Skip to main content

Endoplasmic reticulum stress signaling modulates ischemia/reperfusion injury in the aged heart by regulating mitochondrial maintenance

Abstract

Aging is associated with an increased risk of myocardial ischemia/reperfusion injury (IRI). With an increasing prevalence of cardiovascular diseases such as coronary arteriosclerosis in older people, there has been increasing interest in understanding the mechanisms of myocardial IRI to develop therapeutics that can attenuate its damaging effects. Previous studies identified that abnormal mitochondria, involved in cellar senescence and oxidative stress, are the master subcellular organelle that induces IRI. In addition, endoplasmic reticulum (ER) stress is also associated with IRI. Cellular adaptation to ER stress is achieved by the activation of ER molecular chaperones and folding enzymes, which provide an important link between ER stress and oxidative stress gene programs. In this review, we outline how these ER stress-related molecules affect myocardial IRI via the crosstalk of ER stress and mitochondrial homeostasis and discuss how these may offer promising novel therapeutic targets and strategies against age-related cardiovascular diseases.

Introduction

Coronary revascularization procedures and medical therapies have reduced the global age-standardized prevalence, disability-adjusted life years (DALYs), and mortality from acute myocardial infarction (AMI) and coronary artery disease. However, ischemic heart disease remains a major public health challenge. From 1990 to 2019, the global prevalence of ischemic heart disease peaked in the 85–89 group for males and in the 90–94 age range for females (Safiri et al. 2022). Compared to healthy hearts, aged hearts are more susceptible to ischemic insults and undergo greater damage in response to ischemia/reperfusion (IR) (Zhang et al. 2020a, b). They also gain less protection from ischemic preconditioning against ischemia/reperfusion injury (IRI) (Munckhof et al. 2013). A combination of an aging population combined with the fact that individuals aged over 65 years account for over 80% of ischemic heart disease (Rosamond et al. 2007) has led IRI to become the major cause of global cardiovascular disease-related disability and mortality. IRI is characterized by microvascular perfusion defects, platelet activation, and sequential cardiomyocyte death due to additional ischemic events at the reperfusion stage (Wang et al. 2020a, b, c; Obas and Vasan 1979) (Fig. 1). As such, there is a critical need to explore the structural and functional changes of the aging heart during IRI.

Fig. 1
figure 1

IRI is more likely to occur in the aging heart afflicted with ischemic cardiomyopathy (ICM), such as angina, AMI, arrhythmia, myocardial fibrosis, and heart failure, which are significant causes of cardiovascular disease-related morbidity and mortality

Mitochondria are bioenergetic, biosynthetic, signaling organelles that provide a critical stress-sensing function that helps cells adapt to their environment (Vyas et al. 2016). Over the past few decades, many investigators, including our team, have firmly concluded that the mitochondrion is a central contributor to IRI (Bugger and Pfeil 2020; Wang et al. 2021; Zhang et al. 2021a, b; Kuang et al. 2023). Under ischemic conditions, mitochondrial membrane potential (ΔΨm) is markedly reduced and adenosine triphosphate (ATP) stores become exhausted; this is accompanied by acidosis that is secondary to lactate accumulation, and an increase in the intracellular calcium (Ca2+) concentration. Meanwhile, the outer mitochondrial membrane (OMM) remains intact and the mitochondrial permeability transition pore (mPTP) remains closed. With subsequent reperfusion, the reintroduction of oxygen leads to a rapid normalization of pH and a rapid restoration in ΔΨm, precipitating a range of adverse sequelae including the production of mitochondrial reactive oxygen species (ROS), exacerbating Ca2+ overload, OMM destruction, and mPTP formation (Kulek et al. 2020). Aging aggravates these mitochondrial events that are initiated by IRI (Campbell et al. 2019; Salehpour et al. 2019) (Fig. 2). Most of the cardiomyocyte oxidative damage can be attributed to these mitochondrial dysfunctions (Paradies et al. 2018). In addition, IR stress also induces mitochondrial biosynthesis, mitochondrial dynamics (fusion-fission)/mitophagy, and mitochondrial proteostasis which facilitate mitochondrial quality control (MQC). It is thought that proper MQC is a compensatory molecular mechanism that removes anomalous mitochondrial proteins or entirely injured mitochondria, and restores mitochondrial function and homeostasis in the setting of IR, favoring cardioprotection (Wang and Zhou 2020). In contrast, MOC disruptions accelerate lethal myocardial IRI (Zhu et al. 2021). Aging impairs mitochondrial function, exacerbating mitochondrial dysfunction and imbalances in MQC, thus leading to greater cardiac injury during IR (Chen et al. 2021).

Fig. 2
figure 2

With age, ΔΨm dissipation, loss of ATP production, increasing intracellular Ca2+, toxic ROS generated during ischemia gradually disrupt mitochondrial homeostasis. Subsequent reperfusion opens mPTP, eliciting ΔΨm rising, calcium overload, and overproduction of ROS, thereby contributing to mitochondrial damage and, ultimately, cell death

In addition to the mitochondria and oxidative stress, the endoplasmic reticulum (ER) also plays an important role in IR. ER is a dynamic and stable organelle that assists in protein synthesis, modification, and topology, as well as regulating cellular signal transduction mainly via its function as a dynamic Ca2+ storage compartment (Görlach et al. 2006). IR can alter ER function, triggering the accumulation of unfolded/misfolded proteins and resulting in evolutionarily conserved cell stress responses, namely ER stress (Zhang et al. 2021a, b). ER stress is marked by glucose-regulated protein 78/immunoglobulin heavy chain-binding protein(GRP78/BiP) and C/EBP homologous protein (CHOP) and activates the unfolded protein response (UPR), which involves three principal ER stress sensors: inositol-requiring enzyme 1α (IRE1α), protein kinase R-like ER kinase (PERK), and activating transcription factor 6 (ATF6) (Zhang et al. 2021a, b; He et al. 2018; Sun et al. 2022). These degrade and remove aberrant proteins from the ER lumen (Zhang et al. 2021a, b; He et al. 2018; Sun et al. 2022) (Fig. 3). The resulting ER stress retards protein translation, increases ER chaperone production, and enhances ER-associated protein degradation (ERAD), which together act to restore intracellular homeostasis (Wang et al. 2014). We previously investigated the effects and mechanisms of ER stress concerning the amelioration of IRI (Zhang et al. 2021a, b), finding that well-organized ER stress helps sustain the viability of cardiomyocytes and the integrity of cardiac function. In contrast, aging increases the risk of excessively activated ER stress, which is closely linked to the pathogenesis of cardiac IRI through mechanisms such as calcium imbalance, free radical overproduction, and ER-dependent cardiomyocyte apoptosis (Zhong et al. 2018; Zhu et al. 2018). However, the role of ER stress in aging-associated cardiac IRI remains incompletely understood.

Fig. 3
figure 3

Three response pathways (IRE1α, ATF6, and PERK) of UPR regulate the ER stress response. Upon dimerization and autophosphorylation, IRE1α, a transmembrane RNase, splices XBP1 pre-mRNA to XBP1 mRNA, which is translated into an active transcriptional factor, activating the transcription of UPR genes encoding ER chaperones. IRE1α can also recruit TRAF2 and ASK1, leading to the downstream activation of JNK. The transcription factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis in response to ER stress. Active PERK phosphorylates eIF2α, inhibiting general protein translation. Phosphorylation of eIF2α also allows for the preferential translation of ATF4. Diversified ER chaperone genes are the transcription targets of ATF4

Because ER stress affects the synthesis and modification of nuclear, cytoplasmic, and mitochondrial proteins, it inevitably modulates mitochondrial function and structure via the UPR signaling pathway during IRI (Zhang et al. 2021a, b). Moreover, mitochondria and ER are physically connected through an inner membrane network, called mitochondrial associated membranes (MAMs), which plays a key role in the interaction between Ca2+ communication, mitochondrial dynamics, and ER stress (Tilokani et al. 2018). For example, the inositol 1,4,5-trisphosphate receptor 1/GRP75/voltage-dependent anion channel 1 (IP3R1/GRP75/VDAC1) complex on the MAM is the main channel for calcium transfer from the endoplasmic reticulum to the mitochondria, viz. mPTP (Paillard et al. 2013; Yuan et al. 2022). Since aging increases in ER stress precede mitochondrial dysfunction (Barbero-Camps et al. 2014; Chen et al. 2020), it is now generally accepted that ER stress is a critical trigger of cardiac mitochondrial instability in elderly individuals undergoing IRI (Chen et al. 2020).

The importance of ER stress in the transformation of mitochondrial structure and metabolism, especially in aging and severe stimulation of IR, is reflected by the plethora of studies into ER stress and the associated regulation of mitochondrial maintenance in cardiac IRI (Zhu and Zhou 2021; Yan et al. 2019; Petrovski et al. 2011). In this review, we outline the key molecular pathways and cellular processes initiated by ER stress, and their roles in the mitochondrial modulation of cardiac IRI to identify potential therapeutic targets for cardiac IRI in the aged heart.

