Review Article

Noise, Air, and Heavy Metal Pollution as Risk Factors for Endothelial Dysfunction

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Information image
Average (ratings)
No ratings
Your rating

Abstract

During the last two decades, large epidemiological studies have shown that the physical environment, including noise, air pollution or heavy metals, have a considerable impact on human health. It is known that the most common cardiovascular risk factors are all associated with endothelial dysfunction. Vascular tone, circulation of blood cells, inflammation, and platelet activity are some of the most essential functions regulated by the endothelium that suffer negative effects as a consequence of environmental pollution, causing endothelial dysfunction. In this review, we delineate the impact of environmental risk factors in connection to endothelial function. On a mechanistic level, a significant number of studies suggest the involvement of endothelial dysfunction to fundamentally drive the adverse endothelium health effects of the different pollutants. We focus on well-established studies that demonstrate the negative effects on the endothelium, with a focus on air, noise, and heavy metal pollution. This in-depth review on endothelial dysfunction as a consequence of the physical environment aims to contribute to the associated research needs by evaluating current findings from human and animal studies. From a public health perspective, these findings may also help to reinforce efforts promoting the research for adequate promising biomarkers for cardiovascular diseases since endothelial function is considered a hallmark of environmental stressor health effects.

Disclosure:TM is a section editor on the European Cardiology Review editorial board; this did not influence peer review. All other authors have no conflicts of interest to declare.

Received:

Accepted:

Published online:

Acknowledgements:MTBJ and OH contributed equally and are joint first authors.

Correspondence Details:Omar Hahad, Department of Cardiology, Cardiology I, University Medical Center of Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany. E: omar.hahad@unimedizin-mainz.de

Open Access:

This work is open access under the CC-BY-NC 4.0 License which allows users to copy, redistribute and make derivative works for non-commercial purposes, provided the original work is cited correctly.

Environmental stressors, such as noise, air or heavy metal pollution, represent important risk factors for the development and progression of cardiovascular disease (CVD).1–5 The WHO estimates that at least 1.6 million healthy life years are lost every year in western European countries alone due to the exposure to environmental noise. Additionally, within the last years, the GBD study established that ambient outdoor air pollution due to particulate matter <2.5 μm (PM2.5) was the fifth-ranking global risk factor in 2015, causing 4.2 million deaths annually, with cardiovascular deaths accounting for most of these deaths.6 Importantly, this statistic does not include other occupational and environmental stressors, such as smoking, household air pollution, mental stress or noise, which, taken together, arguably could outrank the global impact of many classical risk factors in combination. In addition, mercury, lead (Pb) or cadmium (Cd) are toxic elements commonly found in the environment, contributing to hypertension.7 According to several reports, high concentrations of heavy metals in different natural systems, including atmosphere, pedosphere, hydrosphere and biosphere, have become a global issue.8 Importantly, these heavy metals have been and are still commonly used in various areas, especially manufacturing and agriculture.9 Alterations in vascular function were the earliest pathophysiological mechanism described in response to noise, air, or heavy metal (e.g. mercury, Pb or Cd) exposure in humans, and indeed disturbances in endothelial function are a critical initiating event that is widely relevant to almost all classical risk factors.10–14 In the present review, we focus on the evidence supporting the impact of different pollutants and, particularly, its molecular mechanism involved in the development of endothelial dysfunction. Additionally, we focus on to what extent alterations in vascular function are responsible for mediating systemic diseases in response to noise, air and heavy metal pollution. Finally, we examine the epidemiological/observational evidence on the association of environmental risk factors and surrogate markers of endothelial dysfunction.

Endothelial Dysfunction is Triggered by the Environment

Endothelial Function

There are three distinct layers in the wall of an artery (Figure 1A). Firstly, the adventitia has primarily a mechanical role to fasten the vessel to the surrounding tissue and driving the inflammation.15,16 Then comes the tunica media layer that regulates the internal diameter of the vessel. In addition, the smooth muscle cells in this layer possess receptors for neurotransmitters and hormones, which allow direct effects on the vasoconstriction of vessels by stress response systems.15,17,18 Finally, the tunica intima has a specialised monolayer of epithelial cells called the endothelium.15,19,20 The endothelium, which is a significant source of nitric oxide (NO) that is produced by nitric oxide synthases (NOS), and its principal function is to control over vasoconstriction and vasodilation.21–23 A healthy endothelium maintains communication with the media through signalling molecules, of which NO plays a prominent role.24 Endothelial NOS (eNOS) uses the amino acid L-arginine as a substrate to generate NO. NO is primarily involved in vasodilation of vascular smooth muscle cells and prevention of platelet/immune cell adhesion to the vascular wall when it is released into the circulation (Figure 1B). eNOS consists of different domains; the carboxy-terminal reductase domain is homologous to the cytochrome P450 reductase and contains a binding site for the nicotinamide adenine dinucleotide phosphate (NADPH), flavin mononucleotide (FMN), flavin adenine dinucleotide (FAD) and calmodulin (CaM).25 Another principal domain is the amino-terminal oxygenase that contains a heme prosthetic group, binding sites for the substrate L-arginine and the cofactor tetrahydrobiopterin (BH4), which is required for the efficient generation of NO by controlling the redox state of the oxy-iron intermediate.26 In the process of constitutive formation of NO, calmodulin activity is modulated by the increase of intracellular Ca2+ levels, thereby enhancing the flow of electrons from NADPH, which represents the signal for activating the reductase function of the enzyme.27,28 The whole regulation of eNOS is shown in Figure 1C. In healthy endothelial cells, eNOS can be activated either by mechanoreceptors that are sensitive to the shear stress generated by blood flow or by acetylcholine (ACh) receptors, all of which leads to altered phosphorylation state of eNOS.28 NO produced by eNOS diffuses to smooth muscle cells and mediates relaxation through cGMP signalling mechanism, finally decreasing the intracellular calcium concentrations in smooth muscle cells and thereby decreasing the contractility of the actinin-myosin-filaments.29 In order to stimulate vascular smooth muscle relaxation, an endothelium-independent approach can be used, where NO is supplied by donor drugs, such as nitroglycerin (GTN). GTN undergoes bioactivation via mitochondrial aldehyde dehydrogenase (ALDH-2) to generate NO, which can directly stimulate vascular smooth muscle relaxation.30

Structure and Role of the Wall of an Artery

Article image

Endothelial Dysfunction Is Induced by Environmental Stress

The inability of a vessel to dilate fully, which arises from a decrease in NO bioavailability caused by reduced production, scavenging by reactive oxygen species (ROS), or both, is known as endothelial dysfunction.24 During the last decades, it has been shown that novel risk factors in the physical environment may facilitate the development of cardiovascular complications such as endothelial dysfunction.31,32 For this reason, endothelial dysfunction in the setting of cardiovascular risk factors, such as hypercholesterolemia, diabetes, chronic smoking or hypertension, is dependent on the production of ROS and the subsequent decrease in vascular bioavailability of NO.14 Additionally, it is known that some stress conditions, such as air or noise pollution, in the setting of cardiovascular diseases (CVD) are characterised by impaired NO signalling and associated with increased circulating stress hormones, cytokines, vasoconstrictors, such as angiotensin II (AngII) and circulating or infiltrating immune cells.33 NO can be scavenged by other free radicals such as superoxide, which ultimately leads to endothelial dysfunction (Figure 1C). When there is a reduction in the bioavailability of BH4, a cofactor of eNOS, because the rate at which it is recycled is slower than the rate of oxidative degradation, it effectively causes eNOS to produce superoxide, rather than NO.26 Reductions in the amount of NO impairs vascular signalling and results in endothelial dysfunction. Bioavailability of BH4 can be altered by the redox status of the cell (i.e. oxidative degradation to BH2) or, since it is endogenously produced, the pathway for BH4 production can be altered. Furthermore, redox modifications to eNOS itself can cause uncoupling, which again leads to superoxide production.26 The disruption of NO synthesis could be affected by several factors, which have a direct correlation with oxidative stress.34

There is evidence that preserving an intact endothelium is a critical component of healthy cardiovascular physiology, since endothelial dysfunction can be detected as an early sign of atherosclerosis and its associated pathologies, hypertension, diabetes and dyslipidemia.31,35–38 Endothelial dysfunction is also present in people with other risk factors for cardiovascular disease, such as in smokers, people with disturbed sleep, chronic inflammation and chronic stress.39–44

Mechanistic Insight into Endothelial Dysfunction Consequences of Noise Exposure and Its Outcomes

