Skip to main content
Toxicology Reports logoLink to Toxicology Reports
. 2022 Mar 22;9:521–533. doi: 10.1016/j.toxrep.2022.03.019

Role of environmental toxicants in the development of hypertensive and cardiovascular diseases

Ehsan Habeeb a, Saad Aldosari a, Shakil A Saghir b,c, Mariam Cheema a, Tahani Momenah a, Kazim Husain d, Yadollah Omidi a, Syed AA Rizvi e, Muhammad Akram f, Rais A Ansari a,
PMCID: PMC8971584  PMID: 35371924

Abstract

The incidence of hypertension with diabetes mellitus (DM) as a co-morbid condition is on the rise worldwide. In 2000, an estimated 972 million adults had hypertension, which is predicted to grow to 1.56 billion by 2025. Hypertension often leads to diabetes mellitus that strongly puts the patients at an increased risk of cardiovascular, kidney, and/or atherosclerotic diseases. Hypertension has been identified as a major risk factor for the development of diabetes; patients with hypertension are at two-to-three-fold higher risk of developing diabetes than patients with normal blood pressure (BP). Causes for the increase in hypertension and diabetes are not well understood, environmental factors (e.g., exposure to environmental toxicants like heavy metals, organic solvents, pesticides, alcohol, and urban lifestyle) have been postulated as one of the reasons contributing to hypertension and cardiovascular diseases (CVD). The mechanism of action(s) of these toxicants in developing hypertension and CVDs is not well defined. Research studies have linked hypertension with the chronic consumption of alcohol and exposure to metals like lead, mercury, and arsenic have also been linked to hypertension and CVD. Workers chronically exposed to styrene have a higher incidence of CVD. Recent studies have demonstrated that exposure to particulate matter (PM) in diesel exhaust and urban air contributes to increased CVD and mortality. In this review, we have imparted the role of environmental toxicants such as heavy metals, organic pollutants, PM, alcohol, and some drugs in hypertension and CVD along with possible mechanisms and limitations in extrapolating animal data to humans.

Keywords: Hypertension, Cardiovascular diseases, Diabetes mellitus, Atherosclerotic diseases, Environmental toxicants, Chronic exposure

Highlights

  • Rising incidence of hypertension may be linked to chronic exposure with environmental toxicants.

  • Urban lifestyle and alcohol intake may be responsible for increased incidence of hypertension among urbanites.

  • Exposure with organic solvent, heavy metals and pesticides could also be contributing to the rise in blood pressure.

1. Introduction

The incidence of hypertension and diabetes is on the rise worldwide [135]. A detailed analysis of hypertension indicates that median systolic blood pressure (BP) in men and women has decreased slightly among the rich industrialized nations in Europe, Central Asia, Middle East, Caribbean, and Latin America between 1975 and 2015. However, an increase in median systolic and diastolic BP is observed among Sub-Saharan Africa, South Asia, and Southeast Asia [135]. The causes for the increase in populations with hypertension are not well understood. It is believed that due to industrial activity and exposure to environmental toxicants such as diesel exhaust particles (DEP), polycyclic aromatic hydrocarbons (PAH), residues of organochlorine insecticides (OCI), polychlorinated biphenyls (PCBs), particulate matters (PM), and heavy metals (Pb, Hg, Cd, and As) may be playing a role (Table 1). Studies have demonstrated that exposure of workers to styrene, an industrial chemical, causes an increased incidence of cardiovascular diseases (CVD) [100]. Recent studies have demonstrated that exposure to DEP and urban PM increases CVD and mortality. In addition, consumption of alcohol may also contribute to hepatotoxicity and an increase in BP [12]. The role of OCI, PCBs, and heavy metals (Pb, Hg, As, and Cd) in increasing BP is not addressed systematically. These xenobiotics cause immune disruption and create inflammation, which might play a role in CVD as in increased incidences of cancer [80], [191]. These xenobiotics increase levels of interleukin (IL-1β, IL-6, and IL-8), tumor necrosis factor-α (TNFα), and C-reactive protein (CRP). The IL-1β, TNFα, and IL-6 are cytokines responsible for acute phase response (APR) from liver. The increase in CRP has been suggested to arise from APR by IL-6 [93], [208]. These cytokines can modulate APR in liver and modify secretion of proteins, like CRP and angiotensinogen (AGT), which play significant role in inflammation and BP regulation.

Table 1.

Examples of Environmental Pollutants and their Mechanism of Causing Hypertension-Related Diseases.

Class of the Compound Examples Risk of Hypertension-Related Diseases Potential Mechanisms Reference
Heavy metals Arsenic Hypertension, Atherosclerosis/Coronary Heart Disease, Stroke Increased Peroxynitrite (RNS) and inflammatory mediator; cyclooxygenase (Cox-2) formation [22]
Cadmium Hypertension, Stroke Oxidative stress, impaired nitric oxide (NO) signaling, modified vascular response to neurotransmitters, disturbed vascular muscle Ca2+ signaling, and interference with the renin-angiotensin system [43], [148]
Mercury Hypertension, Cardiovascular, Mortality Mitochondrial dysfunction, energy production process impaired, mechanism obscure [43]
Chromium Cardiovascular disease Promoting oxidative stress, limiting nitric oxide availability, impairing nitric oxide signaling, increasing adrenergic activity, and endothelin production, altering the renin-angiotensin system [213]
Lead Hypertension Increasing vasoconstrictor and decreasing vasodilator prostaglandins, promoting inflammation, and modifying the vascular response to vasoactive agonists [196]
Vanadium Cardiovascular disease risk factor Not well defined [136]
Cesium Systemic Arterial hypertension Polygenic-not well defined [156]
Methyl mercury Myocardial Infarct Risk, Atherosclerosis/ Coronary Heart Disease Oxidative stress, decrease in heart rate variability adaptation [159]
Persistent Organic Pollutants (POP) Polychlorinated biphenyls (PCBs) Hypertension, Stroke, Myocardial Infarct Risk, Cardiovascular disease Chronic oxidative stress, Inflammation [148]
Dioxins Cardiovascular disease Endocrine disruption [146]
Phthalate Increase in BP Deregulation of endothelial nitric oxide synthase and increased angiotensin-II-angiotensin receptor type-1 signaling [121]
Non-persistent chemicals Bisphenol-A Hypertension, Coronary Heart Disease/ Atherosclerosis, Myocardial Infarct oxidative stress, inflammation, endocrine disruption, and epigenetic change [74], [148]
Organophosphate metabolites Myocardial Infarct Risk Oxidative stress, disruption in autonomic function [14]
Polyaromatic hydrocarbons (PAHs) Myocardial Infarct Risk Aryl hydrocarbon receptor-mediated gene regulation [123]
Gases Carbon disulfide Hypertension, CVD Undefined [41]
Ozone Hypertension, Cardiac arrhythmias Elevated angiotensin-converting enzyme and Endothelin-1 [211]
Freon Various mechanism postulated [162]
Nitrogen dioxide Cardiovascular mortality Reactive nitrogen species (RNS), ROS formation in endothelial cells, reduced NO availability [18]
Cigarette smoke Cardiovascular disease, stroke, aortic aneurysm Persistent stimulation of sympathetic nervous system by tobacco constituent, nicotine, CO-hemoglobin complex causing hypoxemia, lower levels of antioxidants [31]
Vehicular/fuel exhaust CVD, Morbidity, and Mortality Impairment of endothelial function-NO pathway [126]
Alcohol Hypertension Several mechanisms [8], [152], [153], [175]
Vinyl chloride Hypertension, cardiac arrhythmias Portal hypertension, Injury of hepatocytes around the portal vein, and subsequent constriction due to repair, leading to portal vein mediated hypertension [190]
Trichloroethylene Pulmonary hypertension Endothelial cell injury [141], [25]
Chronic nicotine exposure Tobacco smoke Pulmonary hypertension Pulmonary vasculature endothelial dysfunction, reduced eNOS expression, inflammatory CD8 + T lymphocyte infiltration, vascular muscular hyperplasia etc. [96], [138], [170]
Particulate Matter (PM) PM0.2–2.5, PM10 Cardiovascular Mortality, Myocardial Infarct Risk, Stroke Inflammation or oxidative stress [123]
Vehicular exhaust PM Cardiovascular health risks Inflammation and oxidative stress [67]
Second-hand cigarette smoke PM Coronary Heart Disease (CHD) Endothelial dysfunction, inflammation, oxidative stress [11]
Solid & liquid fuel burning for cooking PM Risk of CHD, positive association with hypertension and heart disease especially among women disproportionally exposed due to cooking Increased Oxidative stress & inflammatory cytokines [50], [127]

The BP is the force of blood that is pushed against the walls of different arteries as the heart pumps blood. As per the NIH guideline for blood pressure (European guideline discussed later in comparison to revised guideline in the USA), a normal BP is defined as < 120 mm Hg systolic and < 80 mmHg diastolic BP. The BP is considered elevated when systolic BP is ≥ 120 mm Hg and diastolic is > 80 mm Hg. Prehypertension is defined when systolic BP is > 120 and < 140 while diastolic BP is > 80 but < 89 mm Hg. A stage 1 hypertension is defined when systolic BP is between 140 and 159 mm Hg and diastolic is between 90 and 99 mm Hg. It is stage 2 hypertension when systolic and diastolic BPs are ≥ 160 and ≥ 100 mm Hg, respectively (Table 2) [134].

Table 2.

Blood pressure parameters and stages of hypertension.

BP Category Systolic mm Hg (upper #) Diastolic mm Hg (lower #)
Normal less than 120 and less than 80
Prehypertension 120 – 139 or 80 – 89
High BP (Hypertension) Stage 1 140 – 159 or 90 – 99
High BP (Hypertension) Stage 2 160 or higher or 100 or higher
Hypertensive Crisis (Emergency care needed) Higher than 180 or Higher than 110

Hypertension is amenable with a low-salt diet and antihypertensive agents (e.g., blockers of various steps in the AGT derived peptides, receptors, calcium channel, beta-adrenergic receptor, diuretics, and aldosterone synthase inhibitor). High BP for an extended period can cause cardiac dysfunction, kidney damage, and damage to other vital body organs [125]. Hypertension is classified as primary (essential) and secondary hypertension. The causes or mechanisms of primary hypertension are not well defined. It is believed that primary hypertension involves interaction between genetic loci and environmental factors. Primary hypertension is responsible for affecting more than 90% of hypertensive patients, and therefore is the most common type of hypertension; the prevalence of which increases with age [30], [140]. On the other hand, secondary hypertension is mainly due to known causes or genetic diseases [34], [90]. For example, the secondary hypertension may be due to malfunction of kidneys, lungs, arteries, endocrine system, or the heart. When lungs are involved, it is referred to as pulmonary hypertension [183]. Hepatotoxicants cause liver injury that may lead to portal hypertension from the repair of tissues around the portal vein causing constriction and increasing resistance to blood flow into the liver consequently increasing the BP. An increase in BP during pregnancy (or preeclampsia) is defined as pregnancy-induced hypertension leading to proteinuria at later stages of pregnancy [19]. Simplistically, cause(s) of secondary hypertensions (e.g., Liddle’s syndrome, glucocorticoid remediable hyper-aldosteronism, and G-protein-beta subunit mutant) is known and treatment is easier than primary hypertension, the causes of which are mostly not well defined [161]. One of the concerns of the treatment of hypertension is making it a resistant hypertension refractory to treatments [1].

American College of Cardiology and American Heart Association in November 2017 replaced the Joint National Committee hypertension guidelines [106], [88]. Similarly, the European Society of Cardiology and the European Society of Hypertension brought their recommendation in June 2018 for diagnosing, and management of hypertension [206]. The European guidelines remained the same while American guidelines changed. Previously, both Americans and Europeans defined hypertension when BP readings were ≥ 140/90. Earlier American guideline defined PB > 120/80 mmHg but less than 140/90 mmHg as prehypertension which now has become the first stage of hypertension as per the new guidelines. Now, as per American guidelines, BP ≥ 130/80 mmHg is a hypertension stage 1. This new guideline certainly increased the number of patients who required treatments. The European guidelines continue to define hypertension at BP > 140/90 mmHg. The systolic and diastolic blood pressure readings of the American and the European guidelines are presented in the Table 3.

Table 3.

Classification of Blood Pressure [44].

Category
SBP

DBP
American College of Cardiology/American Heart Association
 Normal < 120 and < 80
 Elevated 120–129 and < 80
 Stage-1 hypertension 130–139 or 80–89
 Stage-2 hypertension ≥ 140 or ≥ 90
European Society of Cardiology and European Society of Hypertension
 Category SBP DBP
 Optimal < 120 and < 80
 Normal 120–129 and/or 80–84
 High-normal 130–139 and/or 85–89
 Stage-1 Hypertension 140–159 and/or 90–99
 Stage-2 hypertension 160–179 and/or 100–109
 Stage-3 hypertension ≥ 180 And/or ≥ 110
 Isolated systolic hypertension ≥ 140 and < 90

SBP, systolic PB; DBP, diastolic BP.

Hypertension is a complex disease that causes many morbidities and mortalities worldwide. Hypertension is considered the major risk factor for CVD. Statistically, high BP affects almost one-third of adults worldwide and contributes to 13.5 million deaths annually [135]. Even with recent advances in science and understanding the pathophysiology of diseases and treatments, the prevalence of hypertension is still on the rise in certain parts of the world [81], [114], [135]. Ethnicity plays an important role in the development and maintenance of high BP which is the reason that one ethnic population respond better to one specific treatment whereas the other ethnic group respond better to another class of antihypertensive drugs [70], [107]. As an example, African Americans always respond better to diuretics and calcium channel blockers (CCB) than Caucasian and selected Asians who respond better to beta-blockers (BB) and angiotensin-converting enzyme I inhibitors (ACEI) [70], [71].

2. Role of aging in hypertension (elderly hypertension)

Studies on monitoring blood pressure in an urban population of developed countries demonstrate a rise in systolic blood pressure and pulse pressure with increasing age [181]. In developed countries, 35–50% of individuals over age 65 are thought to be hypertensive [181]. With increasing age, there is a generalized reduction in organ function [21]. Among older individuals, increased inflammation, arteriolar stiffening, and increased peripheral resistance are common [181]. Inflammation and oxidative stress contribute to endothelial dysfunction, the cells which line the internal walls of circulating blood vessels [20], [147], [158]. The decrease in elasticity of connective tissues increased in oxidative stress (superoxide and hydrogen peroxide) due to increased inflammation with decreased levels of antioxidants in endothelial cells which incapacitates the nitric oxide-mediated smooth muscle relaxation and vascular dysfunction are assumed to be the cause of elderly hypertension [21]. Moreover, decreased sensitivity to beta-2-adrenergic receptors may also be a contributing factor towards the reduced relaxation of smooth muscles [53]. Thus, endothelial dysfunction remains one of the contributing factors in the development of elderly hypertension.

Environmental chemicals and drugs are metabolized by CYP P450 based enzymatic system. It has been difficult to interpret the effects of aging on the levels and activities of P450 isozymes [204]. CYPs produce reactive oxygen species (ROS), which are removed by antioxidants [65], [198]. In addition to CYP450-mediated ROS production, many other biomolecules such as Ang II, endothelin-1, and urotensin II also produce ROS in endothelial cells. Decreasing levels of antioxidants among the elderly and increased ROS can cause vasoconstriction and exacerbate elderly hypertension [171].

