Next Article in Journal
Risk Factors and Cellular Differences in Heart Failure: The Key Role of Sex Hormones
Next Article in Special Issue
Diagnostic Accuracy of Hand-Held Fundus Camera and Artificial Intelligence in Diabetic Retinopathy Screening
Previous Article in Journal
Transient Expression in HEK-293 Cells in Suspension Culture as a Rapid and Powerful Tool: SARS-CoV-2 N and Chimeric SARS-CoV-2N-CD154 Proteins as a Case Study
Previous Article in Special Issue
Advances and Perspectives in Relation to the Molecular Basis of Diabetic Retinopathy—A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of Modern Anti-Diabetic Treatment on Endothelial Progenitor Cells

by
Velimir Altabas
1,2,
Jelena Marinković Radošević
1,
Lucija Špoljarec
1,
Stella Uremović
3 and
Tomislav Bulum
1,4,*
1
Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
2
School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
3
Srebrnjak Children’s Hospital, 10000 Zagreb, Croatia
4
Vuk Vrhovac University Clinic for Diabetes, Endocrinology and Metabolic Diseases, Merkur University Hospital, 10000 Zagreb, Croatia
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(11), 3051; https://doi.org/10.3390/biomedicines11113051
Submission received: 7 October 2023 / Revised: 8 November 2023 / Accepted: 9 November 2023 / Published: 14 November 2023
(This article belongs to the Special Issue Molecular Research and Recent Advances in Diabetic Retinopathy)

Abstract

:
Diabetes is one of the leading chronic diseases globally with a significant impact on mortality. This condition is associated with chronic microvascular and macrovascular complications caused by vascular damage. Recently, endothelial progenitor cells (EPCs) raised interest due to their regenerative properties. EPCs are mononuclear cells that are derived from different tissues. Circulating EPCs contribute to regenerating the vessel’s intima and restoring vascular function. The ability of EPCs to repair vascular damage depends on their number and functionality. Diabetic patients have a decreased circulating EPC count and impaired EPC function. This may at least partially explain the increased risk of diabetic complications, including the increased cardiovascular risk in these patients. Recent studies have confirmed that many currently available drugs with proven cardiovascular benefits have beneficial effects on EPC count and function. Among these drugs are also medications used to treat different types of diabetes. This manuscript aims to critically review currently available evidence about the ways anti-diabetic treatment affects EPC biology and to provide a broader context considering cardiovascular complications. The therapies that will be discussed include lifestyle adjustments, metformin, sulphonylureas, gut glucosidase inhibitors, thiazolidinediones, dipeptidyl peptidase 4 inhibitors, glucagon-like peptide 1 receptor analogs, sodium-glucose transporter 2 inhibitors, and insulin.

1. Introduction

The endothelium is the innermost cell layer of the blood vessel wall, with distinct metabolic features that are critical in maintaining vascular integrity, regulating vascular tone, blood flow, and preventing thrombosis [1]. Under certain conditions, the endothelium may become damaged and dysfunctional, resulting in inappropriate vasomotion, inflammation, and atherosclerotic plaque build-up [2]. Atherosclerotic plaques compromise the blood supply to downstream tissues, either by vessel narrowing or by total vessel obstruction. Consequently, ischemia or infarction may occur leading to end-organ dysfunction and failure. If vital organs are affected, the most dramatic outcome is death. From the epidemiological viewpoint, atherosclerosis and its complications are the leading cause of death worldwide and represent a global burden to human health [3].
Numerous risk factors related to atherosclerosis have been identified so far; the most important include age, gender and genetics, lifestyle factors (such as smoking and diet), and underlying medical conditions, like arterial hypertension, dyslipidemia, diabetes, and obesity [4,5]. Over time, complex strategies to treat atherosclerosis and vascular events have been developed, mainly targeting already-mentioned risk factors, blood clotting, and end-organ protection [3]. Among various approaches to treat atherosclerosis and its deleterious outcomes, none of them were able to restore vascular function completely and reverse atherosclerosis. The progressive nature of this disease and the limitations of currently available treatment options lead to the general opinion that atherosclerosis is an irreversible process, strongly linked to aging [6,7].
Endothelial progenitor cells (EPCs) have recently raised significant interest since these cells can aid in regenerating the damaged vessels’ endothelium and, therefore, restore vascular function. Discovered at the end of the 20th century, they gave new insight into the pathology of atherosclerosis and offered new prospects in curative medicine [8]. EPCs are usually defined as multipotent stem cells with angiogenic potential. These cells are commonly quantified from blood by flow cytometry as mononuclear cells expressing CD34 and VEGFR2+. CD133+ and CD45−/dim are proposed as additional markers for circulating EPCs [9]. The population of EPCs is rather small, with a proportion of up to 0.05% of mononuclear blood cells [10]. Since the proportion of EPCs in blood cells is very low, some technical considerations need to be highlighted. The analysis of rare cells may be compromised with background noise. Therefore, special attention needs to be given to several preanalytical and analytical steps. These include appropriate blood sampling, collection tube choice and handling temperature, the choice of appropriate flow cytometers, erythrocyte depletion and wash/no wash protocols, background control, etc. [9]. However, to determine the proliferation capacity and the ability to form blood vessel-like forms, it is crucial to cultivate EPCs. In cultures, it became apparent that the phenotype and function of EPCs are heterogeneous, and that the cell markers differ from those in EPCs detected by flow cytometry. Originally, these subpopulations were named after the time they appeared in cultures as early and late outgrowth endothelial cells. These cells express distinct cell markers and behave diversely in cultures. They have different abilities to act as paracrine producers or to proliferate and differentiate into mature endothelial cells, contributing to the formation of new blood vessels or repairing the damaged ones. Therefore, the cells were renamed into myeloid angiogenic cells (MACs) and endothelial colony-forming cells (ECFCs). These EPC subpopulations act synergistically in vivo [11,12,13]. It is noteworthy, that there are also technical challenges in stem cell cultivation in general that may have an impact on EPC cultivation, like sample preparation, cell isolation and purification, seeding density, choice of culture media, etc. [14]. The main characteristics of these EPC subtypes isolated in culture are shown in Table 1.
Due to their unique angiogenic features, EPCs seem to be a promising tool to treat conditions like atherosclerosis and atherosclerosis-related conditions such as ischemic heart disease, peripheral artery disease, and diabetic vascular complications [15,16,17]. In various research, improved parameters of EPC biology (e.g., increased EPC blood count quantified by flow cytometry or improved EPC function in cultures) could be correlated with improved flow-mediated dilation, brachial ankle index, or intima media thickness [18,19,20]. Furthermore, EPC biology was found to correlate with clinical outcomes like limb amputation, stroke, myocardial infarction, or death [21,22,23,24].
EPC-based therapies aim to enhance vascular repair and promote the growth of new blood vessels, ultimately improving blood flow to ischemic tissues, but there are challenges to be addressed. These include improving methods for EPC isolation and expansion, enhancing their engraftment and survival in target tissues, and optimizing the timing and delivery of EPC-based therapies [25,26].
It is noteworthy that many currently prescribed drugs have proven beneficial effects on EPC biology, like several antihypertensive drugs, statins, various classes of anti-diabetic medications, some hormones, bisphosphonates, and others [27]. Since at least some of these drugs are proven to decrease the incidence of cardiovascular incidents and mortality, it may be assumed that improved EPC biology may contribute to this effect [28]. In addition, the EPC count in the bloodstream assessed by flow cytometry or altered behavior of EPCs detected in culture can also serve as biomarkers for cardiovascular health. Reduced EPC numbers or impaired EPC function are associated with various cardiovascular diseases, correlating with advanced disease stage and response to treatment [22]. Understanding the biology of EPCs, and the way they react to currently available pharmacologic interventions is essential for unlocking their full potential in clinical practice [29].
The aim of this review is to discuss the impact of modern anti-diabetic treatment on EPC-mediated vascular repair, correlate it to proven clinical effects on vascular health, and discuss the potential effect of EPCs on recent cardiovascular outcome trial results.

2. Endothelial Progenitor Cells in Health and Disease

There are established regenerative cell responses identified that can diminish and even heal vascular injury and re-establish endothelial integrity and function. A prominent role in vascular repair in adult humans is had by resident endothelial progenitor cells, located in the blood vessel wall. These cells may replicate and differentiate into mature endothelial cells in response to vascular injury. In contrast to resident endothelial progenitor cells present in blood vessels’ walls, another type of endothelial progenitor cells may be found in distant tissues like bone marrow, fat tissue, and the spleen. EPCs from distant locations may be mobilized into the bloodstream under certain conditions like ischemia and hypoxia. Under conditions with limited oxygen supply, hypoxia-induced factor 1 (HIF-1) is released, thus inducing stromal-derived factor 1 (SDF-1) production and release. SDF-1 is the key regulator of EPC mobilization. SDF-1 interacts with other mobilizing factors like VEGF and E-selectin, and the PI3K/Akt/eNOS-dependent signal transduction pathway, leading to mobilization of progenitor cells into the blood flow. In addition, there are certain enzymes involved in this EPC mobilization like matrix metalloproteinase 8 and 9 (MMP-9) that inactivate retention factors at the site of EPCs’ origin [30,31]. Blood glucose, erythropoietin, thyroid hormones, and estrogen may modulate EPC mobilization [11,27,32].
EPCs enter the blood circulation, migrate to the place of vessel injury, embed there in a process named homing, and produce various paracrine substances like SDF-1, nitric oxide (NO), vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), insulin-like growth factor (IGF), and others. SDF-1 leads to upregulation of its specific chemokine receptor type 4 (CXCR-4 receptor) on the EPC surface, resulting in enhanced homing of these cells to the injured blood vessel. Other adhesion molecules, like E-selectin, integrins, intercellular adhesion molecule (ICAM), and vascular cell adhesion molecule (VCAM) are also included in this process [30]. Responding to paracrine factors secreted by embedded circulatory EPCs like VEGF, IGF-1, and gasotransmitters, resident endothelial progenitor cells proliferate and differentiate in situ resulting in vascular repair [11,30,32]. The AMPK/Akt/eNOS and AMPK/eNOS signaling pathways with their activators (adipokines, prostaglandins) have an important role in enhancing further EPC differentiation and vessel formation [33,34].
As mentioned earlier, circulating EPCs represent a heterogeneous cell population with varying characteristics. Two types of EPCs could be detected in the circulation and characterized after in vitro cultivation: MACs or early-onset EPCs, and ECFCs, known also as late-onset EPCs. MACs form colonies in cultures under special conditions only, and ECFCs are better differentiated and provide mature endothelial cells as building blocks important for physical endothelial integrity and function. Both progenitor cell types can produce paracrine substances like nitric oxide (NO) and express receptors for growth factors like the vascular endothelial growth factor receptor 2 (VEGF-R2) [35,36,37].
In healthy individuals, both resident and circulating EPCs contribute to the maintenance of vascular health by continuously replenishing and repairing the endothelial lining [38]. Considering the impact of circulating EPCs on vascular repair, it seems that their paracrine function is more important since the resident progenitor cells provide the majority of newly differentiated cells necessary for vascular repair [11,39].
It has to be noticed that the turnover of healthy human endothelial cells is rather low in regions with laminar blood flow, with a cell lifespan of many years [40]. However, in regions with turbulent blood flow, like vessel curvatures and branching points, the lifespan of endothelial cells may be shortened and cell turnover increased. In humans, the endothelial turnover is within the range from 47 to 23,000 days [41,42]. Endothelial damage may occur over time due to various mechanical and chemical stimuli. Mechanical factors like increased blood pressure may directly harm the endothelium, while chemical factors like hyperglycemia or smoking may trigger premature endothelial cell apoptosis. All these mentioned factors disrupt endothelial integrity, resulting in an elevated mature endothelial cell count in peripheral blood, as a marker of endothelial damage. Without the functional innermost layer, NO production is compromised, and vascular tone and blood flow become dysregulated, resulting in endothelial dysfunction and downstream tissue ischemia. In addition, a cascade of inflammatory processes on the site of injury triggers cholesterol accumulation and oxidation promoting further growth of atherosclerotic plaques. Furthermore, cytokines secreted by the damaged tissue attract smooth muscle cells that migrate from the artery’s muscle layer into the vascular intima. These cells further proliferate leading to plaque build-up. Oxidized cholesterol attracts macrophages, further contributing to a pro-inflammatory environment. The plaque surface is covered by a fibrous cap that may become unstable in a pro-inflammatory environment and finally rupture, leading to activation of the clotting cascade, resulting in vessel obstruction and infarction of related tissues [43,44].
In conditions where blood flow is compromised, such as in ischemic diseases (e.g., peripheral artery disease or myocardial infarction), repairing mechanisms are activated: EPCs from distant tissues enter the circulation, providing paracrine factors from myeloid angiogenic cells to stimulate both embedded late outgrowth endothelial progenitor cells and resident endothelial progenitor cells, leading to vascular repair [11,36]. In that way, the presence of EPCs helps restore blood flow by contributing to the formation of collateral vessels, which bypass blocked or narrowed arteries.
Functional EPCs decrease the risks of thrombotic events. This occurs either by replacing aged and injured endothelial cells with EPCs or by releasing substances that inhibit blood clot formation from EPCs to an extent that is still under debate [45,46]. The number and function of EPCs decline with age, which can contribute to the development of age-related vascular diseases, such as atherosclerosis. Gender-specific differences with a protective higher estrogen-dependent EPC count in fertile women during the ovulatory phase compared to age-matched men have been found [47]. EPC dysfunction is also implicated in various other related pathologies, including diabetes, dyslipidemia, and arterial hypertension, highlighting their significance in vascular disease progression [46,48].

