Next Article in Journal
Molecular Signaling to Preserve Mitochondrial Integrity against Ischemic Stress in the Heart: Rescue or Remove Mitochondria in Danger
Previous Article in Journal
Microvalve Bioprinting of MSC-Chondrocyte Co-Cultures
Previous Article in Special Issue
Resveratrol Contrasts LPA-Induced Ovarian Cancer Cell Migration and Platinum Resistance by Rescuing Hedgehog-Mediated Autophagy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Impact of the Main Cardiovascular Risk Factors on Plasma Extracellular Vesicles and Their Influence on the Heart’s Vulnerability to Ischemia-Reperfusion Injury

by
Miłosz Majka
1,†,
Marcin Kleibert
1,† and
Małgorzata Wojciechowska
1,2,*
1
Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland
2
Invasive Cardiology Unit, Independent Public Specialist Western Hospital John Paul II, Daleka 11, 05-825 Grodzisk Mazowiecki, Poland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2021, 10(12), 3331; https://doi.org/10.3390/cells10123331
Submission received: 31 October 2021 / Revised: 20 November 2021 / Accepted: 22 November 2021 / Published: 27 November 2021
(This article belongs to the Collection Cell-to-Cell Metabolic Cross-Talk in Physiology and Pathology)

Abstract

:
The majority of cardiovascular deaths are associated with acute coronary syndrome, especially ST-elevation myocardial infarction. Therapeutic reperfusion alone can contribute up to 40 percent of total infarct size following coronary artery occlusion, which is called ischemia-reperfusion injury (IRI). Its size depends on many factors, including the main risk factors of cardiovascular mortality, such as age, sex, systolic blood pressure, smoking, and total cholesterol level as well as obesity, diabetes, and physical effort. Extracellular vesicles (EVs) are membrane-coated particles released by every type of cell, which can carry content that affects the functioning of other tissues. Their role is essential in the communication between healthy and dysfunctional cells. In this article, data on the variability of the content of EVs in patients with the most prevalent cardiovascular risk factors is presented, and their influence on IRI is discussed.

1. Introduction

Cardiovascular diseases are a major cause of death worldwide; risk estimation and interventions to reduce mortality are essential during management with patients [1]. Initiated in 1984, the Framingham Heart Study assessed the main risk factors of cardiovascular mortality presented in the SCORE charts (Systematic COronary Risk Evaluation) estimating the 10-year risk of death. It takes into account age, sex, systolic blood pressure, smoking, and total cholesterol level [2,3,4]. In addition, increasing average weight, low physical activity, concentration of low-density lipoprotein (LDL), and incidence of diabetes mellitus were identified as major modifiable risk factors in the further analysis of this study [5,6,7]. The majority of cardiovascular deaths are associated with acute coronary syndrome (ACS), especially ST-elevation myocardial infarction (STEMI) [8]. Due to improvements in diagnosis and treatment, the death rate has decreased in the last decades. Unfortunately, despite a further decline in the door-to-balloon time (the time between arrival at an emergency department and reperfusion), the one-year death rate associated with STEMI has not fallen and is on the level of around 4–6% in the latest clinical trials and analysis [9]. Early reperfusion saves the ischemic myocardium from infarction. However, it also induces a specific additional component of irreversible injury, namely ischemia-reperfusion injury (IRI), and this alone can contribute up to 40% of total infarct size following coronary artery occlusion [10]. The mechanism of IRI is not fully understood. Some risk factors make the myocardium more or less sensitive to this damage. Many pathways and molecules are involved, but a few play a crucial role (Figure 1). Further decline in mortality related to myocardial infarction (MI) depends on appropriate interventions preventing or limiting reperfusion injury [11]. One of these is ischemic conditioning (IC)–induction of short ischemic and reperfusion periods of the myocardium before the onset of MI or during reperfusion. A related method is remote ischemic conditioning (RIC), which consists of applying cycles at a remote site [12]. It is not fully understood, but there are an increasing number of studies showing that extracellular vesicles (EVs) can be a crucial player in this process. As EVs can carry content that affects the functioning of recipient cells, their role is essential in the communication between healthy and dysfunctional cells [13]. EVs are present in abundance in the circulation and play an important role in cardiovascular physiology, which they affect in a pleiotropic manner [13,14]. Despite the great interest in these particles, there is not enough data on their role in the pathophysiology and progression of cardiovascular diseases.
In this article, data on the variability of the content of EVs in patients with the most prevalent cardiovascular risk factors is presented. Moreover, the influence of EVs on reperfusion injury is discussed and their potential role in IRI is assessed.

2. Extracellular Vesicles

2.1. Nomenclature and Biogenesis

Extracellular vesicles are a very diverse group due to differences in their origin, release, content, and function, and they can be divided into three main subtypes: exosomes, microvesicles (MVs), and apoptotic bodies. Exosomes and MVs are particularly important in the context of interorgan communication, and they are referred to as EVs in this review (Figure 2). Their size varies within a large range, but it can be assumed that exosomes are vesicles ranging from 30 nm to 150 nm, while microvesicles range from 50 nm to 1 μm [20]. EVs can be released by all cell types. Exosomes are formed by the multivesicular bodies (MVBs) budding into the endosomal lumen, which then fuse with the cell membrane releasing EVs to the extracellular space [21,22,23]. This process in general is regulated by the endosomal sorting complexes required for transport (ESCRT). The formation of microvesicles relies on the outward budding of the cell membrane (Figure 2A). This complex process depends on rearrangements in the cell membrane, in proteins, in the cytoskeleton, and more, which has been discussed by others [24]. The formation route may vary from cell to cell and can follow the above pathways in different ways. These changes in biogenesis associated with various origin translate into different contents and function of individual EVs [24].

2.2. EV Characterization

EVs contain many proteins, enzymes, lipids, and nucleic acids. Their biologically active material is stored inside them or is a part of their lipid bilayer membrane, functioning as receptors or ligands [24]. Some molecules are specific for the vesicles and can be used as markers, while other molecules can be typical for the origin cell (Figure 2C,D). EVs contain a variety of lipids with bioactive or structural and trafficking functions. Their composition may change the properties of EVs, which take part in disease processes [25]. Different contents of cholesterol, sphingomyelin, phosphatidylserine, or unsaturated fatty acids in the membrane are associated with different degrees of stiffness, and thus–resistance in circulation, and the possibility of fusion with cell membranes [26]. The most intriguing content of EVs are the RNAs, especially mRNA and miRNA [27,28,29,30,31]. MicroRNAs are short non-coding RNAs of about 22 nucleotides that mediate gene silencing by guiding Argonaute proteins to target sites in the 3′ untranslated region of the mRNAs. Nucleic acids are the major source of variation of the EV, and the metabolic cell’s status affects both their profile and quantity [32]. Moreover, cells selectively package these molecules under various conditions, but the mechanism is still poorly understood. Recent studies showed that it may depend on the sequence of nucleic acids [33]. Both mRNA, which can be translated into proteins in the target cells, and miRNA keep their functions in the recipient cells. Inflammation, hypoxia, or hyperglycemia impact the miRNA profile inside EVs and may lead to changes in metabolism by activation or suppression of multiple molecular pathways [33,34]. Recent studies showed that the differences in EV status among patients may play a role in the pathogenesis of many cardiovascular diseases and can be a significant factor impacting a patient’s prognosis and outcome [35,36,37,38,39].

2.3. Uptake and Function

The interaction of EVs with cells occurs in several ways: (1) by affecting the cell surface, its receptors and ligands, (2) by implanting membrane fragments, or (3) by delivering the EV cargo to recipient cells by fusion or endocytosis [21,34] (Figure 2B). The internalization of EVs into other cells is still poorly understood, but it probably depends on factors such as the acidity of the microenvironment and the oxygen concentration [26,40,41]. The mechanism by which EVs reach specific cells needs further investigation.
The understanding of the biological function of EVs is still evolving. According to the original hypothesis, EV production was a method for cells to dispose of unnecessary material. Subsequent studies considered EVs mainly in terms of the immune system, cancers, and neurodegenerative diseases [42,43,44]. Over time, EVs became known as the new cell-to-cell communication mechanism, with superiority over other forms due to their ability to transfer bioactive cargo and horizontal gene transfer [14,20,45]. Their function may be various depending on the cellular origin. For example, EVs released by immune cells take part in antigen presentation or modulation of the immune response, while platelet-derived EVs are important factors in angiogenesis and coagulation [41]. EVs have been also described as important factors in cardiac cell-to-cell communication. Their role includes cardiac protection as well as mediating pathological changes including ischemia-reperfusion injury [46].

2.4. Vs and MI

In the case of myocardial infarction, the potential function of EVs has been best studied in the field of regenerative medicine [13,47]. It has been known for a decade that mesenchymal stem cells (MSCs) have great regenerative capabilities. EVs are the important factor responsible for this phenomenon, particularly by delivering microRNA [47,48,49,50,51,52,53,54]. Indeed, stem cell derived EVs have many promising therapeutic effects in the field of chronic coronary syndrome by attenuating reperfusion injury and remodeling as well as increasing cardiomyocyte regeneration and angiogenesis [55].
Moreover, cardiomyocytes and other cells of the heart release different types of EVs under various conditions, including ischemia. They are considered as biomarkers and are of great importance in the pathophysiology of the heart, such as post-MI fibrosis and hypertrophy [32,56,57,58].
Vicencio et al. showed that endogenous plasma exosomes expressing heat shock protein (HSP) 70 have cardioprotective effects against ischemia-reperfusion injury by the activation of the pro-survival signaling pathway involving toll-like receptor (TLR) 4 and various kinases including extracellular signal-regulated kinases, leading to the activation of HSP27 [59]. Circulating exosomes were also previously proposed as a key mediator in transferring the protection effect during RIC [60]. The mechanism of RIC is still not fully understood. Currently, neuronal and humoral signal transfer can be recognized, for which EVs and miRNA are thought to be important. The RIC procedure increases the concentration of circulating exosomes, which attenuate ischemia-reperfusion injury in rat models by transferring miRNA or activating cardioprotective pathways [59,61,62,63]. However, data from randomized controlled trials assessing the number and content of EVs after RIC in humans is inconsistent [64,65,66].
Recent studies show that the protective effect of EVs on myocardial IRI can be reduced or even inverted in the setting of disease [67,68,69]. Indeed, EVs have both harmful and beneficial effects, and some pathologies may contribute to the loss or gain of the functions of EVs [70]. Their relationship with other diseases and with well-known cardiovascular risk factors is still still not understood.

3. Cardiovascular Risk Factors

3.1. Diabetes Mellitus and Obesity

It is known that hyperglycemia or a previous diagnosis of diabetes mellitus (DM) increases susceptibility to IRI [71,72,73]. Patients with DM experience worse clinical outcomes, which partly result from more severe damage associated with reperfusion [74]. This effect may be directly related to hyperglycemia or changes in insulin levels, which has an impact on the number and content of EVs. Due to the different pathogenesis of both types of DM, it is worth discussing their effect on IRI separately. Moreover, obesity is associated with insulin resistance, and a high-fat diet animal model is commonly used to investigate the pathogenesis not only of obesity but also of type 2 diabetes. Therefore, the results of most studies concern the their collective influence.

3.1.1. DM Type 1

This type of DM results from the decreased synthesis of insulin due to the autoimmune damage of pancreatic β-cells. Basic research has shown that type 1 DM (T1DM) could enhance the IRI and attenuate the action of protective mechanisms [75,76]. Streptozotocin-induced diabetic mice had a significantly higher death rate post IRI than wild-type mice (death rate 69% vs. 9%; p < 0.05), and treatment with insulin reduced the damage size [77]. The growing body of data suggests that EVs can play a role in DM pathogenesis as an immune modulator [78,79]. The altered number and cargo of EVs in serum and urine are associated with the progression and development of complications of DM. Indeed, a higher level of HbA1c (glycated haemoglobin) is related to greater differences in EV content [80,81,82]. Studies report that serum obtained from patients with T1DM had a significantly higher number of platelet-derived, monocyte-derived, and endothelium-derived MVs than the serum from healthy volunteers [83,84]. The impact of these differences on IRI is still not understood, however studies show that MVs may play a role in remote conditioning by transporting cardioprotective signals [85]. In addition, an increase in microparticle shedding can be reduced by 10 days of aspirin therapy [86].
Expression of miRNAs inside EVs is dysregulated in T1DM patients [87,88]. Dysregulation of 316 out of 1008 total miRNAs was also observed in diabetic hearts [89]. Some of them can modify the IRI [90,91], but a further assessment of the significance of this is needed as most studies have only been conducted on streptozotocin-induced diabetic models. Furthermore, EVs loaded with miR-21 were assessed as an early biomarker of DM type 1 in children, as their number increases with the duration of the disease and this induced apoptosis of the Langerhans islets [92]. Interestingly, the same miRNA has a protective function against IRI and has been suggested as a potential pharmacological target in IRI in patients with DM type 2 [91]. On the other hand, in vitro studies showed that Langerhans islets incubated with cytokines (the model of T1DM) could release EVs loaded with miR-29b, which can promote apoptosis and the inflammatory process in reperfusion injury [93]. Moreover, EVs obtained from a similar culture can activate the T helper lymphocytes, which take part in the pathogenesis of IRI, especially in late complications such as fibrosis [94,95,96].

3.1.2. DM Type 2 and Obesity

Obesity has become an alarming worldwide health problem. Because obesity causes a crucial dysfunction of the adipose tissue metabolism, it contributes to system-wide pathologies, including type 2 diabetes and cardiovascular problems. Overweight and obesity are important factors contributing to all-cause mortality. Their association with mortality following acute myocardial infarction seems to be inverse, which is known as the obesity paradox [97]. However, overweight, obesity, and type 2 diabetes themselves increase the cardiometabolic risk. Diet or weight management is an essential component of secondary prevention. Moreover, cardiovascular risk further increases when both obesity and diabetes coexist.
Obesity affects the functioning of the body in many ways, one of which is the endocrine and paracrine function of adipose tissue [98]. It involves many bioactive mediators, such as hormones, adipokines, and inflammatory factors. They modify the functioning of the heart and its response to ischemia and reperfusion. Some hormones, such as adiponectin, leptin, TNF-α, plasminogen activator inhibitor-1, and visfatin, are associated with a higher cardiovascular risk and insulin resistance. The distribution of tissue is also an important factor in DM development, as visceral fat secretes more substances than subcutaneous fat, which can modify carbohydrate metabolism and promote insulin resistance [99,100]. In addition, chronic inflammation plays a causal role [101]. The secretion of multiple factors by hypertrophic adipocytes, such as resistin, IL-6, IL-8, and TNF-α leads to the phosphorylation of insulin-related receptors, causing the alteration of its signaling [100].
Adipocyte-derived EVs can act as a mode of communication between adipose tissues and other cells, impairing their function in obesity [102,103]. Adipose tissue is also an important source of exosomal miRNA, regulating metabolism in distant tissues, and considered to be a new form of adipokine [104]. The composition and number of EVs from adipose tissue, including their level in the blood and the profile of their miRNA, differ in obesity and diabetes [105,106,107,108]. The number of EVs is usually increased in patients with type 2 DM, even when it is analysed independently of obesity status. It also correlates with hypertension, arterial stiffness, diabetic kidney disease, and macro-/micro-angiopathy [109,110,111,112,113,114,115,116]. In addition, bariatric surgery and glycaemic control restoration alter the number and cargo of EVs [117,118].
Considering these changes, EVs have become an important element in understanding the mechanism linking obesity and type 2 diabetes with other diseases and complications in different organs [119,120,121,122,123,124,125]. Studies showed that circulating exosomal miRNA, altered by obesity and diabetes, can worsen heart function, and their inhibition has a therapeutic effect [126,127,128]. The cardioprotective effect of plasma exosomes by the HSP70/TLR4 pro-survival axis is dramatically altered in a state of hyperglycaemia as shown by Davidson et al. (2018). These and other pro-survival pathways are diminished in cardiomyocytes from diabetic rats, which would inhibit the proliferation and angiogenesis of cardiomyocytes and promote apoptosis [59,67,129,130,131]. As EVs play a protective role by mediating RIP signals, this study may also explain the failure of protection in the heart by RIP among diabetic animals [69].
As was recently shown by Gan et al. (2020), EVs mediate the detrimental effect of dysfunctional adipose tissue by exacerbating susceptibility of the myocardium to IRI [68]. Exosomes derived from the serum and adipose tissue of high-fat diet mice increase cardiomyocyte death due to ischemia-reperfusion injury. It is associated with the inhibition of antiapoptotic and cardioprotective factors, mainly 5′AMP-activated protein kinase alpha, by miR-130b-3p. The amount of this microRNA is significantly increased in the EVs, the adipose tissue, and the heart of diabetic animals. Moreover, IRI can be attenuated by the administration of the EV generation inhibitor or miR-130b-3p inhibitor, therefore targeting EV-mediated communication between the adipose tissue and the heart may be a novel strategy attenuating the diabetic exacerbation of myocardial infarction size. In contrast, EVs from lean animals had the opposite, beneficial effect by the attenuation of IRI.
There are many other dysregulated circulating miRNAs in obesity and diabetes, and their expression differs between obese patients with and without type 2 diabetes [132]. Some of them were reported to play an essential role in obesity-related heart diseases and cardiovascular complications [87,133]. A list of miRNAs, which have been reported to affect the IRI, are presented in the table below (Table 1 and Table 2). Wang et al. showed that EVs derived from the cardiomyocytes of obese diabetic rats play a role in spreading disease to neighbouring cells and inhibit angiogenesis by transferring higher levels of miR-320 and lower levels of miR-126 than healthy controls [134]. Similar changes in microRNA are observed in EVs derived from the adipose tissue and plasma of obese patients [37,132,135,136]. The overexpression of miR-320 can induce cardiomyocyte apoptosis by targeting A kinase interacting protein 1, HSP20, and insulin-like growth factor-1, whereas its suppression alleviates IRI [137,138,139]. The opposite properties are shown by miR-126, which can attenuate myocardial susceptibility to IRI, and miR-126 downregulation during reperfusion aggravates cardiomyocyte damage by increasing apoptosis [140,141,142,143]. Obesity is also associated with a lower concentration of cardioprotective miR-221 in adipose tissue-derived EVs [104,136,144]. It can prevent cardiac IRI by attenuating apoptosis and reducing infarct size [145,146]. Furthermore, adipose tissue macrophage-derived EVs contain high miR-29a levels, which can promote insulin resistance in the peripheral tissues. This molecule mediates cardiac dysfunction in obesity-related cardiomyopathy and can be responsible for more extensive reperfusion injury. Its inhibition can attenuate heart damage and post-IRI fibrosis [127,147,148,149,150,151,152]. Another important exosomal miRNA in the development of obesity-related diabetes is miR-155, which increases cardiomyocyte apoptosis and the inflammatory response in the course of IRI. Inhibition of its delivery is reported as a promising therapy against both diabetes and cardiac injury [102,131,153,154,155]. A lower miR-26a level was noted in MVs from diabetic patients, which can contribute to more severe IRI due to the weaker inhibition of PTEN (phosphatase and tensin homolog deleted on chromosome ten). It decreases the pro-survival PI3K/AKT/mTOR (phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin) signaling pathway activity and enhances apoptosis in cardiomyocytes in IRI [142,156,157]. Moreover, this molecule can reduce inflammatory cell infiltration and cytokine expression [158]. However, data showing the opposite indicates that knockdown of miR-26a can improve the cardiac function after reperfusion and can decrease cardiac apoptosis [159]. Wang et al. showed that hyperglycemia and hyperinsulinemia lead to the reduction of the cardioprotective miR-24 level [61,77]. This results in increased apoptosis, oxidative stress, a higher IRI area, and mortality [77]. Moreover, some exosomal miRNAs, which have been investigated as a diabetic biomarker (miR-23a, miR-192), can also play a role in apoptosis and oxidation stress regulation, and finally contributes to IRI [160,161,162,163].
Moreover, the therapy for hyperglycemia impacts the concentration of miRNA in EVs [164]. Insulin-induced hypoglycemia stimulates the secretion of endothelial cell-derived EVs (EVs peak in 240 min), which returns to baseline within 24 h [165]. Patients treated with metformin have miRNA profiles more similar to healthy controls than untreated patients [166]. Interestingly, metformin therapy is a well-documented form of protection against IRI in animal model. However, data from clinical trials are inconsistent [167,168,169,170]. Three months of therapy with another antidiabetic drug, acarbose, significantly decreased the number of platelet-derived MVs, which were initially elevated. This change can be associated with better outcomes after reperfusion [171,172].
Exosomal transport of all these molecules may be an important player in IRI in diabetic and obese patients and EV-targeted therapy is a promising method that may alleviate the negative effects of a dysregulated metabolism on the heart. Also, targeting the negative changes in adipose tissue during the obesity and diabetes may diminish IRI affecting the content of EVs originating from this tissue.
Table 1. A list of microRNAs which are increased in obesity and type 2 diabetes mellitus with their potential mechanisms of action and impact on ischemia-reperfusion injury.
Table 1. A list of microRNAs which are increased in obesity and type 2 diabetes mellitus with their potential mechanisms of action and impact on ischemia-reperfusion injury.
Increased
miRNAObDM2Ref.EffectThe Potential Regulatory Mechanism in IRI
miR-15bUp-[136,173,174,175]AggravatingIncreases apoptosis via downregulation of Bcl-2, MAPK3, and KCNJ2 [176,177,178]; inhibits the activity of the JAK2-STAT3 pathway, and promotes ROS production [179].
miR-23UpUp[132,160]AggravatingRegulates glutamine metabolism, promotes the transformation of BMSCs into myocardial cells, suppresses expression of Manganese superoxide dismutase, enhances mitophagy, and inhibits connexin 43 expression [161,163,180,181].
miR-34UpUp[91,136,173,182,183]AggravatingIncreases apoptosis, infarct size, and suppresses angiogenesis silencing sirtuin-1 and Wnt/β-catenin signaling pathway, suppresses cardiomyocyte proliferation and cardiac recovery post-MI regulating cell cycle activity and death via modulation of its targets, including Bcl2, Cyclin D1, and SIRT1 [90,91,184,185,186,187,188].
miR-122UpUp[132,136,173,189,190]AggravatingPromotes cardiomyocyte apoptosis via regulation of caspase 8 [191], inhibits the expression of Bcl-2, and upregulates the expression of HIF-1α, Bax, and caspase 9 via suppression of FOXP2 [192], downregulates the expression of the AKT/mTOR pathway, and upregulates the JNK/p38MAPK pathway [193].
miR-130Up *Up[68,105,132,136,173,174,194,195,196]AggravatingPromotes worse cardiac function recovery, larger infarct size, and greater cardiomyocyte apoptosis targeting AMPKα1/α2, Birc6, and Ucp3 [68], increases NFκB-mediated inflammation and TGF-β1-mediated fibrosis via inhibition of PPAR-γ expression [197].
miR-155UpUp[38,102,131,135,136,173]AggravatingIncreases cardiomyocyte apoptosis in vitro via the downregulation of HIF-1α, RNA-binding protein Quaking, and SIRT1 [153,154,198], enhances the inflammatory response through the activation of the JAK2/STAT1 pathway [155], increases ROS generation during IRI [199].
miR-192UpUp[132,136,160,173,200,201]AggravatingInduces apoptosis targeting FABP3, regulates oxidative stress in IRI [162,202]
miR-320UpUp[37,132,135,200]AggravatingIncreases infarct size and promotes apoptosis via the inhibition of AKIP1, IGF-1, HSP20, and AKT3 [137,138,139,203].
miR-483UpUp[132,204,205]AggravatingDecreases cell viability and increases apoptosis by targeting the MDM4/p53 pathway [206], promotes apoptosis via the IGF-1 signaling pathway [207,208].
miR-21Up *Up *[38,91,132,136,173,209,210]AttenuatingIncreases angiogenesis via silencing the cell death-inducing p53 target protein 1 [211], inhibits apoptosis via p38 downregulation and PI3k/Akt activation, decreases autophagy in the myocardial tissue via the AKT/mTOR pathway [91,152,212,213,214,215,216,217].
miR-26bUpUp[132,136,174,196,218]AttenuatingReduces inflammation in IRI and improves myocardial remodeling via MAPK pathway activation [219], targets High Mobility Group AT-Hook 2 suppressing MI-induced fibrosis [220,221].
miR-142-3pUpUp[105,136,195,200,201,222]AttenuatingInhibits IRI-induced cell apoptosis, autophagy, and fibrosis of the cardiomyocytes by targeting high mobility group box 1 and Rac Family Small GTPase 1 [223,224], improves cardiac function, and attenuates the myocardial inflammatory response targeting IRAK-1 [225].
miR-146Up *Up[91,135,136,144,182,195,200,209,226,227]AttenuatingProtects the myocardium from IRI by inhibition of NF-kB and TRAF6/p-p38/caspase-3 signaling pathways, targeting SMAD4, EGR1, and MED1; suppresses inflammatory cytokine production via IRAK-1 and TRAF6 [228,229,230,231,232], inhibits mitochondrial dysfunction in myocardial infarction by targeting cyclophilin D [233], regulates VEGF expression in the IRI heart [234].
miR-150Up *Up[132,136,173,182,200]AttenuatingAttenuates apoptosis and improves cardiac function via targeting Bax [235,236], reduces myocardial remodeling by downregulating Thioredoxin Interacting Protein [237].
miR-183Up-[132]AttenuatingReduces infarct size and attenuates apoptosis through repressing voltage-dependent anion channel 1 expression, regulation of NF-κB signaling pathway, and suppression of p27 which activates the PI3K/AKT/FOXO3a signaling pathway [238,239,240].
miR-486UpUp[136,174,241,242,243]AttenuatingInhibits apoptosis and improves cardiac function by suppressing PTEN expression, activating the PI3K/AKT signaling pathway [244,245], and targeting NDRG2 to inactivate the JNK/c-jun and NF-κB signaling pathways [246], promotes cardiac angiogenesis via fibroblastic MMP19-VEGFA cleavage signaling [247].
miR-29UpUp *[132,136,173,200,201]AmbiguousAggravating: Increasing apoptosis and fibrosis by suppression of Mcl-2 (Bcl-2 family), IGF-1, follistatin-like 1 protein, JAK2/STAT3 pathway, and the SIRT1/AMPK/PGC1α pathway [148,149,150,151,152,248];Attenuating: Inhibition of oxidative stress, apoptosis, and promotes the viability of cardiomyocytes with IRI in vivo model via downregulation of Cyclin T2 [249].
miR-143Up-[136,189,195]AmbiguousAggravating: Promotes cardiac ischemia-mediated mitochondrial impairment by the inhibition of protein kinase C epsilon [250], inhibits the mitosis of cardiomyocytes [251], promotes fibrosis via targeting sprouty3 [252];Attenuating: Promotes post-MI cell proliferation and reduced cell apoptosis in vitro via cyclooxygenase-2 [253]
miR-223Up *Up[136,173,196,254,255,256]AmbiguousAggravating: Increases cardiomyocyte apoptosis and oxidative stress by targeting KLF1 [257], enhances cardiac fibrosis after MI partially through targeting RASA1 [258], inhibits the angiogenesis of coronary microvascular endothelial cells in the ischemic heart [259]; Attenuating: Protects in vitro cells from hypoxia-induced apoptosis and excessive autophagy via the AKT/mTOR pathway by targeting PARP-1 [260], inhibits I/R-induced cardiac necroptosis at multiple layers [261].
(Ob—Obesity, DM2—diabetes mellitus type 2, *—indicates a contradictory finding where the miRNA was found to be down-regulated in at least one study). AKIP1—Aurora Kinase A Interacting Protein 1, AKT—protein kinase B, AMPKα1/α2—5’AMP-activated protein kinase α1/α2, Bax—Bcl-2-associated X protein, Bcl-2—B-cell lymphoma 2, Birc6—Baculoviral IAP Repeat Containing 6, EGR1—Early growth response protein 1, FABP—fatty acid binding protein, FOX—Forkhead Box Protein, HIF—hypoxia-inducible factor, HSP—heat shock protein, IGF—Insulin Growth Factor, IRAK1—Interleukin-1 receptor associated kinase, JAK—Janus Kinase, JNK—Jun N-terminal kinases, KCNJ2—Potassium Inwardly Rectifying Channel Subfamily J Member 2, KLF1—Kruppel Like Factor 1, MAPK3—Mitogen-Activated Protein Kinase 3, MDM4 – MDM4 Regulator of P53, MED1—Mediator Complex Subunit 1, MMP—matrix metalloproteinases, mTOR—mammalian target of rapamycin, NF-kB—nuclear factor kappa-light-chain-enhancer of activated B cells, NDGR2—NDRG Family Member 2, PGC-1α-Peroxisome proliferator-activated receptor gamma coactivator 1-alpha, PARP—Poly (ADP-ribose) polymerase, PI3K—Phosphoinositide 3-kinases, PPAR—peroxisome proliferator-activated receptor, RASA1—RAS P21 Protein Activator 1, ROS—reactive oxygen species, SIRT1—Sirtuin 1, SMAD4—SMAD Family Member 4, STAT—signal transducer and activator of transcription, TGF—Tumor Growth Factor, Ucp3—Uncoupling Protein 3, TRAF—TNF receptor associated factors, VEGF—Vascular endothelial growth factor.
Table 2. A list of microRNAs which are decreased in obesity and type 2 diabetes mellitus with their potential mechanisms of action and impact on ischemia-reperfusion injury.
Table 2. A list of microRNAs which are decreased in obesity and type 2 diabetes mellitus with their potential mechanisms of action and impact on ischemia-reperfusion injury.
Decreased
miRNAObDM2Ref.EffectThe Potential Regulatory Mechanism in IRI
miR-17-5pDownDown[136,189,200,201]AggravatingPromotes apoptosis induced by ER-stress and oxidative stress injury targeting TSG101 and STAT3 [262,263], increases apoptosis and vascular injury by suppressing the ERK pathway, and via downregulation of Bcl-2 in endothelium cells [264].
miR-24-Down[77,200,265]AttenuatingInhibits apoptosis and excessive O-GlcNAcylation [61,77].
miR-126DownDown[132,136,142,143,182,200,222,265]AttenuatingRegulates oxidative stress and apoptosis via downregulation of ERRFI1 expression, and decline of PI3K/AKT pathway activity [140,141,266], decreases angiogenesis [152].
miR-132Down-[136,189]AttenuatingInhibits apoptosis and ROS production via regulation of the TUG1/miR-132-3p/HDAC3 axis and through IL-1β downregulation [267,268,269], protects against oxygen and glucose deprivation via the inhibition of FOXO3a [270], enhances neovascularization [271].
miR-145DownDown[136,189,272]AttenuatingInhibits IRI-induced apoptosis via regulation of the AKT3/mTOR and CaMKII-mediated ASK1 antiapoptotic signaling pathways, ameliorates inflammation by the NF-κB p65 pathway, and the negative regulation of CD40, protects the heart through induction of autophagy [273,274,275,276,277]; absence of miR-145 results in greater infarct thinning and dilatation [278].
miR-206Down-[136,279]AttenuatingReduces IRI-induced apoptosis, targeting protein tyrosine phosphatase 1B, Gadd45β, and ATG3 (activating PI3K/Akt/mTOR pathway); reduces infarct size and improves cardiac function [236,280,281,282].
miR-221Down-[104,105,136,144,194,241]AttenuatingReduces infarct size and prevents IRI-induced apoptosis via the PUMA/ETS-1 pathway and others [145,146].
miR-26a-Down[142]AmbiguousAttenuating: Improves viability and inhibits apoptosis via regulation of the PTEN/PI3K/AKT signaling pathway, inhibition of high mobility group box 1 protein expression, inflammatory cell infiltration, and cytokine expression [156,158].
Aggravating: Possible proapoptotic mechanism via targeted regulation of the GSK3β/β-catenin signaling pathway [159]
miR-138Down-[136,175,283]AmbiguousAttenuating: Reduces infarct size and myocardial I/R-induced mitochondrial apoptosis by targeting HIF-1α [284], increases the cardiac cells’ viability under hypoxia through targeting PDK1 [285];
Aggravating: Downregulation of miR-138-5p can regulate SIRT1 to inhibit cell pyroptosis and attenuate MI progression [286], miR-138 may mediate inhibition of hypoxia-induced proliferation of endothelial progenitor cells [287].
(Ob—Obesity, DM2—diabetes mellitus type 2). AKT—protein kinase B, ASK1—apoptosis signal-regulating kinase 1, Bcl-2—B-cell lymphoma 2, CaMKII—calcium/calmodulin-dependent protein kinase II, ER-endoplasmic reticulum, ERRFI1—ERBB Receptor Feedback Inhibitor 1, ERK—extracellular signal-regulated kinase, FOX—forkhead Box Protein, GSK3β—glycogen synthase kinase-3 beta, HDAC3—histone deacetylase 3, HIF—hypoxia-inducible factor, mTOR—mammalian target of rapamycin, NF-kB—nuclear factor kappa-light-chain-enhancer of activated B cells, PDK1—Pyruvate Dehydrogenase Kinase 1 PI3K—Phosphoinositide 3-kinases, PTEN—phosphatase and tensin homolog deleted on chromosome ten, SIRT1—Sirtuin 1, STAT—signal transducer and activator of transcription, TSG101—Tumor susceptibility gene 101 protein, TUG1—Taurine up-regulated 1 Gene.

