Abstract
Despite extensive progress in the knowledge and understanding of cardiovascular diseases and significant advances in pharmacological treatments and procedural interventions, cardiovascular diseases (CVD) remain the leading cause of death globally. Mitochondrial dynamics refers to the repetitive cycle of fission and fusion of the mitochondrial network. Fission and fusion balance regulate mitochondrial shape and influence physiology, quality and homeostasis. Mitophagy is a process that eliminates aberrant mitochondria. Melatonin (Mel) is a pineal‐synthesized hormone with a range of pharmacological properties. Numerous nonclinical trials have demonstrated that Mel provides cardioprotection against ischemia/reperfusion, cardiomyopathies, atherosclerosis and cardiotoxicity. Recently, interest has grown in how mitochondrial dynamics contribute to melatonin cardioprotective effects. This review assesses the literature on the protective effects of Mel against CVD via the regulation of mitochondrial dynamics and mitophagy in both in‐vivo and in‐vitro studies. The signalling pathways underlying its cardioprotective effects were reviewed. Mel modulated mitochondrial dynamics and mitophagy proteins by upregulation of mitofusin, inhibition of DRP1 and regulation of mitophagy‐related proteins. The evidence supports a significant role of Mel in mitochondrial dynamics and mitophagy quality control in CVD.
Keywords: cardiovascular disease, dynamin‐related protein 1, heart, melatonin, mitochondrial fission, mitochondrial fusion, mitophagy
1. INTRODUCTION
Cardiovascular disease (CVD) affects the heart, capillaries, arteries and veins. 1 , 2 , 3 CVD and its complications, including ischemia–reperfusion (I/R), septic cardiomyopathies and atherosclerosis, are significant causes of death worldwide. Despite recent advances in treatment, CVD remains a leading public health challenge globally. 1 , 2 , 3 The mitochondria, multifunctional double‐membrane organelles, play an essential role in cells by modulating calcium homeostasis, reactive oxygen species (ROS) production, apoptosis, cellular metabolism and adenosine triphosphate (ATP) production. 4 , 5 , 6
The heart is one of the main energy‐demanding and mitochondrial‐enriched organs and, therefore, sensitive to mitochondrial impairment. 7 Mitochondrial dysfunction plays a major role in CVD pathogenesis. 7 The mitochondria are key regulators of heart function and cardiomyocyte viability. 8 Mitochondrial homeostasis was first described in yeast in the 1990s. 9 Structurally, the mitochondria are highly dynamic organelles that undergo fission and fusion. 5 Fusion occurs when two mitochondria join, resulting in a single, larger mitochondrion, while fission is the opposite process. 5 Excessive fission has been linked to mitochondrial impairments, mainly attributed to elevated stress levels leading to cell death. 5 In conditions like I/R injury, 10 chronic heart failure, 11 cardiomyopathies 12 , 13 and doxorubicin cardiotoxicity 14 , 15 mitochondria in the heart undergo fission. Promoting mitochondrial fusion or inhibiting mitochondrial fission improves their function. Damaged mitochondria are removed by autophagy (mitophagy). 9
These phenomena are necessary for proper mitochondria functioning, and their disruption can lead to various diseases. 3 , 5 Cardiovascular function and adaptation to pathological stressors require a healthy mitochondrial network. 5 , 6 Thus, mitochondrial dynamics and mitophagy are two phenomena that influence mitochondrial health, particularly in cardiomyocytes that are highly vulnerable to ATP depletion. 3 Recent cardioprotective studies have focused on mitochondrial dynamics and mitophagy.
Melatonin (Mel), N‐acetyl‐5‐methoxytryptamine (Figure 1), is a chronobiotic agent first identified in the cow pineal gland. Mel primarily regulates the sleep–wake cycle. 16 , 17 , 18 Mel is also an effective anti‐oxidant, anti‐cancer and anti‐aging hormone. 17 , 18 The parent compound and several metabolites can also scavenge free radicals. 1 Although the pineal gland is the principal site of Mel production and secretion, 17 most organs and tissues can synthesize Mel, including the gastrointestinal tract, retina, ovary and heart. 1
FIGURE 1.
Chemical structure of melatonin.
Plasma Mel levels are reduced in patients with coronary heart disease. 1 , 10 Exogenous Mel has been shown to improve systolic and diastolic blood pressure, 19 maximum flow rate in the internal carotid artery 19 , 20 and decrease platelet aggregation. Accumulating evidence suggests a correlation between a reduction in the circulating level of Mel and its metabolite 6‐sulphatoxymelatonin with CVD such as congestive heart failure, nocturnal hypertension, coronary heart disease and myocardial infarction. 1 , 21
Mel functions via receptor‐ and non‐receptor‐mediated mechanisms. 1 Receptor‐mediated effects of Mel are associated with its affinity for binding sites in the plasma membrane, cytoplasm and nucleolus. Mel targets some orphan nuclear receptors. 1 Specific Mel receptors, MT1, MT2 and MT3, are distributed in cell membrane and nucleus thorough the body in places such as the brain, the retina of the eye and the cardiovascular system. 22 MT1 and MT2 are membrane receptors coupled with G protein, while MT3 is a nuclear quinone reductase involved in oxidative stress reduction. 22 Due to their high expression in ventricles, aorta, coronary arteries and endothelial cells, these receptors are potential targets for CVD treatment. 23
Mel accumulates in mitochondria because of its amphiphilic nature and relatively small size. 24 The concentration of Mel is higher in mitochondria than in blood. 25 , 26 Recent studies have suggested that the mitochondria can produce Mel. 26 , 27 The uptake of Mel by the mitochondria is mediated by the oligopeptide transporters GLUT/SLC2A10 and PEPT1/2 (SLC15A1/2), 25 , 28 suggesting that Mel specifically targets mitochondria. 25 Although the molecular mechanisms involved in regulating mitochondrial dynamics and mitophagy are not fully understood, Mel has been reported to regulate mitochondrial dynamics in various cell lines and animal models of CVD. 29 , 30
Given the role of mitochondrial dynamics and mitophagy in CVD, we hypothesize that Mel may alleviate cardiovascular dysfunction by regulating mitochondrial dynamics and mitophagy. Therefore, this study evaluated the publicly available literature on the effects of Mel on mitochondrial dynamics and mitophagy in CVD in both in‐vivo and in‐vitro experimental models.
1.1. The role of mitochondrial fission in cardiovascular disease
In response to cardiovascular injuries, damaged mitochondria initially convert into small fragments through mitochondrial fission. 31 Subsequently, excessive mitochondrial fission enhances ROS production. It promotes Bcl‐2‐associated X protein (Bax)/Bcl‐2 recruitment, oligomerization, pore formation at the outer mitochondrial membrane (OMM) and leakage of cytochrome c (Cyt c) into the cytoplasm. The mitochondrial fission‐dependent apoptosis is then triggered. 31 Several proteins, including dynamin‐related protein (Drp1), fission factor 1 (Fis1), mitochondrial fission factor (MFF) and mitochondrial dynamics protein 49 and 51 (MiD49 and MiD51) regulate mitochondrial fission. 32 In OMM, Fis1, MFF and MiD49/51 act as Drp1 receptors. 33
Drp1, the master fission regulator, is a member of the GTPase superfamily with four main domains. These include C‐terminal, variable, helical middle and N‐terminal GTPase domains. 31 Several studies have shown that Drp1 regulates mitochondrial fission. In addition, various post‐translational modifications control Drp1 activity during mitochondrial fission. These include SUMOylation, S‐nitrosylation, O‐GlcNAcylation, palmitoylation, ubiquitination and phosphorylation. Among them, phosphorylation plays a key role in regulating Drp1 activity. Phosphorylation occurs at different sites of Drp1 such as Ser‐40, Ser‐44, Ser‐579, Ser‐585, Ser‐616, Ser‐637 and Ser‐693. Ser‐616 and Ser‐637 have been extensively reported among these sites. 34 Drp1 is activated by phosphorylation at Ser‐616 but inactivated by phosphorylation at Ser‐637. 35 The balance of these two phosphorylation sites determines Drp1 activity. 35 Activated Drp1 via phosphorylation migrates toward the surface of the mitochondria and at the predicted division sites along mitochondrial tubules. 35 Several pathways have been reported to modify Drp1 activation, including c‐Jun N‐terminal kinases (JNK), extracellular signal‐regulated kinase (ERK) and protein kinase A (PKA). 5 Excessive mitochondrial fission occurs within 60 min of myocardial reperfusion after transient ischemia, leading to mitochondrial dysfunction and decreased cardiac contractility. 36
Inhibition of mitochondrial fission in CVD is a potential cardioprotective strategy. Researchers have investigated several Drp1 inhibitors, such as Mdivi‐1, Dynasore and P110. Mdivi‐1, a Drp1 inhibitor, has been extensively studied. While it is an efficient Drp1 inhibitor, it shows off‐target effects, such as complex I and complex II inhibition. 37 Therefore, a primary challenge is finely modulating Drp1 activity while reducing possible side effects. Finding more selective Drp1 inhibitors is important to avoid potential off‐target side effects. Recent studies have investigated the role of specific inhibitors such as P110, which inhibits Drp1/Fis1 and P259, which inhibits Drp1/MFF. Targeting mitochondrial fission is a double‐edged problem because, physiologically, mitochondrial fission is required for mitochondrial function.
