Startseite Mitochondrial quality control as a therapeutic target in cardiovascular disease: Mechanistic insights and future directions
Artikel Open Access

Mitochondrial quality control as a therapeutic target in cardiovascular disease: Mechanistic insights and future directions

  • Miao Zhang , Tong Zhang , Rongjun Zou , Kunyang He , Ru Huang , Jingrui Feng , Jinlin Hu , Teng Ge , Xiaoping Fan , Hao Zhou ORCID logo EMAIL logo und Yang Chen ORCID logo EMAIL logo
Veröffentlicht/Copyright: 20. Juni 2025

Abstract

Mitochondrial dysfunction is increasingly recognized as a critical driver in the pathogenesis of cardiovascular diseases. Mitochondrial quality control (MQC) is an ensemble of adaptive mechanisms aimed at maintaining mitochondrial integrity and functionality and is essential for cardiomyocyte viability and optimal cardiac performance under the stress of cardiovascular pathology. The key MQC components include mitochondrial fission, fusion, mitophagy, and mitochondria-dependent cell death, each contributing uniquely to cellular homeostasis. The dynamic interplay among these processes is intricately linked to pathological phenomena, such as redox imbalance, calcium overload, dysregulated energy metabolism, impaired signal transduction, mitochondrial unfolded protein response, and endoplasmic reticulum stress. Aberrant mitochondrial fission is an early marker of mitochondrial injury and cardiomyocyte apoptosis, whereas reduced mitochondrial fusion is frequently observed in stressed cardiomyocytes and is associated with mitochondrial dysfunction and cardiac impairment. Mitophagy is a protective, selective autophagic degradation process that eliminates structurally compromised mitochondria, preserving mitochondrial network integrity. However, dysregulated mitophagy can exacerbate cellular injury, promoting cell death. Beyond their role as the primary energy source of the cell, mitochondria are also central regulators of cardiomyocyte survival, mediating apoptosis and necroptosis in reperfused myocardium. Consequently, MQC impairment may be a determining factor in cardiomyocyte fate. This review consolidates current insights into the regulatory mechanisms and pathological significance of MQC across diverse cardiovascular conditions, highlighting potential therapeutic avenues for the clinical management of heart diseases.

Graphical Abstract

 Healthy cardiomyocytes feature a precise balance between mitochondrial fusion (mediated by MFN1/2 and Opa1) and division (mediated by Drp1). Fusion contributes to mitochondrial network stability and functional restoration, while division ensures mitochondrial renewal and the removal of damaged parts. PGC-1α, NRF1/2 and TFAM regulate mitochondrial biogenesis to ensure mtDNA transcription and replication and maintain mitochondrial quantity and quality. Mitochondrial autophagy selectively removes damaged mitochondria through LC3II, FUNDC1, PINK1-Parkin, and other molecular pathways to prevent the accumulation of cell damage. The cell begins to undergo a series of stress responses when the balance between mitochondrial fusion and division is disrupted or biogenesis and autophagy are impaired. Initially, BAX activates caspase-9, initiating the apoptosis pathway, leading to the activation of caspase-3 and cell death. If mitochondrial damage is aggravated, RIPK3 activates MLKL, which triggers necroptosis through CAMKII mediation, leading to cell structure destruction and loss of function. The interactions between mitochondrial fusion and division, biogenesis and autophagy were demonstrated, and the specific mechanisms of how these processes progressively affect cardiomyocyte survival are described. MFN1/2: mitofusin 1 and 2; Opa1: optic atrophy protein 1; Drp1: dynamin related protein 1; FUNDC1: Fun14 domain-containing protein 1; PINK1: PTEN induced putative kinase 1; RIPK3: receptor-interacting protein kinase 3; MLKL: mixed lineage kinase domain-like protein; CAMKII: Ca2+-calmodulin-dependent protein kinase II.

Healthy cardiomyocytes feature a precise balance between mitochondrial fusion (mediated by MFN1/2 and Opa1) and division (mediated by Drp1). Fusion contributes to mitochondrial network stability and functional restoration, while division ensures mitochondrial renewal and the removal of damaged parts. PGC-1α, NRF1/2 and TFAM regulate mitochondrial biogenesis to ensure mtDNA transcription and replication and maintain mitochondrial quantity and quality. Mitochondrial autophagy selectively removes damaged mitochondria through LC3II, FUNDC1, PINK1-Parkin, and other molecular pathways to prevent the accumulation of cell damage. The cell begins to undergo a series of stress responses when the balance between mitochondrial fusion and division is disrupted or biogenesis and autophagy are impaired. Initially, BAX activates caspase-9, initiating the apoptosis pathway, leading to the activation of caspase-3 and cell death. If mitochondrial damage is aggravated, RIPK3 activates MLKL, which triggers necroptosis through CAMKII mediation, leading to cell structure destruction and loss of function. The interactions between mitochondrial fusion and division, biogenesis and autophagy were demonstrated, and the specific mechanisms of how these processes progressively affect cardiomyocyte survival are described. MFN1/2: mitofusin 1 and 2; Opa1: optic atrophy protein 1; Drp1: dynamin related protein 1; FUNDC1: Fun14 domain-containing protein 1; PINK1: PTEN induced putative kinase 1; RIPK3: receptor-interacting protein kinase 3; MLKL: mixed lineage kinase domain-like protein; CAMKII: Ca2+-calmodulin-dependent protein kinase II.

Introduction

The mitochondria are maternally inherited and originally derived from ancient bacterial ancestors, and are essential double-membraned organelles present in nearly all eukaryotic cells and even certain prokaryotes.[1] Traditionally regarded as the primary source of cellular adenosine 5’-triphosphate (ATP) via oxidative phosphorylation, the mitochondria are pivotal in energy production through glucose and lipid metabolism. However, detection technology advancements have revealed that mitochondrial function extends far beyond energy metabolism. The mitochondria are central hubs for cellular signaling, integration, and amplification, regulating oxidative stress, calcium homeostasis, and apoptosis processes.[2, 3] Mitochondrial dysfunction leads to energetic stress and also disrupts these critical signaling networks, contributing to the pathogenesis of numerous human diseases, including cardiovascular disorders, chronic obstructive pulmonary disease, acute kidney injury,[4] metabolic syndrome,[5] and cancer.[6]

Mitochondrial impairments, such as genetic mutations, compromised membrane integrity, metabolic inactivity, and pro-apoptotic signaling activation, act as independent pathological drivers and also synergize with other molecular mechanisms, aggravating disease processes.[6] Consequently, understanding and targeting mitochondrial damage has become a promising avenue for developing therapeutic strategies that aim to restore mitochondrial health and treat a broad spectrum of human diseases.

In cardiovascular diseases, such as myocardial ischemiareperfusion (I/R) injury, hypertension, coronary atherosclerosis, diabetic cardiomyopathy (DCM), and azithromycin-induced cardiomyopathy, mitochondrial quality control (MQC) is critical in maintaining cardiomyocyte resilience against stress.[7, 8] MQC refers to a suite of adaptive responses that regulate mitochondrial morphology, ATP synthesis, genomic stability, protein homeostasis, and intracellular signaling.[9] In response to mild stresses, such as hypoxia, mitochondrial dynamics, primarily fission, are upregulated to increase mitochondrial mass and match cellular energy demands. During this phase, mild increases in mitochondrial calcium enhance metabolic enzyme activity, adapting mitochondria to shifting energy requirements.[10]

Moderate stressors, such as oxidative stress or inflammation, compromise mitochondrial membrane integrity, leading to outer mitochondrial membrane (OMM) hyperpermeability.[11] This disruption releases mitochondrial signaling molecules into the cytoplasm, potentially initiating inflammatory responses, and also promotes water influx, leading to mitochondrial swelling.[12] Damaged mitochondria can undergo fusion with healthier counterparts, mitigating the accumulation of dysfunctional components within the mitochondrial network. When damage becomes irreversible, the structurally compromised mitochondria are removed through mitophagy, a selective form of autophagy. This recycling process is facilitated by key mitophagy regulators, such as B-cell lymphoma-2 (BCL2)/adenovirus E1B 19 kDa protein-interacting protein 3 (Bnip3), Parkin, and Fun14 domain-containing protein 1 (Fundc1), while mitochondrial biogenesis simultaneously compensates for mitochondrial loss, enabling the generation of a rejuvenated mitochondrial network.[13]

However, the mitochondria initiate the cell death pathways when stress overwhelms these protective mechanisms, including apoptosis, necrosis, and necroptosis. Mitochondrial-mediated cell death represents a crucial cellular decision point: while apoptosis and necroptosis offer potentially reversible outcomes, mitochondrial-driven necrosis is terminal and non-reversible.[14] Therefore, the mitochondria serve as the energy centers of the cell and are also the critical arbiters of cell fate, particularly in the face of severe cardiovascular stressors.

This review consolidates insights into the diverse roles of MQC across various cardiovascular diseases, examining the complex interactions among mitochondrial dynamics, mitophagy, biogenesis, and cell death regulation. Cells respond to fluctuating energy demands, redox imbalance, and calcium dysregulation through mitochondrial fission and fusion, while mitophagy and biogenesis ensure the removal and replenishment of damaged mitochondrial units. Additionally, cell death pathways are explored with a focus on the transition from reversible to irreversible cell death in conditions such as DCM and drug-induced cardiomyopathy. By advancing our understanding of these MQC mechanisms, we aim to highlight potential therapeutic targets that could mitigate mitochondrial dysfunction and enhance cardiomyocyte survival across a range of cardiovascular disorders (Figure 1).

Figure 1 Review of MQC in physiological conditions and cardiovascular diseases. MQC coordinates fission, fusion, mitochondrial autophagy, and mitochondria-controlled cell death to ensure cell homeostasis. Mitochondrial dysfunction is thought to be a major mechanism in cardiovascular disease development, and failure of quality control processes can exacerbate mitochondrial dysfunction. Several potential MQC targets could treat cardiovascular disease by inhibiting mitochondrial division, promoting mitochondrial fusion, moderately activating mitochondrial autophagy, and inhibiting mitochondria-dependent cell death. MQC: mitochondrial quality control; ATP: adenosine 5’-triphosphate.
Figure 1

Review of MQC in physiological conditions and cardiovascular diseases. MQC coordinates fission, fusion, mitochondrial autophagy, and mitochondria-controlled cell death to ensure cell homeostasis. Mitochondrial dysfunction is thought to be a major mechanism in cardiovascular disease development, and failure of quality control processes can exacerbate mitochondrial dysfunction. Several potential MQC targets could treat cardiovascular disease by inhibiting mitochondrial division, promoting mitochondrial fusion, moderately activating mitochondrial autophagy, and inhibiting mitochondria-dependent cell death. MQC: mitochondrial quality control; ATP: adenosine 5’-triphosphate.

Mitochondrial fission

Initially perceived as static structures, the mitochondria are now well recognized as dynamic organelles constantly reshaped by fusion and fission processes. Mitochondrial fission is essential for the targeted removal of dysfunctional mitochondria in cardiomyocytes (Figure 2), with fission activity finely tuned according to the cell metabolic demands. Properly regulated fission generates new mitochondria, supporting the oxidative phosphorylation essential for myocardial development and function.[15] Moreover, fission enables the mitochondria to segregate damaged regions from the mitochondrial network, a crucial process for maintaining mitochondrial homeostasis in cardiomyocytes.[16]

Figure 2 DRP1 and its receptors, including MFF and FIS1, regulate mitochondrial fission. Increased mitochondrial fission is associated with oxidative stress, Ca2+ overload, mtDNA damage, and mitochondrial apoptosis activation. DRP1: dynamin-related protein 1; MFF: mitochondrial fission factor; FIS1: mitochondrial fission 1 protein; ROS: reactive oxygen species; ATP: adenosine 5’-triphosphate.
Figure 2

DRP1 and its receptors, including MFF and FIS1, regulate mitochondrial fission. Increased mitochondrial fission is associated with oxidative stress, Ca2+ overload, mtDNA damage, and mitochondrial apoptosis activation. DRP1: dynamin-related protein 1; MFF: mitochondrial fission factor; FIS1: mitochondrial fission 1 protein; ROS: reactive oxygen species; ATP: adenosine 5’-triphosphate.

Mitochondrial fission is primarily controlled by dynamin-related protein 1 (Drp1) and its receptors anchored in the OMM, including mitochondrial fission factor (Mff), mitochondrial fission 1 protein (Fis1), and mitochondrial dynamics proteins of 49 kDa (Mid49) and 51 kDa (Mid51).[17] Under physiological conditions, Drp1 predominantly resides in the cytoplasm in an inactive form that lacks binding affinity for OMM receptors, maintaining basal fission activity. However, Drp1 undergoes post-translational modifications (ubiquitination, acetylation, phosphorylation) under stress, which induce conformational changes.[18] These modifications expose the Drp1 binding sites, facilitating its translocation to the mitochondrial surface and enhancing its interaction with OMM receptors, which increases fission activity.

Notably, Drp1 transcription levels alone are unreliable indicators of its fission activity,[19] as mass spectrometry-based proteomics (PhosphoSitePlus) have revealed various post-translational modification sites.[20, 21] For example, Drp1 phosphorylation at Ser616 promotes its oligomerization around the OMM, a crucial step in forming the mitochondrial fission ring.[22] Conversely, phosphorylation at Ser637 inhibits Drp1 oligomerization, downregulating fission.[23] Post-translational modifications also affect Drp1 receptors: for example, Mff demonstrates enhanced Drp1 affinity when phosphorylated at Ser146, a modification associated with cardiac microvascular I/R injury.[24] The Fis1 N-terminal arm normally acts in an auto-inhibitory manner to limit Drp1 binding, but phosphorylation at this region enhances Drp1 interaction.[25] Although Mid49 and Mid51 modifications have been less studied, their roles in modulating Drp1-dependent fission merit further exploration.

Role of mitochondrial fission in cardiac I/R injury

Mitochondrial fission is closely linked to mitochondrial damage and cardiomyocyte death in cardiac I/R injury (Figure 2). Mechanistically, reduced phosphorylation of Drp1 at Ser637 leads to its mitochondrial localization, triggering excessive fission, calcium overload, and myocardial contractile dysfunction.[23] Conversely, Drp1 Ser616 phosphorylation is increased after I/R injury, elevating reactive oxygen species (ROS) production and oxidative stress in cardiomyocytes.[26] Enhanced expression and phosphorylation of Mff further promote fission in cardiac microvascular I/R injury, while Mff deletion preserves mitochondrial DNA (mtDNA) integrity and mitochondrial respiration, benefiting endothelial cell viability.[24, 27]

Excessive fission contributes to ATP depletion, cytochrome c (cyt-c) release, mitochondrial permeability transition pore (mPTP) opening, and cardiomyocyte apoptosis.[28] Mitochondrial fission also impairs antioxidant defenses, reducing superoxide dismutase 2 (SOD2) and heme oxygenase 1 (HO-1) levels, and suppresses autophagy, indicated by the lower expression of the LC3-phosphatidylethanolamine (LC3) II/I ratio, Beclin-1, and autophagy related 5/7 gene (ATG5/7).[29] Notably, the extent of the fission correlates with the myocardial infarction size and inversely with cardiac performance, highlighting its role in I/R injury.[29, 30] Pharmacological or genetic inhibition of mitochondrial fission has cardioprotective effects. The Drp1 inhibitor Mdivi-1 reduces Drp1 translocation to the mitochondria,[31] stabilizes membrane potential, prevents mPTP opening,[32, 33] and attenuates mitochondrial apoptosis pathways.[27] Additionally, Mdivi-1 modulates antioxidant activity, increasing manganese superoxide dismutase (SOD) and reducing malondialdehyde (MDA) levels, suggesting the role of fission in redox regulation.[31] Furthermore, fission affects key cardioprotective signaling pathways, including the protein kinase B (PKB), extracellular regulated protein kinases (ERK), adenosine 5’-monophosphate-activated protein kinase (AMPK), and nitric oxide (NO) pathways, which are upstream regulators of Drp1.[34, 35, 36]

However, timing and specificity are critical considerations. Pre-ischemic administration of Mdivi-1 is more effective than later interventions, as physiological fission may benefit cardiac function during ischemia, while pathological fission predominates post-ischemia.[37] Moreover, while inhibiting fission reduces apoptosis, it may inadvertently increase necroptosis, as seen in Mdivi-1-treated models.[27, 38] These insights indicate the need for careful interpretation of the effects of Mdivi-1 and suggest broader implications for targeted mitochondrial fission modulation in cardiac I/R injury.

Role of mitochondrial fission in myocardial infarction

Mitochondrial fission is markedly increased following myocardial infarction (MI) and is driven by hypoxia and oxidative stress. Hypoxia induces profound intracellular environmental changes, activating numerous signaling pathways that alter the expression and activity of key proteins in mitochondrial division and enhancing fission.[39] Elevated mitochondrial division often disrupts mitochondrial function, aggravating cardiomyocyte injury. Excessive fission fragments the mitochondrial network, impairing essential functions such as energy metabolism and molecular transport, which results in cellular energy deficits, metabolite accumulation, and accelerated cardiomyocyte damage and death.[40]

Drp1 is a pivotal mediator of mitochondrial fission, where its expression and activity are frequently upregulated during MI. Increased Drp1 promotes mitochondrial fragmentation, contributing to mitochondrial dysfunction. Drp1 activity in the ischemic myocardium is modulated by hypoxia, oxidative stress, and intracellular calcium fluctuations. For example, hypoxia activates specific signaling pathways that phosphorylate Drp1, increasing its affinity for mitochondrial membrane receptors and facilitating fission.[41, 42] The oxidative stress induced by MI exacerbates mitochondrial dysfunction, with hypoxia-driven redox imbalances generating ROS. ROS directly impair the mitochondrial structure and function and also activate additional signaling cascades that compromise mitochondrial homeostasis, upregulating Drp1 and accelerating fission.[43, 44] Additionally, Drp1 is key in mitochondrial calcium regulation; activated Drp1 translocates to the mitochondria, intensifying fission and ROS production in cardiomyocytes.

Apoptosis is also closely linked to mitochondrial fission, exacerbating cardiomyocyte damage. As central regulators of apoptosis, the mitochondria undergo morphological and functional alterations during apoptotic processes. Certain pro-apoptotic proteins, such as Bax, influence mitochondrial membrane permeability and may increase Drp1 activity, promoting mitochondrial fission and exacerbating cellular damage.[45]

Drp1 inhibitors, such as Mdivi-1, have been explored as therapeutic agents in MI. These inhibitors mitigate cardiomyocyte injury by inhibiting Drp1 activity and reducing mitochondrial division. Mdivi-1 binds to Drp1, preventing it from forming functional fission complexes, reducing mitochondrial fragmentation. Furthermore, Mdivi-1 reduced myocardial injury and improved cardiac function in MI models.[46, 47]

Antioxidants such as vitamins C and E have been investigated as treatments for MI, with evidence suggesting that they support cardiomyocyte function by reducing oxidative stress and improving mitochondrial health. These antioxidants scavenge intracellular ROS, alleviating oxidative damage to the mitochondria and preserving mitochondrial function.[48] Additionally, natural compounds such as ginsenosides and tanshinones have been studied for their protective effects in MI, with results indicating that they improve cardiomyocyte function by reducing oxidative stress, suppressing inflammation, and enhancing

mitochondrial function.[49, 50, 51, 52]

Role of mitochondrial fission in hypertension

Hypertension is a complex disease commonly accompanied by severe complications affecting the vascular systems of the heart, brain, and kidneys, presenting a significant threat to human health. Endothelial damage and vascular wall thickening are key contributors to hypertension onset and progression.[53]

Mitochondrial fission is essential to cellular homeostasis and is primarily regulated by a set of specialized proteins. Drp1 is a central fission regulator that exists in the cytosol in inactive form under physiological conditions. Upon receiving activation signals, Drp1 translocates to the OMM, where it oligomerizes into a helical structure that constricts the mitochondria, leading to their division.[54] Fis1 is located on the OMM and facilitates this process by recruiting Drp1.[55]

Abnormal mitochondrial fission has been observed within endothelial cells in hypertensive conditions, contributing to mitochondrial dysfunction and reduced NO production. NO is a crucial vasodilator, and its diminished availability under hypertensive conditions contributes to increased vascular tone. Drp1 upregulation in hypertensive endothelial cells may be triggered by oxidative stress and inflammatory signals, which disrupt normal mitochondrial structure and function.[56] In spontaneously hypertensive rats (SHR), Drp1 expression was markedly elevated in vascular endothelial cells, accompanied by thickening of the thoracic aorta medial layer and increased inflammatory factor expression.[53]

Mitochondrial fission is also intensified in vascular smooth muscle cells (VSMCs) during hypertension, increasing fragmented mitochondria. These fragmented mitochondria exhibit altered calcium handling and elevated ROS production. The increased ROS activates signaling pathways that drive VSMC contraction and proliferation, promoting vascular wall thickening and elevating blood pressure.[57] Angiotensin II (AngII) stimulation enhances Drp1 phosphorylation at Ser616, prompting its translocation to the mitochondria and initiating mitochondrial fission. This shift transforms VSMCs from a contractile phenotype to a synthetic, proliferative, and migratory state, actively participating in the vascular remodeling associated with hypertension. The mitochondrial morphology in VSMCs was notably fragmented after AngII treatment, with reduced branch length and an increase in mitochondrial count.[58]

Endothelin-1 (ET-1) is a potent endogenous vasoconstrictor implicated in hypertension pathogenesis and induces ROS-dependent activation of Rho-associated protein kinase (ROCK) signaling.[59] ROCK activation promotes mitochondrial fission, and the ROCK inhibitor Y-27632 alleviated ET-1-induced vasoconstriction and inhibited ET-1-induced mitochondrial fragmentation in rat aortic smooth muscle cells.[60]

Pharmacological agents targeting mitochondrial fission-related proteins hold therapeutic potential in hypertension. Compounds such as Mdivi-1 (a Drp1 inhibitor) and Y-27632 (a ROCK inhibitor) may counteract excessive Drp1 activation, restoring mitochondrial function in vascular and renal cells and supporting blood pressure regulation.[31, 58, 60]

Role of mitochondrial fission in DCM

Mitochondrial fission is markedly dysregulated in DCM. Mitochondrial fission becomes excessive in high glucose-induced H9c2 cardiomyoblasts, accompanied by lipid accumulation, oxidative stress, and apoptosis.[61] These results suggest that abnormal mitochondrial fission is closely associated with myocardial cell injury and may be central in DCM pathogenesis. Diabetic mouse models exhibit similar disruptions in mitochondrial structure and function, including increased fission, decreased fusion, reduced membrane potential, elevated ROS levels, increased apoptosis, and impaired cardiac function.[62]

Enhanced mitochondrial fission critically affects cardiomyocyte function by promoting mitochondrial fragmentation, which disrupts energy metabolism as the mitochondria are the primary source of ATP through oxidative phosphorylation. Additionally, excessive mitochondrial fission is linked to increased apoptosis, characterized by the upregulation of pro-apoptotic proteins and downregulation of anti-apoptotic proteins.[35, 61] Drp1 is a key mediator of mitochondrial fission whose expression and phosphorylation in DCM are altered under high-glucose conditions, exacerbating mitochondrial fragmentation. Inhibiting Drp1, such as through the inhibitor Mdivi-1, reduces fission and mitigates myocardial cell damage. Other fission-related proteins, such as Mff and Fis1, may also contribute, with signaling pathways such as protein kinase B-mammalian target of rapamycin (Akt-mTOR) and glycogen synthase kinase-3β (GSK-3β) playing regulatory roles. For example, nimbolide activates the Akt-mTOR pathway, inhibiting mitochondrial fission and ameliorating DCM, while perillaldehyde modulates fission by upregulating miR-133a-3p to inhibit GSK-3β expression.[62, 63]

