Open Access

Role of RIPK3‑CaMKII‑mPTP signaling pathway‑mediated necroptosis in cardiovascular diseases (Review)

  • Authors:
    • Sheng Chen
    • Senhong Guan
    • Zhaohan Yan
    • Fengshan Ouyang
    • Shuhuan Li
    • Lanyuan Liu
    • Jiankai Zhong
  • View Affiliations

  • Published online on: August 30, 2023     https://doi.org/10.3892/ijmm.2023.5301
  • Article Number: 98
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Necroptosis, which is distinct from apoptosis and necrosis, serves a crucial role in ontogeny and the maintenance of homeostasis. In the last decade, it has been demonstrated that the pathogenesis of cardiovascular diseases is also linked to necroptosis. Receptor interaction protein kinase (RIPK) 1, RIPK3 and mixed lineage kinase domain‑like protein serve vital roles in necroptosis. In addition to the aforementioned necroptosis‑related components, calcium/calmodulin‑dependent protein kinase II (CaMKII) has been identified as a novel substrate for RIPK3 that promotes the opening of the mitochondrial permeability transition pore (mPTP), and thus, mediates necroptosis of myocardial cells through the RIPK3‑CaMKII‑mPTP signaling pathway. The present review provides an overview of the current knowledge of the RIPK3‑CaMKII‑mPTP‑mediated necroptosis signaling pathway in cardiovascular diseases, focusing on the role of the RIPK3‑CaMKII‑mPTP signaling pathway in acute myocardial infarction, ischemia‑reperfusion injury, heart failure, abdominal aortic aneurysm, atherosclerosis, diabetic cardiomyopathy, hypertrophic cardiomyopathy, atrial fibrillation, and the cardiotoxicity associated with antitumor drugs and other chemicals. Finally, the present review discusses the research status of drugs targeting the RIPK3‑CaMKII‑mPTP signaling pathway.

1. Introduction

Cardiovascular diseases are associated with a high morbidity rate and are the leading cause of mortality, thus posing a serious threat to human health (1). Numerous factors, including genetic and environmental factors, metabolic disorders, inflammation, oxidative stress, mitochondrial dysfunction and cell death, are involved in the pathogenesis of cardiovascular disease, and myocardial cell loss caused by cell death serves a crucial role in cardiovascular disease (2,3). At present, cell death has been classified into several distinct forms, including apoptosis, autophagy, pyroptosis, ferroptosis and necroptosis (4). Necroptosis is a recently reported pathway of programmed cell necrosis regulated by the kinases receptor interaction protein kinase (RIPK)3 and mixed lineage kinase domain-like protein (MLKL) that, in contrast to apoptosis, induces a proinflammatory state (5). Necroptosis can be experimentally activated under apoptosis-deficient conditions (6,7). When RIPK1 is activated, cells with higher levels of FAS-associated death domain protein (FADD) and caspase8 than MLKL may preferentially induce apoptosis. Conversely, when caspase8 activity is compromised, recruitment of RIPK3 to the ripoptosome complex promotes MLKL-mediated necroptosis, and necroptosis is another form of programmed cell death that closely resembles necrosis and is more proinflammatory than apoptosis (8). Table I shows the differences among necroptosis, apoptosis and necrosis.

Table I

Differences among necroptosis, apoptosis and necrosis.

Table I

Differences among necroptosis, apoptosis and necrosis.

DifferencesNecroptosisApoptosisNecrosis
CauseActivated by death receptor ligands (4,5)Physiological or pathological changes (4)Pathological changes or severe injuries (4)
Gene regulationProgrammed (4,5)Programmed (4,5)Unprogrammed (4)
Cell morphologyCell swelling (5)Cell shrinkage (5)Cell swelling (4)
Cell membraneMembrane rupture (5)Membrane integrity (5)Membrane rupture (4)
OrganelleOrganelle swelling and rupture (5)No obvious changes (5)Organelle swelling and rupture (4)
Apoptotic bodiesNo apoptotic bodies (4,5)Apoptotic bodies (4,5)No apoptotic bodies (4)
Inflammatory responseInflammatory response (5-8)No inflammatory response (5-8)Inflammatory response (4)
Key regulatory proteinsRIPK1, RIPK3, MLKL and CaMKII (5-8)Caspase family (4-8)Unregulated (4)

[i] RIPK1, receptor interaction protein kinase 1; RIPK3, receptor interaction protein kinase 3; MLKL, mixed lineage kinase domain-like protein; CaMKII, calcium/calmodulin-dependent protein kinase II.

Previous studies have reported that the RIPK3/MLKL signaling pathway is a classical necroptosis pathway and serves a critical role in cardiovascular diseases (9-12). Zhang et al (13) demonstrated that, in addition to MLKL as a substrate for RIPK3, calcium/calmodulin-dependent protein kinase II (CaMKII) could also act as a substrate for RIPK3 to mediate necroptosis in myocardial cells via the RIPK3-CaMKII-mitochondrial permeability transition pore (mPTP) signaling pathway. The authors also found that deficiency of RIPK3 prevented ischemia-reperfusion (I/R) injury and reduced doxorubicin (DOX)-induced CaMKII activation, which inhibited necroptosis in the myocardium (13).

With the advancement of research regarding the mechanism of necroptosis, the RIPK3-CaMKII-mPTP-mediated necroptosis signaling pathway has been revealed to serve important roles in other cardiovascular diseases, such as acute myocardial infarction (AMI) (14-16), heart failure (HF) (17), abdominal aortic aneurysm (AAA) (18), atherosclerosis (AS) (19), diabetic cardiomyopathy (DCM) (20,21), hypertrophic cardiomyopathy (HCM) (22,23) and atrial fibrillation (AF) (24), and cardiotoxicity of antitumor drugs and other chemicals (13,25-27), and targeting of the RIPK3-CaMKII-mPTP-mediated necroptosis signaling pathway may provide benefits for the treatment of cardiovascular diseases (13). The present review particularly focuses on novel developments regarding RIPK3-CaMKII-mPTP signaling pathway-mediated necroptosis in cardiovascular diseases, outlining the molecular mechanisms that activate the RIPK3-CaMKII-mPTP signaling pathway in different physiological and pathological contexts. With an understanding of the key components and the regulatory mechanisms of the RIPK3-CaMKII-mPTP signaling pathway, pharmacological intervention strategies targeting the pathway are examined and their therapeutic potential in the treatment of cardiovascular diseases is discussed.

2. Overview of the RIPK3-CaMKII-mPTP signaling pathway

Necroptosis can be initiated by a variety of triggers, including the tumor necrosis factor receptor (TNFR) and Toll-like receptor families, the intracellular DNA/RNA receptor Z-DNA binding protein 1, and interferon (28,29). One of the most well-studied and abundant signaling pathways is the TNF-α-mediated signaling pathway, in which TNF-α binds to TNFR1, RIPK1, DD-containing adaptor protein TNFR1-associated death domain protein (TRADD), TNF receptor-associated factors 2 and 5, and cellular inhibitor of apoptosis proteins 1 and 2, which are recruited to form complex I and initiate necroptosis (Fig. 1) (30,31). RIPK3 is a member of the serine/threonine RIPK family, whose structural components include the N-terminal kinase structural domain and the C-terminal RIP homotypic interaction motif (RHIM) (32). When RHIM-containing RIPK1 is deubiquitinated and activated, RIPK1 assembles a cytoplasmic signaling complex that contains RIPK3, FADD, caspase8 and TRADD (33-35). Under conditions of impaired caspase8 activity, RIPK1 self-phosphorylation promotes the phosphorylation and activation of RIPK3, thereby mediating the phosphorylation and activation of MLKL, which is then oligomerized and transported to the plasma membrane, where the N-terminal four-helix bundle structural domain of MLKL can mediate cell membrane disruption (36), leading to cellular micropore formation and eventually resulting in cell swelling and lysis, and the triggering of subsequent necroptosis (Fig. 1) (35). Sun et al (37) have demonstrated that RIPK3 and MLKL are essential for necroptosis. However, inhibition of MLKL does not completely block cardiomyocyte necroptosis, indicating that there are other signaling pathways involved in necroptosis (13).

Figure 1

Overview of the RIPK3-CaMKII-mPTP signaling pathway. When TNF-α binds to TNFR1, TNFR1 recruits the adaptor protein TRADD, which recruits other proteins to form complex I. When RIPK1 is deubiquitinated and activated, RIPK1 assembles a cytoplasmic signaling complex that includes RIPK3, FADD, caspase8 and TRADD. Under conditions of impaired caspase8 activity, RIPK1 self-phosphorylation promotes the phosphorylation and activation of RIPK3, thereby mediating the phosphorylation and activation of MLKL, which is subsequently oligomerized and transported to the plasma membrane, ultimately leading to the occurrence of necroptosis. In addition, p-RIPK3 can also activate CaMKII. When CaMKII is activated, it causes ER stress, which then leads to Ca2+ leakage. Excess Ca2+ in the cytoplasm enters the mitochondria via the MCU, leading to mitochondrial Ca2+ overload, promoting the opening of mPTP, which leads to the loss of ΔΨm and reduced ATP synthesis, and ultimately leads to necroptosis. RIPK3, receptor interaction protein kinase 3; CaMKII, calcium/calmodulin-dependent protein kinase II; MLKL, mixed lineage kinase domain-like protein; mPTP, mitochondrial permeability transition pore; TNFR1, tumor necrosis factor receptor 1; RIPK1, receptor interaction protein kinase 1; TRADD, DD-containing adaptor protein TNFR1-associated death domain protein; TRAF2/5, TNF receptor-associated factors 2 and 5; cIAP1/2, cellular inhibitor of apoptosis proteins 1 and 2; FADD, FAS-associated death domain protein; P, phosphorylated; ROS, reactive oxygen species; PYGL, glycogen phosphorylase; GLUL, glutamate-ammonia ligase; GLUD1, glutamate dehydrogenase 1; ox-, oxidized; RyR2, ryanodine receptor 2; ER, endoplasmic reticulum; MCU, mitochondrial Ca2+ uniporter; ΔΨm, mitochondrial inner membrane potential.

CaMKII is a serine and threonine kinase with four isoforms encoded by four genes (α, β, γ and δ), namely CaMKIIα, CaMKIIβ, CaMKIIγ and CaMKIIδ, of which CaMKIIδ is mainly expressed in cardiomyocytes (38). CaMKII holoenzymes exist as multimers of 12 subunits, each consisting of three structural domains: The N-terminal catalytic domain, a central regulatory domain and a C-terminal association domain (39). The variable regions are in the central regulatory domain, and their composition differs according to different CaMKII splice variants (39). Normally, CaMKII can regulate various biological functions of the heart, by regulating myocardial cell membrane excitability, maintaining cell Ca2+ homeostasis, participating in excitation contraction coupling between myocardial cells and regulating the cell cycle, so it is an important regulator in cardiomyocytes (38,40). However, sustained CaMKII activation is considered to serve a central role in the pathogenesis of cardiovascular diseases (38,41). Emerging evidence suggests that CaMKII can act as a novel substrate for RIPK3 to mediate necroptosis (13). When RIPK3 binds to CaMKII, it can mediate the phosphorylation of the Thr287 site of CaMKII through its kinase activity, and RIPK3 can also promote the production of reactive oxygen species (ROS) by increasing the activities of glycogen phosphorylase, glutamate-ammonia ligase and glutamate dehydrogenase 1, and then oxidize the Met281/282 site of CaMKII, and both the oxidation and phosphorylation of CaMKII are involved in RIPK3-mediated necroptosis (Fig. 1) (13,42).