1.2. The role of mitochondrial fusion in cardiovascular disease
Mitochondrial fusion occurs when two mitochondria fuse to form one large mitochondrion. In the fusion, mtDNA is mixed, which may be beneficial. Proteins on the outer and inner membranes of the mitochondria mediate mitochondrial fusion. The key proteins that regulate the fusion of the mitochondrial outer membrane are mitofusins 1/2 (MFN1 and 2). 38 Optic atrophy 1 (OPA1) is the key protein that regulates inner mitochondrial membrane fusion. Both belong to a family of dynamin‐related GTPases. 38
The proteins involved in mitochondrial fusion have multiple cellular functions. For instance, OPA1, which exists in both long and short isoforms, is implicated in preserving crista structure and protecting cells from death stimuli. 38 The optimum function of OPA1 depends on its protein expression level and a coordinated balance between its long and short isoforms. 38 OPA1 knock‐out (KO) mice develop mitochondrial defects in the heart and late‐onset cardiomyopathy. Mutations in OPA1 in Drosophila have been associated with decreased heartbeat and cardiac arrhythmia. 39 Charcot–Marie‐Tooth (CMT) and autosomal dominant optic atrophy (ADOA) are inherited neuropathies that lead to blindness. These diseases are associated with mutations in fusion proteins, MFN2 for CMT and OPA1 for ADOA.
1.3. The role of mitochondrial autophagy (mitophagy) in cardiovascular diseases
Excessive aberrant mitochondria can cause serious consequences, leading to cell death. The degradation of aberrant mitochondria through the autophagy pathway is termed mitophagy. 40 Mitophagy is triggered by signals such as hypoxia, loss of mitochondrial membrane potential (MMP), opening of the mitochondrial permeability transition pore (mPTP) and excessive ROS production. 41 Several pathways have been identified for mitophagy. Mitophagy is categorized into ubiquitin‐mediated and receptor‐mediated mitophagy. Ubiquitin‐mediated mitophagy includes PTEN‐induced kinase 1 (PINK1)/Parkin. The receptor‐mediated mitophagy pathways include Bcl‐2 interacting protein 3 (BNIP3)‐mediated mitophagy, FUN14 domain containing 1 (FUNDC1)−mediated mitophagy and lipid‐mediated mitophagy. 41
PINK1 was identified in 2004 as a pathogenic gene of Parkinson's disease. PINK1 is a primary detector of mitochondrial damage. 42 , 43 It contains three domains: the N‐terminal domain, which is a mitochondrial target signal, the hydrophobic transmembrane domain and the C‐terminal domain. 42 , 43 PINK1 is regularly imported into normal healthy mitochondria through the N‐terminal domain and degraded by mitochondrial proteases and proteasomes. 42 , 43 In pathologic conditions, impairment in mitochondrial membrane potential prevents PINK1 from being imported from the cytosol into the mitochondria. This results in the PINK1 stabilization on the OMM in dimers and auto‐phosphorylation at Ser‐228 and Ser‐402. 44 Activated PINK1 phosphorylates ubiquitin and activates Parkin, leading to mitophagy induction. 45
In non‐conventional receptor‐mediated mitophagy, various OMM receptors have been identified, such as Bcl‐2 interacting protein 3 (BNIP3), FUNDC1 and BNIP3L/Nix, a pro‐apoptotic protein. 41 BNIP3L is a mitophagy receptor that binds to Atg8 proteins. 46 Phosphorylation and ubiquitination are the main post‐translational modifications of BNIP3L, which regulates BNIP3L‐mediated mitophagy. FUNDC1 is a newly discovered mitophagy receptor. 47 FUNDC1 contains a LIR motif, three α‐helical stretches and a C‐terminal in the inner mitochondrial membrane. Dephosphorylation plays a key role in FUNDC1's binding affinity with LC3‐II. FUNDC1 and is deactivated when Src kinase and casein kinase 2 phosphorylate it at the tyrosine‐18 and Ser‐13 residues. 48 Among these two sites, phosphorylation of tyrosine‐18 plays a central role, while phosphorylation of Ser‐13 provides a supplementary role in FUNDC1‐mediated mitophagy. 49
Dysregulation of mitophagy has been implicated in CVD pathogenesis, such as atherosclerosis, 30 I/R, 8 cardiomyopathies 50 and heart failure. 51 The mitochondrial function in sustaining cardiomyocytes depends on mitophagy, which plays a dual role in the progression of heart diseases. Accordingly, it has been indicated that adaptive/mild mitophagy is associated with the alleviation of pathologies while maladaptive/excessive mitophagy could exacerbate pathological conditions. 3 So, it seems that induction of mild mitophagy or decline in excessive mitophagy play a suitable strategy for CVDs. Therefore, cardiomyocyte mitophagy needs to be explored in more detail. It may provide theoretical and practical evidence to reduce heart injuries.
2. REGULATION OF MITOCHONDRIAL DYNAMICS AND MITOPHAGY BY MELATONIN IN HEART DISEASE
2.1. Myocardial ischemia/reperfusion
Ischemic heart disease is a major component of CVD and a leading cause of death. 21 Although progress has been made in understanding the cause of ischemic heart disease, the underlying mechanism(s) that occurs during I/R injury have not been fully elucidated and current treatments are limited in effectiveness. 52 Mitochondria play a central role in I/R pathogenesis. 8 In this regard, the effects of Mel on I/R models in the setting of mitochondrial dynamics have been explored. 8 , 20 , 53 The imbalance between mitochondrial fusion and fission contributes to cardiac dysfunction during I/R. 21 During reperfusion, mitochondria undergo fission, while the fusion rate decreases.
Adenosine monophosphate‐activated protein kinase (AMPK), a serine/threonine protein kinase family member, regulates endogenous defensive molecules against various pathological processes in the heart. 54 Evidence supports the promotive role of Mel on AMPK activation. Several dynamic processes of mitochondria, such as fusion, fission and mitophagy, are attributed to the AMPK signalling pathway. 5 , 55 , 56 Moreover, the protective effects of Mel in in‐vivo and in‐vitro models have been attributed to the AMPK signalling pathway. 57 , 58 Zhou et al. found that Mel treatment ameliorated Drp1‐dependent mitochondrial fission through activation of AMPKα which subsequently influenced post‐transitional modification of Drp1 by increasing phosphorylation of Drp1 at Ser‐637 residue and decreasing phosphorylation of Drp1 at Ser‐616 residue 53 (Table 1).
TABLE 1.
Summary of the Mel‐induced protective effects on the cardiovascular system via modulation of mitochondrial fission/fusion in in‐vivo studies.
