and Prevention Committee of the Council on Clinical Cardiology; Council on Epidemiology and Prevention; Council on Peripheral Vascular Disease; Interdisciplinary Council on Quality of Care and Outcomes Research. Clinician’s guide to cardiopulmonary exercise testing in adults: a scientific statement from the American Heart Association.Circulation. 2010; 122:191–225. doi: 10.1161/CIR.0b013e3181e52e69LinkGoogle Scholar
    • 106. Franklin BA, Lavie CJ, Squires RW, Milani RV . Exercise-based cardiac rehabilitation and improvements in cardiorespiratory fitness: implications regarding patient benefit.Mayo Clin Proc. 2013; 88:431–437. doi: 10.1016/j.mayocp.2013.03.009CrossrefMedlineGoogle Scholar
    • 107. De Schutter A, Kachur S, Lavie CJ, Menezes A, Shum KK, Bangalore S, Arena R, Milani RV . Cardiac rehabilitation fitness changes and subsequent survival.Eur Heart J Qual Care Clin Outcomes. 2018; 4:173–179. doi: 10.1093/ehjqcco/qcy018MedlineGoogle Scholar
    • 108. Forman DE, Fleg JL, Kitzman DW, Brawner CA, Swank AM, McKelvie RS, Clare RM, Ellis SJ, Dunlap ME, Bittner V . 6-min walk test provides prognostic utility comparable to cardiopulmonary exercise testing in ambulatory outpatients with systolic heart failure.J Am Coll Cardiol. 2012; 60:2653–2661. doi: 10.1016/j.jacc.2012.08.1010CrossrefMedlineGoogle Scholar
    • 109. Whelton SP, Chin A, Xin X, He J . Effect of aerobic exercise on blood pressure: a meta-analysis of randomized, controlled trials.Ann Intern Med. 2002; 136:493–503.CrossrefMedlineGoogle Scholar
    • 110. Abramson JL, Vaccarino V . Relationship between physical activity and inflammation among apparently healthy middle-aged and older US adults.Arch Intern Med. 2002; 162:1286–1292.CrossrefMedlineGoogle Scholar
    • 111. Perini R, Veicsteinas A . Heart rate variability and autonomic activity at rest and during exercise in various physiological conditions.Eur J Appl Physiol. 2003; 90:317–325. doi: 10.1007/s00421-003-0953-9CrossrefMedlineGoogle Scholar
    • 112. Beck DT, Martin JS, Casey DP, Braith RW . Exercise training reduces peripheral arterial stiffness and myocardial oxygen demand in young prehypertensive subjects.Am J Hypertens. 2013; 26:1093–1102. doi: 10.1093/ajh/hpt080CrossrefMedlineGoogle Scholar
    • 113. Clarkson P, Montgomery HE, Mullen MJ, Donald AE, Powe AJ, Bull T, Jubb M, World M, Deanfield JE . Exercise training enhances endothelial function in young men.J Am Coll Cardiol. 1999; 33:1379–1385.CrossrefMedlineGoogle Scholar
    • 114. Sugawara J, Miyachi M, Moreau KL, Dinenno FA, DeSouza CA, Tanaka H . Age-related reductions in appendicular skeletal muscle mass: association with habitual aerobic exercise status.Clin Physiol Funct Imaging. 2002; 22:169–172.CrossrefMedlineGoogle Scholar
    • 115. Holloszy JO . Exercise-induced increase in muscle insulin sensitivity.J Appl Physiol (1985). 2005; 99:338–343. doi: 10.1152/japplphysiol.00123.2005CrossrefMedlineGoogle Scholar
    • 116. Ohkawara K, Tanaka S, Miyachi M, Ishikawa-Takata K, Tabata I . A dose-response relation between aerobic exercise and visceral fat reduction: systematic review of clinical trials.Int J Obes (Lond). 2007; 31:1786–1797. doi: 10.1038/sj.ijo.0803683CrossrefMedlineGoogle Scholar
    • 117. Lakka TA, Venäläinen JM, Rauramaa R, Salonen R, Tuomilehto J, Salonen JT . Relation of leisure-time physical activity and cardiorespiratory fitness to the risk of acute myocardial infarction.N Engl J Med. 1994; 330:1549–1554. doi: 10.1056/NEJM199406023302201CrossrefMedlineGoogle Scholar