Pharmacological studies have highlighted promising therapeutic avenues. Curcumin-conjugated gold nanoclusters (AuCur) demonstrated potential in reducing lipid accumulation, ROS levels, and mitochondrial fission under hyperlipidemic conditions, possibly through peroxisome proliferators-activated receptors α (PPARα) regulation.[64] Other compounds, such as nimbolide and perillaldehyde, exhibit protective effects in animal models and cellular assays.[63] Molecular mechanistic research has also identified novel targets, such as m6A-mediated phase separation affecting Notch1 expression, which inhibited mitochondrial fission and provided a new therapeutic strategy for diabetic cardiac fibrosis.[65]

Role of mitochondrial fission in drug-induced cardiomyopathy

The heart is an energy-intensive organ reliant on the mitochondria to sustain its function through ATP production via oxidative phosphorylation. The mitochondria are dynamic organelles that continually undergo fusion and fission to preserve their morphology, size, and functionality. However, this balance is often disrupted in drug-induced cardiotoxicity, leading to mitochondrial dysfunction and subsequent cardiomyocyte injury. Under physiological conditions, mitochondrial fission is crucial in MQC by facilitating the removal of damaged or dysfunctional mitochondria via mitophagy, thereby maintaining the overall health of the mitochondrial network. Controlled fission also contributes to key cellular signaling pathways associated with metabolic adaptation and survival, allowing cardiomyocytes to respond to fluctuating energy demands and environmental stressors.[66]

Doxorubicin is a widely used and potent chemotherapeutic agent limited by its severe cardiotoxic effects, which manifest as progressive cardiomyopathy. Evidence indicates that doxorubicin-induced cardiotoxicity is closely associated with increased ROS production, leading to oxidative stress and mitochondrial damage. This damage is frequently characterized by excessive mitochondrial fragmentation, a process tightly linked to mitochondrial fission. Abnormal fission in doxorubicin-treated cardiomyocytes appears to be mediated by Drp1, whose phosphorylation at specific sites promotes its translocation to the mitochondria and drives fission. This dysregulated mitochondrial fragmentation facilitates cyt-c release and caspase activation, culminating in cardiomyocyte apoptosis.[67, 68, 69]

Beyond doxorubicin, other chemotherapeutic agents have been similarly implicated in mitochondrial fission-mediated cardiotoxicity. For example, cisplatin disrupts mitochondrial dynamics, enhancing fission and compromising mitochondrial function, which contributes to cardiomyocyte injury and potentially to heart failure. Similarly, 5-fluorouracil is associated with mitochondrial dysfunction and oxidative stress, potentially mediated by alterations in mitochondrial fission and fusion.[70, 71, 72]

Notably, mitochondrial fission-induced cardiotoxicity is not confined to chemotherapeutics. Certain non-chemotherapeutic agents, including specific antipsychotic drugs, have also been linked to increased risk of cardiomyopathy, potentially through mechanisms involving disrupted mitochondrial fission and altered mitochondrial morphology.[73]

Mitochondrial fusion

Mitochondrial fusion is the converse of fission and is a dynamic process involving the sequential merging of the outer and inner membranes of individual mitochondria to form elongated, interconnected networks.[74, 75] This structural integration facilitates mitochondrial communication and resource sharing, allowing damaged or smaller mitochondrial fragments to merge with healthier mitochondria, supporting mitochondrial repair and functional resilience. Fusion is a protective mechanism that aids in maintaining mitochondrial homeostasis, particularly under stress conditions.[76]

Mechanistically, fusion is regulated by two key guanosine triphosphate hydrolases (GTPases): mitofusin 1 and 2 (MFN1/2), localized on the OMM, and optic atrophy protein 1 (Opa1), which governs inner mitochondrial membrane (IMM) fusion.[77] Fusion begins with MFN1/2 dimerization on adjacent mitochondria, which facilitates tethering with the assistance of ATP and F-actin. Subsequent OMM fusion is promoted by the hydrolysis of cardiolipin, a critical phospholipid within the mitochondrial membrane bilayer.[78] IMM fusion, regulated by Opa1, completes the process, although understanding of this step remains incomplete at the mechanistic level (Figure 3).

Figure 3 Mitochondrial fusion is orchestrated by MFN2, which resides in the OMM, and OPA1, found in the IMM. These proteins are crucial for increasing mitochondrial fusion, inhibiting mitochondrial fission, maintaining mitochondrial membrane potential, promoting mitochondrial bioenergetics, and preventing mitochondrial apoptosis, collectively preserving mitochondrial integrity and function. MFN2: mitofusin 2; OPA1: optic atrophy protein 1; OMM: outer mitochondrial membrane; IMM: inner mitochondrial membrane.
Figure 3

Mitochondrial fusion is orchestrated by MFN2, which resides in the OMM, and OPA1, found in the IMM. These proteins are crucial for increasing mitochondrial fusion, inhibiting mitochondrial fission, maintaining mitochondrial membrane potential, promoting mitochondrial bioenergetics, and preventing mitochondrial apoptosis, collectively preserving mitochondrial integrity and function. MFN2: mitofusin 2; OPA1: optic atrophy protein 1; OMM: outer mitochondrial membrane; IMM: inner mitochondrial membrane.

Mitochondrial fusion confers cytoprotective benefits by counterbalancing fission and mitigating fission-induced apoptosis. Mitochondrial fusion supports a continuous electrochemical gradient across the mitochondrial network, enhancing the ability of the mitochondrial pool to identify and respond to localized damage.[79] Additionally, fusion ensures the even distribution of mitochondrial components, including proteins, lipids, metabolites, and mtDNA, which collectively aid in alleviating localized stress responses and restoring mitochondrial equilibrium.[80]

Pathological conditions, such as myocardial I/R, disrupt mitochondrial fusion. MFN1, MFN2, and Opa1 levels are significantly downregulated in reperfusion models, correlating with reduced mitochondrial integrity, a shortened replicative lifespan, and increased apoptotic rates in endothelial cells.[81] The loss of fusion capacity in such contexts exacerbates mitochondrial fragmentation, ATP depletion, and oxidative stress, further impairing cardiomyocyte viability. Therapeutic strategies aimed at preserving or enhancing mitochondrial fusion may therefore potentially restore mitochondrial homeostasis and protect against mitochondrial-driven cellular damage in cardiovascular disease.

Role of mitochondrial fusion in cardiac I/R injury

While mitochondrial fusion is generally considered protective under physiological conditions, its role in cardiac I/R injury remains debated. MFN1 and Mfn2 are key OMM fusion regulators that appear to have distinct functions in cardiomyocytes. MFN1 deficiency in mice influences cardiac function minimally,[82, 83] whereas mtDNA damage and fragmentation were increased in Mfn2-null hearts.[84] Cardiomyocyte-specific MFN1 knockout (MFN1-KO) models retain respiratory function and demonstrate enhanced resistance to mitochondrial depolarization and oxidative stress, suggesting a protective effect against fission-induced cell injury.[85] Conversely, Mfn2 loss promotes mPTP opening, elevates ROS production, and sensitizes cardiomyocytes to apoptosis.[86, 87] In hypoxia-reoxygenation models, Mfn2 silencing exacerbates apoptosis, which caspase-9 inhibition or Bcl-x (L) overexpression reversed, underscoring its distinct regulatory role.[88] Additionally, MFN1/Mfn2 double knockout leads to the accumulation of defective mitochondria, hinting at an unexplored role of Mfn2 in MQC.[89, 90]

Unlike MFN1/2, the role of Opa1 in cardioprotection is well documented. Opa1 knockdown increases mitochondrial heterogeneity and promotes ventricular dilation with impaired contractile function.[91] Opa1 expression is decreased in myocardial I/R, and its genetic activation suppresses mitochondrial fission and apoptosis.[92] Mechanistically, overlapping activity with m-AAA protease-1 (OMA1)-mediated cleavage of Opa1 during reperfusion leads to fragmentation, cyt-c release, and apoptosis.[93, 94] Furthermore, dysregulating Opa1 impairs mitochondrial bioenergetics and exacerbated oxidative stress, further compromising cardiomyocyte survival.[95] Additionally, Opa1 promotes fatty acid oxidation, reducing ROS generation and preserving mitochondrial structure in heart failure, although this has not been confirmed in I/R models.[96, 97]

MFN1/2 and Opa1 undergo post-translational modifications that modulate fusion activity. The phosphorylation of MFN1 at Ser86 by protein kinase C beta II (βIIPKC) impairs fusion and leads to mitochondrial fragmentation,[98] while other kinases, including mitogen-activated protein kinase (MAPK) and ERK, phosphorylate MFN1 at Tyr562, reducing its oligomerization efficiency and increasing susceptibility to apoptosis.[99] Mfn2 phosphorylation by PINK1 facilitates mitophagy, while stress-induced phosphorylation and degradation by c-Jun N-terminal kinase (JNK) hinder fusion and promote cell death.[100] Although the post-translational regulation of MFN1/2 in I/R is not fully elucidated, MFN1/2 protein levels are both significantly downregulated. Opa1 stability is predominantly controlled by the redox-sensitive proteases OMA1 and ATP-dependent zinc metalloprotease (Yme1L), which are activated under stress to degrade Opa1 during I/R.[96] ROS scavenging extends Opa1 stability,[101] while signal transducer and activator of transcription 3 (STAT3) and RelA transcriptional regulation may influence Opa1 expression, although this has not been fully validated in I/R.[102] Furthermore, Opa1 is subject to acetylation and O-linked-β-N-acetylglucosamine (O-GlcNAcylation) in response to stress, impairing its GTPase activity and contributing to mitochondrial dysfunction and cell death.[103, 104]

Therapeutic strategies to restore mitochondrial fusion have been promising. Sevoflurane postconditioning reduces cardiac I/R injury by upregulating Opa1 and Mfn2, while vagal nerve stimulation improves mitochondrial dynamics in ischemic myocardium.[81] Epigallocatechin gallate stabilizes Opa1 by inhibiting OMA1 degradation, and melatonin enhances Opa1 expression via the AMPK pathway, increasing mitochondrial resistance to I/R injury.[94] These findings underscore the therapeutic potential of targeting Mfn2 and Opa1 to preserve mitochondrial fusion and protect cardiomyocytes during I/R injury.

Role of mitochondrial fusion in MI

During MI, mitochondrial morphology shifts dramatically from an elongated, networked structure to a fragmented form, likely due to an imbalance between mitochondrial fusion and fission. Impaired fusion reduces the mitochondrial capacity to maintain structural and functional connectivity, which is evident in ischemic cardiomyocyte models where mitochondrial fragmentation correlates with cellular damage severity.[97] Dysfunctional fusion disrupts mitochondrial functions, impairing energy metabolism, increasing ROS production, and leading to calcium imbalance. Normal fusion preserves mitochondrial membrane potential stability, supports respiratory chain complex assembly, and maintains the ATP generation essential for the high energy demands of cardiomyocytes. However, disrupted fusion during MI impairs oxidative phosphorylation, leading to ATP deficiency, while excessive ROS production further damages the mitochondria and cardiomyocytes.[105]

Recent studies have suggested that Notch1 signaling influences mitochondrial fusion by modulating Mfn2 expression, enhancing cardiomyocyte resilience to ischemic injury. Activation of the Notch1 intracellular domain (NICD) translocates it to the nucleus, where it binds transcription factors to regulate downstream genes, potentially including Mfn2.[106] Furthermore, Notch1 and MAPK signaling exhibit mutual regulation, with MAPK-linked phosphorylation of mitochondrial fusion proteins affecting fusion capacity.[107] AMPK is activated under MI-induced energy stress and also regulates fusion by phosphorylating Mfn2, enhancing mitochondrial fusion and preserving mitochondrial morphology.[108] Silent mating type information regulation 2 homolog-1 (SIRT1) and SIRT3 modulate mitochondrial fusion through acetylation status, with SIRT3 deacetylating OPA1 to promote its GTPase activity, facilitating fusion and maintaining mitochondrial structure and function.[109]

Several pharmacological interventions have demonstrated promise in modulating mitochondrial fusion to improve MI outcomes. The mitochondrial fission inhibitor Mdivi-1 indirectly promotes fusion by inhibiting Drp1 GTPase activity, reducing infarct size and enhancing cardiac function in myocardial I/R models. However, Mdivi-1 specificity and clinical viability are uncertain, with concerns about off-target effects and unknown human pharmacokinetics.[106] Melatonin has multi-target effects and activates the Notch1-Mfn2 pathway, promoting mitochondrial fusion and NICD and Mfn2 expression, and has antioxidant properties by scavenging free radicals, inhibiting lipid peroxidation, and supporting mitochondrial biogenesis.[57] Resveratrol activates the SIRT1-SIRT3-Mfn2-Parkin-peroxisome proliferators-activated receptor γ coactivator α (PGC1α) pathway, promoting fusion, autophagy, and biogenesis, thereby reducing infarct size in myocardial I/ R models. However, its limited bioavailability presents a challenge.[110] Isosteviol sodium supports fusion by inhibiting fission protein activity, preserving membrane potential, reducing ROS production, and preventing apoptosis in cellular and animal models, although its efficacy and safety should be confirmed with clinical data.[111] Targeted mitochondrial antioxidants such as MitoQ, which accumulate within the mitochondria to clear ROS and protect fusion proteins, remain in the early clinical stages.[112]

Despite advances in targeting mitochondrial fusion for cardioprotection, significant challenges remain in developing clinically viable therapies, necessitating further research into specificity, dosing, safety, and efficacy for optimal therapeutic outcomes.

Role of mitochondrial fusion in hypertension

Mitochondrial fusion is a finely regulated process essential for cellular homeostasis.[113] In mammalian cells, fusion is primarily mediated by OPA1 and MFN1/2.[114] These proteins enable dynamic mitochondrial fusion and division, maintaining a balanced mitochondrial network. OPA1 is localized to the IMM and is central in fusion, regulating cristae integrity and energy efficiency. OPA1 promotes IMM fusion through polymeric complex formation, and its expression and functional status directly influence fusion efficiency.[115] MFN1 and MFN2 are embedded in the OMM and facilitate membrane tethering by forming homotypic or heterotypic dimers essential for mitochondrial material exchange.[116] MFN1/2 activity is further modulated by post-translational modifications: acetylation at specific sites on MFN1 (K222, K491) reduces its GTPase activity, while phosphorylation by the MEK-ERK pathway at T562 impairs oligomerization.[99] Additionally, stress-related truncation at Ser27 reduces MFN2 degradation, promoting elongation and preventing apoptosis.[117]

Efficient mitochondrial fusion supports intracellular energy transfer and metabolic stability, crucial for cell function. Hypertension disrupts this balance, leading to mitochondrial dysfunction, as shown in animal models and clinical studies. Significant mitochondrial abnormalities in SHR, such as swelling, disrupted cristae, and reduced matrix density, indicate impaired mitochondrial function.[118] Hypertension also alters fusion-related protein expression; specifically, reductions in OPA1 and MFN2 lead to fragmented mitochondrial networks, hindering normal metabolic exchange. Clinical studies in hypertensive patients have revealed similar mitochondrial dysfunctions in vascular smooth muscle and cardiomyocytes. Genetic variations in the fusion-related genes may contribute to hypertension pathogenesis by affecting mitochondrial dynamics, intracellular calcium signaling, and ROS production. For example, the OPA1 single-nucleotide polymorphism (SNP) rs7646250 may alter Forkhead Box A2 (FOXA2) transcription factor binding, enhancing mitochondrial fusion and conferring increased resilience against hypertension.[119] Similarly, the PARK2 SNP rs6902041 has been associated with blood pressure regulation.[120]

Therapeutically, several drugs targeting mitochondrial fusion show potential for hypertension management. O-(3-piperidino-2-hydroxy-1-propyl)-nicotinic amidoxime (BGP-15) enhances fusion, inhibits fission, and promotes mitochondrial biogenesis. BGP-15 improved mitochondrial morphology and cardiac function in hypertension-induced heart failure models by upregulating fusion proteins (OPA1, MFN2) and downregulating fission proteins (DRP1, MFF).[121] Acacetin similarly augments mitochondrial fusion via phosphatidylinositol 3-kinase (PI3K)-Akt signaling, elevating MFN2 levels and suppressing DRP1 and MFF expression, protecting against hypertension-induced cardiac damage.[122]

In summary, mitochondrial fusion integrity is critical in hypertension pathophysiology, influencing cellular energy balance, oxidative stress, and apoptosis, and ultimately affecting organ health. Targeting mitochondrial fusion offers a promising therapeutic strategy for hypertension. However, further studies are needed to elucidate the precise regulatory pathways, optimize fusion-targeting therapies, and establish the long-term safety of these interventions. Continued research may unveil effective, fusion-focused treatment strategies, providing novel options for hypertension management and prevention.

Role of mitochondrial fusion in DCM

Mitochondrial fusion is a critical repair process for mildly damaged mitochondria and consists of two key stages: OMM fusion and IMM fusion, which are facilitated by MFN1/2 and Opa1, respectively. During fusion initiation, MFN1/2 on the OMM of adjacent mitochondria interact to stimulate OMM fusion in a GTP-dependent process.[123] Subsequently, Opa1 promotes IMM fusion, with long (L-Opa1) and short (S-Opa1) isoforms working synergistically.[124, 125] The loss of Opa1 impairs fusion capacity and also induces mitochondrial fragmentation and disrupts cristae structure, underscoring its role in mitochondrial integrity. Increased S-Opa1 and reduced L-Opa1 were observed as early as week 5 in diabetic mouse cardiomyocytes, indicating enhanced fission and reduced fusion, which correlated with vacuolar mitochondria and suggested the importance of early intervention.[126]

Mfn2 is pivotal in endoplasmic reticulum (ER)-mitochondrial interactions, which are essential for Ca2+ signaling. Hyperglycemia reduces Mfn2 expression, disrupting ER-mitochondria communication and resulting in mitochondrial Ca2+ overload, ROS accumulation, mPTP opening, and the activation of caspase-dependent apoptotic pathways, leading to myocardial dysfunction.[127] Increased fission and reduced Opa1 expression, coupled with elevated O-GlcNAc glycosylation, drive mitochondrial dysfunction in type 1 DCM.[126]

Several therapeutic agents have demonstrated potential in modulating mitochondrial fusion to counteract DCM. Sodium-glucose cotransporter 2 (SGLT2) inhibitors, such as empagliflozin, inhibited excessive fission and upregulate MFN1 and Opa1 in diabetic mouse cardiomyocytes, improving mitochondrial morphology, reducing myocardial fibrosis, and attenuating cardiac hypertrophy. AMPK activation may mediate this effect by suppressing division-related proteins and promoting fusion.[123, 128] Similarly, glucagon-like peptide-1 (GLP-1) receptor agonists, such as liraglutide, enhanced mitochondrial fusion and autophagy in diabetic models by activating the mTOR-UNC-51-like kinase 1 (ULK1) and AMPK pathways, improving cardiac function and slowing DCM progression.[129, 130]

Melatonin is a natural antioxidant that promotes mitochondrial fusion, biogenesis, and autophagy in diabetic rat cardiomyocytes through the SIRT6-AMPK-PGC1α-Akt pathway, maintaining MQC and alleviating DCM.[131, 132] Paeonol enhanced mitochondrial fusion under hyperglycemic conditions via the casein kinase 2α (CK2α)-Stat3-Opa1 pathway, preventing mitochondrial oxidative stress and preserving respiratory and cardiac function by sustaining Opa1 expression and promoting mitochondrial integrity.[133]

In summary, mitochondrial fusion is essential in cellular homeostasis, and its dysregulation in conditions such as hypertension and DCM highlights its therapeutic potential. Targeting fusion through pharmacological modulation offers a promising approach to managing these cardiovascular diseases, although further studies are needed to elucidate the precise mechanisms, optimize fusion-targeting therapies, and assess their clinical viability for long-term management of DCM and related pathologies.

Role of mitochondrial fusion in drug-induced cardiomyopathy

Doxorubicin (DOX) is a widely used chemotherapeutic agent associated with significant cardiotoxicity, which is largely attributed to mitochondrial fusion disruptions. This cardiotoxicity manifests at both cellular and subcellular levels, exacerbating cardiac dysfunction. Under normal conditions, cardiomyocyte mitochondria form an interconnected tubular or reticular network, supporting efficient energy exchange and material transport essential for cellular homeostasis. However, DOX exposure induces mitochondrial fragmentation, characterized by shortened, dispersed mitochondria that disrupt network integrity, impairing cellular energy metabolism and material transport.[134]

Impaired fusion compromises mitochondrial function at the subcellular level. Following DOX treatment, decreased mitochondrial membrane potential hinders oxidative phosphorylation, reducing ATP production. Furthermore, inhibited fusion elevates ROS levels and disrupts calcium homeostasis, collectively impairing cardiomyocyte contractility and promoting cardiac dysfunction.[135] Trastuzumab is another cardiotoxic drug that similarly downregulates MFN1/2, reducing mitochondrial fusion and contributing to cardiotoxicity in treated patients.[136, 137] Mitochondrial fusion proteins are central in this cardiotoxicity. Mfn2 is essential for both mitochondrial fusion and ER-mitochondria interactions and is downregulated by DOX, disrupting calcium and lipid homeostasis and exacerbating cardiomyocyte injury.[138] Opa1 is responsible for IMM fusion and cristae maintenance, undergoes altered proteolytic processing and post-translational modification in response to DOX, further compromising mitochondrial structure and energy metabolism.[139]

Signaling pathways such as PI3K-Akt and p53 also modulate mitochondrial fusion during cardiotoxicity. DOX inhibits PI3K-Akt, reducing phosphorylation of the downstream GSK-3β, which promotes mPTP opening, decreases membrane potential, and induces cyt-c release and apoptosis.[50] Additionally, DOX activates the p53 pathway, which transcriptionally regulates fusion and fission proteins; p53 activation inhibits Mfn2 and upregulates Drp1, driving excessive fission, apoptosis, and cardiac dysfunction.[140]

Therapeutic interventions targeting mitochondrial fusion demonstrate potential for mitigating DOX-induced cardiotoxicity. Shenmai injection is used clinically for cardiovascular support and alleviates DOX-induced toxicity by activating the PI3K-Akt and AMPK pathways, increasing Mfn2 expression, and promoting fusion. Studies on DOX-treated models demonstrated that Shenmai injection reduced apoptosis, oxidative stress, and mitochondrial fragmentation, ultimately preserving cardiac function.[50] Mitochondria-targeted antioxidants, such as MitoQ, directly scavenge mitochondrial ROS, mitigating oxidative damage and preserving mitochondrial fusion and structural integrity, although their efficacy should be verified in further clinical studies.[112] Exosomes derived from human trophoblast stem cells (TSC-Exos) have emerged as promising therapeutic agents. TSC-Exos increase Mfn2 expression, reduce DOX-induced fission, and decrease cardiomyocyte apoptosis rates, as shown by improved cardiac function and reduced fibrosis in DOX-treated mouse models.[141]

In summary, mitochondrial fusion disruptions are critical drivers of cardiotoxicity. Comprehensive understanding of the mechanisms governing mitochondrial fusion and its dysregulation in drug-induced cardiotoxicity can aid in developing targeted therapeutic strategies. Currently, specific drugs aimed at restoring mitochondrial fusion in cardiomyocytes are lacking, and further exploration is required to optimize combinatory use with cardiotoxic treatments.

