Skip to main content

Therapeutic potential of finerenone for diabetic cardiomyopathy: focus on the mechanisms

Abstract

Diabetic cardiomyopathy (DCM) is a kind of myocardial disease that occurs in diabetes patients and cannot be explained by hypertensive heart disease, coronary atherosclerotic heart disease and other heart diseases. Its pathogenesis may be closely related to programmed cell death, oxidative stress, intestinal microbes and micro-RNAs. The excessive activation of mineralocorticoid receptors (MR) in DCM can cause damage to the heart and kidneys. The third-generation non-steroidal mineralocorticoid receptor antagonist (MRA), finerenone, can effectively block MR, thus playing a role in protecting the heart and kidneys. This review mainly introduces the classification of MRA, and the mechanism of action, applications and limitations of finerenone in DCM, in order to provide reference for the study of treatment plans for DCM patients.

Classification of mineralocorticoid receptor antagonists

Aldosterone is the main mineralocorticoid that can bind to the mineralocorticoid receptor (MR) to maintain water and electrolyte balance and induce pro-inflammatory activity in the body, and ultimately lead to dysfunction and failure of target organs such as the heart and kidneys [1, 2].

Mineralocorticoid receptor antagonists (MRAs) can inhibit the excessive activation of MR, thereby playing a role in protecting the heart and kidneys [3, 4]. According to molecular structure, MRA can be divided into traditional steroidal MRA and new generation non-steroidal MRA [5]. Steroid MRA mainly includes spironolactone and eplerenone, which have steroidal structures. However, spironolactone has low selectivity for MR and a higher incidence of hyperkalemia after administration [6]. Eplerenone has higher selectivity for MR, stronger anti aldosterone activity and lower side effects than spironolactone [7]. Non-steroidal MRA mainly includes finerenone, Esaxerenone, AZD9977, Aparenone, and KBP-5074, which have non steroidal structures [5, 8]. Finerenone has high selectivity for MR and is less prone to side effects such as hyperkalemia [9].

Mechanism and function of finerenone in diabetic cardiomyopathy

Diabetic cardiomyopathy (DCM) is an organic heart disease resulting from abnormal myocardial structure and function in individuals with DM who do not have other conditions, such as coronary artery disease, hypertension, valvular heart disease and congenital heart disease. DCM arises due to dysregulated glucose and lipid metabolism associated with DM, triggering the activation of various inflammatory pathways [10]. Research has found that DCM is closely related to programmed cell death, oxidative stress, intestinal microbiota, and MicroRNAs (miRNAs) [11,12,13,14]. Finerenone is a non-steroidal MRA, and there is extensive research evidence (Phase III study FIDELIO/FIGARO) indicating that finerenone can provide protective effects on the heart and kidneys [15]. As a type of MRA, finerenone can affect programmed cell death [16]. By blocking the MR, finerenone may also inhibit the generation of reactive oxygen species (ROS), which promote oxidative stress in cells, leading to tissue injury [4] (Table 1). However, further research is needed to investigate the relationship between finerenone and intestinal microbiota as well as miRNAs.

Table 1 Basic science trials of finerenone

Programmed cell death and finerenone in diabetic cardiomyopathy

In biology, cell death is broadly classified as necrosis and programmed cell death (PCD). PCD includes apoptosis [23], autophagy [23], pyroptosis [24], ferroptosis [25] and more. More and more evidence has demonstrated that PCD of cardiomyocytes is a major contributor to the development of DCM [24, 26,27,28]. Therefore, it is particularly important to regulate the death of cardiomyocytes in patients with diabetes cardiomyopathy. Some studies have found that finerenone can reduce cell apoptosis, restore autophagy levels and ameliorated cell pyroptosis [12, 16, 29].

Apoptosis

Apoptosis is a programmed and active death process that occurs in cells under the control of specific genes or pathways. It is carried out by proapoptotic caspases (mainly caspase-2/3/6/7/8/9/10), which cleave intracellular substrates, causing cytoplasmic contraction, chromatin concentration, nuclear dissolution, and membrane foaming, ultimately decomposing into membrane encapsulated apoptotic bodies [23]. Studies have shown that long-term hyperglycemia and excessive uptake and accumulation of free fatty acids in diabetes patients can induce cardiomyocyte apoptosis, and apoptosis promotes cardiomyocyte damage in DCM patients through a variety of signal pathways, for example, through extrinsic and intrinsic apoptotic pathways (involving caspase-3/8/9) to cause cardiomyocyte apoptosis [16, 30, 31]. And, there is an upregulation of the renin–angiotensin–aldosterone system in DCM, resulting in an increase in aldosterone levels [32, 33], and aldosterone induces cardiomyocyte apoptosis through dependence on G protein-coupled receptor-kinase (GRK) [34]. In addition, DCM can also cause vascular damage and endothelial dysfunction [30, 35].

Experiments have shown that finerenone can down-regulate the TNFa/TNFR1/CASPASE8 signaling pathway to reduce the apoptosis of cardiomyocytes [16]. And it can improve lipid metabolism in cardiomyocytes and reduce myocardial lipid uptake by down-regulating PPARγ/CD36 to indirectly improve cardiomyocyte apoptosis [16, 30, 36]. On the other hand, as a type of MRA, finerenone can block the MR of the heart, thereby blocking aldosterone induced apoptosis. GRK-5 blocks the cardiac actions of aldosterone via phosphorylation of the MR [37]. Finerenone can induce GRK-5’s phosphorylation and suppress MR basal transcriptional activity in GRK5-overexpressing cardiomyocytes (finerenone’s inverse agonism at the cardiac MR), which plays an important role in blocking cardiomyocyte apoptosis.

In an experiment on vascular injury, non-steroidal MRA finerenone prevents aldosterone-induced smooth muscle cell (SMC) proliferation and endothelial cell (EC) apoptosis [18]. Excessive activation of MR in ECs can lead to endothelial dysfunction, finerenone can block the excessive activation of MR and thus block this process [38, 39].

Autophagy

Autophagy is an intracellular degradation process that encapsulates intracellular substances into double layered membrane vesicles, forming autophagosomes that are then fused by lysosomes to degrade and recycle these substances. The autophagy process is strictly regulated by the body and is crucial for maintaining the homeostasis of the intracellular environment. But abnormal autophagy can lead to cell death [23]. According to current studies, autophagy is regulated mainly by the phosphatidylinositol 3-phosphate kinase-mamma-lian target of rapamycin (PI3K-mTOR) signal transduction pathway upstream of autophagy-associated genes (ATG) and the Beclin1 complex [23, 40]. Research has shown that DCM is closely related to inhibition of cellular autophagy [11, 28]. High fat environment will inhibit myocardial autophagy in patients with diabetes, and in high glucose environment, this autophagy inhibition will worsen [41].

Although the mechanism by which finerenone restores autophagy in cardiomyocytes is not clear, studies have shown that finerenone can attenuate mitochondrial autophagy disruption in renal tubular epithelial cells of patients with diabetes nephropathy by inhibiting MR [19], which may provide guidance.

Pyroptosis

Pyroptosis is a form of PCD that is related to the innate immune response (such as pathogen invasion), and it is usually activated by inflammatory caspases (mainly caspase-1/4/5/11) and caspase-3 and relies on Gasdermin family proteins to form membrane pores, leading to nuclear fragmentation and dissolution, increased cell membrane permeability, swelling and lysis, and release of cellular contents, thereby causing local inflammatory reactions [23, 26, 42]. Moreover, studies have found that pyroptosis is also involved in the formation of DCM [42, 43]. NLRP3 inflammasome activation of caspase-1-mediated pyroptosis plays an important role in the development of diabetic cardiomyopathy [42].

As a type of MRA, finerenone can block inflammation caused by excessive activation of MR [33]. However, whether finerenone can also inhibit NLRP3-mediated pyroptosis in cardiomyocytes remains to be verified.

Oxidative stress and finerenone in diabetic cardiomyopathy

Oxidative stress refers to the imbalance between oxidative and antioxidant effects in the body. The “redox state” is determined by the balance between production of reactive oxygen species (ROS) and their removal by the antioxidant defense system. When this balance is disrupted, excessive ROS production and/or inadequate ROS detoxification may result in ROS-induced damage to DNA, proteins, lipids and micro RNA, leading to irreversible cell damage and death [44, 45]. Meanwhile, studies have shown that reactive nitrogen species (RNS) are also involved in oxidative stress [46, 47].

Oxidative stress is believed to play an important role in DCM. Although pathogenic factors (such as high sugar and high fat) can lead to DCM through different mechanisms, the main contribution of these pathogenic factors to DCM is oxidative stress. And oxidative stress can also mediate programmed cell death, mitochondrial dysfunction, inflammation, and so on [46,47,48]. Due to the abundant energy provided by mitochondria for cardiac activity, when mitochondrial function is impaired, it can have harmful effects on the heart. Multiple signaling pathways are involved in the oxidative stress of DCM [47, 49], and understanding these signaling pathways has beneficial results for antioxidant therapy. And the antioxidant mechanism is another noteworthy issue. The elimination of ROS depends on enzymes such as catalase and superoxide dismutase (SOD) [12]. The MRA, finerenone, can effectively block oxidative stress induced by aldosterone, thereby protecting the heart [17].

Mitochondrial dysfunction

As the energy factory of cells, mitochondria play an important role in the sustained functioning of cells, and mitochondrial dysfunction is closely related to DCM [50]. The heart is an organ with high energy requirements, and most of the ATP it consumes comes from the oxidative metabolism of mitochondria. Mitochondria in the heart account for one-third of the volume of adult cardiomyocytes [51]. Therefore, the heart is greatly affected by mitochondrial dysfunction.

Mitochondria, as producers of intracellular energy, are also the main targets of oxidative stress. There are multiple main sources of ROS production in cardiomyocytes. However, mitochondrial sources of ROS are thought to represent the major ROS burden in the context of diabetes [44]. Persistent hyperglycemia can lead to excessive production of ROS by cardiomyocytes [52]. Increased mitochondrial ROS induce oxidative damage to DNA, proteins and lipids, and may trigger a variety of pathological pathways involved in mitochondrial and cellular damage [53, 54].

In recent years, many studies have shown that oxidative stress can affect mitochondrial function through various factors such as affecting calcium ion levels, mitochondrial membrane potential, and respiratory chain complexes [53, 55,56,57]. When cardiomyocytes are subjected to oxidative stress, the concentration of calcium ions in mitochondria increases, thus inhibiting the generation of mitochondrial ATP [55]. Mitochondrial dysfunction can lead to the generation of ROS, forming a "vicious cycle" of enhanced oxidative stress.

Signaling pathway of finerenone in oxidative stress

Finerenone has certain antioxidant potential. Research has shown that finerenone abrogated oxidative stress in vascular smooth muscle cells from noninfarcted mice incubated with low-dose angiotensin-II [20]. It was also found that finerenone reduced the production of myocardial ROS after short-term administration in Zucker fa/fa rats (a rat model of metabolic syndrome) [21]. In general, finerenone can exert certain benefits in cardiac protection by inhibiting oxidative stress. The analysis of the signaling pathway of finerenone in oxidative stress helps to deepen the understanding of the drug's mechanism of action, thus providing a basis for the formulation of disease treatment strategies.

In rat kidney fibroblast cells, activation of MR induces mitochondrial dysfunction through the PI3K/Akt/eNOS pathway. PI3K phosphorylation stimulates its downstream protein Akt, phosphorylates Akt (p-Akt) and eNOS, regulating a variety of physiological functions, triggering mitochondrial dysfunction. Finerenone normalizes mitochondrial dysfunction by blocking MR, ultimately reducing ROS production [19]. This is helpful for studying the role of finerenone in cardiac oxidative stress.

Finerenone improves cardiomyocyte metabolism and reduces ROS generation through PPARα/CD36 pathway. A nuclear receptor, peroxisome proliferator-activated receptor alpha (PPARα), plays an important role in myocardial substrate metabolism by regulating the transcription of genes involved in FA transport, esterification, and oxidation [47, 58]. Due to insulin resistance or lack of insulin in DCM, the uptake and utilization of glucose in cardiomyocytes are limited, and the expression of CD36 (FFA translocatase) in cardiomyocytes is increased [16], which mediates the entry of FFA into cells, thus activating PPARα, which will promote the β-oxidation (β-ox) of FFA in mitochondria, and thus promote the production of ROS [59,60,61]. The ROS and the expression of PPARγ and CD36 decreased after finerenone treatment, thus effectively blocking oxidative stress [16]. MR activation contributes to aldosterone-mediated activation of NADPH oxidase mediated generation of ROS in the heart and coronary microvascular [62]. Finerenone inhibits this process by blocking MR.

Intestinal microbiota and finerenone in diabetic cardiomyopathy

Maintaining a healthy microbiota in the gut is crucial for maintaining homeostasis. However, when intestinal microbial homeostasis is disrupted, it can induce the development of different diseases [63]. Intestinal microbiota and its metabolites can affect the development of diabetic cardiomyopathy by regulating oxidative stress [64], inflammation [65], insulin resistance [66], apoptosis [67], and autophagy [67, 68]. At present, the relationship between finerenone and intestinal microbiota is not clear, and the specific mechanism needs to be more thoroughly investigated.

MicroRNAs (miRNAs) and finerenone in diabetic cardiomyopathy

MicroRNAs (miRNAs) are a type of noncoding RNAs (ncRNAs) that are approximately 22-nucleotide (nt) long and are encoded by endogenous genes. MiRNAs participate in transcriptional or posttranscriptional regulation by binding to the untranslated regions of target mRNAs, thus participating in the regulation of human pathophysiological processes [14]. Based on previous studies it was found that more than 300 different miRNAs play a role in DCM [69]. For example, experiments have shown that miRNA-373 can participate in the mitogen-activated protein kinase (MAPK) mediated signaling pathway, playing an important role in cardiomyocytes hypertrophy by targeting the hypertrophic protein, MEF2C [70]. MiRNA-503 was involved in the progress of apoptosis in DCM via regulating Nrf2/ARE signaling pathway [71].And miRNA-30c can participate in the PPARα mediated signaling pathway, regulating cardiac oxidative stress by targeting peroxisome proliferator-activated receptor coactivator 1β (PGC-1β) [72]. Therefore, targeting a particular miRNA involved in a specific signaling pathway in the diabetic heart may provide a therapeutic effect to ameliorate diabetic cardiomyopathy. Finerenone can play a certain role in DCM through PPARγ/CD36 pathway [16]. Therefore, it remains to be further confirmed whether there is any relationship between it and miRNA-30c or other miRNAs.

Therapeutic applications and limitations of finerenone in diabetic cardiomyopathy

The data from clinical trials with finerenone has expanded the treatment options for cardiorenal disease management for patients with T2DM (Table 2). The results of the two major studies, FIDELIO-DKD and FIGARO-DKD, are mutually validated, and it is believed that finerenone can improve renal and cardiovascular outcomes, bringing more benefits to patients [15, 73, 74]. Although finerenone has shown positive effects in cardiorenal protection, it may also be accompanied by some side effects. Common side effects include hyperkalemia, headache, nausea, diarrhea and so on. In addition, some patients may experience adverse reactions such as hypoglycemia, and allergies [73,74,75]. Therefore, when using finerenone for disease treatment, it is necessary to pay attention to monitoring the patient's blood pressure and electrolyte levels, and closely observe the patient's condition.

Table 2 Effect of finerenone in clinical treatment

In summary, finerenone is a novel and promising therapeutic drug for patients with chronic kidney disease (CKD), which has received regulatory approval with the indication of cardiorenal protection in patients with CKD associated with type 2 diabetes [76]. And, its indications include cardiovascular related benefits (reducing the risk of cardiovascular death and hospitalization due to heart failure). Although it has not yet been approved for use in DCM, with the expansion of new indications and continuous accumulation of clinical practice in China, finerenone may have broad clinical application prospects in the fields of CKD and chronic cardiovascular disease (CVD).

Conclusions and perspectives

Current studies indicate that finerenone can play an important role in cardiorenal protection. Compared with the first and second generation steroid MRA, the third generation non-steroidal MRA has higher affinity and selectivity for MR, and fewer side effects. A large number of experiments have shown that finerenone can inhibit the overactivation of MR. It can effectively block programmed cell death in the heart, including inhibiting cardiomyocyte apoptosis through the TNFa/TNFR1/CASPASE8 signaling pathway or downregulating PPARγ/CD36 and restoring autophagy in cardiomyocytes. Moreover, finerenone can inhibit oxidative stress, which reduces ROS production through the PPARα/CD36 pathway and inhibition of aldosterone mediated activation of NADPH oxidase (Fig. 1 By Figdraw). At the same time, finerenone can effectively anti-inflammatory and reduce vascular injury. These will lead to a certain therapeutic effect of finerenone in DCM patients (Fig. 2 By Figdraw), but it is also necessary to be alert to its possible side effects. It is worth noting that currently, intestinal microbiota and miRNAs have become relevant factors for the onset of DCM, but further experimental research is needed to investigate the relationship between finerenone and the above two. At the same time, the mechanism of action of finerenone in DCM is not fully understood. Through continuous research in the future, it is expected to become an innovative therapeutic drug in the field of CVD.

Fig. 1
figure 1

Related signaling pathways of finerenone in DCM

Fig. 2
figure 2

Potential protective effects of finerenone in DCM

Availability of data and materials

No datasets were generated or analysed during the current study.

Abbreviations

MR:

Mineralocorticoid receptor

MRAs:

Mineralocorticoid receptor antagonists

DCM:

Diabetic cardiomyopathy

miRNAs:

MicroRNAs

ROS:

Reactive oxygen species

RNS:

Reactive nitrogen species

PCD:

Programmed cell death

GRK:

G protein-coupled receptor-kinase

TNFa:

Tumor necrosis factor alpha

PPAR:

Peroxisome proliferators-activated receptors

FFA:

Free fatty acids

SMC:

Smooth muscle cell

EC:

Endothelial cell

ATG:

Autophagy-associated genes

SOD:

Superoxide dismutase

β-ox:

β-Oxidation

ncRNA:

Noncoding RNAs

PGC-1β:

Peroxisome proliferator-activated receptor coactivator 1β

CKD:

Chronic kidney disease

CVD:

Cardiovascular disease

CytC:

Cytochrome C

Apaf-1:

Apoptotic protease activating factor-1

Aldo:

Aldosterone

References

  1. Goenka L, Padmanaban R, George M. The ascent of mineralocorticoid receptor antagonists in diabetic nephropathy. Curr Clin Pharmacol. 2019;14(2):78–83.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Crompton M, Skinner LJ, Satchell SC, et al. Aldosterone: essential for life but damaging to the vascular endothelium. Biomolecules. 2023;13(6):1004.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Tsujimoto T, Kajio H. Spironolactone use and improved outcomes in patients with heart failure with preserved ejection fraction with resistant hypertension. J Am Heart Assoc. 2020;9(23):e018827.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Kolkhof P, Lawatscheck R, Filippatos G, et al. Nonsteroidal mineralocorticoid receptor antagonism by finerenone—translational aspects and clinical perspectives across multiple organ systems. Int J Mol Sci. 2022;23(16):9243.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Gregg LP, Navaneethan SD. Steroidal or non-steroidal MRAs: should we still enable RAASi use through K binders? Nephrol Dial Transplant. 2023;38(6):1355–65.

    Article  PubMed  CAS  Google Scholar 

  6. Secora AM, Shin J-I, Qiao Y, et al. Hyperkalemia and acute kidney injury with spironolactone use among patients with heart failure. Mayo Clin Proc. 2020;95(11):2408–19.

    Article  PubMed  CAS  Google Scholar 

  7. Naser N, Nalbantic A, Nalbantic N, et al. The effectiveness of eplerenone vs spironolactone on left ventricular systolic function, hospitalization and cardiovascular death in patients with chronic heart failure—HFrEF. Med Arch. 2023;77(2):105.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Kintscher U, Edelmann F. The non-steroidal mineralocorticoid receptor antagonist finerenone and heart failure with preserved ejection fraction. Cardiovasc Diabetol. 2023;22(1):162.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Agarwal R, Kolkhof P, Bakris G, et al. Steroidal and non-steroidal mineralocorticoid receptor antagonists in cardiorenal medicine. Eur Heart J. 2021;42(2):152–61.

    Article  PubMed  CAS  Google Scholar 

  10. Graczyk P, Dach A, Dyrka K, et al. Pathophysiology and advances in the therapy of cardiomyopathy in patients with diabetes mellitus. Int J Mol Sci. 2024;25(9):5027.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Qiao S, Hong L, Zhu Y, et al. RIPK1-RIPK3 mediates myocardial fibrosis in type 2 diabetes mellitus by impairing autophagic flux of cardiac fibroblasts. Cell Death Disease. 2022;13(2):147.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Theofilis P, Vordoni A, Kalaitzidis RG. Oxidative stress management in cardiorenal diseases: focus on novel antidiabetic agents, finerenone, and melatonin. Life. 2022;12(10):1663.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Yang Y, Zhao M, He X, et al. Pyridostigmine protects against diabetic cardiomyopathy by regulating vagal activity, gut microbiota, and branched-chain amino acid catabolism in diabetic mice. Front Pharmacol. 2021;12:647481.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. He X, Kuang G, Wu Y, et al. Emerging roles of exosomal miRNAs in diabetes mellitus. Clin Transl Med. 2021;11(6):e468.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Palanisamy S, Funes Hernandez M, Chang TI, et al. Cardiovascular and renal outcomes with finerenone, a selective mineralocorticoid receptor antagonist. Cardiol Thera. 2022;11(3):337–54.

    Article  Google Scholar 

  16. Jin T, Fu X, Liu M, et al. Finerenone attenuates myocardial apoptosis, metabolic disturbance and myocardial fibrosis in type 2 diabetes mellitus. Diabetol Metab Syndr. 2023;15(1):87.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Pollard CM, Suster MS, Cora N, et al. GRK5 is an essential co-repressor of the cardiac mineralocorticoid receptor and is selectively induced by finerenone. World J Cardiol. 2022;14(4):220–30.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Alvarez de la Rosa D, Dutzmann J, Musmann R-J, et al. The novel mineralocorticoid receptor antagonist finerenone attenuates neointima formation after vascular injury. PLoS ONE. 2017;12(9):e0184888.

    Article  Google Scholar 

  19. Yao L, Liang X, Liu Y, et al. Non-steroidal mineralocorticoid receptor antagonist finerenone ameliorates mitochondrial dysfunction via PI3K/Akt/eNOS signaling pathway in diabetic tubulopathy. Redox Biol. 2023;68: 102946.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Gueret A, Harouki N, Favre J, et al. Vascular smooth muscle mineralocorticoid receptor contributes to coronary and left ventricular dysfunction after myocardial infarction. Hypertension (Dallas, Tex: 1979). 2016;67(4):717–23.

    Article  PubMed  CAS  Google Scholar 

  21. Lachaux M, Barrera-Chimal J, Nicol L, et al. Short- and long-term administration of the non-steroidal mineralocorticoid receptor antagonist finerenone opposes metabolic syndrome-related cardio-renal dysfunction. Diabetes Obes Metab. 2018;20(10):2399–407.

    Article  PubMed  CAS  Google Scholar 

  22. Lima-Posada I, Stephan Y, Soulié M, et al. Benefits of the non-steroidal mineralocorticoid receptor antagonist finerenone in metabolic syndrome-related heart failure with preserved ejection fraction. Int J Mol Sci. 2023;24(3):2536.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Chen Y, Li X, Yang M, et al. Research progress on morphology and mechanism of programmed cell death. Cell Death Disease. 2024;15(5):327.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Liu Z, Chen Y, Mei Y, et al. Gasdermin d-mediated pyroptosis in diabetic cardiomyopathy: molecular mechanisms and pharmacological implications. Molecules. 2023;28(23):7813.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Xie D, Li K, Feng R, et al. Ferroptosis and traditional chinese medicine for type 2 diabetes mellitus. Diabetes Metab Syndr Obes. 2023;16:1915–30.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Wei Y, Yang L, Pandeya A, et al. Pyroptosis-induced inflammation and tissue damage. J Mol Biol. 2022;434(4):167301.