Adverse Cardiovascular Effects of Noise in Animals

The association between noise exposure, endothelial dysfunction and cardiac damage has been suspected for several decades, and investigation into the mechanism by which damage occurs is well underway. An early work in the field was a study where it was demonstrated that cardiac fibrosis and ischaemic myocardial lesions were present in the hearts of rats exposed to relatively low levels of noise (65 dB(A)) over the course of 52 weeks. Herrmann et al. demonstrated that noise stress may also cause a change in microvessel anatomy. Spontaneously hypertensive rats (SHR) were found to have more microvessels with an outer diameter of >19 µm and more severe cardiac fibrosis and extensive ischaemic myocardial lesions, but these effects were not seen in normotensive rats.47 Moreover, in 2013, fibrosis was also observed following noise exposure at 90 dB.48 Atrial fibrosis was reported in conjunction with reductions in connexin 43 levels following 120 dB noise.34 Signs of necrosis and inflammation were also reported in the myocardium of rats exposed to moderate noise levels.49 In terms of impact on the vessels, lumen narrowing was reported in the cochlear vessels following noise exposure in guinea pigs, which normalised several days after noise cessation.50 This report was later confirmed and alleviated in guinea pigs with treatment with etanercept, a TNF inhibitor.51 Other studies established that white noise exposure of rats for periods of 28 weeks (85100 dB(A)) significantly impaired endothelium-dependent vasodilation (measured by acetylcholine-dependent relaxation in thoracic aorta or mesenteric arterial rings), increased the sensitivity to the vasoconstrictor serotonin, decreased the lumen sizes of microvessels, increased systolic blood pressure by 2537 mm Hg and increased circulating markers of oxidative stress.52–55

Noise has been associated with annoyance, stress, sleep disturbance and impaired cognitive performance.56 In some animal noise models, data was generated that may explain the mechanism of aircraft noise-induced vascular dysfunction, oxidative stress and inflammation.57,58 Elevated levels of noradrenaline, adrenaline, angiotensin II (Ang II) and, subsequently, cortisol were present after an overactivation of the sympathetic nervous system. Next, Ang II activates endothelial NADPH oxidase via activation of protein kinase C (PKC), causing oxidative stress, which may induce direct scavenging of NO and eNOS uncoupling through oxidation of BH4 and eNOS S-glutathionylation. ROS play a key role in linking different pathways, including PI3K/Akt signalling, the FOXO transcription factors, TGF-β1 and NF-kB signalling, as well as the endothelin-1 (ET-1) system, increasing the circulating levels of interleukin (IL)-6 and the expression of vascular adhesion molecules. Superoxide and nitric oxide produced by infiltrating immune cells (neutrophils, NK cells and monocytes/macrophages) promote the formation of peroxynitrite and subsequent oxidative damage in the form of 3-nitrotyrosine (3-NT), malondialdehyde (MDA) and 4-hydroxynonenal-(4-HNE) positive proteins and other cellular oxidative damage. The uncoupling of eNOS not only reduces NO production but also potentiates the pre-existing oxidative stress. Endothelial NO production is further reduced by glucocorticoids like cortisol, leading to impaired vasodilation and increased blood pressure. The overproduction of catecholamines (noradrenaline (NA), adrenaline (A)) and ET-1 enhances contraction, which is further potentiated by glucocorticoids. In terms of oxidative stress, in mice (C57BL/6J), aircraft noise induces Nox2 expression and uncoupling of the eNOS, leading to cardiac superoxide formation. Superoxide has been well established as a nitric oxide scavenger resulting in a peroxynitrite production and, correspondingly, a progressive reduction in plasma NO2- was found in our last hypertension study.57–60 In addition, oxidative stress affects the health of the endothelium; this event also affects blood pressure due to the importance of the endothelium as a regulator of vascular tone. In our recent studies, gp91phox deletion and LysM+ cells ablation appear to be effective against developing hypertension from noise exposure.58,61 All these vascular alterations support the development of metabolic disorders as envisaged by increased blood glucose levels, insulin resistance, obesity, hypertension and atherosclerosis in noise-exposed mice.57,60,62 Upon chronic activation of stress response pathways, cardiovascular risk factors can emerge or be exacerbated and lead to cardiovascular or neuronal diseases.63–65 In addition, it was shown that also circadian dysregulation plays a key role in cardiovascular disorders caused by noise exposure.66 More recently, in 2022, it was demonstrated that endothelial dysfunction and inflammation were present during the entire 28 days of aircraft noise exposure (at a mean sound pressure level of 72 dB (A) and peak levels of 85 dB (A)).67 Altogether, these studies provide a solid foundation, given the apparent relationship between noise exposure, endothelial dysfunction and the subsequent vascular and cardiac damage. Moreover, all of the mentioned ones further illustrate health side effects of short and long-term noise exposure and further strengthen a consequent implementation of the WHO noise guidelines in order to prevent the development of noise-related future cardiovascular disease. For a selected overview of the mentioned experimental noise exposure protocols in the context of endothelial dysfunction, see Supplementary Material Table 1, whereas noise exposures for the purposes of studying other pathologies (e.g. acoustic damage) are not included.

Negative Consequences on Endothelial Function Caused by Air Pollution in Animals

Air Pollution

Air pollution is an overarching term used to describe, gaseous, liquid and solid pollutants that are present in the air, and that can arise from both human activity (industry, agriculture, farming, etc.) and natural sources (volcanic dust, soil dust, wild fires, etc).68 Historically, the main focus of the air pollution research field included the gaseous pollutants like nitrogen oxides (NOx), carbon monoxide (CO), sulphur dioxide (SO2) and ozone (O3), since they are common side products of large industrial processes.11 Other gases, such as volatile organic compounds (VOCs), non-methane hydrocarbons and polycyclic aromatic hydrocarbons (PAH), have recently received proper scientific attention. Solid and liquid components of air pollution, termed particulate matter (PM), have recently gained attention from researchers and legislators, as the detrimental effects of PM on human health have been recognised in a growing number of studies.2 Ambient (outdoor) PM was ranked seventh among the risk factors for attributable disability-adjusted life years (DALYs) in the 2019 Lancet’s GBD study, up from 13 place in 1990, but household air pollution fell from fourth to tenth place, showing the importance of this stressor.69 The GBD study also estimated that 6.5 million yearly deaths globally can be attributed to air pollution, which is an increase from the 4.2 million estimated by the WHO in 2016.70 Using novel hazard-response functions, it was recently estimated that up to 8.79 million global premature deaths could be attributed to air pollution every year.71 Non-communicable diseases, especially cardiovascular, are the major contributor to DALYs and global deaths, therefore, it is of great interest for public health that the interactions between air pollution and CVD is well understood.72

Air Pollution and Endothelial Function

When inhaled, pollutants from the air need to reach the circulation to be able to interact with the endothelium. Gaseous components of air pollution can easily enter circulation, bypassing the airblood barrier, where they can cause either direct damage by reacting with the constituents of the endothelium or indirectly by activating the immune system or causing oxidative stress.11,73 Nanometre-scale PM can also penetrate the air-blood barrier and directly migrate into the circulation.74,75 Larger, micrometre-scale PM usually don’t enter circulation, but exert their effects remotely, mostly by causing local inflammation that can later become systemic.76 Oxidative stress and inflammation are the two main risks that lead to onset and progression of endothelial dysfunction.

Oxidative stress can arise either from the free radical and ROS influx from air pollution itself or from the activation of ROS-generating enzymes in the body.77 One of the most prominent sources of ROS in the vascular tissue is the enzyme NADPH oxidase (NOX), which can either be activated in the vascular cells or come from circulating immune cells.78 The presence and activation of NOX from immune cells points to a close interplay between inflammation and oxidative stress, which often work together to progress endothelial dysfunction.79 A study done on Sprague-Dawley rats showed that exposure to concentrated ambient PM caused an impairment in the endothelium-dependent relaxation of isolated aortic rings, as well as activated the NOX in the vasculature.80 A similar study done on mice showed that impairment of the endothelium-dependent relaxation of isolated aortic rings was, in fact, mediated through oxidative stress caused by the upregulation of the monocyte-derived NOX.81 Exposure of animals to gaseous air pollutants was also shown to cause endothelial dysfunction. A study done on mice showed that exposure to ozone caused endothelial dysfunction that was accompanied by an increase in blood pressure and damage to mtDNA in aortic tissue.82 Other studies where animals were exposed to ozone also showed that presence of antioxidant enzymes like SOD and catalase can reverse the endothelial dysfunction.83 Exposure of Wistar rats to a combination of ozone and diesel PM caused upregulation of many different damage markers in aortic tissue, such as markers of oxidative stress (HO-1), of thrombosis (tissue factor and von von Willebrand factor), vasoconstriction (endothelin-1 and eNOS) and proteolysis (matrix metalloproteases-2 and -3).84 Chronic PM exposure of atherosclerosis-prone ApoE(-/-) mice was associated with increased accumulation of oxidatively modified cholesterol (7-ketocholesterol) and oxidised lipids in general, in the aortic plaques.85 The state of oxidative stress leads to direct effects on the vascular signalling through scavenging of NO, mostly by the superoxide radical.86 In rats exposed to ambient PM, it was shown that the impairment of the endothelium-dependent relaxation was associated with lower amounts of cyclic guanosine monophosphate (cGMP) and that NO donors did not improve the vascular function.87 Another study in rats, using diesel exhaust PM, showed that free radicals from the PM can directly scavenge NO, leading to impaired vascular signalling.88 Oxidative stress is not only detrimental to the central vascular system as pulmonary damage can influence pulmonary arteriole damage as well. Different chemical components of PM (e.g. heavy metals, reactive aldehydes, catalytic surfaces that directly produce ROS) can interact directly with the lung lining fluid and deplete antioxidants like glutathione and tocopherol, priming the lungs for oxidative damage.89 More recently, it was shown that also circadian dysregulation plays an important role for cardiovascular disorders caused by air pollution.66

