The endothelium is a thin layer of cells lining the interior surface of blood vessels, playing a crucial physiological role in maintaining vascular homeostasis. It regulates vasodilation and vasoconstriction through the release of nitric oxide (NO) and other factors, acts as a selective barrier controlling vascular permeability, and promotes an antithrombotic and anti-inflammatory environment by expressing anticoagulant proteins like thrombomodulin and inhibiting adhesion molecules.[1]Endothelial dysfunction is a pathophysiological state characterized by impaired endothelial function, primarily involving reduced bioavailability of nitric oxide (NO) and an imbalance between endothelium-derived relaxing and contracting factors, which leads to diminished vasodilation, increased vascular permeability, and a prothrombotic and proinflammatory phenotype.[2] This condition represents an early and reversible marker of vascular injury, often preceding overt atherosclerosis and serving as a key mediator in the initiation and progression of cardiovascular diseases.[3]Endothelial dysfunction is associated with various cardiovascular and systemic conditions and can be assessed through methods like flow-mediated dilation. Therapeutic interventions, such as lifestyle modifications and pharmacological agents, may restore endothelial function and mitigate disease progression.[1]
Overview
Definition
The endothelium is a thin monolayer of specialized endothelial cells that lines the interior surface of all blood vessels, including arteries, veins, and capillaries, as well as the chambers of the heart and the lymphatic system.[4] This cellular layer serves as a dynamic interface between the bloodstream and surrounding tissues, regulating vascular homeostasis through the production of various vasoactive, anti-inflammatory, and anticoagulant factors.[5]Endothelial dysfunction is defined as a pathological shift in endothelial function characterized by impaired vasodilation, a pro-inflammatory state, enhanced prothrombotic properties, and diminished barrier integrity that allows increased vascular permeability.[6] This imbalance often stems from reduced bioavailability of endothelium-derived relaxing factors, such as nitric oxide, leading to a predominance of vasoconstrictive, inflammatory, and thrombogenic signals.[2] Unlike normal endothelial physiology, which promotes vascular relaxation and protection, dysfunction represents an early deviation that can precede overt vascular disease.[7]The term endothelial dysfunction emerged in the 1980s amid research linking endothelial alterations to atherosclerosis, with a pivotal observation in 1986 by Ludmer et al., who reported paradoxical vasoconstriction in response to acetylcholine in human atherosclerotic coronary arteries, highlighting the failure of endothelium-dependent relaxation—a concept advanced by Vanhoutte's foundational studies on endothelial control of vascular tone.[8] This functional impairment is generally reversible through interventions targeting underlying risk factors, distinguishing it from endothelial injury, which involves irreversible structural damage to the cellular layer, such as denudation or apoptosis.[7]
Physiological Role
The vascular endothelium, a monolayer of cells lining the interior of blood vessels, serves as a dynamic interface between the bloodstream and surrounding tissues, performing essential functions that maintain cardiovascular homeostasis. One of its primary roles is barrier regulation, where it controls selective permeability to allow the passage of nutrients, waste products, and immune cells while restricting larger molecules and pathogens. This is achieved through the endothelial surface layer (ESL), a glycocalyx-rich structure typically 0.5–1 µm thick composed of proteoglycans and glycosaminoglycans like heparan sulfate, which acts as a molecular sieve excluding substances larger than 70 kDa.[9] Additionally, the endothelium regulates vascular tone through vasoregulation, synthesizing and releasing key mediators to balance vasodilation and vasoconstriction. It produces vasodilators such as nitric oxide (NO) via endothelial nitric oxide synthase (eNOS), prostacyclin (PGI₂), and endothelium-derived hyperpolarizing factors (EDHFs), which promote smooth muscle relaxation, while also secreting vasoconstrictors like endothelin-1 (ET-1) to fine-tune blood flow in response to physiological demands.[10][9]The endothelium further contributes to hemostatic balance through its anti-thrombotic properties, preventing unwarranted clot formation on the vessel wall. It secretes anticoagulants such as tissue plasminogen activator (tPA) to initiate fibrinolysis and tissue factor pathway inhibitor (TFPI) to inhibit the coagulation cascade, while NO and PGI₂ actively suppress platelet aggregation and adhesion.[10][9] In its anti-inflammatory capacity, the endothelium maintains vascular quiescence by expressing anti-adhesive molecules and low levels of pro-inflammatory adhesion molecules like ICAM-1 and VCAM-1 under normal conditions, thereby repelling leukocytes and minimizing immune cell extravasation. Intercellular junctions, including tight, adherens, and gap junctions, reinforce this barrier to preserve endothelial integrity and limit inflammatory responses.[11][9]Quantitatively, the endothelium represents the body's largest organ, covering an estimated surface area of 3,000–6,000 m² in adults[12] and comprising 1–6 × 10¹³ cells that collectively ensure efficient nutrient delivery and waste removal across the circulatory system.[13] These multifaceted roles underscore the endothelium's integral position in physiological vascular function.
