Next Article in Journal
IFN-β Overexpressing Adipose-Derived Mesenchymal Stem Cells Mitigate Alcohol-Induced Liver Damage and Gut Permeability
Previous Article in Journal
Correlations of Imaging and Therapy in Breast Cancer Based on Molecular Patterns: An Important Issue in the Diagnosis of Breast Cancer
Previous Article in Special Issue
Molecular Basis of Cardiomyopathies in Type 2 Diabetes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Acute Hyperglycemia-Induced Injury in Myocardial Infarction

by
Martino Pepe
1,*,†,
Francesco Addabbo
2,†,
Annagrazia Cecere
3,
Rocco Tritto
1,
Gianluigi Napoli
4,
Palma Luisa Nestola
5,
Plinio Cirillo
6,
Giuseppe Biondi-Zoccai
7,8,
Salvatore Giordano
9 and
Marco Matteo Ciccone
1
1
Division of Cardiology, Department of Interdisciplinary Medicine (D.I.M.), University of Bari “Aldo Moro”, 70100 Bari, Italy
2
ASL Taranto, Local Health Authority of Taranto, Statistics and Epidemiology Unit, 74100 Taranto, Italy
3
Division of Cardiology, Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, 35128 Padua, Italy
4
Division of Cardiology, Villa Verde Clinic, 74121 Taranto, Italy
5
Division of Cardiology, Mater Dei Hospital, 70125 Bari, Italy
6
Department of Advanced Biomedical Sciences, Federico II University of Naples, 80131 Naples, Italy
7
Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
8
Maria Cecilia Hospital, GVM Care & Research, 48032 Cotignola, Italy
9
Division of Cardiology, Department of Medical and Surgical Sciences, “Magna Graecia” University, 88100 Catanzaro, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(15), 8504; https://doi.org/10.3390/ijms25158504
Submission received: 15 May 2024 / Revised: 29 July 2024 / Accepted: 1 August 2024 / Published: 4 August 2024
(This article belongs to the Special Issue Advances in Molecular Research of Diabetic Cardiomyopathy)

Abstract

:
Acute hyperglycemia is a transient increase in plasma glucose level (PGL) frequently observed in patients with ST-elevation myocardial infarction (STEMI). The aim of this review is to clarify the molecular mechanisms whereby acute hyperglycemia impacts coronary flow and myocardial perfusion in patients with acute myocardial infarction (AMI) and to discuss the consequent clinical and prognostic implications. We conducted a comprehensive literature review on the molecular causes of myocardial damage driven by acute hyperglycemia in the context of AMI. The negative impact of high PGL on admission recognizes a multifactorial etiology involving endothelial function, oxidative stress, production of leukocyte adhesion molecules, platelet aggregation, and activation of the coagulation cascade. The current evidence suggests that all these pathophysiological mechanisms compromise myocardial perfusion as a whole and not only in the culprit coronary artery. Acute hyperglycemia on admission, regardless of whether or not in the context of a diabetes mellitus history, could be, thus, identified as a predictor of worse myocardial reperfusion and poorer prognosis in patients with AMI. In order to reduce hyperglycemia-related complications, it seems rational to pursue in these patients an adequate and quick control of PGL, despite the best pharmacological treatment for acute hyperglycemia still remaining a matter of debate.

1. Introduction

Acute hyperglycemia is defined as a transient increase in plasma glucose levels (PGL) and is a common finding in both diabetic and non-diabetic patients admitted for acute myocardial infarction (AMI) [1,2]. As a matter of fact, although hyperglycemia has a higher prevalence in diabetics [3], only 20–25% of hyperglycemic AMI patients have a known history of diabetes [4,5].
Despite acute hyperglycemia being a frequent condition, no uniform and universally accepted cut-off values are available in the literature [6]. Therefore, its prevalence widely ranges from 20 to 50% of AMI patients, depending on the definition. In some cohorts of non-diabetic patients, an even wider variability has been reported with a prevalence of hyperglycemia ranging between 3% and 71% [7], even though about 31% of non-diabetic patients presenting with high PGL reveal afterward an undiagnosed diabetes [8]. Applying a cut-off value of >110 mg/dL, Kosiborod et al. found in a large cohort of about 140,000 elderly patients hospitalized for AMI that 50% suffered from acute hyperglycemia [4] (Table 1). Conversely, the American Heart Association Scientific Statement on Hyperglycemia and Acute Coronary Syndrome suggested an upper limit for admission “random glucose” level of 140 mg/dL since higher values were associated with higher 30-day and 1-year mortality [1,4]. The hypothesis that high PGL in the setting of AMI predicts short and long-term mortality, regardless of diabetes mellitus (DM) history, has been supported by several pieces of evidence over the last two decades [9,10,11,12,13,14,15,16]. Furthermore, hyperglycemia also appears to be associated with well-known negative prognostic predictors, such as anterior wall infarction, multivessel coronary artery disease (CAD), atrial fibrillation, more extensive myocardial necrosis, and worse left ventricular systolic function [17,18,19,20]. We aimed to provide a state-of-the-art literature review of the pathophysiological mechanisms through which hyperglycemia impacts patients’ prognosis, firstly impairing coronary artery flow.

2. Current Evidence: From the Bench to the Bedside

2.1. Pathophysiological Mechanisms Triggered by Hyperglycemic Status in the Setting of Acute Myocardial Infarction

Multiple pathophysiological mechanisms have been proposed to explain the adverse prognostic impact of acute hyperglycemia in AMI patients: platelet hyperactivity; upregulation of the coagulation cascade; induction of a pro-inflammatory state; and, above all, endothelial dysfunction [21].

2.2. Endothelial Dysfunction

Vascular endothelium cannot be considered a mere physical barrier between the circulating blood components and the underlying tissues; it plays a crucial regulatory role in the maintenance of vascular homeostasis [22,23]. Endothelial cells secrete mediators involved in the regulation of the vascular tone, platelet adhesion, leukocyte diapedesis, and coagulation/fibrinolysis balance. Consequently, when present, endothelial dysfunction causes the dysregulation of multiple critical pathways [24,25,26,27,28].
Over the last decades, several studies pointed out that prolonged hyperglycemia and acute fluctuations of PGL are responsible for impaired endothelial function in both macro- and microvascular circulation [29,30,31,32]. Back in 1998, Williams et al. proved that endothelium-dependent vasodilation was transiently decreased in healthy subjects facing hyperglycemic conditions [33]. Moreover, fluctuating plasma glucose levels, compared to persistent hyperglycemia, have been hypothesized to be more harmful in terms of microvascular and macrovascular complications as well as mortality [34]. Three metabolic pathways have been recently involved as the link between hyperglycemia and endothelial dysfunction: oxidative stress; dysregulation of the endothelial nitric oxide synthase (eNOS) activity; and overproduction of leukocyte adhesion molecules.

2.2.1. Oxidative Stress

Increased oxidative stress can be considered the primary cause of endothelial dysfunction and is triggered by both PGL variability and prolonged high PGL [35,36,37]. Multiple mechanisms, mostly mutually interconnected, are accountable for the formation of reactive oxygen species (ROS): (1) enhanced activity of the mitochondrial respiratory chain (MRC); (2) activation of protein kinase C (PKC) isoforms; (3) increased polyol pathway flux associated with a reduction in endogenous antioxidant defenses; and (4) increased formation of advanced glycation end-products (AGEs).
  • The MRC is a protein complex within the internal mitochondrial membrane, which is involved in the redox reactions aimed to produce adenosine triphosphate (ATP) by using oxygen as the final electrons’ acceptor. Some studies identify ROS overproduction by MRC as the principal cause of hyperglycemia-induced tissue damage through the increase in glycolysis, tricarboxylic acid (TCA) cycle activity, ATP/ADP ratio, and the hyperpolarization of the mitochondrial membrane [38,39,40,41]. When the overproduction of electron donors by the TCA cycle occurs, the increased electrochemical potential difference (generated by the proton gradient across the inner mitochondrial membrane) increases the production of superoxide anions by endothelial cells [42]. Moreover, hyperglycemia seems also responsible for the impairment of the mitochondrial ATP-sensitive potassium channels (mKATP), which play a role in the protective preconditioning phenomenon [43,44];
  • Several studies have also pointed out the PKC activation in vascular cells in response to high PGL [45,46,47]. PKC is triggered by intracellular signals, such as diacylglycerol (DAG) or calcium ions (Ca2+), whose concentration in endothelial cells increases during hyperglycemic states. PKC activation, in turn, via numerous different pathways (as described in Figure 1) contributes to the impairment of myocardial perfusion [42,48];
  • In addition, a very suggestive discovery is that hyperglycemia stimulates the production of ROS in the heart through the activation of NADPH oxidase 2 (NOX2) via the sodiummyoinositol cotransporter-1 (SMIT1); the latter is an isoform of the sodium/glucose cotransporters (SGLT) and might explain some of the beneficial effects of the glifozines (SGLT-2 inhibitors) [49,50,51].
Moreover, hyperglycemia enhances the polyol pathway activity, resulting in the accumulation of fructose and sorbitol, which causes an imbalance in the intracellular redox state through an altered NADP+/NADPH ratio. The subsequent decrease in endogenous antioxidant defenses damages vascular homeostasis and intensifies the propensity to oxidative stress [42,46]. The increase in PGL also promotes the production of AGEs through non-enzymatic protein glycosylation and cross-linking reactions. These glycated proteins have been suggested to bind to specific receptors for AGEs (RAGE), which, in turn, activate NADP oxidase and eventually favor intracellular oxidative stress [52]. In detail, RAGEs, through a number of signaling cascades via phosphatidylinositol-3 kinase (PI3K), MAPK (ERK1 and 2), and Ki-Ras pathways, activate the nuclear factor-κB (NF-κB), favoring the production of ROS and the consequent cellular damage and mitochondrial dysfunction. NF-κB sub-unit p65 translocates into the nucleus to transcribe a number of pro-inflammatory cytokines and chemokines, among which are tumor necrosis factor α (TNF α), interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1), which also promote inflammation and stimulate immune cells [53].

