- Review
- Open access
- Published:
The role of glucagon-like peptide-1 receptor (GLP-1R) agonists in enhancing endothelial function: a potential avenue for improving heart failure with preserved ejection fraction (HFpEF)
Cardiovascular Diabetology volume 24, Article number: 70 (2025)
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a prevalent and complex condition with limited effective treatments. Endothelial dysfunction is a significant component of HFpEF pathophysiology, and glucagon-like peptide-1 receptor (GLP-1R) agonists have shown potential benefits in improving endothelial function. This study aims to explore the relationship between endothelial dysfunction in HFpEF and the mechanisms of action of GLP-1R agonists, highlighting their potential therapeutic benefits. A comprehensive review of the literature was conducted to examine the etiology of HFpEF, the role of endothelial dysfunction, and the effects of GLP-1R agonists on endothelial function and heart failure outcomes. The findings indicate that HFpEF is associated with various comorbidities, such as obesity, diabetes mellitus, and hypertension, which contribute to endothelial dysfunction. GLP-1R agonists, including semaglutide and liraglutide, have demonstrated significant cardioprotective effects, such as improving vascular endothelial function, reducing inflammation, and preventing atherosclerosis. Clinical trials, such as the STEP-HFpEF trial, have shown positive results in reducing symptoms and physical restrictions in HFpEF patients. GLP-1R agonists present a promising therapeutic option for HFpEF by targeting endothelial dysfunction and other pathophysiological mechanisms. Further research is needed to elucidate the precise mechanisms through which GLP-1R agonists exert their benefits and to establish their long-term safety and efficacy in diverse HFpEF populations.
Introduction
Heart failure (HF) affects approximately seven million adults in the United States, with one-third to half experiencing heart failure with preserved ejection fraction (HFpEF) [1,2,3,4]. The prevalence of HFpEF is rising, increasing from 33 to 39% between 2005 and 2010, with projections suggesting it will soon become the most common form of HF [5, 6].
The standard treatment for HFpEF includes sodium-glucose cotransporter 2 inhibitors (SGLT2i), such as empagliflozin and dapagliflozin, supported by the EMPEROR-Preserved (2021) and DELIVER (2022) trials [7, 8]. Glucagon-like peptide 1 receptor agonists (GLP-1RAs) have also emerged as a potential therapy. While the LIVE and FIGHT trials explored their effects on heart failure with reduced ejection fraction (HFrEF), the STEP-HFpEF trial demonstrated that GLP-1RAs improve HFpEF outcomes by reducing symptoms, physical limitations, and body weight [9].
Despite these promising results, the STEP-HFpEF trial did not clarify the mechanisms underlying these benefits. Capone et al. discussed the cardiometabolic effects of GLP-1RAs in HFpEF, focusing on obesity [10]. However, HFpEF is multifactorial, and GLP-1RA mechanisms extend beyond obesity management. This study aims to explore the link between endothelial dysfunction, a key component of HFpEF, and the therapeutic actions of GLP-1RAs.
Review
Etiology of HFpEF
Based on population-based studies conducted in the United States, more than 6 million people over the age of 20 have HF at this time. Patients diagnosed with diastolic heart failure (HFpEF) account for approximately half of heart failure hospitalizations. From 2005 (33%) to 2010 (39%), the prevalence increased (from 3.1 cases per person-year in the age group 65–69 years to 14.5 cases per person-year in the age group > 80 years). An increase in the diagnosis of HFpEF was noted in both sexes [3, 4].
The definition of HFpEF includes a combination of previous or current symptoms and/or signs of structural and functional abnormalities with the presence of congestion (including the level of NPs or radiological, ultrasound signs (LVEF ≥ 50%, E/E′ > 15, regurgitant or obstructive valvular injury, ventricular hypertrophy) [1, 2].
There is a shifting trend in the risk factors associated with heart failure in epidemiological studies from classic smoking and hypertension as risk factors for ischemic heart disease and heart failure with reduced ejection fraction, to morbid obesity, diabetes mellitus, and metabolic syndrome in the aging population as risk factors for heart failure with preserved ejection failure [5]. Over the past decade, it has been demonstrated that myocardial structure, cardiomyocyte function, and intramyocardial signaling undergo specific alterations in heart failure with preserved ejection fraction (HFpEF); however, a new paradigm for HFpEF development proposes that a systemic proinflammatory state, induced by comorbidities, is responsible for these myocardial structural and functional changes [11]. The cormorbidities often associated with HFpEF such as obesity, diabetes mellitus, and hypertension as well as the metabolic changes associated with them are known to induce a state of inflammation that affects cardiac metabolism, as well as coronary vascular injury and ischemia [5]. This new HFpEF paradigm thus shifts the focus from LV afterload excess to coronary microvascular inflammation.
Role of endothelial dysfunction in the development of HFpEF
Endothelial dysfunction plays a critical pathophysiological role in heart failure with preserved ejection fraction (HFpEF). This dysfunction affects not only the endothelial cells of the coronary vessels but more crucially those in the intramyocardial capillaries and endocardium, where they interact directly with adjacent cardiomyocytes [12]. Experimental post-infarction models have demonstrated that both cardiac and pulmonary vascular endothelial dysfunction contribute to heart failure development; however, data specifically regarding HFpEF are limited [13, 14].
Key risk factors for HFpEF, such as hypertension and diabetes, are associated with endocardial and myocardial capillary endothelial abnormalities in experimental models [15, 16].
The impact of aging endothelial cells has been explored using mouse models, revealing that mice with accelerated senescence exhibit endothelial cellular dysfunction, impaired relaxation, and interstitial fibrosis, leading to HFpEF. This is correlated with increased endothelial inflammation, characterized by intercellular adhesion molecule 1 expression, and enhanced endothelial senescence at the molecular level. Aged mice in the study showed signs of endothelial dysfunction, increased vascular stiffness, and impaired coronary microcirculation, which are hallmarks of HFpEF [17].
These endothelial abnormalities may account for the impaired left ventricular relaxation observed in pressure-overload hypertrophy [18]. Additionally, the coronary endothelium has been shown to influence left ventricular diastolic function through paracrine effects in both healthy individuals and post-transplant patients [19].
The combined stiffening of the ventricular and vascular systems, including both systemic and pulmonary circulations, significantly contributes to the pathophysiology of HFpEF [20, 21].
In the systemic circulation, endothelial dysfunction, detectable in healthy individuals with normal brachial blood pressure, is associated with increased central pulse pressure and systemic arterial stiffening. This suggests that systemic endothelial dysfunction may be a primary factor in the development of systemic hypertension and its pathological consequences, such as increased left ventricular wall stress, hypertrophy, diastolic dysfunction, and HFpEF [22]; similarly, in the pulmonary circulation, endothelial dysfunction has been identified as an early event leading to the development of pulmonary hypertension in the context of experimental heart failure [21].
A study by Akiyama et al. highlighted the prognostic significance of endothelial dysfunction in HFpEF. The study found that peripheral endothelial dysfunction independently correlated with future cardiovascular events, thus providing additional clinical significance for risk stratification in HFpEF patients. The prognostic impact of the reactive hyperemia index (RHI) in HFpEF patients suggests that endothelial dysfunction may not be merely a passive finding, but rather that endothelial function plays an active and crucial pathophysiological role in HFpEF [23].
Coronary microvascular dysfunction in HFpEF
Understanding the relationship between coronary microvascular dysfunction (CMD) and heart failure with preserved ejection fraction (HFpEF) is crucial. The association between HFpEF and CMD was first proposed in 2013 [11]. Since then, multiple studies have investigated CMD in HFpEF patients. Sucato et al. [24] found that HFpEF patients exhibited more significant microcirculation involvement compared to those without HFpEF. An autopsy study revealed a higher prevalence of coronary microvascular rarefaction, microscopic fibrosis, and hypertrophy in HFpEF patients [25].
Response of the coronary microvascular bed to acetylcholine is diminished in HFpEF. This diminished vasodilator response is correlated with left ventricular diastolic dysfunction [26]. Similar paracrine interactions between the endocardium and myocardium have been previously reported [12]. Recent studies have highlighted the significance of a deficient systemic vasodilator response in contributing to the reduced exercise tolerance observed in HFpEF patients [27].
According to Rush et al [28] CMD is common and often underrecognized in HFpEF patients and may represent a therapeutic target. A metanalysis by Lin et al. summarized and analyzed all research on the prevalence of CMD in HFpEF patients, finding a high pooled prevalence of 71% [29].
GLP-1 agonists and their cardioprotective effects
Incretins are gut hormones that enhance insulin secretion in response to meals, in a glucose- dependent manner. The two most studied incretins, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) stimulate insulin release through specific G-protein-coupled receptors predominantly found on islet β cells. These receptors are also present in non-islet cells and contribute to various metabolic effects in the brain, liver, adipose tissue, and gastrointestinal tract. Two primary strategies to enhance incretin receptor signaling have been explored for type 2 diabetes treatment; one involves inhibiting dipeptidyl peptidase-4 (DPP-4), the enzyme that deactivates GIP and GLP-1; the other strategy includes injectable GLP-1R agonists that mimic human GLP-1 or nonmammalian GLP-1R agonists. The insulin-enhancing effects of GIP and GLP-1 were discovered over 25 years ago, yet new roles for incretin hormones continue to emerge [30].
