Next Article in Journal
A Subset Screen of the Compounds Australia Scaffold Library Identifies 7-Acylaminodibenzoxazepinones as Potent and Selective Hits for Anti-Giardia Drug Discovery
Next Article in Special Issue
Colocalization Analysis of Cytoplasmic Actin Isoforms Distribution in Endothelial Cells
Previous Article in Journal
Disclosure of Genetic Risk Factors for Alzheimer’s Disease to Cognitively Healthy Individuals—From Current Practice towards a Personalised Medicine Scenario
Previous Article in Special Issue
Succinate at the Crossroad of Metabolism and Angiogenesis: Roles of SDH, HIF1α and SUCNR1
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

In Vitro Modeling of Diabetes Impact on Vascular Endothelium: Are Essentials Engaged to Tune Metabolism?

by
Alexander V. Vorotnikov
*,
Asker Y. Khapchaev
,
Alexey V. Nickashin
and
Vladimir P. Shirinsky
National Medical Research Center of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(12), 3181; https://doi.org/10.3390/biomedicines10123181
Submission received: 11 November 2022 / Revised: 1 December 2022 / Accepted: 6 December 2022 / Published: 8 December 2022
(This article belongs to the Special Issue Vascular Endothelial Functions: Insights from Molecular Perspectives)

Abstract

:
Angiopathy is a common complication of diabetes mellitus. Vascular endothelium is among the first targets to experience blood-borne metabolic alterations, such as hyperglycemia and hyperlipidemia, the hallmarks of type 2 diabetes. To explore mechanisms of vascular dysfunction and eventual damage brought by these pathologic conditions and to find ways to protect vasculature in diabetic patients, various research approaches are used including in vitro endothelial cell-based models. We present an analysis of the data available from these models that identifies early endothelial cell apoptosis associated with oxidative stress as the major outcome of mimicking hyperglycemia and hyperlipidemia in vitro. However, the fate of endothelial cells observed in these studies does not closely follow it in vivo where massive endothelial damage occurs mainly in the terminal stages of diabetes and in conjunction with comorbidities. We propose that the discrepancy is likely in missing essentials that should be available to cultured endothelial cells to adjust the metabolic state and withstand the immediate apoptosis. We discuss the role of carnitine, creatine, and AMP-activated protein kinase (AMPK) in suiting the endothelial metabolism for long-term function in diabetic type milieu in vitro. Engagement of these essentials is anticipated to expand diabetes research options when using endothelial cell-based models.

1. Introduction

Vascular endothelium is the primary target of hyperlipidemia, hyperglycemia, and insulin in metabolic disorders and diabetes, leading to endothelial dysfunction and cardiovascular complications at various stages of pathological changes [1,2,3]. Being a frequent comorbidity of atherosclerosis, diabetes accelerates its progression by damaging vital biomolecules through oxidation, glycation, and a plethora of uncontrolled downstream reactions, stimulates inflammation and associated vascular injury leading to atherothrombosis, thus contributing to increased cardiovascular mortality worldwide [4,5,6]. Understanding molecular features and mechanisms that underlie altered physiology of endothelial cells caused by high levels of ambient glucose or free (nonesterified) fatty acids (FFA) is imperative to develop preventive approaches and targeted therapy of both the metabolic and cardiovascular threats. However, the great obstacle to directly studying unique responses of vascular endothelium in vivo or in situ is its fragile, wafer-thin, and hard to reach nature. For this reason, these responses are usually assessed indirectly via changes of the vascular wall reactivity to endothelium-derived substances should they change under disease conditions or those modelled [7]. A proven enhancement to in vivo/in situ research tools is in vitro endothelial cell-based models [8]. Endothelial cells are isolated from tissue biopsies, cultured to propagate, and provide means to manipulate gene expression, uniformly apply stimuli, and enable (patho)physiologic and microstructural studies. The cell-based models provide full control to a researcher and allow the mechanistic studies that are hard to perform in the in vivo setting.
The critical issue that arises in such an approach is whether the culture conditions adequately match those in situ [9]. Considerable efforts have been made to develop culture media suitable for propagation of endothelial cells and resulted in the design of an apparent endothelial basal medium (EBM) prototype also known as MCDB-131 [10]. To enable cell growth, EBM is further furnished with a defined set of growth factors, hormones, vitamins, and other additives to obtain the so-called endothelial growth medium (EGM). Still, EGM contains up to 5% fetal serum to deliver yet uncharacterized factors required for endothelial cells. Compared to the original endothelial growth media, such as M199, DMEM, MEM, F12, etc., supplemented with 10–20% fetal serum, EGM has more defined composition. Several media are now available from various vendors that implement the same or similar EBM/EGM strategy [11]. The most popular vendors include Lonza (Switzerland), Cell Applications (USA), PromoCell (Germany), and ScienCell (USA). The extensive use of low-serum EGM by many laboratories across the globe allowed standardizing the endothelial cell culture conditions and ensured a cross-comparison of the results from various fields of endothelial research.
In particular, metabolic aberrations typical for type 2 diabetes (T2D) have been reproduced in endothelial cell culture in multiple studies in order to reveal how these pathologic conditions affect vascular endothelium. They included exposure of endothelial cells to high environmental glucose or lipid levels typical for T2D progression and implicated in endothelial dysfunction. High content of fatty acids with low carbon number and few/no double bonds in plasma triglycerides and phospholipids are associated with increased risk of diabetes [12]. Palmitic acid, an abundant fatty acid of this type, has been widely used to model hyperlipidemia of prediabetic/diabetic obesity.
In vitro studies promptly identified endothelial apoptosis and cell death as the most common and immediate cell response to high glucose (HG) or palmitate (Figure 1). Overall, these findings have led to a general view on apoptosis as an ultimate outcome of diabetic complications with increased oxidative stress and endothelial dysfunction being just an intermediate step [13,14,15,16]. Yet, it should be stressed that no massive apoptosis of vascular endothelial cells occurs under such metabolic conditions in vivo unless accompanied by inflammation [17,18], atherosclerosis [19], or other diseases [18]. This discrepancy likely stems from the suboptimal composition of endothelial growth media that failed to support endothelium in stringent environment such as that developed in blood in the course of T2D progression.
Below, we review evidence that some essential substances might be missing from the endothelial cell growth media, especially those substances, which are normally produced by other tissues, carried by the blood, and uptaken by vascular endothelium to sustain viability. We suggest that endothelial cells possess an intrinsic mechanism that acts in concert with these essential substances to sense and respond to stressful metabolic conditions in order to tune up metabolism and withstand apoptosis. We discuss metabolic aspects and the key molecules, deteriorated activity of which may lead to vascular endothelium dysfunction.

2. Hyperglycemia Sets Off Multiple Routes to Endothelial Dysfunction and Apoptosis

Table A1 presents the summary of in vitro studies using cultured endothelial cells treated by HG to mimic hyperglycemia. Collectively, they suggest that hyperglycemia induces cell apoptosis via increased reactive oxygen species (ROS) and endothelial nitric oxide synthase (eNOS) uncoupling, oxidative stress, and activation of autophagy (Figure 1). In addition, peroxynitrite accumulation and nitrosative stress accompany oxidative stress, and this mechanism appears important for endothelium since it involves altered eNOS activity. In endothelial cells, insulin signaling that targets eNOS activity through the regulatory phosphorylation of this enzyme also appears to be impaired in hyperglycemia, although this issue was not thoroughly investigated.
Increased ROS generation by endothelial cells is consistently observed in response to both HG and FFA treatment (see below). ROS act as a common upstream event to initiate an array of responses converging into several pathogenic mechanisms [20]. In response to glucose overload, ROS are produced by mitochondria, NADPH-oxidases (NOX), and uncoupled eNOS to trigger DNA double-strand breaks in the nucleus and activation of poly(ADP-ribose) polymerase 1 (PARP-1). PARP-1 depletes intracellular NAD+ and inhibits a key glycolytic enzyme, glyceraldehyde-3-phosphate dehydrogenase, by its polyADP-ribosylation. This causes upstream accumulation of early glycolytic intermediates, which are diverted to activate sorbitol, hexosamine, diacylglycerol/PKC, and advanced glycation end-products (AGE) pathways leading to apoptosis [20]. Depletion of NAD+ also results in SIRT1 inhibition and reduction in AMP-activated protein kinase (AMPK) activity, consistent with lower AMPK activity in insulin-resistant and obese subjects [21,22]. Pharmacological activation of AMPK by AICAR or constitutively active AMPK alleviates the HG-induced oxidant stress and endothelial apoptosis [23].
Measurement of apoptosis in endothelial cell studies requires caution while interpreting the results. Apoptosis is usually assessed indirectly using [3H]thymidine incorporation, DNA laddering, TUNEL or annexin-V assays, expression of pro-apoptotic proteins, such as Bax, Bcl-2, or Bak, and caspase-3 activation [24,25,26,27]. All these approaches seem to suffer from a common drawback of inability to catch the maximum cell response and calculate the true extent of apoptosis. Due to intrinsic heterogeneity of cells in culture, duration of apoptosis can vary between the cells [27]. Some of them can quickly die, disintegrate, and escape the assay, whereas others can survive and manifest only modest responses. Another challenge is that apoptosis can occur at low frequency and the dying endothelial cells can be replaced by proliferation and spreading of adjacent cells, so that the culture can be maintained as a whole [27]. In the majority of studies listed in Table A1 and Table A2, cell apoptosis was measured relative to the untreated cells but not as a fraction of maximum response in the same culture. This leaves uncertainty regarding the true extent of apoptosis in endothelial cell models reproducing both hyperglycemia and hyperlipidemia. To obtain a more complete picture of endothelial cell damage brought by these conditions, it is advantageous, in the course of treatment, to supplement the apoptosis measurements with direct visualization of cells or fluorescent viability assays using low magnification light microscopy to enable wide-field inspection of the endothelial monolayer [27,28]. Additionally, time-lapse imaging of endothelial culture allows observing the dynamics of cell monolayer reparation after the loss of individual apoptotic cells or of wide-range deteriorating events.

3. Hyperlipidemia Threats Endothelium through Increased FFA Levels

Table A2 presents the summary of in vitro studies performed on cultured endothelial cells exposed to FFA (as FFA: albumin complex) with a particular focus on apoptosis. Similar to hyperglycemia studies, these studies collectively suggest that experimental hyperlipidemia induces endothelial cell apoptosis via increased ROS production and oxidative stress, activation of autophagy, and increased inflammation (Figure 1). Furthermore, frequently reported outcomes are the inhibition of insulin signaling and altered eNOS activity. Notably, most of these studies used relatively low FFA concentrations approximately corresponding to those in healthy human subjects (<0.5 mM), perhaps for the reason that higher FFA concentrations acutely triggered endothelial cell death [28]. Based on 50 years of epidemiological studies, plasma FFA levels of 0.7 mM and higher are considered typical for obesity/disease states whereas ~0.6 mM FFA is approximated as a “threshold” level [29,30,31].
Vascular endothelium is exposed to the blood-borne FFA, including the albumin-bound FFA and those liberated by the lipoprotein lipase (LPL) from triacylglycerol (TAG)-enriched lipoproteins at the endothelial surface. About a third of LPL-generated FFA is released into circulation in the process called “spillover” [32]. The lipoprotein-associated TAG are markedly higher in obesity [31] and postprandial chylomicrons are the major source of spilled over FFA [33]. Thus, local FFA concentrations at the blood-endothelial interface would be expected to surpass their mean fasting values in plasma and further increase in obesity or postprandial state.
What makes the external FFA detrimental to vascular endothelium in vitro? Low solubility of long-chain FFA in aqueous solutions is the pivotal factor [34]. In blood plasma, FFA are bound to albumin, which serves both the buffering and transporting functions [35]. Albumin concentration in blood plasma is ~0.6–0.7 mM, and in vitro defatted albumin can non-covalently bind up to seven FFA molecules, although with different affinities. Its three high-affinity binding sites cumulatively provide for effective retention of ~2 mM FFA. In blood plasma, albumin can also complex bilirubin, heme, thyroxine, steroid hormones, bile acids, and a wide array of drugs [35]. Perhaps, due to the presence of other ligands, the measured concentration of albumin-bound FFA in human blood rarely exceeds 2 mM. On the other hand, concentration of FFA unbound to albumin stays in the nanomolar range [34], suggesting that once FFAs dissociate from albumin they should promptly complex with other suitable targets including those on/in endothelial cells. Under pathological conditions, the FFA/albumin molar ratio can rise, reaching as high as six to one, unmasking the unbound FFA lipotoxic effects likely mediated by the inflammatory TLR4 receptors and associated signaling [36]. Thus, it is important to use relevant FFA/albumin molar ratios and clearly indicate them in experimental studies; the ratios greater than five should be avoided as they may potentially induce artifacts [34].

4. Carnitine Is Indispensable for FFA Utilization and Sequestration

Normally, the bulk of FFA traverses the vascular endothelium for ultimate destination in underlying tissues such as skeletal muscle or adipose tissue. A portion of FFA is retained in endothelial cells and may enter various pathways.
The ability of carnitine to enhance oxidation of fatty acids by vascular endothelium in vitro was demonstrated more than three decades ago [37]. The same authors pointed to the propensity of endothelial cells to loose carnitine in culture [38] (Table 1). Exogenous carnitine increased oleate oxidation and ATP generation in human umbilical artery endothelial cells (HUAEC) cultured in the presence of 0.5 mM oleate [37]. Apparently, rich growth medium used in this study (M199, 20% serum and endothelial cell growth supplement (ECGS) from bovine brain) failed to provide sufficient carnitine to support the carnitine-acyl transferase shuttle to deliver the oleic acid to mitochondria for oxidation (Figure 2). When confirmed by subsequent studies [39,40], the idea emerged that the carnitine loss by cultured endothelial cells may produce an artificial impression that vascular endothelium relies predominantly on glucose and glycolysis for its energy needs.
Carnitine supplementation appears important for all standard growth media (M199, DMEM, MEM, F12, MCDB-131, low-serum EGMs) used to culture endothelial cells in stressful conditions that may critically involve oxidation of fatty acids [39]. As emphasized by Ruderman et al., the “high rates of fatty acid oxidation are demonstrable in HUVEC when they are provided with carnitine” [42]. This is consistent with carnitine acting as a key doorkeeper for entry of fatty acids into mitochondria for oxidation via acyl-carnitine shuttles including the major carnitine-palmitoyl transferase, CPT-1 [43] (Figure 2). This implies that many in vitro studies exploring the effects of FFA on endothelium (Table A2) might have experienced an artificially high rate of apoptosis without carnitine supplementation [28].
The key observation made by Ido et al. [23] indicated that this might be the case, because only in the presence of carnitine, additional AMPK-mediated stimulation of fatty acid delivery to mitochondria rescued the cells from apoptosis. From this and collateral studies [39,40,41], the AMPK-mediated mechanism of endothelial protection was suggested to involve decreased levels of intracellular malonyl-CoA, the inhibitor of CPT-1, and to facilitate carnitine-mediated routing of fatty acids to oxidation in mitochondria [42]. Furthermore, it was suggested that this mechanism is common to both the anti-apoptotic and insulin-sensitizing action of AMPK in hyperlipidemia and hyperglycemia [42,44]. If this mechanism is disabled, an increased glucose supply followed by the accumulation of pyruvate and acetyl-CoA would favor malonyl-CoA production via ACC, formation of DAG, and subsequent activation of PKC, NOX, and generation of ROS (Figure 2) to trigger insulin resistance [45]. Similarly, in the absence of FAO flux, accumulating FFA may trigger inflammatory responses [41] via the long-chain acyl-CoA intermediates [46] and/or oxidative/nitrosative stress leading to apoptosis [47]. Overall, as argued by the studies from the Ruderman lab [23,39,40,41,42] and recently supported by the results of Yao et al. [48], carnitine is key for AMPK to manifest its anti-inflammatory, anti-oxidant, and anti-apoptotic activities in endothelial cells.
Additionally, carnitine may participate in maintaining an equilibrium between the pools of the long-chain acyl-CoA (LCACoA) and free HS-CoA in cells. The intracellular concentrations of LCACoA are normally in the 5–160 μM range but vary considerably in different metabolic conditions [49]. The excess of LCACoA is potentially injurious as it might produce a spectrum of aberrant metabolic and signaling switches [46,49] leading to insulin resistance, inflammatory response, dysregulated autophagy, oxidative stress, and apoptosis. Many of these features are observed when endothelial cells are subject to experimental hyperglycemia (Table A1) or hyperlipidemia (Table A2), with carnitine being protective in these settings (reviewed in [50]).
Carnitine is synthesized mainly in the liver and distributed through the blood to other tissues including vascular endothelium. With carnitine concentration in the blood plasma at ~50 μM, it is expected to be about the same in endothelium that is constantly exposed to blood. This is in the range of free and acyl-bound CoA. Thus, due to intrinsic ability to reversibly exchange the acyl groups with many acyl-CoA species ranging from simple acetyl-CoA to LCACoA, carnitine may be essential to maintain the availability of the HS-CoA pool for metabolic needs and to sequester excess LCACoA (Figure 2) in metabolic disorders such as obesity and T2D.

5. AMPK Controls the Malonyl-CoA-Dependent Checkpoint of FFA Utilization

The original discovery that, in addition to being a precursor for the de novo synthesis of fatty acids, malonyl-CoA serves as an acyl-carnitine transferase inhibitor [51] (see [46,52] for explicit reviews), has eventually developed into a key molecular mechanism, mediated by AMPK, that controls both fuel switching by cells and emergence of insulin resistance [42,44,53,54]. Through ACC phosphorylation, AMPK inhibits malonyl-CoA synthesis from acetyl-CoA thereby limiting accumulation of DAG and ceramides that induce ROS generation and apoptosis. Simultaneously, AMPK relieves CPT-1 inhibition by malonyl-CoA. This allows the fatty acid influx into mitochondria (Figure 2), peroxisomes, and endoplasmic reticulum for catabolism [52]. In addition, the skeletal muscle malonyl-CoA-decarboxylase (MCD), which limits the malonyl-CoA amount available for LCACoA formation, is also likely to be under AMPK control [55]. It is unknown whether AMPK similarly regulates MCD and LCACoA levels in vascular endothelium. Despite several other steps of the prototype AMPK/ACC/MCD regulatory mechanism remain to be demonstrated in endothelial cells, it is consistent with the notion that AMPK activity is reduced in diabetic conditions [21,22] and that upregulated FAO may rescue endothelial cells from apoptosis [37,39,48].
Based on the outlined metabolic relationships (Figure 2), it is anticipated that under conditions of excessive glucose or FFA and lack of carnitine and insufficient activity of AMPK in cultured endothelial cells, multiple mechanisms may contribute to endothelial apoptosis. They likely include increased auto-, mito-, or lipophagy [56,57,58,59], the polyol, AGE, and hexosamine pathways branched from glycolysis [20,60], inflammatory signaling and cytokines [61,62], or oxidative/nitrosative stress mediated by NOX or eNOS [47,63,64,65,66,67,68].
In the absence of carnitine and/or with low AMPK activity, the increased malonyl-CoA would be likely rerouted toward de novo synthesis of palmitate, DAG/TAG, and lipid droplet formation. Avoiding the risk of insulin resistance can be achieved via lipid droplet utilization by auto(lipo)phagy. This appears to occur in cells treated with exogenous fatty acids [56,58,59] or high glucose [57]. Inhibiting the early steps in autophagosome formation by 3-methyladenine (3-MA) blocks the Nox4-mediated ROS accumulation, the activating phosphorylation of PKCa [56], and, most importantly, apoptosis [58]. Silencing of vps34, a putative target of 3-MA, produces the same effect [58]. While the mechanism by which auto(lipo)phagy activates the Nox4/ROS response is still elusive, ROS involvement in apoptosis is well conserved in vascular endothelial cells (Table A1 and Table A2).
Whether and to what extent the above-mentioned mechanisms are realized in the presence of carnitine and sufficient AMPK activity and lead to endothelial cell dysfunction and damage needs to be re-evaluated.

6. Mitochondrial Uncoupling as ROS Reduction Option: Potential Mechanisms

Mitochondria are considered the major source of superoxide anion and other ROS, at least in hyperglycemia [65,69,70]. ROS accumulate as by-products of intense aerobic metabolism when the mitochondrial electron transport chain is overloaded and the inner membrane potential increases [57,71]. While there are many multifaceted relationships between mitochondrial ROS and programmed cell death [72], lowering the mitochondrial membrane potential is thought to decrease the production of superoxide anion radicals and to prevent apoptosis. The mitochondrial uncoupling protein UCP-2 belonging to the UCP family [73] was identified in various tissues including vascular endothelium [74,75]. According to suggested models, a member of the UCP family, UCP-1, allows FFA anions to flip from mitochondrial matrix to the outer side of inner mitochondrial membrane where FFA anions are neutralized by accumulated protons and diffuse back into the matrix, where they release protons. This mechanism dissipates the proton gradient (ΔμH+) across the inner mitochondrial membrane and reduces ATP synthase activity [76]. In mammals, UCP-2 also reduces the mitochondrial membrane potential, attenuates mitochondrial ROS production, and protects against oxidative damage [73]. Protective role of mitochondrial uncoupling has been demonstrated by UCP overexpression in endothelial cells. Forced expression of UCP-1, normally found in brown and beige adipocytes, rescues the endothelial cells from high glucose-induced apoptosis [70]. Overexpression of UCP-2 in HAEC inhibits linoleic acid-induced ROS production, NF-kB activation, and apoptosis [77]. High glucose and fatty acids upregulate endogenous UCP-2 expression [73,78], likely demonstrating an adaptive response of endothelial cells to higher mitochondrial respiration. Noteworthy, UCP-2 expression/activity as well as mitochondrial biogenesis stay under AMPK control through the transcriptional peroxisome proliferator coactivator PGC-1a, thus reducing ROS generation by mitochondria in the vascular endothelium [79,80]. In turn, AMPK activity appears to be upregulated by ROS, thus forming a positive feedback circuit to dump mitochondrial ROS production and prevent oxidative stress [68,79]. By this virtue, AMPK seems to function as a dual sensor of both the energy and redox status within a cell [81]. All these findings substantiate an idea that the reduced AMPK activity in prediabetic or diabetic states may aggravate mitochondrial dysfunction and cause insulin resistance [53].
There is also another potential mechanism that may rescue the mitochondrial electron transport chain from overloading. Inability of a cell to spend ATP increases the mitochondrial ROS; restoring the ATP generation lowers the proton electrochemical potential gradient (ΔμH+), increases oxidation of electron carrier pools, and reduces local oxygen to cease superoxide anion radical production in mitochondria [69,82]. Such an ATP-spending mechanism was originally found in neuronal cells and appears to involve either mitochondria-bound hexokinase [83] or creatine kinase [84]. It allows ATP to recycle, which may provide the “mild” uncoupling of mitochondria to prevent ROS formation [82,85]. An increased expression of hexokinase-I has been observed in EA.hy926 endothelial cells treated with high glucose [78]. Whereas it is the hexokinase-II isoform that is generally thought to be mitochondria-targeted, in fact, both isoforms are capable of mitochondria binding and causing “mild” uncoupling [85]. Notably, this mechanism has been implicated as an inherited component of the whole-body anti-aging program [85], but whether it upregulates in endothelial cells to counter the ROS-induced apoptosis is yet unknown.
Similarly, the creatine system (reviewed by [86]) shares much in common with carnitine and the hexokinase mechanism. Endothelial cells contain both creatine transporter (SLC6A8) and substantial amounts of mitochondria-bound creatine kinase [87]. The creatine concentration in the blood plasma is normally ~50 μM but in the typical endothelial EBM-2 culture medium (Bio-Whittaker Inc., USA) containing 5% fetal calf serum, it is ~20-fold less [88]. This suggests that, as with carnitine, the cellular creatine stores may exhaust upon culturing.
A number of studies have elaborated the antioxidant potential of creatine [86], however only a few have addressed whether it controls the mitochondrial membrane potential and ROS production. When human pulmonary endothelial cells were cultured with up to 5 mM extra creatine, the levels of intracellular creatine and phosphocreatine proportionally increased, but the ATP levels remained constant suggesting that mitochondrial ATP generation exceeded initially limited values [88]. Remarkably, this extra creatine inhibited ICAM-1 and E-selectin expression, neutrophil adhesion, and tightened the endothelial barrier by reducing its transient permeability in response to serotonin or hydrogen peroxide, all indicative of the anti-inflammatory effect of creatine. This was substantiated by Meyer et al. in neuronal cells, who directly showed that the ATP⇄ADP recycling by mitochondria-bound creatine kinase decreases ROS generation particularly under high glucose conditions [84]. Studies in cultured HUVEC and C2C12 myoblasts confirm that creatine supplementation protects cells from an oxidant-induced injury and increases cell viability [89,90]. Mechanistically, these effects of creatine could relate to its capability to activate AMPK, PGC-1a, and mitochondrial biogenesis [89].

