Open Access

Role of histone deacetylase inhibitors in diabetic cardiomyopathy in experimental models (Review)

  • Authors:
    • Anna Garmpi
    • Christos Damaskos
    • Nikolaos Garmpis
    • Vaios-Vasileios Kaminiotis
    • Vasiliki Epameinondas Georgakopoulou
    • Demetrios A. Spandidos
    • Petros Papalexis
    • Evangelos Diamantis
    • Alexandros Patsouras
    • George Kyriakos
    • Kyriakos Tarantinos
    • Athanasios Syllaios
    • Georgios Marinos
    • Gregory Kouraklis
    • Dimitrios Dimitroulis
  • View Affiliations

  • Published online on: August 2, 2022     https://doi.org/10.3892/mi.2022.51
  • Article Number: 26
  • Copyright: © Garmpi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In diabetes, metabolic dysregulation, caused by hyperglycemia, leads to both structural and functional changes in cardiomyocytes and subsequently leads to the development of cardiomyopathy. Histone deacetylases (HDAC) are enzymes that regulate gene transcription. Their actions have been examined in the development of multiple disorders, such as cardiovascular diseases and diabetes. The use of HDAC inhibitors (HDACIs), as potential therapeutic agents against disease progression has yielded promising results. The present review article reports preclinical trials identified in which HDACIs were administered to mice suffering from diabetic cardiomyopathy (DCM), and discusses the role and mechanisms of action of HDAC and HDACIs in DCM. A review of the literature was performed using the PubMed database, aiming to identify publications in the English language concerning the role of HDACIs in DCM. More specifically, key words, separately and in various combinations, such as HDACIs, HDAC, diabetes, cardiomyopathy, heart failure and ischemia/reperfusion injury, were used. Furthermore, the references from all the articles were cross‑checked in order to include any other eligible studies. The full‑text articles assessed for eligibility were eight, covering the period from 2015 to 2019; finally, all of them were included. The use of HDACIs exhibited encouraging results against DCM progression through various mechanisms, including the reduction of reactive oxygen species generation, inflammatory cytokine production and fibrosis, and an increase in autophagy and angiogenesis.

1. Introduction

To date, studies have indicated that there is a strong association between diabetes and heart failure. Diabetes mellitus (DM) is one of the most common risk factors for the development of non-ischemic heart failure, and its identification results in the initiation of cardioprotective therapy (1). Therefore, cardiovascular complications constitute the main cause of morbidity and mortality in diabetic patients. In total, 80% of related deaths occur due to cardiovascular diseases in these patients (2). These complications include atherosclerosis, coronary artery disease, and myocardial infarction (3). Diabetic cardiomyopathy (DCM), is another cardiac disorder caused by diabetes (4).

DCM is defined as a heart disorder of a diabetic patient in which abnormal myocardial structures, such as coronary lesions, valvular heart disease and hypertension are absent. This disorder is characterized by diastolic or systolic dysfunction, combined with structural changes, such as fibrosis and hypertrophy (5). The pathophysiology of DCM is complex and involves various mechanisms, such as inflammation, mitochondrial dysfunction, apoptosis, oxidative stress, lipotoxicity, fibrosis and disruptions in insulin signaling (6). Even though extensive research has been conducted concerning diabetic cardiovascular disorders, the efficacy of treatment remains a challenge (7,8).

Multiple contributing mechanisms are associated with diabetic heart disease. It is well established that hyperglycemia plays a critical role in diabetes-related heart failure (9,10). Following a protein modification mechanism and the induction of epigenetic alterations, as well as mitochondrial damage, hyperglycemia results in myocardial dysfunction. Oxidative stress is also associated with both non-cardiac and cardiac diabetes-related complications. Oxidative stress usually provokes impaired cardiomyocyte calcium handling and leads to reduced cardiac contractility and relaxation. Inflammation is another mechanism that leads to the progression of diabetes. Increased inflammatory signaling often results in macrophage infiltration and thus, in cardiac DM-related complications (9).

Moreover, autonomic dysfunction has an impact on myocardial performance, and systemic and coronary vascular function (11). Autonomic dysfunction is a significant complication of DM that is firmly linked to a roughly five-fold increased risk of cardiovascular mortality. Its presentation ranges from resting tachycardia and a fixed heart rate to the development of silent myocardial infarction (12).