Ryanodine receptor 2 (RyR2) is a major calcium release channel on the sarcoplasmic reticulum (SR) of myocardial cells (43). Gui et al (42) indicated that RyR2 is one of the major downstream targets of CaMKII in myocardial cells and serves an important role in regulating Ca2+ homeostasis. When CaMKII is activated, it will phosphorylate the RyR2-Ser2814 site, leading to endoplasmic reticulum (ER) stress and subsequent Ca2+ leak through RyR2 from the SR (42,44,45). CaMKII can also phosphorylate the mitochondrial Ca2+ uniporter (MCU) and increase its ability to transport Ca2+ (46). Excessive Ca2+ in the cytoplasm will enter the mitochondria through the MCU, causing mitochondrial Ca2+ overload, and thus, promoting the opening of the mPTP (13,41,47,48). Recent studies have reported that the mPTP is part of the final pathway of necroptosis (49,50). The opening of the mPTP leads to loss of mitochondrial inner membrane potential (ΔΨm) and the rapid dissipation of proton gradients across the inner membrane, disrupting mitochondrial respiratory function and leading to metabolic shutdown, thereby causing mitochondrial structural and functional disorders that result in mitochondrial damage and mitochondrial swelling; this leads to decreased ATP synthesis and increased ROS generation, ultimately causing organelle swelling, cell swelling and loss of plasma membrane integrity, which leads to the occurrence of necroptosis, ultimately triggering the innate immune inflammatory response and leading to tissue damage (10,13,38,51). The opening of the mPTP and ROS generation can further increase CaMKII activity, thus forming a positive feedback loop and accelerating necroptosis (Fig. 1) (13,40). The opening of the mPTP and the accumulation of ROS also link death receptor-mediated necroptosis to the mitochondrial-dependent necrosis pathway, suggesting that necroptosis can be induced through the mitochondrial pathway, further enriching the mechanism of necroptosis (50,52).

3. Effects of RIPK3-CaMKII-mPTP signaling pathway activation in cardiovascular diseases

AMI and I/R injury

AMI, a common disease, is caused by acute or subacute coronary artery occlusion (53). Prolonged ischemia leads to myocardial cell hypoxia and subsequent myocardial cell death, which is a serious threat to patient health and life (54,55). Reperfusion therapy is the preferred treatment strategy for patients with AMI (56). Although early reperfusion of the ischemic myocardium in coronary artery infarction can rescue the agonal cardiac muscle, it causes subsequent myocardial I/R injury, which lowers the protective effect of reperfusion therapy and restricts the development of this therapy (56,57).

With the continuous development of research, it has been revealed that necroptosis serves an important role in AMI and I/R injury (10,58-60). Luedde et al (61) generated a mouse AMI model by permanent ligation of the left anterior descending coronary artery and subsequently found that the RIPK3 protein level was markedly upregulated in ischemic heart tissue, while the upregulation of RIPK3 could promote myocardial damage, indicating that RIPK3 was activated during AMI. Furthermore, knockdown of RIPK3 improved cardiac function, attenuated the inflammatory response, alleviated mitochondrial injury, decreased ROS production and inhibited cardiac necroptosis (61). Zhang et al (62) found that upregulation of microRNA-325-3p inhibited the progression of AMI in a mouse model by preventing RIPK3 activation and subsequent necroptosis. Oerlemans et al (63) reported that when myocardial I/R injury occurred, RIPK3 phosphorylation was elevated, and inhibition of RIPK3-mediated necroptosis could reduce myocardial cell loss and contribute to increased resistance to I/R injury. Collectively, the aforementioned studies suggest that the RIPK3-MLKL-mediated classical necroptosis pathway serves an important role in I/R injury.

Multiple studies have demonstrated that, in a model of myocardial I/R injury, in addition to the classical RIPK3-MLKL signaling pathway, RIPK3 can mediate the necroptosis of myocardial cells through CaMKII, and inhibition of CaMKII can also prevent the necroptosis of myocardial cells induced by I/R injury (14-16). Thus, these studies suggest that CaMKII serves a crucial role in the pathogenesis of I/R injury. Similarly, studies have demonstrated that the cellular levels of RIPK1, RIPK3 and CaMKII were markedly increased in hypoxic myocardial cells (14,15). When I/R injury occurred, both RIPK3 and CaMKII were present in myocardial cells, and the binding of the two was enhanced, while the phosphorylation of CaMKII increased with the upregulation of RIPK3, indicating that RIPK3 could directly bind and phosphorylate CaMKII; however, after inhibition of CaMKII activation using KN-93, a small-molecule inhibitor of CaMKII, myocardial cell mortality was still reduced even when RIPK3 was upregulated (13). The study further identified two pathways of CaMKII activation by RIPK3: Direct phosphorylation of RIPK3 or indirect ROS-mediated oxidation could activate CaMKII, and inhibition of either CaMKII oxidation or phosphorylation markedly inhibited RIPK3-mediated necroptosis (13). Therefore, direct phosphorylation of RIPK3 and indirect ROS-mediated oxidation mediate RIPK3-induced myocardial cell necroptosis.

A previous study has demonstrated that CaMKII can promote Ca2+ entry into mitochondria by increasing the MCU, thus causing the opening of the mPTP (47). Cyclophilin D (CypD), a peptidylprolyl cis-trans isomerase, is a key component of the mPTP complex, and its activity regulates the open state of the mPTP and is essential for mPTP-induced cell death (64). Cyclosporin A (CsA) is a specific inhibitor of CypD that binds specifically to CypD, thereby preventing mPTP opening (65). The use of CsA has been found to be clinically efficacious in myocardial I/R injury by preventing mPTP opening while reducing the occurrence of mitochondrial disruption and necroptosis (47,66). Therefore, we hypothesized that mPTP opening is part of the final pathway of necroptosis, and it is related to myocardial cell death and myocardial injury caused by I/R injury (34). In a study by Zhang et al (13), upregulation of RIPK3 resulted in depolarization of ΔΨm in a CypD-dependent manner, suggesting that RIPK3 has the ability to promote mPTP opening. Inhibition of CaMKII blocks the RIPK3-induced depolarization of ΔΨm, thus the opening of the mPTP induced by RIPK3 requires CaMKII, thereby demonstrating that the mPTP is an indispensable downstream effector in the conduction of the RIPK3-CaMKII-mediated necroptosis signaling pathway (13,45). Chronic adverse stress induced by chronic pain increases the risk of cardiovascular disease, and it is potentially associated with myocardial I/R injury; however, to the best of our knowledge, the underlying mechanisms are not known (67). In a previous study, chronic pain stimulation was simulated by constructing a reserved nerve injury model, and it was revealed that chronic pain markedly aggravated postischemic myocardial injury and increased RIPK3-dependent CaMKII and MLKL phosphorylation levels, indicating that RIPK3-mediated MLKL and CaMKII activation were two key ways to increase susceptibility to myocardial I/R injury under chronic pain conditions (68). In addition, RIPK3 serves a role in I/R injury by activating mPTP opening through the ER stress/Ca2+ overload/xanthine oxidase/ROS pathway (45). In conclusion, these findings suggest that the RIPK3-CaMKII-mPTP-mediated necroptosis signaling pathway serves an important role in the pathogenesis of myocardial I/R injury and that inhibition of the RIPK3-CaMKII-mPTP signaling pathway may be an important therapeutic target for I/R injury.

HF

HF is the final stage in the development of several cardiovascular diseases, including ischemic cardiomyopathy and dilated cardiomyopathy, rather than an independent disease (69). The loss of cardiomyocytes and subsequent deterioration of systolic function are the main hallmarks of HF (70). Previous studies have demonstrated that the expression levels of RIPK1, RIPK3 and MLKL were markedly increased in mouse models of HF as well as human patients with HF, and were positively associated with the severity of HF (45,71). The targeting of these markers of necroptosis may improve AMI-induced cardiac remodeling, cardiac dysfunction and HF (45,61,72). Collectively, the aforementioned studies suggest that RIPK3-MLKL signaling pathway-mediated necroptosis is involved in the pathogenesis of HF.

Previous studies have demonstrated that the novel necroptosis pathway also serves an important role in HF (17,73). Compared with those in healthy mice with low CaMKII expression, in mice with congestive HF, the expression levels and activity of CaMKII were increased, and myocardial cell death was also increased, which may lead to further worsening of HF (74). A further study has demonstrated that inhibition of CaMKII reduced the levels of proteins involved in necroptosis and improved myocardial contractile function in mouse cardiomyocyte necroptosis induced by myocardial I/R injury, indicating that CaMKII serves a role in the development of HF via necroptosis (73); however, to the best of our knowledge, the upstream substrate of CaMKII remains unclear. Zhang et al (13) reported that CaMKII activation could lead to opening of the mPTP and induction of myocardial necroptosis, ultimately leading to cardiac remodeling and HF. Concurrently, phosphorylation and oxidation of CaMKII were markedly increased in the myocardial cells of HF mice, and this upregulation was markedly reversed in mice with knockout of RIPK3, thus suggesting that RIPK3 is essential for the activation of CaMKII, and CaMKII may be a downstream target or substrate of RIPK3-mediated myocardial injury and necroptosis in HF (17). Further in-depth study revealed that the ultrastructure of myocardial mitochondria was improved in HF mice after inhibition of RIPK3, suggesting that mitochondrial damage may be a potential mechanism for RIPK3-mediated necroptosis in HF (17). In addition, CaMKII also contributes to the development of HF by mechanisms other than necroptosis. Ca2+ leakage in the diastolic SR is an important pathological mechanism in the development of HF, and CaMKII can activate RyR2 on the SR and promote Ca2+ release from the SR, causing cytoplasmic Ca2+ overload, which in turn leads to cardiac diastolic dysfunction (42,75). Overall, these studies suggest that the RIPK3-CaMKII-mPTP-mediated necroptosis signaling pathway is involved in the pathogenesis of HF and may represent a novel target for the treatment of HF.

AAA

AAA is defined as aneurysmal dilatation of the abdominal aorta, which manifests as abnormal dilatation of the abdominal aorta to >50% of its normal diameter (76). The majority of patients with AAA are asymptomatic, AAA is often identified incidentally on physical examination for other reasons and patients with AAA have a potentially fatal risk of rupture that would have a serious impact on the lives of patients (77,78). Currently, there are no effective drugs to slow aneurysm growth or prevent rupture (79). Depletion of smooth muscle cells (SMCs) in the medial layer of the arterial wall is the main pathological feature of AAA; however, the cause of SMC depletion is unknown (80,81). With further research, some studies have demonstrated that the expression levels of RIPK3 were increased in human and mouse atheroma tissues (82,83), and RIPK3 is a key mediator in the regulation of necroptosis, suggesting that necroptosis is involved in the pathogenesis of AAA. Previous studies have demonstrated that inhibition of RIPK3 expression inhibited SMC depletion and aneurysm formation in mice, suggesting that the RIPK3 signaling pathway may promote the progression of AAA by causing SMC necroptosis (82-84). However, to the best of our knowledge, the downstream necroptosis signaling pathway involved in RIPK3-induced SMC death remains unknown. A subsequent study has revealed that both MLKL and CaMKII were activated in mouse atheroma tissue and that inhibition of RIPK3 markedly reduced the phosphorylation of MLKL and CaMKII in aneurysms, demonstrating that the activation of MLKL and CaMKII was dependent on RIPK3 (18). Using TNF-α and Z-Val-Ala-DL-Asp-fluoromethylketone to induce necroptosis in SMCs of the mouse aorta, it was revealed that the levels of phosphorylated MLKL and CaMKII were increased in the mouse aortic aneurysm tissue compared with those in tissues from untreated mice (18). Thus, these findings suggest that both MLKL and CaMKII are involved in SMC necroptosis in vitro and in vivo. However, in the study, coimmunoprecipitation assays did not detect an interaction between RIPK3 and CaMKII during SMC necroptosis, suggesting that RIPK3 may not directly phosphorylate CaMKII, but it cannot be ruled out that the lack of interaction between RIPK3 and CaMKII was caused by the use of SMCs instead of cardiomyocytes in the immunoprecipitation experiment, while silencing MLKL inhibited CaMKII phosphorylation in SMC necroptosis, indicating that CaMKII may be located downstream of MLKL. Further studies are still needed for confirmation (18). In conclusion, RIPK3 can induce SMC necroptosis via MLKL and CaMKII, and inhibition of the RIPK3-MLKL/CaMKII signaling pathway may be a potential way to treat AAA.