3.2. Smoking

Myocardial infarction affects smokers ten years earlier than non-smokers [288]. Recently published research demonstrated an independent and strong association between smoking and intramyocardial hemorrhage occurrence in a group of STEMI patients [289]. The intramyocardial hemorrhage is a marker of severe ischemia-reperfusion injury caused by profound structural damage of the coronary microvasculature, which leads to extravasation of erythrocytes. Studies on cell culture showed that exposure to cigarette smoke affects the release and content of EVs [290,291,292,293,294,295,296,297]. It was noticed in a small group (n = 12) that active smoking of one cigarette can cause an immediate and significant increase in the number of MVs, which may be a sign of acute vascular injury [294]. Cordazzo et al. showed that cigarette smoke extract causes increased intracellular Ca2+ concentration, resulting in a higher generation of MVs [298]. In addition, CS-induced EVs may be enriched in MMP-14, proinflammatory cytokines (IL-1, IL-6, IL-8, MCP-1), procoagulant tissue factor, and phosphatidylserine which can cause destabilization of the atherosclerotic plaque, rapture, and occlusion of the artery as a consequence [299,300,301,302,303,304]. These vesicles can potentially enhance the IRI due to stimulation of inflammasome formation, which can contribute to pyroptosis and other types of cell death [305,306].
Cigarette smoke exposure may also impact the level of miRNA circulating inside EVs. For example, Fujita et al. noticed the upregulation of miR-210 which inhibits autophagy and exacerbates the IRI [307]. In another study, cigarette smoke-induced EVs were enriched in let-7d, miR-191, miR-126, and miR125a. The last two can alleviate the IRI by reducing apoptosis and the myocardial area after reperfusion, as mentioned above [308,309] (Table 1). The other two miRNA should be investigated as they may play a role in the IRI of other organs or may be important factors in cardiovascular diseases [310,311,312]. Further assessment of human serum has revealed that miR-29b is significantly increased and miR-223-3p is significantly decreased in smokers [313]. The role of these mi-RNAs is not clear. However, inhibition of miR-29b by dexmedetomidine was investigated and showed that it can reduce the IRI [314]. In addition, Zhang et al. noticed that LncRNA H19, which can inhibit miR-29b, decreases the reperfusion damage due to mediating the antiapoptotic effect of postconditioning [315]. Overexpression of miR-223 promotes oxidative stress, which can induce apoptosis by inhibiting the expression of FOXO3a [316]. On the other hand, this miRNA can have a cardioprotective effect. Some authors have shown that it could reduce IRI by suppressing necroptosis, apoptosis, and oxidative stress [257,260,261]. Other investigators, however, have not observed any significant difference in miRNA expression between smokers and non-smokers [317].
Studies show that also vaping is harmful to the endothelium, and it can cause a change in the concentration and content of EVs [318,319]. More studies are needed to assess the impact of substances released by electronic devices on IRI.

3.3. Total Cholesterol Level

The number of EVs varies depending on the cholesterol level [122,320]. This correlation may be a result of obesity which is common among patients with hypercholesterolemia. Hypertensive-hypercholesterolemic hamsters have approximately 20 times the number of MVs than the control group with similar body mass, which suggests that the number of EVs can be independent of weight [321]. Nijiati et al. noticed that HDL-C levels and MVs are negatively correlated in humans after acute coronary syndrome [322]. In addition, incubation of platelets with native HDL3 reduces the number of MVs released [323]. Patients with familial hypercholesterolemia and increased oxidized LDL cholesterol levels have a higher number of pro-inflammatory MVs, which can promote fibrosis and necrosis after reperfusion [324].
One of the interventions used to reduce the cardiovascular risk associated with increased cholesterol level is statin therapy. The impact of this treatment was investigated in some studies. Two randomized trials have shown that perioperative therapy with statins can reduce myocardial IRI, but a large-scale study failed to confirm this. [325,326,327]. This effect may be mediated by EVs because, as some authors suggest, statin therapy impacts the level of EVs and consequently the IRI, but again this correlation has not been observed in all studies [328,329,330,331,332,333]. Interestingly, Kocsis et al. showed in an animal study that chronic and acute lovastatin therapy could have a different effect on conditioning. They noticed that chronic therapy decreased the preconditioning efficacy, whereas acute therapy decreased postconditioning efficacy [334]. A lower number of MVs, especially platelets, leukocytes, and endothelial cell-derived MVs have been demonstrated in patients treated with statins (simvastatin, rosuvastatin, atorvastatin) than in untreated patients [328]. This effect was not observed in diabetic patients on pitavastatin therapy [329]. The influence of statins on the shedding of MVs is greater with years of treatment [328]. Also, the effect of statins seems to be dose-dependent. It was shown that the number of MVs was significantly decreased in the 40-mg atorvastatin group compared with the 10-mg atorvastatin group (1683.75 ± 118.5 vs. 1847.98 ± 180.26 after one-year observation, baseline—2098.56 ± 223.73 vs. 2020.36 ± 206.94) [331]. As mentioned above, MVs may play an essential role in IRI, but this should be assessed by considering their origin and cargo, and not only their number.
Simvastatin inhibits exosome production by reducing the levels of Alix, the protein that positively regulates the secretion of EVs [333,335]. In addition, this treatment led to a reduction in the levels of miR-150, which seems to be a key driver of endothelial migration (this process is an integral part of atherosclerotic plaque progression), which can make a patient more prone to MI [333,336]. And miR-150 may also be a regulator of cardiomyocyte survival during cardiac injury by directly repressing the proapoptotic gene EGR2 and reducing cell death after reperfusion [337].
Total cholesterol level and especially LDL level are essential cardiovascular risk factors that lead to death due to CV events. It is well known that statins have a cardioprotective effect, but their impact on IRI, especially with regard to EVs, still requires further studies.

3.4. Systolic Blood Pressure

Although arterial hypertension is one of the most common risk factors of higher mortality due to myocardial infarction, its impact on IRI is poorly studied [338]. Both hypertension and left ventricular hypertrophy induced by elevated arterial pressure are associated with increased myocardium vulnerability to IRI [339,340]. However, antecedent hypertension has no impact on reperfusion efficacy, infarct size, and reperfusion injury in magnetic resonance imaging, and is not significantly associated with one-year mortality [341,342]. Liu et al. characterized the differences in exosomal miRNA between hypertensive and normotensive rats using next-generation sequencing. They noticed that 27 types have significantly different expressions between the groups. Three of them—miR-17-5p, miR-15b-5p, and miR-486-5p—are strongly related to IRI’s myocardial susceptibility in animal studies. miR-17-5p and miR-15b-5p are overexpressed, whereas expression of miR-486-5p is decreased. miR-17-5p and miR-15b-5p aggravate IRI by activation of the MAPK/ERK pathway (mitogen-activated protein kinase/extracellular signal-regulated kinase) or by inhibition of antiapoptotic Potassium Inwardly Rectifying Channel Subfamily J Member 2 while miR-486-5p attenuates IRI by activation of the PI3K/Akt/mTOR pathway [178,244,245,262,264]. Therefore, circulating exosomal miRNA should be studied as a potential link between systolic blood pressure and increased susceptibility of the myocardium to IRI among hypertensive individuals.

3.5. Physical Effort

The lack of physical activity is a strong predictor of mortality, contributing to occurrence myocardial infarction. Physical exercise has many positive effects on cardiovascular health. Exercise training protects the myocardium against ischemia-reperfusion injury, reduces myocardial oxidative damage and improves cardiac function after IRI onset [343,344,345]. Various factors derived from the heart and other tissues are responsible for this effect, but much remains to be clarified. Physical activity increases the level of cardioprotective polypeptides in the heart, such as irisin, FGF21, and neuregulin, as well as non-coding RNA, such as miR-133. Physical activity also decreases the expression of miR-208, which can aggravate IRI [346]. The protective effect of exercise is also mediated by circulating transferable factors in plasma [347]. Recent studies have shown that EVs are one of those factors [346,348,349]. Exercise affects both the composition and the amount of EVs circulating in the serum [349,350,351,352]. The effect varies according to the type of activity as well as its duration and intensity [353]. Exercise-induced EVs attenuate myocardial ischemia-reperfusion injury and studies conducted so far indicate that these mechanisms are related to proteins and miRNAs. Bei et al. showed that the short-term beneficial effect of exercise on myocardial IRI is mainly caused by the increased number of circulating EVs and mediated by the activation of ERK1/2 and HSP27 signaling [59,348].
However, long-term protection provided by exercise-derived circulating EVs is also possible, and it is associated with differentially expressed miRNA. Long-term exercise training increases the level of exosomal miR-342-5p, which inhibits myocardial IRI-induced apoptosis targeting Caspase 9 and JNK2 and enhances survival signaling by PPM1f in cardiomyocytes [354]. Physical activity changes the expression of many other circulating and exosomal miRNAs [355,356,357,358,359], increasing some with a protective function against IRI, such as miR-17-3p, miR-25-3p, and miR-93-5p [360,361,362], or decreasing those aggravating IRI. The impact of exercise on many muscle-specific microRNAs with a significant role in IRI, such as miR-1, miR-21, miR-133, and miR-208, is still unclear and probably dependent on the type of activity [346,357,363,364]. Therefore, EVs are an important vehicle delivering exercise-dependent factors from distant and local tissues to cardiomyocytes. As endurance exercise-derived EVs have a therapeutic potential for metabolic diseases, they have become essential factors in the secondary prevention of myocardial infarction [365,366,367].

3.6. Sex

Males are more susceptible to cardiovascular diseases than females [368]. The impact of sex on the content of EVs has been rarely assessed. There are only a few analyses in which researchers evaluated these groups separately. EVs are more abundant in healthy women than men, but some studies have shown the opposite results [369,370,371]. The number of procoagulants microvesicles (phosphatidylserine and P-selectin positive) is higher in apparently healthy premenopausal women compared with age-matched men (32 ± 8.5 year-old women and 29 ± 3.2 year-old men), but this can be associated with anticonceptive therapy [369]. The growing body of data supports the idea that MVs can play a role in IRI [85,372,373]. The platelet-derived-MVs, which are also increased in age-matched females compared with males, can have a cardioprotective effect [372].
The cargo of EVs differs between the sexes. Bammert et al. showed that miR-125a expression was lower (∼215%; p < 0.05), and miR-34a was higher (∼210%; p < 0.05) in the EV of men than in women [374]. MiR-125a has a potential cardioprotective effect by ameliorating cardiomyocyte autophagy and oxidative injury. These mechanisms are activated by promoting gene expression dysregulation involved in cell death and apoptosis [308,309]. Furthermore, miR-34a, which is also dysregulated in DM and obesity, may play a role in IRI. The artificial inhibition of this miRNA can reduce the damage associated with reperfusion through the SIRT1 protective pathway [90]. This data supports the thesis that men are more susceptible to IRI than women. This can also be part of the explanation why the risk of death due to CV diseases is higher in men.
A notable increase has been observed in cardiovascular risk in women during midlife, a period coincident with menopause transition [375]. The differences between pre-menopausal and post-menopausal women are rarely investigated. The climacteric lowers the level of platelet-derived MVs but has no impact on the number of endothelial-derived MVs [376]. The effect of it on IRI is still not verified.
Interestingly, pregnancy also has an impact on the number and content of EVs. These differences play a role not only in embryo implantation and development [377], but can also regulate the function of the heart [378]. For example, EVs obtained from amniotic fluid mesenchymal stromal cells can reduce the IRI by stimulation of endothelial cell migration [379]. However, pregnant rats are more prone to myocardial IRI than non-pregnant rats due to a higher generation of ROS and activation of apoptosis [380]. The exact role of EVs among pregnant women in IRI is still under investigation.

3.7. Age

There is growing evidence that EVs can play a role in aging and age-related diseases [381]. Basic research has shown that age-related changes in the composition of EVs have an undisputed impact on IRI size.
For example, miR-128-3p in EVs (which inhibit the SMAD5 gene) was markedly upregulated in EVs obtained from older mice, and its inhibition by tongxinluo (a traditional Chinese medicine) reduced IRI in the cell culture of human cardiomyocytes [382,383]. The mechanism is still under investigation, but some data suggest that it can promote the Reperfusion Injury Salvage Kinase (RISK) pathway, which has a cardioprotective effect, by increasing serine/threonine kinase p70s6k1 expression [382,384].
The IL-1 signaling pathway plays an important role in IRI’s multiple mechanisms, including inflammation and apoptosis [385]. It was shown that older rats had a higher level of this protein in their EVs, which can be partly linked to age-related inflammatory conditions [386]. Administration of recombinant human IL-1 receptor antagonist significantly reduces cardiac fibrosis and increases myocardium viability in rats and mice [387,388,389]. It was recently demonstrated that a higher IL-1β level at admission in patients with acute MI was independently associated with the risk of mortality and recurrent MACEs (major adverse cardiovascular events), resulting from greater IRI susceptibility [390].
Fafián-Labora et el. observed a negative correlation between miR-146a, miR-132, and miR-155 levels in EVs and age. It was related to the downregulation of TLR4 in the targeted cells, which is a part of inflammaging [391]. This process can contribute to inflammation imbalance (a decline in the ability to respond to a new pathogen during infection with simultaneously increasing levels of proinflammatory cytokines such as IL-1), which can play a significant role in IRI due to increased apoptosis and fibrosis [392,393]. Unfortunately, there are only a few studies, mentioned above, which have investigated the direct relationship between inflammation and reperfusion injury.
Altered ROS removal appears to be the main reason for increased susceptibility to IRI [394]. EV-mediated delivery ofextracellular nicotinamide phosphoribosyltransferase decreases with aging and promotes the systemic decrease of the NAD+ level due to lower biosynthesis and higher consumption. As a consequence of these changes, ROS elimination is reduced, and oxidative stress is increased, which promotes IRI [395,396,397,398,399].
Only a few studies have evaluated the changes in the concentration and content of EVs with aging in humans. Recently, one research group noted a significant decline in EV concentration with age and increasing internalization by the cells. This change was also related to differences in content. They observed that p53, cleaved caspase-3, and PARP-1 decreased with age when some immune-related antigens such as MUC1, MUCIN-14, NY-ESO, CD-14, and PDL were increased. Apart from comparing these parameters between age groups, the authors checked them in individuals after five years and did not find significant differences in the concentration of 39 out of 46 assessed proteins in EVs. This means that the content is probably more a feature of an individual than the number is. However, the number of EVs significantly declined over this period [293,400]. Bæk et al. showed that the estrogen receptor level in EVs also decreased with aging for both genders, which is statistically significant only among men [293]. It would appear that these changes may be the recently discovered reason for the higher susceptibility of men and the elderly to IRI [12,401].
Finally, it was demonstrated that the mtDNA level encapsulated in EVs also decreases with age. Lazo et al. showed that the addition of EVs obtained from young donors to HeLa cells increases basal and maximal respiration levels. The rise was higher in comparison with cells incubated with EVs from older donors [402]. The data suggest that EVs support the mitochondrial function, and this regulation can change with aging thus influencing the IRI size [403]. The Galectin-3 (Gal-3) concentration was reduced in vesicles derived from the elderly [404]. Studies on animals showed that this protein has a cardioprotective function, and knock-out mice have increased apoptosis and decreased antioxidant defenses compared with WT (wild-type) mice at 24 h after reperfusion. The expression of proinflammatory proteins decreased in Gal-3 knockout mice compared with WT mice [405]. Longer observation (8 days) revealed that, compared with non-treated animals, the inhibition of Gal-3 by modified citrus peptide lowered myocardial inflammation and reduced fibrosis. The perfusion and compliance assessed by MRI was improved, and finally no differences in LV hypertrophy was observed [406]. This paradox is very interesting and suggests multiple roles of Gal-3 in IRI.
The impact of aging on EVs is not in dispute, but the role of these changes on the cardiovascular system should draw the attention of researchers. Further studies may help to find interventions, associated with EVs, that can decrease the risk of cardiovascular death among older people.