Inflammation in the vasculature is not only a source of oxidative stress that impairs vascular signalling but is also responsible for onset and progression of diseases like atherosclerosis. Authors of a study on ApoE-/- mice observed that markers of inflammation TNF-α, MCP 1, IL-6, E-selectin and VCAM-1, were significantly elevated in the mesenteric arteries after exposure to concentrated ambient PM.90 Cardiac IL-6, TNF-α were also associated with 10 exposure of rats, pointing to direct inflammation of the cardiac tissue, and circulating levels of C reactive protein (CRP) pointing to systemic inflammation.91,92 It was also suggested that the inflammation could be mediated through the activation of the NLR family pyrin domain containing 3 inflammasome.93 There are also studies that identified inflammation as the main culprit for PM-induced endothelial dysfunction. In a study done on mice, dexamethasone, an anti-inflammatory substance, reversed the impaired acetylcholine-dependent relaxation of isolated aortic rings, but antioxidants did not.97 Gaseous components of air pollution are also responsible for inflammatory response, as shown in the meta-analysis that associated O3, NO2 and SO2 with increased CRP, but only associated NO2 exposure with elevated TNF-α.94 Most studies associate pulmonary exposure with ozone inhalation, but a study in rats has also demonstrated that systemic inflammation, envisaged by increased circulating IL-6, might accompany the observed endothelial dysfunction (explained in detail in Figure 2).95 In 2022, Wang et al. employed mice to assess vascular homeostasis following a standard urban particulate matter exposure, and they demonstrated that PM aspiration caused thickening of airways, leukocytes infiltration and adhesion to alveolus. In addition, the results showed an induction to vascular cell apoptosis and subcellular dysfunction, to which local and systemic redox biology and inflammation are probably correlated.97 Similar results were showed by Rojas et al., who exposed mice to a mixture of phenanthrene, fluoranthene and pyrene, which are compounds of PM. These exposed mice showed an increase in serum inflammatory cytokines and in expression of markers of endothelial dysfunction, both relevant characteristics associated with the onset of disease atherosclerosis and cardiovascular disease.98 For a selected overview of the mentioned experimental air pollution protocols in the context of endothelial dysfunction, see Supplementary Material Table 1.

Proposed Pathophysiological Mechanisms of Cardiovascular and Neuronal Disease Induced by Air Pollution

Article image

Heavy Metal Pollution and Endothelial Dysfunction

Cadmium (Cd), lead (Pb) and mercury (Hg) are toxic elements commonly found in the environment, contributing to hypertension, as well as these toxic elements cause severe inflammation and oxidative stress.12,99–101 Cd is an especially toxic and persistent heavy metal and a known carcinogen.102,103 The major sources of Cd exposures are food, cigarette smoking and as a byproduct of industrial processes.104 It has been shown that Cd exposure is associated with hypertension, atherosclerosis, diabetes and nephropathy, all of which could be attributable to dysfunctional endothelial and smooth muscle cells.105 In 2015, it was demonstrated that subjects with chronic Cd exposure showed endothelial dysfunction since NO levels were lower in Cd-exposed subjects than non-exposed subjects. Additionally, plasma L-arginine decreased while asymmetric dimethylarginine (ADMA), an endogenous eNOS inhibitor, increased in Cd-exposed subjects.106 More recently, Liang et al. provided novel insight into understanding the alterations of lipid metabolism induced by Cd exposure in endothelial cells. They used human microvascular endothelial cells (HMEC-1) that were exposed to 40-μM Cd for 6, 12 or 24 hours or 10-, 20- or 40-μM Cd for 24 hours, respectively. The Cd exposure accelerated the decomposition of triglyceride and resulted in the accumulation of free fatty acids. These changes stimulated cytotoxicity, induced ROS generation, altered the mitochondrial membrane potential and decreased the ATP content, which eventually led to endothelial dysfunction and cell death.107 In vitro exposure of endothelial cells to Cd (15 μM) resulted in increased permeability, inhibition of cell proliferation and induction of cell death.108 Moreover, in animal studies, Cd induced hypertension, which involved oxidative stress and suppression of eNOS expression in blood vessels leading to impaired Ach-induced vasorelaxation.109 In 2013, it was demonstrated that acute Cd exposure may induce vascular injury through endothelial oxidative stress and increased vasoconstrictor activity by reducing NO bioavailability owing to the increased production of ROS by NADPH oxidase (explained in detail in Figure 3).110 However, further studies are needed to understand much better the pathway by which Cd affects endothelial cell function.111,112 Another key pathomechanism of Cd involves impairment of the circadian clock, also termed “cadmium chronotoxicity”.113

Environmental Pb exposure is an established risk factor for hypertension and a possible risk factor for cardiovascular disease mortality.99 Pb is a highly toxic metal in aquatic environments. Fish are at the top of the food chain in most aquatic environments and are the most susceptible to the toxic effects of Pb exposure.101 Not only is Pb ubiquitous in aquatic environments, but also high levels of Pb exposure can be caused by anthropogenic activities, including the manufacture of batteries, paint and cement, as well as mining and smelting.114 Exposure to Pb, a heavy metal highly toxic to humans when ingested or inhaled, contributed to 900,000 premature deaths, while toxic occupational hazards followed with 870,000 deaths.115 In a recent cohort including 14,289 adults aged 20 years or older who were enrolled in the Third National Health and Nutrition Examination Survey between 1988 and 1994, in 20% of the participants a concentration of Pb in blood of at least 5 μg/dl (≥0.24 μmol/l) was identified, while the geometric mean concentration of Pb in blood was 0.82 µg/dl in 2016.116 An increase in the concentration of Pb in blood from 1.0 to 6.7 μg/dl ([0.0480.324 μmol/l]), which represents the tenth to 90th percentiles, was associated with all-cause mortality (HR 1.37, 95% CI [1.17–1.60]), cardiovascular disease mortality (HR 1.70, 95% CI [1.30–2.22]) and ischaemic heart disease mortality (HR 2.08, 95% CI [1.52–2.85]). The population attributable fraction of the concentration of Pb in blood for all-cause mortality was 18.0% (95% CI [10.9–26.1]), which is equivalent to 412,000 deaths annually. The results demonstrated that low-level environmental Pb exposure is an important risk factor for CVD mortality.117 Tubsakul et al. demonstrated that rats exposed to low level of lead acetate (100 mg/l) in the drinking water for 16 weeks induced had increases in blood pressure and peripheral vascular resistance, and decreases in blood pressure in response to intravenous infusion of acetylcholine. Additionally, rats exposed to Pb showed a downregulation of the eNOS and an upregulation of the NADPH oxidase expression.118 Interestingly, in 2016 an animal study showed an increase in Pb blood levels in rats exposed to lead acetate for 7 days (4 μg/100 g on the 1st day and 0.05 μg/100 g/day i.m. subsequently), as well as an increase in right ventricular pressures. They concluded that short-term and low-level exposure to Pb changes pulmonary hemodynamics and increases oxidative stress since vasodilatation to exogenous NO was reduced by Pb.119 More importantly, these studies have elucidated the cellular and molecular mechanisms of Pb action on cardiovascular/pulmonary systems, but a comprehensive strategy to prevent deaths from CVD should include efforts to reduce lead exposure.

