Skip to main content
European Heart Journal logoLink to European Heart Journal
. 2021 Aug 20;42(43):4420–4430. doi: 10.1093/eurheartj/ehab389

Heart failure with preserved ejection fraction in humans and mice: embracing clinical complexity in mouse models

Coenraad Withaar 1, Carolyn S P Lam 2,3, Gabriele G Schiattarella 4,5,6,7,8, Rudolf A de Boer 9,✉,#, Laura M G Meems 10,#
PMCID: PMC8599003  PMID: 34414416

Abstract

Heart failure (HF) with preserved ejection fraction (HFpEF) is a multifactorial disease accounting for a large and increasing proportion of all clinical HF presentations. As a clinical syndrome, HFpEF is characterized by typical signs and symptoms of HF, a distinct cardiac phenotype and raised natriuretic peptides. Non-cardiac comorbidities frequently co-exist and contribute to the pathophysiology of HFpEF. To date, no therapy has proven to improve outcomes in HFpEF, with drug development hampered, at least partly, by lack of consensus on appropriate standards for pre-clinical HFpEF models. Recently, two clinical algorithms (HFA-PEFF and H2FPEF scores) have been developed to improve and standardize the diagnosis of HFpEF. In this review, we evaluate the translational utility of HFpEF mouse models in the context of these HFpEF scores. We systematically recorded evidence of symptoms and signs of HF or clinical HFpEF features and included several cardiac and extra-cardiac parameters as well as age and sex for each HFpEF mouse model. We found that most of the pre-clinical HFpEF models do not meet the HFpEF clinical criteria, although some multifactorial models resemble human HFpEF to a reasonable extent. We therefore conclude that to optimize the translational value of mouse models to human HFpEF, a novel approach for the development of pre-clinical HFpEF models is needed, taking into account the complex HFpEF pathophysiology in humans.

Keywords: HFpEF, Mouse, Human, Translational, H2FPEF, HFA-PEFF

Graphical Abstract

HFpEF: a heterogeneous disease with multiple disease mechanisms

Heart failure (HF) with preserved ejection fraction (HFpEF) is a complex clinical syndrome that is characterized by both extra-cardiac and cardiac features.1–3 Prevalence is still rising4–8 and survival of patients with HFpEF is poor, with a 5-year survival rate after first hospitalization of 35–40%.9  ,  10 So far no treatment has been proven successful in reducing morbidity and mortality rates in HFpEF, potentially due to the large pathophysiological heterogeneity and diversity in HFpEF phenotypes.11 Recent studies have identified HFpEF as a systemic disease that is associated with, or may be triggered by a wide range of clinical risk factors and comorbidities such as aging, female sex, hypertension,12  ,  13 pulmonary congestion, metabolic syndrome, obesity,7  ,  12  ,  14–16 type 2 diabetes mellitus (T2DM), hyperlipidaemia, renal disease, atrial fibrillation (AF), and skeletal muscle weakness.11 These risk factors and comorbidities give rise to intertwining disease mechanisms in the pathophysiology of HFpEF.17  ,  18 Due to the wide range of comorbidities and clinical presentations, potential underlying aetiology of HFpEF is diverse; HFpEF can result from various structural abnormalities of the myocardium, or may result from abnormal loading conditions, e.g. as seen in hypertension, valvular diseases, volume overload, or rhythm disorders.19

Although HFpEF patients thus represent a heterogeneous group with a broad extent of extra-cardiac features, the cardiac phenotype has less interpatient variability and includes (concentric) left ventricular (LV) hypertrophy,20 LV diastolic dysfunction,21 cardiac stiffening, atrial dilatation, fibrosis,22 (systemic) inflammation, microvascular endothelial dysfunction,23  ,  24 and elevated natriuretic peptides.19  ,  25  ,  26

The definition of HFpEF as a clinical syndrome, based on typical symptoms and signs, presents challenges due to non-specificity of cardinal symptoms such as breathlessness and effort intolerance. Recently, two diagnostic HFpEF algorithms, the HFA-PEFF20 and H2FPEF27 scores, were developed to standardize and improve the accuracy of HFpEF diagnosis. Both of these scores (Figure 1) use a stepwise diagnostic approach to score and evaluate probability of HFpEF presence. The H2FPEF score uses functional echocardiographic data and places emphasis on the presence of comorbidities (e.g. hypertension, obesity) and the effect of age, while not including natriuretic peptide levels. The HFA-PEFF algorithm also assesses pretest probability based on clinical features (including age and comorbidities) and similarly includes a score but based on both functional and structural echocardiographic data, including morphological aspects of the left atrium and LV, as well as levels of natriuretic peptides, such as N-terminal pro brain natriuretic peptide (NT-proBNP).

Figure 1.

Figure 1

Diagnostic HFpEF scoring algorithms used to score HFpEF animal models. Both algorithms first include a pretest assessment to evaluate signs and symptoms and clinical features of HFpEF that include congestion, increased comorbidity burden and reduced exercise tolerance. The second step of the HFA-PEFF19 score assesses three domains that include functional aspects [echocardiographic diastolic function (E/e′ and GLS)], morphological aspects (left atrial enlargement, LV mass and wall thickness and concentric hypertrophy) as well as levels of circulating natriuretic peptides.2  ,  28 The H2FPEF27 score combines clinical and echocardiographic patient characteristics: obesity, hypertension, AF, pulmonary hypertension, age >60 years and diastolic function (E/e′). A higher score represents a higher likelihood of having HFpEF (HFA-PEFF ≥5 points; H2FPEF >6 points), while a lower score is used to rule out HFpEF. For patients with an intermediate score, both algorithms recommend additional testing to refine the diagnosis by exercise echocardiography or invasive measurements of cardiac filling pressures in a non-resting state.19  ,  27 AF, atrial fibrillation; GLS, global longitudinal strain; HF, heart failure; LV, left ventricle; PASP, pulmonary artery systolic pressure.

Both HFpEF scores have recently been validated in various patient cohorts29–33 and communities studies34 and it was concluded that both HFpEF scores categorized patients well, especially in those patients with intermediate and high scores. These scores, however, are not without controversy, with criticisms ranging from over-simplification of the diagnostic challenges to over-complicating the diagnostic process by requiring expensive tests or the scores largely disagree.35  ,  36 In addition, misclassification has been reported, especially in those patients with low HFpEF scores, potentially due to the fact that both scores use resting parameters in a phenotype in which physiological abnormalities augment during exercise.33  ,  37 Nevertheless, both scores have been shown to have prognostic utility in human patients,38  ,  39 suggesting that they capture key pathophysiologic components that determine outcomes in HFpEF.

Of note, the combined considerations of the phenotypic complexity of HFpEF, the interplay of cardiac and non-cardiac comorbidities, and the role that these comorbidities play in the pathophysiology of HFpEF have not been adequately taken into account in the evaluation of pre-clinical models of HFpEF. While the HFA-PEFF and H2FPEF algorithms have been developed to standardize and improve HFpEF diagnosis in patients, these scores may represent a novel approach to improve putative applicability of HFpEF mouse models. Therefore, this review aims to evaluate the translational aspects of currently available pre-clinical mouse models of HFpEF in the context of the HFA-PEFF and H2FPEF scores and proposes a novel approach to the assessment and development of future pre-clinical HFpEF models.

HFpEF in mice: where do we stand?

Over the last decades, development of HFpEF specific treatments has been disappointing. Standard, successful, HF with reduced ejection fraction (HFrEF) treatment options, such as angiotensin-converting enzyme inhibitors (ACEi), angiotensin receptor 1 blockers and mineralocorticoid receptor antagonists (MRA) did not convincingly reduce mortality and morbidity rates in HFpEF patients.40–42 Trials with other types of drugs, such as nitric oxide donors and cyclic guanosine monophosphate (cGMP) stimulating therapies failed to improve clinical status,43–47 or were neutral for the primary endpoint (angiotensin receptor–neprilysin inhibitor, PARAGON-HF trial48  ,  49). To date, no HFpEF specific treatment options exist and there is an unmet need to improve morbidity and mortality rate in these patients.

Drug development typically progresses in stages, from pre-clinical to clinical. Valuable HFpEF animal models presenting clinical HFpEF phenotypes are crucial for the successful design of new therapies. This has been neglected so far, which has led to the failure of many clinical studies. Sildenafil, for example, successfully reduced LV hypertrophy and cardiac remodelling in mice that suffered from angiotensin II (ANGII)-induced or transverse aortic constriction (TAC) induced HF.50  ,  51 Clinical studies of sildenafil in HFpEF patients, however, did not observe these beneficial effects on clinical or hemodynamic parameters.45 Studies with ACEi in myocardial infarction models (MI),52  ,  53 successfully reduced hypertrophy and fibrosis with a concomitant improvement of cardiac function. However, studies in patients with HFpEF have yielded inconsistent results.40 This was also the case for the MRA spironolactone: in pre-clinical studies in diet induced51  ,54 and myocardial infarction (MI)55  ,  56 models this drug improved systolic and diastolic cardiac function. A subsequent large randomized controlled trial on the other hand, remained neutral and did not meet its endpoint.41 The unsuccessful bench-to-bedside translation may, at least partly, be explained by the fact that pre-clinical animals models not fully recapitulate the clinical HFpEF phenotype and TAC or MI models cannot be considered as HFpEF model.

In this review we discuss and score several pre-clinical HFpEF models using the HFA-PEFF and H2FPEF scores. We found that several major discrepancies exist between pre-clinical HF models and clinical HFpEF. Pre-clinical HFpEF models do not always recognize the importance of signs and symptoms of HFpEF, or clinical HFpEF characteristics (graphical abstract). Several so-called HFpEF models would have obtained high scores according to the HFA-PEFF and H2FPEF risk scores (Figure 2) due to functional or morphological features, while signs of lung congestion or exercise impairment were absent and levels of natriuretic peptides low (Table 1). Thus, a model without pulmonary congestion may relate to hypertensive heart disease in humans rather than clinical HFpEF (for example db/db or ob/ob models). The currently developed HFA-PEFF and H2FPEF scores both emphasize typical symptoms and signs of HF, or clinical HFpEF characteristics as key for the diagnosis of HFpEF. Although the assessment of signs and symptoms or diagnostic HF criteria may be more challenging in animals than in humans, it is not impossible. Pulmonary congestion can be demonstrated by increased lung weight, and reduced exercise tolerance can be measured via voluntary or forced exercise testing. Reduced exercise tolerance is one of the hallmarks in human HFpEF and should ideally be part of phenotyping HFpEF animal models.

Figure 2.

Figure 2

HFA-PEFF and H2FPEF scores obtained by HF models. All HF models have been scored for cardiac and extra-cardiac domains of HFA-PEFF and H2FPEF scores. Based upon these scores, mouse HF models are differentiated into more or less likely to fulfil the criteria of the HFA-PEFF or H2FPEF score. If we solely record the scores, several of so-called HFpEF models would have obtained high scores due to functional or morphological features, while signs of lung congestion or exercise impairment were absent and levels of natriuretic peptides low. ANGII, angiotensin II; DOCA, deoxycorticosterone acetate; DOCP, desoxycorticosterone pivalate; db/db, leptin receptor-deficient model; HFpEF, heart failure with preserved ejection fraction; l-NAME, N(ω)-nitro-l-arginine methyl ester; ob/ob, leptin-deficient model.

Table 1.

Validation of HFpEF mouse models by HFA-PEFF and H2FPEF scores

Validation of HFpEF mouse models by HFA-PEFF and H2FPEF scores
Model Pretest assessment of signs and symptoms, clinical HFpEF features and biological factors (age and sex)
HFA-PEFF score
H2FPEF score
Preserved EF Sex Age (months) Lung congestion Impaired Exercise capacity Comorbidity burden
Functional aspects Morphological aspects
Increased natriuretic peptides Total points Obesity Hyper tension Atrial Fibrillation Pulmonary hyper tension Age Diastolic dys function Total points
Hyper tension Obesity T2DM Renal Dys function Diastolic dysfunction Left atrial enlarge ment Left ventricular mass Increased wall thickness Concentric hyper trophy
Low HFpEF likelihood
Aldosterone uninephrectomy mouse Yes M 3 Yes Yes Yes Yes Yes N/A Yes N/A Yes Yes Yes Yes 6 No Yes No N/A No Yes 2
High fat diet/ Western diet Yes M/F 3–16 Yes Yes Yes Yes Yes No Yes N/A Yes Yes Yes No 4 Yes No No N/A No Yes 4
Aged mice (24–30 months) Yes M 24–30 Yes Yes No No No No Yes N/A Yes Yes Yes Yes 6 No No No N/A Yes Yes 2
Angiotensin-II infusion models Yes M/F 3 Yes Yes Yes No No No Yes N/A Yes Yes Yes Yes 6 No Yes No yes No No 2
Accelerated senescence model (SAMP) Yes F 3–12 No Yes Yes No No N/A Yes Yes Yes Yes Yes Yes 6 No Yes No N/A Yes Yes 4
Leptin receptor- deficient model (db/db) Yes M/F 3 No Yes Yes Yes Yes No Yes Yes Yes Yes Yes No 4 Yes Yes No N/A No Yes 4
Leptin-deficient model (ob/ob) Yes M/F 3 No Yes No Yes Yes No Yes No Yes Yes Yes No 4 Yes Yes No Yes No Yes 3
(DOCA) salt-sensitive model Yes M 3 No No Yes No No No Yes N/A Yes Yes Yes Yes 6 No No No No No Yes 1
High fat diet and angiotensin II Yes M 3 No No Yes Yes Yes N/A Yes N/A Yes Yes Yes Yes 6 Yes Yes No N/A No Yes 3
High HFpEF likelihood
High fat diet and L-NAME Yes M/F 3 Yes Yes Yes Yes Yes N/A Yes N/A Yes Yes Yes Yes 6 Yes Yes No N/A No Yes 4
Aging, high fat diet and angiotensin II Yes F 22 Yes Yes Yes Yes Yes N/A Yes Yes Yes Yes Yes Yes 6 Yes Yes No N/A Yes Yes 5
Aging, high fat and DOCP Yes M/F 18 Yes Yes Yes Yes Yes N/A Yes N/A Yes Yes Yes Yes 6 Yes Yes No N/A Yes Yes 5

HF models are scored for signs and symptoms or clinical HFpEF features, included age, sex, as well as cardiac and extra-cardiac domains of HFA-PEFF and H2FPEF scores. Based upon these scores, mouse HF models were differentiated into more or less likely to fulfil the criteria of the human HFpEF situation, with higher scores representing pre-clinical HF models that most resembled clinical HFpEF. Models that presented full signs and symptoms and clinical HFpEF features are shown in the high HFpEF likelihood box.

db/db, leptin receptor-deficient model; DOCA, deoxycorticosterone acetate; DOCP, desoxycorticosterone pivalate; EF, Ejection fraction; l-NAME, N(ω)-nitro-l-arginine methyl ester; ob/ob, leptin-deficient model; T2DM, type 2 diabetes mellitus.

