- Review
- Open access
- Published:
Metabolic rewiring and inter-organ crosstalk in diabetic HFpEF
Cardiovascular Diabetology volume 24, Article number: 155 (2025)
Abstract
Heart failure with preserved ejection fraction (HFpEF) represents a significant and growing clinical challenge. Initially, for an extended period, HFpEF was simply considered as a subset of heart failure, manifesting as haemodynamic disorders such as hypertension, myocardial hypertrophy, and diastolic dysfunction. However, the rising prevalence of obesity and diabetes has reshaped the HFpEF phenotype, with nearly 45% of cases coexisting with diabetes. Currently, it is recognized as a multi-system disorder that involves the heart, liver, kidneys, skeletal muscle, adipose tissue, along with immune and inflammatory signaling pathways. In this review, we summarize the landscape of metabolic rewiring and the crosstalk between the heart and other organs/systems (e.g., adipose, gut, liver and hematopoiesis system) in diabetic HFpEF for the first instance. A diverse array of metabolites and cytokines play pivotal roles in this intricate crosstalk process, with metabolic rewiring, chronic inflammatory responses, immune dysregulation, endothelial dysfunction, and myocardial fibrosis identified as the central mechanisms at the heart of this complex interplay. The liver-heart axis links nonalcoholic steatohepatitis and HFpEF through shared lipid accumulation, inflammation, and fibrosis pathways, while the gut-heart axis involves dysbiosis-driven metabolites (e.g., trimethylamine N-oxide, indole-3-propionic acid and short-chain fatty acids) impacting cardiac function and inflammation. Adipose-heart crosstalk highlights epicardial adipose tissue as a source of local inflammation and mechanical stress, whereas the hematopoietic system contributes via immune cell activation and cytokine release. We contend that, based on the viewpoints expounded in this review, breaking this inter-organ/system vicious cycle is the linchpin of treating diabetic HFpEF.
Graphical abstract

Research insights
Diabetic patients with HFpEF are younger and more obese than their non-diabetic counterparts, and often have a higher rate of hospitalization and poorer prognosis.
AbstractSection What is the key research question?What are the characteristics and mechanisms of diabetic HFpEF?
AbstractSection What is new?This study describes the key cellular mechanisms of this inter-organ crosstalk in diabetic HFpEF. We also discuss the mediators of this crosstalk, such as circulating metabolites, cytokines and other factors, which are either directly released into the circulation or transported by exosomes to their target tissue.
AbstractSection How might this study influence clinical practice?Findings could lead to personalized treatment strategies for diabetic HFpEF.
Introduction
Heart failure (HF) remains a critical global health challenge, affecting over 64 million individuals worldwide. Among its subtypes, heart failure with preserved ejection fraction (HFpEF) has emerged as the predominant form [1,2,3]. This epidemiological shift correlates strongly with aging populations and escalating rates of hypertension, metabolic disorders, obesity, and diabetes [3,4,5]. Advanced phenotyping studies reveal that a substantial proportion of HFpEF patients exhibit a distinct “metabolic-obese” profile [6].
Diabetes coexists in nearly 45% of HFpEF cases [7]. Clinical evidence indicates that diabetic HFpEF patients exhibit greater comorbidity burdens, including severe hypertension, pulmonary complications, and renal dysfunction, alongside more pronounced cardiac structural changes such as elevated ventricular filling pressures and advanced diastolic dysfunction compared to non-diabetic HFpEF patients [8, 9]. Such pathophysiological distinctions suggest the necessity for tailored therapeutic strategies in this subpopulation.
While some reviews have discussed the distinctive features of cardiometabolic HFpEF, the specific pathomechanisms underlying diabetic HFpEF remain inadequately defined [2, 10, 11]. In this review, we systematically describe four pivotal aspects of diabetic HFpEF: (1) biomarkers, (2) characteristic metabolic rewiring patterns, (3) inter-organ crosstalk networks, and (4) currently available therapeutic strategies. Particular emphasis is placed on delineating the mediators and molecular mechanisms underlying inter-organ crosstalk.
Diabetic HFpEF and biomarker
Diabetes mellitus, a multifactorial systemic disorder, exerts heterogeneous effects across multiple tissues yet remains diagnostically defined by hyperglycemia exceeding established thresholds. Notably, diabetic patients with HFpEF demonstrate distinct clinical characteristics-younger age, higher obesity prevalence, elevated hospitalization rates, and unfavorable prognoses compared to non-diabetic HFpEF cohorts [12,13,14,15,16]. Impairments in left ventricular (LV) global longitudinal strain are more pronounced in HFpEF with concurrent type 2 diabetes mellitus (T2DM) [17]. Furthermore, pre-diabetic states frequently coexist with HFpEF, correlating with aggravated clinical manifestations and increased HF-related hospital admissions [18]. Emerging evidence highlights long-term glycemic variability as an independent predictor of adverse outcomes in diabetic HFpEF populations [19], while the stress hyperglycemia ratio (derived from acute-to-chronic glycemic markers) serves as a prognostic indicator for long-term cardiovascular complications [20]. While the contribution of hyperglycemia to diabetes complications is well-documented, contemporary research imperatives demand comprehensive exploration of alternative inter-organ crosstalk in diabetic HFpEF (Fig. 1).
Multisystemic changes in diabetic HFpEF. HFpEF is a multisystemic syndrome featuring prominent extracardiac involvement (e.g., liver, intestine, hematopoietic system and adipose tissue). Comprehensive treatment strategies targeting multiple systems may yield better outcomes. BAT, brown adipose tissue; GMB, gut microbiota; HFpEF, heart failure with preserved ejection fraction; NASH, nonalcoholic steatohepatitis; SCFAs, short-chain fatty acids; WAT, white adipose tissue
HFpEF harbors distinct plasma signatures linked to fluid retention, inflammatory activation, matrix remodeling, and cellular stress-injury axis [21, 22]. Based on the blood preparation and proteomic analysis of 545 diabetic patients (244 with HFpEF and 301 without), Su et al. revealed that the plasma level of integrin subunit alpha 1 (ITGA1) was significantly elevated, which was an independent predictor of cardiac dysfunction in patients with T2DM. Further mechanistic studies demonstrated that an elevated ITGA1 level might affect the cardiac function of patients with diabetes complicated by HFpEF by promoting myocardial fibrosis [23]. Additionally, in a study involving 374 HFpEF patients, Alogna et al. found that high interleukin-6 (IL-6) levels were associated with more severe cardiometabolic disease, with a higher prevalence of obesity and diabetes, worse renal function, and anemia [24]. Distinct biomarker profiles further differentiate obese HFpEF phenotypes, characterized by elevated circulating markers of volume overload (adrenomedullin), fibrotic remodeling (thrombospondin-2), and systemic inflammation (galectin-9, CD4 + T-cell markers), underscoring the need for phenotype-specific therapeutic strategies [25].
The identification and application of biomarkers in HFpEF face significant challenges due to the syndrome’s inherent heterogeneity, which encompasses diverse phenotypes (e.g., metabolic, hypertensive, or renal-driven subtypes) [26], complicating the discovery of universal biomarkers. Overlap with comorbidities such as obesity, diabetes, and chronic kidney disease further obscures biomarker specificity [27], as elevated levels (e.g., fibrosis markers) may reflect systemic dysfunction rather than HFpEF-specific pathophysiology. Addressing these limitations requires combining biomarkers with imaging, clinical context, and novel molecular tools to refine diagnosis and personalized management.
Cardiac substrate metabolism perturbations
HFpEF shares pathophysiological overlap with diabetes and obesity through disrupted energy metabolism, including defective fuel substrate utilization, lipotoxicity, and impaired cytoprotective signaling [5]. Transcriptomic analyses reveal HFpEF-specific upregulation of mitochondrial oxidative phosphorylation pathways-contrasting with downregulation patterns in heart failure with reduced ejection fraction (HFrEF)-while suppression of endoplasmic reticulum stress response, autophagy, and angiogenesis pathways occurs independently of comorbid conditions [28]. Paradoxically, despite prevalent obesity and insulin resistance, HFpEF myocardium demonstrates reduced fatty acid (FA) oxidation capacity and diminished tricarboxylic acid cycle intermediates compared to HFrEF, reflecting impaired metabolic flexibility that remains undetectable in systemic circulation [29]. Concomitant reductions in glycolytic intermediates and regulatory enzyme expression further characterize the metabolic landscape of HFpEF myocardium [30]. The current meta-analysis of metabolites found that higher plasma and serum levels of certain amino acids (branched-chain, aromatic, alanine, glutamate, lysine, and methionine), carbohydrates and energy-related metabolites (mannose, trehalose, and pyruvate), acylcarnitines (C4-DC, C4-OH, C5, C5-OH, and C8:1), the majority of glycerolipids (di- and triacylglycerols), (lyso)phosphatidylethanolamines, and ceramides included in meta-analysis were associated with higher risk of T2DM [31]. In individuals with cardiometabolic HFpEF, profound changes in cardiac metabolism have been identified (Fig. 2).