Effect of Low/High Mercury/Lead/Cadmium Exposure on Endothelium-derived Vasoactive Factors

Article image

Global mercury emission inventories include anthropogenic emissions, contributing via current use or presence of mercury in a variety of products and processes, such as mercury-based artisanal and small-scale gold mining, as well as natural source emissions.120,121 Altogether, the release of this element into the environment leads to the pollution of air, water and soil. In addition, the inorganic form of mercury (Hg2+ ion) has deleterious effects in the kidney, whereas the methylated form of mercury (MeHg), which bioaccumulates in rice since its consumption is the primary pathway for methylmercury exposure at inland mercury (Hg) mining areas of China, targets the central nervous system (CNS) and has been classified as carcinogen since 1993.102,122,123 The most common route of exposure to mercury is through contaminated fish and other seafood.124 An association between the environmental and occupational exposure to mercury and the risk of CVDs was established after follow-up studies of severe cases of poisoning, and studies also showed cardiovascular abnormalities in Iraq and in Minamata, Japan.125,126 As it was introduced, endothelial dysfunction is characterised by a shift in the actions of the endothelium toward reduced vasodilation, a proinflammatory state and prothrombic properties. For this reason, there are some studies that associate chronic exposure to mercury and its derivatives with an increase of vascular resistance and an induction of hypertension.127–129 During the last years, it has been difficult to explain the definitive pathogenesis owing to the fact that the individuals exposed to mercury usually display multiple systemic disorders, including circadian clock dysregulation.113

For this reason, the proposed mechanisms are described by animal models. Golpon et al. demonstrated that mercury chloride (HgCl2) induces an endothelial-dependent vasorelaxation, which was totally blocked by the nitric oxide inhibitor L-NAME using norepinephrine preconstricted rat aorta and pulmonary artery rings with intact endothelium. Therefore, they concluded that HgCl2 alters structure and function of vascular endothelial cells.130 In 2014, similar results were reported that identified mercury exposure as causing a biphasic response: vasorelaxation at lower concentrations and vasoconstriction at higher concentrations. They determined that at low concentrations, mercury caused an endothelium-dependent vasorelaxation and at higher concentrations, mercury changes the composition and functionality of the vascular endothelium.131 These studies indicate that mercury produces a dual/biphasic response. In response to low mercury exposure, an increase in the eNOS activity/expression causes an increase in NO with no effect on prostacyclin (PGl2) and endothelium-derived hyperpolarising factor (EDHF). Both high and low mercury exposure also causes oxidative stress, and a fast interaction between NO and superoxide anions results in the formation of peroxynitrite (ONOO-), which causes vasoconstriction. However, in response to high mercury exposure, a decrease in the eNOS activity/expression causes a decrease in NO with persistence or upregulation of EDHF. Conversely, the endothelium is able to secrete the potent vasoconstrictor ET-1, which through its proinflammatory and mitogenic effects, augments the pathogenesis of CVD (Figure 3).133

Epidemiological/Observational Evidence on the Association of Environmental Risk Factors and Surrogate Markers of Endothelial Dysfunction

Air Pollution

There is mounting evidence from epidemiological/observational studies that exposure to various air pollutants, in particular to fine particulate matter (PM2.5), is associated with impaired endothelial function (Supplementary Material Table 2).11 In the Multi-Ethnic Study of Atherosclerosis (MESA)-Air (Multi-Ethnic Study of Atherosclerosis and Air Pollution), long-term PM2.5 exposure was independently associated with a 0.3% decrease in flow-mediated dilation (FMD per interquartile increase in 3,040 subjects).134 Herein, no meaningful association was observed when investigating the short-term variation in PM2.5. A further study from the MESA cohort also indicated a positive association between PM2.5 and E-selectin.135 Likewise, analyses in 5,112 subjects from the framing cohort revealed that long-term PM2.5 exposure was associated with decreased FMD and hyperemic flow velocity.136 Weaker or no evidence for an association between endothelial dysfunction and PM2.5 was found in the Framingham cohort when investigating the hyperemic response measured by peripheral arterial tonometry.137 This result was also confirmed in a study by Ljungman et al.138 A meta-analysis of six studies by Sun et al. demonstrated that increases in vascular endothelial growth factor (VEGF) were associated with PM2.5 mass concentration.139 A more recent study (n=100) by Riggs et al. showed that a 12.4% decrease in reactive hyperemia index (RHI) was related to increased PM2.5.140 Moreover, PM2.5 was positively associated with VGEF and intracellular adhesion molecule-1 (ICAM-1), whereas a negative association was determined with vascular cell adhesion molecule-1 (VCAM-1). In a study investigating the interplay between endothelial function, oxidative stress and inflammation in young adults, episodic increases in PM2.5 exposure was related to higher levels of endothelial microparticles (annexin V+/CD41-/CD31+) along with increases in markers of endothelial adhesion, i.e. adhesion soluble intercellular adhesion molecule 1 (sICAM-1) and soluble vascular cellular adhesion molecule 1 (sVCAM-1).141 Data from 704 elderly men participating in the Veterans Administration Normative Aging Study suggested an association between short- and intermediate-term air pollution exposure (ozone (O3), particle number, black carbon, nitrogen dioxide (NO2), carbon monoxide (CO), sulfates and/or PM2.5) and ICAM-1 and VCAM-1.142 A study that further investigated the influence of different components of PM2.5 revealed that independent of PM2.5 mass vanadium was related to ICAM-1 and VCAM-1.143 In line, in a panel of 40 healthy adults from China, total PM2.5 from dust/soil and several crustal and transition metals were shown to be associated with increased endothelin 1 (ET-1) and ICAM-1 and decreased VCAM-1.144 Furthermore, CO was associated with higher ICAM-1, whereas nitric oxide (NO) was shown to be associated with decreased ICAM-1. Interestingly, endothelial dysfunction as displayed by increased levels of ET-1, E-selectin and ICAM-1 was shown to partly mediate the effects of personal exposure to PM2.5 components on blood pressure during pregnancy.145 In a recent study using data from the Heredity and Phenotype Intervention (HAPI) Heart Study, exposure to PM2.5 and PM10 was inversely associated with FMD.146

Noise Exposure

Recent comprehensive evidence on the association between noise exposure and endothelial dysfunction comes mainly from a series of field studies that was conducted by our noise research group. Here we could demonstrate that exposure to simulated night-time traffic noise is consistently associated with decreased FMD in healthy adults and patients at increased cardiovascular risk. In a first study, healthy participants were subjected to three different aircraft noise scenarios (0 overflights, 30 and 60 overflights) in a randomised manner.147 Night-time aircraft noise exposure led to decreased FMD (control group: 10.4%; noise 30: 9.7%; noise 60: 9.5%). In addition, pulse transit time was reduced after exposure to noise (control group: 271.8 ms; noise 30: 270.9 ms; noise 60: 264.9 ms), a parameter associated with increased blood pressure, vascular tone and stiffness. The noise-related deterioration of the FMD was particularly pronounced when the subjects were first exposed to the noise scenario 30 and then 60 simulated overflights, so that at the vessel level a noise-related sensitisation rather than a habituation could be observed (priming effect). Interestingly, the worsening of FMD in a small subgroup of subjects (n=5) exposed to noise scenario 60 could be improved by a single dose of vitamin C. This suggests that an increased formation of vascular reactive oxygen species may be at least partly involved in the pathogenesis of endothelial dysfunction. In a follow-up study, patients with present coronary artery disease or at increased risk were randomised to two different noise scenarios (60 overflights with a mean sound pressure level of 46.9 dB and a control scenario with usual ambient noise with a mean sound pressure level of 39.2 dB).148 Interestingly, the negative effects on endothelial function were more pronounced in patients with coronary artery disease (control group: FMD 9.6%; noise 60: FMD 7.9%) when compared to healthy subjects in the previous study. The comparison of 60 versus 120 overflights in a following study resulted in a FMD of 7.27 and 7.21%, respectively, compared with a FMD of 10.02% in the control night.149 The negative effects of simulated railway noise on endothelial function have also been demonstrated.150 Herein, simulated railway noise with a peak noise level of 65 dB and an average sound pressure level of up to 54 dB(A) (30–60 trains per night) led to a substantial deterioration of endothelial function in healthy subjects. As demonstrated previously, endothelial function could be significantly improved by the acute administration of vitamin C (2 g p.o.).

