Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
February-2026 Volume 57 Issue 2

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
February-2026 Volume 57 Issue 2

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Mitochondria-associated endoplasmic reticulum membranes: Emerging regulators of cardiac microvascular ischemia/reperfusion injury (Review)

  • Authors:
    • Yan Wang
    • Baowei Feng
    • Yanting Wu
    • Zongle Sun
    • Hao Yuan
    • Wei Chen
    • Chang Zhao
    • Zhi Liu
  • View Affiliations / Copyright

    Affiliations: College of Integration of Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China, College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China, Changchun Humanities and Sciences College, Changchun, Jilin 130118, P.R. China, College of the First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
    Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 4.0].
  • Article Number: 34
    |
    Published online on: December 2, 2025
       https://doi.org/10.3892/ijmm.2025.5705
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Ischemic heart disease remains the leading cause of global disease burden among cardiovascular disorders. In addition to cardiomyocyte injury, ischemia-reperfusion (I/R)-induced microvascular damage plays a crucial role in determining tissue dysfunction and overall prognosis. Mitochondria-associated endoplasmic reticulum membranes (MAMs), specialized contact sites between the ER and mitochondria, are now recognized as key regulators of cardiovascular pathophysiology. The present review summarized current knowledge of the structure of MAMs and their effects on endothelial cells under hypoxia/reoxygenation conditions. Particular attention was given to their role in regulating mitochondrial quality control processes, including fission, fusion, oxidative stress, mitophagy and Ca2+ homeostasis, within the context of cardiac microvascular I/R injury. Targeting MAMs may represent a promising strategy for microvascular protection in ischemic heart disease.

Introduction

Cardiovascular diseases (CVDs) continue to be the primary cause of death worldwide, with myocardial infarction contributing markedly to global morbidity and mortality (1). The timely restoration of coronary blood flow through thrombolytic therapy or percutaneous coronary intervention (PCI) has been proven to effectively reduce infarct size (2). However, ischemia/reperfusion (I/R) injury induces secondary myocardial damage, mainly affecting cardiomyocytes and cardiac microvascular endothelial cells (CMECs). This secondary injury is characterized by myocardial stunning, microvascular dysfunction, reperfusion arrhythmias and lethal reperfusion injury (3). Microvascular I/R injury involves CMEC apoptosis, microvascular spasm and impaired perfusion, all of which exacerbate myocardial damage after reperfusion (4). Beyond conventional clinical risk scores and left ventricular ejection fraction, microvascular obstruction due to I/R injury has been identified as an independent predictor of infarct size and poor prognosis (5-7).

Mitochondria-associated endoplasmic reticulum membranes (MAMs) serve as key signaling hubs involved in fundamental cellular processes, including calcium (Ca2+) and lipid homeostasis, mitochondrial dynamics and energy production (8). During cardiac microvascular I/R injury, abnormal mitochondrial dynamics and impaired mitophagy (2,9) further exacerbate microvascular dysfunction by compromising vascular patency, altering vascular tone and amplifying inflammatory responses (10). Further, excessive mitochondrial Ca2+ uptake leads to overactivation of the electron transport chain (ETC) and excessive generation of reactive oxygen species (ROS) (11). This pathological cascade leads to the collapse of the mitochondrial membrane potential and promotes the opening of the mitochondrial permeability transition pore (mPTP) (12). Thus, mitochondrial dysfunction represents a central mechanism in microvascular I/R injury (13,14) (Fig. 1).

The pathological functions of
mitochondrial dynamics, mitochondrial oxidative stress, mitophagy
and calcium overload in cardiac microvascular I/R injury. I/R,
ischemia-reperfusion; MAM, mitochondria-associated endoplasmic
reticulum membrane; ROS, reactive oxygen species; mtDNA;
mitochondrial DNA; mROS, mitochondrial ROS.

Figure 1

The pathological functions of mitochondrial dynamics, mitochondrial oxidative stress, mitophagy and calcium overload in cardiac microvascular I/R injury. I/R, ischemia-reperfusion; MAM, mitochondria-associated endoplasmic reticulum membrane; ROS, reactive oxygen species; mtDNA; mitochondrial DNA; mROS, mitochondrial ROS.

The endoplasmic reticulum (ER), the largest cellular organelle composed of a continuous network of tubules and sheets, plays a fundamental role in protein folding, lipid synthesis and Ca2+ storage (15,16). Maintenance of ER proteostasis is vital for secretory function, whereas prolonged ER stress drives cellular dysfunction and contributes to the pathogenesis of CVD (17). Research highlights dynamic crosstalk between the ER and mitochondria as a critical regulator of cellular homeostasis (18). First described by Wilhelm Bernhard in 1956 as the connection between the ER and mitochondria (19), MAMs were later isolated as unique subcellular structures using subcellular isolation techniques and density-gradient centrifugation (20). Disruption of MAM integrity impairs ER-mitochondria communication, leading to homeostatic imbalance and contributing to various diseases, including cancer, neurological disorders and CVDs (21). The present review summarized recent advances in MAM-mediated regulation of mitochondrial quality control and Ca2+ signaling, offering valuable insights for future investigations into the molecular mechanisms with particular emphasis on potential therapeutic strategies.

Mitochondria and MAMs in cardiac microvascular I/R injury

Mitochondria are central to cellular bioenergetics and are traditionally described as the powerhouse of the cell, producing ATP through oxidative phosphorylation driven by the ETC and oxygen reduction (22). As well as ATP generation, mitochondria regulate apoptosis, Ca2+ homeostasis, inflammation and immune responses (23). In microvascular endothelial cells, which primarily depend on glycolysis for energy, mitochondria exhibit a punctate distribution pattern (24). Mitochondria consist of a double membrane defining four compartments: the outer mitochondrial membrane (OMM), the intermembrane space (IMS), the inner mitochondrial membrane (IMM) and the matrix. The OMM contains porins that mediate exchange with other organelles (25), the IMS stores key apoptotic factors (26) and the IMM, with its highly invaginated cristae, harbors the ETC and ATP synthase, enabling efficient ATP synthesis (27). The IMM is characterized by its low permeability and high cardiolipin content (28).

MAMs are dynamic contact sites where the OMM and ER membranes are closely apposed but remain distinct (29). The distance between the two membranes ranges from ~10 nm up to 80-100 nm (29), as identified by electron microscopy, with smooth ER-mitochondria contacts typically closer (10-15 nm) than rough ER contacts (20-30 nm) (30), suggesting that gap width influences the degree of functional coupling (31). Based on the extent of ER-mitochondria membrane contact, MAMs are categorized into three forms: Type I (partial wrapping, ~10% OMM coverage), Type II (extensive contact, ≤50% of the mitochondrial surface) and Type III (complete encapsulation) (32). Type I predominates in most cells, encompassing 10-15% of the OMM (31).

Studies underscore the therapeutic potential of targeting MAMs to mitigate hypoxia-induced endothelial injury (33). Hypoxia directly damages endothelial mitochondria, characterized by elevated mitochondrial ROS (mROS), impaired mitophagy, reduced mitochondrial biogenesis and mitochondrial cristae disruption (34-36). Regulatory proteins and non-coding (nc)RNAs play key roles in this context. MARCH5, a key regulator of mitochondrial dynamics, apoptosis and mitophagy, protects endothelial cells against hypoxia-induced injury via Akt/eNOS signaling (37), whereas RIPK3 exacerbates damage by upregulating 1,4,5-trisphosphate receptors (IP3R) expression, leading to Ca2+ overload and oxidative damage (38). The long (l)ncRNA Malat1 preserves microvascular function after myocardial infarction by regulating mitochondrial dynamics through the miR-26b-5p/mitofusin-1 (MFN1) axis (39). Similarly, the compound FL3 promotes MFN1-dependent mitochondrial fusion, stabilizes Ca2+ homeostasis and alleviates myocardial injury (40). These findings demonstrate that endothelial dysfunction and mitochondrial stress are closely associated with the pathogenesis of microvascular I/R injury. Targeted modulation of MAMs, therefore, represents a promising therapeutic strategy for protecting endothelial integrity and preserving cardiac function (41-43).

Mitochondrial dynamics and mitochondria-associated membranes

Mitochondrial dynamics and MAMs are key factors in the pathogenesis of CVDs

As a crucial component of the mitochondrial quality control (MQC) system, mitochondrial dynamics facilitates the selective removal of damaged organelles and renewal of functional ones (44). Mitochondria continuously undergo fission, fusion and degradation to preserve endothelial homeostasis in response to various cellular cues (45,46). The balance between these fusion and fission processes determines mitochondrial morphology and functionality (47-49). Enhanced fusion or reduced fission promotes the formation of an elongated mitochondrial network, whereas excessive fission or impaired fusion leads to mitochondrial fragmentation (50-52).

MAMs serve as a vital regulatory hub that coordinates mitochondrial by recruiting specific signaling molecules and effector proteins, ensuring precise spatiotemporal regulation of mitochondrial fission and fusion (53,54). Quantitative analyses indicates that >80% of fission and ~60% of fusion events occur at MAMs (55). These findings underscore their role as central hubs that coordinate mitochondrial dynamics and preserve endothelial homeostasis, particularly under stress conditions.

Mitochondrial fission in cardiac microvascular ischemia/reperfusion injury. Mitochondrial fission is markedly upregulated during myocardial I/R injury (56). This process is primarily initiated at MAMs, where the ER outlines the future division site even before dynamin-related protein 1 (Drp1) recruitment (57). Drp1 subsequently accumulates at ER-mitochondria contact sites, where its fission activity is regulated by post-translational modifications (PTMs) such as phosphorylation, ubiquitination and acetylation. These PTMs are mediated by various enzymes, including cyclin-dependent kinases (CDKs) and protein kinase A (PKA) (58-60). For example, phosphorylation of Drp1 at Ser616 promotes its oligomerization and assembly into constrictive rings on the OMM (61), whereas phosphorylation at Ser637 prevents Drp1 translocation to mitochondria (62). Experimental studies have shown that inhibiting Drp1 improves microvascular endothelial function, reduces mROS and attenuates mitochondrial fission (61).

Special attention should be given to the role of mitochondrial fission proteins within the MAM-associated signaling network. During the fission process, Drp1 is actively recruited to the OMM (63). Drp1 then binds to receptors mitochondrial dynamics protein of 49 kDa (MiD49), mitochondrial dynamics protein of 51 kDa (MiD51), mitochondrial fission factor (MFF) and mitochondrial fission protein 1 (Fis1), constricting the mitochondrial membrane in a GTP-dependent manner, thereby dividing a single mitochondrion into two separate organelles (64,65). Immunofluorescence analysis of Drp1 has shown that Fis1 and MFF are key determinants of both the number and size of Drp1 puncta on mitochondria. Either MiD49 or MiD51 can independently mediate Drp1 recruitment and promote mitochondrial fission even in the absence of Fis1 and MFF (66). Modulation of mitochondrial fission protein expression markedly influences mitochondrial morphology: Suppression of MFF disrupts Drp1 foci from the OMM, leading to elongation of the mitochondrial network, whereas MFF overexpression facilitates Drp1 recruitment and promotes mitochondrial fission (67). MFF and Drp1 interact both in vitro and in vivo and MFF-dependent mitochondrial fission occurs independently of Fis1. In endothelial cells, MFF serves as the primary principal adaptor for Drp1 recruitment (67).

FUN14 domain-containing protein 1 (FUNDC1), a recently identified mitochondrial outer membrane protein, localizes to mitochondria-ER contact sites by interacting with the ER membrane protein calnexin under hypoxic conditions (68). During hypoxia, sustained mitophagy disrupts the FUNDC1-calnexin interaction, exposing the cytoplasmic loop of FUNDC1, which subsequently binds to Drp1 and initiates mitochondrial fission (68). Furthermore, at mitochondria-ER contact sites, inverted formin-2 (INF2) becomes activated and promotes actin polymerization (69). Actin filament formation between the ER and mitochondria likely generates mechanical force that facilitates mitochondrial constriction and enhance Drp1 assembly (70). Following fission, the severing and depolymerizing activity of INF2 enables the rapid clearance of actin filaments.

MFF expression is markedly increased following microvascular I/R injury (71) and transcriptional and post-translational mechanisms control both its abundance and activity. Under I/R conditions, activation of the JNK pathway downregulates the dual-specificity phosphatase 1 (DUSP1) (72), thereby enhancing the fission-promoting capacity of MFF. Moreover, I/R upregulates the nuclear receptor 4A1 (NR4A1), which facilitates casein kinase 2α (CK2α)-mediated phosphorylation of MFF, further increasing its fission-inducing activity and exacerbating mitochondrial dysfunction and endothelial cell death (73). A summary of some proteins that mediate MAM structure and mitochondrial quality control is presented in Table I.

Table I

Proteins reported to directly mediate and/or regulate MAM structure and their functions.

Table I

Proteins reported to directly mediate and/or regulate MAM structure and their functions.

RoleProteinFunctions and mechanisms(Refs.)
Mitochondrial fissionDrp 1Phosphorylates at Ser616 and dephosphorylated at Ser637 and forms a potential constriction ring.(61,62)
Interacts with receptors MFF, MiD49/51, and Fis 1 and mediates GTPase-dependent mitochondrial division.(65,66)
FUNDC1Promotes mitochondrial fission through direct interaction with Drp1.(69)
INF2INF2 becomes activated and promotes actin polymerization. Actin filament formation within the MAM likely generates mechanical force that facilitates initial mitochondrial constriction and enhances Drp1 assembly.(70,71)
MFFRecruits Drp1 to mediate mitochondrial fission.(68)
Under I/R conditions, downregulation of DUSP1, improves the fission-promoting capacity of MFF.(73)
I/R upregulates the nuclear receptor NR4A1, which facilitates CK2α-mediated phosphorylation of MFF, further increasing its fission-inducing activity.(74)
Mitochondrial fusionMFN2MFN2 not only mediates OMM fusion but also regulates mitochondria-ER tethering at MAMs.(90,91)
During I/R, knockout of MFN2 limits mPTP opening and increases cardiomyocytes' tolerance to the Ca2+-induced injury.(107)
In other cell types or under pathological stress, silencing or deletion of MFN2 promotes Ca2+ transport into mitochondria, and increases susceptibility to Ca2+-induced cell death.(93)
OPA1OPA1 remodels the IMM, restoring membrane continuity and structural integrity.(89)
Mitochondrial oxidative stressNLRP3MAMs serve as scaffolds that facilitate NLRP3 inflammasome assembly by enabling the interaction between NLRP3 and ASC.(130)
Ero1Ero1 facilitates Ca2+ influx by activating the MCU.(139)
Ero1-PERK complex coordinates mitochondrial fusion, enhances ER-mitochondria interactions, restores mitochondrial bioenergetics, and modulates intracellular ROS levels.(140)
P66ShcUnder oxidative stress, activated PKCβ phosphorylates p66Shc at Ser36, triggering its translocation to mitochondria or the MAM, where it contributes to ROS generation.(137,145)
MitophagyATG5Regulates the formation of autophagosomes and acts as an autophagy marker.(163)
ATG14Interacts with STX17 and localizes to MAMs to facilitate autophagosome formation.(165)
mTORC2Enriched at MAMs, plays a crucial role for autophagy regulation.(164)
PINK1Recruits Parkin, and initiates ubiquitination of OMM proteins such as MFN2 and VDAC, thereby targeting mitochondria for degradation.(167)
Upon mitophagic activation, PINK1 translocates to the MAM and promotes the generation of PI3P. PI3P serves as a critical signal for autophagosome initiation.(168)
Gp78A MAM-resident ubiquitin ligase, also contributes to Parkin-independent mitophagy.(169)
Beclin1Increases autophagic flux and regulates ER-mitochondria interactions.(171)
The serine residue at position 15 of Beclin1 can be phosphorylated by ULK1 and this modification is essential for Beclin1's association with MAMs during the mitophagic process.(172)
FUNDC1Links Drp1-driven fission with mitophagy at the MAMs interface.(69)
Facilitates the removal of damaged mitochondria by interacting with LC3.(176)
FUNDC1 activates the UPRmt to preserve mitochondrial quality control during reperfusion injury.(180)

[i] MAM, mitochondria-associated endoplasmic reticulum membrane; I/R, ischemia-reperfusion; Drp1, dynamin-related protein 1; MFF, mitochondrial fission factor; MiD49, mitochondrial dynamics protein of 49 kDa; MiD51, mitochondrial dynamics protein of 51 kDa; Fis 1, mitochondrial fission 1 protein; FUNDC1, FUN14 domain-containing protein 1; INF2, inverted formin-2; DUSP1, dual-specificity protein phosphatase 1; NR4A1, nuclear receptor 4A1; MFN2, mitofusin-2; OMM, outer mitochondrial membrane; OPA1, optic atrophy 1; NLRP3, pyrin domain-containing receptor 3; ASC, apoptosis-associated speck-like protein containing a CARD; Ero1, endoplasmic reticulum oxidoreductase 1; PERK, PKR-like endoplasmic reticulum kinase; ER, endoplasmic reticulum; ROS, reactive oxygen species; P66Shc, 66-kDa isoform of the growth factor adaptor Shc; PKCβ, protein kinase C beta; ATG5, autophagy-related protein 5; ATG14, autophagy-related protein 14; STX17, syntaxin 17; mTORC2, mechanistic target of rapamycin complex 2; PINK1, PTEN-induced putative kinase 1; PI3P, phosphatidylinositol 3-phosphate; Gp78, glycoprotein 78; ULK1, Unc-51-like autophagy-activating kinase 1; LC3, microtubule-associated proteins 1A/1B light chain 3B; UPRmt, mitochondrial unfolded protein response

MFF-driven excessive fission promotes apoptosis through a number of mechanisms. Pathological fission contributes to mitochondrial outer membrane permeabilization (MOMP) and the subsequent release of cytochrome c into the cytosol (74,75), although whether fission precedes or follows MOMP remains unresolved. Studies suggest three major apoptotic pathways triggered by excessive mitochondrial fission. First, fission induces double-strand breaks in mitochondrial DNA (mtDNA), impairing transcription and replication. mtDNA damage disrupts ETC activity, increases proton leakage and elevates ROS generation (76,77). Second, mROS oxidize cardiolipin, decreasing its binding affinity for cytochrome c and facilitating its release into the cytosol, thereby activating apoptotic signaling (78). Third, mitochondrial fragmentation promotes the polymerization of voltage-dependent anion channel 1 (VDAC1) and displaces hexokinase 2 (HK2) from VDAC1 (79). These changes open the mPTP. The combined effects of oxidative stress, MOMP and mPTP activation exacerbate cardiac microvascular I/R injury, leading to impaired endothelial nitric oxide synthase (eNOS) synthesis, endothelial barrier breakdown, loss of vascular integrity, capillary obstruction and abnormal vascular permeability (80,81). Thus, MFF-mediated mitochondrial fission represents a key pathological mechanism in microvascular I/R injury and represents a potential therapeutic target (78).