Perturbations in cardiac substrate metabolism in diabetic HFpEF. Diabetic HFpEF exhibits impaired cardiac substrate metabolism, marked by reduced metabolic flexibility. Despite systemic insulin resistance, myocardial fatty acid oxidation decreases, contrasting with diabetes-related lipid overload, while glucose utilization is suppressed via GLUT4 downregulation and pyruvate dehydrogenase inhibition. Mitochondrial dysfunction and oxidative stress coexist with disrupted tricarboxylic acid cycle intermediates. Accumulated metabolites (e.g., acetyl-CoA, ceramides) drive lipotoxicity and epigenetic dysregulation. Concurrently, amino acid and ketone metabolism alterations exacerbate energetic inefficiency. AcAc, acetoacetate; ACAT, acetoacetyl CoA thiolase; ATP, adenosine triphosphate; BCAA, branched-chain amino acids; BCATm, mitochondrial BCAA amimotransferase; BCKA, branched-chain keto acids; BCKDH, branched-chain α-ketoacid dehydrogenase; BDH1, b-hydroxybutyrate dehydrogenase 1; CD36, the cluster of differentiation 36; CPT, carnitine palmitoyltransferase; Cyt C, cytochrome C; ETC, electron transport chain; FACS, fatty acid CoA synthetase; LAT, L-type amino acid transporters; LCAD, long-chain acyl-CoA dehydrogenase; LDH, lactate dehydrogenas; MCT1, monocarboxylate transporter 1; GLUT1/4, glucose transporter 1/4; MPC, mitochondrial pyruvate carrier; NAD, nicotinamide adenine dinucleotide; SCOT, succinyl-CoA:3-ketoacid CoA transferase; TCA, tricarboxylic acid; βHAD, 3-OH-acyl-CoA dehydrogenase; βOHB, b-hydroxybutyrate
Clarifying the energy metabolic changes in HFpEF is hindered by the lack of animal models that can fully characterize the complex pathophysiology of HFpEF. Nowadays, metabolic syndrome-related models are established through methods such as a high-fat diet (HFD) combined with streptozotocin (STZ)-induced diabetes, or using leptin/leptin receptor-mutation mice (e.g., db/db or ob/ob strains) [32]. Lipoxin receptor-deficient mice (ALX−/−) might serve as a new experimental model that defines multiple cellular and molecular mechanisms in HFpEF with profound age-associated endothelial dysfunction and inflammation in the spleen, heart, and kidneys [33]. These models offer advantages in mimicking metabolic abnormalities (e.g., obesity, diabetes, dyslipidemia) commonly observed in HFpEF patients and allow for the study of systemic inflammation linked to adipose tissue dysfunction, endothelial dysfunction and myocardial fibrosis [34,35,36]. Limitations involve risks of progression to systolic dysfunction without strict intervention timing, rodent-specific metabolic discrepancies, and challenges in maintaining preserved ejection fraction (requiring precise stressor control) [37,38,39]. Despite these constraints, they remain pivotal for studying metabolic dysregulation’s role in diabetic HFpEF pathophysiology.
In HFD/STZ-induced HFpEF mice, the impairment of endothelial SIRT6 expression links diabetic HFpEF by altering FA transport across the endothelial barrier. Pharmacological strategies to restore SIRT6 function in diabetic endothelium alleviate experimental HFpEF by limiting FA override and improving lipid accumulation [40]. Obesity in ALX−/− mice induce myocardium endothelial dysfunction with the alterations in cluster of differentiation 31/phosphorylated eNOS (CD31/peNOS) pathways [22]. Therapeutic modulation of nicotinamide adenine dinucleotide (NAD+)-a critical cofactor in energy metabolism and cellular repair-through precursor supplementation represents a promising intervention avenue for HFpEF management [41,42,43,44].
Inter-organ crosstalk in diabetic HFpEF
HFpEF is increasingly recognized as a systemic disorder driven by intricate interactions between the heart and distant organs [45,46,47,48]. Emerging evidence highlights how metabolic dysfunction, chronic inflammation, and immune dysregulation propagate through circulating mediators-including metabolites, cytokines, and exosomes-to orchestrate cardiac remodeling and dysfunction. The liver, gut, adipose tissue, and hematopoietic system each contribute distinct yet interconnected mechanisms, forming a pathological network that perpetuates myocardial fibrosis, oxidative stress, and impaired energy metabolism (Fig. 3). This section delineates the critical axes of inter-organ crosstalk.
The inter-organ crosstalk in diabetic HFpEF. In diabetic HFpEF, gut microbiota dysbiosis and metabolites like IPA, TMAO, SCFAs engage in the gut - heart axis. NASH and HFpEF share lipid-accumulation, inflammation, fibrosis mechanisms. Abnormal liver metabolites (Saa1/4, lipid metabolites) and liver-secreted FXI and FGF21 link liver and heart via metabolic inflammation and fibrosis pathways. EAT, near the heart, directly drives HFpEF pathology through local inflammation, lipotoxicity, and mechanical compression. VAT are indirectly involved via systemic metabolic disorders and inflammation. The hematopoietic system contributes through immune cell activation (macrophages, T-cells) and inflammatory cytokines, perpetuating myocardial injury. AhR, aryl hydrocarbon receptor; BMP, bone morphogenetic protein; EAT, epicardial adipose tissue; FGF21, fibroblast growth factor 21; FFA, free fatty acid; FXI, factor XI; GDF15, growth differentiation factor 15; HFpEF, heart failure with preserved ejection fraction; IL, interleukin; IPA, indole-3-propionic acid; LCFAs, long-chain fatty acids; LPL, lipoprotein lipase; MPO, myeloperoxidase; NAD, nicotinamide adenine dinucleotide; NAM, nicotinamide; NASH, nonalcoholic steatohepatitis; NETs, neutrophil extracellular traps; NLRP3, NOD-like receptor family pyrin domain-containing 3; NNMT, nicotinamide N-methyltransferase; PI3K/AKT, phosphoinositide 3-kinase/protein kinase B; Saa1/4, serum amyloid A1/A4; SCFAs, short-chain fatty acids; SIRT3, sirtuin 3; SMAD1/5, SMAD family member 1/5; TGF-β, transforming growth factor-β; TMAO, trimethylamine N-oxide; TNF-α, tumor necrosis factor-α; TNFR1, tumor necrosis factor receptor 1; VAT, visceral adipose tissue
Liver-heart axis
The heart is a “catabolic” organ, and the liver is predominantly “anabolic“ [46]. Multifaceted interactions exist between them [49], with heart diseases often affecting liver diseases and vice versa [50]. In the interplay between liver and heart diseases, nonalcoholic steatohepatitis (NASH) and HFpEF, considered the most common and related [46, 51], share similar risk factors like diabetes and obesity [52]. Up to half of HFpEF patients have NASH [53], and conversely, NASH also increases the risk of developing HFpEF [54]. Pathophysiologically, the combination of obesity and hypertension gives rise to the most common HFpEF phenotype, cardiometabolic HFpEF, characterized by lipid accumulation and abnormal activation of inflammatory pathways, jointly leading to cellular structural and functional damage, fibrosis progression, and organ dysfunction. This bears a striking resemblance to the pathophysiology of NASH, which involves lipid accumulation, inflammation, along with oxidative stress and fibrosis [46]. Additionally, the benefits of sodium-glucose co-transporter 2 inhibitor (SGLT2i), a clinical medication for HFpEF, on NASH have emerged in meta-analyses [55]. Moreover, pirfenidone, an anti-fibrotic drug, is currently undergoing clinical trials in HFpEF patients [56]. All of these similarities sparked significant interest among researchers in the potential liver-heart interactions between HFpEF and NASH. Many studies have been dedicated to this area, uncovering several metabolites involved in such interactions, which we will elaborate on in the subsequent sections.
Through multi-omics integrated analysis, Strocchi et al. for the first time revealed that serum amyloid A1/A4 (Saa1/4) are key candidate mediators of inter-organ crosstalk between the liver and the heart in HFpEF, and they exhibit cross-species conservation characteristics. They indicated that Saa1/4 may exert their functions through metabolic inflammation and myocardial fibrosis [57]. Wegermann et al. analyzed serum metabolites in NASH patients and found 53 related to HFpEF. Lipid metabolites were predominant (73.6%), like glycerophosphocholine and glycerophosphoethanolamine. Fifteen metabolites were significantly linked across different HFpEF phenotypes, mainly lipids and cysteine-related ones. These indicated lipid metabolism disorders as a shared mechanism for NASH and HFpEF [51]. In 2022, Science reported that coagulation factor 11, a protein synthesized and secreted solely by the liver, can activate the bone morphogenetic protein (BMP)-SMAD1/5 pathway in the heart, thereby suppressing inflammation and fibrosis and protecting diastolic cardiac function [58]. The latest study showed that secreted Fibroblast growth factor 21 (FGF21) from the live enhances the production of adiponectin in adipocytes, which in turn indirectly acts on cardiomyocytes, or FGF21 directly targets cardiomyocytes, to negatively regulate pyruvate dehydrogenase kinase 4 production by activating phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) signals, then promoting mitochondrial bioenergetics [59]. Therefore, FGF21 protects against HFpEF via fine-tuning the multiorgan crosstalk among the adipose, liver, and heart [59]. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is mainly synthesized and secreted by the liver. Da Dalt et al. discovered that systemic Pcsk9 knockout mice lead to the development of HFpEF and the accumulation of cardiac lipids, especially cholesterol. Subsequent studies indicated that this is not caused by a reduction in circulating PCSK9, but rather driven by a decrease in cardiac PCSK9 [60]. In addition, myocardial infarction promotes hepatic fibrosis in mice with metabolic dysfunction-associated steatotic liver disease, accompanied by elevated circulating Ly6Chi monocytes and their recruitment to damaged liver tissues [61]. Another study has found the ischemic heart sends signals to the liver through the acute inflammatory IL-6/signal transducer and activator of transcription 3 (STAT3) pathway and that the liver responds by down-regulating mineralocorticoid receptors to promote FGF21 production and to alleviate ischemic injury [62].