2.1. Pulmonary hypertension

Pulmonary hypertension (PH) develops when resistance to blood flow in the lungs is increased. PH is defined when mean pulmonary artery pressure (mPAP) is ≥ 25 mm Hg with pulmonary wedge pressure of ≤ 15 mm Hg [79]. Due to increased resistance of blood flow to blood vessels by narrowing or blockage, increases the resistance to blood flow into lung leading to a rise in the BP in the arteries of lung. To pump enough oxygenated blood to meet the demand, heart puts extra effort which may lead to heart muscle failure. The diagnosis of PH cannot be performed by regular arm-cuff cut measurement. The PAH develops slowly and when developed, symptoms include dyspnea during exercise initially but later even at rest, fatigue, syncope, swelling in the ankles, legs and ascites, and palpitation; cyanosis may also be present in the patients.

Pulmonary arterial hypertension can be differentiated from pulmonary venous hypertension which occurs due to left side of heart disease. The PH due to diseased left-side of the heart can occur from defects in the mitral or aortic valves. It can also occur due to the failure of the left ventricle. The PH due to lung disease can occur when individuals are suffering from chronic obstructive pulmonary disease (COPD), fibrosis of the lung air sacs, obstructive sleep apnea. Individuals with long term exposure with high altitude climbing are also at a risk of pulmonary hypertension (https://www.mayoclinic.org/diseases-conditions/pulmonary-hypertension/symptoms-causes/syc-20350697). A thorough detail on PH definition and its causes is available online for readers to access by simple Google search.

In addition to reasons for PH described above, drugs and exposure with environmental toxicants also cause PH. Use of drugs such as anorexigenics, aminorex fumarate (e.g. amphetamine for suppressing appetite), and fenfluramine can cause increased incidence of PAH and PH [129], [176]. A whole host of other agents, like interferons used to treat viral hepatitis, tyrosine kinase inhibitors to treat various malignancies (e.g., dasatinib, mitomycin C) have also been reported to cause PH [141].

Tobacco smoke inhalation is a risk factor for pulmonary arterial hypertension [170], [96]. Recently, chronic inhalation of the component of tobacco, i.e., nicotine, has been found to cause PH and systemic hypertension (https://www.sciencedaily.com/releases/2020/05/200501125832.htm; [138]. Occupational exposure to trichloroethylene also causes PH [25]. The biochemical mechanisms of the development of PH from these agents in humans are not known; however, shown to be significantly different in humans and animals and therefore, animal models have far less application in studying the pulmonary hypertension by environmental chemicals and drugs [120].

3. Involvement of genes in hypertension: the monogenic hypertension

Liddle's syndrome (LS) is a genetic hypertensive disorder from the malfunction of Na+/K+ ATPase (SCNN1B and SCNN1G genes) due to gain of function mutation in SCNN1B and SCNN1G genes. SCNN1B and SCNN1G encode the beta and gamma subunit of Na+/K+ ATPase; often referred to as epithelial sodium channel [104]. It involves abnormal kidney function with loss of potassium and excess reabsorption of sodium from the renal tubule. In 1963, a new clinical syndrome that looked like primary aldosteronism (pseudoaldosteronism, subsequently named Liddle syndrome) was reported by Liddle and Coppage [113], which affected a 16-year-old Caucasian girl who presented severe resistant hypertension with low renin, metabolic alkalosis, and severe hypokalemia [17], [143]. Biochemical analyses to characterize decreased urinary sodium excretion rate with the absence of effects on aldosterone secretion after low sodium intake led to establish LS [17]. When comparing LS patients with Addison's disease patients (a disorder of adrenal glands not producing enough hormones), it was found that patients with LS have lower urinary sodium indicative of higher renal reabsorption of sodium due to a mechanism independent of mineralocorticoids activity [160]. Thus, LS is a genetic dominant form of low renin arterial hypertension that is caused by a mutation in the SCN genes encoding the non-voltage-gated sodium channel (SCNN1A, SCNN1B, and SCNN1G). The mutation results in an epithelial sodium channel (ENaC) to increase sodium and water reabsorption leading to increased blood volume and hypertension. The genetic disorder is characterized by not only the high BP but also the low plasma level of renin activity, metabolic alkalosis, hypokalemia, and normal to low levels of aldosterone. As a result, it is called pseudoaldosteronism/pseudohyperaldosteronism. In LS, kidney function is characterized by abnormally high sodium reabsorption with potassium loss due to the high activity of ENaC. So, treatment may be initiated with potassium sparing diuretics like amiloride after the confirmation of LS [187], [197].

Glucocorticoid remediable aldosteronism (GRA) is a rare familial autosomal dominant form of primary aldosteronism (PA, also referred to as Familial Hyperaldosteronism (FH) Type I), which comes with increased aldosterone secretion under the feedback control of adrenocorticotropic hormone (ACTH). GRA has a unique clinical response of aldosterone production and hypertension to the administration of glucocorticoids. GRA was first described by Sutherland and colleagues in 1996 in a father and son [173]. It is also characterized by being low renin hypertension with a high aldosterone/renin ratio [92]. GRA is caused by unequal crossing over of the genes encoding steroid 11 β-hydroxylase (CYP11B1) and aldosterone synthase (CYP11B2), which results in chimeric gene CYP11B1/CYP11B2 that has aldosterone synthase activity with CYP11B1 promoter under the regulation of ACTH rather than angiotensin II [109], [115], [145]. GRA patients are represented by mild hypokalemia, low plasma renin, and metabolic alkalosis [40]. As the name suggests, GRA is treatable by glucocorticoids, which inhibit ACTH production that is the activator of aldosterone production [73]. There are other forms of FH, i.e., FH type-II, type-III, and Type-IV, which are very rare and occur due to the gain of functions in channel proteins [110].

The FH type-II occurs due to adenomas or adrenal hyperplasia which is unresponsive to glucocorticoids. Blunting the aldosterone action by unilateral adrenalectomy or mineralocorticoid receptor antagonists are the recommended treatments [110]. The mutation in the chloride channel (ClC-2) is of the gain of function causing increased chloride efflux, resulting in the increased CYP11B2 expression and aldosterone production [110]. In type-III FH, a gain of function mutation in KCNJ5 causes increased calcium entry into adrenal glomerulosa cells due to membrane depolarization causing increased CYP11B2 expression and subsequent aldosterone synthesis. Gene KCNJ5 encodes the Kir 3.4 K+ channel which is an inward rectifier potassium channel [110]. The type-IV FH occurs due to gain of function in the CACNA1H gene [110]. In another form of hypertension referred to as Gordon’s hypertension syndrome, the mutation is observed in WNK1 (with no K (lysine) protein kinase-1) and WNK4 where sodium-chloride cotransporter activity in the kidney is increased [52].

Non-chimeric CYP11B2 has also been shown to cause hypertension and cardiac fibrosis via Angiotensin II type I receptor (AT1R) increasing absorption of Na+ from the kidney, thus increasing blood volume and hypertension. Absorptions of Na+ occurs via aldosterone, which is synthesized by aldosterone synthase (Cytochrome P450 [CYP] 11B2) which is released from the adrenal cortex. Activation of CYP11B2 occurs via K+ resulting in Ca++ release and activation of T and L-type calcium channel and increased CYP11B2 expression in the adrenal cortex [137]. The increased expression of CYPIIB2 is through the upregulation of cyclic-AMP response element (CRE) mediated protein kinase; however, transcriptional regulation of CYP11B2 is not well defined. The CRE and ATF-1 have been shown to bind CRE-element after phosphorylation [137]. Recently, ubiquitin-proteasome inhibitor, bortezomib, is found to suppress the expression of CYP11B2 raising the prospect of the development of antihypertensive drugs [85]. To develop H295R cells as an in vitro cell-based model for screening chemicals for endocrine disruption properties, octyl methoxycinnamate, and acetyl tributyl citrate were found to increase the expression of CYP11B2 [182]. In a study evaluating the endocrine disruption activity of persistent organic pollutants using H295R as in vitro cell-based model, TCCD, PCB, PFOS, HBCD, and BDE-47 were found to increase the secretion of steroid hormone indicating steroid synthesis interference by these compounds. The study did not evaluate effects on the CYP11B2 expression levels [193]. Th use of H295R to study the gene regulation and transcriptional activation of CYP11B2 after xenobiotics exposure and establish a linkage between the environmental toxicants and the regulation of CYP11B2 is insufficient, and thus, the association to hypertension.

4. Alcohol and hypertension

A divergent view on the beneficial and harmful effects of the use of ethanol exists among scientists and researchers. Current and past research suggest that moderate alcohol intake is beneficial to the cardiovascular system lowering BP while excessive use is harmful causing an elevation in BP. Low to moderate alcohol use has been linked to decreased incidence of coronary heart disease and an increase in longevity [48], [94]. However, alcohol is also known to increase BP, especially with the chronic intake. Continued consumption of more than 2 servings of ethanol (30–50 g) per day results in a dose-dependent rise in BP [56]. Heavy alcohol usage (>2 drinks/day) has also been associated with hypertension [56], [69], [101], [188]. The biochemical mechanism of alcohol-mediated hypertension is not unequivocally defined. The alcohol-mediated increase in BP involves several mechanisms including impairment of baroreceptors, increase in sympathetic activity, stimulation of the endothelium to release endothelin, inhibition of endothelium-dependent nitric-oxide production, and stimulation of the renin-angiotensin-aldosterone system (RAAS) [152]. Lowering of alcohol-induced hypertension in rats has been obtained by dexamethasone treatment blocking the activation of the sympathetic nervous system responsible for releasing corticotrophin-releasing hormone [152]. In another study, ethanol exposure increased circulating levels of vasopressin in rats resulting in hypertension [153]. Antihypertensive drugs, e.g., ACEI (zofenoprilat), beta-blocker (carvedilol) and calcium channel blocker (lacidipine) have shown to protect cultured human endothelial cells against alcohol-induced oxidative-stress and endothelial dysfunction [175]. Blunting of oxidative-stress and endothelial dysfunction by zofenoprilat (ACEI) indicates involvement of RAAS. Inhibition of RAAS in experimental animals and clinical studies has proven to effectively lower BP in hypertensive animal models and human subjects [8]. Like alcohol that is responsible for hepatosteatosis and steatohepatitis, OCI and PCBs exposure can produce such effects pointing to (i) a common mechanism of hepatotoxicity and regulation of genes, and (ii) possibly an APR activation after injury to the liver resulting in the production and secretion of proteins from liver into blood. There are two types of APR, (i) APR I that is induced by TNF-α and IL-1β which activate NF-κB, and (ii) APR II that is activated by glucocorticoids and IL-6 via STAT3 activation. Human AGT is an APR II protein, which is triggered by glucocorticoids and IL-6 / STAT3 activation [131]. Metabolism of alcohol in the liver produces oxidative stress from increased ROS levels [51]. In addition, xenobiotics’ metabolism (e.g., OCI and PCBs) in the liver also produces oxidative stress [119]. Liver is the source of many secretory proteins like clotting factors and AGT. The ROS and metabolic products (acetaldehyde in case of ethanol) mediated effects on transcription factors in the liver after xenobiotic exposure likely target the synthesis and secretion of liver-specific genes including AGT, the precursor of vasopressor peptide, angiotensin II. The angiotensin II is released sequentially by proteolytic cleavage of AGT in the kidney by renin and angiotensin-I converting enzyme (ACE) in the lung while it serves as a precursor to other peptide hormones (Fig. 1).

Fig. 1.

Fig. 1

Peptides of Angiotensinogen.

The circulating plasma concentrations of AGT is closed to saturating concentration of aspartyl protease, renin [66]. Therefore, alterations in the circulating AGT levels can cause corresponding changes in the blood angiotensin II levels. Thereby, a rise in blood AGT level can lead to a parallel increase in the formation of angiotensin II causing hypertension [49].

Higher blood AGT levels have been demonstrated more frequently in hypertensive subjects and their children than healthy individuals with normal BP [202]. Expression of the renin-AGT in body organs such as kidneys, heart, placenta, brain, and adrenals are examples of organ-specific BP regulation [26]. The overexpression of human AGT (hAGT) in a humanized mice model was shown to increase BP while the effect was not observed in the AGT gene-knockout mice [99], [185]. Administration of antisense RNA of AGT causes a profound reduction in BP in hypertensive rats [184]. These studies demonstrate that changes in blood AGT levels can measurably alter BP. Similar studies duplicating the ACE gene in mice led to an increase in blood ACE level but no change in BP [105]. The inhibition of RAAS in hypertensive experimental animals and humans has shown to lower BP. Individuals with insulin-dependent diabetes secrete increased levels of AGT in urine from increased RAAS activity in the kidney that is a biomarker of poor glycemic control [133]. Regulation of synthesis and secretion of AGT-related oxidative stress in individuals with diabetes is considered a key step towards reducing AGT synthesis, better glycemic control, and reducing hypertension. The link between environmental toxicants and AGT synthesis and secretion from the liver has not been established. However, results of studies conducted in our laboratory indicate that alcohol can enhance IL-6 mediated AGT secretion from human hepatocytes (unpublished data).

Human AGT possesses several single nucleotide polymorphisms (SNP) in the coding region. Polymorphism resulting from the conversion of methionine into threonine (M235T) causes higher blood AGT levels and hypertension [91]. Similarly, the T174M allele in hAGT is also associated with hypertension [111]. It is known that hAGT SNPs, especially − 6A (−6A/G), − 217A (−271 A/G), and 235 M are associated with hypertension [150]. Due to the association of hAGT with primary hypertension, the role of SNPs in hAGT proximal promoter activity was determined in transgenic mouse models; however, these studies did not provide an unequivocal answer to the role of several proximal promoter SNPs. In one of the studies, Grobe et al. [68] observed no difference in the level of circulating hAGT with either − 6 A or − 6 G nucleotide in context with − 6A/235 T or − 6 G/235 M variant. In another study, Jain et al. [86] found that − 6 A and − 6 G SNP caused an increase in the blood hAGT; − 6 G SNP was less effective than − 6A SNP. Previously, [87] investigated the role of − 217A and − 217 G SNP which is the site of glucocorticoid receptor binding and observed that − 217A increases AGT levels in blood more than − 217 G does. Currently, data on the regulation of hAGT and hepatotoxicants are apparently lacking. However, our laboratory has demonstrated that treatment of human hepatocytes with alcohol results in increased secretion of AGT [7].