In summary, excessive fission serves as a key pathological driver of I/R injury (82). Therapeutic intervention of MAM-associated molecular pathways, therefore, represents a promising strategy for attenuating cardiac microvascular I/R injury by suppressing pathological mitochondrial fission. Several agents have been shown to act through this axis. Shuangshen Ningxin formula, a traditional Chinese medicine compound, alleviates cardiac microvascular I/R injury by improving mitochondrial function through the regulation of the NR4A1/MFF/Drp1 pathway (83). Melatonin exerts protective effects against cardiac microvascular I/R injury by activating AMPKα and promoting inhibitory phosphorylation of Drp1 at Ser637, suppressing fission and enhancing ATP synthase activity. It also attenuates cardiac microvascular injury by targeting the mitochondrial fission-VDAC1-HK2-mPTP-mitophagy cascade (79). Similarly, Bax inhibitor-1 (BI1) preserves mitochondrial integrity under I/R conditions by reducing F-actin-mediated fission, suppressing xanthine oxidase (XO) activity and limiting ROS production, ultimately maintaining endothelial viability and barrier function (84). Furthermore, BI1 is also associated with microvascular protection in I/R injury via repressing Syk-Nox2-Drp1-mitochondrial fission pathways (85). The ROS-JNK-Drp1 pathway has been identified as a primary mechanism driving endothelial cell injury and mitochondrial fission during hypoxia-reoxygenation injury; its inhibition may provide a novel therapeutic strategy to prevent coronary no-reflow injury (86). Empagliflozin protects against microvascular I/R injury by suppressing mitochondrial fission via inactivation of the DNA-PKcs/Fis1 pathway (87). By contrast, pretreatment with 2'-hydroxycinnamaldehyde (HCA) reduces Drp1 expression and improves microvascular function during reperfusion (88).

MAMs exert multifaceted control over mitochondrial fission. Resident proteins and signaling cascades at MAMs facilitate mitochondrial constriction and Drp1 recruitment, promoting fragmentation. Protective mechanisms suppress pro-fission proteins or inhibit associated signaling pathways. These mechanisms collectively reduce mROS, prevent mPTP opening and preserve endothelial cell viability. The MAM-mitochondrial fission axis represents a promising therapeutic target for ameliorating cardiac microvascular I/R injury.

Mitochondrial fusion in cardiac microvascular ischemia/reperfusion injury. Compared with mitochondrial fission, mitochondrial fusion in endothelial cells during myocardial infarction has been less extensively studied. However, mitochondrial fusion is essential for mitochondrial repair and functional recovery, as it facilitates the exchange of matrix and membrane components, preserving mitochondrial integrity and maintaining bioenergetic homeostasis. Mitochondrial fusion involves the coordinated fusion of the OMM and IMM, which is regulated primarily by MFN1/2 and optic atrophy 1 (OPA1), respectively (56,89) (Table I). Mitofusin-2 (MFN2) is particularly important because it not only mediates OMM fusion but also regulates mitochondria-ER tethering at MAMs (90,91). MFN2 plays a crucial role in maintaining normal mitochondrial functions, including fusion, axonal transport, inter-organelle communication and mitosis (92). Unlike MFN1, which is confined to the mitochondrial membrane, MFN2 is distributed across the ER, OMM and MAMs. Homotypic interactions between MFN2 molecules on adjacent membranes, or heterotypic interactions between ER-localized MFN2 and mitochondrial MFN1, form dimeric or multimeric complexes that maintain an optimal distance of 10-30 nm between the two organelles (91,93). Loss or downregulation of MFN2 enhances both structural and functional coupling between the ER and mitochondria. Thus, MFN2 acts as a negative regulator of excessive ER-mitochondria tethering and its reduced expression disrupts this control, promoting aberrant and potentially harmful ER-mitochondria crosstalk (94). OPA1 then remodels the IMM, restoring membrane continuity and structural integrity. This step requires the cardiolipin-binding domain of OPA1.

Fusion of the inner and outer mitochondrial membranes is further regulated by proteolytic processing and ubiquitination (95). At the IMM, long OPA1 (L-OPA1) is cleaved at sites S1 and S2 to form short OPA1 (S-OPA1) (96,97). The AAA+ protease YME1L mediates S2 cleavage under basal conditions, whereas the stress-responsive metalloprotease OMA1 cleaves at S1, thereby disrupting the OPA1 isoform balance (98-100). Under physiological conditions, a near-equivalent ratio of L-OPA1 to S-OPA1 preserves membrane fusion activity (101,102). Pathological stimuli, particularly loss of membrane potential, activate OMA1 (103), shifting the ratio and impairing fusion, leading to fragmentation (104).

MFN1 and MFN2 are not only central to mitochondrial structure but are also closely associated with angiogenic signaling pathways in vascular endothelial cells (105). Loss of MFN1/2 disrupts fusion and mitochondrial networks, leading to reduced membrane potential, impaired endothelial cell survival and reduced responsiveness to VEGF signaling, which then suppresses angiogenesis. Fusion is also essential for maintaining protein synthesis and energy metabolism, as well as preventing mitochondrial DNA loss (106). Despite these protective roles, MFN2 exerts context-dependent regulatory effects on cellular physiological and pathological processes. In cardiomyocytes, MFN2 depletion limits mPTP opening and increases tolerance to Ca2+-induced injury, improving resistance to ischemia during I/R (107). In other cell types or under pathological stress, silencing or deletion of MFN2 enhances mitochondria-ER interactions, promotes Ca2+ transport into mitochondria and increases susceptibility to Ca2+-induced cell death (93). These findings suggest that the function of MFN2 varies according to cell type and disease state. Moreover, MFN2 and OPA1 transcriptional levels are markedly reduced during I/R injury, consistent with functional inhibition of mitochondrial fusion. Regulation of mitochondrial function can mitigate hypoxic-stress-induced apoptosis (108). Overexpression of the sarcoplasmic reticulum Ca2+ pump sarco/endoplasmic reticulum Ca2+ATPase (SERCA) in CMECs reverses MFN2/OPA1 downregulation, increases mitochondrial fusion, reduces fragmentation and improves microvascular integrity by restoring Ca2+ homeostasis (109).

ER stress and impaired intercellular connections are closely associated with mitochondrial fusion-related disorders. This pathogenic cascade is characterized by the loss of cardiac-specific Lon peptidase 1 (LonP1), disruption of the MAM structure, defective mitochondrial fusion and activation of the ER unfolded protein response (UPRER) (110). These interrelated changes contribute to metabolic reprogramming and cardiac structural remodeling. Fusion dysfunction also activates the nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing receptor 3 (NLRP3) inflammasome, triggering vascular inflammatory responses (111). During the early stages of I/R, impaired fusion leads to interstitial edema and endothelial cell swelling, further reducing microcirculatory perfusion (112). In female mouse models, I/R injury disrupts endothelial connections, decreases connexin 43 expression and alters the balance of MMP-3 and TIMP-1 (113,114). Restoration of MFN2 expression reverses these changes and increases endothelial progenitor cell marker expression, including platelet endothelial cell adhesion molecule 1 (CD31), vascular endothelial growth factor receptor 2 (VEGFR2), Fms-related tyrosine kinase 4 (FLT4) and Kinase Insert Domain Receptor (KDR) (114). Loss of cell junctions compromises endothelial barrier function, increasing susceptibility to platelet and coagulation system activation and exacerbating microvascular thrombosis (115). This mechanism impairs myocardial perfusion during reperfusion by promoting microvascular obstruction. Downregulation of MFN2 reduces mitochondrial Ca2+ overload by decreasing CypD-VDAC1-IP3R1 interactions. However, MFN2 broadly supports endothelial integrity, microvascular structure and maintenance of fusion (116). These results suggest that MFN2 exerts a dual role in cardiac microvascular I/R injury by regulating endothelial cell metabolism, apoptosis and inflammatory responses while preserving MAM homeostasis.

Most of these findings are derived from loss-of-function studies using RNA interference to inhibit MFN1/2 or OPA1 expression in endothelial cells in vitro (51,117). However, the protective effects of MFN1/2- or OPA1-mediated fusion have not been conclusively validated in rescue experiments using transgenic mouse models or virus-mediated overexpression. Moreover, interactions between mitochondrial fission and fusion in cardiac microvascular I/R injury remain poorly defined. For example, whether active induction of fusion can directly inhibit fission is unclear. Research suggests that the MFN2/Drp1 expression ratio may reflect the dynamic balance between fusion and fission (118), providing a potential avenue for further investigation; however, its clinical applicability has yet to be established.

Mitochondrial oxidative stress and mitochondria-associated membranes

Oxygen is transported from the bloodstream through endothelial cells to the surrounding perivascular tissues. Research indicates that endothelial dysfunction can impair this process by reducing the rate of oxygen diffusion across the arteriolar wall (119). Under hypoxic conditions, impaired respiration promotes excessive production of mROS (120). The mROS are primarily generated within the ETC of the IMM during oxidative phosphorylation (121,122). While moderate ROS levels act as second messengers in physiological signaling, excessive ROS induces oxidative stress, causing endothelial senescence and cell death. Ischemia- or inflammation-induced oxidative stress activates mitochondria-dependent apoptotic pathways, leading to further cell death (123,124). I/R injury is particularly associated with ROS accumulation. Activation of XO during reperfusion markedly increases ROS, reduces nitric oxide (NO) bioavailability and triggers microvascular spasm and impaired perfusion (125). Furthermore, mitochondrial superoxide oxidizes tetrahydrobiopterin (BH4) to dihydrobiopterin (BH2), preventing BH4 binding to eNOS. This uncoupling of eNOS decreases NO production and exacerbates vascular dysfunction (126).

Protective mechanisms also regulate oxidative stress. MAMs are implicated in multiple cardiovascular disorders, mainly by promoting oxidative stress and inflammation in cardiac tissue (127). They represent the only known binding platform for the NLRP3 inflammasome complex, contributing markedly to ROS-mediated oxidative injury (128). The NLRP3 inflammasome is composed of NLRP3, apoptosis-associated speck-like protein (ASC) and caspase-1 (129). NLRP3 is localized to the ER, whereas ASC is anchored on the OMM. MAMs serve as scaffolds that facilitate NLRP3 inflammasome assembly by enabling the interaction between NLRP3 and ASC (130). mROS are essential regulators of NLRP3 activation. They promote dissociation of thioredoxin-interacting protein (TXNIP) from thioredoxin (TRX), after which TXNIP binds to NLRP3, initiating caspase-1 activation and cleavage of pro-IL-1β and pro-IL-18 (131). This cascade drives robust inflammatory responses. In CMECs, TXNIP-dependent NLRP3 activation has been identified as a novel mechanism of injury during myocardial I/R (129). The extent of myocardial infarction and subsequent functional recovery is strongly affected by the intensity of this inflammatory response (132). Thus, NLRP3 activation serves as a critical mechanistic link between mitochondrial dysfunction and inflammation, playing a key role in microvascular dysfunction following I/R injury (128). Experimental studies demonstrate that early inhibition of NLRP3 inflammasome activity during reperfusion improves cardiac function and reduces infarct size, highlighting it as a promising therapeutic target (133).

Elevated ROS levels at MAMs can trigger the release of mtDNA and the opening of the mPTP. Within MAMs, mtDNA functions as a damage-associated molecular pattern, initiating NLRP3 inflammasome activation and downstream inflammatory signaling (134). During cardiac ischemia-reperfusion, necrotic cardiomyocytes release ATP, which binds to P2X7 receptors on neighboring non-ischemic cardiomyocytes. This interaction induces potassium ion (K+) efflux, lowering cytoplasmic K+ concentration. The resulting hypokalemic state activates NIMA-related kinase 7 (NEK7), promoting inflammasome assembly and NLRP3 activation (135). Moreover, decreased extracellular H+ concentration increases Na+/H+ and Na+/Ca2+ exchange, aggravating intracellular Ca2+ overload (135). This increases mitochondrial Ca2+ accumulation within MAMs and further stimulates ROS production. Excess ROS induces cardiolipin release from the IMM, enabling cardiolipin to bind NLRP3 and promote inflammasome formation. Inhibition of two voltage-dependent anion channel (VDAC1 and VDAC2) isoforms in the OMM markedly reduces NLRP3 inflammasome activation, caspase-1 cleavage and IL-1β production (127). These findings suggest potential therapeutic targets for reducing inflammation and improving heart function following myocardial infarction.

The MAM serves as an important site for ROS generation. Structural proteins located at the MAM, such as endoplasmic reticulum oxidoreductase 1 (Ero1) (136) and 66-kDa isoform of the growth factor adaptor Shc (p66Shc) (137), play pivotal roles in redox signaling between mitochondria and the ER, thereby regulating ROS production (Table I). Ero1α, localized at the MAM (138), facilitates Ca2+ influx by activating the mitochondrial calcium uniporter (MCU) (139). During early stages of ER stress, Ero1α interacts with PKR-like endoplasmic reticulum kinase (PERK), a PKR-like ER kinase, forming the Ero1-PERK complex that coordinates mitochondrial fusion, enhances ER-mitochondria interactions, restores mitochondrial bioenergetics and modulates intracellular ROS levels (140). PERK functions as a MAM-anchoring protein, promoting the formation of MAMs through oligomerization (141) and transmitting ROS signals to mitochondria (142). Furthermore, the Ero1-PERK complex reduces ER Ca2+ levels and promotes ER-to-mitochondria Ca2+ flux (143). Conversely, the absence of PERK impairs Ero1α-mediated IP3R oxidation, thereby disturbing mitochondrial redox homeostasis (144). Similarly, under oxidative stress, activated protein kinase C β (PKCβ) phosphorylates p66Shc at Ser36, triggering its translocation to mitochondria or the MAMs, where it contributes to ROS generation (137,145). Although ROS derived from p66Shc may support short-term cellular repair responses (146), persistent activation can lead to the onset and progression of various cardiovascular diseases (147). NADPH oxidase (Nox), particularly the Nox1, Nox2 and Nox4 isoforms (148), is a major contributor to ROS production in myocardial I/R injury. Nox4 localizes to MAMs, where its expression increases under stress conditions. It functions as an endoplasmic reticulum-localized source of ROS, maintaining basal IP3R oxidation necessary for OXPHOS and regulating redox signaling within the ER under stress conditions (149). At these sites, Nox4 facilitates Akt-mediated phosphorylation of IP3R, which inhibits Ca2+ transfer to mitochondria and prevents mPTP-dependent cell death. This localized redox signaling underlies the protective role of Nox4 in reducing necrosis in both cardiomyocytes and intact hearts following I/R injury (149) (Fig. 2).

MAMs mediate the regulation of
oxidative stress during the pathological process of cardiac
microvascular I/R injury. Following cardiac microvascular ischemia,
excessive Ca2+ transfer from the ER to the mitochondria
induces calcium overload, leading to elevated mROS production.
Excessive ROS stimulation further releases apoptosis-related
proteins into the cytoplasm and activates the apoptotic cascade.
During cardiac ischemia-reperfusion, necrotic cardiomyocytes
release ATP, which binds to P2X7 receptors on neighboring
non-ischemic cardiomyocytes. This interaction induces potassium ion
(K+) efflux, lowering cytoplasmic K+
concentration. The resulting hypokalemic state activates NEK7,
promoting inflammasome assembly and NLRP3 activation. Nox4 is
increased at the MAM during cellular stress, where it encourages
Akt-mediated phosphorylation of IP3R to prevent calcium
transfer and subsequent mPTP-dependent cell death. Furthermore,
MAMs are currently the only known assembly platform for the NLRP3
inflammasome and are important mediators of oxidative damage.
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. MAM, Mitochondria-associated
endoplasmic reticulum membrane; I/R, ischemia-reperfusion; ER mROS,
mitochondrial ROS; ROS, reactive oxygen species; NEK7, NIMA-related
kinase 7; mtDNA; mitochondrial DNA; IP3R,
1,4,5-trisphosphate receptors; NLRP3, pyrin domain-containing
receptor 3; mPTP, mitochondrial permeability transition pore; Ero1,
endoplasmic reticulum oxidoreductase 1; p66Shc, 66-kDa isoform of
the growth factor adaptor Shc.

Figure 2

MAMs mediate the regulation of oxidative stress during the pathological process of cardiac microvascular I/R injury. Following cardiac microvascular ischemia, excessive Ca2+ transfer from the ER to the mitochondria induces calcium overload, leading to elevated mROS production. Excessive ROS stimulation further releases apoptosis-related proteins into the cytoplasm and activates the apoptotic cascade. During cardiac ischemia-reperfusion, necrotic cardiomyocytes release ATP, which binds to P2X7 receptors on neighboring non-ischemic cardiomyocytes. This interaction induces potassium ion (K+) efflux, lowering cytoplasmic K+ concentration. The resulting hypokalemic state activates NEK7, promoting inflammasome assembly and NLRP3 activation. Nox4 is increased at the MAM during cellular stress, where it encourages Akt-mediated phosphorylation of IP3R to prevent calcium transfer and subsequent mPTP-dependent cell death. Furthermore, MAMs are currently the only known assembly platform for the NLRP3 inflammasome and are important mediators of oxidative damage. 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. MAM, Mitochondria-associated endoplasmic reticulum membrane; I/R, ischemia-reperfusion; ER mROS, mitochondrial ROS; ROS, reactive oxygen species; NEK7, NIMA-related kinase 7; mtDNA; mitochondrial DNA; IP3R, 1,4,5-trisphosphate receptors; NLRP3, pyrin domain-containing receptor 3; mPTP, mitochondrial permeability transition pore; Ero1, endoplasmic reticulum oxidoreductase 1; p66Shc, 66-kDa isoform of the growth factor adaptor Shc.

