- Review
- Open access
- Published:
The role of estrogen in the sex difference for the risk factors of heart failure with preserved ejection fraction
Biology Direct volume 20, Article number: 28 (2025)
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major subtype of heart failure, primarily characterized by a normal or mildly reduced left ventricular ejection fraction along with left ventricular diastolic dysfunction. Recent studies have shown that the prevalence of HFpEF is higher in women than that in men, particularly in postmenopausal women. Concurrently, it has been observed that the incidence of risk factors contributing to HFpEF (such as obesity, hypertension, diabetes, and atrial fibrillation) also notably increases post-menopause, affecting the incidence of HFpEF. This review aimed to examine the relationship between estrogen and risk factors associated with HFpEF, clarifying the underlying mechanisms through which estrogen affects these risk factors from epidemiological and pathophysiological perspectives. This review also provides a comprehensive understanding of the association between estrogen and the risk factors for HFpEF, thus helping explore potential targets for HFpEF treatment.
Heart failure (HF) is a global health concern with high incidence and mortality rates. Approximately 2% of the developed Western population suffers from HF, and the prevalence of the disease rapidly increases with age [1,2,3]. Based on the left ventricular ejection fraction (EF) at the time of diagnosis, HF is classified into three types: heart failure with reduced ejection fraction (HFrEF), which refers to an EF less than 40%; heart failure with mid-range ejection fraction (HFmrEF), defined as an EF between 41 and 49%; and heart failure with preserved ejection fraction (HFpEF), ranging from EF greater than 50% [4]. Recent epidemiological studies have found that HFpEF accounts for at least 50% of all HF cases, with an increasing incidence and female preference [5]. Among patients with HFpEF, the number of female patients is nearly twice that of male patients, whereas male patients with HF primarily exhibit a reduced ejection fraction [6]. Data from a European population revealed that the incidence of both types of HF increases with age, and the incidence of HFpEF increases more rapidly with age than that of HFrEF. However, at any age, the incidence of HFpEF is higher in women than that in men, particularly in postmenopausal women [5]. Among individuals aged 25–49 years, the incidence rates, when stratified by age and sex, increased from 0% (males) and 1% (females) to 4–6% in males and 8–10% in females aged 80 years and above [7].
Biological sex is a negligible factor that significantly affects the structure and function of the cardiovascular system. The XY sex chromosome, sex-specific gene expression, sex hormones, and sex reproductive organs show the most significant differences, which may contribute to the pathology of heart diseases [8]. Estrogen is the major sex hormone responsible for this sex-specific difference. Notably, the increased risk of cardiovascular diseases risk in postmenopausal women suggests that changes in estrogen levels may contribute to the occurrence and development of HFpEF in females, further indicating a potential protective role of estrogen in patients with HFpEF [9].
The prevalence of HFpEF is closely associated with risk factors, such as hypertension, obesity, diabetes, and atrial fibrillation (AF), as shown in Fig. 1. The increase in the risk associated with these factors shows an upward trend with age, especially in the female population, significantly increasing with a decline in hormone levels during menopause. Therefore, this review aimed to clarify the relationship between estrogen and these risk factors for HFpEF, providing novel therapeutic targets to combat this devastating disease worldwide from a sex perspective.
Estrogen and HFpEF-related risk factors. Common risk factors of HFpEF include obesity, hypertension, diabetes, and atrial fibrillation. As estrogen levels in women decline, the prevalence and severity of these risk factors continue to increase, directly or indirectly influencing the onset and progression of HFpEF
Interplay among estrogen, obesity, and HFpEF
Obesity is a major risk factor for HFpEF [10]. Globally, nearly 880 million adults are obese, and this number continues to rise annually [11]. The global prevalence of obesity is higher in women than that in men [12], and there is a sex bias in body fat distribution and energy homeostasis [13]. The number of women with severe obesity is approximately as many as twice the number of men (i.e., class 2 obesity with a body mass index (BMI) ≥ 35 kg/m2 and class 3 obesity with a BMI ≥ 40 kg/m2) [14]. Statistics show that the obesity rate among American women aged 40–59 years is 65%, and for women aged 60 years and above, it is as high as 73.8%, demonstrating an increasing trend of being overweight and obese in postmenopausal women [15,16,17]. It is well known that obesity greatly elevates the risks of cardiovascular disease (CVD) and even HF [18], which is particularly evident in HFpEF throughout an individual’s lifetime [19, 20]. Epidemiological studies indicate that nearly 80% of patients with HFpEF are overweight or obese (i.e., BMI ≥ 25 kg/m2) [21, 22]. Among patients with HF, a higher BMI is closely associated with HFpEF [23]. A large study comprising 22,618 individuals from four community cohorts (the Cardiovascular Health Study, PREVENT, Framingham Heart Study, and MESA) revealed that there is a 34% increase in the incidence of HFpEF with every 1 kg/m2 increase in BMI [6, 23, 24]. Obesity and the resulting cardiac metabolic changes significantly elevate the risk of HFpEF in women, implying a stronger link between obesity and HFpEF in women than that in men [23].
Obesity leads to hemodynamic changes, resulting in compensatory alterations in cardiac morphology and ventricular function. These symptoms include increased cardiac output, left ventricular hypertrophy, and impaired diastolic and systolic function in both ventricles [25]. The diastolic function may also be impaired by limiting energy utilization efficiency or increasing myocardial lipid content [26, 27], the latter of which is associated with increased left ventricular diastolic tension [28, 29]. Excess adipose tissue (AT) often leads to plasma volume expansion and impaired left ventricular relaxation, which is likely mediated by systemic inflammation and microvascular rarefaction [30]. These changes can increase left ventricular filling pressure, manifesting as signs and symptoms of HF, and ultimately resulting in restricted ventricular dilation [31, 32].
Moreover, obesity influences the neurohormonal system, promoting the development of HFpEF by increasing aldosterone and leptin levels and suppressing natriuretic peptide activity [33,34,35]. This modulation occurs by activating the renin–angiotensin–aldosterone system (RAAS), which elevates aldosterone synthesis. Such activation triggers a cascade of downstream effects, including sodium retention, plasma volume expansion, elevated systemic inflammation, intensified kidney and heart fibrosis, increased arterial stiffness, and deterioration of left ventricular diastolic function [19, 33, 36, 37]. Patients with obesity and HFpEF are highly likely to experience diastolic dysfunction, as well as cardiac enlargement due to increased epicardial fat, leading to pericardial restraint and enhanced ventricular interaction [19, 38]. Compared with non-obese patients with HFpEF and controls, those with obesity and HFpEF have worse exercise capacity, elevated biventricular filling pressures during exercise, reduced pulmonary arterial vasodilator reserves, inadequate cardiac output, and decreased cardiopulmonary function during exercise [19].
Although it was once believed that overall fat accumulation increases cardiovascular risk, the detrimental effects of obesity on the cardiovascular system may also be linked to AT distribution within the body [39]. Obesity can be classified based on fat distribution into peripheral obesity, with fat predominantly distributed in the hips and limbs as subcutaneous AT, and central obesity, with fat primarily distributed around the abdominal viscera as visceral AT. Central obesity is a major risk factor for CVD, whereas peripheral fat appears to offer protective effects [40,41,42]. AT distribution is influenced by multiple factors, and sex hormone levels play a significant role [43]. Typically, men have a lower total fat mass but a higher proportion of abdominal fat than women. In contrast, women tend to have more total fat distributed predominantly as subcutaneous fat in the hips, thighs, and other peripheral areas than men [44].
In premenopausal women, estrogen (E2) promotes subcutaneous fat accumulation, inhibits visceral fat deposition, and reduces CVD risk [45]. Estrogen acts in synergy with AT genes, increasing the subcutaneous AT mass in reproductive-aged women and reducing the visceral AT mass, thereby producing a protective cardiac metabolic effect. However, the loss of estrogen after menopause increases the total AT mass, partially reversing the protective distribution of AT in women [46]. When estrogen levels become sufficiently low, women experience visceral fat accumulation, possibly due to the direct effects of estrogen on AT distribution [47]. After menopause, subcutaneous AT and visceral AT levels both increase, with the relative rise in visceral AT being twice that of total AT [48]. Evidence has shown that postmenopausal women are three times more likely to develop obesity-related metabolic syndrome than premenopausal women [49]. Fat distribution in women also shifts significantly with changes in estrogen levels, with central obesity becoming more ronounced in postmenopausal women. Metabolic abnormalities are also more closely related to visceral fat accumulation and plasma estrogen levels than to BMI alone [50]. During aging, while subcutaneous fat deposition occurs universally in women, visceral adiposity progressively dominates, posing a heightened risk for metabolic and cardiovascular complications [51].
The epicardial adipose tissue (EAT), a specialized form of visceral fat, is located within the pericardium and closely surrounds the myocardium. They are both metabolically active complex tissues [52]. In addition to storing triglycerides to supply energy to the myocardium, the EAT also secretes pro-inflammatory adipokines and pro-oxidative substances, contributing to adverse cardiac effects [53, 54]. The EAT exerts primarily detrimental effects on the heart by (i) promoting inflammation and driving related complications and (ii) facilitating myocardial interactions that result in diastolic dysfunction [53, 54].