3. Diabetes and Endothelial Progenitor Cells Biology

Diabetes mellitus is a metabolic disorder characterized by chronically elevated blood glucose levels. Depending on the diabetes type, deficient insulin action and insulin resistance may additionally contribute to the occurrence of other metabolic abnormalities like dyslipidemia and abnormal protein metabolism, resulting in inflammation and oxidative stress [49]. These metabolic alterations result in endothelial dysfunction, a key initiating event in vascular complications. Furthermore, diabetes is often accompanied by other conditions affecting endothelial function and vascular health like obesity, arterial hypertension, and dyslipidemia [50]. In addition, diabetes is associated with changes in the coagulation cascade, platelet function, and fibrinolysis, creating a prothrombotic state and increasing the risks of thrombotic events [51]. Over time, chronic diabetic complications may develop. These conditions are linked to vascular dysfunction and structural damage, affecting both large and small blood vessels in the body [52].
An important pathophysiological mechanism linking diabetes and its complications seems to be related to both MACs and ECFCs. Namely, hyperglycemia, hypoglycemia, and increased glucose variability occurring in diabetic patients, regardless of diabetes type, have detrimental effects on EPC biology [53]. Patients with both type 1 and type 2 diabetes have reduced numbers of circulating EPCs. This decrease can be noticed early after the diagnosis of type 1 and type 2 diabetes, regardless of patients’ age [54,55]. Additional studies have shown that in individuals with diabetes, EPCs often exhibit reduced mobilization capacity, shortened survival, and impaired ability to differentiate into mature endothelial cells [37,53,56,57,58].
Aside from the specific metabolic environment determined primarily by blood glucose abnormalities, possible mechanisms involved in the deterioration of EPCs abilities for vascular repair include disruptions in NO pathways, impaired intracellular signaling in other pathways (MAPK/ERK, SDF-1/CXCR-4) and the p53/sirtuin1 (SIRT1)/p66Shc axis, inflammation, reactive oxygen species, accumulation of advanced glycation end products (AGE), low levels of gasotransmitters, imbalance in adipokine production, and direct effects of insulin and IGF-1, as shown in Figure 1 [27,53,59,60,61]. In particular, MAPK causes NF-κB-dependent inflammatory stress response in the bone marrow, disrupting hematopoietic progenitor activity and enhancing inflammation-induced hypoxic injury [62], while p53/sirtuin/p66Shc disruption promotes EPC senescence [63]. Reactive oxidative species impair EPCs viability, increase apoptosis, and negatively impact tube formation, and AGE accumulation disrupts NO production and significantly decreases anti-oxidant enzymes, thus increasing oxidative stress [64]. Impaired gasotransmitter function, including decreased NO, CO, and H2S signaling, may alter extracellular matrix properties, affecting metalloproteinase function [65].
EPC dysfunction results in compromised function of both large and small blood vessels and contributes to the development and progression of diabetes-related vascular complications [53,66,67]. It has been shown that EPC dysfunction is closely linked to the occurrence of diabetic microvascular complications, such as retinopathy, nephropathy, and neuropathy [66,68,69]. These complications may lead over time to vision impairment and blindness, kidney function decline and end-stage renal disease, altered or loss of sensation in the extremities, and delayed wound healing, and are therefore leading causes of invalidity in developed countries [70,71]. In other words, enhanced EPC-mediated angiogenesis and endothelial repair in the renal, retinal, and peripheral vasculature could contribute to better outcomes in diabetic individuals.
Figure 1. Mechanisms of endothelial dysfunction in diabetes [27,53,54,55,57,59,60,61,64,65,72,73,74,75,76,77,78,79,80,81,82,83,84].
Figure 1. Mechanisms of endothelial dysfunction in diabetes [27,53,54,55,57,59,60,61,64,65,72,73,74,75,76,77,78,79,80,81,82,83,84].
Biomedicines 11 03051 g001
EPC dysfunction is also implicated in macrovascular diabetic complications, including coronary artery disease, cerebrovascular disease, and peripheral artery disease. Diminished EPC count and function contribute to impaired endothelial repair mechanisms, leading to endothelial dysfunction and accelerated plaque formation, which increase the risk of major cardiovascular events (MACEs) in diabetic individuals. Even more serious, a decreased EPC function is linked to premature mortality [18,72]. It has been proposed that EPC levels and function could serve as valuable biomarkers for diabetes-related complications. Monitoring EPC parameters may help identify individuals at higher risk of developing vascular complications, allowing early intervention and personalized treatment strategies [22]. However, it is important to note that individual patient factors, including age and the presence of comorbidities, can influence the response of EPCs to treatment and modify their effect on the endothelium [6,46]. Therefore, personalized medicine approaches that consider these factors may be necessary to optimize diabetes management.
Several landmark studies have shown that good blood glucose control decreases the risk of developing chronic diabetic complications and mortality [73,74]. Achieving good blood glucose control requires, in general, both non-pharmacological interventions and drug therapy. Recently, some anti-diabetic medications like sodium–glucose transporter-2 inhibitors and glucagon-like peptide-1 (GLP-1) receptor agonists have proven to have better cardiovascular outcomes in patients with type 2 diabetes, and these medications were highlighted in modern international guidelines [75].

4. Currently Available Diabetes Treatments Affecting EPC Count and Function

4.1. Lifestyle Modification

Lifestyle modifications are integral components of diabetes management. Most importantly, evidence-based strategies encompass dietary adjustments, appropriate exercise programs, body weight optimization, smoking cessation, and prudent alcohol choices. A healthy lifestyle has been proven to benefit blood glucose control and, thus, help to reduce diabetic complications in patients with diabetes [76,77]. Nutrition therapy is an umbrella term used for dietary modifications in diabetes therapy. Quality nutrition choices have been shown to improve blood glucose control and, thus, help to reduce diabetic complications [78,79,80,81,82]. At the time, to our best knowledge, there is a single published paper on a trial regarding EPCs in diabetic patients with a focus on nutrition. The authors found a beneficial effect of a Mediterranean diet compared to a low-fat diet on EPC count (CD34+VEGFR2+ and CD34+VEGFR2+CD133+) and carotid intima-media thickness in patients with type 2 diabetes. The increased EPC count correlated with improvements in blood glucose control, insulin sensitivity, total cholesterol, high-density lipoprotein cholesterol, and systolic blood pressure, decreasing the risk for chronic diabetic complications [83].
Several other studies focused on non-diabetic study populations with risk factors for cardiovascular events or on healthy volunteers. However, because of the complex interplay of diabetes, other risk factors, and cardiovascular disease, these trials are also relevant for diabetic macrovascular complications. Adding vegetables to the diet improved EPC counts in a randomized small study in Japan involving healthy volunteers. From a contemporary viewpoint, a serious limitation of the study is the lack of a precise EPC definition [84]. Better EPC (CD34+/VEGFR2+) counts and improved vasomotor function measured by flow-mediated vasodilation on the brachial artery were detected when flavanols were added to the diet of patients with coronary artery disease [85]. The Mediterranean diet, rich in vegetables and unsaturated fats also increased EPC (CD34+/VEGFR2+/CD133+) counts and reduced endothelial cell microparticles related to mature EC apoptosis in a small group of elderly people with a clinical correlate of improved ischemic reactive hyperemia, indicating reduced endothelial damage and improved endothelial regenerative capacity [86]. Similar results, like improved EPC (CD34+/VEGFR+/CD133+) counts, a decreased level of circulating microparticles, an improved anti-inflammatory profile, and increased flow-mediated vasodilation were found in the larger CORDIOPREV trial involving patients with already diagnosed coronary artery disease who were put on the Mediterranean diet. A simplified dietary approach with fat restriction was statistically inferior in this trial [87]. Calorie restriction in combination with the Mediterranean diet and exercise also had beneficial effects on EPC (CD34+/VEGFR2+) count in patients with metabolic syndrome after three months of intervention. Insulin sensitivity and blood pressure were also improved and body weight loss was promoted. However, endothelial function was improved only in subjects randomized to exercise [88]. Adding polyunsaturated fat from fish was also shown to improve both circulatory EPC (CD34+/VEGFR+; CD133+) count and tube-formation function after a 6-week intervention, but this effect ceased in the following 6 weeks after the intervention was stopped. An improvement in several inflammatory substances like TNF α, interleukin 8 (IL-8), and an improvement in adhesion molecule profile was also detected [89]. In another randomized trial, replacing saturated with monounsaturated and polyunsaturated dietary fats in a larger sample of people at moderate risk for cardiovascular disease increased EPC (CD34+/VEGFR2+) and decreased microparticle numbers, suggesting beneficial effects on endothelial repair and maintenance [90]. Intriguingly, excluding meat and fish from nutrition showed less favorable effects on EPC (CD34+/CD133+/CD45−/dim) than adhering to the full Mediterranean diet in the CARDIVEG study [91]. An ancient type of grain also showed an increase in EPCs (defined as CD34+ or CD133+ cells) in comparison to modern grain varieties after an intervention of 8 weeks. At the same time, significantly better blood glucose control and cholesterol levels were achieved [92].
Besides the approach with the Mediterranean diet, green tea may improve the EPC count (CD34+/VEGFR2+/CD45−/dim) even over the short term in a population of chronic smokers. In the same period, an improvement of vascular function assessed by flow-mediated dilation was detected. However, since there was no control group in the study, definitive conclusions cannot be made [93]. Wise alcohol choices may also benefit EPC biology. In a trial with a cross-over design, the effect of beer and non-alcoholic beer on circulating EPC (CD34+/VEGFR2+/CD133+) levels was tested in patients with high cardiovascular risk, compared to gin. The beer and non-alcoholic beer increased the circulating EPC count and SDF-1 in the peripheral blood. On the other hand, while the same subjects were drinking gin during the control period, there was a decrease in EPC count. A major study limitation is the lack of a control group drinking water [94]. Considering other types of alcohol, red wine has been shown to increase circulating EPC (CD34+/VEGFR2+/CD133+) levels and to improve EPC function (attenuated senescence and improved adhesion, migration, and tube formation) by modifying nitric oxide bioavailability, at least in healthy volunteers who were randomized either on red wine, vodka, beer, or water. Clinically, improved FMD vasodilation was seen in patients drinking red wine [95]. White wine also increases the EPC (CD34+/VEGFR2+, CD133+) count and decreases concentrations of several pro-inflammatory markers in a population with increased cardiovascular risk, but there was also no control group on water [96]. It is unclear if alcohol is the key effect mediator since grape seeds have been shown to also improve CD34+ cell counts. A more precise EPC characterization would be necessary to confirm this result [97].
Essentially, dietary adjustments recommended for patients with diabetes do not only improve blood glucose control but reduce the need for pharmacotherapy. It seems that they may improve vascular health by benefitting several aspects of EPC biology like EPC count, modifying NO production, and reducing the number of endothelial microparticles in circulation. However, the best dietary approach for diabetic patients needs still to be established since there is only one study performed in this population. This study showed the benefits of the Mediterranean diet. Other approaches like adding unsaturated fat, vegetarian diet, caloric restriction, and others still need to be evaluated in diabetic patients. Correlating EPC findings with clinical tests like FMD would enhance the predictive value of such research. However, eating more vegetables, adding more unsaturated fat to the diet, and caloric restriction could positively impact EPCs in non-diabetic persons, but in some parts of these studies, there was no correlate with other clinical findings. The exact mechanisms by which food influences EPC biology remain to be elucidated, but improved insulin sensitivity, decreased inflammatory parameters, and prostaglandins could play a significant role [98,99,100,101]. Intriguingly, alcohol could not show benefits on EPC parameters, with the exception of red wine, and there could not be a definitive conclusion drawn on green tea. In addition, EPCs are not equally defined in these studies, making comparations between studies difficult.
Exercise is another lifestyle factor that affects blood glucose control, but also cardiovascular morbidity and mortality in patients with diabetes. There are several trials dealing with the effect of exercise on EPC biology in diabetic patients. Acute exercise loads promptly increased the EPC (CD34+/CD133+) count in patients with type 2 diabetes with and without nephropathy [102]. In patients with type 2 diabetes and diagnosed coronary artery disease, an elevated level of microparticles and EPC could be detected. In contrast, patients with type 1 diabetes have shown a blunted EPC response to both aerobic and resistance exercise. The authors concluded that these findings may suggest a low reserve of EPC (CD34+/CD45dim and CD34+/VEGFR2+/CD45dim) in the bone marrow in patients with type 1 diabetes [103]. These findings have been confirmed by another trial by an independent group of authors, testing CD34+/VEGFR+ and CD34+/VEGFR2*/CD45dim EPC count [104]. More studies have shown the impact of exercise in non-diabetic populations, with or without other cardiovascular risk factors. Studied populations include healthy volunteers, obese adolescents, patients with renal failure, patients with heart failure, and patients with coronary artery disease [105,106,107,108,109]. In general, moderate exercise increases the EPC count, improves their migratory capacity, and reduces EPC apoptosis, with beneficial effects on vascular healing. Exercise may influence EPC counts and EPC function by several mechanisms that include increased NO production, improved insulin sensitivity, decreased inflammation, and improved hormonal and cytokine signaling [110,111]. However, EPCs are not universally equally defined in these research, making direct comparison hard.
Finally, an important lifestyle factor affecting diabetic and vascular outcomes is smoking. While nicotine is responsible for tobacco addiction, cigarette smoke contains many substances resulting from tobacco combustion. Oxidizing chemicals, carbon monoxide, particulates, heavy metals, nitrosamines, and polycyclic aromatic hydrocarbon carcinogens are drivers of tobacco toxicity and may affect EPC biology. One important mechanism responsible for endothelial damage is compromised tetrahydrobiopterin depletion leading to decreased NO availability, endothelial dysfunction, increased inflammation, and activation of platelets, resulting in accelerated atherogenesis [112]. Smokers are shown to have decreased EPC (CD34+/VEGFR2+) counts and impaired endothelial-dependent vasodilatation compared to non-smokers. The survival of EPC is also shortened [113]. Both active and passive smoking impact EPC biology negatively. Smoking cessation increases EPC (CD34+/VEGFR2+/CD133+) count and function by reducing hypoxia, reactive oxygen species (ROS) generation, and inflammation [113]. Intriguingly, consuming electronic cigarettes increases acutely the count of CD34+/CD309+ cells in regular smokers, maybe indicating an acute bone marrow response to vascular injury. Still, there is not enough evidence to draw definitive conclusions about electronic cigarettes [114]. Important to note, these studies are not consistent in EPC characterization, and there are no studies involving diabetic patients performed up to the present time, making conclusions on this topic elusive.