4. Conclusions

The results of both experimental and clinical studies indicate that modifiable as well as non-modifiable cardiovascular risk factors change the number and content of EVs of various origin. These differences may have a negative impact on intercellular signalling, thus influencing the vulnerability of the heart to IRI. There are many potential components of EVs that may participate in this complicated crosstalk between the peripheral tissue and the heart, however, miRNAs seem to be the most important (Figure 3). Also, some cellular origins of EVs like adipose tissue appear to be more significant than the others.
Based on this information, it can be concluded that non-pharmacological interventions such as physical activity, body weight reduction, smoke cessation, and better control of systolic blood pressure may have beneficial effects on the myocardium and its susceptibility to IRI by modification of the composition and quantity of EVs. Moreover, EVs may be a new target for long-term management in secondary prophylaxis of acute coronary syndrome. Perhaps, the detection of circulating EVs for the prediction and prognosis of patient outcome after MI onset or strategies of blocking abnormal production of EVs could have great clinical implications in the future. However, randomized-controlled trials are needed to verify this hypothesis.

Author Contributions

Conceptualization—M.M. and M.K., resources—M.M. and M.K.; writing—original draft preparation—M.M. and M.K.; writing—review and editing M.M., M.K. and M.W.; visualization, M.M. and M.K.; supervision, M.W.; funding acquisition, M.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Medical University of Warsaw (1MA/N/2021).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jones, D.S.; Podolsky, S.H.; Greene, J.A. The burden of disease and the changing task of medicine. N. Engl. J. Med. 2012, 366, 2333–2338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Dawber, T.R.; Meadors, G.F.; Moore, F.E., Jr. Epidemiological approaches to heart disease: The framingham study. Am. J. Public Health 1951, 41, 279–286. [Google Scholar] [CrossRef] [PubMed]
  3. Piepoli, M.F.; Hoes, A.W.; Agewall, S.; Albus, C.; Brotond, C.; Capatano, A.L.; Cooney, M.-T.; Cosyns, B.; Deaton, C.; Graham, I.; et al. European guidelines on cardiovascular disease prevention in clinical practice: The sixth joint task force of the european society of cardiology and other societies on cardiovascular disease prevention in clinical practice (constituted by representatives of 10 societies and by invited experts)developed with the special contribution of the european association for cardiovascular prevention & rehabilitation (EACPR). Eur. Heart J. 2016, 37, 2315–2381. [Google Scholar] [CrossRef] [PubMed]
  4. Pooling Project Research Group. Relationship of blood pressure, serum cholesterol, smoking habit, relative weight and ECG abnormalities to incidence of major coronary events: Final report of the pooling project. J. Chronic Dis. 1978, 31, 201–306. [Google Scholar] [CrossRef]
  5. Morris, J.; Heady, J.; Raffle, P.; Roberts, C.; Parks, J. Coronary heart-disease and physical activity of work. Lancet 1953, 262, 1053–1057. [Google Scholar] [CrossRef] [Green Version]
  6. Wilson, P.W.; D’Agostino, R.B.; Sullivan, L.; Parise, H.; Kannel, W.B. Overweight and obesity as determinants of cardiovascular risk: The Framingham experience. Arch. Intern. Med. 2002, 162, 1867–1872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Goldschmid, M.G.; Barrett-Connor, E.; Edelstein, S.L.; Wingard, D.L.; Cohn, B.A.; Herman, W.H. Dyslipidemia and ischemic heart disease mortality among men and women with diabetes. Circulation 1994, 89, 991–997. [Google Scholar] [CrossRef] [Green Version]
  8. Ibanez, B.; James, S.; Agewall, S.; Antunes, M.J.; Bucciarelli-Ducci, C.; Bueno, H.; Caforio, A.L.P.; Crea, F.; Goudevenos, J.A.; Halvorsen, S.; et al. 2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: The Task Force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC). Eur. Heart J. 2018, 39, 119–177. [Google Scholar] [CrossRef] [Green Version]
  9. Hofmann, R.; Witt, N.; Lagerqvist, B.; Jernberg, T.; Lindahl, B.; Erlinge, D.; Herlitz, J.; Alfredsson, J.; Linder, R.; Omerovic, E.; et al. Oxygen therapy in ST-elevation myocardial infarction. Eur. Heart J. 2018, 39, 2730–2739. [Google Scholar] [CrossRef]
  10. Abe, H.; Sakurai, A.; Ono, H.; Hayashi, S.; Yoshimoto, S.; Ochi, A.; Ueda, S.; Nishimura, K.; Shibata, E.; Tamaki, M.; et al. Urinary exosomal mRNA of WT1 as diagnostic and prognostic biomarker for diabetic nephropathy. J. Med. Investig. 2018, 65, 208–215. [Google Scholar] [CrossRef] [Green Version]
  11. Hausenloy, D.; Botker, H.E.; Engstrom, T.; Erlinge, D.; Heusch, G.; Ibanez, B.; Kloner, R.A.; Ovize, M.; Yellon, D.; Garcia-Dorado, D. Targeting reperfusion injury in patients with ST-segment elevation myocardial infarction: Trials and tribulations. Eur. Heart J. 2017, 38, 935–941. [Google Scholar] [CrossRef] [Green Version]
  12. Heusch, G. Myocardial ischaemia–reperfusion injury and cardioprotection in perspective. Nat. Rev. Cardiol. 2020, 17, 773–789. [Google Scholar] [CrossRef]
  13. El Andaloussi, S.; Mäger, I.; Breakefield, X.O.; Wood, M.J.A. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 2013, 12, 347–357. [Google Scholar] [CrossRef]
  14. Ludwig, A.-K.; Giebel, B. Exosomes: Small vesicles participating in intercellular communication. Int. J. Biochem. Cell Biol. 2012, 44, 11–15. [Google Scholar] [CrossRef] [PubMed]
  15. Oerlemans, M.I.F.J.; Liu, J.; Arslan, F.; Ouden, K.D.; van Middelaar, B.J.; Doevendans, P.A.; Sluijter, J.P.G. Inhibition of RIP1-dependent necrosis prevents adverse cardiac remodeling after myocardial ischemia-reperfusion in vivo. Basic Res. Cardiol. 2012, 107, 1–13. [Google Scholar] [CrossRef]
  16. Tani, M.; Neely, J.R. Role of intracellular Na+ in Ca2+ overload and depressed recovery of ventricular function of reperfused ischemic rat hearts. Possible involvement of H+-Na+ and Na+-Ca2+ exchange. Circ. Res. 1989, 65, 1045–1056. [Google Scholar] [CrossRef] [Green Version]
  17. Liebert, A.; Krause, A.; Goonetilleke, N.; Bicknell, B.; Kiat, H. A Role for photobiomodulation in the prevention of myocardial ischemic reperfusion injury: A systematic review and potential molecular mechanisms. Sci. Rep. 2017, 7, 42386. [Google Scholar] [CrossRef] [Green Version]
  18. Bernardi, P.; Rasola, A.; Forte, M.; Lippe, G. The mitochondrial permeability transition pore: Channel formation by f-atp synthase, integration in signal transduction, and role in pathophysiology. Physiol. Rev. 2015, 95, 1111–1155. [Google Scholar] [CrossRef]
  19. Bernardi, P.; Di Lisa, F. The mitochondrial permeability transition pore: Molecular nature and role as a target in cardioprotection. J. Mol. Cell. Cardiol. 2015, 78, 100–106. [Google Scholar] [CrossRef] [PubMed]
  20. Doyle, L.M.; Wang, M.Z. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [Green Version]
  21. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [Green Version]
  22. Ostrowski, M.; Carmo, N.B.; Krumeich, S.; Fanget, I.; Raposo, G.; Savina, A.; Moita, C.F.; Schauer, K.; Hume, A.N.; Freitas, R.P.; et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 2010, 12, 19–30. [Google Scholar] [CrossRef] [Green Version]
  23. Wollert, T.; Hurley, J.H. Molecular mechanism of multivesicular body biogenesis by ESCRT complexes. Nature 2010, 464, 864–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  25. Record, M.; Carayon, K.; Poirot, M.; Silvente-Poirot, S. Exosomes as new vesicular lipid transporters involved in cell–cell communication and various pathophysiologies. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2014, 1841, 108–120. [Google Scholar] [CrossRef]
  26. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular vesicles: Composition, biological relevance, and methods of study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [Green Version]
  27. Valadi, H.; Ekstrom, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Nolte-’t Hoen, E.N.; Buermans, H.P.J.; Waasdorp, M.; Stoorvogel, W.; Wauben, M.H.M.; ’t Hoen, P.A. Deep sequencing of RNA from immune cell-derived vesicles uncovers the selective incorporation of small non-coding RNA biotypes with potential regulatory functions. Nucleic Acids Res. 2012, 40, 9272–9285. [Google Scholar] [CrossRef] [Green Version]
  29. Ratajczak, J.; Miękus, K.; Kucia, M.; Zhang, J.; Reca, R.; Dvorak, P.; Ratajczak, M.Z. Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: Evidence for horizontal transfer of mRNA and protein delivery. Leukemia 2006, 20, 847–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Xu, L.; Yang, B.-F.; Ai, J. MicroRNA transport: A new way in cell communication. J. Cell. Physiol. 2013, 228, 1713–1719. [Google Scholar] [CrossRef]
  31. Villarroya-Beltri, C.; Gutierrez-Vazquez, C.; Sanchez-Cabo, F.; Pérez-Hernández, D.; Vázquez, J.; Martin-Cofreces, N.; Martinez-Herrera, D.J.; Pascual-Montano, A.; Mittelbrunn, M.; Sánchez-Madrid, F. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat. Commun. 2013, 4, 2980. [Google Scholar] [CrossRef] [Green Version]
  32. Ibrahim, A.; Marbán, E. Exosomes: Fundamental biology and roles in cardiovascular physiology. Annu. Rev. Physiol. 2016, 78, 67–83. [Google Scholar] [CrossRef] [Green Version]
  33. Groot, M.; Lee, H. Sorting mechanisms for microRNAs into extracellular vesicles and their associated diseases. Cells 2020, 9, 1044. [Google Scholar] [CrossRef] [PubMed]
  34. Maas, S.L.N.; Breakefield, X.O.; Weaver, A.M. Extracellular vesicles: Unique intercellular delivery vehicles. Trends Cell Biol. 2017, 27, 172–188. [Google Scholar] [CrossRef] [Green Version]
  35. Gartz, M.; Strande, J.L. Examining the paracrine effects of exosomes in cardiovascular disease and repair. J. Am. Heart Assoc. 2018, 7, e007954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Akbar, N.; Azzimato, V.; Choudhury, R.P.; Aouadi, M. Extracellular vesicles in metabolic disease. Diabetologia 2019, 62, 2179–2187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Ferrante, S.C.; Nadler, E.P.; Pillai, D.K.; Hubal, M.; Wang, Z.; Wang, J.M.; Gordish-Dressman, H.; Koeck, E.; Sevilla, S.; Wiles, A.A.; et al. Adipocyte-derived exosomal miRNAs: A novel mechanism for obesity-related disease. Pediatr. Res. 2015, 77, 447–454. [Google Scholar] [CrossRef]
  38. Eguchi, A.; Lazic, M.; Armando, A.M.; Phillips, S.A.; Katebian, R.; Maraka, S.; Quehenberger, O.; Sears, D.D.; Feldstein, A.E. Circulating adipocyte-derived extracellular vesicles are novel markers of metabolic stress. J. Mol. Med. 2016, 94, 1241–1253. [Google Scholar] [CrossRef]
  39. Shah, R.; Patel, T.; Freedman, J.E. Circulating extracellular vesicles in human disease. N. Engl. J. Med. 2018, 379, 958–966. [Google Scholar] [CrossRef] [PubMed]
  40. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef] [PubMed]
  41. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Liejendekker, R.; Harding, C.V.; Melief, C.J.; Geuze, H.J. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 1996, 183, 1161–1172. [Google Scholar] [CrossRef]
  43. Zitvogel, L.; Regnault, A.; Lozier, A.; Wolfers, J.; Flament, C.; Tenza, D.; Ricciardi-Castagnoli, P.; Raposo, G.; Amigorena, S. Eradication of established murine tumors using a novel cell-free vaccine: Dendritic cell derived exosomes. Nat. Med. 1998, 4, 594–600. [Google Scholar] [CrossRef]
  44. Bellingham, S.A.; Guo, B.B.; Coleman, B.M.; Hill, A.F. Exosomes: Vehicles for the transfer of toxic proteins associated with neurodegenerative diseases? Front. Physiol. 2012, 3, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Lee, Y.; El Andaloussi, S.; Wood, M.J. Exosomes and microvesicles: Extracellular vesicles for genetic information transfer and gene therapy. Hum. Mol. Genet. 2012, 21, R125–R134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Zhang, J.; Cui, X.; Guo, J.; Cao, C.; Zhang, Z.; Wang, B.; Zhang, L.; Shen, D.; Lim, K.; Tang, J.; et al. Small but significant: Insights and new perspectives of exosomes in cardiovascular disease. J. Cell. Mol. Med. 2020, 24, 8291–8303. [Google Scholar] [CrossRef] [PubMed]
  47. Latifkar, A.; Hur, Y.H.; Sanchez, J.C.; Cerione, R.A.; Antonyak, M.A. New insights into extracellular vesicle biogenesis and function. J. Cell Sci. 2019, 132, jcs222406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Arslan, F.; Lai, R.C.; Smeets, M.B.; Akeroyd, L.; Choo, A.; Aguor, E.N.E.; Timmers, L.; Van Rijen, H.V.; Doevendans, P.A.; Pasterkamp, G.; et al. Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res. 2013, 10, 301–312. [Google Scholar] [CrossRef] [Green Version]
  49. Teng, X.; Chen, L.; Chen, W.; Yang, J.; Yang, Z.; Shen, Z. Mesenchymal stem cell-derived exosomes improve the microenvironment of infarcted myocardium contributing to angiogenesis and anti-inflammation. Cell. Physiol. Biochem. 2015, 37, 2415–2424. [Google Scholar] [CrossRef] [PubMed]
  50. Qiu, G.; Zheng, G.; Ge, M.; Wang, J.; Huang, R.; Shu, Q.; Xu, J. Mesenchymal stem cell-derived extracellular vesicles affect disease outcomes via transfer of microRNAs. Stem Cell Res. Ther. 2018, 9, 320. [Google Scholar] [CrossRef]
  51. Lai, R.C.; Arslan, F.; Lee, M.M.; Sze, N.S.K.; Choo, A.; Chen, T.S.; Salto-Tellez, M.; Timmers, L.; Lee, C.N.; El Oakley, R.M.; et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010, 4, 214–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Ibrahim, A.G.-E.; Cheng, K.; Marbán, E. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Rep. 2014, 2, 606–619. [Google Scholar] [CrossRef] [Green Version]
  53. Wang, Y.; Zhang, L.; Li, Y.; Chen, L.; Wang, X.; Guo, W.; Zhang, X.; Qin, G.; He, S.-H.; Zimmerman, A.; et al. Exosomes/microvesicles from induced pluripotent stem cells deliver cardioprotective miRNAs and prevent cardiomyocyte apoptosis in the ischemic myocardium. Int. J. Cardiol. 2015, 192, 61–69. [Google Scholar] [CrossRef] [Green Version]
  54. Miyahara, Y.; Nagaya, N.; Kataoka, M.; Yanagawa, B.; Tanaka, K.; Hao, H.; Ishino, K.; Ishida, H.; Shimizu, T.; Kangawa, K.; et al. Monolayered mesenchymal stem cells repair scarred myocardium after myocardial infarction. Nat. Med. 2006, 12, 459–465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Chen, G.H.; Xu, J.; Yang, Y.J. Exosomes: Promising sacks for treating ischemic heart disease? Am. J. Physiol. Heart Circ. Physiol. 2017, 313, H508–H523. [Google Scholar] [CrossRef] [PubMed]
  56. Sluijter, J.P.G.; Verhage, V.; Deddens, J.C.; Akker, F.V.D.; Doevendans, P.A. Microvesicles and exosomes for intracardiac communication. Cardiovasc. Res. 2014, 102, 302–311. [Google Scholar] [CrossRef] [Green Version]
  57. Yu, X.; Deng, L.; Wang, D.; Li, N.; Chen, X.; Cheng, X.; Yuan, J.; Gao, X.; Liao, M.; Wang, M.; et al. Mechanism of TNF-alpha autocrine effects in hypoxic cardiomyocytes: Initiated by hypoxia inducible factor 1alpha, presented by exosomes. J. Mol. Cell. Cardiol. 2012, 53, 848–857. [Google Scholar] [CrossRef] [PubMed]
  58. Bang, C.; Batkai, S.; Dangwal, S.; Gupta, S.K.; Foinquinos, A.; Holzmann, A.; Just, A.; Remke, J.; Zimmer, K.; Zeug, A.; et al. Cardiac fibroblast–derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J. Clin. Investig. 2014, 124, 2136–2146. [Google Scholar] [CrossRef]
  59. Vicencio, J.M.; Yellon, D.M.; Sivaraman, V.; Das, D.; Boi-Doku, C.; Arjun, S.; Zheng, Y.; Riquelme, J.A.; Kearney, J.; Sharma, V.; et al. Plasma exosomes protect the myocardium from ischemia-reperfusion injury. J. Am. Coll. Cardiol. 2015, 65, 1525–1536. [Google Scholar] [CrossRef] [Green Version]
  60. Giricz, Z.; Varga, Z.V.; Baranyai, T.; Sipos, P.; Pálóczi, K.; Kittel, Á.; Buzás, E.I.; Ferdinandy, P. Cardioprotection by remote ischemic preconditioning of the rat heart is mediated by extracellular vesicles. J. Mol. Cell. Cardiol. 2014, 68, 75–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Minghua, W.; Zhijian, G.; Chahua, H.; Qiang, L.; Minxuan, X.; Luqiao, W.; Weifang, Z.; Peng, L.; Biming, Z.; Lingling, Y.; et al. Plasma exosomes induced by remote ischaemic preconditioning attenuate myocardial ischaemia/reperfusion injury by transferring miR-24. Cell Death Dis. 2018, 9, 320. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, J.; Zhang, X. Ischaemic preconditioning-induced serum exosomes protect against myocardial ischaemia/reperfusion injury in rats by activating the PI3K/AKT signalling pathway. Cell Biochem. Funct. 2020, 39, 287–295. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, Q.; Huang, M.; Wu, J.; Jiang, Q.; Zheng, X. Exosomes isolated from the plasma of remote ischemic conditioning rats improved cardiac function and angiogenesis after myocardial infarction through targeting Hsp70. Aging 2020, 12, 3682–3693. [Google Scholar] [CrossRef] [PubMed]
  64. Haller, P.M.; Jäger, B.; Piackova, E.; Sztulman, L.; Wegberger, C.; Wojta, J.; Gyöngyösi, M.; Kiss, A.; Podesser, B.K.; Spittler, A.; et al. Changes in circulating extracellular vesicles in patients with st-elevation myocardial infarction and potential effects of remote ischemic conditioning—A randomized controlled trial. Biomedicines 2020, 8, 218. [Google Scholar] [CrossRef] [PubMed]
  65. Frey, U.H.; Klaassen, M.; Ochsenfarth, C.; Murke, F.; Thielmann, M.; Kottenberg, E.; Kleinbongard, P.; Klenke, S.; Engler, A.; Heusch, G.; et al. Remote ischaemic preconditioning increases serum extracellular vesicle concentrations with altered micro-RNA signature in CABG patients. Acta Anaesthesiol. Scand. 2019, 63, 483–492. [Google Scholar] [CrossRef]
  66. Abel, F.; Murke, F.; Gaida, M.; Garnier, N.; Ochsenfarth, C.; Theiss, C.; Thielmann, M.; Kleinbongard, P.; Giebel, B.; Peters, J.; et al. Extracellular vesicles isolated from patients undergoing remote ischemic preconditioning decrease hypoxia-evoked apoptosis of cardiomyoblasts after isoflurane but not propofol exposure. PLoS ONE 2020, 15, e0228948. [Google Scholar] [CrossRef] [PubMed]
  67. Davidson, S.M.; Riquelme, J.A.; Takov, K.; Vicencio, J.M.; Boi-Doku, C.; Khoo, V.; Doreth, C.; Radenkovic, D.; Lavandero, S.; Yellon, D.M. Cardioprotection mediated by exosomes is impaired in the setting of type II diabetes but can be rescued by the use of non-diabetic exosomes in vitro. J. Cell. Mol. Med. 2018, 22, 141–151. [Google Scholar] [CrossRef]
  68. Gan, L.; Xie, D.; Liu, J.; Lau, W.B.; Christopher, T.A.; Lopez, B.; Zhang, L.; Gao, E.; Koch, W.; Ma, X.-L.; et al. Small extracellular microvesicles mediated pathological communications between dysfunctional adipocytes and cardiomyocytes as a novel mechanism exacerbating ischemia/reperfusion injury in diabetic mice. Circulation 2020, 141, 968–983. [Google Scholar] [CrossRef] [PubMed]
  69. Wider, J.; Undyala, V.V.R.; Whittaker, P.; Woods, J.; Chen, X.; Przyklenk, K. Remote ischemic preconditioning fails to reduce infarct size in the Zucker fatty rat model of type-2 diabetes: Role of defective humoral communication. Basic Res. Cardiol. 2018, 113, 16. [Google Scholar] [CrossRef] [PubMed]
  70. Davidson, S.M.; Takov, K.; Yellon, D. Exosomes and cardiovascular protection. Cardiovasc. Drugs Ther. 2017, 31, 77–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Egom, E.E.; Mamas, M.A.; Clark, A.L. The potential role of sphingolipid-mediated cell signaling in the interaction between hyperglycemia, acute myocardial infarction and heart failure. Expert Opin. Ther. Targets 2012, 16, 791–800. [Google Scholar] [CrossRef] [PubMed]