The decline in estrogen levels after menopause exacerbates obesity and inflammation via various mechanisms. Reduced estrogen levels not only directly alter fat distribution and increase visceral fat accumulation but also promote systemic low-grade inflammation and a local pro-inflammatory environment, worsening endothelial dysfunction [55, 56]. Adipose tissues, including EAT, secrete higher levels of pro-inflammatory cytokines, such as TNF-α, IL-6, and CRP [57,58,59], which reduce the bioavailability of nitric oxide (NO). As a result, microvascular dysfunction, impaired diastolic function, and myocardial fibrosis are more pronounced compared to individuals with lower levels of these pro-inflammatory cytokines. [60, 61]. Collectively, these processes drive the development of HFpEF. Estrogen/progesterone hormone replacement therapy has been shown to reduce visceral AT, fasting blood glucose, and insulin levels in postmenopausal women [43, 44]. Therefore, estrogen-related therapy may mitigate the increased cardiovascular risk after menopause by reducing the risk of obesity and its associated detrimental effects, offering a potential strategy for HFpEF prevention and management.
Interplay among estrogen, hypertension, and HFpEF
Patients with long-standing hypertension show a significantly increased risk of HF, which is a significant and highly associated clinical consequence of hypertension-mediated organ damage [62,63,64]. The incidence of hypertension in premenopausal women is usually lower than that in men of the same age; however, after menopause, the incidence of hypertension in women sharply increases, leading to the gradual disappearance of this sex difference [63]. As age increases, women become more susceptible to hypertension than men, especially in the 65–74-year-old age group, where 45% of women and 41% of men are reported to have hypertension, with the incidence in women surpassing that in men [65, 66]. Hypertension is common in females over 60 years of age [67], with sex-associated steroid hormones identified as the key players behind this sex-related difference [68]. It is known that E2 has potent acute and chronic vasodilatory activity, ultimately leading to blood pressure (BP) reduction [69,70,71]. Most animal studies indicate that E2 is involved in various mechanisms preventing hypertension, such as stimulating NO-mediated vasodilator pathway [72, 73]. Previous research suggests that all three types of ERs have protective effects on the cardiovascular system. Specifically, the activation of ERα has been shown to reduce endothelial dysfunction [74], while the activation of ERβ can lower BP, vascular constriction, and vascular resistance, and alleviate cardiac hypertrophy [75, 76]. Additionally, GPR30 lowers BP, stimulates vasodilation, and reduces vascular smooth muscle cell proliferation and migration [77]. The action of estrogen may be beneficial for premenopausal women, exerting a protective effect on the cardiovascular system, as shown in Fig. 2 [71]. In contrast, its loss may render the hearts of postmenopausal women more vulnerable to cardiovascular risks [78].
Mechanisms by which estrogen influences hypertension. E2 binds to membrane GPR30, activating the MAPK/ERK or PI3K pathways, enhancing eNOS activity, and rapidly increasing NO production, leading to vasodilation and blood pressure reduction. Simultaneously, E2 inhibits the NF-κB pathway, reducing the expression of inflammatory mediators and mitigating damage to the vascular endothelium. E2 also interacts with ERα and ERβ, which then translocate to the nucleus and bind directly to estrogen response elements, regulating the transcription of target genes, such as VEGF, to promote angiogenesis and eNOS for vasodilation and blood pressure regulation
This notable sex difference also affects HF progression. In the Framingham Heart Study, after adjusting for age, the risk of HF in hypertensive patients increased twofold in men and 3.2-fold in women [79, 80]. As the incidence of hypertension continues to increase, it primarily leads to a higher occurrence of HFpEF in women and a higher occurrence of HFrEF in men [81, 82]. Proper BP control is widely recognized as the cornerstone in the prevention and clinical management of HFpEF [83,84,85]. Randomized clinical trials have shown that following the strategies outlined in the American College of Cardiology (ACC)/American Heart Association (AHA) hypertension treatment guidelines can reduce the incidence of HFpEF by 40% in the next 2–8 years of life [86, 87]. Further epidemiological investigations have found that the incidence of hypertension in patients with HFpEF ranges from 55 to 90% [88,89,90]. Compared to patients with HFrEF, those with HFpEF are more likely to have a history of hypertension [81, 91]. In patients diagnosed with HFpEF, hypertension may be the sole cause or one of several causes or comorbidities.
Hypertension causes left ventricular hypertrophy, diastolic dysfunction, left ventricular fibrosis, left atrial dilation, and macrovascular and microvascular sclerosis [92,93,94]. In females, hypertension induces concentric hypertrophy of the left ventricle, leading to diastolic dysfunction, and the clinical course of HF is generally benign and often characterized by HFpEF [80, 82]. In males, eccentric hypertrophy with chamber dilatation is more common and ultimately leads to systolic dysfunction [95,96,97]. At any given age, the aortic root diameter in females is smaller than that in males. This may indicate a relatively delayed external remodeling response of the aortic arch during hemodynamic load, potentially leading to increased pulse pressure [98]. Increased load-induced impairment of left ventricular diastolic function makes females more susceptible to the adverse effects of arteriosclerosis and early wave reflection on left ventricular diastolic function [99]. Arterial-ventricular coupling in males remains stable with age, whereas it decreases with age in women [99]. This difference may exacerbate the disadvantages of hypertension and HFpEF in women. Females are also more prone to idiopathic pulmonary arterial hypertension with enhanced pulmonary arterial vasoconstriction and intrinsic pulmonary arterial remodeling, affecting them four times more than males [100]. Compared to HFpEF patients without pulmonary hypertension (PH), patients with PH-HFpEF are more often female, have more severe symptoms, and more frequently exhibit right ventricular hypertrophy and right atrial dilation with higher right atrial pressures [101]. In summary, the interplay between hypertension and HFpEF is influenced by notable sex differences, with females exhibiting unique pathophysiological responses, such as greater susceptibility to diastolic dysfunction and PH, underscoring the need for sex-specific approaches in the prevention and management of HFpEF.
Interplay among estrogen, diabetes, and HFpEF
Approximately 30.3 million individuals in the United States currently have diabetes, with an additional 84.1 million categorized as prediabetic, and the prevalence of diabetes is projected to increase by 54% by 2030 [102, 103]. As of 2023, China, the country with the largest number of patients with diabetes worldwide, has over 118 million people affected by this disease [104]. Individuals with diabetes exhibit a significantly elevated incidence of HF compared with their non-diabetic counterparts, with an approximately 2–fivefold increased risk [105, 106]. It is estimated that diabetes coexists in approximately 30–40% of patients with HF and a deteriorated clinical condition and higher all-cause and cardiovascular mortality than that in patients with HF without type 2 diabetes (T2D) [107].
Significant sex differences have also been observed in the development of diabetes, particularly T2D. Overall, the incidence of T2D is higher in men than in women, especially in the middle-aged and young populations. However, as age increases, the incidence of diabetes in women gradually increases, surpassing that in men after 70Â years of age [108,109,110]. Furthermore, the early onset of diabetes in women may be more insidious; the age-specific prevalence of previously undiagnosed diabetes and impaired glucose tolerance (IGT) defined by isolated post-load hyperglycemia is higher in women than that in men [110]. Additionally, women may be more prone to developing diabetes-related complications, such as CVD, than men [111, 112]. Additionally, gestational diabetes is a risk factor specific to women, as those with gestational diabetes have a higher risk of developing T2D in the future [113]. Moreover, women with a history of GDM appear to have a nearly tenfold higher risk of developing T2D than those with normoglycemic pregnancies [114].
Similarly, significant sex-based differences were observed in the transition from diabetes to HF. Based on a study that followed 5209 men and women aged 30–62 years for 18 years and explored the relationship between the incidence of congestive HF and previous diabetes status, the results showed that diabetes increased the risk of HF more significantly in women than that in men, with the risk increasing fivefold in women compared to a 2.4-fold increase in men [115]. Furthermore, women with diabetes have a significantly higher risk of developing congestive HF than men. Even after adjusting for other factors, such as age, BP, and cholesterol, sex differences remained evident [115]. These findings suggest that the risk of death associated with T2D is significantly higher in women than that in men [116]. Two population-based studies conducted in Scotland and Sweden confirmed this trend, revealing an elevated mortality risk due to T2D in both sexes, with a markedly greater effect in women [117, 118]. Among patients diagnosed with HF and T2DM, nearly half are identified with HFpEF, particularly among older participants and especially in the cohort of females with hypertension and T2D [119].
The molecular mechanisms through which DM contributes to the development of HFpEF are complex and diverse. From a pathophysiological perspective, the occurrence of HFpEF is often highly correlated with pathological features induced by diabetes, such as volume overload, peripheral injury, metabolic disorders, release of pro-inflammatory factors, and skeletal muscle damage. Patients with diabetes often exhibit excessive activation of the neurohormonal system and altered sodium handling, potentially leading to cardiac congestion, cardiorenal syndrome, and reduced diuretic response [120]. In addition, hyperglycemia can lead to the upregulation of sodium-glucose co-transporter-2 (SGLT2), thereby increasing proximal renal sodium reabsorption, resulting in volume expansion and reduced diuretic response [121]. Patients with HFpEF and concomitant diabetes demonstrate manifestations of volume overload, often presenting with higher mechanical ventilation and requirements for dialysis/ultrafiltration, worse renal function, and prolonged hospital stays owing to increased in-hospital volume and poor diuretic response at discharge, leading to an increased rate of HF readmissions [102, 122]. In the EMPA-REG OUTCOME (BI 10773 [Empagliflozin] Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients) trial, empagliflozin was associated with a reduction in major adverse cardiovascular events and a significantly reduced rate of HF hospitalizations [123]. SGLT2 inhibitors (SGLT2is) promote diuresis and natriuresis by restoring sodium delivery to the distal renal tubule without activating the sympathetic nervous system. This achieves natriuretic therapy and alleviates volume overload, thereby reducing the incidence of HFpEF and DM in patients [124]. SGLT2is also reduce myocardial Na/H exchanger activity, reverse electrolyte imbalances, and increase the sodium ion concentration in the mitochondria of patients with HF [125, 126]. In addition, they improve myocardial metabolism by optimizing energy metabolism, enhancing oxygen delivery, promoting ATP storage, increasing mitochondrial oxygen uptake and conversion efficiency, and facilitating energy supply by utilizing glucose-derived ketone bodies [127, 128]. Furthermore, SGLT2is can reduce cardiomyocyte apoptosis and improve myocardial fibrosis by alleviating oxidative stress, decreasing TGF-β production, and regulating macrophage polarization [129, 130]. Insulin resistance in patients with diabetes may lead to increased utilization of free fatty acids by myocardial cells, potentially resulting in mitochondrial dysfunction, production of toxic lipid intermediates, and increased reactive oxygen species [131, 132]. High glucose-induced advanced glycation end products can impair microvascular function and reduce nitric oxide availability [131]. These processes may contribute to structural changes in myocardial cells, endothelial dysfunction, and HF.