Conclusion

In the present review, we provide deep mechanistic insight into the molecular basis of noise, air, or heavy metal pollution-induced endothelial dysfunction. In 2019, the fourth largest risk factor for attributable deaths among males (ambient particulate matter and ambient ozone pollution accounting for 3.75 million deaths or 12.2 % of all male deaths in 2019) and females (2.92 million deaths or 11.3 % of all female deaths) was air pollution.69 There is also a wide range of other risk factors not yet included, such as nitrogen dioxide, heavy metals, environmental noise, sleep, mental (psychological) stress and UV radiation, among others. Furthermore, the WHO even estimates that up to 12.6 million deaths worldwide in 2012 were due to living in unhealthy environments, which means 24% of global deaths (and 28% of deaths among children under five) are due to modifiable environmental factors.151 Accordingly, there are numerous studies in the literature (summarised in Supplementary Material Tables 1 and 2) that have shown that the pollutants mentioned here cause oxidative stress and inflammatory effects on the cardiovascular system, impairing the health of the endothelium. It is known that a healthy endothelium is an important regulator of blood pressure both in terms of vascular tone modulation and as an interactive junction for the immune and cardiovascular systems. Undoubtedly, active research conducted on improved preclinical models associated with various pathways of protection against environmental risk factors, which could prevent the failure of the vascular function that contributes to the development of cardiovascular and neuronal diseases, as well as data obtained from the ongoing clinical research will provide answers to the currently open questions.

Click here to view Supplementary Material.