There is a wide range of cardioprotective benefits to GLP-1R agonists in addition to their metabolic effects on blood glucose, adiposity, serum lipids, and blood pressure [31]. Despite the limited expression of GLP-1 receptors in the cardiac tissue, they tend to enhance the myocardial function through multiple mechanisms, including optimizing the coronary vascular smooth muscle cells, and endothelial function of the coronary microvasculature and the peripheral vessels as well, in addition to improving blood glucose control and providing alternative energy substrates like ketones and lactate. At the level of the vascular smooth muscle cells, they protect against pathogenic, proliferative remodeling of the vascular smooth muscle cells, thus delaying plaque formation and stabilizing existing lesions. They also enhance the cardiac mitochondrial function leading to improved outcomes under hypoxic conditions and reduced pathologic remodeling [31].
The G protein-coupled receptor of GLP-1 (GLP-1R) originally identified in islet β-cells is widely expressed in extrapancreatic tissues as well including the lungs, kidney, brain, the enteric and peripheral nervous system, lymphocytes, smooth muscle cells (SMCs), and atrial cardiomyocytes [32] The specific identity of GLP-1 receptor-positive (GLP-1R+) cells in the heart remains uncertain. Immunohistochemical studies in monkeys and humans found GLP-1R+ cells mainly in the sinoatrial node and atrial tissue, with higher expression in the right atrium. However, varied detection methods show inconsistent GLP-1R localization across heart tissues. While murine models suggest atrial GLP-1R expression, the precise cardiac cell types expressing GLP-1R, especially in humans, are yet to be conclusively identified [33].
Role of GLP-1 agonists in managing comorbidities in HFpEF
Beyond their cardioprotective properties, GLP-1 receptor agonists exert a beneficial impact on HFpEF by influencing other organ systems and mitigating comorbid conditions that contribute to its progression, including metabolic dysfunction-associated steatotic liver disease (MASLD), chronic kidney disease, and pulmonary disorders.
Liver disease in heart failure with preserved ejection fraction
The relationship between heart failure (HF) and liver disease is characterized by bidirectional interactions where one condition exacerbates the other. HF can lead to liver complications such as congestive hepatopathy and ischemic liver injury, primarily through hemodynamic changes, including hepatic congestion and hypoperfusion. Conversely, liver conditions like metabolic dysfunction-associated steatotic liver disease (MASLD), also known as non-alcoholic fatty liver disease (NAFLD) can increase the risk of HF due to systemic inflammation, oxidative stress, and metabolic disturbances [34, 35].
MASLD contributes to HFpEF through several interconnected mechanisms; ectopic fat deposition in the liver and epicardium triggers inflammation, disrupts myocardial energy metabolism, and promotes fibrosis, contributing to cardiac dysfunction and diastolic impairment. Hepatic insulin resistance further exacerbates cardiovascular risk by impairing lipid and glucose metabolism, leading to atherogenic dyslipidemia, vascular inflammation, and endothelial dysfunction. Systemic inflammation driven by steatosis and necro-inflammatory changes in MASLD activates pathways such as toll-like receptors (TLR) and Nuclear Factor-kappa B (NF-κB), causing cardiac and vascular tissue damage. Parallel fibrogenesis in the liver and myocardium adds to ventricular stiffness and diastolic dysfunction, while gut dysbiosis from a Western diet heightens systemic inflammation and endotoxemia, worsening cardiac and hepatic dysfunction. Additionally, genetic factors, including TM6SF2 variants, intensify hepatic fibrosis and dyslipidemia, increasing cardiovascular susceptibility. Collectively, these mechanisms establish a connection between MASLD and diastolic dysfunction, arrhythmias, and a heightened risk of HFpEF [36]. In an animal study, using a mouse model with different age groups, non-alcoholic steatohepatitis (NASH) was induced, revealing age-dependent effects on cardiac function. NASH caused hepatomegaly, fibrosis, and inflammation in all mice, with aged mice showing increased heart weight, left ventricular volumes, and subtle systolic and diastolic dysfunction, thus NASH tends to worsen age-related cardiac dysfunction [37].
In a previous metanalysis, GLP-1 agonists were found to improve hepatic parameters in patients with NAFLD [38]. Multiple studies have demonstrated that patients with MAFLD are at an increased risk of developing heart failure (HF) compared to those without MAFLD. This association persists even in the absence of traditional cardiovascular disease (CVD) risk factors, suggesting a direct relationship between MAFLD and the risk of new-onset HF [39].
GLP-1 receptor agonists (GLP-1RAs) offer therapeutic potential in this interplay. GLP-1RAs improve glucose homeostasis, reduce liver fat, and mitigate hepatic inflammation. These effects help slow the progression of liver fibrosis and reduce cardiovascular risks [34]. Additionally, they may indirectly benefit HF by modulating systemic inflammation and improving metabolic profiles [35].
In a study that examined the effects of GLP-1 receptor agonists (GLP-1 RAs) on liver fibrosis in type 2 diabetes (T2DM) patients with nonalcoholic fatty liver disease (NAFLD). GLP-1 RAs may modestly slow liver fibrosis progression in T2DM patients with NAFLD [40]. In another study, GLP-1 RA use was associated with a lower risk of cirrhosis progression, reduced complications, and lower mortality in non-cirrhotic patients [41].
There is a significant link between NAFLD and an increased risk of new-onset heart failure, regardless of the presence or absence of Type 2 diabetes and other associated cardiometabolic risk factors. The risk magnitude rises with the severity of liver disease in NAFLD, particularly at higher fibrosis stages [42].
GLP-1 receptor agonists (GLP-1 RAs) improve glycemic control, delay gastric emptying, and promote weight loss. These properties make them an attractive therapeutic option for managing non-alcoholic fatty liver disease (NAFLD), particularly in individuals with concurrent diabetes mellitus and obesity.
GLP-1 receptor agonists, such as liraglutide, show promise in treating non-alcoholic fatty liver disease (NAFLD), particularly in individuals with type 2 diabetes (T2DM) and obesity. Clinical trials demonstrate that liraglutide reduces liver fat, improves liver function, and decreases inflammatory markers, though its effects on epicardial adipose tissue (EAT) are inconsistent. GLP-1 receptor agonists also exhibit cardiovascular and anti-inflammatory benefits, potentially reducing the risk of heart failure with preserved ejection fraction (HFpEF). However, further studies are needed to fully understand their effects, particularly on EAT and HFpEF [43].
A multicenter, randomized, double-blind, placebo-controlled trial demonstrated that liraglutide was associated with the resolution of non-alcoholic steatohepatitis (NASH) without worsening fibrosis. Improvements in steatosis and hepatocyte ballooning scores were also observed. Additionally, liraglutide and exenatide have been shown to reduce trunk fat content, particularly in the android region, a critical area linked to NAFLD and cardiovascular disease (CVD) risk [44].
A 12-month interventional study demonstrates the beneficial effects of semaglutide, both oral and subcutaneous, on body weight, insulin resistance, liver function, lipid profile, and hepatic steatosis. Significant improvements were also observed in fibrosis markers, including FIB-4, CAP, and LSM, supporting the potential for broader clinical use of semaglutide in NAFLD patients with type 2 diabetes and obesity.
Further randomized trials with larger sample sizes, including NAFLD patients with and without diabetes and incorporating active control groups, are needed to confirm these findings [45].
Chronic kidney disease in heart failure with preserved ejection fraction
In the pooled analysis of the SUSTAIN 6 and LEADER trials, with median follow-up periods of 2.1 and 3.8 years respectively, semaglutide and liraglutide demonstrated significant renal benefits compared to placebo. Both drugs reduced albuminuria by 24% over two years, with the most substantial reduction observed with semaglutide 1.0 mg (33%). Additionally, the decline in estimated glomerular filtration rate (eGFR) was significantly slowed by 0.87 mL/min/1.73 m2 per year with semaglutide and 0.26 mL/min/1.73 m2 per year with liraglutide. These effects were more pronounced in patients with baseline eGFR below 60 mL/min/1.73 m2. Both drugs also significantly reduced the risk of persistent eGFR reductions of 40% and 50%, while showing similar trends for 30% and 57% reductions. Among patients with baseline eGFR between 30 and < 60 mL/min/1.73 m2, the likelihood of persistent reductions across all thresholds was further decreased, ranging from a 29% to 46% risk reduction depending on the threshold [46].
The “Evaluate Renal Function with Semaglutide Once Weekly” (FLOW) trial, published earlier this year, is the first to specifically investigate kidney outcomes with GLP-1 receptor agonists. Similarly, the “Semaglutide Effects on Cardiovascular Outcomes in People with Overweight or Obesity” (SELECT) trial demonstrated a significant reduction in cardiovascular events in individuals with overweight or obesity, with subsequent analysis addressing prespecified kidney endpoints. Together, these studies offer valuable new evidence supporting the use of GLP-1 receptor agonists for kidney and cardiovascular protection in individuals at risk for or with established chronic kidney disease.
