Abstract
Myocardial ischemia has the highest disease burden among all cardiovascular diseases making it a significant challenge to the global public health. It can result in myocardial cell damage and death due to impaired mitochondrial and endoplasmic reticulum (ER) functions. These two organelles are important regulators of cell death. In recent years, research has shifted from isolated studies of individual organelles to a more integrative approach, with a particular focus on their membrane contact sites—Mitochondria-Associated Endoplasmic Reticulum Membranes (MAMs). These dynamic microdomains play a crucial role in regulating material exchange and signal transduction between the endoplasmic reticulum and mitochondria. This review comprehensively describes the intricate structure of MAMs and their multifaceted roles in cellular pathophysiological processes. Particular focus was directed at the far-reaching effects of MAMs in regulating key pathological events including calcium homeostasis, mitochondrial dysfunction, ER stress, oxidative stress, and autophagy in ischemic heart disease (IHD). The potential treatment targets and regulatory mechanisms of MAMs were discussed and summarized, providing novel research directions and treatment approaches for improving myocardial ischemia-related diseases.
Graphical Abstract
Keywords: Mitochondria-associated endoplasmic reticulum membranes, Myocardial ischemia, Calcium homeostasis, Cellular stress
Introduction
Ischemic Heart Disease (IHD) is a clinical syndrome caused by reduced coronary blood perfusion, resulting in an imbalance between myocardial oxygen supply and demand, dysregulated metabolism in cardiomyocytes, as well as impaired cardiac function. Myocardial ischemia causes irreversible damage to cardiomyocytes and may potentially lead to cell death. Several forms of regulated cell death influence the production of reactive oxygen species (ROS), calcium stress, and inflammatory cascade responses, thereby causing cardiomyocyte loss and exacerbating myocardial ischemia/reperfusion (I/R) injury. These processes trigger adverse cardiac remodeling and functional impairment [1]. IHD has the highest disease burden among all cardiovascular diseases, including acute myocardial infarction, chronic stable angina, chronic IHD, and heart failure related to these conditions [2]. The World Health Organization places IHD as the leading cause of death, out of the total 55.4 million deaths recorded in the past two decade. In this context, it is important to comprehensively elucidate specific mechanisms of myocardial ischemia to explore potential novel treatment strategies.
Mitochondria is a primary site for cellular energy production and play a pivotal role in adjustment to environmental stress as well as regulating metabolism, inflammation, calcium homeostasis, and cell death processes [3, 4]. Their functional integrity is vital for the survival of high-energy-demanding cardiomyocytes under ischemia and hypoxia. As the largest multifunctional organelle within the cell, the endoplasmic reticulum (ER) regulates various physiological processes, such as protein synthesis, folding, and transport, lipid metabolism, and calcium signaling [5, 6]. Impaired ER function can induce ER stress, causing the Unfolded protein response (UPR) and cell apoptosis. This process is closely accompanied with pathological changes including ischemia, hypoxia, oxidative stress, and the depletion of Ca2⁺ stores in the ER [7]. As research advances, the mitochondria and ER are no longer considered isolated structures with independent roles, but as interdependent components within a dynamic, integrated functional region called Mitochondria-associated Endoplasmic Reticulum Membranes (MAMs).
MAMs participate in diverse cellular pathophysiological processes, including Ca2⁺ transport, lipid metabolism, mitochondrial dynamics, cellular stress responses (including ER stress and oxidative stress), and regulation of cell death pathways (including autophagy and apoptosis) [8, 9]. MAMs have gained increasing research attention because of their important and multifaceted roles. Studies have shown that the MAM microdomain significantly affects key pathophysiological mechanisms during myocardial ischemia and reperfusion, including calcium homeostasis, mitochondrial dysfunction, ER stress, oxidative stress, as well as protein–protein crosstalk control [9–11]. Alterations in these processes are recognized as common triggers for various forms of cell death, including apoptosis, autophagy, necrosis, pyroptosis, and ferroptosis [12]. These findings suggest that MAMs can regulate survival and death of cardiomyocytes under pathological conditions including I/R.
Multiple molecules regulate the structural integrity and function of MAMs. Among them is the Phosphofurin Acid Cluster Sorting Protein 2 (PACS-2), which was found to induce severe mitochondrial fragmentation, resulting in mitochondrial fission and separation from the ER, and significantly disrupting the structural integrity of MAMs [13, 14]. Another protein, FUN14 domain-containing protein 1 (FUNDC1) was reported to promote the formation of MAMs by tethering MAMs-specific proteins [15]. Correspondingly, regulatory molecules of MAMs function, including Glycogen Synthase Kinase-3β (GSK3β) and Sigma-1 Receptor (Sig-1R) also play critical roles in myocardial I/R. They can relieve myocardial injury through mechanisms including reducing MAMs-mediated calcium overload and ER stress, thereby exerting cardioprotective effects [16, 17]. With comprehensive research on the role of MAMs in IHD, additional regulatory proteins have been identified, among them CLIC4, Nox4, and ATAD3A, etc. [18–20].
In this review, we systematically describe the complex structure and multifunctional roles of MAMs, focusing on their significant implications in myocardial ischemia. The key regulatory molecules and mechanisms involved in MAMs are discussed, with the aim of identifying potential treatments strategies for myocardial ischemia.
Structure and function of MAMs
The intracellular membrane is the hallmark characteristic of eukaryotic cells, with several organelles or membrane systems interacting through closely connected Multiple Membrane Contact Sites (MCS) to promote material transfer and signal exchange [21, 22]. In 1959, highly ordered tubular structures of the ER were first observed around mitochondria in rat liver cells using transmission electron microscopy although being initially thought to be artifacts of in vitro fixation [23]. With the continuous development of transmission electron microscopy, subcellular fractionation, and mass spectrometry, numerous studies have confirmed the physical relationship between the ER and mitochondria by observing co-sedimentation of ER vesicles with mitochondria and the close link between them and ER vesicles [24, 25]. This physical communication site between the ER and mitochondria is called MAMs; it is a dynamic microdomain maintained by specific tether proteins and spacer proteins [21]. This discovery laid a reference for further research on organelle interactions and their roles in pathophysiological processes.
Morphological structure of MAMs
Mitochondria and the ER, as highly dynamic organelles, the structure of MAMs at their contact sites is not constant. Morphological studies have shown that the contact range between the mitochondria and the ER is approximately 10 to 60 nm, with the two organelles being close but not overlapping. MAMs are considered to be effectively present when the distance between them is less than 30 nm [26]. Chromatographic analysis of isolated mitochondria has also revealed that the distance between the outer mitochondrial membrane (OMM) rough ER (RER) and smooth ER (SER) is different due to the effect of the ribosome [27]. MAMs are also morphologically diverse. Live-cell microscopy and electron tomography investigations have revealed that the tubular structure of ER surrounds the mitochondria in most cells, covering 2% to 5% of the mitochondrial surface area, significantly influencing mitochondrial dynamics [21]. In addition, the ER may partially surround the mitochondria (about 50% of the mitochondrial circumference) or fully envelop the mitochondria.
Proteomic analysis of MAMs has unveiled thousands of proteins with different functions [28, 29]. These proteins modulate various cellular physiological processes (Table 1). Based on their localization within MAMs, these proteins can be divided into three categories: (1) MAMs-resident proteins, which are exclusively localized to MAMs; (2) MAMs-enriched proteins, localized in MAMs but also in other cellular compartments; (3) MAMs-associated proteins, which transiently reside in MAMs during biochemical reactions [29]. The stable and dynamic communication between the ER subregion and the OMM is maintained through a series of protein chains and chains. The link between the ER and mitochondria in yeasts is mediated by the ER-mitochondria encounter structure (ERMES). This complex comprises the ER-resident membrane protein Mmm1, soluble protein Mdm12, and two outer mitochondrial membrane proteins, i.e., Mdm10 and Mdm34 [30]. The associations of ER-mitochondria and their structural organization are more intricate in mammals. Multiple protein complexes play critical roles in mediating communication between two organelles, each regulating different physiological functions (Fig. 1). These protein complexes include the inositol 1,4,5-trisphosphate receptors(IP3Rs)- glucose-regulated protein 75 (Grp75)- voltage-dependent anion channel (VDAC) complex, vesicle-associated membrane protein-associated protein B(VAPB)-protein tyrosine phosphatase interacting protein 51 (PTPIP51) complex, homodimers mitofusin-2 (MFN2)-MFN2 or heterodimers MFN2-MFN1 and B cell receptor-associated protein 31 (Bap31) -mitochondrial fission 1 protein (Fis1) complex, etc. [31, 32]. Based on the complexity of the biological functions mediated by MAMs, we further discussed the composition and role of protein complexes in MAMs in their related different biological functions.
Table 1.
Major related proteins and functions of MAMs
| Role | Protein | Functions and mechanisms | References |
|---|---|---|---|
| MAMs tethers | MFN1/2 | Regulating MAMs formation by functioning as ER-mitochondrial ligation complex | [50] |
| VAPB-PTPIP51 | Modulating MAMs formation by acting as ER-mitochondrial ligation complex | [88] | |
| ERMES | Composed of Mmm1, Mdm10, Mdm12, and Mdm34, mediates Ca2+ transport and phospholipid homeostasis | [30] | |
| Ca2+ transport | IP3R | Regulates the Ca2+ release from ER | [37] |
| VDAC1 | Facilitates mitochondrial uptake of Ca2+ | [38] | |
| Grp75 | Links IP3R and VDAC1 to form a complex that regulates Ca2+ transport | [38] | |
| SERCA | Function as an ER Ca2+ uptake pump that interacts with calnexin to maintain intracellular calcium homeostasis | [98] | |
| Sig-1R | Dissociates from Bip under stress, to influence Ca2+ transport between ER-mitochondria by stabilizing IP3R | [17] | |
| Lipid metabolism | ORP 5/8 | Constitutes the ORP5/8-PTPIP51 complex that mediates PS and sterol transport | [43] |
| ACAT | Modulates the catalytic transformation of intracellular free cholesterol into cholesterol esters | [46] | |
| Mitochondrial dynamics | MFN1/2 | Form a homodimer/heterodimer complex that connects the two organelles and regulates OMM fusion | [32] |
| OPA1 | Regulates the IMM fusion | [48] | |
| DRP1 | Interacts with receptors MFF, MiD49/51 and Fis1 to drive mitochondria division in a GTPase-dependent manner | [53] | |
| ER stress | PERK | Enriched in MAMs and mediates ER stress and URP response | [64] |
| ATF6 | |||
| IRE1 | |||
| MFN2 | Binds to the PERK and inhibits its activity, MFN2 depletion activates PERK and ER stress via the CHOP pathway | [71] | |
| Oxidative stress | Ero1 | Regulates the ROS production in the ER through the protein oxidative folding mechanism | [73] |
| p66Shc | Phosphorylates the Ser36 residue of p66Shc and translocates to the mitochondria/MAMs portion to induce ROS production | [76] | |
| Autophagy | ATG14 | Acts as an autophagy marker and regulates autophagosome formation | [87] |
| ATG5 | |||
| PINK1/Parkin | Facilitates ER-mitochondrial crosstalk and induces autophagy | [89] | |
| Beclin 1 | Mediates the ER-mitochondrial crosstalk and autophagosome formation | [90] | |
| VAPB-PTPIP51 | Loss of VAPB or PTPIP51 induces autophagy by inhibiting mitochondrial Ca2+ uptake | [88] | |
| Apoptosis | Bap31- Fis1 | The combination of the Bap31and Fis1 promotes procaspase 8 activation in response to apoptosis signal and induces apoptosis | [31] |
| CHOP | Mediates ER stress-dependent apoptosis by promoting ROS production | [70] | |
| DRP1 | Enhances MOMP via stimulating BAX oligomerization on mitochondrial membranes to promote apoptosis | [83] |
Fig. 1.
MAMs play a critical role in regulating various cellular physiological functions, including: Calcium homeostasis, by controlling the transfer of Ca2⁺ between the ER and mitochondria; Lipid biosynthesis and metabolism; Mitochondrial dynamics, including the regulation of fission, fusion, and movement; Cellular stress responses, such as ER stress and oxidative stress; Cell death processes, including autophagy and apoptosis
Functions of MAMs
MAMs in Ca2⁺ transfer
As a key second messenger in signal transduction, mitochondrial Ca2⁺ levels influence the various mitochondrial functions, such as signal transduction, ROS generation, oxidative phosphorylation, and apoptosis. Among these, Ca2⁺ transport between mitochondria and ER-mediated by MAMs is a central mechanism in regulating mitochondrial Ca2⁺ levels. Csordás and colleagues discovered that the intermembrane distance of MAMs and the local Ca2⁺ concentration significantly influence the efficiency of mitochondrial Ca2⁺ absorption. The Ca2⁺ transfer efficiency significantly improves when the intermembrane distance of MAMs increases to 15 nm; conversely, the transfer efficiency markedly reduces when the distance decreases to 5 nm [33, 34]. MAMs also provide a high-concentration Ca2⁺ microenvironment for mitochondria, which is a necessary condition for mitochondrial Ca2⁺ uptake [35]. Therefore, the dynamic regulation of MAMs mediates Ca2⁺ transport.
In MAMs, protein bridge allows direct transportation of Ca2⁺ from the ER to the mitochondria, hence promoting the progression of the Krebs cycle in the mitochondrial matrix and ATP synthesis [36]. The major Ca2⁺ release channels in this process include two receptor families: (1) IP3Rs enriched in the ER, and (2) ryanodine receptors (RyRs) enriched in the sarcoplasmic reticulum (SR), a specialized form of ER in cardiac cells. These receptors are located on the ER/SR membrane and mediate Ca2⁺ release, producing local Ca2⁺ concentrations higher than the cytoplasmic level in the MAMs region [37]. On the OMM, VDACs, with high conductance, act as key channels for mitochondrial Ca2⁺ uptake. Grp75, located in the cytoplasm, connects IP3Rs and VDACs via its cytosolic portion, forming the IP3Rs-Grp75-VDAC complex to promote Ca2⁺ transmembrane transport [38]. Furthermore, this complex acts as a multifunctional molecular scaffold, mediating the dynamic regulation of Ca2⁺ transport in MAMs through various regulatory proteins, including Sig-1R, BiP, Bcl-2, and IRBIT [39, 40]. Similarly, VAPB anchored to the ER can be coupled to PTPIP51 on the OMM to regulate Ca2+ transfer by influencing the role of IP3R-VDAC1 to regulate autophagy [41]. Once Ca2⁺ crosses the OMM, it enters the mitochondrial matrix through the MCU on the IMM, a crucial process for maintaining mitochondrial function.
Persistent high levels of Ca2⁺ accumulation can however cause calcium overload, causing apoptosis via the mitochondrial permeability transition pore (mPTP) [42]. This highlights the important role of calcium homeostasis in preserving cellular viability. Recent studies have identified several regulatory factors, including FUNDC1, GSK3β, CLIC4, Nox4, and ATAD3A, that regulate calcium homeostasis mediated by MAMs. The subsequent sections in this work will explore specific roles and regulatory mechanisms of these factors.