In the female population, diabetes and the resulting HFpEF exhibit particularly distinct manifestations. Studies have demonstrated that women with diabetes often show higher levels of insulin resistance [133, 134] and are more susceptible to glucose intolerance and a persistent hyperglycemic state, whereas men with diabetes predominantly exhibit elevated fasting blood glucose levels [135]. Insulin resistance and persistent hyperglycemia can induce a systemic inflammatory response [136], aggravating cardiac fibrosis and diastolic dysfunction [137]. In women, the risk of diabetes is closely linked to a decline in estrogen levels, as lower E2 levels during menopausal transition are associated with an increased risk of diabetes, reflecting the effects of ovarian aging [138]. Research further suggests that postmenopausal women, despite enhanced insulin sensitivity, exhibit reduced non-insulin-dependent glucose-handling capabilities and diminished pancreatic insulin secretion, as indicated by decreased plasma C-peptide levels [139]. Longitudinal studies have highlighted that women undergoing oophorectomy (leading to abrupt and severe E2 deficiency) face a markedly higher risk of developing diabetes than those undergoing natural menopause, with a 57% increased risk of diabetes observed over 9 years [140, 141]. Diabetes research in female rodent models, including oophorectomized Sprague–Dawley rats, revealed worse glucose tolerance than in animals whose ovaries were not removed [142,143,144,145]. Large randomized controlled trials have indicated that menopausal hormone therapy (MHT) can reduce the incidence of T2D in women [142, 143]. However, given the complex balance between risks and benefits and because the impact of MHT on diabetes prevention is not a primary outcome in RCTs, MHT is neither appropriate nor FDA-approved for preventing T2D in women [144]. The role of estrogen under hyperglycemic conditions in female diabetic patients can also lead to sex-specific endothelial dysfunction, which is potentially linked to the estrogen-induced increase in PKCβ expression and O2− production [113], improving vasodilation capacity by elevating NO production, reducing vascular resistance, and aiding in the maintenance of the EF. Moreover, hyperglycemia can alter the balance of ERs, thereby increasing oxidative stress and vasoconstrictor levels [146,147,148].
Previous studies have confirmed that estrogen compounds effectively protect the survival and function of pancreatic β-cells in response to various diabetic injuries [149,150,151,152]. GLP-1 is an agonist that targets pancreatic β-cells and stimulates insulin secretion, thereby lowering blood glucose levels [153]. As both GLP-1 and E2 have beneficial effects in diabetes treatment, some studies have proposed combining these two therapeutic strategies to exploit their synergistic effects, offering a new treatment approach for diabetes and related cardiovascular diseases such as HFpEF.
Compared to single GLP-1 or E2 agonists, GLP1-E2 combination therapy showed greater efficacy in lowering hyperglycemia, protecting β-cells, and alleviating diabetes-related complications [154,155,156]. Research has indicated that this combination therapy significantly enhances glucose-stimulated insulin secretion in human islets, outperforming single agonists [157]. The underlying mechanism of action involves GLP-1 entering β-cells through the GLP-1 receptor (GLP-1R), releasing E2 under lysosomal acidification, and activating ERα, inducing multiple protective signaling pathways. GLP1-E2 significantly enhances anti-apoptotic pathways (e.g., PI3K/Akt signaling), suppresses inflammation and oxidative stress, and improves insulin sensitivity and glucose metabolism, providing substantial protection for pancreatic β-cells [154]. Additionally, by precisely targeting the release of estrogen, GLP1-E2 avoids the systemic side effects associated with conventional estrogen therapy, such as adverse effects on the breast and uterus [158]. GLP1-E2 is particularly suitable for high-risk populations, such as postmenopausal women, and has great potential for preventing and managing diabetes and HFpEF. Future research should validate its safety and efficacy, optimize drug design to enhance therapeutic outcomes, and promote broader clinical applications.
Cardiac hypertrophy, myocardial wall thickening, and increased left ventricular mass are markedly pronounced in women with T2D [159]. A hallmark of diabetic cardiomyopathy is left ventricular hypertrophy, cardiac remodeling, progression of cardiac diastolic dysfunction, and subsequent clinical signs of HF with a normal EF [160]. By promoting the expression of the estrogen receptor ERα, activating the Akt signaling pathway, and inhibiting the TGF-β signaling pathway, estrogen regulates collagen expression and suppresses cardiac hypertrophy and fibrosis, ultimately protecting H9c2 cardiomyocytes from damage caused by advanced glycation end products (AGEs), preventing diabetes-induced cardiac fibrosis in db/db mice, and improving cardiac function [161]. Thus, estrogen and its receptors greatly affect the development of diabetes and associated HF, with changes in estrogen levels potentially increasing the risk of cardiac disease, leading to a increased incidence of HF.
Interplay among estrogen, AF, and HFpEF
Epidemiological studies have shown that the incidence of AF and HFpEF is increasing annually, with an increasing rate of comorbidities. For instance, there is a higher prevalence of AF in patients with HFpEF, ranging from 15 to 40%, depending on the research cohorts and diagnostic methods for AF [81]. AF has also been identified as a major risk factor of new-onset HFpEF, and its presence often predicts the occurrence of HFpEF more effectively than HFrEF [24]. Additionally, the development of AF may have a greater impact on patients with HFpEF than those with HFrEF and is highly associated with adverse outcomes in patients with HFpEF [162,163,164,165,166]. AF is an independent risk factor for new-onset HFpEF in women but not in men [167]. Furthermore, among the participants with AF, the incidence of HFpEF is higher in women than that in men [168].
AF is a common complication in patients with HFpEF, and a strong interplay exists between the two conditions. AF can serve as an early marker of HFpEF, potentially indicating changes in cardiac structure and function [169]. AF can lead to left ventricular dilation, impaired atrial function, and aggravated atrial fibrosis, which may directly trigger HFpEF [170]. Additionally, AF may result in irregular ventricular rates, particularly irregularities owing to variability in atrioventricular conduction, which may lead to decreased ventricular function and cardiac output [171]. More critically, AF may also cause serious complications, such as thrombus formation and embolism, further exacerbating the condition of patients with HFpEF [172].
In contrast, HFpEF promotes the development of AF. A recent study revealed that in a group of 450 patients newly diagnosed with HFpEF, after a median follow-up of 3.7Â years, 32% of patients progressed to AF. This translates to approximately 69 new cases of AF per 1000 patients annually [165]. In patients with HFpEF, the compliance and mechanics of the left atrium (LA) gradually deteriorate, leading to LA enlargement, potentially progressing to permanent AF [169]. The most common mechanism by which HFpEF leads to AF is structural and functional LA remodeling [173, 174]. Compared to age-matched control groups, the LA volume in patients with HFpEF significantly increases, which is more pronounced than that in patients with hypertensive heart disease without HF [173]. LA remodeling manifests differently in different HF subtypes. Although eccentric remodeling is more common in HFrEF, it is characterized by increased LA stiffness, which is more likely to trigger AF [175]. Abnormal ventricular diastolic function in patients with HFpEF may also lead to increased atrial pressure, triggering AF [169].
Although the incidence of AF is higher in men than that in women [176, 177], women exhibit more severe symptoms, including atypical symptoms, and have a worse quality of life. Furthermore, camapered to men, women face a higher risk of adverse events related to AF, such as stroke and death [178, 179]. Notably, women generally develop AF later than men [180]. Data from the Framingham Heart Study show that 74% of female patients with AF are aged ≥ 70 years, while the corresponding proportion in men is only 58% [181]. The incidence of AF is lower in premenopausal women; however, it significantly increases after menopause, especially in women aged > 50 years, suggesting a close association between changes in estrogen levels and the occurrence and development of AF [182]. The effect of estrogen replacement therapy (HRT) on AF has shown contradictory results across studies, indicating that the effect of this treatment on AF risk may be complex. According to long-term follow-up data from the Italian Tamoxifen Study Group, women treated with tamoxifen had a significantly higher risk of developing AF than those in the placebo group, with a relative risk of 1.74 [183], suggesting that certain hormone-related drugs may be associated with AF occurrence. In addition, the Women’s Health Study found that the use of estrogen-only replacement therapy (ERT) was associated with an increased risk of AF, whereas combined estrogen and progesterone therapy did not show a similar association [184]. This indicates that estrogen alone may contribute to an increased risk of AF. A retrospective cohort study further explored the impact of different types of HRT on the risk of AF and revealed that the incidence of AF, stroke, and major adverse cardiac events (MACE) was lower in the estradiol treatment group than that in the conjugated equine estrogen (CEE) treatment group. The incidence of AF was significantly high in women receiving CEE therapy, with an adjusted risk ratio of 1.96, suggesting that CEE may increase the risk of AF [185]. Meanwhile, in women surviving myocardial infarction, the incidence of AF was lower among those receiving HRT, especially in the group aged ≥ 80 years, where the risk of AF was significantly reduced in women receiving overall HRT or vaginal estrogen. This suggests that HRT may help reduce the risk of AF [186]. However, except for estradiol-only HRT, other types of HRT (such as combined estrogen therapy) have been observed to increase the risk of AF in some cases, indicating that the effect of HRT on AF is not only influenced by ERs but may also involve the effects of other female steroids [187].
AF and HFpEF share similar risk factors, including hypertension, obesity, diabetes, and chronic inflammatory states [188, 189]. This is particularly evident in postmenopausal women, wherein a decline in estrogen levels significantly exacerbates these risk factors. The lack of estrogen may contribute to accelerated atrial fibrosis, increased cardiac load, and the promotion of metabolic dysregulation, thereby intensifying the interaction between AF and HFpEF [165, 190,191,192]. Although HRT has not shown survival benefits in large studies, it can improve diastolic function in postmenopausal women [193, 194]. This finding may help explain why, in some cases, HRT has been observed to reduce the incidence of AF and may indirectly or directly impact the occurrence of HFpEF. However, further research is needed to clarify the exact role of HRT and to potentially minimize the side effects of estrogen in the treatment of AF and HFpEF.
Molecular mechanisms of sex differences in animal HFpEF models
The prevalence of HFpEF is characterized by a significant sex imbalance, with a maximum of two-thirds of patients being female in some epidemiological studies; female patients often exhibit more severe symptoms than males [195, 196]. However, some perspectives have questioned the elevated presence of females in HFpEF, suggesting that this might be due to the longer average lifespan of females and their tendency to enter old age, as the risk of HF generally increases with age [5, 99]. However, the exact role of sex in the development of HFpEF remains unclear. To gain a deeper understanding of the association of sex and HFpEF, one research approach involves exploring sex differences through animal models, which allow the control of genetic and environmental factors and the direct study of sex-specific factors in HF, such as through procedures like gonadectomy.
Aging model (age-related HFpEF phenotype)
Age is a key factor in the development of HFpEF, and several models have been used to study how aging affects sex differences in HFpEF. The senescence-accelerated mouse model SAMP8 is one such example, in which aging leads to left ventricular stiffness, diastolic dysfunction, and myocardial fibrosis [197, 198]. Interestingly, variations in left ventricular structure and function in FVB/N inbred mice and Fischer 344 rats are sex-related [197, 199]. Male FVB/N mice exhibit diastolic dysfunction as early as 12Â months of age, whereas female mice do not exhibit similar symptoms. In contrast, aged female Fischer 344 rats are more prone to diastolic dysfunction and left ventricular remodeling than their male counterparts [200, 201].
Spontaneously hypertensive rat (SHR) model (hypertension-induced model)
The SHR model is another commonly used model for studying HFpEF. In this model, hypertension occurs naturally, and the rats exhibit features similar to those of HFpEF, such as left ventricular hypertrophy and diastolic dysfunction. Studies comparing male and female SHR have shown sex-related differences in the progression of HF. For example, male SHRs tend to exhibit a more rapid onset of noticeable aortic dysfunction and left ventricular hypertrophy than females [202]. Additionally, female SHRs showed delayed matrix metalloproteinase-2 activity in the aorta and heart, along with higher levels of AT2R and MasR mRNA, demonstrating relatively less fibrosis and better elastin preservation [202]. These findings suggest that estrogen plays a protective role in the progression of hypertension-related HFpEF.
Diabetic HFpEF model
Diabetes is an important risk factor for HFpEF. Studies using animal models of diabetes, particularly streptozotocin (STZ)-induced T1DM models, have provided crucial experimental evidence for understanding the role of hormonal and metabolic dysregulation in HFpEF development. STZ-induced T1DM has become a widely accepted animal model for studying diabetes-related CVD and HF [203, 204]. Both male and female diabetic mice exhibit characteristic features of HFpEF, including decreased cardiac output and increased cardiac ANP mRNA expression, indicating the onset of HF [205]. Additionally, despite preservation of the EF, STZ-treated rats showed a reduction in left ventricular end-diastolic volume, suggesting left ventricular diastolic dysfunction [205].
Notably, in a diabetic mouse model, diastolic dysfunction was more pronounced in female mice than that in male mice. Specifically, the E/E’ ratio in female mice significantly increased, while no significant change was observed in male mice [204]. This indicates that STZ-induced diabetes makes female mice more sensitive to diastolic dysfunction, even though their glucose levels are lower than those in male mice [204]. These findings highlight the significant role played by sex in diabetes-induced cardiovascular pathological changes.
High-fat diet (HFD) combined with L-NAME (obesity and hypertension model)
One of the most commonly used HFpEF animal models is the HFD combined with L-NAME (an NO synthase inhibitor). This model mimics the common risk factors for obesity and hypertension, both of which are prevalent in patients with HFpEF. In studies using this model, male and female mice showed distinct differences in their responses to the induced HFpEF phenotype. One study showed significant differences in cardiac function and structure between male and female mice in an HFpEF model by feeding C57BL/6N mice an HFD combined with L-NAME for 7 weeks. Male mice exhibited obvious concentric left ventricular hypertrophy and diastolic dysfunction when on this diet, characterized by increased heart mass, left ventricular remodeling index, and E/E′ ratio. In contrast, the changes in female mice were milder, with a smaller increase in the E/E′ ratio and insignificant variations in heart mass and ventricular remodeling index [206]. This finding suggests that in HFpEF, female mice may have a certain degree of protective effect compared to male mice. Ovariectomy surgery was performed and these animals were subsequently subjected to HFpEF treatment for 15 weeks. The reults showed that the heart weight and diastolic function of female mice were similar. This suggests that the protective effect of the female sex on HFpEF may not be entirely mediated by female hormones. However, this experiment was only conducted in young animals, and research on HFpEF in older animals and humans has not yet been conducted. Additionally, using ovariectomy to simulate the physiological menopausal state may not be entirely accurate, and the effects of male hormones have not been thoroughly studied. In another study using the same HFD combined with L-NAME-fed HFpEF C57BL/6 J mouse model, contradictory differences were observed compared to the previous study [207]. Although both male and female mice showed increased body weight, fat mass, left ventricular mass, and fat pad weight, as well as decreased exercise tolerance, the E/A ratio, E/E′ ratio, and lung weight in female mice were higher than those in male mice [206, 207]. The authors explained that this difference might be due to the different genetic backgrounds of the mice and pointed out a significant gene-sex interaction [208]. Mechanistically, in mice and humans, males often show higher mitochondrial DNA content and mitochondrial activity than females, which is partially regulated by sex hormones. RNA-seq analysis further revealed that acyl-CoA synthetase long-chain family member 6 (Acsl6) is a predominant downstream player of sex hormones that regulate mitochondrial activity and metabolic alterations underlying sex bias [207].
Despite employing nearly identical methods, these two studies yielded diametrically opposite results, as shown in Fig. 3. Researchers have suggested that this discrepancy may stem from the use of different mouse strains. However, the specific mouse strain that has the highest correlation with and reflects the pathology of HFpEF in humans remains unclear. Future studies should consider utilizing a more acute animal model as well as age, sex, and genetic background to achieve a more comprehensive understanding of the role of sex in HFpEF in humans.
Molecular mechanisms of sex difference in animal HFpEF models. In the HFpEF model induced by a high-fat diet combined with L-NAME, significant differences in HFpEF phenotypes were observed between male and female mice. In C57BL/6N mice, male mice exhibited concentric left ventricular hypertrophy, diastolic dysfunction, and significant cardiac remodeling, whereas female mice showed relatively milder changes, with a lower increase in the E/E′ ratio and minimal cardiac remodeling. However, in C57BL/6 J mice, although both male and female mice exhibited obesity and reduced exercise tolerance, female mice had higher E/A and E/E′ ratios and increased lung weight compared to males. Estrogen may also influence mitochondrial gene expression in cardiomyocytes, leading to a further reduction in mtDNA content and more pronounced mitochondrial dysfunction in female mice. These two studies yielded contradictory results, and further research is needed to determine which model better reflects the pathological features of human HFpEF
Conclusion
The role of sex in HFpEF is complex and involves multiple factors, necessitating further research to elucidate the specific mechanisms underlying sex differences and how these differences influence the development and progression of HFpEF. Estrogen is closely associated with the occurrence and development of HFpEF. Through various mechanisms, such as regulating fat distribution, lowering BP, improving blood glucose levels, and reducing the occurrence of AF, estrogen reduces the incidence of risk factors, thus affecting HFpEF (Table 1). Therefore, a decline in estrogen levels may interact with these risk factors, leading to the weakening effect of estrogen on the cardiovascular system and collectively increasing the risk of HFpEF in women. Hence, future research should further explore the specific mechanisms of estrogen in various phenotypes of risk factors in HFpEF, as well as the potential value of HRT in the treatment of HFpEF. Overall, this review provides a comprehensive understanding for the role of estrogen in HFpEF in a sex-specific manner.
Availability of data and materials
No datasets were generated or analysed during the current study. The figures were created and licenced by BioRender.Â
Abbreviations
- ACC:
-
American college of cardiology
- AGEs:
-
Advanced glycation end-products
- AHA:
-
American heart association
- AT:
-
Adipose tissue
- BMI:
-
Body mass index
- BP:
-
Blood pressure
- CEE:
-
Conjugated equine estrogen
- CVD:
-
Cardiovascular disease
- EAT:
-
Epicardial adipose tissue
- EF:
-
Ejection fraction
- ER:
-
Estrogen receptor
- ERT:
-
Estrogen-only replacement therapy
- GLP-1:
-
Glucagon-like peptide-1
- HF:
-
Heart failure
- HFmrEF:
-
Heart failure with mid-range ejection fraction
- HFpEF:
-
Heart failure with preserved ejection fraction
- HFrEF:
-
Heart failure with reduced ejection fraction
- HMOD:
-
Hypertension-mediated organ damage
- LA:
-
Left atrium
- MACE:
-
Major adverse cardiac events
- MHT:
-
Menopausal hormone therapy
- NO:
-
Nitric oxide
- PH:
-
Pulmonary hypertension
- RAAS:
-
Renin-angiotensin-aldosterone system
- SGLT2:
-
Sodium-glucose co-transporter-2
- SHR:
-
Spontaneously hypertensive rat
References
Jessup M, Brozena S. Heart failure. N Engl J Med. 2003;348(20):2007–18.
Paulus WJ, et al. How to diagnose diastolic heart failure: a consensus statement on the diagnosis of heart failure with normal left ventricular ejection fraction by the Heart Failure and Echocardiography Associations of the European Society of Cardiology. Eur Heart J. 2007;28(20):2539–50.
Dickstein K, et al. ESC guidelines for the diagnosis and treatment of acute and chronic heart failure 2008: the task force for the diagnosis and treatment of acute and chronic heart failure 2008 of the European Society of Cardiology. Developed in collaboration with the heart failure association of the ESC (HFA) and endorsed by the European society of intensive care medicine (ESICM). Eur J Heart Fail. 2008;10(10):933–89.
Heidenreich PA, et al. 2022 AHA/ACC/HFSA guideline for the management of heart failure: a report of the american college of cardiology/american heart association joint committee on clinical practice guidelines. Circulation. 2022;145(18):e895–1032.
Dunlay SM, Roger VL, Redfield MM. Epidemiology of heart failure with preserved ejection fraction. Nat Rev Cardiol. 2017;14(10):591–602.
Owan TE, et al. Trends in prevalence and outcome of heart failure with preserved ejection fraction. N Engl J Med. 2006;355(3):251–9.
Ceia F, et al. Prevalence of chronic heart failure in Southwestern Europe: the EPICA study. Eur J Heart Fail. 2002;4(4):531–9.
Regitz-Zagrosek V, Gebhard C. Gender medicine: effects of sex and gender on cardiovascular disease manifestation and outcomes. Nat Rev Cardiol. 2023;20(4):236–47.
Sabbatini AR, Kararigas G. Menopause-related estrogen decrease and the pathogenesis of HFpEF: JACC review topic of the week. J Am Coll Cardiol. 2020;75(9):1074–82.
Ndumele CE, et al. Obesity and subtypes of incident cardiovascular disease. J Am Heart Assoc. 2016;5(8):e003921.
Collaboration, N.C.D.R.F., Worldwide trends in underweight and obesity from 1990 to 2022: a pooled analysis of 3663 population-representative studies with 222 million children, adolescents, and adults. Lancet, 2024. 403(10431): p. 1027–1050.
Jaacks LM, et al. The obesity transition: stages of the global epidemic. Lancet Diabetes Endocrinol. 2019;7(3):231–40.
Lovejoy JC, Sainsbury A, and G. Stock Conference Working, Sex differences in obesity and the regulation of energy homeostasis. Obes Rev. 2009; 10(2):154–67.
Kautzky-Willer A, Harreiter J, Pacini G. Sex and gender differences in risk, pathophysiology and complications of type 2 diabetes mellitus. Endocr Rev. 2016;37(3):278–316.
Awa WL, et al. Type 2 diabetes from pediatric to geriatric age: analysis of gender and obesity among 120,183 patients from the German/Austrian DPV database. Eur J Endocrinol. 2012;167(2):245–54.
Wietlisbach V, et al. The relation of body mass index and abdominal adiposity with dyslipidemia in 27 general populations of the WHO MONICA Project. Nutr Metab Cardiovasc Dis. 2013;23(5):432–42.
Flegal KM, et al. Prevalence and trends in obesity among US adults, 1999–2008. JAMA. 2010;303(3):235–41.
Benjamin EJ, et al. Heart disease and stroke statistics-2018 update: a report from the American heart association. Circulation. 2018;137(12):e67–492.
Obokata M, et al. Evidence supporting the existence of a distinct obese phenotype of heart failure with preserved ejection fraction. Circulation. 2017;136(1):6–19.
Borlaug BA. Heart failure with preserved and reduced ejection fraction: different risk profiles for different diseases. Eur Heart J. 2013;34(19):1393–5.
Kitzman DW, et al. Effect of caloric restriction or aerobic exercise training on peak oxygen consumption and quality of life in obese older patients with heart failure with preserved ejection fraction: a randomized clinical trial. JAMA. 2016;315(1):36–46.
Haass M, et al. Body mass index and adverse cardiovascular outcomes in heart failure patients with preserved ejection fraction: results from the Irbesartan in heart failure with preserved ejection fraction (I-PRESERVE) trial. Circ Heart Fail. 2011;4(3):324–31.
Savji N, et al. The association of obesity and cardiometabolic traits with incident HFpEF and HFrEF. JACC Heart Fail. 2018;6(8):701–9.
Ho JE, et al. Predictors of new-onset heart failure: differences in preserved versus reduced ejection fraction. Circ Heart Fail. 2013;6(2):279–86.
Koshino Y, et al. Changes in myocardial mechanics in patients with obesity following major weight loss after bariatric surgery. Obesity (Silver Spring). 2013;21(6):1111–8.
Fox CS, et al. Abdominal visceral and subcutaneous adipose tissue compartments: association with metabolic risk factors in the Framingham heart study. Circulation. 2007;116(1):39–48.
Ferreira I, et al. Central fat mass versus peripheral fat and lean mass: opposite (adverse versus favorable) associations with arterial stiffness? The Amsterdam growth and health longitudinal study. J Clin Endocrinol Metab. 2004;89(6):2632–9.
Alpert MA, et al. Impact of obesity and weight loss on cardiac performance and morphology in adults. Prog Cardiovasc Dis. 2014;56(4):391–400.
Recio-Rodriguez JI, et al. Abdominal obesity vs general obesity for identifying arterial stiffness, subclinical atherosclerosis and wave reflection in healthy, diabetics and hypertensive. BMC Cardiovasc Disord. 2012;12:3.
Triposkiadis F, et al. Obesity, inflammation, and heart failure: links and misconceptions. Heart Fail Rev. 2022;27(2):407–18.
Packer M. Derangements in adrenergic-adipokine signalling establish a neurohormonal basis for obesity-related heart failure with a preserved ejection fraction. Eur J Heart Fail. 2018;20(5):873–8.
Packer M, Kitzman DW. Obesity-related heart failure with a preserved ejection fraction: the mechanistic rationale for combining inhibitors of aldosterone, neprilysin, and sodium-glucose cotransporter-2. JACC Heart Fail. 2018;6(8):633–9.
Pandey A, et al. Physical activity, fitness, and obesity in heart failure with preserved ejection fraction. JACC Heart Fail. 2018;6(12):975–82.
Boustany CM, et al. Activation of the systemic and adipose renin-angiotensin system in rats with diet-induced obesity and hypertension. Am J Physiol Regul Integr Comp Physiol. 2004;287(4):R943–9.
Hall JE, et al. Obesity-induced hypertension: interaction of neurohumoral and renal mechanisms. Circ Res. 2015;116(6):991–1006.
Brown JM. Adverse effects of aldosterone: beyond blood pressure. J Am Heart Assoc. 2024;13(7): e030142.
Kawarazaki W, Fujita T. The role of aldosterone in obesity-related hypertension. Am J Hypertens. 2016;29(4):415–23.
Koepp KE, et al. Hemodynamic and functional impact of epicardial adipose tissue in heart failure with preserved ejection fraction. JACC Heart Fail. 2020;8(8):657–66.
Bays HE, et al. Pathogenic potential of adipose tissue and metabolic consequences of adipocyte hypertrophy and increased visceral adiposity. Expert Rev Cardiovasc Ther. 2008;6(3):343–68.
Ciana P, et al. In vivo imaging of transcriptionally active estrogen receptors. Nat Med. 2003;9(1):82–6.
Chatterjee K, Primary diastolic heart failure. Am J Geriatr Cardiol. 2002; 11(3):178–87; quiz 188–9.
Garza CA, et al. Major weight loss prevents long-term left atrial enlargement in patients with morbid and extreme obesity. Eur J Echocardiogr. 2008;9(5):587–93.
Grodstein F, et al. A prospective, observational study of postmenopausal hormone therapy and primary prevention of cardiovascular disease. Ann Intern Med. 2000;133(12):933–41.
Clement K, et al. Indication for linkage of the human OB gene region with extreme obesity. Diabetes. 1996;45(5):687–90.
Chen GC, et al. Association between regional body fat and cardiovascular disease risk among postmenopausal women with normal body mass index. Eur Heart J. 2019;40(34):2849–55.
Ley CJ, Lees B, Stevenson JC. Sex- and menopause-associated changes in body-fat distribution. Am J Clin Nutr. 1992;55(5):950–4.
Shi H, Seeley RJ, Clegg DJ. Sexual differences in the control of energy homeostasis. Front Neuroendocrinol. 2009;30(3):396–404.
Phillips GB, Jing T, Heymsfield SB. Does insulin resistance, visceral adiposity, or a sex hormone alteration underlie the metabolic syndrome? Studies in women Metabolism. 2008;57(6):838–44.
Eshtiaghi R, Esteghamati A, Nakhjavani M. Menopause is an independent predictor of metabolic syndrome in Iranian women. Maturitas. 2010;65(3):262–6.
Garaulet M, et al. Body fat distribution in pre-and post-menopausal women: metabolic and anthropometric variables. J Nutr Health Aging. 2002;6(2):123–6.
Lovejoy JC, et al. Increased visceral fat and decreased energy expenditure during the menopausal transition. Int J Obes (Lond). 2008;32(6):949–58.
Iacobellis G. Epicardial adipose tissue in contemporary cardiology. Nat Rev Cardiol. 2022;19(9):593–606.
Berg G, et al. Epicardial adipose tissue in cardiovascular disease. Adv Exp Med Biol. 2019;1127:131–43.
Fitzgibbons TP, Czech MP. Epicardial and perivascular adipose tissues and their influence on cardiovascular disease: basic mechanisms and clinical associations. J Am Heart Assoc. 2014;3(2): e000582.
Gersh FL, et al. The renin-angiotensin-aldosterone system in postmenopausal women: the promise of hormone therapy. Mayo Clin Proc. 2021;96(12):3130–41.
Tromp J, et al. Identifying pathophysiological mechanisms in heart failure with reduced versus preserved ejection fraction. J Am Coll Cardiol. 2018;72(10):1081–90.
Kenchaiah S, et al. Pericardial fat and the risk of heart failure. J Am Coll Cardiol. 2021;77(21):2638–52.
Le Jemtel TH, et al. Epicardial adipose tissue and cardiovascular disease. Curr Hypertens Rep. 2019;21(5):36.
Vyas V, et al., Obesity and diabetes are major risk factors for epicardial adipose tissue inflammation. JCI Insight. 2021; 6(16).
Chirinos JA, et al. Multiple plasma biomarkers for risk stratification in patients with heart failure and preserved ejection fraction. J Am Coll Cardiol. 2020;75(11):1281–95.
Xu S, et al. Endothelial dysfunction in atherosclerotic cardiovascular diseases and beyond: from mechanism to pharmacotherapies. Pharmacol Rev. 2021;73(3):924–67.
2018 ESC/ESH Guidelines for the management of arterial hypertension: Erratum. J Hypertens. 2019; 37(1):226.
Neuhauser H, Thamm M, Ellert U. Blood pressure in Germany 2008–2011: results of the German health interview and examination survey for adults (DEGS1). Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz. 2013;56(5–6):795–801.
Messerli FH, Rimoldi SF, Bangalore S. The transition from hypertension to heart failure: contemporary update. JACC Heart Fail. 2017;5(8):543–51.
Hayes SN, Taler SJ. Hypertension in women: current understanding of gender differences. Mayo Clin Proc. 1998;73(2):157–65.
Gasse C, et al. Assessing hypertension management in the community: trends of prevalence, detection, treatment, and control of hypertension in the MONICA Project, Augsburg 1984–1995. J Hum Hypertens. 2001;15(1):27–36.
Yoon SS, Carroll MD, Fryar CD. Hypertension prevalence and control among adults: United States, 2011–2014. NCHS Data Brief. 2015;220:1–8.
Maranon R, Reckelhoff JF. Sex and gender differences in control of blood pressure. Clin Sci. 2013;125(7):311–8.
Meyer MR, Prossnitz ER, Barton M. GPER/GPR30 and regulation of vascular tone and blood pressure. Immunol Endocr Metab Agents Med Chem. 2011;11(4):255–61.
Barton M, Meyer MR. Postmenopausal hypertension: mechanisms and therapy. Hypertension. 2009;54(1):11–8.
Mendelsohn ME, Karas RH. The protective effects of estrogen on the cardiovascular system. N Engl J Med. 1999;340(23):1801–11.
Kim JK, Levin ER. Estrogen signaling in the cardiovascular system. Nucl Recept Signal. 2006;4: e013.
Weiner CP, et al. Induction of calcium-dependent nitric oxide synthases by sex hormones. Proc Natl Acad Sci U S A. 1994;91(11):5212–6.
Widder J, et al. Improvement of endothelial dysfunction by selective estrogen receptor-alpha stimulation in ovariectomized SHR. Hypertension. 2003;42(5):991–6.
Zhu Y, et al. Abnormal vascular function and hypertension in mice deficient in estrogen receptor beta. Science. 2002;295(5554):505–8.
Jazbutyte V, et al. Ligand-dependent activation of ERbeta lowers blood pressure and attenuates cardiac hypertrophy in ovariectomized spontaneously hypertensive rats. Cardiovasc Res. 2008;77(4):774–81.
Fredette NC, Meyer MR, Prossnitz ER. Role of GPER in estrogen-dependent nitric oxide formation and vasodilation. J Steroid Biochem Mol Biol. 2018;176:65–72.
Regitz-Zagrosek V, Brokat S, Tschope C. Role of gender in heart failure with normal left ventricular ejection fraction. Prog Cardiovasc Dis. 2007;49(4):241–51.
Levy D, et al. The progression from hypertension to congestive heart failure. JAMA. 1996;275(20):1557–62.
Regitz-Zagrosek V, Lehmkuhl E. Heart failure and its treatment in women. Role of hypertension, diabetes, and estrogen. Herz. 2005;30(5):356–67.
Lam CS, et al. Epidemiology and clinical course of heart failure with preserved ejection fraction. Eur J Heart Fail. 2011;13(1):18–28.
Arcopinto M, et al. What have we learned so far from the sex/gender issue in heart failure? An overview of current evidence. Intern Emerg Med. 2022;17(6):1589–98.
Ponikowski P, et al. 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: the task force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC)Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J. 2016;37(27):2129–200.
Yancy CW, et al. 2017 ACC/AHA/HFSA focused update of the 2013 ACCF/AHA guideline for the management of heart failure: a report of the American college of cardiology/American heart association task force on clinical practice guidelines and the heart failure society of America. Circulation. 2017;136(6):e137–61.
Okin PM, et al. Regression of electrocardiographic left ventricular hypertrophy is associated with less hospitalization for heart failure in hypertensive patients. Ann Intern Med. 2007;147(5):311–9.
Whelton PK, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: a report of the American college of cardiology/American heart association task force on clinical practice guidelines. Circulation. 2018;138(17):e484–594.
Sciarretta S, et al. Antihypertensive treatment and development of heart failure in hypertension: a Bayesian network meta-analysis of studies in patients with hypertension and high cardiovascular risk. Arch Intern Med. 2011;171(5):384–94.
Bhatia RS, et al. Outcome of heart failure with preserved ejection fraction in a population-based study. N Engl J Med. 2006;355(3):260–9.
Yancy CW, et al. Clinical presentation, management, and in-hospital outcomes of patients admitted with acute decompensated heart failure with preserved systolic function: a report from the acute decompensated heart failure national registry (ADHERE) database. J Am Coll Cardiol. 2006;47(1):76–84.
Lee DS, et al. Relation of disease pathogenesis and risk factors to heart failure with preserved or reduced ejection fraction: insights from the framingham heart study of the national heart, lung, and blood institute. Circulation. 2009;119(24):3070–7.
Kao DP, et al. Characterization of subgroups of heart failure patients with preserved ejection fraction with possible implications for prognosis and treatment response. Eur J Heart Fail. 2015;17(9):925–35.
Slivnick J, Lampert BC. Hypertension and heart failure. Heart Fail Clin. 2019;15(4):531–41.
Gladden JD, Chaanine AH, Redfield MM. Heart failure with preserved ejection fraction. Annu Rev Med. 2018;69:65–79.
Tadic M, et al. The role of arterial hypertension in development heart failure with preserved ejection fraction: just a risk factor or something more? Heart Fail Rev. 2018;23(5):631–9.
Ponikowski P, et al. 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure. Rev Esp Cardiol (Engl Ed). 2016;69(12):1167.
Crabbe DL, et al. Gender differences in post-infarction hypertrophy in end-stage failing hearts. J Am Coll Cardiol. 2003;41(2):300–6.
Aurigemma GP, Gaasch WH. Gender differences in older patients with pressure-overload hypertrophy of the left ventricle. Cardiology. 1995;86(4):310–7.
Lam CS, et al. Aortic root remodeling over the adult life course: longitudinal data from the Framingham heart study. Circulation. 2010;122(9):884–90.
Redfield MM, et al. Age- and gender-related ventricular-vascular stiffening: a community-based study. Circulation. 2005;112(15):2254–62.
Badesch DB, et al. Pulmonary arterial hypertension: baseline characteristics from the REVEAL registry. Chest. 2010;137(2):376–87.
Thenappan T, et al. Clinical characteristics of pulmonary hypertension in patients with heart failure and preserved ejection fraction. Circ Heart Fail. 2011;4(3):257–65.
McHugh K, et al. Heart failure with preserved ejection fraction and diabetes: JACC State-of-the-art review. J Am Coll Cardiol. 2019;73(5):602–11.
Rowley WR, et al. Diabetes 2030: insights from yesterday, today, and future trends. Popul Health Manag. 2017;20(1):6–12.
Collaborators GBDD. Global, regional, and national burden of diabetes from 1990 to 2021, with projections of prevalence to 2050: a systematic analysis for the global burden of disease study 2021. Lancet. 2023;402(10397):203–34.
Nichols GA, et al. The incidence of congestive heart failure in type 2 diabetes: an update. Diabetes Care. 2004;27(8):1879–84.
Bell DS. Heart failure: the frequent, forgotten, and often fatal complication of diabetes. Diabetes Care. 2003;26(8):2433–41.
MacDonald MR, et al. Impact of diabetes on outcomes in patients with low and preserved ejection fraction heart failure: an analysis of the Candesartan in heart failure: assessment of reduction in mortality and morbidity (CHARM) programme. Eur Heart J. 2008;29(11):1377–85.
Kautzky-Willer A, Leutner M, Harreiter J. Correction to: sex differences in type 2 diabetes. Diabetologia. 2023;66(6):1165.
Danaei G, et al. National, regional, and global trends in fasting plasma glucose and diabetes prevalence since 1980: systematic analysis of health examination surveys and epidemiological studies with 370 country-years and 2.7 million participants. Lancet. 2011;378(9785):31–40.