Cardiac disease Model | Animal | Toxic dose of agent, Duration, Route of exposure. | Dose of melatonin, Duration, Route of exposure | Finding(s) | Reference |
---|---|---|---|---|---|
Myocardial ischemia/reperfusion | Male Wistar rats | — | 10 mg/kg/day, 16 weeks, oral | Decreased Mitophagy | 24 |
C57BL/6J mice | — | 20 mg/kg, Single dose, i.p. |
Increased Fusion Increased Mitophagy |
8 | |
C57BL/6J mice | — | 20 mg/kg, Single dose, i.p. | Increased Mitophagy | 20 | |
Male Wistar rats | — | 10 mg/kg, Single dose, i.v. |
Decreased Fission Increased Fusion |
60 | |
C57BL/6J mice | — | 10,20 mg/kg, Single dose, i.p. | Increased Fusion | 29 | |
C57BL/6J mice | — | 20 mg/kg, Single dose, i.p. |
Decreased Fission Decreased Mitophagy |
53 | |
C57BL/6J mice | — | 10 mg/kg/day, 60 days, oral |
Increased Fission Increased Fusion |
66 | |
Myocardial infraction | C57BL/6J mice | — | 10 mg/kg/day, 14 days, i.p. |
Increased Fission |
79 |
Doxorubicin cardiotoxicity | Male Wistar rats |
3 mg/kg i.p. 6 days |
10 mg/kg/day, 30 days, oral |
Decreased Fission Increased Fusion |
7 |
Female Sprague Dawley rats | 4 mg/kg, 3 days, i.p. | 6 mg/kg, 14 days, oral |
Decreased Fission Increased Fusion |
99 | |
Trastuzumab cardiotoxicity |
Male Wistar rats | 4 mg/kg/day, 7 days, i.p. | 10 mg/ kg/day, 7 days, oral |
Decreased Fission Increased Fusion |
101 |
Diabetic cardiomyopathy | C57BL/6J mice | STZ 50 mg/kg/d, 5 days, i.p. | 10 mg/kg, once daily, i.p |
Decreased Fission Increased Fusion |
67 |
C57BL/6J mice | STZ 50 mg/kg/d, 5 days, i.p. | 20 mg/kg, 28 days, oral | Increased Mitophagy | 50 | |
Male Sprague Dawley rats | STZ 40 mg/kg, Single dose, i.p. | 10 mg/kg/d,16 weeks, oral | Decreased Fission | 71 | |
Septic cardiomyopathy | C57BL/6J mice | LPS 15 mg/kg, Single dose, i.p. | 20 mg/kg, Single dose, i.p. |
Decreased Fission Increased Fusion Increased Mitophagy |
73 |
C57BL/6J mice | — | 30 mg/kg, 3 doses, i.p. |
Decreased Fission Increased Fusion Decreased Mitophagy |
75 | |
Post‐traumatic cardiac dysfunction | Male Sprague Dawley rats | — | 30 mg/kg, Single dose, i.p. | Decreased Fission | 77 |
Atherosclerosis | C57BL/6J mice | — | 20 mg/kg/d, 28 days, i.p. | Increased Mitophagy | 30 |
Cardiorenal syndrome type 3 | C57BL/6J mice | — | 20 mg/kg, Single dose, i.p. | Decreased Fission | 102 |
The voltage‐dependent anion channel (VDAC1, mitochondrial porin) is localized on the OMM where its permeability is modulated by protein kinases such as glycerol kinase, creatine kinase and hexokinase. 57 MMP reduction is the trigger for mitophagy activation. Mel treatment induced positive effects by mPTP opening suppression by enhancing the binding of hexokinase 2 to VDAC1. This decreases PINK1/Parkin‐mediated mitophagy. 53 AMPK signalling pathway also contributes to signalling pathways of mitochondrial fusion 8 (Table 1). Mel administration attenuated cardiac I/R damage and relieved mitochondrial stress by activating OPA1‐related mitochondrial fusion and mitophagy in an AMPK‐dependent manner. 8 Mel treatment promoted mitochondrial fusion by enhancing OPA1 protein in cardiomyocytes in a hypoxia/reoxygenation model by increasing cell viability and reducing apoptosis and troponin c secretion. These effects were reversed by silencing the OPA1 gene 10 (Table 2).
TABLE 2.
Summary of the mel‐induced protective effects on the cardiovascular system via modulation of mitochondrial fission/fusion in in‐vitro studies.
Cardiac disease model | Experimental model | Toxic dose of agent Duration | Dose of melatonin, Duration | Finding(s) | Reference |
---|---|---|---|---|---|
In‐vitro studies | |||||
Myocardial ischemia/reperfusion | H9C2 Cells | — | 10 μM | Decreased Mitophagy | 24 |
Isolated perfused rat heart model | — | 0.3,50 μM, 10 min |
Decreased Fission Increased Mitophagy |
36 | |
H9C2 Cells | — | 1000 mM,15 min |
Decreased Fission Increased Fusion |
60 | |
H9C2 Cells | — | — | Increased Fusion | 10 | |
Primary Cardiomyocytes | — | 10,20 μM, 24 h | Increased Fusion | 29 | |
Primary Cardiomyocytes | — | 5 μM, 24 h | Increased Fusion | 65 | |
H9C2 Cells | — | 150 μM, 4 h |
Increased Mitophagy |
24 | |
H9C2 Cells | — | 150 μM, 2 h |
Decreased Fission Decreased Mitophagy |
61 | |
CMEC Cells | — | 5 μmol L, 12 h |
Decreased Fission Decreased Mitophagy |
53 | |
Isolated perfused rat heart model | — | 50 μM, 15 min |
Decreased Fission Increased Fusion |
64 | |
Diabetic cardiomyopathy | H9c2 Cells | — | 100 μmol/L, 4 h |
Decreased Fission Increased Fusion |
67 |
Primary neonatal cardiomyocyte culture | — | 100 μmol/L, 4 h |
Increased Mitophagy |
50 | |
Septic cardiomyopathy | Primary cardiomyocytes |
LPS 5 μg/mL 48 h |
10 μM |
Decreased fission Increased Fusion Increased Mitophagy |
73 |
Post‐traumatic cardiac dysfunction | H9C2 Cells | — |
100 μmol/L |
Decreased Fission |
77 |
Atherosclerosis | RAW264.7 cells |
ox‐LDL 50 mg/mL 24 h |
10 μmol/L, 24 h | Increased Mitophagy | 30 |
Chronic venous disease | HUVEC Cells |
LPS 10 μg/m 12 h |
20 μM, 12 h |
Decreased fission |
92 |
HUVEC Cells | TNF‐α | Not mentioned |
Decreased fission |
103 | |
Vascular Calcification | Isolated VSMC Cells | — |
5 μmol/L, 14 days |
Decreased fission |
89 |
Isolated VSMC Cells | — |
5 μmol/L, 14 days |
Increased Fusion Increased Mitophagy |
90 |
Forkhead box O3a (FOXO3a), a member of the FOX family, is a transcription factor that directly regulates mitophagy by stimulating downstream targets like Parkin. 59 By activating sirtuin 3 (SIRT3)/FOXO3a signalling via its receptor (MT2‐dependent), Mel curbed excessive mitophagy in H9c2 cells caused by anoxia/reoxygenation 24 (Table 2). Singhanat et al. showed that Mel treatment at various time intervals (pretreatment, during ischemia and at the onset of reperfusion) exerted cardioprotective effects against cardiac I/R injury in rats. Mel treatment reduced excessive mitophagy, mitochondrial fission and fusion by activating the MT2 receptor 60 (Table 2).
P62 is the link between LC3 and ubiquitin substrates. The increased expression levels of P62 were correlated with a decrease in mitophagy. Bai et al. demonstrated that Mel treatment reduced mitophagy by P62 upregulation in an anoxia/reoxygenation injury model. The results indicated that Mel post‐conditioning decreased mitophagy through the SIRT3/SOD2 signalling pathway 61 (Table 2).
By regulating mitochondrial dynamics and homeostasis, the Yes‐associated protein (Yap)–Hippo pathway has been associated with cerebral and cardiac IR injury. 62 Mel treatment can activate Yap protein, thereby stabilizing OPA1 and increasing mitochondrial fusion in an I/R model. However, the mechanism by which Yap controls OPA1 expression remains to be elucidated 29 (Table 2).
MiRNAs are small noncoding regulatory RNAs that modify protein‐coding genes post‐transcriptionally. 63 MicroRNAs are also implicated in some mitochondrial‐related processes, such as oxidative phosphorylation, lipid metabolism and mitochondrial dynamics. 64 MicroRNA‐499 (miR‐499) is involved in the modulation of mitochondrial fission by modifying DRP1. 64 The co‐treatment of nicotinamide mononucleotide and melatonin in an isolated I/R rat heart model increased microRNA‐499 expression levels, thereby influencing mitochondrial dynamics proteins by downregulation of DRP1 and upregulation of MFN2 64 (Table 2).
Several non‐conventional mitophagy pathways have been identified. Compelling evidence suggests that a non‐conventional pathway distinct from the PINK1‐Parkin dependent mitophagy pathway plays a role during cardiomyocyte stress. 36 Major components of this pathway are unc‐51 like kinase 1 (ULK1), Rab9, receptor‐interacting serine/threonine protein kinase 1 (Rip1) and Drp1. Mel treatment increased the phosphorylated form of ULK1 and upregulated Rab9 expression. Moreover, Mel reduced mitochondrial fission by increasing DRP‐1 phosphorylation at Ser637 36 (Table 2).