References

  1. Munzel T, Sorensen M, Gori T, et al. Environmental stressors and cardio-metabolic disease: part II – mechanistic insights. Eur Heart J 2017;38:557–64.
    Crossref | PubMed
  2. Munzel T, Sorensen M, Gori T, et al. Environmental stressors and cardio-metabolic disease: part I – epidemiologic evidence supporting a role for noise and air pollution and effects of mitigation strategies. Eur Heart J 2017;38:550–6.
    Crossref | PubMed
  3. Newby DE, Mannucci PM, Tell GS, et al. Expert position paper on air pollution and cardiovascular disease. Eur Heart J 2015;36:83–93b.
    Crossref | PubMed
  4. Claeys MJ, Rajagopalan S, Nawrot TS, Brook RD. Climate and environmental triggers of acute myocardial infarction. Eur Heart J 2017;38:955–60.
    Crossref | PubMed
  5. Brook RD, Rajagopalan S, Pope CA 3rd, et al. Particulate matter air pollution and cardiovascular disease: an update to the scientific statement from the American Heart Association. Circulation 2010;121:2331–78.
    Crossref | PubMed
  6. Cohen AJ, Brauer M, Burnett R, et al. Estimates and 25-year trends of the global burden of disease attributable to ambient air pollution: an analysis of data from the Global Burden of Diseases Study 2015. Lancet 2017;389:1907–18.
    Crossref | PubMed
  7. Fu Z, Xi S. The effects of heavy metals on human metabolism. Toxicol Mech Methods 2020;30:167–76.
    Crossref | PubMed
  8. Rahman Z, Singh VP. The relative impact of toxic heavy metals (THMs) (arsenic (As), cadmium (Cd), chromium (Cr)(VI), mercury (Hg), and lead (Pb)) on the total environment: an overview. Environ Monit Assess 2019;191:419.
    Crossref | PubMed
  9. Alengebawy A, Abdelkhalek ST, Qureshi SR, Wang MQ. Heavy metals and pesticides toxicity in agricultural soil and plants: ecological risks and human health implications. Toxics 2021;9:42.
    Crossref | PubMed
  10. Munzel T, Sorensen M, Daiber A. Transportation noise pollution and cardiovascular disease. Nat Rev Cardiol 2021;18:619–36.
    Crossref | PubMed
  11. Munzel T, Gori T, Al-Kindi S, et al. Effects of gaseous and solid constituents of air pollution on endothelial function. Eur Heart J 2018;39:3543–50.
    Crossref | PubMed
  12. Martins AC, Santos AAD, Lopes ACBA, et al. Endothelial dysfunction induced by cadmium and mercury and its relationship to hypertension. Curr Hypertens Rev 2021;17:14–26.
    Crossref | PubMed
  13. Brook RD, Brook JR, Urch B, et al. Inhalation of fine particulate air pollution and ozone causes acute arterial vasoconstriction in healthy adults. Circulation 2002;105:1534–6.
    Crossref | PubMed
  14. Daiber A, Steven S, Weber A, et al. Targeting vascular (endothelial) dysfunction. Br J Pharmacol 2017;174:1591–619.
    Crossref | PubMed
  15. Gabella G. Complex structure of the common carotid artery of sheep. Anat Rec 1995;243:376–83.
    Crossref | PubMed
  16. Xia N, Horke S, Habermeier A, et al. Uncoupling of endothelial nitric oxide synthase in perivascular adipose tissue of diet-induced obese mice. Arterioscler Thromb Vasc Biol 2016;36:78–85.
    Crossref | PubMed
  17. Wilson DP. Vascular smooth muscle structure and function. In: Fitridge R, Thompson M, eds. Mechanisms of vascular disease: a reference book for vascular specialists. Adelaide: University of Adelaide Press, 2011.
    Crossref
  18. Hanke H, Strohschneider T, Oberhoff M, et al. Time course of smooth muscle cell proliferation in the intima and media of arteries following experimental angioplasty. Circ Res 1990;67:651–9.
    Crossref | PubMed
  19. Swift MR, Weinstein BM. Arterial-venous specification during development. Circ Res 2009;104:576–88.
    Crossref | PubMed
  20. Marziano C, Genet G, Hirschi KK. Vascular endothelial cell specification in health and disease. Angiogenesis 2021;24:213–36.
    Crossref | PubMed
  21. Tousoulis D, Kampoli AM, Tentolouris C, et al. The role of nitric oxide on endothelial function. Curr Vasc Pharmacol 2012;10:4–18.
    Crossref | PubMed
  22. Hirschi KK, Rohovsky SA, D’Amore PA. PDGF, TGF-beta, and heterotypic cell-cell interactions mediate endothelial cell-induced recruitment of 10T1/2 cells and their differentiation to a smooth muscle fate. J Cell Biol 1998;141:805–14.
    Crossref | PubMed
  23. Hirschi KK, Rohovsky SA, Beck LH, et al. Endothelial cells modulate the proliferation of mural cell precursors via platelet-derived growth factor-BB and heterotypic cell contact. Circ Res 1999;84:298–305.
    Crossref | PubMed
  24. Munzel T, Daiber A, Ullrich V, Mulsch A. Vascular consequences of endothelial nitric oxide synthase uncoupling for the activity and expression of the soluble guanylyl cyclase and the cGMP-dependent protein kinase. Arterioscler Thromb Vasc Biol 2005;25:1551–7.
    Crossref | PubMed
  25. Forstermann U, Munzel T. Endothelial nitric oxide synthase in vascular disease: from marvel to menace. Circulation 2006;113:1708–14.
    Crossref | PubMed
  26. Schulz E, Jansen T, Wenzel P, et al. Nitric oxide, tetrahydrobiopterin, oxidative stress, and endothelial dysfunction in hypertension. Antioxid Redox Signal 2008;10:1115–26.
    Crossref | PubMed
  27. Matsubara M, Titani K, Taniguchi H. Interaction of calmodulin-binding domain peptides of nitric oxide synthase with membrane phospholipids: regulation by protein phosphorylation and Ca2+-calmodulin. Biochemistry 1996;35:14651–8.
    Crossref | PubMed
  28. Fleming I. Molecular mechanisms underlying the activation of eNOS. Pflugers Arch 2010;459:793–806.
    Crossref | PubMed
  29. Furchgott RF, Zawadzki JV. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature 1980;288:373–6.
    Crossref | PubMed
  30. Chen Z, Stamler JS. Bioactivation of nitroglycerin by the mitochondrial aldehyde dehydrogenase. Trends Cardiovasc Med 2006;16:259–65.
    Crossref | PubMed
  31. Konukoglu D, Uzun H. Endothelial dysfunction and hypertension. Adv Exp Med Biol 2017;956:511–40.
    Crossref | PubMed
  32. Vita JA, Treasure CB, Nabel EG, et al. Coronary vasomotor response to acetylcholine relates to risk factors for coronary artery disease. Circulation 1990;81:491–7.
    Crossref | PubMed
  33. Balligand JL, Feron O, Dessy C. eNOS activation by physical forces: from short-term regulation of contraction to chronic remodeling of cardiovascular tissues. Physiol Rev 2009;89:481–534.
    Crossref | PubMed
  34. Schulz E, Wenzel P, Munzel T, Daiber A. Mitochondrial redox signaling: interaction of mitochondrial reactive oxygen species with other sources of oxidative stress. Antioxid Redox Signal 2014;20:308–24.
    Crossref | PubMed
  35. Gimbrone MA, Jr, Garcia-Cardena G. Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ Res 2016;118:620–36.
    Crossref | PubMed
  36. Mordi I, Mordi N, Delles C, Tzemos N. Endothelial dysfunction in human essential hypertension. J Hypertens 2016;34:1464–72.
    Crossref | PubMed
  37. Calver A, Collier J, Vallance P. Inhibition and stimulation of nitric oxide synthesis in the human forearm arterial bed of patients with insulin-dependent diabetes. J Clin Invest 1992;90:2548–54.
    Crossref | PubMed
  38. van Boven AJ, Jukema JW, Paoletti R. Endothelial dysfunction and dyslipidemia: possible effects of lipid lowering and lipid modifying therapy. Pharmacol Res 1994;29:261–72.
    Crossref | PubMed
  39. Heitzer T, Brockhoff C, Mayer B, et al. Tetrahydrobiopterin improves endothelium-dependent vasodilation in chronic smokers: evidence for a dysfunctional nitric oxide synthase. Circ Res 2000;86:e36–41.
    Crossref | PubMed
  40. Heitzer T, Just H, Munzel T. Antioxidant vitamin C improves endothelial dysfunction in chronic smokers. Circulation 1996;94:6–9.
    Crossref | PubMed
  41. Budhiraja R, Parthasarathy S, Quan SF. Endothelial dysfunction in obstructive sleep apnea. J Clin Sleep Med 2007;3:409–15.
    Crossref | PubMed
  42. Cherubini JM, Cheng JL, Williams JS, MacDonald MJ. Sleep deprivation and endothelial function: reconciling seminal evidence with recent perspectives. Am J Physiol Heart Circ Physiol 2021;320:H29–35.
    Crossref | PubMed
  43. Steven S, Frenis K, Oelze M, et al. Vascular inflammation and oxidative stress: major triggers for cardiovascular disease. Oxid Med Cell Longev 2019;2019:7092151.
    Crossref | PubMed
  44. Sher LD, Geddie H, Olivier L, et al. Chronic stress and endothelial dysfunction: mechanisms, experimental challenges, and the way ahead. Am J Physiol Heart Circ Physiol 2020;319:H488–506.
    Crossref | PubMed
  45. Forstermann U, Sessa WC. Nitric oxide synthases: regulation and function. Eur Heart J 2012;33:829-837.
    Crossref | PubMed
  46. Gliozzi M, Scicchitano M, Bosco F, et al. Modulation of nitric oxide synthases by oxidized LDLs: role in vascular inflammation and atherosclerosis development. Int J Mol Sci 2019;20.
    Crossref | PubMed
  47. Herrmann HJ, Rohde HG, Schulze W, et al. Effect of noise stress and ethanol intake on hearts of spontaneously hypertensive rats. Basic Res Cardiol 1994;89:510–23.
    Crossref | PubMed
  48. Antunes E, Oliveira P, Borrecho G, et al. Myocardial fibrosis in rats exposed to low frequency noise. Acta Cardiol 2013;68:241–5.
    Crossref | PubMed
  49. Gannouni N, Mhamdi A, El May M, et al. Morphological changes of adrenal gland and heart tissue after varying duration of noise exposure in adult rat. Noise Health 2014;16:416–21.
    Crossref | PubMed
  50. Dengerink H, Miller J, Axelsson A, et al. The recovery of vascular changes following brief noise exposure. Acta Otolaryngol 1985;100:19–25.
    Crossref | PubMed
  51. Arpornchayanon W, Canis M, Ihler F, et al. TNF-alpha inhibition using etanercept prevents noise-induced hearing loss by improvement of cochlear blood flow in vivo. Int J Audiol 2013;52:545–52.
    Crossref | PubMed
  52. Konkle ATM, Keith SE, McNamee JP, Michaud D. Chronic noise exposure in the spontaneously hypertensive rat. Noise Health 2017;19:213–21.
    Crossref | PubMed