5. Role of environmental toxicants in hypertension

We are continuously exposed to environmental pollutants and scientists have shown a strong relationship between exposure to environmental pollutants such as persistent organic pollutants (POP), PM, and chemicals from diesel exhaust and the risk of developing several diseases. Evidence suggests that humans living close to hazardous waste sites and exposure to hazardous waste are more susceptible to developing high BP [61], [76], [172]. However, the biochemical mechanism of these toxicants affecting BP is not completely understood. The major environmental toxicants present in most of these studies were organic pollutants, such as OCI, and PCB. In a cross-sectional epidemiological study conducted in Spain, the POP was found to induce opposite actions on the BP of individuals. The organochlorine cyclodiene insecticide, Aldrin, was negatively associated with hypertension, while dichlorodiphenyltrichloroethane (DDT) metabolite, dichlorodiphenyldichloroethane (DDE), is well known to elevate the BP [55]. These opposing effects were suggested to be from the difference in chemical structures and concentrations. Most POPs play a role in the endocrine function altering hormonal homeostasis that may result in elevating BP [61], [76]. For example, postmenopausal women who take supplement estrogen to control the effects of menopause are at a higher risk of developing hypertension from the hormone, estradiol [179], [180]. Similarly, some of the POPs such as DDE, a known xenoestrogen can modify estrogen homeostasis and impair the regulation of BP [46], [144]. Additionally, POPs can generate free radicals that can lead to triggering proinflammatory cytokine signaling pathways and associated inflammatory diseases including hypertension [146]. In another study, PCBs exposure was associated with hypertension in healthy individuals [29]. The increase in both systolic and diastolic BP was observed, even though the exact reason(s) of PCBs-mediated elevation in BP could not be determined [180]. It has been demonstrated, however, that the dysfunction in the endothelium contributed to the increase in BP [61], [76], [139]. The POPs are not the only environmental pollutants causing hypertension but other air pollutants like those from the exhaust of diesel fuel can also be as bad. A recent study indicated that increased exposure to air pollutants make human more vulnerable to hypertension. When diesel fuel burns, the resulting exhaust, made up of soot consisting of very small particles, and a variety of other harmful chemicals (i.e., carbon dioxide, carbon monoxide, nitrogen dioxide, and nitric oxide) can deposit in the lungs [199]. These air pollutants and the byproducts of burning some organic materials, like tetrachlorodibenzo-p-dioxin (TCDD), are known to induce high BP as they may be an agonist of the aryl hydrocarbon receptor (AhR) [77], [212]. The AhR is a heterodimer of HIF-1α, transcriptionally regulates phase I/II genes in the endothelium [103]. These air pollutants and smoking are further risk factors for CVD. The AhR plays a major role in regulating the level of Angiotensin II from AGT, endothelial nitric oxide synthase (eNOS), and endothelin-1 [212], [214]. In addition to activating AhR, the pollutants may activate the expression of genes involved in the inflammatory response and endothelial dysfunction, thus contributing even further to the development of atherosclerosis, hypertension, and CVD [214], [216].

Other studies have demonstrated a correlation between particulate air pollutants, especially diesel exhaust, and hypertension [59], [166]. Studies conducted on human subjects, who inhaled PM present in the urban and industrial environment, in which the results revealed a correlation between the exposure to PM and the increased BP and CVD [23], [166]. In another study, macrophage U937 cells were used to determine urban dust particles (UDP) and diesel exhaust particulates (DEP) effects on proinflammatory cytokines induction [199]. Both UPD and DEP along with their organic extracts (OE) and stripped particles (sUDP and sDEP) caused induction of proinflammatory cytokines and CRP in macrophage U937 cells [199]. An increase in IL-8, TNF-α, and cyclooxygenase-2 mRNA expressions was noticed after the exposure to OE-UDP, OE-DEP, UDP, and DEP. On the other hand, an increase in the production of CRP and IL-6 mRNA was noticed after the exposure to sUDP, sDEP, UDP and DEP [199]. This inflammatory response is known to play a major role in the pathological process of CVD [112]. A positive association between CRP and coronary artery diseases due to chronic inflammation is well known [9], [47]. Other harmful effects of proinflammatory cytokines include triggering acute vasoconstriction which may lead to hypertension and atherosclerosis [178].

A study conducted by Olea et al. [139] of the University of Chicago, investigating the link between environmental toxicants and the occurrence of elevated BP through a completely different mechanism. They studied different classes of environmental chemicals, i.e., industrial pollutants, phytochemicals, pesticides, waste products, and consumer products - chemicals with different structures, for the mode of action of inducing hypertension. Chemicals they studied could modulate endogenous hormonal signaling pathways (i.e., endocrine-disrupting chemicals [EDCs]). Both, epidemiological and animal data have shown the association between EDCs and metabolic disorders, particularly type 2 diabetes, and CVD. The effect of these EDCs on the cardiovascular system starts with an increase in the prevalence of atherosclerosis plaque and their ability to promote dysregulation of energy metabolism. Unfortunately, many humans and animal studies have failed to fully describe the molecular mechanism by which EDCs exert their effects and only provided some insights into the potential role of EDCs in pathogeneses of macrovascular diseases. High BP is one of the major risk factors that can promote atherosclerosis by enhancing stress and endothelial inflammation mediated by oxidative stress. The regulation of BP is by a host of local and systemic signaling molecules, some of which are modulated by the EDCs. For example, nicotine-exposed rats have shown elevated levels of angiotensin II leading to higher BP. In addition, some chemicals, e.g., organophosphorus pesticides upregulate the activity of adenylate cyclase in neonatal rats. Adenylate cyclase directly signals adrenergic G-protein coupling, which plays a major role in regulating BP through catecholamines [139].

6. Roles of heavy metals in hypertension

The mechanisms of action through which heavy metals, like arsenic (As), mercury (Hg), cadmium (Cd), and lead (Pb) instigate hypertension are still not completely understood; further investigations are needed to enhance the understanding of the relationship between them and CVD including hypertension.

6.1. Arsenic and hypertension

As, as one of the omnipresent metalloids, is found in nature and causes hypertension by several mechanisms including an increase in subclinical atherosclerosis, electrocardiographic abnormalities, oxidative stress, and inflammation [42], [210]. Many cross-sectional epidemiological studies on As exposure and subclinical atherosclerosis are available in the literature. Wang et al. [201] studied 199 adult males and 264 adult females, who were exposed to As, in the southwestern area of Taiwan. Their study examined the duration of consumption of artesian well water, average As concentration in the water consumed, and cumulative As exposure adjusted for factors like age, sex, cholesterol, body mass index (BMI) ratio, diabetes, and hypertension. They found a dose-response relationship between long-term exposure to As and carotid atherosclerosis. Later, Wang et al. [200] studied 280 men and 355 women in the same area of Taiwan. They found a dose-response relationship between the cumulative As exposure and carotid intima-media thickness (cIMT) and prevalence of carotid plaque after adjustment for age, sex, hypertension, diabetes, cholesterol, and triglyceride, BMI, smoking, and alcohol consumption [200]. In addition to Wang et al. [201], [200] studies, Hsieh et al. [83] concluded that the formation of atherosclerosis is increased in people with exposure to high levels of As in drinking well water (>50 μg/l) when they have As metabolic genes, including purine nucleoside phosphorylase (PNP), As (+3) methyltransferase (As3MT), glutathione S-transferase omega 1 (GSTO1), and omega 2 (GSTO2). In a study conducted in Bangladesh, 66 young adults, who consumed well water containing 0.5–439 μg/L As were analyzed. The results showed that the levels of As in their urine ranged 6–209 μg/L, and subjects with higher urinary As levels had higher cIMT (> 0.75 mm) [210]. Another cross-sectional study in Bangladesh of 959 subjects found an effect of long-term As exposure on cIMT which was assessed ~7 years later with a dose-response relationship of urinary As and urinary monomethylarsonic acid with cIMT [36], [38], [39]. Osorio-Yanez et al. [142] found that the concentrations of total speciated As (tAs) were positively associated with cIMT increase. The estimated cIMT diameter was higher in groups with urinary As of 35- to 70-ng/mL and > 70-ng/mL (0.035 mm and 0.058 mm per 1-ng/mL increase in urinary tAs, respectively).

The As-exposed and non-exposed individuals of Bangladesh were assessed for their cardiac status by Ahmad et al. [3]. The subjects of this study were divided into As-exposed individuals with arsenicosis (arsenicosis group), As-exposed persons without arsenicosis (non-arsenicosis group), and individuals not exposed to As (non-exposed group), each group was composed of 50 subjects. The Arsenicosis group had abnormal electrocardiograms findings (58% of total subjects) [3]. In another study conducted in Turkey on 40 men exposed to As with mean As levels in village water of 659 μg/L (ranged 422–1066 μg/L), the study found that exposed individuals had a slight QT prolongation and a higher prevalence of subtle repolarization abnormalities [215]. Mumford et al. [132] found that prolong exposure of As from well-water is associated with the prevalence of QT prolongation in men from Bangladesh and hypothesized that QT prolongation is due to the functional alterations in cardiac cell surface channels in a dose-dependent manner. Mordukhovich et al. [130] conducted a study to assess the relationship between As in toenail and QT, heart rate-corrected QTc, and the effect of modification by calcium channel blocker used by elderly men. They observed a positive association between As in toenail and QT duration; QT and QTc duration was increased by 3.8-millisecond and 2.5-millisecond, respectively in subjects exposed to As [130]. Chen et al. [39] found a prolonged QT in As exposure group assessed on average 6 years after exposure, the association appeared to be present in women but not in men. A positive association between long-term well-water As exposure and plasma levels of soluble ICAM-1 and soluble VCAM-1 (biomarkers to predict future CVD), biomarkers of endothelial dysfunction, and vascular inflammation is observed in an As-exposed population of Araihazar, Bangladesh [15], [37], [155]. Wu et al. [209] found a positive association between As exposure and plasma levels of soluble VCAM-1. They also found an interaction between As exposure and higher BMI in addition to increased plasminogen activator inhibitor-1 (PAI-1) and VCAM levels [209]. In addition, increased concentrations of PAI-1 were associated with acute myocardial infarction in individuals with a high prevalence of coronary heart disease [189]. Karim et al. [95] found significantly higher levels of oxidized LDL (ox-LDL), CRP, ICAM-1, and VCAM-1 in As-endemic subjects than those in nonendemic subjects. They also showed dose-response relationships with As exposure and HDL, ox-LDL, and CRP [95]. A study of 50 subjects in West Bengal, India, who were exposed to As through well water and 41 subjects not exposed to As found that the exposed group had higher catalase (CAT) and myeloperoxidase (MPO) with a higher incidence of chromosomal aberrations (CA) [10]. MPO is linked to atherosclerosis and CVD [169]. Another study found an increase in catalase activity in patients suffering from oxidative stress, CVD, diabetes, tumor, inflammation, dermatological diseases, anemia, and Wilson’s disease [4]. Osorio-Yanez et al. [142] found that tAs was positively associated with plasma asymmetric dimethylarginine (ADMA) levels and cIMT, an indicator of subclinical atherosclerotic. Thus, all observed associations, between As exposure and atherosclerosis leading to CVD, are considered major factors [95]. In a study in Southwest Taiwan of 533 subjects, Tseng et al. [192] suggested that the chronic As exposure, i.e., duration of drinking well water, could positively associate with the serum levels of TC and LDL, which are the risk factors for atherosclerosis and CVDs such as hypertension.

6.2. Mercury and hypertension

The inhalation exposure of metallic mercury causes elevation of heart rate and BP (ATSDR, 1999). Occupational mercury exposure also causes an increase in blood pressure and heart rate. However, a low level of chronic mercury exposure (0–0.27 mg/m3 in one and 0.075 mg/m3 in another study) for 0.5–7 years did not increase the blood pressure or caused abnormal electrocardiograph [2]. Furthermore, estimated exposure with 0.03 mg/m3 of mercury vapors for 5 years caused a reduction in cardiovascular reflexes and increased palpitations [2]. Workers in the thermometer plant were found with hypertension [2]. An increase in the systolic and diastolic pressure has been observed among individuals carrying dental amalgam of mercury, however, the increases in systolic and diastolic pressure were not markedly different from the normal or non-amalgam group [2].

The mechanisms by which Hg causes hypertension are still not completely understood. Hg induces hydrogen peroxide production and mitochondrial dysfunction at the ubiquinone-cytochrome-b site of the mitochondrial respiratory chain [116]. It causes mitochondrial reduced glutathione (GSH) content depletion by more than 50% and increases thiobarbiturate reactive substances (TBARS), an indication of increased mitochondrial lipid peroxidation, by 68% [117]. Following the addition of Hg to mitochondria isolated from kidneys of untreated rats, an increased depolarization of the inner mitochondrial membrane was observed [118], [154]. Oxidation of pyridine nucleotides (NADPH) was also observed in mitochondria incubated with Hg along with significantly increased Ca2+, H2O2, and TBARS due to its effect on the ubiquinone-cytochrome b site of the mitochondrial electron transport chain [117]. These events cause increased oxidative stress and decreased oxidant defense [117]. Increased risk of myocardial infarction and coronary issues resulting from mercury exposure from dietary fish has been recorded in Finnish men [165]. Hg promotes lipid peroxidation by three main sources: Fenton reaction, affinity for sulfhydryl groups, and selenium deficiency [57]. It acts as a catalyst in Fenton-type reactions resulting in the formation of free radicals [57]. In another study, Hg(II) ions in micromolar concentrations increased the production of superoxide anions in human neutrophils [89]. In a relatively newer study, mercuric ions (1–6 μmol/L) caused a concentration-dependent increase (up to 5-fold) in mitochondrial H2O2 production. Hg has been found to enhance iron-stimulated lipid peroxidation in vitro [72]. Mercury’s high affinity for sulfhydryl groups is responsible for most of the antioxidant capacity of plasma such as glutathione, n-acetyl cysteine, and alpha-lipoic acid, which could reduce both membrane and plasma antioxidant defense [58]. Insoluble complexes of Hg with selenium (Hg selenides) reduce selenium availability, a necessary cofactor for glutathione peroxidase (GPx), which is an important scavenger of H2O2 and lipid peroxides. Depletion of selenium increases the risk of CVD and cerebrovascular accidents (CVA) [13], [177]. Hg increases hypertension risk by increasing carotid atherosclerosis. A population-based 4-year prospective study in men in eastern Finland found an increase in mean carotid IMT which was directly related to hair Hg content (P = 0.0007) [164]. An increase in hair Hg by 1 µg equaled a 0.008-mm increase in carotid IMT; a 7.3% increase over the mean value. In addition, Hg hair content was proportional to BP, fibrinogen levels, BMI, and low HDL cholesterol [164]. Hg can interfere with the normal catabolic processing of catecholamines via the cytosolic enzyme catecholamine-O-methyltransferase by inactivating coenzyme S-adenosylmethionine (SAM) which donates the methyl group to catecholamine-O-methyltransferase. As a result, norepinephrine, dopamine, and epinephrine accumulate in blood elevating BP [78]. This catecholamine excess is responsible for the pheochromocytoma-like syndrome [124]. Studies have shown that Hg concentrates in the renal tubules and glomerulus resulting in proteinuria, fibrosis, chronic renal dysfunction, and renal insufficiency [16], [102], [205]. Finally, Hg stimulates proliferation of vascular smooth muscle cells and inactivates paraoxonase, an extracellular antioxidative enzyme related to HDL, and increases the risk of coronary heart disease (CHD), and myocardial infarction (MI) [64], [163].