7. Conclusions

Summing up, the in vitro endothelial models require a thoughtful approach with regard to essential natural components of the culture medium to be available for endothelial cells in order to cope with external metabolic interrogations, such as those occurring in diabetic states. Both high glucose and increased FFA load alter the cellular metabolism to increase intracellular malonyl-CoA and LCACoA and thrust them into the cytosolic lipogenesis flux and/or into mitochondria for the β-Ox/FAO and OxPhos pathways (Figure 2). The choice and the balance are determined by the activities of the rate-limiting enzymes and/or their regulators. Carnitine seems essential for LCACoA buffering and transferring to mitochondria; the lack of carnitine function increases the risk of cell apoptosis. Easy loss and uptake of carnitine appears to be habitual for endothelial cells; therefore, it is essential to maintain endothelial cultures with carnitine supplements, especially in metabolic studies. Unless it is done, LCACoA would be likely rerouted to the lipogenesis pathway, increasing the risk of lipid droplet formation, PKC-mediated insulin resistance and NOX activation, ROS generation, and apoptosis. This flux may also involve the auto(lipo)phagy, which also results in ROS generation by an unknown mechanism(s).
Under high glucose conditions, an increased glycolytic flux triggers three major pathways involved in endothelial cell damage: through polyol/sorbitol or G3P-initiated AGE formation, or F1,6PP-initiated hexosamine pathway (Figure 2). It also increases the levels of dihydroxyacetone phosphate (DHAP) and malonyl-CoA, resulting in both increased de novo synthesis of LCACoA and the lipogenesis flux with all detrimental consequences discussed above.
Involvement of ROS in endothelial dysfunction in diabetic conditions is hardly disputed and seems to be a unifying pathogenic mechanism [14,20]. Yet, differences in the rates of apoptosis in endothelial cell cultures and in the vessels in situ raise the possibility that ROS mechanisms may differ in their extent or contribution. Even further, the ROS-mediated apoptosis may not be the bona fide culprit of diabetic endothelial dysfunction but rather an in vitro side-effect that reflects a primary defect, which may not culminate to such an extreme in vivo until the terminal stages of the disease.
Is there a safeguard regulatory circuit present in cultured/diabetic endothelial cells to protect from detrimental ROS generation? AMPK appears to be an important molecular component of such a circuit. AMPK is an intrinsic double-faced regulator of the cytoplasmic malonyl-CoA and LCACoA levels and mitochondrial ROS (Figure 2). AMPK exerts double control of malonyl-CoA production through ACC inhibition and MCD activation in cytoplasm; AMPK blocks ROS generation through increased mitochondrial biogenesis and respiration. The latter mechanisms are not fully understood but likely include activation of PGC-1α and upregulation of UCP-2. An intriguing question remains as to whether these AMPK effects on ROS involve ATP recycling via mitochondria-bound hexokinase and/or creatine kinase in endothelium. Accumulated data outlined in this review bestow on AMPK a likely potential to be a key regulator of the ROS status in endothelial cells. AMPK activity is lower in insulin-resistant and diabetic states [22,53], making it a promising therapeutic target.
In conclusion, the endothelial cell-based models appear indispensable to explore cellular mechanisms of vascular dysfunction in diabetes-related conditions such as hyperglycemia and/or dyslipidemia. For metabolic studies, the cells need to be constantly supplied with essential factors. Some of them may not yet be fully elaborated, likely because the harsh metabolic conditions had not been envisioned when common cell media were being developed. As evidence accumulates, these essentials may be needed to revive the intrinsic safeguard mechanisms, such as those involving AMPK, to sustain mitochondrial function, combat oxidative stress and prevent artificially high apoptosis. This allows maintaining the cultures for up to several weeks to monitor the long-term progression of cell metabolic dysfunctions. Still, endothelial cell-based models retain limitations with regard to systemic responses imposed by hemodynamic forces or communication with other cells, including blood cells or vascular wall neighbors. These aspects can be also addressed in advanced coculture systems. Clearly, more studies are needed, under the controlled cell culture conditions, to reap the harvest.

Author Contributions

Writing—original draft preparation, A.V.V.; writing—review and editing, V.P.S., A.Y.K., A.V.N. and A.V.V.; funding acquisition, V.P.S. All authors have read and agreed to the published version of the manuscript.

Funding

Supported by Russian Science Foundation grant No. 19-15-00361.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACCAcetyl-CoA carboxylase
AcCoAAcetyl-coenzyme A
ACSLAcyl-CoA synthetase
ADPAdenosine diphosphate
AGEAdvanced glycation end-products
AICARAMPK activator
AktAkt protein kinase
AMPKAMP-activated protein kinase
ATCCAmerican tissue culture collection
ATPAdenosine triphosphate
BAECBovine aortic endothelial cells
BaxBCL2 associated X, apoptosis regulator
BCL-2B-cell lymphoma 2, apoptosis regulator
BIM-1Bisindolylmaleimide 1, protein kinase C inhibitor
BSABovine serum albumin
CarnCarnitine
CCK-8Cell counting kit-8
cGMPCyclic guanosine monophosphate
CK-mtMitochondrial isoform of creatine kinase
COX2Cyclooxygenase 2
CPT-1Carnitine-palmitoyl transferase
CrPCreatine phosphate
DAF2-DADiaminofluorescein diacetate, fluorescent nitric oxide probe
DAGDiacylglycerol
DCFH2DCFDA, a cell-permeant indicator for reactive oxygen species
DHAPDihydroxyacetone phosphate
DMEMCell growth medium formulation
DNADeoxyribonucleic acid
EA.hy926HUVEC-derived endothelial cell line
EBMEndothelial basal medium
ECGSEndothelial cell growth supplement
EGFEpidermal growth factor
EGMEndothelial growth medium
EGM-2mvMicrovascular Endothelial Cell Growth Medium-2 distributed by Lonza
eNOSEndothelial NO synthase
EPCEndothelial progenitor cells
Erk1/2Erk1/2 protein kinase
ER-stressEndoplasmic reticulum stress
ET-1Endothelin-1
ETCElectron transport chain of mitochondria
F1,6PPFructose 1,6 biphosphate
F12Cell growth medium formulation
FADH2Flavin adenine dinucleotide reduced
FAOFatty acid oxidation
FBSFetal bovine serum
FFAFree fatty acid
G3PGlyceraldehyde 3-phosphate
G6PGlucose-6-phosphate
GSHGlutathione, reduced
GSK3βGlycogen synthase kinase-3 beta
HAECHuman aortic endothelial cells
HATHypoxanthine-aminopterin-thymidine, cell growth medium supplement
HCAECHuman coronary artery endothelial cells
HCECHuman corneal endothelial cells
HGHigh glucose
HK I/IIHexokinase I/II isoform
HMEC-1Immortalized human dermal microvascular endothelial cells
HOMA-IRHomeostatic model assessment of insulin resistance
HRECHuman retinal endothelial cells
HS-CoAFree coenzyme A
HUAECHuman umbilical artery endothelial cells
HUVECHuman umbilical vein endothelial cells
ICAM-1Inter-cellular adhesion molecule 1
IKKβInhibitor of NF-kB kinase subunit beta
IL-6Interleukin-6
iNOSInducible NOS
InsInsulin
IκBαNF-κB inhibitor
JNKJNK protein kinase
LCACoALong-chain acyl-coenzyme A
LDlipid droplet
LNAMEL-Nω-nitroarginine methyl ester
LNMAL-Nω-methylarginine, NOS inhibitor
LPLLipoprotein lipase
M199Cell growth medium formulation
MAGMonoacylglycerol
MalCoAMalonyl-coenzyme A
MCDMalonyl-CoA decarboxylase
MCDB 131Cell growth medium formulation
MDAMalondialdehyde
MEMCell growth medium formulation
MGOMethylglyoxal
MHECMouse heart endothelial cells
mTORMammalian target of rapamycin
mTORC1Mammalian target of rapamycin complex 1
MyD88Adaptor protein for Toll-like and interleukin-1 receptors
NACN-acetylcysteine
NAD+Nicotinamide adenine nucleotide oxidized
NADHNicotinamide adenine nucleotide reduced
NADPHNicotinamide adenine nucleotide phosphate reduced
NEFANon-esterified fatty acid
NF-kBNuclear factor kB, transcription factor
NGFNerve growth factor
NONitric oxide
NOXNADPH-oxidases
OxPhosOxidative phosphorylation
p21WAF-1/Cip1Cyclin-dependent kinase inhibitor 1
p38p38 mitogen-activated protein kinases
PAI-1Plasminogen activator inhibitor-1
P-AktPhosphorylated Akt
P-AMPKPhosphorylated AMPK
PARP-1Poly(ADP-ribose) polymerase 1
PCRPolymerase chain reaction
P-eNOSPhosphorylated eNOS
PGC-1αPeroxisome proliferator–activated receptor gamma coactivator-1 alpha
PGE2Prostaglandin E2
PGI2Prostaglandin I2
PGISProstaglandin I2 synthase
PIPropidium iodide, fluorescent intercalating agent for DNA staining
PI3KPhosphatidylinositol 3-kinase
P-IKKPhosphorylated IκB kinase
P-IRSPhosphorylated IRS
P-JNKPhosphorylated JNK
PKCProtein kinase C
PMAPhorbol-12-myristate-13-acetate
P-mTORPhosphorylated mTOR
P-p65Phosphorylated p65
PTENDual specificity phosphatase, dephosphorylates phosphoinositides/proteins
P-TyrPhospho-tyrosine
QCoenzyme Q, ubiquinone
ROSReactive oxygen species
SAPSecreted alkaline phosphatase
SIRT-1Sirtuin-1, deacetylase
SNPSodium nitroprusside
SODSuperoxide dismutase
T2DType 2 diabetes mellitus
TAGTriacylglycerol
TCATricarboxylic acid cycle (Krebs cycle)
TLR4 Toll-like receptor 4
TNFαTumor necrosis factor alpha
TRPV1Transient receptor potential cation channel subfamily V member 1
TUNELDNA fragmentation assay
UCPUncoupling protein
UCP-1Uncoupling protein 1
UCP-2Uncoupling protein 2
VCAM-1Vascular cell adhesion molecule 1
WBWestern blotting
3-MA3-Methyladenine
3-OMG3-O-Methylglucose
β-Oxβ-Oxidation of fatty acids
ΔμH+Membrane electrochemical potential

Appendix A

Table A1. Summary of the outcomes of high glucose effects on endothelial cells (hyperglycemia).
Table A1. Summary of the outcomes of high glucose effects on endothelial cells (hyperglycemia).
Cells and Study DesignCulture ConditionsGlucose Duration and Concentrations, Other EssentialsAssessed ParametersMajor Findings/MechanismRefs
Constant short-term HG treatment (1–3 days)
HUVEC
In vitro
M199, 20% FBS, ECGS5 vs. 30 mM; 24–48–72 h, (also includes the long-term treatment for 13 days)Apoptosis (by DNA fragmentation, [3H]-thymidine incorporation)The phenomenon first reported. Clusterin was not involved[91]
HUVEC
In vitro
M199, 20% FBS, ECGS, passage 45 vs. 30 mM, 48–72 h. Proinsulin or 3-O-methyl- glucose were also usedApoptosis (by cell morphology, flow cytometry, DNA fragmentation)HG, proinsulin, or 3-O-methylglucose induced apoptosis, which was inhibited by PKC activation and increased by PKC inhibition[92]
HUVEC
In vitro
M199, 20% FCS, ECGS, passage 3–55 vs. 19 or 33 mM, 24, 36, 48 hApoptosis, TUNEL assay, cells with hypodiploid DNA by flow cytometry, DNA fragmentation, ROS, intracellular Ca2+Taurine stabilized intracellular Ca2+, blocked HG-induced ROS production and apoptosis[93]
HUVEC
In vitro
M199, 20% FBS, ECGS, passage 3–55 vs. 33 mM, 2–48 hApoptosis, ROS (by DCF), eNOS (by WB), NF-kB, JNK1, caspase-3, PI3K pathwayHG increased eNOS expression that peaked at 6 h, then declined. ROS and apoptosis appeared later (24–48 h), were inhibited by ascorbate, vitamin C, and SNP, and increased by LNAME. HG-induced apoptosis was mediated by NF-kB, JNK1, caspase-3, and counteracted by PI3K > Akt > eNOS activation[94,95,96]
BAEC,
in vitro
DMEM, 10% FCS, passage 5–1010–25 mM HG, 15 min–24 h, 1% FCS, subconfluent cellsROS, apoptosis, cell death (trypan blue), NADPH, GSH, cAMP, glucose-6- phosphate dehydrogenase (G6PD)Rapid (15 min) cAMP-dependent phosphorylation and inhibition of G6PD that decreased GSH and NADPH, increased ROS, and predisposed cells to apoptosis.[97]
HAEC (Clonetics)
In vitro
EGM, 2% FBS, ECGS (Clonetics) for growth, M199, 10% FBS for assays5 vs. 30 mM HG, 6–72 hROS (24 h), apoptosis (Annexin V, PI), GSH, mitochondrial membrane potential (ΔμH+, JC-1 fluorescent probe)HG increased ROS, decreased ΔμH+ and GSH content, increased DNA fragmentation and apoptosis, all of them prevented by 10 mM N-acetylcysteine (NAC)[98]
HUVEC,
in vitro
M199, 20% FBS, ECGS5.5 vs. 30 mM HG, 8–12–48 hApoptosis, viability, COX2, PGE2, NF-kB, ROS, PI3K, caspase-3. HG increased COX2, PGE2, ROS, NF-kB, caspase-3, and apoptosis. All these reduced by blocking PI3K-Akt [99]
HUVEC, in vitro; diabetic mice, in vivoDMEM, 10% FBS, passage 3–75.5 vs. 33 mM HG, 24 hViability (by CCK-8), apoptosis, caspase-3, ROS, autophagy (by LC3-I > LC3-II, mTOR, ULK1)HG increased mitochondrial membrane potential, ROS, caspase-3, apoptosis, but inhibited autophagy via mTORC1, which was rescued by rapamycin or knocking down Raptor.[57]
HUVEC,
in vitro
ECM (ScienCell, 5% FBS, ECGS)25 mM HG, 72 hApoptosis, viability, ROS, SIRT-1/mTOR/AMPK signaling, Bax, Bcl-2, PARP, caspase-3HG induced apoptosis by decreased Bcl-2, increased Bax, caspase-3, PARP cleavage, ROS, lipid peroxidation, P-mTOR (S2448), and decreased P-AMPK (S-485). All prevented by fidarestat, the aldose reductase inhibitor, via Sirt1, AMPK, and mTOR[60]
BAEC (primary cells)
In vitro
DMEM, 10% FCS5 vs. 25 mM HG, 72 h, 2% FCSROS (by ESR), DAG, PKC. HG increased ROS and PKC activity, DPI (2 h), but not LNMA/rotenone, negated ROS and the PKC increase.[64]
HUVEC,
in vitro
ECGM (Cell Applications)5.5 vs. 28 mM HG, 24 h, metabolic glucose, TCA intermediatesROS (O2.−), ER-stress (by secreted alkaline phosphatase, SAP). All intermediates induced ROS, but only glucose and glycolytic metabolites induced ER-stress. NOX and xanthine oxidases not involved[100]
PIEC (porcine ileal artery)
In vitro
DMEM, 10% FBS for growth, EBM for treatment5.5 vs. 30 mM HG, treatment, duration not indicatedROS (dihydroethidium), NO (DAF2-DA), TRPV1, UCP2 expression. Signaling: PKA, AMPK, UCP2, p22phox, eNOS. Capsaicin attenuated HG-induced endothelial dysfunction via TRPV1 activation, upregulation of UCP-2, reduced ROS, and increased NO [101]
HUVEC,
in vitro
EBM-2 (Lonza)5 vs. 25 mM HG, 24 hInsulin signaling. Insulin receptor decreased (both P-Tyr and expression), P-Akt increased, P-eNOS decreased. Erk1/2, p38 and JNK unchanged[102]
Long-term HG treatment (5–14 days), including intermittent (oscillating) protocols
HUVEC
In vitro
M199, 20% heat- inactivated FBS, ECGSConstant 5 vs. 20 mM HG vs. intermittent 5–20 mM glucose, 7–14 daysViability, G0/G1 cell cycle arrest, apoptosis (by DNA fragmentation), Bcl-2/Bax expressionBcl-2 was decreased by constant HG but unchanged by intermittent HG. Bax always increased. Apoptosis increased at >7 days and was greater in intermittent HG[103]
Rat carotid artery, in vivo, glucose infusionNone5 vs. 30 mM HG, 48 h, constant or intermittentBlood insulin, HOMA-IR, Bax, Bcl-2, caspase-3, ROS, p47phox NOX, GSH, MDA, IL-6, TNFa, ICAM-1. Apoptosis (by TUNEL assay)Oscillating or constant HG induced insulin resistance. Oscillating more than constant HG reduced GSH, increased NOX, ROS, Bax/Bcl-2, caspase-3, MDA, inflammatory cytokines, and ICAM-1 expression. Oscillating but not constant HG induced apoptosis[104]
HMEC-1 (dermal)
HAEC (Cambrex)
In vitro
MCDB 131, human serum,
EGM (Cambrex), 2% FBS, ECGS
5 vs. 20 mM HG, 7 daysApoptosis (TUNEL, cell cycle), anoikis, necrosis, adhesion, dicarbonyls/AGEs, modifications by methylglyoxal (MGO)Decreased collagen adhesion upon hyperglycemia-induced collagen IV modification by MGO, increased apoptosis of the detached cells (the attached not affected)[105]
BAEC (Clonetics) or HAEC (ATCC)
In vitro
F12, 10% FBS, ECGS5 vs. 25–30 or 44 mM HG, 3 or 7 dayseNOS dimerization and activity (by arginine to citrulline), peroxynitrite, O2.−, cGMP, PGI2 synthase (PGIS), apoptosis (by DNA fragmentation)Peroxynitrite uncoupled eNOS resulting in O2.− production instead of NO. HG increased Ca2+-induced O2.−, peroxynitrite and PGIS nitration, inhibited cGMP and PGIS acivity, increased apoptosis, which depended on SOD and LNAME[47,68]
MS-1 (the pancreatic islet cells, ATCC)
In vitro
DMEM, 5% FBS5.5 vs. 30 mM HG, 7 daysApoptosis (annexin V), iNOS expression, NO, peroxynitrite, O2.− (dihydroethidium), ROS (DCF), signaling (P-JNK)HG increased iNOS expression, ROS, O2.−, nitrotyrosine, P-JNK, caspase-3 activity, Bax/Bcl-2 ratio and apoptosis, all prevented by JNK and iNOS inhibitors [106]
HAEC (Clonetics)
In vitro
Not clearly indicated5 vs. 22 mM HG, 5 dayseNOS (by PCR/WB), NO (by Griess assay), ROS (O2.−). HG increased ROS (3-fold), eNOS (~2-fold) and NO (~40%)[107]
BAEC,
in vitro
MEM (Gibco), 0.4% FBS5 vs. 30 mM HG, 7 daysROS, PKC, AGEs (MGO), sorbitol, NF-kB. HG increased mitochondrial ROS, which induced PKC, AGEs, aldose reductase-mediated sorbitol, and NF-kB. All prevented by forced expression of UCP-1.[70]
HUVEC,
in vitro
M199, 20% heat- inactivated FBS, ECGSConstant 5 vs. 20 mM HG vs. intermittent 5–20 mM glucose, 7–14 daysSignaling: PKC, NOX, nitrotyrosine, ROS, caspase-3, mitochondrial complex II. HG > 7 days increased PKC activity, 8-OHdG, nitrotyrosine, NOX and caspase-3. All these were negated by PKC inhibitors and MnSOD mimetic. Thenoyltrifluoroacetone blocked nitrotyrosine while caspase-3 increased suggesting mitochondrial origin of ROS[65,66]
EA.hy926,
in vitro
DMEM, 10% FBS5.5 vs. 20 mM HG, 6 daysAerobic metabolism, respiration, hexokinase-1, and UCP-2. HG induced the Crabtree effect, no change in the Krebs cycle or respiratory chain. Upregulated UCP-2 and hexokinase-1[78]
SkMEC (primary cells from rat skeletal muscle)EGM (Cell Applications Inc.)5.5 vs. 11 mM HG, 3 daysCell viability (by trypan blue), mitochondrial respiration and coupling efficiency, H2O2 emission (by Amplex Ultra Red), citrate synthase activity.Cell viability 89–98%, HG lowers mitochondrial respiration and increases H2O2/O2 emission, indicative of mitochondrial dysfunction[108]
In each sub-category, the studies are arranged by their appearance to reflect the research timeline (c.f., Figure 1). The studies where the mechanisms related to apoptosis were investigated are shadowed, whereas apoptosis was not directly measured. They are shown in the end of each subcategory.
Table A2. Summary of the outcomes of FFA action on endothelial cells (experimental hyperlipidemia) in vitro.
Table A2. Summary of the outcomes of FFA action on endothelial cells (experimental hyperlipidemia) in vitro.
Cells and Study DesignCulture ConditionsFFA Type, Duration and Concentrations, Other EssentialsAssessed ParametersMajor Findings/MechanismRefs
Short-term FFA treatment (up to 2 days)
Human skin microvascular ECEGM (Clontech), ECGS0.1 mM palmitate, 4–24 hApoptosis, ROS, DNA fragmentation, [3H]-thymidine incorporationPalmitate increased ROS, DNA fragmentation, [3H]-thymidine incorporation. NAC rescued all these almost completely[109]
HUVECM199, 20% delipidated FBS, 0.4% albumin; human plasma +/− intralipid0.1–0.3 mM, 0.05 vs. 0.37 mM palmitate; 24 or 48 h; also oleate, stearate, linoleate, γ-linoleate, arachidonateApoptosis (time-, dose-, and double-bond dependent), G0/G1 cell cycle arrestUpregulation of pro-apoptotic Bak and Cdk inhibitor p21WAF-1/Cip1. Reduced expression of apoptotic inhibitor Bcl-2, NF-κB inhibitor IκBα, ET-1, and eNOS[110]
HAEC (from Bio Whittaker)EBM, 2% FBS, growth factors (Bio Whittaker)0.3 mM linoleate, 16 hROS (by DCF), ET-1, NO, eNOS, NF-kB, caspase-3, apoptosis (by DNA fragmentation)Linoleate increased ROS, ET-1, reduced NO, activated NF-kB and induced apoptosis; all rescued by UCP-2 adenoviral transduction[77]
HCAECs (coronary artery)EGM-2MV (Clonetics/Bio Whittaker)1 mM, palmitate, also oleate, stearate, palmitoleate, linoleate, 24 h, 2.5 or 5% BSA Apoptosis, NF-kB, caspase-3, cytokine expressionCaspase-3, acyl-CoA formation (triacsin-C), no effect of etomoxir. FFA (but not NEFA) activated NF-kB. Blocking NF-kB or IKK or NEFA added on the top of FFA prevented FFA-induced apoptosis[111]
HUVEC, HAEC, HREC, EPCM199, 20% delipidated FBS, 0.4% albumin0.1–0.3 mM palmitate; 24 h; also oleate, stearate, linoleate, γ-linoleate, arachidonateApoptosis, membrane rigidity, incorporation of FFA into cell membranes PUFA-induced apoptosis was mediated by c-myc in cells other than HUVEC but required caspase activation in all cells[112]
EA.hy926DMEM, 10% FBS0.5 mM palmitate, oleate, 1–28 h; 6:1 to albuminNecroptosis rather than apoptosis, autophagyPA (but not oleate) induced autophagy (by LC3-I > LC3-II), which was rescued by autophagy inhibitors (3-MA, bafilomycin A1, wortmannin)[58]
HUVECDMEM, 10% FBS0.1, 0.2, 0.3 mM; 24 or 48 h; FFA is not specifiedViability, apoptosisFFA increased inflammatory cytokine expression and NF-kB signaling (MyD88, NF-kB, IKKb), all prevented by TLR4 interference[61]
HUVECDMEM, 10% FBS, passage 20–250.3 mM palmitate, 24 hViability, autophagy (by LCI > LCII, loss of p62), cytosolic Ca2+, NO, tube formation, wound healingFFA induced autophagy, increased NOX4, P-PKCa and ROS, decreased NO, wound healing, tube formation and cell viability—all prevented by 3-MA[56]
EA.hy926DMEM, 10% FBS, 1% HAT0.36 mM oleate, 4 hROS, Ca2+, eNOS. Oleate increased ROS, decreased Ca2+ and eNOS activity in response to histamine or ATP[113]
BAEC (primary)DMEM, 10% FBS for growth, EBM for treatment0.1 mM palmitate, oleate, linoleate, 3 h in EBM (Clontech), 30 mM BSAInsulin signaling, IKK (by WB), NO (by DAF). All FFAs inhibited insulin signaling and NO production, and increased IKKb. Dominant-negative IKK reversed the effects of palmitate[114]
HAEC (primary, aortic)EGM, 10% FBS for growth, EBM for treatment0.1–1 mM palmitate, oleate, linoleate, 24 h, 2% BSAIns signaling, eNOS (by Arg > citrulline), JNK/p38, PTEN (ChIP assay). Palmitate and linoleate (but not oleate) inhibited Ins cascade and eNOS activity, increased JNK and p38, upregulated PTEN.[115]
HUVECDMEM, 10% FBS0.1 mM palmitate, 24 hNF-kB, insulin signaling (by WB), NO (by DAF). FFA increased IL-6 and TNF-α production, P-IKKb, P-p65, P-IRS (S307), and decreased P-IRS (Tyr), P-Akt, P-eNOS, NO in response to insulin[116]
HUVECRPMI-1640, 10% FBS0.1 mM palmitate, 0.5, 2 and 12 hNO (by DAF), glucose uptake, insulin signaling (by WB). FFA induced ROS, IL-6, TNF-a (all inhibited by piceatannol, resveratrol analog. FFA reduced P-IRS (Tyr), P-eNOS, NO, glucose uptake[62]
HUVECMedium 200, ECGS, ? % FBS0.25, 0.5, 0.75 mM palmitate, 6–18 hAutophagy, ER-stress, insulin signaling (by WB), glucose uptake. FFA decreased P-Akt, Glut4 exposure and glucose uptake, increased LC3-II, caspase activity, ER-stress, P-IRS (S307), P-JNK[117]
MHEC (mouse heart),
EA.hy926
DMEM, 20% FBS, ECGS
DMEM, 10% FBS
0.1–0.4 mM palmitate, 1–8 hAutophagy (by LCI > LCII), ROS, lipid droplets (by Bodipy), adhesion of leukocytes. PA induced ROS and LD formation, but impaired autophagy. Blocking CPT1 (by etomoxir) promoted, but blocking ACSL (by triacsin-C) inhibited autophagy. Metformin reduced LD and rescued autophagy, the effects blunted by silencing of AMPK[59]
Long-term FFA treatment (more than 2 days)
HUVECM199 or DMEM, 20% FBS, ECGS2 mM oleate/palmitate mix (2:1), 2% albumin, 3 daysApoptosis, insulin signalingCaspase-9 activation, increased bax-a and reduced bcl-2 expression. Decreased P-Akt (S473), P-eNOS. Apoptosis prevented by insulin (10 nM) in PI3K-dependent manner[118]
HUVECM199, 20% FBS0.75 mM oleate/palmitate mix (2:1), 72 h; 0.5, 0.75, 1 mM up to 4 daysApoptosis, proliferation FFA inhibited proliferation and induces apoptosis. The responses were partly rescued by GSK3β knockdown [119]
HUVECEGM-2mv (Lonza), 50 μM carnitine, passage 30.5–1.5 mM palmitate, up to 14 daysCell viability (by light microscopy), monolayer permeability (by ECIS-z), insulin signaling (by WB), ROS (by DCF), NO (by DAF), MDA (by WB)Palmitate dose-dependently induced cell death associated with oxidative/dicarbonyl stress, increased permeability, ROS, NO, MDA. AICAR and carnitine were overwhelmingly protective[28]
BAEC (primary)DMEM, 10% FCS0.2 mM palmitate, 24–72 h, 1% FCS, 3 daysROS (by ESR), DAG and PKC. ROS, DAG, PKC activity increased, DPI (for 2 h) negated the ROS and PKC upregulation responses[64]
SkMEC (primary cells from rat skeletal muscle)EGM (Cell Applications Inc.)1.7 or 2.2 mM palmitate (albumin concentration not indicated), 3 daysCell viability (trypan blue), mitochondrial respiration and coupling efficiency, H2O2 emission (by Amplex Ultra Red), citrate synthase activity.Cell viability 89–98%, palmitate lowers mitochondrial respiration and increases H2O2/O2 emission, indicative of mitochondrial dysfunction[108]
The reference arrangement mode is the same as in Table A1.

References

  1. Del Turco, S.; Gaggini, M.; Daniele, G.; Basta, G.; Folli, F.; Sicari, R.; Gastaldelli, A. Insulin resistance and endothelial dysfunction: A mutual relationship in cardiometabolic risk. Curr. Pharm. Des. 2013, 19, 2420–2431. [Google Scholar] [CrossRef] [PubMed]
  2. Jansson, P.A. Endothelial dysfunction in insulin resistance and type 2 diabetes. J. Intern. Med. 2007, 262, 173–183. [Google Scholar] [CrossRef]
  3. Petrie, J.R.; Guzik, T.J.; Touyz, R.M. Diabetes, Hypertension, and Cardiovascular Disease: Clinical Insights and Vascular Mechanisms. Can. J. Cardiol. 2018, 34, 575–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Jiang, H.; Zhou, Y.; Nabavi, S.M.; Sahebkar, A.; Little, P.J.; Xu, S.; Weng, J.; Ge, J. Mechanisms of Oxidized LDL-Mediated Endothelial Dysfunction and Its Consequences for the Development of Atherosclerosis. Front. Cardiovasc. Med. 2022, 9, 925923. [Google Scholar] [CrossRef] [PubMed]
  5. La Sala, L.; Prattichizzo, F.; Ceriello, A. The link between diabetes and atherosclerosis. Eur. J. Prev. Cardiol. 2019, 26, 15–24. [Google Scholar] [CrossRef] [PubMed]
  6. Laakso, M. Hyperglycemia and cardiovascular disease in type 2 diabetes. Diabetes 1999, 48, 937–942. [Google Scholar] [CrossRef]
  7. Little, P.J.; Askew, C.D.; Xu, S.; Kamato, D. Endothelial Dysfunction and Cardiovascular Disease: History and Analysis of the Clinical Utility of the Relationship. Biomedicines 2021, 9, 699. [Google Scholar] [CrossRef]
  8. Medina-Leyte, D.J.; Domínguez-Pérez, M.; Mercado, I.; Villarreal-Molina, M.T.; Jacobo-Albavera, L. Use of Human Umbilical Vein Endothelial Cells (HUVEC) as a Model to Study Cardiovascular Disease: A Review. Appl. Sci. 2020, 10, 938. [Google Scholar] [CrossRef] [Green Version]
  9. Ham, R.G. Nutritional requirements of primary cultures. a neglected problem of modern biology. In Vitro 1974, 10, 119–129. [Google Scholar] [CrossRef]
  10. Knedler, A.; Ham, R.G. Optimized medium for clonal growth of human microvascular endothelial cells with minimal serum. In Vitro Cell Dev. Biol. 1987, 23, 481–491. [Google Scholar] [CrossRef]
  11. Leopold, B.; Strutz, J.; Weiß, E.; Gindlhuber, J.; Birner-Gruenberger, R.; Hackl, H.; Appel, H.M.; Cvitic, S.; Hiden, U. Outgrowth, proliferation, viability, angiogenesis and phenotype of primary human endothelial cells in different purchasable endothelial culture media: Feed wisely. Histochem. Cell Biol. 2019, 152, 377–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Rhee, E.P.; Cheng, S.; Larson, M.G.; Walford, G.A.; Lewis, G.D.; McCabe, E.; Yang, E.; Farrell, L.; Fox, C.S.; O’Donnell, C.J.; et al. Lipid profiling identifies a triacylglycerol signature of insulin resistance and improves diabetes prediction in humans. J. Clin. Investig. 2011, 121, 1402–1411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Brownlee, M. The pathobiology of diabetic complications: A unifying mechanism. Diabetes 2005, 54, 1615–1625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Evans, J.L.; Goldfine, I.D.; Maddux, B.A.; Grodsky, G.M. Oxidative stress and stress-activated signaling pathways: A unifying hypothesis of type 2 diabetes. Endocr. Rev. 2002, 23, 599–622. [Google Scholar] [CrossRef] [Green Version]
  15. Giugliano, D.; Ceriello, A.; Paolisso, G. Oxidative stress and diabetic vascular complications. Diabetes Care 1996, 19, 257–267. [Google Scholar] [CrossRef] [PubMed]
  16. Schonfeld, P.; Wojtczak, L. Fatty acids as modulators of the cellular production of reactive oxygen species. Free Radic. Biol. Med. 2008, 45, 231–241. [Google Scholar] [CrossRef] [PubMed]
  17. Diez-Roux, G.; Lang, R.A. Macrophages induce apoptosis in normal cells in vivo. Development 1997, 124, 3633–3638. [Google Scholar] [CrossRef]
  18. Winn, R.K.; Harlan, J.M. The role of endothelial cell apoptosis in inflammatory and immune diseases. J. Thromb. Haemost. 2005, 3, 1815–1824. [Google Scholar] [CrossRef]
  19. Best, P.J.; Hasdai, D.; Sangiorgi, G.; Schwartz, R.S.; Holmes, D.R., Jr.; Simari, R.D.; Lerman, A. Apoptosis. Basic concepts and implications in coronary artery disease. Arterioscler. Thromb. Vasc. Biol. 1999, 19, 14–22. [Google Scholar] [CrossRef] [Green Version]
  20. Shah, M.S.; Brownlee, M. Molecular and Cellular Mechanisms of Cardiovascular Disorders in Diabetes. Circ. Res. 2016, 118, 1808–1829. [Google Scholar] [CrossRef]
  21. Gauthier, M.S.; O’Brien, E.L.; Bigornia, S.; Mott, M.; Cacicedo, J.M.; Xu, X.J.; Gokce, N.; Apovian, C.; Ruderman, N. Decreased AMP-activated protein kinase activity is associated with increased inflammation in visceral adipose tissue and with whole-body insulin resistance in morbidly obese humans. Biochem. Biophys. Res. Commun. 2011, 404, 382–387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Xu, X.J.; Gauthier, M.S.; Hess, D.T.; Apovian, C.M.; Cacicedo, J.M.; Gokce, N.; Farb, M.; Valentine, R.J.; Ruderman, N.B. Insulin sensitive and resistant obesity in humans: AMPK activity, oxidative stress, and depot-specific changes in gene expression in adipose tissue. J. Lipid Res. 2012, 53, 792–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ido, Y.; Carling, D.; Ruderman, N. Hyperglycemia-induced apoptosis in human umbilical vein endothelial cells: Inhibition by the AMP-activated protein kinase activation. Diabetes 2002, 51, 159–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Czabotar, P.E.; Lessene, G.; Strasser, A.; Adams, J.M. Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat. Rev. Mol. Cell Biol. 2014, 15, 49–63. [Google Scholar] [CrossRef]
  25. Hu, V.W.; Black, G.E.; Torres-Duarte, A.; Abramson, F.P. 3H-thymidine is a defective tool with which to measure rates of DNA synthesis. FASEB J. 2002, 16, 1456–1457. [Google Scholar] [CrossRef]
  26. Orlov, S.N.; Pchejetski, D.V.; Sarkissian, S.D.; Adarichev, V.; Taurin, S.; Pshezhetsky, A.V.; Tremblay, J.; Maximov, G.V.; deBlois, D.; Bennett, M.R.; et al. [3H]-thymidine labelling of DNA triggers apoptosis potentiated by E1A-adenoviral protein. Apoptosis 2003, 8, 199–208. [Google Scholar] [CrossRef]
  27. Watanabe, M.; Hitomi, M.; van der Wee, K.; Rothenberg, F.; Fisher, S.A.; Zucker, R.; Svoboda, K.K.; Goldsmith, E.C.; Heiskanen, K.M.; Nieminen, A.L. The pros and cons of apoptosis assays for use in the study of cells, tissues, and organs. Microsc. Microanal. 2002, 8, 375–391. [Google Scholar] [CrossRef] [Green Version]
  28. Samsonov, M.V.; Podkuychenko, N.V.; Khapchaev, A.Y.; Efremov, E.E.; Yanushevskaya, E.V.; Vlasik, T.N.; Lankin, V.Z.; Stafeev, I.S.; Skulachev, M.V.; Shestakova, M.V.; et al. AICAR Protects Vascular Endothelial Cells from Oxidative Injury Induced by the Long-Term Palmitate Excess. Int. J. Mol. Sci. 2021, 23, 211. [Google Scholar] [CrossRef]
  29. Arner, P.; Ryden, M. Fatty Acids, Obesity and Insulin Resistance. Obes. Facts 2015, 8, 147–155. [Google Scholar] [CrossRef]
  30. Hales, C.N.; Walker, J.B.; Garland, P.B.; Randle, P.J. Fasting Plasma Concentrations of Insulin, Non-Esterified Fatty Acids, Glycerol, and Glucose in the Early Detection of Diabetes Mellitus. Lancet 1965, 285, 65–67. [Google Scholar] [CrossRef]
  31. McQuaid, S.E.; Hodson, L.; Neville, M.J.; Dennis, A.L.; Cheeseman, J.; Humphreys, S.M.; Ruge, T.; Gilbert, M.; Fielding, B.A.; Frayn, K.N.; et al. Downregulation of adipose tissue fatty acid trafficking in obesity: A driver for ectopic fat deposition? Diabetes 2011, 60, 47–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Miles, J.M.; Park, Y.S.; Walewicz, D.; Russell-Lopez, C.; Windsor, S.; Isley, W.L.; Coppack, S.W.; Harris, W.S. Systemic and forearm triglyceride metabolism: Fate of lipoprotein lipase-generated glycerol and free fatty acids. Diabetes 2004, 53, 521–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Miles, J.M.; Nelson, R.H. Contribution of triglyceride-rich lipoproteins to plasma free fatty acids. Horm. Metab. Res. 2007, 39, 726–729. [Google Scholar] [CrossRef]
  34. Oliveira, A.F.; Cunha, D.A.; Ladriere, L.; Igoillo-Esteve, M.; Bugliani, M.; Marchetti, P.; Cnop, M. In vitro use of free fatty acids bound to albumin: A comparison of protocols. Biotechniques 2015, 58, 228–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Curry, S. Plasma albumin as a fatty acid carrier. Adv. Mol. Cell Biol. 2004, 33, 29–46. [Google Scholar] [CrossRef]
  36. Stafeev, I.S.; Vorotnikov, A.V.; Ratner, E.I.; Menshikov, M.Y.; Parfyonova, Y.V. Latent Inflammation and Insulin Resistance in Adipose Tissue. Int. J. Endocrinol. 2017, 2017, 5076732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Hulsmann, W.C.; Dubelaar, M.L. Aspects of fatty acid metabolism in vascular endothelial cells. Biochimie 1988, 70, 681–686. [Google Scholar] [CrossRef]
  38. Hulsmann, W.C.; Dubelaar, M.L. Carnitine requirement of vascular endothelial and smooth muscle cells in imminent ischemia. Mol. Cell Biochem. 1992, 116, 125–129. [Google Scholar] [CrossRef]
  39. Dagher, Z.; Ruderman, N.; Tornheim, K.; Ido, Y. The effect of AMP-activated protein kinase and its activator AICAR on the metabolism of human umbilical vein endothelial cells. Biochem. Biophys. Res. Commun. 1999, 265, 112–115. [Google Scholar] [CrossRef]
  40. Dagher, Z.; Ruderman, N.; Tornheim, K.; Ido, Y. Acute regulation of fatty acid oxidation and amp-activated protein kinase in human umbilical vein endothelial cells. Circ. Res. 2001, 88, 1276–1282. [Google Scholar] [CrossRef]
  41. Cacicedo, J.M.; Yagihashi, N.; Keaney, J.F., Jr.; Ruderman, N.B.; Ido, Y. AMPK inhibits fatty acid-induced increases in NF-kappaB transactivation in cultured human umbilical vein endothelial cells. Biochem. Biophys. Res. Commun. 2004, 324, 1204–1209. [Google Scholar] [CrossRef] [PubMed]
  42. Ruderman, N.B.; Cacicedo, J.M.; Itani, S.; Yagihashi, N.; Saha, A.K.; Ye, J.M.; Chen, K.; Zou, M.; Carling, D.; Boden, G.; et al. Malonyl-CoA and AMP-activated protein kinase (AMPK): Possible links between insulin resistance in muscle and early endothelial cell damage in diabetes. Biochem. Soc. Trans. 2003, 31, 202–206. [Google Scholar] [CrossRef] [PubMed]
  43. Muoio, D.M.; Noland, R.C.; Kovalik, J.P.; Seiler, S.E.; Davies, M.N.; DeBalsi, K.L.; Ilkayeva, O.R.; Stevens, R.D.; Kheterpal, I.; Zhang, J.; et al. Muscle-specific deletion of carnitine acetyltransferase compromises glucose tolerance and metabolic flexibility. Cell. Metab. 2012, 15, 764–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Ruderman, N.B.; Carling, D.; Prentki, M.; Cacicedo, J.M. AMPK, insulin resistance, and the metabolic syndrome. J. Clin. Investig. 2013, 123, 2764–2772. [Google Scholar] [CrossRef] [Green Version]
  45. Petersen, M.C.; Shulman, G.I. Mechanisms of Insulin Action and Insulin Resistance. Physiol. Rev. 2018, 98, 2133–2223. [Google Scholar] [CrossRef] [Green Version]
  46. McGarry, J.D. Banting lecture 2001: Dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Diabetes 2002, 51, 7–18. [Google Scholar] [CrossRef] [Green Version]
  47. Zou, M.H.; Shi, C.; Cohen, R.A. High glucose via peroxynitrite causes tyrosine nitration and inactivation of prostacyclin synthase that is associated with thromboxane/prostaglandin H(2) receptor–mediated apoptosis and adhesion molecule expression in cultured human aortic endothelial cells. Diabetes 2002, 51, 198–203. [Google Scholar] [CrossRef] [Green Version]
  48. Yao, H.; Gong, J.; Peterson, A.L.; Lu, X.; Zhang, P.; Dennery, P.A. Fatty Acid Oxidation Protects against Hyperoxia-induced Endothelial Cell Apoptosis and Lung Injury in Neonatal Mice. Am. J. Respir. Cell Mol. Biol. 2019, 60, 667–677. [Google Scholar] [CrossRef]
  49. Faergeman, N.J.; Knudsen, J. Role of long-chain fatty acyl-CoA esters in the regulation of metabolism and in cell signalling. Biochem. J. 1997, 323, 1–12. [Google Scholar] [CrossRef] [Green Version]
  50. Karlic, H.; Lohninger, A. Supplementation of L-carnitine in athletes: Does it make sense? Nutrition 2004, 20, 709–715. [Google Scholar] [CrossRef]
  51. McGarry, J.D.; Mannaerts, G.P.; Foster, D.W. A possible role for malonyl-CoA in the regulation of hepatic fatty acid oxidation and ketogenesis. J. Clin. Investig. 1977, 60, 265–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Zammit, V.A. The malonyl-CoA-long-chain acyl-CoA axis in the maintenance of mammalian cell function. Biochem. J. 1999, 343, 505–515. [Google Scholar] [CrossRef] [PubMed]
  53. Ruderman, N.B.; Saha, A.K. Metabolic syndrome: Adenosine monophosphate-activated protein kinase and malonyl coenzyme A. Obesity 2006, 14, 25S–33S. [Google Scholar] [CrossRef] [Green Version]
  54. Saggerson, D. Malonyl-CoA, a key signaling molecule in mammalian cells. Annu. Rev. Nutr. 2008, 28, 253–272. [Google Scholar] [CrossRef]
  55. Park, H.; Kaushik, V.K.; Constant, S.; Prentki, M.; Przybytkowski, E.; Ruderman, N.B.; Saha, A.K. Coordinate regulation of malonyl-CoA decarboxylase, sn-glycerol-3-phosphate acyltransferase, and acetyl-CoA carboxylase by AMP-activated protein kinase in rat tissues in response to exercise. J. Biol. Chem. 2002, 277, 32571–32577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Chen, P.; Liu, H.; Xiang, H.; Zhou, J.; Zeng, Z.; Chen, R.; Zhao, S.; Xiao, J.; Shu, Z.; Chen, S.; et al. Palmitic acid-induced autophagy increases reactive oxygen species via the Ca(2+)/PKCalpha/NOX4 pathway and impairs endothelial function in human umbilical vein endothelial cells. Exp. Ther. Med. 2019, 17, 2425–2432. [Google Scholar] [CrossRef] [Green Version]
  57. Fan, W.; Han, D.; Sun, Z.; Ma, S.; Gao, L.; Chen, J.; Li, X.; Li, X.; Fan, M.; Li, C.; et al. Endothelial deletion of mTORC1 protects against hindlimb ischemia in diabetic mice via activation of autophagy, attenuation of oxidative stress and alleviation of inflammation. Free Radic. Biol. Med. 2017, 108, 725–740. [Google Scholar] [CrossRef]
  58. Khan, M.J.; Rizwan Alam, M.; Waldeck-Weiermair, M.; Karsten, F.; Groschner, L.; Riederer, M.; Hallstrom, S.; Rockenfeller, P.; Konya, V.; Heinemann, A.; et al. Inhibition of autophagy rescues palmitic acid-induced necroptosis of endothelial cells. J. Biol. Chem. 2012, 287, 21110–21120. [Google Scholar] [CrossRef] [Green Version]
  59. Kim, H.S.; Ren, G.; Kim, T.; Bhatnagar, S.; Yang, Q.; Bahk, Y.Y.; Kim, J.A. Metformin reduces saturated fatty acid-induced lipid accumulation and inflammatory response by restoration of autophagic flux in endothelial cells. Sci. Rep. 2020, 10, 13523. [Google Scholar] [CrossRef]
  60. Pal, P.B.; Sonowal, H.; Shukla, K.; Srivastava, S.K.; Ramana, K.V. Aldose reductase regulates hyperglycemia-induced HUVEC death via SIRT1/AMPK-alpha1/mTOR pathway. J. Mol. Endocrinol. 2019, 63, 11–25. [Google Scholar] [CrossRef]
  61. Chen, L.; Yu, C.X.; Song, B.; Cai, W.; Liu, C.; Guan, Q.B. Free fatty acids mediates human umbilical vein endothelial cells inflammation through toll-like receptor-4. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 2421–2431. [Google Scholar] [CrossRef] [PubMed]
  62. Jeong, S.O.; Son, Y.; Lee, J.H.; Cheong, Y.K.; Park, S.H.; Chung, H.T.; Pae, H.O. Resveratrol analog piceatannol restores the palmitic acid-induced impairment of insulin signaling and production of endothelial nitric oxide via activation of anti-inflammatory and antioxidative heme oxygenase-1 in human endothelial cells. Mol. Med. Rep. 2015, 12, 937–944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Colombo, S.L.; Moncada, S. AMPKalpha1 regulates the antioxidant status of vascular endothelial cells. Biochem. J. 2009, 421, 163–169. [Google Scholar] [CrossRef] [Green Version]
  64. 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] [Green Version]
  65. Piconi, L.; Quagliaro, L.; Assaloni, R.; Da Ros, R.; Maier, A.; Zuodar, G.; Ceriello, A. Constant and intermittent high glucose enhances endothelial cell apoptosis through mitochondrial superoxide overproduction. Diabetes/Metab. Res. Rev. 2006, 22, 198–203. [Google Scholar] [CrossRef] [PubMed]
  66. Quagliaro, L.; Piconi, L.; Assaloni, R.; Martinelli, L.; Motz, E.; Ceriello, A. Intermittent high glucose enhances apoptosis related to oxidative stress in human umbilical vein endothelial cells: The role of protein kinase C and NAD(P)H-oxidase activation. Diabetes 2003, 52, 2795–2804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Quintero, M.; Colombo, S.L.; Godfrey, A.; Moncada, S. Mitochondria as signaling organelles in the vascular endothelium. Proc. Natl. Acad. Sci. USA 2006, 103, 5379–5384. [Google Scholar] [CrossRef] [Green Version]
  68. Zou, M.H.; Shi, C.; Cohen, R.A. Oxidation of the zinc-thiolate complex and uncoupling of endothelial nitric oxide synthase by peroxynitrite. J. Clin. Investig. 2002, 109, 817–826. [Google Scholar] [CrossRef]
  69. Green, K.; Brand, M.D.; Murphy, M.P. Prevention of mitochondrial oxidative damage as a therapeutic strategy in diabetes. Diabetes 2004, 53, S110–S118. [Google Scholar] [CrossRef] [Green Version]
  70. 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]
  71. Korshunov, S.S.; Skulachev, V.P.; Starkov, A.A. High protonic potential actuates a mechanism of production of reactive oxygen species in mitochondria. FEBS Lett. 1997, 416, 15–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Skulachev, V.P. Bioenergetic aspects of apoptosis, necrosis and mitoptosis. Apoptosis 2006, 11, 473–485. [Google Scholar] [CrossRef] [PubMed]
  73. Sreedhar, A.; Zhao, Y. Uncoupling protein 2 and metabolic diseases. Mitochondrion 2017, 34, 135–140. [Google Scholar] [CrossRef] [Green Version]
  74. Jezek, P.; Holendova, B.; Garlid, K.D.; Jaburek, M. Mitochondrial Uncoupling Proteins: Subtle Regulators of Cellular Redox Signaling. Antioxid. Redox. Signal. 2018, 29, 667–714. [Google Scholar] [CrossRef] [Green Version]
  75. Pecqueur, C.; Alves-Guerra, C.; Ricquier, D.; Bouillaud, F. UCP2, a metabolic sensor coupling glucose oxidation to mitochondrial metabolism? IUBMB Life 2009, 61, 762–767. [Google Scholar] [CrossRef] [PubMed]
  76. Crichton, P.G.; Lee, Y.; Kunji, E.R. The molecular features of uncoupling protein 1 support a conventional mitochondrial carrier-like mechanism. Biochimie 2017, 134, 35–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Lee, K.U.; Lee, I.K.; Han, J.; Song, D.K.; Kim, Y.M.; Song, H.S.; Kim, H.S.; Lee, W.J.; Koh, E.H.; Song, K.H.; et al. Effects of recombinant adenovirus-mediated uncoupling protein 2 overexpression on endothelial function and apoptosis. Circ. Res. 2005, 96, 1200–1207. [Google Scholar] [CrossRef] [Green Version]
  78. Koziel, A.; Woyda-Ploszczyca, A.; Kicinska, A.; Jarmuszkiewicz, W. The influence of high glucose on the aerobic metabolism of endothelial EA.hy926 cells. Pflügers Arch. Eur. J. Physiol. 2012, 464, 657–669. [Google Scholar] [CrossRef] [Green Version]
  79. Schulz, E.; Dopheide, J.; Schuhmacher, S.; Thomas, S.R.; Chen, K.; Daiber, A.; Wenzel, P.; Munzel, T.; Keaney, J.F., Jr. Suppression of the JNK pathway by induction of a metabolic stress response prevents vascular injury and dysfunction. Circulation 2008, 118, 1347–1357. [Google Scholar] [CrossRef]
  80. Xie, Z.; Zhang, J.; Wu, J.; Viollet, B.; Zou, M.H. Upregulation of mitochondrial uncoupling protein-2 by the AMP-activated protein kinase in endothelial cells attenuates oxidative stress in diabetes. Diabetes 2008, 57, 3222–3230. [Google Scholar] [CrossRef]
  81. Zou, M.H.; Wu, Y. AMP-activated protein kinase activation as a strategy for protecting vascular endothelial function. Clin. Exp. Pharmacol. Physiol. 2008, 35, 535–545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Skulachev, V.P. Role of uncoupled and non-coupled oxidations in maintenance of safely low levels of oxygen and its one-electron reductants. Q. Rev. Biophys. 1996, 29, 169–202. [Google Scholar] [CrossRef] [PubMed]
  83. da-Silva, W.S.; Gomez-Puyou, A.; de Gomez-Puyou, M.T.; Moreno-Sanchez, R.; De Felice, F.G.; de Meis, L.; Oliveira, M.F.; Galina, A. Mitochondrial bound hexokinase activity as a preventive antioxidant defense: Steady-state ADP formation as a regulatory mechanism of membrane potential and reactive oxygen species generation in mitochondria. J. Biol. Chem. 2004, 279, 39846–39855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Meyer, L.E.; Machado, L.B.; Santiago, A.P.; da-Silva, W.S.; De Felice, F.G.; Holub, O.; Oliveira, M.F.; Galina, A. Mitochondrial creatine kinase activity prevents reactive oxygen species generation: Antioxidant role of mitochondrial kinase-dependent ADP re-cycling activity. J. Biol. Chem. 2006, 281, 37361–37371. [Google Scholar] [CrossRef] [Green Version]
  85. Vyssokikh, M.Y.; Holtze, S.; Averina, O.A.; Lyamzaev, K.G.; Panteleeva, A.A.; Marey, M.V.; Zinovkin, R.A.; Severin, F.F.; Skulachev, M.V.; Fasel, N.; et al. Mild depolarization of the inner mitochondrial membrane is a crucial component of an anti-aging program. Proc. Natl. Acad. Sci. USA 2020, 117, 6491–6501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Clarke, H.; Hickner, R.C.; Ormsbee, M.J. The Potential Role of Creatine in Vascular Health. Nutrients 2021, 13, 857. [Google Scholar] [CrossRef]
  87. Decking, U.K.; Alves, C.; Wallimann, T.; Wyss, M.; Schrader, J. Functional aspects of creatine kinase isoenzymes in endothelial cells. Am. J. Physiol. Cell. Physiol. 2001, 281, C320–C328. [Google Scholar] [CrossRef] [Green Version]
  88. Nomura, A.; Zhang, M.; Sakamoto, T.; Ishii, Y.; Morishima, Y.; Mochizuki, M.; Kimura, T.; Uchida, Y.; Sekizawa, K. Anti-inflammatory activity of creatine supplementation in endothelial cells in vitro. Br. J. Pharmacol. 2003, 139, 715–720. [Google Scholar] [CrossRef] [Green Version]
  89. Barbieri, E.; Guescini, M.; Calcabrini, C.; Vallorani, L.; Diaz, A.R.; Fimognari, C.; Canonico, B.; Luchetti, F.; Papa, S.; Battistelli, M.; et al. Creatine Prevents the Structural and Functional Damage to Mitochondria in Myogenic, Oxidatively Stressed C2C12 Cells and Restores Their Differentiation Capacity. Oxid. Med. Cell. Longev. 2016, 2016, 5152029. [Google Scholar] [CrossRef]
  90. Sestili, P.; Martinelli, C.; Bravi, G.; Piccoli, G.; Curci, R.; Battistelli, M.; Falcieri, E.; Agostini, D.; Gioacchini, A.M.; Stocchi, V. Creatine supplementation affords cytoprotection in oxidatively injured cultured mammalian cells via direct antioxidant activity. Free Radic. Biol. Med. 2006, 40, 837–849. [Google Scholar] [CrossRef]
  91. Baumgartner-Parzer, S.M.; Wagner, L.; Pettermann, M.; Grillari, J.; Gessl, A.; Waldhausl, W. High-glucose—Triggered apoptosis in cultured endothelial cells. Diabetes 1995, 44, 1323–1327. [Google Scholar] [CrossRef]
  92. Du, X.L.; Sui, G.Z.; Stockklauser-Farber, K.; Weiss, J.; Zink, S.; Schwippert, B.; Wu, Q.X.; Tschope, D.; Rosen, P. Induction of apoptosis by high proinsulin and glucose in cultured human umbilical vein endothelial cells is mediated by reactive oxygen species. Diabetologia 1998, 41, 249–256. [Google Scholar] [CrossRef] [Green Version]
  93. Wu, Q.D.; Wang, J.H.; Fennessy, F.; Redmond, H.P.; Bouchier-Hayes, D. Taurine prevents high-glucose-induced human vascular endothelial cell apoptosis. Am. J. Physiol. Cell Physiol. 1999, 277, C1229-1238. [Google Scholar] [CrossRef]
  94. Ho, F.M.; Lin, W.W.; Chen, B.C.; Chao, C.M.; Yang, C.R.; Lin, L.Y.; Lai, C.C.; Liu, S.H.; Liau, C.S. High glucose-induced apoptosis in human vascular endothelial cells is mediated through NF-kappaB and c-Jun NH2-terminal kinase pathway and prevented by PI3K/Akt/eNOS pathway. Cell Signal. 2006, 18, 391–399. [Google Scholar] [CrossRef]
  95. Ho, F.M.; Liu, S.H.; Liau, C.S.; Huang, P.J.; Lin-Shiau, S.Y. High glucose-induced apoptosis in human endothelial cells is mediated by sequential activations of c-Jun NH(2)-terminal kinase and caspase-3. Circulation 2000, 101, 2618–2624. [Google Scholar] [CrossRef] [Green Version]
  96. Ho, F.M.; Liu, S.H.; Liau, C.S.; Huang, P.J.; Shiah, S.G.; Lin-Shiau, S.Y. Nitric oxide prevents apoptosis of human endothelial cells from high glucose exposure during early stage. J. Cell. Biochem. 1999, 75, 258–263. [Google Scholar] [CrossRef]
  97. Zhang, Z.; Apse, K.; Pang, J.; Stanton, R.C. High glucose inhibits glucose-6-phosphate dehydrogenase via cAMP in aortic endothelial cells. J. Biol. Chem. 2000, 275, 40042–40047. [Google Scholar] [CrossRef] [Green Version]
  98. Recchioni, R.; Marcheselli, F.; Moroni, F.; Pieri, C. Apoptosis in human aortic endothelial cells induced by hyperglycemic condition involves mitochondrial depolarization and is prevented by N-acetyl-L-cysteine. Metabolism 2002, 51, 1384–1388. [Google Scholar] [CrossRef]
  99. Sheu, M.L.; Ho, F.M.; Yang, R.S.; Chao, K.F.; Lin, W.W.; Lin-Shiau, S.Y.; Liu, S.H. High glucose induces human endothelial cell apoptosis through a phosphoinositide 3-kinase-regulated cyclooxygenase-2 pathway. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 539–545. [Google Scholar] [CrossRef] [Green Version]
  100. Sheikh-Ali, M.; Sultan, S.; Alamir, A.R.; Haas, M.J.; Mooradian, A.D. Hyperglycemia-induced endoplasmic reticulum stress in endothelial cells. Nutrition 2010, 26, 1146–1150. [Google Scholar] [CrossRef]
  101. Sun, J.; Pu, Y.; Wang, P.; Chen, S.; Zhao, Y.; Liu, C.; Shang, Q.; Zhu, Z.; Liu, D. TRPV1-mediated UCP2 upregulation ameliorates hyperglycemia-induced endothelial dysfunction. Cardiovasc. Diabetol. 2013, 12, 69. [Google Scholar] [CrossRef] [Green Version]
  102. De Nigris, V.; Pujadas, G.; La Sala, L.; Testa, R.; Genovese, S.; Ceriello, A. Short-term high glucose exposure impairs insulin signaling in endothelial cells. Cardiovasc. Diabetol. 2015, 14, 114. [Google Scholar] [CrossRef] [Green Version]
  103. Risso, A.; Mercuri, F.; Quagliaro, L.; Damante, G.; Ceriello, A. Intermittent high glucose enhances apoptosis in human umbilical vein endothelial cells in culture. Am. J. Physiol. Endocrinol. Metab. 2001, 281, E924–E930. [Google Scholar] [CrossRef]
  104. Wu, N.; Shen, H.; Liu, H.; Wang, Y.; Bai, Y.; Han, P. Acute blood glucose fluctuation enhances rat aorta endothelial cell apoptosis, oxidative stress and pro-inflammatory cytokine expression in vivo. Cardiovasc. Diabetol. 2016, 15, 109. [Google Scholar] [CrossRef] [Green Version]
  105. Dobler, D.; Ahmed, N.; Song, L.; Eboigbodin, K.E.; Thornalley, P.J. Increased dicarbonyl metabolism in endothelial cells in hyperglycemia induces anoikis and impairs angiogenesis by RGD and GFOGER motif modification. Diabetes 2006, 55, 1961–1969. [Google Scholar] [CrossRef] [Green Version]
  106. Gong, L.; Liu, F.Q.; Wang, J.; Wang, X.P.; Hou, X.G.; Sun, Y.; Qin, W.D.; Wei, S.J.; Zhang, Y.; Chen, L.; et al. Hyperglycemia induces apoptosis of pancreatic islet endothelial cells via reactive nitrogen species-mediated Jun N-terminal kinase activation. Biochim. Biophys. Acta 2011, 1813, 1211–1219. [Google Scholar] [CrossRef] [Green Version]
  107. Cosentino, F.; Hishikawa, K.; Katusic, Z.S.; Luscher, T.F. High glucose increases nitric oxide synthase expression and superoxide anion generation in human aortic endothelial cells. Circulation 1997, 96, 25–28. [Google Scholar] [CrossRef]
  108. Hansen, C.; Olsen, K.; Pilegaard, H.; Bangsbo, J.; Gliemann, L.; Hellsten, Y. High metabolic substrate load induces mitochondrial dysfunction in rat skeletal muscle microvascular endothelial cells. Physiol. Rep. 2021, 9, e14855. [Google Scholar] [CrossRef]
  109. Yamagishi, S.; Okamoto, T.; Amano, S.; Inagaki, Y.; Koga, K.; Koga, M.; Choei, H.; Sasaki, N.; Kikuchi, S.; Takeuchi, M.; et al. Palmitate-induced apoptosis of microvascular endothelial cells and pericytes. Mol. Med. 2002, 8, 179–184. [Google Scholar] [CrossRef] [Green Version]
  110. Artwohl, M.; Roden, M.; Waldhausl, W.; Freudenthaler, A.; Baumgartner-Parzer, S.M. Free fatty acids trigger apoptosis and inhibit cell cycle progression in human vascular endothelial cells. FASEB J. 2004, 18, 146–148. [Google Scholar] [CrossRef]