Importantly, the demonstration of LV diastolic dysfunction has been the cornerstone of validation of a HFpEF animal model; however, the presence of diastolic dysfunction alone is neither synonymous nor sufficient for a diagnosis of HFpEF. Indeed, diastolic dysfunction, as occurs with aging, can exist without the presence of symptomatic HF. Nonetheless, aging is a potent risk factor for HFpEF.7  ,  57  ,  58 Aging itself is associated with ventricular-vascular stiffening and fibrosis, key mechanisms in the pathogenesis of HFpEF.59  ,  60 The aging process also exacerbates chronic systemic inflammation, dysregulation of energy supply61–63 and increased cardiomyocyte stiffness and increased hypertrophy that may all result in HFpEF specific diastolic dysfunction and cardiac remodelling.64  ,  65 We realize that aging itself can have major practical limitations (>20 months to produce the phenotype); however, because it is such an important factor, we encourage researchers to include it.

Another major difference between animal and human HFpEF can be found in disease complexity and disease heterogeneity. In humans, HFpEF is considered a multifactorial and heterogeneous disease with a plethora of clinical manifestations.11 For many years, pre-clinical HFpEF models have relied upon a single perturbation. The development of several recent multifactorial models has shown that it is feasible to develop a HFpEF-like phenotype in mice by using multiple perturbations, and these models may represent a new era of multifactorial pre-clinical HFpEF models.

HFpEF in mice: fundamental checklist

We do not believe that ‘one-size-fits-all’ pre-clinical HFpEF model exists. Several animal models of HFpEF have been developed that only focused on a limited aspect of this multifactorial syndrome. This strategy has been proven unsuccessful and the recent development of combinatory models is very promising.66–68 Although recent multifactorial HFpEF models have been proven valuable, and may improve bench-to-bed translation, these models also focus on specific HFpEF phenotypes and do not recapitulate the entire heterogeneity of the clinical HFpEF syndrome. In addition, technical challenges remain in developing mouse models. AF, for example, has not been included in any of the pre-clinical HFpEF models so far.

We therefore suggest that all pre-clinical HFpEF studies should include a mouse model that fulfils (a majority of) the following requirements in order to perform a reliable and accurate pre-clinical HFpEF study. This has been schematically presented in Figure 3.

Figure 3.

Figure 3

HFpEF in mice: a novel approach to develop a multifactorial pre-clinical HFpEF mouse model. The following clinical HFpEF features are essential to develop a reliable and accurate pre-clinical HFpEF model: (1) pulmonary congestion and elevated natriuretic peptides; (2) a distinct cardiac phenotype with preserved systolic LV function with concentric hypertrophy, fibrosis, atrial enlargement and diastolic dysfunction; (3) extra-cardiac comorbidities such as hypertension, obesity, T2DM and renal dysfunction and skeletal muscle weakness; and (4) incorporate and evaluate the effect of sex and aging. LV, left ventricle. Parts of the figure were drawn by using pictures from Smart Servier Medical Art (http://smart.servier.com), licensed under a Creative Commons Attribution 3.0 Generic License (https://creativecommons.org/licenses/by/3.0/).

Pretest assessment of signs and symptoms and clinical HFpEF features

First of all, ejection fraction should be preserved. Assessment of symptoms such as shortness of breath, fatigue, oedema, tachycardia, and exercise impairment in animals may be less straightforward than in humans, but various parameters are available to provide a global impression if signs and symptoms and clinical HFpEF features are present:

  • Increased natriuretic peptide levels. Natriuretic peptide levels should be measured in plasma or LV tissue. Elevated natriuretic peptide levels play an important part in the HFA-PEFF score and also provide a global impression if HFpEF is likely to be present in animals.

  • Impaired exercise performance. Impaired exercise capacity caused by skeletal muscle weakness, fatigue, or cardiovascular to muscle mismatch should be measured by voluntary or forced exercise. This is a typical feature of HFpEF, and analysis of exercise capacity, including assessment of skeletal muscle function, will provide essential information regarding HFpEF severity.69–71

  • Lung congestion. Analysis of lung weight and pulmonary vasculature will be helpful to determine increased diastolic filling pressures and presence of diastolic dysfunction.

In case surrogate measurements of signs and symptoms and clinical HFpEF features (increased natriuretic peptides, preserved ejection fraction and increased comorbidity burden) are not present, the pre-clinical model does not meet the HFpEF criteria as suggested by the two scores and should therefore not be regarded as a pre-clinical HFpEF model.

A distinct cardiac phenotype with preserved systolic lv function with concentric hypertrophy and diastolic dysfunction

  • Assessment of systolic cardiac function. Systolic cardiac function should be assessed by transthoracic echocardiography and should include measurement of LV dimensions to assess concentric hypertrophy and LV systolic function. Post-mortem analysis (weighing and staining) of the total heart and LV should take place to assess amount of cardiac hypertrophy and fibrosis.

  • Assessment of diastolic function. Diastolic function should be determined by morphological criteria (atrial enlargement) or functional parameters. In mice, evaluation of diastolic function is complex and the E/A and E/e′ ratio is difficult to assess and highly variable.72 Global longitudinal strain (GLS) and reverse peak longitudinal strain rate (RPLSR) are easily obtained, highly reproducible, and have therefore to be integrated as indices of diastolic dysfunction in mice.73  ,  74 Post-mortem analysis (weighing) of atria should take place to evaluate atrial enlargement.

  • Assessment of cardiac hemodynamics. Although considered as gold standard for diagnosis of HFpEF, invasive hemodynamic measurements are performed to a limited scale in humans due to a lack of expertise, availability, risks, and costs. A distinct advantage in animal models is that this gold standard assessment can be done more easily and more frequently but requires experience to be reliable. Invasive hemodynamic measurements provide information on intracardiac volumes, filling pressures, contractile and relaxation forces and derivate measures such as tau, dP/dT of the LV. Although measurements of systolic pulmonary artery pressure and pulmonary capillary wedge pressure yield additional information about diastolic function and pulmonary hypertension, measuring right-sided invasive hemodynamics presents more of a challenge in pre-clinical models and may not be required if gold standard left-sided invasive hemodynamics are already evaluated.

Extra-cardiac comorbidities such as hypertension, obesity, type 2 diabetes mellitus, and renal dysfunction

Assessment of extra-cardiac features of HFpEF should take place in all pre-clinical HFpEF models. This assessment should include evaluation of several comorbidities that are closely related to the development of HFpEF.

  • Hypertension. Assessment of hypertension can be performed in several ways, including invasive hemodynamic measurements at sacrifice or by using tail-cuff measurements or continuous registrations throughout the study period.

  • Renal function. Plasma should be obtained to determine kidney function. Post-mortem analysis of kidneys should take place (weighing + staining).

  • Obesity. Mice should be repeatedly weighed during the experiment. Body mass composition should be determined throughout the experiment and prior to sacrifice.

  • T2DM. Fasting plasma glucose levels or glycated hemoglobin should be obtained throughout the experiment. Glucose tolerance can be evaluated by oral glucose tolerance test and insulin sensitivity can be tested by insulin tolerance test.

  • Skeletal muscle weakness. Post-mortem analysis of skeletal muscle should take place to evaluate reduced mass, and address impaired skeletal oxidative metabolism and abnormal skeletal muscle composition.

AF is a well-known comorbidity for HFpEF and represents an important part of the H2FPEF score (three points if AF is present). Unfortunately, induction of AF in mice is challenging and so far none of the experimental AF models resemble typical clinical HFpEF characteristics.75–77 We therefore excluded AF from this section.

Effect of sex and aging

Epidemiological evidence suggests that HFpEF is highly represented in older women.78 The effect of aging and sex should therefore be taken into account when developing a pre-clinical model.

  • Aging. The life span of a rodent is shorter than humans, and mice are already considered ‘old’ after 18 months and ‘very old’ when >24 months.79 Aging may represent an important contributing factor to the development of HFpEF and should therefore be considered when studying HFpEF.57  ,  80

  • Female sex. Sex-specific differences are known to exist in humans and mice 4  ,  12  ,  81–86 and for various interventions, young female mice have been shown to be less susceptible to develop a cardiac phenotype as compared to young males.87  ,  88 Hormonal differences or hormonal changes (such as menopause) are thought to play an important role in the increased cardiovascular risk profile of older females.21  ,  89 Interestingly, the development of LV hypertrophy may also occur in a sex-specific manner: females more often display concentric remodeling89 while males develop eccentric LV remodeling.90 Since the meaning of these differences are not fully understood yet we strongly advise to develop pre-clinical HFpEF models that take into account the effect sex may have. At the very least, investigators may consider including females rather than performing exclusively male experiments as is often the case.

Validation and translation of the H2FPEF and HFA-PEFF scores in animal models

For most experimental HFpEF models, mice are preferred small animals since they are easy to handle, quick to breed, allow genetic experiments, and are known to produce reliable and highly reproducible outcomes. Larger animal models of HFpEF, such as rat,91–108 dogs109  ,  110 and pigs,111–117 also exist (summarized in Supplementary material online, Table S1); nevertheless, ethical issues, difficulty in introducing high throughput genetic and molecular studies, cost, and duration of study limit large animal models. We included mice models that were widely used in HF research, and are presented as ‘HFpEF’ models, or were used to evaluate several HFpEF treatment options in the pre-clinical phase, often without translational success.

All models were scored for pre-clinical sign and symptoms or clinical HFpEF features (including age and sex), as well as cardiac and extra-cardiac domains of HFA-PEFF and H2FPEF scores (Table 1). Based upon these scores, mouse HF models have been differentiated into more or less likely to fulfil the criteria of the HFA-PEFF or H2FPEF score, schematically presented in Figure 2. In the Graphical abstract, we presented the models in less or high likelihood for HFpEF, including whether models with higher scores also present pre-clinical signs and symptoms or clinical HFpEF characteristics.

graphic file with name ehab389f4.jpg

An in-depth review of existing pre-clinical HFpEF mouse models with validation of their translational value using the HFA-PEFF and H2FPEF scores.

Angiotensin-II infusion models

Chronic stimulation of the ANGII type 1 receptor with ANGII infusion by osmotic mini-pumps is a well-known and reliable model to induce HF with cardiac hypertrophy and increased remodelling. Remodeling takes place with118–122 or without123 hypertension, depending on the dosage of ANGII. The ANGII effects seems to be strain specific: treatment with ANGII in Balb/c124 mice typically results in lung congestion and LV dilatation, whereas treatment with ANGII in C57BL6 mice results in lung congestion, as well as exercise intolerance, concentric remodelling with fibrosis, and increased levels of natriuretic peptides.120  ,  122 ANGII treated mice develop diastolic dysfunction that includes worsening LV isovolumetric relaxation time, increased LV end-diastolic pressure and increased E/e′.50  ,  120–124 In mice, exogenous ANGII administration does not interfere with kidney function,123 but may induce skeletal muscle alterations.125 ANGII models, and especially the ANGII induced hypertension models, resemble cardiac features of human HFpEF to a large extent. Effects of age and obesity, however, are neglected in this model resulting in the following scores:

  • Pretest assessment of signs and symptoms and clinical HFpEF features: lung congestion, hypertension and reduced exercise capacity.

  • Total HFA-PEFF score: 6 (diastolic dysfunction, LV hypertrophy, increased natriuretic peptide levels);

  • Total H2FPEF score: 2 (hypertension and increased filling pressures).

Leptin receptor-deficient model (db/db)

Genetically modified db/db mice have a point mutation in the gene encoding for the leptin receptor that leads to malfunctioning of this receptor.126 These mice are typically used for cardiometabolic research, especially for studies in the field of non-insulin dependent T2DM. Young db/db mice develop obesity, hyperglycaemia and severe dyslipidemia without hypertension.127 The onset of symptoms in mice is severe and early in life, and therefore not directly translatable to the human situation in which progression of obesity and T2DM is a slower and chronic process. db/db mice have been from different strains, different ages and different sex128 and results from studies performed in these mice are therefore not always comparable.

In general, db/db mice develop diastolic dysfunction including atrial enlargement, concentric hypertrophy, and fibrosis at older ages.129  ,  130 LV ejection fraction remains preserved, with decreased GLS rates after 16 weeks. Hypertension may be present, with131  ,  132 or without133  ,  134 ANGII infusion. Development of cardiac hypertrophy may already be present at early age (8–9 weeks133  ,  135) or develops at a later point in time (up to 16 weeks136  ,  137). Most db/db mice develop concentric hypertrophy, although eccentric hypertrophy has been observed as well.138 Signs of congestion are usually not present in these mice, and natriuretic peptide levels are not elevated.139  ,  140

  • Pretest assessment of signs and symptoms and clinical HFpEF features: increased comorbidity burden (obesity and diabetes) and reduced exercise capacity.

  • Total HFA-PEFF score: 4 (diastolic dysfunction, LV hypertrophy);

  • Total H2FPEF score: 4 (obesity, hypertension, diastolic dysfunction).