Group DS, Age- and sex-specific prevalences of diabetes and impaired glucose regulation in 13 European cohorts. Diabetes Care. 2003; 26(1):61–9
Du T, et al. Sex differences in cardiovascular risk profile from childhood to midlife between individuals who did and did not develop diabetes at follow-up: the Bogalusa heart study. Diabetes Care. 2019;42(4):635–43.
Peters SA, Huxley RR, Woodward M. Diabetes as risk factor for incident coronary heart disease in women compared with men: a systematic review and meta-analysis of 64 cohorts including 858,507 individuals and 28,203 coronary events. Diabetologia. 2014;57(8):1542–51.
McIntyre HD, et al. Gestational diabetes mellitus. Nat Rev Dis Primers. 2019;5(1):47.
Vounzoulaki E, et al. Progression to type 2 diabetes in women with a known history of gestational diabetes: systematic review and meta-analysis. BMJ. 2020;369: m1361.
Kannel WB, Hjortland M, Castelli WP. Role of diabetes in congestive heart failure: the Framingham study. Am J Cardiol. 1974;34(1):29–34.
Wang Y, et al. Sex differences in the association between diabetes and risk of cardiovascular disease, cancer, and all-cause and cause-specific mortality: a systematic review and meta-analysis of 5,162,654 participants. BMC Med. 2019;17(1):136.
MacDonald MR, et al. Discordant short- and long-term outcomes associated with diabetes in patients with heart failure: importance of age and sex: a population study of 5.1 million people in Scotland. Circ Heart Fail. 2008;1(4):234–41.
Andersson C, et al. Long-term impact of diabetes in patients hospitalized with ischemic and non-ischemic heart failure. Scand Cardiovasc J. 2010;44(1):37–44.
Aguilar D, et al. Comparison of patients with heart failure and preserved left ventricular ejection fraction among those with versus without diabetes mellitus. Am J Cardiol. 2010;105(3):373–7.
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.
Heerspink HJ, et al. Sodium Glucose cotransporter 2 inhibitors in the treatment of diabetes mellitus: cardiovascular and kidney effects, potential mechanisms, and clinical applications. Circulation. 2016;134(10):752–72.
Smaha LA and American Heart A, The american heart association get with the guidelines program. Am Heart J. 2004; 148(5 Suppl): pp. S46–8.
Zinman B, Lachin JM, Inzucchi SE. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2016;374(11):1094.
Butler J, et al. The potential role and rationale for treatment of heart failure with sodium-glucose co-transporter 2 inhibitors. Eur J Heart Fail. 2017;19(11):1390–400.
Salvatore T, et al., An overview of the cardiorenal protective mechanisms of SGLT2 inhibitors. Int J Mol Sci. 2022; 23(7).
Ni L, et al. SGLT2i: beyond the glucose-lowering effect. Cardiovasc Diabetol. 2020;19(1):98.
Ferrannini E, et al. Shift to fatty substrate utilization in response to sodium-glucose cotransporter 2 inhibition in subjects without diabetes and patients with type 2 diabetes. Diabetes. 2016;65(5):1190–5.
Preda A, et al. SGLT2 inhibitors: from glucose-lowering to cardiovascular benefits. Cardiovasc Res. 2024;120(5):443–60.
Lee TM, Chang NC, Lin SZ. Dapagliflozin, a selective SGLT2 Inhibitor, attenuated cardiac fibrosis by regulating the macrophage polarization via STAT3 signaling in infarcted rat hearts. Free Radic Biol Med. 2017;104:298–310.
Li C, et al. SGLT2 inhibition with empagliflozin attenuates myocardial oxidative stress and fibrosis in diabetic mice heart. Cardiovasc Diabetol. 2019;18(1):15.
Dei Cas A, et al. Impact of diabetes on epidemiology, treatment, and outcomes of patients with heart failure. JACC Heart Fail. 2015;3(2):136–45.
Lopaschuk GD, et al. Myocardial fatty acid metabolism in health and disease. Physiol Rev. 2010;90(1):207–58.
Flanagan DE, et al. Gender differences in the insulin-like growth factor axis response to a glucose load. Acta Physiol (Oxf). 2006;187(3):371–8.
Reichelt ME, et al. Sex, sex steroids, and diabetic cardiomyopathy: making the case for experimental focus. Am J Physiol Heart Circ Physiol. 2013;305(6):H779–92.
Ryden L, et al. Guidelines on diabetes, pre-diabetes, and cardiovascular diseases: executive summary the task force on diabetes and cardiovascular diseases of the European society of cardiology (ESC) and of the European association for the study of diabetes (EASD). Eur Heart J. 2007;28(1):88–136.
Luo B, et al. NLRP3 gene silencing ameliorates diabetic cardiomyopathy in a type 2 diabetes rat model. PLoS One. 2014;9(8): e104771.
Jia G, Hill MA, Sowers JR. Diabetic cardiomyopathy: an update of mechanisms contributing to this clinical entity. Circ Res. 2018;122(4):624–38.
Park SK, et al. Association between changes in oestradiol and follicle-stimulating hormone levels during the menopausal transition and risk of diabetes. Diabet Med. 2017;34(4):531–8.
Walton C, et al. The effects of the menopause on insulin sensitivity, secretion and elimination in non-obese, healthy women. Eur J Clin Invest. 1993;23(8):466–73.
Malacara JM, et al. Menopause in normal and uncomplicated NIDDM women: physical and emotional symptoms and hormone profile. Maturitas. 1997;28(1):35–45.
Appiah D, Winters SJ, Hornung CA. Bilateral oophorectomy and the risk of incident diabetes in postmenopausal women. Diabetes Care. 2014;37(3):725–33.
Espeland MA, et al. Effect of postmenopausal hormone therapy on glucose and insulin concentrations. PEPI investigators postmenopausal estrogen/progestin interventions. Diabetes Care. 1998;21(10):1589–95.
Salpeter SR, et al. Meta-analysis: effect of hormone-replacement therapy on components of the metabolic syndrome in postmenopausal women. Diabetes Obes Metab. 2006;8(5):538–54.
Stuenkel CA, et al. Treatment of symptoms of the menopause: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2015;100(11):3975–4011.
Saengsirisuwan V, et al. Modulation of insulin resistance in ovariectomized rats by endurance exercise training and estrogen replacement. Metabolism. 2009;58(1):38–47.
Donahue RP, et al. Sex differences in endothelial function markers before conversion to pre-diabetes: does the clock start ticking earlier among women? The Western New York study. Diabetes Care. 2007;30(2):354–9.
Wannamethee SG, et al. Do women exhibit greater differences in established and novel risk factors between diabetes and non-diabetes than men? The British regional heart study and British women’s heart health study. Diabetologia. 2012;55(1):80–7.
Hunter I, et al. Cardiovascular function in male and female JCR:LA-cp rats: effect of high-fat/high-sucrose diet. Am J Physiol Heart Circ Physiol. 2017;312(4):H742–51.
Kilic G, et al. The islet estrogen receptor-alpha is induced by hyperglycemia and protects against oxidative stress-induced insulin-deficient diabetes. PLoS One. 2014;9(2): e87941.
Liu S, et al. Oestrogens improve human pancreatic islet transplantation in a mouse model of insulin deficient diabetes. Diabetologia. 2013;56(2):370–81.
Liu S, et al. Importance of extranuclear estrogen receptor-alpha and membrane G protein-coupled estrogen receptor in pancreatic islet survival. Diabetes. 2009;58(10):2292–302.
Xu B, et al. Estrogens promote misfolded proinsulin degradation to protect insulin production and delay diabetes. Cell Rep. 2018;24(1):181–96.
Andersen A, et al. Glucagon-like peptide 1 in health and disease. Nat Rev Endocrinol. 2018;14(7):390–403.
Fuselier T, et al. Efficacy of glucagon-like peptide-1 and estrogen dual agonist in pancreatic islets protection and pre-clinical models of insulin-deficient diabetes. Cell Rep Med. 2022;3(4): 100598.
Schwenk RW, et al. GLP-1-oestrogen attenuates hyperphagia and protects from beta cell failure in diabetes-prone New Zealand obese (NZO) mice. Diabetologia. 2015;58(3):604–14.
Tiano JP, et al. Effect of targeted estrogen delivery using glucagon-like peptide-1 on insulin secretion, insulin sensitivity and glucose homeostasis. Sci Rep. 2015;5:10211.
Sachs S, et al. Targeted pharmacological therapy restores beta-cell function for diabetes remission. Nat Metab. 2020;2(2):192–209.
Purdie DW. Consequences of long-term hormone replacement therapy. Br Med Bull. 2000;56(3):809–23.
Rutter MK, et al. Impact of glucose intolerance and insulin resistance on cardiac structure and function: sex-related differences in the Framingham heart study. Circulation. 2003;107(3):448–54.
Jia G, DeMarco VG, Sowers JR. Insulin resistance and hyperinsulinaemia in diabetic cardiomyopathy. Nat Rev Endocrinol. 2016;12(3):144–53.
Zhang B, et al. Notoginsenoside R1 protects against diabetic cardiomyopathy through activating estrogen receptor alpha and its downstream signaling. Front Pharmacol. 2018;9:1227.
Olsson LG, et al. Atrial fibrillation and risk of clinical events in chronic heart failure with and without left ventricular systolic dysfunction: results from the Candesartan in Heart failure-assessment of reduction in mortality and morbidity (CHARM) program. J Am Coll Cardiol. 2006;47(10):1997–2004.
Linssen GC, et al. Clinical and prognostic effects of atrial fibrillation in heart failure patients with reduced and preserved left ventricular ejection fraction. Eur J Heart Fail. 2011;13(10):1111–20.