MAMs in lipid metabolism
The ER and mitochondria are crucial organelles for lipid biosynthesis, with various intermediate molecules requiring multiple movements between them to complete the entire physiological process of lipid synthesis. However, material transport between the ER and mitochondria does not occur via vesicles despite vesicular transport being the primary mechanism for lipid molecule transfer. Studies have shown that the transport of these lipid molecules between the two organelles primarily depends on the MAM pathway [43]. Various tethering proteins, including oxysterol-binding protein-related protein 5/8 (ORP5/8) and PTPIP51, play key roles in MAMs, participating in the aforementioned transmembrane transport [44]. In addition, several enzymes on the membranes of the ER and mitochondria including phosphatidylserine synthases 1/2 (PSS1/PSS2), phosphatidylserine decarboxylase (PSD), and acyl-coenzyme A, cholesterol acyltransferase/sterol O-acyltransferase (ACAT1/SOAT1) play important catalytic roles in lipid biosynthesis [45].
The “phosphatidylserine (PS) decarboxylation pathway” is a key step in the synthesis of phosphatidylethanolamine (PE). In this process, phosphatidic acid (PA) is converted into PS under the catalysis of PSS1/PSS2, before being transferred to mitochondria via the MAMs pathway. The PS subsequently undergoes decarboxylation to form PE, catalyzed by PSD, via a Ca2⁺-dependent mechanism [45]. Moreover, MAMs has been shown to modulate cholesterol metabolism. ACAT1, which is located on MAMs is a key enzyme that catalyzes the transformation of free cholesterol into cholesterol esters within the cell, playing an important role in maintaining the balance of cholesterol metabolism [46]. Further investigations are still necessary on the mechanisms by which lipid transfer proteins (LTPs) function at membrane contact sites.
Oxysterol-binding protein-related proteins (ORPs), particularly ORP5/8 from the ORP family are localized at MAMs and act as key channels for transporting PS and sterols between the ER and mitochondria [47]. They rely on a functional lipid transfer domain, enabling their interaction with PTPIP51 located on the OMM. Together, they form the ORP5/8-PTPIP51 complex, promoting lipid transport [44]. These MAMs-mediated enzymatic reactions and transmembrane transport mechanisms ensure programmed regulation of lipid biosynthesis, providing important support for the integrity and functional metabolism of the mitochondrial membrane.
MAMs in mitochondrial dynamics
Mitochondria, as highly dynamic organelles, continually reorganize their network through a precise balance of fission and fusion, ensuring the maintenance of genomic integrity and metabolic stability. This complex process is tightly regulated by conserved molecular mechanisms and involves the enrichment of multiple associated proteins in the ER-mitochondria interface. Mitochondrial fusion involves a coordinated merging of the OMM and IMM, with the MFN1/2 complex and optic atrophy 1 (OPA1) acting as important components of this mechanism [48, 49], mediating OMM and IMM fusion, respectively. MFN2 is enriched in the OMM, ER membrane, and MAMs, whereas MFN1 localizes exclusively to the mitochondrial membrane. Together, they form homo- or heterodimeric complexes to connect the two organelles and regulate OMM fusion [32]. Additionally, the efficiency of fusion mediated by MFN1 homodimers, MFN2 homodimers, and MFN1/2 heterodimers varies due to the distinct GTPase activities of MFN1 and MFN2. The MFN1/2 complex also plays a role in the tethering process between the ER and mitochondria [50]. Studies have revealed that the silencing or loss of MFN2 can disrupt the distance between the ER and mitochondria. However, whether the absence of MFN2 loosens the structural connection or increases the contact density between the two organelles remains debatable [51, 52].
MAMs participate in the mitochondrial fission process, specifically in defining the mitochondrial division site [53]. During this process, ER tubules first wrap around the mitochondria, indicating a specific division site, before initiating contraction and fission mechanisms for division. This regulatory pathway involves the sequential action of multiple associated proteins. Mitochondrial contraction begins with the aggregation of actin-myosin assemblies in the MAMs region, a process mediated by inverted formin 2 (INF2) and the mitochondrial actin nucleator, Spire 1c, in the ER [54, 55]. Thereafter, dynamin-related protein 1 (DRP1) is recruited to the OMM, where it interacts with receptors including mitochondrial fission factor (MFF), mitochondrial dynamics 49/51 (MiD49/51), and Fis1. This process mediates mitochondrial constriction and division via a GTPase-dependent mechanism. Furthermore, studies have shown that ER-associated IMM constriction precedes OMM constriction [56]. Active replication of mitochondrial DNA (mtDNA) has also been observed at the mitochondrial constriction and division sites [57]. These results suggest that signals for mitochondrial fission may originate from mtDNA.
Surprisingly, despite undergoing multiple rounds of reorganization, the ER and mitochondria can maintain a consistent overall morphology and remain connected during movement [58]. The bidirectional transport mechanism of these two organelles along microtubules is linked to the motor proteins kinesin-1 and dynein 2 [59, 60]. Nonetheless, research findings on how they maintain their link without being destroyed during movement remain unclear.
MAMs in stress-ER stress and oxidative stress
A critical function of MAMs is to facilitate cellular adaptation to various stress responses, primarily by promoting the coordinated integration of stress signals between the ER and mitochondria, which are interconnected through MAMs. Considering ER stress as an example, the contact between the ER and mitochondria increases, further improving the Ca2⁺ signaling mediated by MAMs, in turn stimulating an increase in mitochondrial oxidative phosphorylation function. However, abnormal regulation of Ca2⁺ signaling ensues if the ER stress response persists for too long, hence causing apoptosis [61]. During oxidative stress, MAMs regulate the production of ROS and the resulting oxidative damage, thus regulating Ca2⁺ signaling and mitochondrial function to finely tune cellular functions [62]. In certain pathological stress conditions, including myocardial ischemia, ER stress, and oxidative stress simultaneously occur and interact with each other.
The ER serves as the primary site for protein folding and secretion in cells. However, when exposed to various pathological stimuli, its capacity for protein processing may be overwhelmed, leading to the accumulation of unfolded or misfolded proteins within the ER and triggering ER stress. This stress response further activates the UPR, relieving the protein load on the ER and restoring cellular homeostasis by expanding the ER membrane and reducing the input of newly synthesized proteins [63]. During this process, three transmembrane proteins in the ER function as stress sensors and play a central role. These proteins include protein kinase RNA-like kinase(PERK), activating transcription factor 6(ATF6) and inosital requiring enzyme 1(IRE1) [64, 65]. Interestingly, these three UPR-related proteins are enriched in MAMs, which are closely involved in the signaling between ER stress and UPR, hence mediating communication between the ER and mitochondria. In these pathological processes, several proteins regulate mitochondrial Ca2⁺ signaling by modulating the activity of Ca2⁺ channels in the MAMs junctions or by influencing ER Ca2⁺ levels, and ultimately cell apoptosis. For instance, IRE1 in MAMs regulates the availability of IP3R, promoting Ca2⁺ transfer to mitochondria and further ATP production [66]. Meanwhile, IRE1α accumulation in MAMs can induce mitochondrial Ca2⁺ overload, resulting in cell death [67]. Additionally, tethering proteins in MAMs, including Grp75 and MFN2regulate the UPR process by targeting Ca2⁺ signaling to participate in the adaptive response to ER stress [68]. In response to ER stress, chaperone proteins within the ER, such as calreticulin, calnexin, and BiP (heat shock protein 70 family protein 5), along with redox-regulatory proteins, can influence oxidative phosphorylation and cell apoptosis by enhancing Ca2⁺ flux within the MAMs region [61, 69]. Another mechanism of the UPR response involves the activation of p38 MAPK and C/EBP homologous protein (CHOP), which mediates ER stress-induced cell apoptosis by promoting ROS production. Verfaillie et al. [70] found that PERK in MAMs participates in the formation of MAMs and regulates the transfer of ROS to mitochondria, thereby regulating intrinsic apoptosis caused by ER stress. Additional investigations revealed that the activity of PERK can be suppressed by interacting with the MAMs tethering protein MFN2 [71]. In addition, MAMs tethering protein VAPB can interact with ATF6, directly suppressing the UPR response [67].
Regulating ROS production and mediating oxidative stress damage are primary pathways by which MAMs regulate cellular function during oxidative stress responses. Booth et al. [62] utilized drug-induced ER-mitochondrial contacts and discovered that hydrogen peroxide nanodomains exist at the MAMs interface, originating from mitochondrial cristae. As part of ER-mitochondria communication, this domain can modulate Ca2⁺ signaling and mitochondrial activity. Surprisingly, oxidative stress can influence the integrity of MAMs and alter Ca2⁺ homeostasis [72]. The production of mitochondrial ROS mainly stems from the activity of the mitochondrial electron transport chain (ETC) during oxidative metabolism and ATP synthesis. MAMs promote ROS production by mediating the excessive transfer of Ca2⁺ to mitochondrial cristae. Meanwhile, ROS can also oxidize the MCU, further promoting the transfer of Ca2⁺ into the mitochondrial matrix [15, 62]. Other structural proteins of MAMs directly participate in the redox cross-talk between the mitochondria and the ER, with significant examples including ER oxidoreductase 1 (Ero1) and the 66 kDa subtype of growth factor adapter Shc (p66Shc). Ero1 promotes the formation of disulfide bonds during protein folding via the oxidation of molecular oxygen, acting as an auxiliary error correction mechanism for protein folding in the ER. However, this oxidative folding process produces numerous ROS, resulting in redox imbalance [73, 74]. Under oxidative conditions, the activation of protein kinase C beta also causes Ser36 phosphorylation on p66Shc [75]. This process promotes the partial translocation of p66Shc to the mitochondria and/or MAMs, where it produces ROS [76]. Interestingly, MAMs regulate cellular function by regulating mitochondrial function during oxidative stress and participate in the genetic mechanisms of damaged mitochondria to regulate cellular functions. Many redox-active proteins, including oxidoreductases TMX1, Ero1α, and Sarco/ER Ca2⁺-ATPase (SERCA) are enriched in MAMs and regulated by ROS [22]. At the MAMs interface, increased ROS activates the mitogen-activated protein kinase (MAPK) pathway and reduces mitochondrial motility, which is crucial for normal mitochondrial inheritance and quality control [77].
MAMs in cell death—apoptosis and autophagy
Studies have extensively reported the link between MAMs and cell death, with various physiological events modulated by MAMs being considered common triggers of cell death mechanisms, including apoptosis, autophagy, necrosis, pyroptosis, and ferroptosis [9, 10, 12]. Wang et al. also showcased the role of MAMs in cell death, specifically in integrating apoptotic signaling [78]. Recent studies have shown that death signals involve bidirectional communication between the ER and mitochondria, a process that comprises various cell death mechanisms, with MAMs playing a critical role in the complex crosstalk between these two organelles.
The high-conductance mPTP, comprising VDAC, ATP synthase oligomycin-sensitivity conferring protein (OSCP), adenine nucleotide translocase (ANT), and cyclophilin D (CypD) drives the process of apoptsis [79]. Studies have shown that MAMs mediates Ca2+ transfer from the ER to mitochondria. Excessive Ca2+ accumulation causes calcium overload, and subsequently, excessive mPTP opening, which promotes cytochrome C release, activates the caspase cascade, ultimately inducing apoptosis [80]. Alterations in MAM tethering are closely associated with cell apoptosis. A key example is the interaction between the mitochondrial protein Fis1 and the ER protein Bap31, which plays a critical role in apoptosis. This interaction facilitates the cleavage of Bap31, transforming it into its pro-apoptotic form. This, in turn, activates procaspase 8, triggering an apoptotic cascade that culminates in cell death [31, 81]. Apoptosis also involves changes in mitochondrial dynamics [82]. During this process, DRP1 mediates mitochondrial fission. However, the role of DRP1 in apoptosis is not limited to this; it may also have functions independent of mitochondrial division. Specifically, DRP1 improves mitochondrial outer membrane permeabilization (MOMP) by promoting BAX oligomerization on the mitochondrial membrane. MOMP is essential for cytochrome c release during apoptosis [83, 84]. In this pathological process, DRP1 and BAX co-localize in MAMs, coordinating MOMP activation.
Autophagy is another important biological role mediated by MAMs. Physiological autophagy is crucial in protein quality control mechanisms and cellular stress response [85]. However, excessive autophagy can over-degrade cellular components, potentially inducing cell death. Although the precise location of autophagosome formation and the sources of proteins and lipids required for its expansion are still controversial, increasing evidence suggests that MAMs may act as important sites for autophagosome biogenesis [86]. In a starvation-induced pathological autophagy model, it was found that the autophagosome marker proteins, ATG14 and ATG5 were found to be recruited to MAMs, where they participates in the formation of autophagosomes [87]. Autophagosome formation is blocked when MAMs are disrupted by knockdown of PACS-2 or MFN2. Gomez-Suaga et al. further reported a correlation between the MAM tether protein VAPB-PTPIP51 complex and autophagy. They discovered that the deletion of VAPB or PTPIP51 caused autophagy by suppressing mitochondrial Ca2⁺ uptake, whereas their overexpression promoted Ca2⁺ flux by tightening the ER-mitochondria connection, hence suppressing autophagosome formation [88]. Moreover, critical autophagy proteins like PINK1/Parkin and Beclin-1 are concentrated at MAMs, where they mediate communication between the ER and mitochondria. These proteins also drive the formation of autophagosomes during mitochondrial autophagy, simultaneously identifying and removing damaged mitochondria through the autophagic process [89, 90]. Research reports have shown that FUNDC1, located at MAMs, mediates mitochondrial fission and mitophagy under hypoxic conditions, and recruits Drp1 to MAMs, further promoting mitochondrial fission during hypoxia [91]. These findings highlight the close link between MAMs and autophagy.
In summary, MAMs mediate the regulation of various cellular processes. In addition to the above-mentioned functions, MAMs modulate other physiological processes, such as inflammation and antiviral responses.
The role of MAMs in myocardial ischemia
The cell biology of the cardiovascular system is tightly regulated, with growing interest being directed towards the role of mitochondria-ER interactions. The importance of these interactions in maintaining cardiovascular health has been emphasized in recent studies. MAMs are broadly present in cardiomyocytes, where they regulate many pathophysiologically relevant signaling processes, including intracellular Ca2⁺ transport, lipid metabolism, mitochondrial dynamics, cellular stress responses, and cell death. Dysfunctions in MAMs are responsible for the onset and progression of cardiovascular diseases including atherosclerosis(AS), myocardial I/R injury, heart failure, and diabetic cardiomyopathy [27]. As a common cardiovascular condition, the incidence of IHD has been on the rise due to the increase in global population aging, hence the first cause of death in human diseases. Currently, there are few interventions for suppressing mortality rates and improving long-term outcomes. Modern medicine can effectively treat ischemic myocardium with timely thrombolysis and interventional procedures; however, these treatments are limited by narrow time windows and the risk of inducing bleeding. Consequently, it is important to explore the potential treatment targets and strategies for IHD.