Leptin-deficient model (ob/ob)

The ob/ob is a leptin-deficient mouse that spontaneously develops obesity (within 4 weeks) and T2DM secondary to hyperglycaemia and hyperinsulinemia.141  ,  142 The mice develop concentric hypertrophy with diastolic dysfunction possible due to lipid accumulation.143 The ejection fraction is preserved without congestion or exercise impairment and natriuretic peptide levels are unchanged or reduced.144–146 The observed maladaptive cardiac alterations appear to be related to the loss of leptin mediated signaling and are reversed by recombinant leptin treatment.129  ,  147 However, obese HFpEF patients with leptin deficiency are rarely observed, so the ob/ob mice do not mimic the human HFpEF phenotype.148

  • Pretest of signs and symptoms and clinical HFpEF features: increased comorbidity burden (obesity and diabetes) and reduced exercise capacity.

  • Total HFA-PEFF score: 4 (diastolic dysfunction, LV hypertrophy);

  • Total H2FPEF score: 3 (obesity, diastolic dysfunction).

High fat diet/western diet

Obesity is an important comorbidity in patients with HFpEF and has been suggested to play an import role in (development of) HFpEF.149  ,  150 In pre-clinical models, unhealthy food consumption is mimicked by a high fat diet (HFD) (>60% fat of daily caloric intake) or by a Western diet (36% fat and 36% sucrose of daily intake). Both of these diets are able to induce an unfavourable cardiometabolic phenotype with obesity and glucose intolerance in young male and female animals138  ,  151–155 albeit in a strain-specific manner.156–159 In older animals, the HFD appears to result in more profound cardiometabolic changes including hyperglycaemia and insulin resistance and more profound inflammation.160  ,  161 There may also be sex-specific effect as female mice tend to gain more weight than age-matched male littermates.81  ,  138  ,  154–164

Besides an unfavourable cardiometabolic phenotype, these models result in concentric LV hypertrophy with preserved ejection fraction, and mild to moderate diastolic dysfunction.154  ,  163  ,  164 Furthermore, pulmonary hypertension has been described as well as increased levels of cardiac fibrosis.164  ,  165 Pulmonary congestion is absent and levels of natriuretic peptides are usually not elevated.166 Renal dysfunction may occur after long term diet (>20 weeks) in young mice or at earlier point in time in aged mice.161  ,  167  ,  168 Mice fed on an HFD or Western diet typically show reduced exercise capacity, most likely related to their obese state as skeletal muscle weakness is not observed in these mice.71  ,  157  ,  169

  • Pretest assessment of signs and symptoms and clinical HFpEF features: increased comorbidity burden (obesity and pre-diabetes) and reduced exercise capacity.

  • Total HFA-PEFF score: 4 (diastolic dysfunction, LV hypertrophy).

  • Total H2FPEF score: 4 (obesity, pulmonary hypertension, diastolic dysfunction)

Aged mice (24–30 months)

Similar to humans, natural aging in mice (with or without dietary intervention) is a main driver of development of a maladaptive cardiac HFpEF phenotype.170 At an age of 24–30 months, mice recapitulate many hallmarks of human HFpEF pathophysiology, including diastolic dysfunction, concentric hypertrophy with fibrosis and reduced exercise capacity.171  ,  172 This mice furthermore have lung congestion and increased natriuretic peptide levels. Hypertension or T2DM, however, have not been described.

  • Pretest assessment of signs and symptoms and clinical HFpEF features: lung congestion, increased natriuretic peptide levels, reduced exercise capacity, but no comorbidity burden.

  • Total HFA-PEFF score: 6 (diastolic dysfunction, LV hypertrophy);

  • Total H2FPEF score: 2 (age, diastolic dysfunction).

Accelerated senescence model (SAMP)

Senescence accelerated prone (SAMP) mice belong to a strain of mice that were generated by selective inbreeding of AKR/J mice.173 These mice show accelerated senescence and age-related pathological phenotypes, similar to aging disorders seen in humans. In addition, they start displaying features of aging at younger age (10 months) than normal mice (8 months).174 Deleterious mutations in the DNA repair genes are to be involved in their genetic vulnerability for enhanced aging, and specific gene analyses show involvement of oxidative and stress response pathways.175 SAMP mice develop age-related diastolic dysfunction with atrial enlargement and adverse cardiac remodelling including LV hypertrophy and fibrosis.102  ,  159  ,  176 Levels of natriuretic peptides are elevated in these mice.159 When fed a Western diet, SAMP mice also develop hypertension and lung congestion, albeit without obesity or T2DM.159 It has not been described if female or male SAMPs age differently.

  • Pretest assessment of signs and symptoms and clinical HFpEF features: increased natriuretic peptide levels, lung congestion and reduced exercise capacity.

  • Total HFA-PEFF score: 6 (diastolic dysfunction, LV hypertrophy, elevated natriuretic peptides).

  • Total H2FPEF score: 4 (hypertension, effect of aging, increased filling pressures).

Progress in pre-clinical HF models: development of multifactorial models

The abovementioned models are mostly unifactorial disease models that use one perturbation to induce HF. More recently, progress has been made in the development of pre-clinical HFpEF models and this has led to multifactorial models that use two or more perturbations to mimic the human HFpEF phenotype. In the following section, we will again use the HFA-PEFF and H2FPEF score to describe and validate a traditional multifactorial model as well as newer multifactorial HFpEF models.

Deoxycorticosterone acetate salt-sensitive model

The deoxycorticosterone acetate salt-sensitive model was already developed in 1969 to study hypertension in young mice and rats.177 This model relies upon a combination of multiple perturbations including administration of deoxycorticosterone acetate, increased salt intake (addition of 1% NaCl to drinking water) and uninephrectomy. This typically results in cardiac hypertrophy with fibrosis, increased levels of natriuretic peptides, while blood pressure remains unchanged or only mildly increased.178  ,  179 LV function remains preserved while moderate diastolic dysfunction can be observed.180 Nevertheless, these mice do not display lung congestion.181 Again, the effect of age and sex has not been described in this model.

  • Pretest assessment of signs and symptoms and clinical HFpEF features: increased natriuretic peptide levels, and reduced exercise capacity.

  • Total HFA-PEFF score: 6 (diastolic dysfunction, LV hypertrophy, increased levels of natriuretic peptides);

  • Total H2FPEF score: 1 (diastolic dysfunction).

Aldosterone uninephrectomy mouse

Impaired renal function is frequently observed in patients with HFpEF. Renal dysfunction may be attributed to fluid overload, blood pressure elevation, and thus congestion.182 In C57BL6 or FB/N background, the combination of uninephrectomy and aldosterone infusion results in the development of hypertension, lung congestion, and reduced exercise capacity without obesity or T2DM.183  ,  184 Preserved LV ejection fraction is observed with concentric remodelling, mild-to-moderate diastolic dysfunction, and increased levels of natriuretic peptides.185–188 The effect of female sex or aging is unknown and obesity or T2DM is not observed.

  • Pretest assessment of signs and symptoms and clinical HFpEF features: lung congestion, increased natriuretic peptide levels and reduced exercise capacity.

  • Total HFA-PEFF score: 6 (diastolic dysfunction, LV hypertrophy, increased levels of natriuretic peptides);

  • Total H2FPEF score: 2 (hypertension, increased filling pressures).

Combinatory model of high fat diet and L-NAME

Schiattarella et al.  169 were the first to present a two-hit pre-clinical mouse model that resembles human HFpEF. In short, C57BL/6N wild-type mice were subjected to a combination of HFD and hypertension that was induced by L-NAME (constitutive nitric oxide synthase inhibitor). They observed that mice that were subjected to both stress factors developed a typical HFpEF phenotype, including lung congestion and reduced exercise tolerance and increased natriuretic peptides. On the contrary, mice that were only exposed to one stressor did not develop this phenotype.169 More recently, sex-dependent effects have also been shown: young female mice were more resilient for development of HFpEF, as the combination of high-fat and L-NAME resulted in a more attenuated cardiac phenotype as compared to young male mice.189 The effect of aging was not studied.

  • Pretest assessment of signs and symptoms and clinical HFpEF features: increased natriuretic peptides, lung congestion, reduced exercise capacity, and increased comorbidity burden (hypertension, obesity and pre-diabetes).

  • Total HFA-PEFF score: 6 points (increased natriuretic peptides, diastolic dysfunction, concentric LV hypertrophy).

  • Total H2FPEF score: 4 (obesity, hypertension, increased filling pressures).

Combinatory model of high fat diet and ANGII infusion

The combination of HFD and ANGII infusion induces hypertension, obesity and T2DM in young male mice.151  ,  190  ,  191 This intervention also results in preserved LV function with diastolic dysfunction, concentric hypertrophy with fibrosis and increased natriuretic peptides. However, signs and symptoms or clinical features of HFpEF, if any, appear to be very mild since lung congestion in young animals is absent and effect on exercise capacity is unknown.151  ,  190–192

  • Pretest assessment of signs and symptoms and clinical HFpEF features: increased natriuretic peptide levels, increased comorbidity burden (hypertension and pre-diabetes);

  • Total HFA-PEFF score: 6 (diastolic dysfunction, LV hypertrophy, elevated levels of natriuretic peptides);

  • Total H2FPEF score: 4 (obesity, hypertension, increased filling pressures).

Combinatory model of aging, high fat diet, and ANGII infusion

We have recently developed a multifactorial mouse model that combines aging (18–22 months) with HFD and ANGII infusion.193 In these older female C57BL6/J mice, a HFpEF-like phenotype is present including concentric LV hypertrophy and LV fibrosis, diastolic dysfunction, lung congestion, increased natriuretic peptide levels, and elevated blood pressures. The effect of sex has not been studied yet.

  • Pretest assessment of signs and symptoms and clinical HFpEF features: lung congestion, increased natriuretic peptide levels, reduced exercise capacity, and increased comorbidity burden (hypertension, obesity and pre-diabetes);

  • Total HFA-PEFF score: 6 (diastolic dysfunction, concentric LV hypertrophy, elevated natriuretic peptide levels);

  • Total H2FPEF score: 5 (obesity, hypertension, elderly, increased filling pressures).

Combinatory model of aging, high fat diet and desoxycorticosterone pivalate

A very recent study by Deng et al.194 used a combinatory model of 16 months of ageing, long-term HFD (13 months) and 3 months of desoxycorticosterone pivalate challenge in mice to induce a HFpEF-like phenotype. Their model resulted in many typical HFpEF features, including lung congestion, hypertension and impaired exercise tolerance. They also showed diastolic dysfunction, LV hypertrophy, fibrosis and increased levels of natriuretic peptides. Both sexes were included but not further studied.

  • Pretest assessment of signs and symptoms and clinical HFpEF features: lung congestion, increased natriuretic peptide levels, reduced exercise capacity and increased comorbidity burden (hypertension, obesity and pre-diabetes);

  • Total HFA-PEFF score: 6 (diastolic dysfunction, LV hypertrophy, elevated natriuretic peptide levels);

  • Total H2FPEF score: 5 (obesity, hypertension, elderly, increased filling pressures).

Conclusion

HFpEF remains a major public health problem worldwide with still increasing prevalence and incidence. So far, HFpEF treatment mostly focuses on symptom reduction since HFpEF-specific drugs do not exist. Despite numerous efforts to develop HFpEF-specific drugs, bench-to-bedside translation has not been successful, and this may, at least partly, be due to the lack of pre-clinical HFpEF models that adequately recapitulate the complexities of the human condition.

HFpEF is a multifactorial disease in which comorbidities contribute to the pathophysiology of the clinical syndrome. While this complicates the development of preclinical models, progress in the field will be aided by consensus on key elements that a HFpEF animal model should manifest. The recent development of two clinical HFpEF scores has led to a novel clinical standard for defining the key clinical features of HFpEF. This state-of-the-art review is the first to apply clinical scores to HFpEF mouse models to improve putative applicability and translational value of pre-clinical HFpEF research. It proposes a novel approach to follow when performing a pre-clinical HFpEF study to optimize bench-to-bed translation and provide a checklist for small HFpEF animal models. Although this checklist may not capture all human HFpEF variables, it will help to provide better and more relevant small animal HFpEF models with better putative application and translational value. So far, most of the pre-clinical models do not fully meet these criteria (presented in Graphical abstract). Of course, pathophysiology of the mouse heart cannot be translated to humans 1 on 1, and translation of pre-clinical findings to human conditions should always be done cautiously. Of note, clinical studies should be challenged as well to account for diverse HFpEF physiology to optimize bench-to-bed translation.

This review furthermore describes some multifactorial models that resemble human HFpEF to a large extent, and suggests that these small animal models remain attractive models for future HFpEF research. Based on this review, we advocate that future HFpEF pre-clinical studies that test potential new therapeutic agents should consider use of multiple HFpEF animal models so that their effects can be tested on multiple HFpEF phenotypes. Following this approach we believe that pre-clinical HFpEF models will be able to help fill major gaps in HFpEF pathophysiology and will eventually facilitate development of novel HFpEF therapeutics.

Supplementary material

Supplementary material is available at European Heart Journal online.

Funding

C.W. and C.S.L are supported by the University of Groningen (Rosalind Franklin fellowship). G.G.S. is supported by the German Center for Cardiovascular Research (DZHK) Junior Research Group Excellence Grant. C.S.L. is supported by a Clinician Scientist Award from the National Medical Research Council of Singapore. This work was supported by grants from the Netherlands Heart Foundation (CVON SHE-PREDICTS-HF, grant 2017-21; CVON RED-CVD, grant 2017-11; CVON PREDICT2, grant 2018-30; and CVON DOUBLE DOSE, grant 2020B005), by a grant from the leDucq Foundation (Cure PhosphoLambaN induced Cardiomyopathy (Cure-PLaN), and by a grant from the European Research Council (ERC CoG 818715, SECRETE-HF).