Pathophysiology
Mechanisms of Impairment
Endothelial dysfunction primarily arises from disruptions in nitric oxide (NO) bioavailability, a core process involving the uncoupling of endothelial nitric oxide synthase (eNOS), where the enzyme produces superoxide anion (O₂⁻) instead of NO due to oxidative stress and imbalances in substrates like L-arginine. This uncoupling is quantified by a reduced L-arginine to asymmetric dimethylarginine (ADMA) ratio or depletion of the cofactor tetrahydrobiopterin (BH₄), leading to peroxynitrite formation that further exacerbates oxidative damage and impairs vascular homeostasis.[14][15]A key manifestation is reduced vasodilation, stemming from decreased NO production via inhibited eNOS activity—such as through substrate deficiency (e.g., low L-arginine) or post-translational modifications like S-glutathionylation—and increased NO degradation by reactive oxygen species (ROS) that form peroxynitrite. This imbalance shifts the endothelium from its normal role in NO-mediated relaxation to a vasoconstrictive state, promoting hypertension and vascular stiffness.[15][16]The dysfunctional endothelium also undergoes a shift to a pro-thrombotic state, characterized by upregulation of von Willebrand factor (vWF) release from Weibel-Palade bodies and increased expression of tissue factor (TF), which initiates the extrinsic coagulation cascade and enhances platelet adhesion. Inflammatory signals activate NF-κB to bind the vWF promoter, amplifying this prothrombotic phenotype in conditions like atherosclerosis.[17][1]Increased vascular permeability results from disruption of tight junctions, including phosphorylation of occludin and reduced claudin-5 or ZO-1 expression, which loosens endothelial barriers and allows paracellular leakage of fluids and solutes, contributing to edema. Pathways involving Rho GTPases and ROS further destabilize these junctions, as seen in inflammatory responses where histamine or VEGF signaling enhances permeability.[18][19]These impairments are perpetuated by feedback loops in which endothelial activation triggers NF-κB signaling, upregulating pro-inflammatory cytokines (e.g., TNF-α, IL-1β) and adhesion molecules (e.g., ICAM-1, VCAM-1) that recruit leukocytes and sustain inflammation. This positive feedback amplifies endothelial dysfunction, as released cytokines further activate NF-κB, creating a self-reinforcing cycle observed in chronic vascular diseases.[20][1]
Molecular Pathways Involved
Endothelial nitric oxide synthase (eNOS) activity is a central regulator of vascular homeostasis, primarily through its phosphorylation by the Akt/protein kinase B (PKB) pathway downstream of phosphoinositide 3-kinase (PI3K) signaling in endothelial cells. Shear stress or agonists like vascular endothelial growth factor (VEGF) activate PI3K, leading to Akt-mediated phosphorylation of eNOS at serine 1177, which enhances enzyme activity and nitric oxide (NO) production to promote vasodilation and inhibit platelet aggregation.[21][1]In endothelial dysfunction, this pathway is impaired, notably by asymmetric dimethylarginine (ADMA), an endogenous inhibitor of eNOS that competitively blocks L-arginine binding, reducing NO synthesis. Elevated plasma ADMA levels exceeding 0.7 μmol/L are associated with increased cardiovascular risk, as they correlate with diminished eNOS activity and progression of atherosclerosis.[22][23]Oxidative stress contributes to endothelial dysfunction via activation of NADPH oxidase (Nox) enzymes, particularly Nox2 and Nox4 isoforms in endothelial cells, which generate superoxide anion (O₂⁻) that scavenges NO to form peroxynitrite (ONOO⁻). Peroxynitrite induces oxidative damage to proteins and lipids, uncouples eNOS by oxidizing its cofactor tetrahydrobiopterin (BH₄), and perpetuates a vicious cycle of reactive oxygen species (ROS) production, impairing endothelial barrier function and promoting vasoconstriction.[24][25][26]Inflammatory cascades exacerbate dysfunction through tumor necrosis factor-alpha (TNF-α)-induced activation of c-Jun N-terminal kinase (JNK) signaling, which upregulates adhesion molecules such as vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) on endothelial surfaces. This JNK-mediated pathway facilitates leukocyte recruitment and transmigration, amplifying local inflammation and contributing to atherogenesis, independent of nuclear factor-kappa B (NF-κB) in some contexts.[27][28]Endothelial-mesenchymal transition (EndMT), driven by transforming growth factor-beta (TGF-β) signaling, represents another key pathway in dysfunction, where endothelial cells lose their phenotype and acquire mesenchymal features, including increased alpha-smooth muscle actin expression and extracellular matrix production. TGF-β activates Smad-dependent transcription, promoting fibrosis in vascular tissues and contributing to stiffening of the arterial wall in conditions like pulmonary hypertension.[29][30]The effective bioavailability of NO can be conceptually represented by the equation:[\text{NO}]_{\text{effective}} = \text{eNOS activity} - (\text{ROS scavenging} + \text{arginase competition})This illustrates how eNOS-derived NO is diminished by ROS-mediated inactivation and competition from arginase for L-arginine substrate, leading to reduced vasodilation and heightened oxidative stress in dysfunctional endothelium.[31][1]
Causes and Risk Factors
Modifiable Factors