  72. Ricci, C.; Jong, C.J.; Schaffer, S.W. Proapoptotic and antiapoptotic effects of hyperglycemia: Role of insulin signaling. Can. J. Physiol. Pharmacol. 2008, 86, 166–172. [Google Scholar] [CrossRef]
  73. Miki, T.; Itoh, T.; Sunaga, D.; Miura, T. Effects of diabetes on myocardial infarct size and cardioprotection by preconditioning and postconditioning. Cardiovasc. Diabetol. 2012, 11, 67. [Google Scholar] [CrossRef] [Green Version]
  74. Donahoe, S.M.; Stewart, G.C.; McCabe, C.H.; Mohanavelu, S.; Murphy, S.A.; Cannon, C.P.; Antman, E.M. Diabetes and mortality following acute coronary syndromes. JAMA 2007, 298, 765–775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Xue, R.; Lei, S.; Xia, Z.-Y.; Wu, Y.; Meng, Q.; Zhan, L.; Su, W.; Liu, H.; Xu, J.; Liu, Z.; et al. Selective inhibition of PTEN preserves ischaemic post-conditioning cardioprotection in STZ-induced Type 1 diabetic rats: Role of the PI3K/Akt and JAK2/STAT3 pathways. Clin. Sci. 2016, 130, 377–392. [Google Scholar] [CrossRef]
  76. Przyklenk, K.; Maynard, M.; Greiner, D.L.; Whittaker, P. Cardioprotection with postconditioning: Loss of efficacy in murine models of type-2 and type-1 diabetes. Antioxid. Redox Signal. 2011, 14, 781–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Wang, D.; Hu, X.; Lee, S.H.; Chen, F.; Jiang, K.; Tu, Z.; Liu, Z.; Du, J.; Wang, L.; Yin, C.; et al. Diabetes exacerbates myocardial ischemia/reperfusion injury by down-regulation of microRNA and up-regulation of o-glcnacylation. JACC Basic Transl. Sci. 2018, 3, 350–362. [Google Scholar] [CrossRef]
  78. Negi, S.; Rutman, A.K.; Paraskevas, S. Extracellular vesicles in type 1 diabetes: Messengers and regulators. Curr. Diabetes Rep. 2019, 19, 69. [Google Scholar] [CrossRef] [PubMed]
  79. Cianciaruso, C.; Phelps, E.A.; Pasquier, M.; Hamelin, R.; Demurtas, D.; Ahmed, M.A.; Piemonti, L.; Hirosue, S.; Swartz, M.A.; De Palma, M.; et al. Primary human and rat β-cells release the intracellular autoantigens gad65, ia-2, and proinsulin in exosomes together with cytokine-induced enhancers of immunity. Diabetes 2017, 66, 460–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Garcia-Contreras, M.; Brooks, R.W.; Boccuzzi, L.; Robbins, P.D.; Ricordi, C. Exosomes as biomarkers and therapeutic tools for type 1 diabetes mellitus. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 2940–2956. [Google Scholar] [PubMed]
  81. Barutta, F.; Tricarico, M.; Corbelli, A.; Annaratone, L.; Pinach, S.; Grimaldi, S.; Bruno, G.; Cimino, D.; Taverna, D.; Deregibus, M.C.; et al. Urinary exosomal microRNAs in incipient diabetic nephropathy. PLoS ONE 2013, 8, e73798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Florijn, B.W.; Duijs, J.M.; Levels, J.H.; Dallinga-Thie, G.M.; Wang, Y.; Boing, A.N.; Yuana, Y.; Stam, W.; Limpens, R.W.; Au, Y.W.; et al. Diabetic nephropathy alters the distribution of circulating angiogenic micrornas among extracellular vesicles, HDL, and Ago-2. Diabetes 2019, 68, 2287–2300. [Google Scholar] [CrossRef]
  83. Bergen, K.; Mobarrez, F.; Jörneskog, G.; Wallén, H.; Tehrani, S. Phosphatidylserine expressing microvesicles in relation to microvascular complications in type 1 diabetes. Thromb. Res. 2018, 172, 158–164. [Google Scholar] [CrossRef]
  84. Zahran, A.M.; Mohamed, I.L.; El Asheer, O.M.; Tamer, D.M.; Abo-Elela, M.G.M.; Abdel-Rahim, M.H.; El-Badawy, O.H.B.; Elsayh, K.I. Circulating endothelial cells, circulating endothelial progenitor cells, and circulating microparticles in type 1 diabetes mellitus. Clin. Appl. Thromb. Hemost. 2019, 25, 1076029618825311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Liu, M.; Wang, Y.; Zhu, Q.; Zhao, J.; Wang, Y.; Shang, M.; Liu, M.; Wu, Y.; Song, J.; Liu, Y. Protective effects of circulating microvesicles derived from ischemic preconditioning on myocardial ischemia/reperfusion injury in rats by inhibiting endoplasmic reticulum stress. Apoptosis 2018, 23, 436–448. [Google Scholar] [CrossRef]
  86. Chiva-Blanch, G.; Suades, R.; Padro, T.; Vilahur, G.; Pena, E.; Ybarra, J.; Pou, J.M.; Badimon, L. Microparticle shedding by erythrocytes, monocytes and vascular smooth muscular cells is reduced by aspirin in diabetic patients. Rev. Esp. Cardiol. 2016, 69, 672–680. [Google Scholar] [CrossRef] [PubMed]
  87. Chang, W.; Wang, J. Exosomes and their noncoding RNA cargo are emerging as new modulators for diabetes mellitus. Cells 2019, 8, 853. [Google Scholar] [CrossRef] [Green Version]
  88. Garcia-Contreras, M.; Shah, S.H.; Tamayo, A.; Robbins, P.; Golberg, R.B.; Mendez, A.J.; Ricordi, C. Plasma-derived exosome characterization reveals a distinct microRNA signature in long duration Type 1 diabetes. Sci. Rep. 2017, 7, 5998. [Google Scholar] [CrossRef]
  89. Costantino, S.; Paneni, F.; Lüscher, T.F.; Cosentino, F. MicroRNA profiling unveils hyperglycaemic memory in the diabetic heart. Eur. Heart J. 2016, 37, 572–576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Fu, B.-C.; Lang, J.-L.; Zhang, D.-Y.; Sun, L.; Chen, W.; Liu, W.; Liu, K.-Y.; Ma, C.-Y.; Jiang, S.-L.; Li, R.-K.; et al. Suppression of miR-34a expression in the myocardium protects against ischemia–reperfusion injury through SIRT1 protective pathway. Stem Cells Dev. 2017, 26, 1270–1282. [Google Scholar] [CrossRef]
  91. Dehaini, H.; Awada, H.; El-Yazbi, A.; Zouein, F.A.; Issa, K.; Eid, A.A.; Ibrahim, M.; Badran, A.; Baydoun, E.; Pintus, G.; et al. MicroRNAs as potential pharmaco-targets in ischemia-reperfusion injury compounded by diabetes. Cells 2019, 8, 152. [Google Scholar] [CrossRef] [Green Version]
  92. Lakhter, A.J.; Pratt, R.E.; Moore, R.E.; Doucette, K.K.; Maier, B.F.; DiMeglio, L.A.; Sims, E.K. Beta cell extracellular vesicle miR-21-5p cargo is increased in response to inflammatory cytokines and serves as a biomarker of type 1 diabetes. Diabetologia 2018, 61, 1124–1134. [Google Scholar] [CrossRef] [Green Version]
  93. Salama, A.; Fichou, N.; Allard, M.; Dubreil, L.; De Beaurepaire, L.; Viel, A.; Jégou, D.; Bosch, S.; Bach, J.-M. MicroRNA-29b modulates innate and antigen-specific immune responses in mouse models of autoimmunity. PLoS ONE 2014, 9, e106153. [Google Scholar] [CrossRef] [PubMed]
  94. Rutman, A.K.; Negi, S.; Gasparrini, M.; Hasilo, C.P.; Tchervenkov, J.; Paraskevas, S. Immune response to extracellular vesicles from human islets of langerhans in patients with type 1 diabetes. Endocrinology 2018, 159, 3834–3847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Liu, L.; Fang, C.; Fu, W.; Jiang, B.; Li, G.; Qin, L.; Rosenbluth, J.; Gong, G.; Xie, C.B.; Yoo, P.; et al. Endothelial cell–derived interleukin-18 released during ischemia reperfusion injury selectively expands t peripheral helper cells to promote alloantibody production. Circulation 2020, 141, 464–478. [Google Scholar] [CrossRef]
  96. Andreadou, I.; Cabrera-Fuentes, H.A.; Devaux, Y.; Frangogiannis, N.G.; Frantz, S.; Guzik, T.; Liehn, E.A.; Gomes, C.P.C.; Schulz, R.; Hausenloy, D.J. Immune cells as targets for cardioprotection: New players and novel therapeutic opportunities. Cardiovasc. Res. 2019, 115, 1117–1130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Wang, L.; Liu, W.; He, X.; Chen, Y.; Lu, J.; Liu, K.; Cao, K.; Yin, P. Association of overweight and obesity with patient mortality after acute myocardial infarction: A meta-analysis of prospective studies. Int. J. Obes. 2016, 40, 220–228. [Google Scholar] [CrossRef]
  98. Van Gaal, L.F.; Mertens, I.L.; De Block, C.E. Mechanisms linking obesity with cardiovascular disease. Nature 2006, 444, 875–880. [Google Scholar] [CrossRef]
  99. Mollard, R.; Sénéchal, M.; MacIntosh, A.C.; Hay, J.; Wicklow, B.A.; Wittmeier, K.D.M.; Sellers, E.A.C.; Dean, H.J.; Ryner, L.; Berard, L.; et al. Dietary determinants of hepatic steatosis and visceral adiposity in overweight and obese youth at risk of type 2 diabetes. Am. J. Clin. Nutr. 2014, 99, 804–812. [Google Scholar] [CrossRef] [Green Version]
  100. Longo, M.; Zatterale, F.; Naderi, J.; Parrilo, L.; Formisano, P.; Raciti, G.A.; Beguinot, F.; Miele, C. Adipose tissue dysfunction as determinant of obesity-associated metabolic complications. Int. J. Mol. Sci. 2019, 20, 2358. [Google Scholar] [CrossRef] [Green Version]
  101. Alcalá, M.; Calderon-Dominguez, M.; Bustos, E.; Ramos, P.; Casals, N.; Serra, D.; Viana, M.; Herrero, L. Increased inflammation, oxidative stress and mitochondrial respiration in brown adipose tissue from obese mice. Sci. Rep. 2017, 7, 16082. [Google Scholar] [CrossRef] [Green Version]
  102. Deng, Z.-B.; Poliakov, A.; Hardy, R.W.; Clements, R.; Liu, C.; Liu, Y.; Wang, J.; Xiang, X.; Zhang, S.; Zhuang, X.; et al. Adipose tissue exosome-like vesicles mediate activation of macrophage-induced insulin resistance. Diabetes 2009, 58, 2498–2505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Crewe, C.; Scherer, P.E. Intercellular and interorgan crosstalk through adipocyte extracellular vesicles. Rev. Endocr. Metab. Disord. 2021, 1–9. [Google Scholar] [CrossRef] [PubMed]
  104. Thomou, T.; Mori, M.A.; Dreyfuss, J.M.; Konishi, M.; Sakaguchi, M.; Wolfrum, C.; Rao, T.N.; Winnay, J.N.; Garcia-Martin, R.; Grinspoon, S.K.; et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature 2017, 542, 450–455. [Google Scholar] [CrossRef] [PubMed]
  105. Ortega, F.J.; Mercader, J.M.; Catalan, V.; Moreno-Navarrete, J.M.; Pueyo, N.; Sabater-Masdeu, M.; Gomez-Ambrosi, J.; Anglada, R.; Formoso, J.A.F.; Ricart, W.; et al. Targeting the circulating MicroRNA signature of obesity. Clin. Chem. 2013, 59, 781–792. [Google Scholar] [CrossRef] [Green Version]
  106. Zhang, B.; Yang, Y.; Xiang, L.; Zhao, Z.; Ye, R. Adipose-derived exosomes: A novel adipokine in obesity-associated diabetes. J. Cell. Physiol. 2019, 234, 16692–16702. [Google Scholar] [CrossRef]
  107. Giannella, A.; Radu, C.M.; Franco, L.; Campello, E.; Simioni, P.; Avogaro, A.; De Kreutzenberg, S.V.; Ceolotto, G. Circulating levels and characterization of microparticles in patients with different degrees of glucose tolerance. Cardiovasc. Diabetol. 2017, 16, 118. [Google Scholar] [CrossRef]
  108. Freeman, D.W.; Hooten, N.N.; Eitan, E.; Green, J.; Mode, N.A.; Bodogai, M.; Zhang, Y.; Lehrmann, E.; Zonderman, A.B.; Biragyn, A.; et al. Altered extracellular vesicle concentration, cargo, and function in diabetes. Diabetes 2018, 67, 2377–2388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Chen, Y.; Feng, B.; Li, X.; Ni, Y.; Luo, Y. Plasma endothelial microparticles and their correlation with the presence of hypertension and arterial stiffness in patients with type 2 diabetes. J. Clin. Hypertens. 2012, 14, 455–460. [Google Scholar] [CrossRef] [PubMed]
  110. Rodrigues, K.F.; Pietrani, N.T.; Fernandes, A.P.; Bosco, A.A.; De Sousa, M.C.R.; Silva, I.D.F.O.; Silveira, J.N.; Campos, F.M.F.; Gomes, K.B. Circulating microparticles levels are increased in patients with diabetic kidney disease: A case-control research. Clin. Chim. Acta 2018, 479, 48–55. [Google Scholar] [CrossRef]
  111. Zhang, X.; McGeoch, S.C.; Johnstone, A.M.; Holtrop, G.; Sneddon, A.; MacRury, S.M.; Megson, I.L.; Pearson, D.W.M.; Abraham, P.; De Roos, B.; et al. Platelet-derived microparticle count and surface molecule expression differ between subjects with and without type 2 diabetes, independently of obesity status. J. Thromb. Thrombol. 2014, 37, 455–463. [Google Scholar] [CrossRef]
  112. Landers-Ramos, R.Q.; Serra, M.C.; Blumenthal, J.B.; Ryan, A.S.; Hafer-Macko, C.E.; Prior, S.J. Type 2 diabetes and older age contribute to elevated plasma microparticle concentrations independent of chronic stroke. Exp. Physiol. 2018, 103, 1560–1570. [Google Scholar] [CrossRef] [Green Version]
  113. Jung, K.-H.; Chu, K.; Lee, S.-T.; Bahn, J.-J.; Kim, J.-H.; Kim, M.; Lee, S.K.; Roh, J.-K. Risk of macrovascular complications in type 2 diabetes mellitus: Endothelial microparticle profiles. Cerebrovasc. Dis. 2011, 31, 485–493. [Google Scholar] [CrossRef]
  114. Gkaliagkousi, E.; Nikolaidou, B.; Gavriilaki, E.; Lazaridis, A.; Yiannaki, E.; Anyfanti, P.; Zografou, I.; Markala, D.; Douma, S. Increased erythrocyte- and platelet-derived microvesicles in newly diagnosed type 2 diabetes mellitus. Diabetes Vasc. Dis. Res. 2019, 16, 458–465. [Google Scholar] [CrossRef]
  115. Nomura, S. Dynamic role of microparticles in type 2 diabetes mellitus. Curr. Diabetes Rev. 2009, 5, 245–251. [Google Scholar] [CrossRef]
  116. Dec-Gilowska, M.; Trojnar, M.; Makaruk, B.; Czop, M.; Przybylska-Kuc, S.; Mosiewicz-Madejska, B.; Dzida, G.; Mosiewicz, J. Circulating endothelial microparticles and aortic stiffness in patients with type 2 diabetes mellitus. Medicina 2019, 55, 596. [Google Scholar] [CrossRef] [Green Version]
  117. Cheng, V.; Kashyap, S.R.; Schauer, P.R.; Kirwan, J.P.; McCrae, K.R. Restoration of glycemic control in patients with type 2 diabetes mellitus after bariatric surgery is associated with reduction in microparticles. Surg. Obes. Relat. Dis. 2013, 9, 207–212. [Google Scholar] [CrossRef] [Green Version]
  118. Hubal, M.; Nadler, E.P.; Ferrante, S.C.; Barberio, M.; Suh, J.-H.; Wang, J.; Dohm, G.L.; Pories, W.J.; Mietus-Snyder, M.; Freishtat, R. Circulating adipocyte-derived exosomal MicroRNAs associated with decreased insulin resistance after gastric bypass. Obesity 2017, 25, 102–110. [Google Scholar] [CrossRef]
  119. Ji, C.; Guo, X. The clinical potential of circulating microRNAs in obesity. Nat. Rev. Endocrinol. 2019, 15, 731–743. [Google Scholar] [CrossRef]
  120. Kumar, A.; Sundaram, K.; Mu, J.; Dryden, G.W.; Sriwastva, M.K.; Lei, C.; Zhang, L.; Qiu, X.; Xu, F.; Yan, J.; et al. High-fat diet-induced upregulation of exosomal phosphatidylcholine contributes to insulin resistance. Nat. Commun. 2021, 12, 213. [Google Scholar] [CrossRef]
  121. Santilli, F.; Marchisio, M.; Lanuti, P.; Boccatonda, A.; Miscia, S.; Davì, G. Microparticles as new markers of cardiovascular risk in diabetes and beyond. Thromb. Haemost. 2016, 116, 220–234. [Google Scholar] [CrossRef]
  122. Kranendonk, M.E.G.; De Kleijn, D.P.V.; Kalkhoven, E.; Kanhai, D.A.; Uiterwaal, C.S.P.M.; Van Der Graaf, Y.; Pasterkamp, G.; Visseren, F.L.J.; Doevendans, P.A.; Algra, A.; et al. Extracellular vesicle markers in relation to obesity and metabolic complications in patients with manifest cardiovascular disease. Cardiovasc. Diabetol. 2014, 13, 37. [Google Scholar] [CrossRef] [Green Version]
  123. Prattichizzo, F.; De Nigris, V.; Sabbatinelli, J.; Giuliani, A.; Castaño, C.; Párrizas, M.; Crespo, I.; Grimaldi, A.; Baranzini, N.; Spiga, R.; et al. CD31+ extracellular vesicles from patients with type 2 diabetes shuttle a mirna signature associated with cardiovascular complications. Diabetes 2021, 70, 240–254. [Google Scholar] [CrossRef]
  124. Ge, Q.; Xie, X.X.; Xiao, X.; Li, X. Exosome-like vesicles as new mediators and therapeutic targets for treating insulin resistance and beta-cell mass failure in type 2 diabetes mellitus. J. Diabetes Res. 2019, 2019, 3256060. [Google Scholar] [CrossRef] [Green Version]
  125. Sáez, T.; Toledo, F.; Sobrevia, L. Impaired signalling pathways mediated by extracellular vesicles in diabesity. Mol. Asp. Med. 2019, 66, 13–20. [Google Scholar] [CrossRef]
  126. Nie, H.; Pan, Y.; Zhou, Y. Exosomal microRNA-194 causes cardiac injury and mitochondrial dysfunction in obese mice. Biochem. Biophys. Res. Commun. 2018, 503, 3174–3179. [Google Scholar] [CrossRef]
  127. Li, F.; Zhang, K.; Xu, T.; Du, W.; Yu, B.; Liu, Y.; Nie, H. Exosomal microRNA-29a mediates cardiac dysfunction and mitochondrial inactivity in obesity-related cardiomyopathy. Endocrine 2019, 63, 480–488. [Google Scholar] [CrossRef]
  128. Zou, T.; Zhu, M.; Ma, Y.-C.; Xiao, F.; Yu, X.; Xu, L.; Ma, L.-Q.; Yang, J.; Dong, J.-Z. MicroRNA-410-5p exacerbates high-fat diet-induced cardiac remodeling in mice in an endocrine fashion. Sci. Rep. 2018, 8, 8780. [Google Scholar] [CrossRef]
  129. Vander Heide, R.S. Increased expression of HSP27 protects canine myocytes from simulated ischemia-reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 2002, 282, H935–H941. [Google Scholar] [CrossRef]
  130. Stępień, E.; Durak-Kozica, M.; Kamińska, A.; Targosz-Korecka, M.; Libera, M.; Tylko, G.; Opalińska, A.; Kapusta, M.; Solnica, B.; Georgescu, A.; et al. Circulating ectosomes: Determination of angiogenic microRNAs in type 2 diabetes. Theranostics 2018, 8, 3874–3890. [Google Scholar] [CrossRef]
  131. Li, S.; Wei, J.; Zhang, C.; Li, X.; Meng, W.; Mo, X.; Zhang, Q.; Liu, Q.; Ren, K.; Du, R.; et al. Cell-derived microparticles in patients with type 2 diabetes mellitus: A systematic review and meta-analysis. Cell. Physiol. Biochem. 2016, 39, 2439–2450. [Google Scholar] [CrossRef]
  132. Kim, H.; Bae, Y.-U.; Lee, H.; Kim, H.; Jeon, J.S.; Noh, H.; Han, D.C.; Byun, D.W.; Kim, S.H.; Park, H.K.; et al. Effect of diabetes on exosomal miRNA profile in patients with obesity. BMJ Open Diabetes Res. Care 2020, 8, e001403. [Google Scholar] [CrossRef]
  133. Prattichizzo, F.; Matacchione, G.; Giuliani, A.; Sabbatinelli, J.; Olivieri, F.; de Candia, P.; De Nigris, V.; Ceriello, A. Extracellular vesicle-shuttled miRNAs: A critical appraisal of their potential as nano-diagnostics and nano-therapeutics in type 2 diabetes mellitus and its cardiovascular complications. Theranostics 2021, 11, 1031–1045. [Google Scholar] [CrossRef]
  134. Wang, X.; Huang, W.; Liu, G.; Cai, W.; Millard, R.W.; Wang, Y.; Chang, J.; Peng, T.; Fan, G.-C. Cardiomyocytes mediate anti-angiogenesis in type 2 diabetic rats through the exosomal transfer of miR-320 into endothelial cells. J. Mol. Cell. Cardiol. 2014, 74, 139–150. [Google Scholar] [CrossRef] [Green Version]
  135. Santamaria-Martos, F.; Benítez, I.D.; Latorre, J.; Lluch, A.; Moreno-Navarrete, J.M.; Sabater, M.; Ricart, W.; de la Torre, M.S.; Mora, S.; Fernández-Real, J.M.; et al. Comparative and functional analysis of plasma membrane-derived extracellular vesicles from obese vs. nonobese women. Clin. Nutr. 2020, 39, 1067–1076. [Google Scholar] [CrossRef]
  136. Withers, S.B.; Dewhurst, T.; Hammond, C.; Topham, C.H. MiRNAs as novel adipokines: Obesity-related circulating MiRNAs influence chemosensitivity in cancer patients. Non-Coding RNA 2020, 6, 5. [Google Scholar] [CrossRef] [Green Version]
  137. Tian, Z.-Q.; Jiang, H.; Lu, Z.-B. MiR-320 regulates cardiomyocyte apoptosis induced by ischemia–reperfusion injury by targeting AKIP1. Cell. Mol. Biol. Lett. 2018, 23, 41. [Google Scholar] [CrossRef] [Green Version]
  138. Song, C.; Liu, B.; Diao, H.-Y.; Shi, Y.-F.; Zhang, J.-C.; Li, Y.-X.; Liu, N.; Yu, Y.-P.; Wang, G.; Wang, J.-P.; et al. Down-regulation of microRNA-320 suppresses cardiomyocyte apoptosis and protects against myocardial ischemia and reperfusion injury by targeting IGF-1. Oncotarget 2016, 7, 39740–39757. [Google Scholar] [CrossRef]
  139. Ren, X.-P.; Wu, J.; Wang, X.; Sartor, M.A.; Qian, J.; Jones, K.; Nicolaou, P.; Pritchard, T.J.; Fan, G.-C. MicroRNA-320 is involved in the regulation of cardiac ischemia/reperfusion injury by targeting heat-shock protein 20. Circulation 2009, 119, 2357–2366. [Google Scholar] [CrossRef] [Green Version]
  140. Wang, W.; Zheng, Y.; Wang, M.; Yan, M.; Jiang, J.; Li, Z. Exosomes derived miR-126 attenuates oxidative stress and apoptosis from ischemia and reperfusion injury by targeting ERRFI1. Gene 2019, 690, 75–80. [Google Scholar] [CrossRef]
  141. Luo, Q.; Guo, D.; Liu, G.; Chen, G.; Hang, M.; Jin, M. Exosomes from MiR-126-Overexpressing adscs are therapeutic in relieving acute myocardial ischaemic injury. Cell. Physiol. Biochem. 2017, 44, 2105–2116. [Google Scholar] [CrossRef]