IRE1α

Upon ER stress, IRE1α is auto-phosphorylated and transformed into an active dimer, catalyzing the excision of an intron from the X box-binding protein 1-unspliced isoform (XBP1u) mRNA (Zhang et al. 2021a, b; Sun et al. 2022). The resulting spliced form of XBP1 (XBP1s) is a highly potent transcription factor, which promotes the transcription of target ER chaperones and molecules that regulate mitochondrial maintenance (Wang et al. 2014; Yan et al. 2019; Matsui et al. 2008; Bi et al. 2018; Zhang et al. 2023). The level of XBP1 is significantly increased in the aging heart; XBP1s is considered an anti-aging factor with the ability to prevent or delay heart failure (Duan et al. 2016; Jiao et al. 2012). During cardiac IR, IRE1α, XBP1s, GRP78/BiP, and CHOP are increased in hypoxic or reoxygenated cardiomyocytes at the mRNA and protein levels, suggesting that activation of the IRE1α-XBP1s axis is the most highly conserved branch in ER stress (Liu and Dudley 2014).

XBP plays a dual role in that it aggravates and defends against IRI. Several studies from our laboratory and others have revealed that IRE1α-XBP1 induces the transcriptional upregulation of hydroxymethylglutaryl reductase degradation 1 (HRD1) (Zhang et al. 2021a, b; Cominacini et al. 2015; Wu et al. 2014). HRD1, also called synoviolin 1 (SYVN1), is an E3 ubiquitin ligase coordinating ERAD that directly catalyzes ubiquitin conjugation onto unfolded or misfolded proteins for proteasomal degradation (Binder et al. 2022). We already know that HRD1 can downregulate nuclear factor erythroid 2-related factor 2 (NRF2 coded by nuclear factor, erythroid 2 like 2 (NFE2L2) through a ubiquitination-degradation pathway, while HRD1 inhibition can prevent the downregulation of NRF2 in cardiomyocytes (Zhang et al. 2021a, b; Wu et al. 2014; Binder et al. 2022). Importantly, the transcription factor NRF2 is best known as an anti-aging element (Cominacini et al. 2015; Zhang et al. 2020a, b), reducing myocardial infarct size and cardiac hypertrophy in the transition to heart failure in animals exposed to IRI (Cominacini et al. 2015). In terms of interaction with mitochondria, NRF2 regulates responses to mitochondrial-derived ROS via its effects on antioxidant and detoxification genes, including NAD(P)H: quinone oxidoreductase (NQO1), heme oxygenase-1 (HO-1), and superoxide dismutase-1 (SOD-1) (Cominacini et al. 2015; Zhang et al. 2020a, b; Chen and Maltagliati 2018). NRF2 transcription can also increase mitochondrial biogenesis through the expression of genes that are essential for mitochondrial biogenesis such as nuclear respiratory factor 1 (Athale et al. 2012; Chen 2021).

In addition, XBP1 couples ER stress with the hexosamine biosynthetic pathway (HBP) by triggering HBP activation of the major HBP enzymes, glutamine fructose-6-phosphate aminotransferase 1 (GFAT1), glucosamine-phosphate N-acetyltransferase (GNPNAT1), phosphoglucomutase 3 (PGM3), and uridine diphosphate-glucose 4-epimease (GalE) (Wang et al. 2014). The IRE1α-XBP1s-HBP axis promotes the synthesis of uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), which is the obligate substrate for O-linked GlcNAc modification (O-GlcNAcylation) on cardiac proteins under IRI conditions (Wang et al. 2014). O-GlcNAcylation, a prosurvival pathway that counterbalances the age-related decline in self-healing capacity (Jiang et al. 2017), can modulate protein stability and function and has been implicated in various IRI-related cardiovascular diseases. Current evidence suggests that increasing O-GlcNAcylation during IRI may represent a unique endogenously recruitable mechanism of cardioprotection via directly O-GlcNAcylating VDAC in mitochondria (Ngoh et al. 2008, 2009; Jones et al. 2008). Because VDAC is a putative member of the mPTP, O-GlcNAcylation of VDAC prevents the formation of the mPTP, alters sensitivity to the loss of ΔΨm, relieves Ca2+ overload-induced mitochondrial swelling, and hence maintains mitochondrial stability (Ngoh et al. 2008).

XBP1 is not the only RNA targeted by the RNase activity of IRE1α. Prolonged ER stress attenuates the expression of XBP1, but continues to engage regulated IRE1-dependent decay (RIDD) and IRE1α-stimulation of the apoptotic-signaling kinase-1 (ASK1) (Nishitoh et al. 2002), which causes activation of Jun-N-terminal kinase (JNK) (Urano et al. 2000). ASK1 participates in the regulation of NRF2 phosphorylation and dissociation from Kelch-like ECH-associated protein 1 (Keap1) (Nishitoh et al. 2002), leading to the nuclear translocation of NRF2. JNK resists cardiac IRI by repressing the mitochondrial fission factor (Mff)-mediated mitochondrial fission and the Bcl-2/adenovirus E1B 19 kDa interacting protein-3 (Bnip-3)-required mitophagy (Jin et al. 2018).

ATF6

ATF6 has two isoforms, ATF6α (90 kDa) and ATF6β (110 kDa), and is a transmembrane sensor protein that becomes a powerful transcription factor upon regulated intramembrane proteolysis-specific cleavage in the Golgi complex (Liu and Dudley 2014). After cleavage, the cytosolic N-terminal fragments of ATF6, containing conserved DNA-binding domains and divergent transcriptional activation domains, translocate into the nucleus, where it binds to several types of regulatory sequences in target ER stress element (ERSE) genes, and activates IRI-mediated ER stress response gene transcription (Tadimalla et al. 2008). The ATF6α arm of ER stress safeguards organelle homeostasis and cellular aging (Wang et al. 2018); it can be activated in ischemia and inactivated upon reperfusion (Liu and Dudley 2014), serving protective roles in the IRI heart through its induction of numerous ER stress-related molecules, especially following ischemic preconditioning (Brooks et al. 2014).

During IR, ATF6 was identified to underlie the altered expression of 18 genes. Twelve oxidative stress response genes are upregulated by ATF6; they encode proteins with strong antioxidant and scavenging effects on free radicals. Six genes that respond to oxidative stress are decreased by ATF6 but do not encode antioxidants (Jin et al. 2017; Glembotski et al. 2020; Marsh et al. 2021). The most well-characterized of these in cardiac myocytes subjected to IR include peroxiredoxin 5 (Prdx5), catalase (CAT), valosin-containing protein-interacting membrane seprotein (Vimp), and fatty acyl-CoA reductase 1 (FAR1) (Jin et al. 2017). Prdx5 is a member of the family of mammalian proteins that neutralize ROS and is highly constitutively expressed in mammalian hearts but its basal levels are reduced in aged hearts (Kropotov et al. 2007; Edwards et al. 2003). In cardiomyocytes, Prdx5 is distributed in several cellular locations including the mitochondria, peroxisomes, cytosol, and nucleus; it possesses mitochondria and peroxisome targeting sequences, and plays a major role in mitochondrial antioxidant defenses by catalyzing the reduction of potentially damaging peroxides and superoxide through thioredoxin (Jin et al. 2017; Agborbesong et al. 2023). CAT is the longest-recognized antioxidant enzyme; it plays a key role in the metabolism of hydrogen peroxide (H2O2) and reactive nitrogen species and is expressed and localized in cardiac peroxisomes, catalase, cytosol, and mitochondria (Jin et al. 2017; Dieterich et al. 2000). A CAT-overexpressing transgene was shown to prolong lifespan and resist cardiac IRI by attenuating ROS levels (Li et al. 1997; Ren et al. 2007). Vimp, an ER-transmembrane protein that is also called selenoprotein S (SelS), is one of the only 25 genes that encodes the 21st amino acid selenocysteine in humans (Schomburg 2011), contributing to selenium-mediated protection from oxidative stress in cardiomyocytes (Jin et al. 2017). Recently, Far1 was demonstrated to be an ATF6-dependent ER stress-inducible gene, which encodes a protein that localizes to peroxisomes of cardiac myocytes during IR (Marsh et al. 2021). Elevated FAR1 induces robust plasmalogen production during the onset of reperfusion and forms plasmalogen catabolism by an excess of ROS generated at the mitochondria during reperfusion. Nevertheless, FAR1 may lead to a lethal accumulation of lysophospholipids and α-hydroxyaldehydes, which impair myocyte viability during IR (Marsh et al. 2021). Given that ATF6-induced antioxidant proteins are present inside proteasomes, and proteasomes have been found to decorate the surface of mitochondria (Jin et al. 2017; Glembotski et al. 2020; Marsh et al. 2021), a molecular mechanism involving ATF6 regulation of mitochondria may involve the neutralization of some of the ROS generated in mitochondria during cardiac IR.

According to our current understanding, the expression of some microRNAs (miRs) is significantly upregulated during myocardial IR and heart failure (Zhao et al. 2022a, b; Hulot and Masurkar 2020). Apart from its ability to transcribe potentially protective genes, ATF6 also regulates miR expression in the mouse heart (Belmont et al. 2012). Recently, a whole-genome miR array analysis of RNA from the hearts of ATF6 transgenic mice was conducted to determine the mechanisms of ATF6-mediated protection (Belmont et al. 2012). ATF6 was found to upregulate miR-721 and miR-130b, which were predicted to target the ATF6-regulated mRNA of mitochondrial uncoupling protein 3 (UCP3). UCP3, which is predominantly expressed in the heart (Bézaire et al. 2007), is essential for the recovery of long-chain fatty acid (LCFA) oxidation, mitochondrial respiratory capacity (MRC), and contractile function after myocardial IR (Edwards et al. 2018). Mice lacking UCP3 have larger infarcts after IR compared to wild-type mice. Adult cardiomyocytes from Ucp3 knockout mice show mitochondrial dysfunction, increased ROS production, and apoptotic cell death when compared with equivalent wild-type cells during in vitro hypoxia (Perrino et al. 2013).