    Article  PubMed  CAS  Google Scholar 

  27. Altamimi JZ, Alfaris NA, Alshammari GM, et al. Esculeoside A decreases diabetic cardiomyopathy in streptozotocin-treated rats by attenuating oxidative stress, inflammation, fibrosis, and apoptosis: impressive role of Nrf2. Medicina. 2023;59(10):1830.

    Article  PubMed  PubMed Central  Google Scholar 

  28. You P, Chen H, Han W, et al. miR-200a-3p overexpression alleviates diabetic cardiomyopathy injury in mice by regulating autophagy through the FOXO3/Mst1/Sirt3/AMPK axis. PeerJ. 2023;11:e15840.

    Article  PubMed  PubMed Central  Google Scholar 

  29. di Lullo L, Lavalle C, Scatena A, et al. Finerenone: questions and answers—the four fundamental arguments on the new-born promising non-steroidal mineralocorticoid receptor antagonist. J Clin Med. 2023;12(12):3992.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Chen Y, Hua Y, Li X, et al. Distinct types of cell death and the implication in diabetic cardiomyopathy. Front Pharmacol. 2020;11:42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Sun S, Yang S, Dai M, et al. The effect of Astragalus polysaccharides on attenuation of diabetic cardiomyopathy through inhibiting the extrinsic and intrinsic apoptotic pathways in high glucose -stimulated H9C2 cells. BMC Complement Altern Med. 2017;17(1):310.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Grubićrotkvić P, Planinić Z, Liberatipršo A-M, et al. The mystery of diabetic cardiomyopathy: from early concepts and underlying mechanisms to novel therapeutic possibilities. Int J Mol Sci. 2021;22(11):5973.

    Article  Google Scholar 

  33. Bernardi S, Michelli A, Zuolo G, et al. Update on RAAS modulation for the treatment of diabetic cardiovascular disease. J Diabetes Res. 2016;2016:1–17.

    Article  Google Scholar 

  34. Cannavo A, Liccardo D, Eguchi A, et al. Myocardial pathology induced by aldosterone is dependent on non-canonical activities of G protein-coupled receptor kinases. Nat Commun. 2016;7(1):10877.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Shi X, Liu C, Chen J, et al. Endothelial MICU1 alleviates diabetic cardiomyopathy by attenuating nitrative stress-mediated cardiac microvascular injury. Cardiovasc Diabetol. 2023;22(1):216.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Morse PT, Arroum T, Wan J, et al. Phosphorylations and acetylations of cytochrome c control mitochondrial respiration, mitochondrial membrane potential, energy, ROS, and apoptosis. Cells. 2024;13(6):493.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Maning J, McCrink K, Pollard C, et al. Antagonistic roles of GRK2 and GRK5 in cardiac aldosterone signaling reveal GRK5-mediated cardioprotection via mineralocorticoid receptor inhibition. Int J Mol Sci. 2020;21(8):2868.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Moss ME, Carvajal B, Jaffe IZ. The endothelial mineralocorticoid receptor: contributions to sex differences in cardiovascular disease. Pharmacol Therapeut. 2019;203:107387.

    Article  CAS  Google Scholar 

  39. Lv R, Xu L, Che L, et al. Cardiovascular-renal protective effect and molecular mechanism of finerenone in type 2 diabetic mellitus. Front Endocrinol. 2023;14:1125693.

    Article  Google Scholar 

  40. Wang H, Wang L, Hu F, et al. Neuregulin-4 attenuates diabetic cardiomyopathy by regulating autophagy via the AMPK/mTOR signalling pathway. Cardiovasc Diabetol. 2022;21(1):205.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Zang H, Wu W, Qi L, et al. Autophagy inhibition enables Nrf2 to exaggerate the progression of diabetic cardiomyopathy in mice. Diabetes. 2020;69(12):2720–34.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Ji N, Qi Z, Wang Y, et al. Pyroptosis: a new regulating mechanism in cardiovascular disease. J Inflamm Res. 2021;14:2647–66.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Lu Y, Lu Y, Meng J, et al. Pyroptosis and its regulation in diabetic cardiomyopathy. Front Physiol. 2022;12:791848.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Byrne NJ, Rajasekaran NS, Abel ED, et al. Therapeutic potential of targeting oxidative stress in diabetic cardiomyopathy. Free Radical Biol Med. 2021;169:317–42.

    Article  CAS  Google Scholar 

  45. de Geest B, Mishra M. Role of oxidative stress in diabetic cardiomyopathy. Antioxidants. 2022;11(4):784.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Liu Q, Wang S, Cai L. Diabetic cardiomyopathy and its mechanisms: role of oxidative stress and damage. J Diabetes Investig. 2014;5(6):623–34.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Peng ML, Fu Y, Wu CW, et al. Signaling pathways related to oxidative stress in diabetic cardiomyopathy. Front Endocrinol (Lausanne). 2022;13: 907757.

    Article  PubMed  Google Scholar 

  48. Sapian S, Taib IS, Latip J, et al. Therapeutic approach of flavonoid in ameliorating diabetic cardiomyopathy by targeting mitochondrial-induced oxidative stress. Int J Mol Sci. 2021;22(21):11616.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Watanabe K, Thandavarayan RA, Harima M, et al. Role of differential signaling pathways and oxidative stress in diabetic cardiomyopathy. Curr Cardiol Rev. 2010;6:280–90.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Jubaidi FF, Zainalabidin S, Mariappan V, et al. Mitochondrial dysfunction in diabetic cardiomyopathy: the possible therapeutic roles of phenolic acids. Int J Mol Sci. 2020;21(17):6043.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Zhou B, Tian R. Mitochondrial dysfunction in pathophysiology of heart failure. J Clin Investig. 2018;128(9):3716–26.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Hamblin M, Friedman DB, Hill S, et al. Alterations in the diabetic myocardial proteome coupled with increased myocardial oxidative stress underlies diabetic cardiomyopathy. J Mol Cell Cardiol. 2007;42(4):884–95.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Gollmer J, Zirlik A, Bugger H. Mitochondrial mechanisms in diabetic cardiomyopathy. Diabetes Metab J. 2020;44(1):33.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Jubaidi FF, Zainalabidin S, Taib IS, et al. The potential role of flavonoids in ameliorating diabetic cardiomyopathy via alleviation of cardiac oxidative stress, inflammation and apoptosis. Int J Mol Sci. 2021;22(10):5094.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Jaquenod de Giusti C, Palomeque J, Mattiazzi A. Ca2+ mishandling and mitochondrial dysfunction: a converging road to prediabetic and diabetic cardiomyopathy. Pflügers Arch Eur J Physiol. 2022;474(1):33–61.

    Article  CAS  Google Scholar 

  56. Galloway CA, Yoon Y. Mitochondrial dynamics in diabetic cardiomyopathy. Antioxid Redox Signal. 2015;22(17):1545–62.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Cai C, Wu F, He J, et al. Mitochondrial quality control in diabetic cardiomyopathy: from molecular mechanisms to therapeutic strategies. Int J Biol Sci. 2022;18(14):5276–90.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Wu L, Wang K, Wang W, et al. Glucagon-like peptide-1 ameliorates cardiac lipotoxicity in diabetic cardiomyopathy via the PPARα pathway. Aging Cell. 2018;17(4): e12763.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Lin Y, Liu R, Huang Y, et al. Reactivation of PPARα alleviates myocardial lipid accumulation and cardiac dysfunction by improving fatty acid β-oxidation in Dsg2-deficient arrhythmogenic cardiomyopathy. Acta Pharmaceut Sin B. 2023;13(1):192–203.

    Article  CAS  Google Scholar 

  60. Son NH, Yu S, Tuinei J, et al. PPARγ-induced cardiolipotoxicity in mice is ameliorated by PPARα deficiency despite increases in fatty acid oxidation. J Clin Investig. 2010;120(10):3443–54.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Cortassa S, Sollott SJ, Aon MA. Mitochondrial respiration and ROS emission during β-oxidation in the heart: an experimental-computational study. PLoS Comput Biol. 2017;13(6): e1005588.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Jia G, Jia Y, Sowers JR. Role of mineralocorticoid receptor activation in cardiac diastolic dysfunction. Biochim Biophys Acta (BBA) Mol Basis Disease. 2017;1863(8):2012–8.

    Article  CAS  Google Scholar 

  63. Huang YL, Xiang Q, Zou JJ, et al. Zuogui Jiangtang Shuxin formula Ameliorates diabetic cardiomyopathy mice via modulating gut-heart axis. Front Endocrinol (Lausanne). 2023;14:1106812.

    Article  PubMed  Google Scholar 

  64. Sah SP, Tirkey N, Kuhad A, et al. Effect of quercetin on lipopolysaccharide induced-sickness behavior and oxidative stress in rats. Indian J Pharmacol. 2011;43(2):192–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Sun X, Jiao X, Ma Y, et al. Trimethylamine N-oxide induces inflammation and endothelial dysfunction in human umbilical vein endothelial cells via activating ROS-TXNIP-NLRP3 inflammasome. Biochem Biophys Res Commun. 2016;481(1–2):63–70.

