Open access peer-reviewed chapter

Experimental Model of Cardiotoxicity

Written By

Oytun Erbaş, İlknur Altuntaş, Özge Çağlar, Elif Özyilmaz, Ece Sari, İlayda Üzümcü and Kaan Erbakan

Submitted: 21 September 2021 Reviewed: 26 October 2021 Published: 07 January 2022

DOI: 10.5772/intechopen.101401

From the Edited Volume

Risk Factors for Cardiovascular Disease

Edited by Johnny Chahine

Chapter metrics overview

355 Chapter Downloads

View Full Metrics

Abstract

The occurrence of heart electrophysiology dysfunction or/and muscle damage is referred to as cardiotoxicity. The heart weakens and becomes less efficient at pumping and hence circulating blood. Cardiomyopathy can be caused by a variety of factors, including viral infections, diseases such as diabetes, ischemia, hypertension, obesity, radiation therapy, antipsychotic drugs, cytotoxic drugs, most notably chemotherapeutic agents; antitumor antibiotics, monoclonal antibodies, tyrosine kinase inhibitors, platinum-based compounds, microtubule inhibitors, vinca alkaloids, antimetabolites, proteasome inhibitors, topoisomerase inhibitors, alkylating agents, corticosteroids. This chapter focuses on the mechanisms of cardiotoxicity, animal models and transgenic methods used in studies, and the effects of therapeutic agents on cardiotoxicity.

Keywords

  • cardiotoxicity
  • cardiomyopathy
  • chemotherapeutics
  • diabetes
  • ischemia
  • radiation therapy
  • toxicants
  • transgenic animal models

1. Introduction

Globally, heart disease is responsible for a third of all deaths [1]. Cardiotoxicity occurs when the heart, in whole or in part, is damaged as a result of factors such as obesity, chemotherapy (CT), alcohol exposure, anorexia, neurosis, unconscious drug use, occupational and environmental heavy metal exposure [2, 3, 4].

Toxicants impair pumping efficiency by reducing the number of active myocytes, cause oxidative damage and lipid peroxidation, which results in cell swelling, altered Ca2+ homeostasis, and irreversible myocyte injury, alter aerobic metabolism, myocardial conduction, cell membrane function, directly damaged myocardium, and induce vascular changes [5]. They also cause QT interval prolongation and ionic channel blockage, which can lead to syncope and ventricular fibrillation. The heart weakens and becomes less efficient at pumping and thus circulating blood. Because of the high energy demands of the heart, it is susceptible to toxins that interfere with oxygen availability, carbohydrate metabolism, and oxidative phosphorylation [6, 7].

Cardiotoxicity is characterized by cardiac dysfunctions, arrhythmia (changes in heart rhythm), hypotension, tachypnea, edema, heart muscle damage (cardiomyopathy), changes in transmission pathways, and toxic effects on the heart [3]. Cardiotoxicity includes changes in resting cardiac measurements as well as dynamic functional evaluations of the cardiovascular (CV) systems [3, 5, 6]. The formation of oxygen free radicals and calcium overload in myocytes, a deficiency of antioxidant systems such as catalase and superoxide dismutase, and a possible immunological reaction triggered by the drug are the main pathophysiological processes of cardiotoxicity [8]. Acute or subacute heart damage includes changes in the ventricular repolarization phase, the duration of the QT interval, arrhythmias, ischemia, acute heart failure (HF), and myocarditis-pericarditis-like syndrome. As a result of chronic (early/late) conditions, patients may have symptoms such as left ventricular (LV) dysfunction, systolic/diastolic impotence, and cardiac death [9, 10].

Cardiotoxicity is a well-known side effect of many cytotoxic drugs that can result in long-term morbidity.

Advertisement

2. Chemotherapy-induced cardiotoxicity (CIC)

The development of cancer screening methods, early diagnosis, and the widespread use of adjuvant chemotherapy can result in a significantly higher positive response rate in cancer treatment. Cancer drugs destroy cancerous cells in a variety of ways. These actions usually result in cell death (cytotoxicity), but they can also prevent the cell from growing without killing it (cytostatic action) [11].

Chemotherapeutics have a variety of modes of action, including alkylation of DNA, disruption of DNA and RNA synthesis by intercalating between base pairs, inhibition of DNA polymerase, stimulation of apoptosis, inhibition of DNA topoisomerase II, and preventing mitosis via altering tubulin polymerization. With this higher positive response, however, the number of people exposed to chemotherapy’s early and late cardiac side effects emerges [12]. A wide range of adverse effects of chemotherapy and radiation on cardiac structure, hemostasis and thrombosis, cardiac dysfunctions and arrhythmias, and toxic effects on the heart have been well-documented [1, 13].

Side effects are common among cytotoxic drugs, notably chemotherapeutic agents; antitumor antibiotics, monoclonal antibodies, tyrosine kinase inhibitors, platinum-based compounds, microtubule inhibitors, vinca alkaloids, antimetabolites, proteasome inhibitors, topoisomerase inhibitors, alkylating agents, corticosteroids, and other drugs. They have the potential to cause long-term morbidity. They are linked to irreversible dilated cardiomyopathy and dose-dependent cardiotoxicity [14, 15].

Anthracyclines, a class of antibiotics derived from Streptomyces spp, have been used to treat a variety of cancers over the last 50 years, including lymphoma, leukemia, bladder cancer, breast cancer, and other metastatic cancers [16, 17]. Cardiotoxicity is a growing concern in clinical oncology due to the increasing use of anthracyclines, the introduction of new antitumor agents with potentially cardiotoxic properties, and the use of combined treatments that may have adverse effects on the heart [18, 19].

Anthracyclines catalyze intracellular oxygen radicals via enzymatic reactions in mitochondria, as well as non-enzymatic iron-mediated free radical reactions which damage DNA. They induce apoptosis in vascular cells and cardiomyocytes by activating caspases and degrading internucleosomal DNA [20]. When compared to other tissues, cardiomyocytes have a 35–40% larger amount of mitochondria. Cardiomyocytes use 90% of the ATP produced by mitochondria [21]. Due to bio-energetic failure, genotoxic stress, and oxidative stress, adenosine monophosphate-activated protein kinase (AMPK) signaling is suppressed during treatment, resulting in increased energy stress and hypertrophy [21]. Serum troponin levels in anthracycline-treated patients are also observed to be higher, indicating cell death [22, 23].

Daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, and valrubicin are some of the most commonly used anthracyclines [17]. These drugs have similar effects on cardiotoxicity [24, 25, 26]. Following the existence of targeted therapy, doxorubicin is still widely used in cancer treatment today [10, 27]. However, anthracycline’s medical use is limited due to dose-dependent and cumulative cardiotoxicity. The clinical efficacy of this drug is restricted due to its side effects, particularly cardiotoxicity when doses exceed 400–700 mg/m2 for adults and 300 mg/m2 for children [6, 7, 28]. Doxorubicin cardiomyopathy is more likely at 400 mg/m2 (5%), at 550 mg/m2 (26%), and at 700 mg/m2, where the risk is as high as 48% [14].

A primary cause of doxorubicin-induced cardiomyocyte damage is assumed to be ROS (reactive oxygen species) generation and lipid peroxidation by inhibition of mitochondrial membrane potential and mitochondrial permeability transition pore [29]. To interfere with DNA replication, doxorubicin inhibits DNA topoisomerase 2-beta (Top2β) whereas a doxorubicin-Top2β DNA complex prevents the repair of damaged DNA and leads to cell death [27, 30, 31]. Doxorubicin also affects adrenergic function and adenylate cyclase inhibition of sarcoplasmic reticulum Ca2+ release, inhibits Ca2+-ATPase activities causing diastolic dysfunction, reduces expression of cardiac-specific genes and down-regulates expression of a variety of cardiac muscle-specific proteins including mitochondrial proteins, contractile proteins, sarcoplasmic reticulum proteins [29]. Treatment with doxorubicin induces the immune system to generate a variety of inflammatory mediators (IL-1, IL-6, IL-7, TNF receptor 2, vascular endothelial growth factor/VEGF, matrix metalloproteinases/MMP2); natural killer cells stop functioning, cytotoxic T lymphocytes responses are triggered, and macrophage differentiation is inhibited [32, 33]. Doxorubicin cardiomyopathy increases oxidative stress, which is connected to an increase in Toll-like receptors 2, induces nuclear factor kappa B (NF-κB), and finally leads to apoptosis [34]. There is also an increase in the level of tumor necrosis factor (TNF-α) due to the toll-like receptor 4 [35].

The human epidermal growth factor receptor 2 (ERBB2) is a transmembrane tyrosine kinase receptor that plays in a variety of cellular processes, including cell survival in normal healthy tissue [36]. As a humanized monoclonal antibody, trastuzumab targets ERBB2 on the surface of tumor cells that overexpress ERBB2 [37]. Trastuzumab-induced cardiac damage was detected in metastatic breast cancer trials for the first time. It is the most common chemotherapeutic agent related to left ventricular dysfunction [38]. Bevacizumab is a humanized monoclonal antibody that inhibits vascular endothelial growth factor (VEGF) activity in patients with metastatic colorectal cancer, non-small cell lung cancer, and breast cancer. Although, congestive heart failure (CHF) has been reported in a study, its overall incidence and relative risk are still unknown [39].

Tyrosine kinase (TK) inhibitors (TKI) are molecules designed to target TKs that are overexpressed in cancer cells, but they also inhibit normal variants of tyrosine kinases in non-cancerous cells, which can cause severe side effects such as left ventricular failure [40].

Imatinib is an ATP-competitive small-molecule ABL kinase inhibitor that was developed primarily for the treatment of malignancies such as chronic myeloid leukemia (CML) [41, 42]. It has been shown that imatinib leads to significant mitochondrial damage, including loss of membrane potential, the release of cytochrome C, and markedly reduced energy production with significant declines in ATP concentration, in studies on cultured cardiomyocytes [43, 44]. Other tyrosine-kinase inhibitors such as dasatinib, nilotinib, sunitinib, sorafenib, and lapatinib have been related to drug-induced cardiotoxicity, although the true extent of the damage remains unknown. The literature mentions only a few cases of asymptomatic QT prolongation, pericardial effusion, acute coronary syndromes [45].

Anticancer drugs based on platinum bind to DNA, causing it to crosslink. End result: cancer cells die through apoptosis because of the crosslinks, which interfere with DNA repair and synthesis in the cancer cells. Cisplatin, carboplatin, and oxaliplatin, platinum-based compounds with severe nephrotoxic, neurotoxic, and ototoxic properties, are frequently used in the treatment of human neoplasms [46, 47]. Vascular toxicity, hypertension, dyslipidemia, early atherosclerosis, and coronary artery disease are the most serious late effects of cisplatin-based chemotherapy in patients [48].

Taxanes (paclitaxel, docetaxel, cabazitaxel, Nab-paclitaxel) are microtubule inhibitors (MIT) or mitotic inhibitors that play an important role in mitosis and have lower cardiotoxicity than anthracyclines. Vinca alkaloids such as vinblastine, vincristine, liposomal vincristine, and vinorelbine are also mitotic inhibitors that are used to treat a variety of cancers such as breast, lung, myelomas, lymphomas, and leukemia. As a result, several trials have been conducted to evaluate their use in combination with anthracyclines [49, 50, 51, 52].

Antimetabolites such as 5-fluorouracil (5-FU), capecitabine, azacitidine, cytarabine, gemcitabine, methotrexate, hydroxyurea, and pentostatin which are commonly used to treat leukemia, ovarian, breast, gastrointestinal, and other solid tumors, damage proliferating cells during the S phase of mitosis by substituting the normal DNA/RNA building blocks [53]. Endothelial injury followed by thrombosis; energy depletion and myocardial ischemia; coronary arterial spasm following myocardial ischemia; and decreased ability of red blood cells to transfer oxygen leading to myocardial ischemia are all associated with antimetabolite toxicity [54, 55].

Proteasome inhibitors (PI), which primarily function as immunosuppressants and inhibit bone resorption, such as bortezomib, carfilzomib, and ixazomib, are a promising new class of drugs for the treatment of multiple myeloma, and they are also being studied for other types of cancer [56]. As non-proliferative cells with increased proteasome activity, cardiomyocytes are particularly sensitive to proteasome inhibition.

DNA topoisomerases (type I and type II) are the enzymes responsible for DNA unlinking, and play critical roles in a variety of biological processes involving DNA. Several topoisomerase I inhibitors (also known as camptothecins) include irinotecan, topotecan, and camptothecin, while topoisomerase II inhibitors (also known as epipodophyllotoxins) include etoposide, mitoxantrone, and teniposide [57]. Topoisomerase inhibitors cause the release of ROS, lead to DNA breaks and prevent ligase repair. The enzymes catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) are antioxidant enzymes in oxidative stress modulation, crucial for efficient removal of ROS. Cardiomyocytes are particularly vulnerable to damage because they have low levels of antioxidant enzymes required to detoxify ROS [58].