The FLOW trial demonstrated clear kidney and cardiovascular protection with GLP-1 receptor agonist (GLP-1RA) treatment in individuals with type 2 diabetes and chronic kidney disease (CKD). It also highlighted significant weight loss benefits, which are particularly valuable for overweight or obese patients with CKD. While the trial showed potential additive benefits when combined with SGLT2 inhibitors (SGLT2i) or finerenone, conclusive evidence for these effects is lacking. However, limitations included the restricted inclusion criteria (eGFR range of 20–75 mL/min/1.73 m2 and albuminuria requirements), which limit generalizability to patients with higher or lower eGFR or preserved eGFR without albuminuria. These findings position GLP-1RAs as a potential fourth pillar of kidney-protective therapy in diabetes, alongside renin-angiotensin system inhibitors (RASi), SGLT2i, and finerenone, and suggest a shift toward rapid initiation of combination therapies for maximum kidney protection.
The SELECT trial provided the first evidence of kidney protection with GLP-1RA treatment in individuals without diabetes, although this was a prespecified secondary endpoint. It showed potential benefits for CKD patients, particularly those who are overweight or obese. However, the trial's low event rate and reliance on serum creatinine to assess eGFR, which may be influenced by muscle mass changes, limit the robustness of its conclusions. Despite these limitations, the findings support recommending GLP-1RAs to facilitate weight loss and improve outcomes in CKD patients with obesity, even in the absence of diabetes.
Together, the FLOW and SELECT trials underscore the potential of GLP-1RAs to address multimorbidity in CKD by impacting obesity, cardiovascular disease, and kidney health. These therapies offer a promising new approach to improving overall health in CKD patients. Further studies and economic evaluations are warranted to refine combination treatment strategies and maximize cost-effectiveness while addressing individual patient needs [47].
Emerging dual agonists, such as survodutide and cotadutide, which target GLP-1 and glucagon receptors, are being explored for treating metabolically associated fatty liver disease and may also address fatty kidney, a key factor in CKD related to visceral obesity [48].
Chronic lung disease in heart failure with preserved ejection fraction
In a nationwide cohort study, it was demonstrated that GLP-1 RAs were associated with a reduced risk of cardiopulmonary complications and all-cause mortality compared to non-GLP-1 RAs in patients with T2D and COPD. GLP-1 RAs may offer a beneficial approach for managing diabetes in individuals with COPD [49]. There is also additional evidence supporting the role of GLP-1 agonists in reducing exacerbations of respiratory diseases [50].
GLP-1RAs may effectively alleviate clinical symptoms in COPD patients, improve airflow limitation, mitigate airway damage, fibrosis, and remodeling, shorten hospital stays, reduce the economic burden, and lower the risk of COPD-related complications. These effects contribute to better long-term prognoses and highlight the potential therapeutic and preventive role of GLP-1RAs in COPD management, offering a promising new target for its treatment and prevention [51].
GLP-1RA markedly enhance lung function in COPD patients, as demonstrated by improved spirometry results. Furthermore, both GLP-1RA and SGLT-2i are linked to a reduced risk of moderate and severe exacerbations in COPD and may lower the incidence of respiratory conditions such as asthma and pneumonia. While the exact mechanisms driving these benefits remain unclear, they likely involve diverse effects, including anti-inflammatory properties and mitigation of oxidative stress [52]. Role in reversing lung fibrosis and improving lung function in experimental models [53, 54].
In preclinical studies, Glucagon-like peptide-1 (GLP-1) has demonstrated its protective effects in lung disease models, potentially mediated by atrial natriuretic peptide (ANP). Using a chronic obstructive pulmonary disease (COPD) mouse model, researchers investigated the relationship between GLP-1 and ANP through genetic GLP-1 receptor (GLP-1R) knockout, pharmaceutical GLP-1R blockade, and exogenous GLP-1 administration. Treatment with a GLP-1R agonist improved lung function and reduced inflammation but did not mitigate emphysema. It also significantly increased ANP expression and reduced endothelin-1, a bronchoconstrictor. Both ANP and GLP-1R agonists showed bronchodilatory effects, with stronger responses in COPD mice. This study suggests a functional link between GLP-1 and ANP in improving lung function and reducing inflammation in COPD, highlighting their therapeutic potential [55].
Effects of GLP-1 agonists on vascular endothelium
Enhancing endothelial function
A review of the literature revealed multiple preclinical studies on GLP-1R agonists in animal models demonstrating the molecular mechanisms behind their pharmacological effects.
One protective mechanism of GLP-1 receptor (GLP1R) agonists offers protection by boosting insulin sensitivity, which subsequently enhances endothelial function. In states of insulin resistance, impairments in phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) signaling pathways can disturb the balance between nitric oxide (NO) production and endothelin-1 secretion, resulting in endothelial dysfunction [56,57,58] (refer to Fig. 1).
Effects of GLP-1 agonists on vascular endothelium*. NOX-4 (NADPH oxidase 4), LOX-1 (lectin-like ox-LDL receptor-1), ROS (reactive oxygen species), oxLDL (oxidized LDL), VCAM-1 (vascular cell adhesion molecule-1), ICAM-1 (intercellular adhesion molecule-1), NO (nitric oxide), eNOS (endothelial nitric oxide synthase), AMPK (AMP-activated protein kinase), NLRP3 (NOD-like receptor family, pyrin domain-containing 3), PPARγ (peroxisome proliferator-activated receptor γ). The GLP-1 agonists effects demonstrated in this figure are mediated through GLP-1 receptors in the vascular endothelium. *This illustration was created using Servier Medical Art for medical illustrations
More clinical evidence can be found on the role of GLP1R agonists such as exenatide can improve vascular endothelial dysfunction in diabetic patients, as has been found in this clinical trial by Hu et al., results suggested that exenatide improved the endothelial dysfunction reflected by digital reactive hyperemia index in newly diagnosed patients with T2DM, and the effect was not inferior to single metformin [59]. In another study that examined the role of exenatide in improving coronary endothelial function in newly diagnosed type 2 diabetic patients in a randomized controlled trial, the coronary endothelial function was assessed by measuring the coronary flow velocity reserve (CFVR) which was improved significantly after 12 weeks of treatment with exenatide, CFVR was correlated inversely with hemoglobin A1c (Hb A1c) and positively with high-density lipoprotein cholesterol (HDL-C), this was also associated with a decrease in serum levels of soluble intercellular adhesion molecule-1 (sICAM-1) and soluble vascular cell adhesion molecule-1 (sVCAM-1) was remarkably decreased in the exenatide treatment group compared with the baseline and the control group emphasizing the anti- inflammatory effects of exenatide. The same study investigated the effects of exenatide in human umbilical vein endothelial cells, and it significantly increased NO production, and endothelial.
NO synthase (eNOS) phosphorylation Furthermore, exenatide reversed homocysteine-induced endothelial dysfunction by decreasing sICAM-1 and reactive oxygen species (ROS) levels and upregulating NO production and eNOS phosphorylation which further contributed the improvement of coronary endothelial function in diabetic patients [58]. Similar findings were demonstrated by liraglutide, which was also found to exert endothelial protective properties in human vascular endothelial cells (hVECs) through inhibition of stimulated plasminogen activator inhibitor type-1 (PAI-1) and vascular adhesion molecule (VAM) expression. Liraglutide treatment also increased endothelial nitric oxide synthase (eNOS) and reduced intercellular adhesion molecule-1 (ICAM-1) expression in the aortic endothelium of atherogenic mice models ApoE−/− mice model [60]. This was supported by another study in which liraglutide attenuated the TNF or high-glucose-mediated induction of PAI-1, ICAM-1, and VCAM-1 expression in human vascular endothelial cells, the proposed molecular mechanism underlying this effect may involve the regulation of NUR77 expression [61], which is a nuclear receptor and transcription factor involved in regulating gene expression related to cell growth, apoptosis, metabolism, and inflammation. It influences glucose and lipid metabolism, modulates inflammatory responses, and impacts vascular endothelial function, playing a role in cardiovascular diseases. In the context of liraglutide treatment, Nur77 regulation may help reduce endothelial cell dysfunction in type 2 diabetes and metabolic syndrome [62].
Another study compared the effects of the glp-1 receptor agonist exenatide and metformin on endothelial function in obese individuals with pre-diabetes. both exenatide and metformin had similar effects on microvascular endothelial function measured by the reactive hyperemic index (RHI), as well as levels of inflammatory and oxidative stress markers including C-reactive protein (CRP), oxidized LDL (oxLDL), and vascular cell adhesion molecule-1 (VCAM-1) levels [63].
Exenatide was found to lower the levels of high sensitivity-C-reactive protein and endothelin-1 in comparison to glargine insulin in another study [63]. Another protective mechanism exerted by exenatide on the endothelium is through its action through opening the KATP channels thus protecting against impairment in endothelium-dependent vasodilatation induced by ischemia–reperfusion (IR) injury [64].