4.2. Anti-Diabetic Agents

Many different anti-diabetic agents are currently used to treat diabetes. Better glucose control itself is associated with improved endothelial function and a decreased risk of microvascular and macrovascular diabetic complications. There is mounting evidence about the impact of currently available anti-diabetic drugs on cardiovascular health. Furthermore, since 2008, every new anti-diabetic drug needs to be tested in a cardiovascular outcome trial if seeking approval from the Food and Drug Agency (FDA). In these trials, new agents are compared to standard treatment, and must at least prove non-inferiority considering cardiovascular outcomes. Cardiovascular outcomes of interest typically include cardiovascular death, non-fatal myocardial infarction, and stroke. Optional other outcomes may be additionally considered, like total mortality, hospitalizations for heart failure, lower limb amputations, albuminuria, etc. A brief overview on anti-diabetic medications, their mode of action, cardiovascular safety/superiority, and clinical trials involving EPC biology is shown in Table 2.
Possible mechanisms of cardiovascular benefits include beneficial effects on EPC health. EPCs may benefit from improved glycemia and insulin sensitivity, enhanced nitric oxide production, reduced oxidative stress, and decreased inflammatory parameters. Some drugs improve EPC homing by other mechanisms, including altered expression of adhesion molecules. Specific effects on EPC may vary according to differences among various classes of anti-diabetic drugs.
Among the drugs with favorable cardiovascular effects is metformin. For many years considered the first-line anti-diabetic therapy for patients with type 2 diabetes, metformin has shown cardiovascular benefits in the landmark UKPDS study [146]. Considering EPC biology, one study found that circulating EPC counts increased after metformin initiation in patients with type 2 diabetes, correlating with increased FMD [115]. Later, an increase in EPCs was proven also in patients with type 1 diabetes, when metformin was given as a supportive treatment in the MERIT trial [116]. Regarding the EPC increase with metformin in patients with type 2 diabetes, this effect could be enhanced when gliclazide, a sulphonylurea, was added [117]. It seems that the most important mechanism affecting EPC biology in metformin-treated patients is increased phosphorylated-eNOS expression and NO production in cultures, as well as altered AMPK function [147].
In general, the effect on EPCs of most other anti-diabetic drugs was tested when these drugs were added to metformin, with few exceptions.
Gliclazide alone was also able to improve EPC biology, correlating with flow-mediated vessel dilatation and improvements in markers of oxidative stress as the key mechanism of action on EPCs [118]. It seems so far that gliclazide is the only sulphonylurea with a proven effect on EPCs. Glibenclamide did not show any increase in EPCs in a trial when it was compared to vildagliptin [119]. The same was shown for glimepiride when compared to sitagliptin [120]. Although hampered by methodological limitations in EPC characterization and the absence of clinical assessment of endothelial function, this finding fits perfectly into the context of better cardiovascular outcomes in patients treated with gliclazide compared to other sulphonylureas after myocardial infarction [148,149].
Another class of oral anti-diabetic drugs are thiazolidinediones, or peroxisome proliferator-activated receptors gamma (PPAR γ) agonists. Although they are used to improve blood glucose control, they have also been shown to affect other cardiovascular risk factors, like dyslipidemia and albuminuria [150,151]. Since these drugs may cause water retention, heart failure may occur [152,153]. Pioglitazone was shown to reduce the incidence of some major adverse cardiovascular events in patients with type 2 diabetes [154,155]. Two members of this class, rosiglitazone and pioglitazone, have been proven to impact EPCs. In patients with type 2 diabetes, rosiglitazone improved EPC re-endothelialization impaired by NADPH oxidase activity, diminishing the effects of oxidative stress [156]. Human studies with pioglitazone have demonstrated increased EPC counts and proliferative ability, decreased EPC apoptosis, and improvements in some metabolic parameters and inflammatory markers [121,122,123,124]. Improved adipokine profile and anti-inflammatory properties were also proposed mechanisms to obtain EPC biology improvement [122]. In some studies, CD34+/VEGFR2+ EPCs were investigated, and these studies showed favorable results on EPC health, but the clinical response on endothelial function remains unknown since it was not tested. It has to be mentioned that in one study, pioglitazone showed no impact on EPC counts, but in this study there is no evidence about the method used for EPC determination [125]. These results justify the need for future research regarding the impact of thiazolidindiones on EPCs and clinically assessed endothelial function.
Incretin-based therapies emerged recently as interesting add-ons to diabetic pharmacotherapy. There are two classes of incretin-modulating drugs. The first one contains inhibitors of dipeptidyl peptidase-4 (DPP-4 inhibitors). The main mode of action is inhibiting the breakdown of endogenous incretins, mainly GLP-1, thus maintaining the concentration of this substance within a physiological range. This incretin then enhances food-triggered insulin secretion. The majority of them like sitagliptin, linagliptin, saxagliptin, and alogliptin were tested in cardiovascular outcome trials and were proved to be non-inferior to standard treatment [157,158,159,160]. In other words, these drugs were as good as standard treatment at that time, like metformin, gliclazide, thiazolidindiones, or insulin. The impacts of vildagliptin and teneligliptin on cardiovascular outcomes were not tested in large, randomized trials.
Considering the effect of DPP-4 inhibitors on EPCs, there are some results published showing the favorable impact of these drugs. Improved glucoregulation, improved oxidative parameters, increased NO and SDF-1 production, and decreased inflammation are the proposed mechanisms for these effects [127,128,131]. Sitagliptin increased EPCs and SDF-1 during 4 weeks as add-on treatment compared to standard treatment involving metformin and/or insulin secretagogues in a small sample of people with type 2 diabetes [126]. Similar results considering EPC count, but with a decrease in SDF-1α during a 12-week trial in patients with type 2 diabetes, were detected when sitagliptin was added to metformin [120]. These results on SDF-1 fit less well into the proposed mechanism of action of DPP-4 inhibitors since DPP-4 degrades SDF-1 [161]. More recently, sitagliptin was tested against metformin in a short trial for three days. The drug increased EPCs, but also SDF-1α and NO concentration, with the most profound impact on patients receiving both drugs [127]. In another trial, in which voglibose was used as the comparator drug, sitagliptin was shown to increase EPCs and improve flow-mediated vasodilation, while voglibose showed no effect [128]. Linagliptin also increased the EPC count acutely in patients with type 2 diabetes during a 4-day trial, with increased concentrations of SDF-1α. Intriguingly, DPP-4 activity was abated by more than 50%, indicating other mechanisms responsible for an increase in SDF-1α [129]. An increase in SDF-1 with linagliptin has been observed even after 6 months of treatment. The effect on EPCs was not statistically significant [130]. However, in patients with chronic kidney disease, linagliptin improved EPC count, the antioxidant level was enhanced, and clinical parameters like augmentation index and pulse wave parameter were improved during a 12-week trial [131].
As mentioned before, vildagliptin also increases the number of EPCs, but with a reduction in SDF-1α levels after 12 months, in comparison to glibenclamide [119]. Alogliptin showed the same effect as gliclazide on the EPC count. The authors concluded that the observed increase in EPCs seemed to be due to the glucose-lowering effect of both drugs [132]. Furthermore, saxagliptin failed to be superior to metformin in two 12-week trials. Although there was an increase in EPC count and improvement in flow-mediated dilation, metformin showed similar effects when these drugs were given as monotherapy, with no add-on effect on EPCs in dual treatment [133,134]. Finally, teneligliptin showed an increasing trend in the number of EPCs, albeit this did not reach statistical significance. However, flow-mediated vasodilatation improved in the study group, suggesting a mechanism different than EPC [135]. In conclusion, there is some evidence that DDP-4 inhibitors improve certain aspects of EPC health; the main limitation is the absence of clinical endothelial assessment in the majority of research, and a short study duration in some of the trials.
In contrast to DPP-4 inhibitors, GLP-1 receptor agonists override the physiological effects of endogenous GLP-1 and exert a stronger effect on GLP-1 receptors. These are potent anti-diabetic drugs with the main mechanism of action that stimulates insulin secretion after an oral glucose load via the incretin effect. Other medical benefits may include delaying gastric emptying, inhibiting glucagon production, decreasing pancreatic beta-cell apoptosis, promoting weight loss, lowering arterial blood pressure and total cholesterol, improving left ventricular ejection fraction, myocardial contractility, coronary blood flow, cardiac output, and endothelial function while reducing infarction size. Some of them like liraglutide, semaglutide, and dulaglutide are proven to be superior to standard anti-diabetic treatment in secondary cardiovascular prevention [162,163,164]. Interestingly, there are limited data about their effects on human EPCs. In a small trial, liraglutide was inferior to exenatide considering EPC counts, but liraglutide was not given in the full dose of 1.8 mg daily. The authors speculated about antioxidative/anti-inflammatory effects as mediators on EPC biology [136]. In a head-to-head trial, comparing liraglutide in the full dose of 1.8 mg daily and sitagliptin, there was a similar effect on EPC count, with a more favorable effect of liraglutide on VEGF and SDF-1α after 26 weeks. However, in this trial, CD 34+/VEGFR2+ EPCs were not investigated [137]. So far, dulaglutide is the single GLP-1 receptor agonist that showed an increase in EPC count and function resulting in improved clinical parameters like brachial-ankle pulse wave velocity. Lower grades of inflammation and increased NO production were the supposed mechanisms for these effects [138]. No data on humans for semaglutide have yet been published. Putting all these data into the context of previously published cardiovascular outcome trials which stated superiority for the majority of these drugs [162,163,164], and borderline significance (p = 0.06) for exenatide [165], it seems that further investigations are justified to prove the beneficial impact of GLP-1 receptor agonists on EPCs.
Sodium-glucose transporter-2 inhibitors (SGLT2i) are the latest introduced class of anti-diabetic treatment. Their main mode of action involves the inhibition of the sodium/glucose co-transporter-2 (SGLT-2) in the kidneys’ proximal tubule, causing glycosuria and lowering blood glucose levels independent of insulin action. Empagliflozin was the first anti-diabetic drug to show cardiovascular superiority over standard anti-diabetic treatment [166]. Later it was recognized that the benefit of the entire drug class goes beyond the glucose-lowering effects, from reducing the risk of MACEs, hospitalization for heart failure, and worsening of chronic kidney disease (CKD) in diabetic patients, to reducing the rate of cardiovascular death and hospitalization for heart failure in nondiabetic patients [167]. There is a limited number of clinical research studies considering the impact of SGLT2i on EPC biology. In the first randomized controlled trial of dapagliflozin vs. placebo with an open-label extension and an open-label observational study of empagliflozin treatment on levels of circulating stem cells (CSCs) and EPCs, results showed a non-significant increase in CSC and EPC after short-term treatment with SGLT-2is. After 1.5 years of dapagliflozin treatment, the EPC count significantly increased. The authors concluded that cardiovascular protection cannot be directly correlated with EPC counts, suggesting protection is dominantly attributable to other factors [139]. In another trial, empagliflozin increased the subpopulations of circulating cells expressing CD133+ following 6 months of treatment, while improving inflammation parameters [141]. In addition, dapagliflozin was shown to improve the vasculogenic capacity of EPCs via activating AMPK-mediated inhibition of inflammation and oxidative stress in a study comparing patients with type 2 diabetes with healthy controls over 3 months [140]. Similarly, a significant better expression of the CXCR4 receptor with an increase in the migratory function of CD34+ cells and an increase in the expression of antioxidants (superoxide dismutase 2, catalase, and glutathione peroxidase) in canagliflozin-treated patients as compared to the placebo group was shown [142]. These results suggest that the action of SGLT-2i may also be in part mediated through the effect on EPCs with consequently beneficial effects that go beyond the glucose-lowering effect, but it is still too early to deduce on the effect of this drug class on EPCs. Although there are only a few studies exploring these issues, they showed new directions in explaining and understanding the beneficial effects of SGLT-2i. Future research would surely benefit from better EPC definition.
The effect of insulin analogs on EPCs has also been investigated. In vitro, insulin can mobilize EPCs. In patients suffering from type 2 diabetes, both insulin glargine and detemir raised EPC counts, with no difference between the two drugs over 6 months [143]. However, it seems that long-acting insulin analogs increased the EPC count to a greater extent in comparison to intermediate-acting human insulin and oral drugs with a trend towards improved intima-media thickness [144]. Intensive insulin therapy enhanced EPC counts over 6 months compared to basal–oral therapy (metformin and/or sulphonylureas) in patients with type 2 diabetes and peripheral artery disease undergoing peripheral angiography and subsequent angioplasty procedure. However, the cumulative incidence of restenosis/amputation/limb salvage procedures/death in patients with type 2 diabetes and chronic limb ischemia patients did not differ between groups at the study end, but there was a significant effect on eNOS gene variants [145]. In addition, hypoglycemia during insulin therapy may negatively impact EPC biology and clinical outcomes [56]. Since there was no inherent effect of both NPH insulin and insulin glargine on EPC count compared to escalated oral therapy, it may be concluded that there is no specific effect of insulin beyond better glucose control. These findings fit into the general accepted fact that novel insulins are not superior to standard treatment in terms of reducing MACEs. Other factors, like eNOS genetic polymorphism and eventual hypoglycemic events, may easily overshadow the beneficial effects of insulin therapy.
Finally, it has to be mentioned that trials involving patients with type 1 diabetes are few. The majority of treatment discussed previously is registered for patients with type 2 diabetes. In an interesting trial involving patients with type 1 diabetes, reducing glucose variability with new technologies like insulin pumps showed beneficial effects on EPC count in a 6-month trial [58]. As mentioned before, adding metformin to insulin treatment in type 1 diabetes may improve EPC count and function [116].

5. Conclusions

EPCs are a captivating and vital component of the vascular system. Their roles in vascular repair, regeneration, and potential therapeutic applications make them a subject of ongoing research and hold promise for improving cardiovascular health and advancing regenerative medicine. Vessel regeneration and repairment are both altered in diabetes mellitus. Consequently, micro and macroangiopathic complications may develop. Therefore, EPCs have become the target of interest for many scientists who are putting an effort into discovering treatment options that can affect their count and function. Many available anti-diabetic drugs like metformin, sulphonylureas, PPAR γ agonists, DPP-4 inhibitors, and insulin are proven to improve, under certain conditions, the low number and functional impairment of EPCs. SGLT-2i and GLP-1 receptor agonists are the newest anti-diabetic drugs with limited evidence of beneficial effects on EPC biology. Future research will likely focus on untangling the complexity of EPC biology and developing innovative approaches to harness their full potential both in type 2 and type 1 diabetes.
To conclude, current data suggest that the low number and dysfunction of EPCs can be improved by treatment of diabetes with currently available drugs, either through drugs’ specific mechanisms or through improving blood glucose control.