PERK

IR-stimulated activation of the PERK signaling pathway is important for mitochondrial homeostasis and cell survival, but the mechanisms remain incompletely understood. PERK activation causes the phosphorylation of the α-subunit of eukaryotic initiation factor 2 (eIF2α) that reduces protein translation, thereby allowing the cell to fold proteins that are already present in the ER (Wang et al. 2020a, b, c; Latorre-Muro et al. 2021). The exception is ATF4, which avoids translational attenuation induced by eIF2α phosphorylation, owing to the upstream open reading frames (ORFs) in its 5′-untranslated region. Under normal conditions, the upstream ORFs prevent the translation of true ATF4, but phosphorylated eIF2α neglects these and increases the selective translation of the mRNA encoding transcription factor ATF4 (Zhang et al. 2017). ATF4 is a translational target of activated PERK in cardiovascular diseases. Young tissues possess an efficient PERK-eIF2α activation adaptive mechanism but this declines with aging (Hussain and Ramaiah 2007; Boriushkin et al. 2015).

The integrated stress response (ISR) is a central signaling network that responds to proteostasis defects by tuning the rates of protein synthesis (Costa-Mattioli and Walter 2020). It thereby protects the heart from IRI by inhibiting protein synthesis (Zhang et al. 2021a, b). The PERK-eIF2α cascade is strongly induced by IR in cardiomyocytes in vitro and in vivo and is the cardinal pathophysiological mechanism underlying ISR (Wang et al. 2020a, b, c; Tian et al. 2021; Sun et al. 2019). PERK-eIF2α inhibits the NADH-ubiquinone oxidoreductase complex assembly factor 2 (NDUFAF2), an assembly factor of the mitochondrial complex I, by directly targeting translational suppression and alleviating mitochondrial complex-derived ROS in response to IR (Zhang et al. 2021a, b). Notably, PERK also phosphorylates an unidentified site that lies within the amino terminus of the Neh2 domain of NRF2, which is vital for its interaction with Keap1. It thereby frees NRF2 from its inhibitor and promotes its nuclear translocation to modulate the transcriptional response to preserve cellular homeostasis, in an eIF2α phosphorylation-independent manner (Yamamoto et al. 2018). Concomitantly, PERK can phosphorylate O-GlcNAc transferase (OGT) which, in turn, O-GlcNAcylates the translocase of the mitochondrial import receptor outer mitochondrial membrane 70 (TOM70). This then enhances mitochondrial protein import 19 (MIC19), causes mitochondrial cristae formation, and sustains cellular bioenergetics during conditions of high energy demand to maintain cellular survival and activity (Latorre-Muro et al. 2021; Fahie et al. 2022).

Furthermore, the PERK located at the contact sites between the ER and the mitochondria creates a PERK-mitochondria axis that allows PERK to detect stress in both organelles, adapt their functions, and prevent apoptosis (Almeida et al. 2022). IRI-initiated PERK strengthens MQC by promoting mitophagy, which leads to the renewal of the mitochondrial network (Matsui et al. 2008; Almeida et al. 2022; Kouroku et al. 2007).

GRP94

GRP94, the only member of the heat shock protein (HSP) 90 family that is present in the ER, is coordinately regulated with the GRP78/BiP chaperone system in protein processing and is inducible by ER stress at the transcriptional level (Santana-Codina et al. 2020; Vitadello et al. 2003; Ni et al. 2011). GRP94 decreases with age leading to a significant decrease in the level of IRE1α-XBP1 and a compensatory upregulation of the ER chaperones GRP78/BiP, calnexin, and calreticulin (Mao et al. 2010; Song et al. 2022). The functions of GRP94 include increasing the folding capacity of the ER, assisting in the targeting of malfolded proteins to ERAD, and storing Ca2+ (Eletto et al. 2010). Similar to GRP78/BiP, GRP94 exerts pro-survival functions in cells subjected to ER stress. Following exposure of cells to ER stress induced by IR, a deregulated increase in intracellular Ca2+ can activate mitochondrial Ca2+ uptake, deteriorating normal mitochondrial function, and increasing the production of mitochondrial-generated ROS (Vitadello et al. 2003; Ni et al. 2011). Accordingly, GRP94 protects cardiomyocytes from calcium overload by counteracting the increase of intracellular Ca2+ during IR (Vitadello et al. 2003), but the specific mechanisms still need to be clarified.

Glycogen synthase kinase-3 (GSK-3β)

GSK-3β, an evolutionarily conserved serine/threonine kinase, regulates numerous signaling transduction systems and pathological states that span across cell growth, inflammation, apoptosis, and heart failure (Sharma et al. 2020). The aged heart shows increased levels of activated GSK-3β compared with hearts from young controls (Zhu et al. 2010). In particular, GSK-3β participates in the apoptosis and necrosis of cardiomyocytes after hypoxia/reoxygenation (H/R) (Juhaszova et al. 2009) and occupies an integration point for ER-derived and mitochondria-derived cell death/survival pathways (Miki et al. 2009; Xie et al. 2016). Many therapeutics have attempted to exploit GSK-3β to treat IRI (Kuang et al. 2023; Sharma et al. 2020). We have determined that methyl eugenol (ME), an analog of the phenolic compound eugenol, binds to the binding pocket of Adenosine 5‘-monophosphate (AMP)-activated protein kinase (AMPK) with high affinity, activating the AMPK/GSK3β axis. This, in turn, blocks the NRF2 nuclear export signal and promotes the transcription of antioxidant target genes that help maintain mitochondrial homeostasis (Kuang et al. 2023). Other therapeutic agents, such as fisetin, were also reported to mitigate myocardial IRI and reduce mitochondrial oxidative stress by inhibiting GSK-3β (Sharma et al. 2020; Shanmugam et al. 2018).

IR elevates ROS levels, mostly within and proximal to mitochondria, and can excite mPTP opening, which directly induces ΔΨm loss, with deleterious consequences for the affected mitochondria (Juhaszova et al. 2009; Sheng et al. 2018). The nonphosphorylated form of GSK-3β is constitutively active and forms a complex with two core subunits of mPTP, adenine nucleotide translocase (ANT) and VDAC, diminishes sensitization of mPTP opening, and exerts cardioprotection after IR (Miki et al. 2009). Unfortunately, studies have shown that ER stress can reverse this protective effect by phosphorylating GSK-3β at Ser9 and reducing its activity and mitochondrial translocation, ultimately augmenting the mPTP opening threshold (Miki et al. 2009; Xie et al. 2016).

ER is regarded as a storage location of intracellular zinc (Zn2+) (Ridlo et al. 2021). Interestingly, stimulation of cardiac IR has been confirmed to provoke the loss of intracellular Zn2+ resulting in the release of Zn2+ from the ER. This disturbs Zn2+ homeostasis which normally helps mediate ER stress and plays a role in the protective action of ER stress inhibition of the inactivation of GSK-3β-mediated suppression of mPTP (Sheng et al. 2018; Wang et al. 2016).

Peroxisome proliferator-activated receptor γ co-activator 1-α (PGC-1α)

PGC-1α belongs to a small family of transcriptional coactivators. It plays a pivotal role as a metabolic sensor, is abundant in oxidative active myocardium, and is responsible for the fine-tuning of transcriptional responses to environmental stimuli, determining mitochondrial composition and content (Ding et al. 2018; Packer 2020). PGC-1α regulates the changes that occur in mitochondria during aging; as such, modulation of PGC-1α may offer a therapeutic target for age-related pathology (Halling et al. 2019). Mice with PGC-1α gene deletion have impaired cardiac reserve under stress conditions, but supraphysiological expression of PGC-1α in the heart also leads to cardiomyopathy and heart failure in mice (Arany et al. 2005; Leone et al. 2005). This U-shaped relationship between expression and function suggests that moderate PGC-1α expression is likely to benefit cardiac IRI.

In the myocardium of diabetic rats subjected to IR, ischemic preconditioning with low-dose ER stress induced the expression of PGC-1α, which regulated mitochondrial biogenesis and mitochondrial energy metabolism by interacting with NRF1/NRF2 and mitochondrial transcription factor A (TFAM) (Chen et al. 2022). Beyond this function, ischemic preconditioning induced PGC-1α-mediated mitochondrial fusion-fission directly by binding to key transcriptional promoters, including mitofusin2 (MFN2), dynamin-related protein 1 (DRP1), phosphatase and tensin homolog (PTEN)-induced putative kinase protein1 (PINK1), and Parkin protein, thereby attenuating IRI (Yan et al. 2019; Chen et al. 2022).