Cells are also prevented from reproducing by alkylating agents such as cisplatin, busulfan, mechlorethamine, temozolomide, dacarbazine, streptozocin, which damage their DNA [59].

Following are a few examples of in vivo models of chemotherapy-induced cardiotoxicity and protective agents based on the literature;

To explore cardiotoxic effects, animal models are being studied, and therapeutic approaches developed. Because considerable pharmacokinetic and pharmacodynamic data from studies examining the anticancer efficacy of drugs are available, rodents are an attractive model for studying cardiotoxicity.

In the initial periods of cardiotoxicity studies, rabbits were thought to be the standard model. In a study, scientists evaluated the protective effect of ICRF-187 (dexrazoxane, a drug used to prevent anthracycline-induced cardiotoxicity) in rabbits against chronic daunorubicin cardiotoxicity. For the experimental model, twenty-four male white rabbits were divided into four groups (group1; 25 mg ICRF-187/kg and 3.2 mg daunorubicin/kg, group2; daunorubicin (3.2mg/kg), group3; ICRF-187 (25 mg/kg), group4; placebo). They treated animals six times at 3-week intervals over an 18-week period. Significantly different cardiotoxic effects were observed in animals treated only with daunorubicin and those treated only with daunorubicin + ICRF-187. Anthracycline cardiotoxicity was significantly reduced by pretreatment with ICRF-187 [60].

In another study, Zhang et al. divided 50 male Sprague-Dawley rats into three groups for a 15-day experiment: control (saline), doxorubicin (3 mg/kg), and doxorubicin+oxymatrine (12.5, 25, and 50 mg/kg) to detect oxymatrine’s protective effects on cardiovascular diseases. Specifically, they found that oxymatrine pretreatment protected against doxorubicin-induced cardiotoxicity in rats’ hearts by inhibiting the apoptotic pathway [61].

Zilinyi et al. investigated metformin’s (anti-diabetic drug) protective role and its effect on autophagy in doxorubicin-induced cardiotoxicity. In the first group of four Sprague-Dawley rats, doxorubicin (3 mg/kg every second day) was administered intraperitoneally, metformin (250 mg/kg/day) was administered via gavage, and the third group received doxorubicin + metformin, while the fourth group was a control group for two weeks. Doxorubicin-treated myocytes were significantly thinner than those in the control group. Myocyte diameters in the doxorubicin + metformin group were nearly identical to those in the control group. According to the histopathological examination of heart tissue samples, metformin normalized autophagy [62].

In a doxorubicin-induced cardiomyopathy model, Erbaş and his colleagues described the therapeutic effects of liraglutide (LIR), oxytocin, and granulocyte colony-stimulating factor. Four groups of 32 rats were given, respectively, group I; placebo 0.9% NaCl saline solution at a dose of 1 ml/kg/day i.p. (doxorubicin + saline), group II; 1.8 mg/kg/day of liraglutide i.p. (doxorubicin + LIR), group III; 160 μg/kg/day oxytocin i.p. (doxorubicin + OX), group IV; 100 μg/kg/day filgrastim i.p. (doxorubicin + G-CSF (Granulocyte colony-stimulating factor)). It was revealed through the study’s findings inflammatory activity and improved tissue integrity were found to be decreased in response to oxytocin treatment. Besides, LIR reduces levels of proinflammatory cytokines, lipid peroxidation products, troponin T, pro-BNP levels, and CASPASE-3 in doxorubicin-treated rats [63].

Arsenic trioxide and imatinib mesilate cardiotoxicity were examined in male Wistar rats. In the experiment, for ten days, arsenic trioxide (5 mg/kg) and imatinib mesilate (30 mg/kg) was given intraperitoneally and orally, respectively. As a result, the cardiac tissue of the combination-treated group showed fibroblastic proliferation, myocardial disorganization, and myocardial necrosis [64].

According to a study by Saleh et al., tadalafil (Tad) might protect against cardiac and vascular damage caused by the chemotherapy drug cisplatin (CDDP). Seventy-two male albino rats were divided into four groups: the control group, the CDDP (4 mg/kg) i.p. group, the Tad (0.4 mg/kg BW Tad i.p. daily) group, and the Tad +CDDP (0.4 mg/kg BW Tad i.p. + 4 mg/kg BW CDDP i.p) group. In the heart homogenate sample from CDDP treated rats, Tad was able to reduce blood pressure, heart rate, and levels of cardiac troponin, malondialdehyde (MDA), while increasing levels of reduced glutathione (GSH) and nitric oxide (NO) [65].

Advertisement

3. Radiation therapy-induced cardiotoxicity

Cardiotoxicity caused by radiation therapy (RT) is not only seen in adults, but also in children. High radiation exposure, being female, higher anthracycline cumulative dose, trisomy 21, and race are all risk factors for cardiotoxicity in children [66]. Since the early 1900s, ionizing radiation therapy has been utilized to treat cancer [67]. Along with medical advancements, the role of imaging modalities in the administration of the treatment process has gradually increased, in addition to surgical and systematic treatment [68, 69]. Considering the significance of these procedures in the treatment process and in enhancing survival, heart problems from radiotherapy remain a risk [69]. Radiation-induced heart disease (RIHD) is an important cause of long-term non-cancer death after thoracic irradiation [70]. Studies have shown that people who live at least 5 years after diagnosis have a high rate of cardiovascular death [71]. Studies have shown that significant cardiovascular events usually occur within 10–15 years [72]. Lung cancer, mediastinal lymphoma, and breast cancer are cancers that are proximal to the heart and have the highest prevalence of RT, which means they have a significant risk of cardiotoxicity [73]. When Saika et al. looked at breast cancer radiotherapy treatment and heart failure risk, they discovered that women who had RT for breast cancer had a 10 times higher risk of heart failure than the control group, regardless of age or cancer type [74]. Cardiotoxicity from RT causes a worsening of cardiac function in a variety of illnesses, including cardiomyopathy, pericardial injury, coronary artery disease, and heart disease [75, 76].

The effects of factors such as the total radiation volume applied to the patient, the patient’s age, the radiation exposure process, and the simultaneous use of cardiotoxic chemotherapeutic drugs such as anthracyclines are seen in radiation-induced heart diseases [1, 4]. Symptoms of these diseases include pericardial and myocardial fibrosis, rhythm disorders, conduction abnormalities, atherosclerosis, and heart valve injuries. Although, cardiac issues do not manifest themselves until later in life in people, evidence of cardiac toxicity can be found after 10–15 years of follow-up [1, 77]. It has been stated in the literature that RT may have detrimental effects on several important tissues in the heart [68]. In studies, it has been explained that the basis of the mechanism of cardiac damage caused by RT is related to microvascular changes and inflammation that causes longer-term fibrotic changes [1, 68, 78].

Following are a few examples of in vivo models of radiation-induced cardiotoxicity and protective agents based on the literature;

In the studies carried out so far, models have been created with different animals to create experimental animal models of cardiotoxicity. Some of these models are Male Albino rats [79, 80], Male Sprague-Dawley rats [63], Albino Wistar rats [81], male C57/BL6 mice [82].

Mezzaroma et al. used three-month-old C57BL/6J male mice in their study, and 12 of them were irradiated with a single 20 Gray (Gy) dose of radiation therapy, while the other six underwent sham-irradiation. They found that when compared to sham non-irradiated mice, radiation therapy-treated mice exhibited a 2-fold higher rate of myocardial interstitial fibrosis after six months [83].

Using Mast cell-deficient rats (Ws/Ws) and mast cell–competent littermate controls (+/+), researchers exposed the rats for six months to 18 Gy localized single-dose irradiation to investigate cardiac function. They found that mast cell–deficient rats had a higher upward/leftward shift in the left ventricular (LV) diastolic pressure-volume relationship, a decrease in vivo LV diastolic area, and a greater rise in the thickness of the LV posterior wall [84].

Dreyfuss et al. in their study aimed to develop a new mouse model to investigate the pathophysiological mechanism of RT-induced cardiotoxicity and to detect clinically targetable biomarkers of cardiac damage. They used 9–11 weeks of female C57BL/6 mice to form the model. Single radiation doses of 20, 40 or 60 Gy were given to the selected mice, with or without the adjacent lung tissue, using conformal radiation therapy to the cardiac apex. When the results were analyzed, perivascular fibrosis was seen 8 and 24 weeks after RT. The developed model can be utilized to incorporate radiomic and biochemical markers of cardiotoxicity to guide early treatment intervention and human translation studies [85].

In another study, Ibrahim et al. aimed to detect cardiac magnetic resonance (CMR) imaging markers of early RT-induced cardiac dysfunction. In the study, the effect of CMR on global and regional cardiac function and myocardial T1/T2 values at 2-time points after RT with the use of CMR in a localized cardiac RT rat model was investigated. Rats that received 24 Gy radiation, whole-heart radiation were compared to sham-treated rats. They concluded that MRI regional myocardial strain is sensitive imaging diagnostic for detecting RT-induced subclinical cardiac damage before global cardiac function was compromised [86].

Advertisement

4. Ischemic cardiomyopathy

Ischemia is another of the most typical cardiotoxic effects [87]. This term comes from the Greek language (isch means restriction and haema means blood) and refers to a situation where there is an imbalance between the demand for blood and the supply in the tissue [88]. Cardiomyocytes, unlike other tissue cells, do not store energy in the form of glycogen, hence the relationship between myocardial oxygen use and the amount of oxygen given to the myocardial cells is extremely delicate. Cardiovascular problems are further increased by myocardial ischemia [89]. Stress, aging, alcohol consumption, and poor nutrition are all risk factors [90]. 5-fluorouracil (5-FU), cisplatin, and capecitabine are the most common chemotherapeutic agents that cause cardiac ischemia as a cardiotoxic side effect [88, 91, 92]. Isoproterenol (ISP) is another drug that is commonly used to treat bradycardia and heart block, but it can cause myocardial ischemia [92]. 5-FU is a common anti-cancer drug that can cause cardiotoxicity and is related to myocardial ischemia. In this case, the potential mechanism that occurs in myocardial ischemia is indirectly induced coronary vasospasm. Coronary vasospasm may be caused by the synthesis of vasoactive compounds, intimal hyperplasia which results in hyperactive coronary arteries, or it can be myocardial cell damage that occurs as an autoimmune reaction in individuals who are susceptible to 5-FU. Eskilsson et al. investigated verapamil to see if it could protect against 5-FU-induced cardiac ischemia, but the results were not significant [88]. Capecitabine, another agent that is used in chemotherapy, is the oral prodrug of 5-FU. In other words, it is the inactive form of 5-FU, which is activated by thymidine phosphorylase in tumor cells. It is used for its advantages compared to 5-FU. However, capecitabine cardiotoxicity is also known to be reported in the literature [92]. In a case of cardiac ischemia associated with capecitabine-induced cardiotoxicity, acute onset of severe anterior chest pain is observed. The development of chest pain and ischemic changes on ECG is no more observed after capecitabine is ceased [93].

Isoproterenol (ISP) is a medication used to treat conditions such as bradycardia, Torsades de pointes (TdP), and heart block. However, ISP also generates free radicals, which cause oxidative stress. Underlying molecular mechanisms in ISP-induced cardiotoxicity include oxidative stress, the renin-angiotensin system (RAS), apoptosis, and DNA damage. All of these causes cell death and, as a result, cardiac injury, including ischemia [90].

Cisplatin is an antineoplastic drug based on platinum. It is used to treat tumors of the lung, ovary, sarcoma, and lymphoma. The most common cardiotoxic complications caused by cisplatin are thromboembolic events, including myocardial ischemia and infarction [94]. Depolarization of the mitochondrial membrane due to structural abnormalities is one of the mechanisms involved in cisplatin-induced myocardial dysfunction. Furthermore, the endoplasmic reticulum stress response is activated, and apoptosis and caspase-3 activity are increased in cardiomyocytes [95].

Arsenic trioxide (As2O3) is an anticancer agent used in patients with acute promyelocytic leukemia. Arsenic is a chemical element that can be consumed or absorbed through the environment, such as through water and air. Arsenic-related cardiopathological consequences include heart failure and arrhythmia. Caspase activation, mitochondrial disruption, and the pk53 and MAPK signaling pathways all contribute to apoptosis in arsenic cardiotoxicity [96].

Following are a few examples of in vivo models of ischemic cardiomyopathy and protective agents based on the literature;

Paclitaxel, a taxoloid drug, is a cardiotoxicity-inducing drug that causes ischemia, with mechanisms including oxidative stress and apoptosis [97]. Studies on mice have shown that l-glutamine protects against the cardiotoxicity of the anticancer drug cantharidin, which is similar to ischemia in its mechanism of action [98]. ISP is used in protective drug research to create acute or progressive cardiotoxicity in animal models. Curcumin, quercetin, coriander, Momordica, and Withania somnifera are plant-based agents that have been shown to reduce myocardial ischemia in ISP models [90]. The extract of the Spondias mombin plant was used as a treatment in an ISP model on rats. The findings strongly imply that the plant could be used as a cardioprotective treatment. Spondias mombin improves the contractility of the ISP model rat hearts, which are unable to pump blood due to ischemia [99]. Another ISP model investigation was conducted by Jain et al., and ferulic acid was found to be a cardioprotective agent for ISP-induced cardiotoxicity [100]. Pituitrin, like ISP, induces myocardial ischemia. Another rat model for cardiotoxicity is being investigated, and a flavonoid named latifolin derived from Lignum dalbergiae odoriferae was shown to protect against acute myocardial ischemia induced by pituitrin and ISP [101]. Zhang et al. investigated latifolin’s cardioprotective effects on doxorubicin-induced cardiotoxicity. They determined that latifolin protects against the cardiotoxic effects of doxorubicin [102].