Excessive platelet activation has been correlated with I/R injuries, and this activation is highly energy consuming and, therefore, relies on the function of mitochondria. 20 A recent study explored the effects of Mel treatment on platelet activity in response to I/R damage. 20 Loss of peroxisome proliferator‐activated receptor γ (PPARγ) is related to FUNDC1 dephosphorylation and activation of the mitophagy process. 20 Mel treatment enhanced platelet activation by restoring PPARγ content in platelets, subsequently inhibiting FUNDC1‐required mitophagy 20 (Table 1). In addition, the role of the peroxisome proliferator‐activated receptor‐gamma coactivator 1 alpha (PGC1α) pathway in I/R model has been investigated. 65 Mel treatment upregulated MFN2 and OPA1, whereas deficiency in PGC1α abolished the upregulation of mitochondrial fusion proteins, indicating that mitochondrial biogenesis restored mitochondrial fusion in an I/R model 65 (Table 2).
Aging is one of the primary risk factors for CVD, and mitochondrial dysfunction is a hallmark of aging. 66 Mel treatment was studied in wild‐type C57BL/6J and NLRP3‐KO mice. Mitochondrial fusion proteins (MFN2 and OPA1) normally decline with aging, while Mel treatment significantly increased mitochondrial fusion 66 (Table 1).
2.2. Cardiomyopathy
2.2.1. Diabetic cardiomyopathy
Diabetes continues to increase worldwide. 67 Cardiovascular complications are a primary cause of morbidity and mortality in diabetic patients. 67 Diabetic cardiomyopathy (DCM) is a major chronic complication of diabetes. 67 These complications are independent of hypertension and other heart diseases. 67 Multiple factors are responsible for the pathogenesis of DCM. Factors including excessive mitochondrial ROS production and resultant mitochondrial dysfunction are pivotal in DCM. 67 An imbalance in mitochondrial fission/fusion dynamics is an early component that increases mitochondrial ROS production and induces mitochondrial dysfunction. 67 Aberrant mitochondria are responsible for excessive ROS generation and cell death‐inducing factors. 5 Adequate clearance of aberrant mitochondria by mitophagy may prevent cardiomyocyte apoptosis. 68 To explore the involvement of mitophagy in the pathogenesis of diabetic cardiomyopathy, Wang et al. showed that a serine/threonine‐protein kinase, mammalian sterile 20‐like kinase 1 (MST1), which is attributed to the pathogenesis of many cardiac diseases, was inhibited by Mel treatment. As a result, Mel activated Parkin and promoted mitophagy in a DCM animal model 50 (Table 1). Peroxisome proliferator‐activated receptor gamma coactivator 1‐alpha (PGC‐1α) is a key protein involved in several mitochondrial functions. 69 , 70 Ding et al. reported that Mel treatment reduced Drp1‐mediated mitochondrial fission by activating the SIRT1/PGC‐1α signalling pathway. 67 Reduction of mitochondrial fission by Mel was associated with a decrease in ROS generation and apoptosis, thereby improving mitochondrial cardiac function 67 (Table 1). Long‐term Mel exposure decreased Drp1 phosphorylation at Ser‐616. Mel possibly enhances SIRT6 in a receptor‐dependent manner, and subsequently, SIRT6 activates AMPK/PGC‐1α/AKT signalling, inhibiting Drp1 phosphorylation 71 (Table 1).
2.2.2. Septic Cardiomyopathy
Septic shock is caused by a severe inflammatory response. Despite the therapeutic approaches used so far, sepsis is the leading cause of mortality and morbidity in hospitalized patients worldwide. 72 Sepsis‐induced myocardial injury, a reversible form of cardiac depression, is characterized by cardiomyocyte death, oxidative stress, an excessive inflammatory response, microvascular vasospasms and ventricular systolic disorder. As a result of excess inflammation, cardiomyocytes are the primary targets of death in septic cardiomyopathy. 72 At the molecular level, two signalling pathways are altered in response to septic shock: mitochondrial injury and endoplasmic reticulum (ER) stress. 72 Mitochondrial dynamic imbalance plays a significant role in septic cardiomyopathy. Fission and fusion balance are potential therapeutic targets for preventing and treating septic cardiomyopathy. 72 Receptor‐interacting protein kinase 3 (RIPK3) is a serine/threonine‐protein kinase closely associated with inflammation pathology. 73 The therapeutic effects of Mel on the sepsis model showed that the imbalance of mitochondrial dynamics was reversed by Mel through the suppression of RIPK3 expression 73 (Table 1). The nucleotide‐binding and leucine‐rich repeat pyrin domains have emerged as key mediators of pathological inflammation in many diseases and are potential drug targets. 74 NLRP3 inflammasome seems to be inhibited by Mel, leading to decreased mitochondrial fission by Drp1 downregulation and increased mitochondrial fusion through MFN2 upregulation. Mel treatment modulated PINK1 and Parkin proteins 75 (Table 1).
2.3. Post‐traumatic cardiac dysfunction
Mechanical trauma, such as that caused by motor vehicle collisions, natural disasters and wars, represents a major economic and medical burden. 76 It is well recognized that mechanical trauma may cause direct heart damage, such as cardiac contusion. 76 However, several studies have indicated that mechanical trauma often results in secondary cardiac dysfunction in the later period of trauma, even without direct cardiac injury during the early period. 76 It is difficult to diagnose secondary cardiac dysfunction caused by mechanical trauma accurately because its clinical features are often obscure. 76 Interestingly, after mechanical trauma induced‐cardiac injury, the plasma level of tumour necrosis factor‐α (TNF‐α) increased, which resulted in Drp1 translocation and initiation of mitochondrial fission. Anti‐TNF‐α reduced Drp1 translocation and prevented Drp1 phosphorylation at the Ser616 residue. Mel treatment also attenuated Drp1‐mediated mitochondrial fission through TNF‐α downregulation 77 (Table 1).
2.4. Myocardial infarction
Myocardial infarction (MI) is a life‐threatening cardiovascular disorder that results mostly from the occlusion of a coronary artery, preventing blood flow to the myocardium. Dysregulation of mitochondrial dynamics and mitophagy are closely associated with MI damage. 78 , 79 The Notch signalling pathway is essential for cell communication. This pathway plays a key role in cardiomyocyte development and cardiac homeostasis 80 and is regulated by ligand‐receptor interaction. The ligands are Delta‐like1,3,4 and Jagged1‐2 and the receptors are Notch1‐4. 80 Mel exposure relieved MI damages by regulating the Notch1/MFN2 pathway 79 (Table 1).
3. REGULATION OF MITOCHONDRIAL DYNAMICS AND MITOPHAGY BY MELATONIN IN VASCULAR DISEASES
3.1. Atherosclerosis
Atherosclerosis involves the accumulation of a sticky substance called plaque inside the arteries. 81 Atherosclerosis may result in severe cardiovascular diseases such as ischemia, myocardial infarction and stroke. 81 Proinflammatory components are released during plaque rupture and thrombosis. Inflammation is thought to be the primary cause of atherosclerosis. In atherosclerosis, mitochondrial dysfunction facilitates the inflammatory response and lesion development. 82 Under pathological conditions, mitochondria in endothelial cells undergo fission. 83
NLRP3 is a pattern recognition receptor in the innate immune system. 30 The NLRP3 inflammasome has gained attention as a potential drug target for diseases underpinned by inflammation. 84 Mel treatment induced Sirt3/FOXO3/Parkin‐mediated mitophagy and reduced ROS levels, leading to diminished NLRP3 inflammasome activation 30 (Table 1).