Conflict of interest: C.W. reports grants from NovoNordisk and AstraZeneca, outside the submitted work. In addition, G.G.S. has a patent PCT/US17/37019 pending. C.S.P.L. reports grants from Boston Scientific, Bayer, Roche Diagnostics, AstraZeneca, Medtronic, and Vifor Pharma, personal fees from Abbott Diagnostics, Amgen, Applied Therapeutics, AstraZeneca, Bayer, Biofourmis, Boehringer Ingelheim, Boston Scientific, Corvia Medical, Cytokinetics, Darma Inc., Us2.ai, JanaCare, Janssen Research & Development LLC, Medtronic, Menarini Group, Merck, MyoKardia, Novartis, Novo Nordisk, Radcliffe Group Ltd, Roche Diagnostics, Sanofi, Stealth BioTherapeutics, The Corpus, Vifor Pharma, and WebMD Global LLC, outside the submitted work. In addition, C.S.P.L. has a patent PCT/SG2016/050217 pending and a patent 16/216,929 issued. R.A.d.B. reports grants from Abbott, grants from AstraZeneca, grants from Boehringer Ingelheim, grants from Cardior Pharmaceuticals Gmbh, grants from Ionis Pharmaceuticals, Inc., grants from Novo Nordisk, grants from Roche, personal fees from Abbott, personal fees from AstraZeneca, personal fees from Bayer, personal fees from Novartis, and personal fees from Roche, outside the submitted work. L.M.G.M. reports grants from Mandema Stipendium (UMCG), outside the submitted work.

Supplementary Material

ehab389_Supplementary_Data

Contributor Information

Coenraad Withaar, Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands.

Carolyn S P Lam, Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands; National Heart Centre, Singapore and Duke-National University of Singapore.

Gabriele G Schiattarella, Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Department of Cardiology, Center for Cardiovascular Research (CCR), Charité - Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany; Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy; Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA.

Rudolf A de Boer, Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands.

Laura M G Meems, Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands.