    • 118. Andersen P, Henriksson J . Capillary supply of the quadriceps femoris muscle of man: adaptive response to exercise.J Physiol. 1977; 270:677–690.CrossrefMedlineGoogle Scholar
    • 119. Koenig W, Sund M, Döring A, Ernst E . Leisure-time physical activity but not work-related physical activity is associated with decreased plasma viscosity. Results from a large population sample.Circulation. 1997; 95:335–341.LinkGoogle Scholar
    • 120. Wipfli BM, Rethorst CD, Landers DM . The anxiolytic effects of exercise: a meta-analysis of randomized trials and dose-response analysis.J Sport Exerc Psychol. 2008; 30:392–410.CrossrefMedlineGoogle Scholar
    • 121. King AC, Oman RF, Brassington GS, Bliwise DL, Haskell WL . Moderate-intensity exercise and self-rated quality of sleep in older adults. A randomized controlled trial.JAMA. 1997; 277:32–37.CrossrefMedlineGoogle Scholar
    • 122. Huai P, Xun H, Reilly KH, Wang Y, Ma W, Xi B . Physical activity and risk of hypertension: a meta-analysis of prospective cohort studies.Hypertension. 2013; 62:1021–1026. doi: 10.1161/HYPERTENSIONAHA.113.01965LinkGoogle Scholar
    • 123. Howe TE, Shea B, Dawson LJ, Downie F, Murray A, Ross C, Harbour RT, Caldwell LM, Creed G . Exercise for preventing and treating osteoporosis in postmenopausal women.Cochrane Database Syst Rev. 2011;(7):CD000333.MedlineGoogle Scholar
    • 124. Mammen G, Faulkner G . Physical activity and the prevention of depression: a systematic review of prospective studies.Am J Prev Med. 2013; 45:649–657. doi: 10.1016/j.amepre.2013.08.001CrossrefMedlineGoogle Scholar
    • 125. Bijlsma JW, Knahr K . Strategies for the prevention and management of osteoarthritis of the hip and knee.Best Pract Res Clin Rheumatol. 2007; 21:59–76. doi: 10.1016/j.berh.2006.08.013CrossrefMedlineGoogle Scholar
    • 126. LaMonte MJ, Barlow CE, Jurca R, Kampert JB, Church TS, Blair SN . Cardiorespiratory fitness is inversely associated with the incidence of metabolic syndrome: a prospective study of men and women.Circulation. 2005; 112:505–512. doi: 10.1161/CIRCULATIONAHA.104.503805LinkGoogle Scholar
    • 127. Barnes DE, Santos-Modesitt W, Poelke G, Kramer AF, Castro C, Middleton LE, Yaffe K . The Mental Activity and eXercise (MAX) trial: a randomized controlled trial to enhance cognitive function in older adults.JAMA Intern Med. 2013; 173:797–804. doi: 10.1001/jamainternmed.2013.189CrossrefMedlineGoogle Scholar
    • 128. Jeon CY, Lokken RP, Hu FB, van Dam RM . Physical activity of moderate intensity and risk of type 2 diabetes: a systematic review.Diabetes Care. 2007; 30:744–752. doi: 10.2337/dc06-1842CrossrefMedlineGoogle Scholar
    • 129. Lee IM . Physical activity and cancer prevention–data from epidemiologic studies.Med Sci Sports Exerc. 2003; 35:1823–1827. doi: 10.1249/01.MSS.0000093620.27893.23CrossrefMedlineGoogle Scholar
    • 130. Katzmarzyk PT, Lee IM, Martin CK, Blair SN . Epidemiology of physical activity and exercise training in the United States.Prog Cardiovasc Dis. 2017; 60:3–10. doi: 10.1016/j.pcad.2017.01.004CrossrefMedlineGoogle Scholar
    • 131. Sui X, Sarzynski MA, Lee DC, Kokkinos PF . Impact of changes in cardiorespiratory fitness on hypertension, dyslipidemia and survival: an overview of the epidemiological evidence.Prog Cardiovasc Dis. 2017; 60:56–66. doi: 10.1016/j.pcad.2017.02.006CrossrefMedlineGoogle Scholar