3.2. Vascular calcification
Vascular calcification, defined by the deposition of calcium‐phosphate complexes in the vascular system, is mainly caused by medical conditions like hypertension, diabetes and chronic kidney disease. It is associated with significant cardiovascular impairments. 85 Mitochondria play a key role in the pathogenesis of vascular calcification. 85 High phosphate levels impair mitochondrial function in vascular smooth muscle cells. This dysfunction is demonstrated by low mitochondrial membrane potential and ATP level, excessive ROS production and induction of apoptosis. It has been shown that mitochondrial dynamics contribute to the underlying molecular mechanisms of vascular calcification. 85 , 86 Notably, imbalances in mitochondrial dynamics contributed to inorganic phosphate‐induced vascular smooth muscle cell (VSMCs) calcification by upregulating Drp1 expression and phosphorylation. 87 This was further supported by a study showing that Drp1 was upregulated in osteogenic differentiation of primary human VSMCs. 88 Chen et al. showed that Mel treatment reduced calcification by enhancing AMPK protein expression. This led to Drp1‐mediated mitochondrial fission inhibition and apoptosis suppression. 89 Using compound C, it was possible to demonstrate the effects of Mel on the AMPK/Drp1 signalling pathway in the VSMC isolated from the aortas of Sprague Dawley rats' model. 89 Chen et al. also demonstrated that Mel treatment increased OPA1 expression, enhanced mitochondrial fusion and mitophagy, and consequently attenuated vascular calcification, oxidative stress, inflammation and apoptosis. 90 Knockout of OPA1 in VSMCs, however, abolished the effects of Mel on mitochondrial fusion/mitophagy. 90 The results indicated that mitochondrial fusion/mitophagy activation was related to AMPK activation. 90
3.3. Chronic Venous Disease
Chronic venous disease (CHVD) is a debilitating condition affecting millions worldwide. 91 CHVD is caused by inflammation within the venous circulation and is subjected to increased hydrostatic pressure, resulting in increased ambulatory venous pressure. 91 Along with being a multifactorial disease, the hallmark of CHVD pathophysiology is inflammation. 91 Dysfunctional endothelium is the consequence of the inflammatory cascade, and it plays a pivotal role in CHVD development and progression. 91 Several studies have demonstrated that mitochondrial function affects inflammation‐related cell damage. Endothelial dysfunction is caused by damaged mitochondrial mass caused by abnormal mitochondrial dynamics and/or decreased mitochondrial autophagy. Using a CHVD model, Caui and colleagues explored the possible role of mitochondrial fission in the inflammatory response. 92 They showed that ROS overproduction contributed to DRP1 post‐transitional modification at Ser 616 residue. 92 Since endothelial cells have lower mitochondrial content, it was suggested that the source of ROS overproduction in endothelial cells may be related to xanthine oxidase (XO). 92 More importantly, they showed that the protective effects of Mel against LPS‐induced fission mediated apoptosis was mediated via increased AMPK activity which was accompanied by the upregulation of sarcoplasmic/ER Ca2+ ATPase 2 which is responsible for the reabsorption of Ca2+ into ER 92 (Table 2).
4. REGULATION OF MITOCHONDRIAL DYNAMICS AND MITOPHAGY BY MELATONIN IN CARDIOVASCULAR TOXICITIES
4.1. Doxorubicin cardiotoxicity
Doxorubicin (Dox) is an anthracycline widely used alone or in combination for the treatment of various types of cancer, such as breast cancer, small‐cell lung cancer, acute lymphoblastic leukaemia and acute myeloid leukaemia. 63 , 93 , 94 , 95 Unfortunately, the clinical use of Dox is limited by its irreversible cardiotoxicity. 96 , 97 Dox‐induced cardiotoxicity is multifactorial and not completely understood. Ample evidence, however, suggests various mechanisms such as excessive ROS generation, apoptosis, disturbance of mitochondrial dynamics and dysregulation of mitophagy are associated with Dox‐induced cardiotoxicity. 5 , 7 Mel exerts its cardioprotective effects against Dox‐induced cardiotoxicity by reducing oxidative damage, inflammation and apoptosis. 54 , 98 Arinno et al. demonstrated that Dox‐treated rats exhibited mitochondrial dysfunction by increasing mitochondrial ROS generation, mitochondrial membrane depolarization (MMP) and impairments in mitochondrial dynamics proteins. 7 Mel treatment upregulated proteins related to the fusion process (OPA1, MFN1 and MFN2) and downregulated mitochondrial fission protein (p‐Drp1Ser616/Drp1) in Dox‐induced cardiotoxicity in rats. 7 Mel treatment not only reduced Dox cardiotoxicity but also enhanced the anticancer activity of Dox and suppressed the tumour's growth. 99 Moreover, Mel treatment enhanced mitochondrial function and morphology. PGC‐1α is a regulator of energy metabolism and mitochondrial biogenesis. 99 Dox treatment reduced PGC‐1α levels and subsequently abolished ATP levels. 99 Since the correlation of mitochondrial dynamics with mitochondrial biogenesis was convincing, it was suggested that Mel enhanced mitochondrial biogenesis and dynamics via PGC‐1α regulation in rats 99 (Table 1).
4.2. Trastuzumab cardiotoxicity
Trastuzumab, the humanized monoclonal antibody that specifically targets the human epidermal growth factor receptor 2 (HER2) receptor, is the first choice for HER2+ breast cancer. Unfortunately, cardiotoxicity has emerged as a significant side effect. The molecular mechanisms involved are poorly understood, but all converge on the mitochondria. 100 A recent study showed that trastuzumab cardiotoxicity is associated with mitochondrial dynamics impairment and mitophagy. The results indicated that trastuzumab cardiotoxicity reduced key mitochondrial proteins such as MFN2 and OPA1. In contrast, the key mitochondrial fission protein p‐Drp1Ser616 was upregulated, indicating that trastuzumab cardiotoxicity leads to impairment of normal mitochondrial dynamics. 101 Mel treatment reversed the imbalance of mitochondrial dynamics by increasing MFN2 and OPA1, while reducing the p‐Drp1Ser616/Drp1 ratio 101 (Table 1).
5. CONCLUSION AND PROSPECTIVE
Mitochondria play a crucial role in all cells, including cardiomyocytes, which are particularly sensitive to mitochondrial dysfunction. Recent studies indicated that mitochondrial dynamics and mitophagy changes may impact cardiovascular biology. Dysregulation of mitochondrial dynamics and mitophagy have been linked to a variety of CVDs, such as I/R, cardiomyopathies, cardiotoxicities and vascular diseases. Mel has recently received attention for its potential to target mitochondria and as a potential CVD treatment. This potential has been supported by detecting Mel in isolated mitochondria and identifying specific transporters in mitochondria.
Mitochondria produce free radicals. Mel has both direct and indirect antioxidant effects because it is an electron‐rich molecule (Figure 1). Mel increased the activity of oxidative stress‐related genes, including SOD2, CAT, GPX, Nrf2 and HO‐1. Considering the effects of Mel against oxidative stress‐induced injuries in various in‐vivo and in‐vitro studies, the beneficial effects of Mel can be attributed to its role in mitochondrial function improvement by alleviating oxidative stress.
Cardiomyocytes undergo mitochondrial fission in pathological conditions such as I/R, cardiomyopathies, cardiotoxicities and atherosclerosis. Mel mitigated mitochondrial fission via multiple pathways. Mel enhanced Drp1 activation by regulating AMPK, sirtuins and miR‐499 (Figure 2). Mel also exhibited cardioprotective effects by regulating mitochondrial fusion proteins and mitophagy. Mel treatment upregulated the mitochondrial fusion‐related proteins MFN1/2 and OPA1. Several reports have suggested that Mel's influence on the fusion rate was mediated by its effects on Notch‐1, YAP and AMPK (Figure 2).
FIGURE 2.
A schematic illustration of the Mel‐induced protective effects against CVDs damage through mitochondrial dynamics and mitophagy regulation. ↑ and → present the promote/activate, ⊥ and ↓ present the inhibitory/suppressive effects. AMPK, Adenosine monophosphate‐activated protein kinase, Drp1, dynamin‐related protein 1; MFN1, mitofusin‐1; MFN2, mitofusin‐2; NLRP3, nucleotide‐binding domain and leucine‐rich repeat pyrin domain containing 3; OPA1, optic atrophy 1; PGC1α, peroxisome proliferator‐activated receptor gamma coactivator 1‐alpha; Yap, yes‐associated protein.
Mel may be useful for treating some mitochondrial‐related heart diseases because it is well‐tolerated, less toxic and relatively inexpensive. Future studies should closely examine Mel's regulatory role on upstream pathways. A more detailed understanding of the molecular basis of the effects of Mel may contribute to developing specific therapeutic interventions for CVD patients.
AUTHOR CONTRIBUTIONS
Sohrab Rahmani: Writing – original draft (equal). Ali Roohbakhsh: Writing – review and editing (equal). Vahid Pourbarkhordar: Writing – review and editing (equal). A. Wallace Hayes: Writing – review and editing (equal). Gholamreza Karimi: Conceptualization (equal).
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest.
ACKNOWLEDGEMENTS
The authors are thankful to Mashhad University of Medical Sciences and National Institute for the Development of Medical Sciences Research (Project No. 4021418) for financial support.
Rahmani S, Roohbakhsh A, Pourbarkhordar V, Hayes AW, Karimi G. Melatonin regulates mitochondrial dynamics and mitophagy: Cardiovascular protection. J Cell Mol Med. 2024;28:e70074. doi: 10.1111/jcmm.70074
DATA AVAILABILITY STATEMENT
Not applicable.