Overall, there are numerous interactions between liver and heart diseases, with NASH and HFpEF being the most classic examples. Many metabolites are involved in the interaction process between them. The commonalities and interactions between these two diseases offer new perspectives for disease management. Addressing lipid accumulation and inflammation may be a win-win strategy.
Gut-heart axis
In cardiometabolic HFpEF, the interplay between the heart and the gut is intricate yet crucial. Cardiac low-output, for one, can induce intestinal ischemia and intestinal barrier damage [63]. Gut microbiota (GMB)-induced metabolite changes, for another, can affect cardiac function and structure [64,65,66]. This forms a vicious cycle. This interaction process will be expounded in detail hereinafter.
The alterations in GMB serve as the cornerstone of the gut-heart axis. Beale et al. discovered that in patients with HFpEF, the α-diversity of GMB decreased, manifested as a decline of the richness. β-diversity analysis indicated that the ratio of Firmicutes to Bacteroidetes (a commonly used biomarker of gut dysbiosis) showed a decreasing trend, and Ruminococcus (which produces short-chain fatty acids (SCFAs)) significantly decreased [45]. There are significant differences in the microbial abundances of Lactobacillaceae, Ruminococcaceae, Erysipelotrichaceae, and Lachnospiraceae between the obesity-related HFpEF rat model and control [67]. Huang et al. elucidated the bridging role of inflammation in the gut - heart axis. They found that in HFpEF, the abundance of pro-inflammatory flora, such as Lactobacillus and Enterococcus, increased, while that of anti-inflammatory flora, including Butyricicoccus, Sutterella, Lachnospira, and Ruminiclostridium, decreased [68].
A variety of metabolites serve as messengers of the gut-heart axis. Indole-3-propionic acid (IPA) is derived from dietary tryptophan through transformation by GMB such as Lactobacillus reuteri, Akkermansia muciniphila, and Clostridium sporogene [69,70,71,72]. Wang et al. discovered that the serum IPA level decreased in two HFpEF cohorts and the mouse model, and it showed a negative correlation with diastolic cardiac function. After IPA supplementation, it could alleviate weight gain and fat accumulation in mice, improve diastolic function and metabolic homeostasis, and also mitigate GMB dysbiosis and intestinal epithelial barrier damage. Further mechanistic investigation indicated that the beneficial effects of IPA on fat accumulation and diastolic function were exerted, at least in part, by inhibiting the expression of nicotinamide N - methyltransferase (NNMT) and restoring the levels of nicotinamide, NAD⁺/NADH, and silent information regulator 3 (SIRT3) [44].
Trimethylamine N-oxide (TMAO), an oxidative amine, occurs naturally in our diet like fish. It can also be generated from its metabolic precursors, including substances like choline and carnitine in food [73]. GMB, such as Firmicutes, Proteus, and Actinomyces, are involved in the metabolic production process of TMAO [74]. Guivala et al. found that in the obesity-related HFpEF rat model, the serum TMAO level increased significantly, and it showed a positive correlation with the degree of diastolic dysfunction [67]. Elevated TMAO demonstrates significant positive correlations with body mass index (BMI), E/e’ ratio, and B-type natriuretic peptide (BNP) levels [75]. Heightened TMAO levels detected at hospital discharge serve as an independent predictor for cardiac events in HFpEF [76]. Combined TMAO and BNP evaluation offers a potential approach for risk stratification in HFpEF patients [75].
SCFAs are organic fatty acids containing fewer than six carbon atoms, such as acetic acid, propionic acid, and butyric acid. They are derived from the fermentation of various dietary fiber substances by GMB [77]. SCFAs can reduce the levels of circulating long-chain fatty acids (LCFAs) and glucose, contributing to the prevention of cardiac steatosis and glucotoxicity, which are regarded as the initiating events in the pathogenesis of obesity-related HFpEF [78, 79]. Additionally, SCFAs possess anti-inflammatory and antioxidant properties [80]. In terms of inflammation regulation, SCFAs can inhibit the up-regulation of pro-inflammatory genes in cardiac tissues, such as IL-1β, NOD-like receptor family pyrin domain containing 3 (NLRP3), and monocyte chemoattractant protein-1 (MCP-1) [81, 82]. Additionally, they can prevent cardiac remodeling by promoting the generation of regulatory T cells [83]. Regarding antioxidant effects, SCFAs can decrease oxidative stress; for example, butyrate can reduce the level of lipid peroxidation [84]. These effects sever the crucial link between metabolic syndrome and HFpEF [64].
In conclusion, the gut-heart axis plays a crucial role in the pathogenesis of cardiometabolic HFpEF, and multiple metabolites (like IPA, TMAO and SCFAs) have been demonstrated to be involved in this inter-organ crosstalk. The development of related drugs may be beneficial for the treatment of cardiometabolic HFpEF.
Adipose-heart axis
Clinical evidence establishes adipose tissue dysfunction as a robust predictor of cardiovascular events and HF progression in populations with obesity, metabolic syndrome, and established T2DM [85, 86]. Visceral adipose tissue releases free fatty acids and pro-inflammatory factors (such as tumor necrosis factor-α (TNF-α) and IL-6), leading to systemic inflammation and insulin resistance, which indirectly exacerbate myocardial fibrosis and diastolic dysfunction [87].
The location of adipose deposits is also likely to be important [88,89,90,91]. Epicardial adipose tissue (EAT), a metabolically active fat depot situated between the myocardium and visceral pericardium, has emerged as a clinically significant entity in cardiovascular medicine [92]. HFpEF exhibits substantially greater EAT thickness compared to both healthy controls and other HF subtypes (including reduced or mid-range ejection fraction) [93, 94]. Excessive EAT deposition correlates with aggravated diastolic dysfunction, manifesting as more severe impairment of ventricular relaxation and maladaptive cardiac remodeling patterns in HFpEF [95,96,97]. Longitudinal cohort evidence further establishes pericardial adiposity as an independent risk factor for predicting de novo cardiovascular events [98]. Notably, EAT demonstrates superior prognostic stratification capacity compared to conventional parameters such as left ventricular ejection fraction (LVEF) and New York Heart Association functional classification [99].
Both elevated EAT thickness and volume were observed in patients with prediabetes and T2DM, which were associated with adverse alterations in cardiac structure and cardiopulmonary performance [100,101,102,103]. In HFpEF, elevated EAT measurements demonstrate strong associations with arterial stiffness, as quantified by brachial-ankle pulse-wave velocity [104]. Asymptomatic HF patients with T2DM exhibit distinct EAT-related cardiac abnormalities, including LV remodeling, functional impairments, and reduced exercise tolerance [105]. Notably, preclinical cardiac dysfunction in obese individuals without overt cardiovascular disease appears linked to EAT accumulation, suggesting its contributory role in subclinical disease progression [106]. Obese patients with excessive EAT deposition experience disproportionately higher rates of adverse cardiovascular events compared to those with lower EAT volumes [107, 108]. Therefore, EAT demonstrates multifaceted interactions with myocardial injury biomarkers, ventricular hypertrophy, elevated intracardiac pressures, systemic inflammation, insulin resistance, and impaired functional capacity - all characteristic hallmarks of HFpEF pathophysiology.
Recent advancements propose EAT radiodensity (rather than volumetric measures alone) as a superior predictor of cardiometabolic risk and prognostic stratification in HFpEF cohorts, with specific relevance to metabolic syndrome components [109,110,111]. Proteomics and transcriptomics analysis of EAT in HFpEF suggested their potential roles in inflammation, metabolic processes, and mitochondrial dysfunction [112,113,114,115,116]. The pathophysiological mechanisms of EAT in cardiometabolic HFpEF involve three principal pathways: (1) paracrine signaling of proinflammatory adipocytokines, (2) myocardial lipid deposition inducing lipotoxic injury, and (3) direct biomechanical compression through pericardial space restriction [88, 117].
All these reveal the diverse pathogenic mechanisms of EAT, unlike conventional adipose depots serving metabolic functions. Targeted therapeutic interventions aimed at modulating EAT quantity and metabolic activity show potential for improving myocardial recovery in HF patients with concurrent T2DM [118, 119].
Hematopoiesis system-heart axis
Emerging evidence supports the pivotal role of systemic inflammation in bridging comorbidities with structural and functional cardiac abnormalities in HFpEF [33, 120]. Proteomic profiling using Olink technology has identified growth differentiation factor-15 (GDF-15), urokinase plasminogen activator receptor (UPAR), tumor necrosis factor receptor-1(TNFR1), insulin-like growth factor binding protein 7 (IGFBP7) as principal mediators linking comorbidity burden with echocardiographic alterations [121]. Critical immune pathways involving NLRP3 inflammasome activation with IL-1β secretion, along with T-cell activation mechanisms, have been implicated in the pathogenesis of metabolic disorders (obesity, hypertension) and cardiac dysfunction associated with HFpEF [87].
Genetic studies using T-cell receptor-deficient mice have established the essential role of T lymphocytes in diastolic impairment and cardiomyocyte hypertrophy. Combinatorial treatment with HFD and Nω-Nitro-L-arginine methyl ester (L-NAME) induces differential cardiac infiltration patterns of immune cells (CD4+/CD8 + T cells, monocytes, neutrophils, B cells) compared to single intervention models [122]. Pharmacologically, SGLT2i exhibits dual therapeutic effects: ameliorating myocardial inflammation in HFpEF [123], while suppressing T-cell effector functions through metabolic rewiring in autoimmune conditions [124].