Advertisement

5. Diabetic cardiomyopathy

Diabetes mellitus (DM) is a heterogeneous metabolic disease characterized by chronic hyperglycemia resulting from defects in insulin action, insulin secretion, or both. Although, some of the patients die from acute metabolic complications such as ketoacidosis, hyperosmolar hyperglycemic state, and hypoglycemia, the main problem is the increased morbidity and mortality resulting from long-term complications of diabetes. Morbidity and mortality are related to decreased life expectancy and decreased quality of life due to diabetes-related complications [103]. The main cause of morbidity and mortality in diabetic patients is cardiovascular complications [104].

40% of DM patients have heart failure and cardiotoxicity. The increase in the incidence of these conditions is because insulin resistance is a risk factor [105]. Insulin desensitization greatly diminishes the important effects of insulin on heart tissue. It is expressed on many cell surfaces, including cardiomyocytes, where insulin receptor, ligand binding, and insulin receptor substrates (IRS) 1 and 2 are taken up. In addition to IRS1 and IRS2, regulation of the PI3K/Akt pathway is also important in the ERK and MAP kinase cascade. They provide control of metabolism and cell survival. One of the Akt isoforms, AKT1 is involved in the survival of cardiomyocytes; AKT2 is required for the modulation of genes involved in cardiac metabolism. AKT2 promotes glucose uptake through mobilization and fusion of GLU4-containing vesicles to the plasma membrane. Short-term activation of AKT shows cardioprotective effects, can increase glycolysis, and decrease oxidative phosphorylation. The long-term activity of AKT1 in the adult heart is associated with a higher risk of cardiac complications and reduced mitochondrial function [106]. Following insulin stimulation, AKT1 phosphorylates and blocks FOXO1 nuclear translocation, inhibiting the expression of proapoptotic proteins belonging to the Bcl-2 family. FOXO1 has emerged as one of the key players in chronic metabolic diseases, promoting hyperglycemia and glucose intolerance [107]. In physiological conditions, pro-survival stimuli were induced by insulin-suppressing FOXO1 activity via the PI3K/AKT1 pathway. Following stress stimuli, FOXO1 translocates in the nucleus and causes negative feedback on the insulin pathway via a JNK-dependent mechanism that greatly reduces IRS-1 activity [108].

The heart of healthy people without DM obtains 60–90% of its energy from free fatty acids (FFA) oxidation and the rest from lactate and glucose [109]. In patients with DM, glucose uptake is greatly reduced, FFA uptake is increased, and the metabolic balance shifts to lipid oxidation. Increased FFA oxidation is complicated by lipotoxicity and high levels of triglyceride synthesis causing myocyte apoptosis. Additionally, in the diabetic heart, increased lipid oxidation increases mitochondrial dissociation and oxidative stress, which can lead to decreased myocardial energy production and myocardial contractile dysfunction [110]. Hyperglycemia is an important component in diabetes-associated cardiotoxicity because glucotoxicity leads to cardiac dysfunction by inducing oxidative stress and producing enhanced glycation end products. In addition, hyperglycemia may activate the renin-angiotensin-aldosterone system (RAAS) and cause an increase in cell necrosis and fibrosis [111]. Another important component is the inflammation that occurs in diabetes. Expression of inflammatory cytokines such as tissue necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) is increased in the myocardium associated with myocardial contractile dysfunction [112]. In a study by Stentz et al., it was reported that acute hyperglycemic crises such as diabetic ketoacidosis (DKA) and hyperosmolar hyperglycemic syndrome (HHS) are associated with the inflammatory state and independently cause changes in proinflammatory cytokines, oxidative stress, and cardiovascular markers [113].

Increased leukocytes in the myocardium also contribute to the relationship between diabetes and cardiotoxicity. Pathological stresses such as hyperglycemia, hyperlipidemia, high RAAS, and advanced glycation end products (AGEs) stimulate the secretion of proinflammatory molecules, adhesion molecules, and danger-associated molecular patterns (DAMPs) from leukocytes. In addition, these triggers induce ROS-mediated endothelial dysfunction, which also causes cardiac remodeling. Secreted proinflammatory cytokines bind to receptors such as TLR4-MyD88 complex, the receptor for AGEs (RAGE), and IL-1R and initiate intracellular signaling pathways. These pathways activate NF-kB, resulting in transcriptional upregulation of inflammatory cytokines and NLRP3 inflammasome. NF-kB activation and increased oxidative stress mature IL-1B and IL-18 with induction of pyroptosis. At the same time, stressed and damaged cardiomyocytes contribute to inflammatory cascades by releasing pro-inflammatory cytokines and DAMPs. The chronic inflammatory cytokine-induced intracellular response causes pathological cardiac remodeling and cardiac dysfunction [114].

In summary, it involves complex and multifactorial mechanisms such as hyperglycemia, hyperinsulinemia, insulin resistance, increased free fatty acids, microvascular damage and inflammatory cytokines, changes cellular metabolic pathways in cardiomyocytes and contributes to cardiotoxicity by impairing heart function.

Following are a few examples of in vivo models of diabetic cardiomyopathy and protective agents based on the literature;

Rodent models of type 1 and type 2 diabetes share many features with human diabetic cardiomyopathy and have greatly advanced our understanding of the underlying pathology of diabetic cardiomyopathy. Each model has certain limitations, and there is no perfect model that fully phenotypes the human condition. Genetic heterogeneity and lifestyle differences among people make it difficult to produce a suitable model. Some studies with these models are given below.

In one study, empagliflozin treatment was applied to investigate the cardiac metabolic profile of Zucker diabetic fatty rats, which is an early-stage DMT2 model. This treatment activated the cardioprotective master regulator of cellular energy homeostasis, AMP-activated protein kinase, and decreased IL-6 and cardiac mRNA levels while increasing autophagy at the cardiac level. In addition, it reduced cardiac levels of the essential glucose mediators 2,3-bisphosphoglycerate and phosphoenolpyruvate, and regulated several amino acids important in the metabolic control of cardiac function, such as glutamic acid. Therefore, it has been proven that empagliflozin has a protective effect on the development of cardiometabolic diseases associated with cardiac bioenergetic dysregulation and cardiac lipidoma dysregulation [115].

In a study at the ERBAS Institute of Experimental Medicine, 12 rats were used to create a diabetic model after receiving an i.p injection of streptozocin. Rats were randomly assigned to one of two groups: the diabetes group, received 1 mL/kg saline, and the second one received 160 g/kg/day i.p oxytocin for 28 days. They found that oxytocin treatment reduced cardiac myocyte thicknesses significantly over a 4-week period. Besides, as plasma TGF-β levels increased in diabetic rats, oxytocin application significantly decreased plasma TGF-β levels [116].

In another study, rats with and without diabetes were used as models. The hearts of those predisposed to diabetes exhibited depressed contractility and ventricular relaxation at high filling pressures, and abnormalities in the contractile performance of these hearts were observed [117].

Diabetic patients suffer from dual stress on the heart: (1) diabetic cardiomyopathy caused by hyperglycemia and (2) cardiotoxicity caused by anti-diabetic drugs. The following drugs are used as a solution to cardiotoxicity [118].

Metformin (Met) is an oral biguanide antihyperglycemic drug commonly used in the treatment of type 2 diabetes. It activates AMPK and induces cardiac autophagy through the AMPK signaling pathway and improves cardiac functions. In other words, metformin activates AMP-activated protein kinases that play an important role in insulin signaling and fat and glucose metabolism [119]. Kobashigawa and colleagues demonstrated that the cardioprotective effect of metformin against DOX-induced toxicity is mediated through the upregulation of AMPK and its downstream target molecules [120]. However, treatment with high doses of metformin induces the same change in the AMPK pathway, but its protective effect is lost. The authors suggested that this may be due to the downregulation of the platelet-derived growth factor receptor. Moreover, silencing of adiponectin receptors suppressed AMPK activation and cell viability in metformin and DOX-treated cells [121]. In another study, metformin was able to activate AMPK, restore autophagy, and improve heart function [122].

Another drug, Pio, is hyperglycemic drug; it is FDA (Food and Drug Administration) approved and does not show liver toxicity, but cardiotoxicity. It stimulates the peroxisome proliferator-activated receptor (PPAR) γ, which controls the storage of fatty acids and glucose metabolism [123].

One study shows that curcumin has the potential to reverse cardiotoxicity caused by the anti-diabetic drugs Pio and Met. It confirms the generation of ROS in cardiomyoblasts upon treatment with anti-diabetic drugs that Pio is more toxic than Met. Curcumin significantly reduced the oxidative stress caused by anti-diabetic drugs and strengthened the built-in oxidative machinery. It also reduces mitochondrial changes and thus reduces apoptotic cell death of cardiomyoblasts in vitro [118].

Advertisement

6. Antipsychotic drug-induced cardiotoxicity

Antipsychotic drugs have been used to treat psychosis caused by a variety of disorders such as bipolar disorder, delirium, paranoia, schizophrenia, substance-induced psychosis, Tourette’s syndrome, dementia, Huntington’s disease, multiple sclerosis, and parkinsonism. They come in a variety of forms and affect dopamine, serotonin, and other receptors as well as physiological systems. This can cause a wide range of negative effects, such as palpitations, akathisia, dystonia, tardive dyskinesia, orthostatic hypotension, tachycardia, arrhythmias, and heart failure [124]. Antipsychotics are grouped into two types: first-generation (typical, conventional or neuroleptics) antipsychotics (FGAs) such as butyrophenones, chlorpromazine, haloperidol, phenothiazines, thioridazine, and thiothixene, and second-generation (atypical) antipsychotics (SGAs) such as aripiprazole, asenapine, brexpiprazole, clozapine, iloperidone, lurasidone, olanzapine, paliperidon, quetiapine, risperidone, ziprasidone. They can have a variety of effects on cardiovascular function, including direct effects such as blocking cardiac muscarinic receptors, blocking 1-adrenoceptors, blocking sodium, potassium, and calcium channels, and blocking calmodulin, causing QT prolongation as well as indirect effects such as blocking 2-adrenoceptors in the central nervous system (CNS). Antipsychotic drug-induced toxic cardiomyopathy has also frequently been linked to myocardial infarction [125, 126].

Clozapine, the only drug approved for resistant schizophrenia, comes with a warning for an increased risk of fatal myocarditis [124, 127].

Cyclic antidepressants, such as tricyclic and tetracyclic forms, were among the first antidepressants to be developed. TCAs (tricyclic antidepressants) inhibit norepinephrine and serotonin reuptake, leading to an overproduction of these neurotransmitters in the presynaptic cleft [128]. They also inhibit postsynaptic histamine, alpha-1 adrenergic, and muscarinic-acetylcholine receptors [129]. These tricyclic antidepressants were approved by the FDA for the treatment of depression and anxiety disorders: amitriptyline, amoxapine, desipramine, doxepin, Imipramine, nortriptyline, protriptyline, and trimipramine [130].

Most mortality from arrhythmias, hypotension, QTc prolongation, myocardial depression, and ventricular fibrillation is caused by cardiac toxicity, which is the most common side effect of TCAs. TCAs lengthen the cardiac action potential by inhibiting cardiac sodium channels and slowing phase 0 depolarization [131].

Amitriptyline (AMT) intoxication is the third most prevalent cause of mortality among prescription medication-related toxicities, after sedative-hypnotic drugs and analgesic drugs [132].

Following are a few examples of in vivo models of antipsychotic drug-induced cardiotoxicity and protective agents based on the literature;

In a study conducted at the ERBAS Institute of Experimental Medicine, they investigated and compared the electrophysiological, immunohistochemical, and biochemical effects of metoprolol, lipid emulsion, and MgSO4 on AMT-induced cardiotoxicity. Thirty male Sprague-Dawley rats were used in the study. Five groups were given the following treatments: saline intraperitoneally (i.p.); AMT 100 mg/kg per os (p.o.) and saline i.p.; AMT 100 mg/kg p.o. and 5 mg/kg metoprolol i.p.; AMT 100 mg/kg p.o. and 20 ml/kg lipid emulsion. As a result of the study, the QT intervals were significantly prolonged in the AMT + saline group than in the other groups. The QT interval was significantly reduced in all the other groups when compared to the AMT + saline group. They reported that AMT has severe cardiotoxic effects and manifests with ECG abnormalities such as prolongation of QTc duration, which is crucial in cardiotoxicity. The study’s findings also demonstrated that MgSO4 was more potent than other treatments in AMT-toxic rats in terms of shortness of QTc prolongation and immunohistochemical/biochemical effects [133].