Mitophagy

Mitophagy is a selective autophagic process targeting damaged mitochondria for degradation and is crucial for maintaining cellular homeostasis, especially in cardiac I/R injury. As with mitochondrial fission and fusion, mitophagy initiation and completion depend on specific adaptor proteins, including Parkin, Bnip3, Fundc 1, and Nix (Figure 4). These adaptors operate via receptor-dependent or-independent pathways depending on their cellular localization. For example, Bnip3, Fundc1, and Nix are anchored on the OMM and facilitate receptor-dependent mitophagy, whereas Parkin, residing in the cytoplasm, translocates to the mitochondria upon activation, initiating receptor-independent mitophagy.[142, 143]

Figure 4 Mitophagy is facilitated through receptor-independent pathways, with key roles played by PARKIN and FUNDC1. Mechanistically, targeted mitochondria are engulfed by autophagy precursors, leading to autophagosome. Thereafter, LC3 conjugates with phosphatidylethanolamine to form LC3II. Ultimately, lysosomes mediate the hydrolytic degradation of autophagosomal contents, including proteins, nucleic acids, carbohydrates, and lipids, which are recycled by the cell to reinstate cellular homeostasis. FUNDC1: Fun14 domain-containing protein 1; BINP3: B-cell lymphoma-2 (BCL2)/adenovirus E1B 19 kDa protein-interacting protein 3; PINK1: PTEN induced putative kinase 1; LC3: microtubule-associated protein 1A/1b-light chain 3.
Figure 4

Mitophagy is facilitated through receptor-independent pathways, with key roles played by PARKIN and FUNDC1. Mechanistically, targeted mitochondria are engulfed by autophagy precursors, leading to autophagosome. Thereafter, LC3 conjugates with phosphatidylethanolamine to form LC3II. Ultimately, lysosomes mediate the hydrolytic degradation of autophagosomal contents, including proteins, nucleic acids, carbohydrates, and lipids, which are recycled by the cell to reinstate cellular homeostasis. FUNDC1: Fun14 domain-containing protein 1; BINP3: B-cell lymphoma-2 (BCL2)/adenovirus E1B 19 kDa protein-interacting protein 3; PINK1: PTEN induced putative kinase 1; LC3: microtubule-associated protein 1A/1b-light chain 3.

Mechanistically, receptor-dependent mitophagy relies on direct interactions between these adaptors and LC3, a key autophagic protein. The N-terminal regions of Bnip3, Fundc1, and Nix contain LC3-interacting regions (LIRs), which enable binding to LC3. This interaction converts LC3I into its phosphatidylethanolamine-conjugated form, LC3II, a critical step in autophagosome formation.[144] Contrastingly, Parkin-mediated mitophagy operates via the PINK1-Parkin pathway. Upon mitochondrial depolarization, PINK1 stabilizes on the OMM, phosphorylating Parkin at Ser65, which triggers Parkin translocation to the mitochondria. Parkin subsequently ubiquitylates numerous OMM proteins, creating ubiquitin chains that recruit LC3 receptors such as prohibitin2 (PHB2), promoting autophagosome formation around damaged mitochondria.[145]

Post-translational modifications, such as phosphorylation, tightly regulate these adaptor-LC3 interactions, ensuring controlled mitophagy activation. For example, Fundc1 phosphorylation at Ser13 by CK2 enhances its binding affinity to LC3, facilitating mitophagy under stress conditions.[146] Similarly, Bnip3 phosphorylation at Ser17 by JNK promotes LC3 interaction,[147] while PINK1-induced phosphorylation of Parkin at Ser65 is essential for its mitochondrial binding and mitophagy initiation.[148]

While mitophagy is generally protective, excessive or dysregulated mitophagy can be maladaptive, leading to cellular energy depletion and cardiomyocyte death. Research has identified three primary mechanisms through which mitophagy may drive cell death. First, selective removal of damaged mitochondria under mild stress supports cell survival. However, severe stress induces widespread mitochondrial damage that overwhelms the mitophagic capacity, resulting in cell death as mitophagy fails to restore mitochondrial homeostasis. Second, excessive mitophagy can deplete mitochondrial mass, impairing ATP production and predisposing cells to necroptosis rather than ATP-dependent apoptosis. Finally, adaptors such as Bnip3 and Nix, which initiate mitophagy by linking LC3 to damaged mitochondria, are also involved in the apoptosis pathways. For example, Bnip3 overexpression sensitizes cells to intrinsic apoptosis, highlighting the fact that the role of mitophagy in cell survival or death often depends on adaptor activity and cell type.[148]

Therapeutic strategies targeting mitophagy regulation demonstrate potential for mitigating cardiac injury. For example, modulating Fundc1 or Bnip3 phosphorylation enhanced controlled mitophagy under ischemic conditions, balancing mitochondrial turnover without tipping towards excessive degradation.[149, 150] Furthermore, understanding cell type-specific mitophagy responses could enable more targeted cardioprotective interventions tailored to optimize MQC without compromising cellular energy reserves.

Role of mitophagy in cardiac I/R injury

Extensive studies have sought to clarify the role of mitophagy in maintaining myocardial function and cardiomyocyte survival in cardiac I/R injury (Figure 4). Current evidence suggests that the influence of mitophagy, whether protective or deleterious, depends largely on the specific adaptor proteins involved. For example, Opa1-mediated mitophagy exacerbates reperfusion-induced cardiomyocyte death due to calcium overload, whereas pharmacological activation of Opa1-induced mitophagy protects against I/R injury. Similarly, ablating Opa1 impaired mitophagy and increased I/R-mediated myocardial damage, underscoring its role in cardioprotection.[93] In contrast, Parkin-mediated mitophagy is harmful in reperfused hearts as it promotes cyclophilin D (CypD)-dependent mPTP opening, a hallmark of necroptosis.[151] Cardiac microvascular I/R injury studies have confirmed that Parkin-mediated mitophagy triggers excessive mitochondrial clearance and ATP depletion, signaling cell death in cardiac microvascular endothelial cells.[152] Bnip3-related mitophagy also exerts lethal effects, as functional abrogation of Bnip3 prevents mitophagy activation and mitigates necrotic cell death in cardiomyocytes.[153]

Conversely, cardiolipin-induced mitophagy appears cardioprotective, attenuating oxidative stress, reducing calcium overload, and enhancing cardiomyocyte survival during I/R injury.[154, 155] Similarly, Fundc 1 (an OMM protein regulated via post-transcriptional modifications) promotes protective mitophagy. Dephosphorylated Fundc1 facilitates mitophagy during ischemia, reversing mitochondrial membrane potential, reducing ROS overproduction, and inhibiting apoptosis in reperfused myocardium.[156, 157] The E3 ubiquitin ligase tumor necrosis factor receptor associated factor-2 (TRAF2) also initiates protective mitophagy, mitigating mitochondrial fragmentation in reperfused hearts.[158, 159]

Despite these advances, little is known about the molecular crosstalk among mitophagy adaptors in cardiac I/R injury, which complicates the assessment of the net effects of mitophagy. While some studies have indicated mitophagy activation during I/R,[151, 160] others have reported its inhibition.[93, 161] This discrepancy may stem from different reperfusion time points, as ischemia/ hypoxia triggers autophagy (mitophagy) initially.[162, 163] During reperfusion, autophagic flux declines within the early phase (0-24 h post-I/R) but increases during the later recovery stages (1-3 days post-I/R).[164, 165] Recent work using autophagy receptor reporter mice (CAG-RFP-EGFP-LC3) subjected to renal I/R injury observed minimal autophagy at 4 h post-reperfusion, followed by autophagosome-lysosome fusion from 1 to 3 days post-reperfusion.[166] These observations suggest that early suppression and later activation of mitophagy may constitute an adaptive protective response. In early reperfusion, when ROS surge and calcium overload precipitate significant cell death, cardiomyocytes may suppress mitophagy to avoid the potential activation of mitophagic cell death, which could exacerbate myocardial damage. In contrast, later-stage mitophagy may aid mitochondrial repair and enhance cardiomyocyte recovery.

Notably, mitophagy adaptors exhibit varied dynamics during reperfusion. For example, Parkin is upregulated,[152] whereas Fundc1 is rapidly inactivated in early reperfusion.[156] This suggests that the net mitophagy response involves complex interplay among adaptors, each with unique timing and roles. Furthermore, the mitophagy regulatory landscape is intricate, with multiple adaptors compensating for each other’s function. For example, germline deletion of Parkin did not eliminate mitophagy entirely, as mitochondrial E3 ubiquitin protein ligase 1 (Mul1) can substitute for Parkin-mediated mitophagy under physiological conditions.[167] Similarly, BCL2L13, the mammalian homologue of yeast Atg32, partially compensated for basal mitophagy in Atg32-deficient yeast,[168] and Mfn2 knockout-induced mitophagy deficiency was mitigated by nonselective autophagy activation.[169]

These compensatory mechanisms ensure sustained mitophagy across diverse cellular contexts, highlighting the complexity of MQC. Research should focus on elucidating the interactive networks and compensatory pathways among mitophagy adaptors in cardiac I/R injury, as understanding these interactions may present new avenues for therapeutic intervention.

Role of mitophagy in MI

Mitochondrial autophagy (mitophagy) undergoes complex dynamic changes following MI, which are initially activated in response to ischemia and hypoxia to remove damaged mitochondria and reduce cellular stress. Autophagy-related proteins such as LC3-II increase rapidly immediately post-infarction, indicating elevated mitophagy flux.[170] However, prolonged ischemia and hypoxia may lead to overactive mitophagy, overwhelming autophagic capacity and causing autophagosome accumulation and mitochondrial damage. This impairs MQC and exacerbates cell death.

MI induces marked alterations in mitochondrial morphology and function, characterized by fragmentation, swelling, and functional decline (reduced membrane potential, ATP depletion, increased ROS production). Dysfunctional mitochondria impair cellular energy homeostasis and also release pro-apoptotic factors, such as cyt-c, further driving cardiomyocyte apoptosis.[171] These mitochondrial changes are intricately linked to mitophagy: damaged mitochondria serve as mitophagy substrates, while excessive ROS from dysfunctional mitochondria can hinder autophagic processes, such as autophagosome-lysosome fusion, reducing autophagy efficiency.

The PINK1-Parkin pathway is pivotal in mitophagy regulation. Under normal conditions, PINK1 is imported into the mitochondria, cleaved by the presenilin associated rhomboid like Gene (PARL) protease, and degraded in the cytosol. During MI, mitochondrial depolarization inhibits PINK1 degradation, allowing its accumulation on the OMM. Activated PINK1 recruits Parkin, which ubiquitinates OMM proteins (voltage dependent anion channel 1 Gene [VDAC1], Mfn2), marking them for recognition by autophagy receptors (p62, optineurin, nuclear dot protein 52 [NDP52]) that mediate autophagosome formation.[172, 173] FUNDC1 is another key mitophagy receptor activated under hypoxia and is regulated by phosphorylation at Tyr18, Ser13, and Ser17. Hypoxic stress dephosphorylates FUNDC1, enhancing its LC3-binding affinity and promoting mitophagy.[174] Similarly, BNIP3 and BNIP3L/NIX are upregulated in hypoxia and ischemic conditions, where they interact with LC3 to drive mitophagy and influence mitochondrial membrane potential and calcium homeostasis.[175]

Beyond the PINK1-Parkin pathway, other mitophagy mechanisms are essential in the myocardial stress response. ULK1 is a central kinase in autophagy initiation that is activated post-infarction via AMPK-mediated phosphorylation independent of mTORC1 inhibition. ULK1 regulates mitophagy receptors, such as phosphorylating FUNDC1 to enhance LC3 interaction, and initiates autophagosome formation.[13, 176] Rab9 is a small GTPase that mediates an alternative mitophagy pathway by forming complexes with ULK1 and DRP1 under pathological conditions. Rab9 is involved in mitochondrial transport and localization, facilitating the delivery of damaged mitochondria to autophagosome assembly sites, which may open avenues for therapeutic intervention.[177, 178]

Several therapeutic strategies have demonstrated potential in targeting mitophagy for MI treatment. For example, melatonin inhibits excessive PINK1-Parkin-mediated mitophagy following ischemic injury, preserving cardiac function.[152] Natural compounds, such as berberine, modulate mitophagy via the hypoxia-inducible factor (HIF)-1α-BNIP3 pathway, attenuating myocardial I/R injury.[179] Gene therapy also presents opportunities: overexpressing mitophagy-promoting genes, such as Parkin and FUNDC1, improved mitochondrial clearance and enhanced cardiac recovery in animal models. However, challenges remain in optimizing gene delivery and ensuring safety.[180]

Mitochondrial autophagy in MI involves complex regulatory networks, balancing protective and potentially deleterious effects. While mitophagy-targeted therapies are promising, further research is necessary to elucidate the mechanistic nuances and optimize strategies for enhanced myocardial recovery.

Role of mitophagy in hypertension

Mitophagy undergoes substantial alterations during the progression of hypertension. Factors such as platelet-derived growth factor (PDGF) induce aberrant mitophagy in VSMCs, which drives VSMC proliferation and migration, ultimately affecting vascular structure and function. PDGF triggers mitochondrial calcium influx and activates Ca2+-calmodulin-dependent protein kinase II (CaMKII), increasing mitochondrial motility and fission while reducing the expression of Mfn2, a protein integral to mitochondrial fusion. These changes collectively promote VSMC proliferation and migration, key features of vascular remodeling in hypertension.[181, 182, 183] In renal cells, hypertension-induced mitophagy dysfunction contributes to pathological processes such as tubulointerstitial fibrosis, although the precise mechanisms remain under investigation.[184] Fine-tuning mitophagy may be neuroprotective in neurons with hypertension-related injury, underscoring tissue-specific mitophagy roles in hypertension.[185]

The PINK1-Parkin pathway is a molecular-level primary mechanism of mitophagy regulation. Under physiological conditions, PINK1 is rapidly degraded within the mitochondria, but accumulates on the OMM upon mitochondrial damage, where it recruits and activates Parkin. Parkin then ubiquitinates several OMM proteins, marking damaged mitochondria for degradation by recruiting autophagy receptors via LC3-interacting region (LIR) motifs, which facilitate autophagosome assembly.[184, 186] Other receptor-dependent mitophagy pathways involve the BNIP3, NIX, and FUNDC1 proteins, which bind LC3 through LIR motifs. BNIP3 is upregulated in hypoxia and promotes mitophagy while regulating ROS production.[175] Similarly, FUNDC1 undergoes dephosphorylation in response to hypoxia or mitochondrial depolarization, enhancing its interaction with LC3 to induce mitophagy.[150]

Signaling pathways are also crucial in mitophagy regulation within hypertensive contexts. The ras homolog family member A-Rho kinase (RhoA-ROCK) pathway is activated by agents such as apatinib and leads to VSMC dysfunction, characterized by heightened proliferation, migration, and anti-apoptotic capacity. Inhibition by ROCK inhibitors, such as Y27632, mitigates these effects, highlighting the significance of the pathway in hypertension-related VSMC behavior.[187] AngII, PDGF, hypoxia, and hyperglycemia influence mitochondrial dynamics and autophagy, affecting VSMC proliferation and migration. For example, AngII activates DRP1 to promote mitochondrial fission, facilitating VSMC proliferation and migration.[182]

Therapeutic strategies targeting mitophagy in hypertension have yielded promising results. The deubiquitinating enzyme inhibitor PR-619 enhanced Parkin-mediated mitophagy in experimental glaucoma models, protecting retinal ganglion cells by reducing ubiquitin-specific protease 15 (USP15) expression, which otherwise inhibits Parkin-dependent mitophagy.[188] In hypertensive rat models, resveratrol and regular exercise improved systolic blood pressure and cardiac function by modulating stress responses, i.e., oxidative stress, ER stress, nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome activation, and mitophagy. These interventions promoted mitochondrial function by upregulating antioxidative genes, relieving ER stress, inhibiting NLRP3 inflammasome activation, and increasing PINK1 and Parkin expression.[189] Furthermore, the ROCK inhibitor Y27632 and RhoA-ROCK pathway gene silencing (e.g., leukemia-associated Rho guanine nucleotide exchange factor [LARG] knockdown) have demonstrated potential in counteracting apatinib-induced VSMC dysfunction, offering a target for mitigating vascular injury associated with ROCK pathway dysregulation.[187]

Drug development targeting mitochondrial dynamics and mitophagy-related molecules also demonstrates therapeutic promise. The DRP1 inhibitor Mdivi-1 suppresses VSMC proliferation and migration, while upregulating Mfn2 or inhibiting miR-93 (which downregulates Mfn2) similarly limits VSMC proliferation and migration. Bioactive compounds, such as liraglutide and tanshinone IIA, modulate mitochondrial dynamics and mitophagy, providing a basis for potential therapeutic options for cardiovascular diseases linked to mitochondrial dysfunction.[182]

Role of mitophagy in DCM

The role of mitochondrial autophagy in DCM remains contentious. While some studies have reported that cardiac and mitochondrial autophagy are activated in DCM, others have suggested the opposite. These discrepancies may be due to differences in research methodologies, animal models, or diabetes progression stages. Cardiac autophagy in type 2 diabetes mellitus (T2DM)-associated DCM undergoes dynamic shifts: cardiac autophagy in a high-fat diet (HFD)-induced diabetic mouse model was initially activated (within 6 weeks) and subsequently declined, whereas mitochondrial autophagy persisted for approximately 2 months post-activation.[10]

The PINK1-Parkin pathway is essential for mitochondrial autophagy and is notably altered in DCM. Several studies have reported downregulation of PINK1 and Parkin downregulation in diabetic hearts, leading to impaired mitophagy. For example, PINK1 and Parkin expression in type 1 diabetes (T1DM) mouse model cardiomyocytes was reduced, decreasing mitophagic flux. Conversely, some studies have indicated that the PINK1-Parkin pathway may be activated in certain contexts, although its precise role in DCM warrants further investigation.[190] FUNDC1 is another mitophagy regulator that has a complex role in DCM progression: FUNDC1 dysfunction inhibits mitophagy, exacerbating cardiac impairment. However, other studies have suggested that reducing FUNDC1 expression alleviates mitochondrial calcium overload and may confer protection in diabetic heart disease, indicating a nuanced role for FUNDC1 in DCM.[174] Research on BNIP3 and NIX in DCM is limited, although increased mitochondrial ROS and inhibited BNIP3 expression have been reported in prediabetic models, implying a potential role for BNIP3 in DCM pathogenesis. Additionally, changes in SIRT3 expression and activity in DCM may alter the acetylation status of mitophagy-related proteins, modulating mitophagy. For example, SIRT3 deacetylates and regulates Parkin, affecting mitophagy and potentially influencing DCM progression.[191]

Several therapeutic agents have demonstrated promise in modulating mitophagy for DCM treatment. Canagliflozin is an SGLT2 inhibitor that traditionally lowers blood glucose by inhibiting renal glucose reabsorption. Canagliflozin treatment significantly improved cardiac function in T2DM mouse models, evidenced by increased left ventricular ejection fraction (LVEF) and shortening fraction (LVFS) and decreased left ventricular end-diastolic and end-systolic diameters (left ventricular internal diameter at end-diastole [LVIDd], LVID at end-systole [LVIDs], respectively). Mechanistically, canagliflozin enhances PINK1-Parkin-dependent mitophagy, increasing PINK1 and Parkin expression, LC3II levels, and mitochondrial respiratory capacity while reducing p62 accumulation and oxidative stress, preserving mitochondrial structure and function.[192] Empagliflozin is another SGLT2 inhibitor that normalizes mitochondrial size and density in diabetic hearts by activating the AMPKα1-ULK1-FUNDC1-mitophagy pathway.[193, 194] Metformin is a classic antidiabetic drug that enhanced cardiac autophagy and improved cardiac function in diabetic OVE26 mice with long-term use.[195] Additionally, coenzyme Q10 is a potent antioxidant that may aid in stabilizing mitochondrial function and mitigating oxidative stress-related mitochondrial damage in DCM treatment.[196]

Despite these advances, challenges remain in optimizing drug therapies targeting mitophagy for DCM. Key issues, such as determining optimal dosing, defining therapeutic windows, and assessing long-term safety, still require clarification through further research.

Role of mitophagy in drug-induced cardiomyopathy

DOX is a well-studied chemotherapeutic agent widely recognized for its potent anti-cancer efficacy and cardiotoxic effects. DOX-induced cardiotoxicity is attributed to mitochondrial damage, dysfunction, disrupted dynamics, excessive autophagy, and both the apoptosis and necrosis of cardiomyocytes, impairing cardiac function. DOX treatment downregulated Parkin expression in mouse hearts and H9c2 cardiomyoblasts, while Parkin overexpression activated mitophagy, reduced apoptosis, and mitigated DOX-induced cardiotoxicity.[197]

Yes-associated protein (YAP) influences Parkin transcriptional regulation. DOX treatment decreases YAP expression; conversely, exogenous YAP inhibits DOX-induced mitochondrial fragmentation and apoptosis, promotes autophagic flux, and upregulates Parkin expression by interacting with TEA domain transcription factor 1 (TEAD1), alleviating DOX-induced cardiotoxicity.[198] Additionally, DOX reduces TANK-binding kinase 1 (TBK1) phosphorylation levels, impairing cardiac function, increasing mortality, and promoting interstitial fibrosis. TBK1 mediates mitochondrial protection through SQSTM1/p62-dependent mitophagy, and its overexpression mitigates DOX-induced mitochondrial damage and cardiotoxicity. Conversely, knocking down TBK1 or inhibiting its phosphorylation exacerbates these deleterious effects.[199]

DOX also activates p53, which interacts with Parkin to inhibit its mitochondrial translocation, blocking mitophagy. This inhibition results in the accumulation of damaged mitochondria and consequent cardiotoxicity. Harpagoside (HAR) inhibits p53-Parkin binding, facilitates Parkin translocation to mitochondria, and restores mitophagy, alleviating DOX-induced cardiotoxicity without compromising the anti-cancer efficacy of DOX.[200]

Furthermore, DOX reduces plasma SIRT6 expression while elevating lactate levels. In vitro, SIRT6 overexpression reduces DOX toxicity in cardiomyocytes and enhances its anti-cancer effects. In vivo, SIRT6 overexpression ameliorates DOX-induced cardiac dysfunction and potentiates its tumor-suppressive activity. SIRT6 enhances mitochondrial biogenesis and mitophagy by inhibiting serum/glucocorticoid-regulated kinase 1 (SGK1), coordinating metabolic remodeling to protect cardiomyocytes from DOX-induced energy depletion.[201]

Mitochondria-dependent cell death

Heart injury is characterized by the rapid loss of functional cardiomyocytes through programmed cell death (PCD), a terminal mechanism of MQC. As a central determinant of cell fate, the mitochondria mediate cardiomyocyte death via two primary routes (Figure 5). The first pathway involves OMM hyperpermeabilization, facilitating cyt-c release into the cytoplasm. Cyt-c subsequently activates caspase-9, which cleaves and activates caspase-3, initiating classical mitochondria-dependent apoptosis.[202, 203] This apoptotic pathway is marked by mitochondrial membrane potential decline, excessive ROS generation, upregulated Bax, and downregulated Bcl-2.[204, 205]

Figure 5 Mitochondrial cell death encompasses apoptosis and necrosis. Apoptosis is regulated by OMM permeabilization, mitochondrial membrane potential reduction, BAX activation, and caspase-9. Necrosis is triggered by RIPK3-MLKL-CAMKII pathway activation and mPTP opening. Subsequently, dysfunction of the mitochondrial electron transport chain and termination of the TCA cycle lead to ATP depletion, cytosolic swelling, and membrane rupture. OMM: outer mitochondrial membrane; RIPK3: receptor-interacting protein kinase 3; MLKL: mixed lineage kinase domain-like protein; mPTP: mitochondrial permeability transition pore; CaMKII: Ca2+-calmodulin-dependent protein kinase II; TCA: tricarboxylic acid; ATP: adenosine 5’-triphosphate.
Figure 5

Mitochondrial cell death encompasses apoptosis and necrosis. Apoptosis is regulated by OMM permeabilization, mitochondrial membrane potential reduction, BAX activation, and caspase-9. Necrosis is triggered by RIPK3-MLKL-CAMKII pathway activation and mPTP opening. Subsequently, dysfunction of the mitochondrial electron transport chain and termination of the TCA cycle lead to ATP depletion, cytosolic swelling, and membrane rupture. OMM: outer mitochondrial membrane; RIPK3: receptor-interacting protein kinase 3; MLKL: mixed lineage kinase domain-like protein; mPTP: mitochondrial permeability transition pore; CaMKII: Ca2+-calmodulin-dependent protein kinase II; TCA: tricarboxylic acid; ATP: adenosine 5’-triphosphate.