References

  • 1. Borlaug  BA.  Evaluation and management of heart failure with preserved ejection fraction. Nat Rev Cardiol  2020;17:559–573. [DOI] [PubMed] [Google Scholar]
  • 2. Ponikowski  P, Voors  AA, Anker  SD, Bueno  H, Cleland  JGF, Coats  AJS, Falk  V, González-Juanatey  JR, Harjola  VP, Jankowska  EA, Jessup  M, Linde  C, Nihoyannopoulos  P, Parissis  JT, Pieske  B, Riley  JP, Rosano  GMC, Ruilope  LM, Ruschitzka  F, Rutten  FH, van der Meer  P; ESC Scientific Document Group. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contibution of the Heart Failure Association (HFA) of the ESC. Eur Heart J  2016;37:2129–2200. [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. Benjamin  EJ, Virani  SS, Callaway  CW, Chamberlain  AM, Chang  AR, Cheng  S, Chiuve  SE, Cushman  M, Delling  FN, Deo  R, De  FS, Ferguson  JF, Fornage  M, Gillespie  C, Isasi  CR, Jiménez  MC, Jordan  LC, Judd  SE, Lackland  D, Lichtman  JH, Lisabeth  L, Liu  S, Longenecker  CT, Lutsey  PL, MacKey  JS, Matchar  DB, Matsushita  K, Mussolino  ME, Nasir  K, O’Flaherty  M, Palaniappan  LP, Pandey  A, Pandey  DK, Reeves  MJ, Ritchey  MD, Rodriguez  CJ, Roth  GA, Rosamond  WD, Sampson  UKA, Satou  GM, Shah  SH, Spartano  NL, Tirschwell  DL, Tsao  CW, Voeks  JH, Willey  JZ, Wilkins  JT, Wu  JH, Alger  HM, Wong  SS, Muntner  P.  Heart disease and stroke statistics - 2018 update: a report from the American Heart Association  . Circulation  2018;137:E67–E492. [DOI] [PubMed] [Google Scholar]
  • 5. Gori  M, Lam  CSP, Gupta  DK, Santos  ABS, Cheng  S, Shah  AM, Claggett  B, Zile  MR, Kraigher-Krainer  E, Pieske  B, Voors  AA, Packer  M, Bransford  T, Lefkowitz  M, McMurray  JJV, Solomon  SD; for the PARAMOUNT Investigators. Sex-specific cardiovascular structure and function in heart failure with preserved ejection fraction. Eur J Heart Fail  2014;16:535–542. [DOI] [PubMed] [Google Scholar]
  • 6. Boonman-de Winter  LJM, Rutten  FH, Cramer  MJM, Landman  MJ, Liem  AH, Rutten  GEHM, Hoes  AW.  High prevalence of previously unknown heart failure and left ventricular dysfunction in patients with type 2 diabetes. Diabetologia  2012;55:2154–2162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Ho  JE, Enserro  D, Brouwers  FP, Kizer  JR, Shah  SJ, Psaty  BM, Bartz  TM, Santhanakrishnan  R, Lee  DS, Chan  C, Liu  K, Blaha  MJ, Hillege  HL, van der Harst  P, van Gilst  WH, Kop  WJ, Gansevoort  RT, Vasan  RS, Gardin  JM, Levy  D, Gottdiener  JS, de Boer  RA, Larson  MG.  Predicting heart failure with preserved and reduced ejection fraction. Circ Heart Fail  2016;9:e003116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Packer  M, Kitzman  DW.  Obesity-related heart failure with a preserved ejection fraction. JACC Heart Fail  2018;6:633–639. [DOI] [PubMed] [Google Scholar]
  • 9. Oktay  AA, Rich  JD, Shah  SJ.  The emerging epidemic of heart failure with preserved ejection fraction. Curr Heart Fail Rep  2013;10:401–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Tribouilloy  C, Rusinaru  D, Mahjoub  H, Soulière  V, Lévy  F, Peltier  M, Slama  M, Massy  Z.  Prognosis of heart failure with preserved ejection fraction: a 5 year prospective population-based study. Eur Heart J  2008;29:339–347. [DOI] [PubMed] [Google Scholar]
  • 11. Shah  SJ, Kitzman  DW, Borlaug  BA, van Heerebeek  L, Zile  MR, Kass  DA, Paulus  WJ.  Phenotype-specific treatment of heart failure with preserved ejection fraction. Circulation  2016;134:73–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Goyal  P, Paul  T, Almarzooq  ZI, Peterson  JC, Krishnan  U, Swaminathan  RV, Feldman  DN, Wells  MT, Karas  MG, Sobol  I, Maurer  MS, Horn  EM, Kim  LK.  Sex- and race-related differences in characteristics and outcomes of hospitalizations for heart failure with preserved ejection fraction. J Am Heart Assoc  2017;6:e003330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Eaton  CB, Pettinger  M, Rossouw  J, Martin  LW, Foraker  R, Quddus  A, Liu  S, Wampler  NS, Hank Wu  W-C, Manson  JE, Margolis  K, Johnson  KC, Allison  M, Corbie-Smith  G, Rosamond  W, Breathett  K, Klein  L.  Risk factors for incident hospitalized heart failure with preserved versus reduced ejection fraction in a multiracial cohort of postmenopausal women. Circ Heart Fail  2016;9:e002883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. 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]
  • 15. De Boer  RA, Nayor  M, DeFilippi  CR, Enserro  D, Bhambhani  V, Kizer  JR, Blaha  MJ, Brouwers  FP, Cushman  M, Lima  JAC, Bahrami  H, Van Der Harst  P, Wang  TJ, Gansevoort  RT, Fox  CS, Gaggin  HK, Kop  WJ, Liu  K, Vasan  RS, Psaty  BM, Lee  DS, Hillege  HL, Bartz  TM, Benjamin  EJ, Chan  C, Allison  M, Gardin  JM, Januzzi  JL, Shah  SJ, Levy  D, Herrington  DM, Larson  MG, Van Gilst  WH, Gottdiener  JS, Bertoni  AG, Ho  JE.  Association of cardiovascular biomarkers with incident heart failure with preserved and reduced ejection fraction. JAMA Cardiol  2018;3:215–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Kanter  R, Caballero  B.  Global gender disparities in obesity: a review. Adv Nutr  2012;3:491–498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Maréchaux  S, Six-Carpentier  MM, Bouabdallaoui  N, Montaigne  D, Bauchart  JJ, Mouquet  F, Auffray  JL, Le Tourneau  T, Asseman  P, Lejemtel  TH, Ennezat  PV.  Prognostic importance of comorbidities in heart failure with preserved left ventricular ejection fraction. Heart Vessels  2011;26:313–320. [DOI] [PubMed] [Google Scholar]
  • 18. Mentz  RJ, Kelly  JP, Von Lueder  TG, Voors  AA, Lam  CSP, Cowie  MR, Kjeldsen  K, Jankowska  EA, Atar  D, Butler  J, Fiuzat  M, Zannad  F, Pitt  B, ’Connor  OCM  Noncardiac comorbidities in heart failure with reduced versus preserved ejection fraction. J Am Coll Cardiol  2014;64:2281–2293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Pieske  B, Tschöpe  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]
  • 20. Tadic  M, Cuspidi  C, Plein  S, Belyavskiy  E, Heinzel  F, Galderisi  M.  Sex and heart failure with preserved ejection fraction: from pathophysiology to clinical studies. J Clin Med  2019;8:792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. De Simone  G, Devereux  RB, Chinali  M, Roman  MJ, Barac  A, Panza  JA, Lee  ET, Howard  BV.  Sex differences in obesity-related changes in left ventricular morphology: the Strong Heart Study. J Hypertens  2011;29:1431–1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Hahn  VS, Yanek  LR, Vaishnav  J, Ying  W, Vaidya  D, Lee  YZJ, Riley  SJ, Subramanya  V, Brown  EE, Hopkins  CD, Ononogbu  S, Perzel Mandell  K, Halushka  MK, Steenbergen  C, Rosenberg  AZ, Tedford  RJ, Judge  DP, Shah  SJ, Russell  SD, Kass  DA, Sharma  K.  Endomyocardial biopsy characterization of heart failure with preserved ejection fraction and prevalence of cardiac amyloidosis. JACC Heart Fail  2020;8:712–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Paulus  WJ, Tschöpe  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]
  • 24. Shah  SJ, Lam  CSP, Svedlund  S, Saraste  A, Hage  C, Tan  RS, Beussink-Nelson  L, Faxén  UL, 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]
  • 25. Chirinos  JA, Orlenko  A, Zhao  L, Basso  MD, Cvijic  ME, Li  Z, Spires  TE, Yarde  M, Wang  Z, Seiffert  DA, Prenner  S, Zamani  P, Bhattacharya  P, Kumar  A, Margulies  KB, Car  BD, Gordon  DA, Moore  JH, Cappola  TP.  Multiple plasma biomarkers for risk stratification in patients with heart failure and preserved ejection fraction. J Am Coll Cardiol  2020;75:1281–1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Pfeffer  MA, Shah  AM, Borlaug  BA.  Heart failure with preserved ejection fraction in perspective. Circ Res  2019;124:1598–1617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Reddy  YNV, Carter  RE, Obokata  M, Redfield  MM, Borlaug  BA.  A simple, evidence-based approach to help guide diagnosis of heart failure with preserved ejection fraction. Circulation  2018;138:861–870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Komajda  M, Lam  CSP.  Heart failure with preserved ejection fraction: a clinical dilemma. Eur Heart J  2014;35:1022–1032. [DOI] [PubMed] [Google Scholar]
  • 29. Faxen  UL, Venkateshvaran  A, Shah  SJ, Lam  CSP, Svedlund  S, Saraste  A, Beussink-Nelson  L, Lagerstrom Fermer  M, Gan  L-M, Hage  C, Lund  LH.  Generalizability of HFA-PEFF and H2FPEF Diagnostic Algorithms and associations with heart failure indices and proteomic biomarkers: insights from PROMIS-HFpEF. J Card Fail  2021;S1071-9164(21)00077-4 doi: 10.1016/j.cardfail.2021.02.005 [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 30. Barandiarán Aizpurua  A, Sanders-van  WS, Brunner-La Rocca  H, Henkens  M, Heymans  S, Beussink‐Nelson  L, Shah  SJ, Empel  VPM.  Validation of the HFA‐PEFF score for the diagnosis of heart failure with preserved ejection fraction. Eur J Heart Fail  2020;22:413–421. [DOI] [PubMed] [Google Scholar]
  • 31. Iwakura  K, Onishi  T, Okada  M, Inoue  K, Koyama  Y, Okamura  A, Yamada  T, Yasumura  Y, Tamaki  S, Hayashi  T, Yano  M, Fujii  K, Hikoso  S, Sakata  Y.  Validation of the HFA-PEFF- and H2FPEF score in Japanese patients with heart failure with preserved ejection fraction [abstract]. Eur Heart J  2020;41:998. [Google Scholar]
  • 32. Myhre  PL, Vaduganathan  M, Claggett  BL, Lam  CSP, Desai  AS, Anand  IS, Sweitzer  NK, Fang  JC, O’Meara  E, Shah  SJ, Shah  AM, Lewis  EF, Rouleau  J, Pitt  B, Solomon  SD.  Application of the H2FPEF score to a global clinical trial of patients with heart failure with preserved ejection fraction: the TOPCAT trial. Eur J Heart Fail  2019;21:1288–1291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Churchill  TW, Li  SX, Curreri  L, Zern  EK, Lau  ES, Liu  EE, Farrell  R, Shoenike  MW, Sbarbaro  J, Malhotra  R, Nayor  M, Tschöpe  C, de Boer  RA, Lewis  GD, Ho  JE.  Evaluation of 2 existing diagnostic scores for heart failure with preserved ejection fraction against a comprehensively phenotyped cohort. Circulation  2021;143:289–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Selvaraj  S, Myhre  PL, Vaduganathan  M, Claggett  BL, Matsushita  K, Kitzman  DW, Borlaug  BA, Shah  AM, Solomon  SD.  Application of diagnostic algorithms for heart failure with preserved ejection fraction to the community. JACC Heart Fail  2020;8:640–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Sanders-van Wijk  S, Barandiarán Aizpurua  A, Brunner-La Rocca  H-P, Henkens  MTHM, Weerts  J, Knackstedt  C, Uszko-Lencer  N, Heymans  S, van Empel  V.  The HFA-PEFF and H2FPEF scores largely disagree in classifying patients with suspected heart failure with preserved ejection fraction. Eur J Heart Fail  2020; doi: 10.1002/ejhf.2019 [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Coats  AJS.  Validating the HFA-PEFF score – or how to define a disease?  Eur J Heart Fail  2020;22:428–431. [DOI] [PubMed] [Google Scholar]
  • 37. Ho  JE, Redfield  MM, Lewis  GD, Paulus  WJ, Lam  CSP.  Deliberating the diagnostic dilemma of heart failure with preserved ejection fraction. Circulation  2020;142:1770–1780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Parcha  V, Kalra  R, Malla  G, Patel  N, Sanders-van Wijk  S, Shah  SJ, Arora  G, Arora  P.  Diagnostic and prognostic implications of heart failure with preserved ejection fraction scoring systems. J Card Fail  2020;26:S38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Sueta  D, Yamamoto  E, Nishihara  T, Tokitsu  T, Fujisue  K, Oike  F, Takae  M, Usuku  H, Takashio  S, Arima  Y, Suzuki  S, Nakamura  T, Ito  M, Kanazawa  H, Sakamoto  K, Kaikita  K, Tsujita  K.  H2FPEF score as a prognostic value in HFpEF patients. Am J Hypertens  2019;32:1082–1090. [DOI] [PubMed] [Google Scholar]
  • 40. Khan  MS, Fonarow  GC, Khan  H, Greene  SJ, Anker  SD, Gheorghiade  M, Butler  J.  Renin–angiotensin blockade in heart failure with preserved ejection fraction: a systematic review and meta-analysis. ESC Heart Fail  2017;4:402–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. 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; TOPCAT Investigators. Spironolactone for heart failure with preserved ejection fraction. N Engl J Med  2014;370:1383–1392. [DOI] [PubMed] [Google Scholar]
  • 42. Beldhuis  IE, Myhre  PL, Claggett  B, Damman  K, Fang  JC, Lewis  EF, O’Meara  E, Pitt  B, Shah  SJ, Voors  AA, Pfeffer  MA, Solomon  SD, Desai  AS.  Efficacy and safety of spironolactone in patients with HFpEF and chronic kidney disease. JACC Heart Fail  2019;7:25–32. [DOI] [PubMed] [Google Scholar]
  • 43. Pieske  B, Maggioni  AP, Lam  CSP, Pieske-Kraigher  E, Filippatos  G, Butler  J, Ponikowski  P, Shah  SJ, Solomon  SD, Scalise  A-V, Mueller  K, Roessig  L, Gheorghiade  M.  Vericiguat in patients with worsening chronic heart failure and preserved ejection fraction: results of the SOluble guanylate Cyclase stimulatoR in heArT failurE patientS with PRESERVED EF (SOCRATES-PRESERVED) study. Eur Heart J  2017;38:1119–1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Redfield  MM, Chen  HH, Borlaug  BA, Semigran  MJ, Lee  KL, Lewis  G, LeWinter  MM, Rouleau  JL, Bull  DA, Mann  DL, Deswal  A, Stevenson  LW, Givertz  EO, Ofili  MM, O’Connor  CM, Felker  GM, Goldsmith  SR, Bart  BA, McNulty  SE, Ibarra  JC, Lin  G, Oh  JK, Patel  MR, Kim  RJ, Tracy  RP, Velazquez  EJ, Anstrom  KJ, Hernandez  AF, Mascette  AM, Braunwald  E.  Effect of phosphodiesterase-5 inhibition on exercise capacity and clinical status in heart failure with preserved ejection fraction: a randomized clinical trial. JAMA  2013;309:1268–1277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Hoendermis  ES, Liu  LCY, Hummel  YM, Van Der Meer  P, De Boer  RA, Berger  RMF, Van Veldhuisen  DJ, Voors  AA.  Effects of sildenafil on invasive haemodynamics and exercise capacity in heart failure patients with preserved ejection fraction and pulmonary hypertension: a randomized controlled trial. Eur Heart J  2015;36:2565–2573. [DOI] [PubMed] [Google Scholar]
  • 46. Borlaug  BA, Anstrom  KJ, Lewis  GD, Shah  SJ, Levine  JA, Koepp  GA, Givertz  MM, Felker  GM, Lewinter  MM, Mann  DL, Margulies  KB, Smith  AL, Tang  WHW, Whellan  DJ, Chen  HH, Davila-Roman  VG, McNulty  S, Desvigne-Nickens  P, Hernandez  AF, Braunwald  E, Redfield  MM.  Effect of inorganic nitrite vs placebo on exercise capacity among patients with heart failure with preserved ejection fraction: the INDIE-HFpEF Randomized Clinical Trial. JAMA  2018;320:1764–1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Komajda  M, Isnard  R, Cohen-Solal  A, Metra  M, Pieske  B, Ponikowski  P, Voors  AA, Dominjon  F, Henon-Goburdhun  C, Pannaux  M, Böhm  M; on behalf of the prEserveD left ventricular ejectIon fraction chronic heart Failure with ivabradine studY (EDIFY) Investigators. Effect of ivabradine in patients with heart failure with preserved ejection fraction: the EDIFY randomized placebo-controlled trial. Eur J Heart Fail  2017;19:1495–1503. [DOI] [PubMed] [Google Scholar]