In their research, Erbas and his colleague examined the impact of metoprolol and diltiazem on ziprasidone-induced QTc prolongation. For the experiment, 24 rats were divided equally into four groups: I, control, II, 3 mg/kg ziprasidone and saline, III, 3 mg/kg ziprasidone and 1 mg/kg metoprolol, IV, and 3 mg/kg ziprasidone and 2 mg/kg diltiazem. As a result, they observed that rats treated solely with antipsychotic drugs developed ECG abnormalities. When the QTc intervals of the four groups were compared, the QTc of the second group (ziprasidone + saline) was significantly prolonged than that of the control group. Moreover, in the study, metoprolol and diltiazem were found to have a beneficial effect on a prolonged QT interval [134].

A study on clozapine-induced cardiotoxicity was conducted using young male Wistar rats that were given clozapine (10, 15, and 25 mg/kg/day, i.p.) for 21 days. Clozapine, particularly at relatively high doses, was found to have a clear cardiotoxic effect after haemodynamic and echocardiographic studies were performed to assess cardiac functions. An increase in the serum activity of CK-MB (creatine kinase-myocardial band) and LDH (lactate dehydrogenase), two markers of cardiotoxicity, supported these findings [135].

Advertisement

7. Transgenic methods in cardiotoxicity research

Cardiotoxicity develops later as a result of stress, chronic diseases, cancer therapies, and immunotherapies in general, but it can occur in vivo and in vitro if the necessary genetic facilities are available. Mimicking proteomic and genetic disorders, in particular, can cause cardiomyopathies, cardiac transmission problems, and a variety of heart diseases. These models are created using a variety of transgenic methods. The method to be used in a study also varies depending on the pathophysiological mechanisms being studied.

The main methods for creating transgenic models are TALEN (transcription activation-like effector nucleases), CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR associated protein 9), sleeping beauty, piggyBac (PB), pronuclear microinjection (PMI), viral transgenesis, RNAi (RNA interference), and hiPSC (human induced pluripotent stem cells). Numerous studies are currently being conducted in which these methods are being used not only to create pathological models, but also to develop new treatment methods.

Following are a few examples of in vivo models of transgenic methods based on the literature;

Genetically modified cell lines are frequently used due to their ease of maintenance and manipulation. Unfortunately, cell culture results do not always accurately reflect human physiology.

hPSCs (human pluripotent stem cells) can be genetically reprogrammed and converted into iPSCs (induced pluripotent stem cells), which can then be used for functional analysis in a variety of studies. Isogenic hPSC lines derived from ZFNs (zinc-finger nucleases), TALENs, or CRISPR/Cas9 add to our understanding of a variety of cardiovascular diseases, particularly cardiomyopathies and electrophysiological disorders [136].

Disease models can be created in a matter of weeks using current gene-editing methods, including knock-in, knock-out or mutation of certain genes in experimental animals, such as rats. CRISPR/Cas9 systems have recently been used to edit DNA more efficiently based on direct injection of genome editing machines into single-celled mouse embryos. Various cardiovascular diseases have been studied in mammalian models, including but not limited to rats, rabbits, and pigs, to date. Even in zebrafish models alone, various cardiac development, cardiac regeneration, vascular development, and hereditary cardiomyopathy were studied. In addition to single-cell embryo studies, it has been demonstrated that somatic in vivo genome editing studies in adult animals using CRISPR/Cas9 delivery via viral vectors and lipid nanoparticles are possible. Adenovirus and adeno-associated viruses (AAVs) are viral vectors that can be used to efficiently present genetic material in adult animals [136].

Several transgenic models of inherited arrhythmias have been described. Long-QT syndrome (LQTS-1/2/3/8/15), Atrial fibrillation (AF), Brugada Syndrome, and Catecholaminergic polymorphic ventricular tachycardia (CPVT) are just a few of the inherited arrhythmias that have been created using the hPSC [137].

LQTS: Ion channel genes KCNQ1 and KCNH2 with dominant-negative mutations that cause LQTS Type 1 and 2, respectively, were successfully integrated into the AAVS1 locus, which is considered a safe haven using ZFN technology and created an LQTS model in a study on iPSC-CMs (iPSC-Cardiomyocytes). The potential duration of action in regulated iPSC-CMs was significantly longer than in control cells that were not regulated as characteristic phenotypes of the long-QT syndrome, according to patch-clamp results [138].

Dermal fibroblasts from two people in a family with LQTS-1 and two healthy people were infected with retroviral vectors encoding the human transcription factors OCT3/4, SOX2, KLF4, and c-MYC and converted into hPSCs in another study [139]. Another research examined the therapeutic potential of new IKs activators in LQTS using dermal fibroblasts differentiated into hPSCs [140].

To create the LQT15 model, an electrophysiological study mimicked mutations in CALM2 from the CALM1, CALM2, and CALM3 genes that encode Calmodulin, a Ca2+ sensor on hPSC [141].

Transgenic models are also frequently used in studies on other types of Long QT Syndrome [142, 143, 144, 145, 146, 147].

Brugada Syndrome: a large class of arrhythmias caused by a mutation in SCN5A (sodium voltage-gated channel alpha subunit 5), a cardiac sodium channel gene. Brugada Syndrome, Bradycardia, Atrioventricular (AV) Blocks, and Ventricular Fibrillation are few examples (VF).

A scn5a+/− (heterozygous knock-out) transgenic rat model was created for therapeutic evaluation, and the Brugada Syndrome phenotype was mimicked in a study [148]. The models created in a study targeting the SCN5A gene demonstrate cardiac conduction slowdowns and ventricular tachycardia (VT) [149].

In an in vitro study, dermal fibroblasts from two patients with Brugada Syndrome and two healthy individuals were differentiated to iPSC-CMs using Sendai virus (SeV) [150].

CPVT: The mutation in the RYR2 gene, which encodes the cardiac ryanodine receptor and is associated with CPVT cases, is modeled on hiPSCs differentiated from dermal fibroblasts via retrovirus [151].

The main cardiomyopathies investigated by developing transgenic models are dilate cardiomyopathy (DCM), hypertrophic cardiomyopathy (HCM), and restrictive cardiomyopathy (RCM).

Dilate cardiomyopathy: A study focused on striated muscle alpha-tropomyosin (α-TM), an essential thin filament protein involved in the pathogenesis of both dilate and hypertrophic cardiomyopathy. The Glu54Lys mutation, one of two prominent mutations in this protein (Glu40Lys and Glu54Lys), was expressed in transgenic rats. In echocardiography examinations, the successful dilate cardiomyopathy phenotype was observed [152].

In an iPSC-based study, an iPSC-CMs model was created from dermal fibroblasts from patients in a DCM family with a spot mutation (R173W) on the gene encoding cardiac troponin T (cTnT), a sarcomric protein [153].

In a study targeting Leiomodin proteins, which play a vital role in muscle thin filament length, the Lmod2 mutation was modeled in rats using the piggyBac transposon. It has been demonstrated that these rats with ventricular arrhythmias and increased postnatal mortality have a typical DCM phenotype [154].

In a recent study, a knock-in mouse model was created in which endogenous genes were altered to include the deletion of three base pairs encoded in cardiac troponin T for K210 in dilate cardiomyopathy patients [155].

In a study focusing on the titin protein, preserved blood samples from three DCM patients with the dominant TTN mutation were reprogrammed, and high-quality iPSC clones were expanded, differentiated, and enriched by metabolic selection to create a culture with >% iPSC-CMs. To analyze the effects of titin mutation on sarcoma structure, the iPSC-CMs method was created [156].

Hypertrophic cardiomyopathy: A transgenic hypertrophic cardiomyopathy model based on the erasure of 468–527 amino acids, which is bridged by the addition of a point mutation (G1445A) and 9 nonmyosin amino acids (SerSerLeuProHisLeuLysLeu) resulting in Arg403Gln, was created in a study targeting two mutations in the myosin heavy chain gene. Transgenic sequences were shown to be extracted from prokaryotic vector sequences, purified on agarose gels, and injected into the pronuclei of fertilized rat eggs [157].

In a comparative study, two different mutations (R92Q and E163R) in the TNNT2 gene, which encodes cardiac troponin T, were used. Echocardiography showed left ventricular hypertrophy, increased contractility, and diastolic dysfunction in both models. These phenotypes, however, were found to be more pronounced in R92Q mice [158]. HCM rat models with these two mutations had previously been described [159, 160].

In a study on the actin protein, the molecular mechanisms of apical hypertrophic changes were tried to be clarified in rat models created by a mutation (E99K) in the cardiac actin gene (ACTC). The phenotypic investigation of the created models was carried out using echocardiography, electrocardiography, magnetic resonance imaging, and a conductance catheter [161].

In a study examining the central role of calcium-related disorders in the disease pathogenesis in HCM, the iPSC-CMs model was created using fibroblasts derived from HCM patients with the Arg663His mutation in the MYH7 gene, which encodes the heavy chain of myosin [162].

A recent study used transgenic mouse models with the cardiac troponin-I mutation (cTnIGly146) to try to demonstrate that the exosomally derived Y-RNA fragment could regress the HCM clinic [163].

Restrictive cardiomyopathy: Mogensen and colleagues first described six different TnI C-terminal mutations linked to restrictive cardiomyopathy (L144Q, R145W, A171T, K178E, D190G, and R192H) in 2003 [164, 165].

In a study in RCM examining troponin mutations, transgenic rat models were explained by focusing on cTnI193His (R193H) and R145W mutations [165].

Numerous transgenic methods from the past to the present have developed in vivo and in vitro models that are commonly utilized to describe the pathophysiology of the disease. Aside from these studies, which look at the phenotypic manifestations of disease molecular mechanisms, the fact that transgenic approaches give hope for the treatment of numerous diseases has sparked a lot of research. Transgenic methods have therapeutic potential, particularly for cardiotoxic conditions induced by proteomic and genetic disorders that cannot be treated with drugs.

Besides these, R14del mutation in the phospholamban gene (PLN) is another important cause of cardiomyopathy development. Correcting this mutation on induced cardiomyocytes (iCMs) using the TALEN vector method resulted in improved Ca2+ handling, hypertrophic phenotype regression, and homogeneous reticular distribution of phospholamban [166]. In another study using 3D human engineering heart tissue technology, the PLN R14del mutation disrupts cardiac contractility; however, the contractile function is restored in this model with TALEN-mediated genetic correction [167].

A study of the human embryo revealed that the heterozygous MYBPC3 mutation associated with HCM is corrected by an endogenous, germline-specific DNA repair response using the homology-directed repair (HDR) with CRISPR/Cas9, an up-to-date genome editing method [168].

Long QT syndrome is caused by three major mutations: KCNQ1 (LQT1), KCNH2 (LQT2), and SCN5A (LQT3) [169]. These mutations disrupt potassium flow or prolong inward storage flows, prolonging the potential duration of cardiac action and reducing repolarization reserve. According to a study that modeled the E637K mutation in KCNH2, potassium currents were also corrected to relatively close levels after transfection with an optimized siRNA targeted against the mutant potassium channel [170].

A study of CASQ2 knockout rat models reported that exogenous CASQ2 expression, provided via intraperitoneal adeno-associated virus serotype 9 (AAV9) vector, improves CPVT phenotype by correcting arrhythmogenic phenotype and ultrastructural abnormalities [171].

In addition to these models, studies on the therapeutic potentials of transgenic studies for heart failure (HF) have been conducted. An earlier study revealed that overexpression of SERCA2a in cells isolated from HF patients improved myocyte contractile function [172]. Furthermore, genetic therapies for Duchenne muscular dystrophy (DMD), which causes HF with cardiomyopathy, were emphasized. Previous research tried to improve dystrophin mutations using transgenic methods such as ZFN, TALEN, and meganucleases [173, 174, 175, 176, 177]. Even more recently, the CRISPR/Cas9 method was used with AAV to treat mice with dystrophin deficiency induced by a spontaneous mutation in the dystrophin gene [178]. The aim was to remove exon 23 from the dystrophin gene, provide partial functional dystrophin expression in skeletal myofibers and heart muscle, and increase muscle strength [179, 180, 181].

Advertisement

8. Conclusion

Cardiomyopathy is a serious disease in which the heart muscle becomes inflamed and does not work as well as it should. Although, that drug administration and radiation therapy for the disease have a higher positive response, the number of patients experiencing early and late cardiac side effects is growing. There are dozens of diseases related to cardiotoxicity described in the literature, each with dozens of distinct proteomic/genetic mechanisms. The difficulty in modeling and treating diseases stems from the fact that disease mechanisms emerge from a variety of causes that interact with one another in a complex structure. New therapeutic agents, advanced genetic editing technologies and the effective revelation of molecular mechanisms every day, however, are a beacon of hope for humanity to overcome these diseases.