  142. Jansen, F.; Wang, H.; Przybilla, D.; Franklin, B.S.; Dolf, A.; Pfeifer, P.; Schmitz, T.; Flender, A.; Endl, E.; Nickenig, G.; et al. Vascular endothelial microparticles-incorporated microRNAs are altered in patients with diabetes mellitus. Cardiovasc. Diabetol. 2016, 15, 49. [Google Scholar] [CrossRef] [Green Version]
  143. Wu, K.; Yang, Y.; Zhong, Y.; Ammar, H.M.; Zhang, P.; Guo, R.; Liu, H.; Cheng, C.; Koroscil, T.M.; Chen, Y.; et al. The effects of microvesicles on endothelial progenitor cells are compromised in type 2 diabetic patients via downregulation of the miR-126/VEGFR2 pathway. Am. J. Physiol. Endocrinol. Metab. 2016, 310, E828–E837. [Google Scholar] [CrossRef] [Green Version]
  144. Ogawa, R.; Tanaka, C.; Sato, M.; Nagasaki, H.; Sugimura, K.; Okumura, K.; Nakagawa, Y.; Aoki, N. Adipocyte-derived microvesicles contain RNA that is transported into macrophages and might be secreted into blood circulation. Biochem. Biophys. Res. Commun. 2010, 398, 723–729. [Google Scholar] [CrossRef]
  145. Zhou, Y.; Richards, A.M.; Wang, P. MicroRNA-221 Is Cardioprotective and anti-fibrotic in a rat model of myocardial infarction. Mol. Ther. Nucleic Acids 2019, 17, 185–197. [Google Scholar] [CrossRef] [Green Version]
  146. Lai, T.-C.; Lee, T.-L.; Chang, Y.-C.; Chen, Y.-C.; Lin, S.-R.; Lin, S.-W.; Pu, C.-M.; Tsai, J.-S.; Chen, Y.-L. MicroRNA-221/222 mediates adsc-exosome-induced cardioprotection against ischemia/reperfusion by targeting puma and ets-1. Front. Cell Dev. Biol. 2020, 8, 1475. [Google Scholar] [CrossRef]
  147. Liu, T.; Sun, Y.-C.; Cheng, P.; Shao, H.-G. Adipose tissue macrophage-derived exosomal miR-29a regulates obesity-associated insulin resistance. Biochem. Biophys. Res. Commun. 2019, 515, 352–358. [Google Scholar] [CrossRef]
  148. Chiricosta, L.; Silvestro, S.; Gugliandolo, A.; Marconi, G.D.; Pizzicannella, J.; Bramanti, P.; Trubiani, O.; Mazzon, E. Extracellular vesicles of human periodontal ligament stem cells contain micrornas associated to proto-oncogenes: Implications in cytokinesis. Front. Genet. 2020, 11, 582. [Google Scholar] [CrossRef]
  149. Wang, L.; Niu, X.; Hu, J.; Xing, H.; Sun, M.; Wang, J.; Jian, Q.; Yang, H. After Myocardial Ischemia-Reperfusion, miR-29a, and Let7 Could Affect Apoptosis through Regulating IGF-1. BioMed Res. Int. 2015, 2015, 245412. [Google Scholar] [CrossRef] [Green Version]
  150. Niu, X.; Pu, S.; Ling, C.; Xu, J.; Wang, J.; Sun, S.; Yao, Y.; Zhang, Z. lncRNA Oip5-as1 attenuates myocardial ischaemia/reperfusion injury by sponging miR-29a to activate the SIRT1/AMPK/PGC1α pathway. Cell Prolif. 2020, 53, e12818. [Google Scholar] [CrossRef]
  151. Ye, Y.; Hu, Z.; Lin, Y.; Zhang, C.; Perez-Polo, J.R. Downregulation of microRNA-29 by antisense inhibitors and a PPAR-γ agonist protects against myocardial ischaemia-reperfusion injury. Cardiovasc. Res. 2010, 87, 535–544. [Google Scholar] [CrossRef] [Green Version]
  152. Kukreja, R.C.; Yin, C.; Salloum, F. MicroRNAs: New players in cardiac injury and protection. Mol. Pharmacol. 2011, 80, 558–564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Chen, J.-G.; Xu, X.-M.; Ji, H.; Sun, B. Inhibiting miR-155 protects against myocardial ischemia/reperfusion injury via targeted regulation of HIF-1α in rats. Iran. J. Basic Med. Sci. 2019, 22, 1050–1058. [Google Scholar] [CrossRef]
  154. Guo, J.; Liu, H.-B.; Sun, C.; Yan, X.-Q.; Hu, J.; Yu, J.; Yuan, Y.; Du, Z.-M. MicroRNA-155 Promotes Myocardial Infarction-Induced Apoptosis by Targeting RNA-binding protein QKI. Oxid. Med. Cell. Longev. 2019, 2019, 4579806–4579814. [Google Scholar] [CrossRef] [PubMed]
  155. Ge, X.; Meng, Q.; Wei, L.; Liu, J.; Li, M.; Liang, X.; Lin, F.; Zhang, Y.; Li, Y.; Liu, Z.; et al. Myocardial ischemia-reperfusion induced cardiac extracellular vesicles harbour proinflammatory features and aggravate heart injury. J. Extracell. Vesicles 2021, 10, e12072. [Google Scholar] [CrossRef]
  156. Xing, X.; Guo, S.; Zhang, G.; Liu, Y.; Bi, S.; Wang, X.; Lu, Q. miR-26a-5p protects against myocardial ischemia/reperfusion injury by regulating the PTEN/PI3K/AKT signaling pathway. Braz. J. Med. Biol. Res. 2020, 53, e9106. [Google Scholar] [CrossRef] [Green Version]
  157. Xu, H.; Du, X.; Xu, J.; Zhang, Y.; Tian, Y.; Liu, G.; Wang, X.; Ma, M.; Du, W.; Liu, Y.; et al. Pancreatic beta cell microRNA-26a alleviates type 2 diabetes by improving peripheral insulin sensitivity and preserving beta cell function. PLoS Biol. 2020, 18, e3000603. [Google Scholar] [CrossRef] [PubMed]
  158. Yao, L.; Lv, X.; Wang, X. MicroRNA 26a inhibits HMGB1 expression and attenuates cardiac ischemia-reperfusion injury. J. Pharmacol. Sci. 2016, 131, 6–12. [Google Scholar] [CrossRef] [Green Version]
  159. Gong, D.D.; Yu, J.; Yu, J.-C.; Diang, X.-D. Effect of miR-26a targeting GSK-3beta/beta-catenin signaling pathway on myocardial apoptosis in rats with myocardial ischemia-reperfusion. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 7073–7082. [Google Scholar] [CrossRef]
  160. Liu, C.; Gao, Y.; Wu, J.; Zou, J. Exosomal miR-23a and miR-192, Potential diagnostic biomarkers for type 2 diabetes. Clin. Lab. 2021, 67, 494–593. [Google Scholar] [CrossRef]
  161. Long, B.; Gan, T.-Y.; Zhang, R.-C.; Zhang, A.Y.-H. miR-23a regulates cardiomyocyte apoptosis by targeting manganese superoxide dismutase. Mol. Cells 2017, 40, 542–549. [Google Scholar] [CrossRef]
  162. Roy, S.; Benz, F.; Alder, J.; Bantel, H.; Janssen, J.; Vucur, M.; Gautheron, J.; Schneider, A.; Schüller, F.; Loosen, S.; et al. Down-regulation of miR-192-5p protects from oxidative stress-induced acute liver injury. Clin. Sci. 2016, 130, 1197–1207. [Google Scholar] [CrossRef]
  163. Wang, L.; Li, Q.; Diao, J.; Lin, L.; Wei, J. MiR-23a is involved in myocardial ischemia/reperfusion injury by directly targeting cx43 and regulating mitophagy. Inflammation 2021, 44, 1581–1591. [Google Scholar] [CrossRef]
  164. Santovito, D.; De Nardis, V.; Marcantonio, P.; Mandolini, C.; Paganelli, C.; Vitale, E.; Buttitta, F.; Bucci, M.; Mezzetti, A.; Consoli, A.; et al. Plasma exosome microrna profiling unravels a new potential modulator of adiponectin pathway in diabetes: Effect of glycemic control. J. Clin. Endocrinol. Metab. 2014, 99, E1681–E1685. [Google Scholar] [CrossRef] [PubMed]
  165. Al-Qaissi, A.; Papageorgiou, M.; Deshmukh, H.; Madden, L.A.; Rigby, A.; Kilpatrick, E.S.; Atkin, S.L.; Sathyapalan, T. Effects of acute insulin-induced hypoglycaemia on endothelial microparticles in adults with and without type 2 diabetes. Diabetes Obes. Metab. 2019, 21, 533–540. [Google Scholar] [CrossRef] [PubMed]
  166. Ghai, V.; Kim, T.-K.; Etheridge, A.; Nielsen, T.; Hansen, T.; Pedersen, O.; Galas, D.; Wang, K. Extracellular vesicle encapsulated micrornas in patients with type 2 diabetes are affected by metformin treatment. J. Clin. Med. 2019, 8, 617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Li, J.; Xu, J.-P.; Zhao, X.-Z.; Sun, X.-J.; Xu, Z.-W.; Song, S.-J. Protective effect of metformin on myocardial injury in metabolic syndrome patients following percutaneous coronary intervention. Cardiology 2014, 127, 133–139. [Google Scholar] [CrossRef]
  168. El Messaoudi, S.; Nederlof, R.; Zuurbier, C.J.; van Swieten, H.A.; Pickkers, P.; Noyez, L.; Dieker, H.-J.; Coenen, M.; Donders, A.R.T.; Vos, A.; et al. Effect of metformin pretreatment on myocardial injury during coronary artery bypass surgery in patients without diabetes (MetCAB): A double-blind, randomised controlled trial. Lancet Diabetes Endocrinol. 2015, 3, 615–623. [Google Scholar] [CrossRef]
  169. El Messaoudi, S.; Schreuder, T.H.; Kengen, R.D.; Rongen, G.A.; Broek, P.H.V.D.; Thijssen, D.H.J.; Riksen, N.P. Impact of metformin on endothelial ischemia-reperfusion injury in humans in vivo: A prospective randomized open, blinded-endpoint study. PLoS ONE 2014, 9, e96062. [Google Scholar] [CrossRef] [Green Version]
  170. Higgins, L.; Palee, S.; Chattipakorn, S.C.; Chattipakorn, N. Effects of metformin on the heart with ischaemia-reperfusion injury: Evidence of its benefits from in vitro, in vivo and clinical reports. Eur. J. Pharmacol. 2019, 858, 172489. [Google Scholar] [CrossRef]
  171. Shimazu, T.; Inami, N.; Satoh, D.; Kajiura, T.; Yamada, K.; Iwasaka, T.; Nomura, S. Effect of acarbose on platelet-derived microparticles, soluble selectins, and adiponectin in diabetic patients. J. Thromb. Thrombol. 2009, 28, 429–435. [Google Scholar] [CrossRef]
  172. Tsimerman, G.; Roguin, A.; Bachar, A.; Melamed, E.; Brenner, B.; Aharon, A. Involvement of microparticles in diabetic vascular complications. Thromb. Haemost. 2011, 106, 310–321. [Google Scholar] [CrossRef]
  173. Thompson, M.D.; Cismowski, M.J.; Serpico, M.; Pusateri, A.; Brigstock, D.R. Elevation of circulating microRNA levels in obese children compared to healthy controls. Clin. Obes. 2017, 7, 216–221. [Google Scholar] [CrossRef]
  174. Cui, X.; You, L.; Zhu, L.; Wang, X.; Zhou, Y.; Li, Y.; Wen, J.; Xia, Y.; Wang, X.; Ji, C.; et al. Change in circulating microRNA profile of obese children indicates future risk of adult diabetes. Metabolism 2018, 78, 95–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Pescador, N.; Pérez-Barba, M.; Ibarra, J.M.; Corbatón-Anchuelo, A.; Martínez-Larrad, M.T.; Serrano-Ríos, M. Serum circulating microrna profiling for identification of potential type 2 diabetes and obesity biomarkers. PLoS ONE 2013, 8, e77251. [Google Scholar] [CrossRef] [Green Version]
  176. Liu, Y.; Yang, L.; Yin, J.; Su, D.; Pan, Z.; Li, P.; Wang, X. MicroRNA-15b deteriorates hypoxia/reoxygenation-induced cardiomyocyte apoptosis by downregulating Bcl-2 and MAPK3. J. Investig. Med. 2018, 66, 39–45. [Google Scholar] [CrossRef] [PubMed]
  177. Liu, L.; Zhang, G.; Liang, Z.; Liu, X.; Li, T.; Fan, J.; Bai, J.; Wang, Y. MicroRNA-15b enhances hypoxia/reoxygenation-induced apoptosis of cardiomyocytes via a mitochondrial apoptotic pathway. Apoptosis 2014, 19, 19–29. [Google Scholar] [CrossRef]
  178. Niu, S.; Xu, L.; Yuan, Y.; Yang, S.; Ning, H.; Qin, X.; Xin, P.; Yuan, D.; Jiao, J.; Zhao, Y. Effect of down-regulated miR-15b-5p expression on arrhythmia and myocardial apoptosis after myocardial ischemia reperfusion injury in mice. Biochem. Biophys. Res. Commun. 2020, 530, 54–59. [Google Scholar] [CrossRef] [PubMed]
  179. Wang, P.; Sun, J.; Lv, S.; Xie, T.; Wang, X. Apigenin alleviates myocardial reperfusion injury in rats by downregulating miR-15b. Med. Sci. Monit. 2019, 25, 2764–2776. [Google Scholar] [CrossRef] [PubMed]
  180. Kou, Y.; Zheng, W.T.; Zhang, Y.R. Inhibition of miR-23 protects myocardial function from ischemia-reperfusion injury through restoration of glutamine metabolism. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 4286–4293. [Google Scholar]
  181. Lu, M.; Xu, Y.; Wang, M.; Guo, T.; Luo, F.; Su, N.; Wang, Z.; Xu, L.; Liu, Z. MicroRNA-23 inhibition protects the ischemia/reperfusion injury via inducing the differentiation of bone marrow mesenchymal stem cells into cardiomyocytes. Int. J. Clin. Exp. Pathol. 2019, 12, 1060. [Google Scholar]
  182. Hijmans, J.G.; Diehl, K.J.; Bammert, T.D.; Kavlich, P.J.; Lincenberg, G.M.; Greiner, J.J.; Stauffer, B.L.; DeSouza, C.A. Influence of overweight and obesity on circulating inflammation-related microRNA. MicroRNA 2018, 7, 148–154. [Google Scholar] [CrossRef] [PubMed]
  183. Manoel Alves, J.; Handerson Gomes Teles, R.; do Valle Gomes Gatto, C.; Muñoz, V.R.; Regina Cominetti, M.; Garcia de Oliveira Duarte, A.C. Mapping research in the obesity, adipose tissue, and microrna field: A bibliometric analysis. Cells 2019, 8, 1581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Yang, Y.; Cheng, H.-W.; Qiu, Y.; Dupee, D.; Noonan, M.; Lin, Y.-D.; Fisch, S.; Unno, K.; Sereti, K.-I.; Liao, R. MicroRNA-34a plays a key role in cardiac repair and regeneration following myocardial infarction. Circ. Res. 2015, 117, 450–459. [Google Scholar] [CrossRef] [PubMed]
  185. Zhao, T.; Li, J.; Chen, A.F. MicroRNA-34a induces endothelial progenitor cell senescence and impedes its angiogenesis via suppressing silent information regulator 1. Am. J. Physiol. Metab. 2010, 299, E110–E116. [Google Scholar] [CrossRef] [Green Version]
  186. Li, J.H.; Dai, J.; Han, B.; Wu, G.-H.; Wang, C.-H. MiR-34a regulates cell apoptosis after myocardial infarction in rats through the Wnt/beta-catenin signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 2555–2562. [Google Scholar] [CrossRef]
  187. Wang, X.; Yuan, B.; Cheng, B.; Liu, Y.; Zhang, B.; Wang, X.; Lin, X.; Yang, B.; Gong, G. Crocin alleviates myocardial ischemia/reperfusion-induced endoplasmic reticulum stress via regulation of mir-34a/sirt1/nrf2 pathway. Shock 2019, 51, 123–130. [Google Scholar] [CrossRef] [PubMed]
  188. Dong, F.-F.; Dong, S.-H.; Liang, Y.; Wang, K.; Qin, Y.-W.; Zhao, X.-X. MiR-34a promotes myocardial infarction in rats by inhibiting the activity of SIRT1. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 7059–7065. [Google Scholar]
  189. Heneghan, H.; Miller, N.; McAnena, O.J.; O’Brien, T.; Kerin, M.J. Differential miRNA Expression in omental adipose tissue and in the circulation of obese patients identifies novel metabolic biomarkers. J. Clin. Endocrinol. Metab. 2011, 96, E846–E850. [Google Scholar] [CrossRef]
  190. Wang, R.; Hong, J.; Cao, Y.; Shi, J.; Gu, W.; Ning, G.; Zhang, Y.; Wang, W. Elevated circulating microRNA-122 is associated with obesity and insulin resistance in young adults. Eur. J. Endocrinol. 2015, 172, 291–300. [Google Scholar] [CrossRef]
  191. Zhang, Z.; Li, H.; Chen, S.; Li, Y.; Cui, Z.; Ma, J. MicroRNA-122 regulates caspase-8 and promotes the apoptosis of mouse cardiomyocytes. Braz. J. Med. Biol. Res. 2017, 50, e5760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Peng, H.; Luo, Y.; Ying, Y. lncRNA XIST attenuates hypoxia-induced H9c2 cardiomyocyte injury by targeting the miR-122-5p/FOXP2 axis. Mol. Cell. Probes 2020, 50, 101500. [Google Scholar] [CrossRef] [PubMed]
  193. Zhang, Z.; Li, H.; Cui, Z.; Zhou, Z.; Chen, S.; Ma, J.; Hou, L.; Pan, X.; Li, Q. Long non-coding RNA UCA1 relieves cardiomyocytes H9c2 injury aroused by oxygen-glucose deprivation via declining miR-122. Artif. Cells Nanomed. Biotechnol. 2019, 47, 3492–3499. [Google Scholar] [CrossRef] [PubMed]
  194. Thomé, J.G.; Mendoza, M.R.; Cheuiche, A.V.; La Porta, V.L.; Silvello, D.; dos Santos, K.G.; Andrades, M.E.; Clausell, N.; Rohde, L.E.; Biolo, A. Circulating microRNAs in obese and lean heart failure patients: A case–control study with computational target prediction analysis. Gene 2015, 574, 1–10. [Google Scholar] [CrossRef] [Green Version]
  195. Al-Rawaf, H.A. Circulating microRNAs and adipokines as markers of metabolic syndrome in adolescents with obesity. Clin. Nutr. 2019, 38, 2231–2238. [Google Scholar] [CrossRef]
  196. Liang, Y.; Li, J.; Xiao, H.; He, Y.; Zhang, L.; Yan, Y. Identification of stress-related microRNA biomarkers in type 2 diabetes mellitus: A systematic review and meta-analysis. J. Diabetes 2020, 12, 633–644. [Google Scholar] [CrossRef] [Green Version]
  197. Chu, X.; Wang, Y.; Pang, L.; Huang, J.; Sun, X.; Chen, X. miR-130 aggravates acute myocardial infarction-induced myocardial injury by targeting PPAR-gamma. J. Cell. Biochem. 2018, 119, 7235–7244. [Google Scholar] [CrossRef]
  198. Huang, G.; Hao, F.; Hu, X. Downregulation of microRNA-155 stimulates sevoflurane-mediated cardioprotection against myocardial ischemia/reperfusion injury by binding to SIRT1 in mice. J. Cell. Biochem. 2019, 120, 15494–15505. [Google Scholar] [CrossRef] [PubMed]
  199. Eisenhardt, S.U.; Weiss, J.B.W.; Smolka, C.; Maxeiner, J.; Pankratz, F.; Bemtgen, X.; Kustermann, M.; Thiele, J.R.; Schmidt, Y.; Stark, G.B.; et al. MicroRNA-155 aggravates ischemia–reperfusion injury by modulation of inflammatory cell recruitment and the respiratory oxidative burst. Basic Res. Cardiol. 2015, 110, 32. [Google Scholar] [CrossRef]
  200. Villard, A.; Marchand, L. Diagnostic value of cell-free circulating micrornas for obesity and type 2 diabetes: A meta-analysis. J. Mol. Biomark. Diagn. 2015, 6, 1–9. [Google Scholar] [CrossRef] [Green Version]
  201. Karolina, D.S.; Tavintharan, S.; Armugam, A.; Sepramaniam, S.; Pek, S.L.T.; Wong, M.T.K.; Lim, S.C.; Sum, C.F.; Jeyaseelan, K. Circulating miRNA profiles in patients with metabolic syndrome. J. Clin. Endocrinol. Metab. 2012, 97, E2271–E2276. [Google Scholar] [CrossRef] [Green Version]
  202. Zhang, Y.; Huang, R.; Zhou, W.; Zhao, Q.; Lü, Z. miR-192-5p mediates hypoxia/reoxygenation-induced apoptosis in H9c2 cardiomyocytes via targeting of FABP3. J. Biochem. Mol. Toxicol. 2017, 31, e21873. [Google Scholar] [CrossRef] [PubMed]
  203. Cao, L.; Chai, S. miR-320-3p is involved in morphine pre-conditioning to protect rat cardiomyocytes from ischemia/reperfusion injury through targeting Akt3. Mol. Med. Rep. 2020, 22, 1480–1488. [Google Scholar] [CrossRef] [PubMed]
  204. Gallo, W.; Esguerra, J.L.S.; Eliasson, L.; Melander, O. miR-483-5p associates with obesity and insulin resistance and independently associates with new onset diabetes mellitus and cardiovascular disease. PLoS ONE 2018, 13, e0206974. [Google Scholar] [CrossRef]
  205. Gallo, W.; Ottosson, F.; Kennbäck, C.; Jujic, A.; Esguerra, J.L.S.; Eliasson, L.; Melander, O. Replication study reveals miR-483-5p as an important target in prevention of cardiometabolic disease. BMC Cardiovasc. Disord. 2021, 21, 162. [Google Scholar] [CrossRef]
  206. Zhang, H.; Wang, J.; Du, A.; Li, Y. MiR-483-3p inhibition ameliorates myocardial ischemia/reperfusion injury by targeting the MDM4/p53 pathway. Mol. Immunol. 2020, 125, 9–14. [Google Scholar] [CrossRef]
  207. Sun, H.; Cai, J.; Xu, L.; Liu, J.; Chen, M.; Zheng, M.; Wang, L.; Yang, X. miR-483-3p regulates acute myocardial infarction by transcriptionally repressing insulin growth factor 1 expression. Mol. Med. Rep. 2018, 17, 4785–4790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Qiao, Y.; Zhao, Y.; Liu, Y.; Ma, N.; Wang, C.; Zou, J.; Liu, Z.; Zhou, Z.; Han, D.; He, J.; et al. miR-483-3p regulates hyperglycaemia-induced cardiomyocyte apoptosis in transgenic mice. Biochem. Biophys. Res. Commun. 2016, 477, 541–547. [Google Scholar] [CrossRef]
  209. Chien, H.Y.; Lee, T.-P.; Chen, C.-Y.; Chiu, Y.-H.; Lin, Y.-C.; Lee, L.-S.; Li, W.-C. Circulating microRNA as a diagnostic marker in populations with type 2 diabetes mellitus and diabetic complications. J. Chin. Med. Assoc. 2015, 78, 204–211. [Google Scholar] [CrossRef] [Green Version]
  210. Sekar, D.; Venugopal, B.; Sekar, P.; Ramalingam, K. Role of microRNA 21 in diabetes and associated/related diseases. Gene 2016, 582, 14–18. [Google Scholar] [CrossRef]
  211. Liao, Z.; Chen, Y.; Duan, C.; Zhu, K.; Huang, R.; Zhao, H.; Hintze, M.; Pu, Q.; Yuan, Z.; Lv, L.; et al. Cardiac telocytes inhibit cardiac microvascular endothelial cell apoptosis through exosomal miRNA-21-5p-targeted cdip1 silencing to improve angiogenesis following myocardial infarction. Theranostics 2021, 11, 268–291. [Google Scholar] [CrossRef] [PubMed]
  212. Cheng, Y.; Liu, X.; Zhang, S.; Lin, Y.; Yang, J.; Zhang, C. MicroRNA-21 protects against the H2O2-induced injury on cardiac myocytes via its target gene PDCD4. J. Mol. Cell. Cardiol. 2009, 47, 5–14. [Google Scholar] [CrossRef] [Green Version]
  213. Cheng, Y.; Zhu, P.; Yang, J.; Liu, X.; Dong, S.; Wang, X.; Chun, B.; Zhuang, J.; Zhang, C. Ischaemic preconditioning-regulated miR-21 protects heart against ischaemia/reperfusion injury via anti-apoptosis through its target PDCD4. Cardiovasc. Res. 2010, 87, 431–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Ma, N.; Bai, J.; Zhang, W.; Luo, H.; Zhang, X.; Liu, D.; Qiao, C. Trimetazidine protects against cardiac ischemia/reperfusion injury via effects on cardiac miRNA-21 expression, Akt and the Bcl-2/Bax pathway. Mol. Med. Rep. 2016, 14, 4216–4222. [Google Scholar] [CrossRef] [Green Version]