Author Contributions

Conceptualization, V.A. and T.B.; formal analysis, V.A., T.B., J.M.R., L.Š. and S.U.; writing—original draft, V.A., J.M.R. and L.Š.; writing—review and editing, V.A. and T.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chang, X.; Lochner, A.; Wang, H.-H.; Wang, S.; Zhu, H.; Ren, J.; Zhou, H. Coronary Microvascular Injury in Myocardial Infarction: Perception and Knowledge for Mitochondrial Quality Control. Theranostics 2021, 11, 6766. [Google Scholar] [CrossRef] [PubMed]
  2. Cavieres, V.; Valdes, K.; Moreno, B.; Moore-Carrasco, R.; Gonzalez, D.R. Vascular Hypercontractility and Endothelial Dysfunction before Development of Atherosclerosis in Moderate Dyslipidemia: Role for Nitric Oxide and Interleukin-6. Am. J. Cardiovasc. Dis. 2014, 4, 114. [Google Scholar] [PubMed]
  3. Hetherington, I.; Totary-Jain, H. Anti-Atherosclerotic Therapies: Milestones, Challenges, and Emerging Innovations. Mol. Ther. 2022, 30, 3106–3117. [Google Scholar] [CrossRef] [PubMed]
  4. Prasad, K. Current Status of Primary, Secondary, and Tertiary Prevention of Coronary Artery Disease. Int. J. Angiol. 2021, 30, 177–186. [Google Scholar] [CrossRef] [PubMed]
  5. Flora, G.D.; Nayak, M.K. A Brief Review of Cardiovascular Diseases, Associated Risk Factors and Current Treatment Regimes. Curr. Pharm. Des. 2019, 25, 4063–4084. [Google Scholar] [CrossRef]
  6. Pacinella, G.; Ciaccio, A.M.; Tuttolomondo, A. Endothelial Dysfunction and Chronic Inflammation: The Cornerstones of Vascular Alterations in Age-Related Diseases. Int. J. Mol. Sci. 2022, 23, 15722. [Google Scholar] [CrossRef]
  7. Chen, M.S.; Lee, R.T.; Garbern, J.C. Senescence Mechanisms and Targets in the Heart. Cardiovasc. Res. 2022, 118, 1173–1187. [Google Scholar] [CrossRef] [PubMed]
  8. Haybar, H.; Shahrabi, S.; Rezaeeyan, H.; Shirzad, R.; Saki, N. Endothelial Cells: From Dysfunction Mechanism to Pharmacological Effect in Cardiovascular Disease. Cardiovasc. Toxicol. 2019, 19, 13–22. [Google Scholar] [CrossRef]
  9. Van Craenenbroeck, E.M.; Van Craenenbroeck, A.H.; Van Ierssel, S.; Bruyndonckx, L.; Hoymans, V.Y.; Vrints, C.J.; Conraads, V.M. Quantification of Circulating CD34+/KDR+/CD45dim Endothelial Progenitor Cells: Analytical Considerations. Int. J. Cardiol. 2013, 167, 1688–1695. [Google Scholar] [CrossRef]
  10. Patel, R.S.; Li, Q.; Ghasemzadeh, N.; Eapen, D.J.; Moss, L.D.; Janjua, A.U.; Manocha, P.; Al Kassem, H.; Veledar, E.; Samady, H.; et al. Circulating CD34+ Progenitor Cells and Risk of Mortality in a Population with Coronary Artery Disease. Circ. Res. 2015, 116, 289–297. [Google Scholar] [CrossRef]
  11. Aquino, J.B.; Sierra, R.; Montaldo, L.A. Diverse Cellular Origins of Adult Blood Vascular Endothelial Cells. Dev. Biol. 2021, 477, 117–132. [Google Scholar] [CrossRef] [PubMed]
  12. Yan, F.; Li, J.; Zhang, W. Transplantation of Endothelial Progenitor Cells: Summary and Prospect. Acta Histochem. 2023, 125, 151990. [Google Scholar] [CrossRef] [PubMed]
  13. Medina, R.J.; Barber, C.L.; Sabatier, F.; Dignat-George, F.; Melero-Martin, J.M.; Khosrotehrani, K.; Ohneda, O.; Randi, A.M.; Chan, J.K.; Yamaguchi, T.; et al. Endothelial Progenitors: A Consensus Statement on Nomenclature. Stem Cells Transl. Med. 2017, 6, 1316–1320. [Google Scholar] [CrossRef]
  14. Mastrolia, I.; Foppiani, E.M.; Murgia, A.; Candini, O.; Samarelli, A.V.; Grisendi, G.; Veronesi, E.; Horwitz, E.M.; Dominici, M. Challenges in Clinical Development of Mesenchymal Stromal/Stem Cells: Concise Review. Stem Cells Transl. Med. 2019, 8, 1135–1148. [Google Scholar] [CrossRef] [PubMed]
  15. Fujita, Y.; Kawamoto, A. Stem Cell-Based Peripheral Vascular Regeneration. Adv. Drug Deliv. Rev. 2017, 120, 25–40. [Google Scholar] [CrossRef] [PubMed]
  16. Xiao, S.-T.; Kuang, C.-Y. Endothelial Progenitor Cells and Coronary Artery Disease: Current Concepts and Future Research Directions. World J. Clin. Cases 2021, 9, 8953. [Google Scholar] [CrossRef]
  17. Rajasekar, P.; O’Neill, C.L.; Eeles, L.; Stitt, A.W.; Medina, R.J. Epigenetic Changes in Endothelial Progenitors as a Possible Cellular Basis for Glycemic Memory in Diabetic Vascular Complications. J. Diabetes Res. 2015, 2015, 436879. [Google Scholar] [CrossRef]
  18. Cassano, V.; Tripepi, G.; Perticone, M.; Miceli, S.; Scopacasa, I.; Armentaro, G.; Greco, M.; Maio, R.; Hribal, M.L.; Sesti, G.; et al. Endothelial Progenitor Cells Predict Vascular Damage Progression in Naive Hypertensive Patients According to Sex. Hypertens. Res. 2021, 44, 1451–1461. [Google Scholar] [CrossRef]
  19. Kumawat, R.; Gulati, S.; Ahluwalia, C.; Bahamania, K.; Varshney, P. Relation of Endothelial Progenitor Cells and Carotid Intima Media Thickness in Indian Rheumatoid Arthritis Patients-A Cross Sectional Study. J. Assoc. Phys. India 2022, 70, 11–12. [Google Scholar]
  20. Wu, W.-Z.; Hu, D.-J.; Wang, Z.-Y.; Liao, L.-S.; Li, C.-C. Endothelial Progenitor Cell Impairment Mediated Vasodilation Dysfunction via Diminishing Nitric Oxide Production in Postmenopausal Females. Mol. Med. Rep. 2019, 19, 2449–2457. [Google Scholar] [CrossRef]
  21. Santillán-Cortez, D.; Vera-Gómez, E.; Hernández-Patricio, A.; Ruíz-Hernández, A.S.; Gutiérrez-Buendía, J.A.; De la Vega-Moreno, K.; Rizo-García, Y.A.; Loman-Zuñiga, O.A.; Escotto-Sánchez, I.; Rodríguez-Trejo, J.M.; et al. Endothelial Progenitor Cells May Be Related to Major Amputation after Angioplasty in Patients with Critical Limb Ischemia. Cells 2023, 12, 584. [Google Scholar] [CrossRef] [PubMed]
  22. Heinisch, P.P.; Bello, C.; Emmert, M.Y.; Carrel, T.; Dreßen, M.; Hörer, J.; Winkler, B.; Luedi, M.M. Endothelial Progenitor Cells as Biomarkers of Cardiovascular Pathologies: A Narrative Review. Cells 2022, 11, 1678. [Google Scholar] [CrossRef] [PubMed]
  23. Pyšná, A.; Bém, R.; Němcová, A.; Fejfarová, V.; Jirkovská, A.; Hazdrová, J.; Jude, E.B.; Dubskỳ, M. Endothelial Progenitor Cells Biology in Diabetes Mellitus and Peripheral Arterial Disease and Their Therapeutic Potential. Stem Cell Rev. Rep. 2019, 15, 157–165. [Google Scholar] [CrossRef] [PubMed]
  24. Kukumberg, M.; Zaw, A.M.; Wong, D.H.; Toh, C.M.; Chan, B.P.; Seet, R.C.; Wong, P.T.; Yim, E.K. Characterization and Functional Assessment of Endothelial Progenitor Cells in Ischemic Stroke Patients. Stem Cell Rev. Rep. 2021, 17, 952–967. [Google Scholar] [CrossRef] [PubMed]
  25. Salybekov, A.A.; Kobayashi, S.; Asahara, T. Characterization of Endothelial Progenitor Cell: Past, Present, and Future. Int. J. Mol. Sci. 2022, 23, 7697. [Google Scholar] [CrossRef]
  26. Chambers, S.E.; Pathak, V.; Pedrini, E.; Soret, L.; Gendron, N.; Guerin, C.L.; Stitt, A.W.; Smadja, D.M.; Medina, R.J. Current Concepts on Endothelial Stem Cells Definition, Location, and Markers. Stem Cells Transl. Med. 2021, 10, S54–S61. [Google Scholar] [CrossRef] [PubMed]
  27. Altabas, V. Diabetes, Endothelial Dysfunction, and Vascular Repair: What Should a Diabetologist Keep His Eye On? Int. J. Endocrinol. 2015, 2015, 848272. [Google Scholar] [CrossRef]
  28. Altabas, V.; Biloš, L.S.K. The Role of Endothelial Progenitor Cells in Atherosclerosis and Impact of Anti-Lipemic Treatments on Endothelial Repair. Int. J. Mol. Sci. 2022, 23, 2663. [Google Scholar] [CrossRef]
  29. Ross, M.D. Endothelial Regenerative Capacity and Aging: Influence of Diet, Exercise and Obesity. Curr. Cardiol. Rev. 2018, 14, 233–244. [Google Scholar] [CrossRef]
  30. Li, J.-H.; Li, Y.; Huang, D.; Yao, M. Role of Stromal Cell-Derived Factor-1 in Endothelial Progenitor Cell-Mediated Vascular Repair and Regeneration. Tissue Eng. Regen. Med. 2021, 18, 747–758. [Google Scholar] [CrossRef]
  31. Klein, G.; Schmal, O.; Aicher, W.K. Matrix Metalloproteinases in Stem Cell Mobilization. Matrix Biol. 2015, 44, 175–183. [Google Scholar] [CrossRef] [PubMed]
  32. Tao, J.; Cao, X.; Yu, B.; Qu, A. Vascular Stem/Progenitor Cells in Vessel Injury and Repair. Front. Cardiovasc. Med. 2022, 9, 845070. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, S.; Miao, J.; Qu, M.; Yang, G.-Y.; Shen, L. Adiponectin Modulates the Function of Endothelial Progenitor Cells via AMPK/eNOS Signaling Pathway. Biochem. Biophys. Res. Commun. 2017, 493, 64–70. [Google Scholar] [CrossRef] [PubMed]
  34. Zhu, Z.; Fu, C.; Li, X.; Song, Y.; Li, C.; Zou, M.; Guan, Y.; Zhu, Y. Prostaglandin E2 Promotes Endothelial Differentiation from Bone Marrow-Derived Cells through AMPK Activation. PLoS ONE 2011, 6, e23554. [Google Scholar] [CrossRef]
  35. Chopra, H.; Hung, M.K.; Kwong, D.L.; Zhang, C.F.; Pow, E.H.N. Insights into Endothelial Progenitor Cells: Origin, Classification, Potentials, and Prospects. Stem Cells Int. 2018, 2018, 9847015. [Google Scholar] [CrossRef]
  36. Yang, J.-X.; Pan, Y.-Y.; Wang, X.-X.; Qiu, Y.-G.; Mao, W. Endothelial Progenitor Cells in Age-Related Vascular Remodeling. Cell Transpl. 2018, 27, 786–795. [Google Scholar] [CrossRef]
  37. Morrone, D.; Picoi, M.E.L.; Felice, F.; De Martino, A.; Scatena, C.; Spontoni, P.; Naccarato, A.G.; Di Stefano, R.; Bortolotti, U.; Dal Monte, M. Endothelial Progenitor Cells: An Appraisal of Relevant Data from Bench to Bedside. Int. J. Mol. Sci. 2021, 22, 12874. [Google Scholar] [CrossRef]
  38. Fadini, G.P.; Mehta, A.; Dhindsa, D.S.; Bonora, B.M.; Sreejit, G.; Nagareddy, P.; Quyyumi, A.A. Circulating Stem Cells and Cardiovascular Outcomes: From Basic Science to the Clinic. Eur. Heart J. 2020, 41, 4271–4282. [Google Scholar] [CrossRef]
  39. Evans, C.E.; Iruela-Arispe, M.L.; Zhao, Y.-Y. Mechanisms of Endothelial Regeneration and Vascular Repair and Their Application to Regenerative Medicine. Am. J. Pathol. 2021, 191, 52–65. [Google Scholar] [CrossRef]
  40. Pearson, J.D. Plasticity of Adult Endothelium: How Frequent and to What Extent? Cardiovasc. Res. 2015, 108, 319–320. [Google Scholar] [CrossRef]
  41. Kliche, K.; Jeggle, P.; Pavenstädt, H.; Oberleithner, H. Role of Cellular Mechanics in the Function and Life Span of Vascular Endothelium. Pflügers Arch. Eur. J. Physiol. 2011, 462, 209–217. [Google Scholar] [CrossRef] [PubMed]
  42. Hobson, B.; Denekamp, J. Endothelial Proliferation in Tumours and Normal Tissues: Continuous Labelling Studies. Br. J. Cancer 1984, 49, 405–413. [Google Scholar] [CrossRef] [PubMed]
  43. Milutinović, A.; Šuput, D.; Zorc-Pleskovič, R. Pathogenesis of Atherosclerosis in the Tunica Intima, Media, and Adventitia of Coronary Arteries: An Updated Review. Bosn. J. Basic Med. Sci. 2020, 20, 21–30. [Google Scholar] [CrossRef]
  44. Beck-Joseph, J.; Lehoux, S. Molecular Interactions between Vascular Smooth Muscle Cells and Macrophages in Atherosclerosis. Front. Cardiovasc. Med. 2021, 8, 737934. [Google Scholar] [CrossRef]
  45. Murad, H.A.; Rafeeq, M.M.; Alqurashi, T.M. Role and Implications of the CXCL12/CXCR4/CXCR7 Axis in Atherosclerosis: Still a Debate. Ann. Med. 2021, 53, 1598–1612. [Google Scholar] [CrossRef] [PubMed]
  46. Bochenek, M.L.; Schütz, E.; Schäfer, K. Endothelial Cell Senescence and Thrombosis: Ageing Clots. Thromb. Res. 2016, 147, 36–45. [Google Scholar] [CrossRef]
  47. Fadini, G.P.; de Kreutzenberg, S.; Albiero, M.; Coracina, A.; Pagnin, E.; Baesso, I.; Cignarella, A.; Bolego, C.; Plebani, M.; Nardelli, G.B.; et al. Gender Differences in Endothelial Progenitor Cells and Cardiovascular Risk Profile: The Role of Female Estrogens. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 997–1004. [Google Scholar] [CrossRef]
  48. Tripolt, N.J.; Narath, S.H.; Eder, M.; Pieber, T.R.; Wascher, T.C.; Sourij, H. Multiple Risk Factor Intervention Reduces Carotid Atherosclerosis in Patients with Type 2 Diabetes. Cardiovasc. Diabetol. 2014, 13, 95. [Google Scholar] [CrossRef]
  49. ElSayed, N.A.; Aleppo, G.; Aroda, V.R.; Bannuru, R.R.; Brown, F.M.; Bruemmer, D.; Collins, B.S.; Hilliard, M.E.; Isaacs, D.; Johnson, E.L.; et al. 2. Classification and Diagnosis of Diabetes: Standards of Care in Diabetes—2023. Diabetes Care 2023, 46, S19–S40. [Google Scholar] [CrossRef]
  50. Jyotsna, F.; Ahmed, A.; Kumar, K.; Kaur, P.; Chaudhary, M.H.; Kumar, S.; Khan, E.; Khanam, B.; Shah, S.U.; Varrassi, G.; et al. Exploring the Complex Connection between Diabetes and Cardiovascular Disease: Analyzing Approaches to Mitigate Cardiovascular Risk in Patients with Diabetes. Cureus 2023, 15, e43882. [Google Scholar] [CrossRef]