    • 132. Kodama S, Saito K, Tanaka S, Maki M, Yachi Y, Asumi M, Sugawara A, Totsuka K, Shimano H, Ohashi Y, Yamada N, Sone H . Cardiorespiratory fitness as a quantitative predictor of all-cause mortality and cardiovascular events in healthy men and women: a meta-analysis.JAMA. 2009; 301:2024–2035. doi: 10.1001/jama.2009.681CrossrefMedlineGoogle Scholar
    • 133. Swift DL, Lavie CJ, Johannsen NM, Arena R, Earnest CP, O’Keefe JH, Milani RV, Blair SN, Church TS . Physical activity, cardiorespiratory fitness, and exercise training in primary and secondary coronary prevention.Circ J. 2013; 77:281–292.CrossrefMedlineGoogle Scholar
    • 134. Berry JD, Willis B, Gupta S, Barlow CE, Lakoski SG, Khera A, Rohatgi A, de Lemos JA, Haskell W, Lloyd-Jones DM . Lifetime risks for cardiovascular disease mortality by cardiorespiratory fitness levels measured at ages 45, 55, and 65 years in men. The Cooper Center Longitudinal Study.J Am Coll Cardiol. 2011; 57:1604–1610. doi: 10.1016/j.jacc.2010.10.056CrossrefMedlineGoogle Scholar
    • 135. Blair SN, Kohl HW, Barlow CE, Paffenbarger RS, Gibbons LW, Macera CA . Changes in physical fitness and all-cause mortality. A prospective study of healthy and unhealthy men.JAMA. 1995; 273:1093–1098.CrossrefMedlineGoogle Scholar
    • 136. Lee DC, Sui X, Artero EG, Lee IM, Church TS, McAuley PA, Stanford FC, Kohl HW, Blair SN . Long-term effects of changes in cardiorespiratory fitness and body mass index on all-cause and cardiovascular disease mortality in men: the Aerobics Center Longitudinal Study.Circulation. 2011; 124:2483–2490. doi: 10.1161/CIRCULATIONAHA.111.038422LinkGoogle Scholar
    • 137. Erikssen G, Liestøl K, Bjørnholt J, Thaulow E, Sandvik L, Erikssen J . Changes in physical fitness and changes in mortality.Lancet. 1998; 352:759–762. doi: 10.1016/S0140-6736(98)02268-5CrossrefMedlineGoogle Scholar
    • 138. Guazzi M, Marenzi G, Alimento M, Contini M, Agostoni P . Improvement of alveolar-capillary membrane diffusing capacity with enalapril in chronic heart failure and counteracting effect of aspirin.Circulation. 1997; 95:1930–1936.LinkGoogle Scholar
    • 139. Lewis GD, Lachmann J, Camuso J, Lepore JJ, Shin J, Martinovic ME, Systrom DM, Bloch KD, Semigran MJ . Sildenafil improves exercise hemodynamics and oxygen uptake in patients with systolic heart failure.Circulation. 2007; 115:59–66. doi: 10.1161/CIRCULATIONAHA.106.626226LinkGoogle Scholar
    • 140. Redfield MM, Chen HH, Borlaug BA, et al ; RELAX Trial. Effect of phosphodiesterase-5 inhibition on exercise capacity and clinical status in heart failure with preserved ejection fraction: a randomized clinical trial.JAMA. 2013; 309:1268–1277. doi: 10.1001/jama.2013.2024CrossrefMedlineGoogle Scholar
    • 141. Edelmann F, Wachter R, Schmidt AG, et al ; Aldo-DHF Investigators. Effect of spironolactone on diastolic function and exercise capacity in patients with heart failure with preserved ejection fraction: the Aldo-DHF randomized controlled trial.JAMA. 2013; 309:781–791. doi: 10.1001/jama.2013.905CrossrefMedlineGoogle Scholar
    • 142. Borlaug BA, Anstrom KJ, Lewis GD, et al ; National Heart, Lung, and Blood Institute Heart Failure Clinical Research Network. Effect of inorganic nitrite vs placebo on exercise capacity among patients with heart failure with preserved ejection fraction: the INDIE-HFpEF randomized clinical trial.JAMA. 2018; 320:1764–1773. doi: 10.1001/jama.2018.14852CrossrefMedlineGoogle Scholar
    • 143. Katzmarzyk PT, Church TS, Blair SN . Cardiorespiratory fitness attenuates the effects of the metabolic syndrome on all-cause and cardiovascular disease mortality in men.Arch Intern Med. 2004; 164:1092–1097. doi: 10.1001/archinte.164.10.1092CrossrefMedlineGoogle Scholar