    Article  PubMed  CAS  Google Scholar 

  66. Saad MJ, Santos A, Prada PO. Linking gut microbiota and inflammation to obesity and insulin resistance. Physiology (Bethesda). 2016;31(4):283–93.

    PubMed  CAS  Google Scholar 

  67. Qiao CM, Sun MF, Jia XB, et al. Sodium butyrate causes α-synuclein degradation by an Atg5-dependent and PI3K/Akt/mTOR-related autophagy pathway. Exp Cell Res. 2020;387(1): 111772.

    Article  PubMed  CAS  Google Scholar 

  68. Zhao G, Zhang X, Wang H, et al. Beta carotene protects H9c2 cardiomyocytes from advanced glycation end product-induced endoplasmic reticulum stress, apoptosis, and autophagy via the PI3K/Akt/mTOR signaling pathway. Ann Transl Med. 2020;8(10):647.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Jakubik D, Fitas A, Eyileten C, et al. MicroRNAs and long non-coding RNAs in the pathophysiological processes of diabetic cardiomyopathy: emerging biomarkers and potential therapeutics. Cardiovasc Diabetol. 2021;20(1):55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Shen E, Diao X, Wang X, et al. MicroRNAs involved in the mitogen-activated protein kinase cascades pathway during glucose-induced cardiomyocyte hypertrophy. Am J Pathol. 2011;179(2):639–50.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Miao Y, Wan Q, Liu X, et al. miR-503 is involved in the protective effect of phase II enzyme inducer (CPDT) in diabetic cardiomyopathy via Nrf2/ARE signaling pathway. Biomed Res Int. 2017;2017:9167450.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Yin Z, Zhao Y, He M, et al. MiR-30c/PGC-1β protects against diabetic cardiomyopathy via PPARα. Cardiovasc Diabetol. 2019;18(1):7.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Bakris GL, Agarwal R, Anker SD, et al. Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. N Engl J Med. 2020;383(23):2219–29.

    Article  PubMed  CAS  Google Scholar 

  74. Pitt B, Filippatos G, Agarwal R, et al. Cardiovascular events with finerenone in kidney disease and type 2 diabetes. N Engl J Med. 2021;385(24):2252–63.

    Article  PubMed  CAS  Google Scholar 

  75. Pitt B, Kober L, Ponikowski P, et al. Safety and tolerability of the novel non-steroidal mineralocorticoid receptor antagonist BAY 94-8862 in patients with chronic heart failure and mild or moderate chronic kidney disease: a randomized, double-blind trial. Eur Heart J. 2013;34(31):2453–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Heinig R, Eissing T. The pharmacokinetics of the nonsteroidal mineralocorticoid receptor antagonist finerenone. Clin Pharmacokinet. 2023;62(12):1673–93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Filippatos G, Anker SD, Böhm M, et al. A randomized controlled study of finerenone vs. eplerenone in patients with worsening chronic heart failure and diabetes mellitus and/or chronic kidney disease. Eur Heart J. 2016;37(27):2105–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Zhang Y, Jiang L, Wang J, et al. Network meta-analysis on the effects of finerenone versus SGLT2 inhibitors and GLP-1 receptor agonists on cardiovascular and renal outcomes in patients with type 2 diabetes mellitus and chronic kidney disease. Cardiovasc Diabetol. 2022;21(1):232.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Funding

This research was funded by grants from Sichuan Science and Technology Program (2022YFS0610), Luzhou Municipal People’s Government—Southwest Medical University Science and Technology Strategic Cooperation (2021LZXNYD-J33), Hejiang County People's Hospital—Southwest Medical University Science and Technology Strategic Cooperation Project (2021HJXNYD13, 2021HJXNYD04 and 2022HJXNYD05), Xuyong County People’s Hospital—Southwest Medical University Science and Technology Strategic Cooperation Project (2024XYXNYD18) and Gulin County People's Hospital—Affiliated Hospital of Southwest Medical University Science and Technology strategic Cooperation (2022GLXNYDFY13), 2022-N-01-33 project of China International Medical Foundation, Provincial-level science and Technology Program Transfer Payment Special Fund project of Panzhihua Science and Technology Bureau (222ZYZF-S-01).

Author information

Authors and Affiliations

Authors

Contributions

JW, HX, and JH conceived, designed, and planned the manuscript. JW, HX, and JH collected and read the literature. JW drafted the manuscript and prepared Figs. 1, 2. LD made extensive revisions to the manuscript during the revision process. JT analyzed the data. JF and YJ conceived, designed, and revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Yang Jiang or Jian Feng.

Ethics declarations

Ethics approval and consent to participate

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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, J., Xue, H., He, J. et al. Therapeutic potential of finerenone for diabetic cardiomyopathy: focus on the mechanisms. Diabetol Metab Syndr 16, 232 (2024). https://doi.org/10.1186/s13098-024-01466-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13098-024-01466-x

Keywords