Hypoxia-inducible factor 1 (HIF-1)

The transcriptional complex HIF-1 is a key element in sensing changes in cellular oxygen tension. It comprises an oxygen-sensitive α-subunit, which can both promote and limit longevity, and a constitutively-expressed β-subunit, which is rapidly degraded under normoxia and stabilized during hypoxic conditions (Leiser and Kaeberlein 2010; Leiser et al. 2011). It has been increasingly recognized that HIF-1 orchestrates protective responses to hypoxia through the transcriptional activation of a series of hypoxia-sensitive genes (Ong et al. 2014). As expected, HIF-1 plays an ascendant role in mediating cardioprotection against IRI (Ong et al. 2014; Fukuda et al. 2007; Semenza 2011; Belaidi et al. 2016; Yang et al. 2016, 2020). Current evidence supports that ER stress is a new myocardial activator of HIF-1 during cardiac IRI (Belaidi et al. 2016).

Under hypoxic conditions, HIF-1 regulates the expression of the cytochrome c oxidase (COX) 4 subunit by activating the transcription of genes encoding COX4-2 and LON, a mitochondrial ATP-dependent protease required for COX4-1 degradation, optimizing the efficiency of mitochondrial respiration and restraining mitochondrial-derived ROS (Fukuda et al. 2007). Additionally, HIF-1 reprograms basal cell metabolism from oxidative phosphorylation (OXPHOS) towards aerobic glycolysis (AG), thus suppressing the production of mitochondrial oxidative stress and mPTP opening during IRI. This occurs via the activation of glycolytic protein, pyruvate dehydrogenase kinase-1 (PDK1), and mitochondrial hexokinase II (HKII) (Ong et al. 2014). Of note, HIF-1 also activates the gene encoding Bnip-3, which triggers selective mitophagy and restores mitochondrial maintenance throughout myocardial IRI (Yang et al. 2020).

Conclusions and perspectives

Our current understanding of myocardial IRI stresses the important influence of ER stress over mitochondrial energy metabolism, Ca2+ influx, MQC, and mitochondrial ROS production through diverse molecular pathways (Fig. 4). As the nexus between IRI and mitochondria in the aged heart, the multiple biological processes involved in ER stress are interdependent, complex, highly orchestrated, and even appear to be contradictory. In general, ER stress triggered by mild IR, such as ischemic preconditioning, is beneficial to preserving mitochondrial function and eliminating injured mitochondria. Correspondingly, overwhelming ER stress, which is more likely to appear with aging cardiac IR, is always accompanied by aberrant mitochondrial homeostasis. This phenomenon has been observed in the heart, kidney, liver, pancreatic islets, and other tissues in our previous explorations (Wang et al. 2021; Zhang et al. 2021a, b; Kuang et al. 2023, 2024; He et al. 2018; Gong et al. 2022; Ni et al. 2023). As an example, XBP1s, the principal effector of the ER stress sensor IRE1α, is of particular interest to us. Following XBP1s deficiency, we noted augmentation of forkhead box O1 (FoxO1)-dependent mitophagy in hepatocytes and reduction in caspase-1-dependent mitochondrial damage in renal tubular epithelial cells (Ni et al. 2023; Kuang et al. 2024). This suggests that controlling the extent and form of ER stress may be used to regulate IRI in various organs including the heart. For the elderly with a higher risk of AMI, diabetes mellitus, and other cardiovascular-related diseases, it may be necessary to perturb ER stress or interrupt the crosstalk between the ER and mitochondria (Sheng et al. 2018; Chen et al. 2019; Tan et al. 2020). Current researchers have endeavored to diminish the formation of MAMs by decreasing the mitochondrial-ER proximity or disrupting the IP3R1-GRP75-VDAC1 complex during ER stress, blocking Ca2+ communication between the ER and mitochondria, ultimately safeguarding cardiomyocytes from IRI (Li et al. 2024; Zhang et al. 2024; Kirshenbaum et al. 2024). Given the adaptive changes in cardiac IRI and the age-dependent decline of certain UPR molecules and their downstream products in the heart, upregulation of these ER stress pathways has become a target of anti-aging cardiac IRI research (Chen et al. 2020; Edwards et al. 2003; Blackwood et al. 2020). In summary, these evolving findings present encouraging implications for the application of ER-mitochondrial correlations in improving cardiac IRI of aging patients in the clinic.

Fig. 4
figure 4

IRE1α, ATF6, PERK, GRP94, GSK-3β, PGC-1α, and HIF-1 are all stimulated by ER stress when the aged heart undergoes IRI. They act on mitochondrial structure and function in cardiac IRI via mechanistically distinct pathways

Admittedly, how IR-induced ER stress modulates mitochondrial maintenance remains incompletely understood. For instance, after testing GSK-3β, PGC-1α, and HIF-1 regulation by ER stress in cardiomyocytes, their regulatory UPR pathway remains unidentified. A number of nascent hypotheses have emerged. It seems that the calcium/calmodulin-dependent protein kinase IIγ (CaMKIIγ) which is capable of pushing mitochondrial Ca2+ uptake, loss of ΔΨm, and release of apoptogens may serve as a unifying link between ER stress and mitochondrial apoptotic pathways in irreversible cardiac IRI (Vila-Petroff et al. 2007; Timmins et al. 2009; Feng and Anderson 2017). Besides, despite preclinical studies indicating the potential effectiveness of plentiful ER stress and mitochondrial-targeted therapies in treating myocardial IRI, their clinical implementation still remains insufficient. Because of notable distinctions among different cardiovascular diseases, scholars are compelled to employ distinct animal models tailored to each condition for a more comprehensive exploration of the underlying pathogenic mechanisms of each disease, and a thorough evaluation of the efficacy of novel therapies, while the applicability of animal models to humans also requires further investigation.

Data availability

Not applicable.

Abbreviations

IRI:

Ischemia/reperfusion injury

ER:

Endoplasmic reticulum

DALYs:

Disability-adjusted life years

AMI:

Acute myocardial infarction

IR:

Ischemia/reperfusion

ΔΨm:

Mitochondrial membrane potential

ATP:

Adenosine triphosphate

OMM:

Outer mitochondrial membrane

mPTP:

Mitochondrial permeability transition pore

ROS:

Reactive oxygen species

MQC:

Mitochondrial quality control

GRP78/BiP:

Glucose-regulated protein 78/immunoglobulin heavy chain-binding protein

CHOP:

C/EBP homologous protein

UPR:

Unfolded protein response

IRE1α:

Inositol-requiring enzyme 1α

PERK:

Protein kinase R-like ER kinase

ATF6:

Activating transcription factor 6

ERAD:

ER-associated protein degradation

MAMs:

Mitochondrial associated membranes

IP3R1:

Inositol 1,4,5-trisphosphate receptor 1

VDAC:

Voltage-dependent anion channel

XBP1:

X box-binding protein 1

HRD1:

Hydroxymethylglutaryl reductase degradation 1

SYVN1:

Synoviolin 1

NRF2:

Nuclear factor erythroid 2-related factor 2

NQO1:

NAD(P)H: quinone oxidoreductase

HO1-:

Heme oxygenase-1

SOD1-:

Superoxide dismutase-1

HBP:

Hexosamine biosynthetic pathway

GFAT1:

Glutamine fructose-6-phosphate aminotransferase 1

GNPNAT1:

Glucosamine-phosphate N-acetyltransferase

PGM3:

Phosphoglucomutase 3

GalE:

Uridine diphosphate-glucose 4-epimease

UDP:

GlcNAc-Uridine diphosphate N-acetylglucosamine

O:

GlcNAcylation-O-linked GlcNAc modification

RIDD:

IRE1-dependent decay

ASK1:

Apoptotic-signaling kinase-1

JNK:

Jun-N-terminal kinase

Keap1:

Kelch-like ECH-associated protein 1

Mff:

Mitochondrial fission factor

Bnip3-:

Bcl-2/adenovirus E1B 19 kDa interacting protein-3

ERSE:

ER stress element

Prdx5:

Peroxiredoxin 5

CAT:

Catalase

Vimp:

Valosin-containing protein-interacting membrane seprotein

FAR1:

fatty acyl-CoA reductase 1

H2O2 :

Hydrogen peroxide

SelS:

Selenoprotein S

miRs:

MicroRNAs

UCP3:

Uncoupling protein 3

LCFA:

Long-chain fatty acid

MRC:

Mitochondrial respiratory capacity

eIF2α:

A-subunit of eukaryotic initiation factor 2

ORFs:

Open reading frames

ISR:

Integrated stress response

NDUFAF2:

NADH-ubiquinone oxidoreductase complex assembly factor 2

OGT:

O-GlcNAc transferase

TOM70:

Translocase of the mitochondrial import receptor outer mitochondrial membrane 70

MIC19:

Mitochondrial protein import 19

HSP:

Heat shock protein

GSK-3β:

Glycogen synthase kinase-3

H/R:

Hypoxia/reoxygenation

ME:

Methyl eugenol

AMP:

Adenosine 5‘-monophosphate

AMPK:

AMP-activated protein kinase

ANT:

Adenine nucleotide translocase

PGC-1α:

Peroxisome proliferator-activated receptor γ co-activator 1-α

TFAM:

Mitochondrial transcription factor A

MFN2:

Mitofusin2

DRP1:

Dynamin-related protein 1

PTEN:

Phosphatase and tensin homolog

PINK1:

PTEN-induced putative kinase protein1

HIF-1:

Hypoxia-inducible factor 1

COX:

Cytochrome c oxidase

OXPHOS:

Oxidative phosphorylation

AG:

Aerobic glycolysis

PDK1:

Pyruvate dehydrogenase kinase-1

HKII:

Hexokinase II

CaMKIIγ:

Calcium/calmodulin-dependent protein kinase IIγ

FoxO1:

Forkhead box O1

References

  • Agborbesong E, Zhou JX, Li LX, Harris PC, Calvet JP, Li X. Prdx5 regulates DNA damage response through autophagy-dependent Sirt2-p53 axis. Hum Mol Genet. 2023;32(4):567–79.