    • 144. Kennedy AB, Lavie CJ, Blair SN . Fitness or fatness: which is more important?JAMA. 2018; 319:231–232. doi: 10.1001/jama.2017.21649CrossrefMedlineGoogle Scholar
    • 145. Lavie CJ, McAuley PA, Church TS, Milani RV, Blair SN . Obesity and cardiovascular diseases: implications regarding fitness, fatness, and severity in the obesity paradox.J Am Coll Cardiol. 2014; 63:1345–1354. doi: 10.1016/j.jacc.2014.01.022CrossrefMedlineGoogle Scholar
    • 146. Barry VW, Caputo JL, Kang M . The joint association of fitness and fatness on cardiovascular disease mortality: a meta-analysis.Prog Cardiovasc Dis. 2018; 61:136–141. doi: 10.1016/j.pcad.2018.07.004CrossrefMedlineGoogle Scholar
    • 147. Lavie CJ, Laddu D, Arena R, Ortega FB, Alpert MA, Kushner RF . Healthy weight and obesity prevention: JACC health promotion series.J Am Coll Cardiol. 2018; 72:1506–1531. doi: 10.1016/j.jacc.2018.08.1037CrossrefMedlineGoogle Scholar
    • 148. Elagizi A, Kachur S, Lavie CJ, Carbone S, Pandey A, Ortega FB, Milani RV . An overview and update on obesity and the obesity paradox in cardiovascular diseases.Prog Cardiovasc Dis. 2018; 61:142–150. doi: 10.1016/j.pcad.2018.07.003CrossrefMedlineGoogle Scholar
    • 149. Lavie CJ, Pandey A, Lau DH, Alpert MA, Sanders P . Obesity and atrial fibrillation prevalence, pathogenesis, and prognosis: effects of weight loss and exercise.J Am Coll Cardiol. 2017; 70:2022–2035. doi: 10.1016/j.jacc.2017.09.002CrossrefMedlineGoogle Scholar
    • 150. McAuley PA, Artero EG, Sui X, Lee DC, Church TS, Lavie CJ, Myers JN, España-Romero V, Blair SN . The obesity paradox, cardiorespiratory fitness, and coronary heart disease.Mayo Clin Proc. 2012; 87:443–451. doi: 10.1016/j.mayocp.2012.01.013CrossrefMedlineGoogle Scholar
    • 151. Lavie CJ, Cahalin LP, Chase P, Myers J, Bensimhon D, Peberdy MA, Ashley E, West E, Forman DE, Guazzi M, Arena R . Impact of cardiorespiratory fitness on the obesity paradox in patients with heart failure.Mayo Clin Proc. 2013; 88:251–258. doi: 10.1016/j.mayocp.2012.11.020CrossrefMedlineGoogle Scholar
    • 152. Pathak RK, Elliott A, Middeldorp ME, Meredith M, Mehta AB, Mahajan R, Hendriks JM, Twomey D, Kalman JM, Abhayaratna WP, Lau DH, Sanders P . Impact of CARDIOrespiratory FITness on arrhythmia recurrence in obese individuals with atrial fibrillation: the CARDIO-FIT study.J Am Coll Cardiol. 2015; 66:985–996. doi: 10.1016/j.jacc.2015.06.488CrossrefMedlineGoogle Scholar
    • 153. Moholdt T, Lavie CJ, Nauman J . Interaction of physical activity and body mass index on mortality in coronary heart disease: data from the Nord-Trøndelag Health Study.Am J Med. 2017; 130:949–957. doi: 10.1016/j.amjmed.2017.01.043CrossrefMedlineGoogle Scholar
    • 154. Moholdt T, Lavie CJ, Nauman J . Sustained physical activity, not weight loss, associated with improved survival in coronary heart disease.J Am Coll Cardiol. 2018; 71:1094–1101. doi: 10.1016/j.jacc.2018.01.011CrossrefMedlineGoogle Scholar
    • 155. Heidenreich PA, Albert NM, Allen LA, Bluemke DA, Butler J, Fonarow GC, Ikonomidis JS, Khavjou O, Konstam MA, Maddox TM, Nichol G, Pham M, Piña IL, Trogdon JG ; American Heart Association Advocacy Coordinating Committee; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Cardiovascular Radiology and Intervention; Council on Clinical Cardiology; Council on Epidemiology and Prevention; Stroke Council. Forecasting the impact of heart failure in the United States: a policy statement from the American Heart Association.Circ Heart Fail. 2013; 6:606–619. doi: 10.1161/HHF.0b013e318291329aLinkGoogle Scholar
