Review article
Roles and mechanisms of SUMOylation on key proteins in myocardial ischemia/reperfusion injury

https://doi.org/10.1016/j.yjmcc.2019.07.009Get rights and content

Highlights

  • Protein SUMOylation plays remarkable roles in myocardial ischemia/reperfusion (MI/R) injury.

  • Sarcoplasmic reticulum Ca2+ ATPase pump SUMOylation alleviates calcium overload. SUMOylation of dynamic-related protein 1 influences the function of mitochondria.

  • SUMOylation of histone deacetylase participates in epigenetic changes, whereas Sirt1 plays cardioprotective roles in the SUMOylated form.

  • SUMOylation of hypoxia-inducible factors is fundamental in oxygen homeostasis and eukaryotic elongation factor 2 SUMOylation induced cardiomyocyte apoptosis.

  • SUMOylation may provide potential targets for the design of novel drugs that used for therapy of MI/R injury.

Abstract

Myocardial ischemia/reperfusion (MI/R) injury has a great influence on the prognosis of patients with acute coronary occlusion. The underlying mechanisms of MI/R injury are complex. While the incidence of MI/R injury is increasing every year, the existing therapies are not satisfactory. Recently, small ubiquitin-related modifier (SUMO), which is a post-translational modification and involved in many cell processes, was found to play remarkable roles in MI/R injury. Several proteins that can be SUMOylated were found to interfere with different mechanisms of MI/R injury. Sarcoplasmic reticulum Ca2+ ATPase pump SUMOylation alleviated calcium overload. Among the histone deacetylase (HDAC) members, SUMOylation of HDAC4 reduced reactive oxygen species generation, whereas Sirt1 played protective roles in the SUMOylated form. Dynamic-related protein 1 modified by different SUMO proteins exerted opposite effects on the function of mitochondria. SUMOylation of hypoxia-inducible factors was fundamental in oxygen homeostasis, while eukaryotic elongation factor 2 SUMOylation induced cardiomyocyte apoptosis. The impact of other SUMOylation substrates in MI/R injury remains unclear. Here we reviewed how these SUMOylated proteins alleviated or exacerbated myocardial impairments by effecting the MI/R injury mechanisms. This may suggest methods for relieving MI/R injury in clinical practice and provide a reference for further study of SUMOylation in MI/R injury.

Introduction

Acute coronary occlusion is one of the leading causes of morbidity and mortality in the world [1]. Recovery of reperfusion is the primary therapy utilized to save viable myocardium, limit the size of myocardial infarction, preserve cardiac systolic function, and delay the development of heart failure [2]. However, recanalization of blood flow not only restores oxygen and nutrient supply but also damages the myocardium. This pathophysiological phenomenon is known as the myocardial ischemia/reperfusion (MI/R) injury. The underlying mechanisms of MI/R injury have not yet been fully elucidated. It is considered that oxidative stress, calcium overload, mitochondrial dysfunction, endoplasmic reticulum stress, activation of apoptotic and autophagic pathways, and epigenetic changes may be involved [3]. Over the past three decades, numerous studies have been devoted to elucidating the molecular mechanisms of MI/R injury, with a view to identifying therapeutic strategies to reduce the severity caused by reperfusion. However, there is still no effective therapy for the prevention of MI/R injury. Therefore, novel cardioprotective strategies are urgently needed to increase the resistance of cardiomyocytes to MI/R injury.

MI/R injury is a complex pathological process that involves many signaling pathways. It may be a better strategy to target a multifunctional cellular process and coordinate multiple signaling pathways involved in MI/R injury. From this point of view, small ubiquitin-related modifier (SUMO), which has been the focus of many studies in recent years, has proven to be a promising candidate.