6.3. Lead and hypertension

The mechanisms of action by which Pb causes hypertension are still not completely understood. Several mechanisms have been suggested for Pb-induced hypertension including oxidative stress, impaired nitric oxide system, inflammation, dysregulation of vasoactive hormones, and alteration of cellular Ca2+ transport and intracellular Ca2+ regulation. Pb can facilitate the production of ROS (e.g., O2 and H2O2) and can produce oxidative stress by acting as the catalyst in Fenton- and Haber-Weiss-type reactions [98]. Khalil-Manesh et al. [98] found dimercaptosuccinic acid (DMSA)-mediated Pb chelation increased cyclic guanosine monophosphate (cGMP) and rapid reduction of BP in rats with Pb-induced hypertension. They proposed that Pb exposure raises arterial BP by promoting ROS production and ROS-mediated inactivation of endothelium-derived relaxing factors. They also proposed that amelioration of high BP is due to attenuation of Pb-induced oxidative stress caused by the strong antioxidant activity of DMSA [97]. Gonick et al. [63] found significant accumulation of the lipid peroxidation product; malondialdehyde and inducible nitric oxide synthase (NOS), in the kidneys of Pb-treated rats. These observations support the presence of oxidative stress in the kidneys of Pb-exposed animals increasing the risk of hypertension [63]. Another study by Ding et al. [45] found a much greater reduction in arterial pressure in Pb-exposed rats than in either control or DMSA-treated Pb-exposed rats after the infusion of L-arginine. The study suggested the reduced nitric oxide production may be due to the oxidative stress. Administration of DMSA for 2 weeks lowered BP and reduced blood Pb concentration in Pb-induced hypertensive rats [45]. Another mechanism of hypertension involves Pb-mediated protein kinase C (PKC) isoforms activation, which is involved in many cellular functions including blood flow, vascular contraction, and cell growth [84]. Hwang et al. [84] assessed blood Pb levels, neurobehavioral effects, and PKC activity in 212 current Pb workers in the Republic of Korea and found elevated erythrocyte PKC activity among them. Increased PKC activity was also found in the microvessels of the rat brain after exposure to Pb at micromolar concentrations [122]. In another study, Watts et al. [203] found PKC in intact and endothelium-denuded rabbit mesenteric artery preparations following exposure of rabbits to Pb acetate at 10−10 to 10−3 M concentrations. They found that Pb-induced vasoconstriction was augmented by a PKC agonist, reduced by a PKC inhibitor, and attenuated by the Ca2+ channel blocker verapamil. In another proposed mechanism, Pb promoted inflammation, fibrosis, and apoptosis by Nuclear Factor-kappa B (NF-κB), which is a transcription factor for numerous proinflammatory cytokines, chemokines, and adhesion molecules. Two studies observed that NF-κB activation is linked to renal tubulointerstitial inflammation and the development of hypertension [157], [195]. Ramesh et al. [151] found activations of NF-κB in the brain of rats exposed to low levels of Pb (50 ppm in drinking water) for 90 days. Recently, Rodriguez-Iturbe et al. [157] reported NF-κB activation, tubulointerstitial accumulation of T cells, macrophages, and angiotensin II-expressing cells, increased number of apoptotic cells, and heavy tyrosine nitration in kidneys of rats with Pb-induced hypertension. Pb exposure also elevates plasma catecholamines and cardiac contractility [32]. Chang et al. [32] found high plasma norepinephrine but normal plasma dopamine and epinephrine levels, pointing to heightened sympathetic nervous system activity. In another study, Chang et al. [33] found increased arterial pressure and plasma norepinephrine without changing plasma epinephrine concentration in Pb-induced hypertensive rats. Another mechanism may be by increased production of endothelins, which are powerful vasoconstrictor peptides, primarily synthesized and secreted by endothelial cells. For instance, Khalil-Manesh et al. [98] found significantly increased arterial pressure, a marked increase in plasma endothelin-3, and decreased endothelin derived relaxing factor (cGMP) levels in rats exposed to the low levels of Pb (0.01% via drinking water) which increased nephropathy without a rise in arterial pressure or plasma endothelin. In another in vitro study, Molero et al. [128] suggested that Pb can increase endothelin activity in the vascular tissue; incubation of isolated rat arteries in the Pb-containing medium led to decreased soluble guanylate cyclase (sGC) and cGMP production. They also found that coincubation with an endothelin type A receptor antagonist partially reverses Pb-induced downregulation of sGC and cGMP production. Another possible mechanism is due to an increase in circulating renin level that eventually elevates BP [194]. In a meta-analysis of the studies published between the late 1970 s and 1990 s, Vander [194] concluded that Pb exposure in young rats for several weeks is sufficient to achieve blood Pb levels in the range of 30–40 μg/dL and elevate plasma and kidney renin activity. Another study by Carmignani et al. [28] found that Pb exposure (60 ppm Pb acetate in water) to young rats for 10 months increased plasma angiotensin-converting enzyme activity as well as plasma kininase-I, kininase-II, and kallikrein activities. A subsequent study by Sharifi et al. [174] reported a steady rise in ACE activity in the plasma, aorta, kidney, and heart in young adult rats exposed to 100 ppm Pb acetate for 2–8 weeks. The initial rise in plasma and tissue ACE activity was followed by a decline to subnormal values by 8 wk., coinciding with a marked elevation of arterial pressure. Another way BP can be increased is by lowering the production of vasodilatory and increasing the production of vasoconstrictive prostaglandins in humans [27]. Cardenas et al. [27] studied a group of Pb workers with elevated blood Pb concentration and compared with control workers, they found increased urinary excretion of the thromboxane metabolite TXB2 and reduced excretion of the vasodilatory prostaglandin metabolite; 6-keto-PGF1. Later in another study by Hotter et al. [82] confirmed the results in a separate group of Pb-exposed workers. However, Gonick et al. [62] failed to find a difference in urinary excretion of these metabolites in rats with Pb-induced hypertension. Another mechanism may involve reduced plasma atrial natriuretic peptide (ANP), which is synthesized and secreted by the cardiac myocytes in response to the distension of cardiac chambers. ANP is also considered a vasodilator and plays a role in regulation of arterial pressure by modulating systemic vascular resistance and blood volume [60]. Giridhar and Isom [60] concluded that Pb-exposed animals exhibited fluid retention, which was paradoxically accompanied by a dose-dependent decline in plasma ANP level.

6.4. Cadmium and Hypertension

Cadmium is a food chain toxicant with a greater rate of soil to plant transfer. Cadmium is also an environmentally persistent toxic metal, which is present in cigarette smoke and polluted air causing human exposure [167]. Cadmium occupational exposure is linked to hypertension; however, the data remains inconsistent [24]. Occupationally exposed individuals with Cd were found to have increased systolic and diastolic pressure [24]. In a study of the Korean National Health and Nutrition Examination Survey, Lee et al. [108] observed increased Cd blood level positively associated with increased blood pressure, indicating risk of hypertension. In a Strong Heart Study (SHS), Franceschini et al. [54] observed that Cd exposure resulted in increased blood pressure. The blood Cd levels but not urine levels are linked to a modest rise in blood pressure. This effect is even more prominent in non-smokers [186]. In this study, it was observed that the effects of Cd exposure to former smokers were intermediate and current smokers were null [186]. In a marked contrast, the 1999–2004 National Health and Nutrition Examination Survey (NHANES) of the USA did not find the link between either blood or urine Cd level with an increase in blood pressure [43]. To decipher Cd-mediated hypertension, Al-Naemi and Das [6] demonstrated abnormal endothelial function in animals as the cause of hypertension. While exposure leads to increased eNOS expression, yet Cd produced oxidative stress by stimulating NOX expression by reducing the bioavailability of nitric oxide (NO) [5]. Satarug and coworkers (2017) have summed up the mechanism of Cd exposure and hypertension. Cadmium, which is absorbed from the GI tract, is transported to the liver by portal vein where Cd binds with metallothionein. After secretion of metallothionein bound Cd in bile and blood it reaches to kidney especially the proximal tubular cells. The Cd-metallothionein complex is broken down in proximal tubular cells. The released Cd causes oxidative stress and inflammation. Thereby, the activity of Na+/K+-ATPase is decreased, in large part due to the oxidation of Na+/K+-ATPase and the changes in the production of 20-hydroxyeicosatetraenoic acid (2-HETE) in proximal tubule causing salt and water retention and hence plasma expansion. Such effects can impose pressure-natriuresis and persistent increased blood pressure and hypertension [168].

7. Role of particulate matter in hypertension

Particulate matter is a complex mixture of extremely small particles and liquid droplets. PM vary extensively in physical (diameter, morphology, surface area, and hygroscopicity) and chemical (organic, inorganic, metallic) characteristics. The diameter ranges from tens of micrometers (µm) to tens of nanometers (nm). Because of this large range, PM of health concern is typically categorized in three size fractions, i.e., ≤ 10 µm in diameter (PM10 or coarse PM), ≤ 2.5 µm in diameter (PM2.5 or fine PM), ≤ 0.1 µm (PM0.1 or ultrafine PM). Two main characteristics of airborne particles dictate the toxicity of exposure – size and chemical composition. Size dictates the fate of airborne particles entering the human respiratory system. Depending on their size, airborne particles can get filtered early on by the respiratory cilia or move past them and get deposited in various regions of the respiratory tract. Larger PM are deposited in the extra thoracic region of the respiratory tract, PM10 in the tracheobronchial region, PM2.5 in the pulmonary region, and PM0.1 in the alveolar region and may exchange with blood [207]. While the deposition fractions may vary as a function of the breathing patterns of a person (i.e., cilia in the head airways are bypassed during mouth breathing; deposition fractions may differ during resting, conversation, or exercise), generally, the smallest fractions reach deeper into the lungs.

Laboratory studies have suggested that exposure to PM2.5 can trigger a combination of pathophysiological responses inducing hypertension. The risk of hypertension associated with long-term exposure to PM2.5 is still sparse and unclear from studies conducted in North America and Europe; however, in epidemiological studies conducted in developing countries with typically higher ambient concentrations of PM2.5 have provided evidence of an association between PM2.5 and hypertension. Huang et al. [75] found an association between long-term exposure to PM2.5 and hypertension in a large-scale prospective study in China comprising of cohorts of 59,456 participants aged ≥ 18 years without hypertension and followed from 2004 to 2015 for ambient PM2.5 and hypertension. Similarly, Prabhakaran et al. [149] found a temporal association between high levels of PM2.5, higher systolic BP, and incident hypertension in Indian. A population-based cohort study (comprising of 35,303 participants) was conducted in Ontario, Canada by Chen et al. [35] to determine whether exposure to ambient PM2.5 is associated with incident hypertension. Results of the study supported an association between PM2.5 and the incidence of hypertension.

8. Conclusions

In today’s life, humans are exposed to various environmental pollutants, which might influence blood pressure in a meaningful way. Several pollutants such as heavy metals, diesel exhausts, and PM have been linked to increased blood pressure. A number of studies have been carried out reporting the linkage between the exposure to environmental pollutants and the increased blood pressure, while no substantial data provided unequivocally unraveling the possible mechanisms involved in such phenomena. However, increased incidence of hypertension worldwide points to the role of environmental pollutants as one of the factors and understanding the mechanisms by which pollutants affect blood pressure is required for meaningful interventions.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Handling Editor: Dr. Aristidis Tsatsakis