    • 156. Malhotra R, Bakken K, D’Elia E, Lewis GD . Cardiopulmonary exercise testing in heart failure.JACC Heart Fail. 2016; 4:607–616. doi: 10.1016/j.jchf.2016.03.022CrossrefMedlineGoogle Scholar
    • 157. Canada JM, Trankle CR, Buckley LF, et al . Severely impaired cardiorespiratory fitness in patients with recently decompensated systolic heart failure.Am J Cardiol. 2017; 120:1854–1857. doi: 10.1016/j.amjcard.2017.07.099CrossrefMedlineGoogle Scholar
    • 158. Garber CE, Blissmer B, Deschenes MR, Franklin BA, Lamonte MJ, Lee IM, Nieman DC, Swain DP ; American College of Sports Medicine. American College of Sports Medicine position stand. Quantity and quality of exercise for developing and maintaining cardiorespiratory, musculoskeletal, and neuromotor fitness in apparently healthy adults: guidance for prescribing exercise.Med Sci Sports Exerc. 2011; 43:1334–1359. doi: 10.1249/MSS.0b013e318213fefbCrossrefMedlineGoogle Scholar
    • 159. Arena R, Sietsema KE . Cardiopulmonary exercise testing in the clinical evaluation of patients with heart and lung disease.Circulation. 2011; 123:668–680. doi: 10.1161/CIRCULATIONAHA.109.914788LinkGoogle Scholar
    • 160. Guazzi M, Bandera F, Ozemek C, Systrom D, Arena R . Cardiopulmonary exercise testing: what is its value?J Am Coll Cardiol. 2017; 70:1618–1636. doi: 10.1016/j.jacc.2017.08.012CrossrefMedlineGoogle Scholar
    • 161. Kondamudi N, Haykowsky M, Forman DE, Berry JD, Pandey A . Exercise training for prevention and treatment of heart failure.Prog Cardiovasc Dis. 2017; 60:115–120. doi: 10.1016/j.pcad.2017.07.001CrossrefMedlineGoogle Scholar
    • 162. O’Connor CM, Whellan DJ, Lee KL, 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.2009.454CrossrefMedlineGoogle Scholar
    • 163. 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.114.001615LinkGoogle Scholar
    • 164. 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.2003.11.033CrossrefMedlineGoogle Scholar
    • 165. Piepoli MF, Davos C, Francis DP, Coats AJ ; ExTraMATCH Collaborative. Exercise training meta-analysis of trials in patients with chronic heart failure (ExTraMATCH).BMJ. 2004; 328:189. doi: 10.1136/bmj.37938.645220.EECrossrefMedlineGoogle Scholar
    • 166. 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.2013.08.006CrossrefMedlineGoogle Scholar
    • 167. Keteyian SJ, Leifer ES, Houston-Miller N, Kraus WE, Brawner CA, O’Connor CM, Whellan DJ, Cooper LS, Fleg JL, Kitzman DW, Cohen-Solal A, Blumenthal JA, Rendall DS, Piña IL ; HF-ACTION Investigators. Relation between volume of exercise and clinical outcomes in patients with heart failure.J Am Coll Cardiol. 2012; 60:1899–1905. doi: 10.1016/j.jacc.2012.08.958CrossrefMedlineGoogle Scholar
    • 168. Fleg JL, Cooper LS, Borlaug BA, Haykowsky MJ, Kraus WE, Levine BD, Pfeffer MA, Piña IL, Poole DC, Reeves GR, Whellan DJ, Kitzman DW ; National Heart, Lung, and Blood Institute Working Group. Exercise training as therapy for heart failure: current status and future directions.Circ Heart Fail. 2015; 8:209–220. doi: 10.1161/CIRCHEARTFAILURE.113.001420LinkGoogle Scholar
    • 169. Tucker WJ, Lijauco CC, Hearon CM, Angadi SS, Nelson MD, Sarma S, Nanayakkara S, La Gerche A, Haykowsky MJ . Mechanisms of the improvement in peak VO2 with exercise training in heart failure with reduced or preserved ejection fraction.Heart Lung Circ. 2018; 27:9–21. doi: 10.1016/j.hlc.2017.07.002CrossrefMedlineGoogle Scholar