Under myocardial ischemic conditions, cardiomyocytes shift to anaerobic metabolism, resulting in rapid ATP depletion, ion imbalance (particularly calcium homeostasis disruption), and intracellular acidosis [92]. ATP depletion, calcium dysregulation, and changes in environmental pH disrupt various intracellular signaling pathways, causing a cascade of pathological events including mitochondrial dysfunction, oxidative stress, ER stress, inflammatory responses, autophagy, and apoptosis. Although rapid reoxygenation after restoring myocardial blood flow can alleviate ATP depletion and pH imbalance, it also causes excessive ROS production and calcium overload. These factors ultimately trigger cardiomyocyte apoptosis and necrosis, contributing to myocardial contractile dysfunction [92, 93]. Several studies have highlighted the signifcance of MAMs tether proteins in the regulation of calcium homeostasis, mitochondrial function, cellular stress, and cell death during myocardial ischemia.
Calcium homeostasis
Calcium signaling regulates post-ischemic cardiac function, encompassing both its physiological role in excitation–contraction coupling (ECC) and its pathological impact during calcium overload. During ECC in cardiomyocytes, a transient increase in cytosolic Ca2⁺ concentration causes the formation of cross-bridges, driving myocardial contraction [94]. Simultaneously, mitochondria absorb Ca2⁺ during contraction to stimulate mitochondrial oxidative phosphorylation and the ETC, enabling ATP synthesis, which is critical to meeting the high energy demands of cardiomyocytes [95]. However, in the post-ischemic state, IP3Rs and RyRs on the ER membrane become overly activated, inducing a massive Ca2⁺ release into the cytoplasm [96, 97]. Although SERCA alleviates cytoplasmic Ca2⁺ overload using ATP to pump Ca2⁺ back into the ER [98], its expression and activity are significantly reduced during ischemia. This suppression contributes to the further increase in intracellular Ca2⁺ levels, excessive mitochondrial ROS production, and activation of mitochondrial fission pathways [99]. These changes exacerbate calcium overload in the cytoplasm and mitochondria, creating a vicious cycle. As discussed in Sect. 2.2.1, MAMs play a critical role in Ca2⁺ transport. Being a bridge between the ER and mitochondria, MAMs tether proteins provide the necessary channels for Ca2⁺ transfer and significantly influence the efficiency of this process (Fig. 2).
Fig. 2.
MAMs regulates calcium homeostasis during myocardial ischemia and reperfusion. Myocardial ischemia induces the overactivation of IP3Rs and RyRs on the endoplasmic reticulum membrane, leading to massive Ca2⁺ efflux, while the activity of SERCA, responsible for Ca2⁺ reuptake into the endoplasmic reticulum, is inhibited. These pathological changes exacerbate calcium overload in the cytoplasm and mitochondria, inducing mPTP-dependent apoptosis. In addition, calcium overload and restored energy production during reperfusion trigger excessive myocardial contraction, leading to mechanical strain-induced mutual damage of adjacent cardiomyocytes
Maintaining Ca2⁺ homeostasis is crucial for protecting cardiomyocytes during myocardial ischemia and reperfusion. mPTP-dependent cell death induced by Ca2⁺ overload is one of the primary mechanisms of cardiomyocyte death [42]. Under ischemic conditions, severe disruption of ionic balance and energy supply in cardiomyocytes triggers the opening of voltage-gated Ca2⁺ channels. Subsequently, massive Ca2⁺ transfer from the ER to mitochondria occurs via the IP3R-Grp75-VDAC complex on MAMs, causing mitochondrial calcium overload [38]. Excessive Ca2⁺ accumulation in mitochondria triggers numerous pathological changes, including mitochondrial membrane potential collapse, mitochondrial swelling, and the release of pro-apoptotic factors including cytochrome C. These changes further activate CypD in the mitochondrial matrix, a key regulator of mPTP activity [100, 101]. Studies have shown that CypD also can interact with the IP3R-Grp75-VDAC complex, directly regulating Ca2⁺ transfer from the SR to mitochondria [102]. These disruptions cause excessive mPTP opening, resulting in increased mitochondrial membrane permeability. The subsequent activation of the mitochondrial apoptotic signaling pathway, through caspase-9 and caspase-3 activation, ultimately induces cardiomyocyte apoptosis. Excessive contraction induced by MAMs-mediated calcium overload represents another critical factor leading to cell death during myocardial I/R. In mitochondria, ETC complexes I, III, IV, and V exhibit pronounced Ca2⁺-dependent phosphorylation regulation [93]. Increased intracellular Ca2⁺ levels improve the conductance of these complexes, significantly boosting ATP production. Although the restored energy supply helps alleviate energy deficit during ischemia, excessive ATP generation paradoxically drives abnormally intense cellular contractions. This hypercontractile activity imposes mechanical strain on myocardial tissue, causing structural disruption and necrosis of adjacent cells [103].
However, MAMs-mediated Ca2⁺ transfer is not entirely harmful in pathological processes like tissue I/R. Göbel et al. demonstrated that after acute brain injury and blood–brain barrier disruption, accumulation of MAMs around blood vessels could promote vascular remodeling. Upon MFN2 deletion, impairing MAMs function, mitochondrial Ca2⁺ uptake was significantly reduced, and the ability to remodel blood vessels was weakened accordingly [104]. This suggests that in acute injuries like myocardial ischemia, appropriately improving the role of MAMs might help maintain ATP production under ischemic conditions by promoting Ca2⁺ transfer, supporting cardiovascular remodeling, and ultimately benefiting myocardial tissue. In summary, a moderate increase of mitochondrial Ca2⁺ induced by MAMs microdomains improves cellular energy metabolism. However, under myocardial I/R pathological conditions, uncontrolled mitochondrial Ca2⁺ accumulation induced by MAMs microdomains can exacerbate myocardial injury. Therefore, interventions targeting molecules involved in Ca2⁺ transfer within MAMs microdomains and maintaining mitochondrial Ca2⁺ homeostasis is an important therapeutic intervention for preventing and alleviating myocardial I/R injury.
Mitochondrial dysfunction
After myocardial ischemia, mitochondria, as sensitive oxygen-sensing organelles, experience significant inhibition of their ETC activity and related ATP production in the hypoxic environment, causing the cell metabolism to shift towards anaerobic glycolysis. Hypoxia weakens mitochondrial oxidative phosphorylation and disrupts mitochondrial ion homeostasis. In this condition, cells attempt to compensate by mechanisms including Na⁺ and Ca2⁺ overload mediated by the Na⁺/H⁺ exchanger and Na⁺/Ca2⁺ antiporter to relieve excess H⁺ [105, 106]. Meanwhile, MAMs play an important role in this process by mediating the transfer of Ca2⁺ from the ER to the mitochondria, further causing mitochondrial Ca2⁺ overload. This Ca2⁺ overload induces excessive mitoROS production and exacerbates mitochondrial damage. Especially during the reperfusion phase, the accumulation of mitoROS triggers the opening of the mPTP, hence mediating myocardial cell death [106–108]. Damage-associated molecular patterns (DAMPs) released from mitochondrial damage mediate the activation of NLRP3 inflammasome and related inflammatory cytokines within MAMs, thereby promoting inflammatory injury of cells and generating harmful effects [109]. Notably, excessive ROS production and mitochondrial dysfunction key factors driving endothelial cell injury toward necrosis or apoptosis. This pathological progression further exacerbates myocardial ischemia/I/R injury [110].
Another alteration in mitochondria following myocardial ischemia is the pronounced increase in fission activity. This process is essential for producing additional mitochondria to support the increased energy demands of myocardial cells during both ischemia and reperfusion. However, studies have discovered that excessive mitochondrial fission is also one of the key pathogenic factors in cardiac I/R injury [111]. MAMs play an important role in maintaining the balance between mitochondrial fission and fusion. Korobova et al. [54] found that mitochondrial fission is significantly reduced when communication at the ER-mitochondria contact sites is interfered with. MAMs tether proteins, specifically the MFN1-MFN2 heterodimer, not only play an important role in stabilizing the ER-mitochondria microdomain but are also vital molecules regulating mitochondrial fission [52]. Research has shown that although combined ablation of MFN1 and MFN2 can cause mitochondrial dysfunction, it can also reduce myocardial cell damage related to mitochondria/SR tethering during I/R [112]. The ablation of MFN1-MFN2 reduces MAMs, alleviating I/R-induced Ca2⁺ overload and ROS production at the same time lowering the sensitivity of mitochondrial mPTP opening. This suggests that disrupting mitochondria/SR tethering may protect against ischemia. As a key regulator of mitochondrial fission, DRP1 is recruited in response to hypoxic stress after myocardial ischemia. However, Ong et al. [111] revealed that DRP1 inactivation suppressed mitochondrial fission and reduced the sensitivity of HL-1 cells to mPTP opening, exerting cardioprotective effects. Under ischemic conditions, the level of OPA1, a key protein in mitochondrial fusion, is reduced. However, studies in the cardiac myogenic cell line H9c2 have shown that both reduced and excessive OPA1 expression can trigger cytochrome c release. This indicates that although OPA1 overexpression improves mitochondrial fusion, it does not offer protection against ischemia-induced apoptosis in myocardial cells [113] (Fig. 3).
Fig. 3.
MAMs regulate mitochondrial dysfunction during myocardial ischemia and reperfusion. Under ischemic and hypoxic conditions, the activity of the ETC and associated ATP production are suppressed, triggering a metabolic shift toward anaerobic glycolysis. This metabolic alteration disrupts ion homeostasis, activating various compensatory mechanisms such as the Na⁺/H⁺ exchanger and Na⁺/Ca2⁺ antiporter. Furthermore, mitochondria undergo excessive fission to meet the energy demands of cardiomyocytes. MAMs contributes to the delicate balance between mitochondrial fission and fusion, a process tightly regulated by MAM-associated tether proteins MFN1-MFN2 heterodimer, DRP1, and OPA1
Cellular stress: ER stress
Under pathological conditions including ischemia and reperfusion, stress responses experienced by myocardial tissue cause damage and necrosis, with ER stress and oxidative stress being prominent features. In this process, MAMs, as critical subcellular structures linking mitochondria and the ER, play a vital role in the transduction of stress response signals and the crosstalk in redox regulation (Fig. 4).
Fig. 4.
MAMs mediate the regulation of ER stress and oxidative stress during the pathological process of myocardial ischemia and reperfusion. Following myocardial ischemia, MAM-mediated Ca2⁺ efflux from the ER triggers ER stress and protein misfolding, which activates the UPR to restore cellular homeostasis. However, prolonged stress can shift UPR signaling from a cardioprotective role to a pro-apoptotic response. Furthermore, excessive Ca2⁺ transfer from the ER to mitochondria induces calcium overload, resulting in elevated mitoROS production. Structural proteins within MAMs, such as Ero1 and p66Shc, play a direct role in redox crosstalk between the mitochondria and ER, further exacerbating ROS generation
ER stress is caused by the imbalance between the protein folding capacity of the ER and the demand for protein processing. Its normal protein folding function relies on ER calcium homeostasis and energy metabolism [114]. After myocardial ischemia, the massive efflux of Ca2+ from the ER-mediated by MAMs, is considered a major factor that contributes to ER stress and decreased protein folding capacity [115]. Excess accumulation of unfolded polypeptides or improperly folded proteins within the ER, exceeding the capacity of ER chaperones stimulates the UPR pathway to restore cell homeostasis and prevent tissue damage [63, 64]. The major function of the UPR is to induce protective ER-targeted proteins to cope with ER stress, including ER chaperones (including GRP78, and calreticulin) and protein disulfide isomerases (PDIs) (including ERp72). These proteins are enriched in MAMs and can re-establish ER calcium as well as redox homeostasis, thereby restoring protein folding functions in the ER and protecting myocardial cells from excessive ER stress under ischemic conditions. Studies suggest that this protective effect is achieved partly by binding UPR-activated ATF6 and X-box binding protein-1 (XBP1) to regulatory elements of ER stress response genes [116]. Nonetheless, ER stress-dependent apoptotic pathways are activated when prolonged stress causes the protein load in the ER to exceed the regulatory capacity of the UPR, resulting in cell death. In this process, UPR signaling switches from a protective role to a pro-apoptotic role via the transcriptional induction of CHOP, activation of c-Jun N-terminal kinase (JNK), and caspase-12-dependent pathways, ultimately causing myocardial cell damage during I/R [117–119].
Sig-1R, a ubiquitously expressed MAMs-resident protein in the heart, normally binds to GRP78 (also known as Bip) to form an inactive complex. Under ER stress or related factors stimulation, the uncoupling of the Sig-1R-GRP78 complex occurs, and Sig-1R shifts to the ER, where it mitigates ER stress-induced myocardial damage by suppressing the UPR response. Studies have shown that overexpression of Sig-1R can attenuate the intensity of the ER stress response by activating PERK and ATF6, whereas Sig-1R knockdown interferes with MAMs integrity and causes mitochondrial dysfunction, ultimately causing apoptosis [17]. According to Alam et al., Sigmar1-siRNA knockdown in myocardial cells (NRCs) significantly upregulated CHOP expression and caused myocardial cytotoxicity via sustained activation of ER stress pathways. Conversely, Sigmar1 overexpression enhanced IRE1α phosphorylation, promoted XBP1s expression and nuclear translocation, and ameliorated these effects [120]. These findings suggest that Sig-1R, being an important component of the adaptive ER stress response is a potentially effective treatment target for myocardial ischemic injury.
Cellular stress: oxidative stress
Oxidative stress is closely linked to the pathophysiological processes of myocardial I/R injury. After myocardial ischemia, MAMs-mediated mitochondrial calcium overload induces the production of mitoROS, subsequently causing mitochondrial damage. Especially during reperfusion, reoxygenation of ischemically injured myocardial tissue further causes massive ROS generation [106, 107]. The accumulated ROS can overwhelm cellular antioxidant defenses, resulting in oxidative damage to DNA, lipids, and proteins, thereby exacerbating myocardial I/R injury.
During the pathophysiology of myocardial I/R, mitochondrial calcium overload causes Ca2+ accumulation within mitochondria, resulting in mitochondrial depolarization and disruptions in oxidative phosphorylation. These abnormalities decouple the mitochondrial ETC from respiratory complexes I and III, hence promoting ROS production [15]. More importantly, excessive superoxide radicals further oxidize RyRs, exacerbating mitochondrial calcium overload and ROS production [121]. Additionally, structural proteins within MAMs, including Ero1 and p66Shc, contribute to redox crosstalk between the mitochondria and ER, playing a significant role in ROS production (Sect. “MAMs in stress-ER stress and oxidative stress”). Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Noxs) are also a major source of ROS in myocardial I/R injury; Nox1, Nox2, and Nox4 are the important isoforms involved in this pathological process [122]. Notably, Beretta et al. discovered that Nox4 is enriched in the MAMs regions of cardiomyocytes, where it regulates redox signaling within MAMs under stress conditions. By improving AKT-dependent phosphorylation of IP3R, Nox4 suppresses excessive Ca2+ flux and reduces mPTP-dependent cell necrosis [20]. In the I/R-injured heart, Nox4 limits infarct size via this mechanism, suggesting that Nox4 upregulation during I/R stress is a protective mechanism for cell survival.