AS

AS is a chronic inflammatory disease (85). Pathologically, initial observations include the formation of fat stripes on the vascular intima, and then the fat stripes evolve into necrotic plaques that are rich in lipids and macrophages (86). In late-stage development, atherosclerotic plaques can form unstable plaques, also known as vulnerable plaques, with thin fibrous caps that are easy to rupture and lead to vascular occlusion, and unstable plaques are a leading cause of AMI, acute cerebral infarction and acute limb ischemia (87). Necroptosis can contribute to the progression of atheromatous plaques and was initially found in a mouse model of AS (88) but has not been studied in humans. Later studies have revealed that RIPK3 and MLKL expression was increased in humans with unstable carotid AS, and MLKL phosphorylation was detected in advanced atheromatous plaques (82,89). It has also been reported that RIPK3 deficiency prevented necroptosis of macrophages in atherosclerotic plaques of mice and delayed the progression of AS (88,90). These findings suggest that the necroptosis pathway mediated by RIPK3-MLKL is involved in the development of human AS. A previous study has demonstrated that CaMKII was highly expressed in atherosclerotic plaques, suggesting that CaMKII is involved in the pathogenesis of AS (19). It has been demonstrated that RIPK3 can lead to SMC necroptosis via CaMKII, leading to AAA progression (18). However, it is unclear whether CaMKII is dependent on RIPK3 to mediate necroptosis in AS, and further research is needed.

DCM

DCM is defined as ventricular dysfunction in the absence of coronary vascular disease or hypertension (91,92). DCM, one of the major cardiovascular complications of diabetes, is characterized by myocardial dysfunction in patients with diabetes (93). Cell death is considered the end point of myocardial cells in DCM (94). Myocardial cell death in diabetes mainly includes apoptosis, autophagy and necroptosis, of which necroptosis serves an important role in DCM (94-96).

Liu et al (97) reported that RIPK3 expression was increased in diabetic rats, suggesting that RIPK3-mediated necroptosis is involved in the development of diabetes. However, whether necroptosis also serves a role in DCM was not determined at the time. In one study, RIPK3 expression was markedly increased in H9C2 cardiomyocytes following 35 mmol/l high-glucose (HG) treatment, while the use of Nec-1, an inhibitor of necroptosis, markedly reduced RIPK3 expression in H9C2 cardiomyocytes exposed to HG and prevented HG-induced myocardial injury (98). In conclusion, the study indicated that HG treatment was a strong stimulus for the induction of necroptosis and that HG treatment may lead to myocardial cell injury via necroptosis, which in turn leads to DCM.

In DCM, the downstream substrates of the RIPK3-mediated necroptosis signaling pathway are gradually being found (20,21). Several previous studies have demonstrated that CaMKII expression is markedly increased in DCM and that HG can induce myocardial cell death via CaMKII (99,100). CaMKII can serve a central role in the pathogenesis of several cardiac diseases as a substrate for RIPK3 (13,17,18). However, it is not clear whether the RIPK3-CaMKII signaling pathway mediates necroptosis of cardiomyocytes in DCM. A previous study has demonstrated that HG increased RIPK3 and CaMKII expression but did not markedly change RIPK1 expression and phosphorylated (p-)MLKL levels, suggesting that RIPK3 and CaMKII mediate HG-induced necroptosis rather than RIPK1 or MLKL (20). Chen et al (21) also revealed that the phosphorylation and oxidation of CaMKII were markedly increased in cardiomyocytes from a mouse model of DCM, but this phenomenon was markedly reversed in RIPK3−/− mice, suggesting that CaMKII is a downstream target or substrate of RIPK3 in DCM. It was also revealed that inhibition of necroptosis by knocking down RIPK3 not only attenuated myocardial injury but also improved cardiac function in DCM mice and improved the mitochondrial ultrastructure in cardiomyocytes, suggesting that mitochondrial damage is involved in necroptosis in DCM (21). In addition, HG can indirectly activate CaMKII and trigger mPTP opening by increasing ROS, ultimately leading to necroptosis (96).

A large amount of advanced glycation end products (AGEs) are known to be present in diabetic patients (101). These AGEs are closely associated with complications in diabetic patients and have the potential to be biomarkers of diabetes and its complications (101,102). Furthermore, CaMKIIδ splicing is strictly regulated, and an imbalance in CaMKIIδ alternative splicing causes cardiomyocyte dysfunction and ultimately cardiovascular diseases (74). Recently, a study has revealed that AGEs increased RIPK3 expression, leading to an imbalance of CaMKIIδ alternative splicing, promoted CaMKII activation and ultimately induced cardiomyocyte necroptosis (103). By contrast, by downregulating RIPK3 or using the RIPK3 inhibitor GSK'872, the imbalance of CaMKIIδ alternative splicing can be corrected, CaMKIIδ activation can be inhibited, oxidative stress can be reduced, necroptosis can be reduced and myocardial cell damage can be reduced (103). Overall, these studies have demonstrated the important role of the RIPK3-CaMKII-mPTP-mediated necroptosis signaling pathway in the pathogenesis of DCM and provide novel ideas for the prevention and treatment of DCM.

HCM

HCM is a myocardial disorder of unknown etiology characterized by abnormal thickening of the left ventricular walls. Its histological manifestations are mainly hypertrophy of myocardial cells (104). The clinical manifestations of HCM include palpitations, exertional dyspnea, precordial pain, syncope, HF or even sudden death, and HCM is one of the main causes of sudden cardiac death in young adults (105). A growing body of evidence has suggested that necroptosis is also involved in the development of cardiac hypertrophy and HCM (22,23).

Previous studies have demonstrated that CaMKII expression was increased in cardiac hypertrophy and have also revealed that the regulation of CaMKIIδ alternative splicing may be related to cardiac function and was involved in cardiac hypertrophy in humans and mice (74,106,107). In a recent animal study, a murine model of HCM was constructed by subcutaneous infusion of angiotensin II using osmotic minipumps (22). The study revealed that, compared with those in the mice without myocardial hypertrophy (infusion of phosphate buffered solution), the levels of RIPK3 in HCM mice were markedly increased, and the levels of both the oxidation and phosphorylation of CaMKIIδ were also increased (22). The study also revealed that, compared with those in untreated mice, the levels of RIPK3, RIPK1, CaMKIIδ and p-MLKL were decreased in mice treated with the RIPK3 inhibitor (GSK'872) (22). In another study by Wang et al (23), the oxidation and phosphorylation levels of CaMKIIδ were increased in myocardial cells in a phenylephrine (PE)-induced cardiac hypertrophy model, while downregulation of RIPK3 markedly inhibited CaMKII activation in PE-stimulated cardiomyocytes. These studies suggest that CaMKII may act as a substrate for RIPK3 and mediate necroptosis of cardiomyocytes in HCM. In addition, upregulation of RIPK3 in HCM mice resulted in CaMKIIδ alternative splicing impairment, a marked decrease in CaMKIIδA and CaMKIIδB expression, and a marked increase in CaMKIIδC expression compared with those of mice without myocardial hypertrophy, which in turn led to necroptosis of cardiomyocytes (22,23). However, compared with wild-type mice (infusion of angiotensin II), knockdown or deletion of RIPK3 inhibited CaMKII activation, corrected CaMKIIδ alternative splicing impairment, alleviated oxidative stress, reduced necroptosis and reversed myocardial injury in HCM mice (22,23). Therefore, targeting the RIPK3-CaMKII-mPTP signaling pathway may protect cardiomyocytes from necroptosis, which may be important for the prevention and treatment of HCM.

AF

AF is a common cardiac arrhythmia. The incidence rate in Chinese individuals >75 years old was 8.6% and the mortality rate of patients with AF with stroke was >50% in a cohort study (108). The pathogenesis of AF has been extensively explored, and structural remodeling of the atria is considered an important part of the pathogenesis of AF (108,109). AF is a common consequence of several diseases that lead to atrial remodeling (110). Atrial structural remodeling is characterized by atrial enlargement and atrial fibrosis (111). A previous study has demonstrated that RIPK3-mediated necroptosis contributes to cardiomyocyte death, which may result in cardiac fibrosis and cardiac remodeling in the long term (61). Sustained CaMKII activation is considered to serve an important role in atrial remodeling and may promote AF (40,112). Several studies have demonstrated that by knocking down RIPK3 or inhibiting its downstream substrate CaMKII, necroptosis signaling can be inhibited, and fibrosis can be prevented (113-115). These studies suggest that necroptosis is involved in the development of fibrosis and is potentially related to atrial fibrosis and atrial structural remodeling. In a recent study, the levels of RIPK1, RIPK3, MLKL, CaMKII and their phosphorylated forms were markedly increased in the atria of AF mice, and myocardial fibrosis and susceptibility to AF caused by acetylcholine-CaCl2 were attenuated by treatment with necrostatin-1, a necrotrophic apoptosis inhibitor (24). In addition, the study has indicated that aerobic swim training of mice inhibited necroptosis signaling, thereby decreasing susceptibility to AF and atrial structural remodeling (24). In conclusion, these results suggest that necroptosis is involved in the development of atrial fibrosis and is an important pathogenesis leading to AF, which provides a novel target for the prevention and treatment of AF.

Cardiotoxicity of antitumor drugs and other chemicals

Chemotherapy is currently one of the leading treatments for cancer; however, improper treatment causes damage to a variety of organs, with the heart being one of the organs frequently affected (116). Prolonged use of antineoplastic drugs can cause cardiotoxicity, triggering excessive loss of cardiomyocytes and leading to cardiovascular complications such as HF, AMI, hypertension, thromboembolism and arrhythmias (117), which adversely affect the prognosis of patients with cancer and are a major cause of treatment termination and drug development failure (118). Excessive loss of cardiomyocytes is known to serve an important role in the cardiotoxicity of antineoplastic drugs (119). As research continues, the role of necroptosis in the cardiotoxicity of various antineoplastic drugs is emerging. The cardiotoxicity of antitumor drugs and other chemicals is summarized in Table II.

Table II

Cardiotoxicity of antitumor drugs and other chemicals.

Table II

Cardiotoxicity of antitumor drugs and other chemicals.

First author/s, yearDrugTargetCardiotoxic effect(Refs.)
Zhang et al, 2016DoxorubicinRIPK3 and CaMKIICardiomyocyte death(13)
Zhou et al, 2021VincristineRIPK1, RIPK3, MLKL and CaMKIICardiomyocyte death(25)
McMullen et al, 2021SunitinibCaMKIICardiac fibroblast death(26)
Reventun et al, 2020BPARIPK3 and CaMKIIEndothelial cell death(27)

[i] RIPK3, receptor interaction protein kinase 3; CaMKII, calcium/calmodulin-dependent protein kinase II; RIPK1, receptor interaction protein kinase 1; MLKL, mixed lineage kinase domain-like protein; BPA, bisphenol A.

DOX is a first-line chemotherapy drug that is widely used to treat a variety of cancer types, including carcinomas, sarcomas and hematological cancers (120) It causes irreversible cardiotoxicity in 30-40% of patients, resulting in the loss of numerous myocardial cells and ultimately leading to cardiomyopathy and HF, thus severely limiting its clinical application (121). It has been reported that the mRNA and protein expression levels of RIPK3 were increased 2-3-fold in DOX-treated mouse cardiomyocytes, and the levels of phosphorylation of the Thr287 site of CaMKII and oxidation of the Met281/282 site of CaMKII were markedly increased, as was the opening of the mPTP (13). RIPK3−/− mice have a certain resistance to the cardiotoxicity of DOX, which is specifically manifested by a reduction in the activation of CaMKII, a reduction in myocardial cell death, and improvement in the heart function and survival of mice (13). These results indicate that DOX is able to activate CaMKII through RIPK3, thereby promoting the opening of the mPTP and ultimately inducing necroptosis of myocardial cells (13).