2.2.2. Nitric Oxide Metabolism

Nitric oxide (NO) is a highly reactive gaseous compound with a very short half-life [54] whose production is catalyzed by the endothelial NO-synthase (eNOS) through the conversion of L-arginine to L-citrulline [55]. NO plays a crucial role in vascular function, being both a potent vasodilator, particularly effective in muscular arteries, and an inhibitor of platelets’ adhesion and aggregation, of leukocytes’ adhesion and migration, and of smooth muscle cell (SMC) proliferation. Under physiological conditions, shear stress is the principal eNOS trigger; nevertheless, the enzyme may also be activated by signaling molecules such as bradykinin, adenosine, vascular endothelial growth factor (in response to hypoxia), adiponectin, insulin, and serotonin [39,56,57]. Acute exposure of the vascular endothelium to elevated PGL results in the dysregulation of eNOS activity and reduced release of NO [30,33]. Several molecular mechanisms have been proposed: (1) the O-linked glycosylation of the eNOS active site [58,59]; (2) the inhibition of the insulin-stimulated expression of eNOS through the activation of the PKC pathway [60]; and (3) the AGEs–RAGEs interaction [61] (Figure 2(1)). Oxidative stress and NO metabolism appear tightly interconnected: the first may impair the enzymatic activity of dimethylarginine-dimethylaminohydrolase (DDAH) and increase asymmetric dimethylarginine (ADMA) levels, which, due to a structural similarity to L-arginine, acts as a competitive antagonist that binds to the catalytic site of eNOS resulting in inhibition of NO production. In particular, a novel model of “subclinical” NOS pharmacological inhibition with an ADMA analog demonstrated an enhanced generation of superoxide anion, which can possibly target the enzymes of the Krebs cycle, causing, in turn, the reduction in the mitochondrial mass and a switch to glycolytic metabolism in the face of normoxia, a first step toward endothelial dysfunction [35].
Seldom, eNOS can also generate superoxide instead of NO; this process, called eNOS uncoupling, is strongly dependent on the availability of eNOS substrate (arginine) and cofactor (tetrahydrobiopterin (BH4)) [62,63]. Hyperglycemia takes part in this vicious circle by determining an excessive production of intracellular ROS, leading to an increased formation of peroxynitrite, which, in turn, may react with the BH4 [64]. In the case of low BH4 concentration, eNOS becomes uncoupled and transfers electrons to molecular oxygen instead of L-arginine, thus producing superoxide rather than NO [35,62,65]. Heitzer and colleagues demonstrated that a diet supplement of BH4 in diabetic patients was able to improve endothelium-dependent vasodilation, thus elegantly proving that uncoupled eNOS plays a role in diabetic endothelial dysfunction [66].

2.3. Impaired Primary Hemostasis and Pro-Thrombotic Status

Elevated PGL drives adverse effects on ischemic myocardium, also favoring a systemic pro-thrombotic state. Different mechanisms have been identified [67], among which are the shortening of the fibrinogen half-life and the increased production of fibrinopeptide A, prothrombin fragments, factor VII, and other coagulation zymogens [1,68,69]. Furthermore, in acute hyperglycemic mouse models, lower tissue plasminogen activator (tPA) and higher plasminogen activator inhibitor (PAI) levels have been clearly demonstrated [70] (Figure 2(2)). Additionally, in non-diabetic subjects, a correlation between AGE formations during hyperglycemia and both PAI-1 and fibrinogen levels supports the interplay between different biochemical pathways triggered by the hyperglycemic status [71]. Besides the positive modulation of the coagulative cascade, hyperglycemia also impairs the fibrinolytic process by inducing structural changes in the fibrin molecules caused by the glycation reactions, with the consequent formation of denser clots, more resistant to fibrinolysis [67].
As concerns primary hemostasis, hyperglycemia modulates platelets’ reactivity, adhesion, and activation through several mechanisms (partly already mentioned) such as the upregulation of surface proteins (P-selectin and GP IIb/IIIa), the decreased membrane fluidity mediated by the glycation of membrane structures, the platelets swelling (an osmotic consequence of the abnormal activation of the polyol pathway), the upregulation of PKC intracellular signaling, and the overproduction of oxidant species [1,72,73] (Figure 2(3)). Moreover, the elevated concentration of cytosolic calcium, observed in platelets exposed to high glucose levels, may also play a role in all the phases of platelet activation, from shape change to granule secretion and thromboxane production [73].
Finally, hyperglycemia also seems to accelerate platelet turnover, as demonstrated by the presence of a higher number of immature reticulated platelets, which are larger, more sensitive, and less responsive to antiplatelet drugs [72,74]. In platelets, because of the absence of a nucleus, the normal lifespan of 7–10 days is largely determined by the mitochondria, which regulate the energy metabolism but also platelet activation and apoptosis [75].

2.4. Pro-Inflammatory Status

Several in vitro and in vivo studies have demonstrated a positive association between PGL and markers of vascular inflammation. Most evidence refers to elevated concentrations of C-reactive protein, interleukin-6, and tumor necrosis factor-alfa. Interestingly, the latter has been shown to induce cardiomyocytes’ apoptosis and correlate with larger infarct size in ischemic animal models [76,77,78]. Clinical evidence supporting these findings has been provided in a study on non-diabetic patients undergoing coronary artery bypass grafting (CABG): a significant reduction in inflammatory markers has been reported in the group undergoing a tight preoperative glycemic control with glucose–insulin–potassium (GIK) solution [79]. Moreover, during hyperglycemic states, an overproduction of leukocyte adhesion molecules [28], such as intercellular adhesion molecule-1 (ICAM-1), and chemokines, such as P-selectin, has also been described. Specifically, P-selectin is a key element in the rolling and diapedesis process, the first step of leukocyte activation. Once activated, the leukocytes, through the release of oxygen-derived free radicals, proteolytic enzymes, and other cytokines, induce a pro-inflammatory state, which, in turn, is capable of sustaining a vicious cycle that leads to further leukocyte activation and endothelial dysfunction [27,80] (Figure 2(4)). The role of leucocytes in the setting of hyperglycemia during acute myocardial ischemia has been confirmed in rats: Hokama et al. demonstrated that early after coronary reperfusion, higher levels of leukocytes become trapped in myocardial capillaries and venules of animals with diabetes than in controls [81].

2.5. Autophagy

Another very promising area of pathophysiologic research is autophagy, which is the ubiquitous cellular process of recycling damaged components [82]. In fact, interventions that leverage autophagy could counteract the detrimental effects of hyperglycemia on the cardiovascular system [83]. While data on this promising field of investigation are still preliminary, it is plausible that, for instance, metformin, SGLT-2 inhibitors, such as dapagliflozin and empagliflozin, and other agents with potent cardiometabolic effects may improve intracellular processes and protect from acute hyperglycemic stress, with effects that may be, at least in part, independent from PGL modulation [20,84,85]. This field of research seems very promising, as demonstrated by the plethora of ongoing clinical trials on the topic (https://clinicaltrials.gov/search?cond=Cardiovascular%20Diseases&intr=SGLT2%20inhibitors%2F(Dapagliflozin%20and%20Empagliflozin); accessed on 10 July 2024).