Smoking is a major modifiable risk factor for endothelial dysfunction, primarily through the actions of nicotine and oxidative components in tobacco smoke that reduce nitric oxide (NO) bioavailability by promoting oxidative stress and impairing endothelial NO synthase activity.[32] This leads to diminished vasodilation and increased vascular inflammation. Smoking cessation rapidly improves endothelial function, with measurable enhancements in flow-mediated dilation observed within weeks to months, even in the presence of potential weight gain.[33][34]Diets high in saturated fats contribute to endothelial dysfunction by promoting the oxidation of low-density lipoprotein (LDL) particles, which triggers inflammatory responses and reduces NO production in endothelial cells.[35][36] In contrast, adherence to a Mediterranean diet, rich in polyphenols from sources like olive oil and fruits, enhances endothelial repair mechanisms by improving NO bioavailability and reducing oxidative damage.[37][38]Physical inactivity, characteristic of a sedentary lifestyle, induces endothelial dysfunction by creating an imbalance in shear stress on vascular walls, leading to decreased NO production and increased oxidative stress.[39] Regular exercise counteracts this by boosting endothelial NO synthase (eNOS) expression, thereby enhancing vasodilation and overall vascular health.[40][41]Hypertension is a key modifiable risk factor that promotes endothelial dysfunction through increased mechanical stress on vessel walls, leading to reduced NO bioavailability, oxidative stress, and inflammation. Effective blood pressure control via lifestyle changes or medications can restore endothelial function and mitigate vascular damage.[42]Dyslipidemia, particularly elevated low-density lipoprotein cholesterol, impairs endothelial function by facilitating LDL oxidation and promoting inflammatory adhesion molecule expression on endothelial cells. Management with statins or dietary interventions improves NO-dependent vasodilation.[43]Diabetes and hyperglycemia contribute to endothelial dysfunction via advanced glycation end-products, oxidative stress, and impaired insulin signaling, which reduce eNOS activity and increase vascular permeability. Glycemic control is essential for preserving endothelial integrity.[44]Obesity drives endothelial dysfunction through the release of pro-inflammatory adipokines such as leptin from adipose tissue, which induces chronic low-grade inflammation and impairs NO-dependent vasodilation.[45][46] Individuals with a body mass index (BMI) greater than 30 are associated with significantly impaired endothelial function compared to those with normal weight, independent of other factors.[47][48]Exposure to fine particulate matter (PM2.5) from air pollution acutely impairs endothelial function by reducing vasodilation through oxidative stress and inflammation, with effects observable even at low concentrations.[49][50] Reducing exposure via environmental measures can mitigate these risks.[51]
Non-Modifiable Factors
Non-modifiable factors contributing to endothelial dysfunction encompass inherent biological and demographic characteristics that cannot be altered through lifestyle or therapeutic interventions. These include aging, genetic predispositions, sex-based differences, family history, and ethnic variations, each influencing endothelial nitric oxide synthase (eNOS) activity, vascular homeostasis, and susceptibility to impairment.[52]Aging represents a primary non-modifiable risk factor for endothelial dysfunction, characterized by a progressive decline in endothelial function due to reduced bioavailability of nitric oxide (NO) and increased oxidative stress. Endothelial cells undergo senescence with advancing age, which impairs vasodilation and promotes vascular stiffness. Telomere shortening in endothelial cells further exacerbates this process by accelerating cellular senescence and reducing regenerative capacity, contributing to chronic low-grade inflammation and diminished angiogenic responses.[53][54]Genetic factors, particularly polymorphisms in the eNOS gene, significantly modulate susceptibility to endothelial dysfunction. The G894T variant (rs1799983) in the eNOS gene is associated with reduced enzyme activity and NO production, increasing the risk of cardiovascular conditions linked to endothelial dysfunction by approximately 1.5-fold in meta-analyses of affected carriers compared to wild-type individuals. This polymorphism disrupts shear stress-induced eNOS expression, leading to heightened oxidative stress and endothelial activation. Other eNOS variants, such as T-786C, similarly contribute to heritable endothelial vulnerability, underscoring the polygenic nature of this risk.[55][56]Sex differences influence endothelial function through hormonal profiles that vary across the lifespan. In women, the loss of estrogen following menopause heightens risk, as estrogen normally enhances eNOS expression and NO-dependent vasodilation; post-menopausal declines correlate with accelerated endothelial dysfunction and increased arterial stiffness. In men, higher testosterone levels are linked to earlier onset of impairment, potentially via androgen-mediated promotion of oxidative stress and reduced antioxidant defenses, with low testosterone paradoxically associated with further endothelial decline in older age. These dimorphic effects highlight sex as a key determinant of endothelial health trajectories.