  53. Wu CC, Chen SJ, Yen MH. Attenuation of endothelium-dependent relaxation in mesenteric artery during noise-induced hypertension. J Biomed Sci 1994;1:49–53.
    Crossref | PubMed
  54. Altura BM, Altura BT, Gebrewold A, et al. Noise-induced hypertension and magnesium in rats: relationship to microcirculation and calcium. J Appl Physiol (1985) 1992;72:194–202.
    Crossref | PubMed
  55. Said MA, El-Gohary OA. Effect of noise stress on cardiovascular system in adult male albino rat: implication of stress hormones, endothelial dysfunction and oxidative stress. Gen Physiol Biophys 2016;35:371–7.
    Crossref | PubMed
  56. Munzel T, Sorensen M, Schmidt F, et al. The adverse effects of environmental noise exposure on oxidative stress and cardiovascular risk. Antioxid Redox Signal 2018;28:873–908.
    Crossref | PubMed
  57. Munzel T, Daiber A, Steven S, et al. Effects of noise on vascular function, oxidative stress, and inflammation: mechanistic insight from studies in mice. Eur Heart J 2017;38:2838–49.
    Crossref | PubMed
  58. Kroller-Schon S, Daiber A, Steven S, et al. Crucial role for Nox2 and sleep deprivation in aircraft noise-induced vascular and cerebral oxidative stress, inflammation, and gene regulation. Eur Heart J 2018;39:3528–39.
    Crossref | PubMed
  59. Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol Rev 2007;87:315–424.
    Crossref | PubMed
  60. Steven S, Frenis K, Kalinovic S, et al. Exacerbation of adverse cardiovascular effects of aircraft noise in an animal model of arterial hypertension. Redox Biol 2020;34:101515.
    Crossref | PubMed
  61. Frenis K, Helmstadter J, Ruan Y, et al. Ablation of lysozyme M-positive cells prevents aircraft noise-induced vascular damage without improving cerebral side effects. Basic Res Cardiol 2021;116:31.
    Crossref | PubMed
  62. Liu L, Huang Y, Fang C, et al. Chronic noise-exposure exacerbates insulin resistance and promotes the manifestations of the type 2 diabetes in a high-fat diet mouse model. PLoS One 2018;13:e0195411.
    Crossref | PubMed
  63. Munzel T, Steven S, Hahad O, Daiber A. Noise and cardiovascular risk: nighttime aircraft noise acutely triggers cardiovascular death. Eur Heart J 2021;42:844–6.
    Crossref | PubMed
  64. Sorensen M, Pershagen G. Transportation noise linked to cardiovascular disease independent from air pollution. Eur Heart J 2019;40:604–6.
    Crossref | PubMed
  65. Hahad O, Bayo Jimenez MT, Kuntic M, et al. Cerebral consequences of environmental noise exposure. Environ Int 2022;165:107306.
    Crossref | PubMed
  66. Daiber A, Frenis K, Kuntic M, et al. Redox regulatory changes of circadian rhythm by the environmental risk factors traffic noise and air pollution. Antioxid Redox Signal 2022;37:679–703.
    Crossref | PubMed
  67. Frenis K, Kalinovic S, Ernst BP, et al. Long-term effects of aircraft noise exposure on vascular oxidative stress, endothelial function and blood pressure: no evidence for adaptation or tolerance development. Front Mol Biosci 2021;8:814921.
    Crossref | PubMed
  68. Kampa M, Castanas E. Human health effects of air pollution. Environ Pollut 2008;151:362–7.
    Crossref | PubMed
  69. Murray CJL, Aravkin AY, Zheng P, et al. Global burden of 87 risk factors in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020;396:1223–49.
    Crossref | PubMed
  70. WHO. Air pollution. 2022. https://www.who.int/health-topics/air-pollution#tab=tab_1 (accessed 31 January 2022).
  71. Lelieveld J, Klingmuller K, Pozzer A, et al. Cardiovascular disease burden from ambient air pollution in Europe reassessed using novel hazard ratio functions. Eur Heart J 2019;40:1590–6.
    Crossref | PubMed
  72. Daiber A, Kuntic M, Hahad O, et al. Effects of air pollution particles (ultrafine and fine particulate matter) on mitochondrial function and oxidative stress – implications for cardiovascular and neurodegenerative diseases. Arch Biochem Biophys 2020;696:108662.
    Crossref | PubMed
  73. Buckley RD, Hackney JD, Clark K, Posin C. Ozone and human blood. Arch Environ Health 1975;30:40–3.
    Crossref | PubMed
  74. Kreyling WG, Hirn S, Schleh C. Nanoparticles in the lung. Nat Biotechnol 2010;28:1275–6.
    Crossref | PubMed
  75. Miller MR, Raftis JB, Langrish JP, et al. Inhaled nanoparticles accumulate at sites of vascular disease. ACS Nano 2017;11:4542–52.
    Crossref | PubMed
  76. Prunicki M, Cauwenberghs N, Ataam JA, et al. Immune biomarkers link air pollution exposure to blood pressure in adolescents. Environ Health 2020;19:108.
    Crossref | PubMed
  77. Dellinger B, Pryor WA, Cueto R, et al. Role of free radicals in the toxicity of airborne fine particulate matter. Chem Res Toxicol 2001;14:1371–7.
    Crossref | PubMed
  78. Drummond GR, Selemidis S, Griendling KK, Sobey CG. Combating oxidative stress in vascular disease: NADPH oxidases as therapeutic targets. Nat Rev Drug Discov 2011;10:453–71.
    Crossref | PubMed
  79. Wenzel P, Kossmann S, Munzel T, Daiber A. Redox regulation of cardiovascular inflammation - immunomodulatory function of mitochondrial and Nox-derived reactive oxygen and nitrogen species. Free Radic Biol Med 2017;109:48–60.
    Crossref | PubMed
  80. Sun Q, Yue P, Ying Z, et al. Air pollution exposure potentiates hypertension through reactive oxygen species-mediated activation of Rho/ROCK. Arterioscler Thromb Vasc Biol 2008;28:1760–6.
    Crossref | PubMed
  81. Kampfrath T, Maiseyeu A, Ying Z, et al. Chronic fine particulate matter exposure induces systemic vascular dysfunction via NADPH oxidase and TLR4 pathways. Circ Res 2011;108:716–26.
    Crossref | PubMed
  82. Chuang GC, Yang Z, Westbrook DG, et al. Pulmonary ozone exposure induces vascular dysfunction, mitochondrial damage, and atherogenesis. Am J Physiol Lung Cell Mol Physiol 2009;297:L209–16.
    Crossref | PubMed
  83. Paffett ML, Zychowski KE, Sheppard L, et al. Ozone inhalation impairs coronary artery dilation via intracellular oxidative stress: evidence for serum-borne factors as drivers of systemic toxicity. Toxicol Sci 2015;146:244–53.
    Crossref | PubMed
  84. Kodavanti UP, Thomas R, Ledbetter AD, et al. Vascular and cardiac impairments in rats inhaling ozone and diesel exhaust particles. Environ Health Perspect 2011;119:312–8.
    Crossref | PubMed
  85. Rao X, Zhong J, Maiseyeu A, et al. CD36-dependent 7-ketocholesterol accumulation in macrophages mediates progression of atherosclerosis in response to chronic air pollution exposure. Circ Res 2014;115:770–80.
    Crossref | PubMed
  86. Pryor WA, Squadrito GL. The chemistry of peroxynitrite: a product from the reaction of nitric oxide with superoxide. Am J Physiol 1995;268:L699–722.
    Crossref | PubMed
  87. Courtois A, Andujar P, Ladeiro Y, et al. Impairment of NO-dependent relaxation in intralobar pulmonary arteries: comparison of urban particulate matter and manufactured nanoparticles. Environ Health Perspect 2008;116:1294–9.
    Crossref | PubMed
  88. Centers for Disease Control and Prevention. Outbreak of lung injury associated with the use of e-cigarette, or vaping, products. 2020. https://www.cdc.gov/tobacco/basic_information/e-cigarettes/severe-lung-disease.html (accessed 31 January 2022).
  89. Poschl U, Shiraiwa M. Multiphase chemistry at the atmosphere-biosphere interface influencing climate and public health in the Anthropocene. Chem Rev 2015;115:4440–75.
    Crossref | PubMed
  90. Ying Z, Xu X, Chen M, et al. A synergistic vascular effect of airborne particulate matter and nickel in a mouse model. Toxicol Sci 2013;135:72–80.
    Crossref | PubMed
  91. Radan M, Dianat M, Badavi M, et al. Gallic acid protects particulate matter (PM10) triggers cardiac oxidative stress and inflammation causing heart adverse events in rats. Environ Sci Pollut Res Int 2019;26:18200–7.
    Crossref | PubMed
  92. Ho CC, Tsai MH, Chen YC, et al. Persistent elevation of blood pressure by ambient coarse particulate matter after recovery from pulmonary inflammation in mice. Environ Toxicol 2019;34:814–24.
    Crossref | PubMed
  93. Guo L, Zhu N, Guo Z, et al. Particulate matter (PM10) exposure induces endothelial dysfunction and inflammation in rat brain. J Hazard Mater 2012;213–214:28–37.
    Crossref | PubMed
  94. Xu Z, Wang W, Liu Q, et al. Association between gaseous air pollutants and biomarkers of systemic inflammation: a systematic review and meta-analysis. Environ Pollut 2022;292:118336.
    Crossref | PubMed
  95. Snow SJ, Henriquez AR, Thompson LC, et al. Pulmonary and vascular effects of acute ozone exposure in diabetic rats fed an atherogenic diet. Toxicol Appl Pharmacol 2021;415:115430.
    Crossref | PubMed
  96. Bayo Jimenez MT, Frenis K, Hahad O, et al. Protective actions of nuclear factor erythroid 2-related factor 2 (NRF2) and downstream pathways against environmental stressors. Free Radic Biol Med 2022;187:72–91.
    Crossref | PubMed
  97. Wang Y, Xiong L, Huang X, et al. Intermittent exposure to airborne particulate matter induces subcellular dysfunction and aortic cell damage in BALB/c mice through multi-endpoint assessment at environmentally relevant concentrations. J Hazard Mater 2022;424:127169.
    Crossref | PubMed
  98. Rojas GA, Saavedra N, Saavedra K, et al. Polycyclic aromatic hydrocarbons (PAHs) exposure triggers inflammation and endothelial dysfunction in BALB/c mice: a pilot study. Toxics 2022;10:1–14.
    Crossref | PubMed
  99. Vaziri ND. Mechanisms of lead-induced hypertension and cardiovascular disease. Am J Physiol Heart Circ Physiol 2008;295:H454–65.
    Crossref | PubMed
  100. Cosselman KE, Navas-Acien A, Kaufman JD. Environmental factors in cardiovascular disease. Nat Rev Cardiol 2015;12:627–42.
    Crossref | PubMed
  101. Lee JW, Choi H, Hwang UK, et al. Toxic effects of lead exposure on bioaccumulation, oxidative stress, neurotoxicity, and immune responses in fish: a review. Environ Toxicol Pharmacol 2019;68:101–8.
    Crossref | PubMed