LCFAs activate multiple inflammatory cytokines released from macrophages, impinging on cardiomyocytes [123]. The pathophysiology of diastolic dysfunction involves elevated circulating monocyte levels and myocardial macrophage accumulation, primarily mediated through enhanced monocyte recruitment from hematopoietic organs (bone marrow and spleen) [125]. Notably, cardiac CCR2+ macrophages demonstrate increased IL-10 production, promoting fibrogenic transformation via autocrine signaling pathways [125]. Bochra Tourki et al. have found the ALX deficiency expands Ly6ChiCCR2+ macrophage population in spleen, primes CCL2 cluster chemokines in the heart, and induces myocardium endothelial dysfunction [22]. These findings collectively highlight macrophage dynamics as a central modulator of diverse HFpEF phenotypic manifestations across experimental models. Neutrophil extracellular traps also contribute to the pathogenesis of HFpEF and HFrEF, which could induced mitochondrial dysfunction in cardiomyocytes, inhibiting mitochondrial biogenesis via the neutrophil elastase-toll-like receptor 4 (NE-TLR4)-mediated suppression of peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α) [126,127,128]. Recent breakthroughs have uncovered novel neuro-immune regulatory mechanisms, where fasting-induced neural signals modulate immune cell activity to establish inter-organ crosstalk networks. These pathways critically regulate pancreatic endocrine function and systemic glucose metabolism [129]. Specifically, neuronal interactions with type 2 innate lymphoid cells (ILC2s) have been identified as key regulators of glucagon secretion and glucose homeostasis [129].
In conclusion, cardiometabolic HFpEF arises from chronic low-grade inflammation and immune cell activation. Metabolic derangements and inflammation form a vicious cycle. Therapeutic strategies targeting immune cell polarization or inflammatory mediators may restore metabolic adaptability and delay disease progression.
Diagnosis and treatment of diabetic HFpEF
Diagnosis of HFpEF is based on clinical symptoms (e.g., dyspnea, fatigue), elevated natriuretic peptides, imaging evidence of preserved LVEF (≥ 50%), and exclusion of alternative causes (e.g., valvular disease, infiltrative cardiomyopathies) using echocardiography, cardiac MRI, or invasive hemodynamic testing [130].
Notably, HFpEF is not a single disease entity but rather a heterogeneous condition characterized by multifactorial pathogenesis and diverse clinical presentations [6, 33]. Peters et al. identified three clinically significant HFpEF phenotypes through phenotype clustering analysis of 20 studies: (1) older, vascular ageing; (2) metabolic, obese; and (3) relatively younger, natriuretic peptide deficiency [6]. This subsection specifically addresses diabetic HFpEF., which accounts for 25–30% of HFpEF cases and has emerged as a distinct metabolic subtype receiving growing research attention [16, 131, 132]. Current treatment focuses on symptom relief, reducing hospitalizations, and managing comorbidities. We have summarized the medications demonstrating therapeutic potential in populations with HFpEF comorbid with obesity and/or diabetes (Table 1).
SGLT2i SGLT2 inhibitors (e.g., empagliflozin, dapagliflozin) are cornerstone therapies, proven in trials (DELIVER, EMPEROR-Preserved) to reduce cardiovascular death and HF hospitalizations across the LVEF spectrum [133, 134]. The EMPEROR-Preserved trial revealed that empagliflozin lowered the composite risk of cardiovascular death or heart failure hospitalization in HFpEF, regardless of diabetes status [134]. SGLT2 inhibitors improve HFpEF through multiple molecular mechanisms: metabolic rewiring of cardiac and renal tissues to favor lipid/ketone utilization over carbohydrates, reducing oxidative stress and inflammation via suppression of pro-inflammatory mediators, and mitigating myocardial fibrosis [135]. They optimize ventricular loading through diuresis and natriuresis, lowering cardiac preload and afterload. Additionally, SGLT2 inhibitors enhance renal hemodynamics by correcting hyperfiltration, albuminuria, and hypoxia, while modulating mitochondrial function and autophagy. These combined effects reduce systemic congestion, improve endothelial dysfunction, and attenuate pathological remodeling, thereby alleviating HFpEF progression [27]. Additionally, Wen et al. indicated that SGLT2i reduces cardiomyocyte senescence and restores cardiac function in diabetic conditions by suppressing forkhead box protein O1 (FOXO1)-mediated angiopoietin-like protein 4 (ANGPTL4) transcription [136].
Glucagon-like peptide-1 receptor agonist (GLP1-RA) In the STEP-HFpEF trial, the GLP1-RA semaglutide alleviated symptoms and physical limitations, improved exercise function, and promoted weight loss in HFpEF with obesity [137]. Similarly, the STEP-HFpEF DM trial demonstrated that semaglutide reduced symptoms and physical limitations while achieving weight loss in individuals with obesity-related HFpEF and concomitant T2DM [138]. HFpEF mouse model induced by advanced age, female, obesity and T2DM, Withaar et al. indicated that semaglutide improves HFpEF through multiple mechanisms beyond weight loss. It enhances LV cytoskeleton function, restores endothelial function, and reduces oxidative stress and systemic inflammation by upregulating antioxidant enzymes and inhibiting pro-inflammatory pathways. Additionally, it attenuates cardiac fibrosis, LV hypertrophy, and lung congestion. Semaglutide also modulates protective immune responses in visceral adipose tissue, promoting T-cell regulation and interferon-γ production [139]. These cardiometabolic effects are mediated via GLP-1 receptor activation, particularly in endothelial cells, improving myocardial performance independently of caloric restriction [139].
Twincretin Tirzepatide, a novel dual agonist targeting both glucose-dependent insulinotropic polypeptide and GLP-1 receptors, has received regulatory approval in multiple regions including the United States, European Union, and Japan [140]. It is indicated as an adjunctive therapy alongside dietary modifications and physical activity to optimize glycemic management in adult patients with T2DM [140]. The SUMMIT trial demonstrated that tirzepatide therapy reduced the composite risk of cardiovascular mortality or heart failure progression compared to placebo and enhanced health-related outcomes in obese individuals with HFpEF [141].
Conclusion
Diabetic HFpEF represents a complex, multisystem disorder driven by metabolic dysregulation, chronic inflammation, immune dysfunction, endothelial dysfunction and myocardial fibrosis. Emerging evidence highlights the pivotal role of inter-organ crosstalk, mediated by metabolites, cytokines, and exosomes, in perpetuating a vicious cycle of cardiac and systemic dysfunction. Key axes include the adipose-heart axis, where epicardial adipose tissue exacerbates inflammation and mechanical stress; the gut-heart axis, involving dysbiosis and metabolites like IPA, TMAO and SCFAs; the liver-heart axis, NASH to shared pathogenic pathways; and hematopoietic system contributions via immune cell activation. Therapeutic strategies targeting these pathways-such as SGLT2i, GLP1-RA, twincretin and NAD⁺ supplementation show promise in disrupting this cycle. Future research should prioritize elucidating organ-specific molecular mechanisms and developing integrated, phenotype-specific therapies to address the heterogeneity of diabetic HFpEF. Breaking the inter-organ vicious cycle remains central to improving outcomes in this challenging syndrome.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- ANGPTL4:
-
Angiopoietin-like protein 4
- BMP:
-
Bone morphogenetic protein
- BNP:
-
B-type natriuretic peptide
- FA:
-
Fatty acid
- FGF21:
-
Fibroblast growth factor 21
- FOXO1:
-
Forkhead box protein O1
- GDF-15:
-
Growth differentiation factor-15
- GLP1-RA:
-
Glucagon-like peptide-1 receptor agonist
- GMB:
-
Gut microbiota
- HFpEF:
-
Heart failure with preserved ejection fraction
- HFrEF:
-
Heart failure with reduced ejection fraction
- HFD:
-
High fat diet
- IGFBP7:
-
Insulin-like growth factor binding protein 7
- IPA:
-
Indole-3-propionic acid
- LCFAs:
-
Long-chain fatty acids
- L-NAME:
-
Nω-Nitro-L-arginine methyl ester
- LV:
-
Left ventricular
- LVEF:
-
Left ventricular ejection fraction
- MCP1:
-
Monocyte chemoattractant protein-1
- NE-TLR4:
-
Neutrophil elastase-toll-like receptor 4
- NLRP3:
-
NOD-like receptor family pyrin domain containing 3
- NASH:
-
Nonalcoholic steatohepatitis
- NNMT:
-
Nicotinamide N-methyltransferase
- PCSK9:
-
Proprotein convertase subtilisin/kexin type 9
- PGC1-α:
-
Peroxisome proliferator-activated receptor gamma coactivator 1-α
- Saa1/4:
-
Serum amyloid A1/A4
- SCFAs:
-
Short-chain fatty acids
- SGLT2i:
-
Sodium-glucose co-transporter 2 inhibitor
- SIRT3:
-
Silent information regulator 3
- T2DM:
-
Type 2 diabetes mellitus
- TNF-α:
-
Tumor necrosis factor-α
- TNFR1:
-
Tumor necrosis factor receptor-1
- UPAR:
-
Urokinase plasminogen activator receptor
References
Campbell P, Rutten FH, Lee MM, Hawkins NM, Petrie MC. Heart failure with preserved ejection fraction: everything the clinician needs to know. Lancet. 2024;403(10431):1083–92.
Hamo CE, DeJong C, Hartshorne-Evans N, Lund LH, Shah SJ, Solomon S, Lam CSP. Heart failure with preserved ejection fraction. Nat Rev Dis Primers. 2024;10(1):55.
Redfield MM, Borlaug BA. Heart failure with preserved ejection fraction: a review. JAMA. 2023;329(10):827–38.