The second route of cell death involves sustained mPTP opening driven by VDAC multimerization, CypD phosphorylation, and upregulation of the adenine nucleotide translocator (ANT), although the exact composition of the mPTP complex remains contentious.[206, 207] mPTP opening induces IMM permeability, leading to mitochondrial swelling, electron transport chain dysfunction, and disruption of the tricarboxylic acid (TCA) cycle.[208, 209] These mitochondrial disturbances lead to ATP depletion, cytoplasmic swelling, membrane rupture, and organelle breakdown, culminating in necroptotic cell death.[210] Unlike apoptosis, necroptosis is non-energy-dependent and characterized by cellular and organelle swelling, extensive mitochondrial disruption, blebbing, and eventual irreversible plasma membrane rupture.[211, 212]

At the molecular level, key regulators of mitochondrial apoptosis and necroptosis are critical for understanding the signal transduction pathways underlying mitochondria-dependent cell death in myocardial injury. Bax is a primary inducer of OMM permeabilization in the apoptotic pathway, whereas Bcl-2 acts as a protective antagonist. Under normal conditions, Bcl-2 binds Bax, forming a heterodimer that neutralizes the pro-apoptotic potential of Bax. Bax expression is upregulated upon stimulation, and cytoplasmic Bax forms homodimers, which migrate to and integrate into the OMM, initiating permeabilization.[213, 214] Consequently, Bcl-2 and Bax levels, and mitochondrial membrane potential reduction, are reliable mitochondrial apoptosis markers.

The critical signaling molecules in necroptosis include receptor-interacting protein kinase 3 (RIPK3), phosphoglycerate mutase 5 (Pgam5), and mixed lineage kinase domain-like protein (Mlkl). During reperfusion, oxidative stress and calcium overload directly or indirectly activate RIPK3, leading to Pgam5 and Mlkl phosphorylation and Mlkl oligomerization at the cell membrane, where Mlkl forms pores that mediate lytic cell death.[215] Recent studies have implicated mPTP opening as a downstream effect of RIPK3 activation in cardiac I/R injury. First, RIPK3 activates CaMKII, which promotes mPTP opening.[216] Additionally, RIPK3 upregulation enhances Pgam5 expression, which phosphorylates CypD, increasing mPTP opening frequency.[217] Together, RIPK3 expression, Mlkl phosphorylation, and mPTP opening present promising targets for modulating mitochondria-driven necroptosis in cardiac injury contexts.

Role of mitochondria-dependent cell death in cardiac I/R injury

Apoptosis has long been considered the principal mechanism of cardiomyocyte death in cardiac I/R injury, contributing significantly to myocyte loss during and post-I/R. However, recent studies have challenged this notion, indicating that the pan-caspase inhibitor Z-Val-Ala-DL-Asp-fluoromethylketone (zVAD) rescues only 30% of cell death in I/R injury, whereas deletion of the necroptosis-related genes, such as RIPK3, reduces cardiomyocyte death by nearly 50%. These results suggest that necroptosis, rather than apoptosis, may be the dominant form of PCD in cardiac I/R injury.[218] This shift in understanding is supported by observations of infarct composition, where the infarct core (“umbra”) exhibits predominantly necroptotic cell death, while the surrounding ischemic penumbra is marked by apoptotic cells.[219, 220]

Despite advances in characterizing the regulatory mechanisms of apoptosis and necroptosis, their interplay remains incompletely understood. Studies have indicated that RIPK3, a key necroptotic effector, is also an upstream activator of caspase-8-dependent apoptosis in MI.[221, 222] Conversely, caspase-8 activation degrades RIPK3, inhibiting necroptosis. Mitochondrial apoptosis inhibitors such as cellular inhibitors of apoptosis 1 and 2 (c-IAP1 and c-IAP2) further influence this crosstalk by promoting RIPK3 ubiquitination and preventing necroptosis activation.[223, 224]

Additional evidence from cellular reperfusion models suggests that RIPK3 may engage in mitochondrial dynamics by activating Drp 1, contributing to mitochondrial potential loss and potentially promoting mitochondrial apoptosis. Interestingly, deleting RIPK3 reversed Fundc1-induced mitophagy, generating an anti-apoptotic signal in reperfused hearts.[218] Conversely, inhibiting autophagy flux triggers necroptotic cardiomyocyte death, underscoring a reciprocal relationship between necroptosis and MQC mechanisms.[225]

Together, these results reveal an intricate overlap between apoptosis and necroptosis, especially in their downstream effects, despite being regulated by distinct upstream pathways. Consequently, cardioprotective strategies aimed at mitigating myocardial I/R injury should consider both anti-apoptotic and anti-necroptotic interventions for comprehensive efficacy.

Role of mitochondria-dependent cell death in MI

Cardiomyocytes receive ischemic and hypoxic damage during MI, which activates the mitochondrial apoptotic pathway. Proapoptotic proteins, such as Bax, translocate from the cytoplasm to the mitochondria, initiating mitochondrial release of cyt-c. This release triggers apoptotic protease activator 1 (Apaf-1) to form apoptosomes, which activate caspase-9 and subsequently the effector caspase-3, leading to apoptosis. Studies using Bax knockout mice have demonstrated reduced cardiomyocyte apoptosis, decreased infarct size, and improved cardiac function, underscoring Bax-mediated mitochondrial apoptosis as a pivotal mechanism in MI pathology.[226, 227]

MI impairs mitochondrial respiratory chain complex activity, decreases ATP production, reduces mitochondrial membrane potential, and increases mitochondrial permeability, facilitating the release of cyt-c and other pro-apoptotic factors. Concurrently, mitochondrial ROS accumulation induces oxidative stress, damaging cell membranes, proteins, and DNA, further compromising cell structure and promoting apoptosis. For example, cardiomyocytes in the infarcted region in MI rat models exhibited significant mitochondrial morphological changes, such as swelling and disrupted cristae, and elevated expression of the apoptosis-related proteins Bax and caspase-3, linking mitochondrial dysfunction closely to apoptosis.[228, 229] Atorvastatin modulated WW domain-containing E3 ubiquitin protein ligase 2 (WWP2) expression and stabilized the Bcl-2-Bax axis within the mitochondrial apoptosis pathway, reducing apoptosis, enhancing cardiac function, and alleviating vascular wall thickening and fibrosis in MI rat models.[230]

Key proteins such as RIPK1, RIPK3, and MLKL are critical in mitochondrial necroptosis. The RIPK3-CaMKII-mPTP signaling axis is activated in MI, with RIPK3-mediated phosphorylation of CaMKII promoting mPTP opening. This causes mitochondrial membrane potential collapse, leading to necroptotic cell death. Treatment with the necroptosis inhibitor necrostatin-1 reduced infarct size and improved cardiac function in MI models, highlighting necroptosis as a significant contributor to MI pathology.[231]

Lipid peroxidation has emerged as a crucial driver of myocardial damage in MI, suggesting a role for ferroptosis in MI pathology. Ferroptosis inhibitors have demonstrated promising cardioprotective effects. For example, the iron chelator deferoxamine (DFO) binds excess iron ions to reduce iron overload and inhibit ferroptosis. DFO treatment reduced myocardial iron levels in MI animal models, mitigated tissue damage, and improved cardiac function.[230] Antioxidants such as vitamin E, which scavenges ROS and inhibits lipid peroxidation, aid in maintaining mitochondrial membrane integrity and provide additional cardioprotection. Post-intervention studies have indicated that vitamin E reduced oxidative stress, limited ferroptotic cell death in cardiomyocytes, and partially restored cardiac function in MI models.[231]

Role of mitochondria-dependent cell death in hypertension

Oxidative stress levels are elevated in hypertension, increasing ROS production. ROS-induced mitochondrial dysfunction in vascular endothelial cells disrupts mitochondrial respiratory chain complex activity, reduces ATP synthesis, and decreases the mitochondrial membrane potential. These disruptions can trigger the mPTP opening, releasing pro-apoptotic factors such as cyt-c, which activates the apoptosis signaling cascade and induces cell death. Studies on SHR models have reported significantly elevated ROS levels in vascular tissues, accompanied by mitochondrial morphological and functional abnormalities, such as swelling and cristae disruption, suggesting that oxidative stress-induced mitochondrial dysfunction is pivotal in hypertension pathogenesis.[232]

AngII is a critical hypertension mediator that exerts its effects by activating the type 1 receptor (AT1R), which activates Ras-MAPK-ERK signaling within the rostral ventrolateral medulla (RVLM) of the brainstem.[233] This pathway increases the expression of the mitochondrial pro-apoptotic proteins Bax and Bad, decreases anti-apoptotic Bcl-2 levels, and activates caspase-3 via the mitochondrial apoptotic pathway, leading to neuronal apoptosis. In the SHR model, heightened Ras, p38 MAPK, and ERK activity in the RVLM are accompanied by increased Bax and Bad expression and enhanced caspase-3 activity, implicating the Ras-MAPK-ERK pathway as a key driver of mitochondrial apoptosis in hypertension-induced sympathetic overactivation.[234]

Emerging studies suggest that ferroptosis (a form of iron-dependent cell death) may also contribute to hypertension pathogenesis. Iron metabolism imbalance in hypertensive states may cause mitochondrial iron overload, amplifying ROS production via the Fenton reaction, thereby exacerbating oxidative stress, compromising mitochondrial membrane integrity, and promoting cell death. Lipid peroxidation is an important facet of ferroptosis, wherein polyunsaturated fatty acids are oxidized under ROS attack, undermining cellular membrane integrity and culminating in cell death. Despite its potential significance, the precise role and regulatory mechanisms of mitochondrial ferroptosis in hypertension remain unclear and warrant further investigation.

Atorvastatin is a commonly prescribed lipid-lowering agent that has demonstrated vascular protective effects relevant to hypertension management. Atorvastatin reduced AngII-induced vascular endothelial injury and apoptosis in cellular models by upregulating the E3 ubiquitin ligase WWP2, which mediates ATP synthase F1 subunit alpha (ATP5A) degradation through the ubiquitin-proteasome pathway. This regulation aids in stabilizing the Bcl-2-Bax axis within the mitochondrial apoptosis pathway, mitigating cell death. Atorvastatin improved endothelial function in hypertensive animal models, lowered blood pressure, and reduced vascular wall thickening and fibrosis, suggesting that its protective effects on endothelial cells are closely linked to enhanced mitochondrial function.[233, 235] Additionally, traditional Chinese medicine (TCM) compounds, such as icariside II, have demonstrated protective effects against hypertension-related damage. Icariside II lowered blood pressure in SHR models, improved left ventricular function, and reduced cardiomyocyte apoptosis and fibrosis potentially by inhibiting the apoptosis signal-regulating kinase 1 (ASK1)-c-Jun N-terminal kinases (JNK)-p38 pathway, thereby reducing oxidative stress and preserving mitochondrial function in cardiomyocytes.[235]

While advances have been made in elucidating the relationship between mitochondrial death and hypertension, significant limitations remain. Mechanistic studies often focus on a limited number of signaling pathways, leaving the broader regulatory network underlying mitochondrial death in hypertension incompletely understood. For example, the specific role of mitochondrial ferroptosis in hypertension and mitochondrial death heterogeneity across different cell types requires further exploration to fully characterize the effects on disease pathology.

Role of mitochondria-dependent cell death in DCM

DCM is closely linked to ferroptosis, which is critical in DCM pathogenesis and progression. Iron overload is a hallmark of diabetes; for example, transferrin receptor 1 (TFR1) upregulation increases cellular iron uptake, while ferroportin (FPN) downregulation reduces iron export and enhances mitochondrial iron accumulation.[236] Excess iron catalyzes ROS production via the Fenton reaction, precipitating mitochondrial dysfunction, as evidenced by decreased respiratory chain complex activity, ATP depletion, reduced mitochondrial membrane potential, and enhanced mPTP opening.[237] These mitochondrial disruptions further elevate ROS levels, creating a vicious cycle that exacerbates cardiomyocyte damage.

Lipid peroxidation is significantly increased in DCM. Hyperglycemia-induced accumulation of advanced glycation end-products (AGEs), heightened oxidative stress, and mitochondrial ROS attack polyunsaturated fatty acids (PUFAs) in cellular membranes, initiating lipid peroxidation. Peroxidation products such as MDA and 4-hydroxynonenal (4-HNE) compromise membrane integrity, alter permeability, and disrupt ion homeostasis, driving ferroptotic cell death. This process further activates inflammatory responses and recruits immune cells, aggravating myocardial tissue damage.[238]

Antioxidant defense system dysfunction also contributes to ferroptosis in DCM. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a master regulator of antioxidant responses and normally sequestered by Keap1 in the cytoplasm. Under oxidative stress, Nrf2 dissociates from Keap 1, translocates to the nucleus, and activates antioxidant gene expression. However, the stability of Keap1 and subsequent Nrf2 activation are impaired in DCM, reducing the expression of downstream antioxidants such as glutathione peroxidase 4 (GPX4) and diminishing cellular defenses against oxidative stress and ferroptosis.[239]

Several pharmacological interventions have demonstrated potential in modulating ferroptosis in DCM. For example, canagliflozin is a SGLT2 inhibitor that attenuates ferroptosis in animal and cell models by restoring cardiac iron homeostasis, enhancing the glutathione (GSH)-GPX4 axis, and activating AMPK signaling, thereby ameliorating oxidative stress and cardiac function.[240] While metformin has demonstrated cardioprotective effects in diabetes-related myocardial injury, its direct role in ferroptosis inhibition within DCM requires further investigation.[241] Additionally, molecular hydrogen (H2) has demonstrated promise in mitigating ferroptosis and mitochondrial apoptosis by promoting SYNV1-Kelch-like ECH-associated protein 1 (Keap1) interaction and regulating the Nrf2-GPx4-GSH pathway, exerting protective effects on diabetic myocardium.[242] Nevertheless, most results have been derived from animal or in vitro studies, underscoring the need for clinical validation and mechanistic elucidation to optimize therapeutic approaches targeting ferroptosis in DCM.

Necroptosis is also markedly elevated in DCM, with increased expression and activity of key necroptotic molecules observed in streptozotocin (STZ)-induced and db/db diabetic mouse models. Elevated RIPK3 expression promotes necrosome formation through RIPK1 binding and MLKL phosphorylation. MLKL oligomerizes and translocates to the cell membrane, disrupting membrane integrity and leading to necroptotic cell death.[243, 244, 245, 246] Mitochondrial dysfunction, including reduced membrane potential and increased oxidative stress, forms a feedback loop with necroptosis in DCM. Mitochondrial impairment amplifies ROS production, which activates necroptotic signaling, while necroptosis-associated mPTP opening and ion imbalance further compromise mitochondrial integrity, perpetuating myocardial injury.[243, 244]

Upregulated RIPK3 in DCM also activates CaMKII via phosphorylation and oxidation, promoting mitochondrial fragmentation and dysfunction, exacerbating cardiomyocyte injury and necroptosis.[216, 243] Additionally, RIPK1 and RIPK3 activate the NLRP3 inflammasome, which recruits and cleaves pro-caspase-1 into active caspase-1, leading to interleukin-1β (IL-1β) maturation and release, amplifying the inflammatory responses.[244, 245]

Therapeutic strategies targeting necroptosis have demonstrated promise. Hydrogen sulfide (H2S) donors, such as sodium hydrosulfide (NaHS), improved cardiac function in db/db and STZ-induced diabetic mice by inhibiting oxidative stress and reducing RIPK1, RIPK3, and MLKL phosphorylation, attenuating necroptosis. H₂S also suppresses NLRP3 inflammasome activation, reducing IL-1β release and subsequent inflammatory damage to cardiomyocytes. Furthermore, H2S promotes the transcription of retinoic acid receptor-related orphan receptor α (RORα), which attenuates oxidative stress and necroptosis through an RORα-dependent pathway, conferring cardioprotection in DCM.[245, 246] Other promising strategies include modulating CaMKIIδ splice variants and activity by targeting protein phosphatase 1 inhibitor 1 (I1PP1), which has demonstrated potential in improving cardiac function and reducing necroptotic myocardial injury in diabetic models. These results suggest that targeting CaMKII activity could be a viable therapeutic avenue for DCM, although further investigation is necessary for drug development and clinical application.[243]

Role of mitochondria-dependent cell death in drug-induced cardiomyopathy

Drug-induced cardiotoxicity studies have described multifaceted mechanisms of mitochondrial death, which encompass various forms of regulated cell death, including mitochondrial apoptosis, necroptosis, ferroptosis, and pyroptosis. For example, the widely used chemotherapeutic agent DOX induces cardiomyocyte apoptosis, impairing cardiac function. Atorvastatin improved cardiac function and reduced vascular wall thickening and fibrosis in DOX-treated models, underscoring its protective role in mitigating drug-induced cardiac damage.[233, 247]

In necroptosis, critical mediators such as RIPK1, RIPK3, and MLKL contribute significantly to mitochondrial necroptotic signaling in drug-induced cardiotoxicity. Treating DOX-induced cardiotoxicity in mice with necrostatin-1, a necroptosis inhibitor, reduced infarct size and improved cardiac function, indicating the pivotal role of necroptosis in drug-related cardiac injury.[243]

Ferroptosis is another pathway implicated in cardiotoxicity and closely associated with dysregulated iron metabolism. The iron chelator DFO binds excess iron ions, reducing iron overload and inhibiting ferroptosis. DFO treatment decreased myocardial iron levels in animal models of DOX-induced cardiotoxicity, alleviated cardiac damage, and improved cardiac function. Similarly, ferristatin-1 was efficacious in inhibiting ferroptosis and mitigating myocardial injury in DOX-treated models.

Pyroptosis in drug-induced cardiotoxicity is intricately linked to inflammasome activation. DOX triggers NLRP3 inflammasome activation, promoting pro-caspase-1 cleavage into active caspase-1 and leading to the maturation and release of inflammatory cytokines such as IL-1β and IL-18.[248] Concurrently, gasdermin D (GSDMD) cleavage facilitates membrane pore formation, a hallmark of pyroptosis, which results in cell swelling, blistering, and eventual lysis.[249] DOX-treated cardiomyocytes exhibited increased expression and activation of NLRP3, caspase-1, and GSDMD, coupled with distinct morphological changes typical of pyroptosis.

Therapeutic interventions targeting these pathways have demonstrated potential. For example, the NLRP3 inflammasome inhibitor MCC950 significantly reduced DOX-induced myocardial injury by inhibiting NLRP3 assembly and activation, curtailing pyroptotic cell death and reducing inflammatory cytokine release.[250] Arbuscular mycorrhiza fungi (AMF) is a mitochondrial function regulator that attenuates DOX-induced pyroptosis and inflammation by suppressing the stimulator of interferon genes-nucleotide-binding domain (NBD), leucine-rich repeat (LRR), and pyrin domain (PYD)-containing protein 3 (STING-NLRP3) signaling pathway, effectively mitigating cardiotoxicity. [251] MitoTEMPO is a mitochondria-targeted antioxidant that has demonstrated cardioprotective effects by reducing mitochondrial damage and indirectly inhibiting pyroptosis.[252] Additionally, SIRT3 overexpression alleviates DOX-induced cardiotoxicity by suppressing NLRP3-mediated pyroptosis, restoring autophagic balance, and dampening inflammatory responses.[244]

Despite these advances, challenges remain in achieving target-specific modulation of mitochondrial death pathways in drug-induced cardiotoxicity. Many agents lack specificity, affecting unrelated physiological pathways and potentially causing adverse effects. For example, some ferroptosis inhibitors may inadvertently disrupt normal iron homeostasis. Furthermore, the complex pharmacokinetics and multi-target effects of these drugs complicate therapeutic outcome stability and reliability, underscoring the need for more precise, targeted approaches in managing mitochondrial dysfunction-associated cardiotoxicity.

Conclusion and future perspectives

MQC represents a critical adaptive system that mitigates cellular damage across various cardiovascular diseases, including MI, DCM, drug-induced cardiomyopathy, and hypertension. MQC preserves mitochondrial integrity, balances energy supply, and maintains cellular homeostasis under physiological and pathophysiological stress through mitochondrial fission, fusion, mitophagy, and regulated cell death pathways. The balance between mitochondrial fission and fusion determines the overall outcome of the mitochondrial network, and is achieved through various mechanisms. For example, mitochondrial fission promotes the isolation and elimination of damaged mitochondria, while fusion allows functional complementarity and mixing of contents, buffering the effects of damaged proteins and maintaining mitochondrial function.[253] Mitochondrial fission and fusion imbalances can lead to mitochondrial dysfunction in different disease states, which affects cell health and survival. For example, increased mitochondrial fission in myocardial I/R injury is strongly associated with mitochondrial damage and cardiomyocyte death.[8] In hypertension, increased mitochondrial fission leads to VSMC proliferation and migration, which affects vascular structure and function.[58] This suggests that regulating mitochondrial fission and fusion is essential for maintaining energy metabolism and cellular stress responses. Mitochondrial division and fusion imbalances lead to mitochondrial dysfunction, which can be partially compensated by autophagy. However, damaged mitochondria can accumulate if autophagy is inhibited, exacerbating mitochondrial dysfunction. Biogenetic processes can replenish mitochondria reduced by autophagy, but the inhibiting these process will result in a further decline in mitochondrial numbers and function. Eventually, an imbalance in these processes can lead to mitochondrial dysfunction, activating the cell death pathway and leading to cardiomyocyte apoptosis and necrosis (Figure 6).