    Article  CAS  PubMed  Google Scholar 

  • Almeida LM, Pinho BR, Duchen MR, Oliveira JMA. The PERKs of mitochondria protection during stress: insights for PERK modulation in neurodegenerative and metabolic diseases. Biol Rev Camb Philos Soc. 2022;97(5):1737–48.

    Article  CAS  PubMed  Google Scholar 

  • Arany Z, He H, Lin J, Hoyer K, Handschin C, Toka O, et al. Transcriptional coactivator PGC-1 alpha controls the energy state and contractile function of cardiac muscle. Cell Metabol. 2005;1(4):259–71.

    Article  CAS  Google Scholar 

  • Athale J, Ulrich A, MacGarvey NC, Bartz RR, Welty-Wolf KE, Suliman HB, et al. Nrf2 promotes alveolar mitochondrial biogenesis and resolution of lung injury in Staphylococcus aureus pneumonia in mice. Free Radic Biol Med. 2012;53(8):1584–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Barbero-Camps E, Fernández A, Baulies A, Martinez L, Fernández-Checa JC, Colell A. Endoplasmic reticulum stress mediates amyloid β neurotoxicity via mitochondrial cholesterol trafficking. Am J Pathol. 2014;184(7):2066–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Belaidi E, Thomas A, Bourdier G, Moulin S, Lemarié E, Levy P, et al. Endoplasmic reticulum stress as a novel inducer of hypoxia inducible factor-1 activity: its role in the susceptibility to myocardial ischemia-reperfusion induced by chronic intermittent hypoxia. Int J Cardiol. 2016;210:45–53.

    Article  PubMed  Google Scholar 

  • Belmont PJ, Chen WJ, Thuerauf DJ, Glembotski CC. Regulation of microRNA expression in the heart by the ATF6 branch of the ER stress response. J Mol Cell Cardiol. 2012;52(5):1176–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Bézaire V, Seifert EL, Harper ME. Uncoupling protein-3: clues in an ongoing mitochondrial mystery. FASEB Journal: Official Publication Federation Am Soc Experimental Biology. 2007;21(2):312–24.

    Article  Google Scholar 

  • Bi X, Zhang G, Wang X, Nguyen C, May HI, Li X, et al. Endoplasmic reticulum chaperone GRP78 protects Heart from Ischemia/Reperfusion Injury through akt activation. Circul Res. 2018;122(11):1545–54.

    Article  CAS  Google Scholar 

  • Binder P, Nguyen B, Collins L, Zi M, Liu W, Christou F, et al. Pak2 regulation of Nrf2 serves as a Novel Signaling Nexus linking ER stress response and oxidative stress in the heart. Front Cardiovasc Med. 2022;9:851419.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Blackwood EA, Bilal AS, Stauffer WT, Arrieta A, Glembotski CC. Designing Novel therapies to mend broken hearts: ATF6 and Cardiac Proteostasis. Cells. 2020;9(3).

  • Boriushkin E, Wang JJ, Li J, Jing G, Seigel GM, Zhang SX. Identification of p58IPK as a novel neuroprotective factor for retinal neurons. Investig Ophthalmol Vis Sci. 2015;56(2):1374–86.

    Article  CAS  Google Scholar 

  • Brooks AC, Guo Y, Singh M, McCracken J, Xuan YT, Srivastava S, et al. Endoplasmic reticulum stress-dependent activation of ATF3 mediates the late phase of ischemic preconditioning. J Mol Cell Cardiol. 2014;76:138–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Bugger H, Pfeil K. Mitochondrial ROS in myocardial ischemia reperfusion and remodeling. Biochim et Biophys acta Mol Basis Disease. 2020;1866(7):165768.

    Article  CAS  Google Scholar 

  • Campbell MD, Duan J, Samuelson AT, Gaffrey MJ, Merrihew GE, Egertson JD, et al. Improving mitochondrial function with SS-31 reverses age-related redox stress and improves exercise tolerance in aged mice. Free Radic Biol Med. 2019;134:268–81.

    Article  CAS  PubMed  Google Scholar 

  • Chen QM. Nrf2 for cardiac protection: pharmacological options against oxidative stress. Trends Pharmacol Sci. 2021;42(9):729–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Chen QM, Maltagliati AJ. Nrf2 at the heart of oxidative stress and cardiac protection. Physiol Genom. 2018;50(2):77–97.

    Article  CAS  Google Scholar 

  • Chen J, Hu Q, Zhang BF, Liu XP, Yang S, Jiang H. Long noncoding RNA UCA1 inhibits ischaemia/reperfusion injury induced cardiomyocytes apoptosis via suppression of endoplasmic reticulum stress. Genes Genomics. 2019;41(7):803–10.

    Article  CAS  PubMed  Google Scholar 

  • Chen Q, Samidurai A, Thompson J, Hu Y, Das A, Willard B, et al. Endoplasmic reticulum stress-mediated mitochondrial dysfunction in aged hearts. Biochim et Biophys acta Mol Basis Disease. 2020;1866(11):165899.

    Article  CAS  Google Scholar 

  • Chen Q, Thompson J, Hu Y, Lesnefsky EJ. Chronic metformin treatment decreases cardiac injury during ischemia-reperfusion by attenuating endoplasmic reticulum stress with improved mitochondrial function. Aging. 2021;13(6):7828–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Chen L, Qin Y, Liu B, Gao M, Li A, Li X, et al. PGC-1α-Mediated mitochondrial Quality Control: Molecular mechanisms and implications for Heart failure. Front cell Dev Biology. 2022;10:871357.

    Article  Google Scholar 

  • Cominacini L, Mozzini C, Garbin U, Pasini A, Stranieri C, Solani E et al. Endoplasmic reticulum stress and Nrf2 signaling in cardiovascular diseases. Free Radic Biol Med. 2015;88(Pt B):233 – 42.

  • Costa-Mattioli M, Walter P. The integrated stress response: from mechanism to disease. Sci (New York NY). 2020;368(6489).

  • Dieterich S, Bieligk U, Beulich K, Hasenfuss G, Prestle J. Gene expression of antioxidative enzymes in the human heart: increased expression of catalase in the end-stage failing heart. Circulation. 2000;101(1):33–9.

    Article  CAS  PubMed  Google Scholar 

  • Ding M, Feng N, Tang D, Feng J, Li Z, Jia M, et al. Melatonin prevents Drp1-mediated mitochondrial fission in diabetic hearts through SIRT1-PGC1α pathway. J Pineal Res. 2018;65(2):e12491.

    Article  PubMed  PubMed Central  Google Scholar 

  • Duan Q, Ni L, Wang P, Chen C, Yang L, Ma B, et al. Deregulation of XBP1 expression contributes to myocardial vascular endothelial growth factor-A expression and angiogenesis during cardiac hypertrophy in vivo. Aging Cell. 2016;15(4):625–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Edwards MG, Sarkar D, Klopp R, Morrow JD, Weindruch R, Prolla TA. Age-related impairment of the transcriptional responses to oxidative stress in the mouse heart. Physiol Genom. 2003;13(2):119–27.

    Article  CAS  Google Scholar 

  • Edwards KS, Ashraf S, Lomax TM, Wiseman JM, Hall ME, Gava FN, et al. Uncoupling protein 3 deficiency impairs myocardial fatty acid oxidation and contractile recovery following ischemia/reperfusion. Basic Res Cardiol. 2018;113(6):47.

    Article  PubMed  PubMed Central  Google Scholar 

  • Eletto D, Dersh D, Argon Y. GRP94 in ER quality control and stress responses. Semin Cell Dev Biol. 2010;21(5):479–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Fahie KMM, Papanicolaou KN, Zachara NE. Integration of O-GlcNAc into stress response pathways. Cells. 2022;11(21).

  • Feng N, Anderson ME. CaMKII is a nodal signal for multiple programmed cell death pathways in heart. J Mol Cell Cardiol. 2017;103:102–9.

    Article  CAS  PubMed  Google Scholar 

  • Fukuda R, Zhang H, Kim JW, Shimoda L, Dang CV, Semenza GL. HIF-1 regulates cytochrome oxidase subunits to optimize efficiency of respiration in hypoxic cells. Cell. 2007;129(1):111–22.

    Article  CAS  PubMed  Google Scholar 

  • Glembotski CC, Arrieta A, Blackwood EA, Stauffer WT. ATF6 as a Nodal Regulator of Proteostasis in the heart. Front Physiol. 2020;11:267.

    Article  PubMed  PubMed Central  Google Scholar 

  • Gong N, Zhao Y, Zhu H, Wang C, Editorial. Novel mechanisms and approaches in kidney/pancreas-kidney transplant-related injury. Front Immunol. 2022;13:1082590.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Görlach A, Klappa P, Kietzmann T. The endoplasmic reticulum: folding, calcium homeostasis, signaling, and redox control. Antioxid Redox Signal. 2006;8(9–10):1391–418.