Omote K, Verbrugge FH, Borlaug BA. Heart failure with preserved ejection fraction: mechanisms and treatment strategies. Annu Rev Med. 2022;73:321–37.
Mishra S, Kass DA. Cellular and molecular pathobiology of heart failure with preserved ejection fraction. Nat Rev Cardiol. 2021;18(6):400–23.
Peters AE, Tromp J, Shah SJ, Lam CSP, Lewis GD, Borlaug BA, Sharma K, Pandey A, Sweitzer NK, Kitzman DW, et al. Phenomapping in heart failure with preserved ejection fraction: insights, limitations, and future directions. Cardiovasc Res. 2023;118(18):3403–15.
Echouffo-Tcheugui JB, Xu H, DeVore AD, Schulte PJ, Butler J, Yancy CW, Bhatt DL, Hernandez AF, Heidenreich PA, Fonarow GC. Temporal trends and factors associated with diabetes mellitus among patients hospitalized with heart failure: findings from get with the guidelines-heart failure registry. Am Heart J. 2016;182:9–20.
Kristensen SL, Mogensen UM, Jhund PS, Petrie MC, Preiss D, Win S, Køber L, McKelvie RS, Zile MR, Anand IS, et al. Clinical and echocardiographic characteristics and cardiovascular outcomes according to diabetes status in patients with heart failure and preserved ejection fraction: a report from the I-preserve trial (Irbesartan in heart failure with preserved ejection fraction). Circulation. 2017;135(8):724–35.
Lindman BR, Dávila-Román VG, Mann DL, McNulty S, Semigran MJ, Lewis GD, de las Fuentes L, Joseph SM, Vader J, Hernandez AF, et al. Cardiovascular phenotype in HFpEF patients with or without diabetes: a RELAX trial ancillary study. J Am Coll Cardiol. 2014;64(6):541–9.
Capone F, Sotomayor-Flores C, Bode D, Wang R, Rodolico D, Strocchi S, Schiattarella GG. Cardiac metabolism in HFpEF: from fuel to signalling. Cardiovasc Res. 2023;118(18):3556–75.
Fuerlinger A, Stockner A, Sedej S, Abdellatif M. Caloric restriction and its mimetics in heart failure with preserved ejection fraction: mechanisms and therapeutic potential. Cardiovasc Diabetol. 2025;24(1):21.
McHugh K, DeVore AD, Wu J, Matsouaka RA, Fonarow GC, Heidenreich PA, Yancy CW, Green JB, Altman N, Hernandez AF. Heart failure with preserved ejection fraction and diabetes: JACC State-of-the-Art review. J Am Coll Cardiol. 2019;73(5):602–11.
Lejeune S, Roy C, Slimani A, Pasquet A, Vancraeynest D, Vanoverschelde JL, Gerber BL, Beauloye C, Pouleur AC. Diabetic phenotype and prognosis of patients with heart failure and preserved ejection fraction in a real life cohort. Cardiovasc Diabetol. 2021;20(1):48.
Kaze AD, Bertoni AG, Fox ER, Hall ME, Mentz RJ, Berry JD, Echouffo-Tcheugui JB. Diabetes, subclinical myocardial injury or stress and risk of heart failure subtypes: the Jackson heart study. Diabetes Care; 2025.
Patel KV, Khan MS, Segar MW, Bahnson JL, Garcia KR, Clark JM, Balasubramanyam A, Bertoni AG, Vaduganathan M, Farkouh ME, et al. Optimal cardiometabolic health and risk of heart failure in type 2 diabetes: an analysis from the look AHEAD trial. Eur J Heart Fail. 2022;24(11):2037–47.
Woolley RJ, Ceelen D, Ouwerkerk W, Tromp J, Figarska SM, Anker SD, Dickstein K, Filippatos G, Zannad F, Metra M, et al. Machine learning based on biomarker profiles identifies distinct subgroups of heart failure with preserved ejection fraction. Eur J Heart Fail. 2021;23(6):983–91.
Lin Y, Xie M, Zhang L, Zhang Y, Zhang P, Chen X, Ji M, Gao L, He Q, Wu Z et al. Prognostic value of LV global longitudinal strain by 2D and 3D speckle-tracking echocardiography in patients with HFpEF. Circ Cardiovasc Imaging 2025:e016975.
Jackson AM, Rørth R, Liu J, Kristensen SL, Anand IS, Claggett BL, Cleland JGF, Chopra VK, Desai AS, Ge J, et al. Diabetes and pre-diabetes in patients with heart failure and preserved ejection fraction. Eur J Heart Fail. 2022;24(3):497–509.
Hsu JC, Yang YY, Chuang SL, Lin LY. Long-term glycemic variability predicts adverse outcomes in diabetic heart failure with preserved ejection fraction. J Clin Endocrinol Metab 2024.
Mohammed AQ, Luo Y, Wang K, Su Y, Liu L, Yin G, Zhang W, Alifu JJ, Mareai RM, Mohammed AA, et al. Stress hyperglycemia ratio as a prognostic indicator for long-term adverse outcomes in heart failure with preserved ejection fraction. Cardiovasc Diabetol. 2024;23(1):67.
Zhang Z, Wang Y, Chen X, Wu C, Zhou J, Chen Y, Liu X, Tang X. The aging heart in focus: the advanced Understanding of heart failure with preserved ejection fraction. Ageing Res Rev. 2024;101:102542.
Tourki B, Kain V, Shaikh SR, Leroy X, Serhan CN, Halade GV. Deficit of resolution receptor magnifies inflammatory leukocyte directed cardiorenal and endothelial dysfunction with signs of cardiomyopathy of obesity. FASEB J. 2020;34(8):10560–73.
Su M, Hou Y, Cai S, Li W, Wei Y, Wang R, Wu M, Liu M, Chang J, Yang K, et al. Elevated ITGA1 levels in type 2 diabetes: implications for cardiac function impairment. Diabetologia. 2024;67(5):850–63.
Alogna A, Koepp KE, Sabbah M, Espindola Netto JM, Jensen MD, Kirkland JL, Lam CSP, Obokata M, Petrie MC, Ridker PM, et al. Interleukin-6 in patients with heart failure and preserved ejection fraction. JACC Heart Fail. 2023;11(11):1549–61.
Kresoja KP, Rommel KP, Wachter R, Henger S, Besler C, Klöting N, Schnelle M, Hoffmann A, Büttner P, Ceglarek U, et al. Proteomics to improve phenotyping in obese patients with heart failure with preserved ejection fraction. Eur J Heart Fail. 2021;23(10):1633–44.
Borlaug BA. Evaluation and management of heart failure with preserved ejection fraction. Nat Rev Cardiol. 2020;17(9):559–73.
Mentz RJ, Brunton SA, Rangaswami J. Sodium-glucose cotransporter-2 Inhibition for heart failure with preserved ejection fraction and chronic kidney disease with or without type 2 diabetes mellitus: a narrative review. Cardiovasc Diabetol. 2023;22(1):316.
Hahn VS, Knutsdottir H, Luo X, Bedi K, Margulies KB, Haldar SM, Stolina M, Yin J, Khakoo AY, Vaishnav J, et al. Myocardial gene expression signatures in human heart failure with preserved ejection fraction. Circulation. 2021;143(2):120–34.
Hahn VS, Petucci C, Kim MS, Bedi KC Jr., Wang H, Mishra S, Koleini N, Yoo EJ, Margulies KB, Arany Z, et al. Myocardial metabolomics of human heart failure with preserved ejection fraction. Circulation. 2023;147(15):1147–61.
Koleini N, Meddeb M, Zhao L, Keykhaei M, Kwon S, Farshidfar F, Hahn VS, Pearce EL, Sharma K, Kass DA. Landscape of glycolytic metabolites and their regulating proteins in myocardium from human heart failure with preserved ejection fraction. Eur J Heart Fail. 2024;26(9):1941–51.
Morze J, Wittenbecher C, Schwingshackl L, Danielewicz A, Rynkiewicz A, Hu FB, Guasch-Ferré M. Metabolomics and type 2 diabetes risk: an updated systematic review and Meta-analysis of prospective cohort studies. Diabetes Care. 2022;45(4):1013–24.
Jankauskas SS, Kansakar U, Varzideh F, Wilson S, Mone P, Lombardi A, Gambardella J, Santulli G. Heart failure in diabetes. Metabolism. 2021;125:154910.
Tourki B, Halade GV. Heart failure syndrome with preserved ejection fraction is a metabolic cluster of Non-resolving inflammation in obesity. Front Cardiovasc Med. 2021;8:695952.
Wen W, Cao Y, Chen P, Li J, Li W, Huang G, Zheng H, Zhu X, Zhang H, Chen Y, et al. A reliable strategy for establishment of an animal model of diabetic cardiomyopathy: induction by a high-fat diet combined with single or multiple injections of low-dose streptozotocin. Life Sci. 2024;358:123161.
Sankaralingam S, Abo Alrob O, Zhang L, Jaswal JS, Wagg CS, Fukushima A, Padwal RS, Johnstone DE, Sharma AM, Lopaschuk GD. Lowering body weight in obese mice with diastolic heart failure improves cardiac insulin sensitivity and function: implications for the obesity paradox. Diabetes. 2015;64(5):1643–57.
Alex L, Russo I, Holoborodko V, Frangogiannis NG. Characterization of a mouse model of obesity-related fibrotic cardiomyopathy that recapitulates features of human heart failure with preserved ejection fraction. Am J Physiol Heart Circ Physiol. 2018;315(4):H934–49.