  215. Qiao, S.; Olson, J.; Paterson, M.; Yan, Y.; Zaja, I.; Liu, Y.; Riess, M.L.; Kersten, J.R.; Liang, M.; Warltier, D.C.; et al. MicroRNA-21 mediates isoflurane-induced cardioprotection against ischemia–reperfusion injury via akt/nitric oxide synthase/mitochondrial permeability transition pore pathway. Anesthesiology 2015, 123, 786–798. [Google Scholar] [CrossRef] [Green Version]
  216. Yang, L.; Wang, B.; Zhou, Q.; Wang, Y.; Liu, X.; Liu, Z.; Zhan, Z. MicroRNA-21 prevents excessive inflammation and cardiac dysfunction after myocardial infarction through targeting KBTBD7. Cell Death Dis. 2018, 9, 769. [Google Scholar] [CrossRef] [Green Version]
  217. Salloum, F.; Yin, C.; Kukreja, R.C. Role of microRNAs in cardiac preconditioning. J. Cardiovasc. Pharmacol. 2010, 56, 581–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Masi, L.N.; Lotufo, P.A.; Ferreira, F.M.; Rodrigues, A.C.; Serdan, T.D.A.; Braga, A.A.; Cury, D.P.; Hirata, M.J.; Curi, R.; Bensenor, I.J.M.; et al. Profiling plasma-extracellular vesicle proteins and microRNAs in diabetes onset in middle-aged male participants in the ELSA-Brasil study. Physiol. Rep. 2021, 9, e14731. [Google Scholar] [CrossRef] [PubMed]
  219. Ruan, Z.; Wang, S.; Yu, W.; Deng, F. LncRNA MALAT1 aggravates inflammation response through regulating PTGS2 by targeting miR-26b in myocardial ischemia-reperfusion injury. Int. J. Cardiol. 2019, 288, 122. [Google Scholar] [CrossRef]
  220. Chen, X.; Ding, Z.; Li, T.; Jiang, W.; Zhang, J.; Deng, X. MicroR-26b Targets High Mobility Group, AT-hook 2 to Ameliorate Myocardial Infarction-induced Fibrosis by Suppression of Cardiac Fibroblasts Activation. Curr. Neurovasc. Res. 2020, 17, 204–213. [Google Scholar] [CrossRef]
  221. Jiang, T.; Liu, Y.; Chen, B.; Si, L. Identification of potential molecular mechanisms and small molecule drugs in myocardial ischemia/reperfusion injury. Braz. J. Med. Biol. Res. 2020, 53. [Google Scholar] [CrossRef]
  222. Ortega, F.J.; Mercader, J.M.; Moreno-Navarrete, J.M.; Rovira, O.; Guerra, E.; Esteve, E.; Xifra, G.; Martínez, C.; Ricart, W.; Rieusset, J.; et al. Profiling of circulating micrornas reveals common micrornas linked to type 2 diabetes that change with insulin sensitization. Diabetes Care 2014, 37, 1375–1383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Su, Q.; Liu, Y.; Lv, X.-W.; Ye, Z.-L.; Sun, Y.-H.; Kong, B.-H.; Qin, Z.-B. Inhibition of lncRNA TUG1 upregulates miR-142-3p to ameliorate myocardial injury during ischemia and reperfusion via targeting HMGB1- and Rac1-induced autophagy. J. Mol. Cell. Cardiol. 2019, 133, 12–25. [Google Scholar] [CrossRef] [PubMed]
  224. Wang, Y.; Ouyang, M.; Wang, Q.; Jian, Z. MicroRNA-142-3p inhibits hypoxia/reoxygenation-induced apoptosis and fibrosis of cardiomyocytes by targeting high mobility group box 1. Int. J. Mol. Med. 2016, 38, 1377–1386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Su, Q.; Lv, X.; Ye, Z.; Sun, Y.; Kong, B.; Qin, Z.; Li, L. The mechanism of miR-142-3p in coronary microembolization-induced myocardiac injury via regulating target gene IRAK-1. Cell Death Dis. 2019, 10, 61. [Google Scholar] [CrossRef] [Green Version]
  226. Cui, X.; He, Z.; Liang, Z.; Chen, Z.; Wang, H.; Zhang, J. Exosomes from adipose-derived mesenchymal stem cells protect the myocardium against ischemia/reperfusion injury through wnt/beta-catenin signaling pathway. J. Cardiovasc. Pharmacol. 2017, 70, 225–231. [Google Scholar] [CrossRef] [Green Version]
  227. Rong, Y.; Bao, W.; Shan, Z.; Liu, J.; Yu, X.; Xia, S.; Gao, H.; Wang, X.; Yao, P.; Hu, F.B.; et al. Increased microRNA-146a levels in plasma of patients with newly diagnosed type 2 diabetes mellitus. PLoS ONE 2013, 8, e73272. [Google Scholar] [CrossRef] [Green Version]
  228. Wang, X.; Ha, T.; Liu, L.; Zou, J.; Zhang, X.; Kalbfleisch, J.; Gao, X.; Williams, D.; Li, C. Increased expression of microRNA-146a decreases myocardial ischaemia/reperfusion injury. Cardiovasc. Res. 2013, 97, 432–442. [Google Scholar] [CrossRef] [Green Version]
  229. Zhang, T.; Ma, Y.; Gao, L.; Mao, C.; Zeng, H.; Wang, X.; Sun, Y.; Gu, J.; Wang, Y.; Chen, K.; et al. MicroRNA-146a protects against myocardial ischaemia reperfusion injury by targeting Med1. Cell. Mol. Biol. Lett. 2019, 24, 62. [Google Scholar] [CrossRef]
  230. Zhang, W.; Shao, M.; He, X.; Wang, B.; Li, Y.; Guo, X. Overexpression of microRNA-146 protects against oxygen-glucose deprivation/recovery-induced cardiomyocyte apoptosis by inhibiting the NF-κB/TNF-α signaling pathway. Mol. Med. Rep. 2017, 17, 1913–1918. [Google Scholar] [CrossRef]
  231. Di, Y.-F.; Li, D.-C.; Shen, Y.-Q.; Wang, C.-L.; Zhang, D.-Y.; Shang, A.; Hu, T. MiR-146b protects cardiomyocytes injury in myocardial ischemia/reperfusion by targeting Smad4. Am. J. Transl. Res. 2017, 9, 656–663. [Google Scholar] [PubMed]
  232. Pan, J.; Alimujiang, M.; Chen, Q.; Shi, H.; Luo, X. Exosomes derived from miR-146a-modified adipose-derived stem cells attenuate acute myocardial infarction−induced myocardial damage via downregulation of early growth response factor 1. J. Cell. Biochem. 2019, 120, 4433–4443. [Google Scholar] [CrossRef]
  233. Su, Q.; Xu, Y.; Cai, R.; Dai, R.; Yang, X.; Liu, Y.; Kong, B. miR-146a inhibits mitochondrial dysfunction and myocardial infarction by targeting cyclophilin D. Mol. Ther. Nucleic Acids 2021, 23, 1258–1271. [Google Scholar] [CrossRef] [PubMed]
  234. Seo, H.-H.; Lee, S.-Y.; Lee, C.Y.; Kim, R.; Kim, P.; Oh, S.; Lee, H.; Lee, M.Y.; Kim, J.; Kim, L.K.; et al. Exogenous miRNA-146a enhances the therapeutic efficacy of human mesenchymal stem cells by increasing vascular endothelial growth factor secretion in the ischemia/reperfusion-injured heart. J. Vasc. Res. 2017, 54, 100–108. [Google Scholar] [CrossRef] [PubMed]
  235. Wu, Z.; Cheng, S.; Wang, S.; Li, W.; Liu, J. BMSCs-derived exosomal microRNA-150-5p attenuates myocardial infarction in mice. Int. Immunopharmacol. 2021, 93, 107389. [Google Scholar] [CrossRef]
  236. Zhou, Y.; Chen, Q.; Lew, K.S.; Richards, A.M.; Wang, P. Discovery of potential therapeutic mirna targets in cardiac ischemia–reperfusion injury. J. Cardiovasc. Pharmacol. Ther. 2016, 21, 296–309. [Google Scholar] [CrossRef] [PubMed]
  237. Ou, H.; Teng, H.; Qin, Y.; Luo, X.; Yang, P.; Zhang, W.; Chen, W.; Lv, D.; Tang, H. Extracellular vesicles derived from microRNA-150-5p-overexpressing mesenchymal stem cells protect rat hearts against ischemia/reperfusion. Aging 2020, 12, 12669–12683. [Google Scholar] [CrossRef]
  238. Lin, D.; Cui, B.; Ma, J.; Ren, J. MiR-183-5p protects rat hearts against myocardial ischemia/reperfusion injury through targeting VDAC1. BioFactors 2020, 46, 83–93. [Google Scholar] [CrossRef]
  239. Xing, J.; Xie, T.; Tan, W.; Li, R.; Yu, C.; Han, X. microRNA-183 improve myocardial damager via NF-kb pathway: In vitro and in vivo study. J. Cell. Biochem. 2018, 120, 10145–10154. [Google Scholar] [CrossRef]
  240. Gong, L.; Xu, H.; Chang, H.; Tong, Y.; Zhang, T.; Guo, G. Knockdown of long non-coding RNA MEG3 protects H9c2 cells from hypoxia-induced injury by targeting microRNA-183. J. Cell. Biochem. 2018, 119, 1429–1440. [Google Scholar] [CrossRef]
  241. Prats-Puig, A.; Ortega, F.J.; Mercader, J.M.; Moreno-Navarrete, J.M.; Moreno, M.; Bonet, N.; Ricart, W.; López-Bermejo, A.; Fernández-Real, J.M. Changes in circulating micrornas are associated with childhood obesity. J. Clin. Endocrinol. Metab. 2013, 98, E1655–E1660. [Google Scholar] [CrossRef]
  242. Taheri, M.; Eghtedarian, R.; Ghafouri-Fard, S.; Omrani, M.D. Non-coding RNAs and type 2 diabetes mellitus. Arch. Physiol. Biochem. 2020, 1–10. [Google Scholar] [CrossRef]
  243. Marzano, F.; Faienza, M.F.; Caratozzolo, M.F.; Brunetti, G.; Chiara, M.; Horner, D.S.; Annese, A.; D’Erchia, A.M.; Consiglio, A.; Pesole, G.; et al. Pilot study on circulating miRNA signature in children with obesity born small for gestational age and appropriate for gestational age. Pediatr. Obes. 2018, 13, 803–811. [Google Scholar] [CrossRef]
  244. Sun, X.-H.; Wang, X.; Zhang, Y.; Hui, J. Exosomes of bone-marrow stromal cells inhibit cardiomyocyte apoptosis under ischemic and hypoxic conditions via miR-486-5p targeting the PTEN/PI3K/AKT signaling pathway. Thromb. Res. 2019, 177, 23–32. [Google Scholar] [CrossRef]
  245. Zhu, H.-H.; Wang, X.-T.; Sun, Y.-H.; He, W.-K.; Liang, J.-B.; Mo, B.-H.; Li, L. MicroRNA-486-5p targeting PTEN Protects Against Coronary Microembolization-Induced Cardiomyocyte Apoptosis in Rats by activating the PI3K/AKT pathway. Eur. J. Pharmacol. 2019, 855, 244–251. [Google Scholar] [CrossRef]
  246. Zhang, X.; Zhang, C.; Wang, N.; Li, Y.; Zhang, D.; Li, Q. MicroRNA-486 alleviates hypoxia-induced damage in h9c2 cells by targeting ndrg2 to inactivate jnk/c-jun and nf-κb signaling pathways. Cell. Physiol. Biochem. 2018, 48, 2483–2492. [Google Scholar] [CrossRef]
  247. Li, Q.; Xu, Y.; Lv, K.; Wang, Y.; Zhong, Z.; Xiao, C.; Zhu, K.; Ni, C.; Wang, K.; Kong, M.; et al. Small extracellular vesicles containing miR-486-5p promote angiogenesis after myocardial infarction in mice and nonhuman primates. Sci. Transl. Med. 2021, 13, 584. [Google Scholar] [CrossRef]
  248. Ren, D.; Li, F.; Gao, A.; Cao, Q.; Liu, Y.; Zhang, J. Hypoxia-induced apoptosis of cardiomyocytes is restricted by ginkgolide B-downregulated microRNA-29. Cell Cycle 2020, 19, 1067–1076. [Google Scholar] [CrossRef]
  249. Tian, R.; Guan, X.; Qian, H.; Wang, L.; Shen, Z.; Fang, L.; Liu, Z. Restoration of NRF2 attenuates myocardial ischemia reperfusion injury through mediating microRNA -29a-3p/ CCNT2 axis. BioFactors 2021, 47, 414–426. [Google Scholar] [CrossRef]
  250. Hong, H.; Tao, T.; Chen, S.; Liang, C.; Qiu, Y.; Zhou, Y.; Zhang, R. MicroRNA-143 promotes cardiac ischemia-mediated mitochondrial impairment by the inhibition of protein kinase Cepsilon. Basic Res. Cardiol. 2017, 112, 60. [Google Scholar] [CrossRef]
  251. Ma, W.-Y.; Song, R.-J.; Xu, B.-B.; Xu, Y.; Wang, X.-X.; Sun, H.-Y.; Li, S.-N.; Liu, S.-Z.; Yu, M.-X.; Yang, F.; et al. Melatonin promotes cardiomyocyte proliferation and heart repair in mice with myocardial infarction via miR-143-3p/Yap/Ctnnd1 signaling pathway. Acta Pharmacol. Sin. 2020, 42, 921–931. [Google Scholar] [CrossRef]
  252. Li, C.; Li, J.; Xue, K.; Zhang, J.; Wang, C.; Zhang, Q.; Chen, X.; Gao, C.; Yu, X.; Sun, L. MicroRNA-143-3p promotes human cardiac fibrosis via targeting sprouty3 after myocardial infarction. J. Mol. Cell. Cardiol. 2019, 129, 281–292. [Google Scholar] [CrossRef]
  253. Liu, K.; Zhao, D.; Wang, D. LINC00528 regulates myocardial infarction by targeting the miR-143-3p/COX-2 axis. Bioengineered 2020, 11, 11–18. [Google Scholar] [CrossRef] [Green Version]
  254. Wen, D.; Qiao, P.; Wang, L. Circulating microRNA-223 as a potential biomarker for obesity. Obes. Res. Clin. Pract. 2015, 9, 398–404. [Google Scholar] [CrossRef]
  255. Sánchez-Ceinos, J.; Rangel-Zuñiga, O.A.; Clemente-Postigo, M.; Podadera-Herreros, A.; Camargo, A.; Alcalá-Diaz, J.F.; Guzmán-Ruiz, R.; López-Miranda, J.; Malagón, M.M. miR-223-3p as a potential biomarker and player for adipose tissue dysfunction preceding type 2 diabetes onset. Mol. Ther. Nucleic Acids 2021, 23, 1035–1052. [Google Scholar] [CrossRef]
  256. Yan, L.-N.; Zhang, X.; Xu, F.; Fan, Y.-Y.; Ge, B.; Guo, H.; Li, Z.-L. Four-microRNA signature for detection of type 2 diabetes. World J. Clin. Cases 2020, 8, 1923–1931. [Google Scholar] [CrossRef]
  257. Tang, Q.; Li, M.-Y.; Su, Y.-F.; Fu, J.; Zou, Z.-Y.; Wang, Y.; Li, S.-N. Absence of miR-223-3p ameliorates hypoxia-induced injury through repressing cardiomyocyte apoptosis and oxidative stress by targeting KLF15. Eur. J. Pharmacol. 2018, 841, 67–74. [Google Scholar] [CrossRef]
  258. Liu, X.; Xu, Y.; Deng, Y.; Li, H. MicroRNA-223 regulates cardiac fibrosis after myocardial infarction by targeting RASA1. Cell. Physiol. Biochem. 2018, 46, 1439–1454. [Google Scholar] [CrossRef]
  259. Dai, G.-H.; Ma, P.-Z.; Song, X.-B.; Liu, N.; Zhang, T.; Wu, B. MicroRNA-223-3p inhibits the angiogenesis of ischemic cardiac microvascular endothelial cells via affecting rps6kb1/hif-1a signal pathway. PLoS ONE 2014, 9, e108468. [Google Scholar] [CrossRef] [Green Version]
  260. Liu, X.; Deng, Y.; Xu, Y.; Jin, W.; Li, H. MicroRNA-223 protects neonatal rat cardiomyocytes and H9c2 cells from hypoxia-induced apoptosis and excessive autophagy via the Akt/mTOR pathway by targeting PARP-1. J. Mol. Cell. Cardiol. 2018, 118, 133–146. [Google Scholar] [CrossRef]
  261. Qin, D.; Wang, X.; Li, Y.; Yang, L.; Wang, R.; Peng, J.; Essandoh, K.; Mu, X.; Peng, T.; Han, Q.; et al. MicroRNA-223-5p and -3p cooperatively suppress necroptosis in ischemic/reperfused hearts. J. Biol. Chem. 2016, 291, 20247–20259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Zhao, L.; Jiang, S.; Wu, N.; Shi, E.; Yang, L.; Li, Q. MiR-17-5p-mediated endoplasmic reticulum stress promotes acute myocardial ischemia injury through targeting Tsg101. Cell Stress Chaperon 2021, 26, 77–90. [Google Scholar] [CrossRef] [PubMed]
  263. Du, W.; Pan, Z.; Chen, X.; Wang, L.; Zhang, Y.; Li, S.; Liang, H.; Xu, C.; Wu, Y.; Shan, H.; et al. By targeting Stat3 microRNA-17-5p promotes cardiomyocyte apoptosis in response to ischemia followed by reperfusion. Cell. Physiol. Biochem. 2014, 34, 955–965. [Google Scholar] [CrossRef] [PubMed]
  264. Yang, S.; Fan, T.; Hu, Q.; Xu, W.; Yang, J.; Xu, C.; Zhang, B.; Chen, J.; Jiang, H. Downregulation of microRNA-17-5p improves cardiac function after myocardial infarction via attenuation of apoptosis in endothelial cells. Mol. Genet. Genom. 2018, 293, 883–894. [Google Scholar] [CrossRef]
  265. Zampetaki, A.; Kiechl, S.; Drozdov, I.; Willeit, P.; Mayr, U.; Prokopi, M.; Mayr, A.; Weger, S.; Oberhollenzer, F.; Bonora, E.; et al. Plasma microrna profiling reveals loss of endothelial mir-126 and other micrornas in type 2 diabetes. Circ. Res. 2010, 107, 810–817. [Google Scholar] [CrossRef]
  266. Ren, Y.; Bao, R.; Guo, Z.; Kai, J.; Cai, C.G.; Li, Z. miR-126-5p regulates H9c2 cell proliferation and apoptosis under hypoxic conditions by targeting IL-17A. Exp. Ther. Med. 2020, 21, 67. [Google Scholar] [CrossRef]
  267. Su, Q.; Liu, Y.; Lv, X.-W.; Dai, R.-X.; Yang, X.-H.; Kong, B.-H. LncRNA TUG1 mediates ischemic myocardial injury by targeting miR-132-3p/HDAC3 axis. Am. J. Physiol. Heart Circ. Physiol. 2020, 318, H332–H344. [Google Scholar] [CrossRef]
  268. Zhao, Z.; Du, S.; Shen, S.; Wang, L. microRNA-132 inhibits cardiomyocyte apoptosis and myocardial remodeling in myocardial infarction by targeting IL-1beta. J. Cell. Physiol. 2020, 235, 2710–2721. [Google Scholar] [CrossRef]
  269. Chen, L.; Wang, G.-Y.; Dong, J.-H.; Cheng, X.-J. MicroRNA-132 improves myocardial remodeling after myocardial infarction. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 6299–6306. [Google Scholar]
  270. Zhang, J.; Xu, H.; Gong, L.; Liu, L. MicroRNA-132 protects H9c2 cells against oxygen and glucose deprivation-evoked injury by targeting FOXO3A. J. Cell. Physiol. 2020, 235, 176–184. [Google Scholar] [CrossRef]
  271. Ma, T.; Chen, Y.; Chen, Y.; Meng, Q.; Sun, J.; Shao, L.; Yu, Y.; Huang, H.; Hu, Y.; Yang, Z.; et al. MicroRNA-132, Delivered by Mesenchymal Stem Cell-Derived Exosomes, Promote Angiogenesis in Myocardial Infarction. Stem Cells Int. 2018, 2018, 3290372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  272. Shahrokhi, S.Z.; Saeidi, L.; Sadatamini, M.; Jafarzadeh, M.; Rahimipour, A.; Kazerouni, F. Can miR-145-5p be used as a marker in diabetic patients? Arch. Physiol. Biochem. 2020, 1–6. [Google Scholar] [CrossRef] [PubMed]
  273. Yan, L.; Guo, N.; Cao, Y.; Zeng, S.; Wang, J.; Lv, F.; Wang, Y.; Cao, X. miRNA-145 inhibits myocardial infarction-induced apoptosis through autophagy via Akt3/mTOR signaling pathway in vitro and in vivo. Int. J. Mol. Med. 2018, 42, 1537–1547. [Google Scholar] [CrossRef] [Green Version]
  274. Liu, Z.; Fan, S.; Tao, B.; Pu, Y.; Xia, H.; Xu, L. microRNA-145 Protects against Myocardial Ischemia Reperfusion Injury via CaMKII-Mediated Antiapoptotic and Anti-Inflammatory Pathways. Oxid. Med. Cell. Longev. 2019, 2019, 8948657. [Google Scholar] [CrossRef] [PubMed]
  275. Yuan, M.; Zhang, L.; You, F.; Zhou, J.; Ma, Y.; Yang, F.; Tao, L. MiR-145-5p regulates hypoxia-induced inflammatory response and apoptosis in cardiomyocytes by targeting CD40. Mol. Cell. Biochem. 2017, 431, 123–131. [Google Scholar] [CrossRef]
  276. Qi, Z.; Li, S.; Su, Y.; Zhang, J.; Kang, Y.; Huang, Y.; Jin, F.; Xing, Q. Role of microRNA-145 in protection against myocardial ischemia/reperfusion injury in mice by regulating expression of GZMK with the treatment of sevoflurane. J. Cell. Physiol. 2019, 234, 16526–16539. [Google Scholar] [CrossRef]
  277. Higashi, K.; Yamada, Y.; Minatoguchi, S.; Baba, S.; Iwasa, M.; Kanamori, H.; Kawasaki, M.; Nishigaki, K.; Takemura, G.; Kumazaki, M.; et al. MicroRNA-145 repairs infarcted myocardium by accelerating cardiomyocyte autophagy. Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H1813–H1826. [Google Scholar] [CrossRef] [Green Version]
  278. Song, H.-F.; He, S.; Li, S.-H.; Wu, J.; Yin, W.; Shao, Z.; Du, G.-Q.; Wu, J.; Li, J.; Weisel, R.D.; et al. Knock-out of MicroRNA 145 impairs cardiac fibroblast function and wound healing post-myocardial infarction. J. Cell. Mol. Med. 2020, 24, 9409–9419. [Google Scholar] [CrossRef]
  279. Iacomino, G.; Siani, A. Role of microRNAs in obesity and obesity-related diseases. Genes Nutr. 2017, 12, 23. [Google Scholar] [CrossRef]
  280. Yan, Y.; Dang, H.; Zhang, X.; Wang, X.; Liu, X. The protective role of MiR-206 in regulating cardiomyocytes apoptosis induced by ischemic injury by targeting PTP1B. Biosci. Rep. 2020, 40, BSR20191000. [Google Scholar] [CrossRef]
  281. Kong, F.; Jin, J.; Lv, X.; Han, Y.; Liang, X.; Gao, Y.; Duan, X. RETRACTED: Long noncoding RNA RMRP upregulation aggravates myocardial ischemia-reperfusion injury by sponging miR-206 to target ATG3 expression. Biomed. Pharmacother. 2019, 109, 716–725. [Google Scholar] [CrossRef]
  282. Zhai, C.; Qian, Q.; Tang, G.; Han, B.; Hu, H.; Yin, D.; Pan, H.; Zhang, S. MicroRNA-206 Protects against Myocardial Ischaemia-Reperfusion Injury in Rats by Targeting Gadd45beta. Mol. Cells 2017, 40, 916–924. [Google Scholar]
  283. Wu, L.; Dai, X.; Zhan, J.; Zhang, Y.; Zhang, H.; Zeng, S.; Xi, W.; Zhang, H. Profiling peripheral microRNAs in obesity and type 2 diabetes mellitus. APMIS 2015, 123, 580–585. [Google Scholar] [CrossRef]
  284. Liu, Y.; Zou, J.; Liu, X.; Zhang, Q. MicroRNA-138 attenuates myocardial ischemia reperfusion injury through inhibiting mitochondria-mediated apoptosis by targeting HIF1-α. Exp. Ther. Med. 2019, 18, 3325–3332. [Google Scholar] [CrossRef] [Green Version]
  285. Zhu, H.; Xue, H.; Jin, Q.-H.; Guo, J.; Chen, Y.-D. MiR-138 protects cardiac cells against hypoxia through modulation of glucose metabolism by targetting pyruvate dehydrogenase kinase 1. Biosci. Rep. 2017, 37, BSR20170296. [Google Scholar] [CrossRef] [Green Version]
  286. Mao, Q.; Liang, X.-L.; Zhang, C.-L.; Pang, Y.-H.; Lu, Y.-X. LncRNA KLF3-AS1 in human mesenchymal stem cell-derived exosomes ameliorates pyroptosis of cardiomyocytes and myocardial infarction through miR-138-5p/Sirt1 axis. Stem Cell Res. Ther. 2019, 10, 393. [Google Scholar] [CrossRef] [PubMed]