  • 48. Wachter  R, Shah  SJ, Cowie  MR, Szecsödy  P, Shi  V, Ibram  G, Zhao  Z, Gong  J, Klebs  S, Pieske  B.  Angiotensin receptor neprilysin inhibition versus individualized RAAS blockade: design and rationale of the PARALLAX trial. ESC Heart Fail  2020;7:856–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. 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, Düngen  H-D, 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]
  • 50. Westermann  D, Becher  PM, Lindner  D, Savvatis  K, Xia  Y, Fröhlich  M, Hoffmann  S, Schultheiss  H-PP, Tschöpe  C.  Selective PDE5A inhibition with sildenafil rescues left ventricular dysfunction, inflammatory immune response and cardiac remodeling in angiotensin II-induced heart failure in vivo. Basic Res Cardiol  2012;107:308. [DOI] [PubMed] [Google Scholar]
  • 51. Takimoto  E, Champion  HC, Li  M, Belardi  D, Ren  S, Rodriguez  ER, Bedja  D, Gabrielson  KL, Wang  Y, Kass  DA.  Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hypertrophy. Nat Med  2005;11:214–222. [DOI] [PubMed] [Google Scholar]
  • 52. Pfeffer  MA, Pfeffer  JM, Steinberg  C, Finn  P.  Survival after an experimental myocardial infarction: beneficial effects of long-term therapy with captopril. Circulation  1985;72:406–412. [DOI] [PubMed] [Google Scholar]
  • 53. Pfeffer  JM, Pfeffer  MA, Braunwald  E.  Influence of chronic captopril therapy on the infarcted left ventricle of the rat. Circ Res  1985;57:84–95. [DOI] [PubMed] [Google Scholar]
  • 54.Bostick B, Habibi J, DeMarco VG, Jia G, Domeier T L, Lambert MD, Aroor AR, Nistala R, Bender SB, Garro M, Hayden MR, Ma L, Manrique C, Sowers JR. Mineralocorticoid receptor blockade prevents western diet-induced diastolic dysfunction in female mice. Am J Physiol – Hear Circ Physiol 2015;308:H1126–H1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wang D, Liu YH, Yang XP, Rhaleb NE, Xu J, Peterson E, Rudolph AE, Carretero OA. Role of a selective aldosterone blocker in mice with chronic heart failure. J Card Fail 2004;10:67–73. [DOI] [PubMed] [Google Scholar]
  • 56.Fraccarollo D, Galuppo P, Sieweke JT, Napp LC, Grobbecker P, Bauersachs J. Efficacy of mineralocorticoid receptor antagonism in the acute myocardial infarction phase: eplerenone versus spironolactone. ESC Hear Fail 2015;2:150–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Kenchaiah  S, Vasan  RS.  Heart failure in women – insights from the Framingham Heart Study. Cardiovasc Drugs Ther  2015;29:377–390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. 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]
  • 59. Lam  CSP, Roger  VL, Rodeheffer  RJ, Bursi  F, Borlaug  BA, Ommen  SR, Kass  DA, Redfield  MM.  Cardiac structure and ventricular-vascular function in persons with heart failure and preserved ejection fraction from Olmsted County, Minnesota. Circulation  2007;115:1982–1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Redfield  MM, Jacobsen  SJ, Borlaug  BA, Rodeheffer  RJ, Kass  DA.  Age- and gender-related ventricular-vascular stiffening: a community-based study. Circulation  2005;112:2254–2262. [DOI] [PubMed] [Google Scholar]
  • 61. Terman  A, Kurz  T, Navratil  M, Arriaga  EA, Brunk  UT.  Mitochondrial turnover and aging of long-lived postmitotic cells: the mitochondrial-lysosomal axis theory of aging. Antioxidants Redox Signal  2010;12:503–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Dai  D-F, Chen  T, Wanagat  J, Laflamme  M, Marcinek  DJ, Emond  MJ, Ngo  CP, Prolla  TA, Rabinovitch  PS.  Age-dependent cardiomyopathy in mitochondrial mutator mice is attenuated by overexpression of catalase targeted to mitochondria. Aging Cell  2010;9:536–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Schriner  SE, Linford  NJ, Martin  GM, Treuting  P, Ogburn  CE, Emond  M, Coskun  PE, Ladiges  W, Wolf  N, Van Remmen  H, Wallace  DC, Rabinovitch  PS.  Extension of murine life span by overexpression of catalase targeted to mitochondria. Science  2005;308:1909–1911. [DOI] [PubMed] [Google Scholar]
  • 64. Dai  DF, Chen  T, Szeto  H, Nieves-Cintrón  M, Kutyavin  V, Santana  LF, Rabinovitch  PS.  Mitochondrial targeted antioxidant peptide ameliorates hypertensive cardiomyopathy. J Am Coll Cardiol  2011;58:73–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Dai  DF, Santana  LF, Vermulst  M, Tomazela  DM, Emond  MJ, MacCoss  MJ, Gollahon  K, Martin  GM, Loeb  LA, Ladiges  WC, Rabinovitch  PS.  Overexpression of catalase targeted to mitochondria attenuates murine cardiac aging. Circulation  2009;119:2789–2797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Conceição  G, Heinonen  I, Lourenço  AP, Duncker  DJ, Falcão-Pires  I.  Animal models of heart failure with preserved ejection fraction. Neth Heart J  2016;24:275–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Valero-Muñoz  M, Backman  W, Sam  F.  Murine models of heart failure with preserved ejection fraction: a “Fishing Expedition”. JACC Basic Transl Sci  2017;2:770–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Roh  J, Houstis  N, Rosenzweig  A.  Why Don’t We Have Proven Treatments for HFpEF?  Circ Res  2017;120:1243–1245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Brunjes  DL, Kennel  PJ, Christian Schulze  P.  Exercise capacity, physical activity, and morbidity. Heart Fail Rev  2017;22:133–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Baltgalvis  KA, White  K, Li  W, Claypool  MD, Lang  W, Alcantara  R, Singh  BK, Friera  AM, McLaughlin  J, Hansen  D, McCaughey  K, Nguyen  H, Smith  IJ, Godinez  G, Shaw  SJ, Goff  D, Singh  R, Markovtsov  V, Sun  T-Q, Jenkins  Y, Uy  G, Li  Y, Pan  A, Gururaja  T, Lau  D, Park  G, Hitoshi  Y, Payan  DG, Kinsella  TM.  Exercise performance and peripheral vascular insufficiency improve with AMPK activation in high-fat diet-fed mice. Am J Physiol Heart Circ Physiol  2014;306:H1128–H1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. 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:H1364–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Schnelle  M, Catibog  N, Zhang  M, Nabeebaccus  AA, Anderson  G, Richards  DA, Sawyer  G, Zhang  X, Toischer  K, Hasenfuss  G, Monaghan  MJ, Shah  AM.  Echocardiographic evaluation of diastolic function in mouse models of heart disease. J Mol Cell Cardiol  2018;114:20–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Ferferieva  V, Van den Bergh  A, Claus  P, Jasaityte  R, La Gerche  A, Rademakers  F, Herijgers  P, D'hooge  J.  Assessment of strain and strain rate by two-dimensional speckle tracking in mice: comparison with tissue Doppler echocardiography and conductance catheter measurements. Eur Heart J Cardiovasc Imaging  2013;14:765–773. [DOI] [PubMed] [Google Scholar]
  • 74. Zacchigna  S, Paldino  A, Falcão-Pires  I, Daskalopoulos  EP, Dal Ferro  M, Vodret  S, Lesizza  P, Cannatà  A, Miranda-Silva  D, Lourenço  AP, Pinamonti  B, Sinagra  G, Weinberger  F, Eschenhagen  T, Carrier  L, Kehat  I, Tocchetti  CG, Russo  M, Ghigo  A, Cimino  J, Hirsch  E, Dawson  D, Ciccarelli  M, Oliveti  M, Linke  WA, Cuijpers  I, Heymans  S, Hamdani  N, de Boer  M, Duncker  DJ, Kuster  D, van der Velden  J, Beauloye  C, Bertrand  L, Mayr  M, Giacca  M, Leuschner  F, Backs  J, Thum  T.  Towards standardization of echocardiography for the evaluation of left ventricular function in adult rodents: a position paper of the ESC Working Group on Myocardial Function. Cardiovasc Res  2021;117:43–59. [DOI] [PubMed] [Google Scholar]
  • 75. Nishida  K, Michael  G, Dobrev  D, Nattel  S.  Animal models for atrial fibrillation: clinical insights and scientific opportunities. Europace  2010;12:160–172. [DOI] [PubMed] [Google Scholar]
  • 76. Zhang  F, Hartnett  S, Sample  A, Schnack  S, Li  Y.  High fat diet induced alterations of atrial electrical activities in mice. Am J Cardiovasc Dis  2016;6:1–9. [PMC free article] [PubMed] [Google Scholar]
  • 77. Nakada  Y, Canseco  DC, Thet  S, Abdisalaam  S, Asaithamby  A, Santos  CX, Shah  AM, Zhang  H, Faber  JE, Kinter  MT, Szweda  LI, Xing  C, Hu  Z, Deberardinis  RJ, Schiattarella  G, Hill  JA, Oz  O, Lu  Z, Zhang  CC, Kimura  W, Sadek  HA.  Hypoxia induces heart regeneration in adult mice. Nature  2017;541:222–227. [DOI] [PubMed] [Google Scholar]
  • 78. Scantlebury  DC, Borlaug  BA.  Why are women more likely than men to develop heart failure with preserved ejection fraction?  Curr Opin Cardiol  2011;26:562–568. [DOI] [PubMed] [Google Scholar]
  • 79. Flurkey  K, Currer  JM, Harrison  DE.  Mpuse models in aging research. In: Fox  J, Barthold  S, Davisson  M, Newcomer  C, Quimby  F, Smith  A, eds. The Mouse in Biomedical Research. 2nd ed. New York, NY: Academic Press; 2007. p637–672. [Google Scholar]
  • 80. Loffredo  FS, Nikolova  AP, Pancoast  JR, Lee  RT.  Heart failure with preserved ejection fraction: molecular pathways of the aging myocardium. Circ Res  2014;115:97–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Salinero  AE, Anderson  BM, Zuloaga  KL.  Sex differences in the metabolic effects of diet-induced obesity vary by age of onset. Int J Obes  2018;42:1088–1091. [DOI] [PubMed] [Google Scholar]
  • 82. Mitchell  SJ, Matute  JM, Scheibye-Knudsen  M, Fang  E, Aon  M, González-Reyes  JA, Cortassa  S, Kaushik  S, Patel  B, Wahl  D, Ali  A, Calvo-Rubio  M, María  I, Guiterrez  V, Ward  TM, Palacios  HH, Cai  H, David  W, Hine  C, Broeskamp  F, Habering  L, Sinclair  DA, Cohen  P, Egan  JM, Mitchell  JR, Baur  JA, Allison  DB, Anson  RM, Villalba  JM, Cuervo  AM, Pearson  KJ, Ingram  DK, Bernier  M.  Effects of sex, strain, and energy intake on hallmarks of aging in mice. Cell Metab  2017;23:1093–1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Piro  M, Della Bona  R, Abbate  A, Biasucci  LM, Crea  F.  Sex-related differences in myocardial remodeling. J Am Coll Cardiol  2010;55:1057–1065. [DOI] [PubMed] [Google Scholar]
  • 84. Krumholz  HM, Larson  M, Levy  D.  Sex differences in cardiac adaptation to isolated systolic hypertension. Am J Cardiol  1993;72:310–313. [DOI] [PubMed] [Google Scholar]
  • 85. Douglas  PS, Katz  SE, Weinberg  EO, Chen  MH, Bishop  SP, Lorell  BH.  Hypertrophic remodeling: gender differences in the early response to left ventricular pressure overload. J Am Coll Cardiol  1998;32:1118–1125. [DOI] [PubMed] [Google Scholar]
  • 86. Harada  E, Mizuno  Y, Kugimiya  F, Shono  M, Maeda  H, Yano  N, Yasue  H.  Sex differences in heart failure with preserved ejection fraction reflected by B-type natriuretic peptide level. Am J Med Sci  2018;356:335–343. [DOI] [PubMed] [Google Scholar]
  • 87. Shioura  KM, Geenen  DL, Goldspink  PH.  Sex-related changes in cardiac function following myocardial infarction in mice. Am J Physiol Integr Comp Physiol  2008;295:R528–R534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Pettersson  US, Waldén  TB, Carlsson  P-O, Jansson  L, Phillipson  M.  Female mice are protected against high-fat diet induced metabolic syndrome and increase the regulatory T cell population in adipose tissue. PLoS One  2012;7:e46057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Kajstura  J, Gurusamy  N, Ogórek  B, Goichberg  P, Clavo-Rondon  C, Hosoda  T, D’Amario  D, Bardelli  S, Beltrami  AP, Cesselli  D, Bussani  R, Del Monte  F, Quaini  F, Rota  M, Beltrami  CA, Buchholz  BA, Leri  A, Anversa  P.  Myocyte turnover in the aging human heart. Circ Res  2010;107:1374–1386. [DOI] [PubMed] [Google Scholar]
  • 90. Kuch  B, Muscholl  M, Luchner  A, Döring  A, Riegger  GAJ, Schunkert  H, Hense  HW.  Gender specific differences in left ventricular adaptation to obesity and hypertension. J Hum Hypertens  1998;12:685–691. [DOI] [PubMed] [Google Scholar]
  • 91. Nguyen  ITN, Brandt  MM, van de Wouw  J, van Drie  RWA, Wesseling  M, Cramer  MJ, de Jager  SCA, Merkus  D, Duncker  DJ, Cheng  C, Joles  JA, Verhaar  MC.  Both male and female obese ZSF1 rats develop cardiac dysfunction in obesity-induced heart failure with preserved ejection fraction. PLoS One  2020;15:e0232399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Lekawanvijit  S, Kompa  AR, Manabe  M, Wang  BH, Langham  RG, Nishijima  F, Kelly  DJ, Krum  H.  Chronic kidney disease-induced cardiac fibrosis is ameliorated by reducing circulating levels of a non-dialysable uremic toxin, indoxyl sulfate. PLoS One  2012;7:e41281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Suzuki  H, Schaefer  L, Ling  H, Schaefer  RM, Dämmrich  J, Teschner  M, Heidland  A.  Prevention of cardiac hypertrophy in experimental chronic renal failure by long-term ACE inhibitor administration: Potential role of lysosomal proteinases. Am J Nephrol  1995;15:129–136. [DOI] [PubMed] [Google Scholar]
  • 94. Kennedy  DJ, Vetteth  S, Periyasamy  SM, Kanj  M, Fedorova  L, Khouri  S, Kahaleh  MB, Xie  Z, Malhotra  D, Kolodkin  NI, Lakatta  EG, Fedorova  OV, Bagrov  AY, Shapiro  JI.  Central role for the cardiotonic steroid marinobufagenin in the pathogenesis of experimental uremic cardiomyopathy. Hypertension  2006;47:488–495. [DOI] [PubMed] [Google Scholar]
  • 95. Bongartz  LG, Braam  B, Verhaar  MC, Cramer  MJ, Goldschmeding  R, Gaillard  CA, Doevendans  PA, Joles  JA.  Transient nitric oxide reduction induces permanent cardiac systolic dysfunction and worsens kidney damage in rats with chronic kidney disease. Am J Physiol Integr Comp Physiol  2010;298:R815–R823. [DOI] [PubMed] [Google Scholar]
  • 96. Hamdani  N, Franssen  C, Lourenço  A, Falca O-Pires  I, Fontoura  D, Leite  S, Plettig  L, Lopez  B, Ottenheijm  CA, Becher  PM, Gonzalez  A, Tscho Pe  C, Diez  J, Linke  WA, Leite-Moreira  AF, Paulus  WJ.  Myocardial titin hypophosphorylation importantly contributes to heart failure with preserved ejection fraction in a rat metabolic risk model. Circ Heart Fail  2013;6:1239–1249. [DOI] [PubMed] [Google Scholar]
  • 97. Van Dijk  CGM, Oosterhuis  NR, Xu  YJ, Brandt  M, Paulus  WJ, Van Heerebeek  L, Duncker  DJ, Verhaar  MC, Fontoura  D, Lourenço  AP, Leite-Moreira  AF, Falcão-Pires  I, Joles  JA, Cheng  C.  Distinct endothelial cell responses in the heart and kidney microvasculature characterize the progression of heart failure with preserved ejection fraction in the obese ZSF1 rat with cardiorenal metabolic syndrome. Circ Heart Fail  2016;9:e002760. [DOI] [PubMed] [Google Scholar]
  • 98. Grobe  JL, Mecca  AP, Mao  H, Katovich  MJ.  Chronic angiotensin-(1–7) prevents cardiac fibrosis in DOCA-salt model of hypertension. Am J Physiol Circ Physiol  2006;290:H2417–H2423. [DOI] [PubMed] [Google Scholar]