Autophagy
Autophagy, a highly conserved intracellular degradation mechanism, has been implicated in the regulation of cardiac homeostasis and function. During myocardial ischemia, autophagy activation is generally considered a protective mechanism that promotes cell survival. It generates substrates needed for ATP regeneration, decreasing the accumulation of misfolded proteins and oxidative stress, and clearing of damaged mitochondria, thereby mitigating ischemic damage and improving the adaptation of the heart to pressure overload (PO) [123, 124]. Under starvation conditions, the autophagosome marker protein ATG14 was found to relocate to the MAMs [87], indicating that ATG14, an indispensable factor in mitophagy activation, may be regulated by MAMs microdomains. However, under certain pathological conditions, such as I/R or the acute phase of PO, excessive autophagy activation may lead to myocardial damage. Evidence shows that autophagy-related protein Beclin 1 can play a detrimental role in I/R injury development. Matsui et al. [125] reported that the Beclin 1 expression was significantly upregulated during cardiac reperfusion through a ROS-dependent mechanism, whereas mice with systemic heterozygous deletion of Beclin 1 showed significant reductions in autophagy and ischemic injury. One plausible mechanism is that autophagy during I/R may induce autosis in cardiomyocytes [126]. This form of cell death is characterized by the excessive formation of autophagosomes, and these autophagosomes are formed at the expense of the cellular membrane rather than relying on lysosomal degradation. This process may cause severe damage to the cellular structure and function, resulting in significant cellular dysfunction and death. As discussed earlier, key autophagy-related proteins such as ATG14, PINK1/Parkin, and Beclin 1 are enriched in MAMs and regulated by ER-mitochondria microdomains [87, 89, 90]. Numerous studies have confirmed that under pathological conditions such as myocardial I/R, these factors mediate pro-survival or pro-apoptotic signals in myocardial injury by modulating mitophagy activation.
Different tether proteins within MAMs are involved in the formation of autophagosomes and clearance of target structures via fine-tuning the contact between the ER and mitochondria. Some MAMs proteins’ specific roles in ischemic injury have gradually been revealed. For example, PTPIP51, a mitochondrial resident protein, was reported to form a complex with VAPB on the ER and is an important component of MAMs. This complex regulates Ca2⁺ transport by modulating ER-mitochondrial contact via the IP3R/VDAC1 pathway [41]. In a mouse model of I/R injury, PTPIP51 was found to be significantly upregulated, and its knockout suppressed myocardial damage and the size of infarct size induced by I/R [127]. Further studies by Sciarretta and colleagues revealed that both downregulation of PTPIP51 or its interacting protein VAPB induced autophagy by lowering mitochondrial Ca2⁺ levels, thus exerting partial protection against I/R injury. However, excessive autophagy during reperfusion may potentially aggravate myocardial damage [128]. In addition, FUNDC1, a mitochondrial autophagy receptor, in located in MAMs and binds to IP3R2 to regulate Ca2⁺ homeostasis, mitochondrial fission, and mitophagy [15]. In a cardiac I/R model, genetic knockout of FUNDC1 compromised the ER-mitochondria microdomain, causing significant decreases in IP3R2 and Ca2⁺ levels in mitochondria. Mice with specific FUNDC1 knockout experienced more severe heart dysfunction. Therefore, autophagy-related proteins located in MAMs can improve ischemic myocardial injury by regulating autophagy, and thus promising treatments for myocardial ischemia.
Potential regulatory molecules targeting MAMs
In summary, this review discusses the critical role of MAMs in myocardial ischemia and reperfusion injury, highlighting their various pathological mechanisms such as calcium homeostasis regulation, mitochondrial dysfunction, ER stress, oxidative stress, and autophagy. As a pivotal hub that coordinates energy metabolism, protein homeostasis, and cell fate regulation, the integrity and functional maintenance of MAMs are important regulators of myocardial protection. Further investigation into the regulatory mechanisms and molecular components associated with MAMs are warranted to not only enhance our understanding of the pathogenesis of myocardial ischemia but also provide valuable insights for uncovering novel therapeutic targets for preventing or mitigating such diseases (Table 2).
Table 2.
The major regulatory molecules of MAMs morphology and function
| Regulatory molecule | Regulation mechanism | References |
|---|---|---|
| PACS-2 |
Regulates the structural and functional integrity of MAMs by affecting mitochondrial fission, thereby participating in Bid-mediated apoptosis; Accumulates in the MAMs of VSMCs and responds to LDL damage, thereby mediating AS; Interferes with MAMs function, playing a key role in calcium homeostasis, formation of ER lipid synthesis centers, and autophagosome formation |
[13, 87, 131, 132] |
| GSK3β |
Interacts with ANT on the IMM, competitively inhibiting the binding of CypD and thereby preventing mPTP opening; Interacts with the CypD-IP3R- Grp75-VDAC complex, thereby modulating IP3R-dependent Ca2+ transport |
[16, 136] |
| FUNDC1 |
Enriched in the MAMs by binding to the ER protein CANX, acting as a central regulatory molecule for mitochondrial fission and autophagy, and mediating the activation of mitochondrial autophagy; Genetic ablation of FUNDC1 disrupts ER-mitochondria coupling by inhibiting ubiquitin-dependent degradation of IP3R2, thereby interfering with mitochondrial Ca2⁺ uptake |
[91, 143, 144] |
| CLIC4 |
Maintaining mitochondrial calcium homeostasis as a regulator of ER/SR-mitochondrial Ca2+ transfer; Inhibits apoptosis and mitochondrial membrane depolarization by suppressing the ROS production in HR injury |
[18] |
| ATAD3A |
Regulates the IP3R1- Grp75-VDAC1 complex to influence the morphology and functional integrity of MAMs Its overexpression can prevent mitochondrial Ca2+ accumulation and ROS generation, thereby reducing mitochondrial oxidative stress and calcium overload |
[19, 153] |
| NOX4 | Regulates the redox signaling within MAMs under stress conditions to increase AKT-dependent phosphorylation of IP3R, thereby inhibiting calcium flux and reducing mPTP-dependent cell necrosis | [20] |
| Sig-1R | Inhibits UPR response by downregulating PERK and ATF6 activation and increasing IRE1α phosphorylation, and ultimately ameliorating ER stress injury | [17, 120] |
PACS-2
The PACS-2, a sorting protein localized on the surface of the ER, is the first MAMs regulatory protein that integrates the ER-mitochondria communication, maintaining ER homeostasis, and regulating apoptotic signaling. Simmen et al. [13] first reported that PACS-2 is involved in Bid-mediated apoptosis by preserving the structural and functional integrity of MAMs through the regulation of mitochondrial fission. Specifically, PACS-2 deficiency can trigger Bap31 cleavage, producing the pro-apoptotic fragment p20, which further triggers early ER Ca2⁺ release into mitochondria and recruits Drp1 to the mitochondrial surface. This process stimulates mitochondrial fission, causes extensive mitochondrial fragmentation and causes the decoupling of the fragmented mitochondria from the ER [13, 129]. This suggests that PACS-2 may suppress Bap31 cleavage through a specific mechanism, thereby effectively preventing excessive mitochondrial fission and its associated dysfunction during myocardial I/R.
AS is the primary pathological process underlying myocardial ischemia. It is marked by the accumulation of LDL in the arterial intima and the phenotypic transformation of vascular smooth muscle cells (VSMCs), both of which play pivotal roles in the progression of AS [130]. The PACS-2 is localized at ER-mitochondria contact sites in human VSMCs (hVSMCs) and responds to LDL-induced damage. Moulis et al. reported that exposure to oxidized LDL (OX-LDL) significantly upregulates PACS-2 expression in the MAMs regions of hVSMCs. Silencing PACS-2 disrupts the structural integrity of MAMs, impairs mitochondrial autophagy, and accelerates apoptosis in VSMCs [131]. Moreover, PACS-2 participates in the maintenance of calcium homeostasis, establishing ER lipid synthesis hubs, and facilitating autophagosome formation by regulating the function of MAMs [13, 87, 132].
Overall, the precise mechanism of PACS-2 action remains to be fully understood. At present, there are no drug agonists or inhibitors specifically targeting PACS-2. While RNAi-based therapies hold potential, they still face significant challenges in terms of safety and efficacy [133]. The phosphorylation status of PACS-2, influences the effectiveness of targeting PACS-2 by acting as a molecular switch. Therefore, when developing chemical modulators of PACS-2 function, an indirect strategy targeting the phosphorylation or degradation of the PACS-2 protein need to be considered.
GSK3β
GSK3 is a serine/threonine kinase with two isoforms, GSK-3α and GSK3β, known to participate in the development of cardiac ischemia and reperfusion. GSK3β is constitutively active in cells, and phosphorylation at the Ser9 site inhibits its activity [16]. It has been reported that the phosphorylated Ser9-GSK3β confers cytoprotection by increasing the threshold for mPTP opening [134, 135], though the precise mechanisms remain unclear. Research has indicated that GSK3β interacts with ANT on the IMM, competitively inhibiting the binding of CypD to prevent mPTP opening [16, 136]. Interestingly, Gomez et al. uncovered an additional regulatory mechanism of GSK3β, in which it partially localizes at the MAMs interface in cardiac cells, where it interacts with the CypD-IP3R- Grp75-VDAC complex, regulating IP3R-dependent Ca2+ transport in cardiomyocytes. During reperfusion, GSK3β inhibition reduces IP3R-mediated Ca2+ leakage at MAMs, limiting mitochondrial calcium overload and subsequent cell death [16]. Furthermore, various strategies to inhibit GSK3β activity have been identified, including the activation of signaling pathways such as AKT, PKC, and protein kinase A [137, 138]. These results suggest that targeting these molecular mechanisms may offer new therapeutic strategies for alleviating cardiac I/R injury. Unexpectedly, Stoica et al. reported a contrasting finding: in neuronal cells, inhibition of GSK3β enhances the VAPB-PTPIP51 interaction and ER-mitochondria association, resulting in elevated mitochondrial Ca2⁺ levels [139]. This may be due to the tissue-specific role of MAMs.
Similarly, Zhai et al. observed that GSK3β has opposing effects in myocardial ischemia and ischemia/I/R injury. Inhibition of GSK3β worsens ischemic injury but helps protect against I/R injury by regulating mTOR and autophagy [140]. In this context, limiting GSK3β inhibition to only reperfusion time is a feasible strategy. GSK3β inhibitors provide a potential therapeutic approach. Jiang et al. [141] reported a list of GSK3β inhibitors currently undergoing clinical studies, which have demonstrated beneficial effects in various disease models, including Alzheimer’s disease, multiple sclerosis, and carotid artery diseases, among others. However, their clinical application in myocardial ischemic diseases has yet to be implemented. This may be attributed to the role of GSK3β in multiple pathophysiological systems, and whether its inhibition may lead to adverse effects on other systems remains to be clarified.
FUNDC1
FUNDC1, a novel hypoxia-induced mitochondrial autophagy receptor, was first reported in 2012. It selectively responds to hypoxic/ischemic stimuli but not to starvation conditions [142]. Wu et al. revealed that under hypoxic conditions, FUNDC1 accumulates at MAMs by interacting with the ER-resident protein calnexin (CANX) and serves as a central regulatory molecule for mitochondrial fission and autophagy during hypoxic responses. In the context of mitochondrial autophagy, the interaction between FUNDC1 and CANX is blocked, exposing the cytosolic loop of FUNDC1, which preferentially recruits DNM1L/DRP1, accelerating mitochondrial fission under hypoxic stress [91, 143]. However, downregulation of the FUNDC1 under hypoxic conditions increases the number of elongated mitochondria, reduces colocalization of autophagosomes with mitochondria, and inhibits mitochondrial fission and autophagy. At the molecular level, genetic ablation of FUNDC1 disrupts ER-mitochondria coupling by inhibiting ubiquitin-dependent degradation of IP3R2, which interferes with mitochondrial Ca2⁺ uptake [144]. Reduced cytosolic Ca2⁺ levels inhibit the binding of cAMP response element-binding protein (CREB) to promoter regions, suppressing Fis1 expression and thus impairing mitochondrial fission [15]. Studies have also demonstrated that FUNDC1 promotes mitochondrial autophagy under ischemic conditions, confering cardioprotection and inhibiting cardiomyocyte apoptosis. Phosphorylation-induced inactivation of FUNDC1 reduces mitochondrial autophagy, which in turn heightens stress responses to unfolded mitochondrial proteins and disrupts mitochondrial homeostasis. This impairment weakens the mitochondrial quality control system, causing the accumulation of damaged mitochondria and triggering the pan-apoptotic program [145–147].
FUNDC1 regulates mitochondrial autophagy by phosphorylating and dephosphorylating three key residues: Ser13, Ser17, and Tyr18 [148]. These sites present promising therapeutic targets for strategies designed to induce protective mitochondrial autophagy. Early research has already shown promising progress in translating FUNDC1 findings into practical applications. For example, cell-permeable functional peptides composed of the HIV-1 Tat protein transduction domain can promote FUNDC1-mediated mitochondrial autophagy in cell experiments. Moreover, intraperitoneal injection of synthesized cell-penetrating peptides has been shown to effectively modulate FUNDC1-mediated mitochondrial autophagy in vivo [149].
CLIC4
Chloride intracellular channel (CLIC) proteins are among the primary intracellular Cl⁻ channels, which function to confer cardioprotective effects. Studies show that blocking CLIC proteins increases myocardial infarction size following I/R injury [150] and abolishes the cardioprotective effects of ischemic preconditioning [151]. Ponnalagu and colleagues further revealed that CLIC4 is expressed in cardiac MAMs, where it regulates Ca2⁺ transfer between the ER/SR and mitochondria, maintaining mitochondrial calcium homeostasis. In cardiomyocytes with CLIC4 gene knockout (clic4 -/-), increased Ca2+ levels can trigger the premature mPTP opening under stress stimuli such as I/R injury. Furthermore, clic4 -/- cardiomyocytes exposed to hypoxia-reoxygenation (HR) injury exhibit significantly increased ROS production, greater mitochondrial membrane depolarization, and higher rates of apoptosis [18]. These findings suggest that the CLIC4 channel in MAMs confers cardioprotection against hypoxia and I/R injury by regulating mitochondrial calcium homeostasis and oxidative stress. This also demonstrates its significant potential as a promising therapeutic target for cardiac protection.
ATAD3A
ATPase Family AAA-domain Containing Protein 3A (ATAD3A) is a mitochondrial membrane-anchored protein that participates in various functions including mitochondrial dynamics, mtDNA expression, and lipid metabolism. Previous studies have shown that ATAD3A prevents mitochondrial disruption and is essential for mantainance of the integrity of the mitochondrial ultrastructure [152]. Furthermore, several studies have indicated that the loss of ATAD3A impairs the interaction between the ER and mitochondria to modulate the formation of MAMs [153]. Building on this, Li et al. found that ATAD3A is localized to the MAMs region and interacts with the IP3R1- Grp75-VDAC1 complex to maintain the morphology and functional integrity of MAMs. They found that ATAD3A overexpression decreased the integrity of the IP3R1- Grp75-VDAC1 complex, inhibiting isoproterenol (ISO)-induced mitochondrial Ca2+ accumulation and ROS generation. This mechanism alleviated mitochondrial oxidative stress and calcium overload, protecting cardiomyocytes from hypertrophic stimuli [19]. Although the specific role of ATAD3A in myocardial I/R models has not been investigated, its role in MAMs microdomains sugggests its potential involvement in myocardial ischemia.