References

  • 1.Acelajado M.C., Hughes Z.H., Oparil S., Calhoun D.A. Treatment of resistant and refractory hypertension. Circ. Res. 2019;124:1061–1070. doi: 10.1161/CIRCRESAHA.118.312156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Agency for Toxic Substances and Disease Registry (ATSDR) 1999. Toxicological Profile for Mercury. U.S. Department of Health and Human Services, ATSDR, Division of Toxicology/Toxicology Information Branch, Altanta, GA, 〈https://www.atsdr.cdc.gov/toxprofiles/tp46.pdf〉.
  • 3.Ahmad S.A., Khatun F., Sayed M.H., Khan M.H., Aziz R., Hossain M.Z., Faruquee M.H. Electrocardiographic abnormalities among arsenic-exposed persons through groundwater in Bangladesh. J. Health Popul. Nutr. 2006;24:221–227. [PubMed] [Google Scholar]
  • 4.Al-Abrash A.S., Al-Quobaili F.A., Al-Akhras G.N. Catalase evaluation in different human diseases associated with oxidative stress. Saudi Med. J. 2000;21:826–830. [PubMed] [Google Scholar]
  • 5.Almenara C.C., Broseghini-Filho G.B., Vescovi M.V., Angeli J.K., Faria Tde O., Stefanon I., Vassallo D.V., Padilha A.S. Chronic cadmium treatment promotes oxidative stress and endothelial damage in isolated rat aorta. PLoS One. 2013;8 doi: 10.1371/journal.pone.0068418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Al-Naemi H.A., Das S.C. Cadmium-induced endothelial dysfunction mediated by asymmetric dimethylarginine. Environ. Sci. Pollut. Res. Int. 2020;27:16246–16253. doi: 10.1007/s11356-020-08116-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ansari R.A., Rizvi S.A.A., Clark M.A. US Society of Toxicology; San Antonio, TX: 2013. Role of Alcohol Dehydrogenase in Regulation of Angiotensinogen in Human Hepatocyte. [Google Scholar]
  • 8.Atlas S.A. The renin-angiotensin aldosterone system: pathophysiological role and pharmacologic inhibition. J. Manag. Care Pharm. 2007;13:9–20. doi: 10.18553/jmcp.2007.13.s8-b.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Auer J., Berent R., Lassnig E., Eber B. C-reactive protein and coronary artery disease. Jpn. Heart J. 2002;43:607–619. doi: 10.1536/jhj.43.607. [DOI] [PubMed] [Google Scholar]
  • 10.Banerjee M., Banerjee N., Ghosh P., Das J.K., Basu S., Sarkar A.K., States J.C., Giri A.K. Evaluation of the serum catalase and myeloperoxidase activities in chronic arsenic-exposed individuals and concomitant cytogenetic damage. Toxicol. Appl. Pharmacol. 2010;249:47–54. doi: 10.1016/j.taap.2010.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Barnoya J., Glantz S.A. Cardiovascular effects of secondhand smoke: nearly as large as smoking. Circulation. 2005;111:2684–2698. doi: 10.1161/CIRCULATIONAHA.104.492215. [DOI] [PubMed] [Google Scholar]
  • 12.Beilin L.J., Puddey I.B. Alcohol and hypertension: an update. Hypertension. 2006;47:1035–1038. doi: 10.1161/01.HYP.0000218586.21932.3c. [DOI] [PubMed] [Google Scholar]
  • 13.Benstoem C., Goetzenich A., Kraemer S., Borosch S., Manzanares W., Hardy G., Stoppe C. Selenium and its supplementation in cardiovascular disease--what do we know? Nutrients. 2015;7:3094–3118. doi: 10.3390/nu7053094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Berg Z.K., Rodriguez B., Davis J., Katz A.R., Cooney R.V., Masaki K. Association between occupational exposure to pesticides and cardiovascular disease incidence: The Kuakini Honolulu Heart Program. J. Am. Heart Assoc. 2019;8 doi: 10.1161/JAHA.119.012569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Blankenberg S., Rupprecht H.J., Bickel C., Peetz D., Hafner G., Tiret L., Meyer J. Circulating cell adhesion molecules and death in patients with coronary artery disease. Circulation. 2001;104:1336–1342. doi: 10.1161/hc3701.095949. [DOI] [PubMed] [Google Scholar]
  • 16.Boffetta P., Sallsten G., Garcia-Gomez M., Pompe-Kirn V., Zaridze D., Bulbulyan M., Caballero J.D., Ceccarelli F., Kobal A.B., Merler E. Mortality from cardiovascular diseases and exposure to inorganic mercury. Occup. Environ. Med. 2001;58:461–466. doi: 10.1136/oem.58.7.461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Botero-Velez M., Curtis J.J., Warnock D.G. Brief report: Liddle’s syndrome revisited--a disorder of sodium reabsorption in the distal tubule. New Engl. J. Med. 1994;330:178–181. doi: 10.1056/NEJM199401203300305. [DOI] [PubMed] [Google Scholar]
  • 18.Bourdrel T., Bind M.A., Bejot Y., Morel O., Argacha J.F. Cardiovascular effects of air pollution. Arch. Cardiovasc. Dis. 2017;110:634–642. doi: 10.1016/j.acvd.2017.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Braunthal S., Brateanu A. Hypertension in pregnancy: pathophysiology and treatment. SAGE Open Med. 2019;7 doi: 10.1177/2050312119843700. 2050312119843700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Brinkley T.E., Leng X., Miller M.E., Kitzman D.W., Pahor M., Berry M.J., Marsh A.P., Kritchevsky S.B., Nicklas B.J. Chronic inflammation is associated with low physical function in older adults across multiple comorbidities. J. Gerontol. A Biol. 2009;64:455–461. doi: 10.1093/gerona/gln038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Buford T.W. Hypertension and aging. Ageing Res. Rev. 2016;26:96–111. doi: 10.1016/j.arr.2016.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bunderson M., Coffin J.D., Beall H.D. Arsenic induces peroxynitrite generation and cyclooxygenase-2 protein expression in aortic endothelial cells: possible role in atherosclerosis. Toxicol. Appl. Pharmacol. 2002;184:11–18. [PubMed] [Google Scholar]
  • 23.Byrd J.B., Morishita M., Bard R.L., Das R., Wang L., Sun Z., Spino C., Harkema J., Dvonch J.T., Rajagopalan S., Brook R.D. Acute increase in blood pressure during inhalation of coarse particulate matter air pollution from an urban location. J. Am. Soc. Hypertens. 2016;10:133–139. doi: 10.1016/j.jash.2015.11.015. e134. [DOI] [PubMed] [Google Scholar]
  • 24.Caciari T., Sancini A., Fioravanti M., Capozzella A., Casale T., Montuori L., Fiaschetti M., Schifano M.P., Andreozzi G., Nardone N., Tomei G., Ciarrocca M., Rosati M.V., Tomei F. Cadmium and hypertension in exposed workers: A meta-analysis. Int. J. Occup. Med. Environ. Health. 2013;26:440–456. doi: 10.2478/s13382-013-0111-5. [DOI] [PubMed] [Google Scholar]
  • 25.Caliez J., Riou M., Manaud G., Nakhleh M.K., Quatredeniers M., Rucker-Martin C., Dorfmüller P., Lecerf F., Vinhas M.C., Khatib S., Haick H., Cohen-Kaminsky S., Humbert M., Montani D., Perros F. Trichloroethylene increases pulmonary endothelial permeability: implication for pulmonary veno-occlusive disease. Pulm. Circ. 2020;10(4) doi: 10.1177/2045894020907884. 2045894020907884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Campbell D.J., Habener J.F. Angiotensinogen gene is expressed and differentially regulated in multiple tissues of the rat. J. Clin. Investig.. 1986;78:31–39. doi: 10.1172/JCI112566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Cardenas A., Roels H., Bernard A.M., Barbon R., Buchet J.P., Lauwerys R.R., Rosello J., Ramis I., Mutti A., Franchini I., et al. Markers of early renal changes induced by industrial pollutants. II. Application to workers exposed to lead. Br. J. Ind. Med. 1993;50:28–36. doi: 10.1136/oem.50.1.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Carmignani M., Boscolo P., Poma A., Volpe A.R. Kininergic system and arterial hypertension following chronic exposure to inorganic lead. Immunopharmacology. 1999;44:105–110. doi: 10.1016/s0162-3109(99)00115-0. [DOI] [PubMed] [Google Scholar]
  • 29.Carpenter, D.O., 2011. Exposure to Polychlorinated Biphenyls Is Associated With an Increased Risk of Hypertension and Cardiovascular Disease, ISEE 22nd Annual Conference. Epidemiology, Seoul, Korea, p. p S147.
  • 30.Carretero O.A., Oparil S. Essential hypertension Part I: definition and etiology. Circulation. 2000;101:329–335. doi: 10.1161/01.cir.101.3.329. [DOI] [PubMed] [Google Scholar]
  • 31.Centers for Disease Control and Prevention (CDC) 2010. How Tobacco Smoke Causes Disease: The Biology and Behavioral Basis for Smoking-Attributable Disease: A Report of the Surgeon General. National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health, Atlanta, GA, 〈https://www.ncbi.nlm.nih.gov/books/NBK53017/〉. [PubMed]
  • 32.Chang H.R., Chen S.S., Chen T.J., Ho C.H., Chiang H.C., Yu H.S. Lymphocyte beta2-adrenergic receptors and plasma catecholamine levels in lead-exposed workers. Toxicol. Appl. Pharmacol. 1996;139:1–5. doi: 10.1006/taap.1996.0136. [DOI] [PubMed] [Google Scholar]
  • 33.Chang H.R., Chen S.S., Tsao D.A., Cheng J.T., Ho C.K., Yu H.S. Change of cardiac beta-adrenoceptors in lead-exposed rats. Toxicology. 1997;123:27–32. doi: 10.1016/s0300-483x(97)00100-5. [DOI] [PubMed] [Google Scholar]
  • 34.Charles L., Triscott J., Dobbs B. Secondary hypertension: discovering the underlying cause. Am. Fam. Physician. 2017;96:453–461. [PubMed] [Google Scholar]
  • 35.Chen H., Burnett R.T., Kwong J.C., Villeneuve P.J., Goldberg M.S., Brook R.D., van Donkelaar A., Jerrett M., Martin R.V., Kopp A., Brook J.R., Copes R. Spatial association between ambient fine particulate matter and incident hypertension. Circulation. 2014;129:562–569. doi: 10.1161/CIRCULATIONAHA.113.003532. [DOI] [PubMed] [Google Scholar]
  • 36.Chen Y., Hakim M.E., Parvez F., Islam T., Rahman A.M., Ahsan H. Arsenic exposure from drinking-water and carotid artery intima-medial thickness in healthy young adults in Bangladesh. J. Health Popul. Nutr. 2006;24:253–257. [PubMed] [Google Scholar]
  • 37.Chen Y., Santella R.M., Kibriya M.G., Wang Q., Kappil M., Verret W.J., Graziano J.H., Ahsan H. Association between arsenic exposure from drinking water and plasma levels of soluble cell adhesion molecules. Environ. Health Perspect. 2007;115:1415–1420. doi: 10.1289/ehp.10277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chen Y., Wu F., Graziano J.H., Parvez F., Liu M., Paul R.R., Shaheen I., Sarwar G., Ahmed A., Islam T., Slavkovich V., Rundek T., Demmer R.T., Desvarieux M., Ahsan H. Arsenic exposure from drinking water, arsenic methylation capacity, and carotid intima-media thickness in Bangladesh. Am. J. Epidemiol. 2013;178:372–381. doi: 10.1093/aje/kwt001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chen Y., Wu F., Parvez F., Ahmed A., Eunus M., McClintock T.R., Patwary T.I., Islam T., Ghosal A.K., Islam S., Hasan R., Levy D., Sarwar G., Slavkovich V., van Geen A., Graziano J.H., Ahsan H. Arsenic exposure from drinking water and QT-interval prolongation: results from the health effects of arsenic longitudinal study. Environ. Health Perspect. 2013;121:427–432. doi: 10.1289/ehp.1205197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Choi K.B. Hypertensive hypokalemic disorders. Electrolyte Blood Press. 2007;5:34–41. doi: 10.5049/EBP.2007.5.1.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chung H., Youn K., Kim K., Park K. Carbon disulfide exposure estimate and prevalence of chronic diseases after carbon disulfide poisoning-related occupational diseases. Ann. Occup. Environ. Med. 2017;29:52. doi: 10.1186/s40557-017-0208-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chung J.Y., Yu S.D., Hong Y.S. Environmental source of arsenic exposure. J. Prev. Med. Public Health. 2014;47:253–257. doi: 10.3961/jpmph.14.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.da Cunha Martins A., Jr., Carneiro M.F.H., Grotto D., Adeyemi J.A., Barbosa F., Jr. Arsenic, cadmium, and mercury-induced hypertension: mechanisms and epidemiological findings. J. Toxicol. Environ. Health B Crit. Rev. 2018;21:61–82. doi: 10.1080/10937404.2018.1432025. [DOI] [PubMed] [Google Scholar]
  • 44.de la Sierra A. New American and European hypertension guidelines, reconciling the differences. Cardiol. Ther. 2019;8:157–166. doi: 10.1007/s40119-019-0144-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ding Y., Vaziri N.D., Gonick H.C. Lead-induced hypertension. II. Response to sequential infusions of L-arginine, superoxide dismutase, and nitroprusside. Environ. Res. 1998;76:107–113. doi: 10.1006/enrs.1997.3796. [DOI] [PubMed] [Google Scholar]
  • 46.Donat-Vargas C., Akesson A., Tornevi A., Wennberg M., Sommar J., Kiviranta H., Rantakokko P., Bergdahl I.A. Persistent organochlorine pollutants in plasma, blood pressure, and hypertension in a longitudinal study. Hypertension. 2018;71:1258–1268. doi: 10.1161/HYPERTENSIONAHA.117.10691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Emerging Risk Factors C., Kaptoge S., Di Angelantonio E., Lowe G., Pepys M.B., Thompson S.G., Collins R., Danesh J. C-reactive protein concentration and risk of coronary heart disease, stroke, and mortality: an individual participant meta-analysis. Lancet. 2010;375:132–140. doi: 10.1016/S0140-6736(09)61717-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Fagrell B., De Faire U., Bondy S., Criqui M., Gaziano M., Gronbaek M., Jackson R., Klatsky A., Salonen J., Shaper A.G. The effects of light to moderate drinking on cardiovascular diseases. J. Intern. Med. 1999;246:331–340. doi: 10.1046/j.1365-2796.1999.00576.x. [DOI] [PubMed] [Google Scholar]
  • 49.Fasola A.F., Martz B.L., Helmer O.M. Renin activity during supine exercise in normotensives and hypertensives. J. Appl. Physiol. 1966;21:1709–1712. doi: 10.1152/jappl.1966.21.6.1709. [DOI] [PubMed] [Google Scholar]
  • 50.Fatmi Z., Coggon D. Coronary heart disease and household air pollution from use of solid fuel: a systematic review. Br. Med. Bull. 2016;118:91–109. doi: 10.1093/bmb/ldw015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Feierman D.E., Winston G.W., Cederbaum A.I. Ethanol oxidation by hydroxyl radicals: role of iron chelates, superoxide, and hydrogen peroxide. Alcohol. Clin. Exp. Res. 1985;9:95–102. doi: 10.1111/j.1530-0277.1985.tb05525.x. [DOI] [PubMed] [Google Scholar]
  • 52.Flatman P.W. Cotransporters, WNKs and hypertension: an update. Curr. Opin. Nephrol. Hypertens. 2008;17:186–192. doi: 10.1097/MNH.0b013e3282f5244e. [DOI] [PubMed] [Google Scholar]
  • 53.Folkow B. The pathophysiology of hypertension. Differences between young and elderly patients. Drugs. 1993;46(Suppl 2):3–7. doi: 10.2165/00003495-199300462-00003. [DOI] [PubMed] [Google Scholar]