SUMO was first discovered by Okura et al. in 1996 [4] and confirmed in the late 1990s as a mechanism conjugating lysine residues of proteins to regulate protein functions [5]. Four distinct human SUMO proteins (SUMO1, 2, 3, and 4) have been identified. SUMO1, 2, and 3 are widely distributed in the body, while SUMO4 is only found in kidney, spleen, and lymph nodes [6] and cannot be conjugated due to the lack of a C-terminal [7]. The expression level of SUMO2/3 protein is much higher than that of SUMO1 [6]. Although most of the SUMO substrates are located in the nucleus, SUMOylation is not limited to nuclear proteins, but can also be detected in extra-nuclear proteins [8]. All SUMOs are expressed as precursor proteins before conjugation. Conjugation requires three reaction steps: SUMO-activation, SUMO-conjugation, and SUMO-ligation. First, mature SUMO is activated by the E1 activating enzyme (SAE1/2). Then, SUMO is transferred to the only E2 conjugating enzyme, UBC9. Finally, SUMO is promoted to conjugate with substrates by the E3 ligase, which includes a protein inhibitor of activated STAT (PIAS) family, RanBR2, polycomb2, mitochondrial-anchored protein ligase (MAPL), and others [9]. SUMO conjugation pathway is a reversible process. Proteins conjugated by SUMO may be deSUMOylated by the SENtrin-specific protease (SENP) and deSUMOylating isopeptidases (DeSI) [10] (Fig. 1). Six members of the SENP family associated with deSUMOylation have been found in humans (SENP1–3, 5–7). SENPs show different specificity for all SUMO proteins and substrates [11]. SENP1 and SENP2 have deSUMOylation effects on all SUMO proteins [12], whereas SENP3, 5, 6, and 7 give priority to SUMO2/3 [13]. DeSUMOylation eliminates the effects of SUMOylation. However, one of the deSUMOylases, SENP, is closely related to SUMO-activation. Specific conditions that trigger SUMOylation and deSUMOylation remain unclear.

Since the discovery of the first SUMO protein, the SUMO conjugation pathway has attracted ample attention due to its different roles in various cellular events. Recent advances in SUMO research have highlighted its roles in heart development, function, and disease. SUMOylation has been shown to occur in many cell types of the heart, such as cardiomyocytes, fibroblasts [14,15], endothelial cells [16,17], smooth muscle cells [18], etc. However, the differences and commonalities of SUMOylation in these cells are still unclear. SUMOylation and some other post-translational modifications (PTM) can also regulate cardiac proteasome function and alter cellular protein homeostasis [19,20]. Previous studies of SUMOylation in cardiovascular disease were principally focused on heart failure. However, few studies have been conducted on SUMOylation in MI/R injury.

Current research suggests that SUMOylation of some proteins could alleviate or exacerbate myocardial damage by inhibiting or promoting the pathological mechanisms of MI/R injury. There is a series of metabolic disorders that occur during MI/R, including dysfunction of a number of proteins. As a type of PTM, SUMOylation or deSUMOylation may partly enhance the protein expression, thereby affecting the mechanisms of MI/R injury through performing their functions. By determining the exact effects of protein SUMOylation on MI/R injury, the damage can be attenuated by promoting or inhibiting SUMOylation. Therefore, SUMOylation in MI/R may provide an effective method to relieve myocardial injury and enhance the curative effects of recanalization.

In this review, we will refer to six key proteins associated with SUMOylation, which have been suggested to play remarkable roles during MI/R injury, as well as describe the following protein characteristics: (1) physiological functions; (2) pathological protein changes during MI/R; (3) influence of altering protein SUMOylation status on MI/R injury and their relevant mechanisms.

Section snippets

SUMOylation of SERCA2a alleviates calcium overload

Calcium overload is one of the most essential mechanisms in MI/R injury. Sarcoplasmic reticulum (SR) Ca2+ ATPase pump (SERCA2a), which is highly expressed in the heart, is a critical ATPase that controls Ca2+ reuptake and replenishment to SR in excitation-contraction coupling. In the period of ischemia and reperfusion, various factors cause the elevation of cytoplasmic Ca2+ levels (Fig. 2). The high concentration of Ca2+ in the cytosol activates several proteases and other proteins that lead to

SUMOylation of HDAC in epigenetic changes

It was confirmed that MI/R could cause epigenetic changes and tissue damage [31]. Epigenetic refers to the regulation of heritable gene expression without DNA sequence changes, including DNA methylation, histone modification, and noncoding RNAs. Histone modifications include acetylation, methylation, ubiquitination, phosphorylation, and crotonylation [32]. A histone octamer contains four different histone proteins (H2A, H2B, H3, and H4) with 146 pairs of DNA wrapped around to form a nucleosome,

SUMOylation of Drp1 in mitochondrial fission

Mitochondria are important organelles for energy production, buffering calcium, ROS generation, and sequestering molecules such as cytochrome c [54]. Mitochondria are composed of the outer and inner membranes. The outer membrane encapsulates the inner membrane, while the inner membrane is the site where most of the electron transport proteins occur [55]. Mitochondria change their morphology constantly through fission and fusion in response to alterations in their intracellular environment in

SUMOylation in HIF-mediated hypoxia-signaling cascade

During myocardial ischemia, the cardiomyocyte energy supply decreases, the metabolism becomes abnormal, and homeostasis is broken. These factors lead to cell dysfunction and even death. Hypoxia-inducible factor (HIF) plays an essential role in cellular and systemic oxygen homeostasis by inducing the transcription of hundreds of hypoxia-responsive genes. These genes are involved in many cell functional events, including angiogenesis, vasomotor control, vascular reactivity and remodeling,

Phosphorylated eEF2 is SUMOylated and induces cardiomyocyte apoptosis

Apoptosis is a basic mechanism of cell death during MI/R injury. It is a programmed cell death mediated by multiple apoptotic mediators. It was confirmed that DNA damage caused by various factors is an important reason for apoptosis [79]. Here, we will describe one of the mechanisms associated with SUMOylation that causes DNA damage.

Eukaryotic elongation factor 2 (eEF2) is one of the protein factors involved in polypeptide chain elongation in protein translation. It was confirmed that eEF2

Conclusion

In this review, we summarized recent studies investigating the effects of SUMOylation of six key proteins on MI/R injury. The modifications by SUMO affect the expression and function of proteins in MI/R and attenuate or exacerbate the myocardium injury via different mechanisms. SEARCA2a SUMOylation relieves Ca2+ overload in MI/R and reduces myocardial injury. SUMOylation promotes degradation of HDAC4 and reduces the production of ROS. Sirt1 mainly exists in the nucleus of cardiomyocytes in the

Author contributions

Jingwen Chen and Yuanyuan Luo contributed equally to this paper. Jingwen Chen and Yuanyuan Luo wrote the manuscript and made the figures. Shuai Wang collected the literature. Dongye Li and Hong Zhu conceived the review and critically revised the manuscript.

Funding

This review did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors. This work was supported by the National Natural Science Foundation of China (81570326).

Declaration of Competing Interest

The authors report no conflict of interest.

References (89)

  • E. Da Silva-Ferrada et al.

    Proteostasis and SUMO in the heart

    Int. J. Biochem. Cell Biol.

    (2016)
  • Y. Kawase et al.

    Reversal of cardiac dysfunction after long-term expression of SERCA2a by gene transfer in a pre-clinical model of heart failure

    J. Am. Coll. Cardiol.

    (2008)
  • S. Sakata et al.

    Restoration of mechanical and energetic function in failing aortic-banded rat hearts by gene transfer of calcium cycling proteins

    J. Mol. Cell. Cardiol.

    (2007)
  • S. Choudhuri et al.

    Molecular targets of epigenetic regulation and effectors of environmental in fluences

    Toxicol. Appl. Pharmacol.

    (2010)
  • D. Thiagarajan et al.

    Mechanisms of transcription factor acetylation and consequences in hearts

    BBA-Mol. Basis Dis.

    (2016)
  • D.J. Herr et al.

    HDAC1 localizes to the mitochondria of cardiac myocytes and contributes to early cardiac reperfusion injury

    J. Mol. Cell. Cardiol.

    (2018)
  • M.H. Tatham et al.

    Polymeric chains of SUMO-2 and SUMO-3 are conjugated to protein substrates by SAE1/SAE2 and Ubc9

    J. Biol. Chem.

    (2001)
  • M. Tanno et al.

    Nucleocytoplasmic shuttling of the NAD+-dependent histone deacetylase SIRT1

    J. Biol. Chem.

    (2007)
  • P. Bayer et al.

    Structure determination of the small ubiquitin-related modifier SUMO-1

    J. Mol. Biol.

    (1998)
  • F. Li et al.

    Aging and dietary restriction effects on ubiquitination, sumoylation, and the proteasome in the heart

    Mech. Ageing Dev.

    (2008)
  • T. Kalogeris et al.

    Cell biology of ischemia/reperfusion injury

    Int. Rev. Cell Mol. Biol.

    (2012)
  • A.M. Labrousse et al.

    C. elegans dynamin-related protein DRP-1 controls severing of the mitochondrial outer membrane

    Mol. Cell

    (1999)
  • K.J. De Vos et al.

    Mitochondrial function and actin regulate dynamin-related protein 1-dependent mitochondrial fission

    Curr. Biol.

    (2005)
  • A. Jourdain et al.

    Mitochondrial outer-membrane permeabilization and remodelling in apoptosis

    Int. J. Biochem. Cell Biol.

    (2009)
  • Y. Shimizu et al.

    DJ-1 protects the heart against ischemia-reperfusion injury by regulating mitochondrial fission

    J. Mol. Cell. Cardiol.

    (2016)
  • L. Gao et al.

    The desumoylating enzyme sentrin-specific protease 3 contributes to myocardial ischemia reperfusion injury

    J. Genet. Genomics.

    (2018)
  • P.H. Maxwell et al.