A number of therapeutic strategies have been reported to alleviate oxidative stress and protect cardiac function. ENPP2 regulates redox balance and mitochondrial activity, reducing hypoxia/reoxygenation injury in CMECs (150). Liproxstatin-1, a ferroptosis inhibitor, protects the myocardium by downregulating VDAC1 and restoring GPX4 (151). In a pressure-overload mouse model, ginkgolide A (GA) reduced oxidative stress and increased NO bioavailability in the heart (152). Glucagon-like peptide-1 (GLP-1) analogs, such as liraglutide, have demonstrated cardioprotective effects in clinical trials by lowering oxidative stress and vascular inflammation, preventing eNOS uncoupling and preserving endothelial function (153). Histone deacetylase 7-derived peptides promote angiogenesis and limit oxidative stress in hind limb ischemia, preserving endothelial integrity (154). Naringin (Nar) improves cardiac microvascular function, potentially by facilitating NADH ubiquinone oxidoreductase core subunit S1 (NDUFS1) translocation to mitochondria, reducing ROS and protecting endothelial cells (155). Furthermore, PSS-carrying nanoparticles minimize oxidative stress and reverse coronary microcirculatory dysfunction (156).

Regulation of ROS production and mitigation of oxidative damage constitute the primary mechanisms by which MAMs modulate cellular functions during oxidative stress responses (157). Importantly, MAM-induced mitochondrial oxidative stress does not occur in isolation; elevated ROS levels drive a shift in mitochondrial dynamics from fusion toward fission. These fragmented mitochondria become major sources of apoptotic signals and excessive mROS generation (158). Excessive ROS stimulation further amplifies mitochondrial dysfunction, leading to swelling, loss of cristae structure and rupture of the outer membrane, ultimately releasing apoptosis-related proteins into the cytoplasm and activating the apoptotic cascade (159). Further research on how MAM-inflammasome crosstalk contributes to oxidative stress is essential for elucidating the mechanisms underlying cardiac microvascular I/R injury and developing effective therapeutic strategies.

Mitophagy and mitochondria-associated membranes

Mitophagy, the selective autophagic degradation of damaged mitochondria, is essential for maintaining cellular homeostasis and regulating energy metabolism. It has emerged as a potential therapeutic target for limiting I/R injury (160) and protecting the microvasculature (48). MAMs play a central role in this process by functioning as specialized subdomains where both initiation and progression of mitophagy occur. The pathway proceeds through different stages: initiation, phagophore elongation, autophagosome closure, lysosomal fusion and degradation and is regulated by a set of autophagy-related genes and proteins (161). Mitophagy signaling depends on receptor proteins at the OMM, including FUNDC1, BCL2/adenovirus E1B 19 kDa interacting protein 3 (BNIP3), Nip3-like protein X (Nix) and mitochondrial E3 ubiquitin protein ligase 1 (Mul1), as well as the E3 ubiquitin ligase Parkin (162). Upstream regulation is closely associated with mitochondrial dynamics. Mitochondrial fission, often a prerequisite for mitophagy, is regulated by proteins such as MFF, whose activity is influenced by DUSP1 (72) and NR4A1 (73). In CMECs, NR4A1 inhibits mitophagy by suppressing FUNDC1 (73), whereas DUSP1 promotes it through BNIP3 phosphorylation (Fig. 3).

During I/R injury, DUSP1 and NR4A1
play distinct roles in the activation of mitophagy: DUSP1 induces
mitophagy by promoting BNIP3 phosphorylation, whereas NR4A1
suppresses mitophagy through inhibition of FUNDC1 activity. These
alterations contribute to mitochondrial energy disorder, ultimately
leading to endothelial cell apoptosis and microvascular
dysfunction. I/R, ischemia-reperfusion; DUSP1, dual-specificity
phosphatase 1; NR4A1, nuclear receptor 4A1; FUNDC1, FUN14
domain-containing protein 1; BNIP3, BCL2/adenovirus E1B 19 kDa
interacting protein 3.

Figure 3

During I/R injury, DUSP1 and NR4A1 play distinct roles in the activation of mitophagy: DUSP1 induces mitophagy by promoting BNIP3 phosphorylation, whereas NR4A1 suppresses mitophagy through inhibition of FUNDC1 activity. These alterations contribute to mitochondrial energy disorder, ultimately leading to endothelial cell apoptosis and microvascular dysfunction. I/R, ischemia-reperfusion; DUSP1, dual-specificity phosphatase 1; NR4A1, nuclear receptor 4A1; FUNDC1, FUN14 domain-containing protein 1; BNIP3, BCL2/adenovirus E1B 19 kDa interacting protein 3.

Numerous key autophagic proteins, including ATG5/14 (163) and mTORC2 (164), are enriched at MAMs, highlighting the crucial role of these contact sites as central hubs for autophagy regulation (Table I). The autophagosome marker ATG14 interacts with STX17 and localizes to MAMs to facilitate autophagosome formation (165). Similarly, PTEN-induced putative kinase 1 (PINK1)/Parkin and Beclin-1 are concentrated at these sites (166), promoting the formation of autophagosomes and aiding in the identification and removal of damaged mitochondria (166). In the PINK1/Parkin pathway, PINK1 accumulates on damaged mitochondria, where it recruits Parkin and initiates the ubiquitination of OMM proteins such as MFN2 and VDAC. This process triggers p97-dependent disassembly of MFN2 complexes from the OMM, thereby marking the damaged mitochondria for degradation (167). Upon mitophagic activation, PINK1 translocates to the MAM and promotes the generation of phosphatidylinositol 3-phosphate (PI3P) (168) by recruiting Beclin1, a key component of the class III PI3-kinase complex. PI3P, a lipid signaling molecule primarily enriched in the ER but also present at MAMs, serves as a critical signal for autophagosome initiation. Gp78, a MAM-resident ubiquitin ligase, also contributes to Parkin-independent mitophagy (169). Mitophagy mediated by the PINK1/Parkin signaling pathway plays a pivotal role in protecting against myocardial injury induced by I/R (170) (Fig. 4).

MAMs play a critical role in
regulating mitophagy during microvascular I/R injury. The mechanism
targets of autophagosome markers (ATG) 5/14 and mTOR2 are key
inducers of autophagy. For example, STX 17, located on the
autophagosome outer membrane can bind to ATG14 and transfer it to
MAM until autophagosome is complete. Under ischemic and hypoxic
conditions, PINK1 accumulates on the damaged OMM and recruits
Parkin from the cytosol, which then ubiquitinates OMM proteins such
as MFN2 and VDAC to promote mitophagy. As a mitochondrial membrane
protein, FUNDC1 interacts with LC3 during hypoxic stress to
facilitate mitochondrial mitophagy. It also recruits Drp1 to MAMs
and stimulates the elimination of defective mitochondria. In
addition, Beclin1, an upstream regulator of mitophagy, protects the
microvascular against I/R injury. MAM, mitochondria-associated
endoplasmic reticulum membrane; I/R, ischemia-reperfusion; STX 17,
syntaxin 17; ATG14, autophagy related 14; OMM, outer mitochondrial
membrane; MFN2, mitofusin-2; VDAC, voltage-dependent anion channel;
Drp1, dynamin-related protein 1

Figure 4

MAMs play a critical role in regulating mitophagy during microvascular I/R injury. The mechanism targets of autophagosome markers (ATG) 5/14 and mTOR2 are key inducers of autophagy. For example, STX 17, located on the autophagosome outer membrane can bind to ATG14 and transfer it to MAM until autophagosome is complete. Under ischemic and hypoxic conditions, PINK1 accumulates on the damaged OMM and recruits Parkin from the cytosol, which then ubiquitinates OMM proteins such as MFN2 and VDAC to promote mitophagy. As a mitochondrial membrane protein, FUNDC1 interacts with LC3 during hypoxic stress to facilitate mitochondrial mitophagy. It also recruits Drp1 to MAMs and stimulates the elimination of defective mitochondria. In addition, Beclin1, an upstream regulator of mitophagy, protects the microvascular against I/R injury. MAM, mitochondria-associated endoplasmic reticulum membrane; I/R, ischemia-reperfusion; STX 17, syntaxin 17; ATG14, autophagy related 14; OMM, outer mitochondrial membrane; MFN2, mitofusin-2; VDAC, voltage-dependent anion channel; Drp1, dynamin-related protein 1

Beclin1, which is also enriched at MAMs, enhances autophagic flux and regulates ER-mitochondria interactions, functioning as a protective factor against I/R injury (171). The serine residue at position 15 of Beclin1 can be phosphorylated by Unc-51-like autophagy-activating kinase 1 (ULK1) (172), a kinase involved in mitophagy (173) and this modification is essential for Beclin1's association with MAMs during the mitophagic process. Localization of Beclin1 at MAMs ensures that autophagosome formation occurs in proximity to damaged mitochondria, facilitating their efficient sequestration and degradation (171). Furthermore, studies have shown that Beclin1-driven autophagy suppresses caspase-4-mediated apoptosis, thereby protecting microvascular endothelial cells from I/R-induced injury (174).

During reperfusion, BNIP3 levels increase and FUNDC1 expression decreases (175). This is paradoxical, as FUNDC1 is recognized as a critical mediator of protective mitophagy under hypoxic conditions. FUNDC1 facilitates the removal of damaged mitochondria by interacting with LC3 (176) (Fig. 4). Activation of FUNDC1 is regulated by multiple key post-translational modifications that act cooperatively to modulate its function. These include phosphoglycerate mutase family member 5 family member 5 (PGAM5)-mediated dephosphorylation at Ser13 (177), ULK1-mediated phosphorylation at Ser17 (178) and MARCH5-mediated ubiquitination at Lys119, all of which play crucial roles in controlling mitochondrial dynamics and mitophagy. Under hypoxic conditions, the ER-resident deubiquitinase USP19 accumulates at MAMs, where it interacts and deubiquitinates FUNDC1. This interaction leads to mitochondrial fission and counteracts the MARCH5-FUNDC1 signaling pathway (179). Under normoxic conditions, MARCH5 facilitates the degradation of excess FUNDC1, thereby preventing uncontrolled mitochondrial fission. The resulting fragmented mitochondria subsequently recruit the ULK1 complex to initiate mitophagy (68). Furthermore, FUNDC1 activates the mitochondrial unfolded protein response (UPRmt) to preserve mitochondrial quality control during reperfusion injury (180). However, during reperfusion, increased RIPK3 phosphorylation of FUNDC1, impairs mitophagy and heightens susceptibility to cell death (181). Collectively, these findings underscore the pivotal role of FUNDC1 in the pathogenesis of I/R injury. Its context-dependent function is shown in divergent therapeutic observations. For instance, the AMP-activated protein kinase α1(AMPKα1)/ULK1/FUNDC1 axis is required to maintain endothelial homeostasis during I/R injury and empagliflozin may exert protective effects through this pathway (182). Melatonin protects against cardiac I/R injury by suppressing FUNDC1-dependent mitophagy in platelets, reducing energy production, platelet hyperactivity and thrombotic complications (183). These findings emphasize that FUNDC1 activity is not uniformly beneficial but is shaped by the cellular and pathological context. Pharmacological studies further support the dual role of mitophagy. Shenlian extract attenuates I/R injury by inhibiting the PINK1/Parkin pathway and reducing mitophagy (184), whereas the mitophagy inducer UA improves overall mitochondrial quality control and confers protection (185). These results suggest that therapeutic strategies should not aim to universally enhance or inhibit mitophagy, but rather focus on fine-tuning specific pathways, including those involving FUNDC1.

Crosstalk among various mitophagy adaptors ultimately determines the net effect of mitophagy. Fission of damaged mitochondria into smaller fragments facilitates their elimination through mitophagy (186). Autophagy and fission occur simultaneously, forming a coordinated response (187). Moderate mitophagy limits excessive fragmentation induced by pathological fission (175,188,189). Thus, fission is both a prerequisite for mitophagy activation and, when excessive, a process that can be counterbalanced by appropriately regulated mitophagy. Taken together, these findings underscore that mitophagy exerts both protective and detrimental roles depending on the signaling context. The precise contribution of specific adaptors in endothelial cells and their interactions with fission machinery remain unclear. Therefore, the controlled regulation of mitophagy represents a key area of investigation for mitigating microvascular I/R injury.

Ca2+ overloading and mitochondria-associated membranes

Mitochondrial Ca2+ overload is a major driver of mitochondrial dysfunction, leading to endothelial impairment and cardiomyocyte death and represents a key contributor to I/R injury (190,191). MAMs function as specialized platforms that tightly regulate intracellular Ca2+ homeostasis during I/R (192). For instance, disruption of MAM integrity has been shown to protect endothelial cells against I/R damage (193), underscoring their dual role in both facilitating signaling and promoting injury under pathological conditions. Ca2+ transfer between the ER and mitochondria occurs through a coordinated sequence of steps mediated by MAM-associated complexes. First, Ca2+ is released from the ER into the cytosol via ryanodine receptors (RyR) and inositol IP3R) (194)). Second, Ca2+ crosses the OMM through the VDAC, the primary conduit for ions and metabolites. Finally, Ca2+ enters the mitochondrial matrix through the mitochondrial calcium uniporter (MCU) complex. Excessive MCU activity is a key determinant of Ca2+ overload and subsequent mitochondrial injury (195,196).

The IP3R is a calcium channel located on the ER membrane and is regulated by the MAM-anchoring protein inositol-requiring enzyme 1 (IRE1). The presence of IRE1 at MAMs is critical for IP3R function, as IRE1 acts as a scaffold that directly interacts with IP3R within MAMs to modulate ER-mitochondria Ca2+ transfer and sustain cellular energy homeostasis (197,198). SERCA, the primary ER calcium pump, is equally important for maintaining calcium homeostasis by sequestering Ca2+ into the ER, maintaining low cytoplasmic concentrations. Experimental evidence shows that inhibiting IP3R or increasing SERCA expression protects against I/R-induced microcirculatory dysfunction (199). These protective effects occur through the reduction of mitochondrial calcium overload and prevention of mPTP-mediated necrosis. Pharmacological interventions further support this concept: dapagliflozin (DAPA) alleviates cytosolic Ca2+ overload by preventing oxidation and inactivation of SERCA2. This action appears to involve inhibition of XO activity, limiting oxidative stress and preserving SERCA2 function (200).

VDAC, an outer mitochondrial membrane protein (117), demonstrates high permeability to Ca2+ and mediates its transfer across the IMS, thereby enabling Ca2+ influx into the IMM (201,202). Within the MAM, IP3R and VDAC do not interact directly, but are functionally connected via the glucose-regulated protein 75 (GRP75) linker protein. The transfer of Ca2+ between the ER and mitochondria is mediated by the VDAC1/GRP75/IP3R1 complex (203). Within this assembly, GRP75 functions as a molecular bridge, tethering IP3R1 on the ER to VDAC1 on the OMM. This complex forms local high-Ca2+ microdomains that strongly promote mitochondrial Ca2+ uptake (203). Moreover, Ca2+ transfer depends on the MCU (204), forming a coordinated complex known as the IP3R-Grp75-VDAC-MCU calcium regulatory axis. In this system, VDAC works in concert with the MCU to regulate Ca2+ influx into mitochondria (205). While essential for physiological signaling, excessive activity of this complex contributes to pathology.

Several molecular regulators modulate this process. Glycogen synthase kinase-3β (GSK-3β) and the Sigma-1 receptor (Sig-1R) are key MAM-associated proteins that alleviate ER stress and reduce Ca2+ uptake, conferring cardioprotection during I/R (206,207). GSK-3β, an enzyme responsible for phosphorylating and inactivating glycogen synthase (208,209), can be recruited to the MAM, where it regulates IP3R1-mediated calcium release. This regulation promotes mitochondrial Ca2+ accumulation and induces the opening of the mPTP. As a result, inhibition of GSK-3β has been suggested to exert cardioprotective effects during I/R injury (206). The Sig-1R is a specific chaperone localized at MAMs (207). Overexpression of Sig-1R enhances ER-to-mitochondria Ca2+ flux and through its interactions with ankyrin and the ER chaperone BiP. Under physiological conditions, Sig-1R remains bound to BiP; however, under ER stress or following ER Ca2+depletion, Sig-1R dissociates from BiP and binds to IP3R instead. This interaction stabilizes IP3R, preventing its degradation and restoring efficient Ca2+ transfer from the ER to mitochondria (207). Moreover, Sig-1R stabilizes MAM structure through its interactions with VDAC1 and IP3R and provides cardioprotective benefits by mitigating MAM-mediated Ca2+ overload and ER stress (207). Under conditions of elevated ROS, Sig-1R also interacts with reactive oxygen species, thereby activating and stabilizing IRE1 (210).

Cyclophilin D (CypD), a key regulator of the mPTP located in the matrix. During H/R, increased interactions within the CypD-VDAC1-GRP75-IP3R1 super-complex drive mitochondrial Ca2+ overload and cardiomyocyte death (126). Genetic ablation of CypD protects against I/R-induced necrosis by limiting this aberrant Ca2+ transfer (211,212). Similarly, deletion of the PPIF gene (encoding CypD), or knockdown of IP3R1 or GRP75, reduces H/R-induced Ca2+ overload and cell death (211). Pharmacological interventions also provide protection; melatonin attenuates oxidative stress and reduces CMEC mortality by inhibiting IP3R-VDAC-mediated mitochondrial Ca2+ influx, increasing endothelial resistance to oxidative damage (77).

MAM tethering is also regulated by the OMM protein PTPIP51 and the ER protein vesicle-associated membrane protein-associated protein-B (VAPB), which form a structural bridge between the two organelles (213). Overexpression of PTPIP51 strengthens this physical connection, enhances ER-mitochondria coupling and increases mitochondrial Ca2+ uptake through MCU activation (214,215). However, pharmacological or genetic inhibition of MCU abolishes this effect and protects cardiomyocytes from apoptosis (211). The ER chaperone calreticulin (CRT), with its high Ca2+-binding capacity, also plays a key role in buffering and storage. Upregulation of CRT restores Ca2+ homeostasis by improving ER Ca2+ reserves (216) (Fig. 5). Collectively, MAM-associated proteins regulate calcium transport through distinct and highly coordinated mechanisms (Table II).