  51. Schneider, D.J. Diabetes and Thrombosis. In Diabetes and Cardiovascular Disease; Springer: Berlin/Heidelberg, Germany, 2023; pp. 99–127. [Google Scholar]
  52. Sartore, G.; Ragazzi, E.; Caprino, R.; Lapolla, A. Long-Term HbA1c Variability and Macro-/Micro-Vascular Complications in Type 2 Diabetes Mellitus: A Meta-Analysis Update. Acta Diabetol. 2023, 60, 721–738. [Google Scholar] [CrossRef] [PubMed]
  53. Wils, J.; Favre, J.; Bellien, J. Modulating Putative Endothelial Progenitor Cells for the Treatment of Endothelial Dysfunction and Cardiovascular Complications in Diabetes. Pharmacol. Ther. 2017, 170, 98–115. [Google Scholar] [CrossRef] [PubMed]
  54. Fadini, G.P.; Boscaro, E.; De Kreutzenberg, S.; Agostini, C.; Seeger, F.; Dimmeler, S.; Zeiher, A.; Tiengo, A.; Avogaro, A. Time Course and Mechanisms of Circulating Progenitor Cell Reduction in the Natural History of Type 2 Diabetes. Diabetes Care 2010, 33, 1097–1102. [Google Scholar] [CrossRef] [PubMed]
  55. Hörtenhuber, T.; Rami-Mehar, B.; Satler, M.; Nagl, K.; Höbaus, C.; Höllerl, F.; Koppensteiner, R.; Schernthaner, G.; Schober, E.; Schernthaner, G.-H. Endothelial Progenitor Cells Are Related to Glycemic Control in Children with Type 1 Diabetes over Time. Diabetes Care 2013, 36, 1647–1653. [Google Scholar] [CrossRef] [PubMed]
  56. Fadini, G.; Albiero, M.; Vigili de Kreutzenberg, S.; Avogaro, A. Hypoglycemia Affects the Changes in Endothelial Progenitor Cell Levels during Insulin Therapy in Type 2 Diabetic Patients. J. Endocrinol. Investig. 2015, 38, 733–738. [Google Scholar] [CrossRef]
  57. Fadini, G.P.; Boscari, F.; Cappellari, R.; Galasso, S.; Rigato, M.; Bonora, B.M.; D’Anna, M.; Bruttomesso, D.; Avogaro, A. Effects of Hypoglycemia on Circulating Stem and Progenitor Cells in Diabetic Patients. J. Clin. Endocrinol. Metab. 2018, 103, 1048–1055. [Google Scholar] [CrossRef]
  58. Maiorino, M.I.; Casciano, O.; Volpe, E.D.; Bellastella, G.; Giugliano, D.; Esposito, K. Reducing Glucose Variability with Continuous Subcutaneous Insulin Infusion Increases Endothelial Progenitor Cells in Type 1 Diabetes: An Observational Study. Endocrine 2016, 52, 244–252. [Google Scholar] [CrossRef]
  59. Qiu, C.; Xie, Q.; Zhang, D.; Chen, Q.; Hu, J.; Xu, L. GM-CSF Induces Cyclin D1 Expression and Proliferation of Endothelial Progenitor Cells via PI3K and MAPK Signaling. Cell. Physiol. Biochem. 2014, 33, 784–795. [Google Scholar] [CrossRef]
  60. Zhou, Y.; Yan, H.; Guo, M.; Zhu, J.; Xiao, Q.; Zhang, L. Reactive Oxygen Species in Vascular Formation and Development. Oxidative Med. Cell. Longev. 2013, 2013, 374963. [Google Scholar] [CrossRef]
  61. Toda, N.; Okamura, T. Obesity Impairs Vasodilatation and Blood Flow Increase Mediated by Endothelial Nitric Oxide: An Overview. J. Clin. Pharmacol. 2013, 53, 1228–1239. [Google Scholar] [CrossRef]
  62. Ramalingam, P.; Poulos, M.G.; Lazzari, E.; Gutkin, M.C.; Lopez, D.; Kloss, C.C.; Crowley, M.J.; Katsnelson, L.; Freire, A.G.; Greenblatt, M.B.; et al. Chronic Activation of Endothelial MAPK Disrupts Hematopoiesis via NFKB Dependent Inflammatory Stress Reversible by SCGF. Nat. Commun. 2020, 11, 666. [Google Scholar] [CrossRef] [PubMed]
  63. Goligorsky, M.; Hirschi, K. Stress-Induced Premature Senescence of Endothelial and Endothelial Progenitor Cells. Adv. Pharmacol. 2016, 77, 281–306. [Google Scholar] [PubMed]
  64. Saad, M.I.; Abdelkhalek, T.M.; Saleh, M.M.; Kamel, M.A.; Youssef, M.; Tawfik, S.H.; Dominguez, H. Insights into the Molecular Mechanisms of Diabetes-Induced Endothelial Dysfunction: Focus on Oxidative Stress and Endothelial Progenitor Cells. Endocrine 2015, 50, 537–567. [Google Scholar] [CrossRef] [PubMed]
  65. Juin, S.K.; Ouseph, R.; Gondim, D.D.; Jala, V.R.; Sen, U. Diabetic Nephropathy and Gaseous Modulators. Antioxidants 2023, 12, 1088. [Google Scholar] [CrossRef]
  66. Rigato, M.; Bittante, C.; Albiero, M.; Avogaro, A.; Fadini, G.P. Circulating Progenitor Cell Count Predicts Microvascular Outcomes in Type 2 Diabetic Patients. J. Clin. Endocrinol. Metab. 2015, 100, 2666–2672. [Google Scholar] [CrossRef]
  67. Makino, H.; Miyamoto, Y.; Kikuchi-Taura, A.; Soma, T.; Taguchi, A.; Kishimoto, I. Decreased Levels of Circulating CD 34+ Cells Are Associated with Coronary Heart Disease in Japanese Patients with Type 2 Diabetes. J. Diabetes Investig. 2015, 6, 473–478. [Google Scholar] [CrossRef]
  68. Yu, C.-G.; Zhang, N.; Yuan, S.-S.; Ma, Y.; Yang, L.-Y.; Feng, Y.-M.; Zhao, D. Endothelial Progenitor Cells in Diabetic Microvascular Complications: Friends or Foes? Stem Cells Int. 2016, 2016, 1803989. [Google Scholar] [CrossRef]
  69. Das, S.K.; Yuan, Y.F.; Li, M.Q. An Overview on Current Issues and Challenges of Endothelial Progenitor Cell-Based Neovascularization in Patients with Diabetic Foot Ulcer. Cell. Reprogramming 2017, 19, 75–87. [Google Scholar] [CrossRef]
  70. Naaman, S.C.; Bakris, G.L. Diabetic Nephropathy: Update on Pillars of Therapy Slowing Progression. Diabetes Care 2023, 46, 1574–1586. [Google Scholar] [CrossRef]
  71. American Diabetes Association. Retinopathy, Neuropathy, and Foot Care: Standards of Care in Diabetes—2023. Diabetes Care 2023, 46, S203–S215. [Google Scholar] [CrossRef]
  72. Berezin, A.E. Endothelial Progenitor Cells Dysfunction and Impaired Tissue Reparation: The Missed Link in Diabetes Mellitus Development. Diabetes Metab. Syndr. Clin. Res. Rev. 2017, 11, 215–220. [Google Scholar] [CrossRef] [PubMed]
  73. Holman, R.R.; Paul, S.K.; Bethel, M.A.; Neil, H.A.W.; Matthews, D.R. Long-Term Follow-up after Tight Control of Blood Pressure in Type 2 Diabetes. N. Engl. J. Med. 2008, 359, 1565–1576. [Google Scholar] [CrossRef] [PubMed]
  74. Fullerton, B.; Jeitler, K.; Seitz, M.; Horvath, K.; Berghold, A.; Siebenhofer, A. Intensive Glucose Control versus Conventional Glucose Control for Type 1 Diabetes Mellitus. Cochrane Database Syst. Rev. 2014, 2016, CD009122. [Google Scholar] [CrossRef] [PubMed]
  75. ElSayed, N.A.; Aleppo, G.; Aroda, V.R.; Bannuru, R.R.; Brown, F.M.; Bruemmer, D.; Collins, B.S.; Hilliard, M.E.; Isaacs, D.; Johnson, E.L.; et al. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Care in Diabetes—2023. Diabetes Care 2023, 46, S140–S157. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, G.; Li, Y.; Pan, A.; Hu, Y.; Chen, S.; Qian, F.; Rimm, E.B.; Manson, J.E.; Stampfer, M.J.; Giatsidis, G.; et al. Adherence to a Healthy Lifestyle in Association with Microvascular Complications Among Adults with Type 2 Diabetes. JAMA Netw. Open 2023, 6, e2252239. [Google Scholar] [CrossRef]
  77. Monaghan, M.; Helgeson, V.; Wiebe, D. Type 1 Diabetes in Young Adulthood. Curr. Diabetes Rev. 2015, 11, 239–250. [Google Scholar] [CrossRef]
  78. Mottalib, A.; Salsberg, V.; Mohd-Yusof, B.-N.; Mohamed, W.; Carolan, P.; Pober, D.M.; Mitri, J.; Hamdy, O. Effects of Nutrition Therapy on HbA1c and Cardiovascular Disease Risk Factors in Overweight and Obese Patients with Type 2 Diabetes. Nutr. J. 2018, 17, 42. [Google Scholar] [CrossRef]
  79. ElSayed, N.A.; Aleppo, G.; Aroda, V.R.; Bannuru, R.R.; Brown, F.M.; Bruemmer, D.; Collins, B.S.; Hilliard, M.E.; Isaacs, D.; Johnson, E.L.; et al. 5. Facilitating Positive Health Behaviors and Well-Being to Improve Health Outcomes: Standards of Care in Diabetes—2023. Diabetes Care 2023, 46, S68–S96. [Google Scholar] [CrossRef]
  80. Dunkler, D.; Kohl, M.; Heinze, G.; Teo, K.K.; Rosengren, A.; Pogue, J.; Gao, P.; Gerstein, H.; Yusuf, S.; Oberbauer, R.; et al. Modifiable Lifestyle and Social Factors Affect Chronic Kidney Disease in High-Risk Individuals with Type 2 Diabetes Mellitus. Kidney Int. 2015, 87, 784–791. [Google Scholar] [CrossRef]
  81. Sala-Vila, A.; Díaz-López, A.; Valls-Pedret, C.; Cofán, M.; García-Layana, A.; Lamuela-Raventós, R.-M.; Castañer, O.; Zanon-Moreno, V.; Martinez-Gonzalez, M.A.; Toledo, E.; et al. Dietary Marine Ømega-3 Fatty Acids and Incident Sight-Threatening Retinopathy in Middle-Aged and Older Individuals with Type 2 Diabetes: Prospective Investigation from the PREDIMED Trial. JAMA Ophthalmol. 2016, 134, 1142–1149. [Google Scholar] [CrossRef]
  82. Storz, M.A.; Küster, O. Plant-Based Diets and Diabetic Neuropathy: A Systematic Review. Lifestyle Med. 2020, 1, e6. [Google Scholar] [CrossRef]
  83. Maiorino, M.I.; Bellastella, G.; Petrizzo, M.; Gicchino, M.; Caputo, M.; Giugliano, D.; Esposito, K. Effect of a Mediterranean Diet on Endothelial Progenitor Cells and Carotid Intima-Media Thickness in Type 2 Diabetes: Follow-up of a Randomized Trial. Eur. J. Prev. Cardiol. 2017, 24, 399–408. [Google Scholar] [CrossRef] [PubMed]
  84. Mano, R.; Ishida, A.; Ohya, Y.; Todoriki, H.; Takishita, S. Dietary Intervention with Okinawan Vegetables Increased Circulating Endothelial Progenitor Cells in Healthy Young Women. Atherosclerosis 2009, 204, 544–548. [Google Scholar] [CrossRef] [PubMed]
  85. Heiss, C.; Jahn, S.; Taylor, M.; Real, W.M.; Angeli, F.S.; Wong, M.L.; Amabile, N.; Prasad, M.; Rassaf, T.; Ottaviani, J.I.; et al. Improvement of Endothelial Function with Dietary Flavanols Is Associated with Mobilization of Circulating Angiogenic Cells in Patients with Coronary Artery Disease. J. Am. Coll. Cardiol. 2010, 56, 218–224. [Google Scholar] [CrossRef] [PubMed]
  86. Marin, C.; Ramirez, R.; Delgado-Lista, J.; Yubero-Serrano, E.M.; Perez-Martinez, P.; Carracedo, J.; Garcia-Rios, A.; Rodriguez, F.; Gutierrez-Mariscal, F.M.; Gomez, P.; et al. Mediterranean Diet Reduces Endothelial Damage and Improves the Regenerative Capacity of Endothelium. Am. J. Clin. Nutr. 2011, 93, 267–274. [Google Scholar] [CrossRef]
  87. Yubero-Serrano, E.M.; Fernandez-Gandara, C.; Garcia-Rios, A.; Rangel-Zuñiga, O.A.; Gutierrez-Mariscal, F.M.; Torres-Peña, J.D.; Marin, C.; Lopez-Moreno, J.; Castaño, J.P.; Delgado-Lista, J.; et al. Mediterranean Diet and Endothelial Function in Patients with Coronary Heart Disease: An Analysis of the CORDIOPREV Randomized Controlled Trial. PLoS Med. 2020, 17, e1003282. [Google Scholar] [CrossRef]
  88. Fernández, J.M.; Rosado-Álvarez, D.; Da Silva Grigoletto, M.E.; Rangel-Zúñiga, O.A.; Landaeta-Díaz, L.L.; Caballero-Villarraso, J.; López-Miranda, J.; Pérez-Jiménez, F.; Fuentes-Jiménez, F. Moderate-to-High-Intensity Training and a Hypocaloric Mediterranean Diet Enhance Endothelial Progenitor Cells and Fitness in Subjects with the Metabolic Syndrome. Clin. Sci. 2012, 123, 361–373. [Google Scholar] [CrossRef]
  89. Spigoni, V.; Lombardi, C.; Cito, M.; Picconi, A.; Ridolfi, V.; Andreoli, R.; Anelli, N.; Gnudi, L.; Goldoni, M.; Zavaroni, I.; et al. N-3 PUFA Increase Bioavailability and Function of Endothelial Progenitor Cells. Food Funct. 2014, 5, 1881–1890. [Google Scholar] [CrossRef]
  90. Weech, M.; Altowaijri, H.; Mayneris-Perxachs, J.; Vafeiadou, K.; Madden, J.; Todd, S.; Jackson, K.G.; Lovegrove, J.A.; Yaqoob, P. Replacement of Dietary Saturated Fat with Unsaturated Fats Increases Numbers of Circulating Endothelial Progenitor Cells and Decreases Numbers of Microparticles: Findings from the Randomized, Controlled Dietary Intervention and VAScular Function (DIVAS) Study. Am. J. Clin. Nutr. 2018, 107, 876–882. [Google Scholar]
  91. Cesari, F.; Dinu, M.; Pagliai, G.; Rogolino, A.; Giusti, B.; Gori, A.M.; Casini, A.; Marcucci, R.; Sofi, F. Mediterranean, but Not Lacto-Ovo-Vegetarian, Diet Positively Influence Circulating Progenitor Cells for Cardiovascular Prevention: The CARDIVEG Study. Nutr. Metab. Cardiovasc. Dis. 2019, 29, 604–610. [Google Scholar] [CrossRef]
  92. Sereni, A.; Cesari, F.; Gori, A.M.; Maggini, N.; Marcucci, R.; Casini, A.; Sofi, F. Cardiovascular Benefits from Ancient Grain Bread Consumption: Findings from a Double-Blinded Randomized Crossover Intervention Trial. Int. J. Food Sci. Nutr. 2017, 68, 97–103. [Google Scholar] [CrossRef] [PubMed]
  93. Kim, W.; Jeong, M.H.; Cho, S.H.; Yun, J.H.; Chae, H.J.; Ahn, Y.K.; Lee, M.C.; Cheng, X.; Kondo, T.; Murohara, T.; et al. Effect of Green Tea Consumption on Endothelial Function and Circulating Endothelial Progenitor Cells in Chronic Smokers. Circ. J. 2006, 70, 1052–1057. [Google Scholar] [CrossRef] [PubMed]