References

  1. 1. Tapio S. Pathology and biology of radiation-induced cardiac disease. Journal of Radiation Research. 2016;57(5):439-448. DOI: 10.1093/jrr/rrw064. Epub 2016 Jul 15
  2. 2. Georgiadis N, Tsarouhas K, Rezaee R, Nepka H, Kass GEN, Dorne JCM, et al. What is considered cardiotoxicity of anthracyclines in animal studies. Oncology Reports. 2020;44(3):798-818. DOI: 10.3892/or.2020.7688. Epub 2020 Jul 14. Erratum in: Oncol Rep. 2020;44(4):1772
  3. 3. Bidulescu DT, Alexandrescu SR. Cardiotoxicology. In: Nriagu JO, editor. Encyclopedia of Environmental Health: Elsevier; 2011. pp. 524-531. ISBN: 9780444522726
  4. 4. Boerma M, Hauer-Jensen M. Preclinical research into basic mechanisms of radiation-induced heart disease. Cardiology Research and Practice. 2010;2011:858262
  5. 5. Hoffman JW Jr, Gilbert TB, Poston RS, Silldorff EP. Myocardial reperfusion injury: etiology, mechanisms, and therapies. Journal of Extra Corporeal Technology. 2004 Dec;36(4):391-411
  6. 6. Li DL, Hill JA. Cardiomyocyte autophagy and cancer chemotherapy. Journal of Molecular and Cellular Cardiology. 2014;71:54-61. DOI: 10.1016/j.yjmcc.2013.11.007. Epub 2013 Nov 14
  7. 7. Sheppard RJ, Berger J, Sebag IA. Cardiotoxicity of cancer therapeutics: Current issues in screening, prevention, and therapy. Frontiers in Pharmacology. 2013;4:19. DOI: 10.3389/fphar.2013.00019
  8. 8. Kaneko M, Matsumoto Y, Hayashi H, Kobayashi A, Yamazaki N. Oxygen free radicals and calcium homeostasis in the heart. Mol Cell Biochem. Jun 1994 15;135(1):99-108. doi: 10.1007/BF00925965. Molecular and Cellular Biochemistry. 1994;139(1):91-100
  9. 9. Antoniou CK, Dilaveris P, Manolakou P, Galanakos S, Magkas N, Gatzoulis K, et al. QT prolongation and malignant arrhythmia: How serious a problem? European Cardiology. 2017;12(2):112-120. DOI: 10.15420/ecr.2017:16:1
  10. 10. Force T, Kolaja KL. Cardiotoxicity of kinase inhibitors: The prediction and translation of preclinical models to clinical outcomes. Nature Reviews. Drug Discovery. 2011;10(2):111-126. DOI: 10.1038/nrd3252
  11. 11. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nature Reviews. Immunology. 2020;20(11):651-668. DOI: 10.1038/s41577-020-0306-5. Epub 2020 May 20
  12. 12. Lenneman CG, Sawyer DB. Cardio-oncology: An update on cardiotoxicity of cancer-related treatment. Circulation Research. 2016;118(6):1008-1020. DOI: 10.1161/CIRCRESAHA.115.303633
  13. 13. Cardinale D, Sandri MT. Role of biomarkers in chemotherapy-induced cardiotoxicity. Progress in Cardiovascular Diseases. 2010;53(2):121-129. DOI: 10.1016/j.pcad.2010.04.002
  14. 14. Swain SM, Whaley FS, Ewer MS. Congestive heart failure in patients treated with doxorubicin: A retrospective analysis of three trials. Cancer. 2003;97(11):2869-2879. DOI: 10.1002/cncr.11407
  15. 15. Yeh ET, Tong AT, Lenihan DJ, Yusuf SW, Swafford J, Champion C, et al. Cardiovascular complications of cancer therapy: Diagnosis, pathogenesis, and management. Circulation. 2004;109(25):3122-3131. DOI: 10.1161/01.CIR.0000133187.74800.B9
  16. 16. Geisberg CA, Sawyer DB. Mechanisms of anthracycline cardiotoxicity and strategies to decrease cardiac damage. Current Hypertension Reports. 2010;12(6):404-410. DOI: 10.1007/s11906-010-0146-y
  17. 17. Venkatesh P, Kasi A. Anthracyclines. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing. p. 2021. Available from: https://www.ncbi.nlm.nih.gov/books/NBK538187/
  18. 18. Barry E, Alvarez JA, Scully RE, Miller TL, Lipshultz SE. Anthracycline-induced cardiotoxicity: Course, pathophysiology, prevention and management. Expert Opinion on Pharmacotherapy. 2007;8(8):1039-1058. DOI: 10.1517/14656566.8.8.1039
  19. 19. Jones RL, Ewer MS. Cardiac and cardiovascular toxicity of nonanthracycline anticancer drugs. Expert Review of Anticancer Therapy. 2006;6(9):1249-1269. DOI: 10.1586/14737140.6.9.1249
  20. 20. Wang L, Ma W, Markovich R, Chen JW, Wang PH. Regulation of cardiomyocyte apoptotic signaling by insulin-like growth factor I. Circulation Research. 1998;83(5):516-522. DOI: 10.1161/01.res.83.5.516
  21. 21. Goffart S, von Kleist-Retzow JC, Wiesner RJ. Regulation of mitochondrial proliferation in the heart: Power-plant failure contributes to cardiac failure in hypertrophy. Cardiovascular Research. 2004;64(2):198-207. DOI: 10.1016/j.cardiores.2004.06.030
  22. 22. Lipshultz SE, Rifai N, Sallan SE, Lipsitz SR, Dalton V, Sacks DB, et al. Predictive value of cardiac troponin T in pediatric patients at risk for myocardial injury. Circulation. 1997;96(8):2641-2648. DOI: 10.1161/01.cir.96.8.2641
  23. 23. Chen W, Liu I, Tomiyasu H, Lee J, Cheng C, Liao AT, et al. Imatinib enhances the anti-tumour effect of doxorubicin in canine B-cell lymphoma cell line. Veterinary Journal. 2019;254:105398
  24. 24. Piska K, Koczurkiewicz P, Bucki A, Wójcik-Pszczoła K, Kołaczkowski M, Pękala E. Metabolic carbonyl reduction of anthracyclines—Role in cardiotoxicity and cancer resistance. Reducing enzymes as putative targets for novel cardioprotective and chemosensitizing agents. Investigational New Drugs. 2017;35(3):375-385. DOI: 10.1007/s10637-017-0443-2. Epub 2017 Mar 10
  25. 25. Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. Anthracyclines: Molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacological Reviews. 2004;56(2):185-229. DOI: 10.1124/pr.56.2.6
  26. 26. Simůnek T, Stérba M, Popelová O, Adamcová M, Hrdina R, Gersl V. Anthracycline-induced cardiotoxicity: Overview of studies examining the roles of oxidative stress and free cellular iron. Pharmacological Reports. 2009;61(1):154-171. DOI: 10.1016/s1734-1140(09)70018-0
  27. 27. Zhang S, Liu X, Bawa-Khalfe T, Lu LS, Lyu YL, Liu LF, et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nature Medicine. 2012;18(11):1639-1642. DOI: 10.1038/nm.2919. Epub 2012 Oct 28
  28. 28. Renu K, Abilash VG, Tirupathi Pichiah PB, Arunachalam S. Molecular mechanism of doxorubicin-induced cardiomyopathy —An update. European Journal of Pharmacology. 2018;818:241-253. DOI: 10.1016/j.ejphar.2017.10.043. Epub 2017 Oct 23
  29. 29. Takemura G, Fujiwara H. Doxorubicin-induced cardiomyopathy from the cardiotoxic mechanisms to management. Progress in Cardiovascular Diseases. 2007;49(5):330-352. DOI: 10.1016/j.pcad.2006.10.002
  30. 30. Capranico G, Tinelli S, Zunino F. Formation, resealing and persistence of DNA breaks produced by 4-demethoxydaunorubicin in P388 leukemia cells. Chemico-Biological Interactions. 1989;72(1-2):113-123. DOI: 10.1016/0009-2797(89)90022-7
  31. 31. Lyu YL, Kerrigan JE, Lin CP, Azarova AM, Tsai YC, Ban Y, et al. Topoisomerase IIbeta mediated DNA double-strand breaks: Implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. Cancer Research. 2007;67(18):8839-8846. DOI: 10.1158/0008-5472.CAN-07-1649
  32. 32. Wu T, Qiao Q, Qin X, Zhang D, Zhang Z. Immunostimulatory cytokine and doxorubicin co-loaded nanovesicles for cancer immunochemotherapy. Nanomedicine. 2019;18:66-77. DOI: 10.1016/j.nano.2019.02.008. Epub 2019 Mar 2
  33. 33. Holopainen T, Räsänen M, Anisimov A, Tuomainen T, Zheng W, Tvorogov D, et al. Endothelial Bmx tyrosine kinase activity is essential for myocardial hypertrophy and remodeling. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(42):13063-13068. DOI: 10.1073/pnas.1517810112. Epub 2015 Oct 1
  34. 34. Nijholt KT, Westenbrink BD, de Boer RA. Mitochondrial therapy for doxorubicin cardiomyopathy: Nuclear factor-κB to the rescue? Cardiovascular Research. 2020;116(6):1092-1094. DOI: 10.1093/cvr/cvz344
  35. 35. Tacar O, Sriamornsak P, Dass CR. Doxorubicin: An update on anticancer molecular action, toxicity and novel drug delivery systems. The Journal of Pharmacy and Pharmacology. 2013;65(2):157-170. DOI: 10.1111/j.2042-7158.2012.01567.x. Epub 2012 Aug 2
  36. 36. Carver JR. Management of trastuzumab-related cardiac dysfunction. Progress in Cardiovascular Diseases. 2010;53(2):130-139. DOI: 10.1016/j.pcad.2010.07.001
  37. 37. Dean L, Kane M. Trastuzumab therapy and ERBB2 genotype. In: Pratt VM, Scott SA, Pirmohamed M, Esquivel B, Kane MS, Kattman BL, Malheiro AJ, editors. Medical Genetics Summaries [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2012
  38. 38. Witteles RM, Fowler MB, Telli ML. Chemotherapy-associated cardiotoxicity: How often does it really occur and how can it be prevented? Heart Failure Clinics. 2011;7(3):333-344. DOI: 10.1016/j.hfc.2011.03.005. Epub 2011 May 14
  39. 39. Choueiri TK, Mayer EL, Je Y, Rosenberg JE, Nguyen PL, Azzi GR, et al. Congestive heart failure risk in patients with breast cancer treated with bevacizumab. Journal of Clinical Oncology. 2011;29(6):632-638. DOI: 10.1200/JCO.2010.31.9129. Epub 2011 Jan 4
  40. 40. Chu TF, Rupnick MA, Kerkela R, Dallabrida SM, Zurakowski D, Nguyen L, et al. Cardiotoxicity associated with tyrosine kinase inhibitor sunitinib. Lancet. 2007;370(9604):2011-2019. DOI: 10.1016/S0140-6736(07)61865-0
  41. 41. Tiribelli M, Medeot M. Cardiotoxicity of imatinib: At the heart of the problem. Leukemia Research. 2011;35(1):36-37. DOI: 10.1016/j.leukres.2010.09.021
  42. 42. Garcia-Alvarez A, Garcia-Albeniz X, Esteve J, Rovira M, Bosch X. Cardiotoxicity of tyrosine-kinase-targeting drugs. Cardiovascular & Hematological Agents in Medicinal Chemistry. 2010;8(1):11-21. DOI: 10.2174/187152510790796192
  43. 43. Dyck JR, Lopaschuk GD. AMPK alterations in cardiac physiology and pathology: Enemy or ally? The Journal of Physiology. 2006;574(Pt 1):95-112. DOI: 10.1113/jphysiol.2006.109389. Epub 2006 May 11
  44. 44. Force T, Krause DS, Van Etten RA. Molecular mechanisms of cardiotoxicity of tyrosine kinase inhibition. Nature Reviews. Cancer. 2007;7(5):332-344. DOI: 10.1038/nrc2106
  45. 45. Talpaz M, Shah NP, Kantarjian H, Donato N, Nicoll J, Paquette R, et al. Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias. The New England Journal of Medicine. 2006;354(24):2531-2541. DOI: 10.1056/NEJMoa055229
  46. 46. Dilruba S, Kalayda GV. Platinum-based drugs: Past, present and future. Cancer Chemotherapy and Pharmacology. 2016;77(6):1103-1124. DOI: 10.1007/s00280-016-2976-z. Epub 2016 Feb 17
  47. 47. Bjerring AW, Fosså SD, Haugnes HS, Nome R, Stokke TM, Haugaa KH, et al. The cardiac impact of cisplatin-based chemotherapy in survivors of testicular cancer: A 30-year follow-up. European Heart Journal Cardiovascular Imaging. 2021;22(4):443-450. DOI: 10.1093/ehjci/jeaa289
  48. 48. Demkow U, Stelmaszczyk-Emmel A. Cardiotoxicity of cisplatin-based chemotherapy in advanced non-small cell lung cancer patients. Respiratory Physiology & Neurobiology. 2013;187(1):64-67. DOI: 10.1016/j.resp.2013.03.013. Epub 2013 Mar 30