  287. Zhou, W.; Zhou, W.; Zeng, Q.; Xiong, J. MicroRNA-138 inhibits hypoxia-induced proliferation of endothelial progenitor cells via inhibition of HIF-1α-mediated MAPK and AKT signaling. Exp. Ther. Med. 2017, 13, 1017–1024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  288. Lisa, K.T.; Gilpin, E.; Ahnve, S.; Henning, H.; Ross, J. Smoking status at the time of acute myocardial infarction and subsequent prognosis. Am. Heart J. 1985, 110, 535–541. [Google Scholar] [CrossRef]
  289. Symons, R.; Masci, P.G.; Francone, M.; Claus, P.; Barison, A.; Carbone, I.; Agati, L.; Galea, N.; Janssens, S.; Bogaert, J. Impact of active smoking on myocardial infarction severity in reperfused ST-segment elevation myocardial infarction patients: The smoker’s paradox revisited. Eur. Heart J. 2016, 37, 2756–2764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  290. Fujita, Y.; Araya, J.; Ito, S.; Kobayashi, K.; Kosaka, N.; Yoshioka, Y.; Kadota, T.; Hara, H.; Kuwano, K.; Ochiya, T. Suppression of autophagy by extracellular vesicles promotes myofibroblast differentiation in COPD pathogenesis. J. Extracell. Vesicles 2015, 4, 28388. [Google Scholar] [CrossRef]
  291. Xu, L.; Deng, X. Tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone induces phosphorylation of mu- and m-calpain in association with increased secretion, cell migration, and invasion. J. Biol. Chem. 2004, 279, 53683–53690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Wu, F.; Yin, Z.; Yang, L.; Fan, J.; Xu, J.; Jin, Y.; Yu, J.; Zhang, D.; Yang, G. Smoking Induced Extracellular Vesicles Release and Their Distinct Properties in Non-Small Cell Lung Cancer. J. Cancer 2019, 10, 3435–3443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Baek, R.; Varming, K.; Jorgensen, M.M. Does smoking, age or gender affect the protein phenotype of extracellular vesicles in plasma? Transfus. Apher. Sci. 2016, 55, 44–52. [Google Scholar] [CrossRef] [Green Version]
  294. Mobarrez, F.; Antoniewicz, L.; Bosson, J.A.; Kuhl, J.; Pisetsky, D.S.; Lundbäck, M. The Effects of Smoking on Levels of Endothelial Progenitor Cells and Microparticles in the Blood of Healthy Volunteers. PLoS ONE 2014, 9, e90314. [Google Scholar] [CrossRef] [Green Version]
  295. Enjeti, A.K.; Ariyarajah, A.; D’Crus, A.; Seldon, M.; Lincz, L.F. Circulating microvesicle number, function and small RNA content vary with age, gender, smoking status, lipid and hormone profiles. Thromb. Res. 2017, 156, 65–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Benedikter, B.J.; Volgers, C.; Van Eijck, P.H.; Wouters, E.F.; Savelkoul, P.H.; Reynaert, N.L.; Haenen, G.R.; Rohde, G.G.; Weseler, A.R.; Stassen, F.R. Cigarette smoke extract induced exosome release is mediated by depletion of exofacial thiols and can be inhibited by thiol-antioxidants. Free Radic. Biol. Med. 2017, 108, 334–344. [Google Scholar] [CrossRef]
  297. Serban, K.A.; Rezania, S.; Petrusca, D.N.; Poirier, C.; Cao, D.; Justice, M.J.; Patel, M.; Tsvetkova, I.; Kamocki, K.; Mikosz, A.; et al. Structural and functional characterization of endothelial microparticles released by cigarette smoke. Sci. Rep. 2016, 6, 31596. [Google Scholar] [CrossRef]
  298. Cordazzo, C.; Petrini, S.; Neri, T.; Lombardi, S.; Carmazzi, Y.; Pedrinelli, R.; Paggiaro, P.; Celi, A. Rapid shedding of proinflammatory microparticles by human mononuclear cells exposed to cigarette smoke is dependent on Ca2+ mobilization. Inflamm. Res. 2014, 63, 539–547. [Google Scholar] [CrossRef]
  299. Li, C.-J.; Liu, Y.; Chen, Y.; Yu, D.; Williams, K.J.; Liu, M.-L. Novel Proteolytic Microvesicles Released from Human Macrophages after Exposure to Tobacco Smoke. Am. J. Pathol. 2013, 182, 1552–1562. [Google Scholar] [CrossRef] [Green Version]
  300. Andres, J.; Smith, L.C.; Murray, A.; Jin, Y.; Businaro, R.; Laskin, J.D.; Laskin, D.L. Role of extracellular vesicles in cell-cell communication and inflammation following exposure to pulmonary toxicants. Cytokine Growth Factor Rev. 2020, 51, 12–18. [Google Scholar] [CrossRef] [PubMed]
  301. Benedikter, B.J.; Bouwman, F.G.; Heinzmann, A.C.A.; Vajen, T.; Mariman, E.C.; Wouters, E.F.M.; Savelkoul, P.H.M.; Koenen, R.R.; Rohde, G.G.U.; Van Oerle, R.; et al. Proteomic analysis reveals procoagulant properties of cigarette smoke-induced extracellular vesicles. J. Extracell. Vesicles 2019, 8, 1585163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  302. Stassen, F.R.M.; van Eijck, P.H.; Savelkoul, P.H.M.; Wouters, E.F.M.; Rohde, G.G.U.; Briede, J.J.; Raynaert, N.L.; de Kok, T.M.; Benedikter, B.J. Cell type- and exposure-specific modulation of cd63/cd81-positive and tissue factor-positive extracellular vesicle release in response to respiratory toxicants. Oxid. Med. Cell. Longev. 2019, 2019, 5204218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  303. Li, M.; Yu, D.; Williams, K.J.; Liu, M.-L. Tobacco smoke induces the generation of procoagulant microvesicles from human monocytes/macrophages. Arter. Thromb. Vasc. Biol. 2010, 30, 1818–1824. [Google Scholar] [CrossRef] [PubMed]
  304. Haque, S.; Kodidela, S.; Sinha, N.; Kumar, P.; Cory, T.J.; Kumar, S. Differential packaging of inflammatory cytokines/ chemokines and oxidative stress modulators in U937 and U1 macrophages-derived extracellular vesicles upon exposure to tobacco constituents. PLoS ONE 2020, 15, e0233054. [Google Scholar] [CrossRef] [PubMed]
  305. Neri, M.; Fineschi, V.; Paolo, M.; Pomara, C.; Riezzo, I.; Turillazzi, E.; Cerretani, D. Cardiac oxidative stress and inflammatory cytokines response after myocardial infarction. Curr. Vasc. Pharmacol. 2015, 13, 26–36. [Google Scholar] [CrossRef]
  306. Toldo, S.; Mauro, A.G.; Cutter, Z.S.; Abbate, A. Inflammasome, pyroptosis, and cytokines in myocardial ischemia-reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 2018, 315, H1553–H1568. [Google Scholar] [CrossRef]
  307. Fujita, Y.; Araya, J.; Ochiya, T. Extracellular vesicles in smoking-related lung diseases. Oncotarget 2015, 6, 43144–43145. [Google Scholar] [CrossRef]
  308. Díaz, I.; Calderón-Sánchez, E.M.; Del Toro, R.; Avila-Medina, J.; Pedro, E.S.D.R.-D.; Domínguez-Rodríguez, A.; Rosado, J.A.; Hmadcha, A.; Ordoñez, A.; Smani, T. miR-125a, miR-139 and miR-324 contribute to Urocortin protection against myocardial ischemia-reperfusion injury. Sci. Rep. 2017, 7, 8898. [Google Scholar] [CrossRef]
  309. Wu, Q.; Shang, Y.; Bai, Y.; Wu, Y.; Wang, H.; Shen, T. Sufentanil preconditioning protects against myocardial ischemia/reperfusion injury via miR-125a/DRAM2 axis. Cell Cycle 2021, 20, 383–391. [Google Scholar] [CrossRef]
  310. Wu, X.Q.; Tian, X.-Y.; Wang, Z.-W.; Wu, X.; Wang, J.-P.; Yan, T.-Z. miR-191 secreted by platelet-derived microvesicles induced apoptosis of renal tubular epithelial cells and participated in renal ischemia-reperfusion injury via inhibiting CBS. Cell Cycle 2019, 18, 119–129. [Google Scholar] [CrossRef] [Green Version]
  311. Pan, W.; Wang, L.; Zhang, X.-D.; Zhang, H.; Zhang, J.; Wang, G.; Xu, P.; Zhang, Y.; Hu, P.; Du, R.-L.; et al. Hypoxia-induced microRNA-191 contributes to hepatic ischemia/reperfusion injury through the ZONAB/Cyclin D1 axis. Cell Death Differ. 2019, 26, 291–305. [Google Scholar] [CrossRef]
  312. Bao, M.-H.; Feng, X.; Zhang, Y.-W.; Lou, X.-Y.; Cheng, Y.; Zhou, H. Let-7 in Cardiovascular diseases, heart development and cardiovascular differentiation from stem cells. Int. J. Mol. Sci. 2013, 14, 23086–23102. [Google Scholar] [CrossRef] [Green Version]
  313. Badrnya, S.; Assinger, A.; Baumgartner, R. Smoking alters circulating plasma microvesicle pattern and microRNA signatures. Thromb. Haemost. 2014, 112, 128–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Zhong, Y.; Li, Y.-P.; Yin, Y.-Q.; Hu, B.-L.; Gao, H. Dexmedetomidine inhibits pyroptosis by down-regulating miR-29b in myocardial ischemia reperfusion injury in rats. Int. Immunopharmacol. 2020, 86, 106768. [Google Scholar] [CrossRef] [PubMed]
  315. Zhang, X.; Cheng, L.; Xu, L.; Zhang, Y.; Yang, Y.; Fu, Q.; Mi, W.; Li, H. The lncRNA, H19 Mediates the protective effect of hypoxia postconditioning against hypoxia-reoxygenation injury to senescent cardiomyocytes by targeting microRNA-29b-3p. Shock 2019, 52, 249–256. [Google Scholar] [CrossRef]
  316. Wang, P.-P.; Zhang, Y.-J.; Xie, T.; Sun, J.; Wang, X.-D. MiR-223 promotes cardiomyocyte apoptosis by inhibiting Foxo3a expression. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 6119–6126. [Google Scholar]
  317. Sundar, I.K.; Li, D.; Rahman, I. Small RNA-sequence analysis of plasma-derived extracellular vesicle miRNAs in smokers and patients with chronic obstructive pulmonary disease as circulating biomarkers. J. Extracell. Vesicles 2019, 8, 1684816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  318. McDonough, S.R.; Rahman, I.; Sundar, I.K. Recent updates on biomarkers of exposure and systemic toxicity in e-cigarette users and EVALI. Am. J. Physiol. Lung Cell. Mol. Physiol. 2021, 320, L661–L679. [Google Scholar] [CrossRef]
  319. Singh, K.P.; Maremanda, K.P.; Li, D.; Rahman, I. Exosomal microRNAs are novel circulating biomarkers in cigarette, waterpipe smokers, E-cigarette users and dual smokers. BMC Med. Genom. 2020, 13, 128. [Google Scholar] [CrossRef]
  320. Martínez, M.C.; Andriantsitohaina, R. Extracellular vesicles in metabolic syndrome. Circ. Res. 2017, 120, 1674–1686. [Google Scholar] [CrossRef]
  321. Georgescu, A.; Alexandru, N.; Andrei, E.; Titorencu, I.; Dragan, E.; Tarziu, C.; Ghiorghe, S.; Badila, E.; Bartos, D.; Popov, D. Circulating microparticles and endothelial progenitor cells in atherosclerosis: Pharmacological effects of irbesartan. J. Thromb. Haemost. 2012, 10, 680–691. [Google Scholar] [CrossRef]
  322. Nijiati, M.; Gao, Y.; Abudureheman, Z.; Yu, X.; Li, G. Relationship between the level of circulating endothelial micro-particles in the blood and blood lipid content in uyghur and han patients with acute coronary syndrome. Clin. Lab. 2015, 61, 1071–1075. [Google Scholar] [CrossRef]
  323. Pienimaeki-Roemer, A.; Fischer, A.; Tafelmeier, M.; Orsó, E.; Konovalova, T.; Böttcher, A.; Liebisch, G.; Reidel, A.; Schmitz, G. High-density lipoprotein 3 and apolipoprotein A-I alleviate platelet storage lesion and release of platelet extracellular vesicles. Transfusion 2014, 54, 2301–2314. [Google Scholar] [CrossRef]
  324. Hjuler Nielsen, M.; Irvine, H.; Vedel, S.; Raungaard, B.; Beck-Nielsen, H.; Handberg, A. Elevated atherosclerosis-related gene expression, monocyte activation and microparticle-release are related to increased lipoprotein-associated oxidative stress in familial hypercholesterolemia. PLoS ONE 2015, 10, e0121516. [Google Scholar] [CrossRef]
  325. Xia, J.; Qu, Y.; Yin, C.; Xu, N. Preoperative rosuvastatin protects patients with coronary artery disease undergoing noncardiac surgery. Cardiology 2015, 131, 30–37. [Google Scholar] [CrossRef]
  326. Mannacio, V.A.; Iorio, D.; DE Amicis, V.; Di Lello, F.; Musumeci, F. Effect of rosuvastatin pretreatment on myocardial damage after coronary surgery: A randomized trial. J. Thorac. Cardiovasc. Surg. 2008, 136, 1541–1548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  327. Tomoda, H.; Elgendy, I.Y.; Mahmoud, A.N.; Bavry, A.A.; Wang, S.; Peng, D.-Q.; Casadei, B.; Collins, R.; Zheng, Z. Perioperative Rosuvastatin in Cardiac Surgery. N. Engl. J. Med. 2016, 375, 1744. [Google Scholar] [CrossRef]
  328. Suades, R.; Padró, T.; Alonso, R.; Mata, P.; Badimon, L. Lipid-lowering therapy with statins reduces microparticle shedding from endothelium, platelets and inflammatory cells. Thromb. Haemost. 2013, 110, 366–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  329. Nomura, S.; Inami, N.; Shouzu, A.; Omoto, S.; Kimura, Y.; Takahashi, N.; Tanaka, A.; Urase, F.; Maeda, Y.; Ohtani, H.; et al. The effects of pitavastatin, eicosapentaenoic acid and combined therapy on platelet-derived microparticles and adiponectin in hyperlipidemic, diabetic patients. Platelets 2009, 20, 16–22. [Google Scholar] [CrossRef]
  330. Camargo, L.; França, C.N.; Izar, M.; Bianco, H.; Lins, L.; Barbosa, S.; Pinheiro, L.; Fonseca, F. Effects of simvastatin/ezetimibe on microparticles, endothelial progenitor cells and platelet aggregation in subjects with coronary heart disease under antiplatelet therapy. Braz. J. Med. Biol. Res. 2014, 47, 432–437. [Google Scholar] [CrossRef] [PubMed]
  331. Huang, B.; Cheng, Y.; Xie, Q.; Lin, G.; Wu, Y.; Feng, Y.; Gao, J.; Xu, D. Effect of 40 mg versus 10 mg of atorvastatin on oxidized low-density lipoprotein, high-sensitivity c-reactive protein, circulating endothelial-derived microparticles, and endothelial progenitor cells in patients with ischemic cardiomyopathy. Clin. Cardiol. 2012, 35, 125–130. [Google Scholar] [CrossRef]
  332. Verbree-Willemsen, L.; Zhang, Y.-N.; Gijsberts, C.M.; Schoneveld, A.H.; Wang, J.-W.; Lam, C.S.; Vernooij, F.; Bots, M.L.; Peelen, L.M.; Grobbee, D.E.; et al. LDL extracellular vesicle coagulation protein levels change after initiation of statin therapy. Findings from the METEOR trial. Int. J. Cardiol. 2018, 271, 247–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  333. Kulshreshtha, A.; Singh, S.; Ahmad, M.; Khanna, K.; Ahmad, T.; Agrawal, A.; Ghosh, B. Simvastatin mediates inhibition of exosome synthesis, localization and secretion via multicomponent interventions. Sci. Rep. 2019, 9, 16373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  334. Kocsis, G.F.; Pipis, J.; Fekete, V.; Kovács-Simon, A.; Odendaal, L.; Molnár, E.; Giricz, Z.; Janáky, T.; Van Rooyen, J.; Csont, T.; et al. Lovastatin interferes with the infarct size-limiting effect of ischemic preconditioning and postconditioning in rat hearts. Am. J. Physiol. Heart Circ. Physiol. 2008, 294, H2406–H2409. [Google Scholar] [CrossRef]
  335. Baietti, M.F.; Zhang, Z.; Mortier, E.; Melchior, A.; Degeest, G.; Geeraerts, A.; Ivarsson, Y.; Depoortere, F.; Coomans, C.; Vermeiren, E.; et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat. Cell Biol. 2012, 14, 677–685. [Google Scholar] [CrossRef]
  336. Zhang, Y.; Liu, D.; Chen, X.; Li, J.; Li, L.; Bian, Z.; Sun, F.; Lu, J.; Yin, Y.; Cai, X.; et al. Secreted monocytic mir-150 enhances targeted endothelial cell migration. Mol. Cell 2010, 39, 133–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  337. Tang, Y.; Wang, Y.; Park, K.-M.; Hu, Q.; Teoh, J.-P.; Broskova, Z.; Ranganathan, P.; Jayakumar, C.; Li, J.; Su, H.; et al. MicroRNA-150 protects the mouse heart from ischaemic injury by regulating cell death. Cardiovasc. Res. 2015, 106, 387–397. [Google Scholar] [CrossRef] [Green Version]
  338. Carrick, D.; Haig, C.; Maznyczka, A.M.; Carberry, J.; Mangion, K.; Ahmed, N.; Petrie, M.C.; Eteiba, H.; Lindsay, M.; Hood, S.; et al. Hypertension, microvascular pathology, and prognosis after an acute myocardial infarction. Hypertension 2018, 72, 720–730. [Google Scholar] [CrossRef]
  339. Galiñanes, M. Role of clinical pathologies in myocardial injury following ischaemia and reperfusion. Cardiovasc. Res. 2004, 61, 512–521. [Google Scholar] [CrossRef]
  340. Mølgaard, S.; Faricelli, B.; Salomonsson, M.; Engstrøm, T.; Treiman, M. Increased myocardial vulnerability to ischemia–reperfusion injury in the presence of left ventricular hypertrophy. J. Hypertens. 2016, 34, 513–523. [Google Scholar] [CrossRef]
  341. Kang, D.G.; Jeong, M.H.; Ahn, Y.; Chae, S.C.; Hur, S.H.; Hong, T.J.; Kim, Y.J.; Seong, I.W.; Chae, J.K.; Rhew, J.Y.; et al. Clinical effects of hypertension on the mortality of patients with acute myocardial infarction. J. Korean Med. Sci. 2009, 24, 800–806. [Google Scholar] [CrossRef]
  342. Reinstadler, S.J.; Stiermaier, T.; Eitel, C.; Saad, M.; Metzler, B.; de Waha, S.; Fuernau, G.; Desch, s.; Thiele, H.; Eitel, I. Antecedent hypertension and myocardial injury in patients with reperfused ST-elevation myocardial infarction. J. Cardiovasc. Magn. Reson. 2017, 18, 80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  343. Powers, S.K.; Lennon, S.L.; Quindry, J.; Mehta, J.L. Exercise and cardioprotection. Curr. Opin. Cardiol. 2002, 17, 495–502. [Google Scholar] [CrossRef] [PubMed]
  344. Powers, S.K.; Quindry, J.C.; Kavazis, A.N. Exercise-induced cardioprotection against myocardial ischemia–reperfusion injury. Free Radic. Biol. Med. 2008, 44, 193–201. [Google Scholar] [CrossRef]
  345. Lee, Y.; Min, K.; Talbert, E.E.; Kavazis, A.N.; Smuder, A.J.; Willis, W.T.; Powers, S.K. Exercise protects cardiac mitochondria against ischemia–reperfusion injury. Med. Sci. Sports Exerc. 2012, 44, 397–405. [Google Scholar] [CrossRef]
  346. Guo, Y.; Chen, J.; Qiu, H. Novel mechanisms of exercise-induced cardioprotective factors in myocardial infarction. Front. Physiol. 2020, 11, 199. [Google Scholar] [CrossRef] [PubMed]
  347. Michelsen, M.M.; Støttrup, N.B.; Schmidt, M.R.; Løfgren, B.; Jensen, R.V.; Tropak, M.; St-Michel, E.J.; Redington, A.N.; Bøtker, H.E. Exercise-induced cardioprotection is mediated by a bloodborne, transferable factor. Basic Res. Cardiol. 2012, 107, 260. [Google Scholar] [CrossRef] [PubMed]
  348. Bei, Y.; Xu, T.; Lu, D.; Yu, P.; Xu, J.; Che, L.; Das, A.; Tigges, J.; Toxavidis, V.; Ghiran, I.; et al. Exercise-induced circulating extracellular vesicles protect against cardiac ischemia–reperfusion injury. Basic Res. Cardiol. 2017, 112, 38. [Google Scholar] [CrossRef]
  349. Oliveira, G.; Porto, W.; Palu, C.; Pereira, L.M.; Petriz, B.; Almeida, J.A.; Viana, J.; Filho, N.N.A.; Franco, O.L.; Pereira, R. Effects of acute aerobic exercise on rats serum extracellular vesicles diameter, concentration and small rnas content. Front. Physiol. 2018, 9, 532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  350. Wiklander, O.P.B.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mäger, I.; Vader, P.; Lee, Y.; Sork, H.; Seow, Y.; et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J. Extracell. Vesicles 2015, 4, 26316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  351. Whitham, M.; Parker, B.L.; Friedrichsen, M.; Hingst, J.R.; Hjorth, M.; Hughes, W.; Egan, C.L.; Cron, L.; Watt, K.; Kuchel, R.P.; et al. Extracellular vesicles provide a means for tissue crosstalk during exercise. Cell Metab. 2018, 27, 237–251.e4. [Google Scholar] [CrossRef] [Green Version]
  352. Wilhelm, E.N.; Mourot, L.; Rakobowchuk, M. Exercise-derived microvesicles: A review of the literature. Sports Med. 2018, 48, 2025–2039. [Google Scholar] [CrossRef] [Green Version]
  353. Frühbeis, C.; Helmig, S.; Tug, S.; Simon, P.; Krämer-Albers, E.-M. Physical exercise induces rapid release of small extracellular vesicles into the circulation. J. Extracell. Vesicles 2015, 4, 28239. [Google Scholar] [CrossRef] [Green Version]
  354. Hou, Z.; Qin, X.; Hu, Y.; Zhang, X.; Li, G.; Wu, J.; Li, J.; Sha, J.; Chen, J.; Xia, J.; et al. Longterm exercise-derived exosomal mir-342-5p. Circ. Res. 2019, 124, 1386–1400. [Google Scholar] [CrossRef] [PubMed]
  355. Barber, J.L.; Zellars, K.N.; Barringhaus, K.G.; Bouchard, C.; Spinale, F.G.; Sarzynski, M.A. The effects of regular exercise on circulating cardiovascular-related micrornas. Sci. Rep. 2019, 9, 7527. [Google Scholar] [CrossRef] [PubMed]
  356. Mooren, F.C.; Viereck, J.; Krüger, K.; Thum, T. Circulating micrornas as potential biomarkers of aerobic exercise capacity. Am. J. Physiol. Heart Circ. Physiol. 2014, 306, H557–H563. [Google Scholar] [CrossRef]
  357. Domańska-Senderowska, D.; Laguette, M.-J.N.; Jegier, A.; Cieszczyk, P.; September, A.V.; Brzeziańska-Lasota, E. MicroRNA profile and adaptive response to exercise training: A review. Endoscopy 2019, 40, 227–235. [Google Scholar] [CrossRef] [PubMed]