  94. Chiva-Blanch, G.; Condines, X.; Magraner, E.; Roth, I.; Valderas-Martínez, P.; Arranz, S.; Casas, R.; Martínez-Huélamo, M.; Vallverdú-Queralt, A.; Quifer-Rada, P.; et al. The Non-Alcoholic Fraction of Beer Increases Stromal Cell Derived Factor 1 and the Number of Circulating Endothelial Progenitor Cells in High Cardiovascular Risk Subjects: A Randomized Clinical Trial. Atherosclerosis 2014, 233, 518–524. [Google Scholar] [CrossRef] [PubMed]
  95. Huang, P.-H.; Chen, Y.-H.; Tsai, H.-Y.; Chen, J.-S.; Wu, T.-C.; Lin, F.-Y.; Sata, M.; Chen, J.-W.; Lin, S.-J. Intake of Red Wine Increases the Number and Functional Capacity of Circulating Endothelial Progenitor Cells by Enhancing Nitric Oxide Bioavailability. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 869–877. [Google Scholar] [CrossRef] [PubMed]
  96. Roth, I.; Casas, R.; Medina-Remón, A.; Lamuela-Raventós, R.M.; Estruch, R. Consumption of Aged White Wine Modulates Cardiovascular Risk Factors via Circulating Endothelial Progenitor Cells and Inflammatory Biomarkers. Clin. Nutr. 2019, 38, 1036–1044. [Google Scholar] [CrossRef]
  97. Corban, M.T.; Widmer, R.J.; Cilluffo, R.; Kazeck, M.A.; Lennon, R.J.; Lerman, L.O.; Lerman, A. The Effect of Polyphenol-Rich Chardonnay Seed Supplements on Peripheral Endothelial Function. Eur. J. Nutr. 2020, 59, 3723–3734. [Google Scholar] [CrossRef]
  98. Shannon, O.M.; Stephan, B.C.; Minihane, A.-M.; Mathers, J.C.; Siervo, M. Nitric Oxide Boosting Effects of the Mediterranean Diet: A Potential Mechanism of Action. J. Gerontol. Ser. A 2018, 73, 902–904. [Google Scholar] [CrossRef]
  99. Medina-Remón, A.; Tresserra-Rimbau, A.; Pons, A.; Tur, J.A.; Martorell, M.; Ros, E.; Buil-Cosiales, P.; Sacanella, E.; Covas, M.I.; Corella, D.; et al. Effects of Total Dietary Polyphenols on Plasma Nitric Oxide and Blood Pressure in a High Cardiovascular Risk Cohort. The PREDIMED Randomized Trial. Nutr. Metab. Cardiovasc. Dis. 2015, 25, 60–67. [Google Scholar] [CrossRef]
  100. Shah, R.; Makarem, N.; Emin, M.; Liao, M.; Jelic, S.; Aggarwal, B. Mediterranean Diet Components Are Linked to Greater Endothelial Function and Lower Inflammation in a Pilot Study of Ethnically Diverse Women. Nutr. Res. 2020, 75, 77–84. [Google Scholar] [CrossRef]
  101. Galié, S.; García-Gavilán, J.; Papandreou, C.; Camacho-Barcía, L.; Arcelin, P.; Palau-Galindo, A.; Rabassa, A.; Bulló, M. Effects of Mediterranean Diet on Plasma Metabolites and Their Relationship with Insulin Resistance and Gut Microbiota Composition in a Crossover Randomized Clinical Trial. Clin. Nutr. 2021, 40, 3798–3806. [Google Scholar] [CrossRef]
  102. Aoki, A.; Murata, M.; Asano, T.; Ikoma, A.; Sasaki, M.; Saito, T.; Otani, T.; Jinbo, S.; Ikeda, N.; Kawakami, M.; et al. Prompt Increases in Retinol-Binding Protein 4 and Endothelial Progenitor Cells during Acute Exercise Load in Diabetic Subjects. Endocr. J. 2012, 59, 1085–1091. [Google Scholar] [CrossRef] [PubMed]
  103. Waclawovsky, G.; Umpierre, D.; Figueira, F.R.; De Lima, E.S.; Alegretti, A.P.; Schneider, L.; Matte, U.S.; Rodrigues, T.C.; Schaan, B.D. Exercise on Progenitor Cells in Healthy Subjects and Patients with Type 1 Diabetes. Med. Sci. Sports Exerc. 2016, 48, 190–199. [Google Scholar] [CrossRef] [PubMed]
  104. Taylor, G.S.; Shaw, A.; Smith, K.; Capper, T.E.; Scragg, J.H.; Cronin, M.; Bashir, A.; Flatt, A.; Campbell, M.D.; Stevenson, E.J.; et al. Type 1 Diabetes Patients Increase CXCR4+ and CXCR7+ Haematopoietic and Endothelial Progenitor Cells with Exercise, but the Response Is Attenuated. Sci. Rep. 2021, 11, 14502. [Google Scholar] [CrossRef] [PubMed]
  105. Bruyndonckx, L.; Hoymans, V.Y.; De Guchtenaere, A.; Van Helvoirt, M.; Van Craenenbroeck, E.M.; Frederix, G.; Lemmens, K.; Vissers, D.K.; Vrints, C.J.; Ramet, J.; et al. Diet, Exercise, and Endothelial Function in Obese Adolescents. Pediatrics 2015, 135, e653–e661. [Google Scholar] [CrossRef] [PubMed]
  106. Eleuteri, E.; Mezzani, A.; Di Stefano, A.; Vallese, D.; Gnemmi, I.; Delle Donne, L.; Taddeo, A.; Della Bella, S.; Giannuzzi, P. Aerobic Training and Angiogenesis Activation in Patients with Stable Chronic Heart Failure: A Preliminary Report. Biomarkers 2013, 18, 418–424. [Google Scholar] [CrossRef]
  107. Möbius-Winkler, S.; Hilberg, T.; Menzel, K.; Golla, E.; Burman, A.; Schuler, G.; Adams, V. Time-Dependent Mobilization of Circulating Progenitor Cells during Strenuous Exercise in Healthy Individuals. J. Appl. Physiol. 2009, 107, 1943–1950. [Google Scholar] [CrossRef]
  108. Manfredini, F.; Rigolin, G.; Malagoni, A.; Catizone, L.; Mandini, S.; Sofritti, O.; Mauro, E.; Soffritti, S.; Boari, B.; Cuneo, A.; et al. Exercise Training and Endothelial Progenitor Cells in Haemodialysis Patients. J. Int. Med. Res. 2009, 37, 534–540. [Google Scholar] [CrossRef]
  109. Kaźmierski, M.; Wojakowski, W.; Michalewska-W\ludarczyk, A.; Podolecka, E.; Kotowski, M.; Machaliński, B.; Tendera, M. Exercise-Induced Mobilisation of Endothelial Progenitor Cells in Patients with Premature Coronary Heart Disease. Kardiol. Pol. Pol. Heart J. 2015, 73, 411–418. [Google Scholar] [CrossRef]
  110. Bajer, B.; Vlcek, M.; Galusova, A.; Imrich, R.; Penesova, A. Exercise Associated Hormonal Signals as Powerful Determinants of an Effective Fat Mass Loss. Endocr. Regul. 2015, 49, 151–163. [Google Scholar] [CrossRef]
  111. Dyakova, E.Y.; Kapilevich, L.V.; Shylko, V.G.; Popov, S.V.; Anfinogenova, Y. Physical Exercise Associated with NO Production: Signaling Pathways and Significance in Health and Disease. Front. Cell Dev. Biol. 2015, 3, 19. [Google Scholar] [CrossRef]
  112. Benowitz, N.L.; Burbank, A.D. Cardiovascular Toxicity of Nicotine: Implications for Electronic Cigarette Use. Trends Cardiovasc. Med. 2016, 26, 515–523. [Google Scholar] [CrossRef] [PubMed]
  113. Kondo, T.; Nakano, Y.; Adachi, S.; Murohara, T. Effects of Tobacco Smoking on Cardiovascular Disease. Circ. J. 2019, 83, 1980–1985. [Google Scholar] [CrossRef] [PubMed]
  114. Antoniewicz, L.; Bosson, J.A.; Kuhl, J.; Abdel-Halim, S.M.; Kiessling, A.; Mobarrez, F.; Lundbäck, M. Electronic Cigarettes Increase Endothelial Progenitor Cells in the Blood of Healthy Volunteers. Atherosclerosis 2016, 255, 179–185. [Google Scholar] [CrossRef] [PubMed]
  115. Liao, Y.; Chen, L.-L.; Zeng, T.; Li, Y.; Yu, F.; Hu, L.; Yue, L. Number of Circulating Endothelial Progenitor Cells as a Marker of Vascular Endothelial Function for Type 2 Diabetes. Vasc. Med. 2010, 15, 279–285. [Google Scholar] [CrossRef] [PubMed]
  116. Ahmed, F.W.; Rider, R.; Glanville, M.; Narayanan, K.; Razvi, S.; Weaver, J.U. Metformin Improves Circulating Endothelial Cells and Endothelial Progenitor Cells in Type 1 Diabetes: MERIT Study. Cardiovasc. Diabetol. 2016, 15, 116. [Google Scholar] [CrossRef] [PubMed]
  117. Chen, L.; Liao, Y.; Zeng, T.; Yu, F.; Li, H.; Feng, Y. Effects of Metformin plus Gliclazide Compared with Metformin Alone on Circulating Endothelial Progenitor Cell in Type 2 Diabetic Patients. Endocrine 2010, 38, 266–275. [Google Scholar] [CrossRef] [PubMed]
  118. Chen, L.-L.; Yu, F.; Zeng, T.; Liao, Y.; Li, Y.; Ding, H. Effects of Gliclazide on Endothelial Function in Patients with Newly Diagnosed Type 2 Diabetes. Eur. J. Pharmacol. 2011, 659, 296–301. [Google Scholar] [CrossRef]
  119. Dei Cas, A.; Spigoni, V.; Cito, M.; Aldigeri, R.; Ridolfi, V.; Marchesi, E.; Marina, M.; Derlindati, E.; Aloe, R.; Bonadonna, R.C.; et al. Vildagliptin, but Not Glibenclamide, Increases Circulating Endothelial Progenitor Cell Number: A 12-Month Randomized Controlled Trial in Patients with Type 2 Diabetes. Cardiovasc. Diabetol. 2017, 16, 27. [Google Scholar] [CrossRef]
  120. Aso, Y.; Jojima, T.; Iijima, T.; Suzuki, K.; Terasawa, T.; Fukushima, M.; Momobayashi, A.; Hara, K.; Takebayashi, K.; Kasai, K.; et al. Sitagliptin, a Dipeptidyl Peptidase-4 Inhibitor, Increases the Number of Circulating CD34+ CXCR4+ Cells in Patients with Type 2 Diabetes. Endocrine 2015, 50, 659–664. [Google Scholar] [CrossRef]
  121. Esposito, K.; Maiorino, M.; Di Palo, C.; Gicchino, M.; Petrizzo, M.; Bellastella, G.; Saccomanno, F.; Giugliano, D. Effects of Pioglitazone versus Metformin on Circulating Endothelial Microparticles and Progenitor Cells in Patients with Newly Diagnosed Type 2 Diabetes—A Randomized Controlled Trial. Diabetes Obes. Metab. 2011, 13, 439–445. [Google Scholar] [CrossRef]
  122. Makino, H.; Okada, S.; Nagumo, A.; Sugisawa, T.; Miyamoto, Y.; Kishimoto, I.; Akie, T.K.; Soma, T.; Taguchi, A.; Yoshimasa, Y. Pioglitazone Treatment Stimulates Circulating CD34-Positive Cells in Type 2 Diabetes Patients. Diabetes Res. Clin. Pract. 2008, 81, 327–330. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, C.-H.; Ting, M.-K.; Verma, S.; Kuo, L.-T.; Yang, N.-I.; Hsieh, I.-C.; Wang, S.-Y.; Hung, A.; Cherng, W.-J. Pioglitazone Increases the Numbers and Improves the Functional Capacity of Endothelial Progenitor Cells in Patients with Diabetes Mellitus. Am. Heart J. 2006, 152, 1051.e1–1051.e8. [Google Scholar] [CrossRef] [PubMed]
  124. Werner, C.; Kamani, C.H.; Gensch, C.; Bohm, M.; Laufs, U. The Peroxisome Proliferator–Activated Receptor-γ Agonist Pioglitazone Increases Number and Function of Endothelial Progenitor Cells in Patients with Coronary Artery Disease and Normal Glucose Tolerance. Diabetes 2007, 56, 2609–2615. [Google Scholar] [CrossRef] [PubMed]
  125. Kampoli, A.-M.; Tousoulis, D.; Pallantza, Z.; Paterakis, G.; Papageorgiou, N.; Oikonomou, E.; Miliou, A.; Vlachopoulou, A.; Stefanadis, C. Comparable Effects of Pioglitazone and Perindopril on Circulating Endothelial Progenitor Cells, Inflammatory Process and Oxidative Stress in Patients with Diabetes Mellitus. Int. J. Cardiol. 2012, 157, 413–415. [Google Scholar] [CrossRef]
  126. Fadini, G.P.; Boscaro, E.; Albiero, M.; Menegazzo, L.; Frison, V.; De Kreutzenberg, S.; Agostini, C.; Tiengo, A.; Avogaro, A. The Oral Dipeptidyl Peptidase-4 Inhibitor Sitagliptin Increases Circulating Endothelial Progenitor Cells in Patients with Type 2 Diabetes: Possible Role of Stromal-Derived Factor-1α. Diabetes Care 2010, 33, 1607–1609. [Google Scholar] [CrossRef]
  127. Xu, M.; Zhao, X.; Zheng, M.; Deng, D.; Zhang, S.; Wang, Y.; Fang, Z.; Chen, M. Acute Effects of Sitagliptin on Progenitor Cells and Soluble Mediators in Newly Diagnosed Type 2 Diabetes. Int. J. Clin. Pharmacol. Ther. 2020, 58, 491. [Google Scholar] [CrossRef]
  128. Nakamura, K.; Oe, H.; Kihara, H.; Shimada, K.; Fukuda, S.; Watanabe, K.; Takagi, T.; Yunoki, K.; Miyoshi, T.; Hirata, K.; et al. DPP-4 Inhibitor and Alpha-Glucosidase Inhibitor Equally Improve Endothelial Function in Patients with Type 2 Diabetes: EDGE Study. Cardiovasc. Diabetol. 2014, 13, 110. [Google Scholar] [CrossRef]
  129. Fadini, G.P.; Bonora, B.M.; Cappellari, R.; Menegazzo, L.; Vedovato, M.; Iori, E.; Marescotti, M.C.; Albiero, M.; Avogaro, A. Acute Effects of Linagliptin on Progenitor Cells, Monocyte Phenotypes, and Soluble Mediators in Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2016, 101, 748–756. [Google Scholar] [CrossRef]
  130. De Boer, S.A.; Reijrink, M.; Abdulahad, W.H.; Hoekstra, E.S.; Slart, R.H.; Heerspink, H.J.; Westra, J.; Mulder, D.J. Angiogenic T Cells Are Decreased in People with Type 2 Diabetes Mellitus and Recruited by the Dipeptidyl Peptidase-4 Inhibitor Linagliptin: A Subanalysis from a Randomized, Placebo-Controlled Trial (RELEASE Study). Diabetes Obes. Metab. 2020, 22, 1220–1225. [Google Scholar] [CrossRef]
  131. Awal, H.B.; Nandula, S.R.; Domingues, C.C.; Dore, F.J.; Kundu, N.; Brichacek, B.; Fakhri, M.; Elzarki, A.; Ahmadi, N.; Safai, S.; et al. Linagliptin, When Compared to Placebo, Improves CD34+ ve Endothelial Progenitor Cells in Type 2 Diabetes Subjects with Chronic Kidney Disease Taking Metformin and/or Insulin: A Randomized Controlled Trial. Cardiovasc. Diabetol. 2020, 19, 72. [Google Scholar] [CrossRef]
  132. Negro, R.; Greco, E.L.; Greco, G. Alogliptin and Gliclazide Similarly Increase Circulating Endothelial Progenitor Cells in Type 2 Diabetes Patients. Exp. Clin. Endocrinol. Diabetes 2019, 127, 215–219. [Google Scholar] [CrossRef] [PubMed]
  133. Li, F.; Chen, J.; Leng, F.; Lu, Z.; Ling, Y. Effect of Saxagliptin on Circulating Endothelial Progenitor Cells and Endothelial Function in Newly Diagnosed Type 2 Diabetic Patients. Exp. Clin. Endocrinol. Diabetes 2017, 125, 400–407. [Google Scholar] [CrossRef] [PubMed]
  134. Dore, F.J.; Domingues, C.C.; Ahmadi, N.; Kundu, N.; Kropotova, Y.; Houston, S.; Rouphael, C.; Mammadova, A.; Witkin, L.; Khiyami, A.; et al. The Synergistic Effects of Saxagliptin and Metformin on CD34+ Endothelial Progenitor Cells in Early Type 2 Diabetes Patients: A Randomized Clinical Trial. Cardiovasc. Diabetol. 2018, 17, 65. [Google Scholar] [CrossRef] [PubMed]
  135. Akashi, N.; Umemoto, T.; Yamada, H.; Fujiwara, T.; Yamamoto, K.; Taniguchi, Y.; Sakakura, K.; Wada, H.; Momomura, S.; Fujita, H. Teneligliptin, a DPP-4 Inhibitor, Improves Vascular Endothelial Function via Divergent Actions Including Changes in Circulating Endothelial Progenitor Cells. Diabetes Metab. Syndr. Obes. 2023, 16, 1043–1054. [Google Scholar] [CrossRef]
  136. De Ciuceis, C.; Agabiti-Rosei, C.; Rossini, C.; Caletti, S.; Coschignano, M.A.; Ferrari-Toninelli, G.; Ragni, G.; Cappelli, C.; Cerudelli, B.; Airò, P.; et al. Microvascular Density and Circulating Endothelial Progenitor Cells before and after Treatment with Incretin Mimetics in Diabetic Patients. High Blood Press. Cardiovasc. Prev. 2018, 25, 369–378. [Google Scholar] [CrossRef]
  137. Ahmad, E.; Waller, H.L.; Sargeant, J.A.; Webb, M.A.; Htike, Z.Z.; McCann, G.P.; Gulsin, G.; Khunti, K.; Yates, T.; Henson, J.; et al. Effects of Liraglutide versus Sitagliptin on Circulating Cardiovascular Biomarkers, Including Circulating Progenitor Cells, in Individuals with Type 2 Diabetes and Obesity: Analyses from the LYDIA Trial. Diabetes Obes. Metab. 2021, 23, 1409–1414. [Google Scholar] [CrossRef]
  138. Xie, D.; Li, Y.; Xu, M.; Zhao, X.; Chen, M. Effects of Dulaglutide on Endothelial Progenitor Cells and Arterial Elasticity in Patients with Type 2 Diabetes Mellitus. Cardiovasc. Diabetol. 2022, 21, 200. [Google Scholar] [CrossRef]
  139. Bonora, B.M.; Cappellari, R.; Albiero, M.; Avogaro, A.; Fadini, G.P. Effects of SGLT2 Inhibitors on Circulating Stem and Progenitor Cells in Patients with Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2018, 103, 3773–3782. [Google Scholar] [CrossRef]
  140. Luo, L.; Dong, B.; Zhang, J.; Qiu, Y.; Liu, X.; Zhou, Z.; He, J.; Zhang, X.; Chen, L.; Xia, W. Dapagliflozin Restores Diabetes-Associated Decline in Vasculogenic Capacity of Endothelial Progenitor Cells via Activating AMPK-Mediated Inhibition of Inflammation and Oxidative Stress. Biochem. Biophys. Res. Commun. 2023, 671, 205–214. [Google Scholar] [CrossRef]
  141. Hess, D.A.; Terenzi, D.C.; Trac, J.Z.; Quan, A.; Mason, T.; Al-Omran, M.; Bhatt, D.L.; Dhingra, N.; Rotstein, O.D.; Leiter, L.A.; et al. SGLT2 Inhibition with Empagliflozin Increases Circulating Provascular Progenitor Cells in People with Type 2 Diabetes Mellitus. Cell Metab. 2019, 30, 609–613. [Google Scholar] [CrossRef]
  142. Nandula, S.R.; Kundu, N.; Awal, H.B.; Brichacek, B.; Fakhri, M.; Aimalla, N.; Elzarki, A.; Amdur, R.L.; Sen, S. Role of Canagliflozin on Function of CD34+ ve Endothelial Progenitor Cells (EPC) in Patients with Type 2 Diabetes. Cardiovasc. Diabetol. 2021, 20, 44. [Google Scholar] [CrossRef]
  143. Fadini, G.; De Kreutzenberg, S.; Mariano, V.; Boscaro, E.; Bertolini, F.; Mancuso, P.; Quarna, J.; Marescotti, M.; Agostini, C.; Tiengo, A.; et al. Optimized Glycaemic Control Achieved with Add-on Basal Insulin Therapy Improves Indexes of Endothelial Damage and Regeneration in Type 2 Diabetic Patients with Macroangiopathy: A Randomized Crossover Trial Comparing Detemir versus Glargine. Diabetes Obes. Metab. 2011, 13, 718–725. [Google Scholar] [CrossRef] [PubMed]
  144. Oikonomou, D.; Kopf, S.; von Bauer, R.; Djuric, Z.; Cebola, R.; Sander, A.; Englert, S.; Vittas, S.; Hidmark, A.; Morcos, M.; et al. Influence of Insulin and Glargine on Outgrowth and Number of Circulating Endothelial Progenitor Cells in Type 2 Diabetes Patients: A Partially Double-Blind, Randomized, Three-Arm Unicenter Study. Cardiovasc. Diabetol. 2014, 13, 137. [Google Scholar] [CrossRef] [PubMed]
  145. Piatti, P.M.; Marone, E.; Mantero, M.; Setola, E.; Galluccio, E.; Lucotti, P.; Shehaj, E.; Villa, V.; Perticone, F.; Venturini, M.; et al. Effect of Normalization of Fasting Glucose by Intensified Insulin Therapy and Influence of eNOS Polymorphisms on the Incidence of Restenosis after Peripheral Angioplasty in Patients with Type 2 Diabetes: A Randomized, Open-Label Clinical Trial. Acta Diabetol. 2013, 50, 373–382. [Google Scholar] [CrossRef] [PubMed]
  146. UK Prospective Diabetes Study (UKPDS) Group. Effect of Intensive Blood-Glucose Control with Metformin on Complications in Overweight Patients with Type 2 Diabetes (UKPDS 34). Lancet 1998, 352, 854–865. [Google Scholar] [CrossRef]
  147. Yu, J.-W.; Deng, Y.-P.; Han, X.; Ren, G.-F.; Cai, J.; Jiang, G.-J. Metformin Improves the Angiogenic Functions of Endothelial Progenitor Cells via Activating AMPK/eNOS Pathway in Diabetic Mice. Cardiovasc. Diabetol. 2016, 15, 88. [Google Scholar] [CrossRef]
  148. Horsdal, H.T.; Johnsen, S.P.; Søndergaard, F.; Jacobsen, J.; Thomsen, R.W.; Schmitz, O.; Sørensen, H.T.; Rungby, J. Sulfonylureas and Prognosis after Myocardial Infarction in Patients with Diabetes: A Population-Based Follow-up Study. Diabetes Metab. Res. Rev. 2009, 25, 515–522. [Google Scholar] [CrossRef]
  149. Johnsen, S.P.; Monster, T.; Olsen, M.L.; Thisted, H.; McLaughlin, J.; Sørensen, H.T.; Lervang, H.; Rungby, J. Risk and Short-Term Prognosis of Myocardial Infarction among Users of Antidiabetic Drugs. Am. J. Ther. 2006, 13, 134–140. [Google Scholar] [CrossRef]
  150. Filipova, E.; Uzunova, K.; Kalinov, K.; Vekov, T. Effects of Pioglitazone Therapy on Blood Parameters, Weight and BMI: A Meta-Analysis. Diabetol. Metab. Syndr. 2017, 9, 90. [Google Scholar] [CrossRef]
  151. Sarafidis, P.A.; Georgianos, P.I.; Lasaridis, A.N. PPAR-γ Agonism for Cardiovascular and Renal Protection. Cardiovasc. Ther. 2011, 29, 377–384. [Google Scholar] [CrossRef]
  152. Lehrke, M.; Marx, N. Diabetes Mellitus and Heart Failure. Am. J. Cardiol. 2017, 120, S37–S47. [Google Scholar] [CrossRef] [PubMed]
  153. Sheikh, I.M.; Hassan, O.A.; Adam, S.M.; Ali, A.I.; Ogedegbe, O.J.; Tabowei, G.; Barbarawi, A.; Yussuf, F.M.; Nor, M.A.; Adam, S.M.; et al. Association of Pioglitazone with Major Adverse Cardiovascular Events, All-Cause Mortality, and Heart Failure Hospitalizations: A Systematic Review. Cureus 2023, 15, e46911. [Google Scholar] [CrossRef] [PubMed]
  154. Erdmann, E.; Dormandy, J.; Wilcox, R.; Massi-Benedetti, M.; Charbonnel, B. PROactive 07: Pioglitazone in the Treatment of Type 2 Diabetes: Results of the PROactive Study. Vasc. Health Risk Manag. 2007, 3, 355–370. [Google Scholar] [PubMed]
  155. Wilcox, R.; Kupfer, S.; Erdmann, E.; PROactive Study Investigators. Effects of Pioglitazone on Major Adverse Cardiovascular Events in High-Risk Patients with Type 2 Diabetes: Results from PROspective pioglitAzone Clinical Trial in Macro Vascular Events (PROactive 10). Am. Heart J. 2008, 155, 712–717. [Google Scholar] [CrossRef]
  156. Sorrentino, S.A.; Bahlmann, F.H.; Besler, C.; Müller, M.; Schulz, S.; Kirchhoff, N.; Doerries, C.; Horváth, T.; Limbourg, A.; Limbourg, F.; et al. Oxidant Stress Impairs in Vivo Reendothelialization Capacity of Endothelial Progenitor Cells from Patients with Type 2 Diabetes Mellitus: Restoration by the Peroxisome Proliferator-Activated Receptor-γ Agonist Rosiglitazone. Circulation 2007, 116, 163–173. [Google Scholar] [CrossRef]
  157. Green, J.B.; Bethel, M.A.; Armstrong, P.W.; Buse, J.B.; Engel, S.S.; Garg, J.; Josse, R.; Kaufman, K.D.; Koglin, J.; Korn, S.; et al. Effect of Sitagliptin on Cardiovascular Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2015, 373, 232–242. [Google Scholar] [CrossRef]
  158. Rosenstock, J.; Kahn, S.E.; Johansen, O.E.; Zinman, B.; Espeland, M.A.; Woerle, H.J.; Pfarr, E.; Keller, A.; Mattheus, M.; Baanstra, D.; et al. Effect of Linagliptin vs Glimepiride on Major Adverse Cardiovascular Outcomes in Patients with Type 2 Diabetes: The CAROLINA Randomized Clinical Trial. JAMA 2019, 322, 1155–1166. [Google Scholar] [CrossRef]
  159. Scirica, B.M.; Bhatt, D.L.; Braunwald, E.; Steg, P.G.; Davidson, J.; Hirshberg, B.; Ohman, P.; Frederich, R.; Wiviott, S.D.; Hoffman, E.B.; et al. Saxagliptin and Cardiovascular Outcomes in Patients with Type 2 Diabetes Mellitus. N. Engl. J. Med. 2013, 369, 1317–1326. [Google Scholar] [CrossRef]
  160. White, W.B.; Bakris, G.L.; Bergenstal, R.M.; Cannon, C.P.; Cushman, W.C.; Fleck, P.; Heller, S.; Mehta, C.; Nissen, S.E.; Perez, A.; et al. EXamination of cArdiovascular outcoMes with alogliptIN versus Standard of carE in Patients with Type 2 Diabetes Mellitus and Acute Coronary Syndrome (EXAMINE): A Cardiovascular Safety Study of the Dipeptidyl Peptidase 4 Inhibitor Alogliptin in Patients with Type 2 Diabetes with Acute Coronary Syndrome. Am. Heart J. 2011, 162, 620–626. [Google Scholar]
  161. Zhong, J.; Rajagopalan, S. Dipeptidyl Peptidase-4 Regulation of SDF-1/CXCR4 Axis: Implications for Cardiovascular Disease. Front. Immunol. 2015, 6, 477. [Google Scholar] [CrossRef]
  162. Marso, S.P.; Daniels, G.H.; Brown-Frandsen, K.; Kristensen, P.; Mann, J.F.; Nauck, M.A.; Nissen, S.E.; Pocock, S.; Poulter, N.R.; Ravn, L.S.; et al. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2016, 375, 311–322. [Google Scholar] [CrossRef] [PubMed]
  163. Gerstein, H.C.; Colhoun, H.M.; Dagenais, G.R.; Diaz, R.; Lakshmanan, M.; Pais, P.; Probstfield, J.; Riesmeyer, J.S.; Riddle, M.C.; Rydén, L.; et al. Dulaglutide and Cardiovascular Outcomes in Type 2 Diabetes (REWIND): A Double-Blind, Randomised Placebo-Controlled Trial. Lancet 2019, 394, 121–130. [Google Scholar] [CrossRef]
  164. Marso, S.P.; Bain, S.C.; Consoli, A.; Eliaschewitz, F.G.; Jódar, E.; Leiter, L.A.; Lingvay, I.; Rosenstock, J.; Seufert, J.; Warren, M.L.; et al. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N. Engl. J. Med. 2016, 375, 1834–1844. [Google Scholar] [CrossRef] [PubMed]
  165. Holman, R.R.; Bethel, M.A.; Mentz, R.J.; Thompson, V.P.; Lokhnygina, Y.; Buse, J.B.; Chan, J.C.; Choi, J.; Gustavson, S.M.; Iqbal, N.; et al. Effects of Once-Weekly Exenatide on Cardiovascular Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2017, 377, 1228–1239. [Google Scholar] [CrossRef] [PubMed]
  166. Zinman, B.; Wanner, C.; Lachin, J.M.; Fitchett, D.; Bluhmki, E.; Hantel, S.; Mattheus, M.; Devins, T.; Johansen, O.E.; Woerle, H.J.; et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N. Engl. J. Med. 2015, 373, 2117–2128. [Google Scholar] [CrossRef] [PubMed]
  167. Ferdinand, K.C.; Arora, N. SGLT2 Inhibition for Outcomes: Is This the Panacea? Am. Heart J. Plus Cardiol. Res. Pract. 2022, 21, 100159. [Google Scholar] [CrossRef]
Table 1. Characteristics of distinct EPC subtypes in cultures [13].
Table 1. Characteristics of distinct EPC subtypes in cultures [13].
LineageMyeloid Angiogenic CellsEndothelial Colony-Forming Cells
positive cell markersCD45, CD31, CD14CD31, CD105, CD 146
VE-cadherin, von Willebrand factor, VEGFR2
CD34+/−
negative cell markersCD146, CD34CD45, CD14
in vitro effectsconditioned media necessary for the endothelial formationintrinsic tube forming capacity
functionprovide paracrine angiogenic factorsprovide cells as building blocks
release of paracrine factors
time of appearance in cultureearlylate
Table 2. Currently available anti-diabetic medications, their main mode of action, cardiovascular effects, and effects on EPCs.
Table 2. Currently available anti-diabetic medications, their main mode of action, cardiovascular effects, and effects on EPCs.
Drug ClassMode of ActionCardiovascular EffectsEffects on EPCsReference
Biguanides:
Metformin
reduces hepatic glucose production
facilitates peripheral glucose uptake and utilization, in part by increasing insulin action
reduces basal hyperinsulinemia
alters glucose turnover in the gut
increases glucose uptake from circulation and decreases absorption from food
increases the release of glucagon-like peptide-1 (GLP-1)
alters the gut microbiome
activates adenosine monophosphate-protein-kinase (AMPK) activator and increases the transport capacity of all types of membrane glucose transporters (GLUTs)
beneficial effects
not proven in CVOTs
↑EPC (CD34+/VEGFR2+/CD45-/dim) count assessed by flow cytometry
↑FMD