  49. 49. Pai VB, Nahata MC. Cardiotoxicity of chemotherapeutic agents: Incidence, treatment and prevention. Drug Safety. 2000;22(4):263-302. DOI: 10.2165/00002018-200022040-00002
  50. 50. Sledge GW, Neuberg D, Bernardo P, Ingle JN, Martino S, Rowinsky EK, et al. Phase III trial of doxorubicin, paclitaxel, and the combination of doxorubicin and paclitaxel as front-line chemotherapy for metastatic breast cancer: An intergroup trial (E1193). Journal of Clinical Oncology. 2003;21(4):588-592. DOI: 10.1200/JCO.2003.08.013
  51. 51. Chatterjee K, Zhang J, Tao R, Honbo N, Karliner JS. Vincristine attenuates doxorubicin cardiotoxicity. Biochemical and Biophysical Research Communications. 2008;373(4):555-560. DOI: 10.1016/j.bbrc.2008.06.067. Epub 2008 Jun 30
  52. 52. Gros R, Hugon V, Thouret JM, Peigne V. Coronary spasm after an injection of vincristine. Chemotherapy. 2017;62(3):169-171. DOI: 10.1159/000455224. Epub 2017 Feb 1
  53. 53. Alter P, Herzum M, Soufi M, Schaefer JR, Maisch B. Cardiotoxicity of 5-fluorouracil. Cardiovascular & Hematological Agents in Medicinal Chemistry. 2006;4(1):1-5. DOI: 10.2174/187152506775268785
  54. 54. Polk A, Vistisen K, Vaage-Nilsen M, Nielsen DL. A systematic review of the pathophysiology of 5-fluorouracil-induced cardiotoxicity. BMC Pharmacology and Toxicology. 2014;15:47. DOI: 10.1186/2050-6511-15-47
  55. 55. Meyer CC, Calis KA, Burke LB, Walawander CA, Grasela TH. Symptomatic cardiotoxicity associated with 5-fluorouracil. Pharmacotherapy. 1997;17(4):729-736
  56. 56. Fricker LD. Proteasome inhibitor drugs. Annual Review of Pharmacology and Toxicology. 2020;60:457-476. DOI: 10.1146/annurev-pharmtox-010919-023603. Epub 2019 Sep 3
  57. 57. Delgado JL, Hsieh CM, Chan NL, Hiasa H. Topoisomerases as anticancer targets. Biochemical Journal. 2018;475(2):373-398. DOI: 10.1042/BCJ20160583
  58. 58. Evison BJ, Sleebs BE, Watson KG, Phillips DR, Cutts SM. Mitoxantrone, more than just another topoisomerase II poison. Medicinal Research Reviews. 2016;36(2):248-299. DOI: 10.1002/med.21364. Epub 2015 Aug 19
  59. 59. Jain D, Aronow W. Cardiotoxicity of cancer chemotherapy in clinical practice. Hospital Practice. 2019;47(1):6-15. DOI: 10.1080/21548331.2018.1530831. Epub 2018 ANTIPSYCHOTICMID: 30270693
  60. 60. Herman EH, Ferrans VJ. Pretreatment with ICRF-187 provides long-lasting protection against chronic daunorubicin cardiotoxicity in rabbits. Cancer Chemotherapy and Pharmacology. 1986;16(2):102-106. DOI: 10.1007/BF00256157
  61. 61. Zhang YY, Yi M, Huang YP. Oxymatrine ameliorates doxorubicin-induced cardiotoxicity in rats. Cellular Physiology and Biochemistry. 2017;43(2):626-635. DOI: 10.1159/000480471. Epub 2017 Sep 21
  62. 62. Zilinyi R, Czompa A, Czegledi A, Gajtko A, Pituk D, Lekli I, et al. The cardioprotective effect of metformin in doxorubicin-induced cardiotoxicity: The role of autophagy. Molecules. 2018;23(5):1184. DOI: 10.3390/molecules23051184
  63. 63. Taşkıran E, Erdoğan MA, Yiğittürk G, Erbaş O. Therapeutic effects of liraglutide, oxytocin and granulocyte colony-stimulating factor in doxorubicin-induced cardiomyopathy model: An experimental animal study. Cardiovascular Toxicology. 2019;19(6):510-517. DOI: 10.1007/s12012-019-09524-x
  64. 64. Saad SY, Alkharfy KM, Arafah MM. Cardiotoxic effects of arsenic trioxide/imatinib mesilate combination in rats. The Journal of Pharmacy and Pharmacology. 2006;58(4):567-573. DOI: 10.1211/jpp.58.4.0017
  65. 65. Saleh RM, Awadin WF, El-Shafei RA, Elseady YY, Wehaish FE, Elshal MF. Cardioprotective role of tadalafil against cisplatin-induced cardiovascular damage in rats. European Journal of Pharmacology. 2015;765:574-581. DOI: 10.1016/j.ejphar.2015.09.015. Epub 2015 Sep 14
  66. 66. Lipshultz SE, Sambatakos P, Maguire M, Karnik R, Ross SW, Franco VI, et al. Cardiotoxicity and cardioprotection in childhood cancer. Acta Haematologica. 2014;132(3-4):391-399
  67. 67. Gianfaldoni S, Gianfaldoni R, Wollina U, Lotti J, Tchernev G, Lotti T. An overview on radiotherapy: From its history to its current applications in dermatology. Open Access Macedonian Journal of Medical Sciences. 2017;5(4):521-525
  68. 68. Ell P, Martin JM, Cehic DA, Ngo DTM, Sverdlov AL. Cardiotoxicity of radiation therapy: Mechanisms, management, and mitigation. Current Treatment Options in Oncology. 2021;22(8):70
  69. 69. Wei T, Cheng Y. The cardiac toxicity of radiotherapy—A review of characteristics, mechanisms, diagnosis, and prevention. International Journal of Radiation Biology. 2021;97:1-8
  70. 70. Yi P, Li H, Fang Y, Su J, Xu C, Cao L, et al. Administration of trastuzumab with heart irradiation induced acute cardiotoxicity in mice. American Journal of Cancer Research. 2020;10(2):536-544
  71. 71. Koczwara B, Meng R, Miller MD, Clark RA, Kaambwa B, Marin T, et al. Late mortality in people with cancer: A population-based Australian study. The Medical Journal of Australia. 2021;214(7):318-323. DOI: 10.5694/mja2.50879. Epub 2020 Dec 9
  72. 72. Adams MJ, Hardenbergh PH, Constine LS, Lipshultz SE. Radiation-associated cardiovascular disease. Critical Reviews Oncology Hematologyl. 2003 Jan;45(1):55-75. DOI: 10.1016/s1040-8428(01)00227-x
  73. 73. Ratosa I, Ivanetic PM. Cardiotoxicity of mediastinal radiotherapy. Reports of Practical Oncology and Radiotherapy. 2019;24(6):629-643. DOI: 10.1016/j.rpor.2019.09.002. Epub 2019 Oct 30
  74. 74. Saiki H, Petersen IA, Scott CG, Bailey KR, Dunlay SM, Finley RR, et al. Risk of heart failure with preserved ejection fraction in older women after contemporary radiotherapy for breast cancer. Circulation. 2017;135(15):1388-1396
  75. 75. Frankart AJ, Nagarajan R, Pater L. The impact of proton therapy on cardiotoxicity following radiation treatment. Journal of Thrombosis and Thrombolysis. 2021;51(4):877-883
  76. 76. Hoppe BS, Flampouri S, Su Z, Latif N, Dang NH, Lynch J, et al. Effective dose reduction to cardiac structures using protons compared with 3DCRT and IMRT in mediastinal Hodgkin lymphoma. International Journal of Radiation Oncology, Biology, Physics. 2012;84(2):449-455
  77. 77. Giraud P, Cosset JM. Toxicité cardiaque de la radiothérapie: physiopathologie, données cliniques [Radiation toxicity to the heart: Physiopathology and clinical data]. Bulletin du Cancer. 2004;91(Suppl 3):147-153. French
  78. 78. Yusuf SW, Venkatesulu BP, Mahadevan LS, Krishnan S. Radiation-induced cardiovascular disease: A clinical perspective. Frontiers in Cardiovascular Medicine. 2017;4:66
  79. 79. Elkady AA, Mohamed ET. Possible role of Withania somnifera against gamma radiation induced cardiotoxicity in male albino rats. Pakistan Journal of Zoology. 2016;48(2):539-545
  80. 80. Mohamed ET, Elkady AA, Mohamed S, Said E, Kiki E. Protective effect of Symplocos racemosa Roxb against gamma radiation induced cardiotoxicity in male albino rats. Pakistan Journal of Zoology. 2016;48(6):1831-1837
  81. 81. Mansour HH, Eid M, El-Arnaouty MB. Effect of silver nanoparticles synthesized by gamma radiation on the cytotoxicity of doxorubicin in human cancer cell lines and experimental animals. Human & Experimental Toxicology. 2018;37(1):38-50
  82. 82. EBCTCG (Early Breast Cancer Trialists' Collaborative Group), McGale P, Taylor C, Correa C, Cutter D, Duane F, et al. Effect of radiotherapy after mastectomy and axillary surgery on 10-year recurrence and 20-year breast cancer mortality: Meta-analysis of individual patient data for 8135 women in 22 randomised trials. Lancet. 2014;383(9935):2127-2135. DOI: 10.1016/S0140-6736(14)60488-8. Epub 2014 Mar 19. Erratum in: Lancet. 2014 Nov 22;384(9957):1848
  83. 83. Mezzaroma E, Di X, Graves P, Toldo S, Van Tassell BW, Kannan H, et al. A mouse model of radiation-induced cardiomyopathy. International Journal of Cardiology. 2012;156(2):231-233. DOI: 10.1016/j.ijcard.2012.01.038. Epub 2012 Feb 16
  84. 84. Boerma M, Wang J, Wondergem J, Joseph J, Qiu X, Kennedy RH, et al. Influence of mast cells on structural and functional manifestations of radiation-induced heart disease. Cancer Research. 2005;65(8):3100-3107. DOI: 10.1158/0008-5472.CAN-04-4333
  85. 85. Dreyfuss AD, Goia D, Shoniyozov K, Shewale SV, Velalopoulou A, Mazzoni S, et al. A novel mouse model of radiation-induced cardiac injury reveals biological and radiological biomarkers of cardiac dysfunction with potential clinical relevance. Clinical Cancer Research. 2021;27(8):2266-2276. DOI: 10.1158/1078-0432.CCR-20-3882. Epub 2021 Feb 4
  86. 86. Ibrahim EH, Baruah D, Croisille P, Stojanovska J, Rubenstein JC, Frei A, et al. Cardiac magnetic resonance for early detection of radiation therapy-induced cardiotoxicity in a small animal model. JACC: CardioOncology. 2021;3(1):113-130. DOI: 10.1016/j.jaccao.2020.12.006. Epub 2021 Mar 16
  87. 87. Frank A, Bonney M, Bonney S, Weitzel L, Koeppen M, Eckle T. Myocardial ischemia reperfusion injury: From basic science to clinical bedside. Seminars in Cardiothoracic and Vascular Anesthesia. 2012;16(3):123-132
  88. 88. Eskilsson J, Albertsson M. Failure of preventing 5-fluorouracil cardiotoxicity by prophylactic treatment with verapamil. Acta Oncologica. 1990;29(8):1001-1003
  89. 89. Pagliaro BR, Cannata F, Stefanini GG, Bolognese L. Myocardial ischemia and coronary disease in heart failure. Heart Failure Reviews. 2020;25(1):53-65
  90. 90. Allawadhi P, Khurana A, Sayed N, Kumari P, Godugu C. Isoproterenol-induced cardiac ischemia and fibrosis: Plant-based approaches for intervention. Phytotherapy Research. 2018;32(10):1908-1932
  91. 91. Oun R, Rowan E. Cisplatin induced arrhythmia; electrolyte imbalance or disturbance of the SA node? European Journal of Pharmacology. 2017;811:125-128
  92. 92. Tunio MA, Hashmi A, Shoaib M. Capecitabine induced cardiotoxicity: A case report and review of literature. Pakistan Journal of Pharmaceutical Sciences. 2012;25(1):277-281
  93. 93. McGoldrick TA, Jodrell D, Clive S. Capecitabine (oral 5-fluorouracil pro-drug) treatment for colorectal carcinoma causing ischaemic chest pain. BML Case Reports. 2009;2009:bcr07.2008.0449
  94. 94. Bursac D. Myocardial ischemia during combined chemotherapy with etoposide and cisplatin for non-small lung cancer. Oncology & Cancer Case Reports. 2018;4:141
  95. 95. Varga ZV, Ferdinandy P, Liaudet L, Pacher P. Drug-induced mitochondrial dysfunction and cardiotoxicity. American Journal of Physiology. Heart and Circulatory Physiology. 2015;309(9):H1453-H1467