A recent study demonstrated the protective role of GLP-1R receptors against hyperglycemia- induced endothelial to-mesenchymal transformation which is a crucial component of diabetic endothelial injury in cardiomyopathy, nephropathy, and retinopathy; as liraglutide was found to suppress neointima formation through upregulating the endothelial AMPK-nitric oxide pathway [65].
Recent studies have demonstrated new anti-inflammatory properties for GLP1-R agonists; one example is through the suppression of the NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome, which is an inflammatory complex containing three components, including NLRP3, caspase-1, and apoptosis-associated speck-like protein (ASC), resulting in the maturation and secretion of the proinflammatory cytokines Activation of the NLRP3 inflammasome plays an important role in high glucose-induced endothelial dysfunction in patients with type 2 diabetes mellitus and cardiovascular disorders. Dulaglutide was found to suppress this complex by reducing the expression of NLRP3, ASC, and cleaved caspase 1 (p10), in addition to mitigating oxidative stress by inhibiting the expression of NADPH oxidase 4 (NOX-4) and lowering the levels of reactive oxygen species (ROS) as has been demonstrated in human umbilical vein endothelial cells [66] (refer to Fig. 1).
GLP1-R agonists have been shown to provide anti-inflammatory and endothelial protective benefits through the activation of peroxisome proliferator-activated receptor γ (PPARγ). This activation inhibits intracellular serine/threonine kinases, like JNK, which in turn reduces insulin receptor substrate (IRS)-1 serine phosphorylation, a major contributor to insulin resistance, and decreases atherosclerosis. Additionally, PPARγ activation inhibits transcription factors like NF- κB, preventing atherosclerosis by suppressing the expression of cytokines and adhesion molecules in endothelial cells. This effect was observed in human umbilical vein endothelial cells treated with exendin-4 [67].
Preventing atherosclerosis
In a study that examined the effects exerted by liraglutide on the endothelium in mice models with early onset, low-burden atherosclerotic disease, liraglutide treatment in apolipoprotein E deficient (ApoE−/−) mice inhibited the progression of early onset, low-burden atherosclerotic disease and enhanced plaque stability in a partially GLP-1R-dependent manner, without significantly affecting late onset, high-burden atherosclerotic disease. Liraglutide significantly reduced lipid deposition and increased vascular smooth muscle cell and collagen content in plaques, thus enhancing plaque stability. The additional, indirect, GLP1R agonist effects on both weight and blood pressure further contributed to mitigating the progression of atherosclerosis [60].
In another study that examined the molecular mechanisms behind the effects of liraglutide on oxidized low-density lipoprotein in endothelial cells [68], liraglutide was found to suppress LDL-induced immune cell adhesion to the endothelial cells by inhibiting the expression of vascular adhesion molecules including E-selectin and VCAM-1 both of which have been related to the development of atherosclerosis [68,69,70,71] (refer to Figure one). Similar effects were exerted by Exendin-4 [72].
Liraglutide has also been found to protect against LDL-induced endothelial hyperpermeability by influencing the expression of the cellular junction protein Occludin and thus maintaining vascular permeability. It also maintains the vascular tone by enhancing the levels of endothelial nitric oxide (NO). Liraglutide also antagonizes the effects of oxidized LDL in hyperlipidemia through the ERK-5-KLF2 signals axis which is a transcription regulator with a wide range of regulatory functions in the vascular endothelium including tone, permeability, and inflammation [73]. Dulaglutide has shown similar effects, potentially preventing the atherosclerotic impacts of ox-LDL by inhibiting the suppression of KLF2 by the p53 protein in human aortic endothelial cells which is known to play a crucial role in protecting vascular endothelial cells from damage caused by ox-LDL. R Liraglutide also protects against the effects of oxidized LDL by downregulating lectin-like ox-LDL receptor-1(LOX-1) expression thus inhibiting LOX-1- mediated oxidative stress and inflammation [68].
One key component of atherosclerosis is vascular smooth muscle proliferation, previous studies have demonstrated the effects of GLP-1 agonists in reducing vascular remodeling in murine models. This effect appears to be driven by anti-inflammatory and proliferative mechanisms independent of receptor activation/signaling in the resident vascular cells [74,75,76].
GLP-1R agonists in HFpEF; review of literature
Given the pathophysiological heterogeneity of HFpEF, our understanding has been constrained by limited access to human samples and the absence of animal models that fully replicate the human HFpEF phenotype. While existing animal models, which replicate specific phenotypes by targeting individual organ systems, offer the closest approximation to the HFpEF pathology, they still fall short of fully recapitulating the human condition. Nonetheless, these animal models remain valuable research tools for elucidating the mechanistic underpinnings of the disease [77, 78].
The following is a review of the preclinical and clinical studies examining the role of GLP-1R agonists on the vascular endothelium and HFpEF:
Data from preclinical studies
In a study on non-diabetic mice models with arterial hypertension (simulating preconditions for HFpEF), Liraglutide treatment improved blood pressure and cardiac hypertrophy in angiotensin II-induced arterial hypertension independent of glucose/insulin modulation leading to the reversal of cardiac hypertrophy. Liraglutide also decreased angiotensin II-induced oxidative stress by increasing the levels of NADPH oxidase activity, increasing endothelial NO availability, and reducing immune cell infiltration by reducing the levels of F4/80 (a macrophage marker F4/80 with proinflammatory properties) expression in cardiac tissue thus reducing the overall level of oxidative stress in hypertensive heart disease. Liraglutide has been shown to exhibit antioxidant and anti-inflammatory properties that reduce eNOS uncoupling and increase NO bioavailability, NO bioavailability is thereby maintained, endothelium-mediated vasorelaxation is preserved, and vascular remodeling and fibrosis are prevented. Liraglutide also ameliorated ATII-induced rolling and infiltration of leukocytes by downregulation of central proinflammatory mediators (NF-κb, TNF-α, and IL-1β) with concomitant reduced expression of adhesion molecules (VCAM-1, ICAM-1, and P-selectin) [79] (refer to Fig. 1).
In a separate study utilizing angiotensin II-induced hypertensive mouse models to examine the effects of liraglutide on diastolic dysfunction in HFpEF, improvement in diastolic dysfunction was linked to alterations in amino acid levels within the cardiac tissue. These changes were associated with enhanced markers of protein translation, indicative of increased protein turnover and amino acid uptake, which provide protection against fibrotic remodeling and diastolic dysfunction [80].
Findings from this study were supported by another study in which the cascade of hypertension- induced vascular injury was mitigated by the use of liraglutide and a dipeptidyl peptidase-4 inhibitor to prevent the endogenous degradation of GLP-1, linagliptin; this was shown to protect against Ang II-induced injury in the heart and aorta, potentially associated with inhibition of NOX4 expression and preservation of mitochondrial integrity, reduced the protein levels of NADPH oxidase 4 (NOX4) and TGFβ1 and expression of monocyte chemoattractant protein 1, and attenuated the proliferation of myofibroblasts culminating in reduced aortic wall thickness [81].
Another study examined the effects of Liraglutide in obesity mice models, and it has demonstrated anti-inflammatory properties, treatment with Liraglutide has reduced markers of inflammation TNF-α, nuclear translocation of nuclear factor kappa B (NFκB), prevented adhesion of human monocytes to tumor necrosis factor-α-activated human endothelial cells in vitro. maintained endothelial function by correction of obesity-induced decreases in endothelial nitric oxide synthase (eNOS), reduced markers of hypertrophy and fibrosis. liraglutide protects isolated mouse neonatal cardiomyocytes and human coronary smooth muscle cells from palmitate-induced lipotoxicity. Liraglutide improved the cardiac endoplasmic reticulum stress response and also improved cardiac function by an AMP-activated protein kinase-dependent mechanism [82].
In another study performed to examine the potential cardioprotective effects in elderly hearts, a model of aging rats was used in which the effects were tested in aged rats. liraglutide treatment significantly mitigated the prolongation of QRS duration and elevated both systolic and diastolic blood pressure, accompanied by a restoration of plasma oxidant and antioxidant statuses. The prolonged action potential durations and depolarized membrane potentials in isolated cardiomyocytes from aged rats were normalized through the recovery of K+ channel currents with liraglutide treatment. Additionally, alterations in Ca2+ regulation, including leaky ryanodine receptors (RyR2), were ameliorated via the recovery of Na+/Ca2+ exchanger currents. Direct liraglutide treatment of isolated aged rat cardiomyocytes restored the depolarized mitochondrial membrane potential, reduced reactive oxygen and nitrogen species (ROS and RNS), and normalized cytosolic Na+ levels, despite the Na+ channel currents remaining unaffected by aging. Notably, liraglutide treatment significantly inhibited activated sodium-glucose co- transporter-2 (SGLT2) restored depressed insulin receptor substrate 1 (IRS1), and increased protein kinase G (PKG) levels. The recovery of the phospho-endothelial nitric oxide synthase (pNOS3) to NOS3 protein ratio in liraglutide-treated cardiomyocytes suggests that liraglutide- associated inhibition of oxidative stress-induced injury occurs via the IRS1-eNOS-PKG pathway in the aging heart [83].