2.6. The Close Relationship between Acute Hyperglycemia, Coronary Flow and Myocardial Perfusion

Many epidemiological studies have found a high prevalence of acute hyperglycemia at presentation in patients with ST-segment elevation myocardial infarction (STEMI), regardless of DM history [86,87]. High PGL on admission is frequently associated with impaired coronary flow, which can be angiographically detected as “slow-flow” at presentation (TIMI flow grade 0–1) and/or “no-reflow”. The latter is defined as inadequate myocardial reperfusion after primary percutaneous coronary intervention (pPCI) despite effective treatment of the culprit lesion in the epicardial coronary artery with no angiographic evidence of epicardial obstruction, flow-limiting dissection, or vasospasm [12,88]. The impaired coronary flow has also been measured using the corrected TIMI frame count (cTFC): an easy, quantitative, and reproducible parameter reflecting both the epicardial flow and the microvascular function was used to more accurately assess the effectiveness of myocardial reperfusion after AMI [89]. Several pieces of evidence reported a higher cTFC in patients with acute hyperglycemia as proof of its negative impact on myocardial reperfusion and microvascular dysfunction [90] (Figure 3). This could be partially explained by the recruitment of activated leukocytes in coronary vessels, the subsequent pro-inflammatory status, and the impairment of myocardium oxygenation due to free radical overproduction, as previously described [81,91,92,93]. Moreover, the strict connection between hyperglycemia and no-reflow may also be due to additional mechanisms: first of all, a reduction in the collateral flow to the infarcted area through a NO-mediated mechanism, resulting in greater myocardial damage before reperfusion [94]; secondly, the hyperglycemia-induced platelet-dependent thrombosis [21], especially at capillaries level [95]; lastly, the detrimental impact of acute hyperglycemia on the cardioprotective mechanism of ischemic preconditioning (IPC), a powerful endogenous reaction against myocardial ischemia and reperfusion injury [96]. DM and acute hyperglycemia have been shown to counteract the positive effects of both ischemic and pharmacological preconditioning in animals and humans by inhibiting Akt phosphorylation. Noteworthy, the normalization of PGL determined by insulin administration has been conversely proved to be able to restore the cardioprotective effect of IPC [97].
Intriguingly, it was shown that both short- and long-term hyperglycemia increased the permeability and the barrier function of the endothelial glycocalyx and also decreased the functional capillary density and deformability of the red blood cells (RBCs). Glycocalyx also seems to have a putative role in myocardial tissue edema; thus, it could represent a potential early target of hyperglycemia [98].
Furthermore, in the course of AMI, the microvascular dysfunction in hyperglycemic patients is not limited to the culprit coronary artery since a 45% reduction in non-culprit coronary flow has also been demonstrated [99]. These data suggest a widespread dysfunction involving the whole coronary circulation rather than the thrombotic culprit lesion single-handedly. This hypothesis has been recently sustained in a selected cohort of STEMI patients divided into two groups based on PGL (< or >140 mg/dL): hyperglycemic patients showed higher cTFC values in both the culprit and non-culprit vessels regardless of preexisting diabetes. Moreover, a linear relationship between cTFC values and PGL at admission was also found [100]. The above-mentioned independence of the hyperglycemia-related microvascular damage from the possible concomitant presence of DM has been repeatedly demonstrated in the recent literature [101]. Planer et al. evaluated the prognostic significance of high PGL, regardless of DM diagnosis, in patients enrolled in the HORIZONS-AMI trial, a large-scale prospective study on STEMI patients. Patients with PGL > 200 mg/dL on admission showed a higher long-term mortality risk than the remaining population but comparable to subjects with a known history of DM [102]. Similar results had been previously reported by Stranders et al. [103]. Both studies concluded that high PGL at admission impacts short and long-term mortality independently of DM diagnosis, so hyperglycemia could be considered a supplementary risk factor. Conversely, in a recent analysis on 2958 consecutive STEMI patients, after stratification for DM history, hyperglycemia resulted as an independent predictor of mortality only in patients without DM [104]. The authors suggested, as a possible but speculative explanation of this finding, that chronic exposure to elevated glucose levels in diabetic patients could set the stage for a sort of protective “glucidic preconditioning” mechanism, which acts as a modulator of the detrimental effects of acute hyperglycemia during STEMI.

2.7. Glucose Control Strategy in Patients with AMI

The recognized correlation between acute hyperglycemia on admission and impaired vascular homeostasis suggests that insulin, due to its anti-inflammatory, anti-apoptotic, and pro-fibrinolytic properties, may be useful in the AMI setting to both achieve glycemic control and improve coronary flow [105,106]. Numerous preclinical studies supported the protective effect of early administration of insulin: possible mechanisms include the activation of eNOS and the eventual increase in NO synthesis [107,108], stimulation of angiogenesis, and inhibition of myocardial cell apoptosis [109]. Clinical data on insulin treatment are nevertheless non-univocal; it is known that chronic insulin treatment, within diabetic cohorts, is associated with worse prognosis in patients undergoing PCI because of either possible drug-related adverse effects (e.g., neointimal tissue proliferation, augmented plaque vulnerability, platelet dysfunction, resistance to antiplatelet agents, and vascular smooth muscle cell proliferation) or because insulin assumption may merely represent a marker of more advanced diabetic disease [39,110,111,112]. Evidence regarding the effectiveness of insulin use in the acute setting of AMI is also controversial: a sub-analysis of the DIGAMI study had shown that an intensive insulin regimen in diabetic patients with AMI [113] reduced long-term mortality compared to routine anti-diabetic therapy. Nevertheless, the attempt to confirm this finding failed with the following DIGAMI-2 study [114], which randomly assigned 1253 Type 2 DM (T2DM) patients with suspected acute coronary syndrome (ACS) to three different glycemic control strategies and showed no clinical benefit from an intensive insulin therapy. In the absence of conclusive data, the PGL management of these patients is still based on clinical common sense: before the publication of the 2023 European guidelines for the management of ACS [115] and Cardiovascular Disease in patients with Diabetes [116], the previous recommendation was to start a glucose-lowering therapy for glucose values above 180 mg/dL (class of recommendation IIa, level of evidence B) [117,118]. However, because of the lack of definite data, any cut-off value has been removed from the current guidelines and replaced with the broader definition of “persistent hyperglycemia” as an indication of pharmacological glycemic control [115,116]. According to this document, glycemic status should be assessed at initial evaluation in all patients with ACS and frequently monitored in patients with known diabetes or glucose levels exceeding 200 mg/dL.

3. Conclusions

Hyperglycemia on admission, regardless of DM history, has been identified as a strong predictor of impaired myocardial reperfusion and, thus, of worse outcomes in patients with AMI. High PGL should not be considered a mere stress-related epiphenomenon but an additional risk factor, which impacts short- and long-term prognosis through mechanisms involving endothelial dysfunction, platelet aggregation, inflammation, and oxidative stress [4,119,120]. In addition, in line with the above-described pathophysiology, high PGL at admission seems to impair the whole coronary circulation and not only the culprit vessel. To reduce hyperglycemia-related complications, it appears rational to pursue adequate glyco-metabolic control; anyhow, the best glycemic control strategy in the acute phase of AMI remains unknown and further studies are needed and strongly advocated.

Funding

This research received no external funding.

Conflicts of Interest

Giuseppe Biondi-Zoccai has consulted for Aleph, Amarin, Balmed, Cardionovum, Crannmedical, Endocore Lab, Eukon, Guidotti, Innovheart, Meditrial, Menarini, Microport, Opsens Medical, Terumo, and Translumina, outside the present work. The authors declare no conflict of interest.

References

  1. Deedwania, P.; Kosiborod, M.; Barrett, E.; Ceriello, A.; Isley, W.; Mazzone, T.; Raskin, P. Hyperglycemia and acute coronary syndrome: A scientific statement from the american heart association diabetes committee of the council on nutrition, physical activity, and metabolism. Circulation 2008, 117, 1610–1619. [Google Scholar] [CrossRef] [PubMed]
  2. Kosiborod, M.; Deedwania, P. An overview of glycemic control in the coronary care unit with recommendations for clinical management. J. Diabetes Sci. Technol. 2009, 3, 1342–1351. [Google Scholar] [CrossRef] [PubMed]
  3. Kosuge, M.; Kimura, K.; Kojima, S.; Sakamoto, T.; Matsui, K.; Ishihara, M.; Asada, Y.; Tei, C.; Miyazaki, S.; Sonoda, M.; et al. Effects of glucose abnormalities on in-hospital outcome after coronary intervention for acute myocardial infarction. Circ. J. 2005, 69, 375–379. [Google Scholar] [CrossRef] [PubMed]
  4. Kosiborod, M.; Rathore, S.S.; Inzucchi, S.E.; Masoudi, F.A.; Wang, Y.; Havranek, E.P.; Krumholz, H.M. Admission glucose and mortality in elderly patients hospitalized with acute myocardial infarction: Implications for patients with and without recognized diabetes. Circulation 2005, 111, 3078–3086. [Google Scholar] [CrossRef] [PubMed]
  5. Wahab, N.N.; Cowden, E.A.; Pearce, N.J.; Gardner, M.J.; Merry, H.; Cox, J.L. Is blood glucose an independent predictor of mortality in acute myocardial infarction in the thrombolytic era? J. Am. Coll. Cardiol. 2002, 40, 1748–1754. [Google Scholar] [CrossRef] [PubMed]
  6. Alkatiri, A.H.; Qalby, N.; Mappangara, I.; Zainal, A.T.F.; Cramer, M.J.; Doevendans, P.A.; Qanitha, A. Stress hyperglycemia and poor outcomes in patients with st-elevation myocardial infarction: A systematic review and meta-analysis. Front. Cardiovasc. Med. 2024, 11, 1303685. [Google Scholar] [CrossRef] [PubMed]
  7. Capes, S.E.; Hunt, D.; Malmberg, K.; Gerstein, H.C. Stress hyperglycaemia and increased risk of death after myocardial infarction in patients with and without diabetes: A systematic overview. Lancet 2000, 355, 773–778. [Google Scholar] [CrossRef] [PubMed]
  8. Ishihara, M. Acute hyperglycemia in patients with acute myocardial infarction. Circ. J. 2012, 76, 563–571. [Google Scholar] [CrossRef]
  9. Finfer, S.; Chittock, D.R.; Su, S.Y.; Blair, D.; Foster, D.; Dhingra, V.; Bellomo, R.; Cook, D.; Dodek, P.; Henderson, W.R.; et al. Intensive versus conventional glucose control in critically ill patients. N. Engl. J. Med. 2009, 360, 1283–1297. [Google Scholar] [CrossRef]
  10. Goldberg, R.J.; Kramer, D.G.; Lessard, D.; Yarzebski, J.; Gore, J.M. Serum glucose levels and hospital outcomes in patients with acute myocardial infarction without prior diabetes: A community-wide perspective. Coron. Artery Dis. 2007, 18, 125–131. [Google Scholar] [CrossRef]
  11. Hoebers, L.P.; Damman, P.; Claessen, B.E.; Vis, M.M.; Baan, J., Jr.; van Straalen, J.P.; Fischer, J.; Koch, K.T.; Tijssen, J.G.; de Winter, R.J.; et al. Predictive value of plasma glucose level on admission for short and long term mortality in patients with st-elevation myocardial infarction treated with primary percutaneous coronary intervention. Am. J. Cardiol. 2012, 109, 53–59. [Google Scholar] [CrossRef] [PubMed]
  12. Iwakura, K.; Ito, H.; Ikushima, M.; Kawano, S.; Okamura, A.; Asano, K.; Kuroda, T.; Tanaka, K.; Masuyama, T.; Hori, M.; et al. Association between hyperglycemia and the no-reflow phenomenon in patients with acute myocardial infarction. J. Am. Coll. Cardiol. 2003, 41, 1–7. [Google Scholar] [CrossRef] [PubMed]
  13. Krinsley, J.S.; Egi, M.; Kiss, A.; Devendra, A.N.; Schuetz, P.; Maurer, P.M.; Schultz, M.J.; van Hooijdonk, R.T.; Kiyoshi, M.; Mackenzie, I.M.; et al. Diabetic status and the relation of the three domains of glycemic control to mortality in critically ill patients: An international multicenter cohort study. Crit. Care 2013, 17, R37. [Google Scholar] [CrossRef] [PubMed]
  14. Mladenović, V.; Zdravković, V.; Jović, M.; Vucić, R.; Irić-Cupić, V.; Rosić, M. Influence of admission plasma glucose level on short- and long-term prognosis in patients with st-segment elevation myocardial infarction. Vojnosanit. Pregl. 2010, 67, 291–295. [Google Scholar] [CrossRef] [PubMed]
  15. Zhao, C.J.; Hao, Z.X.; Liu, R.; Liu, Y. Admission glucose and risk of early death in non-diabetic patients with st-segment elevation myocardial infarction: A meta-analysis. Med. Sci. Monit. 2015, 21, 1387–1394. [Google Scholar] [CrossRef] [PubMed]
  16. Paolisso, P.; Foà, A.; Bergamaschi, L.; Angeli, F.; Fabrizio, M.; Donati, F.; Toniolo, S.; Chiti, C.; Rinaldi, A.; Stefanizzi, A.; et al. Impact of admission hyperglycemia on short and long-term prognosis in acute myocardial infarction: Minoca versus mioca. Cardiovasc. Diabetol. 2021, 20, 192. [Google Scholar] [CrossRef] [PubMed]
  17. Ishihara, M.; Inoue, I.; Kawagoe, T.; Shimatani, Y.; Kurisu, S.; Nishioka, K.; Umemura, T.; Nakamura, S.; Yoshida, M. Impact of acute hyperglycemia on left ventricular function after reperfusion therapy in patients with a first anterior wall acute myocardial infarction. Am. Heart J. 2003, 146, 674–678. [Google Scholar] [CrossRef]
  18. Timmer, J.R.; van der Horst, I.C.; Ottervanger, J.P.; Henriques, J.P.; Hoorntje, J.C.; de Boer, M.J.; Suryapranata, H.; Zijlstra, F. Prognostic value of admission glucose in non-diabetic patients with myocardial infarction. Am. Heart J. 2004, 148, 399–404. [Google Scholar] [CrossRef]
  19. Singh, K.; Hibbert, B.; Singh, B.; Carson, K.; Premaratne, M.; Le May, M.; Chong, A.-Y.; Arstall, M.; So, D. Meta-analysis of admission hyperglycaemia in acute myocardial infarction patients treated with primary angioplasty: A cause or a marker of mortality? Eur. Heart J.-Cardiovasc. Pharmacother. 2015, 1, 220–228. [Google Scholar] [CrossRef]
  20. Li, M.; Gao, Y.; Guo, K.; Wu, Z.; Lao, Y.; Li, J.; Huang, X.; Feng, L.; Dong, J.; Yuan, Y. Association between fasting hyperglycemia and new-onset atrial fibrillation in patients with acute myocardial infarction and the impact on short- and long-term prognosis. Front. Cardiovasc. Med. 2021, 8, 667527. [Google Scholar] [CrossRef]
  21. Shechter, M.; Merz, C.N.; Paul-Labrador, M.J.; Kaul, S. Blood glucose and platelet-dependent thrombosis in patients with coronary artery disease. J. Am. Coll. Cardiol. 2000, 35, 300–307. [Google Scholar] [CrossRef] [PubMed]
  22. Krüger-Genge, A.; Blocki, A.; Franke, R.P.; Jung, F. Vascular endothelial cell biology: An update. Int. J. Mol. Sci. 2019, 20, 4411. [Google Scholar] [CrossRef] [PubMed]
  23. Alexander, Y.; Osto, E.; Schmidt-Trucksäss, A.; Shechter, M.; Trifunovic, D.; Duncker, D.J.; Aboyans, V.; Bäck, M.; Badimon, L.; Cosentino, F.; et al. Endothelial function in cardiovascular medicine: A consensus paper of the european society of cardiology working groups on atherosclerosis and vascular biology, aorta and peripheral vascular diseases, coronary pathophysiology and microcirculation, and thrombosis. Cardiovasc. Res. 2021, 117, 29–42. [Google Scholar] [CrossRef] [PubMed]
  24. Cosentino, F.; Lüscher, T.F. Endothelial function in coronary artery disease. Cardiologia 1997, 42, 1221–1227. [Google Scholar] [PubMed]
  25. He, Z.; King, G.L. Microvascular complications of diabetes. Endocrinol. Metab. Clin. N. Am. 2004, 33, 215–238. [Google Scholar] [CrossRef] [PubMed]
  26. Lüscher, T.F.; Barton, M. Biology of the endothelium. Clin. Cardiol. 1997, 20, II-3-10. [Google Scholar] [CrossRef] [PubMed]
  27. Marfella, R.; Esposito, K.; Giunta, R.; Coppola, G.; De Angelis, L.; Farzati, B.; Paolisso, G.; Giugliano, D. Circulating adhesion molecules in humans: Role of hyperglycemia and hyperinsulinemia. Circulation 2000, 101, 2247–2251. [Google Scholar] [CrossRef] [PubMed]
  28. Marfella, R.; Siniscalchi, M.; Esposito, K.; Sellitto, A.; De Fanis, U.; Romano, C.; Portoghese, M.; Siciliano, S.; Nappo, F.; Sasso, F.C.; et al. Effects of stress hyperglycemia on acute myocardial infarction: Role of inflammatory immune process in functional cardiac outcome. Diabetes Care 2003, 26, 3129–3135. [Google Scholar] [CrossRef] [PubMed]
  29. European Diabetes Epidemiology Group. Glucose tolerance and mortality: Comparison of who and american diabetes association diagnostic criteria. The decode study group. Diabetes epidemiology: Collaborative analysis of diagnostic criteria in europe. Lancet 1999, 354, 617–621. [Google Scholar] [CrossRef]
  30. Lash, J.M.; Nase, G.P.; Bohlen, H.G. Acute hyperglycemia depresses arteriolar no formation in skeletal muscle. Am. J. Physiol. 1999, 277, H1513–H1520. [Google Scholar] [CrossRef]
  31. Monnier, L.; Mas, E.; Ginet, C.; Michel, F.; Villon, L.; Cristol, J.P.; Colette, C. Activation of oxidative stress by acute glucose fluctuations compared with sustained chronic hyperglycemia in patients with type 2 diabetes. JAMA 2006, 295, 1681–1687. [Google Scholar] [CrossRef] [PubMed]
  32. Taylor, P.D.; Poston, L. The effect of hyperglycaemia on function of rat isolated mesenteric resistance artery. Br. J. Pharmacol. 1994, 113, 801–808. [Google Scholar] [CrossRef] [PubMed]
  33. Williams, S.B.; Goldfine, A.B.; Timimi, F.K.; Ting, H.H.; Roddy, M.A.; Simonson, D.C.; Creager, M.A. Acute hyperglycemia attenuates endothelium-dependent vasodilation in humans in vivo. Circulation 1998, 97, 1695–1701. [Google Scholar] [CrossRef] [PubMed]
  34. Sun, B.; Luo, Z.; Zhou, J. Comprehensive elaboration of glycemic variability in diabetic macrovascular and microvascular complications. Cardiovasc. Diabetol. 2021, 20, 9. [Google Scholar] [CrossRef] [PubMed]
  35. Addabbo, F.; Ratliff, B.; Park, H.C.; Kuo, M.C.; Ungvari, Z.; Csiszar, A.; Krasnikov, B.; Sodhi, K.; Zhang, F.; Nasjletti, A.; et al. The krebs cycle and mitochondrial mass are early victims of endothelial dysfunction: Proteomic approach. Am. J. Pathol. 2009, 174, 34–43. [Google Scholar] [CrossRef] [PubMed]
  36. Ciccone, M.M.; Scicchitano, P.; Zito, A.; Cortese, F.; Boninfante, B.; Falcone, V.A.; Quaranta, V.N.; Ventura, V.A.; Zucano, A.; Di Serio, F.; et al. Correlation between inflammatory markers of atherosclerosis and carotid intima-media thickness in obstructive sleep apnea. Molecules 2014, 19, 1651–1662. [Google Scholar] [CrossRef] [PubMed]
  37. Pepe, M.; Napoli, G.; Carulli, E.; Moscarelli, M.; Forleo, C.; Nestola, P.L.; Biondi-Zoccai, G.; Giordano, A.; Favale, S. Autoimmune diseases in patients undergoing percutaneous coronary intervention: A risk factor for in-stent restenosis? Atherosclerosis 2021, 333, 24–31. [Google Scholar] [CrossRef] [PubMed]
  38. Inoguchi, T.; Li, P.; Umeda, F.; Yu, H.Y.; Kakimoto, M.; Imamura, M.; Aoki, T.; Etoh, T.; Hashimoto, T.; Naruse, M.; et al. High glucose level and free fatty acid stimulate reactive oxygen species production through protein kinase c—Dependent activation of nad(p)h oxidase in cultured vascular cells. Diabetes 2000, 49, 1939–1945. [Google Scholar] [CrossRef] [PubMed]
  39. Potenza, M.A.; Addabbo, F.; Montagnani, M. Vascular actions of insulin with implications for endothelial dysfunction. Am. J. Physiol. Endocrinol. Metab. 2009, 297, E568–E577. [Google Scholar] [CrossRef]
  40. Du, X.L.; Edelstein, D.; Rossetti, L.; Fantus, I.G.; Goldberg, H.; Ziyadeh, F.; Wu, J.; Brownlee, M. Hyperglycemia-induced mitochondrial superoxide overproduction activates the hexosamine pathway and induces plasminogen activator inhibitor-1 expression by increasing sp1 glycosylation. Proc. Natl. Acad. Sci. USA 2000, 97, 12222–12226. [Google Scholar] [CrossRef]
  41. Nishikawa, T.; Edelstein, D.; Du, X.L.; Yamagishi, S.; Matsumura, T.; Kaneda, Y.; Yorek, M.A.; Beebe, D.; Oates, P.J.; Hammes, H.P.; et al. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature 2000, 404, 787–790. [Google Scholar] [CrossRef]
  42. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001, 414, 813–820. [Google Scholar] [CrossRef]
  43. Asano, G.; Takashi, E.; Ishiwata, T.; Onda, M.; Yokoyama, M.; Naito, Z.; Ashraf, M.; Sugisaki, Y. Pathogenesis and protection of ischemia and reperfusion injury in myocardium. J. Nippon Med. Sch. 2003, 70, 384–392. [Google Scholar] [CrossRef] [PubMed]
  44. Kersten, J.R.; Montgomery, M.W.; Ghassemi, T.; Gross, E.R.; Toller, W.G.; Pagel, P.S.; Warltier, D.C. Diabetes and hyperglycemia impair activation of mitochondrial k(atp) channels. Am. J. Physiol. Heart Circ. Physiol. 2001, 280, H1744–H1750. [Google Scholar] [CrossRef]
  45. Inoguchi, T.; Xia, P.; Kunisaki, M.; Higashi, S.; Feener, E.P.; King, G.L. Insulin’s effect on protein kinase c and diacylglycerol induced by diabetes and glucose in vascular tissues. Am. J. Physiol. 1994, 267, E369–E379. [Google Scholar] [CrossRef]
  46. Tesfamariam, B. Free radicals in diabetic endothelial cell dysfunction. Free Radic. Biol. Med. 1994, 16, 383–391. [Google Scholar] [CrossRef] [PubMed]
  47. Wolf, B.A.; Williamson, J.R.; Easom, R.A.; Chang, K.; Sherman, W.R.; Turk, J. Diacylglycerol accumulation and microvascular abnormalities induced by elevated glucose levels. J. Clin. Investig. 1991, 87, 31–38. [Google Scholar] [CrossRef] [PubMed]
  48. Williams, B.; Schrier, R.W. Characterization of glucose-induced in situ protein kinase c activity in cultured vascular smooth muscle cells. Diabetes 1992, 41, 1464–1472. [Google Scholar] [CrossRef]
  49. Baartscheer, A.; Schumacher, C.A.; Wüst, R.C.; Fiolet, J.W.; Stienen, G.J.; Coronel, R.; Zuurbier, C.J. Empagliflozin decreases myocardial cytoplasmic na(+) through inhibition of the cardiac na(+)/h(+) exchanger in rats and rabbits. Diabetologia 2017, 60, 568–573. [Google Scholar] [CrossRef]
  50. Van Steenbergen, A.; Balteau, M.; Ginion, A.; Ferté, L.; Battault, S.; Ravenstein, C.M.; Balligand, J.L.; Daskalopoulos, E.P.; Gilon, P.; Despa, F.; et al. Sodium-myoinositol cotransporter-1, smit1, mediates the production of reactive oxygen species induced by hyperglycemia in the heart. Sci. Rep. 2017, 7, 41166. [Google Scholar] [CrossRef]
  51. Balteau, M.; Tajeddine, N.; de Meester, C.; Ginion, A.; Rosiers, C.D.; Brady, N.R.; Sommereyns, C.; Horman, S.; Vanoverschelde, J.L.; Gailly, P.; et al. Nadph oxidase activation by hyperglycaemia in cardiomyocytes is independent of glucose metabolism but requires sglt1. Cardiovasc. Res. 2011, 92, 237–246. [Google Scholar] [CrossRef] [PubMed]
  52. Wautier, M.P.; Chappey, O.; Corda, S.; Stern, D.M.; Schmidt, A.M.; Wautier, J.L. Activation of nadph oxidase by age links oxidant stress to altered gene expression via rage. Am. J. Physiol. Endocrinol. Metab. 2001, 280, E685–E694. [Google Scholar] [CrossRef] [PubMed]
  53. Gugliucci, A.; Menini, T. The axis age-rage-soluble rage and oxidative stress in chronic kidney disease. Adv. Exp. Med. Biol. 2014, 824, 191–208. [Google Scholar] [CrossRef] [PubMed]
  54. Furchgott, R.F.; Zawadzki, J.V. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature 1980, 288, 373–376. [Google Scholar] [CrossRef] [PubMed]
  55. Griffith, O.W.; Stuehr, D.J. Nitric oxide synthases: Properties and catalytic mechanism. Annu. Rev. Physiol. 1995, 57, 707–736. [Google Scholar] [CrossRef] [PubMed]
  56. Addabbo, F.; Nacci, C.; De Benedictis, L.; Leo, V.; Tarquinio, M.; Quon, M.J.; Montagnani, M. Globular adiponectin counteracts vcam-1-mediated monocyte adhesion via adipor1/nf-κb/cox-2 signaling in human aortic endothelial cells. Am. J. Physiol. Endocrinol. Metab. 2011, 301, E1143–E1154. [Google Scholar] [CrossRef] [PubMed]
  57. Govers, R.; Rabelink, T.J. Cellular regulation of endothelial nitric oxide synthase. Am. J. Physiol. Renal. Physiol. 2001, 280, F193–F206. [Google Scholar] [CrossRef] [PubMed]