  102. IARC Working Group. Beryllium, cadmium, mercury, and exposures in the glass manufacturing industry. Lyon: WHO, 1993;58:1–415.
  103. McElroy JA, Kruse RL, Guthrie J, et al. Cadmium exposure and endometrial cancer risk: a large midwestern U.S. population-based case-control study. PLoS One 2017;12:e0179360.
    Crossref | PubMed
  104. Suria A, Nasreen R, Saeed SA. GABA involvement in naloxone induced reversal of respiratory paralysis produced by thiopental. Life Sci 1988;42:643–50.
    Crossref | PubMed
  105. Kukongviriyapan U, Apaijit K, Kukongviriyapan V. Oxidative stress and cardiovascular dysfunction associated with cadmium exposure: beneficial effects of curcumin and tetrahydrocurcumin. Tohoku J Exp Med 2016;239:25–38.
    Crossref | PubMed
  106. Lukkhananan P, Thawonrachat N, Srihirun S, et al. Endothelial dysfunction in subjects with chronic cadmium exposure. J Toxicol Sci 2015;40:605–13.
    Crossref | PubMed
  107. Liang H, Yue R, Zhou C, et al. Cadmium exposure induces endothelial dysfunction via disturbing lipid metabolism in human microvascular endothelial cells. J Appl Toxicol 2021;41:775–88.
    Crossref | PubMed
  108. Messner B, Knoflach M, Seubert A, et al. Cadmium is a novel and independent risk factor for early atherosclerosis mechanisms and in vivo relevance. Arterioscler Thromb Vasc Biol 2009;29:1392–8.
    Crossref | PubMed
  109. Messner B, Bernhard D. Cadmium and cardiovascular diseases: cell biology, pathophysiology, and epidemiological relevance. Biometals 2010;23:811–22.
    Crossref | PubMed
  110. Angeli JK, Pereira CA, C.A, de Oliveira Faria T, et al. Cadmium exposure induces vascular injury due to endothelial oxidative stress: the role of local angiotensin II and COX-2. Free Radic Biol Med 2013;65:838–48.
    Crossref | PubMed
  111. Donpunha W, Kukongviriyapan U, Sompamit K, et al. Protective effect of ascorbic acid on cadmium-induced hypertension and vascular dysfunction in mice. Biometals 2011;24:105–15.
    Crossref | PubMed
  112. Yoopan N, Watcharasit P, Wongsawatkul O, et al. Attenuation of eNOS expression in cadmium-induced hypertensive rats. Toxicol Lett 2008;176:157–61.
    Crossref | PubMed
  113. Li H, Kilgallen AB, Munzel T, et al. Influence of mental stress and environmental toxins on circadian clocks: implications for redox regulation of the heart and cardioprotection. Br J Pharmacol 2020;177:5393–412.
    Crossref | PubMed
  114. Kim JH, Kang JC. The immune responses in juvenile rockfish, Sebastes schlegelii for the stress by the exposure to the dietary lead (II). Environ Toxicol Pharmacol 2016;46:211–6.
    Crossref | PubMed
  115. Landrigan PJ, Fuller R, Acosta NJR, et al. The Lancet Commission on pollution and health. Lancet 2018;391:462–512.
    Crossref | PubMed
  116. Wang T, Zhou YP, Sun Y, Zheng YX. Trends in blood lead levels in the U.S. From 1999 to 2016. Am J Prev Med 2021;60:e179–87.
    Crossref | PubMed
  117. Lanphear BP, Rauch S, Auinger P, et al. Low-level lead exposure and mortality in US adults: a population-based cohort study. Lancet Public Health 2018;3:e177–84.
    Crossref | PubMed
  118. Tubsakul A, Sangartit W, Pakdeechote P, et al. Curcumin mitigates hypertension, endothelial dysfunction and oxidative stress in rats with chronic exposure to lead and cadmium. Tohoku J Exp Med 2021;253:69–76.
    Crossref | PubMed
  119. Covre EP, Freire DD, Jr, Dalfior BM, et al. Low-level lead exposure changes endothelial modulation in rat resistance pulmonary arteries. Vascul Pharmacol 2016;85:21–8.
    Crossref | PubMed
  120. Esdaile LJ, Chalker JM. The mercury problem in artisanal and small-scale gold mining. Chemistry 2018;24:6905–16.
    Crossref | PubMed
  121. Kocman D, Horvat M, Pirrone N, Cinnirella S. Contribution of contaminated sites to the global mercury budget. Environ Res 2013;125:160–70.
    Crossref | PubMed
  122. Wang Z, Sun T, Driscoll CT, et al. Mechanism of accumulation of methylmercury in rice (Oryza sativa L.) in a mercury mining area. Environ Sci Technol 2018;52:9749–57.
    Crossref | PubMed
  123. Carocci A, Rovito N, Sinicropi MS, Genchi G. Mercury toxicity and neurodegenerative effects. Rev Environ Contam Toxicol 2014;229:1–18.
    Crossref
  124. Sheehan MC, Burke TA, Navas-Acien A, et al. Global methylmercury exposure from seafood consumption and risk of developmental neurotoxicity: a systematic review. Bull World Health Organ 2014;92:254–69f.
    Crossref | PubMed
  125. Jalili MA, Abbasi AH. Poisoning by ethyl mercury toluene sulphonanilide. Br J Ind Med 1961;18:303–8.
    Crossref | PubMed
  126. Eto K. Minamata disease. Neuropathology 2000;20(Suppl):S14–9.
    Crossref | PubMed
  127. McGill SN, Ahmed NA, Christou NV. Endothelial cells: role in infection and inflammation. World J Surg 1998;22:171–8.
    Crossref | PubMed
  128. Garcia Gomez M, Boffetta P, Caballero Klink JD, et al. Cardiovascular mortality in mercury miners. Med Clin (Barc) 2007;128:766–71.
    Crossref | PubMed
  129. Torres AD, Rai AN, Hardiek ML. Mercury intoxication and arterial hypertension: report of two patients and review of the literature. Pediatrics 2000;105:e34.
    Crossref | PubMed
  130. Golpon HA, Puchner A, Barth P, et al. Nitric oxide-dependent vasorelaxation and endothelial cell damage caused by mercury chloride. Toxicology 2003;192:179–88.
    Crossref | PubMed
  131. Omanwar S, Saidullah B, Ravi K, Fahim M. Vasorelaxant effects of mercury on rat thoracic aorta: the nitric oxide signaling mechanism. Hum Exp Toxicol 2014;33:904–10.
    Crossref | PubMed
  132. Omanwar S, Fahim M. Mercury exposure and endothelial dysfunction: an interplay between nitric oxide and oxidative stress. Int J Toxicol 2015;34:300–7.
    Crossref | PubMed
  133. Bohm F, Pernow J. The importance of endothelin-1 for vascular dysfunction in cardiovascular disease. Cardiovasc Res 2007;76:8–18.
    Crossref | PubMed
  134. Krishnan RM, Adar SD, Szpiro AA, et al. Vascular responses to long- and short-term exposure to fine particulate matter: MESA Air (Multi-Ethnic Study of Atherosclerosis and Air Pollution). J Am Coll Cardiol 2012;60:2158–66.
    Crossref | PubMed
  135. Hajat A, Allison M, Diez-Roux AV, et al. Long-term exposure to air pollution and markers of inflammation, coagulation, and endothelial activation: a repeat-measures analysis in the Multi-Ethnic Study of Atherosclerosis (MESA). Epidemiology 2015;26:310–20.
    Crossref | PubMed
  136. Wilker EH, Ljungman PL, Rice MB, et al. Relation of long-term exposure to air pollution to brachial artery flow-mediated dilation and reactive hyperemia. Am J Cardiol 2014;113:2057–63.
    Crossref | PubMed
  137. Ljungman PL, Wilker EH, Rice MB, et al. Short-term exposure to air pollution and digital vascular function. Am J Epidemiol 2014;180:482–9.
    Crossref | PubMed
  138. Ljungman PL, Wilker EH, Rice MB, et al. The impact of multipollutant clusters on the association between fine particulate air pollution and microvascular function. Epidemiology 2016;27:194–201.
    Crossref | PubMed
  139. Sun Y, Wang Y, Yuan S, et al. Exposure to PM2.5 via vascular endothelial growth factor relationship: meta-analysis. PLoS One 2018;13:e0198813.
    Crossref | PubMed
  140. Riggs DW, Zafar N, Krishnasamy S, et al. Exposure to airborne fine particulate matter is associated with impaired endothelial function and biomarkers of oxidative stress and inflammation. Environ Res 2020;180:108890.
    Crossref | PubMed
  141. Pope CA, 3rd, Bhatnagar A, McCracken JP, et al. Exposure to fine particulate air pollution is associated with endothelial injury and systemic inflammation. Circ Res 2016;119:1204–14.
    Crossref | PubMed
  142. Bind MA, Baccarelli A, Zanobetti A, et al. Air pollution and markers of coagulation, inflammation, and endothelial function: associations and epigene-environment interactions in an elderly cohort. Epidemiology 2012;23:332–40.
    Crossref | PubMed
  143. Dai L, Bind MA, Koutrakis P, et al. Fine particles, genetic pathways, and markers of inflammation and endothelial dysfunction: analysis on particulate species and sources. J Expo Sci Environ Epidemiol 2016;26:415–21.
    Crossref | PubMed
  144. Wu S, Yang D, Pan L, et al. Chemical constituents and sources of ambient particulate air pollution and biomarkers of endothelial function in a panel of healthy adults in Beijing, China. Sci Total Environ 2016;560–561:141–9.
    Crossref | PubMed
  145. Xia B, Zhou Y, Zhu Q, et al. Personal exposure to PM2.5 constituents associated with gestational blood pressure and endothelial dysfunction. Environ Pollut 2019;250:346–56.
    Crossref | PubMed
  146. Salimi S, Yanosky JD, Huang D, et al. Long-term exposure to particulate air pollution and brachial artery flow-mediated dilation in the Old Order Amish. Environ Health 2020;19:50.
    Crossref | PubMed
  147. Schmidt FP, Basner M, Kroger G, et al. Effect of nighttime aircraft noise exposure on endothelial function and stress hormone release in healthy adults. Eur Heart J 2013;34:3508–14a.
    Crossref | PubMed
  148. Schmidt F, Kolle K, Kreuder K, et al. Nighttime aircraft noise impairs endothelial function and increases blood pressure in patients with or at high risk for coronary artery disease. Clin Res Cardiol 2015;104:23–30.
    Crossref | PubMed
  149. Schmidt FP, Herzog J, Schnorbus B, et al. The impact of aircraft noise on vascular and cardiac function in relation to noise event number: a randomized trial. Cardiovasc Res 2021;117:1382–90.
    Crossref | PubMed
  150. Herzog J, Schmidt FP, Hahad O, et al. Acute exposure to nocturnal train noise induces endothelial dysfunction and pro-thromboinflammatory changes of the plasma proteome in healthy subjects. Basic Res Cardiol 2019;114:46.
    Crossref | PubMed
  151. World Health Organization. Preventing disease through healthy environments: a global assessment of the burden of disease from environmental risks. 2018. https://www.who.int/publications/i/item/9789241565196 (accessed 3 January 2022).