[57][58]Family history reflects the heritable component of endothelial function, with estimates indicating 20-30% heritability for metrics such as flow-mediated dilation (FMD), a key indicator of endothelial health. Twin and parent-offspring studies demonstrate that genetic influences on vasomotor responses and NO signaling pathways account for this variance, independent of shared environmental factors. This heritability underscores the role of familial aggregation in predisposing individuals to endothelial dysfunction, often manifesting as subclinical vascular changes in at-risk lineages.[59][60]Ethnicity also plays a non-modifiable role, with South Asians exhibiting higher prevalence of endothelial dysfunction linked to inherent patterns of insulin resistance. Compared to other groups, South Asians display greater visceral adiposity and β-cell dysfunction from younger ages, leading to impaired NO production and elevated inflammatory markers that compromise endothelial integrity. Population studies confirm this disparity, showing 2- to 3-fold higher insulin resistance in South Asian cohorts, which correlates with reduced FMD and accelerated vascular aging, independent of traditional risk factors.[61][62]
Clinical Associations
Cardiovascular Diseases
Endothelial dysfunction serves as an early and pivotal marker in the development of atherosclerosis, often preceding visible plaque formation by several years. Impaired flow-mediated dilation (FMD) of the brachial artery, a key indicator of endothelial health, has been shown to correlate with subclinical atherosclerosis in multiple vascular beds, reflecting reduced nitric oxide bioavailability that promotes vascular inflammation and oxidative stress. This dysfunction facilitates monocyte adhesion to the endothelium via upregulated expression of adhesion molecules such as vascular cell adhesion molecule-1 (VCAM-1), thereby initiating the atherogenic process. Studies have demonstrated that individuals with endothelial dysfunction exhibit a significantly higher risk of plaque progression, with FMD values below 10% associated with a 2-3 fold increased incidence of cardiovascular events over 5-10 years.In hypertension, endothelial dysfunction contributes to increased vascular resistance through endothelial stiffening and impaired vasorelaxation, affecting approximately 70% of patients with essential hypertension. The reduced production of endothelium-derived relaxing factors, particularly nitric oxide, leads to heightened vasoconstriction and structural remodeling of resistance arteries, exacerbating blood pressure elevation. Longitudinal data indicate that endothelial dysfunction in hypertensive patients is linked to a 1.5-2 times greater risk of target organ damage, including left ventricular hypertrophy. This association underscores the endothelium's role in the vicious cycle of hypertension, where chronic shear stress from elevated pressure further impairs endothelial function.Endothelial dysfunction is intimately linked to coronary artery disease (CAD), where it manifests as reduced coronary flow reserve, contributing to myocardial ischemia and angina symptoms. In patients with stable CAD, endothelial-dependent vasodilation in epicardial coronary arteries is often diminished by 50% or more compared to healthy controls, limiting the ability to meet increased myocardial oxygen demands during stress. This impairment promotes thrombus formation at lesion sites due to prothrombotic shifts in endothelial phenotype, increasing the likelihood of acute coronary syndromes. Clinical trials have reported that improving endothelial function through statins correlates with enhanced coronary flow reserve and reduced angina episodes.In heart failure, endothelial dysfunction drives diastolic dysfunction by promoting myocardial fibrosis through the release of profibrotic cytokines and transforming growth factor-beta (TGF-β) from dysfunctional endothelial cells. This leads to extracellular matrix accumulation and stiffening of the ventricular wall, impairing relaxation and filling during diastole. Observational studies in patients with preserved ejection fraction heart failure show that endothelial biomarkers like asymmetric dimethylarginine (ADMA) levels predict worse diastolic parameters, with dysfunction present in up to 80% of cases. The resultant fibrotic remodeling perpetuates a cycle of endothelial injury due to neurohormonal activation.Endothelial dysfunction is a strong predictor of ischemic stroke, with impaired brachial FMD associated with an odds ratio of approximately 2.5 for future cerebrovascular events in at-risk populations.[63] This predictive value stems from the endothelium's role in regulating local thrombosis and plaque stability in the carotid bifurcation, where dysfunction accelerates atheroma progression and embolization. Prospective cohort studies have confirmed that brachial FMD below 5% identifies individuals with a 3-4 fold higher stroke risk over 5 years, independent of traditional risk factors.[64]
Systemic Conditions