    • 170. Haykowsky MJ, Tomczak CR, Scott JM, Paterson DI, Kitzman DW . Determinants of exercise intolerance in patients with heart failure and reduced or preserved ejection fraction.J Appl Physiol (1985). 2015; 119:739–744. doi: 10.1152/japplphysiol.00049.2015CrossrefMedlineGoogle Scholar
    • 171. Haykowsky MJ, Liang Y, Pechter D, Jones LW, McAlister FA, Clark AM . A meta-analysis of the effect of exercise training on left ventricular remodeling in heart failure patients: the benefit depends on the type of training performed.J Am Coll Cardiol. 2007; 49:2329–2336. doi: 10.1016/j.jacc.2007.02.055CrossrefMedlineGoogle Scholar
    • 172. Sullivan MJ, Higginbotham MB, Cobb FR . Exercise training in patients with severe left ventricular dysfunction. Hemodynamic and metabolic effects.Circulation. 1988; 78:506–515.LinkGoogle Scholar
    • 173. Abbate A, Van Tassell BW, Canada JM, Dixon DL, Arena RA, Biondi-Zoccai G . Pharmacologic and surgical interventions to improve functional capacity in heart failure.Heart Fail Clin. 2015; 11:117–124. doi: 10.1016/j.hfc.2014.08.005CrossrefMedlineGoogle Scholar
    • 174. Sanganalmath SK, Bolli R . Cell therapy for heart failure: a comprehensive overview of experimental and clinical studies, current challenges, and future directions.Circ Res. 2013; 113:810–834. doi: 10.1161/CIRCRESAHA.113.300219LinkGoogle Scholar
    • 175. 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.2015.17346CrossrefMedlineGoogle Scholar
    • 176. 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.2011.02.055CrossrefMedlineGoogle Scholar
    • 177. Edelmann F, Gelbrich G, Düngen HD, Fröhling S, Wachter R, Stahrenberg R, Binder L, Töpper A, Lashki DJ, Schwarz S, Herrmann-Lingen C, Löffler M, Hasenfuss G, Halle M, Pieske B . 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.2011.06.054CrossrefMedlineGoogle Scholar
    • 178. Del Buono MG, Buckley L, Abbate A . Primary and secondary diastolic dysfunction in heart failure with preserved ejection fraction.Am J Cardiol. 2018; 122:1578–1587. doi: 10.1016/j.amjcard.2018.07.012CrossrefMedlineGoogle Scholar
    • 179. Borlaug BA . The pathophysiology of heart failure with preserved ejection fraction.Nat Rev Cardiol. 2014; 11:507–515. doi: 10.1038/nrcardio.2014.83CrossrefMedlineGoogle Scholar
    • 180. Abbate A, Arena R, Abouzaki N, Van Tassell BW, Canada J, Shah K, Biondi-Zoccai G, Voelkel NF . Heart failure with preserved ejection fraction: refocusing on diastole.Int J Cardiol. 2015; 179:430–440. doi: 10.1016/j.ijcard.2014.11.106CrossrefMedlineGoogle Scholar
    • 181. Ponikowski P, Voors AA, Anker SD, et al ; Authors/Task Force Members; Document Reviewers. 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: the task force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC.Eur J Heart Fail. 2016; 18:891–975. doi: 10.1002/ejhf.592CrossrefMedlineGoogle Scholar
    • 182. Yancy CW, Jessup M, Bozkurt B, et al ; American College of Cardiology Foundation; American Heart Association Task Force on Practice Guidelines. 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology Foundation/American Heart Association task force on practice guidelines.J Am Coll Cardiol. 2013; 62:e147–e239. doi: 10.1016/j.jacc.2013.05.019CrossrefMedlineGoogle Scholar