Vincristine is also a first-line chemotherapy drug, and myocardial injury induced by vincristine has been reported continuously since the launch of the drug on the market (122,123). However, to the best of our knowledge, the potential mechanisms leading to myocardial injury remain unclear. A recent study has demonstrated that activation of CaMKII was also detected in myocardial necroptosis induced by vincristine, and both in vivo and ex vivo experiments with vincristine demonstrated that the levels of RIPK1, RIPK3, MLKL and p-CaMKII were all increased in cardiomyocytes, suggesting that vincristine-induced necroptosis of cardiomyocytes can be mediated not only by MLKL but also by CaMKII (25). The study has also revealed that the oxidation of the Met281/282 site of CaMKII did not markedly change after treatment of H9C2 cells with vincristine (25). By contrast, both the phosphorylation and oxidation levels of CaMKII were increased in DOX-mediated necroptosis of myocardial cells (13), indicating that different antineoplastic drugs may lead to cardiac toxicity via different mechanisms.

In addition to myocardial cells, the damage caused by antineoplastic drugs to cardiac fibroblasts in the heart may also mediate cardiotoxicity (26). A recent study has demonstrated that the oxidation of CaMKII was increased in cardiac fibroblasts after sunitinib treatment, and cell death was correspondingly increased (26). However, because the study did not evaluate the expression levels of key components of necroptosis, such as RIPK3 and MLKL, it is still unclear whether CaMKII-mediated cell death is dependent on RIPK3.

In addition to the aforementioned antitumor drugs, some studies have demonstrated that other chemicals can cause cardiotoxicity through the RIPK3-CaMKII-mediated necroptosis signaling pathway. Bisphenol A (BPA) is a chemical substance widely used in daily life. Because of its widespread use, individuals are often exposed to it, so its safety has become a focus of public attention (124,125). Epidemiological studies have demonstrated that BPA increased the incidence rate and mortality of cardiovascular diseases (126,127). In a recent study, the necroptosis of coronary artery endothelial cells was increased, and the mice also had severe complications such as coronary artery injury, vascular leakage, myocardial injury and even HF after the mice were treated with BPA (27). The study further explored the underlying mechanisms in depth and found that the activation of CaMKII in endothelial cells was increased, and inhibition of CaMKII activity with KN-93 could reduce endothelial necroptosis but did not affect the increased expression levels of RIPK3 induced by BPA treatment (27). However, knockdown of RIPK3 prevented the phosphorylation of CaMKII and the transduction of necroptosis signals (27). Therefore, these results indicated that BPA can induce necroptosis through the RIPK3-CaMKII-mPTP signaling pathway, thus leading to cardiotoxicity.

In summary, the RIPK3-CaMKII-mPTP-mediated necroptosis signaling pathway serves an important role in the cardiotoxicity of antitumor drugs and other chemicals, and targeting this necroptosis signaling pathway is important to reduce the cardiotoxicity of drugs.

4. Therapeutic targeting of the RIPK3-CaMKII-mPTP signaling pathway

At present, the therapeutic options for the treatment of cardiovascular diseases remain unsatisfactory, necessitating the development of more precise targeted therapeutic drugs. The aforementioned findings (13-27) on the involvement of RIPK3-CaMKII-mPTP-mediated necroptosis in a variety of cardiovascular diseases suggest that the RIPK3-CaMKII-mPTP pathway has high potential as a drug target. The emergence of drugs targeting RIPK3, CaMKII and mPTP enables the development of clinical candidates. The present review discusses the inhibitors that target RIPK3, CaMKII and mPTP, and these are summarized in Table III.

Table III

Summary of inhibitors of necroptosis in cardiovascular diseases.

Table III

Summary of inhibitors of necroptosis in cardiovascular diseases.

First author/s, yearInhibitorTargetCardiovascular diseases(Refs.)
Zhang et al, 2022GSK'872RIPK3HCM(22)
Oerlemans et al, 2012; Lim et al, 2007; Smith et al, 2007Necrostatin-1RIPK3AMI and I/R injury(63,131,132)
Liang et al, 2017Necrostatin-1RIPK3DCM(98)
Wei et al, 20223-T1AMRIPK1, RIPK3 and CaMKIII/R injury(15)
Yang et al, 2018MelatoninRIPK3 and CaMKIII/R injury(68)
Chang et al, 2019ZYZ-803RIPK3 and CaMKIIAMI(136)
Feng and Anderson, 2017; Adameova et al, 2012KN-93CaMKIII/R injury(38,137)
Ikeda et al, 2019NifedipineCaMKIIDOX-induced cardiomyopathy(140)
Ikeda et al, 2019AmlodipineCaMKIIDOX-induced cardiomyopathy(140)
Liu et al, 2019CaMKIInCaMKIIAF(112)
Sun et al, 2019I1PP1CaMKIIDCM(20)
Xu et al, 2018; Joiner et al, 2012; Lim et al, 2012; Piot et al, 2008; Cung et al, 2015; Ottani et al, 2016CsAmPTPI/R injury(16,47,141-144)
Zhang et al, 2019 CsA@PLGA-PEG-SS31mPTPI/R injury(146)
Yao et al, 2010H2SmPTPI/R injury(147)

[i] RIPK3, receptor interaction protein kinase 3; HCM, hypertrophic cardiomyopathy; AMI, acute myocardial infarction; I/R, ischemia-reperfusion; DCM, diabetic cardiomyopathy; 3-T1AM, 3-iodothyronamine; RIPK1, receptor interaction protein kinase 1; CaMKII, calcium/calmodulin-dependent protein kinase II; DOX, doxorubicin; AF, atrial fibrillation; I1PP1, inhibitor-1 of protein phosphatase 1; CsA, cyclosporin A; mPTP, mitochondrial permeability transition pore; H2S, hydrogen sulfide.

Inhibitors targeting RIPK3

RIPK3 kinase activity serves an important role in necroptosis, and the kinase domain of RIPK3 is an important target of small-molecule inhibitor intervention (128). Numerous RIPK3 kinase inhibitors have been reported, including GSK'840, GSK'843 and GSK'872, which can bind to the RIPK3 kinase domain with high affinity and inhibit RIPK3 kinase activity, thereby inhibiting RIPK3-induced necroptosis (129,130). GSK'840 and GSK'843 have been reported to be therapeutic interventions for viral infection, inflammatory diseases and cancer by inhibiting necroptosis (130). GSK'872 has been reported to correct the imbalance of CaMKII alternative splicing and to prevent necroptosis, thus reducing myocardial cell damage (103). A previous study has demonstrated that GSK'872 also had a marked protective effect on Alzet miniosmotic pumps (ALZA Corporation) and perfused with angiotensin II-induced cardiac hypertrophy in mice, which may make it a potential drug candidate for HCM (22). Necrostatin-1 is a classic necroptosis inhibitor. It has been demonstrated to reduce the area of myocardial infarction (131,132) and to prevent adverse cardiac remodeling after I/R injury in mice (63). A previous study has also demonstrated that necrostatin-1 reduced RIPK3 expression induced by HG, thereby increasing cardiomyocyte viability, reducing ROS production and attenuating ΔΨm, and ultimately exerting a protective effect against DCM (98). 3-Iodothyronamine (3-T1AM) is a metabolite of decarboxylated and deiodinated thyroid hormone. Previous studies have demonstrated that 3-T1AM could attenuate cardiomyocyte apoptosis induced by I/R injury (133). A recent study has revealed that 3-T1AM could also reduce the expression levels of RIPK1, RIPK3 and CaMKII in myocardial cells with I/R injury and could alleviate necroptosis of myocardial cells, thus protecting myocardial cells from I/R injury (15). Melatonin is a hormone produced by the pineal gland that is mainly involved in the regulation of sleep and the circadian rhythm (134). A previous study has reported that melatonin could inhibit the phosphorylation of RIPK3 and the downstream molecule CaMKII, and reduced the occurrence of necroptosis, thus improving cardiac function after I/R injury (68). ZYZ-803 is a novel type of hydrogen sulfide (H2S)-nitric oxide (NO) coupling donor that can be decomposed into H2S and NO (135). A study has reported that it could correct the imbalance in these two small gas molecules, inhibit the interaction between RIPK3 and CaMKII, and inhibit the phosphorylation of CaMKII, thereby inhibiting the RIPK3-CaMKII-mPTP signaling pathway, reducing the necroptosis of myocardial cells and having a protective effect in AMI (136).

Inhibitors targeting CaMKII

Sustained CaMKII activation has an important role in the necroptosis pathway and is implicated in the pathogenesis of several cardiovascular diseases, thus CaMKII can be an important target for the inhibition of necroptosis (38). KN-93 is a small-molecule inhibitor of CaMKII. During myocardial I/R injury, treatment of the heart with KN-93 reduces cell death under RIPK3 upregulation conditions, improves postischemic cardiac contractile function and inhibits the development of malignant arrhythmias (38,137). KN-93 could also downregulate CaMKIIδ expression, exhibiting potential therapeutic value for I/R injury (38,137). The L-type Ca2+ channel is the main pathway of Ca2+ entry into myocardial cells and is essential for myocardial contraction (138). L-Type Ca2+ channel blockers, such as nifedipine and amlodipine, are known to block this pathway and are commonly used to treat hypertension (139). A previous study has demonstrated that nifedipine and amlodipine could inhibit the activity of CaMKII and reduce myocardial cell death by reducing the levels of Ca2+ in myocardial cells, thus serving a protective role in DOX-induced cardiomyopathy (140). However, the study noted that L-type Ca2+ channel blockers suppressed DOX-induced cardiomyocyte injury by reducing cellular apoptosis. Nevertheless, their role in necroptosis was not investigated. CaMKIIn, a specific inhibitor of CaMKII, has been found to reduce atrial structural remodeling and to decrease the incidence of AF in an animal model of HF (112). RIPK3-mediated necroptosis in myocardial cells was markedly inhibited by inhibiting CaMKII activation or oxidation using inhibitor-1 of protein phosphatase 1 (I1PP1) (20). Furthermore, I1PP1 can reverse CaMKIIδ alternative splicing, reduce CaMKII activity and attenuate the necroptosis of cardiomyocytes induced by HG, which may provide benefits for the treatment of DCM (20).

Inhibitors targeting mPTP

Increasing evidence indicates that the opening of the mPTP is a part of the final pathway of necroptosis and is related to the death of myocardial cells and myocardial damage caused by necroptosis (49-51). CsA is an mPTP antagonist that acts mainly by inhibiting the activity of CypD, thus preventing mPTP opening, reducing ΔΨm damage and preventing necroptosis caused by mitochondrial damage and I/R damage (32,48). In an animal study and a small clinical trial (141,142), CsA has been demonstrated to reduce the area of AMI, which is conducive to reducing I/R injury. However, in two large clinical trials, CsA was injected intravenously before percutaneous coronary intervention, and it was found that the clinical outcome and long-term prognosis of the experimental group were similar to those of the placebo group (143,144). Based on the aforementioned research conclusions, the clinical application of CsA has been severely limited. The potential reasons for this phenomenon may include some known confounding factors, such as age, diabetes, interference of clopidogrel and other drugs, and insufficient CsA reaching the mitochondria of the ischemic myocardium (145,146). In this regard, the CsA@ PLGA-PEG-SS31 nanoparticles successfully overcome the limitations of CsA by effectively accumulating in ischemic myocardial tissue and accurately targeting the mitochondria of ischemic myocardial cells, thus exerting a protective effect on the ischemic myocardium (146). This method represents a prospective clinical strategy in which CsA can be used to treat I/R injury after AMI in the future. H2S is a gaseous molecule. As an endogenous antioxidant, it can regulate cardiovascular function and has a potential protective effect on the heart (147). A previous study has demonstrated that H2S could prevent the opening of the mPTP in cardiomyopathy induced by I/R injury and could prevent apoptosis of cardiomyocytes (147). Previous studies have indicated that exogenous administration of H2S can inhibit necroptosis, alleviate the dysfunction of cardiomyocytes induced by HG and improve heart damage caused by diabetes, suggesting that the restoration of normal levels of H2S in the body may be a potential treatment for DCM; however, it is unclear whether H2S can also prevent the opening of the mPTP in DCM, which needs to be further confirmed (148,149).