To date, preclinical HFpEF models have primarily utilized single perturbations to replicate the human HFpEF phenotype. However, these models have faced challenges in reproducing the wide range of symptoms observed in human HFpEF, since usually one organ system is targeted and the pathology is replicated in a relatively acute setting in comparison to the natural course of HFpEF in humans.
A recent study investigated the effects of GLP-1R agonists in a multi-hit mouse model in which the combination of a high-fat diet and angiotensin II-induced a cardiometabolic HFpEF phenotype that resembles the human HFpEF phenotype to a large extent characterized by obesity, impaired glucose handling, and metabolic dysregulation accompanied by inflammation. This multifactorial approach resulted in the manifestation of typical clinical HFpEF features, including ventricular hypertrophy, increased collagen deposition and fibrosis, elevated blood pressure, and diastolic dysfunction; as well as left atrial enlargement, pulmonary congestion, and raised natriuretic peptides all features of clinical HFpEF. Liraglutide treatment in this model has alleviated cardiometabolic dysregulation and enhanced cardiac function, resulting in reduced cardiac hypertrophy and fibrosis, as well as diminished atrial weight, natriuretic peptide levels, and lung congestion [84].
Study | Animal model | Drugs used | Results | Mechanism |
---|---|---|---|---|
Helmstädter et al. [60] | Hypertensive mouse model murine, nondiabetic model of angiotensin II-induced arterial hypertension Wild-type (C57BL/6 J), global (Glp1r−/−), as well as endothelial (Glp1rflox/ floxxCdh5cre) and myeloid cell–specific knockout mice (Glp1rflox/ floxxLysMcre) of the GLP-1R | Liraglutide | Improvement in blood pressure and cardiac hypertrophy Reduction in cardiac and whole blood oxidative stress Improvement ATII-induced endothelial dysfunction and vascular fibrosis Amelioration of ATII-induced inflammation of the vasculature | Suppression of inflammation and NADPH oxidase activity Reduction of vascular oxidative stress and recoupling of eNOS Prevention of ly6g−ly6c+- and ly6g+ ly6c+ cell infiltration to the vessel wall |
Rutledge et al. [80] | Hypertensive mouse model mouse model of angiotensin II-mediated diastolic dysfunction | Liraglutide | Protection against AngII-mediated diastolic dysfunction | Promoting amino acid uptake and protein turnover in the heart |
Banks et al. [81] | Hypertensive model Sprague–Dawley rat model of Ang II infusion | Liraglutide and dipeptidyl peptides-4 inhibitor linagliptin | Protection against Ang II-induced injury in the heart and aorta A significant reduction in cardiac fibrosis.Reduction in aortic wall thickness and fibrotic areaReduction in mean blood pressure | Downregulation of the expression of NOX4 and intercellular adhesion molecule 1 Enhanced endothelial NOS expression Reduction in the protein levels of NOX4 and TGFβ1 and Reduced expression of monocyte chemoattractant protein 1, Attenuation in the proliferation of myofibroblasts Decrease in the |
Noyan-Ashraf et al. [82] | Obesity model Obesity mice model C57Bl6 mice were placed on a 45% high-fat diet or a regular chow diet | Liraglutide | Reversed insulin resistance, cardiac tumor necrosis factor-α expression, nuclear factor kappa B translocation, Reversed obesity-induced perturbations in cardiac endothelial nitric oxide synthase, connexin-43, and markers of hypertrophy and fibrosis Reduction in cardiac endoplasmic reticulum stress response Improved cardiac function | AMP-activated protein kinase– dependent mechanism Prevention of palmitate- induced lipotoxicity in isolated mouse cardiomyocytes and primary human coronary smooth muscle cells Preventing the adhesion of human monocytes to tumor necrosis factor-α– activated human endothelial cells in vitro |
Durak and Turan [83] | Aged rats | Liraglutide | Reduction in the prolongation of QRS duration Increase in both systolic and diastolic blood pressure Recovery in plasma oxidant and antioxidant statuses At the cellular level, normalizing the action potential (AP) parameters, ionic currents, and Ca2+ regulation in freshly isolated | Recoveries in K+ channel currents and Na+/Ca2+ exchanger currents Inhibition of activated sodium-glucose co-transporter-2 (SGLT2) Recoveries in the depressed insulin receptor substrate 1 (IRS1) and increased protein kinase G (PKG) Recovery in the ratio of phosphor endothelial nitric oxide (pNOS3) level to NOS3 |
Withaar et al. [84] | Multi-hit mouse model aged female C57BL/6 J mice were administered a high-fat diet and angiotensin II. The combined treatment of high-fat diet and angiotensin II (HFD + ANGII) induced a cardiometabolic phenotype consistent with heart failure with preserved ejection fraction (HFpEF) | Liraglutide and Dapagliflozin | Treatment with liraglutide ameliorated cardiometabolic dysregulation Enhancing cardiac function by reducing cardiac hypertrophy and myocardial fibrosis Attenuated atrial weight, natriuretic peptide levels, and lung congestion Dapagliflozin treatment improved glucose metabolism but had only mild effects on the HFpEF | Liraglutide alleviated left ventricular load, enhanced diastolic function, and exhibited anti- atherosclerotic properties via an anti- inflammatory mechanism The effectiveness of SGLT2 inhibitors can be attributed, at least in part, to their natriuretic effects |
Data from clinical studies
In an observational study involving 20 diabetic subjects receiving background metformin therapy, exenatide treatment over 16 weeks resulted in improved flow-mediated vasodilation of the brachial artery following 5 min of forearm ischemia, as assessed by ultrasound echocardiography, compared to patients treated with glimepiride [85].
In another study, exenatide's impact on postprandial endothelial function (EF) in type 2 diabetes and its mechanisms of vasodilation were investigated. Two crossover studies were conducted: participants with type 2 diabetes received exenatide vs. controls, and their EF, glucose, and lipid responses to meals were measured. Ex vivo studies were also conducted on human subcutaneous adipose tissue arterioles and endothelial cells. Results showed that subcutaneous exenatide increased postprandial EF independently of glucose and triglyceride reductions. Intravenous exenatide improved fasting EF, an effect nullified by exendin-9 (GLP-1R antagonist). Exenatide activated eNOS and NO production in endothelial cells, induced dose-dependent vasorelaxation, and mitigated high-glucose or lipid-induced endothelial dysfunction in arterioles ex vivo, effects which were reduced by AMPK (AMP-activated protein kinase) inhibition [86].
A recent metanalysis [87] has assessed the impact of oral antidiabetic drugs (OADs) and GLP-1 receptor agonists on left ventricular diastolic function in patients with type 2 diabetes, the findings indicated that, compared to placebo and OADs, only liraglutide significantly improved left ventricular diastolic function. This was supported by a recent metanalysis in which, twenty- two randomized controlled trials involving 61,412 patients who were either T2DM patients with or without cardiovascular disease or patients with cardiovascular disease alone, were included in the meta-analysis, and results have shown the diastolic function according to echocardiography was significantly improved by GLP-1R agonists [88].
In a 2022 metanalysis [89] of previous randomized controlled trials, 20 papers from 19 randomized placebo-controlled trials were included, comprising a total sample size of 2062.
patients on the potential effects of GLP1R agonists on the cardiovascular system. In patients with type 2 DM, GLP-1R agonists have demonstrated a reduction in circumferential strain and improvement in markers of diastolic function, a key component of diabetic cardiomyopathy and the risk of developing heart failure; this effect was plausibly exerted through the diuretic and natriuretic effect of GLP-1R agonists reducing preload. In heart failure patients, there was a lack of changes in cardiac imaging parameters correlates with the lack of impact on outcomes among patients with pre-existing heart failure in major GLP-1 receptor agonist trials. Findings were supported by a recently updated metanalysis of 15 trials involving 898 patients were included in this analysis, GLP-1R agonists were found to have a positive impact on left ventricle diastolic function, hypertrophy, and exercise capacity [90].
Data from the Semaglutide Treatment Effect in People with Obesity (STEP) trials, which examined the weight loss effects of semaglutide, demonstrated a 30–40% reduction in C-reactive protein levels, indicating a significant anti-inflammatory effect. Recently, the STEP-HFpEF trial enrolled 529 non-diabetic patients with HFpEF and obesity, who were randomly assigned to receive either once-weekly semaglutide (2.4 mg) or placebo for 52 weeks. This trial observed statistically significant improvements in quality of life scores, weight loss, 6-min walk distance, C-reactive protein levels, and N-terminal pro–B-type natriuretic peptide levels. These findings were supported by another study in which liraglutide improved diastolic function in diabetic patients [91, 92].
Additionally, the Semaglutide Effects on Cardiovascular Outcomes in People with Overweight or Obesity trial demonstrated that semaglutide consistently reduced major cardiovascular event endpoints by approximately 20% compared to placebo over an approximately 3-year follow-up period in patients with overweight or obesity and cardiovascular disease (Table 1), but without diabetes [93].