Riehle C, Bauersachs J. Of mice and men: models and mechanisms of diabetic cardiomyopathy. Basic Res Cardiol. 2018;114(1):2.
Heather LC, Gopal K, Srnic N, Ussher JR. Redefining diabetic cardiomyopathy: perturbations in substrate metabolism at the heart of its pathology. Diabetes. 2024;73(5):659–70.
Xie S-Y, Liu S-Q, Zhang T, Shi W-K, Xing Y, Fang W-X, Zhang M, Chen M-Y, Xu S-C, Fan M-Q, et al. USP28 serves as a key suppressor of mitochondrial morphofunctional defects and cardiac dysfunction in the diabetic heart. Circulation. 2024;149(9):684–706.
Wu X, Liu H, Brooks A, Xu S, Luo J, Steiner R, Mickelsen DM, Moravec CS, Jeffrey AD, Small EM, et al. SIRT6 mitigates heart failure with preserved ejection fraction in diabetes. Circ Res. 2022;131(11):926–43.
Tong D, Schiattarella GG, Jiang N, Altamirano F, Szweda PA, Elnwasany A, Lee DI, Yoo H, Kass DA, Szweda LI, et al. NAD + repletion reverses heart failure with preserved ejection fraction. Circ Res. 2021;128(11):1629–41.
Abdellatif M, Trummer-Herbst V, Koser F, Durand S, Adão R, Vasques-Nóvoa F, Freundt JK, Voglhuber J, Pricolo M-R, Kasa M et al. Nicotinamide for the treatment of heart failure with preserved ejection fraction. Sci Transl Med 2021, 13(580).
Abdellatif M, Sedej S, Kroemer G. NAD + metabolism in cardiac health, aging, and disease. Circulation. 2021;144(22):1795–817.
Wang Y-C, Koay YC, Pan C, Zhou Z, Tang W, Wilcox J, Li XS, Zagouras A, Marques F, Allayee H, et al. Indole-3-propionic acid protects against heart failure with preserved ejection fraction. Circ Res. 2024;134(4):371–89.
Beale AL, O’Donnell JA, Nakai ME, Nanayakkara S, Vizi D, Carter K, Dean E, Ribeiro RV, Yiallourou S, Carrington MJ, et al. The gut Microbiome of heart failure with preserved ejection fraction. J Am Heart Assoc. 2021;10(13):e020654.
Capone F, Vettor R, Schiattarella GG. Cardiometabolic HFpEF: NASH of the heart. Circulation. 2023;147(6):451–3.
Borlaug BA, Jensen MD, Kitzman DW, Lam CSP, Obokata M, Rider OJ. Obesity and heart failure with preserved ejection fraction: new insights and pathophysiological targets. Cardiovasc Res. 2023;118(18):3434–50.
Thorp EB, Filipp M. Contributions of inflammation to cardiometabolic heart failure with preserved ejection fraction. Annu Rev Pathol. 2025;20(1):143–67.
Møller S, Bernardi M. Interactions of the heart and the liver. Eur Heart J. 2013;34(36):2804–11.
Baskin KK, Bookout AL, Olson EN. The heart-liver metabolic axis: defective communication exacerbates disease. EMBO Mol Med. 2014;6(4):436–8.
Wegermann K, Fudim M, Henao R, Howe CF, McGarrah R, Guy C, Abdelmalek MF, Diehl AM, Moylan CA. Serum metabolites are associated with HFpEF in biopsy-proven nonalcoholic fatty liver disease. J Am Heart Assoc. 2023;12(14):e029873.
Vernon G, Baranova A, Younossi ZM. Systematic review: the epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther. 2011;34(3):274–85.
Miller A, McNamara J, Hummel SL, Konerman MC, Tincopa MA. Prevalence and staging of non-alcoholic fatty liver disease among patients with heart failure with preserved ejection fraction. Sci Rep. 2020;10(1):12440.
Packer M. Atrial fibrillation and heart failure with preserved ejection fraction in patients with nonalcoholic fatty liver disease. Am J Med. 2020;133(2):170–7.
Mo M, Huang Z, Liang Y, Liao Y, Xia N. The safety and efficacy evaluation of sodium-glucose co-transporter 2 inhibitors for patients with non-alcoholic fatty liver disease: an updated meta-analysis. Dig Liver Dis. 2022;54(4):461–8.
Lewis GA, Dodd S, Clayton D, Bedson E, Eccleson H, Schelbert EB, Naish JH, Jimenez BD, Williams SG, Cunnington C, et al. Pirfenidone in heart failure with preserved ejection fraction: a randomized phase 2 trial. Nat Med. 2021;27(8):1477–82.
Strocchi S, Liu L, Wang R, Häseli SP, Capone F, Bode D, Nambiar N, Eroglu T, Santiago Padilla L, Farrelly C, et al. Systems biology approach uncovers candidates for liver-heart interorgan crosstalk in HFpEF. Circ Res. 2024;135(8):873–6.
Cao Y, Wang Y, Zhou Z, Pan C, Jiang L, Zhou Z, Meng Y, Charugundla S, Li T, Allayee H, et al. Liver-heart cross-talk mediated by coagulation factor XI protects against heart failure. Science. 2022;377(6613):1399–406.
Zhang K, Gan J, Wang B, Lei W, Zhen D, Yang J, Wang N, Wen C, Gao X, Li X, et al. FGF21 protects against HFpEF by improving cardiac mitochondrial bioenergetics in mice. Nat Commun. 2025;16(1):1661.
Da Dalt L, Castiglioni L, Baragetti A, Audano M, Svecla M, Bonacina F, Pedretti S, Uboldi P, Benzoni P, Giannetti F, et al. PCSK9 deficiency rewires heart metabolism and drives heart failure with preserved ejection fraction. Eur Heart J. 2021;42(32):3078–90.
Xie W, Gan J, Zhou X, Tian H, Pan X, Liu W, Li X, Du J, Xu A, Zheng M, et al. Myocardial infarction accelerates the progression of MASH by triggering immunoinflammatory response and induction of Periosti. Cell Metab. 2024;36(6):1269–e12861269.
Sun JY, Du LJ, Shi XR, Zhang YY, Liu Y, Wang YL, Chen BY, Liu T, Zhu H, Liu Y, et al. An IL-6/STAT3/MR/FGF21 axis mediates heart-liver cross-talk after myocardial infarction. Sci Adv. 2023;9(14):eade4110.
Groschwitz KR, Hogan SP. Intestinal barrier function: molecular regulation and disease pathogenesis. J Allergy Clin Immunol 2009, 124(1).
Hu T, Wu Q, Yao Q, Jiang K, Yu J, Tang Q. Short-chain fatty acid metabolism and multiple effects on cardiovascular diseases. Ageing Res Rev. 2022;81:101706.
Zhao M, Wei H, Li C, Zhan R, Liu C, Gao J, Yi Y, Cui X, Shan W, Ji L, et al. Gut microbiota production of trimethyl-5-aminovaleric acid reduces fatty acid oxidation and accelerates cardiac hypertrophy. Nat Commun. 2022;13(1):1757.
Romano KA, Nemet I, Prasad Saha P, Haghikia A, Li XS, Mohan ML, Lovano B, Castel L, Witkowski M, Buffa JA, et al. Gut microbiota-generated phenylacetylglutamine and heart failure. Circ Heart Fail. 2023;16(1):e009972.
Guivala SJ, Bode KA, Okun JG, Kartal E, Schwedhelm E, Pohl LV, Werner S, Erbs S, Thiele H, Büttner P. Interactions between the gut microbiome, associated metabolites and the manifestation and progression of heart failure with preserved ejection fraction in ZSF1 rats. Cardiovasc Diabetol. 2024;23(1):299.
Huang Z, Mei X, Jiang Y, Chen T, Zhou Y. Gut microbiota in heart failure patients with preserved ejection fraction (GUMPTION Study). Front Cardiovasc Med. 2021;8:803744.
Zhang LS, Davies SS. Microbial metabolism of dietary components to bioactive metabolites: opportunities for new therapeutic interventions. Genome Med. 2016;8(1):46.
Rothhammer V, Mascanfroni ID, Bunse L, Takenaka MC, Kenison JE, Mayo L, Chao C-C, Patel B, Yan R, Blain M, et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the Aryl hydrocarbon receptor. Nat Med. 2016;22(6):586–97.
Li Y, Xu W, Zhang F, Zhong S, Sun Y, Huo J, Zhu J, Wu C. The gut microbiota-produced indole-3-propionic acid confers the antihyperlipidemic effect of mulberry-derived 1-deoxynojirimycin. mSystems 2020, 5(5).
Dodd D, Spitzer MH, Van Treuren W, Merrill BD, Hryckowian AJ, Higginbottom SK, Le A, Cowan TM, Nolan GP, Fischbach MA, et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017;551(7682):648–52.
Cho CE, Caudill MA. Trimethylamine-N-Oxide: friend, foe, or simply caught in the cross-fire? Trends Endocrinol Metab. 2017;28(2):121–30.
Simó C, García-Cañas V. Dietary bioactive ingredients to modulate the gut microbiota-derived metabolite TMAO. New opportunities for functional food development. Food Funct. 2020;11(8):6745–76.
Salzano A, Israr MZ, Yazaki Y, Heaney LM, Kanagala P, Singh A, Arnold JR, Gulsin GS, Squire IB, McCann GP, et al. Combined use of trimethylamine N-oxide with BNP for risk stratification in heart failure with preserved ejection fraction: findings from the DIAMONDHFpEF study. Eur J Prev Cardiol. 2020;27(19):2159–62.