    Article  PubMed  Google Scholar 

  • Halling JF, Jessen H, Nøhr-Meldgaard J, Buch BT, Christensen NM, Gudiksen A, et al. PGC-1α regulates mitochondrial properties beyond biogenesis with aging and exercise training. Am J Physiol Endocrinol Metabolism. 2019;317(3):E513–25.

    Article  CAS  Google Scholar 

  • He Y, Zhang D, Zeng Y, Ma J, Wang J, Guo H, et al. Bone marrow-derived mesenchymal stem cells protect islet grafts against endoplasmic reticulum stress-Induced apoptosis during the early stage after transplantation. Stem cells (Dayton. Ohio). 2018;36(7):1045–61.

    CAS  Google Scholar 

  • Hulot JS, Masurkar N. miRNA-Based therapeutics for heart failure: why not? J Am Coll Cardiol. 2020;75(15):1801–3.

    Article  PubMed  Google Scholar 

  • Hussain SG, Ramaiah KV. Reduced eIF2alpha phosphorylation and increased proapoptotic proteins in aging. Biochem Biophys Res Commun. 2007;355(2):365–70.

    Article  CAS  PubMed  Google Scholar 

  • Jiang M, Yu S, Yu Z, Sheng H, Li Y, Liu S, et al. XBP1 (x-box-binding Protein-1)-Dependent O-GlcNAcylation is neuroprotective in ischemic stroke in Young Mice and its impairment in aged mice is rescued by Thiamet-G. Stroke. 2017;48(6):1646–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Jiao Q, Takeshima H, Ishikawa Y, Minamisawa S. Sarcalumenin plays a critical role in age-related cardiac dysfunction due to decreases in SERCA2a expression and activity. Cell Calcium. 2012;51(1):31–9.

    Article  CAS  PubMed  Google Scholar 

  • Jin JK, Blackwood EA, Azizi K, Thuerauf DJ, Fahem AG, Hofmann C, et al. ATF6 decreases myocardial Ischemia/Reperfusion damage and links ER stress and oxidative stress signaling pathways in the heart. Circul Res. 2017;120(5):862–75.

    Article  CAS  Google Scholar 

  • Jin Q, Li R, Hu N, Xin T, Zhu P, Hu S, et al. DUSP1 alleviates cardiac ischemia/reperfusion injury by suppressing the Mff-required mitochondrial fission and Bnip3-related mitophagy via the JNK pathways. Redox Biol. 2018;14:576–87.

    Article  CAS  PubMed  Google Scholar 

  • Jones SP, Zachara NE, Ngoh GA, Hill BG, Teshima Y, Bhatnagar A, et al. Cardioprotection by N-acetylglucosamine linkage to cellular proteins. Circulation. 2008;117(9):1172–82.

    Article  CAS  PubMed  Google Scholar 

  • Juhaszova M, Zorov DB, Yaniv Y, Nuss HB, Wang S, Sollott SJ. Role of glycogen synthase kinase-3beta in cardioprotection. Circul Res. 2009;104(11):1240–52.

    Article  CAS  Google Scholar 

  • Kirshenbaum LA, Dhingra R, Bravo-Sagua R, Lavandero S. DIAPH1-MFN2 interaction decreases the endoplasmic reticulum-mitochondrial distance and promotes cardiac injury following myocardial ischemia. Nat Commun. 2024;15(1):1469.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Kouroku Y, Fujita E, Tanida I, Ueno T, Isoai A, Kumagai H, et al. ER stress (PERK/eIF2alpha phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ. 2007;14(2):230–9.

    Article  CAS  PubMed  Google Scholar 

  • Kropotov A, Usmanova N, Serikov V, Zhivotovsky B, Tomilin N. Mitochondrial targeting of human peroxiredoxin V protein and regulation of PRDX5 gene expression by nuclear transcription factors controlling biogenesis of mitochondria. FEBS J. 2007;274(22):5804–14.

    Article  CAS  PubMed  Google Scholar 

  • Kuang BC, Wang ZH, Hou SH, Zhang J, Wang MQ, Zhang JS, et al. Methyl eugenol protects the kidney from oxidative damage in mice by blocking the Nrf2 nuclear export signal through activation of the AMPK/GSK3β axis. Acta Pharmacol Sin. 2023;44(2):367–80.

    Article  CAS  PubMed  Google Scholar 

  • Kuang B, Wang M, Yan H, Jiang Q, Wang Z, Ni H et al. Genetic and pharmacological targeting of XBP1 alleviates hepatic ischemia reperfusion injury by enhancing FoxO1-dependent mitophagy. Translational Research: J Lab Clin Med. 2024.

  • Kulek AR, Anzell A, Wider JM, Sanderson TH, Przyklenk K. Mitochondrial Quality Control: role in Cardiac models of Lethal Ischemia-Reperfusion Injury. Cells. 2020;9(1).

  • Latorre-Muro P, O’Malley KE, Bennett CF, Perry EA, Balsa E, Tavares CDJ, et al. A cold-stress-inducible PERK/OGT axis controls TOM70-assisted mitochondrial protein import and cristae formation. Cell Metabol. 2021;33(3):598–e6147.

    Article  CAS  Google Scholar 

  • Leiser SF, Kaeberlein M. The hypoxia-inducible factor HIF-1 functions as both a positive and negative modulator of aging. Biol Chem. 2010;391(10):1131–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Leiser SF, Begun A, Kaeberlein M. HIF-1 modulates longevity and healthspan in a temperature-dependent manner. Aging Cell. 2011;10(2):318–26.

    Article  CAS  PubMed  Google Scholar 

  • Leone TC, Lehman JJ, Finck BN, Schaeffer PJ, Wende AR, Boudina S, et al. PGC-1alpha deficiency causes multi-system energy metabolic derangements: muscle dysfunction, abnormal weight control and hepatic steatosis. PLoS Biol. 2005;3(4):e101.

    Article  PubMed  PubMed Central  Google Scholar 

  • Li G, Chen Y, Saari JT, Kang YJ. Catalase-overexpressing transgenic mouse heart is resistant to ischemia-reperfusion injury. Am J Physiol. 1997;273(3 Pt 2):H1090–5.

    CAS  PubMed  Google Scholar 

  • Li Z, Hu O, Xu S, Lin C, Yu W, Ma D, et al. The SIRT3-ATAD3A axis regulates MAM dynamics and mitochondrial calcium homeostasis in cardiac hypertrophy. Int J Biol Sci. 2024;20(3):831–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Liu M, Dudley SC. Jr. Targeting the unfolded protein response in heart diseases. Expert Opin Ther Targets. 2014;18(7):719–23.

    Article  PubMed  PubMed Central  Google Scholar 

  • Mao C, Wang M, Luo B, Wey S, Dong D, Wesselschmidt R, et al. Targeted mutation of the mouse Grp94 gene disrupts development and perturbs endoplasmic reticulum stress signaling. PLoS ONE. 2010;5(5):e10852.

    Article  PubMed  PubMed Central  Google Scholar 

  • Marsh KG, Arrieta A, Thuerauf DJ, Blackwood EA, MacDonnell L, Glembotski CC. The peroxisomal enzyme, FAR1, is induced during ER stress in an ATF6-dependent manner in cardiac myocytes. Am J Physiol Heart Circ Physiol. 2021;320(5):H1813–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Matsui Y, Kyoi S, Takagi H, Hsu CP, Hariharan N, Ago T, et al. Molecular mechanisms and physiological significance of autophagy during myocardial ischemia and reperfusion. Autophagy. 2008;4(4):409–15.

    Article  CAS  PubMed  Google Scholar 

  • Miki T, Miura T, Hotta H, Tanno M, Yano T, Sato T, et al. Endoplasmic reticulum stress in diabetic hearts abolishes erythropoietin-induced myocardial protection by impairment of phospho-glycogen synthase kinase-3beta-mediated suppression of mitochondrial permeability transition. Diabetes. 2009;58(12):2863–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Ngoh GA, Watson LJ, Facundo HT, Dillmann W, Jones SP. Non-canonical glycosyltransferase modulates post-hypoxic cardiac myocyte death and mitochondrial permeability transition. J Mol Cell Cardiol. 2008;45(2):313–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Ngoh GA, Facundo HT, Hamid T, Dillmann W, Zachara NE, Jones SP. Unique hexosaminidase reduces metabolic survival signal and sensitizes cardiac myocytes to hypoxia/reoxygenation injury. Circul Res. 2009;104(1):41–9.

    Article  CAS  Google Scholar 

  • Ni M, Zhang Y, Lee AS. Beyond the endoplasmic reticulum: atypical GRP78 in cell viability, signalling and therapeutic targeting. Biochem J. 2011;434(2):181–8.

    Article  CAS  PubMed  Google Scholar 

  • Ni H, Ou Z, Wang Y, Liu Y, Sun K, Zhang J, et al. XBP1 modulates endoplasmic reticulum and mitochondria crosstalk via regulating NLRP3 in renal ischemia/reperfusion injury. Cell Death Discovery. 2023;9(1):69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Nishitoh H, Matsuzawa A, Tobiume K, Saegusa K, Takeda K, Inoue K, et al. ASK1 is essential for endoplasmic reticulum stress-induced neuronal cell death triggered by expanded polyglutamine repeats. Genes Dev. 2002;16(11):1345–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Obas V, Vasan RS. The aging heart. Clinical science (London, England: 1979). 2018;132(13):1367-82.