The currently ongoing SUMMIT trial (A Study of Tirzepatide in Participants With Heart Failure With Preserved Ejection Fraction and Obesity; NCT04847557) seeks to evaluate the effect of tirzepatide (is a dual agonist for the glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptors) over 52 weeks in 700 patients with heart failure with preserved ejection fraction (HFpEF) and obesity.
A limitation of these studies is the absence of stratification of heart failure patients based on ejection fraction, therefore additional clinical research specifically investigating the effects of GLP-1 receptor agonists in patients with heart failure with preserved ejection fraction (HFpEF) is warranted [94].
Conclusion
Glucagon-like peptide-1 receptor agonists (GLP-1RAs) represent a promising therapeutic avenue for heart failure with preserved ejection fraction (HFpEF). By targeting endothelial dysfunction—a central component of HFpEF pathophysiology—GLP-1RAs offer cardioprotective benefits that extend beyond their established metabolic effects. These include improving vascular function, reducing systemic inflammation, enhancing coronary microvascular function, and mitigating comorbid conditions like metabolic dysfunction-associated steatotic liver disease (MASLD) and chronic kidney disease (CKD). Emerging clinical evidence, such as the STEP-HFpEF and FLOW trials, underscores the efficacy of GLP-1RAs in alleviating symptoms, improving quality of life, and slowing disease progression in HFpEF patients.
Limitations
Despite their promise, several limitations exist. Current evidence is derived primarily from preclinical studies and a limited number of clinical trials, which may not fully represent the diverse HFpEF population. Many trials lack stratification based on ejection fraction, hindering the ability to determine the differential effects of GLP-1RAs in various heart failure subtypes. Furthermore, the precise mechanisms by which GLP-1RAs confer their cardioprotective effects remain incompletely understood, necessitating further investigation. Lastly, questions about long-term safety, efficacy, and optimal patient selection criteria remain unanswered.
Areas of future research
Future studies should focus on addressing these gaps. Large-scale, randomized controlled trials with robust stratification of HFpEF phenotypes are needed to confirm the efficacy and safety of GLP-1RAs in this population. Mechanistic studies should aim to elucidate how GLP-1RAs influence endothelial function, inflammation, and myocardial remodeling in HFpEF. Exploring potential synergistic effects of GLP-1RAs with other established therapies, such as SGLT2 inhibitors, could unlock combination approaches for improved outcomes. Finally, economic evaluations are necessary to assess cost-effectiveness and accessibility in clinical practice.
As our understanding of HFpEF evolves, GLP-1RAs have the potential to become a cornerstone of therapy, addressing the multifactorial nature of this condition and improving outcomes in a challenging patient population.
Availability of data and materials
No datasets were generated or analysed during the current study.
References
Bozkurt B, Ahmad T, Alexander KM, Baker WL, Bosak K, Breathett K, et al. Heart failure epidemiology and outcomes statistics: a report of the heart failure society of America. J Card Fail. 2023;29(10):1412–51.
Adamczak DM, Oduah MT, Kiebalo T, Nartowicz S, Bęben M, Pochylski M, Ciepłucha A, Gwizdała A, Lesiak M, Straburzyńska-Migaj E. Heart failure with preserved ejection fraction—a concise review. Curr Cardiol Rep. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11886-020-01349-3.
Heinzel FR, Shah SJ. The future of heart failure with preserved ejection fraction. Herz. 2022;47:308–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00059-022-05124-8.
Owan TE, Hodge DO, Herges RM, Jacobsen SJ, Roger VL, Redfield MM. Trends in prevalence and outcome of heart failure with preserved ejection fraction. NEJM. 2006. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa052256.
Stoicescu L, Crisan D, Morgovan C, Avram L, Ghibu S. Heart failure with preserved ejection fraction: the pathophysiological mechanisms behind the clinical phenotypes and the therapeutic approach. Int J Mol Sci. 2024;25(2):794.
Abebe TB, Gebreyohannes EA, Tefera YG, Abegaz TM. Patients with HFpEF and HFrEF have different clinical characteristics but similar prognosis: a retrospective cohort study. BMC Cardiovasc Disord. 2016;16(1):232.
Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Böhm M, Brunner-La Rocca HP, Choi DJ, Chopra V, Chuquiure-Valenzuela E, Giannetti N. Empagliflozin in heart failure with a preserved ejection fraction. NEJM. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa2107038.
Solomon SD, McMurray JJV, Claggett B, de Boer RA, DeMets D, Hernandez AF, et al. Dapagliflozin in heart failure with mildly reduced or preserved ejection fraction. N Engl J Med. 2022;387(12):1089–98.
Kosiborod MN, Verma S, Borlaug BA, Butler J, Davies MJ, Jon Jensen T, Rasmussen S, Erlang Marstrand P, Petrie MC, Shah SJ, Ito H. Effects of semaglutide on symptoms, function, and quality of life in patients with heart failure with preserved ejection fraction and obesity: a prespecified analysis of the STEP-HFpEF trial. Circulation. 2024;149:204–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/CIRCULATIONAHA.123.067505.
Capone F, Nambiar N, Schiattarella GG. Beyond weight loss: the emerging role of incretin-based treatments in cardiometabolic HFpEF. Curr Opin Cardiol. 2024;39:148–53.
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(4):263–71.
Brutsaert DL. Cardiac endothelial-myocardial signaling: its role in cardiac growth, contractile performance, and rhythmicity. Physiol Rev. 2003;83(1):59–115.
Qi XL, Stewart DJ, Gosselin H, Azad A, Picard P, Andries L, et al. Improvement of endocardial and vascular endothelial function on myocardial performance by captopril treatment in postinfarct rat hearts. Circulation. 1999;100(12):1338–45.
Hemodynamic stresses induce endothelial dysfunction and remodeling of pulmonary artery in experimental compensated heart failure—PubMed. Available from: https://pubmed.ncbi.nlm.nih.gov/10851216/
Chu GX, Ling Q, Guo ZG. Effects of endocardial endothelium in myocardial mechanics of hypertrophied myocardium of rats. Zhongguo Yao Li Xue Bao. 1995;16(4):352–6.
The pathomorphological alterations of endocardial endothelium in experimental diabetes and diabetes associated with hyperlipidemia—PubMed [cited 2024 Jul 24]. Available from: https://pubmed.ncbi.nlm.nih.gov/8777284/
Gevaert AB, Shakeri H, Leloup AJ, Van Hove CE, De Meyer GR, Vrints CJ, et al. Endothelial senescence contributes to heart failure with preserved ejection fraction in an aging mouse model. Circ Heart Fail. 2017;10(6):e003806.
Lam CSP, Brutsaert DL. Endothelial dysfunction. J Am Coll Cardiol. 2012;60(18):1787–9.
Impaired endothelium-dependent regulation of ventricular relaxation in pressure-overload cardiac hypertrophy—PubMed [cited 2024 Jul 24]. Available from: https://pubmed.ncbi.nlm.nih.gov/10769288/
Cardiac structure and ventricular-vascular function in persons with heart failure and preserved ejection fraction from Olmsted County, Minnesota—PubMed. [cited 2024 Jul 24]. Available from: https://pubmed.ncbi.nlm.nih.gov/17404159/
Lam CS, Roger VL, Rodeheffer RJ, Borlaug BA, Enders FT, Redfield MM. Pulmonary hypertension in heart failure with preserved ejection fraction: a community-based study. J Am Coll Cardiol. 2008;53:1119–26. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jacc.2008.11.051.
McEniery CM, Wallace S, Mackenzie IS, McDonnell B, Yasmin null, Newby DE, et al. Endothelial function is associated with pulse pressure, pulse wave velocity, and augmentation index in healthy humans. Hypertens Dallas Tex 1979. 2006;48(4):602–8.
Akiyama E, Sugiyama S, Matsuzawa Y, Konishi M, Suzuki H, Nozaki T, Ohba K, Matsubara J, Maeda H, Horibata Y, Sakamoto K. Incremental prognostic significance of peripheral endothelial dysfunction in patients with heart failure with normal left ventricular ejection fraction. J Am Coll Cardiol. 2012. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jacc.2012.07.036.
Sucato V, Evola S, Novo G, Sansone A, Quagliana A, Andolina G, et al. Angiographic evaluation of coronary microvascular dysfunction in patients with heart failure and preserved ejection fraction. Microcirc N Y N. 1994;22(7):528–33.
Coronary microvascular rarefaction and myocardial fibrosis in heart failure with preserved ejection fraction. PubMed. Available from: https://pubmed.ncbi.nlm.nih.gov/25552356/
Tschope C, Bock CT, Kasner M, Noutsias M, Westermann D, Schwimmbeck PL, Pauschinger M, Poller WC, Kuhl U, Kandolf R, Schultheiss HP. High prevalence of cardiac parvovirus B19 infection in patients with isolated left ventricular diastolic dysfunction. Circulation. 2005. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/01.CIR.0000155615.68924.B3.