[115]


↑EPC
(CD34+/VEGFR2+/CD45-/dim) count assessed by flow cytometry
↑ECFC colonies number
↑adhesion capability of proangiogenic cells using fibronectin adhesion assay
[116] *
↑EPC (CD34+/VEGFR2+/CD45-/dim) count assessed by flow cytometry
[117]
1Sulphonylureas:
Gliclazide
Glimepiride
Gliquidon
stimulates insulin secretion from the β-cells of the islets of Langerhans
increases insulin and C-peptide secretion
second generation sulphonylureas are superior to first generation
finding not proven in CVOTs
gliclazide only:
↑EPC (CD34+/VEGFR2+/CD45-/dim) count assessed by flow cytometry
↑FMD

[118]
no proven effects for other sulphonylureas[119,120]
Thiazolidinediones:
Pioglitazone
Rosiglitazone
reduces insulin resistance and reduces insulin concentrations
activates peroxisome proliferator-activated receptor gamma
increases insulin sensitivity of liver, fat, and skeletal muscle cells
reduces hepatic glucose output
increases peripheral glucose disposal
improves some MACEs
not proven in CVOTs
increased risk of heart failure
pioglitazone:
↑EPC (CD34+/VEGFR2+) count assessed by flow cytometry

pioglitazone:

[121]

↑ circulating CD34+ cell count[122]
pioglitazone:
↑EPC (CD34+) count assessed by flow cytometry
↑increased migratory response and adhesion capacity to fibronectin and collagen in culture
[123]
pioglitazone:
↑EPC (CD34+/VEGFR2+) count assessed by flow cytometry
↑ SDF1 induced migratory capacity
↑ ECFC in cultures
[124]
pioglitazone:
no effect on EPC count
[125]
DPP-4 inhibitors
Sitagliptin
Linagliptin
Alogliptin
Saxagliptin
Vildagliptin
Teneligliptin
inhibits dipeptidyl peptidase 4 (DPP-4)
enhances the levels of glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) in a glucose-dependent manner
improves beta cell responsiveness to glucose and stimulates insulin biosynthesis and release
lowers glucagon secretion
reduces hepatic glucose production
non-inferiority to standard treatment/class effect
proven in CVOTs except for vildagliptin and teneligliptin
increased risk for heart failure for saxagliptin
sitagliptin:
↑EPC (CD34+/VEGFR2+) count assessed by flow cytometry
↑ SDF1 blood concentrations
[126]
sitagliptin:
↑EPC (CD34+/CXCR4+) count assessed by flow cytometry
↓ SDF1 blood concentrations
[120]
sitagliptin:
↑EPC (CD34+/VEGFR2+ and CD34+/VEGFR2+/CD133+) count assessed by flow cytometry
↑GLP-1, NO and SDF-1 blood concentrations
[127]
sitagliptin:
↑EPC (CD34+) count assessed by flow cytometry
↑FMD
[128]
linagliptin:
↑EPC (CD34+/VEGFR2+ and CD34+/CD133+) count
↑GLP-1, and SDF-1 blood concentrations
[129]
linalgiptin
no effect on EPCs
↑SDF-1 blood concentrations
[130]
linagliptin↑CD34+/CD184+ EPC
improved arterial stiffness and pulse wave velocity
[131]
vildagliptin:
↑EPC (CD34+/VEGFR2 +/CD133+) count assessed by flow cytometry
↓SDF1 blood concentrations
[119]
alogliptin:
↑EPC (CD34+/VEGFR2 +/CD45-/dim) count assessed by flow cytometry
[132]
saxagliptin:
↑EPC (CD34+/VEGFR +/CD133+) count assessed by flow cytometry
↑FMD
[133]
saxagliptin:
no effect on EPC if added to metformin
improved migratory capacity
[134]
teneligliptin:
no significant effect on EPC
↑FMD
[135]
GLP-1 receptor agonists
Exenatide
Lixisenatide
Liraglutide
Dulaglutide
Semaglutide
activates the GLP-1 receptor
stimulates insulin secretion and lowers glucagon secretion in a glucose-dependent manner
delays gastric emptying in the early postprandial phase.
liraglutide, dulaglutide, and semaglutide showed cardiovascular superiority in CVOTs or equivalent studiesexenatide in full dose superior to medium dosed liraglutide in ↑EPC (CD34+/VEGFR2 +)
[136]
liraglutide superior to sitagliptin in ↑VEGF and SDF-1[137]
dulaglutide:
↑EPC (CD34+/VEGFR2 +/CD133+)
enhanced EPC proliferation, adhesion, migration, and tubule formation abilities
improved brachial-ankle pulse wave velocity
[138]
SGLT-2 inhibitors
Empagliflozin
Dapagliflozin
Canagliflozin
inhibits the sodium-glucose cotransporter-2 by dapagliflozin in the proximal renal tubule
improves both fasting and postprandial plasma glucose levels
cardiovascular superiority proven in CVOTs
beneficial in heart failure and renal failure
black box warning for lower limb amputations
dapagliflozin lead to a late increase in EPC count (CD34+/VEGFR2+)
no change in EPCs in the empagliflozin group
[139]
dapagliflozin treatment activated AMPK signaling in EPCs[140]
empagliflozin increases CD133+EPC count[141]
canagliflozin improved CXCR receptors on EPCs, improving their migratory capacity[142]
Insulin and insulin analogsbinds to insulin receptorsproven non-inferiority to standard therapy in CVOTs for newer insulin analogsdetemir and glargin both increased EPC count (CD34+/VEGFR2+ and CD34+/VEGFR2+/CD133+)
decreased adhesion molecules

[143]
no difference in CD34*/VEGFR2+ EPCs between groups receiving NPH insulin, insulin glargin, or oral therapy
improved ECFC growth with NPH insulin and glargin
decrease in intima media thickness
[144]
intensive insulin therapy increased EPC count (CD34+/VEGFR2+)
no effect on clinical outcomes
[145]
reduced glucovariability by using insulin pumps increased the EPC count (CD34+/VEGFR+) compared to intensified insulin therapy[58] *
* indicates studies of patients with type 1 diabetes, ↑ increased, ↓ reduced.
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

Altabas, V.; Marinković Radošević, J.; Špoljarec, L.; Uremović, S.; Bulum, T. The Impact of Modern Anti-Diabetic Treatment on Endothelial Progenitor Cells. Biomedicines 2023, 11, 3051. https://doi.org/10.3390/biomedicines11113051

AMA Style

Altabas V, Marinković Radošević J, Špoljarec L, Uremović S, Bulum T. The Impact of Modern Anti-Diabetic Treatment on Endothelial Progenitor Cells. Biomedicines. 2023; 11(11):3051. https://doi.org/10.3390/biomedicines11113051

Chicago/Turabian Style

Altabas, Velimir, Jelena Marinković Radošević, Lucija Špoljarec, Stella Uremović, and Tomislav Bulum. 2023. "The Impact of Modern Anti-Diabetic Treatment on Endothelial Progenitor Cells" Biomedicines 11, no. 11: 3051. https://doi.org/10.3390/biomedicines11113051

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