REFERENCES
- 1. Imenshahidi M, Karimi G, Hosseinzadeh H. Effects of melatonin on cardiovascular risk factors and metabolic syndrome: a comprehensive review. Naunyn Schmiedeberg's Arch Pharmacol. 2020;393(4):521‐536. [DOI] [PubMed] [Google Scholar]
- 2. Molaei A, Molaei E, Sadeghnia H, Hayes AW, Karimi G. LKB1: an emerging therapeutic target for cardiovascular diseases. Life Sci. 2022;306:120844. [DOI] [PubMed] [Google Scholar]
- 3. Ajoolabady A, Chiong M, Lavandero S, Klionsky DJ, Ren J. Mitophagy in cardiovascular diseases: molecular mechanisms, pathogenesis, and treatment. Trends Mol Med. 2022;28(10):836‐849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Knowlton A, Liu T. Mitochondrial dynamics and heart failure. Compr Physiol. 2015;6(1):507‐526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Rahmani S, Roohbakhsh A, Karimi G. Inhibition of Drp1‐dependent mitochondrial fission by natural compounds as a therapeutic strategy for organ injuries. Pharmacol Res. 2023;188:106672. [DOI] [PubMed] [Google Scholar]
- 6. Rahmani S, Rezaei M. Toxicity of fluoride on isolated rat liver mitochondria. J Fluor Chem. 2020;239:109636. [Google Scholar]
- 7. Arinno A, Maneechote C, Khuanjing T, et al. Cardioprotective effects of melatonin and metformin against doxorubicin‐induced cardiotoxicity in rats are through preserving mitochondrial function and dynamics. Biochem Pharmacol. 2021;192:114743. [DOI] [PubMed] [Google Scholar]
- 8. Zhang Y, Wang Y, Xu J, et al. Melatonin attenuates myocardial ischemia‐reperfusion injury via improving mitochondrial fusion/mitophagy and activating the AMPK‐OPA1 signaling pathways. J Pineal Res. 2019;66(2):e12542. [DOI] [PubMed] [Google Scholar]
- 9. Wang SH, Zhu XL, Wang F, et al. LncRNA H19 governs mitophagy and restores mitochondrial respiration in the heart through Pink1/parkin signaling during obesity. Cell Death Dis. 2021;12(6):557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Ma X, Wang S, Cheng H, Ouyang H, Ma X. Melatonin attenuates ischemia/reperfusion‐induced oxidative stress by activating mitochondrial fusion in cardiomyocytes. Oxidative Med Cell Longev. 2022;2022:7105181. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 11. Sui YB, Xiu J, Wei JX, Pan PP, Sun BH, Liu L. Shen Qi Li Xin formula improves chronic heart failure through balancing mitochondrial fission and fusion via upregulation of PGC‐1α. J Physiol Sci. 2021;71(1):32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Li E, Li X, Huang J, et al. BMAL1 regulates mitochondrial fission and mitophagy through mitochondrial protein BNIP3 and is critical in the development of dilated cardiomyopathy. Protein Cell. 2020;11(9):661‐679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Tan X, Chen YF, Zou SY, et al. ALDH2 attenuates ischemia and reperfusion injury through regulation of mitochondrial fusion and fission by PI3K/AKT/mTOR pathway in diabetic cardiomyopathy. Free Radic Biol Med. 2023;195:219‐230. [DOI] [PubMed] [Google Scholar]
- 14. Catanzaro MP, Weiner A, Kaminaris A, et al. Doxorubicin‐induced cardiomyocyte death is mediated by unchecked mitochondrial fission and mitophagy. FASEB J. 2019;33(10):11096‐11108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Zhou L, Li R, Liu C, et al. Foxo3a inhibits mitochondrial fission and protects against doxorubicin‐induced cardiotoxicity by suppressing MIEF2. Free Radic Biol Med. 2017;104:360‐370. [DOI] [PubMed] [Google Scholar]
- 16. Lerner AB, Case JD, Takahashi Y, Lee TH, Mori W. Isolation of melatonin, the pineal gland factor that lightens melanocyteS1. J Am Chem Soc. 1958;80(10):2587. [Google Scholar]
- 17. Roohbakhsh A, Shamsizadeh A, Hayes AW, Reiter RJ, Karimi G. Melatonin as an endogenous regulator of diseases: the role of autophagy. Pharmacol Res. 2018;133:265‐276. [DOI] [PubMed] [Google Scholar]
- 18. Yarmohammadi F, Barangi S, Aghaee‐Bakhtiari SH, et al. Melatonin ameliorates arsenic‐induced cardiotoxicity through the regulation of the Sirt1/Nrf2 pathway in rats. Biofactors. 2023;49(3):620‐635. [DOI] [PubMed] [Google Scholar]
- 19. Pandi‐Perumal SR, BaHammam AS, Ojike NI, et al. Melatonin and human cardiovascular disease. J Cardiovasc Pharmacol Ther. 2017;22(2):122‐132. [DOI] [PubMed] [Google Scholar]
- 20. Zhou H, Li D, Zhu P, et al. Melatonin suppresses platelet activation and function against cardiac ischemia/reperfusion injury via PPARγ/FUNDC1/mitophagy pathways. J Pineal Res. 2017;63(4). [DOI] [PubMed] [Google Scholar]
- 21. Randhawa PK, Gupta MK. Melatonin as a protective agent in cardiac ischemia‐reperfusion injury: vision/illusion? Eur J Pharmacol. 2020;885:173506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Lacoste B, Angeloni D, Dominguez‐Lopez S, et al. Anatomical and cellular localization of melatonin MT1 and MT2 receptors in the adult rat brain. J Pineal Res. 2015;58(4):397‐417. [DOI] [PubMed] [Google Scholar]
- 23. Lochner A, Marais E, Huisamen B. Melatonin and cardioprotection against ischaemia/reperfusion injury: What's new? A review. J Pineal Res. 2018;65(1):e12490. [DOI] [PubMed] [Google Scholar]
- 24. Wu J, Yang Y, Gao Y, Wang Z, Ma J. Melatonin attenuates anoxia/reoxygenation injury by inhibiting excessive mitophagy through the MT2/SIRT3/FoxO3a signaling pathway in H9c2 cells. Drug Des Devel Ther. 2020;14:2047‐2060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Tan DX, Manchester LC, Qin L, Reiter RJ. Melatonin: a mitochondrial targeting molecule involving mitochondrial protection and dynamics. Int J Mol Sci. 2016;17(12):2124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Reiter RJ, Rosales‐Corral S, Tan DX, Jou MJ, Galano A, Xu B. Melatonin as a mitochondria‐targeted antioxidant: one of evolution's best ideas. Cell Mol Life Sci. 2017;74(21):3863‐3881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Tan DX, Manchester LC, Esteban‐Zubero E, Zhou Z, Reiter RJ. Melatonin as a potent and inducible endogenous antioxidant: synthesis and metabolism. Molecules. 2015;20(10):18886‐18906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Huo X, Wang C, Yu Z, et al. Human transporters, PEPT1/2, facilitate melatonin transportation into mitochondria of cancer cells: an implication of the therapeutic potential. J Pineal Res. 2017;62(4):e12390. [DOI] [PubMed] [Google Scholar]
- 29. Ma S, Dong Z. Melatonin attenuates cardiac reperfusion stress by improving OPA1‐related mitochondrial fusion in a yap‐hippo pathway‐dependent manner. J Cardiovasc Pharmacol. 2019;73(1):27‐39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Ma S, Chen J, Feng J, et al. Melatonin ameliorates the progression of atherosclerosis via mitophagy activation and NLRP3 inflammasome inhibition. Oxidative Med Cell Longev. 2018;2018:9286458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Jin JY, Wei XX, Zhi XL, Wang XH, Meng D. Drp1‐dependent mitochondrial fission in cardiovascular disease. Acta Pharmacol Sin. 2021;42(5):655‐664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Chang CR, Manlandro CM, Arnoult D, et al. A lethal de novo mutation in the middle domain of the dynamin‐related GTPase Drp1 impairs higher order assembly and mitochondrial division. J Biol Chem. 2010;285(42):32494‐32503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Serasinghe MN, Chipuk JE. Mitochondrial fission in human diseases. Handb Exp Pharmacol. 2017;240:159‐188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Qi Z, Huang Z, Xie F, Chen L. Dynamin‐related protein 1: a critical protein in the pathogenesis of neural system dysfunctions and neurodegenerative diseases. J Cell Physiol. 2019;234(7):10032‐10046. [DOI] [PubMed] [Google Scholar]