Several studies have revealed various important roles of MAMs regulatory targets in cardiovascular protection. However, its translation into clinical settings need to be further explored. Currently, there is a lack of research on certain regulatory targets of MAMs in relevant translational fields, but some of their mechanisms provide directions for our exploration. For example, since the multiple functions of Nox4 may depend on its subcellular localization, position-specific regulation of Nox4 could be considered in the treatment of I/R injury. Steroids may act as endogenous ligands that bind to Sig-1R, inducing its inhibition or activation, among which dehydroepiandrosterone (DHEA) and its sulfate ester DHEAS have been shown to be strong Sig-1R agonists [154]. Exploring endogenous ligands of Sig-1R is a feasible direction. Moreover, the effects of various chemical molecules or drugs on MAMs and their regulatory targets have been investigated in diverse studies. Notably, sulforaphane (SFN) was shown to regulate Ca2⁺ flux through IP3R and MCU, helping to maintain calcium homeostasis within MAMs [155]. Pravastatin, a drug used for the treatment of hypercholesterolemia, has been recently found to reverse hypercholesterolemia-induced changes in bone marrow-derived macrophages (BMDMs), including affecting alterations in MAMs, ox-LDL uptake, and phagocytic contact points, while enhancing the expression of mitochondrial dynamics-related genes, Mfn2 and Fis1 [156]. Furthermore, fluvoxamine, a selective serotonin reuptake inhibitor (SSRI) with high affinity for Sig-1R, can also act as a Sig-1R agonist to protect against transverse aortic constriction (TAC)-induced cardiac dysfunction [157]. Future studies are needed to identify the potential roles and targets of MAMs to promote the development of MAMs-based therapeutic approaches.
Discussion and perspectives
MAMs are important reglators of the cellular processes in the mitochondria and the endoplasmic reticulum. Due to the changes in MAMs and their significant pathophysiological correlation in ischemic heart disease (IHD), further understanding of the upstream and downstream regulatory mechanisms of MAMs is expected to provide new solutions in this field. However, the complexity and dynamic nature of MAMs present significant challenges for further research. Emerging tools and technologies, such as split-GFP-based contact site sensors (SPLICS), offer promising solutions for real-time in vitro monitoring of MAMs, providing valuable support for advancing studies in this field [158, 159]. Similarly, the development of Contact-ID and OrthoID methods is expected to promote proteomic analysis of MAMs in living cells [160, 161]. Furthermore, several functional detection tools for MAMs need to be developed. Notably, Cho et al. developed the MAM-Calflux, a MAM-specific BRET-based Ca2⁺ indicator, which facilitates ratiometric measurement of dynamic intercellular calcium signaling [162]. Further development of these tools to investigate MAMs alterations in pathophysiology plays a crucial role in gaining new insights into the mechanisms of MAMs in related diseases.
Overall, in the pathological responses associated with myocardial ischemia and subsequent reperfusion, the MAMs microdomain serves as a critical signaling platform that coordinating the regulation of calcium homeostasis, mitochondrial dysfunction, oxidative stress, ER stress, autophagy, and apoptosis. These signaling molecules rely on the MAMs microdomain to transmit signals, thereby collectively mediating myocardial injury and protection. Building on this foundation, therapies that modulate the homeostasis of MAM microdomains are emerging as crucial targets for the clinical treatment of myocardial ischemia. In this review, we have systematically examined key regulatory molecules of MAMs, including PACS-2, GSK3β, and FUNDC1, etc., exploring their mechanisms of action and highlighting recent advancements in translational medicine research related to these targets. However, the development of MAMs-targeted therapies for myocardial ischemia-related conditions still faces several significant challenges. First, some MAMs-related regulatory mechanisms are still being debated, such as the effect of MFN2 deficiency on the structure between mitochondria and the endoplasmic reticulum, the bidirectional regulatory mechanism of GSK3β on mitochondrial Ca2⁺ levels, and the tissue-specific roles of MAMs. Second, the communication between organelles is complex and dual-faceted. MAMs-mediated ER Ca2⁺ transfer to mitochondria enhances ATP production, while excessive mitochondrial calcium accumulation can trigger mPTP-dependent cell death. Similarly, autophagy activation during myocardial ischemia acts as a protective survival mechanism; however, excessive autophagy can exacerbate myocardial damage. Consequently, it is essential to investigate how to precisely regulate its formation and function. Such understanding will help navigate the complexity of this process and inform the development of targeted therapeutic strategies.
Acknowledgements
Not applicable.
Abbreviations
- IHD
Ischemic heart disease
- ROS
Reactive oxygen species
- I/R
Ischemia/reperfusion
- ER
Endoplasmic reticulum
- UPR
Unfolded protein response
- MAMs
Mitochondria-associated endoplasmic reticulum membranes
- PACS-2
Phosphofurin acid cluster sorting protein 2
- FUNDC1
FUN14 domain-containing protein 1
- GSK3β
Glycogen synthase kinase-3β
- Sig-1R
Sigma-1 Receptor
- MCS
Membrane contact sites
- OMM
Outer mitochondrial membrane
- RER
Rough ER
- SER
Smooth ER
- ERMES
ER-mitochondria encounter structure
- IP3Rs
Inositol 1,4,5-trisphosphate receptors
- Grp75
Glucose-regulated protein 75
- VDAC
Voltage-dependent anion channel
- VAPB
Vesicle-associated membrane protein-associated protein B
- PTPIP51
Protein tyrosine phosphatase interacting protein 51
- MFN2
Mitofusin-2
- Bap31
B cell receptor-associated protein 31
- Fis1
Fission 1 protein
- RyRs
Ryanodine receptors
- SR
Sarcoplasmic reticulum
- MCU
Mitochondrial calcium uniporter
- IMM
Inner mitochondrial membrane
- mPTP
Mitochondrial permeability transition pore
- ORP5/8
Oxysterol-binding protein-related protein 5/8
- PSS1/PSS2
Phosphatidylserine synthases 1/2
- PSD
Phosphatidylserine decarboxylase
- ACAT1/SOAT1
Acyl-coenzyme A, cholesterol acyltransferase/sterol O-acyltransferase
- PS
Phosphatidylserine
- PE
Phosphatidylethanolamine
- PA
Phosphatidic acid
- LTPs
Lipid transfer proteins
- OPA1
Optic atrophy 1
- INF2
Inverted formin 2
- DRP1
Dynamin-related protein 1
- MFF
Mitochondrial fission factor
- MiD49/51
Mitochondrial dynamics 49/51
- mtDNA
Mitochondrial DNA
- PERK
Protein kinase RNA-like kinase
- ATF6
Activating transcription factor 6
- IRE1
Inosital requiring enzyme 1
- BiP/GRP78
Heat shock protein 70 family protein 5
- CHOP
C/EBP homologous protein
- ETC
Electron transport chain
- Ero1
ER oxidoreductase 1
- p66Shc
66 KDa subtype of growth factor adapter Shc
- SERCA
Sarco/ER Ca2⁺-ATPase
- MAPK
Mitogen-activated protein kinase
- OSCP
ATP synthase oligomycin-sensitivity conferring protein
- ANT
Adenine nucleotide translocase
- CypD
Cyclophilin D
- MOMP
Mitochondrial outer membrane permeabilization
- AS
Atherosclerosis
- ECC
Excitation–contraction coupling
- DAMPs
Damage-associated molecular patterns
- PDIs
Protein disulfide isomerases
- XBP1
X-box binding protein-1
- JNK
C-Jun N-terminal kinase
- Noxs
Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases
- PO
Pressure overload
- VSMCs
Vascular smooth muscle cells
- hVSMCs
Human VSMCs
- OX-LDL
Oxidized LDL
- CANX
Calnexin
- CREB
CAMP response element-binding protein
- CLIC
Chloride intracellular channel
- clic4 -/-
CLIC4 gene knockout
- HR
Hypoxia-reoxygenation
- ATAD3A
ATPase family AAA-domain containing protein 3A
Author contributions
CC designed research topics and wrote manuscripts, FMX and WXM conducted literature search and preliminary screening, FMX and NKB revised manuscript language readability, CC and WXM created schematics and reviewed manuscripts. DGH and GWL are the co-corresponding authors responsible for the overall conception, framework design, and revision of the review. All authors read and approved the fnal manuscript.
Funding
The work was supported by National Natural Science Foundation of China (No.82204942); Natural Science Foundation of Shandong Province (No.ZR2022QH123); China Postdoctoral Science Foundation (No. 2022M721998); Shandong Taishan Scholar Project (No.tsqn202408383).
Availability of data and materials
Not applicable.
Declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Contributor Information
Guohua Dai, Email: daigh2004@163.com.
Wulin Gao, Email: gaowulin05@sina.com.
References
- 1.Jensen RV, Hjortbak MV, Bøtker HE. Ischemic heart disease: an update. Semin Nucl Med. 2020;50(3):195–207. 10.1053/j.semnuclmed.2020.02.007. [DOI] [PubMed] [Google Scholar]
- 2.Roth GA, Mensah GA, Johnson CO, Addolorato G, Ammirati E, Baddour LM, et al. Global burden of cardiovascular diseases and risk factors, 1990–2019: update from the GBD 2019 study. J Am Coll Cardiol. 2020;76(25):2982–3021. 10.1016/j.jacc.2020.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Lopez-Crisosto C, Pennanen C, Vasquez-Trincado C, Morales PE, Bravo-Sagua R, Quest AFG, Chiong M, Lavandero S. Sarcoplasmic reticulum-mitochondria communication in cardiovascular pathophysiology. Nat Rev Cardiol. 2017;14(6):342–60. 10.1038/nrcardio.2017.23. [DOI] [PubMed] [Google Scholar]
- 4.Chang X, Liu R, Li R, Peng Y, Zhu P, Zhou H. Molecular mechanisms of mitochondrial quality control in ischemic cardiomyopathy. Int J Biol Sci. 2023;19(2):426–48. 10.7150/ijbs.76223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Schwarz DS, Blower MD. The endoplasmic reticulum: structure, function and response to cellular signaling. Cell Mol Life Sci. 2016;73(1):79–94. 10.1007/s00018-015-2052-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Namba T. Regulation of endoplasmic reticulum functions. Aging (Albany NY). 2015;7(11):901–2. 10.18632/aging.100851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wiseman RL, Mesgarzadeh JS, Hendershot LM. Reshaping endoplasmic reticulum quality control through the unfolded protein response. Mol Cell. 2022;82(8):1477–91. 10.1016/j.molcel.2022.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Szymański J, Janikiewicz J, Michalska B, Patalas-Krawczyk P, Perrone M, Ziółkowski W, Duszyński J, Pinton P, Dobrzyń A, Więckowski MR. Interaction of mitochondria with the endoplasmic reticulum and plasma membrane in calcium homeostasis, lipid trafficking and mitochondrial structure. Int J Mol Sci. 2017;18(7):1576. 10.3390/ijms18071576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Saheki Y, De Camilli P. Endoplasmic reticulum-plasma membrane contact sites. Annu Rev Biochem. 2017;20(86):659–84. 10.1146/annurev-biochem-061516-044932. [DOI] [PubMed] [Google Scholar]
- 10.Rowland AA, Voeltz GK. Endoplasmic reticulum-mitochondria contacts: function of the junction. Nat Rev Mol Cell Biol. 2012;13(10):607–25. 10.1038/nrm3440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhou H, Wang S, Hu S, Chen Y, Ren J. ER-mitochondria microdomains in cardiac ischemia-reperfusion injury: a fresh perspective. Front Physiol. 2018;15(9):755. 10.3389/fphys.2018.00755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Giamogante F, Poggio E, Barazzuol L, Covallero A, Calì T. Apoptotic signals at the endoplasmic reticulum-mitochondria interface. Adv Protein Chem Struct Biol. 2021;126:307–43. 10.1016/bs.apcsb.2021.02.007. [DOI] [PubMed] [Google Scholar]
- 13.Simmen T, Aslan JE, Blagoveshchenskaya AD, Thomas L, Wan L, Xiang Y, Feliciangeli SF, Hung CH, Crump CM, Thomas G. PACS-2 controls endoplasmic reticulum-mitochondria communication and Bid-mediated apoptosis. EMBO J. 2005;24(4):717–29. 10.1038/sj.emboj.7600559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Arruda AP, Pers BM, Parlakgül G, Güney E, Inouye K, Hotamisligil GS. Chronic enrichment of hepatic endoplasmic reticulum-mitochondria contact leads to mitochondrial dysfunction in obesity. Nat Med. 2014;20(12):1427–35. 10.1038/nm.3735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wu S, Lu Q, Wang Q, Ding Y, Ma Z, Mao X, Huang K, Xie Z, Zou MH. Binding of FUN14 domain containing 1 with inositol 1,4,5-trisphosphate receptor in mitochondria-associated endoplasmic reticulum membranes maintains mitochondrial dynamics and function in hearts in vivo. Circulation. 2017;136(23):2248–66. 10.1161/CIRCULATIONAHA.117.030235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Gomez L, Thiebaut PA, Paillard M, Ducreux S, Abrial M, Crola Da Silva C, Durand A, Alam MR, Van Coppenolle F, Sheu SS, Ovize M. The SR/ER-mitochondria calcium crosstalk is regulated by GSK3β during reperfusion injury. Cell Death Differ. 2016. 10.1038/cdd.2015.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hayashi T, Su TP. Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate Ca(2+) signaling and cell survival. Cell. 2007;131(3):596–610. 10.1016/j.cell.2007.08.036. [DOI] [PubMed] [Google Scholar]