  58. Du, X.L.; Edelstein, D.; Dimmeler, S.; Ju, Q.; Sui, C.; Brownlee, M. Hyperglycemia inhibits endothelial nitric oxide synthase activity by posttranslational modification at the akt site. J. Clin. Investig. 2001, 108, 1341–1348. [Google Scholar] [CrossRef] [PubMed]
  59. Federici, M.; Menghini, R.; Mauriello, A.; Hribal, M.L.; Ferrelli, F.; Lauro, D.; Sbraccia, P.; Spagnoli, L.G.; Sesti, G.; Lauro, R. Insulin-dependent activation of endothelial nitric oxide synthase is impaired by o-linked glycosylation modification of signaling proteins in human coronary endothelial cells. Circulation 2002, 106, 466–472. [Google Scholar] [CrossRef]
  60. Potashnik, R.; Bloch-Damti, A.; Bashan, N.; Rudich, A. Irs1 degradation and increased serine phosphorylation cannot predict the degree of metabolic insulin resistance induced by oxidative stress. Diabetologia 2003, 46, 639–648. [Google Scholar] [CrossRef]
  61. Bucala, R.; Tracey, K.J.; Cerami, A. Advanced glycosylation products quench nitric oxide and mediate defective endothelium-dependent vasodilatation in experimental diabetes. J. Clin. Investig. 1991, 87, 432–438. [Google Scholar] [CrossRef]
  62. Vásquez-Vivar, J.; Kalyanaraman, B.; Martásek, P.; Hogg, N.; Masters, B.S.; Karoui, H.; Tordo, P.; Pritchard, K.A., Jr. Superoxide generation by endothelial nitric oxide synthase: The influence of cofactors. Proc. Natl. Acad. Sci. USA 1998, 95, 9220–9225. [Google Scholar] [CrossRef]
  63. Wever, R.M.; van Dam, T.; van Rijn, H.J.; de Groot, F.; Rabelink, T.J. Tetrahydrobiopterin regulates superoxide and nitric oxide generation by recombinant endothelial nitric oxide synthase. Biochem. Biophys. Res. Commun. 1997, 237, 340–344. [Google Scholar] [CrossRef] [PubMed]
  64. Milstien, S.; Katusic, Z. Oxidation of tetrahydrobiopterin by peroxynitrite: Implications for vascular endothelial function. Biochem. Biophys. Res. Commun. 1999, 263, 681–684. [Google Scholar] [CrossRef]
  65. Xia, Y.; Tsai, A.L.; Berka, V.; Zweier, J.L. Superoxide generation from endothelial nitric-oxide synthase. A ca2+/calmodulin-dependent and tetrahydrobiopterin regulatory process. J. Biol. Chem. 1998, 273, 25804–25808. [Google Scholar] [CrossRef]
  66. Heitzer, T.; Krohn, K.; Albers, S.; Meinertz, T. Tetrahydrobiopterin improves endothelium-dependent vasodilation by increasing nitric oxide activity in patients with type ii diabetes mellitus. Diabetologia 2000, 43, 1435–1438. [Google Scholar] [CrossRef] [PubMed]
  67. Grant, P.J. Diabetes mellitus as a prothrombotic condition. J. Intern. Med. 2007, 262, 157–172. [Google Scholar] [CrossRef]
  68. Ceriello, A.; Giugliano, D.; Quatraro, A.; Russo, P.D.; Marchi, E.; Torella, R. Hyperglycemia may determine fibrinopeptide a plasma level increase in humans. Metabolism 1989, 38, 1162–1163. [Google Scholar] [CrossRef] [PubMed]
  69. Ceriello, A.; Giugliano, D.; Quatraro, A.; Russo, P.D.; Torella, R. Blood glucose may condition factor vii levels in diabetic and normal subjects. Diabetologia 1988, 31, 889–891. [Google Scholar] [CrossRef]
  70. Pandolfi, A.; Giaccari, A.; Cilli, C.; Alberta, M.M.; Morviducci, L.; De Filippis, E.A.; Buongiorno, A.; Pellegrini, G.; Capani, F.; Consoli, A. Acute hyperglycemia and acute hyperinsulinemia decrease plasma fibrinolytic activity and increase plasminogen activator inhibitor type 1 in the rat. Acta. Diabetol. 2001, 38, 71–76. [Google Scholar] [CrossRef]
  71. Enomoto, M.; Adachi, H.; Yamagishi, S.; Takeuchi, M.; Furuki, K.; Hino, A.; Hiratsuka, A.; Takajo, Y.; Imaizumi, T. Positive association of serum levels of advanced glycation end products with thrombogenic markers in humans. Metabolism 2006, 55, 912–917. [Google Scholar] [CrossRef]
  72. Ferreiro, J.L.; Angiolillo, D.J. Diabetes and antiplatelet therapy in acute coronary syndrome. Circulation 2011, 123, 798–813. [Google Scholar] [CrossRef]
  73. Gaiz, A.; Mosawy, S.; Colson, N.; Singh, I. Thrombotic and cardiovascular risks in type two diabetes; role of platelet hyperactivity. Biomed. Pharmacother. 2017, 94, 679–686. [Google Scholar] [CrossRef]
  74. Guthikonda, S.; Alviar, C.L.; Vaduganathan, M.; Arikan, M.; Tellez, A.; DeLao, T.; Granada, J.F.; Dong, J.F.; Kleiman, N.S.; Lev, E.I. Role of reticulated platelets and platelet size heterogeneity on platelet activity after dual antiplatelet therapy with aspirin and clopidogrel in patients with stable coronary artery disease. J. Am. Coll. Cardiol. 2008, 52, 743–749. [Google Scholar] [CrossRef]
  75. Melchinger, H.; Jain, K.; Tyagi, T.; Hwa, J. Role of platelet mitochondria: Life in a nucleus-free zone. Front. Cardiovasc. Med. 2019, 6, 153. [Google Scholar] [CrossRef]
  76. Esposito, K.; Nappo, F.; Marfella, R.; Giugliano, G.; Giugliano, F.; Ciotola, M.; Quagliaro, L.; Ceriello, A.; Giugliano, D. Inflammatory cytokine concentrations are acutely increased by hyperglycemia in humans: Role of oxidative stress. Circulation 2002, 106, 2067–2072. [Google Scholar] [CrossRef]
  77. Li, D.; Zhao, L.; Liu, M.; Du, X.; Ding, W.; Zhang, J.; Mehta, J.L. Kinetics of tumor necrosis factor alpha in plasma and the cardioprotective effect of a monoclonal antibody to tumor necrosis factor alpha in acute myocardial infarction. Am. Heart J. 1999, 137, 1145–1152. [Google Scholar] [CrossRef]
  78. Morohoshi, M.; Fujisawa, K.; Uchimura, I.; Numano, F. Glucose-dependent interleukin 6 and tumor necrosis factor production by human peripheral blood monocytes in vitro. Diabetes 1996, 45, 954–959. [Google Scholar] [CrossRef]
  79. Visser, L.; Zuurbier, C.J.; Hoek, F.J.; Opmeer, B.C.; de Jonge, E.; de Mol, B.A.; van Wezel, H.B. Glucose, insulin and potassium applied as perioperative hyperinsulinaemic normoglycaemic clamp: Effects on inflammatory response during coronary artery surgery. Br. J. Anaesth. 2005, 95, 448–457. [Google Scholar] [CrossRef]
  80. Booth, G.; Stalker, T.J.; Lefer, A.M.; Scalia, R. Elevated ambient glucose induces acute inflammatory events in the microvasculature: Effects of insulin. Am. J. Physiol. Endocrinol. Metab. 2001, 280, E848–E856. [Google Scholar] [CrossRef]
  81. Hokama, J.Y.; Ritter, L.S.; Davis-Gorman, G.; Cimetta, A.D.; Copeland, J.G.; McDonagh, P.F. Diabetes enhances leukocyte accumulation in the coronary microcirculation early in reperfusion following ischemia. J. Diabetes Complicat. 2000, 14, 96–107. [Google Scholar] [CrossRef]
  82. Sekar, M.; Thirumurugan, K. Autophagy: A molecular switch to regulate adipogenesis and lipolysis. Mol. Cell Biochem. 2022, 477, 727–742. [Google Scholar] [CrossRef]
  83. Bharath, L.P.; Rockhold, J.D.; Conway, R. Selective autophagy in hyperglycemia-induced microvascular and macrovascular diseases. Cells 2021, 10, 2114. [Google Scholar] [CrossRef]
  84. García-Díez, E.; Pérez-Jiménez, J.; Martín, M.Á.; Ramos, S. (−)-epicatechin and colonic metabolite 2,3-dihydroxybenzoic acid, alone or in combination with metformin, protect cardiomyocytes from high glucose/high palmitic acid-induced damage by regulating redox status, apoptosis and autophagy. Food Funct. 2024, 15, 2536–2549. [Google Scholar] [CrossRef]
  85. Saad, R.; Tadmor, H.; Ertracht, O.; Nakhoul, N.; Nakhoul, F.; Evgeny, F.; Atar, S. The molecular effects of sglt2i empagliflozin on the autophagy pathway in diabetes mellitus type 2 and its complications. J. Diabetes Res. 2022, 2022, 8337823. [Google Scholar] [CrossRef]
  86. Hao, Y.; Lu, Q.; Li, T.; Yang, G.; Hu, P.; Ma, A. Admission hyperglycemia and adverse outcomes in diabetic and non-diabetic patients with non-st-elevation myocardial infarction undergoing percutaneous coronary intervention. BMC Cardiovasc. Disord. 2017, 17, 6. [Google Scholar] [CrossRef]