  111. Staiger, K.; Staiger, H.; Weigert, C.; Haas, C.; Haring, H.U.; Kellerer, M. Saturated, but not unsaturated, fatty acids induce apoptosis of human coronary artery endothelial cells via nuclear factor-kappaB activation. Diabetes 2006, 55, 3121–3126. [Google Scholar] [CrossRef]
  112. Artwohl, M.; Lindenmair, A.; Sexl, V.; Maier, C.; Rainer, G.; Freudenthaler, A.; Huttary, N.; Wolzt, M.; Nowotny, P.; Luger, A.; et al. Different mechanisms of saturated versus polyunsaturated FFA-induced apoptosis in human endothelial cells. J. Lipid Res. 2008, 49, 2627–2640. [Google Scholar] [CrossRef] [Green Version]
  113. Esenabhalu, V.E.; Schaeffer, G.; Graier, W.F. Free fatty acid overload attenuates Ca2+ signaling and NO production in endothelial cells. Antioxid. Redox Signal. 2003, 5, 147–153. [Google Scholar] [CrossRef]
  114. Kim, F.; Tysseling, K.A.; Rice, J.; Pham, M.; Haji, L.; Gallis, B.M.; Baas, A.S.; Paramsothy, P.; Giachelli, C.M.; Corson, M.A.; et al. Free fatty acid impairment of nitric oxide production in endothelial cells is mediated by IKKbeta. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 989–994. [Google Scholar] [CrossRef] [Green Version]
  115. Wang, X.L.; Zhang, L.; Youker, K.; Zhang, M.X.; Wang, J.; LeMaire, S.A.; Coselli, J.S.; Shen, Y.H. Free fatty acids inhibit insulin signaling-stimulated endothelial nitric oxide synthase activation through upregulating PTEN or inhibiting Akt kinase. Diabetes 2006, 55, 2301–2310. [Google Scholar] [CrossRef] [Green Version]
  116. Liu, K.; Zhao, W.; Gao, X.; Huang, F.; Kou, J.; Liu, B. Diosgenin ameliorates palmitate-induced endothelial dysfunction and insulin resistance via blocking IKKbeta and IRS-1 pathways. Atherosclerosis 2012, 223, 350–358. [Google Scholar] [CrossRef]
  117. Ye, M.; Qiu, H.; Cao, Y.; Zhang, M.; Mi, Y.; Yu, J.; Wang, C. Curcumin Improves Palmitate-Induced Insulin Resistance in Human Umbilical Vein Endothelial Cells by Maintaining Proteostasis in Endoplasmic Reticulum. Front. Pharmacol. 2017, 8, 148. [Google Scholar] [CrossRef] [Green Version]
  118. Piro, S.; Spampinato, D.; Spadaro, L.; Oliveri, C.E.; Purrello, F.; Rabuazzo, A.M. Direct apoptotic effects of free fatty acids on human endothelial cells. Nutr. Metab. Cardiovasc. Dis. 2008, 18, 96–104. [Google Scholar] [CrossRef]
  119. Zhu, P.; Chen, G.; You, T.; Yao, J.; Jiang, Q.; Lin, X.; Shen, X.; Qiao, Y.; Lin, L. High FFA-induced proliferation and apoptosis in human umbilical vein endothelial cell partly through Wnt/beta-catenin signal pathway. Mol. Cell. Biochem. 2010, 338, 123–131. [Google Scholar] [CrossRef]
Figure 1. Schematic view of research progress using endothelial cell-based models of hyperglycemia (high glucose, top) or hyperlipidemia (palmitate, underneath). The legends next to the years depict major advances in identifying the key detrimental events and molecular players further detailed in the text. Abbreviations: AMPK, AMP-activated protein kinase; eNOS, endothelial NO-synthase; FAO, fatty acid oxidation; mTOR, mammalian target of rapamycin; NO, nitric oxide; ROS, reactive oxygen species.
Figure 1. Schematic view of research progress using endothelial cell-based models of hyperglycemia (high glucose, top) or hyperlipidemia (palmitate, underneath). The legends next to the years depict major advances in identifying the key detrimental events and molecular players further detailed in the text. Abbreviations: AMPK, AMP-activated protein kinase; eNOS, endothelial NO-synthase; FAO, fatty acid oxidation; mTOR, mammalian target of rapamycin; NO, nitric oxide; ROS, reactive oxygen species.
Biomedicines 10 03181 g001
Figure 2. Endothelial metabolic wires altered by hyperlipidemia and hyperglycemia. The major pathways are indicated as grey boxes. Molecular actors in main focus are marked in red. The scheme embraces cytosolic carbohydrate metabolism (top left) and fatty acid turnover (top right); utilization of these substrates in mitochondria for energy production is boxed underneath. Abbreviations: ACC, acetyl-CoA carboxylase; AcCoA, acetyl-coenzyme A; ADP, adenosine diphosphate; AMPK, AMP-activated protein kinase; ATP, adenosine triphosphate; Carn, carnitine; CK-mt, mitochondrial isoform of creatine kinase; CPT-1, carnitine-palmitoyl transferase; CrP, creatine phosphate; DAG, diacylglycerol; DHAP, dihydroxyacetone phosphate; ETC, electron transport chain of mitochondria; F1,6PP, fructose 1,6 biphosphate; FADH2, flavin adenine dinucleotide reduced; FAO, fatty acid oxidation; FFA, free fatty acid; G3P, glyceraldehyde 3-phosphate; G6P, glucose-6-phosphate; HK I/II, hexokinase I/II isoform; LCACoA, long-chain acyl-coenzyme A; MAG, monoacylglycerol; MalCoA, malonyl-coenzyme A; MCD, malonyl-CoA decarboxylase; NADH, nicotinamide adenine nucleotide reduced; NADPH, nicotinamide adenine nucleotide phosphate reduced; NOX, NADPH-oxidases; OxPhos, oxidative phosphorylation; PKC, protein kinase C; Q, coenzyme Q, ubiquinone; ROS, reactive oxygen species; TAG, triacylglycerol; UCP-2, uncoupling protein 2; β-Ox, β-oxidation of fatty acids; ΔμH+, membrane electrochemical potential; and I, II, III, IV, complexes I–IV of ETC.
Figure 2. Endothelial metabolic wires altered by hyperlipidemia and hyperglycemia. The major pathways are indicated as grey boxes. Molecular actors in main focus are marked in red. The scheme embraces cytosolic carbohydrate metabolism (top left) and fatty acid turnover (top right); utilization of these substrates in mitochondria for energy production is boxed underneath. Abbreviations: ACC, acetyl-CoA carboxylase; AcCoA, acetyl-coenzyme A; ADP, adenosine diphosphate; AMPK, AMP-activated protein kinase; ATP, adenosine triphosphate; Carn, carnitine; CK-mt, mitochondrial isoform of creatine kinase; CPT-1, carnitine-palmitoyl transferase; CrP, creatine phosphate; DAG, diacylglycerol; DHAP, dihydroxyacetone phosphate; ETC, electron transport chain of mitochondria; F1,6PP, fructose 1,6 biphosphate; FADH2, flavin adenine dinucleotide reduced; FAO, fatty acid oxidation; FFA, free fatty acid; G3P, glyceraldehyde 3-phosphate; G6P, glucose-6-phosphate; HK I/II, hexokinase I/II isoform; LCACoA, long-chain acyl-coenzyme A; MAG, monoacylglycerol; MalCoA, malonyl-coenzyme A; MCD, malonyl-CoA decarboxylase; NADH, nicotinamide adenine nucleotide reduced; NADPH, nicotinamide adenine nucleotide phosphate reduced; NOX, NADPH-oxidases; OxPhos, oxidative phosphorylation; PKC, protein kinase C; Q, coenzyme Q, ubiquinone; ROS, reactive oxygen species; TAG, triacylglycerol; UCP-2, uncoupling protein 2; β-Ox, β-oxidation of fatty acids; ΔμH+, membrane electrochemical potential; and I, II, III, IV, complexes I–IV of ETC.
Biomedicines 10 03181 g002
Table 1. Effects of carnitine on endothelial cells in vitro.
Table 1. Effects of carnitine on endothelial cells in vitro.
Cells Culture ConditionsFFA Duration and Concentrations, Other EssentialsAssessed ParametersMajor Findings/MechanismReference
HUAEC
(arterial)
M199, 10% FCS, 10% HS, ECGS, passage 2–40.5 mM oleate, 50 μM carnitineFAO, ATP generationCarnitine stimulates FAO and ATP production by ~2.5-fold [37]
HUVECEBM-2 (Clonetics), 50 μM carnitine (in metabolic studies), passage 3–50.11 mM palmitate in metabolic studies; 0.2–2 mM AICAR, 0.5–2 hFAO vs. Glucose metabolismIn the presence of carnitine, AMPK activation by AICAR reverts energy production from glycolysis to FAO[39]
HUVECEBM-2 (Clonetics), 50 μM carnitine, passage 3–50, 5, or 30 mM glucose, 2 h; 0.1 mM palmitate, 24 h in metabolic studiesFAO vs. Glucose metabolismAICAR reduces malonyl-CoA and glycolysis, but increases FAO ~3-fold only in the presence of carnitine[40]
HUVECEGM-2 (Clonetics) (for cell growth); M199, 10% FBS (for assays); 50 μM carnitine (in metabolic studies), passage 5–65 vs. 30 mM glucose, 24–72 h, 1 mM AICAR, 24 h; 0.1 mM palmitate, 24 h in metabolic studiesApoptosis (TUNEL), caspase-3, P-Akt (S473, WB), FAO vs. glucose oxidation, ATP, DAG, ceramide, malonyl-CoAAICAR or AMPK forced expression inhibits apoptosis, decreases malonyl-CoA and DAG, increases FAO and P-Akt, reduces lactate, pyruvate, and glucose oxidation[23]
HUVECEBM-2 (Cambrex), M199, 5% FBS, 50 μM carnitine with FFA, passage 4–65 vs. 25 mM glucose, 24 h, 0.4 mM palmitate, 16–24 h NF-kB, VCAM-1 expressionPalmitate (but not HG) increases, but AICAR decreases both NF-kB activation and VCAM-1 expression[41]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Vorotnikov, A.V.; Khapchaev, A.Y.; Nickashin, A.V.; Shirinsky, V.P. In Vitro Modeling of Diabetes Impact on Vascular Endothelium: Are Essentials Engaged to Tune Metabolism? Biomedicines 2022, 10, 3181. https://doi.org/10.3390/biomedicines10123181

AMA Style

Vorotnikov AV, Khapchaev AY, Nickashin AV, Shirinsky VP. In Vitro Modeling of Diabetes Impact on Vascular Endothelium: Are Essentials Engaged to Tune Metabolism? Biomedicines. 2022; 10(12):3181. https://doi.org/10.3390/biomedicines10123181

Chicago/Turabian Style

Vorotnikov, Alexander V., Asker Y. Khapchaev, Alexey V. Nickashin, and Vladimir P. Shirinsky. 2022. "In Vitro Modeling of Diabetes Impact on Vascular Endothelium: Are Essentials Engaged to Tune Metabolism?" Biomedicines 10, no. 12: 3181. https://doi.org/10.3390/biomedicines10123181

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