Borlaug BA, Olson TP, Lam CS, Flood KS, Lerman A, Johnson BD, Redfield MM. Global cardiovascular reserve dysfunction in heart failure with preserved ejection fraction. J Am Coll Cardiol. 2010. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jacc.2010.03.077.
Rush CJ, Berry C, Oldroyd KG, Rocchiccioli JP, Lindsay MM, Touyz RM, et al. Prevalence of coronary artery disease and coronary microvascular dysfunction in patients with heart failure with preserved ejection fraction. JAMA Cardiol. 2021;6(10):1130–43.
Lin X, Wu G, Wang S, Huang J. The prevalence of coronary microvascular dysfunction (CMD) in heart failure with preserved ejection fraction (HFpEF): a systematic review and meta-analysis. Heart Fail Rev. 2023;29:405–16.
Campbell JE, Drucker DJ. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 2013;17(6):819–37.
Berndt J, Ooi SL, Pak SC. What is the mechanism driving the reduction of cardiovascular events from glucagon-like peptide-1 receptor agonists?-A mini review. Molecules. 2021;26(16):4822.
Bullock BP, Heller RS, Habener JF. Tissue distribution of messenger ribonucleic acid encoding the rat glucagon-like peptide-1 receptor. Endocrinology. 1996;137(7):2968–78.
McLean BA, Wong CK, Campbell JE, Hodson DJ, Trapp S, Drucker DJ. Revisiting the complexity of GLP-1 action from sites of synthesis to receptor activation. Endocr Rev. 2020;42(2):101–32.
Fatima A, Maryam A, Nadeem MS, Asmat KN. GLP-1 receptor agonists: a new frontier in combating metabolic liver disease complications. Aliment Pharmacol Ther. 2024;60:1499–500.
Xanthopoulos A, Starling RC, Kitai T, Triposkiadis F. Heart failure and liver disease: cardiohepatic interactions. JACC Heart Fail. 2019;7(2):87–97.
Inciardi RM, Mantovani A, Targher G. Non-alcoholic fatty liver disease as an emerging risk factor for heart failure. Curr Heart Fail Rep. 2023;20(4):308–19.
Kucsera D, Ruppert M, Sayour NV, Tóth VE, Kovács T, Hegedűs ZI, et al. NASH triggers cardiometabolic HFpEF in aging mice. GeroScience. 2024;46(5):4517–31.
Zafar Y, Rashid AM, Siddiqi AK, Ellahi A, Ahmed A, Hussain H, et al. Effect of novel glucose lowering agents on non-alcoholic fatty liver disease: a systematic review and meta-analysis. Clin Res Hepatol Gastroenterol. 2022;46(7):101970.
Badmus OO, Hinds TD, Stec DE. Mechanisms linking metabolic-associated fatty liver disease (MAFLD) to cardiovascular disease. Curr Hypertens Rep. 2023;25(8):151–62.
Wood M, Kennedy AG, Khan S, Hitt JR, Davis K, Reddy SS, et al. Impact of GLP-1 receptor agonist use in patients with steatotic liver disease and type 2 diabetes: a retrospective cohort study. J Pharm Pract. 2024;37(6):1297–302.
Kanwal F, Kramer JR, Li L, Yang YX, Cao Y, Yu X, et al. GLP-1 receptor agonists and risk for cirrhosis and related complications in patients with metabolic dysfunction-associated steatotic liver disease. JAMA Intern Med. 2024;16: e244661.
Mantovani A, Byrne CD, Benfari G, Bonapace S, Simon TG, Targher G. Risk of heart failure in patients with nonalcoholic fatty liver disease: JACC review topic of the week. J Am Coll Cardiol. 2022;79(2):180–91.
Yang Z, Tian R, Zhang XJ, Cai J, She ZG, Li H. Effects of treatment of non-alcoholic fatty liver disease on heart failure with preserved ejection fraction. Front Cardiovasc Med. 2023;12(9):1120085.
Arriola-Montenegro J, Beas R, Cerna-Viacava R, Chaponan-Lavalle A, Hernandez Randich K, Chambergo-Michilot D, et al. Therapies for patients with coexisting heart failure with reduced ejection fraction and non-alcoholic fatty liver disease. World J Cardiol. 2023;15(7):328–41.
Gad AI, Ibrahim NF, Almadani N, Mahfouz R, Nofal HA, El-Rafey DS, et al. Therapeutic effects of semaglutide on nonalcoholic fatty liver disease with type 2 diabetes mellitus and obesity: an open-label controlled trial. Diseases. 2024;12(8):186.
Shaman AM, Bain SC, Bakris GL, Buse JB, Idorn T, Mahaffey KW, et al. Effect of the glucagon-like peptide-1 receptor agonists semaglutide and liraglutide on kidney outcomes in patients with type 2 diabetes: pooled analysis of SUSTAIN 6 and LEADER. Circulation. 2022;145(8):575–85.
Taal MW, Selby NM. Glucagon-like peptide-1 receptor agonists: new evidence of kidney and cardiovascular protection from the FLOW and SELECT Trials. Am J Kidney Dis. 2024. Available from: https://www.ajkd.org/article/S0272-6386(24)00975-2/fulltext
Kanbay M, Copur S, Guldan M, Ozbek L, Mallamaci F, Zoccali C. Glucagon and glucagon-like peptide-1 dual agonist therapy: a possible future towards fatty kidney disease. Eur J Clin Invest. 2025;55(1):e14330.
Yen FS, Hsu CC, Wei JCC, Tsai FJ, Huang Y, Yu TS, et al. Glucagon-like peptide-1 receptor agonists may benefit cardiopulmonary outcomes in patients with COPD. Thorax. 2024;79(11):1017–23.
Albogami Y, Cusi K, Daniels MJ, Wei YJJ, Winterstein AG. Glucagon-like peptide 1 receptor agonists and chronic lower respiratory disease exacerbations among patients with type 2 diabetes. Diabetes Care. 2021;44(6):1344–52.
Wang W, Mei A, Qian H, Li D, Xu H, Chen J, et al. The role of glucagon-like peptide-1 receptor agonists in chronic obstructive pulmonary disease. Int J Chronic Obstr Pulm Dis. 2023;18:129–37.
Kantreva K, Katsaounou P, Saltiki K, Trakada G, Ntali G, Stratigou T, et al. The possible effect of anti-diabetic agents GLP-1RA and SGLT-2i on the respiratory system function. Endocrine. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12020-024-04033-6.
Fandiño J, Toba L, González-Matías LC, Diz-Chaves Y, Mallo F. GLP-1 receptor agonist ameliorates experimental lung fibrosis. Sci Rep. 2020;10(1):18091.
Viby NE, Isidor MS, Buggeskov KB, Poulsen SS, Hansen JB, Kissow H. Glucagon-like peptide-1 (GLP-1) reduces mortality and improves lung function in a model of experimental obstructive lung disease in female mice. Endocrinology. 2013;154(12):4503–11.
Balk-Møller E, Windeløv JA, Svendsen B, Hunt J, Ghiasi SM, Sørensen CM, et al. Glucagon-like peptide 1 and atrial natriuretic peptide in a female mouse model of obstructive pulmonary disease. J Endocr Soc. 2020;4(1):034.
Muniyappa R, Sowers JR. Role of insulin resistance in endothelial dysfunction. Rev Endocr Metab Disord. 2013;14(1):5–12.
Del Turco S, Gaggini M, Daniele G, Basta G, Folli F, Sicari R, et al. Insulin resistance and endothelial dysfunction: a mutual relationship in cardiometabolic risk. Curr Pharm Des. 2013;19(13):2420–31.
Wei R, Ma S, Wang C, Ke J, Yang J, Li W, et al. Exenatide exerts direct protective effects on endothelial cells through the AMPK/Akt/eNOS pathway in a GLP-1 receptor-dependent manner. Am J Physiol Endocrinol Metab. 2016;310(11):E947-957.
Hu Y, Liu J, Wang G, Xu Y. The effects of exenatide and metformin on endothelial function in newly diagnosed type 2 diabetes mellitus patients: a case-control study. Diabetes Ther. 2018;9(3):1295–305.
Gaspari T, Welungoda I, Widdop RE, Simpson RW, Dear AE. The GLP-1 receptor agonist liraglutide inhibits progression of vascular disease via effects on atherogenesis, plaque stability and endothelial function in an ApoE−/− mouse model. Diabetes Vasc Dis Res. 2013;10(4):353–60.
Liu H, Dear AE, Knudsen LB, Simpson RW. A long-acting glucagon-like peptide-1 analogue attenuates induction of plasminogen activator inhibitor type-1 and vascular adhesion molecules. J Endocrinol. 2009;201(1):59–66.
Maxwell MA, Muscat GEO. The NR4A subgroup: immediate early response genes with pleiotropic physiological roles. Nucl Recept Signal. 2006;4:e002.
Gurkan E, Tarkun I, Sahin T, Cetinarslan B, Canturk Z. Evaluation of exenatide versus insulin glargine for the impact on endothelial functions and cardiovascular risk markers. Diabetes Res Clin Pract. 2014;106(3):567–75.