  • Ong SG, Lee WH, Theodorou L, Kodo K, Lim SY, Shukla DH, et al. HIF-1 reduces ischaemia-reperfusion injury in the heart by targeting the mitochondrial permeability transition pore. Cardiovascular Res. 2014;104(1):24–36.

    Article  CAS  Google Scholar 

  • Packer M. Cardioprotective effects of Sirtuin-1 and its downstream effectors: potential role in mediating the heart failure benefits of SGLT2 (sodium-Glucose cotransporter 2) inhibitors. Circulation Heart Fail. 2020;13(9):e007197.

    Article  CAS  Google Scholar 

  • Paillard M, Tubbs E, Thiebaut PA, Gomez L, Fauconnier J, Da Silva CC, et al. Depressing mitochondria-reticulum interactions protects cardiomyocytes from lethal hypoxia-reoxygenation injury. Circulation. 2013;128(14):1555–65.

    Article  CAS  PubMed  Google Scholar 

  • Paradies G, Paradies V, Ruggiero FM, Petrosillo G. Mitochondrial bioenergetics and cardiolipin alterations in myocardial ischemia-reperfusion injury: implications for pharmacological cardioprotection. Am J Physiol Heart Circ Physiol. 2018;315(5):H1341–52.

    Article  PubMed  Google Scholar 

  • Perrino C, Schiattarella GG, Sannino A, Pironti G, Petretta MP, Cannavo A, et al. Genetic deletion of uncoupling protein 3 exaggerates apoptotic cell death in the ischemic heart leading to heart failure. J Am Heart Association. 2013;2(3):e000086.

    Article  Google Scholar 

  • Petrovski G, Das S, Juhasz B, Kertesz A, Tosaki A, Das DK. Cardioprotection by endoplasmic reticulum stress-induced autophagy. Antioxid Redox Signal. 2011;14(11):2191–200.

    Article  CAS  PubMed  Google Scholar 

  • Ren J, Li Q, Wu S, Li SY, Babcock SA. Cardiac overexpression of antioxidant catalase attenuates aging-induced cardiomyocyte relaxation dysfunction. Mech Ageing Dev. 2007;128(3):276–85.

    Article  CAS  PubMed  Google Scholar 

  • Ridlo MR, Kim GA, Taweechaipaisankul A, Kim EH, Lee BC. Zinc supplementation alleviates endoplasmic reticulum stress during porcine oocyte in vitro maturation by upregulating zinc transporters. J Cell Physiol. 2021;236(4):2869–80.

    Article  CAS  PubMed  Google Scholar 

  • Rosamond W, Flegal K, Friday G, Furie K, Go A, Greenlund K, et al. Heart disease and stroke statistics–2007 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation. 2007;115(5):e69–171.

    Article  PubMed  Google Scholar 

  • Safiri S, Karamzad N, Singh K, Carson-Chahhoud K, Adams C, Nejadghaderi SA, et al. Burden of ischemic heart disease and its attributable risk factors in 204 countries and territories, 1990–2019. Eur J Prev Cardiol. 2022;29(2):420–31.

    Article  PubMed  Google Scholar 

  • Salehpour F, Farajdokht F, Mahmoudi J, Erfani M, Farhoudi M, Karimi P, et al. Photobiomodulation and Coenzyme Q(10) treatments attenuate cognitive impairment Associated with Model of transient global brain ischemia in artificially aged mice. Front Cell Neurosci. 2019;13:74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Santana-Codina N, Muixí L, Foj R, Sanz-Pamplona R, Badia-Villanueva M, Abramowicz A, et al. GRP94 promotes brain metastasis by engaging pro-survival autophagy. Neurooncology. 2020;22(5):652–64.

    CAS  Google Scholar 

  • Schomburg L. Selenium, selenoproteins and the thyroid gland: interactions in health and disease. Nat Reviews Endocrinol. 2011;8(3):160–71.

    Article  Google Scholar 

  • Semenza GL. Hypoxia-inducible factor 1: regulator of mitochondrial metabolism and mediator of ischemic preconditioning. Biochim Biophys Acta. 2011;1813(7):1263–8.

    Article  CAS  PubMed  Google Scholar 

  • Shanmugam K, Ravindran S, Kurian GA, Rajesh M. Fisetin confers Cardioprotection against Myocardial Ischemia Reperfusion Injury by suppressing mitochondrial oxidative stress and mitochondrial dysfunction and inhibiting glycogen synthase kinase 3β activity. Oxid Med Cell Longev. 2018;2018:9173436.

    Article  PubMed  PubMed Central  Google Scholar 

  • Sharma AK, Thanikachalam PV, Bhatia S. The signaling interplay of GSK-3β in myocardial disorders. Drug Discovery Today. 2020;25(4):633–41.

    Article  CAS  PubMed  Google Scholar 

  • Sheng M, Zhang G, Wang J, Yang Q, Zhao H, Cheng X, et al. Remifentanil induces Cardio Protection Against Ischemia/Reperfusion Injury by inhibiting endoplasmic reticulum stress through the maintenance of Zinc Homeostasis. Anesth Analg. 2018;127(1):267–76.

    Article  CAS  PubMed  Google Scholar 

  • Song J, Ni Q, Sun J, Xie J, Liu J, Ning G, et al. Aging impairs adaptive unfolded protein response and drives Beta cell dedifferentiation in humans. J Clin Endocrinol Metab. 2022;107(12):3231–41.

    Article  PubMed  PubMed Central  Google Scholar 

  • Sun J, Yu X, Huangpu H, Yao F. Ginsenoside Rb3 protects cardiomyocytes against hypoxia/reoxygenation injury via activating the antioxidation signaling pathway of PERK/Nrf2/HMOX1. Biomed Pharmacother. 2019;109:254–61.

    Article  CAS  PubMed  Google Scholar 

  • Sun K, Fan C, Zhang J, Ni H, Wang M, Zhang J, et al. Prevention of alloimmune rejection using XBP1-deleted bone marrow-derived dendritic cells in heart transplantation. J Heart lung Transplantation: Official Publication Int Soc Heart Transplantation. 2022;41(12):1660–71.

    Article  Google Scholar 

  • Tadimalla A, Belmont PJ, Thuerauf DJ, Glassy MS, Martindale JJ, Gude N, et al. Mesencephalic astrocyte-derived neurotrophic factor is an ischemia-inducible secreted endoplasmic reticulum stress response protein in the heart. Circul Res. 2008;103(11):1249–58.

    Article  CAS  Google Scholar 

  • Tan Y, Mui D, Toan S, Zhu P, Li R, Zhou H. SERCA overexpression improves mitochondrial Quality Control and attenuates Cardiac Microvascular Ischemia-Reperfusion Injury. Mol Therapy Nucleic Acids. 2020;22:696–707.

    Article  CAS  Google Scholar 

  • Tian X, Zhang S, Zhou L, Seyhan AA, Hernandez Borrero L, Zhang Y, et al. Targeting the Integrated stress response in Cancer Therapy. Front Pharmacol. 2021;12:747837.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Tilokani L, Nagashima S, Paupe V, Prudent J. Mitochondrial dynamics: overview of molecular mechanisms. Essays Biochem. 2018;62(3):341–60.

    Article  PubMed  PubMed Central  Google Scholar 

  • Timmins JM, Ozcan L, Seimon TA, Li G, Malagelada C, Backs J, et al. Calcium/calmodulin-dependent protein kinase II links ER stress with Fas and mitochondrial apoptosis pathways. J Clin Investig. 2009;119(10):2925–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP, et al. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Volume 287. New York, NY): Science; 2000. pp. 664–6. 5453.

    Google Scholar 

  • van den Munckhof I, Riksen N, Seeger JP, Schreuder TH, Borm GF, Eijsvogels TM, et al. Aging attenuates the protective effect of ischemic preconditioning against endothelial ischemia-reperfusion injury in humans. Am J Physiol Heart Circ Physiol. 2013;304(12):H1727–32.

    Article  PubMed  Google Scholar 

  • Vila-Petroff M, Salas MA, Said M, Valverde CA, Sapia L, Portiansky E, et al. CaMKII inhibition protects against necrosis and apoptosis in irreversible ischemia-reperfusion injury. Cardiovascular Res. 2007;73(4):689–98.

    Article  CAS  Google Scholar 

  • Vitadello M, Penzo D, Petronilli V, Michieli G, Gomirato S, Menabò R, et al. Overexpression of the stress protein Grp94 reduces cardiomyocyte necrosis due to calcium overload and simulated ischemia. FASEB Journal: Official Publication Federation Am Soc Experimental Biology. 2003;17(8):923–5.

    Article  CAS  Google Scholar 

  • Vyas S, Zaganjor E, Haigis MC. Mitochondria Cancer Cell. 2016;166(3):555–66.

    CAS  Google Scholar 

  • Wang J, Zhou H. Mitochondrial quality control mechanisms as molecular targets in cardiac ischemia-reperfusion injury. Acta Pharm Sinica B. 2020;10(10):1866–79.