    • 183. Shah SJ, Kitzman DW, Borlaug BA, van Heerebeek L, Zile MR, Kass DA, Paulus WJ . Phenotype-specific treatment of heart failure with preserved ejection fraction: a multiorgan roadmap.Circulation. 2016; 134:73–90. doi: 10.1161/CIRCULATIONAHA.116.021884LinkGoogle Scholar
    • 184. Pandey A, Patel KV, Vaduganathan M, Sarma S, Haykowsky MJ, Berry JD, Lavie CJ . Physical activity, fitness, and obesity in heart failure with preserved ejection fraction.JACC Heart Fail. 2018; 6:975–982. doi: 10.1016/j.jchf.2018.09.006CrossrefMedlineGoogle Scholar
    • 185. Wohlfahrt P, Redfield MM, Lopez-Jimenez F, Melenovsky V, Kane GC, Rodeheffer RJ, Borlaug BA . Impact of general and central adiposity on ventricular-arterial aging in women and men.JACC Heart Fail. 2014; 2:489–499. doi: 10.1016/j.jchf.2014.03.014CrossrefMedlineGoogle Scholar
    • 186. Obokata M, Reddy YNV, Melenovsky V, Pislaru S, Borlaug BA . Deterioration in right ventricular structure and function over time in patients with heart failure and preserved ejection fraction [published online December 12, 2018].Eur Heart J. doi: 10.1093/eurheartj/ehy809. https://academic.oup.com/eurheartj/advance-article-abstract/doi/10.1093/eurheartj/ehy809/5240922?redirectedFrom=fulltext.Google Scholar
    • 187. Obokata M, Reddy YNV, Pislaru SV, Melenovsky V, Borlaug BA . Evidence supporting the existence of a distinct obese phenotype of heart failure with preserved ejection fraction.Circulation. 2017; 136:6–19. doi: 10.1161/CIRCULATIONAHA.116.026807LinkGoogle Scholar
    • 188. Carbone S, Canada JM, Buckley LF, Trankle CR, Dixon DL, Buzzetti R, Arena R, Van Tassell BW, Abbate A . Obesity contributes to exercise intolerance in heart failure with preserved ejection fraction.J Am Coll Cardiol. 2016; 68:2487–2488. doi: 10.1016/j.jacc.2016.08.072CrossrefMedlineGoogle Scholar
    • 189. Carbone S, Popovic D, Lavie CJ, Arena R . Obesity, body composition and cardiorespiratory fitness in heart failure with preserved ejection fraction [published online August 10, 2017].Future Cardiol. doi: 10.2217/fca-2017-0023. https://www.futuremedicine.com/doi/abs/10.2217/fca-2017-0023.Google Scholar
    • 190. 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.117.030617LinkGoogle Scholar
    • 191. Tate CW, Robertson AD, Zolty R, Shakar SF, Lindenfeld J, Wolfel EE, Bristow MR, Lowes BD . Quality of life and prognosis in heart failure: results of the Beta-Blocker Evaluation of Survival Trial (BEST).J Card Fail. 2007; 13:732–737. doi: 10.1016/j.cardfail.2007.07.001CrossrefMedlineGoogle Scholar
    • 192. Flynn KE, Piña IL, Whellan DJ, et al ; HF-ACTION Investigators. 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.2009.457CrossrefMedlineGoogle Scholar
    • 193. Omar W, Pandey A, Haykowsky MJ, Berry JD, Lavie CJ . The evolving role of cardiorespiratory fitness and exercise in prevention and management of heart failure.Curr Heart Fail Rep. 2018; 15:75–80. doi: 10.1007/s11897-018-0382-zCrossrefMedlineGoogle Scholar
    • 194. 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.2013.12.031CrossrefMedlineGoogle Scholar
    • 195. Molina AJ, Bharadwaj MS, Van Horn C, Nicklas BJ, Lyles MF, Eggebeen J, Haykowsky MJ, Brubaker PH, Kitzman DW . Skeletal muscle mitochondrial content, oxidative capacity, and Mfn2 expression are reduced in older patients with heart failure and preserved ejection fraction and are related to exercise intolerance.JACC Heart Fail. 2016; 4:636–645. doi: 10.1016/j.jchf.2016.03.011CrossrefMedlineGoogle Scholar
    • 196. 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.00004.2014CrossrefMedlineGoogle Scholar