MAMs regulate calcium homeostasis
during microvascular I/R. During hypoxia/reoxygenation, an
increased interaction of the CYPD-VDAC1-GRP75-IP3R1
Ca2+ complex leads to mitochondrial Ca2+
overload and cardiomyocyte mortality. The activity of SERCA and
CRT, responsible for Ca2+ reuptake into the endoplasmic
reticulum and Ca2+ storage in the ER, is inhibited. The
complex of VAPB-PTPIP51-MCU mediates the transport of
Ca2+ from the endoplasmic reticulum to mitochondria.
GSK-3β can be recruited to the MAM, where it regulates
IP3R1-mediated calcium release. This regulation promotes
mitochondrial Ca2+ accumulation and induces the opening
of the mPTP. Sig-1R is a specific chaperone localized at MAMs and
is functionally associated with the IP3R-GRP75-VDAC-MCU
calcium signaling axis. Therapeutically, TMEM215 protects
endothelial cells by inhibiting BIK. Similarly, HINT2 protects
CMECs during ischemia by directly inhibiting the MCU complex,
reducing mitochondrial Ca2+ overload and preserving
cardiac function. MAM, mitochondria-associated endoplasmic
reticulum membrane; I/R, ischemia-reperfusion; CYPD, cyclophilin D;
VDAC1, voltage-dependent anion channel 1; GRP75, glucose-regulated
protein 75; IP3R, 1,4,5-trisphosphate receptors; SERCA,
sarco/endoplasmic reticulum Ca2+ ATPase; CRT,
calreticulin; ER, endoplasmic reticulum; VAPB, vesicle-associated
membrane protein-associated protein-B; PTPIP51, protein tyrosine
phosphatase-interacting protein-51; MCU, mitochondrial calcium
uniporter; GSK-3β, glycogen synthase kinase-3β; mPTP, mitochondrial
permeability transition pore; Sig-1R, σ-1 receptor; TMEM215,
transmembrane protein 215; BIK, Bcl-2-interacting killer; HINT2,
histidine triad nucleotide-binding protein 2; CMECs, cardiac
microvascular endothelial cells.

Figure 5

MAMs regulate calcium homeostasis during microvascular I/R. During hypoxia/reoxygenation, an increased interaction of the CYPD-VDAC1-GRP75-IP3R1 Ca2+ complex leads to mitochondrial Ca2+ overload and cardiomyocyte mortality. The activity of SERCA and CRT, responsible for Ca2+ reuptake into the endoplasmic reticulum and Ca2+ storage in the ER, is inhibited. The complex of VAPB-PTPIP51-MCU mediates the transport of Ca2+ from the endoplasmic reticulum to mitochondria. GSK-3β can be recruited to the MAM, where it regulates IP3R1-mediated calcium release. This regulation promotes mitochondrial Ca2+ accumulation and induces the opening of the mPTP. Sig-1R is a specific chaperone localized at MAMs and is functionally associated with the IP3R-GRP75-VDAC-MCU calcium signaling axis. Therapeutically, TMEM215 protects endothelial cells by inhibiting BIK. Similarly, HINT2 protects CMECs during ischemia by directly inhibiting the MCU complex, reducing mitochondrial Ca2+ overload and preserving cardiac function. MAM, mitochondria-associated endoplasmic reticulum membrane; I/R, ischemia-reperfusion; CYPD, cyclophilin D; VDAC1, voltage-dependent anion channel 1; GRP75, glucose-regulated protein 75; IP3R, 1,4,5-trisphosphate receptors; SERCA, sarco/endoplasmic reticulum Ca2+ ATPase; CRT, calreticulin; ER, endoplasmic reticulum; VAPB, vesicle-associated membrane protein-associated protein-B; PTPIP51, protein tyrosine phosphatase-interacting protein-51; MCU, mitochondrial calcium uniporter; GSK-3β, glycogen synthase kinase-3β; mPTP, mitochondrial permeability transition pore; Sig-1R, σ-1 receptor; TMEM215, transmembrane protein 215; BIK, Bcl-2-interacting killer; HINT2, histidine triad nucleotide-binding protein 2; CMECs, cardiac microvascular endothelial cells.

Table II

Proteins in the MAM associated with calcium homeostasis.

Table II

Proteins in the MAM associated with calcium homeostasis.

ProteinFunction(Refs.)
IP3RReleases Ca2+ from the ER into the mitochondria, which in turn affects oxidative metabolism and energy production.(194)
IRE1MAM-anchoring protein IRE1 interacts with IP3R within MAMs to modulate ER-mitochondria Ca2+ transfer.(197,198)
SERCAOverexpression of SERCA in CMECs reverses MFN2/OPA1 downregulation, increases mitochondrial fusion and improves microvascular integrity by restoring Ca2+ homeostasis.(109)
The primary ER calcium pump, is equally important for maintaining calcium homeostasis by sequestering Ca2+ into the ER.(199)
VDACMitochondrial fragmentation promotes the polymerization of VDAC1 and displaces HK2 from VDAC1. These changes result in the opening of the mPTP and exacerbate cardiac microvascular I/R injury.(63)
Inhibition of VDAC1 and VDAC2 in the OMM significantly reduces NLRP3 inflammasome activation, caspase-1 cleavage, and IL-1β production.(127)
Demonstrates high permeability to Ca2+ and mediates its transfer across the IMS, thereby enabling Ca2+ influx into the IMM.(201,202)
MCUCa2+ enters the mitochondrial matrix through the (MCU) complex. Excessive MCU activity is a key determinant of Ca2+ overload and subsequent mitochondrial injury.(195,196)
CypDA key regulator of the mPTP located in the matrix. During H/R, increased interactions within the CypD-VDAC1-GRP75-IP3R1 super-complex drive mitochondrial Ca2+ overload and cardiomyocyte death.(126)
GSK-3βCan be recruited to the MAM, where it regulates IP3R1-mediated calcium release.(208,209)
Sig-1RSig-1R is a specific chaperone localized at MAMs.(207)
Sig-1R stabilizes MAM structure through its interactions with VDAC1 and IP3R. Under conditions of elevated ROS, Sig-1R also interacts with reactive oxygen species, thereby activating and stabilizing IRE1.(210)
VAPBThe OMM protein PTPIP51 and the ER protein VAPB form a structural bridge between the two organelles.
Disruption of this complex promotes mitophagy.
(213)
PTPIP51Overexpression of PTPIP51 strengthens the physical connection between ER and mitochondrial, enhances ER-mitochondria coupling, and increases mitochondrial Ca2+ uptake through MCU activation.(214,215)
CRTPlays a key role in buffering and storage with its high Ca2+-binding capacity.(216)

[i] MAM, mitochondria-associated endoplasmic reticulum membrane; IP3R, inositol 1,4,5-trisphosphate receptor; ER, endoplasmic reticulum; IRE1, inositol-requiring enzyme 1; SERCA, sarco/endoplasmic reticulum Ca2+ ATPase; MFN2, mitofusin-2; OPA1, optic atrophy 1; VDAC1, voltage-dependent anion channel 1; VDAC2, voltage-dependent anion channel 2; HK2, hexokinase 2; NLRP3, pyrin domain-containing receptor 3; caspase-1, cysteinyl aspartate specific proteinase-1; IL-1β, interleukin-1 beta; IMS, intermembrane space; IMM, inner mitochondrial membrane; mPTP, mitochondrial permeability transition pore; MCU, mitochondrial calcium uniporter; CypD, cyclophilin D; GRP75, glucose-regulated protein 75; GSK-3β, glycogen synthase kinase-3β; Sig-1R, σ-1 receptor; VAPB, vesicle-associated membrane protein-associated protein-B; PTPIP51, protein tyrosine phosphatase-interacting protein-51; CRT, calreticulin

Therapeutically, pinacidil has been shown to preserve CRT expression, reduce endothelial Ca2+ overload and prevent mitochondrial apoptosis in CMECs, ultimately improving microvascular density, increasing blood flow, reducing infarct size and attenuating the no-reflow phenomenon following I/R injury (217). Other protective mechanisms involve ER-transmembrane proteins such as transmembrane protein 215 (TMEM215), which protect endothelial cells by inhibiting BCL-2-interacting killer (BIK). Since BIK promotes ER-to-mitochondria Ca2+ transfer, its suppression by TMEM215 reduces pathological Ca2+ flux and supports cell survival during reperfusion (218).

Other targets act more directly on calcium-handling machinery. For example, Li et al (219) demonstrated that histidine triad nucleotide-binding protein 2 (HINT2) protects CMECs during ischemia by directly inhibiting the MCU complex, reducing mitochondrial Ca2+ overload and preserving cardiac function. Regulating MAM-associated molecules plays a critical role in microvascular ischemia/reperfusion injury (Table III). Similarly, acetylcholine has been shown to protect endothelial cells by destabilizing ER-mitochondrial contacts and reducing intracellular and mitochondrial Ca2+ overload (193).

Table III

Role of regulating MAM-related molecules in microvascular I/R injury.

Table III

Role of regulating MAM-related molecules in microvascular I/R injury.

ProteinFunction(Refs.)
MARCH5Protects endothelial cells against hypoxia-induced injury via Akt/eNOS signaling.(37)
RIPK3Exacerbates hypoxia-induced microvascular damage by upregulating IP3R expression, leading to Ca2+ overload and oxidative damage.(38)
Increased RIPK3 phosphorylation of FUNDC1 impairs mitophagy and heightens susceptibility to cell death(181)
Malat1Preserves microvascular function after myocardial infarction by regulating mitochondrial dynamics through the miR-26b-5p/MFN1 axis.(39)
BI1Preserves mitochondrial integrity under I/R conditions by reducing F-actin-mediated fission, suppressing XO activity, and limiting ROS production, ultimately maintaining endothelial viability and barrier function.(84)
BI1 is also associated with microvascular protection in I/R injury viarepressing Syk-Nox2-Drp1-mitochondrial fission pathways.(85)
LonP1Loss of cardiac-specific LonP1 is associated with ER stress and impaired intercellular connections.(110)
MFN2Restores microcirculatory perfusion and cellular junction functions by increasing the endothelial progenitor cell marker expression and maintaining the balance of MMP-3 and TIMP-1.(113,114)
ENPP2Regulates redox balance and mitochondrial activity, reducing hypoxia/reoxygenation injury in CMECs.(150)
TMEM215Protects endothelial cells by inhibiting BIK and reducing pathological Ca2+ flux.(218)
HINT2Protects cardiac function during ischemia by directly inhibiting the MCU complex, reducing mitochondrial Ca2+ overload.(219)

[i] MAM, mitochondria-associated endoplasmic reticulum membrane; I/R, ischemia-reperfusion; MARCH5, mitochondrial E3 ubiquitin ligase; eNOS, endothelial nitric-oxide synthase; IP3R, inositol 1,4,5-trisphosphate receptor; RIPK3, receptor-interacting protein kinase 3; FUNDC1, FUN14 domain-containing protein 1; MFN1, mitofusin-1; BI1, Bax inhibitor-1; Syk, spleen tyrosine kinase; Nox2, NADPH oxidases 2; Drp1, dynamin-related protein 1; XO, Xanthine Oxidase; LonP1, Lon peptidase 1; MFN2, mitofusin-2; MMP-3, matrix metalloproteinase-3; TIMP-1, tissue inhibitor of metalloproteinases 1; ENPP2, ectonucleotide pyrophosphatase/phosphodiesterase 2; TMEM215, transmembrane protein 215; BIK, Bcl-2-interacting killer; HINT2, histidine triad nucleotide-binding protein 2; MCU, mitochondrial calcium uniporter.

Inhibiting ER-to-mitochondria Ca2+ transfer in endothelial cells improves cardiac microcirculation, enhances perfusion, reduces microvascular obstruction and limits inflammatory cell infiltration. An important mechanism underlying this protection is the preservation of mitochondrial morphology and function (198). In conclusion, MAMs represent a critical hub in the pathogenesis of microvascular I/R injury. Dysregulated Ca2+ transport through MAMs not only disrupts mitochondrial homeostasis but also increases ROS production and activates cell death pathways. A deeper understanding of MAM-regulated Ca2+ signaling may provide novel therapeutic opportunities to mitigate cardiac microvascular dysfunction during reperfusion.

Conclusions

Damage to this microvascular network during I/R can result in interstitial hemorrhage and edema (220). The MAM functions as a pivotal signaling hub that orchestrates mitochondrial dynamics, oxidative stress responses, mitophagy and calcium homeostasis. Importantly, these mitochondrial alterations do not occur in isolation but are intricately interconnected (Fig. 6). Core MAM-associated proteins such as Drp1, MFN2, FUNDC1, Ero1α, IP3R, VDAC1 and Sig-1R play essential roles in maintaining normal microvascular physiology. By integrating these key MAM components, molecular mechanisms and signaling pathways, advances in understanding cardiac microvascular I/R injury have provided new perspectives for addressing clinical conditions such as the no-reflow phenomenon and microvascular angina (Fig. 7).

Mitochondrial dysfunction plays a
central role in the apoptosis of endothelial cells during cardiac
microvascular I/R injury. Specifically, ischemia/reperfusion
induces mitochondrial oxidative stress and structural remodeling in
microvascular endothelial cells, characterized by excessive
mitochondrial fission, impaired fusion and overproduction of mROS.
These alterations facilitate the opening of the mPTP and calcium
overload, leading to the release of Cyt-C, the activation of
caspase-9 and caspase-3 and ultimately, endothelial apoptosis and
microvascular damage. Furthermore, potential therapeutic
interventions, such as the Shuangshen Ningxin formula, melatonin,
empagliflozin, Shenlian extract and pinacidil have been shown to
mitigate mROS generation, stabilize mPTP function and restore
mitochondrial dynamics, thereby protecting endothelial cells and
alleviating cardiac microvascular I/R injury. I/R,
ischemia-reperfusion; m/mitoROS, mitochondrial reactive oxygen
species; mPTP, mitochondrial permeability transition pore; Cyt-C,
cytochrome c.

Figure 6

Mitochondrial dysfunction plays a central role in the apoptosis of endothelial cells during cardiac microvascular I/R injury. Specifically, ischemia/reperfusion induces mitochondrial oxidative stress and structural remodeling in microvascular endothelial cells, characterized by excessive mitochondrial fission, impaired fusion and overproduction of mROS. These alterations facilitate the opening of the mPTP and calcium overload, leading to the release of Cyt-C, the activation of caspase-9 and caspase-3 and ultimately, endothelial apoptosis and microvascular damage. Furthermore, potential therapeutic interventions, such as the Shuangshen Ningxin formula, melatonin, empagliflozin, Shenlian extract and pinacidil have been shown to mitigate mROS generation, stabilize mPTP function and restore mitochondrial dynamics, thereby protecting endothelial cells and alleviating cardiac microvascular I/R injury. I/R, ischemia-reperfusion; m/mitoROS, mitochondrial reactive oxygen species; mPTP, mitochondrial permeability transition pore; Cyt-C, cytochrome c.

MAMs play a central role in
maintaining mitochondrial homeostasis during cardiac microvascular
I/R injury. MAMs function as structural and signaling hubs that
coordinate mitochondrial dynamics, oxidative stress, mitophagy and
Ca2+ homeostasis. Specifically, the Drp1 and MFN1/2-OPA1
complexes mediate mitochondrial fission and fusion. MFN1 and MFN2
are not only central to mitochondrial structure but are also
closely associated with angiogenic signaling pathways in vascular
endothelial cells. MAM-localized p66Shc and Ero1α contribute to
reactive oxygen species (ROS) generation and Ca2+
influx, activating NLRP3 inflammasome-mediated inflammatory
cascades. The IP3R-Grp75-VDAC1 complex regulates
Ca2+ transfer between the endoplasmic reticulum and
mitochondria, while Sig-1R and VAPB-PTPIP51 tethers stabilize
inter-organelle communication and prevent Ca2+ overload.
Additionally, MAMs serve as key platforms for PINK1/Parkin- and
LC3-dependent mitophagy, mediated through FUNDC1, NIX and BNIP3.
Dysregulation of these pathways leads to excessive ROS
accumulation, Ca2+ imbalance and mitochondrial
dysfunction, ultimately contributing to endothelial apoptosis and
impaired microvascular repair following cardiac
ischemia-reperfusion (I/R) injury. MAM, mitochondria-associated
endoplasmic reticulum membrane; I/R, ischemia-reperfusion; MFN1,
mitofusin-1; MFN2, mitofusin-2; OPA1, optic atrophy 1.

Figure 7

MAMs play a central role in maintaining mitochondrial homeostasis during cardiac microvascular I/R injury. MAMs function as structural and signaling hubs that coordinate mitochondrial dynamics, oxidative stress, mitophagy and Ca2+ homeostasis. Specifically, the Drp1 and MFN1/2-OPA1 complexes mediate mitochondrial fission and fusion. MFN1 and MFN2 are not only central to mitochondrial structure but are also closely associated with angiogenic signaling pathways in vascular endothelial cells. MAM-localized p66Shc and Ero1α contribute to reactive oxygen species (ROS) generation and Ca2+ influx, activating NLRP3 inflammasome-mediated inflammatory cascades. The IP3R-Grp75-VDAC1 complex regulates Ca2+ transfer between the endoplasmic reticulum and mitochondria, while Sig-1R and VAPB-PTPIP51 tethers stabilize inter-organelle communication and prevent Ca2+ overload. Additionally, MAMs serve as key platforms for PINK1/Parkin- and LC3-dependent mitophagy, mediated through FUNDC1, NIX and BNIP3. Dysregulation of these pathways leads to excessive ROS accumulation, Ca2+ imbalance and mitochondrial dysfunction, ultimately contributing to endothelial apoptosis and impaired microvascular repair following cardiac ischemia-reperfusion (I/R) injury. MAM, mitochondria-associated endoplasmic reticulum membrane; I/R, ischemia-reperfusion; MFN1, mitofusin-1; MFN2, mitofusin-2; OPA1, optic atrophy 1.

Moreover, several pharmacological agents, including empagliflozin, Shuangshen Ningxin Pill and melatonin, have been shown to exert protective effects against microvascular I/R injury through MAM-related mechanisms (Table IV). However, most of the current evidence remains confined to cellular and animal models. Future research, focusing on the safety, controllability and efficacy of MAM-targeted interventions at the human microvascular level may enable the development of more effective therapeutic strategies.

Table IV

Role of MAM-targeted drugs in cardiac microvascular ischemia-reperfusion injury.

Table IV

Role of MAM-targeted drugs in cardiac microvascular ischemia-reperfusion injury.