In diabetes mellitus, hyperglycemia promotes the formation of advanced glycation end-products (AGEs), which bind to receptors on endothelial cells, leading to reduced nitric oxide (NO) production and bioavailability through mechanisms such as suppression of endothelial NO synthase activity and increased oxidative stress.[65] This impairment contributes to endothelial dysfunction, a key factor in the development of microvascular complications, including retinopathy, nephropathy, and neuropathy, where diminished vasodilation exacerbates tissue ischemia.[66] Studies in type 2 diabetes patients demonstrate that elevated AGE levels correlate with more severe endothelial impairment compared to newly diagnosed cases.[65]Chronic kidney disease (CKD) is characterized by uremia, which elevates levels of asymmetric dimethylarginine (ADMA), an endogenous inhibitor of NO synthase that accumulates due to reduced renal clearance and contributes to widespread endothelial dysfunction.[67] This dysfunction is prevalent in advanced CKD, affecting the microvasculature and promoting cardiovascular risk through impaired vasodilation and prothrombotic states, with evidence of its presence in the majority of patients progressing to later stages.[68] Uremic toxins like indoxyl sulfate further exacerbate this by inducing inflammation and oxidative damage to endothelial cells.[67]In COVID-19, the SARS-CoV-2 spike protein binds to angiotensin-converting enzyme 2 (ACE2) receptors abundantly expressed on endothelial cells, triggering direct viral entry and subsequent endothelialitis, particularly in the pulmonary vasculature, which leads to inflammation, thrombosis, and barrier disruption.[69] Autopsy studies from 2020 onward have confirmed widespread endothelial damage, contributing to acute respiratory distress syndrome and systemic coagulopathy.[70] Research between 2020 and 2023 indicates that endothelial dysfunction in COVID-19 patients is linked to higher mortality rates, with biomarkers of endothelial injury such as elevated von Willebrand factor associated with severe outcomes and increased risk of death.[69]Autoimmune diseases, such as rheumatoid arthritis (RA), involve the production of anti-endothelial cell antibodies (AECAs) that target endothelial antigens, inducing complement activation, cytokine release, and apoptosis, which promote vasculitis and chronic vascular inflammation.[71] In RA patients, these antibodies correlate with disease activity and contribute to endothelial dysfunction by enhancing adhesion molecule expression and leukocyte recruitment, facilitating the progression to systemic vasculitis.[72] This immune-mediated damage underscores the role of endothelial cells as both targets and amplifiers in autoimmune vasculopathy.[73]Sepsis induces a cytokine storm, characterized by excessive release of pro-inflammatory mediators like tumor necrosis factor-alpha and interleukin-6, which acutely impair endothelial function by increasing permeability, promoting leukocyte adhesion, and reducing NO production, culminating in multi-organ failure.[74] This endothelial barrier dysfunction leads to vascular leakage, microvascular thrombosis, and tissue hypoperfusion, with studies highlighting its centrality in sepsis pathogenesis and high mortality.[75] Reviews emphasize that sepsis-associated endothelial injury is a key driver of remote organ damage beyond the initial infection site.[76]Endothelial dysfunction has also been implicated in neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease, where blood-brain barrier disruption and reduced cerebral blood flow contribute to neuronal damage and disease progression. Oxidative stress and inflammation from dysfunctional endothelium exacerbate amyloid-beta accumulation and tau pathology in Alzheimer's, while in Parkinson's, it promotes alpha-synuclein aggregation and dopaminergicneuron loss.[77] Studies indicate that endothelial biomarkers correlate with cognitive decline and neurodegeneration severity.[78]
Diagnosis
Assessment Methods
Assessment of endothelial dysfunction relies on functional tests that evaluate the endothelium's ability to respond to stimuli, such as shear stress or pharmacological agents, through vasodilation or other vasomotor responses. These methods provide direct insights into endothelial health by measuring dynamic vascular function, distinguishing them from static imaging or molecular analyses. Non-invasive techniques are preferred in clinical settings for their safety and reproducibility, while invasive approaches offer higher precision for coronary evaluation. In vitro models complement these by isolating cellular responses to mechanical forces.Flow-mediated dilation (FMD) is a widely adopted non-invasive ultrasound-based technique to assess conduit artery endothelial function. It involves inflating a cuff around the forearm to suprasystolic pressure for 5 minutes to induce ischemia, followed by release to create reactive hyperemia and shear stress on the brachial artery endothelium. The resulting nitric oxide-dependent vasodilation is quantified as the percentage increase in arterial diameter from baseline, measured via high-resolution ultrasound. Normal FMD values exceed 7%, while values below 4% indicate significant dysfunction, correlating with cardiovascular risk. This method, first described in seminal work, has become a standard for peripheral endothelial assessment due to its prognostic value in predicting adverse events.[79][80]Peripheral arterial tonometry (PAT) offers a user-friendly, non-invasive alternative for evaluating microvascular endothelial function using fingertip probes. The technique measures pulse wave amplitude in the digital arteries before and after a 5-minute brachial artery occlusion, calculating the reactive hyperemia index (RHI) as the ratio of post- to pre-occlusion amplitudes. An RHI greater than 1.67 reflects normal endothelial reactivity, whereas values below this threshold signify dysfunction, often linked to reduced nitric oxide bioavailability. Developed as an automated system, PAT correlates well with invasive measures and is advantageous for its simplicity and low operator dependency in clinical research.