  • 54.Franceschini N., Fry R.C., Balakrishnan P., Navas-Acien A., Oliver-Williams C., Howard A.G., Cole S.A., Haack K., Lange E.M., Howard B.V., Best L.G., Francesconi K.A., Goessler W., Umans J.G., Tellez-Plaza M. Cadmium body burden and increased blood pressure in middle-aged American Indians: the Strong Heart Study. J. Hum. Hypertens. 2017;31:225–230. doi: 10.1038/jhh.2016.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Fregly M.J. Effect of o,p'-DDD and metyrapone (SU-4885) on development of renal hypertension in rats. Toxicol. Appl. Pharmacol. 1968;12:548–559. doi: 10.1016/0041-008x(68)90161-0. [DOI] [PubMed] [Google Scholar]
  • 56.Fuchs F.D., Chambless L.E., Whelton P.K., Nieto F.J., Heiss G. Alcohol consumption and the incidence of hypertension: The Atherosclerosis Risk in Communities Study. Hypertension. 2001;37:1242–1250. doi: 10.1161/01.hyp.37.5.1242. [DOI] [PubMed] [Google Scholar]
  • 57.Ganther H.E. Interactions of vitamin E and selenium with mercury and silver. Ann. N. Y. Acad. Sci. 1980;355:212–226. doi: 10.1111/j.1749-6632.1980.tb21340.x. [DOI] [PubMed] [Google Scholar]
  • 58.Genchi G., Sinicropi M.S., Carocci A., Lauria G., Catalano A. Mercury exposure and heart diseases. Int. J. Environ. Res. Public. Health. 2017:14. doi: 10.3390/ijerph14070761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Giorgini P., Di Giosia P., Grassi D., Rubenfire M., Brook R.D., Ferri C. Air pollution exposure and blood pressure: an updated review of the literature. Curr. Pharm. Des. 2016;22:28–51. doi: 10.2174/1381612822666151109111712. [DOI] [PubMed] [Google Scholar]
  • 60.Giridhar J., Isom G.E. Interaction of lead acetate with atrial natriuretic factor in rats. Life Sci. 1990;46:569–576. doi: 10.1016/0024-3205(90)90124-a. [DOI] [PubMed] [Google Scholar]
  • 61.Goncharov A., Pavuk M., Foushee H.R., Carpenter D.O., Anniston Environmental Health Reseach C. Blood pressure in relation to concentrations of PCB congeners and chlorinated pesticides. Environ. Health Perspect. 2011;119:319–325. doi: 10.1289/ehp.1002830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Gonick H.C., Ding Y., Vaziri N.D. Effect of low lead exposure on eicosanoid excretion in rats. Prostaglandins Other Lipid Mediat. 1998;55:77–82. [Google Scholar]
  • 63.Gonick H.C., Ding Y., Bondy S.C., Ni Z., Vaziri N.D. Lead-induced hypertension: interplay of nitric oxide and reactive oxygen species. Hypertension. 1997;30:1487–1492. doi: 10.1161/01.hyp.30.6.1487. [DOI] [PubMed] [Google Scholar]
  • 64.Gonzalvo M.C., Gil F., Hernandez A.F., Villanueva E., Pla A. Inhibition of paraoxonase activity in human liver microsomes by exposure to EDTA, metals and mercurials. Chem. Biol. Interact. 1997;105:169–179. doi: 10.1016/s0009-2797(97)00046-x. [DOI] [PubMed] [Google Scholar]
  • 65.Gorni D., Finco A. Oxidative stress in elderly population: a prevention screening study. Aging Med. 2020;3:205–213. doi: 10.1002/agm2.12121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gould A.B., Green D. Kinetics of the human renin and human substrate reaction. Cardiovasc. Res. 1971;5:86–89. doi: 10.1093/cvr/5.1.86. [DOI] [PubMed] [Google Scholar]
  • 67.Grahame T.J., Schlesinger R.B. Cardiovascular health and particulate vehicular emissions: a critical evaluation of the evidence. Air Qual. Atmos. Health. 2010;3:3–27. doi: 10.1007/s11869-009-0047-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Grobe J.L., Dickson M.E., Park S., Davis D.R., Born E.J., Sigmund C.D. Cardiovascular consequences of genetic variation at -6/235 in human angiotensinogen using "humanized" gene-targeted mice. Hypertension. 2010;56:981–987. doi: 10.1161/HYPERTENSIONAHA.110.157354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Grogan J.R., Kochar M.S. Alcohol and hypertension. Arch. Fam. Med. 1994;3:150–154. doi: 10.1001/archfami.3.2.150. [DOI] [PubMed] [Google Scholar]
  • 70.Gu A., Yue Y., Desai R.P., Argulian E. Racial and ethnic differences in antihypertensive medication use and blood pressure control among US adults with hypertension: The National Health and Nutrition Examination Survey, 2003 to 2012. Circ. Cardiovasc. Qual. Outcomes. 2017:10. doi: 10.1161/CIRCOUTCOMES.116.003166. [DOI] [PubMed] [Google Scholar]
  • 71.Gupta A.K. Racial differences in response to antihypertensive therapy: does one size fits all? Int. J. Prev. Med. 2010;1:217–219. [PMC free article] [PubMed] [Google Scholar]
  • 72.B.A.G. Halliwel, J.M.C., 2015. Free Radicals in Biology and Medicine Oxford University Press, Oxford Scholarship Online.
  • 73.Halperin F., Dluhy R.G. Glucocorticoid-remediable aldosteronism. Endocrinol. Metab. Clin. North Am. 2011;40:333–341. doi: 10.1016/j.ecl.2011.01.012. [DOI] [PubMed] [Google Scholar]
  • 74.Han C., Hong Y.C. Bisphenol A, hypertension, and cardiovascular diseases: epidemiological, laboratory, and clinical trial evidence. Curr. Hypertens. Rep. 2016;18:11. doi: 10.1007/s11906-015-0617-2. [DOI] [PubMed] [Google Scholar]
  • 75.Huang K., Yang X., Liang F., Liu F., Li J., Xiao Q., Chen J., Liu X., Cao J., Shen C., Yu L., Lu F., Wu X., Zhao L., Wu X., Li Y., Hu D., Huang J., Liu Y., Lu X., Gu D. Long-term exposure to fine particulate matter and hypertension incidence in China. Hypertension. 2019;73:1195–1201. doi: 10.1161/HYPERTENSIONAHA.119.12666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hennig B., Ormsbee L., McClain C.J., Watkins B.A., Blumberg B., Bachas L.G., Sanderson W., Thompson C., Suk W.A. Nutrition can modulate the toxicity of environmental pollutants: implications in risk assessment and human health. Environ. Health Perspect. 2012;120:771–774. doi: 10.1289/ehp.1104712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Henriquez-Hernandez L.A., Luzardo O.P., Zumbado M., Camacho M., Serra-Majem L., Alvarez-Leon E.E., Boada L.D. Blood pressure in relation to contamination by polychlorobiphenyls and organochlorine pesticides: results from a population-based study in the Canary Islands (Spain) Environ. Res. 2014;135:48–54. doi: 10.1016/j.envres.2014.05.036. [DOI] [PubMed] [Google Scholar]
  • 78.Heyer N.J., Echeverria D., Martin M.D., Farin F.M., Woods J.S. Catechol O-methyltransferase (COMT) VAL158MET functional polymorphism, dental mercury exposure, and self-reported symptoms and mood. J. Toxicol. Environ. Health A. 2009;72:599–609. doi: 10.1080/15287390802706405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Hoeper M.M., Humbert M. The new haemodynamic definition of pulmonary hypertension: evidence prevails, finally! Eur. Respir. J. 2019;53 doi: 10.1183/13993003.00038-2019. [DOI] [PubMed] [Google Scholar]
  • 80.Hogg J.C., van Eeden S. Pulmonary and systemic response to atmospheric pollution. Respirology. 2009;14:336–346. doi: 10.1111/j.1440-1843.2009.01497.x. [DOI] [PubMed] [Google Scholar]
  • 81.Hottenga J.J., Boomsma D.I., Kupper N., Posthuma D., Snieder H., Willemsen G., de Geus E.J. Heritability and stability of resting blood pressure. Twin Res. Hum. Genet. 2005;8:499–508. doi: 10.1375/183242705774310123. [DOI] [PubMed] [Google Scholar]
  • 82.Hotter G., Fels L.M., Closa D., Rosello J., Stolte H., Gelpi E. Altered levels of urinary prostanoids in lead-exposed workers. Toxicol. Lett. 1995;77:309–312. doi: 10.1016/0378-4274(95)03311-4. [DOI] [PubMed] [Google Scholar]
  • 83.Hsieh Y.C., Lien L.M., Chung W.T., Hsieh F.I., Hsieh P.F., Wu M.M., Tseng H.P., Chiou H.Y., Chen C.J. Significantly increased risk of carotid atherosclerosis with arsenic exposure and polymorphisms in arsenic metabolism genes. Environ. Res. 2011;111:804–810. doi: 10.1016/j.envres.2011.05.003. [DOI] [PubMed] [Google Scholar]
  • 84.Hwang K.Y., Lee B.K., Bressler J.P., Bolla K.I., Stewart W.F., Schwartz B.S. Protein kinase C activity and the relations between blood lead and neurobehavioral function in lead workers. Environ. Health Perspect. 2002;110:133–138. doi: 10.1289/ehp.02110133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Ito R., Sato I., Tsujita T., Yokoyama A., Sugawara A. A ubiquitin-proteasome inhibitor bortezomib suppresses the expression of CYP11B2, a key enzyme of aldosterone synthesis. Biochem. Biophys. Res. Commun. 2017;489:21–28. doi: 10.1016/j.bbrc.2017.05.109. [DOI] [PubMed] [Google Scholar]
  • 86.Jain S., Tillinger A., Mopidevi B., Pandey V.G., Chauhan C.K., Fiering S.N., Warming S., Kumar A. Transgenic mice with -6A haplotype of the human angiotensinogen gene have increased blood pressure compared with -6G haplotype. J. Biol. Chem. 2010;285:41172–41186. doi: 10.1074/jbc.M110.167585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Jain S., Vinukonda G., Fiering S.N., Kumar A. A haplotype of human angiotensinogen gene containing -217A increases blood pressure in transgenic mice compared with -217G. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2008;295:R1849–R1857. doi: 10.1152/ajpregu.90637.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.James P.A., Oparil S., Carter B.L., Cushman W.C., Dennison-Himmelfarb C., Handler J., Lackland D.T., LeFevre M.L., MacKenzie T.D., Ogedegbe O., Smith S.C., Jr., Svetkey L.P., Taler S.J., Townsend R.R., Wright J.T., Narva A.S., Ortiz E. Evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the eighth joint national committee (JNC 8) JAMA. 2014;311(5):507–520. doi: 10.1001/jama.2013.284427. [DOI] [PubMed] [Google Scholar]
  • 89.Jansson G., Harms-Ringdahl M. Stimulating effects of mercuric- and silver ions on the superoxide anion production in human polymorphonuclear leukocytes. Free Radic. Res. Commun. 1993;18:87–98. doi: 10.3109/10715769309147345. [DOI] [PubMed] [Google Scholar]
  • 90.Japanese Society of Hypertension (JSH) Guidelines (JSH 2014), Chapter 13. Secondary hypertension. Hypertens. Res. 2014;37:349–361. [Google Scholar]
  • 91.Jeunemaitre X., Soubrier F., Kotelevtsev Y.V., Lifton R.P., Williams C.S., Charru A., Hunt S.C., Hopkins P.N., Williams R.R., Lalouel J.M., et al. Molecular basis of human hypertension: role of angiotensinogen. Cell. 1992;71:169–180. doi: 10.1016/0092-8674(92)90275-h. [DOI] [PubMed] [Google Scholar]
  • 92.Jonsson J.R., Klemm S.A., Tunny T.J., Stowasser M., Gordon R.D. A new genetic test for familial hyperaldosteronism type I aids in the detection of curable hypertension. Biochem. Biophys. Res. Commun. 1995;207:565–571. doi: 10.1006/bbrc.1995.1225. [DOI] [PubMed] [Google Scholar]
  • 93.Kaplan B.L., Li J., LaPres J.J., Pruett S.B., Karmaus P.W. Contributions of nonhematopoietic cells and mediators to immune responses: implications for immunotoxicology. Toxicol. Sci. 2015;145:214–232. doi: 10.1093/toxsci/kfv060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kaplan N.M. Alcohol and hypertension. Lancet. 1995;345:1588–1589. doi: 10.1016/s0140-6736(95)90110-8. [DOI] [PubMed] [Google Scholar]
  • 95.Karim M.R., Rahman M., Islam K., Mamun A.A., Hossain S., Hossain E., Aziz A., Yeasmin F., Agarwal S., Hossain M.I., Saud Z.A., Nikkon F., Hossain M., Mandal A., Jenkins R.O., Haris P.I., Miyataka H., Himeno S., Hossain K. Increases in oxidized low-density lipoprotein and other inflammatory and adhesion molecules with a concomitant decrease in high-density lipoprotein in the individuals exposed to arsenic in Bangladesh. Toxicol. Sci. 2013;135:17–25. doi: 10.1093/toxsci/kft130. [DOI] [PubMed] [Google Scholar]
  • 96.Keusch S., Hildenbrand F.F., Bollmann T., Halank M., Held M., Kaiser R., Kovacs G., Lange T.J., Seyfarth H.J., Speich R., Ulrich S. Tobacco smoke exposure in pulmonary arterial and thromboembolic pulmonary hypertension. Respiration. 2014;88(1):38–45. doi: 10.1159/000359972. [DOI] [PubMed] [Google Scholar]
  • 97.Khalil-Manesh F., Gonick H.C., Weiler E.W., Prins B., Weber M.A., Purdy R., Ren Q. Effect of chelation treatment with dimercaptosuccinic acid (DMSA) on lead-related blood pressure changes. Environ. Res. 1994;65:86–99. doi: 10.1006/enrs.1994.1023. [DOI] [PubMed] [Google Scholar]
  • 98.Khalil-Manesh F., Gonick H.C., Weiler E.W., Prins B., Weber M.A., Purdy R.E. Lead-induced hypertension: possible role of endothelial factors. Am. J. Hypertens. 1993;6:723–729. doi: 10.1093/ajh/6.9.723. [DOI] [PubMed] [Google Scholar]
  • 99.Kim H.S., Krege J.H., Kluckman K.D., Hagaman J.R., Hodgin J.B., Best C.F., Jennette J.C., Coffman T.M., Maeda N., Smithies O. Genetic control of blood pressure and the angiotensinogen locus. Proc. Natl. Acad. Sci. U.S.A. 1995;92:2735–2739. doi: 10.1073/pnas.92.7.2735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Kim K.W., Won Y.L., Ko K.S., Heo K.H., Chung Y.H. The effects of hazardous chemical exposure on cardiovascular disease in chemical products manufacturing workers. Toxicol Res. 2012;28:269–277. doi: 10.5487/TR.2012.28.4.269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Klatsky A.L. Alcohol-associated hypertension: when one drinks makes a difference. Hypertension. 2004;44:805–806. doi: 10.1161/01.HYP.0000146538.26193.60. [DOI] [PubMed] [Google Scholar]
  • 102.Kobal A.B., Flisar Z., Miklavcic V., Dizdarevic T., Sesek-Briski A. Renal function in miners intermittently exposed to elemental mercury vapour. Arh. Hig. Rada Toksikol. 2000;51:369–380. [PubMed] [Google Scholar]
  • 103.Kohle C., Bock K.W. Coordinate regulation of Phase I and II xenobiotic metabolisms by the Ah receptor and Nrf2. Biochem. Pharmacol. 2007;73:1853–1862. doi: 10.1016/j.bcp.2007.01.009. [DOI] [PubMed] [Google Scholar]
  • 104.Kozina A.A., Trofimova T.A., Okuneva E.G., Baryshnikova N.V., Obuhova V.A., Krasnenko A.Y., Tsukanov K.Y., Klimchuk O.I., Surkova E.I., Shatalov P.A., Ilinsky V.V. Liddle syndrome due to a novel mutation in the gamma subunit of the epithelial sodium channel (ENaC) in family from Russia: a case report. BMC Nephrol. 2019;20:389. doi: 10.1186/s12882-019-1579-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Krege J.H., Kim H.S., Moyer J.S., Jennette J.C., Peng L., Hiller S.K., Smithies O. Angiotensin-converting enzyme gene mutations, blood pressures, and cardiovascular homeostasis. Hypertension. 1997;29:150–157. doi: 10.1161/01.hyp.29.1.150. [DOI] [PubMed] [Google Scholar]