  • 99. Schauer  A, Adams  V, Augstein  A, Jannasch  A, Draskowski  R, Kirchhoff  V, Goto  K, Mittag  J, Galli  R, Männel  A, Barthel  P, Linke  A, Winzer  EB.  Sacubitril/valsartan improves diastolic function but not skeletal muscle function in a rat model of HFpEF. Int J Mol Sci  2021;22:3570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Bode  D, Rolim  NPL, Guthof  T, Hegemann  N, Wakula  P, Primessnig  U, Berre  AMO, Adams  V, Wisløff  U, Pieske  BM, Heinzel  FR, Hohendanner  F.  Effects of different exercise modalities on cardiac dysfunction in heart failure with preserved ejection fraction. ESC Heart Fail  2021;8:1806–1818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Bode  D, Semmler  L, Wakula  P, Hegemann  N, Primessnig  U, Beindorff  N, Powell  D, Dahmen  R, Ruetten  H, Oeing  C, Alogna  A, Messroghli  D, Pieske  BM, Heinzel  FR, Hohendanner  F.  Dual SGLT-1 and SGLT-2 inhibition improves left atrial dysfunction in HFpEF. Cardiovasc Diabetol  2021;20:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Penumatsa  KC, Falcão-Pires  I, Leite  S, Leite-Moreira  A, Bhedi  CD, Nasirova  S, Ma  J, Sutliff  RL, Fanburg  BL.  Increased transglutaminase 2 expression and activity in rodent models of obesity/metabolic syndrome and aging. Front Physiol  2020;11:560019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Hohendanner  F, Bode  D, Primessnig  U, Guthof  T, Doerr  R, Jeuthe  S, Reimers  S, Zhang  K, Bach  D, Wakula  P, Pieske  BM, Heinzel  FR.  Cellular mechanisms of metabolic syndrome-related atrial decompensation in a rat model of HFpEF. J Mol Cell Cardiol  2018;115:10–19. [DOI] [PubMed] [Google Scholar]
  • 104. Brenner  DA, Apstein  CS, Saupe  KW.  Exercise training attenuates age-associated diastolic dysfunction in rats. Circulation  2001;104:221–226. [DOI] [PubMed] [Google Scholar]
  • 105. Elkholey  K, Morris  L, Niewiadomska  M, Houser  J, Ramirez  M, Tang  M, Humphrey  MB, Stavrakis  S.  Sex differences in the incidence and mode of death in rats with heart failure with preserved ejection fraction. Exp Physiol  2021;106:673–682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Bustamante  M, Garate-Carrillo  A, R. Ito  B, Garcia  R, Carson  N, Ceballos  G, Ramirez-Sanchez  I, Omens  J, Villarreal  F.  Unmasking of oestrogen-dependent changes in left ventricular structure and function in aged female rats: a potential model for pre-heart failure with preserved ejection fraction. J Physiol  2019;597:1805–1817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Curl  CL, Danes  VR, Bell  JR, Raaijmakers  AJA, Ip  WTK, Chandramouli  C, Harding  TW, Porrello  ER, Erickson  JR, Charchar  FJ, Kompa  AR, Edgley  AJ, Crossman  DJ, Soeller  C, Mellor  KM, Kalman  JM, Harrap  SB, Delbridge  LMD.  Cardiomyocyte functional etiology in heart failure with preserved ejection fraction is distinctive—a new preclinical model. J Am Heart Assoc  2018;7:e007451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Goto  K, Schauer  A, Augstein  A, Methawasin  M, Granzier  H, Halle  M, Craenenbroeck  EMV, Rolim  N, Gielen  S, Pieske  B, Winzer  EB, Linke  A, Adams  V.  Muscular changes in animal models of heart failure with preserved ejection fraction: what comes closest to the patient?  ESC Heart Fail  2021;8:139–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Munagala  VK, Hart  CYT, Burnett  JC, Meyer  DM, Redfield  MM.  Ventricular structure and function in aged dogs with renal hypertension: a model of experimental diastolic heart failure. Circulation  2005;111:1128–1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Zakeri  R, Moulay  G, Chai  Q, Ogut  O, Hussain  S, Takahama  H, Lu  T, Wang  X-L, Linke  WA, Lee  H-C, Redfield  MM.  Left atrial remodeling and atrioventricular coupling in a canine model of early heart failure with preserved ejection fraction. Circ Heart Fail  2016;9:1128–1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Schwarzl  M, Hamdani  N, Seiler  S, Alogna  A, Manninger  M, Reilly  S, Zirngast  B, Kirsch  A, Steendijk  P, Verderber  J, Zweiker  D, Eller  P, Höfler  G, Schauer  S, Eller  K, Maechler  H, Pieske  BM, Linke  WA, Casadei  B, Post  H.  A porcine model of hypertensive cardiomyopathy: implications for heart failure with preserved ejection fraction. Am J Physiol Heart Circ Physiol  2015;309:H1407–H1418. [DOI] [PubMed] [Google Scholar]
  • 112. Sharp  TE, Scarborough  AL, Li  Z, Polhemus  DJ, Hidalgo  HA, Schumacher  JD, Matsuura  TR, Jenkins  JS, Kelly  DP, Goodchild  TT, Lefer  DJ.  Novel Göttingen Miniswine model of heart failure with preserved ejection fraction integrating multiple comorbidities. JACC Basic Transl Sci  2021;6:154–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Zhang  N, Feng  B, Ma  X, Sun  K, Xu  G, Zhou  Y.  Dapagliflozin improves left ventricular remodeling and aorta sympathetic tone in a pig model of heart failure with preserved ejection fraction. Cardiovasc Diabetol  2019;18:107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Olver  TD, Edwards  JC, Jurrissen  TJ, Veteto  AB, Jones  JL, Gao  C, Rau  C, Warren  CM, Klutho  PJ, Alex  L, Ferreira-Nichols  SC, Ivey  JR, Thorne  PK, McDonald  KS, Krenz  M, Baines  CP, Solaro  RJ, Wang  Y, Ford  DA, Domeier  TL, Padilla  J, Rector  RS, Emter  CA.  Western diet-fed, aortic-banded Ossabaw Swine: a preclinical model of cardio-metabolic heart failure. JACC Basic Transl Sci  2019;4:404–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Charles  CJ, Lee  P, Li  RR, Yeung  T, Ibraham Mazlan  SM, Tay  ZW, Abdurrachim  D, Teo  XQ, Wang  WH, de Kleijn  DPV, Cozzone  PJ, Lam  CSP, Richards  AM.  A porcine model of heart failure with preserved ejection fraction: magnetic resonance imaging and metabolic energetics. ESC Heart Fail  2020;7:92–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Gyöngyösi  M, Pavo  N, Lukovic  D, Zlabinger  K, Spannbauer  A, Traxler  D, Goliasch  G, Mandic  L, Bergler-Klein  J, Gugerell  A, Jakab  A, Szankai  Z, Toth  L, Garamvölgyi  R, Maurer  G, Jaisser  F, Zannad  F, Thum  T, Bátkai  S, Winkler  J.  Porcine model of progressive cardiac hypertrophy and fibrosis with secondary postcapillary pulmonary hypertension. J Transl Med  2017;15:202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Sorop  O, Heinonen  I, van Kranenburg  M, van de Wouw  J, de Beer  VJ, Nguyen  ITN, Octavia  Y, van Duin  RWB, Stam  K, van Geuns  R-J, Wielopolski  PA, Krestin  GP, van den Meiracker  AH, Verjans  R, van Bilsen  M, Danser  AHJ, Paulus  WJ, Cheng  C, Linke  WA, Joles  JA, Verhaar  MC, van der Velden  J, Merkus  D, Duncker  DJ.  Multiple common comorbidities produce left ventricular diastolic dysfunction associated with coronary microvascular dysfunction, oxidative stress, and myocardial stiffening. Cardiovasc Res  2018;114:954–964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Shen  Y, Cheng  F, Sharma  M, Merkulova  Y, Raithatha  SA, Parkinson  LG, Zhao  H, Westendorf  K, Bohunek  L, Bozin  T, Hsu  I, Ang  LS, Williams  SJ, Bleackley  RC, Eriksson  JE, Seidman  MA, McManus  BM, Granville  DJ.  Granzyme B deficiency protects against angiotensin II-induced cardiac fibrosis. Am J Pathol  2016;186:87–100. [DOI] [PubMed] [Google Scholar]
  • 119. Ichihara  S, Senbonmatsu  T, Price  E, Ichiki  T, Gaffney  FA, Inagami  T.  Angiotensin II type 2 receptor is essential for left ventricular hypertrophy and cardiac fibrosis in chronic angiotensin II-induced hypertension. Circulation  2001;104:346–351. [DOI] [PubMed] [Google Scholar]
  • 120. Glenn  DJ, Cardema  MC, Ni  W, Zhang  Y, Yeghiazarians  Y, Grapov  D, Fiehn  O, Gardner  DG.  Cardiac steatosis potentiates angiotensin II effects in the heart. Am J Physiol Heart Circ Physiol  2015;308:H339–H350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Murdoch  CE, Chaubey  S, Zeng  L, Yu  B, Ivetic  A, Walker  SJ, Vanhoutte  D, Heymans  S, Grieve  DJ, Cave  AC, Brewer  AC, Zhang  M, Shah  AM.  Endothelial NADPH oxidase-2 promotes interstitial cardiac fibrosis and diastolic dysfunction through proinflammatory effects and endothelial-mesenchymal transition. J Am Coll Cardiol  2014;63:2734–2741. [DOI] [PubMed] [Google Scholar]
  • 122. Becher  PM, Lindner  D, Miteva  K, Savvatis  K, Zietsch  C, Schmack  B, Van Linthout  S, Westermann  D, Schultheiss  HP, Tschöpe  C.  Role of heart rate reduction in the prevention of experimental heart failure: comparison between if-channel blockade and β-receptor blockade. Hypertension  2012;59:949–957. [DOI] [PubMed] [Google Scholar]
  • 123. Regan  JA, Mauro  AG, Carbone  S, Marchetti  C, Gill  R, Mezzaroma  E, Raleigh  JV, Salloum  FN, Van Tassell  BW, Abbate  A, Toldo  S, Mauro  AG, Carbone  S, Marchetti  C, Gill  R, Mezzaroma  E, Valle Raleigh  J, Salloum  FN, Van Tassell  BW, Abbate  A, Toldo  S.  A mouse model of heart failure with preserved ejection fraction due to chronic infusion of a low subpressor dose of angiotensin II. Am J Physiol Heart Circ Physiol  2015;309:H771–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Peng  H, Yang  X-P, Carretero  OA, Nakagawa  P, D’Ambrosio  M, Leung  P, Xu  J, Peterson  EL, González  GE, Harding  P, Rhaleb  N-E.  Angiotensin II-induced dilated cardiomyopathy in Balb/c but not C57BL/6J mice. Exp Physiol  2011;96:756–764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Kadoguchi  T, Kinugawa  S, Takada  S, Fukushima  A, Furihata  T, Homma  T, Masaki  Y, Mizushima  W, Nishikawa  M, Takahashi  M, Yokota  T, Matsushima  S, Okita  K, Tsutsui  H.  Angiotensin II can directly induce mitochondrial dysfunction, decrease oxidative fibre number and induce atrophy in mouse hindlimb skeletal muscle. Exp Physiol  2015;100:312–322. [DOI] [PubMed] [Google Scholar]
  • 126. Chen  H, Charlat  O, Tartaglia  LA, Woolf  EA, Weng  X, Ellis  SJ, Lakey  ND, Culpepper  J, More  KJ, Breitbart  RE, Duyk  GM, Tepper  RI, Morgenstern  JP.  Evidence that the diabetes gene encodes the leptin receptor: identification of a mutation in the leptin receptor gene in db/db mice. Cell  1996;84:491–495. [DOI] [PubMed] [Google Scholar]
  • 127. Reil  J-C, Hohl  M, Reil  G-H, Granzier  HL, Kratz  MT, Kazakov  A, Fries  P, Müller  A, Lenski  M, Custodis  F, Gräber  S, Fröhlig  G, Steendijk  P, Neuberger  H-R, Böhm  M.  Heart rate reduction by If-inhibition improves vascular stiffness and left ventricular systolic and diastolic function in a mouse model of heart failure with preserved ejection fraction. Eur Heart J  2013;34:2839–2849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Alex  L, Russo  I, Holoborodko  V, Frangogiannis  NG.  Characterization of a mouse model of obesity-related fibrotic cardiomyopathy that recapitulates features of human heart failure with preserved ejection fraction. Am J Physiol Heart Circ Physiol  2018;315:H934–H949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Barouch  LA, Berkowitz  DE, Harrison  RW, O’Donnell  CP, Hare  JM.  Disruption of leptin signaling contributes to cardiac hypertrophy independently of body weight in mice. Circulation  2003;108:754–759. [DOI] [PubMed] [Google Scholar]
  • 130. Van den Bergh  A, Flameng  W, Herijgers  P.  Type II diabetic mice exhibit contractile dysfunction but maintain cardiac output by favourable loading conditions. Eur J Heart Fail  2006;8:777–783. [DOI] [PubMed] [Google Scholar]
  • 131. Arow  M, Waldman  M, Yadin  D, Nudelman  V, Shainberg  A, Abraham  NG, Freimark  D, Kornowski  R, Aravot  D, Hochhauser  E, Arad  M.  Sodium-glucose cotransporter 2 inhibitor Dapagliflozin attenuates diabetic cardiomyopathy. Cardiovasc Diabetol  2020;19:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Van Bilsen  M, Daniels  A, Brouwers  O, Janssen  BJAA, Derks  WJAA, Brouns  AE, Munts  C, Schalkwijk  CG, Van Der Vusse  GJ, Van Nieuwenhoven  FA.  Hypertension is a conditional factor for the development of cardiac hypertrophy in type 2 diabetic mice. PLoS One  2014;9:e85078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Sukumaran  V, Tsuchimochi  H, Tatsumi  E, Shirai  M, Pearson  JT.  Azilsartan ameliorates diabetic cardiomyopathy in young db/db mice through the modulation of ACE-2/ANG 1–7/Mas receptor cascade. Biochem Pharmacol  2017;144:90–99. [DOI] [PubMed] [Google Scholar]
  • 134. Habibi  J, Aroor  AR, Sowers  JR, Jia  G, Hayden  MR, Garro  M, Barron  B, Mayoux  E, Rector  RS, Whaley-Connell  A, DeMarco  VG.  Sodium glucose transporter 2 (SGLT2) inhibition with empagliflozin improves cardiac diastolic function in a female rodent model of diabetes. Cardiovasc Diabetol  2017;16:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Yue  P, Arai  T, Terashima  M, Sheikh  AY, Cao  F, Charo  D, Hoyt  G, Robbins  RC, Ashley  EA, Wu  J, Yang  PC, Tsao  PS.  Magnetic resonance imaging of progressive cardiomyopathic changes in the db/db mouse. Am J Physiol Heart Circ Physiol  2007;292:H2106–H2118. [DOI] [PubMed] [Google Scholar]
  • 136. Regitz-Zagrosek  V, Brokat  S, Tschope  C.  Role of gender in heart failure with normal left ventricular ejection fraction. Prog Cardiovasc Dis  2007;49:241–251. [DOI] [PubMed] [Google Scholar]
  • 137. Hutchinson  KR, Lord  CK, West  TA, Stewart  JA.  Jr.,  Cardiac fibroblast-dependent extracellular matrix accumulation is associated with diastolic stiffness in type 2 diabetes. PLoS One  2013;8:e72080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Bostick  B, Habibi  J, Ma  L, Aroor  A, Rehmer  N, Hayden  MR, Sowers  JR.  Dipeptidyl peptidase inhibition prevents diastolic dysfunction and reduces myocardial fibrosis in a mouse model of Western diet induced obesity. Metabolism  2014;63:1000–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Gutkowska  J, Broderick  TL, Bogdan  D, Wang  D, Lavoie  JM, Jankowski  M.  Downregulation of oxytocin and natriuretic peptides in diabetes: Possible implications in cardiomyopathy. J Physiol  2009;587:4725–4736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Broderick  TL, Jankowski  M, Wang  D, Danalache  BA, Parrott  CR, Gutkowska  J.  Downregulation in GATA4 and downstream structural and contractile genes in the db/db mouse heart. ISRN Endocrinol  2012;2012:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Lindström  P.  The physiology of obese-hyperglycemic mice [ob/ob mice]. ScientificWorldJournal  2007;7:666–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Ingalls  AM, Dickie  MM, Snell  GD.  Obese, a new mutation in the house mouse. J Hered  1950;41:317–318. [DOI] [PubMed] [Google Scholar]
  • 143. Christoffersen  C, Bollano  E, Lindegaard  MLS, Bartels  ED, Goetze  JP, Andersen  CB, Nielsen  LB.  Cardiac lipid accumulation associated with diastolic dysfunction in obese mice. Endocrinology  2003;144:3483–3490. [DOI] [PubMed] [Google Scholar]
  • 144. Broderick  TL, Wang  D, Jankowski  M, Gutkowska  J.  Unexpected effects of voluntary exercise training on natriuretic peptide and receptor mRNA expression in the ob/ob mouse heart. Regul Pept  2014;188:52–59. [DOI] [PubMed] [Google Scholar]