NameTargetModels(Refs.)
Shuangshen Ningxin formulaAlleviates cardiac microvascular I/R injury by improving mitochondrial function through the regulation of the NR4A1/MFF/Drp1 pathway.Rat cardiac microvascular I/R injury(83)
MelatoninExerts protective effects against cardiac microvascular I/R injury by promoting inhibitory phosphorylation of Drp1 at Ser637, suppressing fission.
Attenuates cardiac microvascular injury by targeting the mitochondrial fission-VDAC1-HK2-mPTP-mitophagy cascade.
Attenuates oxidative stress and reduces CMEC mortality by inhibiting IP3R-VDAC-mediated mitochondrial Ca2+ influx.
Protects against cardiac I/R injury by inhibition of the PPAR/FUNDC1/mitophagy pathway.
Mouse cardiac microvascular I/R injury(63)
H/R cardiac microvascular endothelial cells (CMEC)(78)
H/R CMECs Mouse I/R injury(183)
EmpagliflozinProtects against microvascular I/R injury by suppressing mitochondrial fission via through the inactivation of the DNA-PKcs/Fis1 pathway.
Exerts protective effects through activating AMPKα1/ULK1/FUNDC1 axis.
Mouse cardiac microvascular I/R injury(87)
Mouse cardiac microvascular I/R injury(182)
HCA (2'Hydroxycin-namaldehyde)Pretreatment with HCA reduces Drp1 expression and improves microvascular function during reperfusion.Rat cardiac microvascular I/R injury
H2O2-Induced H9C2 Cells
(88)
Liproxstatin-1Protects the myocardium by downregulating VDAC1 and restoring GPX4.Mouse I/R injury(151)
Ginkgolide AReduces oxidative stress and increased NO bioavailability in the heart.Mouse with pressure overload(152)
NaringinImproves cardiac microvascular function, potentially by facilitating NDUFS1 translocation to mitochondria and reducing ROS.Rat cardiac microvascular I/R injury(155)
Shenlian extractAttenuates I/R injury by inhibiting the PINK1/Parkin pathway and reducing mitophagy.Rat cardiac microvascular I/R injury(184)
Urolithin AMitophagy inducer, improves overall mitochondrial quality control and confers protection.H/R human umbilical vein endothelial cells (HUVECs)(185)
AcetylcholineProtects endothelial cells by destabilizing ER-mitochondrial contacts, and reducing the intracellular and mitochondrial Ca2+ overload.H/R HUVECs(193)
DapagliflozinReduces cardiac microvascular damage and endothelial dysfunction through inhibition of the XO-SERCA2-CaMKII-cofilin pathway.Mouse cardiac microvascular I/R injury
H/R human coronary artery endothelial cells (HCAECs)
(200)
PinacidilPreserves CRT expression, reduces endothelial Ca2+ overload, and prevents mitochondrial apoptosis in CMECs.Mouse cardiac microvascular I/R injury(217)

[i] MAM, mitochondria-associated endoplasmic reticulum membrane; I/R, ischemia-reperfusion; H/R, hypoxia/reoxygenation; NR4A1, nuclear receptor 4A1; MFF, mitochondrial fission factor; Drp1, dynamin-related protein 1; VDAC1, voltage-dependent anion channel 1; HK2, hexokinase 2; mPTP, mitochondrial permeability transition pore; IP3R, inositol 1,4,5-trisphosphate receptor; PPAR, peroxisome proliferator-activated receptor; FUNDC1, FUN14 domain-containing protein 1; DNA-PKcs, DNA-dependent protein kinase; Fis1, mitochondrial fission protein 1; AMPKα1, AMP-activated protein kinase alpha1; ULK1, Unc-51-like autophagy-activating kinase 1.

Despite these advances, the inherent complexity and dynamic nature of MAMs present major challenges to further investigation. Our current understanding of MAMs remains limited, particularly regarding how their morphology, inter-organelle distance, membrane thickness and number of contact sites influence cellular energy metabolism and contribute to disease pathogenesis. Beyond serving as a structural bridge between mitochondria and the ER, the bidirectional regulatory mechanisms by which MAMs coordinate organelle function remain incompletely understood, for instance, the role of MAM-localized MFN2. It also remains unclear whether MAMs show conserved structural and functional features across different cardiovascular cell types, how their formation can be selectively modulated for therapeutic benefit and whether they might serve as early biomarkers of microvascular injury. Furthermore, the temporal dynamics, spatial organization and hierarchical signaling networks that govern MAM remodeling in endothelial cells under stress conditions are still poorly characterized.

Future perspectives

Future research should aim to develop multiscale integrative models of MAMs to elucidate the causal relationships between their dynamic remodeling and microvascular pathological responses. Employing advanced in vivo multi-omics and imaging technologies, such as the Split-GFP contact sensor (221,222) and serial block-face scanning electron microscopy (223), may provide deeper insights into their structural and functional characteristics. Given the involvement of MAMs in key cellular processes, including energy metabolism, inflammation and cell death, their dysregulation likely contributes to the pathogenesis of cardiovascular, metabolic and neurological disorders. Therefore, clarifying the molecular mechanisms underlying MAM function could not only enhance our understanding of cardiovascular microcirculatory pathology but also reveal broadly applicable therapeutic targets for multiple diseases. However, pharmacological strategies targeting MAMs alone may be limited by individual metabolic variations and comorbid conditions; thus, future investigations should emphasize combinatorial regulatory approaches, such as dual-target therapies that modulate both MAMs and mitochondrial metabolism.

Availability of data and materials

Not applicable.

Authors' contributions

YW and B F conceived the idea for the present review and drafted the manuscript. YtW and ZS created the figures and contributed to the writing of the manuscript. HY, WC and CZ reviewed and modified the manuscript and screened literature. ZL provided valuable guidance and analysis during the present review. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Jilin Provincial Natural Science Foundation (grant no. YDZJ202401052ZYTS).

References

1 

Marin W, Marin D, Ao X and Liu Y: Mitochondria as a therapeutic target for cardiac ischemia-reperfusion injury (Review). Int J Mol Med. 47:485–499. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Yang M, Linn BS, Zhang Y and Ren J: Mitophagy and mitochondrial integrity in cardiac ischemia-reperfusion injury. Biochim Biophys Acta Mol Basis Dis. 1865:2293–2302. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Zhao BH, Ruze A, Zhao L, Li QL, Tang J, Xiefukaiti N, Gai MT, Deng AX, Shan XF and Gao XM: The role and mechanisms of microvascular damage in the ischemic myocardium. Cell Mol Life Sci. 80:3412023. View Article : Google Scholar : PubMed/NCBI

4 

Abbate A, Kontos MC and Biondi-Zoccai GGL: No-reflow: The next challenge in treatment of ST-elevation acute myocardial infarction. Eur Heart J. 29:1795–1797. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Davidson SM, Arjun S, Basalay MV, Bell RM, Bromage DI, Bøtker HE, Carr RD, Cunningham J, Ghosh AK, Heusch G, et al: The 10th biennial hatter cardiovascular institute workshop: Cellular protection-evaluating new directions in the setting of myocardial infarction, ischaemic stroke, and cardio-oncology. Basic Res Cardiol. 113:432018. View Article : Google Scholar : PubMed/NCBI

6 

de Waha S, Patel MR, Granger CB, Ohman EM, Maehara A, Eitel I, Ben-Yehuda O, Jenkins P, Thiele H and Stone GW: Relationship between microvascular obstruction and adverse events following primary percutaneous coronary intervention for ST-segment elevation myocardial infarction: An individual patient data pooled analysis from seven randomized trials. Eur Heart J. 38:3502–3510. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Eitel I, de Waha S, Wöhrle J, Fuernau G, Lurz P, Pauschinger M, Desch S, Schuler G and Thiele H: Comprehensive prognosis assessment by CMR imaging after ST-segment elevation myocardial infarction. J Am Coll Cardiol. 64:1217–1226. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Gao P, Yan Z and Zhu Z: Mitochondria-associated endoplasmic reticulum membranes in cardiovascular diseases. Front Cell Dev Biol. 8:6042402020. View Article : Google Scholar : PubMed/NCBI

9 

Kuznetsov AV, Javadov S, Margreiter R, Grimm M, Hagenbuchner J and Ausserlechner MJ: The role of mitochondria in the mechanisms of cardiac ischemia-reperfusion injury. Antioxidants (Basel). 8:4542019. View Article : Google Scholar : PubMed/NCBI

10 

Kluge MA, Fetterman JL and Vita JA: Mitochondria and endothelial function. Circ Res. 112:1171–1188. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Bertero E, Popoiu TA and Maack C: Mitochondrial calcium in cardiac ischemia/reperfusion injury and cardioprotection. Basic Res Cardiol. 119:569–585. 2024. View Article : Google Scholar : PubMed/NCBI

12 

Wong R, Steenbergen C and Murphy E: Mitochondrial permeability transition pore and calcium handling. Methods Mol Biol. 810:235–242. 2012. View Article : Google Scholar :

13 

Pang B, Dong G, Pang T, Sun X, Liu X, Nie Y and Chang X: Emerging insights into the pathogenesis and therapeutic strategies for vascular endothelial injury-associated diseases: Focus on mitochondrial dysfunction. Angiogenesis. 27:623–639. 2024. View Article : Google Scholar : PubMed/NCBI

14 

Luo Z, Yao J, Wang Z and Xu J: Mitochondria in endothelial cells angiogenesis and function: Current understanding and future perspectives. J Transl Med. 21:4412023. View Article : Google Scholar : PubMed/NCBI

15 

Parkkinen I, Their A, Asghar MY, Sree S, Jokitalo E and Airavaara M: Pharmacological regulation of endoplasmic reticulum structure and calcium dynamics: Importance for neurodegenerative diseases. Pharmacol Rev. 75:959–978. 2023. View Article : Google Scholar : PubMed/NCBI

16 

Zhou X, Jiang Y, Wang Y, Fan L, Zhu Y, Chen Y, Wang Y, Zhu Y, Wang H, Pan Z, et al: Endothelial FIS1 DeSUMOylation protects against hypoxic pulmonary hypertension. Circ Res. 133:508–531. 2023. View Article : Google Scholar : PubMed/NCBI

17 

Ren J, Bi Y, Sowers JR, Hetz C and Zhang Y: Endoplasmic reticulum stress and unfolded protein response in cardiovascular diseases. Nat Rev Cardiol. 18:499–521. 2021. View Article : Google Scholar : PubMed/NCBI

18 

Lackner LL: The expanding and unexpected functions of mitochondria contact sites. Trends Cell Biol. 29:580–590. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Copeland DE and Dalton AJ: An association between mitochondria and the endoplasmic reticulum in cells of the pseudobranch gland of a teleost. J Biophys Biochem Cytol. 5:393–396. 1959. View Article : Google Scholar : PubMed/NCBI

20 

Vance JE: Phospholipid synthesis in a membrane fraction associated with mitochondria. J Biol Chem. 265:7248–7256. 1990. View Article : Google Scholar : PubMed/NCBI

21 

Ronayne CT and Latorre-Muro P: Navigating the landscape of mitochondrial-ER communication in health and disease. Front Mol Biosci. 11:13565002024. View Article : Google Scholar : PubMed/NCBI

22 

Anderson AJ, Jackson TD, Stroud DA and Stojanovski D: Mitochondria-hubs for regulating cellular biochemistry: Emerging concepts and networks. Open Biol. 9:1901262019. View Article : Google Scholar : PubMed/NCBI

23 

Mishra SR, Mahapatra KK, Behera BP, Patra S, Bhol CS, Panigrahi DP, Praharaj PP, Singh A, Patil S, Dhiman R and Bhutia SK: Mitochondrial dysfunction as a driver of NLRP3 inflammasome activation and its modulation through mitophagy for potential therapeutics. Int J Biochem Cell Biol. 136:1060132021. View Article : Google Scholar : PubMed/NCBI

24 

Leung SWS and Shi Y: The glycolytic process in endothelial cells and its implications. Acta Pharmacol Sin. 43:251–259. 2022. View Article : Google Scholar :

25 

Kühlbrandt W: Structure and function of mitochondrial membrane protein complexes. BMC Biol. 13:892015. View Article : Google Scholar : PubMed/NCBI

26 

Vázquez-Meza H, Vilchis-Landeros MM, Vázquez-Carrada M, Uribe-Ramírez D and Matuz-Mares D: Cellular compartmentalization, glutathione transport and its relevance in some pathologies. Antioxidants (Basel). 12:8342023. View Article : Google Scholar : PubMed/NCBI

27 

Jagtap YA, Kumar P, Kinger S, Dubey AR, Choudhary A, Gutti RK, Gutti RK, Singh S, Jha HC, Poluri KM and Mishra A: Disturb mitochondrial associated proteostasis: Neurodegeneration and imperfect ageing. Front Cell Dev Biol. 11:11465642023. View Article : Google Scholar : PubMed/NCBI

28 

Schiaffarino O, Valdivieso González D, García-Pérez IM, Peñalva DA, Almendro-Vedia VG, Natale P and López-Montero I: Mitochondrial membrane models built from native lipid extracts: Interfacial and transport properties. Front Mol Biosci. 9:9109362022. View Article : Google Scholar : PubMed/NCBI

29 

Giacomello M and Pellegrini L: The coming of age of the mitochondria-ER contact: A matter of thickness. Cell Death Differ. 23:1417–1427. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Csordás G, Renken C, Várnai P, Walter L, Weaver D, Buttle KF, Balla T, Mannella CA and Hajnóczky G: Structural and functional features and significance of the physical linkage between ER and mitochondria. J Cell Biol. 174:915–921. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Filadi R, Theurey P and Pizzo P: The endoplasmic reticulum-mitochondria coupling in health and disease: Molecules, functions and significance. Cell Calcium. 62:1–15. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Wang N, Wang C, Zhao H, He Y, Lan B, Sun L and Gao Y: The MAMs structure and its role in cell death. Cells. 10:6572021. View Article : Google Scholar : PubMed/NCBI

33 

Mohan AA and Talwar P: MAM kinases: Physiological roles, related diseases, and therapeutic perspectives-a systematic review. Cell Mol Biol Lett. 30:352025. View Article : Google Scholar : PubMed/NCBI

34 

Adesina SE, Kang BY, Bijli KM, Ma J, Cheng J, Murphy TC, Michael Hart C and Sutliff RL: Targeting mitochondrial reactive oxygen species to modulate hypoxia-induced pulmonary hypertension. Free Radic Biol Med. 87:36–47. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Haslip M, Dostanic I, Huang Y, Zhang Y, Russell KS, Jurczak MJ, Mannam P, Giordano F, Erzurum SC and Lee PJ: Endothelial uncoupling protein 2 regulates mitophagy and pulmonary hypertension during intermittent hypoxia. Arterioscler Thromb Vasc Biol. 35:1166–1178. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Ye JX, Wang SS, Ge M and Wang DJ: Suppression of endothelial PGC-1α is associated with hypoxia-induced endothelial dysfunction and provides a new therapeutic target in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol. 310:L1233–L1242. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Lei W, Li J, Li C, Chen L, Huang F, Xiao D, Zhang J, Zhao J, Li G, Qu T, et al: MARCH5 restores endothelial cell function against ischaemic/hypoxia injury via Akt/eNOS pathway. J Cell Mol Med. 25:3182–3193. 2021. View Article : Google Scholar : PubMed/NCBI

38 

Zhou H, Wang J, Zhu P, Hu S and Ren J: Ripk3 regulates cardiac microvascular reperfusion injury: The role of IP3R-dependent calcium overload, XO-mediated oxidative stress and F-action/filopodia-based cellular migration. Cell Signal. 45:12–22. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Chen Y, Li S, Zhang Y, Wang M, Li X, Liu S, Xu D, Bao Y, Jia P, Wu N, et al: The lncRNA Malat1 regulates microvascular function after myocardial infarction in mice via miR-26b-5p/Mfn1 axis-mediated mitochondrial dynamics. Redox Biol. 41:1019102021. View Article : Google Scholar : PubMed/NCBI

40 

Zhong Z, Hou Y, Zhou C, Wang J, Gao L, Wu X, Zhou G, Liu S, Xu Y and Yang W: FL3 mitigates cardiac ischemia-reperfusion injury by promoting mitochondrial fusion to restore calcium homeostasis. Cell Death Discov. 11:3042025. View Article : Google Scholar : PubMed/NCBI

41 

Groschner LN, Waldeck-Weiermair M, Malli R and Graier WF: Endothelial mitochondria-less respiration, more integration. Pflugers Arch. 464:63–76. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Wang J, Toan S and Zhou H: New insights into the role of mitochondria in cardiac microvascular ischemia/reperfusion injury. Angiogenesis. 23:299–314. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Pang B, Dong G, Pang T, Sun X, Liu X, Nie Y and Chang X: Advances in pathogenesis and treatment of vascular endothelial injury-related diseases mediated by mitochondrial abnormality. Front Pharmacol. 15:14226862024. View Article : Google Scholar : PubMed/NCBI

44 

Dorn GW II: Evolving concepts of mitochondrial dynamics. Annu Rev Physiol. 81:1–17. 2019. View Article : Google Scholar

45 

Murphy E and Steenbergen C: Mechanisms underlying acute protection from cardiac ischemia-reperfusion injury. Physiol Rev. 88:581–609. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Wang HH, Wu YJ, Tseng YM, Su CH, Hsieh CL and Yeh HI: Mitochondrial fission protein 1 up-regulation ameliorates senescence-related endothelial dysfunction of human endothelial progenitor cells. Angiogenesis. 22:569–582. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Liu Y, Huo JL, Ren K, Pan S, Liu H, Zheng Y, Chen J, Qiao Y, Yang Y and Feng Q: Mitochondria-associated endoplasmic reticulum membrane (MAM): A dark horse for diabetic cardiomyopathy treatment. Cell Death Discov. 10:1482024. View Article : Google Scholar : PubMed/NCBI

48 

Tagaya M and Arasaki K: Regulation of mitochondrial dynamics and autophagy by the mitochondria-associated membrane. Adv Exp Med Biol. 997:33–47. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Bernal AF, Mota N, Pamplona R, Area-Gomez E and Portero-Otin M: Hakuna MAM-Tata: Investigating the role of mitochondrial-associated membranes in ALS. Biochim Biophys Acta Mol Basis Dis. 1869:1667162023. View Article : Google Scholar : PubMed/NCBI

50 

Chan DC: Fusion and fission: Interlinked processes critical for mitochondrial health. Annu Rev Genet. 46:265–287. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Chen W, Zhao H and Li Y: Mitochondrial dynamics in health and disease: Mechanisms and potential targets. Signal Transduct Target Ther. 8:3332023. View Article : Google Scholar : PubMed/NCBI

52 

Ferrier V: Mitochondrial fission in life and death. Nat Cell Biol. 3:E2692001. View Article : Google Scholar