Figure 6 Role of mitochondrial dynamic imbalance, impaired mitochondrial autophagy, and cell death in cardiomyopathy. The balance between mitochondrial division and fusion is disrupted, impairing the mitochondrial network structure and function. Increased division and decreased fusion lead to mitochondrial fragmentation, which decreases ATP energy production, elevates mitochondrial ROS levels, and causes imbalanced calcium ion regulation. The increased damaged mitochondria cannot be effectively cleared by mitochondrial autophagy, which aggravates mitochondrial dysfunction and cell stress. Cardiomyocyte survival is threatened and they may die by apoptosis or necroptosis. The apoptotic pathway involves key proteins such as BAX, caspase-9, and caspase-3, while necrotic apoptosis is closely related to molecules such as RIPK3, MLKL, and CAMKII. ATP: adenosine 5’-triphosphate; mtROS: mitochondrial reactive oxygen species; RIPK3: receptor-interacting protein kinase 3; MLKL: mixed lineage kinase domain-like protein; CaMKII: Ca2+-calmodulin-dependent protein kinase II.
Figure 6

Role of mitochondrial dynamic imbalance, impaired mitochondrial autophagy, and cell death in cardiomyopathy. The balance between mitochondrial division and fusion is disrupted, impairing the mitochondrial network structure and function. Increased division and decreased fusion lead to mitochondrial fragmentation, which decreases ATP energy production, elevates mitochondrial ROS levels, and causes imbalanced calcium ion regulation. The increased damaged mitochondria cannot be effectively cleared by mitochondrial autophagy, which aggravates mitochondrial dysfunction and cell stress. Cardiomyocyte survival is threatened and they may die by apoptosis or necroptosis. The apoptotic pathway involves key proteins such as BAX, caspase-9, and caspase-3, while necrotic apoptosis is closely related to molecules such as RIPK3, MLKL, and CAMKII. ATP: adenosine 5’-triphosphate; mtROS: mitochondrial reactive oxygen species; RIPK3: receptor-interacting protein kinase 3; MLKL: mixed lineage kinase domain-like protein; CaMKII: Ca2+-calmodulin-dependent protein kinase II.

The rapid activation of MQC mechanisms in MI and I/R injury can remove damaged mitochondrial fragments, while timely fusion and mitophagy aid in restoring mitochondrial network function, protecting cardiomyocytes from oxidative and ischemic damage. Similarly, MQC is critical in DCM in addressing mitochondrial iron overload, elevated ROS, and the metabolic imbalances that drive disease progression. In drug-induced cardiomyopathy, such as that caused by DOX, dysregulated MQC contributes to excessive mitochondrial fission, increased ROS production, and cardiomyocyte apoptosis, leading to cardiac dysfunction. Protective strategies targeting specific MQC pathways, such as inhibiting fission or enhancing mitophagy, have therapeutic potential for mitigating the cardiotoxic effects of chemotherapy agents. Furthermore, MQC mechanisms are essential for preserving endothelial and VSMC function in hypertensive cardiovascular disease, where mitochondrial dynamics and autophagy imbalances exacerbate oxidative stress, endothelial dysfunction, and inflammation, contributing to vascular remodeling and hypertension progression (Table 1).

Table 1

Compounds or drugs that target MQC in cardiovascular disease

Name Mechanism Target Disease Reference
Mdivi-1 Mitochondrial fission Drp1 Myocardial I/R injury 31
Mdivi-1 Mitochondrial fission Drp1 MI 46, 47
Mdivi-1 Mitochondrial fission Drp1 Hypertension 58
Y-27632 (ROCK inhibitor) Mitochondrial fission Drp1 Hypertension 60
Nimbolide Mitochondrial fission Akt/mTOR DCM 62
Perillaldehyde Mitochondrial fission miR-133a-3p DCM 63
AuCur Mitochondrial fission PPARα DCM 64
Sevoflurane Mitochondrial fusion OPA1/MFN2 Myocardial I/R injury 81
Epigallocatechin gallate Mitochondrial fusion OPA1 Myocardial I/R injury 94
Melatonin Mitochondrial fusion Notch1/Mfn2 MI 57
Resveratrol Mitochondrial fusion Sirt1/Sirt3-Mfn2-Parkin-PGC1α MI 110
BGP-15 Mitochondrial fusion OPA1/MFN2 Hypertension 121
Acacetin Mitochondrial fusion MFN2 Hypertension 122
Liraglutide Mitochondrial fusion AMPK/OPA1 DCM 129
Shenmai injection Mitochondrial fusion AMPK/Mfn2 Drug cardiomyotoxicity 50
Melatonin Mitophagy PINK1/Parkin MI 153
Berberine Mitophagy HIF-1α/BNIP3 MI 180
Resveratrol Mitophagy PINK1/Parkin Hypertension 190
Y27632 Mitophagy RhoA/ROCK Hypertension 188
Canagliflozin Mitophagy PINK1/Parkin DCM 193
Empagliflozin Mitophagy AMPKα1/ULK1/FUNDC1 DCM 194
Harpagoside Mitophagy p53/Parkin Drug cardiomyotoxicity 201
Atorvastatin Apoptosis Bcl-2/Bax MI 231
Atorvastatin Apoptosis WWP2/Bcl-2/Bax Hypertension 234
Icariside II Apoptosis ASK1-JNK/p38 Hypertension 236
H2 Ferroptosis Nrf2/GPx4/GSH DCM 243
MCC950 Pyroptosis NLRP3 Drug cardiomyotoxicity 251
  1. DRP1: dynamin-related protein 1; I/R: ischemia-reperfusion; MI: myocardial infarction; ROCK: Rho-associated protein kinase; Akt/mTOR: protein kinase B/ mammalian target of rapamycin; DCM: diabetic cardiomyopathy; AuCur: Curcumin-AuNCs; PPARα: peroxisome proliferators-activated receptors α; OPA1: optic atrophy protein 1; MFN2: mitofusin 2; SIRT1: silent mating type information regulation 2 homolog-1; PGC1α: peroxisome proliferators-activated receptor γ coactivator α; AMPK: adenosine 5’-monophosphate-activated protein kinase; PINK1: PTEN induced putative kinase 1. HIF-1α: hypoxia-inducible factor-1 alpha; Bcl-2: B-cell lymphoma-2; BNIP3: BCL2/adenovirus E1B 19 kDa protein-interacting protein 3; ULK1: UNC-51-like kinase 1. FUNDC1: Fun14 domain-containing protein 1; WWP2: WW domain-containing E3 ubiquitin protein ligase 2; ASK1: apoptosis signal-regulating kinase 1; JNK: c-Jun N-terminal kinases; Nrf2: Nuclear factor erythroid 2-related factor 2; GPx4: glutathione peroxidase 4; GSH: glutathione; NLRP3: nucleotide-binding oligomerization domain-like receptor protein 3.

Future research should focus on precisely modulating these MQC pathways to enhance their cardioprotective effects while minimizing the detrimental outcomes associated with prolonged or excessive activation. Fine-tuning mitophagy, fission, and fusion responses to specific stressors could enhance resilience to injury and improve outcomes across various cardiomyopathies. Furthermore, an integrated therapeutic approach targeting both apoptosis and necroptosis in combination with MQC modulation may present a synergistic strategy for conditions such as MI and DCM. Emerging pharmacological and genetic interventions directed at specific MQC proteins, such as those involved in the PINK1-Parkin pathway, Drp1-mediated fission, and antioxidant responses, are promising. However, translating these promising preclinical findings into effective clinical therapies presents several challenges. One major challenge is the complexity of mitochondrial biology and the specificity of targeting the MQC pathway without causing adverse off-target effects. Developing drugs that can selectively regulate MQC components, such as Drp1 inhibitors or mitosis enhancers, requires a deep understanding of the molecular mechanisms involved and their effects on human physiology. Patient population heterogeneity and disease progression variability complicate the treatment response prediction and treatment strategy optimization.

Another challenge is delivering the therapeutic agent to the mitochondria inside the heart muscle cells. Ensuring that the drug achieves its intended goal while minimizing systemic adverse effects is a key aspect of developing effective MQC targeted therapies. Additionally, the long-term safety and efficacy of these treatments must be rigorously evaluated in clinical trials to determine their usefulness in routine clinical practice. Current clinical trials have reported that ABI-009 (rapamycin derivative, nanoparticle albumin-binding sirolimus) treats Leigh or Leigh-like syndrome by targeting mitochondrial autophagy (NCT03747328). A phase Ia/Ib trial of KL1333 (NCT03888716), a cellular nicotinamide adenine dinucleotide (NAD+) level modulator, is being conducted in people with primary mitochondrial diseases.[254]

Mitochondrial transplantation is an emerging approach for treating mitochondrial diseases by using isolated functional mitochondria to restore dysfunctional mitochondria in defective cells. Baharvand et al.’s study of platelet-derived mitochondrial transplantation in 30 patients with acute ST-elevation myocardial infarction (STEMI) observed a slightly larger improvement in the LVEF over 40 days in the intervention group compared to the control group.[255] Coupled with technological innovations in drug delivery and precision medicine, advances in understanding the molecular basis of MQC are poised to facilitate the transition from the laboratory to the bedside. While a large body of relevant data currently supports an association between MQC and cardiovascular disease, relying primarily on relevant data to establish causality is subject to limitations. While these data can reveal associations between changes in MQC and cardiovascular disease phenotypes, they do not fully prove causation. Future research can overcome these limitations by relying more on genetic models, mechanism studies, and clinical trials to directly manipulate the MQC process and validate these findings in human samples. This approach will contribute to a more accurate understanding of the role of MQC in cardiovascular disease and provide a solid foundation for developing new treatment strategies.

In conclusion, while the road to clinical application is challenging, the preclinical and emerging clinical data on MQC modulation offer a glimpse into a future where targeted therapies could significantly affect the treatment landscape of cardiovascular diseases. Additionally, advancements in biomarker-based diagnostics to monitor mitochondrial health in real-time could enable more personalized MQC-targeted therapies. Understanding the interplay of these pathways within different cellular environments, such as cardiomyocytes, endothelial cells, and smooth muscle cells, will be critical for developing highly specific and safe treatments. Innovative therapeutic avenues will likely emerge as the complex dynamics of MQC are decoded further, with the potential to significantly improve outcomes in a broad spectrum of cardiovascular diseases, from acute ischemic events to chronic metabolic and hypertensive heart disease.


Address for Correspondence: Hao Zhou, Department of Cardiology, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
Address for Correspondence: Yang Chen, College of Pharmacy, Guangzhou University of Chinese Medicine, 232 Outer Ring East Road, Guangzhou University City, Panyu District, Guangzhou 510405, Guangdong Province, China.
#

These authors contributed equally to this work.


Funding statement: This work was supported by the National Natural Science Foundation of China (NO. 82300315; NO. 82374240), Guangdong Province Basic and Applied Basic Research Fund Project (No. 2024A1515012174; No. 2024A1515013184). National Administration of Traditional Chinese Medicine Research Project (No. 0102023703), Project of the State Key Laboratory of Dampness Syndrome of Traditional Chinese Medicine jointly established by the province and the ministry (No. SZ2022KF10), Scientific Research Initiation Project of Guangdong Provincial Hospital of Traditional Chinese Medicine (No. 2021KT1709), Research Project of Guangdong Provincial Bureau of Traditional Chinese Medicine (No. 20241120), Guangdong Provincial Key Laboratory of Research on Emergency in TCM (No. 2023B1212060062; 2023 KT15450), Excellent Young Talents Program of Guangdong Provincial Hospital of Traditional Chinese Medicine (No. SZ2024QN05) and Basic Clinical Collaborative Innovation Program of Guangdong Provincial Hospital of Traditional Chinese Medicine and School of Biomedical Sciences, The Chinese University of Hong Kong (No. YN2024HK01). National Traditional Chinese Medicine (TCM) Heritage, Innovation, and Development Demonstration Pilot Project-Guangzhou University of Chinese Medicine and Zhongshan Traditional Chinese Medicine Hospital (Tenth Clinical Medical College) High-Level Hospital Construction Project (GZYZS2024G15).

Acknowledgements

All figures were created with BioRender.com.

  1. Author Contributions

    Miao Zhang, Tong Zhang and Rongjun Zou: Writing—Original draft. Kunyang He, Ru Huang, Jingrui Feng, Jinlin Hu and Teng Ge: Resources. Hao Zhou, Xiaoping Fan and Yang Chen: Writing—Review and Editing. All authors have read and approved the final version of the manuscript.

  2. Ethical Approval

    Not applicable.

  3. Informed Consent

    Not applicable.

  4. Conflict of Interest

    Authors state no conflict of interest.

  5. Use of Large Language Models, AI and Machine Learning Tools

    None declared.

  6. Data Availability Statement

    No additional data.

References

1 Focusing on mitochondrial form and function. Nat Cell Biol 2018;20:735.10.1038/s41556-018-0139-7Suche in Google Scholar PubMed

2 Zhou H, Wang SY, Hu SY, Chen YD, Ren J. ER-mitochondria microdomains in cardiac ischemia-reperfusion injury: a fresh perspective. Front Physiol 2018;9:755.10.3389/fphys.2018.00755Suche in Google Scholar PubMed PubMed Central

3 Wang MJ, Smith K, Yu Q, Miller C, Singh K, Sen CK. Mitochondrial connexin 43 in sex-dependent myocardial responses and estrogen-mediated cardiac protection following acute ischemia/reperfusion injury. Basic Res Cardiol 2019;115:1.10.1007/s00395-019-0759-5Suche in Google Scholar PubMed PubMed Central

4 Livingston MJ, Dong Z. Autophagy in acute kidney injury. Semin Nephrol 2014;34:17-26.10.1016/j.semnephrol.2013.11.004Suche in Google Scholar PubMed PubMed Central

5 Claret M, Garcia-Roves PM. Editorial: dissecting the role of mitochondria in the pathophysiology of type-2 diabetes and obesity: novel concepts and challenges. Curr Diabetes Rev 2017;13:337.10.2174/157339981304170725150725Suche in Google Scholar PubMed

6 Zong Y, Li H, Liao P, Chen L, Pan Y, Zheng YQ, et al. Mitochondrial dysfunction: mechanisms and advances in therapy. Signal Transduct Target Ther 2024;9:124.10.1038/s41392-024-01839-8Suche in Google Scholar PubMed PubMed Central

7 Chang X, Li YK, Cai C, Wu F, He J, Zhang YY, et al. Mitochondrial quality control mechanisms as molecular targets in diabetic heart. Metabolism 2022;137:155313.10.1016/j.metabol.2022.155313Suche in Google Scholar PubMed

8 Peng JF, Salami OM, Lei C, Ni D, Habimana O, Yi GH. Targeted mitochondrial drugs for treatment of myocardial ischaemia-reperfusion injury. J Drug Target 2022;30:833-844.10.1080/1061186X.2022.2085728Suche in Google Scholar PubMed

9 Campos JC, Bozi LHM, Bechara LRG, Lima VM, Ferreira JCB. Mitochondrial quality control in cardiac diseases. Front Physiol 2016;7:479.10.3389/fphys.2016.00479Suche in Google Scholar PubMed PubMed Central

10 Ketenci M, Zablocki D, Sadoshima J. Mitochondrial quality control mechanisms during diabetic cardiomyopathy. JMAJ 2022;5:407-15.10.31662/jmaj.2022-0155Suche in Google Scholar PubMed PubMed Central

11 Chang X, Liu RX, Li RB, Peng YY, Zhu PJ, Zhou H. Molecular mechanisms of mitochondrial quality control in ischemic cardiomyopathy. Int J Biol Sci 2023;19:426-448.10.7150/ijbs.76223Suche in Google Scholar PubMed PubMed Central

12 Horvath C, Young M, Jarabicova I, Kindernay L, Ferenczyova K, Ravingerova T, et al. Inhibition of cardiac RIP3 mitigates early reperfusion injury and calcium-induced mitochondrial swelling without altering necroptotic signalling. Int J Mol Sci 2021;22:7983.10.3390/ijms22157983Suche in Google Scholar PubMed PubMed Central

13 Zhang RH, Krigman J, Luo HK, Ozgen S, Yang MC, Sun N. Mitophagy in cardiovascular homeostasis. Mech Ageing Dev 2020;188:111245.10.1016/j.mad.2020.111245Suche in Google Scholar PubMed PubMed Central

14 Li P, Dong XR, Zhang B, Zhang XT, Liu JZ, Ma DS, et al. Molecular mechanism and therapeutic targeting of necrosis, apoptosis, pyroptosis, and autophagy in cardiovascular disease. Chin Med J (Engl) 2021;134:26472655.10.1097/CM9.0000000000001772Suche in Google Scholar PubMed PubMed Central

15 Mughal W, Martens M, Field J, Chapman D, Huang J, Rattan S, et al. Myocardin regulates mitochondrial calcium homeostasis and prevents permeability transition. Cell Death Differ 2018;25:1732-1748.10.1038/s41418-018-0073-zSuche in Google Scholar PubMed PubMed Central

16 Wang HH, Wu YJ, Tseng YM, Su CH, Hsieh CL, Yeh HI. Mitochondrial fission protein 1 up-regulation ameliorates senescence-related endothelial dysfunction of human endothelial progenitor cells. Angiogenesis 2019;22:569-582.10.1007/s10456-019-09680-2Suche in Google Scholar PubMed

17 Scarpelli PH, Tessarin-Almeida G, Viçoso KL, Lima WR, Borges-Pereira L, Meissner KA, et al. Melatonin activates FIS1 DYN1 and DYN2 Plasmodium falciparum related-genes for mitochondria fission: Mitoemerald-GFP as a tool to visualize mitochondria structure. J Pineal Res 2019;66:e12484.10.1111/jpi.12484Suche in Google Scholar PubMed PubMed Central

18 Rosdah AA, Holien JK, Delbridge LMD, Dusting GJ, Lim SY. Mitochondrial fission—a drug target for cytoprotection or cytodestruction? Pharmacol Res Perspect 2016;4:e00235.10.1002/prp2.235Suche in Google Scholar PubMed PubMed Central

19 Dorn GW. Mitochondrial fission/ fusion and cardiomyopathy. Curr Opin Genet Dev 2016;38:38-44.10.1016/j.gde.2016.03.001Suche in Google Scholar PubMed PubMed Central

20 Hornbeck PV, Zhang B, Murray B, Kornhauser JM, Latham V, Skrzypek E. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res 2015;43:D512-D520.10.1093/nar/gku1267Suche in Google Scholar PubMed PubMed Central

21 Jhun BS, Jin OU, Adaniya SM, Cypress MW, Yoon Y. Adrenergic regulation of Drp1-driven mitochondrial fission in cardiac physio-pathology. Antioxidants (Basel) 2018;7:195.10.3390/antiox7120195Suche in Google Scholar PubMed PubMed Central

22 Xu SC, Wang P, Zhang HL, Gong GH, Gutierrez Cortes N, Zhu WZ, et al. CaMKII induces permeability transition through Drp1 phosphorylation during chronic β-AR stimulation. Nat Commun 2016;7:13189.10.1038/ncomms13189Suche in Google Scholar PubMed PubMed Central

23 Sharp WW, Fang YH, Han M, Zhang HJ, Hong Z, Banathy A, et al. Dynamin-related protein 1 (Drp1)-mediated diastolic dysfunction in myocardial ischemia-reperfusion injury: therapeutic benefits of Drp1 inhibition to reduce mitochondrial fission. FASEB J 2014;28:316-326.10.1096/fj.12-226225Suche in Google Scholar PubMed PubMed Central

24 Zhou H, Wang J, Zhu PJ, Zhu H, Toan S, Hu SY, et al. NR4A1 aggravates the cardiac microvascular ischemia reperfusion injury through suppressing FUNDC1-mediated mitophagy and promoting Mff-required mitochondrial fission by CK2α. Basic Res Cardiol 2018;113:23.10.1007/s00395-018-0682-1Suche in Google Scholar PubMed

25 Wells RC, Picton LK, Williams SCP, Tan FJ, Blake Hill R. Direct binding of the dynamin-like GTPase, Dnm1, to mitochondrial dynamics protein Fis1 is negatively regulated by the Fis1 N-terminal arm. J Biol Chem 2007;282:33769-33775.10.1074/jbc.M700807200Suche in Google Scholar PubMed PubMed Central

26 Zaja I, Bai XW, Liu YN, Kikuchi C, Dosenovic S, Yan YS, et al. Cdk1, PKCδ and calcineurin-mediated Drp1 pathway contributes to mitochondrial fission-induced cardiomyocyte death. Biochem Biophys Res Commun 2014;453:710-721.10.1016/j.bbrc.2014.09.144Suche in Google Scholar PubMed PubMed Central

27 Zhou H, Hu SY, Jin QH, Shi C, Zhang Y, Zhu PJ, et al. Mff-dependent mitochondrial fission contributes to the pathogenesis of cardiac microvas-culature ischemia/reperfusion injury via induction of mROS-mediated cardiolipin oxidation and HK2/VDAC1 disassociation-involved mPTP opening. J Am Heart Assoc 2017;6:e005328.10.1161/JAHA.116.005328Suche in Google Scholar PubMed PubMed Central

28 Luo T, Yue RC, Hu HX, Zhou Z, Yiu KH, Zhang S, et al. PD150606 protects against ischemia/reperfusion injury by preventing μ-calpain-induced mitochondrial apoptosis. Arch Biochem Biophys 2015;586:1-9.10.1016/j.abb.2015.06.005Suche in Google Scholar PubMed

29 Yu P, Zhang J, Yu SC, Luo ZZ, Hua FZ, Yuan LH, et al. Protective effect of sevoflurane postconditioning against cardiac ischemia/reperfusion injury via ameliorating mitochondrial impairment, oxidative stress and rescuing autophagic clearance. PLoS One 2015;10:e0134666.10.1371/journal.pone.0134666Suche in Google Scholar PubMed PubMed Central

30 Su HH, Liao JM, Wang YH, Chen KM, Lin CW, Lee IH, et al. Exogenous GDF11 attenuates non-canonical TGF-β signaling to protect the heart from acute myocardial ischemia-reperfusion injury. Basic Res Cardiol 2019;114:20.10.1007/s00395-019-0728-zSuche in Google Scholar PubMed

31 Ding MG, Dong QQ, Liu ZH, Liu Z, Qu YX, Li X, et al. Inhibition of dynamin-related protein 1 protects against myocardial ischemia-reperfusion injury in diabetic mice. Cardiovasc Diabetol 2017;16:19.10.1186/s12933-017-0501-2Suche in Google Scholar PubMed PubMed Central

32 Yu J, Maimaitili Y, Xie P, Wu JJ, Wang J, Yang YN, et al. High glucose concentration abrogates sevoflurane post-conditioning cardioprotection by advancing mitochondrial fission but dynamin-related protein 1 inhibitor restores these effects. Acta Physiol (Oxf) 2017;220:83-98.10.1111/apha.12812Suche in Google Scholar PubMed

33 Schreiber T, Salhöfer L, Quinting T, Fandrey J. Things get broken: the hypoxia-inducible factor prolyl hydroxylases in ischemic heart disease. Basic Res Cardiol 2019;114:16.10.1007/s00395-019-0725-2Suche in Google Scholar PubMed

34 Gharanei M, Hussain A, Janneh O, Maddock H. Attenuation of doxo-rubicin-induced cardiotoxicity by mdivi-1: a mitochondrial division/ mitophagy inhibitor. PLoS One 2013;8:e77713.10.1371/journal.pone.0077713Suche in Google Scholar PubMed PubMed Central

35 Xue RQ, Sun L, Yu XJ, Li DL, Zang WJ. Vagal nerve stimulation improves mitochondrial dynamics via an M3 receptor/CaMKKβ/AMPK pathway in isoproterenol-induced myocardial ischaemia. J Cell Mol Med 2017;21:58-71.10.1111/jcmm.12938Suche in Google Scholar PubMed PubMed Central

36 Totzeck M, Hendgen-Cotta UB, Rassaf T. Nitrite-nitric oxide signaling and cardioprotection. In: Santulli G, ed. Mitochondrial Dynamics in Cardiovascular Medicine. Springer International Publishing; 2017: 335346.10.1007/978-3-319-55330-6_18Suche in Google Scholar PubMed

37 Maneechote C, Palee S, Kerdphoo S, Jaiwongkam T, Chattipakorn SC, Chattipakorn N. Differential temporal inhibition of mitochondrial fission by Mdivi-1 exerts effective cardioprotection in cardiac ischemia/ reperfusion injury. Clin Sci (Lond) 2018;132:1669-1683.10.1042/CS20180510Suche in Google Scholar PubMed

38 Ikeda Y, Shirakabe A, Maejima Y, Zhai P, Sciarretta S, Toli J, et al. Endogenous Drp1 mediates mitochondrial autophagy and protects the heart against energy stress. Circ Res 2015;116:264-278.10.1161/CIRCRESAHA.116.303356Suche in Google Scholar PubMed

39 Kaur S, Khullar N, Navik U, Bali A, Bhatti GK, Bhatti JS. Multifaceted role of dynamin-related protein 1 in cardiovascular disease: From mitochondrial fission to therapeutic interventions. Mitochondrion 2024;78:101904.10.1016/j.mito.2024.101904Suche in Google Scholar PubMed

40 Andreux PA, Blanco-Bose W, Ryu D, Burdet F, Ibberson M, Aebischer P, et al. The mitophagy activator urolithin A is safe and induces a molecular signature of improved mitochondrial and cellular health in humans. Nat Metab 2019;1:595-603.10.1038/s42255-019-0073-4Suche in Google Scholar PubMed

41 Adaniya SM, Jin OU, Cypress MW, Kusakari Y, Jhun BS. Posttranslational modifications of mitochondrial fission and fusion proteins in cardiac physiology and pathophysiology. Am J Physiol Cell Physiol 2019;316:C583-C604.10.1152/ajpcell.00523.2018Suche in Google Scholar PubMed PubMed Central

42 Kim YM, Youn SW, Sudhahar V, Das A, Chandhri R, Cuervo Grajal H, et al. Redox regulation of mitochondrial fission protein Drp1 by protein disulfide isomerase limits endothelial senescence. Cell Rep 2018;23:35653578.10.1016/j.celrep.2018.05.054Suche in Google Scholar PubMed PubMed Central

43 Kluge MA, Fetterman JL, Vita JA. Mitochondria and endothelial function. Circ Res 2013;112:1171-1188.10.1161/CIRCRESAHA.111.300233Suche in Google Scholar PubMed PubMed Central

44 Los M, Mozoluk M, Ferrari D, Stepczynska A, Stroh C, Renz A, et al. Activation and caspase-mediated inhibition of PARP: a molecular switch between fibroblast necrosis and apoptosis in death receptor signaling. Mol Biol Cell 2002;13:978-988.10.1091/mbc.01-05-0272Suche in Google Scholar PubMed PubMed Central

45 Guo X, Sesaki H, Qi X. Drp1 stabilizes p53 on the mitochondria to trigger necrosis under oxidative stress conditions in vitro and in vivo. Biochem J 2014;461:137-146.10.1042/BJ20131438Suche in Google Scholar PubMed PubMed Central

46 Liu R, Wang SC, Li M, Ma XH, Jia XN, Bu Y, et al. An inhibitor of DRP1 (mdivi-1) alleviates LPS-induced septic AKI by inhibiting NLRP3 inflammasome activation. Biomed Res Int 2020;2020:2398420.10.1155/2020/8493938Suche in Google Scholar PubMed PubMed Central

47 Gawlowski T, Suarez J, Scott B, Torres-Gonzalez M, Wang H, Schwappacher R, et al. Modulation of dynamin-related protein 1 (DRP1) function by increased O-linked-β-N-acetylglucosamine modification (O-GlcNAc) in cardiac myocytes. J Biol Chem 2012;287:30024-30034.10.1074/jbc.M112.390682Suche in Google Scholar PubMed PubMed Central

48 Frei B. Ascorbic acid protects lipids in human plasma and low-density lipoprotein against oxidative damage. Am J Clin Nutr 1991;54:1113S-1118S.10.1093/ajcn/54.6.1113sSuche in Google Scholar PubMed

49 Wang QW, Jiang F, Zhao CL, Song JX, Hu MY, Lv YC, et al. miR-21-5p prevents doxorubicin-induced cardiomyopathy by downregulating BTG2. Heliyon 2023;9:e15451.10.1016/j.heliyon.2023.e15451Suche in Google Scholar PubMed PubMed Central

50 Li L, Li JH, Wang QL, Zhao X, Yang DL, Niu L, et al. Shenmai injection protects against doxorubicin-induced cardiotoxicity via maintaining mitochondrial homeostasis. Front Pharmacol 2020;11:815.10.3389/fphar.2020.00815Suche in Google Scholar PubMed PubMed Central

51 Qin YT, Lv C, Zhang XX, Ruan WB, Xu XY, Chen C, et al. Neuraminidase1 inhibitor protects against doxorubicin-induced cardiotoxicity via suppressing Drp1-dependent mitophagy. Front Cell Dev Biol 2021;9:802502.10.3389/fcell.2021.802502Suche in Google Scholar PubMed PubMed Central

52 Liang XY, Wang SY, Wang LF, Ceylan AF, Ren J, Zhang YM. Mitophagy inhibitor liensinine suppresses doxorubicin-induced cardiotoxicity through inhibition of Drp 1-mediated maladaptive mitochondrial fission. Pharmacol Res 2020;157:104846.10.1016/j.phrs.2020.104846Suche in Google Scholar PubMed

53 Liu XH, Tan HW, Liu XQ, Wu Q. Correlation between the expression of Drp1 in vascular endothelial cells and inflammatory factors in hypertension rats. Exp Ther Med 2018;15:3892-3898.10.3892/etm.2018.5899Suche in Google Scholar PubMed PubMed Central

54 Westermann B. Mitochondrial fusion and fission in cell life and death. Nat Rev Mol Cell Biol 2010;11:872-884.10.1038/nrm3013Suche in Google Scholar PubMed

55 Jin JY, Wei XX, Zhi XL, Wang XH, Meng D. Drp1-dependent mitochondrial fission in cardiovascular disease. Acta Pharmacol Sin 2021;42:655664.10.1038/s41401-020-00518-ySuche in Google Scholar PubMed PubMed Central

56 Cai H, Harrison DG. Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circ Res 2000;87:840-844.10.1161/01.RES.87.10.840Suche in Google Scholar PubMed

57 Wang L, Yu TZ, Lee HJ, O’Brien DK, Sesaki H, Yoon YS. Decreasing mitochondrial fission diminishes vascular smooth muscle cell migration and ameliorates intimal hyperplasia. Cardiovasc Res 2015;106:272-283.10.1093/cvr/cvv005Suche in Google Scholar PubMed PubMed Central

58 Deng Y, Li SY, Chen ZZ, Wang WJ, Geng B, Cai J. Mdivi- 1, a mitochondrial fission inhibitor, reduces angiotensin-II-induced hypertension by mediating VSMC phenotypic switch. Biomed Pharmacother 2021;140:111689.10.1016/j.biopha.2021.111689Suche in Google Scholar PubMed

59 Jernigan NL, Walker BR, Resta TC. Reactive oxygen species mediate RhoA/Rho kinase-induced Ca2+ sensitization in pulmonary vascular smooth muscle following chronic hypoxia. Am J Physiol Lung Cell Mol Physiol 2008;295:L515-L529.10.1152/ajplung.00355.2007Suche in Google Scholar PubMed PubMed Central

60 Chen C, Gao JL, Liu MY, Li SL, Xuan XC, Zhang XZ, et al. Mitochondrial fission inhibitors suppress endothelin- 1-induced artery constriction. Cell Physiol Biochem 2017;42:1802-1811.10.1159/000479536Suche in Google Scholar PubMed

61 Song XG, Fan CX, Wei C, Yu WH, Tang JC, Ma F, et al. Mitochondria fission accentuates oxidative stress in hyperglycemia-induced H9c2 cardiomyoblasts in vitro by regulating fatty acid oxidation. Cell Biol Int 2024;48:1378-1391.10.1002/cbin.12204Suche in Google Scholar PubMed

62 Zhang HT, Zhao XL, Wei W, Shen CJ. Nimbolide protects against diabetic cardiomyopathy by regulating endoplasmic reticulum stress and mitochondrial function via the Akt/mTOR pathway. Tissue Cell 2024;90:102478.10.1016/j.tice.2024.102478Suche in Google Scholar PubMed

63 Yu YN, Ren YY, Shao ZL, Chen BL, Cui BY, Chao CY, et al. Perillaldehyde improves diabetic cardiomyopathy by upregulating miR-133a-3p to regulate GSK-3β. Eur J Pharmacol 2023;953:175836.10.1016/j.ejphar.2023.175836Suche in Google Scholar PubMed

64 Wei DZ, Li D, Zheng DM, An ZN, Xing XJ, Jiang DW, et al. Curcumin conjugated gold nanoclusters as perspective therapeutics for diabetic cardiomyopathy. Front Chem 2021;9:763892.10.3389/fchem.2021.763892Suche in Google Scholar PubMed PubMed Central

65 Liu ZY, Lin LC, Liu ZY, Song K, Tu B, Sun H, et al. N6-Methyladenosine-mediated phase separation suppresses NOTCH1 expression and promotes mitochondrial fission in diabetic cardiac fibrosis. Cardiovasc Diabetol 2024;23:347.10.1186/s12933-024-02444-3Suche in Google Scholar PubMed PubMed Central

66 Twig G, Elorza A, Molina AJA, Mohamed H, Wikstrom JD, Walzer G, et al. Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. EMBO J 2008;27:433-446.10.1038/sj.emboj.7601963Suche in Google Scholar PubMed PubMed Central

67 Doxorubicin-induced cardiomyopathy. N Engl J Med 1999;340:653-655.10.1056/NEJM199902253400813Suche in Google Scholar PubMed

68 Zhang S, Liu XB, Bawa-Khalfe T, Lu LS, Lyu YL, Liu LF, et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med 2012;18:1639-1642.10.1038/nm.2919Suche in Google Scholar PubMed

69 Ichikawa Y, Ghanefar M, Bayeva M, Wu R, Khechaduri A, Naga Prasad SV, et al. Cardiotoxicity of doxorubicin is mediated through mitochondrial iron accumulation. J Clin Invest 2014;124:617-630.10.1172/JCI72931Suche in Google Scholar PubMed PubMed Central

70 Lee YJ, Jeong SY, Karbowski M, Smith CL, Youle RJ. Roles of the mammalian mitochondrial fission and fusion mediators Fis1, Drp1, and Opa1 in apoptosis. Mol Biol Cell 2004;15:5001-5011.10.1091/mbc.e04-04-0294Suche in Google Scholar PubMed PubMed Central

71 Wang YH, Feng WK, Xue WL, Tan Y, Hein DW, Li XK, et al. Inactivation of GSK-3beta by metallothionein prevents diabetes-related changes in cardiac energy metabolism, inflammation, nitrosative damage, and remodeling. Diabetes 2009;58:1391-1402.10.2337/db08-1697Suche in Google Scholar PubMed PubMed Central

72 Khaliq NU, Sandra FC, Park DY, Lee JY, Oh KS, Kim D, et al. Doxorubicin/heparin composite nanoparticles for caspase-activated prodrug chemotherapy. Biomaterials 2016;101:131-142.10.1016/j.biomaterials.2016.05.056Suche in Google Scholar PubMed

73 Vaziri N, Marques D, Greenway SC, Bousman CA. The cellular mechanism of antipsychotic-induced myocarditis: a systematic review. Schizophr Res 2023;261:206-215.10.1016/j.schres.2023.09.039Suche in Google Scholar PubMed

74 Meyer JN, Leuthner TC, Luz AL. Mitochondrial fusion, fission, and mitochondrial toxicity. Toxicology 2017;391:42-53.10.1016/j.tox.2017.07.019Suche in Google Scholar PubMed PubMed Central

75 Park M, Sandner P, Krieg T. cGMP at the centre of attention: emerging strategies for activating the cardioprotective PKG pathway. Basic Res Cardiol 2018;113:24.10.1007/s00395-018-0679-9Suche in Google Scholar PubMed PubMed Central

76 Guo YJ, Zhang H, Yan C, Shen BR, Zhang Y, Guo XY, et al. Small molecule agonist of mitochondrial fusion repairs mitochondrial dysfunction. Nat Chem Biol 2023;19:468-477.10.1038/s41589-022-01224-ySuche in Google Scholar PubMed

77 Cohen MM, Tareste D. Recent insights into the structure and function of Mitofusins in mitochondrial fusion. F1000Res 2018;7:1983.10.12688/f1000research.16629.1Suche in Google Scholar PubMed PubMed Central

78 MacVicar T, Langer T. OPA1 processing in cell death and disease—the long and short of it. J Cell Sci 2016;129:2297-2306.10.1242/jcs.159186Suche in Google Scholar PubMed

79 Ban T, Ishihara T, Kohno H, Saita S, Ichimura A, Maenaka K, et al. Molecular basis of selective mitochondrial fusion by heterotypic action between OPA1 and cardiolipin. Nat Cell Biol 2017;19:856-863.10.1038/ncb3560Suche in Google Scholar PubMed

80 Dudek J. Role of cardiolipin in mitochondrial signaling pathways. Front Cell Dev Biol 2017;5:90.10.3389/fcell.2017.00090Suche in Google Scholar PubMed PubMed Central

81 Yu J, Wu JJ, Xie P, Maimaitili Y, Wang J, Xia ZY, et al. Sevoflurane post-conditioning attenuates cardiomyocyte hypoxia/reoxygenation injury via restoring mitochondrial morphology. PeerJ 2016;4:e2659.10.7717/peerj.2659Suche in Google Scholar PubMed PubMed Central

82 Chen HC, Ren SX, Clish C, Jain M, Mootha V, Michael McCaffery J, et al. Titration of mitochondrial fusion rescues Mff-deficient cardiomyopathy. J Cell Biol 2015;211:795-805.10.1083/jcb.201507035Suche in Google Scholar PubMed PubMed Central

83 Chen Y, Csordás G, Jowdy C, Schneider TG, Csordás N, Wang W, et al. Mitofusin 2-containing mitochondrial-reticular microdomains direct rapid cardiomyocyte bioenergetic responses via interorganelle Ca2+ crosstalk. Circ Res 2012;111:863-875.10.1161/CIRCRESAHA.112.266585Suche in Google Scholar PubMed PubMed Central

84 Chen Y, Sparks M, Bhandari P, Matkovich SJ, Dorn GW 2nd. Mitochondrial genome linearization is a causative factor for cardiomyopathy in mice and Drosophila. Antioxid Redox Signal 2014;21:1949-1959.10.1089/ars.2013.5432Suche in Google Scholar PubMed PubMed Central

85 Papanicolaou KN, Ngoh GA, Dabkowski ER, O’Connell KA, Ribeiro RF Jr, Stanley WC, et al. Cardiomyocyte deletion of mitofusin-1 leads to mitochondrial fragmentation and improves tolerance to ROS-induced mitochondrial dysfunction and cell death. Am J Physiol Heart Circ Physiol 2012;302:H167-H179.10.1152/ajpheart.00833.2011Suche in Google Scholar PubMed PubMed Central

86 Papanicolaou KN, Khairallah RJ, Ngoh GA, Chikando A, Luptak I, O’Shea KM, et al. Mitofusin-2 maintains mitochondrial structure and contributes to stress-induced permeability transition in cardiac myocytes. Mol Cell Biol 2011;31:1309-1328.10.1128/MCB.00911-10Suche in Google Scholar PubMed PubMed Central

87 Ndongson-Dongmo B, Lang GP, Mece O, Hechaichi N, Lajqi T, Hoyer D, et al. Reduced ambient temperature exacerbates SIRS-induced cardiac autonomic dysregulation and myocardial dysfunction in mice. Basic Res Cardiol 2019;114:26.10.1007/s00395-019-0734-1Suche in Google Scholar PubMed

88 Shen T, Zheng M, Cao CM, Chen CL, Tang J, Zhang WR, et al. Mitofusin-2 is a major determinant of oxidative stress-mediated heart muscle cell apoptosis. J Biol Chem 2007;282:23354-23361.10.1074/jbc.M702657200Suche in Google Scholar PubMed

89 Song M, Mihara K, Chen Y, Scorrano L, Dorn GW. Mitochondrial fission and fusion factors reciprocally orchestrate mitophagic culling in mouse hearts and cultured fibroblasts. Cell Metab 2015;21:273-286.10.1016/j.cmet.2014.12.011Suche in Google Scholar PubMed PubMed Central

90 Mouton AJ, DeLeon-Pennell KY, Rivera Gonzalez OJ, Flynn ER, Freeman TC, Saucerman JJ, et al. Mapping macrophage polarization over the myocardial infarction time continuum. Basic Res Cardiol 2018;113:26.10.1007/s00395-018-0686-xSuche in Google Scholar PubMed PubMed Central

91 Dorn GW 2nd, Clark CF, Eschenbacher WH, Kang MY, Engelhard JT, Warner SJ, et al. MARF and Opa1 control mitochondrial and cardiac function in Drosophila. Circ Res 2011;108:12-17.10.1161/CIRCRESAHA.110.236745Suche in Google Scholar PubMed PubMed Central

92 Guan LC, Che ZM, Meng XD, Yu Y, Li MH, Yu ZQ, et al. MCU Up-regulation contributes to myocardial ischemia-reperfusion Injury through calpain/OPA-1-mediated mitochondrial fusion/mitophagy Inhibition. J Cell Mol Med 2019;23:7830-7843.10.1111/jcmm.14662Suche in Google Scholar PubMed PubMed Central

93 Zhang Y, Wang Y, Xu JN, Tian F, Hu SY, Chen YD, 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:e12542.10.1111/jpi.12542Suche in Google Scholar PubMed

94 Nan JL, Nan CJ, Ye J, Qian L, Geng Y, Xing DW, et al. EGCG protects cardiomyocytes against hypoxia-reperfusion injury through inhibition of OMA1 activation. J Cell Sci 2019;132:jcs220871.10.1242/jcs.220871Suche in Google Scholar PubMed

95 Ma SX, Dong ZM. Melatonin attenuates cardiac reperfusion stress by improving OPA1-related mitochondrial fusion in a Yap-hippo pathway-dependent manner. J Cardiovasc Pharmacol. 2019;73:27-39.10.1097/FJC.0000000000000626Suche in Google Scholar PubMed PubMed Central

96 Guo YZ, Wang Z, Qin XH, Xu J, Hou ZX, Yang HY, et al. Enhancing fatty acid utilization ameliorates mitochondrial fragmentation and cardiac dysfunction via rebalancing optic atrophy 1 processing in the failing heart. Cardiovasc Res 2018;114:979-991.10.1093/cvr/cvy052Suche in Google Scholar PubMed

97 Moore JB 4th, Tang XL, Zhao J, Fischer AG, Wu WJ, Uchida S, et al. Epigenetically modified cardiac mesenchymal stromal cells limit myocardial fibrosis and promote functional recovery in a model of chronic ischemic cardiomyopathy. Basic Res Cardiol 2018;114:3.10.1007/s00395-018-0710-1Suche in Google Scholar PubMed PubMed Central

98 Ferreira JCB, Campos JC, Qvit N, Qi X, Bozi LHM, Bechara LRG, et al. A selective inhibitor of mitofusin 1-βIIPKC association improves heart failure outcome in rats. Nat Commun 2019;10:329.10.1038/s41467-018-08276-6Suche in Google Scholar PubMed PubMed Central

99 Pyakurel A, Savoia C, Hess D, Scorrano L. Extracellular regulated kinase phosphorylates mitofusin 1 to control mitochondrial morphology and apoptosis. Mol Cell 2015;58:244-254.10.1016/j.molcel.2015.02.021Suche in Google Scholar PubMed PubMed Central

100 Chen Y, Dorn GW 2nd. PINK1-phosphorylated mitofusin 2 is a Parkin receptor for culling damaged mitochondria. Science 2013;340:471-475.10.1126/science.1231031Suche in Google Scholar PubMed PubMed Central

101 Tsushima K, Bugger H, Wende AR, Soto J, Jenson GA, Tor AR, et al. Mitochondrial reactive oxygen species in lipotoxic hearts induce post-translational modifications of AKAP121, DRP 1, and OPA1 that promote mitochondrial fission. Circ Res 2018;122:58-73.10.1161/CIRCRESAHA.117.311307Suche in Google Scholar PubMed PubMed Central

102 Nan JL, Hu HX, Sun Y, Zhu LL, Wang YC, Zhong ZW, et al. TNFR2 stimulation promotes mitochondrial fusion via Stat3- and NF-kB-dependent activation of OPA1 expression. Circ Res. 2017;121(4):392-410.10.1161/CIRCRESAHA.117.311143Suche in Google Scholar PubMed PubMed Central

103 Samant SA, Zhang HJ, Hong Z, Pillai VB, Sundaresan NR, Wolfgeher D, et al. SIRT3 deacetylates and activates OPA1 to regulate mitochondrial dynamics during stress. Mol Cell Biol 2014;34:807-819.10.1128/MCB.01483-13Suche in Google Scholar PubMed PubMed Central

104 Makino A, Suarez J, Gawlowski T, Han WL, Wang H, Scott BT, et al. Regulation of mitochondrial morphology and function by O-GlcNAcylation in neonatal cardiac myocytes. Am J Physiol Regul Integr Comp Physiol 2011;300:R1296-1302.10.1152/ajpregu.00437.2010Suche in Google Scholar PubMed PubMed Central

105 Ramachandra CJA, Hernandez-Resendiz S, Crespo-Avilan GE, Lin YH, Hausenloy DJ. Mitochondria in acute myocardial infarction and cardioprotection. EBioMedicine 2020;57:102884.10.1016/j.ebiom.2020.102884Suche in Google Scholar PubMed PubMed Central

106 Pei HF, Du J, Song XF, He L, Zhang YF, Li XC, et al. Melatonin prevents adverse myocardial infarction remodeling via Notch1/Mfn2 pathway. Free Radic Biol Med 2016;97:408-417.10.1016/j.freeradbiomed.2016.06.015Suche in Google Scholar PubMed

107 MacGrogan D, Nus M, de la Pompa JL. Notch signaling in cardiac development and disease. Curr Top Dev Biol 2010;92:333-365.10.1016/S0070-2153(10)92011-5Suche in Google Scholar PubMed

108 Du JX, Li HC, Song JJ, Wang TT, Dong YB, Zhan A, et al. AMPK activation alleviates myocardial ischemia-reperfusion injury by regulating Drp1-mediated mitochondrial dynamics. Front Pharmacol 2022;13:862204.10.3389/fphar.2022.862204Suche in Google Scholar PubMed PubMed Central

109 Shi T, Wang F, Stieren E, Tong Q. SIRT3, a mitochondrial sirtuin deacetylase, regulates mitochondrial function and thermogenesis in brown adipocytes. J Biol Chem 2005;280:13560-13567.10.1074/jbc.M414670200Suche in Google Scholar PubMed