[81]Invasive coronary testing, considered the gold standard for direct coronary endothelial assessment, employs intracoronary acetylcholine infusion during cardiac catheterization. Acetylcholine stimulates endothelial muscarinic receptors to release nitric oxide, causing vasodilation in healthy vessels; in dysfunctional endothelium, it paradoxically induces vasoconstriction due to unopposed smooth muscle effects. Vasoconstriction greater than 20% in epicardial segments or less than 50% increase in coronary blood flow indicates impairment, assessed via quantitative angiography or Doppler flow velocity. This method, pioneered in early studies, is reserved for high-risk patients owing to its procedural risks but provides critical insights into coronary-specific dysfunction.Nailfold capillaroscopy visualizes microvascular endothelial changes non-invasively using a stereomicroscope or videocapillaroscope on the nailfold skin. It detects abnormalities such as reduced capillary density (normal: 9-10 per millimeter), avascular areas, or hemorrhages, which reflect endothelial damage in conditions like connective tissue diseases. The procedure involves applying oil to the nailfold and imaging under 200-300x magnification to classify patterns from normal to scleroderma-like, aiding early detection of microvascular dysfunction. Widely used in rheumatology, this technique's simplicity supports its role in monitoring endothelial integrity without radiation exposure.Shear stress models in vitro replicate hemodynamic forces on endothelial cells using cultured monolayers in parallel-plate flow chambers or microfluidic devices. Cells are exposed to controlled laminar or oscillatory shear stress (e.g., 10-20 dyn/cm² for physiological flow), assessing responses like nitric oxide production via fluorescence assays or barrier integrity through permeability measurements. Dysfunction manifests as diminished vasodilation analogs or increased permeability, providing mechanistic insights into endothelial mechanotransduction. These assays, foundational since early cell culture studies, are essential for preclinical evaluation of endothelial responses under simulated pathological conditions.
Biomarkers and Imaging
Circulating biomarkers provide a non-invasive means to assess endothelial dysfunction by reflecting molecular alterations in endothelial cells. Asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide synthase, is elevated in conditions associated with endothelial impairment and correlates with cardiovascular risk.[82]Von Willebrand factor (vWF), a glycoprotein released from endothelial cells, serves as a marker of endothelial damage and activation; elevated plasma concentrations are linked to increased endothelial stress and thrombotic propensity in vascular disorders.[83] Similarly, soluble E-selectin, shed from activated endothelial surfaces, rises in inflammatory states, with elevated levels signifying adhesion molecule upregulation and endothelial inflammation.[84]Endothelial progenitor cells (EPCs) offer insight into the endothelium's regenerative capacity, as their reduced numbers signal impaired vascular repair. EPCs are typically identified by co-expression of CD34 and vascular endothelial growth factor receptor 2 (VEGF-R2); reduced counts suggest a deficit in progenitor mobilization, commonly observed in patients with endothelial dysfunction and associated with higher atherosclerosis progression.[85] This quantification via flow cytometry highlights the role of EPCs in maintaining endothelial integrity, with low levels predicting adverse outcomes in cardiovascular disease.[86]Advanced imaging modalities enable direct visualization of endothelial alterations, complementing biochemical markers. Contrast-enhanced ultrasound (CEUS) assesses microvascular perfusion by tracking microbubble transit, revealing impaired endothelial-dependent vasodilation in dysfunctional vessels through reduced replenishment rates post-destruction.[87]Magnetic resonance imaging (MRI) evaluates vascular compliance by measuring pulse wave velocity (PWV), where values greater than 10 m/s denote increased arterial stiffness and cardiovascular risk.[88]Positron emission tomography (PET) using 18F-fluorodeoxyglucose (FDG) quantifies endothelial inflammation via uptake in activated macrophages and endothelial cells, with heightened standardized uptake values indicating inflammatory burden in atherosclerotic plaques.[89]Omics approaches, particularly proteomics, have advanced biomarker discovery for endothelial dysfunction since 2020, identifying multi-marker panels for enhanced risk stratification. Proteomic profiling of plasma has revealed panels of over 10 endothelial-specific proteins, such as those involved in barrier integrity and inflammation, that collectively predict dysfunction with superior sensitivity compared to single markers, aiding in early identification of at-risk individuals. Recent advances as of 2025 include integration of multi-omicsdata for novel signatures in conditions like cerebral small vessel disease.[90][91] These panels integrate data from mass spectrometry to uncover novel signatures, supporting personalized cardiovascular risk assessment.[92]
Management and Treatment
Pharmacological Options