  • 106.Lackland D.T. Hypertension, joint national committee on detection, evaluation, and treatment of high blood pressure guidelines. Curr. Opin. Neurol. 2013;26:8–12. doi: 10.1097/WCO.0b013e32835c4f54. [DOI] [PubMed] [Google Scholar]
  • 107.Lackland D.T. Racial differences in hypertension: implications for high blood pressure management. Am. J. Med. Sci. 2014;348:135–138. doi: 10.1097/MAJ.0000000000000308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Lee M.S., Park S.K., Hu H., Lee S. Cadmium exposure and cardiovascular disease in the 2005 Korea National Health and Nutrition Examination Survey. Environ. Res. 2011;111:171–176. doi: 10.1016/j.envres.2010.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.LeHoux J.G., Dupuis G., Lefebvre A. Control of CYP11B2 gene expression through differential regulation of its promoter by atypical and conventional protein kinase C isoforms. J. Biol. Chem. 2001;276:8021–8028. doi: 10.1074/jbc.M009495200. [DOI] [PubMed] [Google Scholar]
  • 110.Lenzini L., Prisco S., Caroccia B., Rossi G.P. Saga of familial hyperaldosteronism: yet a new channel. Hypertension. 2018;71:1010–1014. doi: 10.1161/HYPERTENSIONAHA.118.11150. [DOI] [PubMed] [Google Scholar]
  • 111.Liao X., Yang Z., Peng D., Dai H., Lei Y., Zhao Q., Han Y., Wang W. Association of T174M polymorphism of angiotensinogen gene with essential hypertension: a meta-analysis. Genet. Mol. Biol. 2014;37:473–479. doi: 10.1590/s1415-47572014000400001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Libby P. Inflammation in atherosclerosis. Nature. 2002;420:868–874. doi: 10.1038/nature01323. [DOI] [PubMed] [Google Scholar]
  • 113.Liddle G.W., B.T, Coppage W.S.J. A familial renal disorder simulating primary aldosteronism but with negligible aldosterone secretion. Trans. Assoc. Am. Phys. 1963;76:199–213. [Google Scholar]
  • 114.Liddle G.W.Ba.C., W.S. J A familial renal disorder simulating primary aldosteronism but with negligible aldosterone secretion. Trans. Assoc. Am. Phys. 1963;76:199–213. [Google Scholar]
  • 115.Lifton R.P., Dluhy R.G., Powers M., Rich G.M., Cook S., Ulick S., Lalouel J.M. A chimaeric 11 beta-hydroxylase/aldosterone synthase gene causes glucocorticoid-remediable aldosteronism and human hypertension. Nature. 1992;355:262–265. doi: 10.1038/355262a0. [DOI] [PubMed] [Google Scholar]
  • 116.Lund B.O., Miller D.M., Woods J.S. Mercury-induced H2O2 production and lipid peroxidation in vitro in rat kidney mitochondria. Biochem. Pharmacol. 1991;42 Suppl:S181–S187. doi: 10.1016/0006-2952(91)90408-w. [DOI] [PubMed] [Google Scholar]
  • 117.Lund B.O., Miller D.M., Woods J.S. Studies on Hg(II)-induced H2O2 formation and oxidative stress in vivo and in vitro in rat kidney mitochondria. Biochem. Pharmacol. 1993;45:2017–2024. doi: 10.1016/0006-2952(93)90012-l. [DOI] [PubMed] [Google Scholar]
  • 118.Ma L., Bi K.D., Fan Y.M., Jiang Z.Y., Zhang X.Y., Zhang J.W., Zhao J., Jiang F.L., Dong J.X. In vitro modulation of mercury-induced rat liver mitochondria dysfunction. Toxicol. Res. 2018;7:1135–1143. doi: 10.1039/c8tx00060c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Ma Q. Transcriptional responses to oxidative stress: pathological and toxicological implications. Pharmacol. Ther. 2010;125:376–393. doi: 10.1016/j.pharmthera.2009.11.004. [DOI] [PubMed] [Google Scholar]
  • 120.Maarman G., Lecour S., Butrous G., Thienemann F., Sliwa K. A comprehensive review: the evolution of animal models in pulmonary hypertension research; are we there yet? Pulm. Circ. 2013;3:739–756. doi: 10.1086/674770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Mariana M., Cairrao E. Phthalates implications in the cardiovascular system. J. Cardiovasc. Dev. Dis. 2020:7. doi: 10.3390/jcdd7030026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Markovac J., Goldstein G.W. Picomolar concentrations of lead stimulate brain protein kinase C. Nature. 1988;334:71–73. doi: 10.1038/334071a0. [DOI] [PubMed] [Google Scholar]
  • 123.Marris C.R., Kompella S.N., Miller M.R., Incardona J.P., Brette F., Hancox J.C., Sorhus E., Shiels H.A. Polyaromatic hydrocarbons in pollution: a heart-breaking matter. J. Physiol. 2020;598:227–247. doi: 10.1113/JP278885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Mathews, C.K., van Holde, K.E., and Ahern, K.G., 1999. Biochemistry. Benjamin-Cummings.
  • 125.Mensah G.A. Hypertension and target organ damage: don’t believe everything you think! Ethn. Dis. 2016;26:275–278. doi: 10.18865/ed.26.3.275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Miller M.R., Newby D.E. Air pollution and cardiovascular disease: car sick. Cardiovasc. Res. 2020;116:279–294. doi: 10.1093/cvr/cvz228. [DOI] [PubMed] [Google Scholar]
  • 127.Mitter S.S., Vedanthan R., Islami F., Pourshams A., Khademi H., Kamangar F., Abnet C.C., Dawsey S.M., Pharoah P.D., Brennan P., Fuster V., Boffetta P., Malekzadeh R. Household fuel use and cardiovascular disease mortality: golestan cohort study. Circulation. 2016;133:2360–2369. doi: 10.1161/CIRCULATIONAHA.115.020288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Molero L., Carrasco C., Marques M., Vaziri N.D., Mateos-Caceres P.J., Casado S., Macaya C., Barrientos A., Lopez-Farre A.J. Involvement of endothelium and endothelin-1 in lead-induced smooth muscle cell dysfunction in rats. Kidney Int. 2006;69:685–690. doi: 10.1038/sj.ki.5000103. [DOI] [PubMed] [Google Scholar]
  • 129.Montani D., Seferian A., Savale L., Simonneau G., Humbert M. Drug-induced pulmonary arterial hypertension: a recent outbreak. Eur. Respir. Rev. 2013;22:244–250. doi: 10.1183/09059180.00003313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Mordukhovich I., Wright R.O., Amarasiriwardena C., Baja E., Baccarelli A., Suh H., Sparrow D., Vokonas P., Schwartz J. Association between low-level environmental arsenic exposure and QT interval duration in a general population study. Am. J. Epidemiol. 2009;170:739–746. doi: 10.1093/aje/kwp191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Moshage H. Cytokines and the hepatic acute phase response. J. Pathol. 1997;181:257–266. doi: 10.1002/(SICI)1096-9896(199703)181:3<257::AID-PATH756>3.0.CO;2-U. [DOI] [PubMed] [Google Scholar]
  • 132.Mumford J.L., Wu K., Xia Y., Kwok R., Yang Z., Foster J., Sanders W.E. Chronic arsenic exposure and cardiac repolarization abnormalities with QT interval prolongation in a population-based study. Environ. Health Perspect. 2007;115:690–694. doi: 10.1289/ehp.9686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Nakatani S., Ishimura E., Naganuma T., Nakatani A., Ichii M., Fukumoto S., Mori K., Emoto M., Nakatani T., Inaba M. Poor glycemic control and decreased renal function are associated with increased intrarenal RAS activity in Type 2 diabetes mellitus. Diabetes Res. Clin. Pract. 2014;105:40–46. doi: 10.1016/j.diabres.2014.04.019. [DOI] [PubMed] [Google Scholar]
  • 134.National Institutes of Health (NIH) 2003. Your Guide to Lowering Blood pressure. U.S. Department of Health and Human Services, NIH, National Heart, Lung, and Blood Institute, NIH Publication No. 03–5232, 〈https://www.nhlbi.nih.gov/files/docs/public/heart/hbp_low.pdf〉.
  • 135.NCD Risk Factor Collaboration (NCD-RisC) Worldwide trends in blood pressure from 1975 to 2015: a pooled analysis of 1479 population-based measurement studies with 19·1 million participants. Lancet. 2017;389:37–55. doi: 10.1016/S0140-6736(16)31919-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Nigra A.E., Ruiz-Hernandez A., Redon J., Navas-Acien A., Tellez-Plaza M. Environmental metals and cardiovascular disease in adults: a systematic review beyond lead and cadmium. Curr. Environ. Health Rep. 2016;3:416–433. doi: 10.1007/s40572-016-0117-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Nogueira E.F., Rainey W.E. Regulation of aldosterone synthase by activator transcription factor/cAMP response element-binding protein family members. Endocrinology. 2010;151:1060–1070. doi: 10.1210/en.2009-0977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Oakes J.M., Xu J., Morris T.M., Fried N.D., Pearson C.S., Lobell T.D., Gilpin N.W., Lazartigues E., Gardner J.D., Yue X. Effects of chronic nicotine inhalation on systemic and pulmonary blood pressure and right ventricular remodeling in mice. Hypertension. 2020;75(5):1305–1314. doi: 10.1161/HYPERTENSIONAHA.119.14608. (May) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Olea N., Pazos P., Exposito J. Inadvertent exposure to xenoestrogens. Eur. J. Cancer Prev. 1998;7(1):S17–S23. doi: 10.1097/00008469-199802001-00005. [DOI] [PubMed] [Google Scholar]
  • 140.Oparil S., Acelajado M.C., Bakris G.L., Berlowitz D.R., Cifkova R., Dominiczak A.F., Grassi G., Jordan J., Poulter N.R., Rodgers A., Whelton P.K. Hypertension. Nat. Rev. Dis. Prim. 2018;4:18014. doi: 10.1038/nrdp.2018.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Orcholski M.E., Yuan K., Rajasingh C., Tsai H., Shamskhou E.A., Dhillon N.K., et al. Drug-induced pulmonary arterial hypertension: a primer for clinicians and scientists. Am. J. Physiol. Lung Cell. Mol. Physiol. 2018;314:L967–L983. doi: 10.1152/ajplung.00553.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Osorio-Yanez C., Ayllon-Vergara J.C., Aguilar-Madrid G., Arreola-Mendoza L., Hernandez-Castellanos E., Barrera-Hernandez A., De Vizcaya-Ruiz A., Del Razo L.M. Carotid intima-media thickness and plasma asymmetric dimethylarginine in Mexican children exposed to inorganic arsenic. Environ. Health Perspect. 2013;121:1090–1096. doi: 10.1289/ehp.1205994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Palmer B.F., Alpern R.J. Liddle's syndrome. Am. J. Med. 1998;104:301–309. doi: 10.1016/s0002-9343(98)00018-7. [DOI] [PubMed] [Google Scholar]
  • 144.Park S.H., Lim J.E., Park H., Jee S.H. Body burden of persistent organic pollutants on hypertension: a meta-analysis. Environ. Sci. Pollut. Res. Int. 2016;23:14284–14293. doi: 10.1007/s11356-016-6568-6. [DOI] [PubMed] [Google Scholar]
  • 145.Pascoe L., Curnow K.M., Slutsker L., Connell J.M., Speiser P.W., New M.I., White P.C. Glucocorticoid-suppressible hyperaldosteronism results from hybrid genes created by unequal crossovers between CYP11B1 and CYP11B2. Proc. Natl. Acad. Sci. U.S.A. 1992 doi: 10.1073/pnas.89.17.8327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Perkins J.T., Petriello M.C., Newsome B.J., Hennig B. Polychlorinated biphenyls and links to cardiovascular disease. Environ. Sci. Pollut. Res. Int. 2016;23:2160–2172. doi: 10.1007/s11356-015-4479-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Pierce G.L., Lesniewski L.A., Lawson B.R., Beske S.D., Seals D.R. Nuclear factor-{kappa}B activation contributes to vascular endothelial dysfunction via oxidative stress in overweight/obese middle-aged and older humans. Circulation. 2009;119:1284–1292. doi: 10.1161/CIRCULATIONAHA.108.804294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Pizzorno, J., 2020. Environmental Toxins and Cardiovascular Disease. Thoracic Key.
  • 149.Prabhakaran D., Mandal S., Krishna B., Magsumbol M., Singh K., Tandon N., Narayan K., Shivashankar R., Kondal D., Ali M.K., Srinath Reddy K., Schwartz J.D., GeoHealth Hub Study investigators, COE-CARRS Study investigators Exposure to particulate matter is associated with elevated blood pressure and incident hypertension in urban India. Hypertension. 2020;76:1289–1298. doi: 10.1161/HYPERTENSIONAHA.120.15373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Purkait P., Halder K., Thakur S., Ghosh Roy A., Raychaudhuri P., Bhattacharya S., Sarkar B.N., Naidu J.M. Association of angiotensinogen gene SNPs and haplotypes with risk of hypertension in eastern Indian population. Clin. Hypertens. 2017;23:12. doi: 10.1186/s40885-017-0069-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Ramesh G.T., Manna S.K., Aggarwal B.B., Jadhav A.L. Lead exposure activates nuclear factor kappa B, activator protein-1, c-Jun N-terminal kinase and caspases in the rat brain. Toxicol. Lett. 2001;123:195–207. doi: 10.1016/s0378-4274(01)00395-2. [DOI] [PubMed] [Google Scholar]
  • 152.Randin D., Vollenweider P., Tappy L., Jequier E., Nicod P., Scherrer U. Suppression of alcohol-induced hypertension by dexamethasone. New Engl. J. Med. 1995;332:1733–1737. doi: 10.1056/NEJM199506293322601. [DOI] [PubMed] [Google Scholar]
  • 153.Resstel L.B., Scopinho A.A., Lopes da Silva A., Antunes-Rodrigues J., Correa F.M. Increased circulating vasopressin may account for ethanol-induced hypertension in rats. Am. J. Hypertens. 2008;21:930–935. doi: 10.1038/ajh.2008.189. [DOI] [PubMed] [Google Scholar]
  • 154.Rice K.M., Walker E.M., Jr., Wu M., Gillette C., Blough E.R. Environmental mercury and its toxic effects. J. Prev. Med. Public Health. 2014;47:74–83. doi: 10.3961/jpmph.2014.47.2.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Ridker P.M., Hennekens C.H., Roitman-Johnson B., Stampfer M.J., Allen J. Plasma concentration of soluble intercellular adhesion molecule 1 and risks of future myocardial infarction in apparently healthy men. Lancet. 1998;351:88–92. doi: 10.1016/S0140-6736(97)09032-6. [DOI] [PubMed] [Google Scholar]
  • 156.Rodrigues J.V.R., Pinto M.M., Figueredo R.M.P., Lima H., Souto R., Sacchetim S.C. Systemic arterial hypertension in patients exposed to cesium-137 in goiania-GO: prevalence study. Arq. Bras. Cardiol. 2017;108:533–538. doi: 10.5935/abc.20170062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Rodriguez-Iturbe B., Vaziri N.D., Herrera-Acosta J., Johnson R.J. Oxidative stress, renal infiltration of immune cells, and salt-sensitive hypertension: all for one and one for all. Am. J. 2004;286:F606–F616. doi: 10.1152/ajprenal.00269.2003. [DOI] [PubMed] [Google Scholar]
  • 158.Rodriguez-Manas L., El-Assar M., Vallejo S., Lopez-Doriga P., Solis J., Petidier R., Montes M., Nevado J., Castro M., Gomez-Guerrero C., Peiro C., Sanchez-Ferrer C.F. Endothelial dysfunction in aged humans is related with oxidative stress and vascular inflammation. Aging Cell. 2009;8:226–238. doi: 10.1111/j.1474-9726.2009.00466.x. [DOI] [PubMed] [Google Scholar]