110 Zheng MS, Bai YL, Sun XY, Fu R, Liu LY, Liu MS, et al. Resveratrol reestablishes mitochondrial quality control in myocardial ischemia/ reperfusion injury through Sirt1/Sirt3-Mfn2-parkin-PGC-1α pathway. Molecules 2022;27:5545.10.3390/molecules27175545Suche in Google Scholar PubMed PubMed Central

111 Sun XO, Yang YY, Xie YX, Shi XJ, Huang LJ, Tan W. Protective role of STVNa in myocardial ischemia reperfusion injury by inhibiting mitochondrial fission. Oncotarget 2017;9:1898-1905.10.18632/oncotarget.22969Suche in Google Scholar PubMed PubMed Central

112 Kelso GF, Porteous CM, Coulter CV, Hughes G, Porteous WK, Ledgerwood EC, et al. Selective targeting of a redox-active ubiquinone to mitochondria within cells: antioxidant and antiapoptotic properties. J Biol Chem 2001;276:4588-4596.10.1074/jbc.M009093200Suche in Google Scholar PubMed

113 Chen W, Zhao HK, Li YS. Mitochondrial dynamics in health and disease: mechanisms and potential targets. Signal Transduct Target Ther 2023;8:333.10.1038/s41392-023-01547-9Suche in Google Scholar PubMed PubMed Central

114 Chen HC, Detmer SA, Ewald AJ, Griffin EE, Fraser SE, Chan DC. Mitofusins MFN1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J Cell Biol 2003;160:189-200.10.1083/jcb.200211046Suche in Google Scholar PubMed PubMed Central

115 Ishihara N, Fujita Y, Oka T, Mihara K. Regulation of mitochondrial morphology through proteolytic cleavage of OPA1. EMBO J 2006;25:29662977.10.1038/sj.emboj.7601184Suche in Google Scholar PubMed PubMed Central

116 Franco A, Kitsis RN, Fleischer JA, Gavathiotis E, Kornfeld OS, Gong G, et al. Correcting mitochondrial fusion by manipulating mitofusin conformations. Nature 2016;540:74-79.10.1038/nature20156Suche in Google Scholar PubMed PubMed Central

117 Leboucher GP, Tsai YC, Yang M, Shaw KC, Zhou M, Veenstra TD, et al. Stress-induced phosphorylation and proteasomal degradation of mitofusin 2 facilitates mitochondrial fragmentation and apoptosis. Mol Cell 2012;47:547-557.10.1016/j.molcel.2012.05.041Suche in Google Scholar PubMed PubMed Central

118 Wang ZG, Niu QL, Peng XY, Li M, Liu K, Liu Y, et al. Candesartan cilexetil attenuated cardiac remodeling by improving expression and function of mitofusin 2 in SHR. Int J Cardiol 2016;214:348-357.10.1016/j.ijcard.2016.04.007Suche in Google Scholar PubMed

119 Jin HS, Sober S, Hong KW, Org E, Kim BY, Laan M, et al. Age-dependent association of the polymorphisms in the mitochondria-shaping gene, OPA1 with blood pressure and hypertension in Korean population. Am J Hypertens 2011;24:1127-1135.10.1038/ajh.2011.131Suche in Google Scholar PubMed

120 Jin HS, Hong KW, Kim BY, Kim J, Yoo YH, Oh B, et al. Replicated association between genetic variation in the PARK2 gene and blood pressure. Clin Chim Acta 2011;412:1673-1677.10.1016/j.cca.2011.05.026Suche in Google Scholar PubMed

121 Longevity OMAC. Retracted: modulation of mitochondrial quality control processes by BGP-15 in oxidative stress scenarios: from cell culture to heart failure. Oxid Med Cell Longev 2023;2023:9864208.10.1155/2023/9864208Suche in Google Scholar PubMed PubMed Central

122 Yuan PP, Zhang Q, Fu Y, Hou Y, Gao LY, Wei YX, et al. Acacetin inhibits myocardial mitochondrial dysfunction by activating PI3K/AKT in SHR rats fed with fructose. J Nat Med 2023;77:262-275.10.1007/s11418-022-01666-7Suche in Google Scholar PubMed

123 Ishihara N, Eura Y, Mihara K. Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity. J Cell Sci 2004;117:65356546.10.1242/jcs.01565Suche in Google Scholar PubMed

124 Malka F, Guillery O, Cifuentes-Diaz C, Guillou E, Belenguer P, Lombès A, et al. Separate fusion of outer and inner mitochondrial membranes. EMBO Rep 2005;6:853-859.10.1038/sj.embor.7400488Suche in Google Scholar PubMed PubMed Central

125 Cipolat S, Martins de Brito O, Dal Zilio B, Scorrano L. OPA1 requires mitofusin 1 to promote mitochondrial fusion. Proc Natl Acad Sci U S A 2004;101:15927-15932.10.1073/pnas.0407043101Suche in Google Scholar PubMed PubMed Central

126 Zhou YT, Suo WD, Zhang XN, Liang JJ, Zhao WZ, Wang Y, et al. Targeting mitochondrial quality control for diabetic cardiomyopathy: Therapeutic potential of hypoglycemic drugs. Biomed Pharmacother 2023;168:115669.10.1016/j.biopha.2023.115669Suche in Google Scholar PubMed

127 Wu QR, Zheng DL, Liu PM, Yang H, Li LA, Kuang SJ, et al. High glucose induces Drp1-mediated mitochondrial fission via the Orai1 calcium channel to participate in diabetic cardiomyocyte hypertrophy. Cell Death Dis 2021;12:216.10.1038/s41419-021-03502-4Suche in Google Scholar PubMed PubMed Central

128 Patten D, Harper ME, Boardman N. Harnessing the protective role of OPA1 in diabetic cardiomyopathy. Acta Physiol (Oxf) 2020;229:e13466.10.1111/apha.13466Suche in Google Scholar PubMed

129 Yang Y, Fang H, Xu G, Zhen YF, Zhang YZ, Tian JL, et al. Liraglutide improves cognitive impairment via the AMPK and PI3K/Akt signaling pathways in type 2 diabetic rats. Mol Med Rep 2018;18:2449-2457.10.3892/mmr.2018.9180Suche in Google Scholar PubMed

130 Zhang L, Zhang HM, Xie XZ, Tie RP, Shang XL, Zhao QQ, et al. Empagliflozin ameliorates diabetic cardiomyopathy via regulated branched-chain amino acid metabolism and mTOR/p-ULK1 signaling pathway-mediated autophagy. Diabetol Metab Syndr 2023;15:93.10.1186/s13098-023-01061-6Suche in Google Scholar PubMed PubMed Central

131 Wu T, Qu YW, Xu SJ, Wang Y, Liu X, Ma DF. SIRT6: a potential therapeutic target for diabetic cardiomyopathy. FASEB J 2023;37:e23099.10.1096/fj.202301012RSuche in Google Scholar PubMed

132 Yu LM, Gong B, Duan WX, Fan CX, Zhang J, Li Z, et al. Melatonin ameliorates myocardial ischemia/reperfusion injury in type 1 diabetic rats by preserving mitochondrial function: role of AMPK-PGC-1α-SIRT3 signaling. Sci Rep 2017;7:41337.10.1038/srep41337Suche in Google Scholar PubMed PubMed Central

133 Liu CY, Han YH, Gu XM, Li M, Du YY, Feng N, et al. Paeonol promotes Opa1-mediated mitochondrial fusion via activating the CK2α-Stat3 pathway in diabetic cardiomyopathy. Redox Biol 2021;46:102098.10.1016/j.redox.2021.102098Suche in Google Scholar PubMed PubMed Central

134 Catanzaro MP, Weiner A, Kaminaris A, Li CR, Cai F, Zhao FY, et al. Doxorubicin-induced cardiomyocyte death is mediated by unchecked mitochondrial fission and mitophagy. FASEB J 2019;33:11096-11108.10.1096/fj.201802663RSuche in Google Scholar PubMed PubMed Central

135 Dhingra R, Margulets V, Chowdhury SR, Thliveris J, Jassal D, Fernyhough P, et al. Bnip3 mediates doxorubicin-induced cardiac myocyte necrosis and mortality through changes in mitochondrial signaling. Proc Natl Acad Sci U S A 2014;111:E5537-5544.10.1073/pnas.1414665111Suche in Google Scholar PubMed PubMed Central

136 Maneechote C, Khuanjing T, Ongnok B, Arinno A, Prathumsap N, Chunchai T, et al. Upregulation of mitochondrial fusion as potential cardioprotective strategies against trastuzumab-induced cardiotoxicity in rats. Eur Heart J 2023;44:ehad655.3124.10.1093/eurheartj/ehad655.3124Suche in Google Scholar

137 Méndez-Valdés G, Gómez-Hevia F, Bragato MC, Lillo-Moya J, Rojas-Solé C, Saso L, et al. Antioxidant protection against trastuzumab cardiotoxicity in breast cancer therapy. Antioxidants (Basel) 2023;12:457.10.3390/antiox12020457Suche in Google Scholar PubMed PubMed Central

138 Ding MG, Shi R, Cheng SL, Li M, De DM, Liu CY, et al. Mfn2-mediated mitochondrial fusion alleviates doxorubicin-induced cardiotoxicity with enhancing its anticancer activity through metabolic switch. Redox Biol 2022;52:102311.10.1016/j.redox.2022.102311Suche in Google Scholar PubMed PubMed Central

139 Semenzato M, Kohr MJ, Quirin C, Menabò R, Alanova P, Alan L, et al. Oxidization of optic atrophy 1 cysteines occurs during heart ischemiareperfusion and amplifies cell death by oxidative stress. Redox Biol 2023;63:102755.10.1016/j.redox.2023.102755Suche in Google Scholar PubMed PubMed Central

140 Vousden KH, Lane DP. p53 in health and disease. Nat Rev Mol Cell Biol 2007;8:275-283.10.1038/nrm2147Suche in Google Scholar PubMed

141 Duan JF, Liu XL, Shen S, Tan X, Wang Y, Wang L, et al. Trophoblast stem-cell-derived exosomes alleviate cardiotoxicity of doxorubicin via improving Mfn2-mediated mitochondrial fusion. Cardiovasc Toxicol 2023;23:23-31.10.1007/s12012-022-09774-2Suche in Google Scholar PubMed PubMed Central

142 Liu K, Zhao Q, Sun HY, Liu L, Wang CQ, Li Z, et al. BNIP3 (BCL2 interacting protein 3) regulates pluripotency by modulating mitochondrial homeostasis via mitophagy. Cell Death Dis 2022;13:334.10.1038/s41419-022-04795-9Suche in Google Scholar PubMed PubMed Central

143 Turkieh A, El Masri Y, Pinet F, Dubois-Deruy E. Mitophagy regulation following myocardial infarction. Cells 2022;11:199.10.3390/cells11020199Suche in Google Scholar PubMed PubMed Central

144 Springer MZ, MacLeod KF. In Brief: Mitophagy: mechanisms and role in human disease. J Pathol 2016;240:253-255.10.1002/path.4774Suche in Google Scholar PubMed PubMed Central

145 Oshima Y, Verhoeven N, Cartier E, Karbowski M. The OMM-severed and IMM-ubiquitinated mitochondria are intermediates of mitochondrial proteotoxicity-induced autophagy in PRKN/parkin-deficient cells. Autophagy 2021;17:3884-3886.10.1080/15548627.2021.1964887Suche in Google Scholar PubMed PubMed Central

146 Li KQ, Xia X, Tong Y. Multiple roles of mitochondrial autophagy receptor FUNDC1 in mitochondrial events and kidney disease. Front Cell Dev Biol 2024;12:1453365.10.3389/fcell.2024.1453365Suche in Google Scholar PubMed PubMed Central

147 He YL, Li J, Gong SH, Cheng X, Zhao M, Cao Y, et al. BNIP3 phosphorylation by JNK1/2 promotes mitophagy via enhancing its stability under hypoxia. Cell Death Dis 2022;13:966.10.1038/s41419-022-05418-zSuche in Google Scholar PubMed PubMed Central

148 Kazlauskaite A, Kondapalli C, Gourlay R, Campbell DG, Ritorto MS, Hofmann K, et al. Parkin is activated by PINK1-dependent phosphorylation of ubiquitin at Ser65. Biochem J 2014;460:127-139.10.1042/BJ20140334Suche in Google Scholar PubMed PubMed Central

149 Zhu YY, Massen S, Terenzio M, Lang V, Chen-Lindner S, Eils R, et al. Modulation of serines 17 and 24 in the LC3-interacting region of Bnip3 determines pro-survival mitophagy versus apoptosis. J Biol Chem 2013;288:1099-1113.10.1074/jbc.M112.399345Suche in Google Scholar PubMed PubMed Central

150 Liu L, Feng D, Chen G, Chen M, Zheng QX, Song PP, et al. Mitochondrial outer-membrane protein FUNDC1 mediates hypoxia-induced mitophagy in mammalian cells. Nat Cell Biol 2012;14:177-185.10.1038/ncb2422Suche in Google Scholar PubMed

151 Sun T, Ding W, Xu T, Ao X, Yu T, Li MY, et al. Parkin regulates programmed necrosis and myocardial ischemia/reperfusion injury by targeting cyclophilin-D. Antioxid Redox Signal 2019;31:1177-1193.10.1089/ars.2019.7734Suche in Google Scholar PubMed

152 Zhou H, Zhang Y, Hu SY, Shi C, Zhu PJ, Ma Q, 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:e12413.10.1111/jpi.12413Suche in Google Scholar PubMed PubMed Central

153 Dhingra A, Jayas R, Afshar P, Guberman M, Maddaford G, Gerstein J, et al. Ellagic acid antagonizes Bnip3-mediated mitochondrial injury and necrotic cell death of cardiac myocytes. Free Radic Biol Med 2017;112:411-422.10.1016/j.freeradbiomed.2017.08.010Suche in Google Scholar PubMed

154 Paradies G, Paradies V, Ruggiero FM, Petrosillo G. Mitochondrial bioenergetics and cardiolipin alterations in myocardial ischemia-reperfusion injury: implications for pharmacological cardioprotection. Am J Physiol Heart Circ Physiol 2018;315:H1341-352.10.1152/ajpheart.00028.2018Suche in Google Scholar PubMed

155 Morton AB, Smuder AJ, Wiggs MP, Hall SE, Ahn B, Hinkley JM, et al. Increased SOD2 in the diaphragm contributes to exercise-induced protection against ventilator-induced diaphragm dysfunction. Redox Biol 2019;20:402-413.10.1016/j.redox.2018.10.005Suche in Google Scholar PubMed PubMed Central

156 Zhou H, Zhu PJ, Wang J, Zhu H, Ren J, Chen YD. Pathogenesis of cardiac ischemia reperfusion injury is associated with CK2α-disturbed mitochondrial homeostasis via suppression of FUNDC1-related mitophagy. Cell Death Differ 2018;25:1080-1093.10.1038/s41418-018-0086-7Suche in Google Scholar PubMed PubMed Central

157 Zhou H, Li DD, Zhu PJ, Hu SY, Hu N, Ma S, et al. Melatonin suppresses platelet activation and function against cardiac ischemia/reperfusion injury via PPARγ/FUNDC1/mitophagy pathways. J Pineal Res 2017;63. Epub 2017 Aug 21.10.1111/jpi.12438Suche in Google Scholar PubMed

158 Yang KC, Ma XC, Liu HY, Murphy J, Barger PM, Mann DL, et al. Tumor necrosis factor receptor-associated factor 2 mediates mitochondrial autophagy. Circ Heart Fail 2015;8:175-187.10.1161/CIRCHEARTFAILURE.114.001635Suche in Google Scholar PubMed PubMed Central

159 Flórido A, Saraiva N, Cerqueira S, Almeida N, Parsons M, Batinic-Haberle I, et al. The manganese(III) porphyrin MnTnHex-2-PyP5+ modulates intracellular ROS and breast cancer cell migration: Impact on doxorubicin-treated cells. Redox Biol 2019;20:367-378.10.1016/j.redox.2018.10.016Suche in Google Scholar PubMed PubMed Central

160 Jin QH, Li RB, Hu N, Xin T, Zhu PJ, Hu SY, et al. DUSP1 alleviates cardiac ischemia/reperfusion injury by suppressing the Mff-required mitochondrial fission and Bnip3-related mitophagy via the JNK pathways. Redox Biol 2018;14:576-587.10.1016/j.redox.2017.11.004Suche in Google Scholar PubMed PubMed Central

161 Zhang J, Nadtochiy SM, Urciuoli WR, Brookes PS. The cardioprotective compound cloxyquin uncouples mitochondria and induces autophagy. Am J Physiol Heart Circ Physiol 2016;310:H29-H38.10.1152/ajpheart.00926.2014Suche in Google Scholar PubMed PubMed Central

162 Esposti DD, Domart MC, Sebagh M, Harper F, Pierron G, Brenner C, et al. Autophagy is induced by ischemic preconditioning in human livers formerly treated by chemotherapy to limit necrosis. Autophagy 2010;6:172-174.10.4161/auto.6.1.10699Suche in Google Scholar PubMed

163 Otani H. Ischemic preconditioning: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal 2008;10:207-247.10.1089/ars.2007.1679Suche in Google Scholar PubMed

164 Ji J, Zhou XS, Xu P, Li YF, Shi HL, Chen DX, et al. Deficiency of apoptosisstimulating protein two of p53 ameliorates acute kidney injury induced by ischemia reperfusion in mice through upregulation of autophagy. J Cell Mol Med 2019;23:2457-2467.10.1111/jcmm.14094Suche in Google Scholar PubMed PubMed Central

165 Feng JX, Li HY, Zhang YF, Wang Q, Zhao SL, Meng P, et al. Mammalian STE20-like kinase 1 deletion alleviates renal ischaemia-reperfusion injury via modulating mitophagy and the AMPK-YAP signalling pathway. Cell Physiol Biochem 2018;51:2359-2376.10.1159/000495896Suche in Google Scholar PubMed

166 Li L, Wang ZV, Hill JA, Lin FM. New autophagy reporter mice reveal dynamics of proximal tubular autophagy. J Am Soc Nephrol 2014;25:305315.10.1681/ASN.2013040374Suche in Google Scholar PubMed PubMed Central

167 Yun JN, Puri R, Yang H, Lizzio MA, Wu CL, Sheng ZH, et al. MUL1 acts in parallel to the PINK1/parkin pathway in regulating mitofusin and compensates for loss of PINK1/parkin. eLife 2014;3:e01958.10.7554/eLife.01958Suche in Google Scholar PubMed PubMed Central

168 Mao K, Wang K, Zhao MT, Xu T, Klionsky DJ. Two MAPK-signaling pathways are required for mitophagy in Saccharomyces cerevisiae. J Cell Biol 2011;193:755-767.10.1083/jcb.201102092Suche in Google Scholar PubMed PubMed Central

169 Song MS, Chen Y, Gong GH, Murphy E, Rabinovitch PS, Dorn GW 2nd. Super-suppression of mitochondrial reactive oxygen species signaling impairs compensatory autophagy in primary mitophagic cardiomyopathy. Circ Res 2014;115:348-353.10.1161/CIRCRESAHA.115.304384Suche in Google Scholar PubMed PubMed Central

170 Yang YB, Li TY, Li ZB, Liu N, Yan YY, Liu B. Role of mitophagy in cardiovascular disease. Aging Dis 2020;11:419-437.10.14336/AD.2019.0518Suche in Google Scholar PubMed PubMed Central

171 Bugger H, Pfeil K. Mitochondrial ROS in myocardial ischemia reperfusion and remodeling. Biochim Biophys Acta Mol Basis Dis 2020;1866:165768.10.1016/j.bbadis.2020.165768Suche in Google Scholar PubMed

172 Bingol B, Sheng M. Mechanisms of mitophagy: PINK1, parkin, USP30 and beyond. Free Radic Biol Med 2016;100:210-222.10.1016/j.freeradbiomed.2016.04.015Suche in Google Scholar PubMed

173 Geisler S, Holmström KM, Skujat D, Fiesel FC, Rothfuss OC, Kahle PJ, et al. PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol 2010;12:119-131.10.1038/ncb2012Suche in Google Scholar PubMed

174 Zhang WL, Siraj S, Zhang R, Chen Q. Mitophagy receptor FUNDC1 regulates mitochondrial homeostasis and protects the heart from I/R injury. Autophagy 2017;13:1080-1081.10.1080/15548627.2017.1300224Suche in Google Scholar PubMed PubMed Central

175 Zhang J, Ney PA. Role of BNIP3 and NIX in cell death, autophagy, and mitophagy. Cell Death Differ 2009;16:939-946.10.1038/cdd.2009.16Suche in Google Scholar PubMed PubMed Central

176 Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, et al. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science 2011;331:456-461.10.1126/science.1196371Suche in Google Scholar PubMed PubMed Central

177 Saito T, Nah J, Oka SI, Mukai R, Monden Y, Maejima Y, et al. An alternative mitophagy pathway mediated by Rab9 protects the heart against ischemia. J Clin Invest 2019;129:802-819.10.1172/JCI122035Suche in Google Scholar PubMed PubMed Central

178 Hammerling BC, Najor RH, Cortez MQ, Shires SE, Leon LJ, Gonzalez ER, et al. A Rab5 endosomal pathway mediates Parkin-dependent mitochondrial clearance. Nat Commun 2017;8:14050.10.1038/ncomms14050Suche in Google Scholar PubMed PubMed Central

179 Zhu N, Li J, Li YL, Zhang YW, Du QB, Hao PY, et al. Berberine protects against simulated ischemia/reperfusion injury-induced H9C2 cardiomyocytes apoptosis in vitro and myocardial ischemia/reperfusion-induced apoptosis in vivo by regulating the mitophagy-mediated HIF-1α/BNIP3 pathway. Front Pharmacol 2020;11:367.10.3389/fphar.2020.00367Suche in Google Scholar PubMed PubMed Central

180 Li GY, Li JL, Shao RC, Zhao JH, Chen M. FUNDC1: a promising mitophagy regulator at the mitochondria-associated membrane for cardiovascular diseases. Front Cell Dev Biol 2021;9:788634.10.3389/fcell.2021.788634Suche in Google Scholar PubMed PubMed Central

181 Salabei JK, Hill BG. Mitochondrial fission induced by platelet-derived growth factor regulates vascular smooth muscle cell bioenergetics and cell proliferation. Redox Biol 2013;1:542-551.10.1016/j.redox.2013.10.011Suche in Google Scholar PubMed PubMed Central

182 Huynh DTN, Heo KS. Role of mitochondrial dynamics and mitophagy of vascular smooth muscle cell proliferation and migration in progression of atherosclerosis. Arch Pharm Res 2021;44:1051-1061.10.1007/s12272-021-01360-4Suche in Google Scholar PubMed

183 Zhao Y, Lv WT, Piao HY, Chu XJ, Wang H. Role of platelet-derived growth factor-BB (PDGF-BB) in human pulmonary artery smooth muscle cell proliferation. J Recept Signal Transduct Res 2014;34:254-260.10.3109/10799893.2014.908915Suche in Google Scholar PubMed

184 Ma YL, Zhou XJ, Gui MT, Yao L, Li JH, Chen XZ, et al. Mitophagy in hypertension-mediated organ damage. Front Cardiovasc Med 2024;10:1309863.10.3389/fcvm.2023.1309863Suche in Google Scholar PubMed PubMed Central