Statins represent a cornerstone in the pharmacological management of endothelial dysfunction, exerting beneficial effects independent of their lipid-lowering properties. These agents upregulate endothelial nitric oxide synthase (eNOS) expression and activity primarily through inhibition of RhoA geranylgeranylation, which enhances eNOS mRNA stability and translocation to caveolae.[93] For example, atorvastatin at a dose of 40 mg daily has been shown to significantly improve flow-mediated dilation (FMD), a key noninvasive marker of endothelial function, by approximately 2-3% in patients with metabolic syndrome after short-term treatment, reflecting enhanced nitric oxide bioavailability.[94]Angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs) address endothelial dysfunction by attenuating angiotensin II-induced oxidative stress, which otherwise uncouples eNOS and reduces nitric oxide production. These drugs promote eNOS phosphorylation and diminish reactive oxygen species generation, thereby restoring vascular tone. Losartan, a widely used ARB, increases nitric oxide bioavailability and improves endothelium-dependent vasodilation in patients with hypertension and atherosclerosis, as evidenced by enhanced FMD responses in clinical evaluations.[95]Antiplatelet therapies play a supportive role in preventing the prothrombotic phenotype of dysfunctional endothelium. Aspirin inhibits cyclooxygenase-1 to block thromboxane A2 synthesis, thereby reducing platelet aggregation and preserving endothelial integrity against inflammatory and oxidative insults that promote a thrombotic shift.[96] In high-risk scenarios, such as post-coronary stent implantation, clopidogrel offers targeted benefits by further suppressing platelet reactivity and improving endothelial function through reduced inflammation and enhanced nitric oxide pathways.[97]Emerging pharmacological strategies focus on novel agents that directly target eNOS uncoupling and substrate limitations. Tetrahydrobiopterin (BH4), an essential eNOS cofactor, supplementation recouples the enzyme to favor nitric oxide over superoxide production, thereby ameliorating oxidative stress in the endothelium. Clinical trials, including those evaluating oral BH4 in patients with coronary artery disease and hypertension, have demonstrated improvements in endothelial function, underscoring its potential despite ongoing needs for larger-scale validation.[98]Endothelial-specific interventions, such as anti-asymmetric dimethylarginine (ADMA) therapies, aim to counteract ADMA-mediated eNOS inhibition, which elevates in cardiovascular disease and impairs nitric oxide synthesis. These approaches, including pegylated arginase inhibitors that reduce arginase activity and alleviate L-arginine competition with ADMA, have shown promise in early-phase clinical trials and have demonstrated gains in vasodilation independent of glycemic control in conditions like type 2 diabetes.[99]Sodium-glucose cotransporter 2 (SGLT2) inhibitors, such as empagliflozin, have also shown benefits in improving endothelial function, particularly in patients with type 2 diabetes and heart failure, by reducing oxidative stress and inflammation as demonstrated in studies up to 2025.[100]
Lifestyle Interventions
Lifestyle interventions represent essential non-pharmacological strategies for ameliorating endothelial dysfunction by addressing underlying vascular stressors and promoting nitric oxide (NO) bioavailability. These approaches, including exercise, dietary adjustments, weight control, smoking cessation, and stress management, have been validated through clinical trials and meta-analyses, demonstrating improvements in endothelial-dependent vasodilation as measured by flow-mediated dilation (FMD).Aerobic exercise training is a primary recommendation, with guidelines advocating at least 150 minutes of moderate-intensity activity per week, such as brisk walking or cycling. This regimen induces hemodynamic shear stress on endothelial cells, activating endothelial nitric oxide synthase (eNOS) to enhance NO production and reduce oxidative stress.[101] Meta-analyses of randomized controlled trials confirm that continuous aerobic exercise significantly improves FMD by 1-3%, a clinically meaningful gain associated with reduced cardiovascular risk.[102][103]Dietary modifications focusing on anti-inflammatory nutrients further support endothelial repair. Daily supplementation with 1-2 g of omega-3 fatty acids (eicosapentaenoic acid [EPA] and docosahexaenoic acid [DHA]) attenuates inflammation by modulating cytokine production and improves FMD in populations with metabolic risks.[104] Complementing this, antioxidant-rich foods like berries—such as blueberries (e.g., 150-200 g daily)—counteract reactive oxygen species, preserving endothelial integrity and enhancing vascular function over 4-12 weeks.[105][106]Effective weight management targets obesity-induced endothelial impairment, a common modifiable factor. A 5-10% body weight loss, achieved via balanced caloric restriction combined with exercise, reverses microvascular and conduit artery dysfunction, with improvements in FMD evident after 3-6 months.[107][108]Smoking cessation programs, often aided by nicotine replacement therapy, yield prompt vascular benefits. Within 1-3 months of abstinence, endothelial function recovers, as indicated by increased FMD and decreased arterial stiffness, due to diminished oxidative damage and restored NO signaling.[109]Stress reduction via mindfulness-based techniques, such as mindfulness-based stress reduction (MBSR) programs, indirectly bolsters endothelial health by lowering cortisol, a hormone that impairs NO production under chronic elevation. Randomized controlled trials from the 2020s show MBSR reduces cortisol levels, and earlier studies indicate mindfulness interventions can enhance vascular function in stressed individuals.[110][111]