Consequently, myocardial hypertrophy, fibrosis and myocardial dysfunction lead to the progression of heart failure. Cardiac complications, such as disrupted insulin signaling, the renin-angiotensin-aldosterone system, and small and large-vessel disease lead to alterations in myocardial energetics, cardiac remodeling and impaired perfusion (11). Myocardial energy depletion in individuals suffering from DM is multifactorial and is associated with limitations in the uptake and utilization of substrates, mitochondrial dysfunction and affected energy transfer from mitochondria to myofibrils. These metabolic alterations combined with affected myocardial perfusion, may result in the reduced ability of the heart to cope with acute increases in workload (13).

As a result, myocardial dysfunction, cardiac stiffness and myocardial fibrosis occur. Finally, cytosolic calcium trafficking and gene expression cause excitation-contraction decoupling, and lead to cardiomyocyte contraction and relaxation (11).

Histone deacetylases (HDACs) are enzymes that exert their activities through the alteration of chromatin regulation. The balance between HDACs and histone acetyltransferases (HATs) controls that regulation. As their name suggests, they remove acetyl-groups from histones. They actually regulate the interaction between the negatively charged DNA and positively charged histones. Thus, they serve as a repressor of transcription (14) (Fig. 1). The 18 HDACs are divided into four classes as follows: The class I Rpd3-like proteins (HDAC1, HDAC2, HDAC3 and HDAC8); the class II Hda1-like proteins (HDAC4, HDAC5, HDAC6, HDAC7, HDAC9 and HDAC10); the class III Sir2-like proteins [sirtuin (SIRT)1, SIRT2, SIRT3, SIRT4, SIRT5, SIRT6 and SIRT7]; and the class IV protein (HDAC11) (15).

The HDACs of classes I, II and IV exhibit a zinc-dependent activity. On the contrary, class III HDACs or SIRTs (SIRT1-7) require nicotinamide adenine dinucleotide (NAD) for catalysis (Table I) (14). Their crucial roles in epigenetics and gene expression have rendered them potential targets in various diseases, including both metabolic and cardiovascular entities (16).

Table I

HDAC classification.

Table I

HDAC classification.

Class IClass IIClass IIIClass IV
Zn+-dependentNAD-dependent Zn+-dependent
HDAC1IIASIRT1HDAC11
HDAC2      HDAC4SIRT2 
HDAC3      HDAC5SIRT3 
HDAC8      HDAC7SIRT4 
       HDAC9SIRT5 
  SIRT6 
  SIRT7 
 IIB  
       HDAC6  
       HDAC10  

[i] HDAC, histone deacetylase; SIRT, sirtuin; NAD, nicotinamide adenine dinucleotide.

HDAC inhibitors (HDACIs) are already used as anticancer agents. Of note, four of them have already been approved by the US Food and Drug Administration (FDA) against hematological malignancies (14). Additionally, their anticancer activity has been reported extensively in the literature, including colorectal, hepatocellular, pancreatic, breast, thyroid, lung, endometrial, and other cancers such as uveal and cutaneous melanoma (17-27). Furthermore, their role appear to be crucial in fighting other diseases, including viral infections and both inflammatory and neurological disorders (28,29). Hyperglycemia is a crucial factor that contributes to the development of oxidative stress, mitochondrial injury, reactive oxygen species (ROS) and myocardial inflammation (30,31). Thus, HDACIs appear to be promising therapeutic agents against cardiovascular diseases, including DCM.

The present review article reports pre-clinical trials identified in which HDACIs were tested in animal models of DCM. The roles and mechanisms of action of HDACIs in metabolic and cardiovascular diseases were also discussed.

2. Role of HDACIs in experimental models of DCM

In 2015, Chen et al (31) examined the role of sodium butyrate, which is a specific HDACI, in preventing myocardial dysfunction in diabetic mice. They established a model of diabetes using mice by injecting the mice intraperitoneally with streptozocin. The next step was the administration of sodium butyrate to certain subgroups of these mice. The echocardiography findings revealed that cardiac dysfunction was attenuated in he diabetic mice receiving sodium butyrate. Additionally, the attenuation of cardiac remodeling and interstitial fibrosis were noted. These findings were associated with an increase in cardiac angiogenesis, and a decrease in both apoptosis and active caspase 3. Glucose metabolism was improved through the upregulation of glucose transporter (GLUT)-1 and -4. Finally, sodium butyrate exerted an antioxidant effect on the myocardium by elevating superoxide dismutase levels (31).