- 18.Ponnalagu D, Hamilton S, Sanghvi S, Antelo D, Schwieterman N, Hansra I, Xu X, Gao E, Edwards JC, Bansal SS, Wold LE, Terentyev D, Janssen PML, Hund TJ, Khan M, Kohut AR, Koch WJ, Singh H. CLIC4 localizes to mitochondrial-associated membranes and mediates cardioprotection. Sci Adv. 2022. 10.1126/sciadv.abo1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Li Z, Hu O, Xu S, Lin C, Yu W, Ma D, Lu J, Liu P. The SIRT3-ATAD3A axis regulates MAM dynamics and mitochondrial calcium homeostasis in cardiac hypertrophy. Int J Biol Sci. 2024;20(3):831–47. 10.7150/ijbs.89253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Beretta M, Santos CX, Molenaar C, Hafstad AD, Miller CC, Revazian A, Betteridge K, Schröder K, Streckfuß-Bömeke K, Doroshow JH, Fleck RA, Su TP, Belousov VV, Parsons M, Shah AM. Nox4 regulates InsP3 receptor-dependent Ca2+ release into mitochondria to promote cell survival. EMBO J. 2020;39(19): e103530. 10.15252/embj.2019103530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wu H, Carvalho P, Voeltz GK. Here, there, and everywhere: The importance of ER membrane contact sites. Science. 2018. 10.1126/science.aan5835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Prinz WA, Toulmay A, Balla T. The functional universe of membrane contact sites. Nat Rev Mol Cell Biol. 2020;21(1):7–24. 10.1038/s41580-019-0180-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Copeland DE, Dalton AJ. An association between mitochondria and the endoplasmic reticulum in cells of the pseudobranch gland of a teleost. J Biophys Biochem Cytol. 1959;5(3):393–6. 10.1083/jcb.5.3.393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Shore GC, Tata JR. Two fractions of rough endoplasmic reticulum from rat liver I. Recovery of rapidly sedimenting endoplasmic reticulum in association with mitochondria. J Cell Biol. 1977. 10.1083/jcb.72.3.714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Mannella CA, Buttle K, Rath BK, Marko M. Electron microscopic tomography of rat-liver mitochondria and their interaction with the endoplasmic reticulum. BioFactors. 1998;8(3–4):225–8. 10.1002/biof.5520080309. [DOI] [PubMed] [Google Scholar]
- 26.Achleitner G, Gaigg B, Krasser A, Kainersdorfer E, Kohlwein SD, Perktold A, Zellnig G, Daum G. Association between the endoplasmic reticulum and mitochondria of yeast facilitates interorganelle transport of phospholipids through membrane contact. Eur J Biochem. 1999;264(2):545–53. 10.1046/j.1432-1327.1999.00658.x. [DOI] [PubMed] [Google Scholar]
- 27.Silva-Palacios A, Zazueta C, Pedraza-Chaverri J. ER membranes associated with mitochondria: possible therapeutic targets in heart-associated diseases. Pharmacol Res. 2020;156: 104758. 10.1016/j.phrs.2020.104758. [DOI] [PubMed] [Google Scholar]
- 28.Hung V, Lam SS, Udeshi ND, Svinkina T, Guzman G, Mootha VK, Carr SA, Ting AY. Proteomic mapping of cytosol-facing outer mitochondrial and ER membranes in living human cells by proximity biotinylation. Elife. 2017;25(6): e24463. 10.7554/eLife.24463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Poston CN, Krishnan SC, Bazemore-Walker CR. In-depth proteomic analysis of mammalian mitochondria-associated membranes (MAM). J Proteomics. 2013;21(79):219–30. 10.1016/j.jprot.2012.12.018. [DOI] [PubMed] [Google Scholar]
- 30.Lang A, John Peter AT, Kornmann B. ER-mitochondria contact sites in yeast: beyond the myths of ERMES. Curr Opin Cell Biol. 2015;35:7–12. 10.1016/j.ceb.2015.03.002. [DOI] [PubMed] [Google Scholar]
- 31.Iwasawa R, Mahul-Mellier AL, Datler C, Pazarentzos E, Grimm S. Fis1 and Bap31 bridge the mitochondria-ER interface to establish a platform for apoptosis induction. EMBO J. 2011;30(3):556–68. 10.1038/emboj.2010.346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Filadi R, Greotti E, Pizzo P. Highlighting the endoplasmic reticulum-mitochondria connection: focus on Mitofusin 2. Pharmacol Res. 2018;128:42–51. 10.1016/j.phrs.2018.01.003. [DOI] [PubMed] [Google Scholar]
- 33.Csordás G, Várnai P, Golenár T, Roy S, Purkins G, Schneider TG, Balla T, Hajnóczky G. Imaging interorganelle contacts and local calcium dynamics at the ER-mitochondrial interface. Mol Cell. 2010;39(1):121–32. 10.1016/j.molcel.2010.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Shin DM, Muallem S. What the mitochondria see. Mol Cell. 2010;39(1):6–7. 10.1016/j.molcel.2010.06.030. [DOI] [PubMed] [Google Scholar]
- 35.De Stefani D, Rizzuto R, Pozzan T. Enjoy the trip: calcium in mitochondria back and forth. Annu Rev Biochem. 2016;2(85):161–92. 10.1146/annurev-biochem-060614-034216. [DOI] [PubMed] [Google Scholar]
- 36.Dorn GW 2nd, Maack C. SR and mitochondria: calcium cross-talk between kissing cousins. J Mol Cell Cardiol. 2013;55:42–9. 10.1016/j.yjmcc.2012.07.015. [DOI] [PubMed] [Google Scholar]
- 37.Clapham DE. Calcium signaling. Cell. 2007;131(6):1047–58. 10.1016/j.cell.2007.11.028. [DOI] [PubMed] [Google Scholar]
- 38.Szabadkai G, Bianchi K, Várnai P, De Stefani D, Wieckowski MR, Cavagna D, Nagy AI, Balla T, Rizzuto R. Chaperone-mediated coupling of endoplasmic reticulum and mitochondrial Ca2+ channels. J Cell Biol. 2006;175(6):901–11. 10.1083/jcb.200608073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Chai J, Xiong Q, Zhang P, Zheng R, Peng J, Jiang S. Induction of Ca2+ signal mediated apoptosis and alteration of IP3R1 and SERCA1 expression levels by stress hormone in differentiating C2C12 myoblasts. Gen Comp Endocrinol. 2010;166(2):241–9. 10.1016/j.ygcen.2009.08.011. [DOI] [PubMed] [Google Scholar]
- 40.Yadav VR, Song T, Mei L, Joseph L, Zheng YM, Wang YX. PLCγ1-PKCε-IP3R1 signaling plays an important role in hypoxia-induced calcium response in pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2018;314(5):L724–35. 10.1152/ajplung.00243.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Stoica R, De Vos KJ, Paillusson S, Mueller S, Sancho RM, Lau KF, Vizcay-Barrena G, Lin WL, Xu YF, Lewis J, Dickson DW, Petrucelli L, Mitchell JC, Shaw CE, Miller CC. ER-mitochondria associations are regulated by the VAPB-PTPIP51 interaction and are disrupted by ALS/FTD-associated TDP-43. Nat Commun. 2014;3(5):3996. 10.1038/ncomms4996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bauer TM, Murphy E. Role of mitochondrial calcium and the permeability transition pore in regulating cell death. Circ Res. 2020;126(2):280–93. 10.1161/CIRCRESAHA.119.316306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Stone SJ, Levin MC, Zhou P, Han J, Walther TC, Farese RV Jr. The endoplasmic reticulum enzyme DGAT2 is found in mitochondria-associated membranes and has a mitochondrial targeting signal that promotes its association with mitochondria. J Biol Chem. 2009;284(8):5352–61. 10.1074/jbc.M805768200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Galmes R, Houcine A, van Vliet AR, Agostinis P, Jackson CL, Giordano F. ORP5/ORP8 localize to endoplasmic reticulum-mitochondria contacts and are involved in mitochondrial function. EMBO Rep. 2016;17(6):800–10. 10.15252/embr.201541108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Janikiewicz J, Szymański J, Malinska D, Patalas-Krawczyk P, Michalska B, Duszyński J, Giorgi C, Bonora M, Dobrzyn A, Wieckowski MR. Mitochondria-associated membranes in aging and senescence: structure, function, and dynamics. Cell Death Dis. 2018;9(3):332. 10.1038/s41419-017-0105-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Shi W, Wu H, Liu S, Wu Z, Wu H, Liu J, Hou Y. Progesterone suppresses cholesterol esterification in app/ps1 mice and a cell model of Alzheimer’s disease. Brain Res Bull. 2021;173:162–73. 10.1016/j.brainresbull.2021.05.020. [DOI] [PubMed] [Google Scholar]
- 47.Drin G, Moser von Filseck J, Čopič A. New molecular mechanisms of inter-organelle lipid transport. Biochem Soc Trans. 2016;44(2):486–92. 10.1042/BST20150265. [DOI] [PubMed] [Google Scholar]
- 48.Mishra P, Carelli V, Manfredi G, Chan DC. Proteolytic cleavage of Opa1 stimulates mitochondrial inner membrane fusion and couples fusion to oxidative phosphorylation. Cell Metab. 2014;19(4):630–41. 10.1016/j.cmet.2014.03.011.Erratum.In:CellMetab.2014May6;19(5):891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Al Ojaimi M, Salah A, El-Hattab AW. Mitochondrial fission and fusion: molecular mechanisms, biological functions, and related disorders. Membranes (Basel). 2022;12(9):893. 10.3390/membranes12090893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Naon D, Zaninello M, Giacomello M, Varanita T, Grespi F, Lakshminaranayan S, Serafini A, Semenzato M, Herkenne S, Hernández-Alvarez MI, Zorzano A, De Stefani D, DornScorrano GWL. Critical reappraisal confirms that Mitofusin 2 is an endoplasmic reticulum-mitochondria tether. Proc Natl Acad Sci U S A. 2016;113(40):11249–54. 10.1073/pnas.1606786113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Filadi R, Greotti E, Turacchio G, Luini A, Pozzan T, Pizzo P. Mitofusin 2 ablation increases endoplasmic reticulum-mitochondria coupling. Proc Natl Acad Sci U S A. 2015;112(17):E2174–81. 10.1073/pnas.1504880112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.de Brito OM, Scorrano L. Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature. 2008;456(7222):605–10. 10.1038/nature07534. [DOI] [PubMed] [Google Scholar]
- 53.Friedman JR, Lackner LL, West M, DiBenedetto JR, Nunnari J, Voeltz GK. ER tubules mark sites of mitochondrial division. Science. 2011;334(6054):358–62. 10.1126/science.1207385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Korobova F, Ramabhadran V, Higgs HN. An actin-dependent step in mitochondrial fission mediated by the ER-associated formin INF2. Science. 2013;339(6118):464–7. 10.1126/science.1228360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Manor U, Bartholomew S, Golani G, Christenson E, Kozlov M, Higgs H, Spudich J, Lippincott-Schwartz J. A mitochondria-anchored isoform of the actin-nucleating spire protein regulates mitochondrial division. Elife. 2015;25(4): e08828. 10.7554/eLife.08828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Cho B, Cho HM, Jo Y, Kim HD, Song M, Moon C, Kim H, Kim K, Sesaki H, Rhyu IJ, Kim H, Sun W. Constriction of the mitochondrial inner compartment is a priming event for mitochondrial division. Nat Commun. 2017;9(8):15754. 10.1038/ncomms15754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lewis SC, Uchiyama LF, Nunnari J. ER-mitochondria contacts couple mtDNA synthesis with mitochondrial division in human cells. Science. 2016. 10.1126/science.aaf5549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Friedman JR, Webster BM, Mastronarde DN, Verhey KJ, Voeltz GK. ER sliding dynamics and ER-mitochondrial contacts occur on acetylated microtubules. J Cell Biol. 2010;190(3):363–75. 10.1083/jcb.200911024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Pilling AD, Horiuchi D, Lively CM, Saxton WM. Kinesin-1 and Dynein are the primary motors for fast transport of mitochondria in drosophila motor axons. Mol Biol Cell. 2006;17(4):2057–68. 10.1091/mbc.e05-06-0526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Woźniak MJ, Bola B, Brownhill K, Yang YC, Levakova V, Allan VJ. Role of kinesin-1 and cytoplasmic dynein in endoplasmic reticulum movement in VERO cells. J Cell Sci. 2009;122(Pt 12):1979–89. 10.1242/jcs.041962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Gutiérrez T, Simmen T. Endoplasmic reticulum chaperones tweak the mitochondrial calcium rheostat to control metabolism and cell death. Cell Calcium. 2018;70:64–75. 10.1016/j.ceca.2017.05.015. [DOI] [PubMed] [Google Scholar]
- 62.Booth DM, Enyedi B, Geiszt M, Várnai P, Hajnóczky G. Redox nanodomains are induced by and control calcium signaling at the ER-mitochondrial interface. Mol Cell. 2016;63(2):240–8. 10.1016/j.molcel.2016.05.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hetz C. The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol. 2012;13(2):89–102. 10.1038/nrm3270. [DOI] [PubMed] [Google Scholar]
- 64.Hetz C, Papa FR. The unfolded protein response and cell fate control. Mol Cell. 2018;69(2):169–81. 10.1016/j.molcel.2017.06.017. [DOI] [PubMed] [Google Scholar]
- 65.Song S, Tan J, Miao Y, Zhang Q. Crosstalk of ER stress-mediated autophagy and ER-phagy: Involvement of UPR and the core autophagy machinery. J Cell Physiol. 2018;233(5):3867–74. 10.1002/jcp.26137. [DOI] [PubMed] [Google Scholar]
- 66.Carreras-Sureda A, Jaña F, Urra H, Durand S, Mortenson DE, Sagredo A, et al. Non-canonical function of IRE1α determines mitochondria-associated endoplasmic reticulum composition to control calcium transfer and bioenergetics. Nat Cell Biol. 2019;21(6):755–67. 10.1038/s41556-019-0329-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Shuda M, Kondoh N, Imazeki N, Tanaka K, Okada T, Mori K, Hada A, Arai M, Wakatsuki T, Matsubara O, Yamamoto N, Yamamoto M. Activation of the ATF6, XBP1 and grp78 genes in human hepatocellular carcinoma: a possible involvement of the ER stress pathway in hepatocarcinogenesis. J Hepatol. 2003;38(5):605–14. 10.1016/s0168-8278(03)00029-1. [DOI] [PubMed] [Google Scholar]
- 68.Yuan M, Gong M, Zhang Z, Meng L, Tse G, Zhao Y, Bao Q, Zhang Y, Yuan M, Liu X, Li G, Liu T. Hyperglycemia induces endoplasmic reticulum stress in atrial cardiomyocytes, and mitofusin-2 downregulation prevents mitochondrial dysfunction and subsequent cell death. Oxid Med Cell Longev. 2020;22(2020):6569728. 10.1155/2020/6569728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lynes EM, Raturi A, Shenkman M, Ortiz Sandoval C, Yap MC, Wu J, Janowicz A, Myhill N, Benson MD, Campbell RE, Berthiaume LG, Lederkremer GZ, Simmen T. Palmitoylation is the switch that assigns calnexin to quality control or ER Ca2+ signaling. J Cell Sci. 2013;126(Pt 17):3893–903. 10.1242/jcs.125856. [DOI] [PubMed] [Google Scholar]
- 70.Verfaillie T, Rubio N, Garg AD, Bultynck G, Rizzuto R, Decuypere JP, Piette J, Linehan C, Gupta S, Samali A, Agostinis P. PERK is required at the ER-mitochondrial contact sites to convey apoptosis after ROS-based ER stress. Cell Death Differ. 2012;19(11):1880–91. 10.1038/cdd.2012.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Muñoz JP, Ivanova S, Sánchez-Wandelmer J, Martínez-Cristóbal P, Noguera E, Sancho A, Díaz-Ramos A, Hernández-Alvarez MI, Sebastián D, Mauvezin C, Palacín M, Zorzano A. Mfn2 modulates the UPR and mitochondrial function via repression of PERK. EMBO J. 2013;32(17):2348–61. 10.1038/emboj.2013.168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Rodríguez LR, Calap-Quintana P, Lapeña-Luzón T, Pallardó FV, Schneuwly S, Navarro JA, Gonzalez-Cabo P. Oxidative stress modulates rearrangement of endoplasmic reticulum-mitochondria contacts and calcium dysregulation in a Friedreich’s ataxia model. Redox Biol. 2020;37: 101762. 10.1016/j.redox.2020.101762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Frand AR, Kaiser CA. Ero1p oxidizes protein disulfide isomerase in a pathway for disulfide bond formation in the endoplasmic reticulum. Mol Cell. 1999;4(4):469–77. 10.1016/s1097-2765(00)80198-7. [DOI] [PubMed] [Google Scholar]
- 74.Landeta C, Boyd D, Beckwith J. Disulfide bond formation in prokaryotes. Nat Microbiol. 2018;3(3):270–80. 10.1038/s41564-017-0106-2. [DOI] [PubMed] [Google Scholar]
- 75.Pinton P, Rimessi A, Marchi S, Orsini F, Migliaccio E, Giorgio M, Contursi C, Minucci S, Mantovani F, Wieckowski MR, Del Sal G, Pelicci PG, Rizzuto R. Protein kinase C beta and prolyl isomerase 1 regulate mitochondrial effects of the life-span determinant p66Shc. Science. 2007;315(5812):659–63. 10.1126/science.1135380. [DOI] [PubMed] [Google Scholar]
- 76.Giorgio M, Migliaccio E, Orsini F, Paolucci D, Moroni M, Contursi C, Pelliccia G, Luzi L, Minucci S, Marcaccio M, Pinton P, Rizzuto R, Bernardi P, Paolucci F, Pelicci PG. Electron transfer between cytochrome c and p66Shc generates reactive oxygen species that trigger mitochondrial apoptosis. Cell. 2005;122(2):221–33. 10.1016/j.cell.2005.05.011. [DOI] [PubMed] [Google Scholar]
- 77.Debattisti V, Gerencser AA, Saotome M, Das S, Hajnóczky G. ROS control mitochondrial motility through p38 and the motor adaptor Miro/Trak. Cell Rep. 2017;21(6):1667–80. 10.1016/j.celrep.2017.10.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wang N, Wang C, Zhao H, He Y, Lan B, Sun L, Gao Y. The MAMs structure and its role in cell death. Cells. 2021;10(3):657. 10.3390/cells10030657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Malhotra JD, Kaufman RJ. ER stress and its functional link to mitochondria: role in cell survival and death. Cold Spring Harb Perspect Biol. 2011;3(9): a004424. 10.1101/cshperspect.a004424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Marchi S, Patergnani S, Missiroli S, Morciano G, Rimessi A, Wieckowski MR, Giorgi C, Pinton P. Mitochondrial and endoplasmic reticulum calcium homeostasis and cell death. Cell Calcium. 2018;69:62–72. 10.1016/j.ceca.2017.05.003. [DOI] [PubMed] [Google Scholar]
- 81.Wang B, Nguyen M, Chang NC, Shore GC. Fis1, Bap31 and the kiss of death between mitochondria and endoplasmic reticulum. EMBO J. 2011;30(3):451–2. 10.1038/emboj.2010.352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Hoppins S, Nunnari J. Cell biology. Mitochondrial dynamics and apoptosis–the ER connection. Science. 2012. 10.1126/science.1224709. [DOI] [PubMed] [Google Scholar]
- 83.Montessuit S, Somasekharan SP, Terrones O, Lucken-Ardjomande S, Herzig S, Schwarzenbacher R, Manstein DJ, Bossy-Wetzel E, Basañez G, Meda P, Martinou JC. Membrane remodeling induced by the dynamin-related protein Drp1 stimulates Bax oligomerization. Cell. 2010;142(6):889–901. 10.1016/j.cell.2010.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Mazur M, Kmita H, Wojtkowska M. The diversity of the mitochondrial outer membrane protein import channels: emerging targets for modulation. Molecules. 2021;26(13):4087. 10.3390/molecules26134087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ichimiya T, Yamakawa T, Hirano T, Yokoyama Y, Hayashi Y, Hirayama D, Wagatsuma K, Itoi T, Nakase H. Autophagy and autophagy-related diseases: a review. Int J Mol Sci. 2020;21(23):8974. 10.3390/ijms21238974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ji C, Zhang Z, Li Z, She X, Wang X, Li B, Xu X, Song D, Zhang D. Mitochondria-associated endoplasmic reticulum membranes: inextricably linked with autophagy process. Oxid Med Cell Longev. 2022;23(2022):7086807. 10.1155/2022/7086807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hamasaki M, Furuta N, Matsuda A, Nezu A, Yamamoto A, Fujita N, Oomori H, Noda T, Haraguchi T, Hiraoka Y, Amano A, Yoshimori T. Autophagosomes form at ER-mitochondria contact sites. Nature. 2013;495(7441):389–93. 10.1038/nature11910. [DOI] [PubMed] [Google Scholar]
- 88.Gomez-Suaga P, Paillusson S, Stoica R, Noble W, Hanger DP, Miller CCJ. The ER-mitochondria tethering complex VAPB-PTPIP51 regulates autophagy. Curr Biol. 2017;27(3):371–85. 10.1016/j.cub.2016.12.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Barazzuol L, Giamogante F, Brini M, Calì T. PINK1/Parkin mediated mitophagy, Ca2+ signalling, and ER-mitochondria contacts in parkinson’s disease. Int J Mol Sci. 2020;21(5):1772. 10.3390/ijms21051772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Gelmetti V, De Rosa P, Torosantucci L, Marini ES, Romagnoli A, Di Rienzo M, Arena G, Vignone D, Fimia GM, Valente EM. PINK1 and BECN1 relocalize at mitochondria-associated membranes during mitophagy and promote ER-mitochondria tethering and autophagosome formation. Autophagy. 2017;13(4):654–69. 10.1080/15548627.2016.1277309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wu W, Li W, Chen H, Jiang L, Zhu R, Feng D. FUNDC1 is a novel mitochondrial-associated-membrane (MAM) protein required for hypoxia-induced mitochondrial fission and mitophagy. Autophagy. 2016;12(9):1675–6. 10.1080/15548627.2016.1193656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Morciano G, Giorgi C, Bonora M, Punzetti S, Pavasini R, Wieckowski MR, Campo G, Pinton P. Molecular identity of the mitochondrial permeability transition pore and its role in ischemia-reperfusion injury. J Mol Cell Cardiol. 2015;78:142–53. 10.1016/j.yjmcc.2014.08.015. [DOI] [PubMed] [Google Scholar]
- 93.Agyapong ED, Pedriali G, Ramaccini D, Bouhamida E, Tremoli E, Giorgi C, Pinton P, Morciano G. Calcium signaling from sarcoplasmic reticulum and mitochondria contact sites in acute myocardial infarction. J Transl Med. 2024;22(1):552. 10.1186/s12967-024-05240-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Smith GL, Eisner DA. Calcium buffering in the heart in health and disease. Circulation. 2019;139(20):2358–71. 10.1161/CIRCULATIONAHA.118.039329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kohlhaas M, Maack C. Calcium release microdomains and mitochondria. Cardiovasc Res. 2013;98(2):259–68. 10.1093/cvr/cvt032. [DOI] [PubMed] [Google Scholar]
- 96.Fuping Z, Wuping L, Linhua W, Chengxi P, Fuqiang Z, Yi Z, Aijun W. Tao-Hong-Si-Wu decoction reduces ischemia reperfusion rat myoblast cells calcium overloading and inflammation through the Wnt/IP3R/CAMKII pathway. J Cell Biochem. 2019;120(8):13095–106. 10.1002/jcb.28582. [DOI] [PubMed] [Google Scholar]
- 97.Zhao H, Liu D, Yan Q, Bian X, Yu J, Wang J, Cheng X, Xu Z. Endoplasmic reticulum Stress/Ca2+-calmodulin-dependent protein kinase/signal transducer and activator of transcription 3 pathway plays a role in the regulation of cellular zinc deficiency in myocardial ischemia/reperfusion injury. Front Physiol. 2022;5(12): 736920. 10.3389/fphys.2021.736920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Gambardella J, Trimarco B, Iaccarino G, Santulli G. New insights in cardiac calcium handling and excitation-contraction coupling. Adv Exp Med Biol. 2018;1067:373–85. 10.1007/5584_2017_106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Zhang Y, Zhou H, Wu W, Shi C, Hu S, Yin T, Ma Q, Han T, Zhang Y, Tian F, Chen Y. Liraglutide protects cardiac microvascular endothelial cells against hypoxia/reoxygenation injury through the suppression of the SR-Ca(2+)-XO-ROS axis via activation of the GLP-1R/PI3K/Akt/survivin pathways. Free Radic Biol Med. 2016;95:278–92. 10.1016/j.freeradbiomed.2016.03.035. [DOI] [PubMed] [Google Scholar]
- 100.Elrod JW, Molkentin JD. Physiologic functions of cyclophilin D and the mitochondrial permeability transition pore. Circ J. 2013;77(5):1111–22. 10.1253/circj.cj-13-0321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Ong SB, Samangouei P, Kalkhoran SB, Hausenloy DJ. The mitochondrial permeability transition pore and its role in myocardial ischemia reperfusion injury. J Mol Cell Cardiol. 2015;78:23–34. 10.1016/j.yjmcc.2014.11.005. [DOI] [PubMed] [Google Scholar]
- 102.Paillard M, Tubbs E, Thiebaut PA, Gomez L, Fauconnier J, Da Silva CC, Teixeira G, Mewton N, Belaidi E, Durand A, Abrial M, Lacampagne A, Rieusset J, Ovize M. Depressing mitochondria-reticulum interactions protects cardiomyocytes from lethal hypoxia-reoxygenation injury. Circulation. 2013;128(14):1555–65. 10.1161/CIRCULATIONAHA.113.001225. [DOI] [PubMed] [Google Scholar]
- 103.Garcia-Dorado D, Ruiz-Meana M, Inserte J, Rodriguez-Sinovas A, Piper HM. Calcium-mediated cell death during myocardial reperfusion. Cardiovasc Res. 2012;94(2):168–80. 10.1093/cvr/cvs116. [DOI] [PubMed] [Google Scholar]
- 104.Gӧbel J, Engelhardt E, Pelzer P, Sakthivelu V, Jahn HM, Jevtic M, Folz-Donahue K, Kukat C, Schauss A, Frese CK, Giavalisco P, Ghanem A, Conzelmann KK, Motori E, Bergami M. Mitochondria-endoplasmic reticulum contacts in reactive astrocytes promote vascular remodeling. Cell Metab. 2020;31(4):791-808.e8. 10.1016/j.cmet.2020.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Pike MM, Luo CS, Clark MD, Kirk KA, Kitakaze M, Madden MC, Cragoe EJ Jr, Pohost GM. NMR measurements of Na+ and cellular energy in ischemic rat heart: role of Na(+)-H+ exchange. Am J Physiol. 1993;265(6 Pt 2):H2017–26. 10.1152/ajpheart.1993.265.6.H2017. [DOI] [PubMed] [Google Scholar]
- 106.Bonora M, Giorgi C, Pinton P. Molecular mechanisms and consequences of mitochondrial permeability transition. Nat Rev Mol Cell Biol. 2022;23(4):266–85. 10.1038/s41580-021-00433-y. [DOI] [PubMed] [Google Scholar]
- 107.Morciano G, Naumova N, Koprowski P, Valente S, Sardão VA, Potes Y, Rimessi A, Wieckowski MR, Oliveira PJ. The mitochondrial permeability transition pore: an evolving concept critical for cell life and death. Biol Rev Camb Philos Soc. 2021;96(6):2489–521. 10.1111/brv.12764. [DOI] [PubMed] [Google Scholar]
- 108.Chang X, Zhou S, Liu J, Wang Y, Guan X, Wu Q, Liu Z, Liu R. Zishenhuoxue decoction-induced myocardial protection against ischemic injury through TMBIM6-VDAC1-mediated regulation of calcium homeostasis and mitochondrial quality surveillance. Phytomedicine. 2024;132: 155331. 10.1016/j.phymed.2023.155331. [DOI] [PubMed] [Google Scholar]
- 109.Thoudam T, Jeon JH, Ha CM, Lee IK. Role of mitochondria-associated endoplasmic reticulum membrane in inflammation-mediated metabolic diseases. Mediators Inflamm. 2016;2016:1851420. 10.1155/2016/1851420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Pang B, Dong G, Pang T, Sun X, Liu X, Nie Y, Chang X. Emerging insights into the pathogenesis and therapeutic strategies for vascular endothelial injury-associated diseases: focus on mitochondrial dysfunction. Angiogenesis. 2024;27(4):623–39. 10.1007/s10456-024-09938-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Ong SB, Subrayan S, Lim SY, Yellon DM, Davidson SM, Hausenloy DJ. Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation. 2010;121(18):2012–22. 10.1161/CIRCULATIONAHA.109.906610. [DOI] [PubMed] [Google Scholar]
- 112.Hall AR, Burke N, Dongworth RK, Kalkhoran SB, Dyson A, Vicencio JM, Dorn GW II, Yellon DM, Hausenloy DJ. Hearts deficient in both Mfn1 and Mfn2 are protected against acute myocardial infarction. Cell Death Dis. 2016;7(5): e2238. 10.1038/cddis.2016.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Chen L, Gong Q, Stice JP, Knowlton AA. Mitochondrial OPA1, apoptosis, and heart failure. Cardiovasc Res. 2009;84(1):91–9. 10.1093/cvr/cvp181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Zhang IX, Raghavan M, Satin LS. The endoplasmic reticulum and calcium homeostasis in pancreatic beta cells. Endocrinology. 2020. 10.1210/endocr/bqz028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Xu Y, Chen J, Chen J, Teng J. EI24 promotes cell adaption to ER stress by coordinating IRE1 signaling and calcium homeostasis. EMBO Rep. 2022;23(3): e51679. 10.15252/embr.202051679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Thuerauf DJ, Marcinko M, Gude N, Rubio M, Sussman MA, Glembotski CC. Activation of the unfolded protein response in infarcted mouse heart and hypoxic cultured cardiac myocytes. Circ Res. 2006;99(3):275–82. 10.1161/01.RES.0000233317.70421.03. [DOI] [PubMed] [Google Scholar]
- 117.Hu H, Tian M, Ding C, Yu S. The C/EBP homologous protein (CHOP) transcription factor functions in endoplasmic reticulum stress-induced apoptosis and microbial infection. Front Immunol. 2019;4(9):3083. 10.3389/fimmu.2018.03083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Hetz C, Russelakis-Carneiro M, Maundrell K, Castilla J, Soto C. Caspase-12 and endoplasmic reticulum stress mediate neurotoxicity of pathological prion protein. EMBO J. 2021;40(18): e109151. 10.15252/embj.2021109151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Arshad M, Ye Z, Gu X, Wong CK, Liu Y, Li D, Zhou L, Zhang Y, Bay WP, Yu VC, Li P. RNF13, a RING finger protein, mediates endoplasmic reticulum stress-induced apoptosis through the inositol-requiring enzyme (IRE1α)/c-Jun NH2-terminal kinase pathway. J Biol Chem. 2013;288(12):8726–36. 10.1074/jbc.M112.368829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Alam S, Abdullah CS, Aishwarya R, Orr AW, Traylor J, Miriyala S, Panchatcharam M, Pattillo CB, Bhuiyan MS. Sigmar1 regulates endoplasmic reticulum stress-induced C/EBP-homologous protein expression in cardiomyocytes. 2017. Biosci Rep. 10.1042/BSR20170898. [DOI] [PMC free article] [PubMed]
- 121.Tomczyk M, Kraszewska I, Szade K, Bukowska-Strakova K, Meloni M, Jozkowicz A, Dulak J, Jazwa A. Splenic Ly6Chi monocytes contribute to adverse late post-ischemic left ventricular remodeling in heme oxygenase-1 deficient mice. Basic Res Cardiol. 2017;112(4):39. 10.1007/s00395-017-0629-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Braunersreuther V, Montecucco F, Asrih M, Pelli G, Galan K, Frias M, Burger F, Quinderé AL, Montessuit C, Krause KH, Mach F, Jaquet V. Role of NADPH oxidase isoforms NOX1, NOX2 and NOX4 in myocardial ischemia/reperfusion injury. J Mol Cell Cardiol. 2013;64:99–107. 10.1016/j.yjmcc.2013.09.007. [DOI] [PubMed] [Google Scholar]
- 123.Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med. 2013;368(7):651–62. 10.1056/NEJMra1205406. [DOI] [PubMed] [Google Scholar]
- 124.Kroemer G. Autophagy: a druggable process that is deregulated in aging and human disease. J Clin Invest. 2015;125(1):1–4. 10.1172/JCI78652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Matsui Y, Takagi H, Qu X, Abdellatif M, Sakoda H, Asano T, Levine B, Sadoshima J. Distinct roles of autophagy in the heart during ischemia and reperfusion: roles of AMP-activated protein kinase and Beclin 1 in mediating autophagy. Circ Res. 2007;100(6):914–22. 10.1161/01.RES.0000261924.76669.36. [DOI] [PubMed] [Google Scholar]
- 126.Liu Y, Shoji-Kawata S, Sumpter RM Jr, Wei Y, Ginet V, Zhang L, Posner B, Tran KA, Green DR, Xavier RJ, Shaw SY, Clarke PG, Puyal J, Levine B. Autosis is a Na+, K+-ATPase-regulated form of cell death triggered by autophagy-inducing peptides, starvation, and hypoxia-ischemia. Proc Natl Acad Sci U S A. 2013;110(51):20364–71. 10.1073/pnas.1319661110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Qiao X, Jia S, Ye J, Fang X, Zhang C, Cao Y, Xu C, Zhao L, Zhu Y, Wang L, Zheng M. PTPIP51 regulates mouse cardiac ischemia/reperfusion through mediating the mitochondria-SR junction. Sci Rep. 2017;27(7):45379. 10.1038/srep45379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Sciarretta S, Maejima Y, Zablocki D, Sadoshima J. The role of autophagy in the heart. Annu Rev Physiol. 2018;10(80):1–26. 10.1146/annurev-physiol-021317-121427. [DOI] [PubMed] [Google Scholar]
- 129.Breckenridge DG, Stojanovic M, Marcellus RC, Shore GC. Caspase cleavage product of BAP31 induces mitochondrial fission through endoplasmic reticulum calcium signals, enhancing cytochrome c release to the cytosol. J Cell Biol. 2003;160(7):1115–27. 10.1083/jcb.200212059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Ference BA, Ginsberg HN, Graham I, Ray KK, Packard CJ, Bruckert E, et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European atherosclerosis society consensus Panel. Eur Heart J. 2017. 10.1093/eurheartj/ehx144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Moulis M, Grousset E, Faccini J, Richetin K, Thomas G, Vindis C. The multifunctional sorting protein PACS-2 controls mitophagosome formation in human vascular smooth muscle cells through mitochondria-ER contact sites. Cells. 2019;8(6):638. 10.3390/cells8060638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Li C, Li L, Yang M, Zeng L, Sun L. PACS-2: A key regulator of mitochondria-associated membranes (MAMs). Pharmacol Res. 2020;160: 105080. 10.1016/j.phrs.2020.105080. [DOI] [PubMed] [Google Scholar]
- 133.Setten RL, Rossi JJ, Han SP. The current state and future directions of RNAi-based therapeutics. Nat Rev Drug Discov. 2019;18(6):421–46. 10.1038/s41573-019-0017-4. [DOI] [PubMed] [Google Scholar]
- 134.Gomez L, Paillard M, Thibault H, Derumeaux G, Ovize M. Inhibition of GSK3beta by postconditioning is required to prevent opening of the mitochondrial permeability transition pore during reperfusion. Circulation. 2008;117(21):2761–8. 10.1161/CIRCULATIONAHA.107.755066. [DOI] [PubMed] [Google Scholar]
- 135.Wang S, Zhang F, Zhao G, Cheng Y, Wu T, Wu B, Zhang YE. Mitochondrial PKC-ε deficiency promotes I/R-mediated myocardial injury via GSK3β-dependent mitochondrial permeability transition pore opening. J Cell Mol Med. 2017;21(9):2009–21. 10.1111/jcmm.13121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Nishihara M, Miura T, Miki T, Tanno M, Yano T, Naitoh K, Ohori K, Hotta H, Terashima Y, Shimamoto K. Modulation of the mitochondrial permeability transition pore complex in GSK-3beta-mediated myocardial protection. J Mol Cell Cardiol. 2007;43(5):564–70. 10.1016/j.yjmcc.2007.08.010. [DOI] [PubMed] [Google Scholar]
- 137.Juhaszova M, Zorov DB, Kim SH, Pepe S, Fu Q, Fishbein KW, Ziman BD, Wang S, Ytrehus K, Antos CL, Olson EN, Sollott SJ. Glycogen synthase kinase-3beta mediates convergence of protection signaling to inhibit the mitochondrial permeability transition pore. J Clin Invest. 2004;113(11):1535–49. 10.1172/JCI19906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Sharma AK, Thanikachalam PV, Bhatia S. The signaling interplay of GSK-3β in myocardial disorders. Drug Discov Today. 2020;25(4):633–41. 10.1016/j.drudis.2020.01.017. [DOI] [PubMed] [Google Scholar]
- 139.Stoica R, Paillusson S, Gomez-Suaga P, Mitchell JC, Lau DH, Gray EH, Sancho RM, Vizcay-Barrena G, De Vos KJ, Shaw CE, Hanger DP, Noble W, Miller CC. ALS/FTD-associated FUS activates GSK-3β to disrupt the VAPB-PTPIP51 interaction and ER-mitochondria associations. EMBO Rep. 2016;17(9):1326–42. 10.15252/embr.201541726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Zhai P, Sciarretta S, Galeotti J, Volpe M, Sadoshima J. Differential roles of GSK-3β during myocardial ischemia and ischemia/reperfusion. Circ Res. 2011;109(5):502–11. 10.1161/CIRCRESAHA.111.249532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Jiang T, Ruan N, Luo P, Wang Q, Wei X, Li Y, Dai Y, Lin L, Lv J, Liu Y, Zhang C. Modulation of ER-mitochondria tethering complex VAPB-PTPIP51: novel therapeutic targets for aging-associated diseases. Ageing Res Rev. 2024;98: 102320. 10.1016/j.arr.2024.102320. [DOI] [PubMed] [Google Scholar]
- 142.Liu L, Feng D, Chen G, Chen M, Zheng Q, Song P, Ma Q, Zhu C, Wang R, Qi W, Huang L, Xue P, Li B, Wang X, Jin H, Wang J, Yang F, Liu P, Zhu Y, Sui S, Chen Q. Mitochondrial outer-membrane protein FUNDC1 mediates hypoxia-induced mitophagy in mammalian cells. Nat Cell Biol. 2012;14(2):177–85. 10.1038/ncb2422. [DOI] [PubMed] [Google Scholar]
- 143.Wu W, Lin C, Wu K, Jiang L, Wang X, Li W, Zhuang H, Zhang X, Chen H, Li S, Yang Y, Lu Y, Wang J, Zhu R, Zhang L, Sui S, Tan N, Zhao B, Zhang J, Li L, Feng D. FUNDC1 regulates mitochondrial dynamics at the ER-mitochondrial contact site under hypoxic conditions. EMBO J. 2016;35(13):1368–84. 10.15252/embj.201593102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Wu S, Lu Q, Ding Y, Wu Y, Qiu Y, Wang P, Mao X, Huang K, Xie Z, Zou MH. Hyperglycemia-Driven inhibition of AMP-activated protein kinase α2 induces diabetic cardiomyopathy by promoting mitochondria-associated endoplasmic reticulum membranes in vivo. Circulation. 2019;139(16):1913–36. 10.1161/CIRCULATIONAHA.118.033552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Li Y, Liu Z, Zhang Y, Zhao Q, Wang X, Lu P, Zhang H, Wang Z, Dong H, Zhang Z. PEDF protects cardiomyocytes by promoting FUNDC1-mediated mitophagy via PEDF-R under hypoxic condition. Int J Mol Med. 2018;41(6):3394–404. 10.3892/ijmm.2018.3536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Zhou H, Zhu P, Guo J, Hu N, Wang S, Li D, Hu S, Ren J, Cao F, Chen Y. Ripk3 induces mitochondrial apoptosis via inhibition of FUNDC1 mitophagy in cardiac IR injury. Redox Biol. 2017. 10.1016/j.redox.2017.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Wang J, Zhuang H, Jia L, He X, Zheng S, Ji K, Xie K, Ying T, Zhang Y, Li C, Chang X. Nuclear receptor subfamily 4 group A member 1 promotes myocardial ischemia/reperfusion injury through inducing mitochondrial fission factor-mediated mitochondrial fragmentation and inhibiting FUN14 domain containing 1-depedent mitophagy. Int J Biol Sci. 2024;20(11):4458–75. 10.7150/ijbs.95853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Lv M, Wang C, Li F, Peng J, Wen B, Gong Q, Shi Y, Tang Y. Structural insights into the recognition of phosphorylated FUNDC1 by LC3B in mitophagy. Protein Cell. 2017;8(1):25–38. 10.1007/s13238-016-0328-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Li G, Li J, Shao R, Zhao J, Chen M. FUNDC1: a promising mitophagy regulator at the mitochondria-associated membrane for cardiovascular diseases. Front Cell Dev Biol. 2021;16(9): 788634. 10.3389/fcell.2021.788634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Ponnalagu D, Hussain AT, Thanawala R, Meka J, Bednarczyk P, Feng Y, Szewczyk A, GururajaRao S, Bopassa JC, Khan M, Singh H. Chloride channel blocker IAA-94 increases myocardial infarction by reducing calcium retention capacity of the cardiac mitochondria. Life Sci. 2019;15(235): 116841. 10.1016/j.lfs.2019.116841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Diaz RJ, Losito VA, Mao GD, Ford MK, Backx PH, Wilson GJ. Chloride channel inhibition blocks the protection of ischemic preconditioning and hypo-osmotic stress in rabbit ventricular myocardium. Circ Res. 1999;84(7):763–75. 10.1161/01.res.84.7.763. [DOI] [PubMed] [Google Scholar]
- 152.Zhao Y, Sun X, Hu D, Prosdocimo DA, Hoppel C, Jain MK, Ramachandran R, Qi X. ATAD3A oligomerization causes neurodegeneration by coupling mitochondrial fragmentation and bioenergetics defects. Nat Commun. 2019;10(1):1371. 10.1038/s41467-019-09291-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Issop L, Fan J, Lee S, Rone MB, Basu K, Mui J, Papadopoulos V. Mitochondria-associated membrane formation in hormone-stimulated Leydig cell steroidogenesis: role of ATAD3. Endocrinology. 2015;156(1):334–45. 10.1210/en.2014-1503. [DOI] [PubMed] [Google Scholar]
- 154.Ehmke H. The sigma-1 receptor: a molecular chaperone for the heart and the soul? Cardiovasc Res. 2012;93(1):6–7. 10.1093/cvr/cvr313. [DOI] [PubMed] [Google Scholar]
- 155.Tian S, Lei P, Zhang J, Sun Y, Li B, Shan Y. Sulforaphane balances Ca2+ homeostasis injured by excessive fat via mitochondria-associated membrane (MAM). Mol Nutr Food Res. 2021;65(14): e2001076. 10.1002/mnfr.202001076. [DOI] [PubMed] [Google Scholar]
- 156.Assis LHP, Dorighello GG, Rentz T, de Souza JC, Vercesi AE, de Oliveira HCF. In Vivo pravastatin treatment reverses hypercholesterolemia induced mitochondria-associated membranes contact sites, foam cell formation, and phagocytosis in macrophages. Front Mol Biosci. 2022;15(9): 839428. 10.3389/fmolb.2022.839428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Tagashira H, Bhuiyan S, Shioda N, Hasegawa H, Kanai H, Fukunaga K. Sigma1-receptor stimulation with fluvoxamine ameliorates transverse aortic constriction-induced myocardial hypertrophy and dysfunction in mice. Am J Physiol Heart Circ Physiol. 2010;299(5):H1535–45. 10.1152/ajpheart.00198.2010. [DOI] [PubMed] [Google Scholar]
- 158.Vallese F, Catoni C, Cieri D, Barazzuol L, Ramirez O, Calore V, Bonora M, Giamogante F, Pinton P, Brini M, Calì T. An expanded palette of improved SPLICS reporters detects multiple organelle contacts in vitro and in vivo. Nat Commun. 2020;11(1):6069. 10.1038/s41467-020-19892-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Calì T, Brini M. Quantification of organelle contact sites by split-GFP-based contact site sensors (SPLICS) in living cells. Nat Protoc. 2021;16(11):5287–308. 10.1038/s41596-021-00614-1. [DOI] [PubMed] [Google Scholar]
- 160.Kwak C, Shin S, Park JS, Jung M, Nhung TTM, Kang MG, Lee C, Kwon TH, Park SK, Mun JY, Kim JS, Rhee HW. Contact-ID, a tool for profiling organelle contact sites, reveals regulatory proteins of mitochondrial-associated membrane formation. Proc Natl Acad Sci U S A. 2020;117(22):12109–20. 10.1073/pnas.1916584117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Lee A, Sung G, Shin S, Lee SY, Sim J, Nhung TTM, Nghi TD, Park SK, Sathieshkumar PP, Kang I, Mun JY, Kim JS, Rhee HW, Park KM, Kim K. OrthoID: profiling dynamic proteomes through time and space using mutually orthogonal chemical tools. Nat Commun. 2024;15(1):1851. 10.1038/s41467-024-46034-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Cho E, Woo Y, Suh Y, Suh BK, Kim SJ, Nhung TTM, Yoo JY, Nghi TD, Lee SB, Mun DJ, Park SK. Ratiometric measurement of MAM Ca2+ dynamics using a modified CalfluxVTN. Nat Commun. 2023;14(1):3586. 10.1038/s41467-023-39343-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Citations
- Alam S, Abdullah CS, Aishwarya R, Orr AW, Traylor J, Miriyala S, Panchatcharam M, Pattillo CB, Bhuiyan MS. Sigmar1 regulates endoplasmic reticulum stress-induced C/EBP-homologous protein expression in cardiomyocytes. 2017. Biosci Rep. 10.1042/BSR20170898. [DOI] [PMC free article] [PubMed]
Data Availability Statement
Not applicable.