5. Conclusions and perspectives

In the last decade, necroptosis mediated by RIPK3 and MLKL has been recognized as the classical necroptosis pathway. The necrosome components RIPK1, RIPK3 and MLKL are key regulators of necroptosis. RIPK3 activates MLKL and promotes MLKL localization to the plasma membrane, ultimately leading to plasma membrane permeabilization and cell death. CaMKII has been found to act as a novel substrate for RIPK3 in mediating necroptosis of cardiomyocytes via the RIPK3-CaMKII-mPTP signaling pathway, which causes ATP depletion by inducing mitochondrial damage, ultimately leading to necroptosis. This adds to the complexity of the necroptosis pathway by linking death receptor-mediated necroptosis with the mitochondrial necrosis pathway.

As aforementioned, RIPK3-CaMKII-mPTP pathway-mediated necroptosis serves an important role in a variety of cardiovascular diseases, including AMI, HF, AAA, DCM, HCM and AF, and cardiotoxicity of antitumor drugs and other chemicals. The expression levels of CaMKII are increased in AS and sunitinib-induced cardiotoxicity. Additionally, the oxidation levels of CaMKII are also increased in sunitinib-induced cardiotoxicity, and cell death has been demonstrated to increase with the oxidation of CaMKII. However, to the best of our knowledge, there are currently no studies evaluating the role of necroptosis components such as RIPK3 in AS and sunitinib-induced cardiotoxicity. Therefore, it remains uncertain whether CaMKII-mediated cell death is dependent on RIPK3. Further studies are needed.

Since RIPK3-CaMKII-mPTP-mediated necroptosis serves a role in the pathogenesis of several cardiovascular diseases, targeting the RIPK3-CaMKII-mPTP pathway is a potential therapeutic approach for several cardiovascular diseases. Several inhibitors of the necroptosis pathway, such as necrostatin-1, KN-93 and CsA, have been developed. However, most studies on these inhibitors are based on in vitro experiments or animal models, and their clinical feasibility requires validation in vivo and in clinical trials. Future work will focus on conducting clinical studies to verify the clinical application value of these inhibitors, as well as a more precise understanding of the RIPK3-CaMKII-mPTP signaling pathway in the pathogenesis of cardiovascular diseases. In addition, to improve the selectivity and safety of the drugs, the value of necroptosis inhibitors in clinical applications should be carefully investigated.

Availability of data and materials

Not applicable.

Authors' contributions

JZ contributed to the conception, design and drafting of the manuscript. SC, SG, ZY, FO, SL and LL contributed to data collection and manuscript drafting. Data authentication is not applicable. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

RIPK1

receptor interaction protein kinase 1

RIPK3

receptor interaction protein kinase 3

MLKL

mixed lineage kinase domain-like protein

CaMKII

calcium/calmodulin-dependent protein kinase II

mPTP

mitochondrial permeability transition pore

AMI

acute myocardial infarction

I/R

ischemia-reperfusion

HF

heart failure

AAA

abdominal aortic aneurysm

AS

atherosclerosis

DCM

diabetic cardiomyopathy

HCM

hypertrophic cardiomyopathy

AF

atrial fibrillation

FADD

FAS-associated death domain protein

TNFR

tumor necrosis factor receptor

RHIM

RIP homotypic interaction motif

ROS

reactive oxygen species

RyR2

ryanodine receptor 2

SR

sarcoplasmic reticulum

ER

endoplasmic reticulum

MCU

mitochondrial Ca2+ uniporter

ΔΨm

mitochondrial inner membrane potential

CypD

cyclophilin D

CsA

cyclosporin A

SMC

smooth muscle cell

HG

high-glucose

AGE

advanced glycation end product

BPA

bisphenol A

3-T1AM

3-iodothyronamine

H2S

hydrogen sulfide

NO

nitric oxide

I1PP1

inhibitor-1 of protein phosphatase 1

DOX

doxorubicin

Acknowledgments

Not applicable.

Funding

This work was supported by the Medical Science and Technology Research Foundation of Guangdong Province of China (grant no. A2023151), Science and Technology Project of Foshan (grant no. 2220001003865), Scientific Research Start Plan of Shunde Hospital, Southern Medical University (grant no. SRSP2019004), Natural Science Foundation of Guangdong Province of China (grant no. 2018A030313067), Science and Technology Innovation Project of Foshan, Guangdong (grant nos. FS0AA-KJ218-1301-0006 and FS0AA-KJ218-1301-0010) and Key Specialist Department Training Project of Foshan, Guangdong Province of China (grant no. Fspy 3-2015034).

References

1 

Mathers CD and Loncar D: Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 3:e4422006. View Article : Google Scholar :

2 

Del Re DP, Amgalan D, Linkermann A, Liu Q and Kitsis RN: Fundamental mechanisms of regulated cell death and implications for heart disease. Physiol Rev. 99:1765–1817. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Ruparelia N, Chai JT, Fisher EA and Choudhury RP: Inflammatory processes in cardiovascular disease: A route to targeted therapies. Nat Rev Cardiol. 14:133–144. 2017. View Article : Google Scholar :

4 

D'Arcy MS: Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 43:582–592. 2019. View Article : Google Scholar

5 

Bertheloot D, Latz E and Franklin BS: Necroptosis, pyroptosis and apoptosis: An intricate game of cell death. Cell Mol Immunol. 18:1106–1121. 2021. View Article : Google Scholar :

6 

He S, Wang L, Miao L, Wang T, Du F, Zhao L and Wang X: Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell. 137:1100–1111. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Koo GB, Morgan MJ, Lee DG, Kim WJ, Yoon JH, Koo JS, Kim SI, Kim SJ, Son MK, Hong SS, et al: Methylation-dependent loss of RIP3 expression in cancer represses programmed necrosis in response to chemotherapeutics. Cell Res. 25:707–725. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Yatim N, Jusforgues-Saklani H, Orozco S, Schulz O, Barreira da Silva R, Reis e Sousa C, Green DR, Oberst A and Albert ML: RIPK1 and NF-κB signaling in dying cells determines cross-priming of CD8+ T cells. Science. 350:328–334. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Gupta K, Phan N, Wang Q and Liu B: Necroptosis in cardiovascular disease-a new therapeutic target. J Mol Cell Cardiol. 118:26–35. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Kung G, Konstantinidis K and Kitsis RN: Programmed necrosis, not apoptosis, in the heart. Circ Res. 108:1017–1036. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Zhe-Wei S, Li-Sha G and Yue-Chun L: The role of necroptosis in cardiovascular disease. Front Pharmacol. 9:7212018. View Article : Google Scholar : PubMed/NCBI

12 

Ruan ZH, Xu ZX, Zhou XY, Zhang X and Shang L: Implications of necroptosis for cardiovascular diseases. Curr Med Sci. 39:513–522. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Zhang T, Zhang Y, Cui M, Jin L, Wang Y, Lv F, Liu Y, Zheng W, Shang H, Zhang J, et al: CaMKII is a RIP3 substrate mediating ischemia- and oxidative stress-induced myocardial necroptosis. Nat Med. 22:175–182. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Wang W, Wang B, Sun S, Cao S, Zhai X, Zhang C, Zhang Q, Yuan Q, Sun Y, Xue M, et al: Inhibition of adenosine kinase attenuates myocardial ischaemia/reperfusion injury. J Cell Mol Med. 25:2931–2943. 2021. View Article : Google Scholar : PubMed/NCBI

15 

Wei B, Zhao H, Hu B, Dai L, Zhang G, Mo L, Huang N, Zou C, Zhang B, Zhou H, et al: T1AM attenuates the hypoxia/reoxygenation-induced necroptosis of H9C2 cardiomyocytes via RIPK1/RIPK3 pathway. Biomed Res Int. 2022:48337912022. View Article : Google Scholar : PubMed/NCBI

16 

Xu T, Ding W, Tariq MA, Wang Y, Wan Q, Li M and Wang J: Molecular mechanism and therapy application of necrosis during myocardial injury. J Cell Mol Med. 22:2547–2557. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Cao J, Zhang J, Qian J, Wang X, Zhang W and Chen X: Ca2+/calmodulin-dependent protein kinase II regulation by RIPK3 alleviates necroptosis in transverse arch constriction-induced heart failure. Front Cardiovasc Med. 9:8473622022. View Article : Google Scholar

18 

Zhou T, DeRoo E, Yang H, Stranz A, Wang Q, Ginnan R, Singer HA and Liu B: MLKL and CaMKII are involved in RIPK3-mediated smooth muscle cell necroptosis. Cells. 10:23972021. View Article : Google Scholar : PubMed/NCBI

19 

Maione AS, Cipolletta E, Sorriento D, Borriello F, Soprano M, Rusciano MR, D'Esposito V, Markabaoui AK, De Palma GD, Martino G, et al: Cellular subtype expression and activation of CaMKII regulate the fate of atherosclerotic plaque. Atherosclerosis. 256:53–61. 2017. View Article : Google Scholar

20 

Sun L, Chen Y, Luo H, Xu M, Meng G and Zhang W: Ca2+/calmodulin-dependent protein kinase II regulation by inhibitor 1 of protein phosphatase 1 alleviates necroptosis in high glucose-induced cardiomyocytes injury. Biochem Pharmacol. 163:194–205. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Chen Y, Li X, Hua Y, Ding Y, Meng G and Zhang W: RIPK3-mediated necroptosis in diabetic cardiomyopathy requires CaMKII activation. Oxid Med Cell Longev. 2021:66178162021.PubMed/NCBI

22 

Zhang J, Qian J, Cao J, Wang X, Zhang W and Gu X: Ca2+/calmodulin-dependent protein kinase II regulation by inhibitor of RIPK3 protects against cardiac hypertrophy. Oxid Med Cell Longev. 2022:79413742022.