Lee et al (32,33) conducted research on diabetic rats who received low doses of streptozocin. MPT0E014, which is a pan-HDACI, was administered to a subgroup of diabetic rats. The left-ventricular end-diastolic diameter was smaller in the diabetic rats receiving the HDACI compared to the diabetic rats not receiving the treatment. The reduction in blood sugar and triglyceride levels revealed the anti-hyperglycemic and hypolipidemic effects of MPT0E014. At the cellular level, GLUT-4 expression was augmented through HDACI administration via an insulin-independent pathway. The cleavage of poly(ADP-ribose) polymerase 1 (PARP-1) was decreased, inhibiting apoptosis. The levels of pro-inflammatory cytokines, such as tumor necrosis factor α (TNF-α) or interleukin (IL)-6 were reduced following treatment with MPT0E014(32). Finally, the procedure of autophagy, which is necessary for the development and homeostasis of cells, was accelerated following the administration of this HDACI (33).

Wu et al (34) investigated whether the suppression of HDACs in diabetic rat hearts could protect the rats from myocardial ischemia/reperfusion (MI/R) injury mediated via the PI3K/Akt pathway (34). This pathway results in the activation of anti-apoptotic proteins that inhibit mitochondrial dysfunction during MI/R injury (34,35). In their experiment, diabetic rats underwent 45 min of ischemia followed by 3 h of reperfusion. They used trichostatin A (TSA), a selective I and II HDACI, both in vitro and in vivo experiments. Their study was the first to demonstrate that MI/R injury and diabetes increased HDAC activity in rat hearts. However, TSA protected the function and integrity of mitochondria both in vivo and in vitro through the expression of p-Akt via the increased phosphorylation of p-Foxo3a and Foxo3a in the cytoplasm of myocardial cells (34).

Leng et al (36) used the same methods and experimental model as those in the study by Wu et al (34); however, they concentrated on the inhibition of HDAC6 activity using tubastatin A (Tub A), a highly selective HDAC6 inhibitor. Their research demonstrated that both in vivo and in vitro, Tub A exhibited its cardioprotective actions in diabetic rat hearts by modulating peroxiredoxin 1 (Prdx1) acetylation and attenuating ROS generation and subsequent cellular injury (36).

Another group of researchers examined the association between the inhibition of HDAC3 and DCM. RGFP966, a selective HDAC3 inhibitor, and valproic acid were administered to mice with type 1 diabetes (37). Treatment with RGFP966 prevented cardiac hypertrophy through various mechanisms. Firstly, a reduction in heart hypertrophy and fibrosis markers was noted. Histologically, cardiomyocyte size, fibrosis and the accumulation of collagen were suppressed. Insulin resistance, oxidative stress and inflammatory processes were all diminished. TNF-α, plasminogen activator inhibitor-1 and ROS production were downregulated, whereas the expression of GLUT-4 was upregulated. The dual specificity phosphatase 5 (DUSP5), which is an extracellular signal-regulated kinase 1 and 2 (ERK1/2) nuclear phosphatase, inhibits phosphorylated extracellular signal-regulated kinases ERK1/2, which accelerates cardiac hypertrophy (37). The administration of RGFP966 exerted its cardioprotective effects through an increase in the levels of DUSP5. Last but not least, they demonstrated that these effects lasted for a period of time, since the protective activity of RGFP966 remained for 3 months after the end of its administration (37).

Zhang et al (7) performed a study in which they administered sodium butyrate to mice with type 2 diabetes. Treatment with sodium butyrate reduced obesity, hypercholesterolemia, and glucose intolerance. In addition, the wall thickness and the internal dimension of the left ventricle were smaller in the diabetic group treated with sodium butyrate, compared with the untreated diabetic group. Both the size of the cardiomyocytes and collagen presence were limited in the mice receiving the HDACI. The production of superoxide and active caspase-3, which is an apoptotic agent, were significantly reduced. Angiogenesis and capillary formation were enhanced using sodium butyrate. Finally, sodium butyrate can activate signaling pathways, such as mitogen-activated protein kinase 3 (MKK3)/p38/regulated-activated kinase (PRAK) and Akt-1, in order to regulate proper cardiac function (7).

Bocchi et al (38) reported the action of suberoylanilide hydroxamic acid (SAHA), a pan-HDACI, in the cardiomyocytes of diabetic rats. Ventricular cardiomyocytes were extracted in order to measure their contractility and calcium dynamics. These cells were either treated with SAHA for 90 min or left untreated. The group treated with SAHA exhibited higher contractility and calcium dynamics. This was a result of the upregulation in ryanodine receptors and a decrease in intracellular ROS levels (38).

Table II summarizes the findings of all the aforementioned studies reporting the potential role of HDACIs against DCM in experimental models (5,31-34,36-38).

Table II

Preclinical studies of histone deacetylase inhibitors in diabetic cardiomyopathy.