Kinugasa Y, Nakamura K, Kamitani H, Hirai M, Yanagihara K, Kato M, Yamamoto K. Trimethylamine N-oxide and outcomes in patients hospitalized with acute heart failure and preserved ejection fraction. ESC Heart Fail. 2021;8(3):2103–10.
Silva YP, Bernardi A, Frozza RL. The role of short-chain fatty acids from gut microbiota in gut-brain communication. Front Endocrinol (Lausanne). 2020;11:25.
den Besten G, Bleeker A, Gerding A, van Eunen K, Havinga R, van Dijk TH, Oosterveer MH, Jonker JW, Groen AK, Reijngoud D-J, et al. Short-chain fatty acids protect against high-fat diet-induced obesity via a PPARγ-dependent switch from lipogenesis to fat oxidation. Diabetes. 2015;64(7):2398–408.
Gao Z, Yin J, Zhang J, Ward RE, Martin RJ, Lefevre M, Cefalu WT, Ye J. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes. 2009;58(7):1509–17.
Challa AA, Lewandowski ED. Short-chain carbon sources: exploiting pleiotropic effects for heart failure therapy. JACC Basic Transl Sci. 2022;7(7):730–42.
Millard AL, Mertes PM, Ittelet D, Villard F, Jeannesson P, Bernard J. Butyrate affects differentiation, maturation and function of human monocyte-derived dendritic cells and macrophages. Clin Exp Immunol. 2002;130(2):245–55.
Nastasi C, Candela M, Bonefeld CM, Geisler C, Hansen M, Krejsgaard T, Biagi E, Andersen MH, Brigidi P, Ødum N, et al. The effect of short-chain fatty acids on human monocyte-derived dendritic cells. Sci Rep. 2015;5:16148.
Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504(7480):451–5.
Huang W, Guo H-L, Deng X, Zhu T-T, Xiong J-F, Xu Y-H, Xu Y. Short-Chain fatty acids inhibit oxidative stress and inflammation in mesangial cells induced by high glucose and lipopolysaccharide. Exp Clin Endocrinol Diabetes 2017, 125(2).
Berezin AE, Berezin AA, Lichtenauer M. Emerging role of adipocyte dysfunction in inducing heart failure among obese patients with prediabetes and known diabetes mellitus. Front Cardiovasc Med. 2020;7:583175.
Crum Y, Hoendermis ES, van Veldhuisen DJ, van Woerden G, Lobeek M, Dickinson MG, Meems LMG, Voors AA, Rienstra M, Gorter TM. Epicardial adipose tissue and pericardial constraint in heart failure with preserved ejection fraction. ESC Heart Fail. 2024;11(3):1698–706.
Alcaide P, Kallikourdis M, Emig R, Prabhu SD. Myocardial inflammation in heart failure with reduced and preserved ejection fraction. Circ Res. 2024;134(12):1752–66.
Dronkers J, van Veldhuisen DJ, van der Meer P, Meems LMG. Heart failure and obesity: unraveling molecular mechanisms of excess adipose tissue. J Am Coll Cardiol. 2024;84(17):1666–77.
Paterek A, Załęska-Kocięcka M, Wojdyńska Z, Kalisz M, Litwiniuk A, Leszek P, Mączewski M. Epicardial fat in heart failure-friend, foe, or bystander. Obes Rev. 2024;25(12):e13820.
Salvatore T, Galiero R, Caturano A, Vetrano E, Rinaldi L, Coviello F, Di Martino A, Albanese G, Colantuoni S, Medicamento G et al. Dysregulated epicardial adipose tissue as a risk factor and potential therapeutic target of heart failure with preserved ejection fraction in diabetes. Biomolecules 2022, 12(2).
Ayton SL, Gulsin GS, McCann GP, Moss AJ. Epicardial adipose tissue in obesity-related cardiac dysfunction. Heart. 2022;108(5):339–44.
Iacobellis G. Epicardial adipose tissue in contemporary cardiology. Nat Rev Cardiol. 2022;19(9):593–606.
Pugliese NR, Paneni F, Mazzola M, De Biase N, Del Punta L, Gargani L, Mengozzi A, Virdis A, Nesti L, Taddei S, et al. Impact of epicardial adipose tissue on cardiovascular haemodynamics, metabolic profile, and prognosis in heart failure. Eur J Heart Fail. 2021;23(11):1858–71.
Jin X, Hung CL, Tay WT, Soon D, Sim D, Sung KT, Loh SY, Lee S, Jaufeerally F, Ling LH, et al. Epicardial adipose tissue related to left atrial and ventricular function in heart failure with preserved versus reduced and mildly reduced ejection fraction. Eur J Heart Fail. 2022;24(8):1346–56.
Schulz A, Backhaus SJ, Lange T, Evertz R, Kutty S, Kowallick JT, Hasenfuß G, Schuster A. Impact of epicardial adipose tissue on cardiac function and morphology in patients with diastolic dysfunction. ESC Heart Fail. 2024;11(4):2013–22.
Wu A, Yang Z, Zhang X, Lin Z, Lu H. Association between epicardial adipose tissue and left atrial and ventricular function in patients with heart failure: a systematic review and Meta-Analysis. Curr Probl Cardiol. 2023;48(12):101979.
Tromp J, Bryant JA, Jin X, van Woerden G, Asali S, Yiying H, Liew OW, Ching JCP, Jaufeerally F, Loh SY, et al. Epicardial fat in heart failure with reduced versus preserved ejection fraction. Eur J Heart Fail. 2021;23(5):835–8.
Rämö JT, Kany S, Hou CR, Friedman SF, Roselli C, Nauffal V, Koyama S, Karjalainen J, Maddah M, Palotie A, et al. Cardiovascular significance and genetics of epicardial and pericardial adiposity. JAMA Cardiol. 2024;9(5):418–27.
Wang W, Gao Y, Wang J, Ji C, Gu H, Yuan X, Yang S, Wang X. Prognostic value of epicardial adipose tissue in heart failure with Mid-Range and preserved ejection fraction: a multicenter study. J Am Heart Assoc. 2024;13(24):e036789.
Hsu JC, Huang KC, Lin TT, Lee JK, Su MM, Juang JJ, Wu CK, Lin LY. Epicardial adipose tissue is associated with geometry alteration and diastolic dysfunction in prediabetic cardiomyopathy. J Clin Endocrinol Metab 2024.
Nesti L, Pugliese NR, Chiriacò M, Trico D, Baldi S, Natali A. Epicardial adipose tissue thickness is associated with reduced peak oxygen consumption and systolic reserve in patients with type 2 diabetes and normal heart function. Diabetes Obes Metab. 2023;25(1):177–88.
Uygur B, Çelik Ö, Demir AR, Karakayalı M, Arslan Ç, Otcu Temur H, Alis D, Yıldırım C, Çörekçioğlu B, Ertürk M. Epicardial adipose tissue volume predicts long term major adverse cardiovascular events in patients with type 2 diabetes. Turk Kardiyol Dern Ars. 2021;49(2):127–34.
Dhore-Patil A, Urina-Jassir D, Samson R, Le Jemtel TH, Oparil S. Epicardial adipose tissue thickness and preserved ejection fraction heart failure. Curr Hypertens Rep. 2024;26(9):381–8.
Liu Z, Hu W, Zhang H, Tao H, Lei P, Liu J, Yu Y, Dong Q, Gao L, Zhang D. EAT thickness as a predominant feature for evaluating arterial stiffness in patients with heart failure with preserved ejection fraction. Diabetes Metab Syndr Obes. 2022;15:1217–26.
Sugita Y, Ito K, Sakurai S, Sakai S, Kuno S. Epicardial adipose tissue is tightly associated with exercise intolerance in patients with type 2 diabetes mellitus with asymptomatic left ventricular structural and functional abnormalities. J Diabetes Complications. 2020;34(5):107552.
Chin JF, Aga YS, Abou Kamar S, Kroon D, Snelder SM, van de Poll SWE, Kardys I, Brugts JJ, de Boer RA, van Dalen BM. Association between epicardial adipose tissue and cardiac dysfunction in subjects with severe obesity. Eur J Heart Fail. 2023;25(11):1936–43.
van Woerden G, van Veldhuisen DJ, Manintveld OC, van Empel VPM, Willems TP, de Boer RA, Rienstra M, Westenbrink BD, Gorter TM. Epicardial adipose tissue and outcome in heart failure with Mid-Range and preserved ejection fraction. Circ Heart Fail. 2022;15(3):e009238.
Obokata M, Reddy YNV, Pislaru SV, Melenovsky V, Borlaug BA. Evidence supporting the existence of a distinct obese phenotype of heart failure with preserved ejection fraction. Circulation. 2017;136(1):6–19.
Liu J, Yu Q, Li Z, Zhou Y, Liu Z, You L, Tao L, Dong Q, Zuo Z, Gao L, et al. Epicardial adipose tissue density is a better predictor of cardiometabolic risk in HFpEF patients: a prospective cohort study. Cardiovasc Diabetol. 2023;22(1):45.
Ayton SL, Yeo JL, Gulsin GS, Dattani A, Bilak J, Deshpande A, Arnold JR, Singh A, Graham-Brown MPM, Ng L, et al. Association of epicardial adipose tissue with early structural and functional cardiac changes in type 2 diabetes. Eur J Radiol. 2024;174:111400.
Choy M, Huang Y, Peng Y, Liang W, He X, Chen C, Li J, Zhu W, Wei FF, Dong Y, et al. Association between epicardial adipose tissue and incident heart failure mediating by alteration of natriuretic peptide and myocardial strain. BMC Med. 2023;21(1):117.