Preventive effects of exenatide on endothelial dysfunction induced by ischemia-reperfusion injury via KATP channels—PubMed. Available from: https://pubmed.ncbi.nlm.nih.gov/22155457/
Tsai TH, Lee CH, Cheng CI, Fang YN, Chung SY, Chen SM, et al. Liraglutide inhibits endothelial-to-mesenchymal transition and attenuates neointima formation after endovascular injury in streptozotocin-induced diabetic mice. Cells. 2019;8(6):589.
Luo X, Hu Y, He S, Ye Q, Lv Z, Liu J, et al. Dulaglutide inhibits high glucose- induced endothelial dysfunction and NLRP3 inflammasome activation. Arch Biochem Biophys. 2019;15(671):203–9.
Onuma H, Inukai K, Kitahara A, Moriya R, Nishida S, Tanaka T, et al. The glucagon-like peptide 1 receptor agonist enhances intrinsic peroxisome proliferator-activated receptor γ activity in endothelial cells. Biochem Biophys Res Commun. 2014;451(2):339–44.
Chang W, Zhu F, Zheng H, Zhou Z, Miao P, Zhao L, et al. Glucagon-like peptide-1 receptor agonist dulaglutide prevents ox-LDL-induced adhesion of monocytes to human endothelial cells: An implication in the treatment of atherosclerosis. Mol Immunol. 2019;116:73–9.
Galkina E, Ley K. Vascular adhesion molecules in atherosclerosis. Arterioscler Thromb Vasc Biol. 2007;27(11):2292–301.
Cybulsky MI, Iiyama K, Li H, Zhu S, Chen M, Iiyama M, et al. A major role for VCAM-1, but not ICAM-1, in early atherosclerosis. J Clin Invest. 2001;107(10):1255–62.
Di Tomo P, Lanuti P, Di Pietro N, Baldassarre MPA, Marchisio M, Pandolfi A, et al. Liraglutide mitigates TNF-α induced pro-atherogenic changes and microvesicle release in HUVEC from diabetic women. Diabetes Metab Res Rev. 2017;33(8):e2925.
Arakawa M, Mita T, Azuma K, Ebato C, Goto H, Nomiyama T, et al. Inhibition of monocyte adhesion to endothelial cells and attenuation of atherosclerotic lesion by a glucagon-like peptide-1 receptor agonist, exendin-4. Diabetes. 2010;59(4):1030–7.
Yue W, Li Y, Ou D, Yang Q. The GLP-1 receptor agonist liraglutide protects against oxidized LDL-induced endothelial inflammation and dysfunction via KLF2. IUBMB Life. 2019;71(9):1347–54.
Jensen DM, Skovsted GF, Bonde MFB, Bentzon JF, Rolin B, Franck G, et al. Semaglutide treatment attenuates vessel remodelling in ApoE-/- mice following vascular injury and blood flow perturbation. Atheroscler Plus. 2022;49:32–41.
Eriksson L, Saxelin R, Röhl S, Roy J, Caidahl K, Nyström T, et al. Glucagon-like peptide-1 receptor activation does not affect re-endothelialization but reduces intimal hyperplasia via direct effects on smooth muscle cells in a nondiabetic model of arterial injury. J Vasc Res. 2015;52(1):41–52.
Goto H, Nomiyama T, Mita T, Yasunari E, Azuma K, Komiya K, et al. Exendin-4, a glucagon-like peptide-1 receptor agonist, reduces intimal thickening after vascular injury. Biochem Biophys Res Commun. 2011;405(1):79–84.
Conceição G, Heinonen I, Lourenço AP, Duncker DJ, Falcão-Pires I. Animal models of heart failure with preserved ejection fraction. Neth Heart J Mon J Neth Soc Cardiol Neth Heart Found. 2016;24(4):275–86.
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(6):770–89.
Helmstadter J, Frenis K, Filippou K, Grill A, Dib M, Kalinovic S, et al. Endothelial GLP-1 (Glucagon-Like Peptide-1) receptor mediates cardiovascular protection by liraglutide in mice with experimental arterial hypertension. Arterioscler Thromb Vasc Biol. 2020;40(1):145–58.
Rutledge C, Enriquez A, Redding K, Lopez M, Mullett S, Gelhaus SL, et al. Liraglutide protects against diastolic dysfunction and improves ventricular protein translation. Cardiovasc Drugs Ther. 2023.
Banks TE, Rajapaksha M, Zhang LH, Bai F, Wang NP, Zhao ZQ. Suppression of angiotensin II-activated NOX4/NADPH oxidase and mitochondrial dysfunction by preserving glucagon-like peptide-1 attenuates myocardial fibrosis and hypertension. Eur J Pharmacol. 2022;15(927): 175048.
Noyan-Ashraf MH, Shikatani EA, Schuiki I, Mukovozov I, Wu J, Li RK, et al. A glucagon-like peptide-1 analog reverses the molecular pathology and cardiac dysfunction of a mouse model of obesity. Circulation. 2013;127(1):74–85.
Durak A, Turan B. Liraglutide provides cardioprotection through the recovery of mitochondrial dysfunction and oxidative stress in aging hearts. J Physiol Biochem. 2023;79(2):297–311.
Withaar C, Meems LMG, Markousis-Mavrogenis G, Boogerd CJ, Sillje HHW, Schouten EM, et al. 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. 2021;117(9):2108–24.
Irace C, De Luca S, Shehaj E, Carallo C, Loprete A, Scavelli F, et al. Exenatide improves endothelial function assessed by flow mediated dilation technique in subjects with type 2 diabetes: results from an observational research. Diabetes Vasc Dis Res. 2013;10(1):72–7.
Koska J, Sands M, Burciu C, D’Souza KM, Raravikar K, Liu J, et al. Exenatide Protects against glucose- and lipid-induced endothelial dysfunction: evidence for direct vasodilation effect of GLP-1 receptor agonists in humans. Diabetes. 2015;64(7):2624–35.
Ida S, Kaneko R, Imataka K, Okubo K, Shirakura Y, Azuma K, et al. Effects of oral antidiabetic drugs and glucagon-like peptide-1 receptor agonists on left ventricular diastolic function in patients with type 2 diabetes mellitus: a systematic review and network meta-analysis. Heart Fail Rev. 2021;26(5):1151–8.
Huixing L, Di F, Daoquan P. Effect of glucagon-like peptide-1 receptor agonists on prognosis of heart failure and cardiac function: a systematic review and meta-analysis of randomized controlled trials. Clin Ther. 2023;45(1):17–30.
Wong SY, Lee A, Sia AHJ, Wo YJ, Teo YH, Teo YN, et al. Effects of glucagon-like peptide-1 receptor agonist (GLP-1RA) on cardiac structure and function: a systematic review and meta-analysis of randomized-controlled trials. Cardiovasc Drugs Ther. 2022;38:371–89.
Zhang Y, Yang D, Jia Q, Yan J, An F. The effect of glucagon-like peptide-1 receptor agonists on cardiac function and structure in patients with or without type 2 diabetes mellitus: an updated systematic review and meta-analysis. Diabetes Obes Metab. 2024;26:2401–11. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/dom.15557.
Yagi K, Imamura T, Tada H, Chujo D, Liu J, Shima Y, et al. Diastolic cardiac function improvement by liraglutide is mainly body weight reduction dependent but independently contributes to B-type natriuretic peptide reduction in patients with type 2 diabetes with preserved ejection fraction. J Diabetes Res. 2021;2021:8838026.
Bizino MB, Jazet IM, Westenberg JJM, van Eyk HJ, Paiman EHM, Smit JWA, et al. Effect of liraglutide on cardiac function in patients with type 2 diabetes mellitus: randomized placebo-controlled trial. Cardiovasc Diabetol. 2019;18(1):55.
Temporelli PL. Role of glucagon-like peptide-1 agonists in obesity and heart failure with preserved ejection fraction. Eur Heart J Suppl. 2024;26(Supplement_1):127–530.
Ferreira JP, Sharma A, Butler J, Packer M, Zannad F, Vasques-Nóvoa F, et al. Glucagon-like peptide-1 receptor agonists across the spectrum of heart failure. J Clin Endocrinol Metab. 2023;109(1):4–9.
Acknowledgements
None to given at this time.
Funding
This study was not supported by any sponsor or funder.
Author information
Authors and Affiliations
Contributions
Darshan Hullon—Concept, Writer, editor Ghasaq K Subeh—Concept, writer, editor Yelizaveta Volkova—Writer Karolina Janiec—Writer editor Adam Trach—Writer Ruslan Mnevets—Writer.
Corresponding authors
Ethics declarations
Ethical approval and consent to participate
Not applicable. Our study is a review and does not involve patients.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Hullon, D., Subeh, G.K., Volkova, Y. et al. The role of glucagon-like peptide-1 receptor (GLP-1R) agonists in enhancing endothelial function: a potential avenue for improving heart failure with preserved ejection fraction (HFpEF). Cardiovasc Diabetol 24, 70 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12933-025-02607-w
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12933-025-02607-w