Table II

Preclinical studies of histone deacetylase inhibitors in diabetic cardiomyopathy.

Authors, yearSpecimenHDACIOutcome(Refs.)
Chen et al, 2015• Control miceSodium butyrate (inhibitor of HDAC class I and IIA)Reduction of apoptosis, oxidative stress, cardiac remodeling, fibrosis. Upregulation of GLUT-1 and -4, and angiogenesis.(31)
• Control receiving sodium butyrate mice
• Mice with streptozocin-induced diabetes
• Mice with streptozocin-induced diabetes receiving sodium butyrate
Zhang et al, 2017• Mice fed a high-fat dietSodium butyrateReduction of obesity, hypercholesterolemia, hyperglycemia, apoptosis, oxidative stress, left ventricular wall thickness and internal diameter. Increase in angiogenesis.(7)
• Mice fed a high-fat diet and treated with sodium butyrate
• Mice fed a standard chow diet
• Mice fed a standard chow diet and treated with sodium butyrate
Wu et al, 2017• Control ratsTrichostatin AProtected against myocardial ischemia/reperfusion injury and hypoxia/reoxygenation injury under diabetic. conditions(34)
• Control rats subjected to myocardial ischemia/reperfusion injury
• Diabetic rats
• Diabetic rats subjected to myocardial ischemia/reperfusion injury
Xu et al, 2017• Mice with type 1 diabetesRGFP966 (Selective HDAC-3 inhibitor), VPA (pan-HDACI)Reduction of cardiac hypertrophy, cardiomyocyte size, fibrosis, insulin resistance, inflammation and oxidative stress. Increase in levels of DUSP5.(37)
• Mice with type 1 diabetes treated with VPA
• Mice with type 1 diabetes treated with RGFP966
• Control
• Control and VPA
• Control and RGFP966
Lee et al, 2016 and 2018• ControlMPT0E014 (pan-HDACI)Reduction of glucose, triglycerides, inflammatory cytokines and apoptosis. Up-regulation of autophagy, GLUT-4 and homeostasis.(32,33)
• Rats fed a high-fat diet with streptozocin-induced T2D and treated with MPT0E014
• Rats fed a high-fat diet with streptozocin-induced T2D
Leng et al, 2018• Control ratsTubastatin AProtected against myocardial ischemia/reperfusion injury under diabetic conditions(36)
• Control rats with ischemia/reperfusion injury
• Diabetic rats
• Diabetic rats with ischemia/reperfusion injury
Bocchi et al, 2019• ControlSAHA (pan-HDACI)Increase in contractility and calcium dynamics. Reduction in intracellular oxidative stress.(38)
• Mice with streptozocin-induced type 2 diabetes
• Mice with streptozocin-induced type 2 diabetes and treated with SAHA

[i] DCM, diabetic cardiomyopathy; HDACI, histone deacetylase inhibitor; HDAC, histone deacetylase; GLUT, glucose transporter; T2D, type 2 diabetes; VPA, valproic acid; DUSP5, dual specificity phosphatase 5; SAHA, suberoylanilide hydroxamic acid.

3. Mechanisms of prevention of cardiac hypertrophy via the inhibition of HDACs

HDACIs are divided according to their actions on HDACs. They are divided into hydroxamic acids, short-chain fatty acids, cyclic peptides and benzamides. They are mostly studied and used against malignancies (8). However, extensive research into the possible therapeutic effects of HDACIs on DCM has been performed (5,31-34,36-38). HDAsC contribute to the development of DCM through various metabolic and hypertrophic pathways (39).

The inhibition of HDACs prevents cardiac hypertrophy a number of mechanisms. As described above, active caspase-3 is a protein which triggers apoptosis. The anti-apoptotic effects of HDACIs are demonstrated through the reduction of apoptosis (31). The inflammatory process is reduced through the downregulation of pro-inflammatory cytokines, such as TNF-α, IL-1, IL-6 and IFN-γ (40). Angiogenesis is accomplished through the increased production of vascular endothelial growth factor (VEGF) (41). The reduction of ROS generation diminishes oxidative stress (37) and can be accomplished by the modulation of acetylation of Prdx1(36). Autophagy is an adaptive mechanism, whose absence induces intracellular death (42). As Lee et al (32,33) demonstrated, HDACIs can increase autophagy and protect the myocardium. Wu et al (34) demonstrated that the protection of the function of mitochondria via the regulation of P13K/Akt expression decreased cell apoptosis and protected the diabetic heart. The transcription of GLUT-4 is regulated by class II HDAC in skeletal muscles (43,44). The upregulation of GLUT by HDACIs leads to the entrance of glucose into cells and to a reduction in both insulin resistance and hyperglycemia (32,45). Fibrosis is reduced by inhibiting TGF-β and the angiotensin type 2 receptor (46). The mechanisms of action of HDACIσ against DCM are reported in Table III and are illustrated in Fig. 2.