- 35. Tong M, Zablocki D, Sadoshima J. The role of Drp1 in mitophagy and cell death in the heart. J Mol Cell Cardiol. 2020;142:138‐145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Dube K, Dhanabalan K, Salie R, Blignaut M, Huisamen B, Lochner A. Melatonin has profound effects on mitochondrial dynamics in myocardial ischaemia/reperfusion. Heliyon. 2019;5(10):e02659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Zhang H, Wang P, Bisetto S, et al. A novel fission‐independent role of dynamin‐related protein 1 in cardiac mitochondrial respiration. Cardiovasc Res. 2017;113(2):160‐170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Samant SA, Zhang HJ, Hong Z, et al. SIRT3 deacetylates and activates OPA1 to regulate mitochondrial dynamics during stress. Mol Cell Biol. 2014;34(5):807‐819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Piquereau J, Caffin F, Novotova M, et al. Down‐regulation of OPA1 alters mouse mitochondrial morphology, PTP function, and cardiac adaptation to pressure overload. Cardiovasc Res. 2012;94(3):408‐417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Titus AS, Sung EA, Zablocki D, Sadoshima J. Mitophagy for cardioprotection. Basic Res Cardiol. 2023;118(1):42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Li Y, Zheng W, Lu Y, et al. BNIP3L/NIX‐mediated mitophagy: molecular mechanisms and implications for human disease. Cell Death Dis. 2021;13(1):14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Lazarou M, Jin SM, Kane LA, Youle RJ. Role of PINK1 binding to the TOM complex and alternate intracellular membranes in recruitment and activation of the E3 ligase parkin. Dev Cell. 2012;22(2):320‐333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Yamano K, Youle RJ. PINK1 is degraded through the N‐end rule pathway. Autophagy. 2013;9(11):1758‐1769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Narendra D, Tanaka A, Suen DF, Youle RJ. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol. 2008;183(5):795‐803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Gladkova C, Maslen SL, Skehel JM, Komander D. Mechanism of parkin activation by PINK1. Nature. 2018;559(7714):410‐414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Novak I, Kirkin V, McEwan DG, et al. Nix is a selective autophagy receptor for mitochondrial clearance. EMBO Rep. 2010;11(1):45‐51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Liu L, Feng D, Chen G, et al. Mitochondrial outer‐membrane protein FUNDC1 mediates hypoxia‐induced mitophagy in mammalian cells. Nat Cell Biol. 2012;14(2):177‐185. [DOI] [PubMed] [Google Scholar]
- 48. Li A, Gao M, Liu B, et al. Mitochondrial autophagy: molecular mechanisms and implications for cardiovascular disease. Cell Death Dis. 2022;13(5):444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Kuang Y, Ma K, Zhou C, et al. Structural basis for the phosphorylation of FUNDC1 LIR as a molecular switch of mitophagy. Autophagy. 2016;12(12):2363‐2373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Wang S, Zhao Z, Feng X, et al. Melatonin activates parkin translocation and rescues the impaired mitophagy activity of diabetic cardiomyopathy through Mst1 inhibition. J Cell Mol Med. 2018;22(10):5132‐5144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Shires SE, Gustafsson ÅB. Mitophagy and heart failure. J Mol Med (Berl). 2015;93(3):253‐262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Matsuda T, Zhai P, Sciarretta S, et al. NF2 activates hippo signaling and promotes ischemia/reperfusion injury in the heart. Circ Res. 2016;119(5):596‐606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Zhou H, Zhang Y, Hu S, et al. Melatonin protects cardiac microvasculature against ischemia/reperfusion injury via suppression of mitochondrial fission‐VDAC1‐HK2‐mPTP‐mitophagy axis. J Pineal Res. 2017;63(1):e12413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Liu D, Ma Z, Di S, et al. AMPK/PGC1α activation by melatonin attenuates acute doxorubicin cardiotoxicity via alleviating mitochondrial oxidative damage and apoptosis. Free Radic Biol Med. 2018;129:59‐72. [DOI] [PubMed] [Google Scholar]
- 55. Liang J, Xu ZX, Ding Z, et al. Myristoylation confers noncanonical AMPK functions in autophagy selectivity and mitochondrial surveillance. Nat Commun. 2015;6:7926. [DOI] [PubMed] [Google Scholar]
- 56. Wang Q, Zhang M, Torres G, et al. Metformin suppresses diabetes‐accelerated atherosclerosis via the inhibition of Drp1‐mediated mitochondrial fission. Diabetes. 2017;66(1):193‐205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Shoshan‐Barmatz V, De Pinto V, Zweckstetter M, Raviv Z, Keinan N, Arbel N. VDAC, a multi‐functional mitochondrial protein regulating cell life and death. Mol Asp Med. 2010;31(3):227‐285. [DOI] [PubMed] [Google Scholar]
- 58. Wang X, Xue GX, Liu WC, et al. Melatonin alleviates lipopolysaccharide‐compromised integrity of blood‐brain barrier through activating AMP‐activated protein kinase in old mice. Aging Cell. 2017;16(2):414‐421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Gupta P, Sharma G, Lahiri A, Barthwal MK. FOXO3a acetylation regulates PINK1, mitophagy, inflammasome activation in murine palmitate‐conditioned and diabetic macrophages. J Leukoc Biol. 2022;111(3):611‐627. [DOI] [PubMed] [Google Scholar]
- 60. Singhanat K, Apaijai N, Jaiwongkam T, Kerdphoo S, Chattipakorn SC, Chattipakorn N. Melatonin as a therapy in cardiac ischemia‐reperfusion injury: potential mechanisms by which MT2 activation mediates cardioprotection. J Adv Res. 2021;29:33‐44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Bai Y, Yang Y, Gao Y, Lin D, Wang Z, Ma J. Melatonin postconditioning ameliorates anoxia/reoxygenation injury by regulating mitophagy and mitochondrial dynamics in a SIRT3‐dependent manner. Eur J Pharmacol. 2021;904:174157. [DOI] [PubMed] [Google Scholar]
- 62. Geng C, Wei J, Wu C. Yap‐hippo pathway regulates cerebral hypoxia‐reoxygenation injury in neuroblastoma N2a cells via inhibiting ROCK1/F‐actin/mitochondrial fission pathways. Acta Neurol Belg. 2020;120(4):879‐892. [DOI] [PubMed] [Google Scholar]
- 63. Yarmohammadi F, Ebrahimian Z, Karimi G. MicroRNAs target the PI3K/Akt/p53 and the Sirt1/Nrf2 signaling pathways in doxorubicin‐induced cardiotoxicity. J Biochem Mol Toxicol. 2023;37(2):e23261. [DOI] [PubMed] [Google Scholar]
- 64. Mokhtari B, Hosseini L, Høilund‐Carlsen PF, Salehinasab R, Rajabi M, Badalzadeh R. The additive effects of nicotinamide mononucleotide and melatonin on mitochondrial biogenesis and fission/fusion, autophagy, and microRNA‐499 in the aged rat heart with reperfusion injury. Naunyn Schmiedeberg's Arch Pharmacol. 2023;396(8):1701‐1711. [DOI] [PubMed] [Google Scholar]
- 65. Qi X, Wang J. Melatonin improves mitochondrial biogenesis through the AMPK/PGC1α pathway to attenuate ischemia/reperfusion‐induced myocardial damage. Aging. 2020;12(8):7299‐7312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Fernández‐Ortiz M, Sayed RKA, Fernández‐Martínez J, et al. Melatonin/Nrf2/NLRP3 connection in mouse heart mitochondria during aging. Antioxidants (Basel). 2020;9(12):1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Ding M, Feng N, Tang D, et al. Melatonin prevents Drp1‐mediated mitochondrial fission in diabetic hearts through SIRT1‐PGC1α pathway. J Pineal Res. 2018;65(2):e12491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Saito T, Nah J, Oka SI, et al. An alternative mitophagy pathway mediated by Rab9 protects the heart against ischemia. J Clin Invest. 2019;129(2):802‐819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Xu W, Yan J, Ocak U, et al. Melanocortin 1 receptor attenuates early brain injury following subarachnoid hemorrhage by controlling mitochondrial metabolism via AMPK/SIRT1/PGC‐1α pathway in rats. Theranostics. 2021;11(2):522‐539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Helli B, Navabi SP, Hosseini SA, et al. The protective effects of syringic acid on bisphenol A‐induced neurotoxicity possibly through AMPK/PGC‐1α/Fndc5 and CREB/BDNF signaling pathways. Mol Neurobiol. 2024;7767‐7784. [DOI] [PubMed] [Google Scholar]
