scholarly journals Characterization of a mouse model of obesity-related fibrotic cardiomyopathy that recapitulates features of human heart failure with preserved ejection fraction

2018 ◽  
Vol 315 (4) ◽  
pp. H934-H949 ◽  
Author(s):  
Linda Alex ◽  
Ilaria Russo ◽  
Volodymir Holoborodko ◽  
Nikolaos G Frangogiannis

Heart failure with preserved ejection fraction (HFpEF) is caused, or exacerbated by, a wide range of extracardiac conditions. Diabetes, obesity, and metabolic dysfunction are associated with a unique HFpEF phenotype, characterized by inflammation, cardiac fibrosis, and microvascular dysfunction. Development of new therapies for HFpEF is hampered by the absence of reliable animal models. The leptin-resistant db/ db mouse has been extensively studied as a model of diabetes-associated cardiomyopathy; however, data on the functional and morphological alterations in db/ db hearts are conflicting. In the present study, we report a systematic characterization of the cardiac phenotype in db/ db mice, focusing on the time course of functional and histopathological alterations and on the identification of sex-specific cellular events. Although both male and female db/ db mice developed severe obesity, increased adiposity, and hyperglycemia, female mice had more impressive weight gain and exhibited a modest but significant increase in blood pressure. db/ db mice had hypertrophic ventricular remodeling and diastolic dysfunction with preserved ejection fraction; the increase in left ventricular mass was accentuated in female mice. Histological analysis showed that both male and female db/ db mice had cardiomyocyte hypertrophy and interstitial fibrosis, associated with marked thickening of the perimysial collagen, and expansion of the periarteriolar collagen network, in the absence of replacement fibrosis. In vivo and in vitro experiments showed that fibrotic changes in db/ db hearts were associated with increased collagen synthesis by cardiac fibroblasts, in the absence of periostin, α-smooth muscle actin, or fibroblast activation protein overexpression. Male db/ db mice exhibited microvascular rarefaction. In conclusion, the db/ db mouse model recapitulates functional and histological features of human HFpEF associated with metabolic dysfunction. Development of fibrosis in db/ db hearts, in the absence of myofibroblast conversion, suggests that metabolic dysfunction may activate an alternative profibrotic pathway associated with accentuated extracellular matrix protein synthesis. NEW & NOTEWORTHY We provide a systematic analysis of the sex-specific functional and structural myocardial alterations in db/ db mice. Obese diabetic C57BL6J db/ db mice exhibit diastolic dysfunction with preserved ejection fraction, associated with cardiomyocyte hypertrophy, interstitial/perivascular fibrosis, and microvascular rarefaction, thus recapitulating aspects of human obesity-related heart failure with preserved ejection fraction. Myocardial fibrosis in db/ db mice is associated with a matrix-producing fibroblast phenotype, in the absence of myofibroblast conversion, suggesting an alternative mechanism of activation.

Circulation ◽  
2015 ◽  
Vol 132 (suppl_3) ◽  
Author(s):  
Jessica A Regan ◽  
Adolofo G Mauro ◽  
Salvatore Carbone ◽  
Carlo Marchetti ◽  
Eleonora Mezzaroma ◽  
...  

Background: Heart failure with preserved ejection fraction (HFpEF) is characterized by elevated left ventricular (LV) filling pressures due to impaired LV diastolic function. Low-dose infusion of angiotensin 2 (AT2) in the mouse induces a HFpEF phenotype without increasing blood pressure. AT2 infusion induces expression of Interleukin-18 (IL-18) in the heart. We therefore tested whether IL-18 mediated AT2-induced LV diastolic dysfunction in this model. Methods: We infused subcutaneously AT2 (0.2 mg/Kg/day) or a matching volume of vehicle via osmotic pumps surgically implanted in the interscapular space in adult wild-type (WT) male mice and IL-18 knock-out mice (IL-18KO). We also treated WT mice with daily intraperitoneal injections of recombinant murine IL-18 binding protein (IL-18bp, a naturally occurring IL-18 blocker) at 3 different doses (0.1, 0.3 and 1.0 mg/kg) or vehicle for 25 days starting on day 3. We performed a Doppler-echocardiography study before implantation and at 28 days to measure LV dimensions, mass, and systolic and diastolic function in all mice. LV catheterization was performed prior to sacrifice to measure LV end-diastolic pressure (LVEDP) using a Millar catheter. Results: AT2 induces a significant increase in isovolumetric relaxation time (IRT) and myocardial performance index (MPI) at Doppler echocardiography and elevation of LVEDP at catheterization, indicative of impaired LV diastolic function, in absence of any measurable effects on systolic blood pressure nor LV dimensions, mass, or systolic function. Mice with genetic deletion of IL-18 (IL-18 KO) or WT mice treated with IL-18bp had no significant increase in IRT, MPI or LVEDP with AT2 infusion. Conclusion: Genetic or pharmacologic IL-18 blockade prevent diastolic dysfunction in a mouse model of HFpEF induced by low dose AT2 infusion, suggesting a critical role of IL-18 in the pathophysiology of HFpEF.