He S, Zhu H, Zhang J, Wu X, Zhao L, Yang X. Proteomic analysis of epicardial adipose tissue from heart disease patients with concomitant heart failure with preserved ejection fraction. Int J Cardiol. 2022;362:118–25.
He S, Zhao L, Zhang J, Yang X, Zhu H. Identification of molecular signatures in epicardial adipose tissue in heart failure with preserved ejection fraction. ESC Heart Fail. 2024;11(5):2510–20.
Zhao L, Peng Y, Su P. Expression profiles and functional analysis of tRNA-derived small RNAs in epicardial adipose tissue of patients with heart failure. Ann Med. 2023;55(2):2267981.
Zheng ML, Du XP, Zhao L, Yang XC. Expression profile of circular RNAs in epicardial adipose tissue in heart failure. Chin Med J Engl. 2020;133(21):2565–72.
Gao Q, He S, Peng Y, Su P, Zhao L. Proteomic profiling of epicardial fat in heart failure with preserved versus reduced and mildly reduced ejection fraction. J Cell Mol Med. 2023;27(5):727–35.
van Woerden G, van Veldhuisen DJ, Westenbrink BD, de Boer RA, Rienstra M, Gorter TM. Connecting epicardial adipose tissue and heart failure with preserved ejection fraction: mechanisms, management and modern perspectives. Eur J Heart Fail. 2022;24(12):2238–50.
Yang CD, Quan JW, Tay GP, Feng S, Yuan H, Amuti A, Tang SY, Wu XR, Yuan RS, Lu L, et al. Epicardial adipose tissue volume and density are associated with heart failure with improved ejection fraction. Cardiovasc Diabetol. 2024;23(1):283.
Shi YJ, Dong GJ, Guo M. Targeting epicardial adipose tissue: a potential therapeutic strategy for heart failure with preserved ejection fraction with type 2 diabetes mellitus. World J Diabetes. 2023;14(6):724–40.
Pugliese NR, Pellicori P, Filidei F, De Biase N, Maffia P, Guzik TJ, Masi S, Taddei S, Cleland JGF. Inflammatory pathways in heart failure with preserved left ventricular ejection fraction: implications for future interventions. Cardiovasc Res. 2023;118(18):3536–55.
Sanders-van Wijk S, Tromp J, Beussink-Nelson L, Hage C, Svedlund S, Saraste A, Swat SA, Sanchez C, Njoroge J, Tan RS, et al. Proteomic evaluation of the comorbidity-inflammation paradigm in heart failure with preserved ejection fraction: results from the PROMIS-HFpEF study. Circulation. 2020;142(21):2029–44.
Smolgovsky S, Bayer AL, Kaur K, Sanders E, Aronovitz M, Filipp ME, Thorp EB, Schiattarella GG, Hill JA, Blanton RM et al. Impaired T cell IRE1α/XBP1 signaling directs inflammation in experimental heart failure with preserved ejection fraction. J Clin Invest 2023, 133(24).
Panico C, Felicetta A, Kunderfranco P, Cremonesi M, Salvarani N, Carullo P, Colombo F, Idini A, Passaretti M, Doro R, et al. Single-cell RNA sequencing reveals metabolic stress-dependent activation of cardiac macrophages in a model of dyslipidemia-induced diastolic dysfunction. Circulation. 2024;150(19):1517–32.
Jenkins BJ, Blagih J, Ponce-Garcia FM, Canavan M, Gudgeon N, Eastham S, Hill D, Hanlon MM, Ma EH, Bishop EL, et al. Canagliflozin impairs T cell effector function via metabolic suppression in autoimmunity. Cell Metab. 2023;35(7):1132–e11461139.
Hulsmans M, Sager HB, Roh JD, Valero-Muñoz M, Houstis NE, Iwamoto Y, Sun Y, Wilson RM, Wojtkiewicz G, Tricot B, et al. Cardiac macrophages promote diastolic dysfunction. J Exp Med. 2018;215(2):423–40.
Zhang XL, Wang TY, Chen Z, Wang HW, Yin Y, Wang L, Wang Y, Xu B, Xu W. HMGB1-Promoted neutrophil extracellular traps contribute to cardiac diastolic dysfunction in mice. J Am Heart Assoc. 2022;11(4):e023800.
Mang G, Chen J, Sun P, Ma R, Du J, Wang X, Cui J, Yang M, Tong Z, Yan X, et al. Von Willebrand factor exacerbates heart failure through formation of neutrophil extracellular traps. Eur Heart J. 2024;45(37):3853–67.
Yang K, Gao R, Chen H, Hu J, Zhang P, Wei X, Shi J, Chen Y, Zhang L, Chen J, et al. Myocardial reperfusion injury exacerbation due to ALDH2 deficiency is mediated by neutrophil extracellular traps and prevented by leukotriene C4 Inhibition. Eur Heart J. 2024;45(18):1662–80.
Šestan M, Raposo B, Rendas M, Brea D, Pirzgalska R, Rasteiro A, Aliseychik M, Godinho I, Ribeiro H, Carvalho T, et al. Neuronal-ILC2 interactions regulate pancreatic glucagon and glucose homeostasis. Science. 2025;387(6731):eadi3624.
Pieske B, Tschöpe C, de Boer RA, Fraser AG, Anker SD, Donal E, Edelmann F, Fu M, Guazzi M, Lam CSP, et al. How to diagnose heart failure with preserved ejection fraction: the HFA-PEFF diagnostic algorithm: a consensus recommendation from the heart failure association (HFA) of the European society of cardiology (ESC). Eur J Heart Fail. 2020;22(3):391–412.
Shah SJ, Katz DH, Selvaraj S, Burke MA, Yancy CW, Gheorghiade M, Bonow RO, Huang C-C, Deo RC. Phenomapping for novel classification of heart failure with preserved ejection fraction. Circulation. 2015;131(3):269–79.
Cohen JB, Schrauben SJ, Zhao L, Basso MD, Cvijic ME, Li Z, Yarde M, Wang Z, Bhattacharya PT, Chirinos DA, et al. Clinical phenogroups in heart failure with preserved ejection fraction: detailed phenotypes, prognosis, and response to spironolactone. JACC Heart Fail. 2020;8(3):172–84.
Solomon SD, McMurray JJV, Claggett B, de Boer RA, DeMets D, Hernandez AF, Inzucchi SE, Kosiborod MN, Lam CSP, Martinez F, et al. Dapagliflozin in heart failure with mildly reduced or preserved ejection fraction. N Engl J Med. 2022;387(12):1089–98.
Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Böhm M, Brunner-La Rocca H-P, Choi D-J, Chopra V, Chuquiure-Valenzuela E, et al. Empagliflozin in heart failure with a preserved ejection fraction. N Engl J Med. 2021;385(16):1451–61.
Salvatore T, Galiero R, Caturano A, Rinaldi L, Di Martino A, Albanese G, Di Salvo J, Epifani R, Marfella R, Docimo G et al. An overview of the cardiorenal protective mechanisms of SGLT2 inhibitors. Int J Mol Sci 2022, 23(7).
Wen Y, Zhang X, Liu H, Ye H, Wang R, Ma C, Duo T, Wang J, Yang X, Yu M, et al. SGLT2 inhibitor downregulates ANGPTL4 to mitigate pathological aging of cardiomyocytes induced by type 2 diabetes. Cardiovasc Diabetol. 2024;23(1):430.
Kosiborod MN, Abildstrøm SZ, Borlaug BA, Butler J, Rasmussen S, Davies M, Hovingh GK, Kitzman DW, Lindegaard ML, Møller DV, et al. Semaglutide in patients with heart failure with preserved ejection fraction and obesity. N Engl J Med. 2023;389(12):1069–84.
Kosiborod MN, Petrie MC, Borlaug BA, Butler J, Davies MJ, Hovingh GK, Kitzman DW, Møller DV, Treppendahl MB, Verma S, et al. Semaglutide in patients with obesity-related heart failure and type 2 diabetes. N Engl J Med. 2024;390(15):1394–407.
Withaar C, Meems LMG, Nollet EE, Schouten EM, Schroeder MA, Knudsen LB, Niss K, Madsen CT, Hoegl A, Mazzoni G, et al. The cardioprotective effects of semaglutide exceed those of dietary weight loss in mice with HFpEF. JACC Basic Transl Sci. 2023;8(10):1298–314.
France NL, Syed YY. Tirzepatide: a review in type 2 diabetes. Drugs. 2024;84(2):227–38.
Packer M, Zile MR, Kramer CM, Baum SJ, Litwin SE, Menon V, Ge J, Weerakkody GJ, Ou Y, Bunck MC, et al. Tirzepatide for heart failure with preserved ejection fraction and obesity. N Engl J Med. 2025;392(5):427–37.
Acknowledgements
We would like to express our gratitude to the Figdraw for providing drawing materials.
Funding
The work was supported by the National Natural Science Foundation of China (No. 82400428 to LD) and the Natural Science Foundation Program of Hubei Province (No.2024AFB062 to LD).
Author information
Authors and Affiliations
Contributions
LD contributes to the conception and design of the work; YYZ and LYL have drafted the work; LD, YYZ and LYL substantively revised it. All authors have approved the submitted version.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
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
Luo, L., Zuo, Y. & Dai, L. Metabolic rewiring and inter-organ crosstalk in diabetic HFpEF. Cardiovasc Diabetol 24, 155 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12933-025-02707-7
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12933-025-02707-7