Zakeri R, et al. Impact of atrial fibrillation on exercise capacity in heart failure with preserved ejection fraction: a RELAX trial ancillary study. Circ Heart Fail. 2014;7(1):123–30.
Zakeri R, et al. Temporal relationship and prognostic significance of atrial fibrillation in heart failure patients with preserved ejection fraction: a community-based study. Circulation. 2013;128(10):1085–93.
Zafrir B, et al. Prognostic implications of atrial fibrillation in heart failure with reduced, mid-range, and preserved ejection fraction: a report from 14 964 patients in the European society of cardiology heart failure long-term registry. Eur Heart J. 2018;39(48):4277–84.
Meyer S, et al. Sex differences in new-onset heart failure. Clin Res Cardiol. 2015;104(4):342–50.
Providencia R, Lambiase PD. Letter by providencia and lambiase regarding article, atrial fibrillation begets heart failure and vice versa: temporal associations and differences in preserved versus reduced ejection fraction. Circulation. 2016;133(23): e691.
Reddy YNV, et al. Atrial dysfunction in patients with heart failure with preserved ejection fraction and atrial fibrillation. J Am Coll Cardiol. 2020;76(9):1051–64.
Manning WJ, et al. Impaired left atrial mechanical function after cardioversion: relation to the duration of atrial fibrillation. J Am Coll Cardiol. 1994;23(7):1535–40.
Clark DM, et al. Hemodynamic effects of an irregular sequence of ventricular cycle lengths during atrial fibrillation. J Am Coll Cardiol. 1997;30(4):1039–45.
Pozzoli M, et al. Predictors of primary atrial fibrillation and concomitant clinical and hemodynamic changes in patients with chronic heart failure: a prospective study in 344 patients with baseline sinus rhythm. J Am Coll Cardiol. 1998;32(1):197–204.
Melenovsky V, et al. Cardiovascular features of heart failure with preserved ejection fraction versus nonfailing hypertensive left ventricular hypertrophy in the urban Baltimore community: the role of atrial remodeling/dysfunction. J Am Coll Cardiol. 2007;49(2):198–207.
Knackstedt C, et al. Association of echocardiographic atrial size and atrial fibrosis in a sequential model of congestive heart failure and atrial fibrillation. Cardiovasc Pathol. 2008;17(5):318–24.
Melenovsky V, et al. Left atrial remodeling and function in advanced heart failure with preserved or reduced ejection fraction. Circ Heart Fail. 2015;8(2):295–303.
Ball J, et al. Women versus men with chronic atrial fibrillation: insights from the standard versus atrial fibrillation spEcific managemenT studY (SAFETY). PLoS One. 2013;8(5): e65795.
Heeringa J, et al. Prevalence, incidence and lifetime risk of atrial fibrillation: the Rotterdam study. Eur Heart J. 2006;27(8):949–53.
Scheuermeyer FX, et al., There are sex differences in the demographics and risk profiles of emergency department (ED) patients with atrial fibrillation and flutter, but no apparent differences in ED management or outcomes. Acad Emerg Med. 2015; 22(9):1067–75.
Benjamin EJ, et al. Impact of atrial fibrillation on the risk of death: the Framingham heart study. Circulation. 1998;98(10):946–52.
Magnussen C, et al. Sex differences and similarities in atrial fibrillation epidemiology, risk factors, and mortality in community cohorts: results from the biomarCaRE consortium (Biomarker for Cardiovascular Risk Assessment in Europe). Circulation. 2017;136(17):1588–97.
Schnabel RB, et al. 50 year trends in atrial fibrillation prevalence, incidence, risk factors, and mortality in the Framingham heart study: a cohort study. Lancet. 2015;386(9989):154–62.
Ko D, et al. Atrial fibrillation in women: epidemiology, pathophysiology, presentation, and prognosis. Nat Rev Cardiol. 2016;13(6):321–32.
Correction to: Tamoxifen for the prevention of breast cancer: late results of the italian randomized tamoxifen prevention trial among women with hysterectomy. J Natl Cancer Inst. 2024; 116(6):995.
Wong JA, et al. Menopausal age, postmenopausal hormone therapy and incident atrial fibrillation. Heart. 2017;103(24):1954–61.
Tsai WC, et al. Hormone replacement therapy and risk of atrial fibrillation in Taiwanese menopause women: a nationwide cohort study. Sci Rep. 2016;6:24132.
Bretler DM, et al. Hormone replacement therapy and risk of new-onset atrial fibrillation after myocardial infarction–a nationwide cohort study. PLoS One. 2012;7(12): e51580.
Lee J, et al., Clinical impact of hormone replacement therapy on atrial fibrillation in postmenopausal women: a nationwide cohort study. J Clin Med. 2021; 10(23).
Pan Y, et al. The common characteristics and mutual effects of heart failure and atrial fibrillation: initiation, progression, and outcome of the two aging-related heart diseases. Heart Fail Rev. 2022;27(3):837–47.
Jurica J, et al. Obesity as a risk factor in atrial fibrillation and heart failure. J Diabetes Metab Disord. 2024;23(1):125–34.
O’Neal WT, et al. Gender differences in the risk of adverse outcomes in patients with atrial fibrillation and heart failure with preserved ejection fraction. Am J Cardiol. 2017;119(11):1785–90.
Carr MC. The emergence of the metabolic syndrome with menopause. J Clin Endocrinol Metab. 2003;88(6):2404–11.
Odening KE, et al. Mechanisms of sex differences in atrial fibrillation: role of hormones and differences in electrophysiology, structure, function, and remodelling. Europace. 2019;21(3):366–76.
Gokce M, et al. Left ventricular diastolic function assessment by tissue Doppler echocardiography in relation to hormonal replacement therapy in postmenopausal women with diastolic dysfunction. Am J Ther. 2003;10(2):104–11.
Rossouw JE, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288(3):321–33.
Beale AL, et al. Sex differences in cardiovascular pathophysiology: why women are overrepresented in heart failure with preserved ejection fraction. Circulation. 2018;138(2):198–205.
Kessler EL, et al. Sex-specific influence on cardiac structural remodeling and therapy in cardiovascular disease. Biol Sex Differ. 2019;10(1):7.
Reed AL, et al. Diastolic dysfunction is associated with cardiac fibrosis in the senescence-accelerated mouse. Am J Physiol Heart Circ Physiol. 2011;301(3):H824–31.
Gevaert AB, et al., Endothelial senescence contributes to heart failure with preserved ejection fraction in an aging mouse model. Circ Heart Fail. 2017; 10(6).
Koch SE, et al. Age- and gender-related changes in ventricular performance in wild-type FVB/N mice as evaluated by conventional and vector velocity echocardiography imaging: a retrospective study. Ultrasound Med Biol. 2013;39(11):2034–43.
No MH, et al. Effects of aging and exercise training on mitochondrial function and apoptosis in the rat heart. Pflugers Arch. 2020;472(2):179–93.
Hacker TA, et al. Age-related changes in cardiac structure and function in Fischer 344 x Brown Norway hybrid rats. Am J Physiol Heart Circ Physiol. 2006;290(1):H304–11.
Al-Gburi S, et al. Sex-specific differences in age-dependent progression of aortic dysfunction and related cardiac remodeling in spontaneously hypertensive rats. Am J Physiol Regul Integr Comp Physiol. 2017;312(5):R835–49.
Joffe II, et al. Abnormal cardiac function in the streptozotocin-induced non-insulin-dependent diabetic rat: noninvasive assessment with doppler echocardiography and contribution of the nitric oxide pathway. J Am Coll Cardiol. 1999;34(7):2111–9.
Chandramouli C, et al. Diastolic dysfunction is more apparent in STZ-induced diabetic female mice, despite less pronounced hyperglycemia. Sci Rep. 2018;8(1):2346.
Huang JP, et al., Exosomal microRNAs miR-30d-5p and miR-126a-5p Are associated with heart failure with preserved ejection fraction in STZ-induced type 1 diabetic rats. Int J Mol Sci. 2022; 23(14).
Tong D, et al. Female sex is protective in a preclinical model of heart failure with preserved ejection fraction. Circulation. 2019;140(21):1769–71.
Cao Y, et al. Sex differences in heart mitochondria regulate diastolic dysfunction. Nat Commun. 2022;13(1):3850.
Norheim F, et al., Gene-by-sex interactions in mitochondrial functions and cardio-metabolic traits. Cell Metab. 2019; 29(4):932–949 e4.
Funding
National Natural Science Foundation of China, 82070264, 82100764, 82070264,82100764, Xi ‘an Science and Technology Bureau Project, 24YXYJ0138, Department of Science and Technology of Shaanxi Province, 2024SF-LCZX05, 2022ZDLSF01-09,2023-CX-PT-06, 2024SF-LCZX05, 2022ZDLSF01-09, 2023-CX-PT-06, Xijing Research Boosting Program, XJZT24LY40.
Author information
Authors and Affiliations
Contributions
Jun Du.: Writing—original draft. Jiaqi Liu: Writing—original draft. Xiaoya Wang: Writing—review and editing. Xiaowu Wang: Writing—review and editing. Yu Ma.: Writing—review and editing. Sipan Zhang: Writing—review and editing. Zilin Li:Writing—review and editing. Jipeng Ma.: Conceptualization, Writing—review and editing, Supervision. Jincheng Liu.:Conceptualization, Writing—review and editing, Supervision, Funding acquisition. All authors have read and agreed to the published version of the manuscript.
Corresponding authors
Ethics declarations
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
Du, J., Liu, J., Wang, X. et al. The role of estrogen in the sex difference for the risk factors of heart failure with preserved ejection fraction. Biol Direct 20, 28 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13062-025-00618-x
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13062-025-00618-x