- 71. Yu LM, Dong X, Xue XD, et al. Melatonin attenuates diabetic cardiomyopathy and reduces myocardial vulnerability to ischemia‐reperfusion injury by improving mitochondrial quality control: role of SIRT6. J Pineal Res. 2021;70(1):e12698. [DOI] [PubMed] [Google Scholar]
- 72. Cimolai MC, Alvarez S, Bode C, Bugger H. Mitochondrial mechanisms in septic cardiomyopathy. Int J Mol Sci. 2015;16(8):17763‐17778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Zhong J, Tan Y, Lu J, et al. Therapeutic contribution of melatonin to the treatment of septic cardiomyopathy: a novel mechanism linking Ripk3‐modified mitochondrial performance and endoplasmic reticulum function. Redox Biol. 2019;26:101287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Zahid A, Li B, Kombe AJK, Jin T, Tao J. Pharmacological inhibitors of the NLRP3 inflammasome. Front Immunol. 2019;10:2538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Rahim I, Sayed RK, Fernández‐Ortiz M, et al. Melatonin alleviates sepsis‐induced heart injury through activating the Nrf2 pathway and inhibiting the NLRP3 inflammasome. Naunyn Schmiedeberg's Arch Pharmacol. 2021;394(2):261‐277. [DOI] [PubMed] [Google Scholar]
- 76. Weber B, Lackner I, Gebhard F, Miclau T, Kalbitz M. Trauma, a matter of the heart‐molecular mechanism of post‐traumatic cardiac dysfunction. Int J Mol Sci. 2021;22(2):737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Ding M, Ning J, Feng N, et al. Dynamin‐related protein 1‐mediated mitochondrial fission contributes to post‐traumatic cardiac dysfunction in rats and the protective effect of melatonin. J Pineal Res. 2018;64(1):e12447. [DOI] [PubMed] [Google Scholar]
- 78. Turkieh A, El Masri Y, Pinet F, Dubois‐Deruy E. Mitophagy regulation following myocardial infarction. Cells. 2022;11(2):199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Pei H, Du J, Song X, et al. Melatonin prevents adverse myocardial infarction remodeling via Notch1/Mfn2 pathway. Free Radic Biol Med. 2016;97:408‐417. [DOI] [PubMed] [Google Scholar]
- 80. Luxán G, D'Amato G, MacGrogan D, de la Pompa JL. Endocardial notch signaling in cardiac development and disease. Circ Res. 2016;118(1):e1‐e18. [DOI] [PubMed] [Google Scholar]
- 81. Khwaja B, Thankam FG, Agrawal DK. Mitochondrial DAMPs and altered mitochondrial dynamics in OxLDL burden in atherosclerosis. Mol Cell Biochem. 2021;476(4):1915‐1928. [DOI] [PubMed] [Google Scholar]
- 82. Markin AM, Khotina VA, Zabudskaya XG, et al. Disturbance of mitochondrial dynamics and mitochondrial therapies in atherosclerosis. Life (Basel, Switzerland). 2021;11(2):165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Chehaitly A, Guihot AL, Proux C, et al. Altered mitochondrial Opa1‐related fusion in mouse promotes endothelial cell dysfunction and atherosclerosis. Antioxidants (Basel, Switzerland). 2022;11(6):1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Coll RC, Schroder K, Pelegrín P. NLRP3 and pyroptosis blockers for treating inflammatory diseases. Trends Pharmacol Sci. 2022;43(8):653‐668. [DOI] [PubMed] [Google Scholar]
- 85. Lee SJ, Lee IK, Jeon JH. Vascular calcification‐new insights into its mechanism. Int J Mol Sci. 2020;21(8):2685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Ma WQ, Sun XJ, Wang Y, Zhu Y, Han XQ, Liu NF. Restoring mitochondrial biogenesis with metformin attenuates β‐GP‐induced phenotypic transformation of VSMCs into an osteogenic phenotype via inhibition of PDK4/oxidative stress‐mediated apoptosis. Mol Cell Endocrinol. 2019;479:39‐53. [DOI] [PubMed] [Google Scholar]
- 87. Cui L, Li Z, Chang X, Cong G, Hao L. Quercetin attenuates vascular calcification by inhibiting oxidative stress and mitochondrial fission. Vasc Pharmacol. 2017;88:21‐29. [DOI] [PubMed] [Google Scholar]
- 88. Rogers MA, Maldonado N, Hutcheson JD, et al. Dynamin‐related protein 1 inhibition attenuates cardiovascular calcification in the presence of oxidative stress. Circ Res. 2017;121(3):220‐233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Chen WR, Zhou YJ, Sha Y, Wu XP, Yang JQ, Liu F. Melatonin attenuates vascular calcification by inhibiting mitochondria fission via an AMPK/Drp1 signalling pathway. J Cell Mol Med. 2020;24(11):6043‐6054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Chen WR, Zhou YJ, Yang JQ, Liu F, Wu XP, Sha Y. Melatonin attenuates calcium deposition from vascular smooth muscle cells by activating mitochondrial fusion and mitophagy via an AMPK/OPA1 signaling pathway. Oxidative Med Cell Longev. 2020;2020:5298483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Raffetto JD, Mannello F. Pathophysiology of chronic venous disease, international angiology: a journal of the international union of. Angiology. 2014;33(3):212‐221. [PubMed] [Google Scholar]
- 92. Cui J, Li Z, Zhuang S, et al. Melatonin alleviates inflammation‐induced apoptosis in human umbilical vein endothelial cells via suppression of Ca(2+)‐XO‐ROS‐Drp1‐mitochondrial fission axis by activation of AMPK/SERCA2a pathway. Cell Stress Chaperones. 2018;23(2):281‐293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Tabrizi FB, Yarmohammadi F, Hayes AW, Karimi G. The modulation of SIRT1 and SIRT3 by natural compounds as a therapeutic target in doxorubicin‐induced cardiotoxicity: a review. J Biochem Mol Toxicol. 2022;36(1):e22946. [DOI] [PubMed] [Google Scholar]
- 94. Yarmohammadi F, Rezaee R, Karimi G. Natural compounds against doxorubicin‐induced cardiotoxicity: a review on the involvement of Nrf2/ARE signaling pathway. Phytother Res. 2021;35(3):1163‐1175. [DOI] [PubMed] [Google Scholar]
- 95. Rahbardar MG, Eisvand F, Rameshrad M, Razavi BM, Hosseinzadeh H. In vivo and in vitro protective effects of Rosmarinic acid against doxorubicin‐induced cardiotoxicity. Nutr Cancer. 2022;74(2):747‐760. [DOI] [PubMed] [Google Scholar]
- 96. Rahmani S, Naraki K, Roohbakhsh A, Hayes AW, Karimi G. The protective effects of rutin on the liver, kidneys, and heart by counteracting organ toxicity caused by synthetic and natural compounds. Food Sci Nutr. 2023;11(1):39‐56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Eisvand F, Imenshahidi M, Ghasemzadeh Rahbardar M, et al. Cardioprotective effects of alpha‐mangostin on doxorubicin‐induced cardiotoxicity in rats. Phytother Res. 2022;36(1):506‐524. [DOI] [PubMed] [Google Scholar]
- 98. Koşar PA, Nazıroğlu M, Övey İS, Çiğ B. Synergic effects of doxorubicin and melatonin on apoptosis and mitochondrial oxidative stress in MCF‐7 breast cancer cells: involvement of TRPV1 channels. J Membr Biol. 2016;249(1–2):129‐140. [DOI] [PubMed] [Google Scholar]
- 99. Govender J, Loos B, Marais E, Engelbrecht AM. Melatonin improves cardiac and mitochondrial function during doxorubicin‐induced cardiotoxicity: a possible role for peroxisome proliferator‐activated receptor gamma coactivator 1‐alpha and sirtuin activity? Toxicol Appl Pharmacol. 2018;358:86‐101. [DOI] [PubMed] [Google Scholar]
- 100. Pecoraro M, Pinto A, Popolo A. Trastuzumab‐induced cardiotoxicity and role of mitochondrial connexin43 in the adaptive response. Toxicol In Vitro. 2020;67:104926. [DOI] [PubMed] [Google Scholar]
- 101. Arinno A, Maneechote C, Khuanjing T, et al. Melatonin and metformin ameliorated trastuzumab‐induced cardiotoxicity through the modulation of mitochondrial function and dynamics without reducing its anticancer efficacy. Biophys Acta Mol Basis Dis. 2023;1869(2):166618. [DOI] [PubMed] [Google Scholar]
- 102. Wang J, Toan S, Li R, Zhou H. Melatonin fine‐tunes intracellular calcium signals and eliminates myocardial damage through the IP3R/MCU pathways in cardiorenal syndrome type 3. Biochem Pharmacol. 2020;174:113832. [DOI] [PubMed] [Google Scholar]
- 103. Lu K, Liu X, Guo W. Melatonin attenuates inflammation‐related venous endothelial cells apoptosis through modulating the MST1‐MIEF1 pathway. J Cell Physiol. 2019;234(12):23675‐23684. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Not applicable.