Table III

Preventive mechanisms of action of HDACIs against DCM.

Table III

Preventive mechanisms of action of HDACIs against DCM.

ReductionIncrease
ROSAutophagy
Inflammatory cytokines (IL-1, IL-6, TNF-α, IFN-γ)GLUT-1 and -4
Apoptosis (caspase-3)Angiogenesis
FibrosisContractility
Left ventricular thickness and internal diameter Insulin resistanceCalcium dynamics

[i] DCM, diabetic cardiomyopathy; ROS, reactive oxygen species; GLUT, glucose transporters.

4. Conclusion and future perspectives

Conventionally, HDACIs are considered as anticancer drugs. However, they appear to be useful in the treatment of other non-malignant diseases. The present review article reported numerous preclinical trials, which examined the association between HDACs, HDACIs and DCM. The use of HDACIs has yielded encouraging results against disease progression. Research has markedly enhance current knowledge on HDACs and their roles in metabolic and cardiovascular dysfunction. Additionally, HDACs are considered as possible therapeutic targets against heart failure in general. Due to the novelty of this approach, further research into the mechanisms of action and biochemical pathways of HDACs and their inhibitors is warranted prior to any clinical applications. Extending current knowledge regarding the therapeutic potential of HDACIs against these diseases will pave the way for the discovery of novel targeted epigenetic drugs. Thus, millions of individuals will be beneficially affected worldwide in the future.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

AG, CD, NG and VVK conceptualized the study. VEG, DAS, PP, ED, AP, AS, KT, GM, Gky, GKo and DD analyzed the data from the literature to be included in the review, and wrote and prepared the draft of the manuscript. GKo and DD provided critical revisions. ED, AS and AP prepared the figure and the tables. All authors contributed to manuscript revision and have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Wang Y, Yang H, Huynh Q, Nolan M, Negishi K and Marwick TH: Diagnosis of nonischemic stage B heart failure in type 2 diabetes mellitus. JACC Cardiovasc Imaging. 11:1390–1400. 2018.PubMed/NCBI View Article : Google Scholar

2 

Hölscher ME, Bode C and Bugger H: Diabetic cardiomyopathy: Does the type of diabetes matter? Int J Mol Sci. 17(2136)2016.PubMed/NCBI View Article : Google Scholar

3 

Yilmaz S, Canpolat U, Aydogdu S and Abboud HE: Diabetic cardiomyopathy; Summary of 41 years. Korean Circ J. 45:266–272. 2015.PubMed/NCBI View Article : Google Scholar

4 

Avogaro A, de Kreutzenberg SV, Negut C, Tiengo A and Scognamiglio R: Diabetic cardiomyopathy: A metabolic perspective. Am J Cardiol. 93:13A–16A. 2004.PubMed/NCBI View Article : Google Scholar

5 

Jia G, Hill MA and Sowers JR: Diabetic cardiomyopathy: An update of mechanisms contributing to this clinical entity. Circ Res. 122:624–638. 2018.PubMed/NCBI View Article : Google Scholar

6 

Dinarello CA: Anti-inflammatory agents: Present and future. Cell. 140:935–950. 2010.PubMed/NCBI View Article : Google Scholar

7 

Zhang L, Du J, Yano N, Wang H, Zhao YT, Dubielecka PM, Zhuang S, Chin YE, Qin G and Zhao TC: Sodium butyrate protects-against high fat diet-induced cardiac dysfunction and metabolic disorders in type II diabetic mice. J Cell Biochem. 118:2395–2408. 2017.PubMed/NCBI View Article : Google Scholar

8 

Jia G, Whaley-Connell A and Sowers JR: Diabetic cardiomyopathy: A hyperglycaemia- and insulin-resistance-induced heart disease. Diabetologia. 61:21–28. 2018.PubMed/NCBI View Article : Google Scholar

9 

Dhingra R and Vasan RS: Diabetes and the risk of heart failure. Heart Fail Clin. 8:125–133. 2012.PubMed/NCBI View Article : Google Scholar

10 

Rosano GM, Vitale C and Seferovic P: Heart failure in patients with diabetes mellitus. Card Fail Rev. 3:52–55. 2017.PubMed/NCBI View Article : Google Scholar