23 

Wang X, Zhang J, Qian J, Cao J, Zhang W and Jiang Y: The regulatory mechanism and effect of receptor-interacting protein kinase 3 on phenylephrine-induced cardiomyocyte hypertrophy. J Cardiovasc Pharmacol. 80:236–250. 2022. View Article : Google Scholar : PubMed/NCBI

24 

Fu Y, Jiang T, Sun H, Li T, Gao F, Fan B, Li X, Qin X and Zheng Q: Necroptosis is required for atrial fibrillation and involved in aerobic exercise-conferred cardioprotection. J Cell Mol Med. 25:8363–8375. 2021. View Article : Google Scholar : PubMed/NCBI

25 

Zhou H, Liu L, Ma X, Wang J, Yang J, Zhou X, Yang Y and Liu H: RIP1/RIP3/MLKL-mediated necroptosis contributes to vinblastine-induced myocardial damage. Mol Cell Biochem. 476:1233–1243. 2021. View Article : Google Scholar :

26 

McMullen CJ, Chalmers S, Wood R, Cunningham MR and Currie S: Sunitinib and imatinib display differential cardiotoxicity in adult rat cardiac fibroblasts that involves a role for calcium/calmodulin dependent protein kinase II. Front Cardiovasc Med. 7:6304802021. View Article : Google Scholar : PubMed/NCBI

27 

Reventun P, Sanchez-Esteban S, Cook A, Cuadrado I, Roza C, Moreno-Gomez-Toledano R, Muñoz C, Zaragoza C, Bosch RJ and Saura M: Bisphenol A induces coronary endothelial cell necroptosis by activating RIP3/CamKII dependent pathway. Sci Rep. 10:41902020. View Article : Google Scholar : PubMed/NCBI

28 

Galluzzi L, Kepp O and Kroemer G: RIP kinases initiate programmed necrosis. J Mol Cell Biol. 1:8–10. 2009. View Article : Google Scholar

29 

Ingram JP, Thapa RJ, Fisher A, Tummers B, Zhang T, Yin C, Rodriguez DA, Guo H, Lane R, Williams R, et al: ZBP1/DAI drives RIPK3-mediated cell death induced by IFNs in the absence of RIPK1. J Immunol. 203:1348–1355. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Garvin AM, Jackson MA and Korzick DH: Inhibition of programmed necrosis limits infarct size through altered mitochondrial and immune responses in the aged female rat heart. Am J Physiol Heart Circ Physiol. 315:H1434–H1442. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Khoury MK, Gupta K, Franco SR and Liu B: Necroptosis in the pathophysiology of disease. Am J Pathol. 190:272–285. 2020. View Article : Google Scholar :

32 

Cuny GD and Degterev A: RIPK protein kinase family: Atypical lives of typical kinases. Semin Cell Dev Biol. 109:96–105. 2021. View Article : Google Scholar :

33 

Li J, McQuade T, Siemer AB, Napetschnig J, Moriwaki K, Hsiao YS, Damko E, Moquin D, Walz T, McDermott A, et al: The RIP1/RIP3 necrosome forms a functional amyloid signaling complex required for programmed necrosis. Cell. 150:339–350. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Moquin DM, McQuade T and Chan FK: CYLD deubiquitinates RIP1 in the TNFα-induced necrosome to facilitate kinase activation and programmed necrosis. PLoS One. 8:e768412013. View Article : Google Scholar

35 

Zhang J, Yang Y, He W and Sun L: Necrosome core machinery: MLKL. Cell Mol Life Sci. 73:2153–2163. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Meng Y, Davies KA, Fitzgibbon C, Young SN, Garnish SE, Horne CR, Luo C, Garnier JM, Liang LY, Cowan AD, et al: Human RIPK3 maintains MLKL in an inactive conformation prior to cell death by necroptosis. Nat Commun. 12:67832021. View Article : Google Scholar : PubMed/NCBI

37 

Sun L, Wang H, Wang Z, He S, Chen S, Liao D, Wang L, Yan J, Liu W, Lei X and Wang X: Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell. 148:213–227. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Feng N and Anderson ME: CaMKII is a nodal signal for multiple programmed cell death pathways in heart. J Mol Cell Cardiol. 103:102–109. 2017. View Article : Google Scholar :

39 

Hudmon A and Schulman H: Structure-function of the multifunctional Ca2+/calmodulin-dependent protein kinase II. Biochem J. 364:593–611. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Luczak ED and Anderson ME: CaMKII oxidative activation and the pathogenesis of cardiac disease. J Mol Cell Cardiol. 73:112–116. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Beckendorf J, van den Hoogenhof MMG and Backs J: Physiological and unappreciated roles of CaMKII in the heart. Basic Res Cardiol. 113:292018. View Article : Google Scholar : PubMed/NCBI

42 

Gui L, Guo X, Zhang Z, Xu H, Ji YW, Wang RJ, Zhu JH and Chen QH: Activation of CaMKIIδA promotes Ca2+ leak from the sarcoplasmic reticulum in cardiomyocytes of chronic heart failure rats. Acta Pharmacol Sin. 39:1604–1612. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Szentesi P, Pignier C, Egger M, Kranias EG and Niggli E: Sarcoplasmic reticulum Ca2+ refilling controls recovery from Ca2+-induced Ca2+ release refractoriness in heart muscle. Circ Res. 95:807–813. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Chelu MG, Sarma S, Sood S, Wang S, van Oort RJ, Skapura DG, Li N, Santonastasi M, Müller FU, Schmitz W, et al: Calmodulin kinase II-mediated sarcoplasmic reticulum Ca2+ leak promotes atrial fibrillation in mice. J Clin Invest. 119:1940–1951. 2009.

45 

Zhu P, Hu S, Jin Q, Li D, Tian F, Toan S, Li Y, Zhou H and Chen Y: Ripk3 promotes ER stress-induced necroptosis in cardiac IR injury: A mechanism involving calcium overload/XO/ROS/mPTP pathway. Redox Biol. 16:157–168. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Gonano LA and Petroff MV: Subcellular mechanisms underlying digitalis-induced arrhythmias: Role of calcium/calmodulin-dependent kinase II (CaMKII) in the transition from an inotropic to an arrhythmogenic effect. Heart Lung Circ. 23:1118–1124. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Joiner ML, Koval OM, Li J, He BJ, Allamargot C, Gao Z, Luczak ED, Hall DD, Fink BD, Chen B, et al: CaMKII determines mitochondrial stress responses in heart. Nature. 491:269–273. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Bonora M, Giorgi C and Pinton P: Molecular mechanisms and consequences of mitochondrial permeability transition. Nat Rev Mol Cell Biol. 23:266–285. 2022. View Article : Google Scholar

49 

Zhou Y, Liao J, Mei Z, Liu X and Ge J: Insight into crosstalk between ferroptosis and necroptosis: Novel therapeutics in ischemic stroke. Oxid Med Cell Longev. 2021:99910012021. View Article : Google Scholar : PubMed/NCBI

50 

Kist M and Vucic D: Cell death pathways: Intricate connections and disease implications. EMBO J. 40:e1067002021. View Article : Google Scholar :

51 

Halestrap AP: A pore way to die: The role of mitochondria in reperfusion injury and cardioprotection. Biochem Soc Trans. 38:841–860. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Naryzhnaya NV, Maslov LN and Oeltgen PR: Pharmacology of mitochondrial permeability transition pore inhibitors. Drug Dev Res. 80:1013–1030. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Thygesen K, Alpert JS, Jaffe AS, Simoons ML, Chaitman BR, White HD; Joint ESC/ACCF/AHA/WHF Task Force for the Universal Definition of Myocardial Infarction; Katus HA, Lindahl B, Morrow DA, et al: Third universal definition of myocardial infarction. Circulation. 126:2020–2035. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Pollard TJ: The acute myocardial infarction. Prim Care. 27:631–649. vi2000. View Article : Google Scholar : PubMed/NCBI

55 

Lindsey ML, Bolli R, Canty JM Jr, Du XJ, Frangogiannis NG, Frantz S, Gourdie RG, Holmes JW, Jones SP, Kloner RA, et al: Guidelines for experimental models of myocardial ischemia and infarction. Am J Physiol Heart Circ Physiol. 314:H812–H838. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Cabrera-Fuentes HA, Aragones J, Bernhagen J, Boening A, Boisvert WA, Bøtker HE, Bulluck H, Cook S, Di Lisa F, Engel FB, et al: From basic mechanisms to clinical applications in heart protection, new players in cardiovascular diseases and cardiac theranostics: Meeting report from the third international symposium on 'New frontiers in cardiovascular research'. Basic Res Cardiol. 111:692016. View Article : Google Scholar

57 

Hausenloy DJ and Yellon DM: Myocardial ischemia-reperfusion injury: A neglected therapeutic target. J Clin Invest. 123:92–100. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Adameova A, Horvath C, Abdul-Ghani S, Varga ZV, Suleiman MS and Dhalla NS: Interplay of oxidative stress and necrosis-like cell death in cardiac ischemia/reperfusion injury: A focus on necroptosis. Biomedicines. 10:1272022. View Article : Google Scholar : PubMed/NCBI

59 

Mishra PK, Adameova A, Hill JA, Baines CP, Kang PM, Downey JM, Narula J, Takahashi M, Abbate A, Piristine HC, et al: Guidelines for evaluating myocardial cell death. Am J Physiol Heart Circ Physiol. 317:H891–H922. 2019. View Article : Google Scholar : PubMed/NCBI

60 

DeRoo E, Zhou T and Liu B: The role of RIPK1 and RIPK3 in cardiovascular disease. Int J Mol Sci. 21:81742020. View Article : Google Scholar : PubMed/NCBI

61 

Luedde M, Lutz M, Carter N, Sosna J, Jacoby C, Vucur M, Gautheron J, Roderburg C, Borg N, Reisinger F, et al: RIP3, a kinase promoting necroptotic cell death, mediates adverse remodelling after myocardial infarction. Cardiovasc Res. 103:206–216. 2014. View Article : Google Scholar : PubMed/NCBI

62 

Zhang DY, Wang BJ, Ma M, Yu K, Zhang Q and Zhang XW: MicroRNA-325-3p protects the heart after myocardial infarction by inhibiting RIPK3 and programmed necrosis in mice. BMC Mol Biol. 20:172019. View Article : Google Scholar : PubMed/NCBI

63 

Oerlemans MI, Liu J, Arslan F, den Ouden K, van Middelaar BJ, Doevendans PA and Sluijter JP: Inhibition of RIP1-dependent necrosis prevents adverse cardiac remodeling after myocardial ischemia-reperfusion in vivo. Basic Res Cardiol. 107:2702012. View Article : Google Scholar : PubMed/NCBI

64 

Nakagawa T, Shimizu S, Watanabe T, Yamaguchi O, Otsu K, Yamagata H, Inohara H, Kubo T and Tsujimoto Y: Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death. Nature. 434:652–658. 2005. View Article : Google Scholar : PubMed/NCBI

65 

Clarke SJ, McStay GP and Halestrap AP: Sanglifehrin A acts as a potent inhibitor of the mitochondrial permeability transition and reperfusion injury of the heart by binding to cyclophilin-D at a different site from cyclosporin A. J Biol Chem. 277:34793–34799. 2002. View Article : Google Scholar : PubMed/NCBI

66 

Parks RJ, Menazza S, Holmström KM, Amanakis G, Fergusson M, Ma H, Aponte AM, Bernardi P, Finkel T and Murphy E: Cyclophilin D-mediated regulation of the permeability transition pore is altered in mice lacking the mitochondrial calcium uniporter. Cardiovasc Res. 115:385–394. 2019. View Article : Google Scholar :

67 

Torrance N, Elliott AM, Lee AJ and Smith BH: Severe chronic pain is associated with increased 10 year mortality. A cohort record linkage study. Eur J Pain. 14:380–386. 2010. View Article : Google Scholar

68 

Yang Z, Li C, Wang Y, Yang J, Yin Y, Liu M, Shi Z, Mu N, Yu L and Ma H: Melatonin attenuates chronic pain related myocardial ischemic susceptibility through inhibiting RIP3-MLKL/CaMKII dependent necroptosis. J Mol Cell Cardiol. 125:185–194. 2018. View Article : Google Scholar : PubMed/NCBI

69 

Szobi A, Gonçalvesová E, Varga ZV, Leszek P, Kuśmierczyk M, Hulman M, Kyselovič J, Ferdinandy P and Adameová A: Analysis of necroptotic proteins in failing human hearts. J Transl Med. 15:862017. View Article : Google Scholar :

70 

Lázár E, Sadek HA and Bergmann O: Cardiomyocyte renewal in the human heart: Insights from the fall-out. Eur Heart J. 38:2333–2342. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Chen Y, Li X, Lai W, Zhu F, Tan X, Xian W, Kang P and Wang H: RIP1/RIP3-MLKL signaling pathway correlates with occurrence, progression and prognosis of chronic heart failure. Nan Fang Yi Ke Da Xue Xue Bao. 41:1534–1539. 2021.In Chinese.