2021 ◽  
Vol 129 (Suppl_1) ◽  
Author(s):  
Naixin Zhang ◽  
Katherine A Murphy ◽  
Dylan J Gyberg ◽  
Timothy D O'Connell

Non-resolving inflammation is central to the pathogenesis of heart failure (HF). Heart failure preserved ejection fraction (HFpEF) is a type of HF that is particularly associated with inflammation provoked by metabolic syndrome (MetS). The G-protein coupled receptor, free fatty acid receptor 4 (Ffar4), is a receptor for medium and long chain fatty acids (FA) that regulates metabolism and attenuates inflammation. Ffar4 is expressed in the human heart, and downregulated in heart failure. Furthermore, polymorphisms in Ffar4 have been associated with eccentric remodeling in a patient cohort. Previously, Ffar4 was shown to protect the heart from pathologic stress by attenuating oxidative stress in a mouse model of pressure overload. Here, we tested the hypothesis that Ffar4 would attenuate the development of heart failure using a mouse model of MetS-induced HFpEF. Metabolic syndrome was induced in mice by feeding a high-fat, high-sucrose diet (42% fat, 30% sucrose) to produce obesity and delivering the nitric oxide synthase inhibitor, L-NAME, in the drinking water to induce hypertension. The combined intervention (referred to as HFpEF diet) resulted in mice developing excessive adiposity, glucose intolerance (in males only), and mild hypertension. After 20 weeks on the HFpEF diet, both male and female WT mice, developed diastolic dysfunction (increased E/A and E/e’) and preserved ejection fraction (EF), consistent with clinical HFpEF. In Ffar4KO male mice HFpEF diet induced a greater degree of diastolic dysfunction compared to WT mice, despite equivalent metabolic parameters. Female Ffar4KO mice fed the HFpEF diet had a greater increase in weight gain and adiposity compared to WT female mice. Surprisingly, diastolic function was equivalent between WT and FFAR4KO female mice, suggesting a sex-based difference in FFAR4 cardioprotection. Our data show that Ffar4 attenuates HFpEF secondary to MetS.


Hypertension ◽  
2020 ◽  
Vol 76 (Suppl_1) ◽  
Author(s):  
Charles D Smart ◽  
Vineet Agrawal ◽  
Anna R Hemnes ◽  
Meena S Madhur

Deoxycorticosterone acetate (DOCA)-salt is a common hypertension model in mice and has recently been used to study heart failure with preserved ejection fraction (HFpEF) in rats. Our goal was to validate DOCA-salt as a mouse model of HFpEF and determine how DOCA-salt alters the cardiac immunological landscape to identify novel therapeutic targets for this disease. DOCA-salt mice underwent uninephrectomy, implantion of a DOCA pellet, and supplementation of the drinking water with 1% NaCl water for three weeks. Control mice underwent a sham procedure and received normal water. Compared to control mice, DOCA-salt mice exhibited elevated systolic BP, increased heart weight to body weight ratios (5.6 vs 7.1), increased lung wet to dry weight ratios (4.4 vs 4.8) indicative of pulmonary congestion, and decreased time to exhaustion upon treadmill exercise testing (23.0 vs. 18.5 seconds). On conscious echocardiography, DOCA-salt mice exhibited a preserved ejection fraction. Invasive hemodynamic studies revealed an increased tau constant (5.7 vs 8.2) and increased end-diastolic pressures in DOCA-salt mice (1.7 vs 2.6), consistent with diastolic dysfunction. CITE-seq, a novel technique to obtain transcriptomic and surface marker expression on single cells, was performed on a total of 4,359 and 7,600 cells sorted live CD45+ leukocytes from four sham and four DOCA-salt left ventricles, respectively. Unsupervised computational analysis revealed 29 clusters of immune cells. Six clusters containing natural killer, T lymphocyte and myeloid cell populations were overrepresented and five B cell clusters were underrepresented in DOCA-salt samples. Differential expression analysis of CD11b+CD64+ cardiac macrophages revealed transcriptional changes between groups with 146 significantly upregulated and 111 downregulated genes. Gene set enrichment analysis showed upregulation of leukocyte migration, response to type I interferon, and cytokine-mediated signaling pathways in DOCA-salt macrophages. In conclusion, the DOCA-salt mouse model recapitulates key features of HFpEF including diastolic dysfunction with preserved ejection fraction, cardiac hypertrophy, and pulmonary congestion and is associated with an altered cardiac immune cell profile.