  87. Kosiborod, M.; Inzucchi, S.E.; Krumholz, H.M.; Xiao, L.; Jones, P.G.; Fiske, S.; Masoudi, F.A.; Marso, S.P.; Spertus, J.A. Glucometrics in patients hospitalized with acute myocardial infarction: Defining the optimal outcomes-based measure of risk. Circulation 2008, 117, 1018–1027. [Google Scholar] [CrossRef]
  88. Timmer, J.R.; Ottervanger, J.P.; de Boer, M.J.; Dambrink, J.H.; Hoorntje, J.C.; Gosselink, A.T.; Suryapranata, H.; Zijlstra, F.; Hof, A.W.V. Hyperglycemia is an important predictor of impaired coronary flow before reperfusion therapy in st-segment elevation myocardial infarction. J. Am. Coll Cardiol. 2005, 45, 999–1002. [Google Scholar] [CrossRef]
  89. Gibson, C.M.; Cannon, C.P.; Daley, W.L.; Dodge, J.T., Jr.; Alexander, B., Jr.; Marble, S.J.; McCabe, C.H.; Raymond, L.; Fortin, T.; Poole, W.K.; et al. Timi frame count: A quantitative method of assessing coronary artery flow. Circulation 1996, 93, 879–888. [Google Scholar] [CrossRef]
  90. Eshraghi, A.; Talasaz, A.H.; Salamzadeh, J.; Bahremand, M.; Salarifar, M.; Nozari, Y.; Jenab, Y.; Boroumand, M.A.; Vaseghi, G.; Eshraghi, N. Study of the possible medical and medication explanatory factors of angiographic outcomes in patients with acute st elevation myocardial infarction undergoing primary percutaneous intervention. Adv. Biomed. Res. 2014, 3, 186. [Google Scholar] [CrossRef]
  91. Engler, R.L.; Dahlgren, M.D.; Morris, D.D.; Peterson, M.A.; Schmid-Schönbein, G.W. Role of leukocytes in response to acute myocardial ischemia and reflow in dogs. Am. J. Physiol. 1986, 251, H314–H323. [Google Scholar] [CrossRef]
  92. McDonagh, P.F.; Hokama, J.Y.; Copeland, J.G.; Reynolds, J.M. The blood contribution to early myocardial reperfusion injury is amplified in diabetes. Diabetes 1997, 46, 1859–1867. [Google Scholar] [CrossRef]
  93. Nakamura, T.; Ako, J.; Kadowaki, T.; Funayama, H.; Sugawara, Y.; Kubo, N.; Momomura, S. Impact of acute hyperglycemia during primary stent implantation in patients with st-elevation myocardial infarction. J. Cardiol. 2009, 53, 272–277. [Google Scholar] [CrossRef]
  94. Kersten, J.R.; Toller, W.G.; Tessmer, J.P.; Pagel, P.S.; Warltier, D.C. Hyperglycemia reduces coronary collateral blood flow through a nitric oxide-mediated mechanism. Am. J. Physiol. Heart Circ. Physiol. 2001, 281, H2097–H2104. [Google Scholar] [CrossRef]
  95. Montalescot, G.; Barragan, P.; Wittenberg, O.; Ecollan, P.; Elhadad, S.; Villain, P.; Boulenc, J.M.; Morice, M.C.; Maillard, L.; Pansiéri, M.; et al. Platelet glycoprotein iib/iiia inhibition with coronary stenting for acute myocardial infarction. N. Engl. J. Med. 2001, 344, 1895–1903. [Google Scholar] [CrossRef]
  96. Opie, L.H. Preconditioning and metabolic anti-ischaemic agents. Eur. Heart J. 2003, 24, 1854–1856. [Google Scholar] [CrossRef]
  97. Yang, Z.; Tian, Y.; Liu, Y.; Hennessy, S.; Kron, I.L.; French, B.A. Acute hyperglycemia abolishes ischemic preconditioning by inhibiting akt phosphorylation: Normalizing blood glucose before ischemia restores ischemic preconditioning. Oxid. Med. Cell Longev. 2013, 2013, 329183. [Google Scholar] [CrossRef]
  98. Zuurbier, C.J.; Demirci, C.; Koeman, A.; Vink, H.; Ince, C. Short-term hyperglycemia increases endothelial glycocalyx permeability and acutely decreases lineal density of capillaries with flowing red blood cells. J. Appl. Physiol. 2005, 99, 1471–1476. [Google Scholar] [CrossRef]
  99. Gibson, C.M.; Ryan, K.A.; Murphy, S.A.; Mesley, R.; Marble, S.J.; Giugliano, R.P.; Cannon, C.P.; Antman, E.M.; Braunwald, E. Impaired coronary blood flow in nonculprit arteries in the setting of acute myocardial infarction. The timi study group. Thrombolysis in myocardial infarction. J. Am. Coll Cardiol. 1999, 34, 974–982. [Google Scholar] [CrossRef] [PubMed]
  100. Pepe, M.; Zanna, D.; Cafaro, A.; Marchese, A.; Addabbo, F.; Navarese, E.P.; Napodano, M.; Cecere, A.; Resta, F.; Paradies, V.; et al. Role of plasma glucose level on myocardial perfusion in st-segment elevation myocardial infarction patients. J. Diabetes Complicat. 2018, 32, 764–769. [Google Scholar] [CrossRef] [PubMed]
  101. Ferreira, J.A.; Baptista, R.M.; Monteiro, S.R.; Gonçalves, F.M.; Monteiro, P.F.; Gonçalves, L.M. Admission hyperglycemia and all-cause mortality in diabetic and non-diabetic patients with acute myocardial infarction: A tertiary center analysis. Intern. Emerg. Med. 2021, 16, 2109–2119. [Google Scholar] [CrossRef]
  102. Planer, D.; Witzenbichler, B.; Guagliumi, G.; Peruga, J.Z.; Brodie, B.R.; Xu, K.; Fahy, M.; Mehran, R.; Stone, G.W. Impact of hyperglycemia in patients with st-segment elevation myocardial infarction undergoing percutaneous coronary intervention: The horizons-ami trial. Int. J. Cardiol. 2013, 167, 2572–2579. [Google Scholar] [CrossRef]
  103. Stranders, I.; Diamant, M.; van Gelder, R.E.; Spruijt, H.J.; Twisk, J.W.; Heine, R.J.; Visser, F.C. Admission blood glucose level as risk indicator of death after myocardial infarction in patients with and without diabetes mellitus. Arch. Intern. Med. 2004, 164, 982–988. [Google Scholar] [CrossRef]
  104. Demarchi, A.; Cornara, S.; Somaschini, A.; Fortuni, F.; Mandurino-Mirizzi, A.; Crimi, G.; Ferlini, M.; Gnecchi, M.; De Servi, S.; Visconti, L.O.; et al. Has hyperglycemia a different prognostic role in stemi patients with or without diabetes? Nutr. Metab. Cardiovasc. Dis. 2021, 31, 528–531. [Google Scholar] [CrossRef]
  105. Chaudhuri, A.; Janicke, D.; Wilson, M.F.; Tripathy, D.; Garg, R.; Bandyopadhyay, A.; Calieri, J.; Hoffmeyer, D.; Syed, T.; Ghanim, H.; et al. Anti-inflammatory and profibrinolytic effect of insulin in acute st-segment-elevation myocardial infarction. Circulation 2004, 109, 849–854. [Google Scholar] [CrossRef] [PubMed]
  106. Lautamäki, R.; Airaksinen, K.E.; Seppänen, M.; Toikka, J.; Härkönen, R.; Luotolahti, M.; Borra, R.; Sundell, J.; Knuuti, J.; Nuutila, P. Insulin improves myocardial blood flow in patients with type 2 diabetes and coronary artery disease. Diabetes 2006, 55, 511–516. [Google Scholar] [CrossRef] [PubMed]
  107. McNulty, P.; Pfau, S.; Deckelbaum, L. Effect of plasma insulin level on myocardial blood flow and its mechanism of action. Am. J. Cardiol. 2000, 85, 161–165. [Google Scholar] [CrossRef] [PubMed]
  108. Scherrer, U.; Randin, D.; Vollenweider, P.; Vollenweider, L.; Nicod, P. Nitric oxide release accounts for insulin’s vascular effects in humans. J. Clin. Investig. 1994, 94, 2511–2515. [Google Scholar] [CrossRef]
  109. Iliadis, F.; Kadoglou, N.; Didangelos, T. Insulin and the heart. Diabetes Res. Clin. Pract. 2011, 93 (Suppl. S1), S86–S91. [Google Scholar] [CrossRef]
  110. Iguchi, T.; Hasegawa, T.; Otsuka, K.; Matsumoto, K.; Yamazaki, T.; Nishimura, S.; Nakata, S.; Ehara, S.; Kataoka, T.; Shimada, K.; et al. Insulin resistance is associated with coronary plaque vulnerability: Insight from optical coherence tomography analysis. Eur. Heart J. Cardiovasc. Imaging 2014, 15, 284–291. [Google Scholar] [CrossRef]
  111. Wang, C.C.; Goalstone, M.L.; Draznin, B. Molecular mechanisms of insulin resistance that impact cardiovascular biology. Diabetes 2004, 53, 2735–2740. [Google Scholar] [CrossRef] [PubMed]
  112. Pepe, M.; Sardella, G.; Stefanini, G.G.; Corcione, N.; Nestola, P.L.; Morello, A.; Briguori, C.; Tamburino, C.; Fabbiocchi, F.; Rotolo, F.L.; et al. Impact of insulin-treated and noninsulin-treated diabetes mellitus in all-comer patients undergoing percutaneous coronary interventions with polymer-free biolimus-eluting stent (from the rudi-free registry). Am. J. Cardiol. 2019, 124, 1518–1527. [Google Scholar] [CrossRef] [PubMed]