  96. 96. Alamolhodaei NS, Shirani K, Karimi G. Arsenic cardiotoxicity: An overview. Environmental Toxicology and Pharmacology. 2015;40(3):1005-1014
  97. 97. Lamberti M, Giovane G, Garzillo EM, Avino F, Feola A, Porto S, et al. Animal models in studies of cardiotoxicity side effects from antiblastic drugs in patients and occupational exposed workers. BioMed Research International. 2014;2014:240642
  98. 98. Shao H, Dong L, Feng Y, Wang C, Tong H. The protective effect of L-glutamine against acute cantharidin-induced cardiotoxicity in the mice. BMC Pharmacology and Toxicology. 2020;21(1):71
  99. 99. Akinmoladun AC, Obuotor EM, Barthwal MK, Dikshit M, Farombi EO. Ramipril-like activity of Spondias mombin linn against no-flow ischemia and isoproterenol-induced cardiotoxicity in rat heart. Cardiovascular Toxicology. 2010;10(4):295-305
  100. 100. Jain PG, Mahajan UB, Shinde SD, Surana SJ. Cardioprotective role of FA against isoproterenol induced cardiac toxicity. Molecular Biology Reports. 2018;45(5):1357-1365
  101. 101. Li XL, Chen LY, Guan ZY, Luo Y, Cui YR, Liu RH, et al. Effect of neoflavonoid latifolin isolated from Dalbergia odorifera on acute myocardial ischemia in rats and its mechanism of Nrf2 signaling pathway. Zhongguo Zhong Yao Za Zhi. 2017;42(20):3974-3982. Chinese. DOI: 10.19540/j.cnki.cjcmm.20170907.009
  102. 102. Zhang N, Shou B, Chen L, Lai X, Luo Y, Meng X, et al. Cardioprotective effects of latifolin against doxorubicin-induced cardiotoxicity by macrophage polarization in mice. Journal of Cardiovascular Pharmacology. 2020;75(6):564-572
  103. 103. Tuomi T, Santoro N, Caprio S, Cai M, Weng J, Groop L. The many faces of diabetes: A disease with increasing heterogeneity. Lancet. 2014;383(9922):1084-1094
  104. 104. Seferović PM, Petrie MC, Filippatos GS, Anker SD, Rosano G, Bauersachs J, et al. Type 2 diabetes mellitus and heart failure: A position statement from the Heart Failure Association of the European Society of Cardiology. European Journal of Heart Failure. 2018;20(5):853-872
  105. 105. Tromp J, Voors AA, Sharma A, Ferreira JP, Ouwerkerk W, Hillege HL, et al. Distinct pathological pathways in patients with heart failure and diabetes. JACC: Heart Failure. 2020;8:234-242
  106. 106. DeBosch B, Sambandam N, Weinheimer C, Courtois M, Muslin AJ. Akt2 regulates cardiac metabolism and cardiomyocyte survival. The Journal of Biological Chemistry. 2006;281(43):32841-32851
  107. 107. Riehle C, Abel ED. Insulin signaling and heart failure. Circulation Research. 2016;118(7):1151-1169
  108. 108. Eijkelenboom A, Burgering BM. FOXOs: Signalling integrators for homeostasis maintenance. Nature Reviews. Molecular Cell Biology. 2013;14(2):83-97
  109. 109. Stanley WC, Chandler MP. Energy metabolism in the normal and failing heart: Potential for therapeutic interventions. Heart Failure Reviews. 2002;7:115-130
  110. 110. Aneja A, Tang WH, Bansilal S, Garcia MJ, Farkouh ME. Diabetic cardiomyopathy: Insights into pathogenesis, diagnostic challenges, and therapeutic options. The American Journal of Medicine. 2008;121(9):748-757
  111. 111. Schilling JD, Mann DL. Diabetic cardiomyopathy: Bench to bedside. Heart Failure Clinics. 2012;8(4):619-631
  112. 112. Westermann D, Van Linthout S, Dhayat S, Dhayat N, Schmidt A, Noutsias M, et al. Tumor necrosis factor-alpha antagonism protects from myocardial inflammation and fibrosis in experimental diabetic cardiomyopathy. Basic Research in Cardiology. 2007;102(6):500-507. DOI: 10.1007/s00395-007-0673-0. Epub 2007 Oct 5
  113. 113. Stentz FB, Umpierrez GE, Cuervo R, Kitabchi AE. Proinflammatory cytokines, markers of cardiovascular risks, oxidative stress, and lipid peroxidation in patients with hyperglycemic crises. Diabetes. 2004;53(8):2079-2086
  114. 114. Kaur N, Guan Y, Raja R, Ruiz-Velasco A, Liu W. Mechanisms and therapeutic prospects of diabetic cardiomyopathy through the inflammatory response. Frontiers in Physiology. 2021;21(12):694864
  115. 115. Aragón-Herrera A, Feijóo-Bandín S, Otero Santiago M, Barral L, Campos-Toimil M, Gil-Longo J, et al. Empagliflozin reduces the levels of CD36 and cardiotoxic lipids while improving autophagy in the hearts of Zucker diabetic fatty rats. Biochemical Pharmacology. 2019;170:113677
  116. 116. Çavuşoğlu T, Çiftçi ÖD, Çağıltay E, Meral A, Kızıloğlu İ, Gurgul S, et al. Histological and biochemical examination of the effect of oxytocin on diabetic cardiomyopathy rat model. Dicle Medical Journal. 2017;44(2):135-143
  117. 117. Broderick TL, Hutchison AK. Cardiac dysfunction in the euglycemic diabetic-prone BB Wor rat. Metabolism. 2004;53(11):1391-1394
  118. 118. Jain A, Rani V. Curcumin-mediated effects on anti-diabetic drug-induced cardiotoxicity. 3 Biotech. 2018;8(9):399
  119. 119. Hostalek U, Gwilt M, Hildemann S. Therapeutic use of metformin in prediabetes and diabetes prevention. Drugs. 2015;75(10):1071-1094
  120. 120. Kobashigawa LC, Xu YC, Padbury JF, Tseng YT, Yano N. Metformin protects cardiomyocyte from doxorubicin induced cytotoxicity through an AMP-activated protein kinase dependent signaling pathway: An in vitro study. PLoS ONE. 2014;9(8):e104888
  121. 121. Asensio-López MC, Lax A, Pascual-Figal DA, Valdés M, Sánchez-Más J. Metformin protects against doxorubicin-induced cardiotoxicity: Involvement of the adiponectin cardiac system. Free Radical Biology & Medicine. 2011;51(10):1861-1871
  122. 122. Rena G, Hardie DG, Pearson ER. The mechanisms of action of metformin. Diabetologia. 2017;60(9):1577-1585. DOI: 10.1007/s00125-017-4342-z. Epub 2017 Aug 3
  123. 123. Smith U. Pioglitazone: Mechanism of action. International Journal of Clinical Practice. Supplement. 2001;121:13-18
  124. 124. Khasawneh FT, Shankar GS. Minimizing cardiovascular adverse effects of atypical antipsychotic drugs in patients with schizophrenia. Cardiology Research and Practice. 2014;2014:273060. DOI: 10.1155/2014/273060. Epub 2014 Feb 4
  125. 125. Buckley NA, Sanders P. Cardiovascular adverse effects of antipsychotic drugs. Drug Safety. 2000;23(3):215-228. DOI: 10.2165/00002018-200023030-00004
  126. 126. Harrigan EP, Miceli JJ, Anziano R, Watsky E, Reeves KR, Cutler NR, et al. A randomized evaluation of the effects of six antipsychotic agents on QTc, in the absence and presence of metabolic inhibition. Journal of Clinical Psychopharmacology. 2004;24(1):62-69. DOI: 10.1097/01.jcp.0000104913.75206.62
  127. 127. Kilian JG, Kerr K, Lawrence C, Celermajer DS. Myocarditis and cardiomyopathy associated with clozapine. Lancet. 1999;354(9193):1841-1845. DOI: 10.1016/s0140-6736(99)10385-4
  128. 128. Schneider J, Patterson M, Jimenez XF. Beyond depression: Other uses for tricyclic antidepressants. Cleveland Clinic Journal of Medicine. 2019;86(12):807-814. DOI: 10.3949/ccjm.86a.19005
  129. 129. Obata H. Analgesic mechanisms of antidepressants for neuropathic pain. International Journal of Molecular Sciences. 2017;18(11):2483. DOI: 10.3390/ijms18112483
  130. 130. Moraczewski J, Aedma KK. Tricyclic Antidepressants. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing. p. 2021. Available from: https://www.ncbi.nlm.nih.gov/books/NBK557791/
  131. 131. Foianini A, Joseph Wiegand T, Benowitz N. What is the role of lidocaine or phenytoin in tricyclic antidepressant-induced cardiotoxicity? Clinical Toxicology (Philadelphia, Pa.). 2010;48(4):325-330. DOI: 10.3109/15563650.2010.487050
  132. 132. Bateman DN, Chick J, Good AM, Kelly CA, Masterton G. Are selective serotonin re-uptake inhibitors associated with an increased risk of self-harm by antidepressant overdose? European Jouirnal of Clinical Pharmacology. 2004;60(3):221-224. DOI: 10.1007/s00228-004-0748-x
  133. 133. Bora S, Erdoğan MA, Yiğittürk G, Erbaş O, Parlak İ. The effects of lipid emulsion, magnesium sulphate and metoprolol in amitriptyline-induced cardiovascular toxicity in rats. Cardiovascular Toxicology. 2018;18(6):547-556. DOI: 10.1007/s12012-018-9466-y
  134. 134. Erbas O, Yilmaz M. Metoprolol and diltiazem ameliorate ziprasidone-induced prolonged corrected QT interval in rats. Toxicology and Industrial Health. 2015;31(12):1152-1157. DOI: 10.1177/0748233713487249. Epub 2013 Apr 26
  135. 135. Abdel-Wahab BA, Metwally ME. Clozapine-induced cardiotoxicity in rats: Involvement of tumour necrosis factor alpha, NF-κβ and caspase-3. Toxicology Reports. 2014;1:1213-1223. DOI: 10.1016/j.toxrep.2014.11.012
  136. 136. Chadwick AC, Musunuru K. Genome editing for the study of cardiovascular diseases. Current Cardiology Reports. 2017;19(3):22. DOI: 10.1007/s11886-017-0830-5
  137. 137. Christidi E, Huang HM, Brunham LR. CRISPR/Cas9-mediated genome editing in human stem cell-derived cardiomyocytes: Applications for cardiovascular disease modelling and cardiotoxicity screening. Drug Discovery Today: Technologies. 2018;28:13-21. DOI: 10.1016/j.ddtec.2018.06.002. Epub 2018 Jun 25
  138. 138. Wang Y, Liang P, Lan F, Wu H, Lisowski L, Gu M, et al. Genome editing of isogenic human induced pluripotent stem cells recapitulates long QT phenotype for drug testing. Journal of the American College of Cardiology. 2014;64(5):451-459. DOI: 10.1016/j.jacc.2014.04.057
  139. 139. Moretti A, Bellin M, Welling A, Jung CB, Lam JT, Bott-Flügel L, et al. Patient-specific induced pluripotent stem-cell models for long-QT syndrome. The New England Journal of Medicine. 2010;363(15):1397-1409. DOI: 10.1056/NEJMoa0908679. Epub 2010 Jul 21
  140. 140. Ma D, Wei H, Lu J, Huang D, Liu Z, Loh LJ, et al. Characterization of a novel KCNQ1 mutation for type 1 long QT syndrome and assessment of the therapeutic potential of a novel IKs activator using patient-specific induced pluripotent stem cell-derived cardiomyocytes. Stem Cell Research & Therapy. 2015;6(1):39. DOI: 10.1186/s13287-015-0027-z
  141. 141. Yamamoto Y, Makiyama T, Harita T, Sasaki K, Wuriyanghai Y, Hayano M, et al. Allele-specific ablation rescues electrophysiological abnormalities in a human iPS cell model of long-QT syndrome with a CALM2 mutation. Human Molecular Genetics. 2017;26(9):1670-1677. DOI: 10.1093/hmg/ddx073
  142. 142. Matsa E, Rajamohan D, Dick E, Young L, Mellor I, Staniforth A, et al. Drug evaluation in cardiomyocytes derived from human induced pluripotent stem cells carrying a long QT syndrome type 2 mutation. European Heart Journal. 2011;32(8):952-962. DOI: 10.1093/eurheartj/ehr073. Epub 2011 Mar 2
  143. 143. Bellin M, Casini S, Davis RP, D'Aniello C, Haas J, Ward-van Oostwaard D, et al. Isogenic human pluripotent stem cell pairs reveal the role of a KCNH2 mutation in long-QT syndrome. The EMBO Journal. 2013;32(24):3161-3175. DOI: 10.1038/emboj.2013.240. Epub 2013 Nov 8