2021 ◽  
Vol 35 (S1) ◽  
Author(s):  
Sasha Smolgovsky ◽  
Francisco Carrillo‐Salinas ◽  
Marina Anastasiou ◽  
Kuljeet Kaur ◽  
Mark Aronovitz ◽  
...  

Circulation ◽  
2020 ◽  
Vol 142 (Suppl_4) ◽  
Author(s):  
Yang Cao

Introduction: Heart failure with preserved ejection fraction (HFpEF) is a complex syndrome involving diastolic dysfunction and metabolic disorders. Women are twice as likely to develop HFpEF as compared to men and present with a greater symptom burden. However, the sex-specific biology of HFpEF is unknown. This study aims to dissect sex-specific molecular signatures of diastolic dysfunction in HFpEF. Methods: We developed a ‘two-hit’ HFpEF model with HFD and l-NAME (Nω-nitrol-arginine methyl ester) in C57BL/6J male and female mice. RNA-seq for heart tissue from mice under HFpEF versus Chow condition was performed and results were integrated with multi-omics datasets including hybrid mouse diversity panel (HMDP, a panel of more than 100 inbred strains of mice) and human heart failure cohort to dissect novel gene-by-sex interactions in the heart. Results: HFD and l-NAME induced diastolic heart failure phenotypes in both male and female mice. We observed higher E/A (1.68 ± 0.09 vs. 1.40 ± 0.04, p < 0.05 under chow diet), E/E’ (35.82 ± 2.05 vs. 26.13±1.11, p < 0.05 under chow diet; 53.78 ± 3.94 vs. 43.92±2.08, p < 0.05 after HFpEF) and lung weight (wet/dry ratio, 4.51 ± 0.09 vs. 4.15 ± 0.09, p < 0.05 after HFpEF) in females compared to males. Pathway analysis revealed that compared with male mice, female mice exhibited a greater decrease in fatty acid metabolism, insulin signaling, heart development and an enhanced increase in cell cycle, inflammation, cholesterol efflux and serine synthesis upon HFpEF. Together with our high fat/high sucrose diet-induced metabolic disorder HMDP cohort and isoproterenol-induced heart failure HMDP cohort, HFpEF mice revealed that female hearts contained reduced mtDNA copy number relative to males (3102.56 ± 85.70 vs. 3770.43 ± 77.44, p = 4.51E-08). Cardiac mtDNA was inversely correlated with diastolic dysfunction (bicor = -0.307, p = 0.005) and was regulated by gonadal hormones. Conclusions: We utilize our systems genetics approach that integrates multi-omics datasets and dissect novel gene-by-sex interactions in the heart. Females exhibit more severe metabolic disorders and mitochondrial dysfunction relative to males, which are responsible for greater diastolic dysfunction of HFpEF.


2021 ◽  
Vol 35 (S1) ◽  
Author(s):  
Francisco Carrillo‐Salinas ◽  
Sasha Smolgovsky ◽  
Marina Anastasiou ◽  
Kuljeet Kaur ◽  
Mark Aronovitz ◽  
...  

2021 ◽  
Vol 129 (Suppl_1) ◽  
Author(s):  
Sasha Smolgovsky ◽  
Francisco Carrillo-Salinas ◽  
Marina Anastasiou ◽  
Kuljeet Kaur ◽  
Abraham Bayer ◽  
...  