    Oxygen sensors and angiogenesis

    Semin. Cell Dev. Biol.

    (2002)
  • G.L. Wang et al.

    Purification and characterization of Hypoxia-inducible Factor 1

    J. Biol. Chem.

    (1995)
  • M. Ikeda et al.

    cDNA cloning and tissue-specific expression of a novel basic helix-loop-helix/PAS protein (BMAL1) and identification of alternatively spliced variants with alternative translation initiation site usage 1

    Biochem. Biophys. Res. Commun.

    (1997)
  • R. Shao et al.

    Increase of SUMO-1 expression in response to hypoxia: direct interaction with HIF-1α in adult mouse brain and heart in vivo

    FEBS Lett.

    (2004)
  • J. Cheng et al.

    SUMO-specific protease 1 is essential for stabilization of HIF1α during hypoxia

    Cell.

    (2007)
  • F. Bayram et al.

    Increased genome instability and oxidative DNA damage and their association with IGF-1 levels in patients with active acromegaly

    Growth Hormon. IGF Res.

    (2014)
  • Q. Yao et al.

    C-terminal src kinase (CSK)-mediated phosphorylation of eukaryotic elongation factor 2 (EEF2) promotes proteolytic cleavage and nuclear translocation of EEF2

    J. Biol. Chem.

    (2014)
  • G. Leprivier et al.

    The eEF2 kinase confers resistance to nutrient deprivation by blocking translation elongation

    Cell.

    (2013)
  • C. Zhang et al.

    Phosphorylated eEF2 is SUMOylated and induces cardiomyocyte apoptosis during myocardial ischemia reperfusion

    J. Cardiol.

    (2017)
  • T. Maruyama et al.

    SENP1 and SENP2 regulate SUMOylation of amyloid precursor protein

    Heliyon.

    (2018)
  • H. Rodríguez-magadán et al.

    Gene expression patterns sexually dimorphic gene expression of the Zimp7 and Zimp10 genes in embryonic gonads

    Gene Expr. Patterns

    (2010)
  • V.L. Roger et al.

    Heart disease and stroke statistics-2012 update: a report from the American heart association

    Circulation

    (2012)
  • M.A. De Wood

    Prevalence of total coronary occlusion during the early hours of transmural myocardial infarction

    N. Engl. J. Med.

    (2010)
  • W.W. Brooks et al.

    Ischaemia in intact rat heart: role of intracellular calcium

    Cardiovasc. Res.

    (1995)
  • T. Okura et al.

    Protection against Fas/APO-1- and tumor necrosis factor-mediated cell death by a novel protein, sentrin

    J. Immunol.

    (1996)
  • R. Geiss-friedlander et al.

    Concepts in sumoylation: a decade on

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • D. Guo et al.

    A functional variant of SUMO4, a new IκBα modifier, is associated with type diabetes

    Nat. Genet.

    (2004)
  • Z. Xu et al.

    SUMO proteases: redox regulation and biological consequences

    Antioxid. Redox Signal.

    (2009)
  • Cited by (25)

    • SUMOylation of SIRT1 activating PGC-1α/PPARα pathway mediates the protective effect of LncRNA-MHRT in cardiac hypertrophy

      2022, European Journal of Pharmacology
      Citation Excerpt :

      Further, peroxisome proliferator-activated receptor γ coactivator-1 α (PGC-1α) and peroxisome proliferator-activated receptor-α (PPARα) are involved in cardiac energy metabolism. SUMOylation of SIRT1 significantly upregulated its downstream target PGC1-α by decreasing acetylation of PGC1-α and enhancing interaction between nuclear SIRT1 and PGC1-α (Chen et al., 2019). Whether MHRT inhibits hypertrophy by regulating the SUMOylation of SIRT1 to activate PGC1-α/PPAR-α is still unknown.

    • TANK-binding kinase 1 alleviates myocardial ischemia/reperfusion injury through regulating apoptotic pathway

      2020, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      For the past few years, percutaneous coronary intervention (PCI) develops extremely fast and has become an effective treatment to restore blood flow [5]. However, recanalization of artery can also damage or even deteriorate heart tissue, which was described as ischemia/reperfusion (I/R) injury by Jennings (1960) for the first time [6,7]. As a complicated pathophysiological process, the underlying mechanisms of MI/R injury are still elusive, including mitochondrial dysfunction, activation of apoptotic pathways, oxidative stress, calcium overload and so on [8].

    • Function and regulation of ubiquitin-like SUMO system in heart

      2023, Frontiers in Cell and Developmental Biology
    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text