11 

Marwick TH, Ritchi R, Shaw JE and Kaye D: Implications of underlying mechanisms for the recognition and management of diabetic cardiomyopathy. J Am Coll Cardiol. 71:339–351. 2018.PubMed/NCBI View Article : Google Scholar

12 

Serhiyenko VA and Serhiyenko AA: Cardiac autonomic neuropathy: Risk factors, diagnosis and treatment. World J Diabetes. 9:1–24. 2018.PubMed/NCBI View Article : Google Scholar

13 

Levelt E, Rodgers CT, Clarke WT, Mahmod M, Ariga R, Francis JM, Liu A, Wijesurendra RS, Dass S, Sabharwal N, et al: Cardiac energetics, oxygenation, and perfusion during increased workload in patients with type 2 diabetes mellitus. Eur Heart J. 37:3461–3469. 2016.PubMed/NCBI View Article : Google Scholar

14 

Garmpi A, Garmpis N, Damaskos C, Valsami S, Spartalis E, Lavaris A, Patelis N, Margonis GA, Apostolou KG, Spartalis M, et al: Histone deacetylase inhibitors as a new anticancer option: How far can we go with expectations? J BUON. 23:846–861. 2018.PubMed/NCBI

15 

Seto E and Yoshida M: Erasers of histone acetylation: The histone deacetylase enzymes. Cold Spring Harb Perspect Biol. 6(a018713)2014.PubMed/NCBI View Article : Google Scholar

16 

Zwergel C, Stazi G, Valente S and Mai A: Histone deacetylase inhibitors: Updated studies in various epigenetic-related diseases. J Clin Epigenet. 2(1)2016.

17 

Garmpis N, Damaskos C, Garmpi A, Nonni A, Georgakopoulou VE, Antoniou E, Schizas D, Sarantis P, Patsouras A, Syllaios A, et al: Histone deacetylases and their inhibitors in colorectal cancer therapy: Current evidence and future considerations. Curr Med Chem. 29:2979–2994. 2022.PubMed/NCBI View Article : Google Scholar

18 

Garmpis N, Damaskos C, Garmpi A, Georgakopoulou VE, Sarantis P, Antoniou EA, Karamouzis MV, Nonni A, Schizas D, Diamantis E, et al: Histone deacetylase inhibitors in the treatment of hepatocellular carcinoma: Current evidence and future opportunities. J Pers Med. 11(223)2021.PubMed/NCBI View Article : Google Scholar

19 

Damaskos C, Garmpis N, Karatzas T, Nikolidakis L, Kostakis ID, Garmpi A, Karamaroudis S, Boutsikos G, Damaskou Z, Kostakis A and Kouraklis G: Histone deacetylase (HDAC) inhibitors: Current evidence for therapeutic activities in pancreatic cancer. Anticancer Res. 35:3129–3135. 2015.PubMed/NCBI

20 

Damaskos C, Garmpis N, Valsami S, Kontos M, Spartalis E, Kalampokas T, Kalampokas E, Athanasiou A, Moris D, Daskalopoulou A, et al: Histone deacetylase inhibitors: An attractive therapeutic strategy against breast cancer. Anticancer Res. 37:35–46. 2017.PubMed/NCBI View Article : Google Scholar

21 

Garmpis N, Damaskos C, Garmpi A, Kalampokas E, Kalampokas T, Spartalis E, Daskalopoulou A, Valsami S, Kontos M, Nonni A, et al: Histone deacetylases as new therapeutic targets in triple-negative breast cancer: Progress and promises. Cancer Genomics Proteomics. 14:299–313. 2017.PubMed/NCBI View Article : Google Scholar

22 

Damaskos C, Garmpis N, Valsami S, Spartalis E, Antoniou EA, Tomos P, Karamaroudis S, Zoumpou T, Pergialiotis V, Stergios K, et al: Histone deacetylase inhibitors: A novel therapeutic weapon against medullary thyroid cancer? Anticancer Res. 36:5019–5024. 2016.PubMed/NCBI View Article : Google Scholar

23 

Spartalis E, Athanasiadis DI, Chrysikos D, Spartalis M, Boutzios G, Schizas D, Garmpis N, Damaskos C, Paschou SA, Ioannidis A, et al: Histone deacetylase inhibitors and anaplastic thyroid carcinoma. Anticancer Res. 39:1119–1127. 2019.PubMed/NCBI View Article : Google Scholar

24 

Damaskos C, Tomos I, Garmpis N, Karakatsani A, Dimitroulis D, Garmpi A, Spartalis E, Kampolis CF, Tsagkari E, Loukeri AA, et al: Histone deacetylase inhibitors as a novel targeted therapy against non-small cell lung cancer: Where are we now and what should we expect? Anticancer Res. 38:37–43. 2018.PubMed/NCBI View Article : Google Scholar