Introduction: Heart Failure with Preserved Ejection Fraction (HFpEF) is a prevalent cardiovascular syndrome with no curative therapies, characterized by diastolic dysfunction, preserved systolic function, and decreased expression of unfolded protein response (UPR) genes in the heart. Obesity and hypertension are risk factors for HFpEF and are intimately associated with systemic inflammation. However, the inflammatory mechanisms driving HFpEF remain largely unexplored. Hypothesis: We hypothesized that nitrosative stress induced by obesity and hypertension programs T cells to infiltrate the heart and drive cardiac pathology in HFpEF. Methods: Using a recently established model of HFpEF, we modeled obesity and hypertension in male C57/BL6 (wild-type, WT), T cell receptor alpha-deficient ( Tcra-/-), and Nur77-GFP reporter mice for T cell receptor engagement, using a high-fat diet (HFD) and L-NAME for 5 weeks, or standard chow (STD) as controls. Invasive hemodynamic analyses were used to assess cardiac function, and the heart and lymphoid organs were harvested to characterize immune cell populations by flow cytometry, histology, and gene expression of cardiac remodeling. Results: In WT mice, HFD/L-NAME induced significant cardiac infiltration of T cells alongside the hallmarks of HFpEF. HFD/L-NAME significantly expanded CD62 lo CD44 hi effector T cells in the mediastinal lymph nodes and spleen. Nur77-GFP mice revealed no antigen engagement by T cells in the heart, however, T cells sorted out of the lymphoid organs of HFpEF mice had significantly decreased gene expression of the UPR gene spliced X box-binding protein 1 (XBP1s) compared to controls, suggesting a T cell intrinsic dysregulation of the UPR, and T cell phenotypic changes independent of TCR engagement in the heart. Strikingly, Tcra-/- mice did not develop diastolic dysfunction or cardiomyocyte hypertrophy, demonstrating a novel role for T cells in this experimental model of HFpEF. Conclusions: We conclude diastolic dysfunction and cardiomyocyte hypertrophy in HFpEF is T cell dependent. Ongoing studies are determining whether the observed intrinsic T cell changes in XBP1s prime T cells for cardiac infiltration and effector function that results in diastolic dysfunction and HFpEF.


2017 ◽  
Vol 313 (2) ◽  
pp. H392-H407 ◽  
Author(s):  
Hirofumi Saiki ◽  
Gilles Moulay ◽  
Adam J. Guenzel ◽  
Weibin Liu ◽  
Teresa D. Decklever ◽  
...  

Breast cancer radiotherapy increases the risk of heart failure with preserved ejection fraction (HFpEF). Cardiomyocytes are highly radioresistant, but radiation specifically affects coronary microvascular endothelial cells, with subsequent microvascular inflammation and rarefaction. The effects of radiation on left ventricular (LV) diastolic function are poorly characterized. We hypothesized that cardiac radiation exposure may result in diastolic dysfunction without reduced EF. Global cardiac expression of the sodium-iodide symporter (NIS) was induced by cardiotropic gene (adeno-associated virus serotype 9) delivery to 5-wk-old rats. SPECT/CT (125I) measurement of cardiac iodine uptake allowed calculation of the 131I doses needed to deliver 10- or 20-Gy cardiac radiation at 10 wk of age. Radiated (Rad; 10 or 20 Gy) and control rats were studied at 30 wk of age. Body weight, blood pressure, and heart rate were similar in control and Rad rats. Compared with control rats, Rad rats had impaired exercise capacity, increased LV diastolic stiffness, impaired LV relaxation, and elevated filling pressures but similar LV volume, EF, end-systolic elastance, preload recruitable stroke work, and peak +dP/d t. Pathology revealed reduced microvascular density, mild concentric cardiomyocyte hypertrophy, and increased LV fibrosis in Rad rats compared with control rats. In the Rad myocardium, oxidative stress was increased and in vivo PKG activity was decreased. Experimental cardiac radiation exposure resulted in diastolic dysfunction without reduced EF. These data provide insight into the association between cardiac radiation exposure and HFpEF risk and lend further support for the importance of inflammation-related coronary microvascular compromise in HFpEF. NEW & NOTEWORTHY Cardiac radiation exposure during radiotherapy increases the risk of heart failure with preserved ejection fraction. In a novel rodent model, cardiac radiation exposure resulted in coronary microvascular rarefaction, oxidative stress, impaired PKG signaling, myocardial fibrosis, mild cardiomyocyte hypertrophy, left ventricular diastolic dysfunction, and elevated left ventricular filling pressures despite preserved ejection fraction.


2019 ◽  
Vol 94 (7) ◽  
pp. 1199-1209 ◽  
Author(s):  
Yogesh N.V. Reddy ◽  
Gregory D. Lewis ◽  
Sanjiv J. Shah ◽  
Masaru Obokata ◽  
Omar F. Abou-Ezzedine ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document