  113. Malmberg, K.; Norhammar, A.; Wedel, H.; Rydén, L. Glycometabolic state at admission: Important risk marker of mortality in conventionally treated patients with diabetes mellitus and acute myocardial infarction: Long-term results from the diabetes and insulin-glucose infusion in acute myocardial infarction (digami) study. Circulation 1999, 99, 2626–2632. [Google Scholar] [CrossRef] [PubMed]
  114. Malmberg, K.; Rydén, L.; Wedel, H.; Birkeland, K.; Bootsma, A.; Dickstein, K.; Efendic, S.; Fisher, M.; Hamsten, A.; Herlitz, J.; et al. Intense metabolic control by means of insulin in patients with diabetes mellitus and acute myocardial infarction (digami 2): Effects on mortality and morbidity. Eur. Heart J. 2005, 26, 650–661. [Google Scholar] [CrossRef] [PubMed]
  115. Byrne, R.A.; Rossello, X.; Coughlan, J.J.; Barbato, E.; Berry, C.; Chieffo, A.; Claeys, M.J.; Dan, G.A.; Dweck, M.R.; Galbraith, M.; et al. 2023 esc guidelines for the management of acute coronary syndromes. Eur. Heart J. 2023, 44, 3720–3826. [Google Scholar] [CrossRef] [PubMed]
  116. Marx, N.; Federici, M.; Schütt, K.; Müller-Wieland, D.; Ajjan, R.A.; Antunes, M.J.; Christodorescu, R.M.; Crawford, C.; Di Angelantonio, E.; Eliasson, B.; et al. 2023 esc guidelines for the management of cardiovascular disease in patients with diabetes. Eur. Heart J. 2023, 44, 4043–4140. [Google Scholar] [CrossRef] [PubMed]
  117. Cosentino, F.; Grant, P.J.; Aboyans, V.; Bailey, C.J.; Ceriello, A.; Delgado, V.; Federici, M.; Filippatos, G.; Grobbee, D.E.; Hansen, T.B.; et al. 2019 esc guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the easd. Eur. Heart J. 2020, 41, 255–323. [Google Scholar] [CrossRef] [PubMed]
  118. Collet, J.P.; Thiele, H.; Barbato, E.; Barthélémy, O.; Bauersachs, J.; Bhatt, D.L.; Dendale, P.; Dorobantu, M.; Edvardsen, T.; Folliguet, T.; et al. 2020 esc guidelines for the management of acute coronary syndromes in patients presenting without persistent st-segment elevation. Eur. Heart J. 2021, 42, 1289–1367. [Google Scholar] [CrossRef] [PubMed]
  119. Ekmekci, A.; Cicek, G.; Uluganyan, M.; Gungor, B.; Osman, F.; Ozcan, K.S.; Bozbay, M.; Ertas, G.; Zencirci, A.; Sayar, N.; et al. Admission hyperglycemia predicts inhospital mortality and major adverse cardiac events after primary percutaneous coronary intervention in patients without diabetes mellitus. Angiology 2014, 65, 154–159. [Google Scholar] [CrossRef]
  120. Meier, J.J.; Deifuss, S.; Klamann, A.; Launhardt, V.; Schmiegel, W.H.; Nauck, M.A. Plasma glucose at hospital admission and previous metabolic control determine myocardial infarct size and survival in patients with and without type 2 diabetes: The langendreer myocardial infarction and blood glucose in diabetic patients assessment (lambda). Diabetes Care 2005, 28, 2551–2553. [Google Scholar] [CrossRef]
Figure 1. Effects of PKC activation induced by hyperglycemia. Activation of PKC inhibits eNOS expression and increases ET-1 activity, resulting in increased permeability of endothelium coupled with an improved expression of the permeability-enhancing factor VEGF in smooth muscle cells. PKC also contributes to increased microvascular matrix protein production through the expression of TGF-β and overexpression of the fibrinolytic inhibitor PAI-1. ENOS, endothelial nitric oxide synthetase; ET-1, endothelin-1; NAD(P)H, nicotinamide adenine dinucleotide phosphate; NFkB, nuclear factor kappa-light-chain-enhancer of activated B cells; PAI-1, plasminogen activator inhibitor-1; PKC, protein kinase C; TGF, transforming growth factor; VEGF, vascular endothelial growth factor.
Figure 1. Effects of PKC activation induced by hyperglycemia. Activation of PKC inhibits eNOS expression and increases ET-1 activity, resulting in increased permeability of endothelium coupled with an improved expression of the permeability-enhancing factor VEGF in smooth muscle cells. PKC also contributes to increased microvascular matrix protein production through the expression of TGF-β and overexpression of the fibrinolytic inhibitor PAI-1. ENOS, endothelial nitric oxide synthetase; ET-1, endothelin-1; NAD(P)H, nicotinamide adenine dinucleotide phosphate; NFkB, nuclear factor kappa-light-chain-enhancer of activated B cells; PAI-1, plasminogen activator inhibitor-1; PKC, protein kinase C; TGF, transforming growth factor; VEGF, vascular endothelial growth factor.
Ijms 25 08504 g001
Figure 2. Mechanism of hyperglycemia-induced injury in myocardial infarction. ADMA, asymmetric dimethylarginine; AGE, advanced glycation end-products; CRP, C-reactive protein; DAG, diacylglycerol; eNOS, endothelial nitric oxide synthase; FVII, coagulation factor VII; GP, glycoprotein; ICAM, intercellular adhesion molecule; IL, interleukin; MRC, mitochondrial respiratory chain; NO, nitric oxide; PAI, plasminogen activator inhibitor; PKC, protein kinase C; PTL, platelet; TNF, tumor necrosis factor; tPA, tissue plasminogen activator; TXA, thromboxane-A.
Figure 2. Mechanism of hyperglycemia-induced injury in myocardial infarction. ADMA, asymmetric dimethylarginine; AGE, advanced glycation end-products; CRP, C-reactive protein; DAG, diacylglycerol; eNOS, endothelial nitric oxide synthase; FVII, coagulation factor VII; GP, glycoprotein; ICAM, intercellular adhesion molecule; IL, interleukin; MRC, mitochondrial respiratory chain; NO, nitric oxide; PAI, plasminogen activator inhibitor; PKC, protein kinase C; PTL, platelet; TNF, tumor necrosis factor; tPA, tissue plasminogen activator; TXA, thromboxane-A.
Ijms 25 08504 g002
Figure 3. Elevated cTFC in patients with acute hyperglycemia (A). Coronary angiogram (CA) at the 25th cine frame (utilizing 30 fps acquisition), revealing contrast opacification only up to the proximal segment of LAD and CX. (B) CA at the 50th cine frame. (C) CA at the 75th cine frame. (D) CA at the 100th cine frame cTFC, showing full opacification of left coronary artery (normal value ≤ 25 frames).
Figure 3. Elevated cTFC in patients with acute hyperglycemia (A). Coronary angiogram (CA) at the 25th cine frame (utilizing 30 fps acquisition), revealing contrast opacification only up to the proximal segment of LAD and CX. (B) CA at the 50th cine frame. (C) CA at the 75th cine frame. (D) CA at the 100th cine frame cTFC, showing full opacification of left coronary artery (normal value ≤ 25 frames).
Ijms 25 08504 g003
Table 1. Correlation between blood glucose level at presentation and 30-day/1 year mortality, according to Kosiborod et al. [4].
Table 1. Correlation between blood glucose level at presentation and 30-day/1 year mortality, according to Kosiborod et al. [4].
Blood Glucose Level
at Presentation
% of Population30-Days Mortality (%)1 Year Mortality (%)
≤110 mg/dL15.071122.8
111–140 mg/dL26.7813.425.4
141–170 mg/dL17.8117.730.4
171–240 mg/dL19.8622.437.5
≥240 mg/dL20.4827.844.6
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pepe, M.; Addabbo, F.; Cecere, A.; Tritto, R.; Napoli, G.; Nestola, P.L.; Cirillo, P.; Biondi-Zoccai, G.; Giordano, S.; Ciccone, M.M. Acute Hyperglycemia-Induced Injury in Myocardial Infarction. Int. J. Mol. Sci. 2024, 25, 8504. https://doi.org/10.3390/ijms25158504

AMA Style

Pepe M, Addabbo F, Cecere A, Tritto R, Napoli G, Nestola PL, Cirillo P, Biondi-Zoccai G, Giordano S, Ciccone MM. Acute Hyperglycemia-Induced Injury in Myocardial Infarction. International Journal of Molecular Sciences. 2024; 25(15):8504. https://doi.org/10.3390/ijms25158504

Chicago/Turabian Style

Pepe, Martino, Francesco Addabbo, Annagrazia Cecere, Rocco Tritto, Gianluigi Napoli, Palma Luisa Nestola, Plinio Cirillo, Giuseppe Biondi-Zoccai, Salvatore Giordano, and Marco Matteo Ciccone. 2024. "Acute Hyperglycemia-Induced Injury in Myocardial Infarction" International Journal of Molecular Sciences 25, no. 15: 8504. https://doi.org/10.3390/ijms25158504

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