25 

Garmpis N, Damaskos C, Garmpi A, Spartalis E, Kalampokas E, Kalampokas T, Margonis GA, Schizas D, Andreatos N, Angelou A, et al: Targeting histone deacetylases in endometrial cancer: A paradigm-shifting therapeutic strategy? Eur Rev Med Pharmacol Sci. 22:950–960. 2018.PubMed/NCBI View Article : Google Scholar

26 

Moschos MM, Dettoraki M, Androudi S, Kalogeropoulos D, Lavaris A, Garmpis N, Damaskos C, Garmpi A and Tsatsos M: The role of histone deacetylase inhibitors in uveal melanoma: Current evidence. Anticancer Res. 38:3817–3824. 2018.PubMed/NCBI View Article : Google Scholar

27 

Garmpis N, Damaskos C, Garmpi A, Dimitroulis D, Spartalis E, Margonis GA, Schizas D, Deskou I, Doula C, Magkouti E, et al: Targeting histone deacetylases in malignant melanoma: A future therapeutic agent or just great expectations? Anticancer Res. 37:5355–5362. 2017.PubMed/NCBI View Article : Google Scholar

28 

Gray SG: Epigenetic treatment of neurological disease. Epigenomics. 3:431–450. 2011.PubMed/NCBI View Article : Google Scholar

29 

Savi M, Bocchi L, Sala R, Frati C, Lagrasta C, Madeddu D, Falco A, Pollino S, Bresciani L, Miragoli M, et al: Parenchymal and stromal cells contribute to pro-inflammatory myocardial environment at early stages of diabetes: Protective role of resveratrol. Nutrients. 8(729)2016.PubMed/NCBI View Article : Google Scholar

30 

Bugger H and Abel ED: Molecular mechanisms of diabetic cardiomyopathy. Diabetologia. 57:660–671. 2014.PubMed/NCBI View Article : Google Scholar

31 

Chen Y, Du J, Zhao YT, Zhang L, Lv G, Zhuang S, Qin G and Zhao TC: Histone deacetylase (HDAC) inhibition improves myocardial function and prevents cardiac remodeling in diabetic mice. Cardiovasc Diabetol. 14(99)2015.PubMed/NCBI View Article : Google Scholar

32 

Lee TI, Kao YH, Tsai WC, Chung CC, Chen YC and Chen YJ: HDAC inhibition modulates cardiac PPARs and fatty acid metabolism in diabetic cardiomyopathy. PPAR Res. 2016(5938740)2016.PubMed/NCBI View Article : Google Scholar

33 

Lee TI, Bai KJ, Chen YC, Lee TW, Chung CC, Tsai WC, Tsao SY and Kao YH: Histone deacetylase inhibition of cardiac autophagy in rats on a high-fat diet with low-dose streptozotocin-induced type 2 diabetes mellitus. Mol Med Rep. 17:594–601. 2018.PubMed/NCBI View Article : Google Scholar

34 

Wu Y, Leng Y, Meng Q, Xue R, Zhao B, Zhan L and Xia Z: Suppression of excessive histone deacetylases activity in diabetic hearts attenuates myocardial ischemia/reperfusion injury via mitochondria apoptosis pathway. J Diabetes Res. 2017(8208065)2017.PubMed/NCBI View Article : Google Scholar

35 

Ban K, Cooper AJ, Samuel S, Bhatti A, Patel M, Izumo S, Penninger JM, Backx PH, Oudit GY and Tsushima RG: Phosphatidylinositol 3-kinase gamma is a critical mediator of myocardial ischemic and adenosine-mediated preconditioning. Circ Res. 103:643–653. 2008.PubMed/NCBI View Article : Google Scholar

36 

Leng Y, Wu Y, Lei S, Zhou B, Qiu Z, Wang K and Xia Z: Inhibition of HDAC6 activity alleviates myocardial ischemia/reperfusion injury in diabetic rats: Potential role of peroxiredoxin 1 acetylation and redox regulation. Oxid Med Cell Longev. 2018(9494052)2018.PubMed/NCBI View Article : Google Scholar

37 

Xu Z, Tong Q, Zhang Z, Wang S, Zheng Y, Liu Q, Qian LB, Chen SY, Sun J and Cai L: Inhibition of HDAC3 prevents diabetic cardiomyopathy in OVE26 mice via epigenetic regulation of DUSP5-ERK1/2 pathway. Clin Sci (Lond). 131:1841–1857. 2017.PubMed/NCBI View Article : Google Scholar