    • 197. Prado CM, Heymsfield SB . Lean tissue imaging: a new era for nutritional assessment and intervention.JPEN J Parenter Enteral Nutr. 2014; 38:940–953. doi: 10.1177/0148607114550189CrossrefMedlineGoogle Scholar
    • 198. Cicoira M, Zanolla L, Franceschini L, Rossi A, Golia G, Zamboni M, Tosoni P, Zardini P . Skeletal muscle mass independently predicts peak oxygen consumption and ventilatory response during exercise in noncachectic patients with chronic heart failure.J Am Coll Cardiol. 2001; 37:2080–2085.CrossrefMedlineGoogle Scholar
    • 199. Bekfani T, Pellicori P, Morris DA, et al . Sarcopenia in patients with heart failure with preserved ejection fraction: Impact on muscle strength, exercise capacity and quality of life.Int J Cardiol. 2016; 222:41–46. doi: 10.1016/j.ijcard.2016.07.135CrossrefMedlineGoogle Scholar
    • 200. Upadhya B, Haykowsky MJ, Eggebeen J, Kitzman DW . Sarcopenic obesity and the pathogenesis of exercise intolerance in heart failure with preserved ejection fraction.Curr Heart Fail Rep. 2015; 12:205–214. doi: 10.1007/s11897-015-0257-5CrossrefMedlineGoogle Scholar
    • 201. Carbone S, Lavie CJ, Arena R . Obesity and heart failure: focus on the obesity paradox.Mayo Clin Proc. 2017; 92:266–279. doi: 10.1016/j.mayocp.2016.11.001CrossrefMedlineGoogle Scholar
    • 202. Lavie CJ, Forman DE, Arena R . Bulking up skeletal muscle to improve heart failure prognosis.JACC Heart Fail. 2016; 4:274–276. doi: 10.1016/j.jchf.2015.12.005CrossrefMedlineGoogle Scholar
    • 203. Ventura HO, Carbone S, Lavie CJ . Muscling up to improve heart failure prognosis.Eur J Heart Fail. 2018; 20:1588–1590. doi: 10.1002/ejhf.1314CrossrefMedlineGoogle Scholar
    • 204. Pocock SJ, McMurray JJ, Dobson J, Yusuf S, Granger CB, Michelson EL, Ostergren J, Pfeffer MA, Solomon SD, Anker SD, Swedberg KB . Weight loss and mortality risk in patients with chronic heart failure in the candesartan in heart failure: assessment of reduction in mortality and morbidity (CHARM) programme.Eur Heart J. 2008; 29:2641–2650. doi: 10.1093/eurheartj/ehn420CrossrefMedlineGoogle Scholar
    • 205. Lavie CJ, Arena R, Blair SN . A call to increase physical activity across the globe in the 21st century.Future Cardiol. 2016; 12:605–607. doi: 10.2217/fca-2016-0055CrossrefMedlineGoogle Scholar
    • 206. Carlson SA, Fulton JE, Schoenborn CA, Loustalot F . Trend and prevalence estimates based on the 2008 physical activity guidelines for Americans.Am J Prev Med. 2010; 39:305–313. doi: 10.1016/j.amepre.2010.06.006CrossrefMedlineGoogle Scholar
    • 207. Tucker JM, Welk GJ, Beyler NK . Physical activity in U.S.: adults compliance with the physical activity guidelines for Americans.Am J Prev Med. 2011; 40:454–461. doi: 10.1016/j.amepre.2010.12.016CrossrefMedlineGoogle Scholar
    • 208. Lee DC, Lavie CJ, Sui X, Blair SN . Running and mortality: is more actually worse?Mayo Clin Proc. 2016; 91:534–536. doi: 10.1016/j.mayocp.2016.01.013CrossrefMedlineGoogle Scholar
    • 209. Lew WY . Exercise: commitment to a young heart.J Am Coll Cardiol. 2014; 64:1267–1269. doi: 10.1016/j.jacc.2014.04.082CrossrefMedlineGoogle Scholar
    • 210. Karlsen T, Aamot IL, Haykowsky M, Rognmo Ø . High intensity interval training for maximizing health outcomes.Prog Cardiovasc Dis. 2017; 60:67–77. doi: 10.1016/j.pcad.2017.03.006CrossrefMedlineGoogle Scholar

    eLetters(0)

    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.

    Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.