Prevention and Research
Strategies for Risk Reduction
Population-level screening for endothelial dysfunction involves non-invasive assessments such as flow-mediated dilation (FMD) of the brachial artery or biomarker analysis in high-risk groups, including individuals over 40 years with cardiovascular risk factors like hypertension or dyslipidemia, to enable early identification and risk stratification before overt disease develops.[112] These methods, particularly FMD, provide prognostic value for adverse cardiovascular outcomes in asymptomatic populations, supporting their integration into routine clinical evaluations for targeted prevention.[113]Early interventions, such as vaccinations against infections known to impair endothelial function, play a key role in averting acute vascular insults. For instance, SARS-CoV-2vaccination protects against COVID-19-induced endothelial dysfunction by mitigating inflammatory responses and preserving vascular integrity during infection.[114] Similarly, vaccines targeting other viruses like dengue have demonstrated prevention of endothelial permeability and vascular leak, highlighting the broader utility of immunization in maintaining endothelial health.[115]Public health policies aimed at reducing modifiable risk factors significantly contribute to preventing endothelial dysfunction at a societal level. Anti-smoking campaigns, as part of comprehensive tobacco control strategies, lower smoking prevalence and thereby reduce exposure to tobacco smoke, which directly impairs endothelial function through oxidative stress and inflammation; sustained cessation efforts have been shown to restore vascular endothelial responses over time.[116] Additionally, urban planning initiatives that enhance walkability—such as developing pedestrian-friendly infrastructure—promote active transportation, decrease reliance on vehicles, and consequently lower air pollution exposure, which is a potent inducer of endothelial perturbation and early cardiovascular damage.[117][118]Personalized prevention strategies leverage genetic insights to tailor interventions for at-risk individuals. Testing for variants in the endothelial nitric oxide synthase (eNOS) gene, such as the -786T>C polymorphism, identifies those with reduced nitric oxide bioavailability and heightened susceptibility to endothelial dysfunction, enabling customized lifestyle recommendations like enhanced physical activity or dietary modifications to mitigate cardiovascular risk.[119]Long-term monitoring through regular vascular health assessments is essential for high-risk populations, particularly those with diabetes. The 2023 European Society of Cardiology guidelines recommend annual screening for chronic kidney disease using estimated glomerular filtration rate and albumin-to-creatinine ratio, alongside regular clinical evaluation and ankle-brachial index measurement for lower-extremity artery disease, to detect early vascular changes and guide preventive measures in diabetic patients.[120]
Current and Emerging Studies
Recent studies from 2023 to 2025 have highlighted the persistence of endothelial dysfunction in long COVID, with elevated markers of vascular damage and microcirculatory impairments observed up to a year post-infection, contributing to ongoing symptoms such as fatigue and chest pain.[121][122] In cohort analyses, COVID-19 vaccination has been associated with reduced risks of post-infectious cardiovascular complications, including vascular events linked to endothelial damage, underscoring its protective role against long-term vascular sequelae.[123]Advances in nanotechnology have introduced endothelial-targeted nanoparticles for precise drug delivery, with lipid nanoparticle systems designed to deliver mRNA therapeutics directly to endothelial cells in models of acute respiratory distress syndrome, showing promise in restoring vascular integrity.[124] Preclinical evaluations in 2025 demonstrated that LRP1-targeted nanoparticles loaded with simvastatin effectively reduced endothelial inflammation and improved barrier function in Alzheimer's disease models.[125]Research on the gut microbiome has established links between dysbiosis and endothelial dysfunction through metabolites like trimethylamine N-oxide (TMAO), which promotes vascular inflammation and atherosclerosis progression.[126] In animal models, fecal microbiota transplantation from healthy donors has restored endothelial function by lowering TMAO levels and mitigating arterial stiffness, as evidenced in murine studies of hypertension and atherosclerosis.[127][128]Integration of artificial intelligence with multi-omics data has enabled machine learning models to predict endothelial dysfunction, with plasma proteomics and transcriptomics analyses achieving predictive accuracies around 76% in 2025 cohorts at risk for hyperglycemia-related vascular damage.[129] These models identify key biomarkers from endothelial cell exposure data to forecast dysfunction onset, enhancing early intervention strategies.Despite these advances, significant research gaps persist, including limited data on endothelial dysfunction in pediatric populations, where studies primarily focus on obesity-related risks but lack longitudinal insights into developmental impacts.[130] Additionally, diverse ethnic groups remain underrepresented in trials, with few investigations addressing variations in endothelial responses among African ancestry children, necessitating inclusive studies to address disparities in vascular disease prevalence.[131]