  358. Terlecki-Zaniewicz, L.; Lämmermann, I.; Latreille, J.; Bobbili, M.R.; Pils, V.; Schosserer, M.; Weinmüllner, R.; Dellago, H.; Skalicky, S.; Pum, D.; et al. Small extracellular vesicles and their miRNA cargo are anti-apoptotic members of the senescence-associated secretory phenotype. Aging 2018, 10, 1103–1132. [Google Scholar] [CrossRef]
  359. Huang, X.; Yuan, T.; Tschannen, M.; Sun, Z.; Jacob, H.; Du, M.; Liang, M.; Dittmar, R.L.; Liu, Y.; Liang, M.; et al. Characterization of human plasma-derived exosomal RNAs by deep sequencing. BMC Genom. 2013, 14, 319. [Google Scholar] [CrossRef] [Green Version]
  360. Shi, J.; Bei, Y.; Kong, X.; Liu, X.; Lei, Z.; Xu, T.; Wang, H.; Xuan, Q.; Chen, P.; Xu, J.; et al. miR-17-3p contributes to exercise-induced cardiac growth and protects against myocardial ischemia-reperfusion injury. Theranostics 2017, 7, 664–676. [Google Scholar] [CrossRef]
  361. Peng, Y.; Zhao, J.-L.; Peng, Z.-Y.; Xu, W.-F.; Yu, G.-L. Exosomal miR-25-3p from mesenchymal stem cells alleviates myocardial infarction by targeting pro-apoptotic proteins and EZH2. Cell Death Dis. 2020, 11, 317. [Google Scholar] [CrossRef] [PubMed]
  362. Liu, J.; Jiang, M.; Deng, S.; Lu, J.; Huang, H.; Zhang, Y.; Gong, P.; Shen, X.; Ruan, H.; Jin, M.; et al. miR-93-5p-containing exosomes treatment attenuates acute myocardial infarction-induced myocardial damage. Mol. Ther. Nucleic Acids 2018, 11, 103–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  363. Gomes, C.P.; Oliveira, G.P., Jr.; Madrid, B.; Almeida, J.A.; Franco, O.L.; Pereira, R.W. Circulating miR-1, miR-133a, and miR-206 levels are increased after a half-marathon run. Biomarkers 2014, 19, 585–589. [Google Scholar] [CrossRef]
  364. Clauss, S.; Wakili, R.; Hildebrand, B.; Kaab, S.; Hoster, E.; Klier, I.; Martens, E.; Hanley, A.; Hanssen, H.; Halle, M.; et al. MicroRNAs as Biomarkers for acute atrial remodeling in marathon runners (the mirathon study—A sub-study of the Munich marathon study). PLoS ONE 2016, 11, e0148599. [Google Scholar] [CrossRef] [PubMed]
  365. Safdar, A.; Saleem, A.; Tarnopolsky, M.A. The potential of endurance exercise-derived exosomes to treat metabolic diseases. Nat. Rev. Endocrinol. 2016, 12, 504–517. [Google Scholar] [CrossRef]
  366. Eichner, N.Z.M.; Erdbrügger, U.; Malin, S.K. Extracellular vesicles: A novel target for exercise-mediated reductions in type 2 diabetes and cardiovascular disease risk. J. Diabetes Res. 2018, 2018, 7807245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  367. Safdar, A.; Tarnopolsky, M.A. Exosomes as mediators of the systemic adaptations to endurance exercise. Cold Spring Harb. Perspect. Med. 2018, 8, a029827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  368. Go, A.S.; Mozaffarian, D.; Roger, V.L.; Benjamin, E.J.; Berry, J.D.; Blaha, M.J.; Dai, S.; Ford, E.S.; Fox, C.S.; Franco, S.; et al. Heart disease and stroke statistics—2014 update: A report from the American Heart Association. Circulation 2014, 129, e28–e292. [Google Scholar] [CrossRef] [Green Version]
  369. Gustafson, C.M.; Shepherd, A.J.; Miller, V.M.; Jayachandran, M. Age- and sex-specific differences in blood-borne microvesicles from apparently healthy humans. Biol. Sex Differ. 2015, 6, 10. [Google Scholar] [CrossRef] [Green Version]
  370. Kobayashi, Y.; Eguchi, A.; Tempaku, M.; Honda, T.; Togashi, K.; Iwasa, M.; Hasegawa, H.; Takei, Y.; Sumida, Y.; Taguchi, O. Circulating extracellular vesicles are associated with lipid and insulin metabolism. Am. J. Physiol. Endocrinol. Metab. 2018, 315, E574–E582. [Google Scholar] [CrossRef]
  371. Noulsri, E.; Palasuwan, A. Effects of donor age, donor sex, blood-component processing, and storage on cell-derived microparticle concentrations in routine blood-component preparation. Transfus. Apher. Sci. 2018, 57, 587–592. [Google Scholar] [CrossRef]
  372. Ma, F.; Liu, H.; Shen, Y.; Zhang, Y.; Pan, S. Platelet-derived microvesicles are involved in cardio-protective effects of remote preconditioning. Int. J. Clin. Exp. Pathol. 2015, 8, 10832–10839. [Google Scholar] [PubMed]
  373. Tushuizen, M.E.; Diamant, M.; Sturk, A.; Nieuwland, R. Cell-derived microparticles in the pathogenesis of cardiovascular disease: Friend or foe? Arterioscler. Thromb. Vasc. Biol. 2011, 31, 4–9. [Google Scholar] [CrossRef] [Green Version]
  374. Bammert, T.D.; Hijmans, J.G.; Kavlich, P.J.; Lincenberg, G.M.; Reiakvam, W.R.; Fay, R.T.; Greiner, J.J.; Stauffer, B.L.; DeSouze, C.A. Influence of sex on the number of circulating endothelial microparticles and microRNA expression in middle-aged adults. Exp. Physiol. 2017, 102, 894–900. [Google Scholar] [CrossRef] [PubMed]
  375. El Khoudary, S.R.; Aggarwal, B.; Beckie, T.M.; Hodis, H.N.; Johnson, A.E.; Langer, R.D.; Limacher, M.C.; Manson, J.E.; Stefanick, M.L.; Allison, M.A.; et al. Menopause Transition and cardiovascular disease risk: Implications for timing of early prevention: A scientific statement from the american heart association. Circulation 2020, 142, e506–e532. [Google Scholar] [CrossRef]
  376. Rank, A.; Nieuwland, R.; Roesner, S.; Nikolajek, K.; Hiller, E.; Toth, B. Climacteric lowers plasma levels of platelet-derived microparticles: A pilot study in pre- versus postmenopausal women. Acta Haematol. 2012, 128, 53–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  377. Zhang, J.; Li, H.; Fan, B.; Xu, W.; Zhang, X. Extracellular vesicles in normal pregnancy and pregnancy-related diseases. J. Cell. Mol. Med. 2020, 24, 4377–4388. [Google Scholar] [CrossRef] [Green Version]
  378. Gill, M.; Motta-Mejia, C.; Kandzija, N.; Cooke, W.; Zhang, W.; Cerdeira, A.S.; Bastie, C.; Redman, C.; Vatish, M. Placental syncytiotrophoblast-derived extracellular vesicles carry active nep (neprilysin) and are increased in preeclampsia. Hypertension 2019, 73, 1112–1119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  379. Takov, K.; He, Z.; Johnston, H.; Timms, J.; Guillot, P.V.; Yellon, D.; Davidson, S.M. Small extracellular vesicles secreted from human amniotic fluid mesenchymal stromal cells possess cardioprotective and promigratory potential. Basic Res. Cardiol. 2020, 115, 26. [Google Scholar] [CrossRef]
  380. Li, J.; Umar, S.; Iorga, A.; Youn, J.-Y.; Wang, Y.; Regitz-Zagrosek, V.; Cai, H.; Eghbali, M. Cardiac vulnerability to ischemia/reperfusion injury drastically increases in late pregnancy. Basic Res. Cardiol. 2012, 107, 271. [Google Scholar] [CrossRef] [Green Version]
  381. Panagiotou, N.; Neytchev, O.; Selman, C.; Shiels, P.G. Extracellular vesicles, ageing, and therapeutic interventions. Cells 2018, 7, 110. [Google Scholar] [CrossRef] [Green Version]
  382. Chen, G.H.; Xu, C.-S.; Zhang, J.; Li, Q.; Cui, H.-H.; Li, X.-D.; Chang, L.; Tang, R.-J.; Xu, J.-Y.; Tian, X.-Q.; et al. Inhibition of miR-128-3p by tongxinluo protects human cardiomyocytes from ischemia/reperfusion injury via upregulation of p70s6k1/p-p70s6k1. Front. Pharmacol. 2017, 8, 775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  383. Xu, T.; Luo, Y.; Wang, J.; Zhang, N.; Gu, C.; Li, L.; Qian, D.; Cai, W.; Fan, J.; Yin, G. Exosomal miRNA-128-3p from mesenchymal stem cells of aged rats regulates osteogenesis and bone fracture healing by targeting Smad5. J. Nanobiotechnol. 2020, 18, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  384. Rossello, X.; Yellon, D.M. The RISK pathway and beyond. Basic Res. Cardiol. 2018, 113, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  385. Merhi-Soussi, F.; Kwak, B.; Magne, D.; Chadjichristos, C.; Berti, M.; Pelli, G.; James, R.W.; Mach, F.; Gabay, C. Interleukin-1 plays a major role in vascular inflammation and atherosclerosis in male apolipoprotein E-knockout mice. Cardiovasc. Res. 2005, 66, 583–593. [Google Scholar] [CrossRef] [Green Version]
  386. Gomes de Andrade, G.; Cechinel, L.R.; Bertoldi, K.; Galvão, F.; Worm, P.V.; Siqueira, I.R. The Aging Process Alters IL-1beta and CD63 levels differently in extracellular vesicles obtained from the plasma and cerebrospinal fluid. Neuroimmunomodulation 2018, 25, 18–22. [Google Scholar] [CrossRef]
  387. Zhu, J.; Huang, J.; Dai, D.; Wang, X.; Gao, J.; Han, W.; Zhang, R. Recombinant human interleukin-1 receptor antagonist treatment protects rats from myocardial ischemia–reperfusion injury. Biomed. Pharmacother. 2019, 111, 1–5. [Google Scholar] [CrossRef]
  388. Toldo, S.; Schatz, A.M.; Mezzaroma, E.; Chawla, R.; Stallard, T.W.; Stallard, W.C.; Jahangiri, A.; Van Tassell, B.W.; Abbate, A. Recombinant human interleukin-1 receptor antagonist provides cardioprotection during myocardial ischemia reperfusion in the mouse. Cardiovasc. Drugs Ther. 2012, 26, 273–276. [Google Scholar] [CrossRef]
  389. Grothusen, C.; Hagemann, A.; Attmann, T.; Braesen, J.; Broch, O.; Cremer, J.; Schoettler, J. Impact of an interleukin-1 receptor antagonist and erythropoietin on experimental myocardial ischemia/reperfusion injury. Sci. World J. 2012, 2012, 737585. [Google Scholar] [CrossRef] [Green Version]
  390. Silvain, J.; Kerneis, M.; Zeitouni, M.; Lattuca, B.; Mertens, E.; Procopi, N.; Suc, G.; Salloum, T.; Frisdal, E.; Le Goff, W.; et al. Interleukin-1Beta and risk of premature death and MACE in patients with myocardial infarction. J. Am. Coll. Cardiol. 2020, 41, ehaa946.1697. [Google Scholar] [CrossRef]
  391. Fafian-Labora, J.A.; Lesende-Rodriguez, I.; Fernández-Pernas, P.; Sangiao-Alvarellos, S.; Monserrat, L.; Arntz, O.J.; Van De Loo, F.A.J.; Mateos, J.; Arufe, M.C. Effect of age on pro-inflammatory miRNAs contained in mesenchymal stem cell-derived extracellular vesicles. Sci. Rep. 2017, 7, 43923. [Google Scholar] [CrossRef] [Green Version]
  392. Olivieri, F.; Rippo, M.R.; Prattichizzo, F.; Babini, L.; Graciotti, L.; Recchioni, R.; Procopio, A.D. Toll like receptor signaling in “inflammaging”: MicroRNA as new players. Immun. Ageing 2013, 10, 11. [Google Scholar] [CrossRef] [Green Version]
  393. Lopez-Otin, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  394. González-Montero, J.; Brito, R.; Gajardo, A.I.; Rodrigo, R. Myocardial reperfusion injury and oxidative stress: Therapeutic opportunities. World J. Cardiol. 2018, 10, 74–86. [Google Scholar] [CrossRef] [PubMed]
  395. Yoshida, M.; Satoh, A.; Lin, J.B.; Mills, K.F.; Sasaki, Y.; Rensing, N.; Wong, M.; Apte, R.S.; Imai, S.-I. Extracellular vesicle-contained enampt delays aging and extends lifespan in mice. Cell Metab. 2019, 30, 329–342.e5. [Google Scholar] [CrossRef] [PubMed]
  396. Chouchani, E.T.; Pell, V.R.; Gaude, E.; Sundier, S.Y.; Robb, E.L.; Logan, A.; Nadtochiy, S.M.; Ord, E.N.J.; Smith, A.C.; Eyassu, F.; et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature 2014, 515, 431–435. [Google Scholar] [CrossRef] [Green Version]
  397. Yoshino, J.; Mills, K.F.; Yoon, M.J.; Imai, S.-I. Nicotinamide Mononucleotide, a Key NAD+ Intermediate, Treats the Pathophysiology of Diet- and Age-Induced Diabetes in Mice. Cell Metab. 2011, 14, 528–536. [Google Scholar] [CrossRef] [Green Version]
  398. Imai, S.-I.; Guarente, L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 2014, 24, 464–471. [Google Scholar] [CrossRef]
  399. Imai, S.-I. Nicotinamide phosphoribosyltransferase (nampt): A link between nad biology, metabolism, and diseases. Curr. Pharm. Des. 2009, 15, 20–28. [Google Scholar] [CrossRef] [Green Version]
  400. Eitan, E.; Green, J.; Bodogai, M.; Mode, N.A.; Bæk, R.; Jørgensen, M.M.; Freeman, D.W.; Witwer, K.; Zonderman, A.B.; Biragyn, A.; et al. Age-related changes in plasma extracellular vesicle characteristics and internalization by leukocytes. Sci. Rep. 2017, 7, 1342. [Google Scholar] [CrossRef] [Green Version]
  401. Deschamps, A.; Murphy, E.; Sun, J. Estrogen receptor activation and cardioprotection in ischemia reperfusion injury. Trends Cardiovasc. Med. 2010, 20, 73–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  402. Lazo, S.; Hooten, N.N.; Green, J.; Eitan, E.; Mode, N.A.; Liu, Q.; Zonderman, A.B.; Ezike, N.; Mattson, M.P.; Ghosh, P.; et al. Mitochondrial DNA in extracellular vesicles declines with age. Aging Cell 2021, 20, e13283. [Google Scholar] [CrossRef] [PubMed]
  403. Picca, A.; Guerra, F.; Calvani, R.; Bucci, C.; Monaco, M.R.L.; Bentivoglio, A.R.; Coelho-Júnior, H.J.; Landi, F.; Bernabei, R.; Marzetti, E. Mitochondrial dysfunction and aging: Insights from the analysis of extracellular vesicles. Int. J. Mol. Sci. 2019, 20, 805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  404. Weilner, S.; Keider, V.; Winter, M.; Harreither, E.; Salzer, B.; Weiss, F.; Schraml, E.; Messner, P.; Pietschmann, P.; Hildner, F.; et al. Vesicular Galectin-3 levels decrease with donor age and contribute to the reduced osteo-inductive potential of human plasma derived extracellular vesicles. Aging 2016, 8, 16–33. [Google Scholar] [CrossRef] [Green Version]
  405. Al-Salam, S.; Hashmi, S. Myocardial ischemia reperfusion injury: Apoptotic, inflammatory and oxidative stress role of galectin-3. Cell. Physiol. Biochem. 2018, 50, 1123–1139. [Google Scholar] [CrossRef]
  406. Ibarrola, J.F.; Matilla, L.; Martínez-Martínez, E.; Gueret, A.; Fernández-Celis, A.; Henry, J.-P.; Nicol, L.; Jaisser, F.; Mulder, P.; Ouvrard-Pascaud, A.; et al. Myocardial injury after ischemia/reperfusion is attenuated by pharmacological galectin-3 inhibition. Sci. Rep. 2019, 9, 607. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Molecular mechanisms involved in ischemia-reperfusion injury. All cell death types are involved in IRI, including regulated such as necroptosis (A), pyroptosis (B), apoptosis (C), and unregulated such as necrosis (D). (A) The activation of TNF-α (Tumor Necrosis Factor-alpha) receptors and TLRs (Toll-like Receptors) by DAMPs (Damage-associated Molecular Patterns), which are released during mitochondria and cell rapture, promote necroptosis. Stimulation of Tumor Necrosis Factor receptor recruits RIPKs (receptor-interacting serine/threonine-protein kinases) by proteins associated with the receptor. The RIPKs form the necrosome which phosphorylate the MLKL (mixed-lineage kinase domain-like proteins). Phosphorylated MLKL activates the pores which permeabilize plasma [15]. (B) DAMPs stimulate inflammasome formation, which activates caspases, leading to the formation of gasdermin-dependent pores in the membrane. (C) The lack of oxygen induces failure of ion pumps, acidosis, and Ca2+ overload [16]. In anaerobic glycolysis, the production of H+ is increased. The 2Na+/Ca2+ and the Na+/H+ ion exchangers remove sodium excess and increase calcium and hydrogen levels [17]. The elevated Ca2+ load induces reactive oxygen species (ROS) production and activates phospholipases and proteolytic enzymes which stimulate the release of cytochrome c from the mitochondria and apoptosis in consequence. In addition, elevated Ca2+ and inorganic phosphate levels stimulate the opening of the mitochondrial permeability transition pore (mPTP), which in turn causes mitochondrial matrix swelling and outer membrane rupture [18,19]. (D) Elevated ion levels and swelling of the cell promote rupture of the cell membrane and necrosis. These pathways and many others not mentioned due to this article’s limitations are responsible for IRI. Heusch recently published a more extensive description of the IRI mechanism [12]. Reperfusion injury can be regulated at any stage by many molecules, such as miRNA (micro ribonucleic acid/miR) and proteins. Created with BioRender.com.
Figure 1. Molecular mechanisms involved in ischemia-reperfusion injury. All cell death types are involved in IRI, including regulated such as necroptosis (A), pyroptosis (B), apoptosis (C), and unregulated such as necrosis (D). (A) The activation of TNF-α (Tumor Necrosis Factor-alpha) receptors and TLRs (Toll-like Receptors) by DAMPs (Damage-associated Molecular Patterns), which are released during mitochondria and cell rapture, promote necroptosis. Stimulation of Tumor Necrosis Factor receptor recruits RIPKs (receptor-interacting serine/threonine-protein kinases) by proteins associated with the receptor. The RIPKs form the necrosome which phosphorylate the MLKL (mixed-lineage kinase domain-like proteins). Phosphorylated MLKL activates the pores which permeabilize plasma [15]. (B) DAMPs stimulate inflammasome formation, which activates caspases, leading to the formation of gasdermin-dependent pores in the membrane. (C) The lack of oxygen induces failure of ion pumps, acidosis, and Ca2+ overload [16]. In anaerobic glycolysis, the production of H+ is increased. The 2Na+/Ca2+ and the Na+/H+ ion exchangers remove sodium excess and increase calcium and hydrogen levels [17]. The elevated Ca2+ load induces reactive oxygen species (ROS) production and activates phospholipases and proteolytic enzymes which stimulate the release of cytochrome c from the mitochondria and apoptosis in consequence. In addition, elevated Ca2+ and inorganic phosphate levels stimulate the opening of the mitochondrial permeability transition pore (mPTP), which in turn causes mitochondrial matrix swelling and outer membrane rupture [18,19]. (D) Elevated ion levels and swelling of the cell promote rupture of the cell membrane and necrosis. These pathways and many others not mentioned due to this article’s limitations are responsible for IRI. Heusch recently published a more extensive description of the IRI mechanism [12]. Reperfusion injury can be regulated at any stage by many molecules, such as miRNA (micro ribonucleic acid/miR) and proteins. Created with BioRender.com.
Cells 10 03331 g001
Figure 2. (A) Biogenesis of extracellular vesicles (EVs) and their interactions with recipient cells (B). (C,D) The main components of EV membrane and EV cargo; CD—Clusters of Differentiation, MHC—Major Histocompatibility Complex. Created with BioRender.
Figure 2. (A) Biogenesis of extracellular vesicles (EVs) and their interactions with recipient cells (B). (C,D) The main components of EV membrane and EV cargo; CD—Clusters of Differentiation, MHC—Major Histocompatibility Complex. Created with BioRender.
Cells 10 03331 g002
Figure 3. Impact of the main cardiovascular risk factors and physical activity on EVs’ expression of miRNAs and its potential influence on myocardial susceptibility to IRI. (upward arrow—increased level, downward arrow—decreased level, green—attenuating IRI, red—aggravating IRI).
Figure 3. Impact of the main cardiovascular risk factors and physical activity on EVs’ expression of miRNAs and its potential influence on myocardial susceptibility to IRI. (upward arrow—increased level, downward arrow—decreased level, green—attenuating IRI, red—aggravating IRI).
Cells 10 03331 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Majka, M.; Kleibert, M.; Wojciechowska, M. Impact of the Main Cardiovascular Risk Factors on Plasma Extracellular Vesicles and Their Influence on the Heart’s Vulnerability to Ischemia-Reperfusion Injury. Cells 2021, 10, 3331. https://doi.org/10.3390/cells10123331

AMA Style

Majka M, Kleibert M, Wojciechowska M. Impact of the Main Cardiovascular Risk Factors on Plasma Extracellular Vesicles and Their Influence on the Heart’s Vulnerability to Ischemia-Reperfusion Injury. Cells. 2021; 10(12):3331. https://doi.org/10.3390/cells10123331

Chicago/Turabian Style

Majka, Miłosz, Marcin Kleibert, and Małgorzata Wojciechowska. 2021. "Impact of the Main Cardiovascular Risk Factors on Plasma Extracellular Vesicles and Their Influence on the Heart’s Vulnerability to Ischemia-Reperfusion Injury" Cells 10, no. 12: 3331. https://doi.org/10.3390/cells10123331

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop