Skip to main content
Cardiovascular Research logoLink to Cardiovascular Research
. 2022 Jul 26;118(18):3434–3450. doi: 10.1093/cvr/cvac120

Obesity and heart failure with preserved ejection fraction: new insights and pathophysiological targets

Barry A Borlaug 1,, Michael D Jensen 2, Dalane W Kitzman 3, Carolyn S P Lam 4, Masaru Obokata 5, Oliver J Rider 6,2
PMCID: PMC10202444  PMID: 35880317

Abstract

Obesity and heart failure with preserved ejection fraction (HFpEF) represent two intermingling epidemics driving perhaps the greatest unmet health problem in cardiovascular medicine in the 21st century. Many patients with HFpEF are either overweight or obese, and recent data have shown that increased body fat and its attendant metabolic sequelae have widespread, protean effects systemically and on the cardiovascular system leading to symptomatic HFpEF. The paucity of effective therapies in HFpEF underscores the importance of understanding the distinct pathophysiological mechanisms of obese HFpEF to develop novel therapies. In this review, we summarize the current understanding of the cardiovascular and non-cardiovascular features of the obese phenotype of HFpEF, how increased adiposity might pathophysiologically contribute to the phenotype, and how these processes might be targeted therapeutically.

Keywords: Adiposity, Heart failure, Inflammation, Obesity, phenotype, HFpEF


This article is part of the Spotlight Issue on heart failure.

1. Introduction

Heart failure (HF) is an enormous public health problem, with worldwide prevalence of over 64 million.1 In the USA alone, over 6 million adults have HF, and by 2030 this number is projected to swell to 8 million,2 with similar trends evident worldwide. Over half of patients with HF have preserved ejection fraction (HFpEF), and the prevalence is increasing relative to HF with reduced ejection fraction.3,4 The epidemic of HFpEF is related to increases in mean age and rising trends in the prevalence of hypertension, metabolic syndrome, diabetes, and atrial fibrillation; all comorbidities that are intimately tied to the development of excess body fat.5

Obesity has doubled in more than 70 countries since 1980. Prevalence in the USA now exceeds 40%, a staggering increase from 15% as recently as the 1970s,5 and it is now projected that 1 in 2 adults in the USA will be obese by 2030.6 Epidemiological studies have identified a strong, independent relationship between obesity and development of HFpEF.7–10 In contrast to atherosclerotic vascular diseases, the link between obesity and HF is not explainable by traditional cardiovascular risk factors.11 Despite increasing recognition, HFpEF remains too often neglected as a cardiovascular complication among patients with excess body fat, leading to underdiagnosis of obesity-related HFpEF, and therefore, undertreatment.12

New data have changed our thinking about the contribution of obesity to HFpEF—from being a mechanical cause of dyspnoea, to a comorbid bystander, to the primary, direct cause of HFpEF, mediated via multiple deleterious effects on myocardial structure, function, and metabolism, as well as effects on lung, skeletal muscle, kidney, and liver, collectively mediated by systemic inflammation, neurohormonal activation, autonomic dys-regulation, and altered haemodynamic load.13–17

Patients with obese HFpEF are often excluded from clinical trials, either directly because of restrictive body mass index (BMI) eligibility cutoffs, or indirectly through the requirement for elevated plasma natriuretic peptide (NP) levels, which are characteristically depressed in obesity.12,15,17–19 The distinct characteristics of obese HFpEF suggest that it may require unique therapies that could differ from non-obese patients with HFpEF. In this review, we summarize the current understanding of how increased adipose tissue pathophysiologically contributes to HFpEF, and consider specific treatment approaches that merit study in future trials.

2. Biology of excess body fat

While BMI is traditionally used to define obesity (≥30 kg/m2), a high BMI may not correspond to the same degree of body fat, or cardiovascular risk, in different individuals. Indeed, beyond quantity of excess fat, the distribution and quality of the excess fat also influences cardiovascular risk. Excess body fat is always the result of positive energy balance. The ability of mammals to store energy as triglycerides in adipocytes provides an extremely efficient means of retaining large amounts of energy that serves as fuel. However, excess body fat, and especially upper body (central) obesity, is associated with greater risk for dyslipidaemia, hypertension, Type 2 diabetes, and of course, HFpEF.7–10 Many investigators have found that visceral adipose tissue (VAT) is an even better predictor of abnormal metabolic profiles than upper body subcutaneous fat, leading to the hypothesis that VAT is directly causing insulin resistance, inflammation, accelerated senescence, and mitochondrial dysfunction.20–23

Gains in body fat occur by adipocyte expansion (hypertrophy) and/or the generation of new adipocytes (hyperplasia), even in adults (Figure 1). These processes vary from person to person and depot to depot.24 Mean adipocyte size increases as a function of obesity, up to a point.25 The balance between hypertrophy and hyperplasia appears linked to the metabolic consequences of obesity; enlarged abdominal subcutaneous adipocytes are associated with an increased cardiometabolic risk.26

Figure 1.

Figure 1

Patterns of fat gain through hypertrophy and hyperplasia in obesity, and association with free fatty acid (FFA) release. See text for details.

2.1. Distribution of body fat: regional fat depots

The major fat depots have heterogenous and distinct characteristics. Upper body subcutaneous fat, which includes superficial and deep truncal depots,27,28 is usually the largest. Lower body subcutaneous fat includes gluteal, femoral, and calf adipose tissue, i.e. all adipose tissue caudal to the inguinal ligament anteriorly and the ileac crest posteriorly. Intra-abdominal fat includes mesenteric and omental (visceral) depots, both of which drain into the portal vein; perinephric fat is intra-abdominal, but because it drains into the systemic circulation, it is not considered visceral. Adipose depots differ in terms of the fat cell size,29 responsiveness to pro- and antilipolytic hormones,30 as well as the ability to store fatty acids in triglycerides.31,32

2.2. Adipocyte hypertrophy vs. hyperplasia

In 2000, it was posited that the adipocyte hypertrophy in abdominal subcutaneous fat in those predisposed to Type 2 diabetes was due to a failure of preadipocytes to differentiate as needed to store extra body fat.33 This is important because hypertrophic adipocytes are insulin resistant and have greater rates of lipolysis. Figure 1 depicts the relative extremes of potential fat gain, ranging from a fat storage solely into hypertrophic subcutaneous adipocytes (lower right), with substantial gain of VAT, to excess fat storage solely from hyperplasia, with maintenance of normal subcutaneous fat cell size and function (lower left) and little gain of VAT.

Much has been learned about the process of the creation of new adipocytes in humans.34,35 Peroxisome proliferator-activated receptor gamma (PPARϒ) is the master regulator of adipogenesis and required for adipocyte differentiation/survival; bone morphogenic protein-4 (BMP-4) promotes processes that activates PPARϒ, allowing the creation of new adipocytes from adipose mesenchymal stem cells. However, this process of adipogenic commitment and early differentiation is regulated by both pro- and anti-adipogenic pathways. The pro-adipogenic signals involve secreted proteins such as Dickkopf 1 and the secreted inhibitory protein gremlin-1 (GREM-1), which inhibits BMP-4 signalling. Humans with hypertrophic obesity appear to produce excess GREM-1, which suppresses adipogenesis, thereby forcing adipose lipid accumulation via hypertrophy.34,35 This may be due to epigenetic and genetic factors that regulate adipogenesis. This ability or inability may then determine the relative gain in VAT. Inherited or acquired defects in these processes may explain the tendency of some people to gain body fat predominantly by adipocyte hypertrophy rather than hyperplasia. Evidence for this theory comes from the study of volunteers who were overfed in order to study adipose responses to fat gain.36 Adults who created larger numbers of new leg adipocytes had less gain of VAT than those who gained fat by increasing abdominal subcutaneous adipocyte size.

2.3. Functions of adipose tissue

The major functions of adipose tissue include the storage and release of fatty acids. Adipose tissue stores the circulating triglyceride fatty acids and free fatty acids (FFAs) that are not used by lean tissue for essential processes. Healthy adipose tissue releases FFA into the circulation at rates tightly linked to energy needs.37 Increases in upper body/visceral fat in obesity is associated with excess release of FFA into the circulation,38 and this may have important and highly relevant deleterious effects on the heart in obese HFpEF. In contrast, humans who preferentially gain fat in their lower body have a lesser risk of metabolic abnormalities,39 and more normal FFA metabolism.38

In humans, insulin-stimulated glucose disposal is strongly correlated with insulin-suppressed FFA concentrations/flux,40,41 and nadir FFA concentrations in response to insulin independently predict insulin sensitivity.42 Because skeletal muscle is the dominant site of glucose disposal in humans,43 muscle insulin resistance with respect to glucose uptake is a major metabolic defect in obesity. Mechanistically, excess FFA can create insulin resistance affecting glucose metabolism in skeletal muscle and liver, as well as stimulate hepatic very low–density lipoprotein-triglyceride production. Artificially elevating FFA in healthy adults induces insulin resistance,44 while obese mice that are haploinsufficient for hormone-sensitive lipase in adipose tissue have reduced lipolysis.45 Elevated FFA can decrease tissue glucose metabolism by forcing the preferential oxidation of fatty acids (the Randle cycle),46 and can impair insulin signalling via lipid intermediates.47

Endocrine properties of visceral fat may directly contribute to metabolic health and cardiac structure/function. Cytokine concentration gradients have been demonstrated from portal venous to arterial blood, suggesting release from visceral fat,48 whereas others have measured cytokine concentrations in the venous effluent of abdominal subcutaneous fat.49

Humans with defects in the ability of adipocyte precursors to proliferate and differentiate in response to the need for more fat storage develop dysfunctional, hypertrophic subcutaneous adipocytes, which expose peripheral tissues to excess FFA.50 Over time, excess FFA can drive the storage of fat in ectopic depots such as visceral fat, pericardial fat, skeletal muscle, liver, and intramyocellular triglycerides.51 We suggest that the most likely cause of excess visceral fat gain is the forced storage of fatty acids, possibly from excess fasting and postprandial FFA release, as a result of insulin resistant subcutaneous fat. Humans with greater amounts of visceral fat are less, not more efficient in storing fatty acids. The greatest efficiency of fatty acid storage in omental fat is seen in lean adults with the smallest visceral fat depots.32,52 The efficiency of fatty acid storage in omental fat decreases as a function of visceral fat mass and there is down-regulation of proteins and enzymes responsible for fatty acid transport into the cells in those with visceral obesity.52 This finding contrasts with the tendency of fat storage (and adipocyte fatty acid storage factors) to remain stable (abdominal fat) or actually increase (femoral fat) as a function of depot size.32

In order for VAT to continue to increase in mass despite down-regulation of fatty acid storage pathways, there must be an even greater down-regulation of lipolysis pathways. Because visceral obesity is associated with failure to normally suppress lipolysis after meals53,54 as well as higher postprandial chylomicron concentrations,54 it is possible that the higher FFA and chylomicron-triglyceride concentrations drive fatty acids into ectopic depots, including visceral fat, and potentially epicardial fat, despite lesser expression of the proteins and enzymes that facilitate fat storage, and this may importantly contribute to development of HFpEF through abnormalities in cardiac and non-cardiac structure and function.

3. Diagnostic challenges of HFpEF in people with obesity

Obesity is an important cause of HFpEF but can also be associated with symptoms of dyspnoea that are not caused by cardiac dysfunction. Diagnosis of HFpEF requires objective demonstration of congestion, and this is most commonly evaluated by physician examination, echocardiography, and NP testing.55,56 Each of these are more challenging in patients with obesity. Physical signs such as jugular distention may be masked by increased neck girth. NP levels are well known to be lower and often ‘normal’ (lower than traditional diagnostic thresholds) in patients with HFpEF who are obese,19 and recent data have revealed that even echo-Doppler indicators such as the E/e′ ratio underestimate circulatory congestion in patients with obesity.12 Several factors appear to contribute to lower NP levels in obese HFpEF, including increased external constraint on the heart, mitigating increases in wall stress.12,15 Sex hormones may also contribute. When compared with premenopausal women, postmenopausal women, and men display increasing testosterone levels, which are associated with progressively lower NP levels,57 further masking the presence of circulatory congestion. The lower NP levels in obese HFpEF also raise the possibility that many of these patients may suffer from an ‘NP deficiency’. Invasive haemodynamic exercise testing is frequently necessary in equivocal cases to determine whether HFpEF is present or absent.55,56

4. Diabetes, obesity, and HFpEF

As described, Type 2 diabetes and metabolic syndrome are causally tied to excess body fat, blurring the lines between these conditions such that many refer to the common syndrome of diabesity.5 Approximately 40–50% of patients with HFpEF have diabetes, and another 15–20% have prediabetes.58,59 When compared with those without diabetes, patients with HFpEF and diabetes have poorer patient-reported quality of life, worse exercise capacity, increased markers of inflammation, fibrosis, and endothelial dysfunction, worse congestion, worse renal function, higher left ventricular (LV) filling pressures, and increased mortality and HF admissions.58

5. Organ-level pathophysiology of obese HFpEF

Even among patients without HF, obesity is associated with preclinical alterations that are known to lead to HFpEF, including LV hypertrophic remodelling, increases in chamber stiffness, prolonged myocardial relaxation, altered substrate utilization, reduced myocardial ATP availability, and mild systolic ventricular dysfunction.60–71 Obesity is also associated with volume expansion, neurohormonal activation, systemic inflammation, and dysfunction of multiple extra-cardiac organ systems of relevance to HFpEF, including skeletal muscle, liver, and kidney.

5.1. Ventricular remodelling and dysfunction

In the absence of HF, the higher cardiac output and total blood volume (TBV) resulting from increased fat mass that accompanies obesity leads to LV cavity dilatation and subsequent hypertrophy secondary to increased wall stress.72,73 Although this accounts for an eccentric hypertrophic pattern, it is evident that concentric remodelling occurs commonly in obesity.73–75 Concentric remodelling appears to be more strongly related to insulin resistance,76 diabetes,76 hyperleptinaemia,77 myocardial steatosis,78 as well as VAT expansion.68,73,79 Patients with obese HFpEF have more LV concentric remodelling than those with non-obese HFpEF, with increased LV mass-to-volume ratio.15 Similar to the LV, obesity is associated with right ventricular (RV) remodelling in patients with and without HFpEF.15,77,80

LV diastolic function assessed using echocardiography is impaired in patients with obesity,60,68 and weight gain is strongly correlated with increasing diastolic chamber stiffness over time (Figure 2).62 These effects are even more pronounced with increases in central adiposity.62,66 Despite preserved EF, patients with HFpEF display subtle impairment in LV contractility at rest and severe impairments in systolic reserve with exercise.81–83 Impaired systolic reserve worsens diastolic reserve, since the failure to contract to a smaller end systolic volume reduces elastic recoil at the onset of diastole, further worsening the elevation in pulmonary capillary pressures (Figure 2). Community-based studies have reported that LV systolic mechanics decrease as BMI, waist circumference, and waist-to-hip ratio increase.63,65,84 Interestingly, calcium-activated force in skinned cardiomyocytes from RV septal endomyocardial biopsies was recently shown to be substantially depressed in patients with obesity-related HFpEF, with a direct correlation between sarcomere dysfunction and fat mass estimated by BMI (Figure 2).85

Figure 2.

Figure 2

(A) Increasing weight gain is associated with progressively greater increases in estimated left ventricular (LV) end diastolic chamber stiffness (ΔLV Eed) over time in community-dwelling adults, predisposing to HFpEF. (B) Impairments in the ability to enhance LV early diastolic relaxation velocity (ΔLV e′) with exercise are associated with greater increases in pulmonary capillary wedge pressure (PCWP), and enhancement in LV e′ with exercise is substantially blunted in obese HFpEF (C). The ability to augment LV e′ is strongly correlated with enhancement in LV systolic function (LV s′), demonstrating the cross talk between systolic and diastolic reserve (D). When compared with cardiomyocytes sampled from healthy hearts and hypertensive HFpEF, individuals with obese HFpEF display reduced Ca2+ activated maximal tension development (Tmax) at the level of the sarcomere, which is inversely related to body mass index (BMI) (E and F). Panel A adapted from Wohlfahrt et al.,62 Panels BD adapted from Borlaug et al.,83 and Panels E and F are from Aslam et al.85

5.2. Atrial pathophysiology

Patients with HFpEF display left atrial (LA) remodelling and dysfunction that leads to increased risk for atrial fibrillation, a triad referred to as LA myopathy.86–88 Obesity is also associated with greater risk of atrial remodelling89 and atrial fibrillation.90 However, in studies comparing patients with obese HFpEF to those with non-obese HFpEF, atrial remodelling is less marked, and atrial fibrillation burden is less in those with obese HFpEF.15,91 This may relate in part to the fact that the latter patients with obese HFpEF are roughly a decade younger than those with non-obese HFpEF. Mean BMI decreases sequentially from patients with HFpEF and no AF to those with paroxysmal AF, to those with permanent AF, though all groups display higher BMI than controls.86 Thus, while LA myopathy is an important pathophysiological contributor in obese HFpEF, its role may be less pivotal in this group than among non-obese HFpEF.

5.3. Epicardial adipose and pericardial restraint

HFpEF is associated with increases in total heart volume, and patients with obese HFpEF display even greater cardiomegaly due to chamber dilatation, increased wall thickness, and greater increases in epicardial adipose tissue (EAT; Figure 3).15,92,93 This increases coupling between the heart and pericardium, amplifying ventricular interdependence, such that intracavitary pressures are higher for any net distending pressure.94 This increase in external constraint on the heart at least partially explains why NP levels and echo-Doppler estimates of filling pressures are much lower in patients with HFpEF with obesity than those without obesity (Figure 3).12

Figure 3.

Figure 3

(A) Gross pathology showing heart from an 82-year-old woman with HFpEF at autopsy revealing marked increase in epicardial fat. (B) When compared with healthy controls and non-obese HFpEF, patients with obese HFpEF display increased epicardial fat thickness, which is associated with flattening of the interventricular septum (C), increased ventricular interdependence reflected by higher LV eccentricity index (D), and higher pulmonary capillary wedge pressures (PCWP) (E). The increase in external constraint on the heart in patients with obesity results in a higher PCWP for any LV transmural distending pressure (LVTMP) (F). See text for details. Panel A reproduced from Borlaug and Maleszewski,93 Panels B–D from Obokata et al.,15 Panel E from Koepp et al.,92 and Panel F from Obokata et al.12

In addition to mechanical effects, EAT may directly influence myocardial structure and function through secretion of soluble factors.95–98 Epicardial adipose (like VAT) has been shown to release proinflammatory cytokines, such as interleukin (IL)-1β, IL-6, and tumour necrosis factor (TNF)-α, which may directly cause remodelling and dysfunction in both cardiac and skeletal muscle through mitochondrial dysfunction, capillary rarefaction, or other processes.99 In a study where EAT samples were harvested from patients at cardiac surgery, incubating rat atria with the EAT secretome from patients induced fibrosis, coupled with increased secretion of the adipo-fibrokine activin A.97

The importance of EAT has been emphasized by recent studies showing that increases in EAT in HFpEF are associated with more severe haemodynamic perturbations, greater abnormalities in RV–pulmonary artery (PA) coupling, more LV fibrosis, more severely impaired exercise capacity, and increased risk of HF hospitalization or death.92,100–104 Interestingly, some of the same studies have shown thinning of EAT in HF with reduced ejection fraction (as opposed to thickening in HFpEF), and an association of EAT thinning with worse exercise capacity and outcomes in HF with reduced ejection fraction.103,104 These contrasting associations of EAT with cardiac function and outcomes in HF with preserved vs. reduced ejection fraction may reflect divergent quality and/or quantity of EAT in the types of HF [proinflammatory in HFpEF, depleted in HF with reduced ejection fraction (HFrEF)].

5.4. Cardiac metabolism and substrate use

In the normal heart under fasting conditions, the majority (70–90%) of ATP derives from FFA (Figure 4), with a lesser (10–30%) contribution from glucose, and a much smaller proportion from ketones and lactate (<1%).5,105,106 In the setting of insulin resistance, like obesity, the heart is exposed to increased circulating FFA by increasing PPARα. As a result, initially a compensatory increase in myocardial FFA uptake and oxidation is driven by increased PPARα expression.107 However, despite this initial protective mechanism, cardiac lipotoxicity occurs. With an increasing intracellular pool of long-chain fatty acyl-CoA, which accompanies increases FFA uptake, comes a larger pool of fatty acid substrate for non-oxidative processes, including the synthesis of triacylglycerol, diacylglycerol, and ceramides.108 In a murine model of HFpEF, cardiomyocyte steatosis is coupled with increases in the activity of the transcription factor Forkhead box protein O1,109 a key transcription factor involved in cell metabolism promoting abnormal unfolded protein response, a recently demonstrated cellular mechanism of HFpEF.110

Figure 4.

Figure 4

(A) Normal cardiac metabolism where substrate selection produces ∼30% glucose oxidation and ∼70% fatty acid oxidation providing nicotinamide adenine dinucleotide and flavin adenine dinucleotide (NADH/FADH2) to the electron transport chain for adenosine triphosphate (ATP) synthesis, and a normal phosphocreatine (PCR) to ATP ratio. (B) Cardiac metabolism in obesity where the balance of substrate selection is towards overreliance on fatty acid oxidation, with decreased glucose uptake (from insulin resistance) and oxidation due to pyruvate dehydrogenase (PDH) inhibition. The increased FADH2/NADH ratio is highlighted with additional uncoupling protein expression (UCP), leading to reduced PCr/ATP ratio. Increased fatty acid flux may also increase production of reactive oxygen species and non-oxidative metabolism of fatty acids resulting in lipotoxicity.

Whether significant lipotoxicity occurs in humans is less clear but increased levels of intramyocardial lipid has been shown in patients with obesity and diabetes,111 with the degree of steatosis related to concentric LV remodelling and diastolic impairment.112 A recent study has further showed increased myocardial fat in human HFpEF.113 This would imply that ectopic myocardial triglyceride plays at least some role in cardiac dysfunction and remodelling in humans.

While no study has yet evaluated cardiac metabolism specifically in obesity-related HFpEF, the available evidence suggests that FFA use is likely increased in this disorder (Figure 4). For example, in obesity without HF,71,114,115 as well as conditions associated with HFpEF such as diabetes116,117 and LV hypertrophy due to aortic stenosis,118 there is increased cardiac FFA uptake and oxidation. Increases in circulating FFA levels, uptake, and flux due to excessive lipolysis in obesity have deleterious effects on metabolism,38,53 especially in the heart.106,117 In obese women without HF, higher BMI is correlated with increased myocardial FFA uptake and reduced efficiency.71 Higher FFA use is associated with decreased glucose oxidation, which is the more efficient fuel source (higher P/O ratio, or ATP produced relative to O2 consumed) when compared with fat.5,119 The importance of the increase in FFA metabolism lies in the fact that only 50% of the reducing equivalents produced in the process of β-oxidation are able to donate electrons at Complex I of the electron transport chain, whereas the remaining half are donated by FADH2 at the flavoprotein site further downstream at Complex II, resulting in this relative O2 inefficiency (Figure 4). Elevations in FFA levels also increase mitochondrial uncoupling protein 3 expression, leading to reduced ATP production,120 with impaired excitation-contraction coupling121 and reduced myocardial glucose uptake.122 The increase in FFA flux and myocardial VO2 in obesity-related HFpEF may also promote generation of reactive oxygen and lipid species that further impair myocyte function.123,124

The surfeit of FFA in obese HFpEF may play a fundamental role in causing myocardial dysfunction. In obese patients without HF, imaging studies have shown that weight loss is associated with a decrease in cardiac FFA utilization,125–127 and with a reduction in myocardial steatosis.107 A small study (n = 13) reported that myocardial glucose oxidation was improved following weight loss,128 and another found increased insulin-stimulated glucose uptake,127 but neither study was performed in patients with HFpEF. There may be important differences in HFpEF, as recent studies have shown that myocardial ischaemia and injury are common in HFpEF,129 even in the absence of epicardial coronary disease.130,131 This is important because FFA use is reduced during ischaemia as the heart preferentially shifts to glucose utilization as it requires less O2.132 Thus, deficits in glucose uptake would be expected to further exacerbate cardiac dysfunction during exercise in obese HFpEF.

5.5. Myocardial energetics in obese HFpEF

As a continual ATP delivery to the myocardium is required for both contraction and relaxation, the increased ATP demand in the context of this metabolic inefficiency in obesity is a potential explanation obesity-related HFpEF. In line with this, using 31P magnetic resonance spectroscopy, the PCr/ATP ratio (a marker of energetic state of the myocardium) has been shown to be lower in patients with obesity, and correlated with diastolic function.133 This is in line with the concept that any impairment in ATP metabolism will initially affect the function of the sarcoplasmic reticular calcium ATPase (SERCA) to lower cytosolic Ca2+ during diastole (as it is energetically most demanding of all enzymes in the contractile apparatus). However, in obesity creatine kinase activity is increased to compensate for the reduced PCr/ATP ratio and maintain ATP delivery at rest.70 This compensatory mechanism is also seen in obese HFrEF.134 Successful weight loss in the absence of HF is associated with a reduced CK flux70 and with increased PCr/ATP.135 In HFrEF, normalization of CK flux is also seen,134 but no study has yet evaluated effects of weight loss on myocardial energetics in HFpEF.

5.6. Pulmonary vascular disease and right-sided HF

Pulmonary hypertension (PH) is common in HFpEF and associated with increased mortality,136 especially when pulmonary vascular disease (PVD; i.e. elevated pulmonary vascular resistance) is present.137 PA pressures are similar at rest in patients with obesity-related HFpEF and non-obese HFpEF, but with the increase in lung blood flow accompanying exercise, obese patients with HFpEF display impaired pulmonary vasodilation, and greater elevations in PA pressure.15 Recent studies in animal models have shed new light. Obese ZSF-1 leptin-receptor knockout rats had normal PA pressures at rest, but elevated pressures during exercise, similar to what is commonly observed in human HFpEF.138 Cultured PA smooth muscle cells treated with FFA, glucose, and insulin showed increased mitochondrial reactive oxygen species and reduced cyclic guanosine monophosphate-dependent signalling, which favourably modified following treatment with the SGLT2i empagliflozin.139 Recent studies in porcine banded models have identified novel pathways activated in PVD due to HFpEF, and further research is needed to further delineate how obesity and insulin resistance might affect these pathways.140

Sustained exposure to PH and PVD leads to RV dysfunction in patients with HFpEF.141,142 A recent meta-analysis has revealed that the higher BMI is associated with RV dysfunction in HFpEF.143 In a study in which patients with obese and non-obese HFpEF were directly compared, RV systolic function and RV-PA coupling were more impaired in obesity-related HFpEF compared with non-obese HFpEF,15 and in a longitudinal study, higher body weight was associated with greater risk for new onset RV dysfunction in patients with HFpEF.142

5.7. Effects beyond the heart

5.7.1. Neurohormone activation and volume distribution

Plasma volume expansion is typical of obesity, and elevation in cardiac filling pressures is directly correlated with estimated plasma volume in patients with obese HFpEF.15 Two mechanisms likely contribute to the TBV expansion in obese HFpEF: systemic vasodilation and sodium retention. Because of increased metabolic demands in obese subjects, excessive systemic vasodilation occurs which is associated with arterial underfilling, contributing to neurohormonal activation, and increases in sodium retention.144 Perivascular adipose tissues also release hormones such as leptin to induce vasodilation.145 The ensuing reduction in vascular resistance coupled with these changes may lead to the rarer entity of obesity-related high output HF in some patients,146 but most patients with obesity-related HFpEF have normal output.15

Obesity, especially when associated with increased VAT, is associated with activation of the sympathetic nervous system (SNS) and renin–angiotensin–aldosterone system (RAAS), both through increased synthesis of adipokines that activate these systems (e.g. leptin, aldosterone) and through mechanical compression of the kidneys.147,148 Sodium reabsorption, plasma volume expansion, SNS activation, and development of hypertension are even more strongly related to increases in visceral fat.149,150 Hyperfiltration and activation of inflammatory pathways may lead to obesity-related glomerulopathy, further contributing to volume overload.151

In addition to TBV expansion, recent studies have shown that the distribution of blood volume is altered in patients with obesity. TBV can be divided into two functional compartments, the unstressed blood volume (UBV), defined as the volume of blood necessary to fill the entire vascular space, and the stressed blood volume (SBV), which is the volume in excess of unstressed volume that increases transmural pressure in the vasculature (Figure 5). The relative distribution of these volumes is importantly regulated by the SNS, whereby sympathetic venoconstriction increases SBV and the SBV/TBV ratio, a teleologically important adaptation to augment venous return to the heart during stress. Sorimachi et al.152 have recently shown that SBV is increased in patients with HFpEF compared with controls, and notably both SBV and SBV/TBV ratio were directly correlated with BMI, suggesting that abnormalities in venous capacitance play an important role in obesity-related HFpEF (Figure 5).

Figure 5.

Figure 5

Altered venous capacitance in obese HFpEF. (A) Total blood volume (TBV) can be functionally divided into an UBV, defined as the volume of blood necessary to fill the vascular space, and a SBV, which is defined as the volume in excess of UBV that increases wall tension and vascular pressure. Under normal conditions SBV constitutes only 20% of TBV, but with venoconstriction, blood is translocated from the unstressed to stressed functional compartments, increasing SBV and the SBV/TBV ratio. (B) When compared with controls, patients with HFpEF display an increase in both absolute SBV and SBV/TBV ratio, indicating reduced venous capacitance, both at rest and during exercise. (C) In patients with HFpEF and non-cardiac dyspnoea (NCD), increasing body mass index (BMI) is associated with greater impairment in venous capacitance during exercise, estimated by the SBV/TBV. Panel A modified from Fudim et al.153 Panels B and C from Sorimachi et al.152

Neurohormonal activation may contribute to cardiac structural remodelling by inducing cardiac fibrosis and sodium retention-mediated blood pressure rise and plasma volume expansion. Studies have shown that leptin is an important mediator of neurohormonal activation, and hyperleptinaemia contributes to obesity-related cardiovascular disease.154–156 Leptin enhances both RAAS and renal SNS, and stimulates aldosterone synthesis from the adrenal glands. Leptin receptors have been reported to be expressed in the heart,157 and experimental studies have demonstrated that leptin can promote cardiomyocyte hypertrophy and fibrosis by mediating angiotensin II, mineralocorticoid, and endothelin-1 receptors.155,158

5.7.2. Systemic inflammation

A detailed review of obesity-related inflammation is beyond the scope of this review, but growing evidence suggest that metabo-inflammation plays an important role in the pathophysiology of HFpEF, especially when coupled with increases in visceral fat.110,159–161 Infiltration of macrophages causes adipose tissue inflammation,162 promoting up-regulation of proinflammatory adipokines (e.g. leptin, TNF-α, IL-6, and resistin) and the down-regulation of anti-inflammatory adipokines (e.g. adiponectin, omentin-1), leading to chronic and low-grade systemic inflammation.163 Obesity may combine with hypertension to induce proinflammatory macrophage polarization (M1 phenotype) that contributes to the alterations in myocardial substrate availability and utilization described above.164 Chronic activation of these proinflammatory pathways is believed to increase systemic inflammation and nitrosative stress, and lead to capillary rarefaction and mitochondrial dysfunction, and impairment endothelium-dependent vasodilation, contributing to abnormalities in the heart, vasculature, skeletal muscle, and other organs in HFpEF through a wide variety of pathways, as recently reviewed.159,165

5.8. Regional adipose distribution and HFpEF

5.8.1. Abdominal visceral fat

In addition to increases in EAT described above, a number of recent studies have pointed to a pivotal role for VAT in obese HFpEF.98,166,167 In a multinational prospective study, central adiposity, as measured by waist circumference and reflecting VAT, was directly related with worse outcomes in patients with HFpEF, whereas an inverse relationship was found with BMI.168 A population-based study has shown that the risk of HFpEF is most strongly tied to increases in VAT rather than subcutaneous fat (Figure 6).10 This may relate to the higher degrees of adipocyte hypertrophy, insulin resistance, and FFA elevation among patients with increased VAT. In this regard, Wang and colleagues23 recently showed in mice that a unique cell line present in adipose tissue (p21Cip1 highly expressing cells) importantly contributes to insulin resistance, and treatment with senolytic drugs eliminates these cells in human fat while improving insulin sensitivity. Insulin resistance, accumulation of perivascular adipose tissue-derived adipokines and vascular inflammation interfere with vascular nitric oxide bioavailability to limit increases in blood flow during exercise.169,170 Such abnormalities are commonly observed in patients with HFpEF in both the peripheral and coronary microcirculations,14,131 potentially leading to impaired O2 delivery and utilization in skeletal muscle171–173 as well as myocardium to cause ischaemia and injury during stress.129 VAT expansion may also influence myocardiac structure via secreted profibrotic factors. Recent data from mouse models have shown that VAT-secreted osteopontin may play an important role in driving cardiac senescence.174 Osteopontin levels increased in wildtype animals during ageing, and VAT removal decreased circulating osteopontin, restored cardiac function, and decreased myocardial fibrosis and transforming growth factor β levels, collectively supporting a novel role for VAT-derived secretome in cardiac changes with ageing and HFpEF.

Figure 6.

Figure 6

(A) Hazard ratio (HR) plotted development of incident HFpEF in community-dwelling adults for measures of adiposity, showing greatest risk with increasing visceral rather than subcutaneous fat. (B and E) When compared with age and BMI-matched control women, women with HFpEF display 35% higher visceral adipose tissue (VAT) area, whereas there were no statistically significant differences observed in men. (C and F) Greater VAT in women was associated with higher exercise pulmonary capillary wedge pressure in women but not men, with significant sex interaction. (D) The estimated causal odds ratio (OR) for hypertension and diabetes with increasing VAT significantly exceeds unity in both men and women, but is substantially greater for women, particularly for diabetes. Panel A plotted from data in Rao et al.,10 Panels B, C, E, and F adapted from Sorimachi et al.,167 and Panel D plotted from data in Karlsson et al.175

VAT expansion may be even more detrimental for women (Figure 6), and may help explain the strong sex differential in prevalence of HFpEF.98 A Mendelian randomization study showed that increases in VAT are associated with greater risk for diabetes, hypertension, and hyperlipidaemia in both sexes, but the risk was substantially greater in women.175 A large community-based study in Taiwan showed that higher BMI and larger waist circumference were associated with subclinical LV contractile dysfunction (detected by myocardial strain imaging), more pronounced in women than men (P for interaction < 0.05), with detectable LV dysfunction at low anthropometric cutoffs in Asian women that are far lower than typically considered pathological in western countries (BMI > 23.4 kg/m2, waist circumference > 83 cm).84

Fat distribution is known to vary in women and men,176 with more gluteofemoral fat in women and more VAT in men, but around the time of menopause, VAT increases in women, which may contribute to the greater risk of HFpEF in women compared with men.177 This sex difference in the effects of ageing on body composition may be very important in HFpEF. A recent study showed that women with HFpEF display significantly greater VAT when compared with control women, even after matching for age and BMI; no such differences were present in men (Figure 6).167 In addition to these quantitative sex differences in VAT, there were qualitative differences as well. Increases in VAT in women were directly related to greater elevation in pulmonary capillary pressures during exercise, even as the absolute volume of VAT was lower compared with men, whereas in men there was no significant relationship present.167 A more recent study has confirmed this finding using waist circumference alone as a less robust, but more practical method to evaluate for central obesity, showing relationships with HFpEF severity in women but not men with HFpEF.178

5.8.2. Effects in kidneys, liver, and skeletal muscle

Increased renal fat deposition has been shown to be associated with microvascular proliferation and hyperfiltration in the kidney, with increased inflammatory and oxidative stress.179 Renal sodium avidity may be enhanced through compression by visceral, perinephric, and renal sinus fat, as well as neurohormonal activation.180,181 Visceral adiposity is often accompanied by increased fat deposition in the liver, further promoting inflammation, dyslipidaemia, and insulin resistance.161 Obesity is associated with increased inflammation in skeletal muscle, in which immune cell infiltration and proinflammatory activation occur in inter- and perimuscular adipose tissue.182 Recent studies have identified several critical abnormalities in the skeletal muscle, including adipose infiltration, capillary rarefaction, and mitochondrial dysfunction that importantly lead to symptoms and exercise intolerance in HFpEF,165,173,183,184 and many of these may be related to excess body fat and its impact on skeletal muscle.

6. The obesity paradox in HFpEF

Like most chronic diseases, obesity paradoxically protects against adverse outcomes in patients with established HFpEF, despite being a well-established risk factor for incident HFpEF.185–187 However, these findings come from cross-sectional studies, which are limited by survival bias (i.e. sicker obese patients die prior to assessment), and reverse causation (patients with the most severe HF have lost weight due to cachexia). A Mendelian randomization study, which overcomes the limitations of cross-sectional analyses, has raised questions with the obesity paradox in HF.188 The hazard ratio for HF mortality per 1 kg/m2 increase in BMI was 1.04 [95% confidence interval (CI), 1.03–1.06] in the observational cohort, suggesting a relatively minor risk. In contrast, the causal risk ratio for the same 1 kg/m2 increase in BMI via Mendelian randomization revealed a potentially stronger causal relationship between high BMI and HF mortality (causal risk ratio 1.18; 95% CI, 1.00–1.38), though differences in population characteristics might contribute to the differential relationships observed.188 Other studies have shown that when more specific measures of adiposity, such as weight circumference, are used in place of BMI, there is a continuous increase in risk of all-cause and cardiovascular mortality with greater body fat, with no paradox.189 The importance of different adiposity parameters for the assessment of outcomes in established HF was also demonstrated in a longitudinal multinational study, where higher BMI was associated with better outcomes, yet larger waist circumference was associated with worse outcomes, in patients with HF.168

6.1. Treatment

6.1.1. Diuretics

As described above, patients with obesity-related HFpEF often display volume expansion,15 and diuretics are effective to reduce filling pressures and congestion in these patients. However, patient with obese HFpEF may tolerate diuresis more poorly than non-obese HFpEF, with greater risk of worsening renal function during decongestion,190 despite the presence of more marked volume expansion.191

6.1.2. Mineralocorticoid antagonists

There is evidence that mineralocorticoid receptor antagonists (MRAs) may be of greater benefit in obese HFpEF.192 In mice, treatment with MRA was shown to reduce expression of proinflammatory factors in adipocytes.193 In the TOPCAT trial, treatment with spironolactone had no significant effect on the primary endpoint of HF hospitalization, cardiovascular death, or aborted sudden death in the overall trial cohort of patients with HF and EF ≥ 45%, but there was regional variation in treatment response, which was more favourable in patients with genuine HFpEF, and signal for benefit in patients with BMI above the median (interaction P = 0.11).194,195 Post hoc analyses from TOPCAT showed that patients with lower NTproBNP levels, a group that typically displays more prominent central adiposity,178 derived greater benefit from MRA,196 and patients with HFrEF and greater waist circumference displayed more favourable responses to eplerenone than those without central obesity.197 Further insight into the role for MRA in obese HFpEF will be provided by ongoing trials evaluating MRA in HFpEF (NCT02901184, NCT04727073, NCT04435626).

6.1.3. Neprilysin inhibitors

In the PARAGON-HF trial, treatment with the dual neprilysin/angiotensin receptor antagonist sacubitril valsartan tended to reduce the rate of HF hospitalizations or cardiovascular death, narrowly missing statistical significance (rate ratio, 0.87; P = 0.06).198 Neprilysin degrades BNP and other vasoactive peptides, and is expressed on adipocytes, suggesting excessive breakdown by adipocyte-derived neprilysin could contribute to obese HFpEF.192,199 There was no subgroup analysis in PARAGON-HF stratified by obesity status, but there was no significant treatment interaction between patients with NTproBNP levels above and below the median.198 A subsequent analysis from PARAGON-HF reported on regional differences in HFpEF characteristics, showing highest rates of obesity in HFpEF in North America, but treatment effects did not vary by geographic region.200

6.1.4. Statins

Statins reduce systemic inflammation,201 which is strongly associated with the pathogenesis of obese HFpEF.159 Treatment with statins induces EAT regression in a lipoprotein-lowering independent manner, which may be related to anti-inflammatory effects.202 Statins have been associated with lower event rates in observational studies in HFpEF,203,204 though randomized trial data are lacking.

6.1.5. SGLT2 inhibitors

Epicardial fat thickening and inflammation may also play a role in HFpEF, and sodium-glucose co-transporter 2 inhibitors (SGLT2i) have been shown to increase adipocyte glucose uptake, reduce the secretion of proinflammatory chemokines, and improve the differentiation of EAT cells in adipose samples of patient undergoing heart surgery.205 One study showed that SGLT2i treatment for 6 months decreased EAT and TNF-α levels compared with usual therapy in patients with diabetes and coronary disease (n = 40),206 while another placebo-controlled trial (n = 56) reported that treatment with SGLT2i for 12 weeks did not affect EAT, intramyocardial fat, or PCr/ATP, but resulted in favourable reductions in VAT and liver fat.207 In patients with HFpEF (EF > 40%), treatment with SGLT2i has been shown to reduce risk of HF hospitalization or cardiovascular death in the EMPEROR-preserved trial,208 and to improve patient-reported health status in EMPEROR and other trials.208–210

Submaximal functional capacity assessed by the 6 min walk test was improved in the PRESERVED-HF trial, conducted within the USA,209 whereas no such benefit was observed in the similarly sized EMPERIAL trial.211 The reasons for the discrepant results are not clear, but may relate to differences in baseline characteristics in the participants studied: patients in EMPERIAL were notably less obese (BMI 29.6 vs. 34.9 kg/m2), and less likely to be women (43 vs. 57%) when compared with PRESERVED-HF.209,211 Because health status,212 functional capacity,15,91 and haemodynamic abnormalities152 are so strongly tied to excess body fat in HFpEF (especially among women167), this could explain the disparate results. There was no significant treatment effect interaction by obesity status in EMPEROR preserved, but this trial was largely conducted outside the USA, with mean BMI of <30 kg/m2.208 The mechanisms responsible for benefits from SGLT2i in HFpEF remain unknown. Weight loss is typically observed following treatment with SGLT2i, but the magnitude is modest (1–2 kg) and unlikely to account for the clinical improvements observed in obese or non-obese HFpEF.

6.1.6. Weight loss

An analysis from 4 community-based cohorts showed that risk for HFpEF increased by 34% for each 1 standard deviation increase in BMI, suggesting that weight loss might reduce risk for HFpEF, or attenuate HFpEF severity among patients with prevalent disease.213 Weight loss can be achieved through lifestyle interventions including diet and exercise training, pharmacotherapy, and bariatric surgery. Gepner et al.214 recently showed that the combination of Mediterranean/low-carbohydrate diet mobilized specific fat depots including EAT and VAT, and exercise training also interpedently contributed to reduction in VAT. In a landmark trial, Kitzman and colleagues215 demonstrated that either weight loss via caloric restriction (prepared meals) or aerobic exercise reduced LV mass and inflammatory markers, improved exercise capacity, and enhanced quality of life in patients with obese HFpEF, while the combination of training and weight loss was additive (Figure 7).216

Figure 7.

Figure 7

(A) Pooled hazard ratio for cardiovascular outcomes following bariatric surgery vs. control in a meta-analysis of observational studies. (B) Changes in aerobic capacity (peak oxygen consumption, VO2) from a randomized controlled trial in obese HFpEF with diet-induced weight loss, exercise training, or combination. In observational studies of patients without heart failure, weight loss is associated with improved haemodynamics (C) and myocardial energetics, assessed by phosphocreatine/ATP ratio (D). Panel A plotted from data in van Veldhuisen et al.,217 Panel B adapted from Upadhya et al.,216 Panel C from Reddy et al.,218 and Panel D from Rayner et al.70

Bariatric surgery leads to the most dramatic and sustained weight loss, and while no randomized trial has been performed to date, observational data strongly indicate a reduction in the risk for new onset HF.217 One small, non-controlled study in patients with mild HFpEF showed improvements in symptom severity with signal for improved diastolic relaxation, despite no effect on intramyocardial fat.219 Reddy et al.218 showed in a meta-analysis of invasive studies that weight loss decreases total body O2 consumption, heart rate, and blood pressure, along with 20–25% reductions in cardiac filling pressures and PA pressures (Figure 7). While these evaluations were not carried out in patients with HFpEF, each of these effects would be expected to improve clinical status in such patients. Three randomized clinical trials are currently underway evaluating the role for pharmacological weight loss therapies in HFpEF, two using the glucagon-like peptide-1 (GLP-1) receptor agonist semaglutide and one the dual glucose-dependent insulinotropic polypeptide (GIP), GLP-1 agonist tirzepatide (NCT04788511, NCT04916470, NCT04847557).

7. Future directions

Patients with obese HFpEF may be better positioned to respond to therapies under development and evaluation in HFpEF owing to key pathophysiological features. For example, the greater abnormalities in venous capacitance observed in obese HFpEF discussed above152 may position these patients to respond more favourably to novel treatments targeting volume distribution, as with splanchnic nerve ablation.153 Because patients with obese HFpEF display increased EAT and ventricular interdependence, novel therapies to reduce pericardial restraint, such as minimally invasive pericardiotomy, may also be more efficacious in this patient population.220,221

Other novel treatments may target fat reduction through different pathways, alter the consequences of excess body fat on organ function, modify the adipocyte secretome, or target cells in obesity that promote disease progression, such as senescent cells.23,222 In addition to the GLP-1 and combined GIP/GLP-1 agonists, other novel treatments to facilitate weight loss through decreased appetite and/or increased satiety are at varying stages of development. Other potential treatments may target weight loss through increased catabolism. Novel classes of drugs may target proton transport across the inner mitochondrial membrane, essentially lead to increased fat breakdown by uncoupling oxidative phosphorylation from the electron transport chain, increasing resting metabolic rate.223,224 The prototypic compound is 2,4-dinitrophenol, which was used in the 1930s as a highly effective weight loss drug, but was withdrawn because of the high risk of malignant hyperthermia. A related compound with wider therapeutic index (HU6) is under development. Mishra et al.225 recently demonstrated that inhibition of phosphodiesterase 9A in mice reduced body (and myocardial) fat and stimulated mitochondrial activity in a PPARα-dependent manner, without altering activity levels or food intake. Notably, favourable cardiometabolic and weight loss effects were exclusively observed in male animals and ovariectomized females, additional evidence indicating a strong sexual dimorphism in treatment response.

Other drugs may target the metabolic derangements in obesity that lead to cardiac and non-cardiac organ dysfunction, particularly the excess use of circulating FFA as fuel, such as partial fatty acid oxidation inhibitors, carnitine palmitoyltransferase 1 inhibitors, and mitochondrial-targeted antioxidants.226 In HFrEF and hypertrophic cardiomyopathy, treatment with the carnitine palmitoyltransferase-1 inhibitor perhexiline was shown to improve PCr/ATP ratio and functional capacity, presumably by reducing myocardial fat oxidation and shifting to more favourable glucose dependence.227,228 Therapies targeting obesity-associated inflammation may also hold promise to improve clinical status in obesity-related HFpEF, such as the IL-6 inhibitor ziltivekimab,229 and warrant testing in obesity-related HFpEF given the evidence of systemic inflammation in this cohort.91

8. Conclusion

Obesity is the one of the most important causes of the clinical syndrome of HFpEF, leading to symptomatic HF through a wide variety of pathophysiological mechanisms. While there have been important advances in our understanding over the past decade, many knowledge gaps persist, and further study is needed to advance our understanding about this disease, ranging from cellular mechanisms to clinical trials, in order to improve outcomes for the ever expanding group of patients with obesity and HFpEF.

Contributor Information

Barry A Borlaug, Department of Cardiovascular Diseases, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN 55905, USA.

Michael D Jensen, Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA.

Dalane W Kitzman, Department of Internal Medicine, Section on Cardiology, Wake Forest School of Medicine, Winston-Salem, NC, USA.

Carolyn S P Lam, Duke-National University of Singapore, Singapore, Singapore.

Masaru Obokata, Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.

Oliver J Rider, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK.

Funding

Supported in part by NIH grants: R01 HL128526 and U01 HL160226 (B.A.B.), R01AG045551; R01AG18915; P30AG021332; U24AG059624; 1U01HL160272 (D.W.K.), by the British Heart Foundation FS/16/70/32157 (O.J.R.), and by the US Department of Defense: W81XWH2210245 (B.A.B.).

Data availability

Data will be made available upon request to the corresponding author.

References

  • 1. GBD 2017 Disease and Injury Incidence and Prevalence Collaborators . Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 2018;392:1789–1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Tsao  CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Ferguson JF, Generoso G, Ho JE, Kalani R, Khan SS, Kissela BM, Knutson KL, Levine DA, Lewis TT, Liu J, Loop MS, Ma J, Mussolino ME, Navaneethan SD, Perak AM, Poudel R, Rezk-Hanna M, Roth GA, Schroeder EB, Shah SH, Thacker EL, VanWagner LB, Virani SS, Voecks JH, Wang NY, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2022 Update: a report from the American Heart Association. Circulation 2022;145:e153–e639. [DOI] [PubMed] [Google Scholar]
  • 3. Dunlay  SM, Roger VL, Redfield MM. Epidemiology of heart failure with preserved ejection fraction. Nat Rev Cardiol 2017;14:591–602. [DOI] [PubMed] [Google Scholar]
  • 4. Tsao  CW, Lyass A, Enserro D, Larson MG, Ho JE, Kizer JR, Gottdiener JS, Psaty BM, Vasan RS. Temporal trends in the incidence of and mortality associated with heart failure with preserved and reduced ejection fraction. JACC Heart Fail 2018;6:678–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Ng  ACT, Delgado V, Borlaug BA, Bax JJ. Diabesity: the combined burden of obesity and diabetes on heart disease and the role of imaging. Nat Rev Cardiol 2021;18:291–304. [DOI] [PubMed] [Google Scholar]
  • 6. Ward  ZJ, Bleich SN, Cradock AL, Barrett JL, Giles CM, Flax C, Long MW, Gortmaker SL. Projected U.S. state-level prevalence of adult obesity and severe obesity. N Engl J Med 2019;381:2440–2450. [DOI] [PubMed] [Google Scholar]
  • 7. Ho  JE, Lyass A, Lee DS, Vasan RS, Kannel WB, Larson MG, Levy D. Predictors of new-onset heart failure: differences in preserved versus reduced ejection fraction. Circ Heart Fail 2013;6:279–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Brouwers  FP, de Boer RA, van der Harst P, Voors AA, Gansevoort RT, Bakker SJ, Hillege HL, van Veldhuisen DJ, van Gilst WH. Incidence and epidemiology of new onset heart failure with preserved vs. reduced ejection fraction in a community-based cohort: 11-year follow-up of PREVEND. Eur Heart J 2013;34:1424–1431. [DOI] [PubMed] [Google Scholar]
  • 9. Pandey  A, LaMonte M, Klein L, Ayers C, Psaty BM, Eaton CB, Allen NB, de Lemos JA, Carnethon M, Greenland P, Berry JD. Relationship between physical activity, body mass index, and risk of heart failure. J Am Coll Cardiol 2017;69:1129–1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Rao  VN, Zhao D, Allison MA, Guallar E, Sharma K, Criqui MH, Cushman M, Blumenthal RS, Michos ED. Adiposity, incident heart failure, its subtypes: MESA (multi-ethnic study of atherosclerosis). JACC Heart Fail 2018;6:999–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Ndumele  CE, Matsushita K, Lazo M, Bello N, Blumenthal RS, Gerstenblith G, Nambi V, Ballantyne CM, Solomon SD, Selvin E, Folsom AR, Coresh J. Obesity and subtypes of incident cardiovascular disease. J Am Heart Assoc 2016;5:e003921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Obokata  M, Reddy YNV, Melenovsky V, Sorimachi H, Jarolim P, Borlaug BA. Uncoupling between intravascular and distending pressures leads to underestimation of circulatory congestion in obesity. Eur J Heart Fail 2022;24:353–361. [DOI] [PubMed] [Google Scholar]
  • 13. Paulus  WJ, Tschope C. A novel paradigm for heart failure with preserved ejection fraction: comorbidities drive myocardial dysfunction and remodeling through coronary microvascular endothelial inflammation. J Am Coll Cardiol 2013;62:263–271. [DOI] [PubMed] [Google Scholar]
  • 14. Borlaug  BA, Olson TP, Lam CS, Flood KS, Lerman A, Johnson BD, Redfield MM. Global cardiovascular reserve dysfunction in heart failure with preserved ejection fraction. J Am Coll Cardiol 2010;56:845–854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Obokata  M, Reddy YN, 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] [PMC free article] [PubMed] [Google Scholar]
  • 16. Mohammed  SF, Hussain S, Mirzoyev SA, Edwards WD, Maleszewski JJ, Redfield MM. Coronary microvascular rarefaction and myocardial fibrosis in heart failure with preserved ejection fraction. Circulation 2015;131:550–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Kitzman  DW, Lam CSP. Obese heart failure with preserved ejection fraction phenotype: from Pariah to Central Player. Circulation 2017;136:20–23. [DOI] [PubMed] [Google Scholar]
  • 18. Kitzman  DW, Shah SJ. The HFpEF obesity phenotype: the elephant in the room. J Am Coll Cardiol 2016;68:200–203. [DOI] [PubMed] [Google Scholar]
  • 19. Verbrugge  FH, Omote K, Reddy YNV, Sorimachi H, Obokata M, Borlaug BA. Heart failure with preserved ejection fraction in patients with normal natriuretic peptide levels is associated with increased morbidity and mortality. Eur Heart J 2022;43:1941–1951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Cefalu  WT, Wang ZQ, Werbel S, Bell-Farrow A, Crouse JR 3rd, Hinson WH, Terry JG, Anderson R. Contribution of visceral fat mass to the insulin resistance of aging. Metabolism 1995;44:954–959. [DOI] [PubMed] [Google Scholar]
  • 21. Seidell  JC, Bjorntorp P, Sjostrom L, Kvist H, Sannerstedt R. Visceral fat accumulation in men is positively associated with insulin, glucose, and C-peptide levels, but negatively with testosterone levels. Metabolism 1990;39:897–901. [DOI] [PubMed] [Google Scholar]
  • 22. Kuk  JL, Katzmarzyk PT, Nichaman MZ, Church TS, Blair SN, Ross R. Visceral fat is an independent predictor of all-cause mortality in men. Obesity (Silver Spring) 2006;14:336–341. [DOI] [PubMed] [Google Scholar]
  • 23. Wang  L, Wang B, Gasek NS, Zhou Y, Cohn RL, Martin DE, Zuo W, Flynn WF, Guo C, Jellison ER, Kim T, Prata L, Palmer AK, Li M, Inman CL, Barber LS, Al-Naggar IMA, Zhou Y, Du W, Kshitiz KG, Meves A, Tchkonia T, Kirkland JL, Robson P, Xu M. Targeting p21(Cip1) highly expressing cells in adipose tissue alleviates insulin resistance in obesity. Cell Metab 2022;34:75–89.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Haykowsky  MJ, Nicklas BJ, Brubaker PH, Hundley WG, Brinkley TE, Upadhya B, Becton JT, Nelson MD, Chen H, Kitzman DW. Regional adipose distribution and its relationship to exercise intolerance in older obese patients who have heart failure with preserved ejection fraction. JACC Heart Fail 2018;6:640–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Arner  E, Westermark PO, Spalding KL, Britton T, Ryden M, Frisen J, Bernard S, Arner P. Adipocyte turnover: relevance to human adipose tissue morphology. Diabetes 2010;59:105–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. O’Connell  J, Lynch L, Cawood TJ, Kwasnik A, Nolan N, Geoghegan J, McCormick A, O’Farrelly C, O’Shea D. The relationship of omental, subcutaneous adipocyte size to metabolic disease in severe obesity. PLoS One 2010;5:e9997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Goodpaster  BH, Thaete FL, Simoneau JA, Kelley DE. Subcutaneous abdominal fat and thigh muscle composition predict insulin sensitivity independently of visceral fat. Diabetes 1997;46:1579–1585. [DOI] [PubMed] [Google Scholar]
  • 28. Jensen  MD, Johnson CM. Contribution of leg and splanchnic free fatty acid (FFA) kinetics to postabsorptive FFA flux in men and women. Metabolism 1996;45:662–666. [DOI] [PubMed] [Google Scholar]
  • 29. Tchoukalova  YD, Koutsari C, Karpyak MV, Votruba SB, Wendland E, Jensen MD. Subcutaneous adipocyte size and body fat distribution. Am J Clin Nutr 2008;87:56–63. [DOI] [PubMed] [Google Scholar]
  • 30. Richelsen  B, Pedersen SB, Moller-Pedersen T, Bak JF. Regional differences in triglyceride breakdown in human adipose tissue: effects of catecholamines, insulin, and prostaglandin E2. Metabolism 1991;40:990–996. [DOI] [PubMed] [Google Scholar]
  • 31. Votruba  SB, Jensen MD. Sex-specific differences in leg fat uptake are revealed with a high-fat meal. Am J Physiol Endocrinol Metab 2006;291:E1115–E1123. [DOI] [PubMed] [Google Scholar]
  • 32. Votruba  SB, Mattison RS, Dumesic DA, Koutsari C, Jensen MD. Meal fatty acid uptake in visceral fat in women. Diabetes 2007;56:2589–2597. [DOI] [PubMed] [Google Scholar]
  • 33. Danforth E  J. Failure of adipocyte differentiation causes type II diabetes mellitus? Nat Genet 2000;26:13. [DOI] [PubMed] [Google Scholar]
  • 34. Hammarstedt  A, Graham TE, Kahn BB. Adipose tissue dysregulation and reduced insulin sensitivity in non-obese individuals with enlarged abdominal adipose cells. Diabetol Metab Syndr 2012;4:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Hammarstedt  A, Gogg S, Hedjazifar S, Nerstedt A, Smith U. Impaired adipogenesis and dysfunctional adipose tissue in human hypertrophic obesity. Physiol Rev 2018;98:1911–1941. [DOI] [PubMed] [Google Scholar]
  • 36. Tchoukalova  YD, Votruba SB, Tchkonia T, Giorgadze N, Kirkland JL, Jensen MD. Regional differences in cellular mechanisms of adipose tissue gain with overfeeding. Proc Natl Acad Sci U S A 2010;107:18226–18231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Nielsen  S, Guo Z, Albu JB, Klein S, O’Brien PC, Jensen MD. Energy expenditure, sex, and endogenous fuel availability in humans. J Clin Invest 2003;111:981–988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Jensen  MD, Haymond MW, Rizza RA, Cryer PE, Miles JM. Influence of body fat distribution on free fatty acid metabolism in obesity. J Clin Invest 1989;83:1168–1173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Snijder  MB, Dekker JM, Visser M, Bouter LM, Stehouwer CD, Yudkin JS, Heine RJ, Nijpels G, Seidell JC. Trunk fat and leg fat have independent and opposite associations with fasting and postload glucose levels: the Hoorn study. Diabetes Care 2004;27:372–377. [DOI] [PubMed] [Google Scholar]
  • 40. Magkos  F, Fabbrini E, Conte C, Patterson BW, Klein S. Relationship between adipose tissue lipolytic activity and skeletal muscle insulin resistance in nondiabetic women. J Clin Endocrinol Metab 2012;97:E1219–E1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Shadid  S, Kanaley JA, Sheehan MT, Jensen MD. Basal and insulin-regulated free fatty acid and glucose metabolism in humans. Am J Physiol Endocrinol Metab 2007;292:E1770–E1774. [DOI] [PubMed] [Google Scholar]
  • 42. Bush  NC, Basu R, Rizza RA, Nair KS, Khosla S, Jensen MD. Insulin-mediated FFA suppression is associated with triglyceridemia and insulin sensitivity independent of adiposity. J Clin Endocrinol Metab 2012;97:4130–4138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. DeFronzo  RA. Pathogenesis of type 2 diabetes mellitus. Med Clin North Am 2004;88:787–835, ix. [DOI] [PubMed] [Google Scholar]
  • 44. McGarry  JD. Banting lecture 2001: dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Diabetes 2002;51:7–18. [DOI] [PubMed] [Google Scholar]
  • 45. Girousse  A, Tavernier G, Valle C, Moro C, Mejhert N, Dinel AL, Houssier M, Roussel B, Besse-Patin A, Combes M, Mir L, Monbrun L, Bezaire V, Prunet-Marcassus B, Waget A, Vila I, Caspar-Bauguil S, Louche K, Marques MA, Mairal A, Renoud ML, Galitzky J, Holm C, Mouisel E, Thalamas C, Viguerie N, Sulpice T, Burcelin R, Arner P, Langin D. Partial inhibition of adipose tissue lipolysis improves glucose metabolism and insulin sensitivity without alteration of fat mass. PLoS Biol 2013;11:e1001485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Randle  PJ, Garland PB, Hales CN, Newsholme EA. The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1963;1:785–789. [DOI] [PubMed] [Google Scholar]
  • 47. Kanaley  JA, Shadid S, Sheehan MT, Guo Z, Jensen MD. Relationship between plasma free fatty acid, intramyocellular triglycerides and long-chain acylcarnitines in resting humans. J Physiol 2009;587:5939–5950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Fontana  L, Eagon JC, Trujillo ME, Scherer PE, Klein S. Visceral fat adipokine secretion is associated with systemic inflammation in obese humans. Diabetes 2007;56:1010–1013. [DOI] [PubMed] [Google Scholar]
  • 49. Mohamed-Ali  V, Goodrick S, Rawesh A, Katz DR, Miles JM, Yudkin JS, Klein S, Coppack SW. Subcutaneous adipose tissue releases interleukin-6, but not tumor necrosis factor-alpha, in vivo. J Clin Endocrinol Metab 1997;82:4196–4200. [DOI] [PubMed] [Google Scholar]
  • 50. Espinosa De Ycaza  AE, Sondergaard E, Morgan-Bathke M, Carranza Leon BG, Lytle KA, Ramos P, Kirkland JL, Tchkonia T, Jensen MD. Senescent cells in human adipose tissue: a cross-sectional study. Obesity (Silver Spring) 2021;29:1320–1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Goodpaster  BH, Thaete FL, Kelley DE. Thigh adipose tissue distribution is associated with insulin resistance in obesity and in type 2 diabetes mellitus. Am J Clin Nutr 2000;71:885–892. [DOI] [PubMed] [Google Scholar]
  • 52. Ali  AH, Koutsari C, Mundi M, Stegall MD, Heimbach JK, Taler SJ, Nygren J, Thorell A, Bogachus LD, Turcotte LP, Bernlohr D, Jensen MD. Free fatty acid storage in human visceral and subcutaneous adipose tissue: role of adipocyte proteins. Diabetes 2011;60:2300–2307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Roust  LR, Jensen MD. Postprandial free fatty acid kinetics are abnormal in upper body obesity. Diabetes 1993;42:1567–1573. [DOI] [PubMed] [Google Scholar]
  • 54. Guo  Z, Hensrud DD, Johnson CM, Jensen MD. Regional postprandial fatty acid metabolism in different obesity phenotypes. Diabetes 1999;48:1586–1592. [DOI] [PubMed] [Google Scholar]
  • 55. Borlaug  BA. Evaluation and management of heart failure with preserved ejection fraction. Nat Rev Cardiol 2020;17:559–573. [DOI] [PubMed] [Google Scholar]
  • 56. Pieske  B, Tschope C, de Boer RA, Fraser AG, Anker SD, Donal E, Edelmann F, Fu M, Guazzi M, Lam CSP, Lancellotti P, Melenovsky V, Morris DA, Nagel E, Pieske-Kraigher E, Ponikowski P, Solomon SD, Vasan RS, Rutten FH, Voors AA, Ruschitzka F, Paulus WJ, Seferovic P, Filippatos G. How to diagnose heart failure with preserved ejection fraction: the HFA-PEFF diagnostic algorithm: a consensus recommendation from the Heart Failure Association (HFA) of the European Society of Cardiology (ESC). Eur Heart J 2019;40:3297–3317. [DOI] [PubMed] [Google Scholar]
  • 57. Lam  CS, Cheng S, Choong K, Larson MG, Murabito JM, Newton-Cheh C, Bhasin S, McCabe EL, Miller KK, Redfield MM, Vasan RS, Coviello AD, Wang TJ. Influence of sex and hormone status on circulating natriuretic peptides. J Am Coll Cardiol 2011;58:618–626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. McHugh  K, DeVore AD, Wu J, Matsouaka RA, Fonarow GC, Heidenreich PA, Yancy CW, Green JB, Altman N, Hernandez AF. Heart failure with preserved ejection fraction and diabetes: JACC state-of-the-art review. J Am Coll Cardiol 2019;73:602–611. [DOI] [PubMed] [Google Scholar]
  • 59. Jackson  AM, Rorth R, Liu J, Kristensen SL, Anand IS, Claggett BL, Cleland JGF, Chopra VK, Desai AS, Ge J, Gong J, Lam CSP, Lefkowitz MP, Maggioni AP, Martinez F, Packer M, Pfeffer MA, Pieske B, Redfield MM, Rizkala AR, Rouleau JL, Seferovic PM, Tromp J, Van Veldhuisen DJ, Yilmaz MB, Zannad F, Zile MR, Kober L, Petrie MC, Jhund PS, Solomon SD, McMurray JJV, PARAGON-HF Committees and Investigators . Diabetes and pre-diabetes in patients with heart failure and preserved ejection fraction. Eur J Heart Fail 2022;24:497–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Russo  C, Jin Z, Homma S, Rundek T, Elkind MS, Sacco RL, Di Tullio MR. Effect of obesity and overweight on left ventricular diastolic function: a community-based study in an elderly cohort. J Am Coll Cardiol 2011;57:1368–1374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. 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] [PMC free article] [PubMed] [Google Scholar]
  • 62. 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] [PMC free article] [PubMed] [Google Scholar]
  • 63. Russo  C, Sera F, Jin Z, Palmieri V, Homma S, Rundek T, Elkind MS, Sacco RL, Di Tullio MR. Abdominal adiposity, general obesity, and subclinical systolic dysfunction in the elderly: a population-based cohort study. Eur J Heart Fail 2016;18:537–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Schwarzl  M, Ojeda F, Zeller T, Seiffert M, Becher PM, Munzel T, Wild PS, Blettner M, Lackner KJ, Pfeiffer N, Beutel ME, Blankenberg S, Westermann D. Risk factors for heart failure are associated with alterations of the LV end-diastolic pressure-volume relationship in non-heart failure individuals: data from a large-scale, population-based cohort. Eur Heart J 2016;37:1807–1814. [DOI] [PubMed] [Google Scholar]
  • 65. Bello  NA, Cheng S, Claggett B, Shah AM, Ndumele CE, Roca GQ, Santos AB, Gupta D, Vardeny O, Aguilar D, Folsom AR, Butler KR, Kitzman DW, Coresh J, Solomon SD. Association of weight and body composition on cardiac structure and function in the ARIC study (Atherosclerosis Risk in Communities). Circ Heart Fail 2016;9:e002978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Selvaraj  S, Martinez EE, Aguilar FG, Kim KY, Peng J, Sha J, Irvin MR, Lewis CE, Hunt SC, Arnett DK, Shah SJ. Association of central adiposity with adverse cardiac mechanics: findings from the hypertension genetic epidemiology network study. Circ Cardiovasc Imaging 2016;9:e004396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Fernandes-Silva  MM, Shah AM, Claggett B, Cheng S, Tanaka H, Silvestre OM, Nadruz W, Borlaug BA, Solomon SD. Adiposity, body composition and ventricular-arterial stiffness in the elderly: the Atherosclerosis Risk in Communities Study. Eur J Heart Fail 2018;20:1191–1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Wong  CY, O’Moore-Sullivan T, Leano R, Byrne N, Beller E, Marwick TH. Alterations of left ventricular myocardial characteristics associated with obesity. Circulation 2004;110:3081–3087. [DOI] [PubMed] [Google Scholar]
  • 69. Peterson  LR, Waggoner AD, Schechtman KB, Meyer T, Gropler RJ, Barzilai B, Davila-Roman VG. Alterations in left ventricular structure and function in young healthy obese women: assessment by echocardiography and tissue Doppler imaging. J Am College Cardiol 2004;43:1399–1404. [DOI] [PubMed] [Google Scholar]
  • 70. Rayner  JJ, Peterzan MA, Watson WD, Clarke WT, Neubauer S, Rodgers CT, Rider OJ. Myocardial energetics in obesity: enhanced ATP delivery through creatine kinase with blunted stress response. Circulation 2020;141:1152–1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Peterson  LR, Herrero P, Schechtman KB, Racette SB, Waggoner AD, Kisrieva-Ware Z, Dence C, Klein S, Marsala J, Meyer T, Gropler RJ. Effect of obesity and insulin resistance on myocardial substrate metabolism and efficiency in young women. Circulation 2004;109:2191–2196. [DOI] [PubMed] [Google Scholar]
  • 72. Alpert  MA. Obesity cardiomyopathy: pathophysiology and evolution of the clinical syndrome. Am J Med Sci 2001;321:225–236. [DOI] [PubMed] [Google Scholar]
  • 73. Turkbey  EB, McClelland RL, Kronmal RA, Burke GL, Bild DE, Tracy RP, Arai AE, Lima JA, Bluemke DA. The impact of obesity on the left ventricle: the Multi-Ethnic Study of Atherosclerosis (MESA). JACC Cardiovasc Imaging 2010;3:266–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Rider  OJ, Francis JM, Ali MK, Byrne J, Clarke K, Neubauer S, Petersen SE. Determinants of left ventricular mass in obesity; a cardiovascular magnetic resonance study. J Cardiovasc Magn Reson 2009;11:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Rider  OJ, Lewandowski A, Nethononda R, Petersen SE, Francis JM, Pitcher A, Holloway CJ, Dass S, Banerjee R, Byrne JP, Leeson P, Neubauer S. Gender-specific differences in left ventricular remodelling in obesity: insights from cardiovascular magnetic resonance imaging. Eur Heart J 2013;34:292–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Athithan  L, Gulsin GS, McCann GP, Levelt E. Diabetic cardiomyopathy: pathophysiology, theories and evidence to date. World J Diabetes 2019;10:490–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Rider  OJ, Petersen SE, Francis JM, Ali MK, Hudsmith LE, Robinson MR, Clarke K, Neubauer S. Ventricular hypertrophy and cavity dilatation in relation to body mass index in women with uncomplicated obesity. Heart 2011;97:203–208. [DOI] [PubMed] [Google Scholar]
  • 78. Levelt  E, Mahmod M, Piechnik SK, Ariga R, Francis JM, Rodgers CT, Clarke WT, Sabharwal N, Schneider JE, Karamitsos TD, Clarke K, Rider OJ, Neubauer S. Relationship between left ventricular structural and metabolic remodeling in type 2 diabetes. Diabetes 2016;65:44–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Levelt  E, Pavlides M, Banerjee R, Mahmod M, Kelly C, Sellwood J, Ariga R, Thomas S, Francis J, Rodgers C, Clarke W, Sabharwal N, Antoniades C, Schneider J, Robson M, Clarke K, Karamitsos T, Rider O, Neubauer S. Ectopic and visceral fat deposition in lean and obese patients with type 2 diabetes. J Am Coll Cardiol 2016;68:53–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Wong  CY, O’Moore-Sullivan T, Leano R, Hukins C, Jenkins C, Marwick TH. Association of subclinical right ventricular dysfunction with obesity. J Am Coll Cardiol 2006;47:611–616. [DOI] [PubMed] [Google Scholar]
  • 81. Shah  AM, Claggett B, Sweitzer NK, Shah SJ, Anand IS, Liu L, Pitt B, Pfeffer MA, Solomon SD. Prognostic importance of impaired systolic function in heart failure with preserved ejection fraction and the impact of spironolactone. Circulation 2015;132:402–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Borlaug  BA, Lam CS, Roger VL, Rodeheffer RJ, Redfield MM. Contractility and ventricular systolic stiffening in hypertensive heart disease insights into the pathogenesis of heart failure with preserved ejection fraction. J Am Coll Cardiol 2009;54:410–418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Borlaug  BA, Kane GC, Melenovsky V, Olson TP. Abnormal right ventricular-pulmonary artery coupling with exercise in heart failure with preserved ejection fraction. Eur Heart J 2016;37:3293–3302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Lai  YH, Liu ME, Su CH, Yun CH, Liu CY, Hou CJ, Hu KC, Hung CL, Yeh HI, Lam CSP. Obesity-related changes in cardiac structure and function among Asian men and women. J Am Coll Cardiol 2017;69:2876–2878. [DOI] [PubMed] [Google Scholar]
  • 85. Aslam  MI, Hahn VS, Jani V, Hsu S, Sharma K, Kass DA. Reduced right ventricular sarcomere contractility in heart failure with preserved ejection fraction and severe obesity. Circulation 2021;143:965–967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Reddy  YNV, Obokata M, Verbrugge FH, Lin G, Borlaug BA. Atrial dysfunction in patients with heart failure with preserved ejection fraction and atrial fibrillation. J Am Coll Cardiol 2020;76:1051–1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Khan  MS, Memon MM, Murad MH, Vaduganathan M, Greene SJ, Hall M, Triposkiadis F, Lam CSP, Shah AM, Butler J, Shah SJ. Left atrial function in heart failure with preserved ejection fraction: a systematic review and meta-analysis. Eur J Heart Fail 2020;22:472–485. [DOI] [PubMed] [Google Scholar]
  • 88. Melenovsky  V, Hwang SJ, Redfield MM, Zakeri R, Lin G, Borlaug BA. Left atrial remodeling and function in advanced heart failure with preserved or reduced ejection fraction. Circ Heart Fail 2015;8:295–303. [DOI] [PubMed] [Google Scholar]
  • 89. Gottdiener  JS, Reda DJ, Williams DW, Materson BJ. Left atrial size in hypertensive men: influence of obesity, race and age. Department of Veterans Affairs Cooperative Study Group on Antihypertensive Agents. J Am Coll Cardiol 1997;29:651–658. [DOI] [PubMed] [Google Scholar]
  • 90. Wang  TJ, Parise H, Levy D, D’Agostino RB Sr, Wolf PA, Vasan RS, Benjamin EJ. Obesity and the risk of new-onset atrial fibrillation. JAMA 2004; 292:2471–2477. [DOI] [PubMed] [Google Scholar]
  • 91. Reddy  YNV, Lewis GD, Shah SJ, Obokata M, Abou-Ezzedine OF, Fudim M, Sun JL, Chakraborty H, McNulty S, LeWinter MM, Mann DL, Stevenson LW, Redfield MM, Borlaug BA. Characterization of the obese phenotype of heart failure with preserved ejection fraction: a RELAX trial ancillary study. Mayo Clin Proc 2019;94:1199–1209. [DOI] [PubMed] [Google Scholar]
  • 92. Koepp  KE, Obokata M, Reddy YNV, Olson TP, Borlaug BA. Hemodynamic and functional impact of epicardial adipose tissue in heart failure with preserved ejection fraction. JACC Heart Fail 2020;8:657–666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Borlaug  BA, Maleszewski JJ. The heavy heart of HFpEF. Eur Heart J 2020;41:3447. [DOI] [PubMed] [Google Scholar]
  • 94. Borlaug  BA, Reddy YNV. The role of the pericardium in heart failure: implications for pathophysiology and treatment. JACC Heart Fail 2019;7:574–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Mazurek  T, Zhang L, Zalewski A, Mannion JD, Diehl JT, Arafat H, Sarov-Blat L, O’Brien S, Keiper EA, Johnson AG, Martin J, Goldstein BJ, Shi Y. Human epicardial adipose tissue is a source of inflammatory mediators. Circulation 2003;108:2460–2466. [DOI] [PubMed] [Google Scholar]
  • 96. Patel  VB, Mori J, McLean BA, Basu R, Das SK, Ramprasath T, Parajuli N, Penninger JM, Grant MB, Lopaschuk GD, Oudit GY. ACE2 deficiency worsens epicardial adipose tissue inflammation and cardiac dysfunction in response to diet-induced obesity. Diabetes 2016;65:85–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Venteclef  N, Guglielmi V, Balse E, Gaborit B, Cotillard A, Atassi F, Amour J, Leprince P, Dutour A, Clement K, Hatem SN. Human epicardial adipose tissue induces fibrosis of the atrial myocardium through the secretion of adipo-fibrokines. Eur Heart J 2015;36:795–805a. [DOI] [PubMed] [Google Scholar]
  • 98. Packer  M, Lam CSP, Lund LH, Maurer MS, Borlaug BA. Characterization of the inflammatory-metabolic phenotype of heart failure with a preserved ejection fraction: a hypothesis to explain influence of sex on the evolution and potential treatment of the disease. Eur J Heart Fail 2020;22:1551–1567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Kitzman  DW, Upadhya B, Vasu S. What the dead can teach the living: systemic nature of heart failure with preserved ejection fraction. Circulation 2015;131:522–524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Pugliese  NR, Paneni F, Mazzola M, De Biase N, Del Punta L, Gargani L, Mengozzi A, Virdis A, Nesti L, Taddei S, Flammer A, Borlaug BA, Ruschitzka F, Masi S. Impact of epicardial adipose tissue on cardiovascular haemodynamics, metabolic profile, and prognosis in heart failure. Eur J Heart Fail 2021;23:1858–1871. [DOI] [PubMed] [Google Scholar]
  • 101. Gorter  TM, van Woerden G, Rienstra M, Dickinson MG, Hummel YM, Voors AA, Hoendermis ES, van Veldhuisen DJ. Epicardial adipose tissue and invasive hemodynamics in heart failure with preserved ejection fraction. JACC Heart Fail 2020;8:667–676. [DOI] [PubMed] [Google Scholar]
  • 102. van Woerden  G, van Veldhuisen DJ, Manintveld OC, van Empel VPM, Willems TP, de Boer RA, Rienstra M, Westenbrink BD, Gorter TM. Epicardial adipose tissue and outcome in heart failure with mid-range and preserved ejection fraction. Circ Heart Fail 2021;15:e009238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Tromp  J, Packer M, Lam CS. The diverging role of epicardial adipose tissue in heart failure with reduced and preserved ejection fraction: not all fat is created equal. Eur J Heart Fail 2021;23:1872–1874. [DOI] [PubMed] [Google Scholar]
  • 104. Tromp  J, Bryant JA, Jin X, van Woerden G, Asali S, Yiying H, Liew OW, Ching JCP, Jaufeerally F, Loh SY, Sim D, Lee S, Soon D, Tay WT, Packer M, van Veldhuisen DJ, Chin C, Richards AM, Lam CSP. Epicardial fat in heart failure with reduced versus preserved ejection fraction. Eur J Heart Fail 2021;23:835–838. [DOI] [PubMed] [Google Scholar]
  • 105. Most  AS, Brachfeld N, Gorlin R, Wahren J. Free fatty acid metabolism of the human heart at rest. J Clin Invest 1969;48:1177–1188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Wisneski  JA, Gertz EW, Neese RA, Mayr M. Myocardial metabolism of free fatty acids. Studies with 14C-labeled substrates in humans. J Clin Invest 1987;79:359–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Rayner  JJ, Abdesselam I, Peterzan MA, Akoumianakis I, Akawi N, Antoniades C, Tomlinson JW, Neubauer S, Rider OJ. Very low calorie diets are associated with transient ventricular impairment before reversal of diastolic dysfunction in obesity. Int J Obes (Lond) 2019;43:2536–2544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Chiu  HC, Kovacs A, Ford DA, Hsu FF, Garcia R, Herrero P, Saffitz JE, Schaffer JE. A novel mouse model of lipotoxic cardiomyopathy. J Clin Invest 2001;107:813–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Schiattarella  GG, Altamirano F, Kim SY, Tong D, Ferdous A, Piristine H, Dasgupta S, Wang X, French KM, Villalobos E, Spurgin SB, Waldman M, Jiang N, May HI, Hill TM, Luo Y, Yoo H, Zaha VG, Lavandero S, Gillette TG, Hill JA. Xbp1s-FoxO1 axis governs lipid accumulation and contractile performance in heart failure with preserved ejection fraction. Nat Commun 2021;12:1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Schiattarella  GG, Altamirano F, Tong D, French KM, Villalobos E, Kim SY, Luo X, Jiang N, May HI, Wang ZV, Hill TM, Mammen PPA, Huang J, Lee DI, Hahn VS, Sharma K, Kass DA, Lavandero S, Gillette TG, Hill JA. Nitrosative stress drives heart failure with preserved ejection fraction. Nature 2019;568:351–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Sharma  S, Adrogue JV, Golfman L, Uray I, Lemm J, Youker K, Noon GP, Frazier OH, Taegtmeyer H. Intramyocardial lipid accumulation in the failing human heart resembles the lipotoxic rat heart. FASEB J 2004;18:1692–1700. [DOI] [PubMed] [Google Scholar]
  • 112. Banerjee  R, Rial B, Holloway CJ, Lewandowski AJ, Robson MD, Osuchukwu C, Schneider JE, Leeson P, Rider OJ, Neubauer S. Evidence of a direct effect of myocardial steatosis on LV hypertrophy and diastolic dysfunction in adult and adolescent obesity. JACC Cardiovasc Imaging 2015;8:1468–1470. [DOI] [PubMed] [Google Scholar]
  • 113. Wu  C-K, Lee J-K, Hsu J-C, Su M-M, Wu Y-F, Lin T-T, Lan C-W, Hwang J-J, Lin L-Y. Myocardial adipose deposition and the development of heart failure with preserved ejection fraction. Eur J Heart Fail 2020;22:445–454. [DOI] [PubMed] [Google Scholar]
  • 114. Peterson  LR, Soto PF, Herrero P, Mohammed BS, Avidan MS, Schechtman KB, Dence C, Gropler RJ. Impact of gender on the myocardial metabolic response to obesity. JACC Cardiovasc Imaging 2008;1:424–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Peterson  LR, Herrero P, Coggan AR, Kisrieva-Ware Z, Saeed I, Dence C, Koudelis D, McGill JB, Lyons MR, Novak E, Davila-Roman VG, Waggoner AD, Gropler RJ. Type 2 diabetes, obesity, and sex difference affect the fate of glucose in the human heart. Am J Physiol Heart Circ Physiol 2015;308:H1510–H1516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Stanley  WC, Lopaschuk GD, McCormack JG. Regulation of energy substrate metabolism in the diabetic heart. Cardiovasc Res 1997;34:25–33. [DOI] [PubMed] [Google Scholar]
  • 117. Herrero  P, Peterson LR, McGill JB, Matthew S, Lesniak D, Dence C, Gropler RJ. Increased myocardial fatty acid metabolism in patients with type 1 diabetes mellitus. J Am Coll Cardiol 2006;47:598–604. [DOI] [PubMed] [Google Scholar]
  • 118. Voros  G, Ector J, Garweg C, Droogne W, Van Cleemput J, Peersman N, Vermeersch P, Janssens S. Increased cardiac uptake of ketone bodies and free fatty acids in human heart failure and hypertrophic left ventricular remodeling. Circ Heart Fail 2018;11:e004953. [DOI] [PubMed] [Google Scholar]
  • 119. Korvald  C, Elvenes OP, Myrmel T. Myocardial substrate metabolism influences left ventricular energetics in vivo. Am J Physiol Heart Circ Physiol 2000;278:H1345–H1351. [DOI] [PubMed] [Google Scholar]
  • 120. Murray  AJ, Anderson RE, Watson GC, Radda GK, Clarke K. Uncoupling proteins in human heart. Lancet 2004;364:1786–1788. [DOI] [PubMed] [Google Scholar]
  • 121. Haim  TE, Wang W, Flagg TP, Tones MA, Bahinski A, Numann RE, Nichols CG, Nerbonne JM. Palmitate attenuates myocardial contractility through augmentation of repolarizing Kv currents. J Mol Cell Cardiol 2010;48:395–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Kim  G, Jo K, Kim KJ, Lee YH, Han E, Yoon HJ, Wang HJ, Kang ES, Yun M. Visceral adiposity is associated with altered myocardial glucose uptake measured by (18)FDG-PET in 346 subjects with normal glucose tolerance, prediabetes, and type 2 diabetes. Cardiovasc Diabetol 2015;14:148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Listenberger  LL, Ory DS, Schaffer JE. Palmitate-induced apoptosis can occur through a ceramide-independent pathway. J Biol Chem 2001;276:14890–14895. [DOI] [PubMed] [Google Scholar]
  • 124. Unger  RH, Orci L. Diseases of liporegulation: new perspective on obesity and related disorders. FASEB J 2001;15:312–321. [DOI] [PubMed] [Google Scholar]
  • 125. Viljanen  AP, Karmi A, Borra R, Parkka JP, Lepomaki V, Parkkola R, Lautamaki R, Jarvisalo M, Taittonen M, Ronnemaa T, Iozzo P, Knuuti J, Nuutila P, Raitakari OT. Effect of caloric restriction on myocardial fatty acid uptake, left ventricular mass, and cardiac work in obese adults. Am J Cardiol 2009;103:1721–1726. [DOI] [PubMed] [Google Scholar]
  • 126. Lin  CH, Kurup S, Herrero P, Schechtman KB, Eagon JC, Klein S, Davila-Roman VG, Stein RI, Dorn GW 2nd, Gropler RJ, Waggoner AD, Peterson LR. Myocardial oxygen consumption change predicts left ventricular relaxation improvement in obese humans after weight loss. Obesity 2011;19:1804–1812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Hannukainen  JC, Lautamaki R, Parkka J, Strandberg M, Saunavaara V, Hurme S, Soinio M, Dadson P, Virtanen KA, Gronroos T, Forsback S, Salminen P, Iozzo P, Nuutila P. Reversibility of myocardial metabolism and remodelling in morbidly obese patients 6 months after bariatric surgery. Diabetes Obes Metab 2018;20:963–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Madigan  MJ  Jr, Racette SB, Coggan AR, Stein RI, McCue LM, Gropler RJ, Peterson LR. Weight loss affects intramyocardial glucose metabolism in obese humans. Circ Cardiovasc Imaging 2019;12:e009241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Obokata  M, Reddy YNV, Melenovsky V, Kane GC, Olson TP, Jarolim P, Borlaug BA. Myocardial injury and cardiac reserve in patients with heart failure and preserved ejection fraction. J Am Coll Cardiol 2018;72:29–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Shah  SJ, Lam CSP, Svedlund S, Saraste A, Hage C, Tan RS, Beussink-Nelson L, Fermer ML, Broberg MA, Gan LM, Lund LH. Prevalence and correlates of coronary microvascular dysfunction in heart failure with preserved ejection fraction: PROMIS-HFpEF. Eur Heart J 2018;39:3439–3450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Yang  JH, Obokata M, Reddy YNV, Redfield MM, Lerman A, Borlaug BA. Endothelium-dependent and independent coronary microvascular dysfunction in patients with heart failure with preserved ejection fraction. Eur J Heart Fail 2020;22:432–441. [DOI] [PubMed] [Google Scholar]
  • 132. Taegtmeyer  H. Ischemia and glucose metabolism. Circulation 1997;96:3810–3811. [PubMed] [Google Scholar]
  • 133. Rider  OJ, Francis JM, Ali MK, Holloway C, Pegg T, Robson MD, Tyler D, Byrne J, Clarke K, Neubauer S. Effects of catecholamine stress on diastolic function and myocardial energetics in obesity. Circulation 2012;125:1511–1519. [DOI] [PubMed] [Google Scholar]
  • 134. Rayner  JJ, Peterzan MA, Clarke WT, Rodgers CT, Neubauer S, Rider OJ. Obesity modifies the energetic phenotype of dilated cardiomyopathy. Eur Heart J 2021;43:868–877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Rider  OJ, Francis JM, Tyler D, Byrne J, Clarke K, Neubauer S. Effects of weight loss on myocardial energetics and diastolic function in obesity. Int J Cardiovasc Imaging 2013;29:1043–1050. [DOI] [PubMed] [Google Scholar]
  • 136. Lam  CS, Roger VL, Rodeheffer RJ, Borlaug BA, Enders FT, Redfield MM. Pulmonary hypertension in heart failure with preserved ejection fraction: a community-based study. J Am Coll Cardiol 2009;53:1119–1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Vanderpool  RR, Saul M, Nouraie M, Gladwin MT, Simon MA. Association between hemodynamic markers of pulmonary hypertension and outcomes in heart failure with preserved ejection fraction. JAMA Cardiol 2018;3:298–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Borlaug  BA, Nishimura RA, Sorajja P, Lam CS, Redfield MM. Exercise hemodynamics enhance diagnosis of early heart failure with preserved ejection fraction. Circ Heart Fail 2010;3:588–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Satoh  T, Wang L, Espinosa-Diez C, Wang B, Hahn SA, Noda K, Rochon ER, Dent MR, Levine AR, Baust JJ, Wyman S, Wu YL, Triantafyllou GA, Tang Y, Reynolds M, Shiva S, Hilaire CS, Gomez D, Goncharov DA, Goncharova EA, Chan SY, Straub AC, Lai YC, McTiernan CF, Gladwin MT. Metabolic syndrome mediates ROS-miR-193b-NFYA-dependent downregulation of soluble guanylate cyclase and contributes to exercise-induced pulmonary hypertension in heart failure with preserved ejection fraction. Circulation 2021;144:615–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Fayyaz  AU, Sabbah MS, Dasari S, Griffiths LG, DuBrock HM, Wang Y, Charlesworth MC, Borlaug BA, Jenkins SM, Edwards WD, Redfield MM. Histologic and proteomic remodeling of the pulmonary veins and arteries in a porcine model of chronic pulmonary venous hypertension. Cardiovasc Res 2022:cvac005. doi: 10.1093/cvr/cvac005. Published online ahead of print 12 January 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Melenovsky  V, Hwang SJ, Lin G, Redfield MM, Borlaug BA. Right heart dysfunction in heart failure with preserved ejection fraction. Eur Heart J 2014;35:3452–3462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. 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. Eur Heart J 2019;40:689–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Gorter  TM, Hoendermis ES, van Veldhuisen DJ, Voors AA, Lam CS, Geelhoed B, Willems TP, van Melle JP. Right ventricular dysfunction in heart failure with preserved ejection fraction: a systematic review and meta-analysis. Eur J Heart Fail 2016;18:1472–1487. [DOI] [PubMed] [Google Scholar]
  • 144. Messerli  FH, Christie B, DeCarvalho JG, Aristimuno GG, Suarez DH, Dreslinski GR, Frohlich ED. Obesity, essential hypertension. Hemodynamics, intravascular volume, sodium excretion, plasma renin activity. Arch Intern Med 1981;141:81–85. [DOI] [PubMed] [Google Scholar]
  • 145. Gollasch  M. Vasodilator signals from perivascular adipose tissue. Br J Pharmacol 2012;165:633–642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Reddy  YN, Melenovsky V, Redfield MM, Nishimura RA, Borlaug BA. High-output heart failure: a 15-year experience. J Am Coll Cardiol 2016;68:473–482. [DOI] [PubMed] [Google Scholar]
  • 147. Hall  JE, do Carmo JM, da Silva AA, Wang Z, Hall ME. Obesity-induced hypertension: interaction of neurohumoral, renal mechanisms. Circ Res 2015;116:991–1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Heymsfield  SB, Wadden TA. Mechanisms, pathophysiology, and management of obesity. N Engl J Med 2017;376:254–266. [DOI] [PubMed] [Google Scholar]
  • 149. Alvarez  GE, Beske SD, Ballard TP, Davy KP. Sympathetic neural activation in visceral obesity. Circulation 2002;106:2533–2536. [DOI] [PubMed] [Google Scholar]
  • 150. Chandra  A, Neeland IJ, Berry JD, Ayers CR, Rohatgi A, Das SR, Khera A, McGuire DK, de Lemos JA, Turer AT. The relationship of body mass and fat distribution with incident hypertension: observations from the Dallas Heart Study. J Am Coll Cardiol 2014;64:997–1002. [DOI] [PubMed] [Google Scholar]
  • 151. Camara  NO, Iseki K, Kramer H, Liu ZH, Sharma K. Kidney disease and obesity: epidemiology, mechanisms and treatment. Nat Rev Nephrol 2017;13:181–190. [DOI] [PubMed] [Google Scholar]
  • 152. Sorimachi  H, Burkhoff D, Verbrugge FH, Omote K, Obokata M, Reddy YNV, Takahashi N, Sunagawa K, Borlaug BA. Obesity, venous capacitance, and venous compliance in heart failure with preserved ejection fraction. Eur J Heart Fail 2021;23:1648–1658. [DOI] [PubMed] [Google Scholar]
  • 153. Fudim  M, Kaye DM, Borlaug BA, Shah SJ, Rich S, Kapur NK, Costanzo MR, Brener MI, Sunagawa K, Burkhoff D. Venous tone and stressed blood volume in heart failure: JACC review topic of the week. J Am Coll Cardiol 2022;79:1858–1869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Lim  K, Burke SL, Head GA. Obesity-related hypertension and the role of insulin and leptin in high-fat-fed rabbits. Hypertension 2013;61:628–634. [DOI] [PubMed] [Google Scholar]
  • 155. Huby  AC, Antonova G, Groenendyk J, Gomez-Sanchez CE, Bollag WB, Filosa JA, Belin de Chantemele EJ. Adipocyte-derived hormone leptin is a direct regulator of aldosterone secretion, which promotes endothelial dysfunction and cardiac fibrosis. Circulation 2015;132:2134–2145. [DOI] [PubMed] [Google Scholar]
  • 156. Xue  B, Yu Y, Zhang Z, Guo F, Beltz TG, Thunhorst RL, Felder RB, Johnson AK. Leptin mediates high-fat diet sensitization of angiotensin II-elicited hypertension by upregulating the brain renin-angiotensin system and inflammation. Hypertension 2016;67:970–976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Purdham  DM, Zou MX, Rajapurohitam V, Karmazyn M. Rat heart is a site of leptin production and action. Am J Physiol Heart Circ Physiol 2004;287:H2877–H2884. [DOI] [PubMed] [Google Scholar]
  • 158. Sweeney  G. Cardiovascular effects of leptin. Nat Rev Cardiol 2010;7:22–29. [DOI] [PubMed] [Google Scholar]
  • 159. Schiattarella  GG, Rodolico D, Hill JA. Metabolic inflammation in heart failure with preserved ejection fraction. Cardiovasc Res 2021;117:423–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Sanders-van Wijk  S, Tromp J, Beussink-Nelson L, Hage C, Svedlund S, Saraste A, Swat SA, Sanchez C, Njoroge J, Tan RS, Fermer ML, Gan LM, Lund LH, Lam CSP, Shah SJ. Proteomic evaluation of the comorbidity-inflammation paradigm in heart failure with preserved ejection fraction: results from the PROMIS-HFpEF study. Circulation 2020;142:2029–2044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Fuster  JJ, Ouchi N, Gokce N, Walsh K. Obesity-induced changes in adipose tissue microenvironment and their impact on cardiovascular disease. Circ Res 2016;118:1786–1807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Amano  SU, Cohen JL, Vangala P, Tencerova M, Nicoloro SM, Yawe JC, Shen Y, Czech MP, Aouadi M. Local proliferation of macrophages contributes to obesity-associated adipose tissue inflammation. Cell Metab 2014;19:162–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Ouchi  N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nat Rev Immunol 2011;11:85–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Mouton  AJ, Li X, Hall ME, Hall JE. Obesity, hypertension, and cardiac dysfunction: novel roles of immunometabolism in macrophage activation and inflammation. Circ Res 2020;126:789–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Pandey  A, Shah SJ, Butler J, Kellogg DL Jr, Lewis GD, Forman DE, Mentz RJ, Borlaug BA, Simon MA, Chirinos JA, Fielding RA, Volpi E, Molina AJA, Haykowsky MJ, Sam F, Goodpaster BH, Bertoni AG, Justice JN, White JP, Ding J, Hummel SL, LeBrasseur NK, Taffet GE, Pipinos II, Kitzman D. Exercise intolerance in older adults with heart failure with preserved ejection fraction: JACC state-of-the-art review. J Am Coll Cardiol 2021;78:1166–1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Kitzman  DW, Nicklas BJ. Pivotal role of excess intra-abdominal adipose in the pathogenesis of metabolic/obese HFpEF. JACC Heart Fail 2018;6:1008–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Sorimachi  H, Obokata M, Takahashi N, Reddy YNV, Jain CC, Verbrugge FH, Koepp KE, Khosla S, Jensen MD, Borlaug BA. Pathophysiologic importance of visceral adipose tissue in women with heart failure and preserved ejection fraction. Eur Heart J 2021;42:1595–1605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Chandramouli  C, Tay WT, Bamadhaj NS, Tromp J, Teng TK, Yap JJL, MacDonald MR, Hung CL, Streng K, Naik A, Wander GS, Sawhney J, Ling LH, Richards AM, Anand I, Voors AA, Lam CSP, ASIAN-HF Investigators . Association of obesity with heart failure outcomes in 11 Asian regions: a cohort study. PLoS Med 2019;16:e1002916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Virdis  A, Duranti E, Rossi C, Dell’Agnello U, Santini E, Anselmino M, Chiarugi M, Taddei S, Solini A. Tumour necrosis factor-alpha participates on the endothelin-1/nitric oxide imbalance in small arteries from obese patients: role of perivascular adipose tissue. Eur Heart J 2015;36:784–794. [DOI] [PubMed] [Google Scholar]
  • 170. Greenstein  AS, Khavandi K, Withers SB, Sonoyama K, Clancy O, Jeziorska M, Laing I, Yates AP, Pemberton PW, Malik RA, Heagerty AM. Local inflammation and hypoxia abolish the protective anticontractile properties of perivascular fat in obese patients. Circ Heart Fail 2009;119:1661–1670. [DOI] [PubMed] [Google Scholar]
  • 171. Dhakal  BP, Malhotra R, Murphy RM, Pappagianopoulos PP, Baggish AL, Weiner RB, Houstis NE, Eisman AS, Hough SS, Lewis GD. Mechanisms of exercise intolerance in heart failure with preserved ejection fraction: the role of abnormal peripheral oxygen extraction. Circ Heart Fail 2015;8:286–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Lee  JF, Barrett-O’Keefe Z, Nelson AD, Garten RS, Ryan JJ, Nativi-Nicolau JN, Richardson RS, Wray DW. Impaired skeletal muscle vasodilation during exercise in heart failure with preserved ejection fraction. Int J Cardiol 2016;211:14–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. 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] [PMC free article] [PubMed] [Google Scholar]
  • 174. Sawaki  D, Czibik G, Pini M, Ternacle J, Suffee N, Mercedes R, Marcelin G, Surenaud M, Marcos E, Gual P, Clement K, Hue S, Adnot S, Hatem SN, Tsuchimochi I, Yoshimitsu T, Henegar C, Derumeaux G. Visceral adipose tissue drives cardiac aging through modulation of fibroblast senescence by osteopontin production. Circulation 2018;138:809–822. [DOI] [PubMed] [Google Scholar]
  • 175. Karlsson  T, Rask-Andersen M, Pan G, Hoglund J, Wadelius C, Ek WE, Johansson A. Contribution of genetics to visceral adiposity and its relation to cardiovascular and metabolic disease. Nat Med 2019;25:1390–1395. [DOI] [PubMed] [Google Scholar]
  • 176. Krotkiewski  M, Bjorntorp P, Sjostrom L, Smith U. Impact of obesity on metabolism in men and women. Importance of regional adipose tissue distribution. J Clin Invest 1983;72:1150–1162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Greendale  GA, Sternfeld B, Huang M, Han W, Karvonen-Gutierrez C, Ruppert K, Cauley JA, Finkelstein JS, Jiang SF, Karlamangla AS. Changes in body composition and weight during the menopause transition. JCI Insight 2019;4:e124865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Sorimachi  H, Omote K, Omar M, Popovic D, Verbrugge FH, Reddy YNV, Lin G, Obokata M, Miles JM, Jensen MD, Borlaug BA. Sex and central obesity in heart failure with preserved ejection fraction. Eur J Heart Fail 2022;24:1359–1370. [DOI] [PubMed] [Google Scholar]
  • 179. Li  Z, Woollard JR, Wang S, Korsmo MJ, Ebrahimi B, Grande JP, Textor SC, Lerman A, Lerman LO. Increased glomerular filtration rate in early metabolic syndrome is associated with renal adiposity and microvascular proliferation. Am J Physiol Renal Physiol 2011;301:F1078–F1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Hall  JE, do Carmo JM, da Silva AA, Wang Z, Hall ME. Obesity, kidney dysfunction, hypertension: mechanistic links. Nat Rev Nephrol 2019;15:367–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Boorsma  EM, Ter Maaten JM, Voors AA, van Veldhuisen DJ. Renal compression in heart failure: the renal tamponade hypothesis. JACC Heart Fail 2022;10:175–183. [DOI] [PubMed] [Google Scholar]
  • 182. Wu  H, Ballantyne CM. Skeletal muscle inflammation and insulin resistance in obesity. J Clin Invest 2017;127:43–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. 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] [PMC free article] [PubMed] [Google Scholar]
  • 184. 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] [PMC free article] [PubMed] [Google Scholar]
  • 185. Haass  M, Kitzman DW, Anand IS, Miller A, Zile MR, Massie BM, Carson PE. Body mass index and adverse cardiovascular outcomes in heart failure patients with preserved ejection fraction: results from the Irbesartan in heart failure with preserved ejection fraction (I-PRESERVE) trial. Circ Heart Fail 2011;4:324–331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Gustafsson  F, Kragelund CB, Torp-Pedersen C, Seibaek M, Burchardt H, Akkan D, Thune JJ, Kober L, DIAMOND study group . Effect of obesity and being overweight on long-term mortality in congestive heart failure: influence of left ventricular systolic function. Eur Heart J 2005;26:58–64. [DOI] [PubMed] [Google Scholar]
  • 187. Padwal  R, McAlister FA, McMurray JJ, Cowie MR, Rich M, Pocock S, Swedberg K, Maggioni A, Gamble G, Ariti C, Earle N, Whalley G, Poppe KK, Doughty RN, Bayes-Genis A. The obesity paradox in heart failure patients with preserved versus reduced ejection fraction: a meta-analysis of individual patient data. Int J Obes (Lond) 2014;38:1110–1114. [DOI] [PubMed] [Google Scholar]
  • 188. Benn  M, Marott SCW, Tybjaerg-Hansen A, Nordestgaard BG. Obesity increases heart failure incidence and mortality: observational and Mendelian randomization studies totalling over 1 million individuals. Cardiovasc Res 2022;118:3576–3585. [DOI] [PubMed] [Google Scholar]
  • 189. Tsujimoto  T, Kajio H. Abdominal obesity is associated with an increased risk of all-cause mortality in patients with HFpEF. J Am Coll Cardiol 2017;70:2739–2749. [DOI] [PubMed] [Google Scholar]
  • 190. Reddy  YNV, Obokata M, Testani JM, Felker GM, Tang WHW, Abou-Ezzeddine OF, Sun JL, Chakrabothy H, McNulty S, Shah SJ, Lewis GD, Stevenson LW, Redfield MM, Borlaug BA. Adverse renal response to decongestion in the obese phenotype of heart failure with preserved ejection fraction. J Card Fail 2020;26:101–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Miller  WL, Borlaug BA. Impact of obesity on volume status in patients with ambulatory chronic heart failure. J Card Fail 2020;26:112–117. [DOI] [PubMed] [Google Scholar]
  • 192. Packer  M. Obesity-associated heart failure as a theoretical target for treatment with mineralocorticoid receptor antagonists. JAMA Cardiol 2018;3:883–887. [DOI] [PubMed] [Google Scholar]
  • 193. Guo  C, Ricchiuti V, Lian BQ, Yao TM, Coutinho P, Romero JR, Li J, Williams GH, Adler GK. Mineralocorticoid receptor blockade reverses obesity-related changes in expression of adiponectin, peroxisome proliferator-activated receptor-gamma, and proinflammatory adipokines. Circulation 2008;117:2253–2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Pitt  B, Pfeffer MA, Assmann SF, Boineau R, Anand IS, Claggett B, Clausell N, Desai AS, Diaz R, Fleg JL, Gordeev I, Harty B, Heitner JF, Kenwood CT, Lewis EF, O’Meara E, Probstfield JL, Shaburishvili T, Shah SJ, Solomon SD, Sweitzer NK, Yang S, McKinlay SM. Spironolactone for heart failure with preserved ejection fraction. N Engl J Med 2014;370:1383–1392. [DOI] [PubMed] [Google Scholar]
  • 195. Pfeffer  MA, Claggett B, Assmann SF, Boineau R, Anand IS, Clausell N, Desai AS, Diaz R, Fleg JL, Gordeev I, Heitner JF, Lewis EF, O’Meara E, Rouleau JL, Probstfield JL, Shaburishvili T, Shah SJ, Solomon SD, Sweitzer NK, McKinlay SM, Pitt B. Regional variation in patients and outcomes in the Treatment of Preserved Cardiac Function Heart Failure With an Aldosterone Antagonist (TOPCAT) trial. Circulation 2015;131:34–42. [DOI] [PubMed] [Google Scholar]
  • 196. Anand  IS, Claggett B, Liu J, Shah AM, Rector TS, Shah SJ, Desai AS, O’Meara E, Fleg JL, Pfeffer MA, Pitt B, Solomon SD. Interaction between spironolactone and natriuretic peptides in patients with heart failure and preserved ejection fraction: from the TOPCAT trial. JACC Heart Fail 2017;5:241–252. [DOI] [PubMed] [Google Scholar]
  • 197. Olivier  A, Pitt B, Girerd N, Lamiral Z, Machu JL, McMurray JJV, Swedberg K, van Veldhuisen DJ, Collier TJ, Pocock SJ, Rossignol P, Zannad F, Pizard A. Effect of eplerenone in patients with heart failure and reduced ejection fraction: potential effect modification by abdominal obesity. Insight from the EMPHASIS-HF trial. Eur J Heart Fail 2017;19:1186–1197. [DOI] [PubMed] [Google Scholar]
  • 198. Solomon  SD, McMurray JJV, Anand IS, Ge J, Lam CSP, Maggioni AP, Martinez F, Packer M, Pfeffer MA, Pieske B, Redfield MM, Rouleau JL, van Veldhuisen DJ, Zannad F, Zile MR, Desai AS, Claggett B, Jhund PS, Boytsov SA, Comin-Colet J, Cleland J, Dungen HD, Goncalvesova E, Katova T, Kerr Saraiva JF, Lelonek M, Merkely B, Senni M, Shah SJ, Zhou J, Rizkala AR, Gong J, Shi VC, Lefkowitz MP, PARAGON-HF Investigators and Committees . Angiotensin-neprilysin inhibition in heart failure with preserved ejection fraction. N Engl J Med 2019;381:1609–1620. [DOI] [PubMed] [Google Scholar]
  • 199. Packer  M, Kitzman DW. Obesity-related heart failure with a preserved ejection fraction: the mechanistic rationale for combining inhibitors of aldosterone, neprilysin, and sodium-glucose cotransporter-2. JACC Heart Fail 2018;6:633–639. [DOI] [PubMed] [Google Scholar]
  • 200. Tromp  J, Claggett BL, Liu J, Jackson AM, Jhund PS, Kober L, Widimsky J, Boytsov SA, Chopra VK, Anand IS, Ge J, Chen CH, Maggioni AP, Martinez F, Packer M, Pfeffer MA, Pieske B, Redfield MM, Rouleau JL, Van Veldhuisen DJ, Zannad F, Zile MR, Rizkala AR, Inubushi-Molessa A, Lefkowitz MP, Shi VC, McMurray JJV, Solomon SD, Lam CSP, PARAGON-HF Investigators . Global differences in heart failure with preserved ejection fraction: the PARAGON-HF trial. Circ Heart Fail 2021;14:e007901. [DOI] [PubMed] [Google Scholar]
  • 201. Jain  MK, Ridker PM. Anti-inflammatory effects of statins: clinical evidence and basic mechanisms. Nat Rev Drug Discov 2005;4:977–987. [DOI] [PubMed] [Google Scholar]
  • 202. Alexopoulos  N, Melek BH, Arepalli CD, Hartlage GR, Chen Z, Kim S, Stillman AE, Raggi P. Effect of intensive versus moderate lipid-lowering therapy on epicardial adipose tissue in hyperlipidemic post-menopausal women: a substudy of the BELLES trial (Beyond Endorsed Lipid Lowering with EBT Scanning). J Am Coll Cardiol 2013;61:1956–1961. [DOI] [PubMed] [Google Scholar]
  • 203. Fukuta  H, Sane DC, Brucks S, Little WC. Statin therapy may be associated with lower mortality in patients with diastolic heart failure: a preliminary report. Circulation 2005;112:357–363. [DOI] [PubMed] [Google Scholar]
  • 204. Alehagen  U, Benson L, Edner M, Dahlstrom U, Lund LH. Association between use of statins and mortality in patients with heart failure and ejection fraction of >/=50. Circ Heart Fail 2015;8:862–870. [DOI] [PubMed] [Google Scholar]
  • 205. Diaz-Rodriguez  E, Agra RM, Fernandez AL, Adrio B, Garcia-Caballero T, Gonzalez-Juanatey JR, Eiras S. Effects of dapagliflozin on human epicardial adipose tissue: modulation of insulin resistance, inflammatory chemokine production, and differentiation ability. Cardiovasc Res 2018;114:336–346. [DOI] [PubMed] [Google Scholar]
  • 206. Sato  T, Aizawa Y, Yuasa S, Kishi S, Fuse K, Fujita S, Ikeda Y, Kitazawa H, Takahashi M, Sato M, Okabe M. The effect of dapagliflozin treatment on epicardial adipose tissue volume. Cardiovasc Diabetol 2018;17:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Gaborit  B, Ancel P, Abdullah AE, Maurice F, Abdesselam I, Calen A, Soghomonian A, Houssays M, Varlet I, Eisinger M, Lasbleiz A, Peiretti F, Bornet CE, Lefur Y, Pini L, Rapacchi S, Bernard M, Resseguier N, Darmon P, Kober F, Dutour A. Effect of empagliflozin on ectopic fat stores and myocardial energetics in type 2 diabetes: the EMPACEF study. Cardiovasc Diabetol 2021;20:57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Anker  SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Bohm M, Brunner-La Rocca HP, Choi DJ, Chopra V, Chuquiure-Valenzuela E, Giannetti N, Gomez-Mesa JE, Janssens S, Januzzi JL, Gonzalez-Juanatey JR, Merkely B, Nicholls SJ, Perrone SV, Pina IL, Ponikowski P, Senni M, Sim D, Spinar J, Squire I, Taddei S, Tsutsui H, Verma S, Vinereanu D, Zhang J, Carson P, Lam CSP, Marx N, Zeller C, Sattar N, Jamal W, Schnaidt S, Schnee JM, Brueckmann M, Pocock SJ, Zannad F, Packer M, Investigators EM-PT. Empagliflozin in heart failure with a preserved ejection fraction. N Engl J Med 2021;385:1451–1461. [DOI] [PubMed] [Google Scholar]
  • 209. Nassif  ME, Windsor SL, Borlaug BA, Kitzman DW, Shah SJ, Tang F, Khariton Y, Malik AO, Khumri T, Umpierrez G, Lamba S, Sharma K, Khan SS, Chandra L, Gordon RA, Ryan JJ, Chaudhry SP, Joseph SM, Chow CH, Kanwar MK, Pursley M, Siraj ES, Lewis GD, Clemson BS, Fong M, Kosiborod MN. The SGLT2 inhibitor dapagliflozin in heart failure with preserved ejection fraction: a multicenter randomized trial. Nat Med 2021;27:1954–1960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210. Spertus  JA, Birmingham MC, Nassif M, Damaraju CV, Abbate A, Butler J, Lanfear DE, Lingvay I, Kosiborod MN, Januzzi JL. The SGLT2 inhibitor canagliflozin in heart failure: the CHIEF-HF remote, patient-centered randomized trial. Nat Med 2022;28:809–813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Abraham  WT, Lindenfeld J, Ponikowski P, Agostoni P, Butler J, Desai AS, Filippatos G, Gniot J, Fu M, Gullestad L, Howlett JG, Nicholls SJ, Redon J, Schenkenberger I, Silva-Cardoso J, Stork S, Krzysztof Wranicz J, Savarese G, Brueckmann M, Jamal W, Nordaby M, Peil B, Ritter I, Ustyugova A, Zeller C, Salsali A, Anker SD. Effect of empagliflozin on exercise ability and symptoms in heart failure patients with reduced and preserved ejection fraction, with and without type 2 diabetes. Eur Heart J 2021;42:700–710. [DOI] [PubMed] [Google Scholar]
  • 212. Reddy  YNV, Rikhi A, Obokata M, Shah SJ, Lewis GD, AbouEzzedine OF, Dunlay S, McNulty S, Chakraborty H, Stevenson LW, Redfield MM, Borlaug BA. Quality of life in heart failure with preserved ejection fraction: importance of obesity, functional capacity, and physical inactivity. Eur J Heart Fail 2020;22:1009–1018. [DOI] [PubMed] [Google Scholar]
  • 213. Savji  N, Meijers WC, Bartz TM, Bhambhani V, Cushman M, Nayor M, Kizer JR, Sarma A, Blaha MJ, Gansevoort RT, Gardin JM, Hillege HL, Ji F, Kop WJ, Lau ES, Lee DS, Sadreyev R, van Gilst WH, Wang TJ, Zanni MV, Vasan RS, Allen NB, Psaty BM, van der Harst P, Levy D, Larson M, Shah SJ, de Boer RA, Gottdiener JS, Ho JE. The association of obesity and cardiometabolic traits with incident HFpEF and HFrEF. JACC Heart Fail 2018;6:701–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Gepner  Y, Shelef I, Schwarzfuchs D, Zelicha H, Tene L, Yaskolka Meir A, Tsaban G, Cohen N, Bril N, Rein M, Serfaty D, Kenigsbuch S, Komy O, Wolak A, Chassidim Y, Golan R, Avni-Hassid H, Bilitzky A, Sarusi B, Goshen E, Shemesh E, Henkin Y, Stumvoll M, Bluher M, Thiery J, Ceglarek U, Rudich A, Stampfer MJ, Shai I. Effect of distinct lifestyle interventions on mobilization of fat storage pools: CENTRAL magnetic resonance imaging randomized controlled trial. Circulation 2018;137:1143–1157. [DOI] [PubMed] [Google Scholar]
  • 215. 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] [PMC free article] [PubMed] [Google Scholar]
  • 216. Upadhya  B, Pisani B, Kitzman DW. Evolution of a geriatric syndrome: pathophysiology and treatment of heart failure with preserved ejection fraction. J Am Geriatr Soc 2017;65:2431–2440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. van Veldhuisen  SL, Gorter TM, van Woerden G, de Boer RA, Rienstra M, Hazebroek EJ, van Veldhuisen DJ. Bariatric surgery and cardiovascular disease: a systematic review and meta-analysis. Eur Heart J 2022;43:1955–1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218. Reddy  YNV, Anantha-Narayanan M, Obokata M, Koepp KE, Erwin P, Carter RE, Borlaug BA. Hemodynamic effects of weight loss in obesity: a systematic review and meta-analysis. JACC Heart Fail 2019;7:678–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Mikhalkova  D, Holman SR, Jiang H, Saghir M, Novak E, Coggan AR, O’Connor R, Bashir A, Jamal A, Ory DS, Schaffer JE, Eagon JC, Peterson LR. Bariatric surgery-induced cardiac and lipidomic changes in obesity-related heart failure with preserved ejection fraction. Obesity (Silver Spring) 2018;26:284–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. Borlaug  BA, Schaff HV, Pochettino A, Pedrotty DM, Asirvatham SJ, Abel MD, Carter RE, Mauermann WJ. Pericardiotomy enhances left ventricular diastolic reserve with volume loading in humans. Circulation 2018;138:2295–2297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221. Borlaug  BA, Carter RE, Melenovsky V, DeSimone CV, Gaba P, Killu A, Naksuk N, Lerman L, Asirvatham SJ. Percutaneous pericardial resection: a novel potential treatment for heart failure with preserved ejection fraction. Circ Heart Fail 2017;10:e003612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Gasek  NS, Kuchel GA, Kirkland JL, Xu M. Strategies for targeting senescent cells in human disease. Nat Aging 2021;1:870–879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Berry  BJ, Trewin AJ, Amitrano AM, Kim M, Wojtovich AP. Use the protonmotive force: mitochondrial uncoupling and reactive oxygen species. J Mol Biol 2018;430:3873–3891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Modriansky  M, Gabrielova E. Uncouple my heart: the benefits of inefficiency. J Bioenerg Biomembr 2009;41:133–136. [DOI] [PubMed] [Google Scholar]
  • 225. Mishra  S, Sadagopan N, Dunkerly-Eyring B, Rodriguez S, Sarver DC, Ceddia RP, Murphy SA, Knutsdottir H, Jani VP, Ashok D, Oeing CU, O’Rourke B, Gangoiti JA, Sears DD, Wong GW, Collins S, Kass DA. Inhibition of phosphodiesterase type 9 reduces obesity and cardiometabolic syndrome in mice. J Clin Invest 2021;131:e148798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Noordali  H, Loudon BL, Frenneaux MP, Madhani M. Cardiac metabolism – a promising therapeutic target for heart failure. Pharmacol Ther 2018;182:95–114. [DOI] [PubMed] [Google Scholar]
  • 227. Beadle  RM, Williams LK, Kuehl M, Bowater S, Abozguia K, Leyva F, Yousef Z, Wagenmakers AJ, Thies F, Horowitz J, Frenneaux MP. Improvement in cardiac energetics by perhexiline in heart failure due to dilated cardiomyopathy. JACC Heart Fail 2015;3:202–211. [DOI] [PubMed] [Google Scholar]
  • 228. Abozguia  K, Elliott P, McKenna W, Phan TT, Nallur-Shivu G, Ahmed I, Maher AR, Kaur K, Taylor J, Henning A, Ashrafian H, Watkins H, Frenneaux M. Metabolic modulator perhexiline corrects energy deficiency and improves exercise capacity in symptomatic hypertrophic cardiomyopathy. Circulation 2010;122:1562–1569. [DOI] [PubMed] [Google Scholar]
  • 229. Ridker  PM, Rane M. Interleukin-6 signaling and anti-interleukin-6 therapeutics in cardiovascular disease. Circ Res 2021;128:1728–1746. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data will be made available upon request to the corresponding author.


Articles from Cardiovascular Research are provided here courtesy of Oxford University Press

RESOURCES