72 

Piamsiri C, Maneechote C, Siri-Angkul N, Chattipakorn SC and Chattipakorn N: Targeting necroptosis as therapeutic potential in chronic myocardial infarction. J Biomed Sci. 28:252021. View Article : Google Scholar : PubMed/NCBI

73 

Szobi A, Rajtik T, Carnicka S, Ravingerova T and Adameova A: Mitigation of postischemic cardiac contractile dysfunction by CaMKII inhibition: Effects on programmed necrotic and apoptotic cell death. Mol Cell Biochem. 388:269–276. 2014. View Article : Google Scholar

74 

Zhang T and Brown JH: Role of Ca2+/calmodulin-dependent protein kinase II in cardiac hypertrophy and heart failure. Cardiovasc Res. 63:476–486. 2004. View Article : Google Scholar : PubMed/NCBI

75 

Kerfant BG, Verheule S and Schotten U: Leaky ryanodine receptors in the failing heart: The root of all evil? Cardiovasc Res. 90:399–401. 2011. View Article : Google Scholar : PubMed/NCBI

76 

Sakalihasan N, Limet R and Defawe OD: Abdominal aortic aneurysm. Lancet. 365:1577–1589. 2005. View Article : Google Scholar : PubMed/NCBI

77 

Baxter BT, Terrin MC and Dalman RL: Medical management of small abdominal aortic aneurysms. Circulation. 117:1883–1889. 2008. View Article : Google Scholar : PubMed/NCBI

78 

Legg JS and Legg LM: Abdominal aortic aneurysms. Radiol Technol. 88:145–163. 2016.

79 

Jiang F, Jones GT and Dusting GJ: Failure of antioxidants to protect against angiotensin II-induced aortic rupture in aged apolipoprotein(E)-deficient mice. Br J Pharmacol. 152:880–890. 2007. View Article : Google Scholar : PubMed/NCBI

80 

López-Candales A, Holmes DR, Liao S, Scott MJ, Wickline SA and Thompson RW: Decreased vascular smooth muscle cell density in medial degeneration of human abdominal aortic aneurysms. Am J Pathol. 150:993–1007. 1997.PubMed/NCBI

81 

Guo DC, Papke CL, He R and Milewicz DM: Pathogenesis of thoracic and abdominal aortic aneurysms. Ann N Y Acad Sci. 1085:339–352. 2006. View Article : Google Scholar : PubMed/NCBI

82 

Wang Q, Liu Z, Ren J, Morgan S, Assa C and Liu B: Receptor-interacting protein kinase 3 contributes to abdominal aortic aneurysms via smooth muscle cell necrosis and inflammation. Circ Res. 116:600–611. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Zhou T, Wang Q, Phan N, Ren J, Yang H, Feldman CC, Feltenberger JB, Ye Z, Wildman SA, Tang W and Liu B: Identification of a novel class of RIP1/RIP3 dual inhibitors that impede cell death and inflammation in mouse abdominal aortic aneurysm models. Cell Death Dis. 10:2262019. View Article : Google Scholar : PubMed/NCBI

84 

Wang Q, Zhou T, Liu Z, Ren J, Phan N, Gupta K, Stewart DM, Morgan S, Assa C, Kent KC and Liu B: Inhibition of receptor-interacting protein kinase 1 with necrostatin-1s ameliorates disease progression in elastase-induced mouse abdominal aortic aneurysm model. Sci Rep. 7:421592017. View Article : Google Scholar : PubMed/NCBI

85 

Adamson PD, Dweck MR and Newby DE: The vulnerable atherosclerotic plaque: In vivo identification and potential therapeutic avenues. Heart. 101:1755–1766. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Finney AC, Funk SD, Green JM, Yurdagul A Jr, Rana MA, Pistorius R, Henry M, Yurochko A, Pattillo CB, Traylor JG, et al: EphA2 expression regulates inflammation and fibroproliferative remodeling in atherosclerosis. Circulation. 136:566–582. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Virmani R, Burke AP, Farb A and Kolodgie FD: Pathology of the vulnerable plaque. J Am Coll Cardiol. 47(8 Suppl): C13–C18. 2006. View Article : Google Scholar

88 

Lin J, Li H, Yang M, Ren J, Huang Z, Han F, Huang J, Ma J, Zhang D, Zhang Z, et al: A role of RIP3-mediated macrophage necrosis in atherosclerosis development. Cell Rep. 3:200–210. 2013. View Article : Google Scholar : PubMed/NCBI

89 

Karunakaran D, Geoffrion M, Wei L, Gan W, Richards L, Shangari P, DeKemp EM, Beanlands RA, Perisic L, Maegdefessel L, et al: Targeting macrophage necroptosis for therapeutic and diagnostic interventions in atherosclerosis. Sci Adv. 2:e16002242016. View Article : Google Scholar : PubMed/NCBI

90 

Liu C, Jiang Z, Pan Z and Yang L: The function, regulation and mechanism of programmed cell death of macrophages in atherosclerosis. Front Cell Dev Biol. 9:8095162021. View Article : Google Scholar

91 

Jang WB, Park JH, Ji ST, Lee NK, Kim DY, Kim YJ, Jung SY, Kang S, Lamichane S, Lamichane BD, et al: Cytoprotective roles of a novel compound, MHY-1684, against hyperglycemia-induced oxidative stress and mitochondrial dysfunction in human cardiac progenitor cells. Oxid Med Cell Longev. 2018:45281842018. View Article : Google Scholar : PubMed/NCBI

92 

Poornima IG, Parikh P and Shannon RP: Diabetic cardiomyopathy: The search for a unifying hypothesis. Circ Res. 98:596–605. 2006. View Article : Google Scholar

93 

Task Force on diabetes, pre-diabetes, and cardiovascular diseases of the European Society of Cardiology (ESC); European Association for the Study of Diabetes (EASD); Rydén L, Grant PJ, Anker SD, Berne C, Cosentino F, Danchin N, Deaton C, Escaned J, et al: ESC guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD-summary. Diab Vasc Dis Res. 11:133–173. 2014. View Article : Google Scholar

94 

Wei J, Zhao Y, Liang H, Du W and Wang L: Preliminary evidence for the presence of multiple forms of cell death in diabetes cardiomyopathy. Acta Pharm Sin B. 12:1–17. 2022. View Article : Google Scholar : PubMed/NCBI

95 

Frustaci A, Kajstura J, Chimenti C, Jakoniuk I, Leri A, Maseri A, Nadal-Ginard B and Anversa P: Myocardial cell death in human diabetes. Circ Res. 87:1123–1132. 2000. View Article : Google Scholar : PubMed/NCBI

96 

Chen Y, Hua Y, Li X, Arslan IM, Zhang W and Meng G: Distinct types of cell death and the implication in diabetic cardiomyopathy. Front Pharmacol. 11:422020. View Article : Google Scholar : PubMed/NCBI

97 

Liu YS, Huang ZW, Wang L, Liu XX, Wang YM, Zhang Y and Zhang M: Sitagliptin alleviated myocardial remodeling of the left ventricle and improved cardiac diastolic dysfunction in diabetic rats. J Pharmacol Sci. 127:260–274. 2015. View Article : Google Scholar

98 

Liang W, Chen M, Zheng D, He J, Song M, Mo L, Feng J and Lan J: A novel damage mechanism: Contribution of the interaction between necroptosis and ROS to high glucose-induced injury and inflammation in H9c2 cardiac cells. Int J Mol Med. 40:201–208. 2017. View Article : Google Scholar : PubMed/NCBI

99 

Luo M, Guan X, Luczak ED, Lang D, Kutschke W, Gao Z, Yang J, Glynn P, Sossalla S, Swaminathan PD, et al: Diabetes increases mortality after myocardial infarction by oxidizing CaMKII. J Clin Invest. 123:1262–1274. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Hegyi B, Bers DM and Bossuyt J: CaMKII signaling in heart diseases: Emerging role in diabetic cardiomyopathy. J Mol Cell Cardiol. 127:246–259. 2019. View Article : Google Scholar : PubMed/NCBI

101 

Yamagishi S, Fukami K and Matsui T: Evaluation of tissue accumulation levels of advanced glycation end products by skin autofluorescence: A novel marker of vascular complications in high-risk patients for cardiovascular disease. Int J Cardiol. 185:263–268. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Simó-Servat O, Planas A, Ciudin A, Simó R and Hernández C: Assessment of advanced glycation end-products as a biomarker of diabetic outcomes. Endocrinol Diabetes Nutr (Engl Ed). 65:540–545. 2018.PubMed/NCBI

103 

Hua Y, Qian J, Cao J, Wang X, Zhang W and Zhang J: Ca2+/calmodulin-dependent protein kinase II regulation by inhibitor of receptor interacting protein kinase 3 alleviates necroptosis in glycation end products-induced cardiomyocytes injury. Int J Mol Sci. 23:69882022. View Article : Google Scholar

104 

Fang L, Beale A, Ellims AH, Moore XL, Ling LH, Taylor AJ, Chin-Dusting J and Dart AM: Associations between fibrocytes and postcontrast myocardial T1 times in hypertrophic cardiomyopathy. J Am Heart Assoc. 2:e0002702013. View Article : Google Scholar : PubMed/NCBI

105 

Maron BJ and Maron MS: Hypertrophic cardiomyopathy. Lancet. 381:242–255. 2013. View Article : Google Scholar

106 

Yao J, Qin X, Zhu J and Sheng H: Dyrk1A-ASF-CaMKIIδ signaling is involved in valsartan inhibition of cardiac hypertrophy in renovascular hypertensive rats. Cardiology. 133:198–204. 2016. View Article : Google Scholar

107 

Li C, Cai X, Sun H, Bai T, Zheng X, Zhou XW, Chen X, Gill DL, Li J and Tang XD: The δA isoform of calmodulin kinase II mediates pathological cardiac hypertrophy by interfering with the HDAC4-MEF2 signaling pathway. Biochem Biophys Res Commun. 409:125–130. 2011. View Article : Google Scholar : PubMed/NCBI

108 

Staerk L, Sherer JA, Ko D, Benjamin EJ and Helm RH: Atrial fibrillation: Epidemiology, pathophysiology, and clinical outcomes. Circ Res. 120:1501–1517. 2017. View Article : Google Scholar : PubMed/NCBI

109 

Nattel S and Harada M: Atrial remodeling and atrial fibrillation: Recent advances and translational perspectives. J Am Coll Cardiol. 63:2335–2345. 2014. View Article : Google Scholar

110 

Wakili R, Voigt N, Kääb S, Dobrev D and Nattel S: Recent advances in the molecular pathophysiology of atrial fibrillation. J Clin Invest. 121:2955–2968. 2011. View Article : Google Scholar : PubMed/NCBI

111 

Calvo D, Filgueiras-Rama D and Jalife J: Mechanisms and drug development in atrial fibrillation. Pharmacol Rev. 70:505–525. 2018. View Article : Google Scholar : PubMed/NCBI

112 

Liu Z, Finet JE, Wolfram JA, Anderson ME, Ai X and Donahue JK: Calcium/calmodulin-dependent protein kinase II causes atrial structural remodeling associated with atrial fibrillation and heart failure. Heart Rhythm. 16:1080–1088. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Lee JM, Yoshida M, Kim MS, Lee JH, Baek AR, Jang AS, Kim DJ, Minagawa S, Chin SS, Park CS, et al: Involvement of alveolar epithelial cell necroptosis in idiopathic pulmonary fibrosis pathogenesis. Am J Respir Cell Mol Biol. 59:215–224. 2018. View Article : Google Scholar : PubMed/NCBI

114 

Wei S, Zhou H, Wang Q, Zhou S, Li C, Liu R, Qiu J, Shi C and Lu L: RIP3 deficiency alleviates liver fibrosis by inhibiting ROCK1-TLR4-NF-κB pathway in macrophages. FASEB J. 33:11180–11193. 2019. View Article : Google Scholar : PubMed/NCBI