    Article  Google Scholar 

  • Wang ZV, Deng Y, Gao N, Pedrozo Z, Li DL, Morales CR, et al. Spliced X-box binding protein 1 couples the unfolded protein response to hexosamine biosynthetic pathway. Cell. 2014;156(6):1179–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Wang G, Huang H, Zheng H, He Y, Zhang Y, Xu Z, et al. Zn(2+) and mPTP mediate endoplasmic reticulum stress inhibition-Induced Cardioprotection Against Myocardial Ischemia/Reperfusion Injury. Biol Trace Elem Res. 2016;174(1):189–97.

    Article  CAS  PubMed  Google Scholar 

  • Wang S, Hu B, Ding Z, Dang Y, Wu J, Li D, et al. ATF6 safeguards organelle homeostasis and cellular aging in human mesenchymal stem cells. Cell Discovery. 2018;4:2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Wang J, Toan S, Zhou H. New insights into the role of mitochondria in cardiac microvascular ischemia/reperfusion injury. Angiogenesis. 2020a;23(3):299–314.

    Article  PubMed  Google Scholar 

  • Wang J, Lu L, Chen S, Xie J, Lu S, Zhou Y, et al. Up-regulation of PERK/Nrf2/HO-1 axis protects myocardial tissues of mice from damage triggered by ischemia-reperfusion through ameliorating endoplasmic reticulum stress. Cardiovasc Diagnosis Therapy. 2020b;10(3):500–11.

    Article  Google Scholar 

  • Wang J, Lu L, Chen S, Xie J, Lu S, Zhou Y, et al. PERK overexpression-mediated Nrf2/HO-1 pathway alleviates Hypoxia/Reoxygenation-Induced Injury in neonatal murine cardiomyocytes via improving endoplasmic reticulum stress. Biomed Res Int. 2020c;2020:6458060.

    PubMed  PubMed Central  Google Scholar 

  • Wang M, Zhang J, Zhang J, Sun K, Li Q, Kuang B, et al. Methyl eugenol attenuates liver ischemia reperfusion injury via activating PI3K/Akt signaling. Int Immunopharmacol. 2021;99:108023.

    Article  CAS  PubMed  Google Scholar 

  • Wu T, Zhao F, Gao B, Tan C, Yagishita N, Nakajima T, et al. Hrd1 suppresses Nrf2-mediated cellular protection during liver cirrhosis. Genes Dev. 2014;28(7):708–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Xie Y, He Y, Cai Z, Cai J, Xi M, Zhang Y, et al. Tauroursodeoxycholic acid inhibits endoplasmic reticulum stress, blocks mitochondrial permeability transition pore opening, and suppresses reperfusion injury through GSK-3ß in cardiac H9c2 cells. Am J Translational Res. 2016;8(11):4586–97.

    CAS  Google Scholar 

  • Yamamoto M, Kensler TW, Motohashi H. The KEAP1-NRF2 system: a thiol-based sensor-effector Apparatus for maintaining Redox Homeostasis. Physiol Rev. 2018;98(3):1169–203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Yan B, Liu S, Li X, Zhong Y, Tong F, Yang S. Preconditioning with endoplasmic reticulum stress alleviated heart ischemia/reperfusion injury via modulating IRE1/ATF6/RACK1/PERK and PGC-1α in diabetes mellitus. Biomed Pharmacother. 2019;118:109407.

    Article  CAS  PubMed  Google Scholar 

  • Yang L, Xie P, Wu J, Yu J, Yu T, Wang H, et al. Sevoflurane postconditioning improves myocardial mitochondrial respiratory function and reduces myocardial ischemia-reperfusion injury by up-regulating HIF-1. Am J Translational Res. 2016;8(10):4415–24.

    CAS  Google Scholar 

  • Yang L, Xie P, Wu J, Yu J, Li X, Ma H, et al. Deferoxamine Treatment Combined with Sevoflurane Postconditioning attenuates myocardial ischemia-reperfusion Injury by restoring HIF-1/BNIP3-Mediated mitochondrial autophagy in GK rats. Front Pharmacol. 2020;11:6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Yuan M, Gong M, He J, Xie B, Zhang Z, Meng L, et al. IP3R1/GRP75/VDAC1 complex mediates endoplasmic reticulum stress-mitochondrial oxidative stress in diabetic atrial remodeling. Redox Biol. 2022;52:102289.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhang M, Liu X, Wang Q, Ru Y, Xiong X, Wu K, et al. NDRG2 acts as a PERK co-factor to facilitate PERK branch and ERS-induced cell death. FEBS Lett. 2017;591(21):3670–81.

    Article  CAS  PubMed  Google Scholar 

  • Zhang J, Ren D, Fedorova J, He Z, Li J. SIRT1/SIRT3 modulates Redox Homeostasis during Ischemia/Reperfusion in the Aging Heart. Antioxid (Basel Switzerland). 2020a;9(9).

  • Zhang Y, Zhang JJ, Liu XH, Wang L. CBX7 suppression prevents ischemia-reperfusion injury-induced endoplasmic reticulum stress through the Nrf-2/HO-1 pathway. Am J Physiol Ren Physiol. 2020b;318(6):F1531–8.

    Article  CAS  Google Scholar 

  • Zhang J, Zhang J, Ni H, Wang Y, Katwal G, Zhao Y, et al. Downregulation of XBP1 protects kidney against ischemia-reperfusion injury via suppressing HRD1-mediated NRF2 ubiquitylation. Cell Death Discovery. 2021a;7(1):44.

    Article  PubMed  PubMed Central  Google Scholar 

  • Zhang G, Wang X, Li C, Li Q, An YA, Luo X, et al. Integrated stress response couples mitochondrial protein translation with oxidative stress control. Circulation. 2021b;144(18):1500–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhang J, Zhao Y, Gong N. XBP1 modulates the aging Cardiorenal System by regulating oxidative stress. Antioxid (Basel Switzerland). 2023;12(11).

  • Zhang C, Liu B, Sheng J, Wang J, Zhu W, Xie C, et al. Potential targets for the treatment of MI: GRP75-mediated ca(2+) transfer in MAM. Eur J Pharmacol. 2024;971:176530.

    Article  CAS  PubMed  Google Scholar 

  • Zhao Y, Du D, Chen S, Chen Z, Zhao J. New insights into the functions of MicroRNAs in Cardiac Fibrosis: from mechanisms to therapeutic strategies. Genes. 2022a;13:8.

    Article  Google Scholar 

  • Zhao H, Chen X, Hu G, Li C, Guo L, Zhang L, et al. Small extracellular vesicles from Brown Adipose tissue mediate Exercise Cardioprotection. Circul Res. 2022b;130(10):1490–506.

    Article  CAS  Google Scholar 

  • Zhong H, Song R, Pang Q, Liu Y, Zhuang J, Chen Y, et al. Propofol inhibits parthanatos via ROS-ER-calcium-mitochondria signal pathway in vivo and vitro. Cell Death Dis. 2018;9(10):932.

    Article  PubMed  PubMed Central  Google Scholar 

  • Zhu H, Zhou H. Novel insight into the role of endoplasmic reticulum stress in the Pathogenesis of Myocardial Ischemia-Reperfusion Injury. Oxidative Med Cell Longev. 2021;2021:5529810.

    Article  Google Scholar 

  • Zhu J, Rebecchi MJ, Tan M, Glass PS, Brink PR, Liu L. Age-associated differences in activation of Akt/GSK-3beta signaling pathways and inhibition of mitochondrial permeability transition pore opening in the rat heart. Journals Gerontol Ser Biol Sci Med Sci. 2010;65(6):611–9.

    Article  Google Scholar 

  • Zhu P, Hu S, Jin Q, Li D, Tian F, Toan S, et al. Ripk3 promotes ER stress-induced necroptosis in cardiac IR injury: a mechanism involving calcium overload/XO/ROS/mPTP pathway. Redox Biol. 2018;16:157–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhu H, Tan Y, Du W, Li Y, Toan S, Mui D, et al. Phosphoglycerate mutase 5 exacerbates cardiac ischemia-reperfusion injury through disrupting mitochondrial quality control. Redox Biol. 2021;38:101777.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to express their gratitude to EditSprings (https://www.editsprings.cn) for the expert linguistic services provided.

Funding

This work was supported by grants to NQ Gong from the National Natural Science Foundation of China (No. 82370759 and 82170772), to NQ Gong from the Major State Basic Research Development Program of China (NO.2013CB530803, 973), to XP Chen from the Special Project of the Ministry of Health (201302009), and to NQ Gong from the Clinical Research Physician Program of Tongji Medical College, HUST.

Author information

Authors and Affiliations

Authors

Contributions

JZ, YZ and NG conceived and drafted the paper. JZ and NG prepared the tables and figures. JZ, YZ and NG were involved in the compilation of the references. YZ and NG instructed and revised the manuscript. All authors contributed to the review and approved the submitted version.

Corresponding author

Correspondence to Nianqiao Gong.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All authors give consent for publication.

Competing interests

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, J., Zhao, Y. & Gong, N. Endoplasmic reticulum stress signaling modulates ischemia/reperfusion injury in the aged heart by regulating mitochondrial maintenance. Mol Med 30, 107 (2024). https://doi.org/10.1186/s10020-024-00869-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s10020-024-00869-w

Keywords