53 

Gatti P, Schiavon C, Cicero J, Manor U and Germain M: Mitochondria- and ER-associated actin are required for mitochondrial fusion. Nat Commun. 16:4512025. View Article : Google Scholar : PubMed/NCBI

54 

Hemel IMGM, Sarantidou R and Gerards M: It takes two to tango: The essential role of ER-mitochondrial contact sites in mitochondrial dynamics. Int J Biochem Cell Biol. 141:1061012021. View Article : Google Scholar : PubMed/NCBI

55 

Guo Y, Li D, Zhang S, Yang Y, Liu JJ, Wang X, Liu C, Milkie DE, Moore RP, Tulu US, et al: Visualizing intracellular organelle and cytoskeletal interactions at nanoscale resolution on millisecond timescales. Cell. 175:1430–1442.e17. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Al Ojaimi M, Salah A and El-Hattab AW: Mitochondrial fission and fusion: Molecular mechanisms, biological functions, and related disorders. Membranes (Basel). 12:8932022. View Article : Google Scholar : PubMed/NCBI

57 

Friedman JR, Lackner LL, West M, DiBenedetto JR, Nunnari J and Voeltz GK: ER tubules mark sites of mitochondrial division. Science. 334:358–362. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Hyun HW, Min SJ and Kim JE: CDK5 inhibitors prevent astroglial apoptosis and reactive astrogliosis by regulating PKA and DRP1 phosphorylations in the rat hippocampus. Neurosci Res. 119:24–37. 2017. View Article : Google Scholar : PubMed/NCBI

59 

Ganesan V, Willis SD, Chang KT, Beluch S, Cooper KF and Strich R: Cyclin C directly stimulates Drp1 GTP affinity to mediate stress-induced mitochondrial hyperfission. Mol Biol Cell. 30:302–311. 2019. View Article : Google Scholar :

60 

Bravo-Sagua R, Parra V, Ortiz-Sandoval C, Navarro-Marquez M, Rodríguez AE, Diaz-Valdivia N, Sanhueza C, Lopez-Crisosto C, Tahbaz N, Rothermel BA, et al: Author correction: Caveolin-1 impairs PKA-DRP1-mediated remodelling of ER-mitochondria communication during the early phase of ER stress. Cell Death Differ. 26:24942019. View Article : Google Scholar : PubMed/NCBI

61 

Giedt RJ, Yang C, Zweier JL, Matzavinos A and Alevriadou BR: Mitochondrial fission in endothelial cells after simulated ischemia/reperfusion: Role of nitric oxide and reactive oxygen species. Free Radic Biol Med. 52:348–356. 2012. View Article : Google Scholar

62 

Ko AR, Hyun HW, Min SJ and Kim JE: The differential DRP1 phosphorylation and mitochondrial dynamics in the regional specific astroglial death induced by status epilepticus. Front Cell Neurosci. 10:1242016. View Article : Google Scholar : PubMed/NCBI

63 

Otera H, Ishihara N and Mihara K: New insights into the function and regulation of mitochondrial fission. Biochim Biophys Acta. 1833:1256–1268. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Ding Y, Liu N, Zhang D, Guo L, Shang Q, Liu Y, Ren G and Ma X: Mitochondria-associated endoplasmic reticulum membranes as a therapeutic target for cardiovascular diseases. Front Pharmacol. 15:13983812024. View Article : Google Scholar : PubMed/NCBI

65 

Ilamathi HS and Germain M: ER-mitochondria contact sites in mitochondrial DNA dynamics, maintenance, and distribution. Int J Biochem Cell Biol. 166:1064922024. View Article : Google Scholar

66 

Losón OC, Song Z, Chen H and Chan DC: Fis1, Mff, MiD49, and MiD51 mediate Drp1 recruitment in mitochondrial fission. Mol Biol Cell. 24:659–667. 2013. View Article : Google Scholar : PubMed/NCBI

67 

Otera H, Wang C, Cleland MM, Setoguchi K, Yokota S, Youle RJ and Mihara K: Mff is an essential factor for mitochondrial recruitment of Drp1 during mitochondrial fission in mammalian cells. J Cell Biol. 191:1141–1158. 2010. View Article : Google Scholar : PubMed/NCBI

68 

Wu W, Lin C, Wu K, Jiang L, Wang X, Li W, Zhuang H, Zhang X, Chen H, Li S, et al: FUNDC1 regulates mitochondrial dynamics at the ER-mitochondrial contact site under hypoxic conditions. EMBO J. 35:1368–1384. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Korobova F, Ramabhadran V and Higgs HN: An actin-dependent step in mitochondrial fission mediated by the ER-associated formin INF2. Science. 339:464–467. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Ji WK, Chakrabarti R, Fan X, Schoenfeld L, Strack S and Higgs HN: Receptor-mediated Drp1 oligomerization on endoplasmic reticulum. J Cell Biol. 216:4123–4139. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Li S, Li J, Li Y, Ye Q, Wang R, Liu X, Li H, Peng D and Duan X: Regulation of NR4A1 by Taohong Siwu decoction inhibits endothelial cell apoptosis in cerebral ischemia-reperfusion injury. J Ethnopharmacol. 353:1202852025. View Article : Google Scholar : PubMed/NCBI

72 

Jin Q, Li R, Hu N, Xin T, Zhu P, Hu S, Ma S, Zhu H, Ren J and Zhou H: DUSP1 alleviates cardiac ischemia/reperfusion injury by suppressing the Mff-required mitochondrial fission and Bnip3-related mitophagy via the JNK pathways. Redox Biol. 14:576–587. 2018. View Article : Google Scholar

73 

Zhou H, Wang J, Zhu P, Zhu H, Toan S, Hu S, Ren J and Chen Y: NR4A1 aggravates the cardiac microvascular ischemia reperfusion injury through suppressing FUNDC1-mediated mitophagy and promoting Mff-required mitochondrial fission by CK2α. Basic Res Cardiol. 113:232018. View Article : Google Scholar

74 

Landes T and Martinou JC: Mitochondrial outer membrane permeabilization during apoptosis: The role of mitochondrial fission. Biochim Biophys Acta. 1813:540–545. 2011. View Article : Google Scholar : PubMed/NCBI

75 

Adebayo M, Singh S, Singh AP and Dasgupta S: Mitochondrial fusion and fission: The fine-tune balance for cellular homeostasis. FASEB J. 35:e216202021. View Article : Google Scholar : PubMed/NCBI

76 

Hu SY, Zhang Y, Zhu PJ, Zhou H and Chen YD: Liraglutide directly protects cardiomyocytes against reperfusion injury possibly via modulation of intracellular calcium homeostasis. J Geriatr Cardiol. 14:57–66. 2017.PubMed/NCBI

77 

Zhu H, Jin Q, Li Y, Ma Q, Wang J, Li D, Zhou H and Chen Y: Melatonin protected cardiac microvascular endothelial cells against oxidative stress injury via suppression of IP3R-[Ca2+] c/VDAC-[Ca2+]m axis by activation of MAPK/ERK signaling pathway. Cell Stress Chaperones. 23:101–113. 2018. View Article : Google Scholar

78 

Zhou H, Hu S, Jin Q, Shi C, Zhang Y, Zhu P, Ma Q, Tian F and Chen Y: Mff-dependent mitochondrial fission contributes to the pathogenesis of cardiac microvasculature ischemia/reperfusion injury via induction of mROS-mediated cardiolipin oxidation and HK2/VDAC1 disassociation-involved mPTP opening. J Am Heart Assoc. 6:e0053282017. View Article : Google Scholar : PubMed/NCBI

79 

Zhou H, Zhang Y, Hu S, Shi C, Zhu P, Ma Q, Jin Q, Cao F, Tian F and Chen Y: Melatonin protects cardiac microvasculature against ischemia/reperfusion injury via suppression of mitochondrial fission-VDAC1-HK2-mPTP-mitophagy axis. J Pineal Res. 63:e124132017. View Article : Google Scholar : PubMed/NCBI

80 

Shenouda SM, Widlansky ME, Chen K, Xu G, Holbrook M, Tabit CE, Hamburg NM, Frame AA, Caiano TL, Kluge MA, et al: Altered mitochondrial dynamics contributes to endothelial dysfunction in diabetes mellitus. Circulation. 124:444–453. 2011. View Article : Google Scholar : PubMed/NCBI

81 

Qu K, Yan F, Qin X, Zhang K, He W, Dong M and Wu G: Mitochondrial dysfunction in vascular endothelial cells and its role in atherosclerosis. Front Physiol. 13:10846042022. View Article : Google Scholar

82 

Ong SB, Subrayan S, Lim SY, Yellon DM, Davidson SM and Hausenloy DJ: Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation. 121:2012–2022. 2010. View Article : Google Scholar : PubMed/NCBI

83 

Liu Z, Han X, You Y, Xin G, Li L, Gao J, Meng H, Cao C, Liu J, Zhang Y, et al: Shuangshen ningxin formula attenuates cardiac microvascular ischemia/reperfusion injury through improving mitochondrial function. J Ethnopharmacol. 323:1176902024. View Article : Google Scholar : PubMed/NCBI

84 

Zhou H, Wang J, Hu S, Zhu H, Toanc S and Ren J: BI1 alleviates cardiac microvascular ischemia-reperfusion injury via modifying mitochondrial fission and inhibiting XO/ROS/F-actin pathways. J Cell Physiol. 234:5056–5069. 2019. View Article : Google Scholar

85 

Zhou H, Shi C, Hu S, Zhu H, Ren J and Chen Y: BI1 is associated with microvascular protection in cardiac ischemia reperfusion injury via repressing Syk-Nox2-Drp1-mitochondrial fission pathways. Angiogenesis. 21:599–615. 2018. View Article : Google Scholar : PubMed/NCBI

86 

Chen Y, Liu C, Zhou P, Li J, Zhao X, Wang Y, Chen R, Song L, Zhao H and Yan H: Coronary endothelium no-reflow injury is associated with ROS-modified mitochondrial fission through the JNK-Drp1 signaling pathway. Oxid Med Cell Longev. 2021:66995162021. View Article : Google Scholar : PubMed/NCBI

87 

Zou R, Shi W, Qiu J, Zhou N, Du N, Zhou H, Chen X and Ma L: Empagliflozin attenuates cardiac microvascular ischemia/reperfusion injury through improving mitochondrial homeostasis. Cardiovasc Diabetol. 21:1062022. View Article : Google Scholar : PubMed/NCBI

88 

Cheng YH, Chiang CY, Wu CH and Chien CT: 2'-Hydroxycinnamaldehyde, a natural product from cinnamon, alleviates ischemia/reperfusion-induced microvascular dysfunction and oxidative damage in rats by upregulating cytosolic BAG3 and Nrf2/HO-1. Int J Mol Sci. 25:129622024. View Article : Google Scholar : PubMed/NCBI

89 

Mishra P, Carelli V, Manfredi G and Chan DC: Proteolytic cleavage of Opa1 stimulates mitochondrial inner membrane fusion and couples fusion to oxidative phosphorylation. Cell Metab. 19:630–641. 2014. View Article : Google Scholar : PubMed/NCBI

90 

Zhao L, Zhuang J, Wang Y, Zhou D, Zhao D, Zhu S, Pu J, Zhang H, Yin M, Zhao W, et al: Propofol ameliorates H9c2 cells apoptosis induced by oxygen glucose deprivation and reperfusion injury via inhibiting high levels of mitochondrial fusion and fission. Front Pharmacol. 10:612019. View Article : Google Scholar : PubMed/NCBI

91 

de Brito OM and Scorrano L: Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature. 456:605–610. 2008. View Article : Google Scholar : PubMed/NCBI

92 

Stuppia G, Rizzo F, Riboldi G, Del Bo R, Nizzardo M, Simone C, Comi GP, Bresolin N and Corti S: MFN2-related neuropathies: Clinical features, molecular pathogenesis and therapeutic perspectives. J Neurol Sci. 356:7–18. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Filadi R, Greotti E, Turacchio G, Luini A, Pozzan T and Pizzo P: Mitofusin 2 ablation increases endoplasmic reticulum-mitochondria coupling. Proc Natl Acad Sci USA. 112:E2174–E2181. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Prinz WA: Bridging the gap: Membrane contact sites in signaling, metabolism, and organelle dynamics. J Cell Biol. 205:759–769. 2014. View Article : Google Scholar : PubMed/NCBI

95 

Yang F, Wu R, Jiang Z, Chen J, Nan J, Su S, Zhang N, Wang C, Zhao J, Ni C, et al: Leptin increases mitochondrial OPA1 via GSK3-mediated OMA1 ubiquitination to enhance therapeutic effects of mesenchymal stem cell transplantation. Cell Death Dis. 9:5562018. View Article : Google Scholar : PubMed/NCBI

96 

Sprenger HG and Langer T: The good and the bad of mitochondrial breakups. Trends Cell Biol. 29:888–900. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Romanello V, Scalabrin M, Albiero M, Blaauw B, Scorrano L and Sandri M: Inhibition of the fission machinery mitigates OPA1 impairment in adult skeletal muscles. Cells. 9:5972019. View Article : Google Scholar

98 

Yin W, Li R, Feng X and James Kang Y: The involvement of cytochrome c oxidase in mitochondrial fusion in primary cultures of neonatal rat cardiomyocytes. Cardiovasc Toxicol. 19:365–373. 2018. View Article : Google Scholar

99 

Anderson CJ, Kahl A, Fruitman H, Qian L, Zhou P, Manfredi G and Iadecola C: Prohibitin levels regulate OMA1 activity and turnover in neurons. Cell Death Differ. 27:1896–1906. 2020. View Article : Google Scholar :

100 

Schulman JJ, Szczesniak LM, Bunker EN, Nelson HA, Roe MW, Wagner LE II, Yule DI and Wojcikiewicz RJH: Bok regulates mitochondrial fusion and morphology. Cell Death Differ. 26:2682–2694. 2019. View Article : Google Scholar : PubMed/NCBI

101 

Ding M, Liu C, Shi R, Yu M, Zeng K, Kang J, Fu F and Mi M: Mitochondrial fusion promoter restores mitochondrial dynamics balance and ameliorates diabetic cardiomyopathy in an optic atrophy 1-dependent way. Acta Physiol (Oxf). 229:e134282020. View Article : Google Scholar

102 

Hong Y, Tak H, Kim C, Kang H, Ji E, Ahn S, Jung M, Kim HL, Lee JH, Kim W and Lee EK: RNA binding protein HuD contributes to β-cell dysfunction by impairing mitochondria dynamics. Cell Death Differ. 27:1633–1643. 2020. View Article : Google Scholar

103 

Meyer JN, Leuthner TC and Luz AL: Mitochondrial fusion, fission, and mitochondrial toxicity. Toxicology. 391:42–53. 2017. View Article : Google Scholar : PubMed/NCBI

104 

Wu W, Zhao D, Shah SZA, Zhang X, Lai M, Yang D, Wu X, Guan Z, Li J, Zhao H, et al: OPA1 overexpression ameliorates mitochondrial cristae remodeling, mitochondrial dysfunction, and neuronal apoptosis in prion diseases. Cell Death Dis. 10:7102019. View Article : Google Scholar : PubMed/NCBI

105 

Lugus JJ, Ngoh GA, Bachschmid MM and Walsh K: Mitofusins are required for angiogenic function and modulate different signaling pathways in cultured endothelial cells. J Mol Cell Cardiol. 51:885–893. 2011. View Article : Google Scholar : PubMed/NCBI

106 

Sabouny R and Shutt TE: The role of mitochondrial dynamics in mtDNA maintenance. J Cell Sci. 134:jcs2589442021. View Article : Google Scholar : PubMed/NCBI

107 

Papanicolaou KN, Khairallah RJ, Ngoh GA, Chikando A, Luptak I, O'Shea KM, Riley DD, Lugus JJ, Colucci WS, Lederer WJ, et al: Mitofusin-2 maintains mitochondrial structure and contributes to stress-induced permeability transition in cardiac myocytes. Mol Cell Biol. 31:1309–1328. 2011. View Article : Google Scholar : PubMed/NCBI

108 

Peng C, Rao W, Zhang L, Wang K, Hui H, Wang L, Su N, Luo P, Hao YL, Tu Y, et al: Corrigendum to 'Mitofusin 2 ameliorates hypoxia-induced apoptosis via mitochondrial function and signaling pathways title of article' [International Journal of Biochemistry and Cell Biology 69 (2015) 29-40]. Int J Biochem Cell Biol. 73:1372016. View Article : Google Scholar : PubMed/NCBI

109 

Tan Y, Mui D, Toan S, Zhu P, Li R and Zhou H: SERCA overexpression improves mitochondrial quality control and attenuates cardiac microvascular ischemia-reperfusion injury. Mol Ther Nucleic Acids. 22:696–707. 2020. View Article : Google Scholar : PubMed/NCBI

110 

Li Y, Huang D, Jia L, Shangguan F, Gong S, Lan L, Song Z, Xu J, Yan C, Chen T, et al: LonP1 links mitochondria-er interaction to regulate heart function. Research (Wash D C). 6:01752023.PubMed/NCBI

111 

Liu P, Xie Q, Wei T, Chen Y, Chen H and Shen W: Activation of the NLRP3 inflammasome induces vascular dysfunction in obese OLETF rats. Biochem Biophys Res Commun. 468:319–325. 2015. View Article : Google Scholar : PubMed/NCBI

112 

Jung M, Dodsworth M and Thum T: Inflammatory cells and their non-coding RNAs as targets for treating myocardial infarction. Basic Res Cardiol. 114:42018. View Article : Google Scholar : PubMed/NCBI

113 

Xue J, Yan X, Yang Y, Chen M, Wu L, Gou Z, Sun Z, Talabieke S, Zheng Y and Luo D: Connexin 43 dephosphorylation contributes to arrhythmias and cardiomyocyte apoptosis in ischemia/reperfusion hearts. Basic Res Cardiol. 114:402019. View Article : Google Scholar : PubMed/NCBI

114 

Veeranki S and Tyagi SC: Mdivi-1 induced acute changes in the angiogenic profile after ischemia-reperfusion injury in female mice. Physiol Rep. 5:e132982017. View Article : Google Scholar : PubMed/NCBI

115 

Rouault P, Guimbal S, Cornuault L, Bourguignon C, Foussard N, Alzieu P, Choveau F, Benoist D, Chapouly C, Gadeau AP, et al: Thrombosis in the coronary microvasculature impairs cardiac relaxation and induces diastolic dysfunction. Arterioscler Thromb Vasc Biol. 44:e1–e18. 2024. View Article : Google Scholar

116 

Paillard M, Tubbs E, Thiebaut PA, Gomez L, Fauconnier J, Da Silva CC, Teixeira G, Mewton N, Belaidi E, Durand A, et al: Depressing mitochondria-reticulum interactions protects cardiomyocytes from lethal hypoxia-reoxygenation injury. Circulation. 128:1555–1565. 2013. View Article : Google Scholar : PubMed/NCBI

117 

Hall AR, Burke N, Dongworth RK, Kalkhoran SB, Dyson A, Vicencio JM, Dorn GW II, Yellon DM and Hausenloy DJ: Hearts deficient in both Mfn1 and Mfn2 are protected against acute myocardial infarction. Cell Death Dis. 7:e22382016. View Article : Google Scholar : PubMed/NCBI

118 

Hu C, Huang Y and Li L: Drp1-dependent mitochondrial fission plays critical roles in physiological and pathological progresses in mammals. Int J Mol Sci. 18:1442017. View Article : Google Scholar : PubMed/NCBI

119 

He P, Talukder MAH and Gao F: Oxidative stress and microvessel barrier dysfunction. Front Physiol. 11:4722020. View Article : Google Scholar : PubMed/NCBI

120 

Ivanina AV, Nesmelova I, Leamy L, Sokolov EP and Sokolova IM: Intermittent hypoxia leads to functional reorganization of mitochondria and affects cellular bioenergetics in marine molluscs. J Exp Biol. 219:1659–1674. 2016. View Article : Google Scholar : PubMed/NCBI

121 

Zhao RZ, Jiang S, Zhang L and Yu ZB: Mitochondrial electron transport chain, ROS generation and uncoupling (Review). Int J Mol Med. 44:3–15. 2019.PubMed/NCBI

122 

Qing G, Huang C, Pei J and Peng B: Alteration of cardiac energetics and mitochondrial function in doxorubicin-induced cardiotoxicity: Molecular mechanism and prospective implications (Review). Int J Mol Med. 56:1832025. View Article : Google Scholar :

123 

Potier E, Ferreira E, Meunier A, Sedel L, Logeart-Avramoglou D and Petite H: Prolonged hypoxia concomitant with serum deprivation induces massive human mesenchymal stem cell death. Tissue Eng. 13:1325–1331. 2007. View Article : Google Scholar : PubMed/NCBI

124 

Zhou H, Yang J, Xin T, Li D, Guo J, Hu S, Zhou S, Zhang T, Zhang Y, Han T and Chen Y: Exendin-4 protects adipose-derived mesenchymal stem cells from apoptosis induced by hydrogen peroxide through the PI3K/Akt-Sfrp2 pathways. Free Radic Biol Med. 77:363–375. 2014. View Article : Google Scholar : PubMed/NCBI

125 

Granger DN and Kvietys PR: Reperfusion injury and reactive oxygen species: The evolution of a concept. Redox Biol. 9:524–551. 2015. View Article : Google Scholar

126 

Kar S and Kavdia M: Modeling of biopterin-dependent pathways of eNOS for nitric oxide and superoxide production. Free Radic Biol Med. 51:1411–1427. 2011. View Article : Google Scholar : PubMed/NCBI

127 

Giorgi C, Missiroli S, Patergnani S, Duszynski J, Wieckowski MR and Pinton P: Mitochondria-associated membranes: Composition, molecular mechanisms, and physio-pathological implications. Antioxid Redox Signal. 22:995–1019. 2015. View Article : Google Scholar : PubMed/NCBI

128 

Missiroli S, Patergnani S, Caroccia N, Pedriali G, Perrone M, Previati M, Wieckowski MR and Giorgi C: Mitochondria-associated membranes (MAMs) and inflammation. Cell Death Dis. 9:3292018. View Article : Google Scholar : PubMed/NCBI

129 

Liu Y, Lian K, Zhang L, Wang R, Yi F, Gao C, Xin C, Zhu D, Li Y, Yan W, et al: TXNIP mediates NLRP3 inflammasome activation in cardiac microvascular endothelial cells as a novel mechanism in myocardial ischemia/reperfusion injury. Basic Res Cardiol. 109:4152014. View Article : Google Scholar : PubMed/NCBI

130 

Ovciarikova J, Shikha S, Lacombe A, Courjol F, McCrone R, Hussain W, Maclean A, Lemgruber L, Martins-Duarte ES, Gissot M and Sheiner L: Two ancient membrane pores mediate mitochondrial-nucleus membrane contact sites. J Cell Biol. 223:e2023040752024. View Article : Google Scholar : PubMed/NCBI

131 

Yan YR, Zhang L, Lin YN, Sun XW, Ding YJ, Li N, Li HP, Li SQ, Zhou JP and Li QY: Chronic intermittent hypoxia-induced mitochondrial dysfunction mediates endothelial injury via the TXNIP/NLRP3/IL-1β signaling pathway. Free Radic Biol Med. 165:401–410. 2021. View Article : Google Scholar : PubMed/NCBI

132 

Francisco J and Del Re DP: Inflammation in myocardial ischemia/reperfusion injury: Underlying mechanisms and therapeutic potential. Antioxidants (Basel). 12:19442023. View Article : Google Scholar : PubMed/NCBI

133 

van Hout GPJ, Bosch L, Ellenbroek GHJM, de Haan JJ, van Solinge WW, Cooper MA, Arslan F, de Jager SC, Robertson AAB, Pasterkamp G and Hoefer IE: The selective NLRP3-inflammasome inhibitor MCC950 reduces infarct size and preserves cardiac function in a pig model of myocardial infarction. Eur Heart J. 38:828–836. 2017.

134 

Zhang X, Zeng W, Zhang Y, Yu Q, Zeng M, Gan J, Zhang W, Jiang X and Li H: Focus on the role of mitochondria in NLRP3 inflammasome activation: A prospective target for the treatment of ischemic stroke (Review). Int J Mol Med. 49:742022. View Article : Google Scholar : PubMed/NCBI

135 

Toldo S, Mauro AG, Cutter Z and Abbate A: Inflammasome, pyroptosis, and cytokines in myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 315:H1553–H1568. 2018. View Article : Google Scholar : PubMed/NCBI

136 

Frand AR and Kaiser CA: Ero1p oxidizes protein disulfide isomerase in a pathway for disulfide bond formation in the endoplasmic reticulum. Mol Cell. 4:469–477. 1999. View Article : Google Scholar : PubMed/NCBI

137 

Giorgio M, Migliaccio E, Orsini F, Paolucci D, Moroni M, Contursi C, Pelliccia G, Luzi L, Minucci S, Marcaccio M, et al: Electron transfer between cytochrome c and p66Shc generates reactive oxygen species that trigger mitochondrial apoptosis. Cell. 122:221–233. 2005. View Article : Google Scholar : PubMed/NCBI

138 

Gilady SY, Bui M, Lynes EM, Benson MD, Watts R, Vance JE and Simmen T: Ero1alpha requires oxidizing and normoxic conditions to localize to the mitochondria-associated membrane (MAM). Cell Stress Chaperones. 15:619–629. 2010. View Article : Google Scholar : PubMed/NCBI

139 

Anelli T, Bergamelli L, Margittai E, Rimessi A, Fagioli C, Malgaroli A, Pinton P, Ripamonti M, Rizzuto R and Sitia R: Ero1α regulates Ca(2+) fluxes at the endoplasmic reticulum-mitochondria interface (MAM). Antioxid Redox Signal. 16:1077–1087. 2012. View Article : Google Scholar

140 

Bassot A, Chen J, Takahashi-Yamashiro K, Yap MC, Gibhardt CS, Le GNT, Hario S, Nasu Y, Moore J, Gutiérrez T, et al: The endoplasmic reticulum kinase PERK interacts with the oxidoreductase ERO1 to metabolically adapt mitochondria. Cell Rep. 42:1118992023. View Article : Google Scholar : PubMed/NCBI

141 

Muñoz JP, Ivanova S, Sánchez-Wandelmer J, Martínez-Cristóbal P, Noguera E, Sancho A, Díaz-Ramos A, Hernández-Alvarez MI, Sebastián D, Mauvezin C, et al: Mfn2 modulates the UPR and mitochondrial function via repression of PERK. EMBO J. 32:2348–2361. 2013. View Article : Google Scholar : PubMed/NCBI

142 

van Vliet AR, Giordano F, Gerlo S, Segura I, Van Eygen S, Molenberghs G, Rocha S, Houcine A, Derua R, Verfaillie T, et al: The ER stress sensor PERK coordinates ER-plasma membrane contact site formation through interaction with filamin-A and F-actin remodeling. Mol Cell. 65:885–899.e6. 2017. View Article : Google Scholar : PubMed/NCBI

143 

Verfaillie T, Rubio N, Garg AD, Bultynck G, Rizzuto R, Decuypere JP, Piette J, Linehan C, Gupta S, Samali A and Agostinis P: PERK is required at the ER-mitochondrial contact sites to convey apoptosis after ROS-based ER stress. Cell Death Differ. 19:1880–1891. 2012. View Article : Google Scholar : PubMed/NCBI

144 

Li G, Mongillo M, Chin KT, Harding H, Ron D, Marks AR and Tabas I: Role of ERO1-alpha-mediated stimulation of inositol 1,4,5-triphosphate receptor activity in endoplasmic reticulum stress-induced apoptosis. J Cell Biol. 186:783–792. 2009. View Article : Google Scholar : PubMed/NCBI

145 

Pinton P, Rimessi A, Marchi S, Orsini F, Migliaccio E, Giorgio M, Contursi C, Minucci S, Mantovani F, Wieckowski MR, et al: Protein kinase C beta and prolyl isomerase 1 regulate mitochondrial effects of the life-span determinant p66Shc. Science. 315:659–663. 2007. View Article : Google Scholar : PubMed/NCBI

146 

Akhmedov A, Montecucco F, Braunersreuther V, Camici GG, Jakob P, Reiner MF, Glanzmann M, Burger F, Paneni F, Galan K, et al: Genetic deletion of the adaptor protein p66Shc increases susceptibility to short-term ischaemic myocardial injury via intracellular salvage pathways. Eur Heart J. 36:516–526a. 2015. View Article : Google Scholar

147 

Carreras-Sureda A, Jaña F, Urra H, Durand S, Mortenson DE, Sagredo A, Bustos G, Hazari Y, Ramos-Fernández E, Sassano ML, et al: Publisher correction: Non-canonical function of IRE1α determines mitochondria-associated endoplasmic reticulum composition to control calcium transfer and bioenergetics. Nat Cell Biol. 21:9132019. View Article : Google Scholar

148 

Braunersreuther V, Montecucco F, Asrih M, Pelli G, Galan K, Frias M, Burger F, Quinderé AL, Montessuit C, Krause KH, et al: Role of NADPH oxidase isoforms NOX1, NOX2 and NOX4 in myocardial ischemia/reperfusion injury. J Mol Cell Cardiol. 64:99–107. 2013. View Article : Google Scholar : PubMed/NCBI

149 

Beretta M, Santos CX, Molenaar C, Hafstad AD, Miller CC, Revazian A, Betteridge K, Schröder K, Streckfuß-Bömeke K, Doroshow JH, et al: Nox4 regulates InsP3 receptor-dependent Ca2+ release into mitochondria to promote cell survival. EMBO J. 39:e1035302020. View Article : Google Scholar

150 

Fang G, Shen Y and Liao D: ENPP2 alleviates hypoxia/reoxygenation injury and ferroptosis by regulating oxidative stress and mitochondrial function in human cardiac microvascular endothelial cells. Cell Stress Chaperones. 28:253–263. 2023. View Article : Google Scholar : PubMed/NCBI

151 

Feng Y, Madungwe NB, Imam Aliagan AD, Tombo N and Bopassa JC: Liproxstatin-1 protects the mouse myocardium against ischemia/reperfusion injury by decreasing VDAC1 levels and restoring GPX4 levels. Biochem Biophys Res Commun. 520:606–611. 2019. View Article : Google Scholar : PubMed/NCBI

152 

You W, Wu Z, Ye F and Wu X: Ginkgolide A protects adverse cardiac remodeling through enhancing antioxidation and nitric oxide utilization in mice with pressure overload. Pharmazie. 74:698–702. 2019.PubMed/NCBI

153 

Helmstädter J, Frenis K, Filippou K, Grill A, Dib M, Kalinovic S, Pawelke F, Kus K, Kröller-Schön S, Oelze M, et al: Endothelial GLP-1 (glucagon-like peptide-1) receptor mediates cardiovascular protection by liraglutide in mice with experimental arterial hypertension. Arterioscler Thromb Vasc Biol. 40:145–158. 2020. View Article : Google Scholar :

154 

Yang J, Moraga A, Xu J, Zhao Y, Luo P, Lao KH, Margariti A, Zhao Q, Ding W, Wang G, et al: A histone deacetylase 7-derived peptide promotes vascular regeneration via facilitating 14-3-3γ phosphorylation. Stem Cells. 38:556–573. 2020. View Article : Google Scholar

155 

Guan X, Yang Z, Wang J, Lu W, Wang S, Yang M, Sun P, Hu W, Yang L and Li H: Naringin attenuates myocardial ischemia-reperfusion injury by promoting mitochondrial translocation of NDUFS1 and suppressing cardiac microvascular endothelial cell ferroptosis. J Nutr Biochem. 145:1100192025. View Article : Google Scholar : PubMed/NCBI

156 

Mao Y, Hu Y, Feng W, Yu L, Li P, Cai B, Li C and Guan H: Effects and mechanisms of PSS-loaded nanoparticles on coronary microcirculation dysfunction in streptozotocin-induced diabetic cardiomyopathy rats. Biomed Pharmacother. 121:1092802020. View Article : Google Scholar

157 

Booth DM, Enyedi B, Geiszt M, Várnai P and Hajnóczky G: Redox nanodomains are induced by and control calcium signaling at the ER-mitochondrial interface. Mol Cell. 63:240–248. 2016. View Article : Google Scholar : PubMed/NCBI

158 

Ježek J, Cooper KF and Strich R: The impact of mitochondrial fission-stimulated ROS production on pro-apoptotic chemotherapy. Biology (Basel). 10:332021.

159 

Li B, Yang WW, Yao BC, Chen QL, Zhao LL, Song YQ, Jiang N and Guo ZG: Liriodendrin alleviates myocardial ischemia-reperfusion injury via partially attenuating apoptosis, inflammation and mitochondria damage in rats. Int J Mol Med. 55:652025. View Article : Google Scholar :

160 

Chen Z, Liu T, Xiong L and Liu Z: Shen-fu injection modulates HIF-1α/BNIP3-mediated mitophagy to alleviate myocardial ischemia-reperfusion injury. Cardiovasc Toxicol. 25:898–914. 2025. View Article : Google Scholar : PubMed/NCBI

161 

Ashrafi G and Schwarz TL: The pathways of mitophagy for quality control and clearance of mitochondria. Cell Death Differ. 20:31–42. 2013. View Article : Google Scholar

162 

Puri R, Cheng XT, Lin MY, Huang N and Sheng ZH: Mul1 restrains Parkin-mediated mitophagy in mature neurons by maintaining ER-mitochondrial contacts. Nat Commun. 10:36452019. View Article : Google Scholar : PubMed/NCBI

163 

Martens S and Fracchiolla D: Activation and targeting of ATG8 protein lipidation. Cell Discov. 6:232020. View Article : Google Scholar : PubMed/NCBI

164 

Condon KJ and Sabatini DM: Nutrient regulation of mTORC1 at a glance. J Cell Sci. 132:jcs2225702019. View Article : Google Scholar : PubMed/NCBI

165 

Hamasaki M, Furuta N, Matsuda A, Nezu A, Yamamoto A, Fujita N, Oomori H, Noda T, Haraguchi T, Hiraoka Y, et al: Autophagosomes form at ER-mitochondria contact sites. Nature. 459:389–393. 2013. View Article : Google Scholar

166 

Barazzuol L, Giamogante F, Brini M and Calì T: PINK1/Parkin mediated mitophagy, Ca2+ signalling, and ER-mitochondria contacts in Parkinson's disease. Int J Mol Sci. 21:17722020. View Article : Google Scholar

167 

McLelland GL, Goiran T, Yi W, Dorval G, Chen CX, Lauinger ND, Krahn AI, Valimehr S, Rakovic A, Rouiller I, et al: Mfn2 ubiquitination by PINK1/parkin gates the p97-dependent release of ER from mitochondria to drive mitophagy. Elife. 7:e328662018. View Article : Google Scholar : PubMed/NCBI

168 

Gelmetti V, De Rosa P, Torosantucci L, Marini ES, Romagnoli A, Di Rienzo M, Arena G, Vignone D, Fimia GM and Valente EM: PINK1 and BECN1 relocalize at mitochondria-associated membranes during mitophagy and promote ER-mitochondria tethering and autophagosome formation. Autophagy. 13:654–669. 2017. View Article : Google Scholar : PubMed/NCBI

169 

Fu M, St-Pierre P, Shankar J, Wang PT, Joshi B and Nabi IR: Regulation of mitophagy by the Gp78 E3 ubiquitin ligase. Mol Biol Cell. 24:1153–1162. 2013. View Article : Google Scholar : PubMed/NCBI

170 

Zhao X, Wang Z, Wang L, Jiang T, Dong D and Sun M: The PINK1/Parkin signaling pathway-mediated mitophagy: A forgotten protagonist in myocardial ischemia/reperfusion injury. Pharmacol Res. 209:1074662024. View Article : Google Scholar : PubMed/NCBI

171 

Quiles JM, Najor RH, Gonzalez E, Jeung M, Liang W, Burbach SM, Zumaya EA, Diao RY, Lampert MA and Gustafsson ÅB: Deciphering functional roles and interplay between Beclin1 and Beclin2 in autophagosome formation and mitophagy. Sci Signal. 16:eabo44572023. View Article : Google Scholar : PubMed/NCBI

172 

Russell RC, Tian Y, Yuan H, Park HW, Chang YY, Kim J, Kim H, Neufeld TP, Dillin A and Guan KL: ULK1 induces autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase. Nat Cell Biol. 15:741–750. 2013. View Article : Google Scholar : PubMed/NCBI

173 

Kundu M, Lindsten T, Yang CY, Wu J, Zhao F, Zhang J, Selak MA, Ney PA and Thompson CB: Ulk1 plays a critical role in the autophagic clearance of mitochondria and ribosomes during reticulocyte maturation. Blood. 112:1493–1502. 2008. View Article : Google Scholar : PubMed/NCBI

174 

Sun W, Lu H, Dong S, Li R, Chu Y, Wang N, Zhao Y, Zhang Y, Wang L, Sun L and Lu D: Beclin1 controls caspase-4 inflammsome activation and pyroptosis in mouse myocardial reperfusion-induced microvascular injury. Cell Commun Signal. 19:1072021. View Article : Google Scholar : PubMed/NCBI

175 

Lampert MA, Orogo AM, Najor RH, Hammerling BC, Leon LJ, Wang BJ, Kim T, Sussman MA and Gustafsson ÅB: BNIP3L/NIX and FUNDC1-mediated mitophagy is required for mitochondrial network remodeling during cardiac progenitor cell differentiation. Autophagy. 15:1182–1198. 2019. View Article : Google Scholar : PubMed/NCBI

176 

Wang C, Dai X, Wu S, Xu W, Song P, Huang K and Zou MH: FUNDC1-dependent mitochondria-associated endoplasmic reticulum membranes are involved in angiogenesis and neoangiogenesis. Nat Commun. 12:26162021. View Article : Google Scholar : PubMed/NCBI

177 

Chen G, Han Z, Feng D, Chen Y, Chen L, Wu H, Huang L, Zhou C, Cai X, Fu C, et al: A regulatory signaling loop comprising the PGAM5 phosphatase and CK2 controls receptor-mediated mitophagy. Mol Cell. 54:362–377. 2014. View Article : Google Scholar : PubMed/NCBI

178 

Wu W, Tian W, Hu Z, Chen G, Huang L, Li W, Zhang X, Xue P, Zhou C, Liu L, et al: ULK1 translocates to mitochondria and phosphorylates FUNDC1 to regulate mitophagy. EMBO Rep. 15:566–575. 2014. View Article : Google Scholar : PubMed/NCBI

179 

Zhang Y, Zhuang H, Liu H and Feng D: Molecular regulations of FUNDC1 at ER-mitochondria contacts under hypoxic stress. Contact (Thousand Oaks). 5:251525642210924872022.PubMed/NCBI

180 

Ji H, Wang J, Muid D, Song W, Jiang Y and Zhou H: FUNDC1 activates the mitochondrial unfolded protein response to preserve mitochondrial quality control in cardiac ischemia/reperfusion injury. Cell Signal. 92:1102492022. View Article : Google Scholar : PubMed/NCBI

181 

Zhou H, Zhu P, Guo J, Hu N, Wang S, Li D, Hu S, Ren J, Cao F and Chen Y: Ripk3 induces mitochondrial apoptosis via inhibition of FUNDC1 mitophagy in cardiac IR injury. Redox Biol. 13:498–507. 2017. View Article : Google Scholar : PubMed/NCBI

182 

Cai C, Guo Z, Chang X, Li Z, Wu F, He J, Cao T, Wang K, Shi N, Zhou H, et al: Empagliflozin attenuates cardiac microvascular ischemia/reperfusion through activating the AMPKα1/ULK1/FUNDC1/mitophagy pathway. Redox Biol. 52:1022882022. View Article : Google Scholar

183 

Zhou H, Li D, Zhu P, Hu S, Hu N, Ma S, Zhang Y, Han T, Ren J, Cao F and Chen Y: Melatonin suppresses platelet activation and function against cardiac ischemia/reperfusion injury via PPARγ/FUNDC1/mitophagy pathways. J Pineal Res. 63:e124382017. View Article : Google Scholar

184 

Li JJ, Wang YJ, Wang CM, Li YJ, Yang Q, Cai WY, Chen Y and Zhu XX: Shenlian extract decreases mitochondrial autophagy to regulate mitochondrial function in microvascular to alleviate coronary artery no-reflow. Phytother Res. 37:1864–1882. 2023. View Article : Google Scholar : PubMed/NCBI

185 

Liu S, Faitg J, Tissot C, Konstantopoulos D, Laws R, Bourdier G, Andreux PA, Davey T, Gallart-Ayala H, Ivanisevic J, et al: Urolithin A provides cardioprotection and mitochondrial quality enhancement preclinically and improves human cardiovascular health biomarkers. iScience. 28:1118142025. View Article : Google Scholar : PubMed/NCBI

186 

Wang J, Toan S and Zhou H: Mitochondrial quality control in cardiac microvascular ischemia-reperfusion injury: New insights into the mechanisms and therapeutic potentials. Pharmacol Res. 156:1047712020. View Article : Google Scholar : PubMed/NCBI

187 

Shirihai OS, Song M and Dorn GW II: How mitochondrial dynamism orchestrates mitophagy. Circ Res. 116:1835–1849. 2015. View Article : Google Scholar : PubMed/NCBI

188 

Wong YC, Ysselstein D and Krainc D: Mitochondria-lysosome contacts regulate mitochondrial fission via RAB7 GTP hydrolysis. Nature. 554:382–386. 2018. View Article : Google Scholar : PubMed/NCBI

189 

Wang J, Zhu P, Li R, Ren J and Zhou H: Fundc1-dependent mitophagy is obligatory to ischemic preconditioning-conferred renoprotection in ischemic AKI via suppression of Drp1-mediated mitochondrial fission. Redox Biol. 30:1014152020. View Article : Google Scholar : PubMed/NCBI

190 

Huang CH, Chiang CY, Pen RH, Tsai MS, Chen HW, Hsu CY, Wang TD, Ma MH, Chen SC and Chen WJ: Hypothermia treatment preserves mitochondrial integrity and viability of cardiomyocytes after ischaemic reperfusion injury. Injury. 46:233–239. 2015. View Article : Google Scholar

191 

Wang Z, Liu D, Varin A, Nicolas V, Courilleau D, Mateo P, Caubere C, Rouet P, Gomez AM, Vandecasteele G, et al: A cardiac mitochondrial cAMP signaling pathway regulates calcium accumulation, permeability transition and cell death. Cell Death Dis. 7:e21982016. View Article : Google Scholar : PubMed/NCBI

192 

Jiang RQ, Li QQ and Sheng R: Mitochondria associated ER membranes and cerebral ischemia: Molecular mechanisms and therapeutic strategies. Pharmacol Res. 191:1067612023. View Article : Google Scholar : PubMed/NCBI

193 

He X, Bi XY, Lu XZ, Zhao M, Yu XJ, Sun L, Xu M, Wier WG and Zang WJ: Reduction of mitochondria-endoplasmic reticulum interactions by acetylcholine protects human umbilical vein endothelial cells from hypoxia/reoxygenation injury. Arterioscler Thromb Vasc Biol. 35:1623–1634. 2015. View Article : Google Scholar : PubMed/NCBI

194 

Wu S, Lu Q, Wang Q, Ding Y, Ma Z, Mao X, Huang K, Xie Z and 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. 136:2248–2266. 2017. View Article : Google Scholar : PubMed/NCBI

195 

Li C, Ma Q, Toan S, Wang J, Zhou H and Liang J: SERCA overexpression reduces reperfusion-mediated cardiac microvascular damage through inhibition of the calcium/MCU/mPTP/necroptosis signaling pathways. Redox Biol. 36:1016592020. View Article : Google Scholar : PubMed/NCBI

196 

Yang R, Zhang X, Xing P, Zhang S, Zhang F, Wang J, Yu J, Zhu X and Chang P: Grpel2 alleviates myocardial ischemia/reperfusion injury by inhibiting MCU-mediated mitochondrial calcium overload. Biochem Biophys Res Commun. 609:169–175. 2022. View Article : Google Scholar : PubMed/NCBI

197 

Carreras-Sureda A, Jaña F, Urra H, Durand S, Mortenson DE, Sagredo A, Bustos G, Hazari Y, Ramos-Fernández E, Sassano ML, et al: Non-canonical function of IRE1α determines mitochondria-associated endoplasmic reticulum composition to control calcium transfer and bioenergetics. Nat Cell Biol. 21:755–767. 2019. View Article : Google Scholar : PubMed/NCBI

198 

Goodman JB, Qin F, Morgan RJ, Chambers JM, Croteau D, Siwik DA, Hobai I, Panagia M, Luptak I, Bachschmid M, et al: Redox-Resistant SERCA [Sarco(endo)plasmic Reticulum Calcium ATPase] attenuates oxidant-stimulated mitochondrial calcium and apoptosis in cardiac myocytes and pressure overload-induced myocardial failure in mice. Circulation. 142:2459–2469. 2020. View Article : Google Scholar : PubMed/NCBI

199 

Chen LT, Xu TT, Qiu YQ, Liu NY, Ke XY, Fang L, Yan JP and Zhu DY: Homocysteine induced a calcium-mediated disruption of mitochondrial function and dynamics in endothelial cells. J Biochem Mol Toxicol. 35:e227372021. View Article : Google Scholar : PubMed/NCBI

200 

Ma L, Zou R, Shi W, Zhou N, Chen S, Zhou H, Chen X and Wu Y: SGLT2 inhibitor dapagliflozin reduces endothelial dysfunction and microvascular damage during cardiac ischemia/reperfusion injury through normalizing the XO-SERCA2-CaMKII-coffilin pathways. Theranostics. 12:5034–5050. 2022. View Article : Google Scholar : PubMed/NCBI

201 

Hoppe UC: Mitochondrial calcium channels. FEBS Lett. 584:1975–1981. 2010. View Article : Google Scholar : PubMed/NCBI

202 

Israelson A, Abu-Hamad S, Zaid H, Nahon E and Shoshan-Barmatz V: Localization of the voltage-dependent anion channel-1 Ca2+-binding sites. Cell Calcium. 41:235–244. 2007. View Article : Google Scholar

203 

Szabadkai G, Bianchi K, Várnai P, De Stefani D, Wieckowski MR, Cavagna D, Nagy AI, Balla T and Rizzuto R: Chaperone-mediated coupling of endoplasmic reticulum and mitochondrial Ca2+ channels. J Cell Biol. 175:901–911. 2006. View Article : Google Scholar : PubMed/NCBI

204 

Li Y, Hu H, Chu C and Yang J: Mitochondrial calcium uniporter complex: An emerging therapeutic target for cardiovascular diseases (Review). Int J Mol Med. 55:402025. View Article : Google Scholar : PubMed/NCBI

205 

Xu H, Guan N, Ren YL, Wei QJ, Tao YH, Yang GS, Liu XY, Bu DF, Zhang Y and Zhu SN: IP3R-Grp75-VDAC1-MCU calcium regulation axis antagonists protect podocytes from apoptosis and decrease proteinuria in an Adriamycin nephropathy rat model. BMC Nephrol. 19:1402018. View Article : Google Scholar

206 

Gomez L, Thiebaut PA, Paillard M, Ducreux S, Abrial M, Crola Da Silva C, Durand A, Alam MR, Van Coppenolle F, Sheu SS and Ovize M: The SR/ER-mitochondria calcium crosstalk is regulated by GSK3β during reperfusion injury. Cell Death Differ. 23:313–322. 2016. View Article : Google Scholar

207 

Hayashi T and Su TP: Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate Ca(2+) signaling and cell survival. Cell. 131:596–610. 2007. View Article : Google Scholar : PubMed/NCBI

208 

Embi N, Rylatt DB and Cohen P: Glycogen synthase kinase-3 from rabbit skeletal muscle. Separation from cyclic-AMP-dependent protein kinase and phosphorylase kinase. Eur J Biochem. 107:519–527. 1980. View Article : Google Scholar : PubMed/NCBI

209 

Woodgett JR and Cohen P: Multisite phosphorylation of glycogen synthase. Molecular basis for the substrate specificity of glycogen synthase kinase-3 and casein kinase-II (glycogen synthase kinase-5). Biochim Biophys Acta. 788:339–347. 1984. View Article : Google Scholar : PubMed/NCBI

210 

Thoudam T, Jeon JH, Ha CM and Lee IK: Role of mitochondria-associated endoplasmic reticulum membrane in inflammation-mediated metabolic diseases. Mediators Inflamm. 2016:18514202016. View Article : Google Scholar

211 

Elrod JW and Molkentin JD: Physiologic functions of cyclophilin D and the mitochondrial permeability transition pore. Circ J. 77:1111–1122. 2013. View Article : Google Scholar : PubMed/NCBI

212 

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

213 

Stoica R, De Vos KJ, Paillusson S, Mueller S, Sancho RM, Lau KF, Vizcay-Barrena G, Lin WL, Xu YF, Lewis J, et al: ER-mitochondria associations are regulated by the VAPB-PTPIP51 interaction and are disrupted by ALS/FTD-associated TDP-43. Nat Commun. 5:39962014. View Article : Google Scholar : PubMed/NCBI

214 

Gomez-Suaga P, Paillusson S, Stoica R, Noble W, Hanger DP and Miller CCJ: The ER-mitochondria tethering complex VAPB-PTPIP51 regulates autophagy. Curr Biol. 27:371–385. 2017. View Article : Google Scholar : PubMed/NCBI

215 

De Vos KJ, Mórotz GM, Stoica R, Tudor EL, Lau KF, Ackerley S, Warley A, Shaw CE and Miller CC: VAPB interacts with the mitochondrial protein PTPIP51 to regulate calcium homeostasis. Hum Mol Genet. 21:1299–1311. 2012. View Article : Google Scholar :

216 

Fucikova J, Spisek R, Kroemer G and Galluzzi L: Calreticulin and cancer. Cell Res. 31:5–16. 2021. View Article : Google Scholar :

217 

Liu M, Li S, Yin M, Li Y, Chen J, Chen Y, Zhou Y, Li Q, Xu F, Dai C, et al: Pinacidil ameliorates cardiac microvascular ischemia-reperfusion injury by inhibiting chaperone-mediated autophagy of calreticulin. Basic Res Cardiol. 119:113–131. 2024. View Article : Google Scholar : PubMed/NCBI

218 

Zhang J, Wang L, Xie W, Hu S, Zhou H, Zhu P, Zhu P and Zhu H: Melatonin attenuates ER stress and mitochondrial damage in septic cardiomyopathy: A new mechanism involving BAP31 upregulation and MAPK-ERK pathway. J Cell Physiol. 235:2847–2856. 2020. View Article : Google Scholar

219 

Li S, Chen J, Liu M, Chen Y, Wu Y, Li Q, Ma T, Gao J, Xia Y, Fan M, et al: Protective effect of HINT2 on mitochondrial function via repressing MCU complex activation attenuates cardiac microvascular ischemia-reperfusion injury. Basic Res Cardiol. 116:652021. View Article : Google Scholar : PubMed/NCBI

220 

Zhou H and Toan S: Pathological roles of mitochondrial oxidative stress and mitochondrial dynamics in cardiac microvascular ischemia/reperfusion injury. Biomolecules. 10:852020. View Article : Google Scholar : PubMed/NCBI

221 

Vallese F, Catoni C, Cieri D, Barazzuol L, Ramirez O, Calore V, Bonora M, Giamogante F, Pinton P, Brini M and Calì T: An expanded palette of improved SPLICS reporters detects multiple organelle contacts in vitro and in vivo. Nat Commun. 11:60692020. View Article : Google Scholar : PubMed/NCBI

222 

Calì T and Brini M: Quantification of organelle contact sites by split-GFP-based contact site sensors (SPLICS) in living cells. Nat Protoc. 16:5287–5308. 2021. View Article : Google Scholar : PubMed/NCBI

223 

Marshall AG, Neikirk K, Stephens DC, Vang L, Vue Z, Beasley HK, Crabtree A, Scudese E, Lopez EG, Shao B, et al: Serial block face-scanning electron microscopy as a burgeoning technology. Adv Biol (Weinh). 7:e23001392023. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Wang Y, Feng B, Wu Y, Sun Z, Yuan H, Chen W, Zhao C and Liu Z: Mitochondria-associated endoplasmic reticulum membranes: Emerging regulators of cardiac microvascular ischemia/reperfusion injury (Review). Int J Mol Med 57: 34, 2026.
APA
Wang, Y., Feng, B., Wu, Y., Sun, Z., Yuan, H., Chen, W. ... Liu, Z. (2026). Mitochondria-associated endoplasmic reticulum membranes: Emerging regulators of cardiac microvascular ischemia/reperfusion injury (Review). International Journal of Molecular Medicine, 57, 34. https://doi.org/10.3892/ijmm.2025.5705
MLA
Wang, Y., Feng, B., Wu, Y., Sun, Z., Yuan, H., Chen, W., Zhao, C., Liu, Z."Mitochondria-associated endoplasmic reticulum membranes: Emerging regulators of cardiac microvascular ischemia/reperfusion injury (Review)". International Journal of Molecular Medicine 57.2 (2026): 34.
Chicago
Wang, Y., Feng, B., Wu, Y., Sun, Z., Yuan, H., Chen, W., Zhao, C., Liu, Z."Mitochondria-associated endoplasmic reticulum membranes: Emerging regulators of cardiac microvascular ischemia/reperfusion injury (Review)". International Journal of Molecular Medicine 57, no. 2 (2026): 34. https://doi.org/10.3892/ijmm.2025.5705
Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y, Feng B, Wu Y, Sun Z, Yuan H, Chen W, Zhao C and Liu Z: Mitochondria-associated endoplasmic reticulum membranes: Emerging regulators of cardiac microvascular ischemia/reperfusion injury (Review). Int J Mol Med 57: 34, 2026.
APA
Wang, Y., Feng, B., Wu, Y., Sun, Z., Yuan, H., Chen, W. ... Liu, Z. (2026). Mitochondria-associated endoplasmic reticulum membranes: Emerging regulators of cardiac microvascular ischemia/reperfusion injury (Review). International Journal of Molecular Medicine, 57, 34. https://doi.org/10.3892/ijmm.2025.5705
MLA
Wang, Y., Feng, B., Wu, Y., Sun, Z., Yuan, H., Chen, W., Zhao, C., Liu, Z."Mitochondria-associated endoplasmic reticulum membranes: Emerging regulators of cardiac microvascular ischemia/reperfusion injury (Review)". International Journal of Molecular Medicine 57.2 (2026): 34.
Chicago
Wang, Y., Feng, B., Wu, Y., Sun, Z., Yuan, H., Chen, W., Zhao, C., Liu, Z."Mitochondria-associated endoplasmic reticulum membranes: Emerging regulators of cardiac microvascular ischemia/reperfusion injury (Review)". International Journal of Molecular Medicine 57, no. 2 (2026): 34. https://doi.org/10.3892/ijmm.2025.5705
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team