38 

Bocchi L, Motta BM, Savi M, Vilella R, Meraviglia V, Rizzi F, Galati S, Buschini A, Lazzaretti M, Pramstaller PP, et al: The histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) restores cardiomyocyte contractility in a rat model of early diabetes. Int J Mol Sci. 20:1873–1885. 2019.PubMed/NCBI View Article : Google Scholar

39 

Bagshi RA and Weeks KL: Histone deacetylases in cardiovascular and metabolic diseases. J Mol Cell Cardiol. 130:151–159. 2019.PubMed/NCBI View Article : Google Scholar

40 

Larsen L, Tonnesen M, Ronn SG, Størling J, Jørgensen S, Mascagni P, Dinarello CA, Billestrup N and Mandrup-Poulsen T: Inhibition of histone deacetylases prevents cytokine-induced toxicity in beta cells. Diabetologia. 50:779–789. 2007.PubMed/NCBI View Article : Google Scholar

41 

Bogaard HJ, Mizuno S, Hussaini AA, Toldo S, Abbate A, Kraskauskas D, Kasper M, Natarajan R and Voelkel NF: Suppression of histone deacetylases worsens right ventricular dysfunction after pulmonary artery banding in rats. Am J Respir Crit Cure Med. 183:1402–1410. 2011.PubMed/NCBI View Article : Google Scholar

42 

Hur KY, Jung HS and Lee MS: Role of autophagy in β-cell function and mass. Diabetes Obes Metab. 2 (12 Suppl):S20–S26. 2010.PubMed/NCBI View Article : Google Scholar

43 

Weems J and Olson AL: Class II histone deacetylases limit GLUT4 gene expression during adipocyte differentiation. J Biol Chem. 286:460–468. 2011.PubMed/NCBI View Article : Google Scholar

44 

Thai MV, Guruswamy S, Cao KT, Pessin JE and Olson AL: Myocyte enhancer factor 2 (MEF2)-binding site is required for GLUT4 gene expression in transgenic mice. Regulation of MEF2 DNA binding activity in insulin-deficient diabetes. J Biol Chem. 273:14285–14292. 1998.PubMed/NCBI View Article : Google Scholar

45 

Shao D and Tian R: Glucose transporters in cardiac metabolism and hypertrophy. Compr Physiol. 6:331–351. 2015.PubMed/NCBI View Article : Google Scholar

46 

Kao YH, Liou JP, Chung CC, Lien GS, Kuo CC, Chen SA and Chen YJ: Histone deacetylase inhibition improved cardiac functions with direct antifibrotic activity in heart failure. Int J Cardiol. 168:4178–4183. 2013.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

July-August 2022
Volume 2 Issue 4

Print ISSN: 2754-3242
Online ISSN:2754-1304

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Garmpi A, Damaskos C, Garmpis N, Kaminiotis V, Georgakopoulou VE, Spandidos DA, Papalexis P, Diamantis E, Patsouras A, Kyriakos G, Kyriakos G, et al: Role of histone deacetylase inhibitors in diabetic cardiomyopathy in experimental models (Review). Med Int 2: 26, 2022
APA
Garmpi, A., Damaskos, C., Garmpis, N., Kaminiotis, V., Georgakopoulou, V.E., Spandidos, D.A. ... Dimitroulis, D. (2022). Role of histone deacetylase inhibitors in diabetic cardiomyopathy in experimental models (Review). Medicine International, 2, 26. https://doi.org/10.3892/mi.2022.51
MLA
Garmpi, A., Damaskos, C., Garmpis, N., Kaminiotis, V., Georgakopoulou, V. E., Spandidos, D. A., Papalexis, P., Diamantis, E., Patsouras, A., Kyriakos, G., Tarantinos, K., Syllaios, A., Marinos, G., Kouraklis, G., Dimitroulis, D."Role of histone deacetylase inhibitors in diabetic cardiomyopathy in experimental models (Review)". Medicine International 2.4 (2022): 26.
Chicago
Garmpi, A., Damaskos, C., Garmpis, N., Kaminiotis, V., Georgakopoulou, V. E., Spandidos, D. A., Papalexis, P., Diamantis, E., Patsouras, A., Kyriakos, G., Tarantinos, K., Syllaios, A., Marinos, G., Kouraklis, G., Dimitroulis, D."Role of histone deacetylase inhibitors in diabetic cardiomyopathy in experimental models (Review)". Medicine International 2, no. 4 (2022): 26. https://doi.org/10.3892/mi.2022.51