115 

Mukherjee S, Sheng W, Sun R and Janssen LJ: Ca2+/calmodulin-dependent protein kinase IIβ and IIδ mediate TGFβ-induced transduction of fibronectin and collagen in human pulmonary fibroblasts. Am J Physiol Lung Cell Mol Physiol. 312:L510–L519. 2017. View Article : Google Scholar

116 

Cui H, Zuo S, Liu Z, Liu H, Wang J, You T, Zheng Z, Zhou Y, Qian X, Yao H, et al: The support of genetic evidence for cardiovascular risk induced by antineoplastic drugs. Sci Adv. 6:eabb85432020. View Article : Google Scholar : PubMed/NCBI

117 

Yeh ETH and Bickford CL: Cardiovascular complications of cancer therapy: Incidence, pathogenesis, diagnosis, and management. J Am Coll Cardiol. 53:2231–2247. 2009. View Article : Google Scholar

118 

Curigliano G, Cardinale D, Dent S, Criscitiello C, Aseyev O, Lenihan D and Cipolla CM: Cardiotoxicity of anticancer treatments: Epidemiology, detection, and management. CA Cancer J Clin. 66:309–325. 2016. View Article : Google Scholar

119 

Liang Z, He Y and Hu X: Cardio-oncology: Mechanisms, drug combinations, and reverse cardio-oncology. Int J Mol Sci. 23:106172022. View Article : Google Scholar : PubMed/NCBI

120 

Carvalho C, Santos RX, Cardoso S, Correia S, Oliveira PJ, Santos MS and Moreira PI: Doxorubicin: The good, the bad and the ugly effect. Curr Med Chem. 16:3267–3285. 2009. View Article : Google Scholar

121 

Fer reira AL, Matsubara LS and Matsubara BB: Anthracycline-induced cardiotoxicity. Cardiovasc Hematol Agents Med Chem. 6:278–281. 2008. View Article : Google Scholar

122 

Stefenelli T, Kuzmits R, Ulrich W and Glogar D: Acute vascular toxicity after combination chemotherapy with cisplatin, vinblastine, and bleomycin for testicular cancer. Eur Heart J. 9:552–556. 1988. View Article : Google Scholar

123 

Lejonc JL, Vernant JP, Macquin J and Castaigne A: Myocardial infarction following vinblastine treatment. Lancet. 2:6921980. View Article : Google Scholar : PubMed/NCBI

124 

Calafat AM, Ye X, Wong LY, Reidy JA and Needham LL: Exposure of the U.S. population to bisphenol A and 4-tertiary-octylphenol: 2003-2004. Environ Health Perspect. 116:39–44. 2008. View Article : Google Scholar : PubMed/NCBI

125 

Vandenberg LN, Hauser R, Marcus M, Olea N and Welshons WV: Human exposure to bisphenol A (BPA). Reprod Toxicol. 24:139–177. 2007. View Article : Google Scholar : PubMed/NCBI

126 

Gao X and Wang HS: Impact of bisphenol a on the cardiovascular system-epidemiological and experimental evidence and molecular mechanisms. Int J Environ Res Public Health. 11:8399–8413. 2014. View Article : Google Scholar : PubMed/NCBI

127 

Moon S, Yu SH, Lee CB, Park YJ, Yoo HJ and Kim DS: Effects of bisphenol A on cardiovascular disease: An epidemiological study using national health and nutrition examination survey 2003-2016 and meta-analysis. Sci Total Environ. 763:1429412021. View Article : Google Scholar

128 

Conrad M, Angeli JP, Vandenabeele P and Stockwell BR: Regulated necrosis: Disease relevance and therapeutic opportunities. Nat Rev Drug Discov. 15:348–366. 2016. View Article : Google Scholar : PubMed/NCBI

129 

Kaiser WJ, Sridharan H, Huang C, Mandal P, Upton JW, Gough PJ, Sehon CA, Marquis RW, Bertin J and Mocarski ES: Toll-like receptor 3-mediated necrosis via TRIF, RIP3, and MLKL. J Biol Chem. 288:31268–31279. 2013. View Article : Google Scholar : PubMed/NCBI

130 

Mandal P, Berger SB, Pillay S, Moriwaki K, Huang C, Guo H, Lich JD, Finger J, Kasparcova V, Votta B, et al: RIP3 induces apoptosis independent of pronecrotic kinase activity. Mol Cell. 56:481–495. 2014. View Article : Google Scholar : PubMed/NCBI

131 

Lim SY, Davidson SM, Mocanu MM, Yellon DM and Smith CCT: The cardioprotective effect of necrostatin requires the cyclophilin-D component of the mitochondrial permeability transition pore. Cardiovasc Drugs Ther. 21:467–469. 2007. View Article : Google Scholar : PubMed/NCBI

132 

Smith CCT, Davidson SM, Lim SY, Simpkin JC, Hothersall JS and Yellon DM: Necrostatin: A potentially novel cardioprotective agent? Cardiovasc Drugs Ther. 21:227–233. 2007. View Article : Google Scholar : PubMed/NCBI

133 

Zhou H, Hu B and Liu X: Thyroid hormone metabolite 3-iodothyronamine (T1AM) alleviates hypoxia/reoxygenation-induced cardiac myocyte apoptosis via Akt/FoxO1 pathway. Med Sci Monit. 26:e9231952020.PubMed/NCBI

134 

Meng Y, Tao Z, Zhou S, Da W and Tao L: Research hot spots and trends on melatonin from 2000 to 2019. Front Endocrinol (Lausanne). 12:7539232021. View Article : Google Scholar : PubMed/NCBI

135 

Hu Q, Wu D, Ma F, Yang S, Tan B, Xin H, Gu X, Chen X, Chen S, Mao Y and Zhu YZ: Novel angiogenic activity and molecular mechanisms of ZYZ-803, a slow-releasing hydrogen sulfide-nitric oxide hybrid molecule. Antioxid Redox Signal. 25:498–514. 2016. View Article : Google Scholar

136 

Chang L, Wang Z, Ma F, Tran B, Zhong R, Xiong Y, Dai T, Wu J, Xin X, Guo W, et al: ZYZ-803 Mitigates endoplasmic reticulum stress-related necroptosis after acute myocardial infarction through downregulating the RIP3-CaMKII signaling pathway. Oxid Med Cell Longev. 2019:61736852019. View Article : Google Scholar : PubMed/NCBI

137 

Adameova A, Carnicka S, Rajtik T, Szobi A, Nemcekova M, Svec P and Ravingerova T: Upregulation of CaMKIIδ during ischaemia-reperfusion is associated with reperfusion-induced arrhythmias and mechanical dysfunction of the rat heart: Involvement of sarcolemmal Ca2+-cycling proteins. Can J Physiol Pharmacol. 90:1127–1134. 2012. View Article : Google Scholar : PubMed/NCBI

138 

Pallien T and Klussmann E: New aspects in cardiac L-type Ca2+ channel regulation. Biochem Soc Trans. 48:39–49. 2020. View Article : Google Scholar : PubMed/NCBI

139 

Gao H, Wang F, Wang W, Makarewich CA, Zhang H, Kubo H, Berretta RM, Barr LA, Molkentin JD and Houser SR: Ca(2+) influx through L-type Ca(2+) channels and transient receptor potential channels activates pathological hypertrophy signaling. J Mol Cell Cardiol. 53:657–667. 2012. View Article : Google Scholar : PubMed/NCBI

140 

Ikeda S, Matsushima S, Okabe K, Ikeda M, Ishikita A, Tadokoro T, Enzan N, Yamamoto T, Sada M, Deguchi H, et al: Blockade of L-type Ca2+ channel attenuates doxorubicin-induced cardiomyopathy via suppression of CaMKII-NF-κB pathway. Sci Rep. 9:98502019. View Article : Google Scholar

141 

Lim WY, Messow CM and Berry C: Cyclosporin variably and inconsistently reduces infarct size in experimental models of reperfused myocardial infarction: A systematic review and meta-analysis. Br J Pharmacol. 165:2034–2043. 2012. View Article : Google Scholar

142 

Piot C, Croisille P, Staat P, Thibault H, Rioufol G, Mewton N, Elbelghiti R, Cung TT, Bonnefoy E, Angoulvant D, et al: Effect of cyclosporine on reperfusion injury in acute myocardial infarction. N Engl J Med. 359:473–481. 2008. View Article : Google Scholar : PubMed/NCBI

143 

Cung TT, Morel O, Cayla G, Rioufol G, Garcia-Dorado D, Angoulvant D, Bonnefoy-Cudraz E, Guérin P, Elbaz M, Delarche N, et al: Cyclosporine before PCI in patients with acute myocardial infarction. N Engl J Med. 373:1021–1031. 2015. View Article : Google Scholar : PubMed/NCBI

144 

Ottani F, Latini R, Staszewsky L, La Vecchia L, Locuratolo N, Sicuro M, Masson S, Barlera S, Milani V, Lombardi M, et al: Cyclosporine A in reperfused myocardial infarction: The multicenter, controlled, open-label CYCLE trial. J Am Coll Cardiol. 67:365–374. 2016. View Article : Google Scholar : PubMed/NCBI

145 

Gill RS, Bigam DL and Cheung PY: The role of cyclosporine in the treatment of myocardial reperfusion injury. Shock. 37:341–347. 2012. View Article : Google Scholar : PubMed/NCBI

146 

Zhang CX, Cheng Y, Liu DZ, Liu M, Cui H, Zhang BL, Mei QB and Zhou SY: Mitochondria-targeted cyclosporin A delivery system to treat myocardial ischemia reperfusion injury of rats. J Nanobiotechnology. 17:182019. View Article : Google Scholar : PubMed/NCBI

147 

Yao LL, Huang XW, Wang YG, Cao YX, Zhang CC and Zhu YC: Hydrogen sulfide protects cardiomyocytes from hypoxia/reoxygenation-induced apoptosis by preventing GSK-3beta-dependent opening of mPTP. Am J Physiol Heart Circ Physiol. 298:H1310–H1319. 2010. View Article : Google Scholar : PubMed/NCBI

148 

Kar S, Kambis TN and Mishra PK: Hydrogen sulfide-mediated regulation of cell death signaling ameliorates adverse cardiac remodeling and diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol. 316:H1237–H1252. 2019. View Article : Google Scholar :

149 

Sun HJ, Wu ZY, Nie XW, Wang XY and Bian JS: An updated insight into molecular mechanism of hydrogen sulfide in cardiomyopathy and myocardial ischemia/reperfusion injury under diabetes. Front Pharmacol. 12:6518842021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2023
Volume 52 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen S, Guan S, Yan Z, Ouyang F, Li S, Liu L and Zhong J: Role of RIPK3‑CaMKII‑mPTP signaling pathway‑mediated necroptosis in cardiovascular diseases (Review). Int J Mol Med 52: 98, 2023
APA
Chen, S., Guan, S., Yan, Z., Ouyang, F., Li, S., Liu, L., & Zhong, J. (2023). Role of RIPK3‑CaMKII‑mPTP signaling pathway‑mediated necroptosis in cardiovascular diseases (Review). International Journal of Molecular Medicine, 52, 98. https://doi.org/10.3892/ijmm.2023.5301
MLA
Chen, S., Guan, S., Yan, Z., Ouyang, F., Li, S., Liu, L., Zhong, J."Role of RIPK3‑CaMKII‑mPTP signaling pathway‑mediated necroptosis in cardiovascular diseases (Review)". International Journal of Molecular Medicine 52.4 (2023): 98.
Chicago
Chen, S., Guan, S., Yan, Z., Ouyang, F., Li, S., Liu, L., Zhong, J."Role of RIPK3‑CaMKII‑mPTP signaling pathway‑mediated necroptosis in cardiovascular diseases (Review)". International Journal of Molecular Medicine 52, no. 4 (2023): 98. https://doi.org/10.3892/ijmm.2023.5301