  • 145. Christoffersen  C, Bartels  ED, Nielsen  LB.  Heart specific up-regulation of genes for B-type and C-type natriuretic peptide receptors in diabetic mice. Eur J Clin Invest  2006;36:69–75. [DOI] [PubMed] [Google Scholar]
  • 146. Manolescu  DC, Jankowski  M, Danalache  BA, Wang  D, Broderick  TL, Chiasson  JL, Gutkowska  J.  All-trans retinoic acid stimulates gene expression of the cardioprotective natriuretic peptide system and prevents fibrosis and apoptosis in cardiomyocytes of obese ob/ob mice. Appl Physiol Nutr Metab  2014;39:1127–1136. [DOI] [PubMed] [Google Scholar]
  • 147. Dong  F, Zhang  X, Yang  X, Esberg  LB, Yang  H, Zhang  Z, Culver  B, Ren  J.  Impaired cardiac contractile function in ventricular myocytes from leptin-deficient ob/ob obese mice. J Endocrinol  2006;188:25–36. [DOI] [PubMed] [Google Scholar]
  • 148. Clément  K.  Genetics of human obesity. C R Biol  2006;329:608–622. [DOI] [PubMed] [Google Scholar]
  • 149. 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]
  • 150. Withaar  C, Meems  LMG, De Boer  RA.  Fighting HFpEF in women: taking aim at belly fat. Eur Heart J  2021;42:1606–1608. [DOI] [PubMed] [Google Scholar]
  • 151. Piek  A, Koonen  DPYY, Schouten  E-MM, Lindtstedt  EL, Michaëlsson  E, de Boer  RA, Silljé  HHWW.  Pharmacological myeloperoxidase (MPO) inhibition in an obese/hypertensive mouse model attenuates obesity and liver damage, but not cardiac remodeling. Sci Rep  2019;9:18765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Ternacle  J, Wan  F, Sawaki  D, Surenaud  M, Pini  M, Mercedes  R, Ernande  L, Audureau  E, Dubois-Rande  JL, Adnot  S, Hue  S, Czibik  G, Derumeaux  G.  Short-term high-fat diet compromises myocardial function: a radial strain rate imaging study. Eur Heart J Cardiovasc Imaging  2017;18:1283–1291. [DOI] [PubMed] [Google Scholar]
  • 153. Bostick  B, Aroor  AR, Habibi  J, Durante  W, Ma  L, DeMarco  VG, Garro  M, Hayden  MR, Booth  FW, Sowers  JR.  Daily exercise prevents diastolic dysfunction and oxidative stress in a female mouse model of western diet induced obesity by maintaining cardiac heme oxygenase-1 levels. Metabolism  2017;66:14–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Aroor  AR, Habibi  J, Kandikattu  HK, Garro-Kacher  M, Barron  B, Chen  D, Hayden  MR, Whaley-Connell  A, Bender  SB, Klein  T, Padilla  J, Sowers  JR, Chandrasekar  B, DeMarco  VG.  Dipeptidyl peptidase-4 (DPP-4) inhibition with linagliptin reduces western diet-induced myocardial TRAF3IP2 expression, inflammation and fibrosis in female mice. Cardiovasc Diabetol  2017;16:61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Luptak  I, Sverdlov  AL, Panagia  M, Qin  F, Pimentel  DR, Croteau  D, Siwik  DA, Ingwall  JS, Bachschmid  MM, Balschi  JA, Colucci  WS.  Decreased ATP production and myocardial contractile reserve in metabolic heart disease. J Mol Cell Cardiol  2018;116:106–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Agrawal  V, Fortune  N, Yu  S, Fuentes  J, Shi  F, Nichols  D, Gleaves  L, Poovey  E, Wang  TJ, Brittain  EL, Collins  S, West  JD, Hemnes  AR.  Natriuretic peptide receptor C contributes to disproportionate right ventricular hypertrophy in a rodent model of obesity-induced heart failure with preserved ejection fraction with pulmonary hypertension. Pulm Circ  2019;9:204589401987859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Aboumsallem  JP, Muthuramu  I, Mishra  M, De Geest  B.  Cholesterol-lowering gene therapy prevents heart failure with preserved ejection fraction in obese type 2 diabetic mice. Int J Mol Sci  2019;20:2222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Carbone  S, Mauro  AG, Mezzaroma  E, Kraskauskas  D, Marchetti  C, Buzzetti  R, Van Tassell  BW, Abbate  A, Toldo  S.  A high-sugar and high-fat diet impairs cardiac systolic and diastolic function in mice. Int J Cardiol  2015;198:66–69. [DOI] [PubMed] [Google Scholar]
  • 159. Gevaert  AB, Shakeri  H, Leloup  AJ, Van Hove  CE, De Meyer  GRY, Vrints  CJ, Lemmens  K, Van Craenenbroeck  EM.  Endothelial senescence contributes to heart failure with preserved ejection fraction in an aging mouse model. Circ Heart Fail  2017;10:e003806. [DOI] [PubMed] [Google Scholar]
  • 160. Zhong  J, Gupta  GK, Chiurchiù  V, Ahnstedt  H, Ahnstedt  H, Reilly  R-O, Ms  S, As  M, Bravo-Alegria  J, Chauhan  A, Aronowski  J, Sp  M, Ld  M, Roy-O ’reilly  M, Spychala  MS, Mobley  AS, Bravo-Alegria  J, Chauhan  A, Aronowski  J, Marrelli  SP, Mccullough  LD.  Sex differences in adipose tissue CD8+ T cells and regulatory T cells in middle-aged mice. Front Immunol  2018;9:659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. van der Heijden  RA, Bijzet  J, Meijers  WC, Yakala  GK, Kleemann  R, Nguyen  TQ, de Boer  RA, Schalkwijk  CG, Hazenberg  BPC, Tietge  UJF, Heeringa  P.  Obesity-induced chronic inflammation in high fat diet challenged C57BL/6J mice is associated with acceleration of age-dependent renal amyloidosis. Sci Rep  2015;5:16474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Ingvorsen  C, Karp  NA, Lelliott  CJ.  The role of sex and body weight on the metabolic effects of high-fat diet in C57BL/6N mice. Nutr Diabetes  2017;7:e261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Manrique  C, DeMarco  VG, Aroor  AR, Mugerfeld  I, Garro  M, Habibi  J, Hayden  MR, Sowers  JR.  Obesity and insulin resistance induce early development of diastolic dysfunction in young female mice fed a western diet. Endocrinology  2013;154:3632–3642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Meng  Q, Lai  YC, Kelly  NJ, Bueno  M, Baust  JJ, Bachman  TN, Goncharov  D, Vanderpool  RR, Radder  JE, Hu  J, Goncharova  E, Morris  AM, Mora  AL, Shapiro  SD, Gladwin  MT.  Development of a mouse model of metabolic syndrome, pulmonary hypertension, and heart failure with preserved ejection fraction. Am J Respir Cell Mol Biol  2017;56:497–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Cannon  MV, Silljé  HHW, Sijbesma  JWA, Khan  MAF, Steffensen  KR, van Gilst  WH, de Boer  RA.  LXRα improves myocardial glucose tolerance and reduces cardiac hypertrophy in a mouse model of obesity-induced type 2 diabetes. Diabetologia  2016;59:634–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Bartels  ED, Nielsen  JM, Bisgaard  LS, Goetze  JP, Nielsen  LB.  Decreased expression of natriuretic peptides associated with lipid accumulation in cardiac ventricle of obese mice. Endocrinology  2010;151:5218–5225. [DOI] [PubMed] [Google Scholar]
  • 167. Bruder-Nascimento  T, Ekeledo  OJ, Anderson  R, Le  HB, Belin de Chantemele  EJ.  Long Term High Fat Diet Treatment: an Appropriate Approach to Study the Sex-Specificity of the Autonomic and Cardiovascular Responses to Obesity in Mice. Front Physiol  2017;8: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Gai  Z, Hiller  C, Chin  SH, Hofstetter  L, Stieger  B, Konrad  D, Kullak-Ublick  GA.  Uninephrectomy augments the effects of high fat diet induced obesity on gene expression in mouse kidney. Biochim Biophys Acta Mol Basis Dis  2014;1842:1870–1878. [DOI] [PubMed] [Google Scholar]
  • 169. 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]
  • 170. Aurich  AC, Niemann  B, Pan  R, Gruenler  S, Issa  H, Silber  RE, Rohrbach  S.  Age-dependent effects of high fat-diet on murine left ventricles: role of palmitate. Basic Res Cardiol  2013;108:1–17. [DOI] [PubMed] [Google Scholar]
  • 171. Roh  JD, Houstis  N, Yu  A, Chang  B, Yeri  A, Li  H, Hobson  R, Lerchenmüller  C, Vujic  A, Chaudhari  V, Damilano  F, Platt  C, Zlotoff  D, Lee  RT, Shah  R, Jerosch-Herold  M, Rosenzweig  A.  Exercise training reverses cardiac aging phenotypes associated with heart failure with preserved ejection fraction in male mice. Aging Cell  2020;19:e13159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Roh  JD, Hobson  R, Chaudhari  V, Quintero  P, Yeri  A, Benson  M, Xiao  C, Zlotoff  D, Bezzerides  V, Houstis  N, Platt  C, Damilano  F, Lindman  BR, Elmariah  S, Biersmith  M, Lee  SJ, Seidman  CE, Seidman  JG, Gerszten  RE, Lach-Trifilieff  E, Glass  DJ, Rosenzweig  A.  Activin type II receptor signaling in cardiac aging and heart failure. Sci Transl Med  2019;11:8680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Takeda  T, Hosokawa  M, Higuchi  K.  Senescence-accelerated mouse (SAM): a novel murine model of senescence. Exp Gerontol  1997;32:105–109. [DOI] [PubMed] [Google Scholar]
  • 174. Karuppagounder  V, Arumugam  S, Babu  SS, Palaniyandi  SS, Watanabe  K, Cooke  JP, Thandavarayan  RA.  The senescence accelerated mouse prone 8 (SAMP8): a novel murine model for cardiac aging. Ageing Res Rev  2017;35:291–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Tanisawa  K, Mikami  E, Fuku  N, Honda  Y, Honda  S, Ohsawa  I, Ito  M, Endo  S, Ihara  K, Ohno  K, Kishimoto  Y, Ishigami  A, Maruyama  N, Sawabe  M, Iseki  H, Okazaki  Y, Hasegawa-Ishii  S, Takei  S, Shimada  A, Hosokawa  M, Mori  M, Higuchi  K, Takeda  T, Higuchi  M, Tanaka  M.  Exome sequencing of senescence-accelerated mice (SAM) reveals deleterious mutations in degenerative disease-causing genes. BMC Genomics  2013;14:248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Reed  AL, Tanaka  A, Sorescu  D, Liu  H, Jeong  E-M, Sturdy  M, Walp  ER, Dudley  SC, Sutliff  RL.  Diastolic dysfunction is associated with cardiac fibrosis in the senescence-accelerated mouse. Am J Physiol Circ Physiol  2011;301:H824–H831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Willard  PW.  A model for evaluation of thiazide-induced hypotension. J Pharm Pharmacol  1969;21:406–408. [DOI] [PubMed] [Google Scholar]
  • 178. Lovelock  JD, Monasky  MM, Jeong  EM, Lardin  HA, Liu  H, Patel  BG, Taglieri  DM, Gu  L, Kumar  P, Pokhrel  N, Zeng  D, Belardinelli  L, Sorescu  D, Solaro  RJ, Dudley  SC.  Ranolazine improves cardiac diastolic dysfunction through modulation of myofilament calcium sensitivity. Circ Res  2012;110:841–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Mohammed  SF, Ohtani  T, Korinek  J, Lam  CSP, Larsen  K, Simari  RD, Valencik  ML, Burnett  JC, Redfield  MM.  Mineralocorticoid accelerates transition to heart failure with preserved ejection fraction via “nongenomic effects”. Circulation  2010;122:370–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Jeong  EM, Monasky  MM, Gu  L, Taglieri  DM, Patel  BG, Liu  H, Wang  Q, Greener  I, Dudley  SC, Solaro  RJ.  Tetrahydrobiopterin improves diastolic dysfunction by reversing changes in myofilament properties. J Mol Cell Cardiol  2013;56:44–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Bowen  TS, Eisenkolb  S, Drobner  J, Fischer  T, Werner  S, Linke  A, Mangner  N, Schuler  G, Adams  V.  High-intensity interval training prevents oxidantmediated diaphragm muscle weakness in hypertensive mice. FASEB J  2017;31:60–71. [DOI] [PubMed] [Google Scholar]
  • 182. ter Maaten  JM, Damman  K, Verhaar  MC, Paulus  WJ, Duncker  DJ, Cheng  C, van Heerebeek  L, Hillege  HL, Lam  CSPP, Navis  G, Voors  AA.  Connecting heart failure with preserved ejection fraction and renal dysfunction: the role of endothelial dysfunction and inflammation. Eur J Heart Fail  2016;18:588–598. [DOI] [PubMed] [Google Scholar]
  • 183. Tanaka  K, Valero-Muñoz  M, Wilson  RM, Essick  EE, Fowler  CT, Nakamura  K, van den Hoff  M, Ouchi  N, Sam  F.  Follistatin-like 1 regulates hypertrophy in heart failure with preserved ejection fraction. JACC Basic Transl Sci  2016;1:207–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Valero-Munõz  M, Li  S, Wilson  RM, Hulsmans  M, Aprahamian  T, Fuster  JJ, Nahrendorf  M, Scherer  PE, Sam  F.  Heart failure with preserved ejection fraction induces Beiging in adipose tissue. Circ Heart Fail  2016;9:e002724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Valero-Munoz  M, Li  S, Wilson  RM, Boldbaatar  B, Iglarz  M, Sam  F.  Dual endothelin-A/endothelin-B receptor blockade and cardiac remodeling in heart failure with preserved ejection fraction. Circ Heart Fail  2016;9:e003381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Wilson  RM, De Silva  DS, Sato  K, Izumiya  Y, Sam  F.  Effects of fixed-dose isosorbide dinitrate/hydralazine on diastolic function and exercise capacity in hypertension-induced diastolic heart failure. Hypertension  2009;54:583–590. [DOI] [PubMed] [Google Scholar]
  • 187. Garcia  AG, Wilson  RM, Heo  J, Murthy  NR, Baid  S, Ouchi  N, Sam  F.  Interferon-γ ablation exacerbates myocardial hypertrophy in diastolic heart failure. Am J Physiol Heart Circ Physiol  2012;303:H587–H596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Tanaka  K, Wilson  RM, Essick  EE, Duffen  JL, Scherer  PE, Ouchi  N, Sam  F.  Effects of adiponectin on calcium-handling proteins in heart failure with preserved ejection fraction. Circ Heart Fail  2014;7:976–985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Tong  D, Schiattarella  GG, Jiang  N, May  HI, Lavandero  S, Gillette  TG, Hill  JA.  Female sex is protective in a preclinical model of heart failure with preserved ejection fraction. Circulation  2019;140:1769–1771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Reddy  SS, Agarwal  H, Barthwal  MK.  Cilostazol ameliorates heart failure with preserved ejection fraction and diastolic dysfunction in obese and non-obese hypertensive mice. J Mol Cell Cardiol  2018;123:46–57. [DOI] [PubMed] [Google Scholar]
  • 191. Gaspari  T, Brdar  M, Lee  HW, Spizzo  I, Hu  Y, Widdop  RE, Simpson  RW, Dear  AE.  Molecular and cellular mechanisms of glucagon-like peptide-1 receptor agonist-mediated attenuation of cardiac fibrosis. Diabetes Vasc Dis Res  2016;13:56–68. [DOI] [PubMed] [Google Scholar]
  • 192. Du  W, Piek  A, Marloes Schouten  E, van de Kolk  CWA, Mueller  C, Mebazaa  A, Voors  AA, de Boer  RA, Silljé  HHW.  Plasma levels of heart failure biomarkers are primarily a reflection of extracardiac production. Theranostics  2018;8:4155–4169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Withaar  C, Meems  LMG, Markousis-Mavrogenis  G, Boogerd  CJ, Silljé  HHW, Schouten  EM, Dokter  MM, Voors  AA, Westenbrink  BD, Lam  CSP, de Boer  RA.  The effects of liraglutide and dapagliflozin on cardiac function and structure in a multi-hit mouse model of heart failure with preserved ejection fraction. Cardiovasc Res  2020;cvaa256. doi: 10.1093/cvr/cvaa256 [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Deng  Y, Xie  M, Li  Q, Xu  X, Ou  W, Zhang  Y, Xiao  H, Yu  H, Zheng  Y, Liang  Y, Jiang  C, Chen  G, Du  D, Zheng  W, Wang  S, Gong  M, Chen  Y, Tian  R, Li  T.  Targeting mitochondria-inflammation circuit by β-hydroxybutyrate mitigates HFpEF. Circ Res  2021;128:232–245. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

ehab389_Supplementary_Data

Articles from European Heart Journal are provided here courtesy of Oxford University Press

RESOURCES