  144. 144. Mehta A, Sequiera GL, Ramachandra CJ, Sudibyo Y, Chung Y, Sheng J, et al. Re-trafficking of hERG reverses long QT syndrome 2 phenotype in human iPS-derived cardiomyocytes. Cardiovascular Research. 2014;102(3):497-506. DOI: 10.1093/cvr/cvu060. Epub 2014 Mar 12
  145. 145. Terrenoire C, Wang K, Tung KW, Chung WK, Pass RH, Lu JT, et al. Induced pluripotent stem cells used to reveal drug actions in a long QT syndrome family with complex genetics. The Journal of General Physiology. 2013;141(1):61-72. DOI: 10.1085/jgp.201210899
  146. 146. Spencer CI, Baba S, Nakamura K, Hua EA, Sears MA, Fu CC, et al. Calcium transients closely reflect prolonged action potentials in iPSC models of inherited cardiac arrhythmia. Stem Cell Reports. 2014;3(2):269-281. DOI: 10.1016/j.stemcr.2014.06.003. Epub 2014 Jul 4
  147. 147. Limpitikul WB, Dick IE, Tester DJ, Boczek NJ, Limphong P, Yang W, et al. A precision medicine approach to the rescue of function on malignant calmodulinopathic long-QT syndrome. Circulation Research. 2017;120(1):39-48. DOI: 10.1161/CIRCRESAHA.116.309283. Epub 2016 Oct 20
  148. 148. Stokoe KS, Balasubramaniam R, Goddard CA, Colledge WH, Grace AA, Huang CL. Effects of flecainide and quinidine on arrhythmogenic properties of Scn5a+/− murine hearts modelling the Brugada syndrome. The Journal of Physiology. 2007;581(Pt 1):255-275. DOI: 10.1113/jphysiol.2007.128785. Epub 2007 Feb 15
  149. 149. Papadatos GA, Wallerstein PM, Head CE, Ratcliff R, Brady PA, Benndorf K, et al. Slowed conduction and ventricular tachycardia after targeted disruption of the cardiac sodium channel gene Scn5a. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(9):6210-6215. DOI: 10.1073/pnas.082121299. Epub 2002 Apr 23
  150. 150. Liang P, Sallam K, Wu H, Li Y, Itzhaki I, Garg P, et al. Patient-specific and genome-edited induced pluripotent stem cell-derived cardiomyocytes elucidate single-cell phenotype of Brugada Syndrome. Journal of the American College of Cardiology. 2016;68(19):2086-2096. DOI: 10.1016/j.jacc.2016.07.779
  151. 151. Itzhaki I, Maizels L, Huber I, Gepstein A, Arbel G, Caspi O, et al. Modeling of catecholaminergic polymorphic ventricular tachycardia with patient-specific human-induced pluripotent stem cells. Journal of the American College of Cardiology. 2012;60(11):990-1000. DOI: 10.1016/j.jacc.2012.02.066. Epub 2012 Jun 27
  152. 152. Rajan S, Ahmed RP, Jagatheesan G, Petrashevskaya N, Boivin GP, Urboniene D, et al. Dilated cardiomyopathy mutant tropomyosin mice develop cardiac dysfunction with significantly decreased fractional shortening and myofilament calcium sensitivity. Circulation Research. 2007;101(2):205-214. DOI: 10.1161/CIRCRESAHA.107.148379. Epub 2007 Jun 7. Erratum in: Circ Res. 2007 Sep 14;101(6):e80
  153. 153. Sun N, Yazawa M, Liu J, Han L, Sanchez-Freire V, Abilez OJ, et al. Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy. Science Translational Medicine. 2012;4(130):130ra47. DOI: 10.1126/scitranslmed.3003552
  154. 154. Li S, Mo K, Tian H, Chu C, Sun S, Tian L, et al. Lmod2 piggyBac mutant mice exhibit dilated cardiomyopathy. Cell & Bioscience. 2016;6:38. DOI: 10.1186/s13578-016-0101-y
  155. 155. Du CK, Morimoto S, Nishii K, Minakami R, Ohta M, Tadano N, et al. Knock-in mouse model of dilated cardiomyopathy caused by troponin mutation. Circulation Research. 2007;101(2):185-194. DOI: 10.1161/CIRCRESAHA.106.146670. Epub 2007 Jun 7
  156. 156. Hinson JT, Chopra A, Nafissi N, Polacheck WJ, Benson CC, Swist S, et al. HEART DISEASE. Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy. Science. 2015;349(6251):982-986. DOI: 10.1126/science.aaa5458
  157. 157. Vikstrom KL, Factor SM, Leinwand LA. Mice expressing mutant myosin heavy chains are a model for familial hypertrophic cardiomyopathy. Molecular Medicine. 1996;2(5):556-567
  158. 158. Ferrantini C, Coppini R, Pioner JM, Gentile F, Tosi B, Mazzoni L, et al. Pathogenesis of hypertrophic cardiomyopathy is mutation rather than disease specific: A comparison of the cardiac troponin T E163R and R92Q mouse models. Journal of the American Heart Association. 2017;6(7):e005407. DOI: 10.1161/JAHA.116.005407
  159. 159. Tardiff JC, Hewett TE, Palmer BM, Olsson C, Factor SM, Moore RL, et al. Cardiac troponin T mutations result in allele-specific phenotypes in a mouse model for hypertrophic cardiomyopathy. The Journal of Clinical Investigation. 1999;104(4):469-481. DOI: 10.1172/JCI6067
  160. 160. Moore RK, Abdullah S, Tardiff JC. Allosteric effects of cardiac troponin TNT1 mutations on actomyosin binding: A novel pathogenic mechanism for hypertrophic cardiomyopathy. Archives of Biochemistry and Biophysics. 2014;552-553:21-28. DOI: 10.1016/j.abb.2014.01.016. Epub 2014 Jan 28
  161. 161. Song W, Dyer E, Stuckey DJ, Copeland O, Leung MC, Bayliss C, et al. Molecular mechanism of the E99K mutation in cardiac actin (ACTC Gene) that causes apical hypertrophy in man and mouse. The Journal of Biological Chemistry. 2011;286(31):27582-27593. DOI: 10.1074/jbc.M111.252320. Epub 2011 May 26
  162. 162. Lan F, Lee AS, Liang P, Sanchez-Freire V, Nguyen PK, Wang L, et al. Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell. 2013;12(1):101-113. DOI: 10.1016/j.stem.2012.10.010
  163. 163. Huang F, Na N, Ijichi T, Wu X, Miyamoto K, Ciullo A, et al. Exosomally derived Y RNA fragment alleviates hypertrophic cardiomyopathy in transgenic mice. Molecular Therapy—Nucleic Acids. 2021;24:951-960. DOI: 10.1016/j.omtn.2021.04.014
  164. 164. Mogensen J, Kubo T, Duque M, Uribe W, Shaw A, Murphy R, et al. Idiopathic restrictive cardiomyopathy is part of the clinical expression of cardiac troponin I mutations. Journal of Clinical Investigation. 2003;111(2):209-216. DOI: 10.1172/JCI16336. Erratum in: The Journal of Clinical Investigation 2003 Mar;111(6):925
  165. 165. Liu X, Zhang L, Pacciulli D, Zhao J, Nan C, Shen W, et al. Restrictive cardiomyopathy caused by troponin mutations: Application of disease animal models in translational studies. Frontiers in Physiology. 2016;7:629. DOI: 10.3389/fphys.2016.00629
  166. 166. Karakikes I, Stillitano F, Nonnenmacher M, Tzimas C, Sanoudou D, Termglinchan V, et al. Correction of human phospholamban R14del mutation associated with cardiomyopathy using targeted nucleases and combination therapy. Nature Communications. 2015;6:6955. DOI: 10.1038/ncomms7955
  167. 167. Stillitano F, Turnbull IC, Karakikes I, Nonnenmacher M, Backeris P, Hulot JS, et al. Genomic correction of familial cardiomyopathy in human engineered cardiac tissues. European Heart Journal. 2016;37(43):3282-3284. DOI: 10.1093/eurheartj/ehw307. Epub 2016 Jul 22
  168. 168. Ma H, Marti-Gutierrez N, Park SW, Wu J, Lee Y, Suzuki K, et al. Correction of a pathogenic gene mutation in human embryos. Nature. 2017;548(7668):413-419. DOI: 10.1038/nature23305. Epub 2017 Aug 2
  169. 169. Mizusawa Y, Horie M, Wilde AA. Genetic and clinical advances in congenital long QT syndrome. Circulation Journal. 2014;78(12):2827-2833. DOI: 10.1253/circj.cj-14-0905. Epub 2014 Oct 1
  170. 170. Lu X, Yang X, Huang X, Huang C, Sun HH, Jin L, et al. RNA interference targeting E637K mutation rescues hERG channel currents and restores its kinetic properties. Heart Rhythm. 2013;10(1):128-136. DOI: 10.1016/j.hrthm.2012.09.124. Epub 2012 Sep 26
  171. 171. Denegri M, Avelino-Cruz JE, Boncompagni S, De Simone SA, Auricchio A, Villani L, et al. Viral gene transfer rescues arrhythmogenic phenotype and ultrastructural abnormalities in adult calsequestrin-null mice with inherited arrhythmias. Circulation Research. 2012;110(5):663-668. DOI: 10.1161/CIRCRESAHA.111.263939. Epub 2012 Jan 31
  172. 172. del MonteF HSE, Schmidt U, Matsui T, Kang ZB, Dec GW, Gwathmey JK, et al. Restoration of contractile function in isolated cardiomyocytes from failing human hearts by gene transfer of SERCA2a. Circulation. 1999;100(23):2308-2311. DOI: 10.1161/01.cir.100.23.2308
  173. 173. Chapdelaine P, Pichavant C, Rousseau J, Pâques F, Tremblay JP. Meganucleases can restore the reading frame of a mutated dystrophin. Gene Therapy. 2010;17(7):846-858. DOI: 10.1038/gt.2010.26. Epub 2010 Apr 15
  174. 174. Ousterout DG, Perez-Pinera P, Thakore PI, Kabadi AM, Brown MT, Qin X, et al. Reading frame correction by targeted genome editing restores dystrophin expression in cells from Duchenne muscular dystrophy patients. Molecular Therapy. 2013;21(9):1718-1726. DOI: 10.1038/mt.2013.111, . Epub 2013 Jun 4. Erratum in: Mol Ther. 2013 Nov;21(11):2130
  175. 175. Ousterout DG, Kabadi AM, Thakore PI, Perez-Pinera P, Brown MT, Majoros WH, et al. Correction of dystrophin expression in cells from Duchenne muscular dystrophy patients through genomic excision of exon 51 by zinc finger nucleases. Molecular Therapy. 2015;23(3):523-532. DOI: 10.1038/mt.2014.234. Epub 2014 Dec 10
  176. 176. Popplewell L, Koo T, Leclerc X, Duclert A, Mamchaoui K, Gouble A, et al. Gene correction of a duchenne muscular dystrophy mutation by meganuclease-enhanced exon knock-in. Human Gene Therapy. 2013;24(7):692-701. DOI: 10.1089/hum.2013.081
  177. 177. Cordova G, Negroni E, Cabello-Verrugio C, Mouly V, Trollet C. Combined therapies for duchenne muscular dystrophy to optimize treatment efficacy. Frontiers in Genetics. 2018;9:114. DOI: 10.3389/fgene.2018.00114
  178. 178. Nance ME, Duan D. Perspective on adeno-associated virus capsid modification for duchenne muscular dystrophy gene therapy. Human Gene Therapy. 2015;26(12):786-800. DOI: 10.1089/hum.2015.107. Epub 2015 Oct 15
  179. 179. Nelson CE, Hakim CH, Ousterout DG, Thakore PI, Moreb EA, Castellanos Rivera RM, et al. In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystrophy. Science. 2016;351(6271):403-407. DOI: 10.1126/science.aad5143. Epub 2015 Dec 31
  180. 180. Wang JZ, Wu P, Shi ZM, Xu YL, Liu ZJ. The AAV-mediated and RNA-guided CRISPR/Cas9 system for gene therapy of DMD and BMD. Brain & Development. 2017;39(7):547-556. DOI: 10.1016/j.braindev.2017.03.024. Epub 2017 Apr 5
  181. 181. Zhu P, Wu F, Mosenson J, Zhang H, He TC, Wu WS. CRISPR/Cas9-mediated genome editing corrects dystrophin mutation in skeletal muscle stem cells in a mouse model of muscle dystrophy. Molecular Therapy—Nucleic Acids. 2017;7:31-41. DOI: 10.1016/j.omtn.2017.02.007. Epub 2017 Feb 28

Written By

Oytun Erbaş, İlknur Altuntaş, Özge Çağlar, Elif Özyilmaz, Ece Sari, İlayda Üzümcü and Kaan Erbakan

Submitted: 21 September 2021 Reviewed: 26 October 2021 Published: 07 January 2022