185 Forte M, Bianchi F, Cotugno M, Marchitti S, De Falco E, Raffa S, et al. Pharmacological restoration of autophagy reduces hypertension-related stroke occurrence. Autophagy 2020;16:1468-1481.10.1080/15548627.2019.1687215Suche in Google Scholar PubMed PubMed Central

186 Yoo SM, Jung YK. A molecular approach to mitophagy and mitochondrial dynamics. Mol Cells 2018;41:18-26.Suche in Google Scholar

187 Wang WJ, He QJ, Zhuang CC, Zhang HD, Fan X, Wang QY, et al. Apatinib through activating the RhoA/ROCK signaling pathway to cause dysfunction of vascular smooth muscle cells. Appl Biochem Biotechnol 2022;194:5367-5385.10.1007/s12010-022-04020-5Suche in Google Scholar PubMed

188 Hu XX, Zhang JT, Ma HX, Lian W, Song WQ, Du C, et al. The broad-spectrum deubiquitinating enzyme inhibitor PR-619 protects retinal ganglion cell and augments parkin-mediated mitophagy in experimental glaucoma. Sci Rep 2024;14:24654.10.1038/s41598-024-75562-3Suche in Google Scholar PubMed PubMed Central

189 Bal NB, Bostanci A, Sadi G, Dönmez MO, Uludag MO, Demirel-Yilmaz E. Resveratrol and regular exercise may attenuate hypertension-induced cardiac dysfunction through modulation of cellular stress responses. Life Sci 2022;296:120424.10.1016/j.lfs.2022.120424Suche in Google Scholar PubMed

190 Yu WJ, Gao BL, Li N, Wang JX, Qiu CT, Zhang GY, et al. Sirt3 deficiency exacerbates diabetic cardiac dysfunction: Role of Foxo3A-Parkin-mediated mitophagy. Biochim Biophys Acta Mol Basis Dis 2017;1863:1973-1983.10.1016/j.bbadis.2016.10.021Suche in Google Scholar PubMed

191 Sun W, Liu CX, Chen QH, Liu N, Yan YY, Liu B. SIRT3: a new regulator of cardiovascular diseases. Oxid Med Cell Longev 2018;2018:7293861.10.1155/2018/7293861Suche in Google Scholar PubMed PubMed Central

192 Yang CR, Xiao C, Ding ZR, Zhai XJ, Liu JY, Yu M. Canagliflozin mitigates diabetic cardiomyopathy through enhanced PINK1-parkin mitophagy. Int J Mol Sci 2024;25:7008.10.3390/ijms25137008Suche in Google Scholar PubMed PubMed Central

193 Cai C, Guo ZZ, Chang X, Li ZY, Wu F, He J, et al. Empagliflozin attenuates cardiac microvascular ischemia/reperfusion through activating the AMPKα1/ULK1/FUNDC1/mitophagy pathway. Redox Biol 2022;52:102288.10.1016/j.redox.2022.102288Suche in Google Scholar PubMed PubMed Central

194 Mizuno M, Kuno A, Yano T, Miki T, Oshima H, Sato T, et al. Empagliflozin normalizes the size and number of mitochondria and prevents reduction in mitochondrial size after myocardial infarction in diabetic hearts. Physiol Rep 2018;6:e13741.10.14814/phy2.13741Suche in Google Scholar PubMed PubMed Central

195 Xie ZL, Lau K, Eby B, Lozano P, He CY, Pennington B, et al. Improvement of cardiac functions by chronic metformin treatment is associated with enhanced cardiac autophagy in diabetic OVE26 mice. Diabetes 2011;60:1770-1778.10.2337/db10-0351Suche in Google Scholar PubMed PubMed Central

196 Luan Y, Luan Y, Feng Q, Chen X, Ren KD, Yang Y. Emerging role of mitophagy in the heart: therapeutic potentials to modulate mitophagy in cardiac diseases. Oxid Med Cell Longev 2021;2021:3259963.10.1155/2021/3259963Suche in Google Scholar PubMed PubMed Central

197 Zhou JR, Lu Y, Li ZL, Wang ZH, Kong WH, Zhao J. Sphingosylphos-phorylcholine ameliorates doxorubicin-induced cardiotoxicity in zebrafish and H9c2 cells by reducing excessive mitophagy and mitochondrial dysfunction. Toxicol Appl Pharmacol 2022;452:116207.10.1016/j.taap.2022.116207Suche in Google Scholar PubMed

198 Xiao DD, Chang WG, Ding W, Wang Y, Fa HG, Wang JX. Enhanced mitophagy mediated by the YAP/Parkin pathway protects against DOX-induced cardiotoxicity. Toxicol Lett 2020;330:96-107.10.1016/j.toxlet.2020.05.015Suche in Google Scholar PubMed

199 Yu WJ, Deng DW, Li Y, Ding KH, Qian QF, Shi HJ, et al. Cardiomyocytespecific Tbk1 deletion aggravated chronic doxorubicin cardiotoxicity via inhibition of mitophagy. Free Radic Biol Med 2024;222:244-258.10.1016/j.freeradbiomed.2024.06.009Suche in Google Scholar PubMed

200 Li WL, Wang XP, Liu TH, Zhang Q, Cao J, Jiang YY, et al. Harpagoside protects against doxorubicin-induced cardiotoxicity via P53-parkinmediated mitophagy. Front Cell Dev Biol 2022;10:813370.10.3389/fcell.2022.813370Suche in Google Scholar PubMed PubMed Central

201 Peng KZ, Zeng CY, Gao YQ, Liu BL, Li LY, Xu K, et al. Overexpressed SIRT6 ameliorates doxorubicin-induced cardiotoxicity and potentiates the therapeutic efficacy through metabolic remodeling. Acta Pharm Sin B 2023;13:2680-2700.10.1016/j.apsb.2023.03.019Suche in Google Scholar PubMed PubMed Central

202 Amanakis G, Kleinbongard P, Heusch G, Skyschally A. Attenuation of ST-segment elevation after ischemic conditioning maneuvers reflects cardioprotection online. Basic Res Cardiol 2019;114:22.10.1007/s00395-019-0732-3Suche in Google Scholar PubMed

203 Honda T, He Q, Wang FF, Redington AN. Acute and chronic remote ischemic conditioning attenuate septic cardiomyopathy, improve cardiac output, protect systemic organs, and improve mortality in a lipopolysaccharide-induced sepsis model. Basic Res Cardiol 2019;114:15.10.1007/s00395-019-0724-3Suche in Google Scholar PubMed

204 Opferman JT, Kothari A. Anti-apoptotic BCL-2 family members in development. Cell Death Differ 2018;25:37-45.10.1038/cdd.2017.170Suche in Google Scholar PubMed PubMed Central

205 Pohl SÖ, Pervaiz S, Dharmarajan A, Agostino M. Gene expression analysis of heat-shock proteins and redox regulators reveals combinatorial prognostic markers in carcinomas of the gastrointestinal tract. Redox Biol 2019;25:101060.10.1016/j.redox.2018.11.018Suche in Google Scholar PubMed PubMed Central

206 Someda M, Kuroki S, Miyachi H, Tachibana M, Yonehara S. Caspase-8, receptor-interacting protein kinase 1 (RIPK1), and RIPK3 regulate retinoic acid-induced cell differentiation and necroptosis. Cell Death Differ 2020;27:1539-1553.10.1038/s41418-019-0434-2Suche in Google Scholar PubMed PubMed Central

207 Xiong Y, Li LL, Zhang LT, Cui YY, Wu CY, Li H, et al. The bromodomain protein BRD4 positively regulates necroptosis via modulating MLKL expression. Cell Death Differ 2019;26:1929-1941.10.1038/s41418-018-0262-9Suche in Google Scholar PubMed PubMed Central

208 Morciano G, Bonora M, Campo G, Aquila G, Rizzo P, Giorgi C, et al. Mechanistic role of mPTP in ischemia-reperfusion injury. Adv Exp Med Biol 2017:982:169-189.10.1007/978-3-319-55330-6_9Suche in Google Scholar PubMed

209 Pozzer D, Varone E, Chernorudskiy A, Schiarea S, Missiroli S, Giorgi C, et al. A maladaptive ER stress response triggers dysfunction in highly active muscles of mice with SELENON loss. Redox Biol 2019;20:354-366.10.1016/j.redox.2018.10.017Suche in Google Scholar PubMed PubMed Central

210 Frank T, Tuppi M, Hugle M, Dötsch V, van Wijk SJL, Fulda S. Cell cycle arrest in mitosis promotes interferon-induced necroptosis. Cell Death Differ 2019;26:2046-2060.10.1038/s41418-019-0298-5Suche in Google Scholar PubMed PubMed Central

211 Robinson N, Ganesan R, Hegedűs C, Kovács K, Kufer TA, Virág L. Programmed necrotic cell death of macrophages: Focus on pyroptosis, necroptosis, and parthanatos. Redox Biol 2019;26:101239.10.1016/j.redox.2019.101239Suche in Google Scholar PubMed PubMed Central

212 Zhou X, Wu YJ, Ye LF, Wang YT, Zhang KM, Wang LJ, et al. Aspirin alleviates endothelial gap junction dysfunction through inhibition of NLRP3 inflammasome activation in LPS-induced vascular injury. Acta Pharm Sin B 2019;9:711-723.10.1016/j.apsb.2019.02.008Suche in Google Scholar PubMed PubMed Central

213 Kumar D, Jugdutt BI. Apoptosis and oxidants in the heart. J Lab Clin Med 2003;142:288-297.10.1016/S0022-2143(03)00148-3Suche in Google Scholar PubMed

214 Kleinbongard P, Skyschally A, Gent S, Pesch M, Heusch G. STAT3 as a common signal of ischemic conditioning: a lesson on “rigor and reproducibility” in preclinical studies on cardioprotection. Basic Res Cardiol 2017;113:3.10.1007/s00395-017-0660-zSuche in Google Scholar PubMed

215 Quispe RL, Jaramillo ML, Galant LS, Engel D, Dafre AL, Teixeira da Rocha JB, et al. Diphenyl diselenide protects neuronal cells against oxidative stress and mitochondrial dysfunction: Involvement of the glutathionedependent antioxidant system. Redox Biol 2019;20:118-129.10.1016/j.redox.2018.09.014Suche in Google Scholar PubMed PubMed Central

216 Zhang T, Zhang Y, Cui MY, Jin L, Wang YM, Lv FX, et al. CaMKII is a RIP3 substrate mediating ischemia- and oxidative stress-induced myocardial necroptosis. Nat Med 2016;22:175-182.10.1038/nm.4017Suche in Google Scholar PubMed

217 Zhou H, Li DD, Zhu PJ, Ma Q, Toan S, Wang J, et al. Inhibitory effect of melatonin on necroptosis via repressing the Ripk3-PGAM5-CypD-mPTP pathway attenuates cardiac microvascular ischemia-reperfusion injury. J Pineal Res 2018;65:e12503.10.1111/jpi.12503Suche in Google Scholar PubMed

218 Zhou H, Zhu PJ, Guo J, Hu N, Wang SY, Li DD, et al. Ripk3 induces mitochondrial apoptosis via inhibition of FUNDC1 mitophagy in cardiac IR injury. Redox Biol 2017;13:498-507.10.1016/j.redox.2017.07.007Suche in Google Scholar PubMed PubMed Central

219 Oerlemans MIFJ, Koudstaal S, Chamuleau SA, de Kleijn DP, Doevendans PA, Sluijter JPG. Targeting cell death in the reperfused heart: pharmacological approaches for cardioprotection. Int J Cardiol 2013;165:410-422.10.1016/j.ijcard.2012.03.055Suche in Google Scholar PubMed

220 Schmidt HM, Kelley EE, Straub AC. The impact of xanthine oxidase (XO) on hemolytic diseases. Redox Biol 2019;21:101072.10.1016/j.redox.2018.101072Suche in Google Scholar PubMed PubMed Central

221 Luedde M, Lutz M, Carter N, Sosna J, Jacoby C, Vucur M, et al. RIP3, a kinase promoting necroptotic cell death, mediates adverse remodelling after myocardial infarction. Cardiovasc Res 2014;103:206-216.10.1093/cvr/cvu146Suche in Google Scholar PubMed

222 Lalaoui N, Lindqvist LM, Sandow JJ, Ekert PG. The molecular relationships between apoptosis, autophagy and necroptosis. Semin Cell Dev Biol 2015;39:63-69.10.1016/j.semcdb.2015.02.003Suche in Google Scholar PubMed

223 Bertrand MJM, Milutinovic S, Dickson KM, Ho WC, Boudreault A, Durkin J, et al. cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination. Mol Cell 2008;30:689-700.10.1016/j.molcel.2008.05.014Suche in Google Scholar PubMed

224 Graczyk-Jarzynka A, Goral A, Muchowicz A, Zagozdzon R, Winiarska M, Bajor M, et al. Inhibition of thioredoxin-dependent H2O2 removal sensitizes malignant B-cells to pharmacological ascorbate. Redox Biol 2019;21:101062.10.1016/j.redox.2018.11.020Suche in Google Scholar PubMed PubMed Central

225 Ogasawara M, Yano T, Tanno M, Abe K, Ishikawa S, Miki T, et al. Suppression of autophagic flux contributes to cardiomyocyte death by activation of necroptotic pathways. J Mol Cell Cardiol 2017;108:203-213.10.1016/j.yjmcc.2017.06.008Suche in Google Scholar PubMed

226 Tian M, Yuan YC, Li JY, Gionfriddo MR, Huang RC. Tumor necrosis factor-α and its role as a mediator in myocardial infarction: a brief review. Chronic Dis Transl Med 2015;1:18-26.10.1016/j.cdtm.2015.02.002Suche in Google Scholar PubMed PubMed Central

227 Bulluck H, Rosmini S, Abdel-Gadir A, White SK, Bhuva AN, Treibel TA, et al. Residual myocardial iron following intramyocardial hemorrhage during the convalescent phase of reperfused ST-segment-elevation myocardial infarction and adverse left ventricular remodeling. Circ Cardiovasc Imaging 2016;9:e004940.10.1161/CIRCIMAGING.116.004940Suche in Google Scholar PubMed PubMed Central

228 Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci U S A 2016;113:E4966-E4975.10.1073/pnas.1603244113Suche in Google Scholar PubMed PubMed Central

229 Fang XX, Wang H, Han D, Xie EJ, Yang X, Wei JY, et al. Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci U S A 2019;116:2672-2680.10.1073/pnas.1821022116Suche in Google Scholar PubMed PubMed Central

230 Shen YH, Shen XY, Wang SL, Zhang YY, Wang Y, Ding Y, et al. Protective effects of Salvianolic acid B on rat ferroptosis in myocardial infarction through upregulating the Nrf2 signaling pathway. Int Immunopharmacol 2022;112:109257.10.1016/j.intimp.2022.109257Suche in Google Scholar PubMed

231 Li N, Jiang WY, Wang W, Xiong R, Wu XJ, Geng Q. Ferroptosis and its emerging roles in cardiovascular diseases. Pharmacol Res 2021;166:105466.10.1016/j.phrs.2021.105466Suche in Google Scholar PubMed

232 Han X, Zhang J, Liu J, Wang H, Du F, Zeng X, et al. Targeting ferroptosis: a novel insight against myocardial infarction and ischemia-reperfusion injuries. Apoptosis 2023;28:108-123.10.1007/s10495-022-01785-2Suche in Google Scholar PubMed

233 Yin ZY, You SL, Zhang S, Zhang LL, Wu BQ, Huang XY, et al. Atorvastatin rescues vascular endothelial injury in hypertension by WWP 2-mediated ubiquitination and degradation of ATP5A. Biomed Pharmacother 2023;166:115228.10.1016/j.biopha.2023.115228Suche in Google Scholar PubMed

234 Zahedi N, Pourajam S, Zaker E, Kouhpayeh S, Mirbod SM, Tavangar M, et al. The potential therapeutic impacts of trehalose on cardiovascular diseases as the environmental-influenced disorders: an overview of contemporary findings. Environ Res 2023;226:115674.10.1016/j.envres.2023.115674Suche in Google Scholar PubMed

235 Song ZC, Wang JY, Zhang LJ. Ferroptosis: a new mechanism in diabetic cardiomyopathy. Int J Med Sci 2024;21:612-622.10.7150/ijms.88476Suche in Google Scholar PubMed PubMed Central

236 Banerjee S, Lu SL, Jain A, Wang I, Tao H, Srinivasan S, et al. Targeting PKCα alleviates iron overload in diabetes and hemochromatosis through the inhibition of ferroportin. Blood 2024;144:1433-1444.10.1182/blood.2024023829Suche in Google Scholar PubMed PubMed Central

237 Chen YC, Meng ZH, Li Y, Liu SB, Hu P, Luo E. Advanced glycation end products and reactive oxygen species: uncovering the potential role of ferroptosis in diabetic complications. Mol Med 2024;30:141.10.1186/s10020-024-00905-9Suche in Google Scholar PubMed PubMed Central

238 Jin EJ, Jo Y, Wei SB, Rizzo M, Ryu D, Gariani K. Ferroptosis and iron metabolism in diabetes: pathogenesis, associated complications, and therapeutic implications. Front Endocrinol (Lausanne) 2024;15:1447148.10.3389/fendo.2024.1447148Suche in Google Scholar PubMed PubMed Central

239 Xiao JL, Liu HY, Sun CC, Tang CF. Regulation of Keap1-Nrf2 signaling in health and diseases. Mol Biol Rep 2024;51:809.10.1007/s11033-024-09771-4Suche in Google Scholar PubMed

240 Huang K, Luo X, Liao B, Li G, Feng J. Insights into SGLT2 inhibitor treatment of diabetic cardiomyopathy: focus on the mechanisms. Cardiovasc Diabetol 2023;22:86.10.1186/s12933-023-01816-5Suche in Google Scholar PubMed PubMed Central

241 Tan Y, Zhang ZG, Zheng C, Wintergerst KA, Keller BB, Cai L. Mechanisms of diabetic cardiomyopathy and potential therapeutic strategies: preclinical and clinical evidence. Nat Rev Cardiol 2020;17:585-607.10.1038/s41569-020-0339-2Suche in Google Scholar PubMed PubMed Central

242 Wang MY, Tang JY, Zhang SW, Pang KM, Zhao YJ, Liu N, et al. Exogenous H2S initiating Nrf2/GPx4/GSH pathway through promoting Syvn1-Keap1 interaction in diabetic hearts. Cell Death Discov 2023;9:394.10.1038/s41420-023-01690-wSuche in Google Scholar PubMed PubMed Central

243 Chen Y, Li XS, Hua YY, Ding Y, Meng GL, Zhang W. RIPK3-mediated necroptosis in diabetic cardiomyopathy requires CaMKII activation. Oxid Med Cell Longev 2021;2021:6617816.10.1155/2021/6617816Suche in Google Scholar PubMed PubMed Central

244 Song S, Ding Y, Dai GL, Zhang Y, Xu MT, Shen JR, et al. Sirtuin 3 deficiency exacerbates diabetic cardiomyopathy via necroptosis enhancement and NLRP3 activation. Acta Pharmacol Sin 2021;42:230-241.10.1038/s41401-020-0490-7Suche in Google Scholar PubMed PubMed Central

245 Gong WW, Zhang SP, Chen Y, Shen JR, Zheng YY, Liu X, et al. Protective role of hydrogen sulfide against diabetic cardiomyopathy via alleviating necroptosis. Free Radic Biol Med 2022;181:29-42.10.1016/j.freeradbiomed.2022.01.028Suche in Google Scholar PubMed

246 Zhang SP, Shen JR, Zhu Y, Zheng YY, San WQ, Cao DY, et al. Hydrogen sulfide promoted retinoic acid-related orphan receptor α transcription to alleviate diabetic cardiomyopathy. Biochem Pharmacol 2023;215:115748.10.1016/j.bcp.2023.115748Suche in Google Scholar PubMed

247 Wang X, Chen XX, Zhou WQ, Men HB, Bao T, Sun YK, et al. Ferroptosis is essential for diabetic cardiomyopathy and is prevented by sulforaphane via AMPK/NRF2 pathways. Acta Pharm Sin B 2022;12:708-722.10.1016/j.apsb.2021.10.005Suche in Google Scholar PubMed PubMed Central

248 Hwang S, Kim SH, Yoo KH, Chung MH, Lee JW, Son KH. Exogenous 8-hydroxydeoxyguanosine attenuates doxorubicin-induced cardiotoxicity by decreasing pyroptosis in H9c2 cardiomyocytes. BMC Mol Cell Biol 2022;23:55.10.1186/s12860-022-00454-1Suche in Google Scholar PubMed PubMed Central

249 Christidi E, Brunham LR. Regulated cell death pathways in doxorubicin-induced cardiotoxicity. Cell Death Dis 2021;12:339.10.1038/s41419-021-03614-xSuche in Google Scholar PubMed PubMed Central

250 Zhang L, Jiang YH, Fan CD, Zhang Q, Jiang YH, Li Y, et al. MCC950 attenuates doxorubicin-induced myocardial injury in vivo and in vitro by inhibiting NLRP3-mediated pyroptosis. Biomed Pharmacother 2021;143:112133.10.1016/j.biopha.2021.112133Suche in Google Scholar PubMed

251 Fang GY, Li XC, Yang FY, Huang T, Qiu CM, Peng K, et al. Amentoflavone mitigates doxorubicin-induced cardiotoxicity by suppressing cardiomyocyte pyroptosis and inflammation through inhibition of the STING/NLRP3 signalling pathway. Phytomedicine 2023;117:154922.10.1016/j.phymed.2023.154922Suche in Google Scholar PubMed

252 Tambe PK, Mathew AJ, Bharati S. Cardioprotective potential of mitochondria-targeted antioxidant, mito-TEMPO, in 5-fluorouracil-induced cardiotoxicity. Cancer Chemother Pharmacol 2023;91:389-400.10.1007/s00280-023-04529-4Suche in Google Scholar PubMed PubMed Central

253 Sabouny R, Shutt TE. Reciprocal regulation of mitochondrial fission and fusion. Trends Biochem Sci 2020;45:564-577.10.1016/j.tibs.2020.03.009Suche in Google Scholar PubMed

254 Pitceathly RDS, Keshavan N, Rahman J, Rahman S. Moving towards clinical trials for mitochondrial diseases. J Inherit Metab Dis 2021;44:22-41.10.1002/jimd.12281Suche in Google Scholar PubMed PubMed Central

255 Baharvand F, Habibi Roudkenar M, Pourmohammadi-Bejarpasi Z, Najafi-Ghalehlou N, Feizkhah A, Bashiri Aliabadi S, et al. Safety and efficacy of platelet-derived mitochondrial transplantation in ischaemic heart disease. Int J Cardiol 2024;410:132227.10.1016/j.ijcard.2024.132227Suche in Google Scholar PubMed

Published Online: 2025-06-20

© 2025 Miao Zhang, Tong Zhang, Rongjun Zou, Kunyang He, Ru Huang, Jingrui Feng, Jinlin Hu, Teng Ge, Xiaoping Fan, Hao Zhou, Yang Chen, published by De Gruyter on behalf of the SMP

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

Heruntergeladen am 19.11.2025 von https://www.degruyterbrill.com/document/doi/10.1515/jtim-2025-0030/html?lang=de
Button zum nach oben scrollen