  • 159.Roman H.A., Walsh T.L., Coull B.A., Dewailly E., Guallar E., Hattis D., Marien K., Schwartz J., Stern A.H., Virtanen J.K., Rice G. Evaluation of the cardiovascular effects of methylmercury exposures: current evidence supports development of a dose-response function for regulatory benefits analysis. Environ. Health Perspect. 2011;119:607–614. doi: 10.1289/ehp.1003012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Rossier B.C., Schild L. Epithelial sodium channel: mendelian versus essential hypertension. Hypertension. 2008;52:595–600. doi: 10.1161/HYPERTENSIONAHA.107.097147. [DOI] [PubMed] [Google Scholar]
  • 161.Rosskopf D., Schurks M., Rimmbach C., Schafers R. Genetics of arterial hypertension and hypotension. Naunyn. Schmiedebergs Arch. Pharmacol. 2007;374:429–469. doi: 10.1007/s00210-007-0133-2. [DOI] [PubMed] [Google Scholar]
  • 162.Sabik L.M., Abbas R.A., Ismail M.M., El-Refaei S. Cardiotoxicity of Freon among refrigeration services workers: comparative cross-sectional study. Environ. Health. 2009;8:31. doi: 10.1186/1476-069X-8-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Salonen J.T., Malin R., Tuomainen T.P., Nyyssonen K., Lakka T.A., Lehtimaki T. Polymorphism in high density lipoprotein paraoxonase gene and risk of acute myocardial infarction in men: prospective nested case-control study. BMJ. 1999;319:487–489. doi: 10.1136/bmj.319.7208.487. discussion 490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Salonen J.T., Seppanen K., Lakka T.A., Salonen R., Kaplan G.A. Mercury accumulation and accelerated progression of carotid atherosclerosis: a population-based prospective 4-year follow-up study in men in eastern Finland. Atherosclerosis. 2000;148:265–273. doi: 10.1016/s0021-9150(99)00272-5. [DOI] [PubMed] [Google Scholar]
  • 165.Salonen J.T., Seppanen K., Nyyssonen K., Korpela H., Kauhanen J., Kantola M., Tuomilehto J., Esterbauer H., Tatzber F., Salonen R. Intake of mercury from fish, lipid peroxidation, and the risk of myocardial infarction and coronary, cardiovascular, and any death in eastern Finnish men. Circulation. 1995;91:645–655. doi: 10.1161/01.cir.91.3.645. [DOI] [PubMed] [Google Scholar]
  • 166.Sanidas E., Papadopoulos D.P., Grassos H., Velliou M., Tsioufis K., Barbetseas J., Papademetriou V. Air pollution and arterial hypertension. A new risk factor is in the air. J. Am. Soc. Hypertens. 2017;11:709–715. doi: 10.1016/j.jash.2017.09.008. [DOI] [PubMed] [Google Scholar]
  • 167.Satarug S. Dietary cadmium intake and its effects on kidneys. Toxics. 2018:6. doi: 10.3390/toxics6010015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Satarug S., Vesey D.A., Gobe G.C. Kidney cadmium toxicity, diabetes and high blood pressure: the perfect storm. Tohoku J. Exp. Med. 2017;241:65–87. doi: 10.1620/tjem.241.65. [DOI] [PubMed] [Google Scholar]
  • 169.Schindhelm R.K., van der Zwan L.P., Teerlink T., Scheffer P.G. Myeloperoxidase: a useful biomarker for cardiovascular disease risk stratification? Clin. Chem. 2009;55:1462–1470. doi: 10.1373/clinchem.2009.126029. [DOI] [PubMed] [Google Scholar]
  • 170.Schiess R., Senn O., Fischler M., Huber L.C., Vatandaslar S., Speich R., Ulrich S. Tobacco smoke: a risk factor for pulmonary arterial hypertension? A case-control study. Chest. 2010;138(5):1086–1092. doi: 10.1378/chest.09-2962. (Nov) [DOI] [PubMed] [Google Scholar]
  • 171.Senoner T., Dichtl W. Oxidative Stress in Cardiovascular Diseases: Still a Therapeutic Target? Nutrients. 2019:11. doi: 10.3390/nu11092090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Sergeev A.V., Carpenter D.O. Hospitalization rates for coronary heart disease in relation to residence near areas contaminated with persistent organic pollutants and other pollutants. Environ. Health Perspect. 2005;113:756–761. doi: 10.1289/ehp.7595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Shahrrava A., Moinuddin S., Boddu P., Shah R. A Case of Glucocorticoid Remediable Aldosteronism and Thoracoabdominal Aneurysms. Case Rep Endocrinol. 2016;2016 doi: 10.1155/2016/2017571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Sharifi A.M., Darabi R., Akbarloo N., Larijani B., Khoshbaten A. Investigation of circulatory and tissue ACE activity during development of lead-induced hypertension. Toxicol. Lett. 2004;153:233–238. doi: 10.1016/j.toxlet.2004.04.013. [DOI] [PubMed] [Google Scholar]
  • 175.Soardo G., Donnini D., Moretti M., Milocco C., Catena C., Sechi L.A. Effects of antihypertensive drugs on alcohol-induced functional responses of cultured human endothelial cells. Hypertens. Res. 2008;31:345–351. doi: 10.1291/hypres.31.345. [DOI] [PubMed] [Google Scholar]
  • 176.Souza R., Humbert M., Sztrymf B., Jais X., Yaici A., Le Pavec J., et al. Pulmonary arterial hypertension associated with fenfluramine exposure: report of 109 cases. Eur. Respir. J. 2008;31:343–348. doi: 10.1183/09031936.00104807. [DOI] [PubMed] [Google Scholar]
  • 177.Spiller H.A. Rethinking mercury: the role of selenium in the pathophysiology of mercury toxicity. Clin. Toxicol. 2018;56:313–326. doi: 10.1080/15563650.2017.1400555. [DOI] [PubMed] [Google Scholar]
  • 178.Sprague A.H., Khalil R.A. Inflammatory cytokines in vascular dysfunction and vascular disease. Biochem. Pharmacol. 2009;78:539–552. doi: 10.1016/j.bcp.2009.04.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Staessen J., Bulpitt C.J., Fagard R., Lijnen P., Amery A. The influence of menopause on blood pressure. J. Hum. Hypertens. 1989;3:427–433. [PubMed] [Google Scholar]
  • 180.Staessen J.A., Byttebier G., Buntinx F., Celis H., O'Brien E.T., Fagard R. Antihypertensive treatment based on conventional or ambulatory blood pressure measurement. A randomized controlled trial. Ambulatory Blood Pressure Monitoring and Treatment of Hypertension Investigators. JAMA. 1997;278:1065–1072. [PubMed] [Google Scholar]
  • 181.Stott D.J., Bowman A. Blood pressure and aging. J. Hum. Hypertens. 2000;14:771–772. doi: 10.1038/sj.jhh.1001108. [DOI] [PubMed] [Google Scholar]
  • 182.Strajhar P., Tonoli D., Jeanneret F., Imhof R.M., Malagnino V., Patt M., Kratschmar D.V., Boccard J., Rudaz S., Odermatt A. Steroid profiling in H295R cells to identify chemicals potentially disrupting the production of adrenal steroids. Toxicology. 2017;381:51–63. doi: 10.1016/j.tox.2017.02.010. [DOI] [PubMed] [Google Scholar]
  • 183.Sysol J.R., Machado R.F. Classification and pathophysiology of pulmonary hypertension. Continuing Cardiol. Educ. 2018;4:2–12. [Google Scholar]
  • 184.Tang X., Mohuczy D., Zhang Y.C., Kimura B., Galli S.M., Phillips M.I. Intravenous angiotensinogen antisense in AAV-based vector decreases hypertension. Am. J. Physiol. 1999;277:H2392–H2399. doi: 10.1152/ajpheart.1999.277.6.H2392. [DOI] [PubMed] [Google Scholar]
  • 185.Tanimoto K., Sugiyama F., Goto Y., Ishida J., Takimoto E., Yagami K., Fukamizu A., Murakami K. Angiotensinogen-deficient mice with hypotension. J. Biol. Chem. 1994;269:31334–31337. [PubMed] [Google Scholar]
  • 186.Tellez-Plaza M., Navas-Acien A., Crainiceanu C.M., Guallar E. Cadmium exposure and hypertension in the 1999-2004 National Health and Nutrition Examination Survey (NHANES) Environ. Health Perspect. 2008;116:51–56. doi: 10.1289/ehp.10764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Tetti M., Monticone S., Burrello J., Matarazzo P., Veglio F., Pasini B., Jeunemaitre X., Mulatero P. Liddle Syndrome: review of the literature and description of a new case. Int. J. Mol. Sci. 2018:19. doi: 10.3390/ijms19030812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Thakker K.D. An overview of health risks and benefits of alcohol consumption. Alcohol. Clin. Exp. Res. 1998;22:285S–298S. doi: 10.1097/00000374-199807001-00003. [DOI] [PubMed] [Google Scholar]
  • 189.Thogersen A.M., Jansson J.H., Boman K., Nilsson T.K., Weinehall L., Huhtasaari F., Hallmans G. High plasminogen activator inhibitor and tissue plasminogen activator levels in plasma precede a first acute myocardial infarction in both men and women: evidence for the fibrinolytic system as an independent primary risk factor. Circulation. 1998;98:2241–2247. doi: 10.1161/01.cir.98.21.2241. [DOI] [PubMed] [Google Scholar]
  • 190.Thomas L.B., Popper H., Berk P.D., Selikoff I., Falk H. Vinyl-chloride-induced liver disease. From idiopathic portal hypertension (Banti’s syndrome) to Angiosarcomas. New Engl. J. Med. 1975;292:17–22. doi: 10.1056/NEJM197501022920104. [DOI] [PubMed] [Google Scholar]
  • 191.Thompson P.A., Khatami M., Baglole C.J., Sun J., Harris S.A., Moon E.Y., Al-Mulla F., Al-Temaimi R., Brown D.G., Colacci A., Mondello C., Raju J., Ryan E.P., Woodrick J., Scovassi A.I., Singh N., Vaccari M., Roy R., Forte S., Memeo L., Salem H.K., Amedei A., Hamid R.A., Lowe L., Guarnieri T., Bisson W.H. Environmental immune disruptors, inflammation and cancer risk. Carcinogenesis. 2015;36(1):S232–S253. doi: 10.1093/carcin/bgv038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Tseng C.H., Chong C.K., Chen C.J., Tai T.Y. Lipid profile and peripheral vascular disease in arseniasis-hyperendemic villages in Taiwan. Angiology. 1997;48:321–335. doi: 10.1177/000331979704800405. [DOI] [PubMed] [Google Scholar]
  • 193.van den Dungen M.W., Rijk J.C., Kampman E., Steegenga W.T., Murk A.J. Steroid hormone related effects of marine persistent organic pollutants in human H295R adrenocortical carcinoma cells. Toxicol. in Vitro. 2015;29:769–778. doi: 10.1016/j.tiv.2015.03.002. [DOI] [PubMed] [Google Scholar]
  • 194.Vander A.J. Chronic effects of lead on the renin-angiotensin system. Environ. Health Perspect. 1988;78:77–83. doi: 10.1289/ehp.887877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Vaziri N.D. Mechanisms of lead-induced hypertension and cardiovascular disease. Am. J. Physiol. Heart Circ. Physiol. 2008;295:H454–H465. doi: 10.1152/ajpheart.00158.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Vaziri N.D., Sica D.A. Lead-induced hypertension: role of oxidative stress. Curr. Hypertens. Rep. 2004;6:314–320. doi: 10.1007/s11906-004-0027-3. [DOI] [PubMed] [Google Scholar]
  • 197.Vehaskari V.M. Heritable forms of hypertension. Pediatr. Nephrol. 2009;24:1929–1937. doi: 10.1007/s00467-007-0537-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Veith A., Moorthy B. Role of cytochrome P450s in the generation and metabolism of reactive oxygen species. Curr. Opin. Toxicol. 2018;7:44–51. doi: 10.1016/j.cotox.2017.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Vogel C.F., Sciullo E., Wong P., Kuzmicky P., Kado N., Matsumura F. Induction of proinflammatory cytokines and C-reactive protein in human macrophage cell line U937 exposed to air pollution particulates. Environ. Health Perspect. 2005;113:1536–1541. doi: 10.1289/ehp.8094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Wang C.H., Chen C.L., Hsiao C.K., Chiang F.T., Hsu L.I., Chiou H.Y., Hsueh Y.M., Wu M.M., Chen C.J. Increased risk of QT prolongation associated with atherosclerotic diseases in arseniasis-endemic area in southwestern coast of Taiwan. Toxicol. Appl. Pharmacol. 2009;239:320–324. doi: 10.1016/j.taap.2009.06.017. [DOI] [PubMed] [Google Scholar]
  • 201.Wang C.H., Jeng J.S., Yip P.K., Chen C.L., Hsu L.I., Hsueh Y.M., Chiou H.Y., Wu M.M., Chen C.J. Biological gradient between long-term arsenic exposure and carotid atherosclerosis. Circulation. 2002;105:1804–1809. doi: 10.1161/01.cir.0000015862.64816.b2. [DOI] [PubMed] [Google Scholar]
  • 202.Watt G.C., Harrap S.B., Foy C.J., Holton D.W., Edwards H.V., Davidson H.R., Connor J.M., Lever A.F., Fraser R. Abnormalities of glucocorticoid metabolism and the renin-angiotensin system: a four-corners approach to the identification of genetic determinants of blood pressure. J. Hypertens. 1992;10:473–482. doi: 10.1097/00004872-199205000-00011. [DOI] [PubMed] [Google Scholar]
  • 203.Watts S.W., Chai S., Webb R.C. Lead acetate-induced contraction in rabbit mesenteric artery: interaction with calcium and protein kinase C. Toxicology. 1995;99:55–65. doi: 10.1016/0300-483x(94)03003-k. [DOI] [PubMed] [Google Scholar]
  • 204.Wauthier V., Verbeeck R.K., Calderon P.B. The effect of ageing on cytochrome p450 enzymes: consequences for drug biotransformation in the elderly. Curr. Med. Chem. 2007;14:745–757. doi: 10.2174/092986707780090981. [DOI] [PubMed] [Google Scholar]
  • 205.Wiggers G.A., Pecanha F.M., Briones A.M., Perez-Giron J.V., Miguel M., Vassallo D.V., Cachofeiro V., Alonso M.J., Salaices M. Low mercury concentrations cause oxidative stress and endothelial dysfunction in conductance and resistance arteries. Am. J. Physiol. Heart Circ. Physiol. 2008;295:H1033–H1043. doi: 10.1152/ajpheart.00430.2008. [DOI] [PubMed] [Google Scholar]
  • 206.Williams B., Mancia G., Spiering W., Agabiti Rosei E., Azizi M., Burnier M., Clement D.L., Coca A., de Simone G., Dominiczak A., Kahan T., Mahfoud F., Redon J., Ruilope L., Zanchetti A., Kerins M., Kjeldsen S.E., Kreutz R., Laurent S., Lip G.Y.H., McManus R., Narkiewicz K., Ruschitzka F., Schmieder R.E., Shlyakhto E., Tsioufis C., Aboyans V., Desormais I., ESC Scientific Document Group 2018 ESC/ESH Guidelines for the management of arterial hypertension. Eur. Heart J. 2018;39:3021–3104. doi: 10.1093/eurheartj/ehy339. [DOI] [PubMed] [Google Scholar]
  • 207.World Health Organization, "WHO air quality guidelines for Europe, 2nd edition, 2000 (CD ROM version)", 〈https://www.euro.who.int/en/health-topics/environment-and-health/air-quality/publications/pre2009/who-air-quality-guidelines-for-europe,-2nd-edition,-2000-cd-rom-version〉 (accessed July 17, 2021).
  • 208.Woolbright B.L., Jaeschke H. Xenobiotic and endobiotic mediated interactions between the cytochrome P450 system and the inflammatory response in the liver. Adv. Pharmacol. 2015;74:131–161. doi: 10.1016/bs.apha.2015.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Wu F., Jasmine F., Kibriya M.G., Liu M., Wojcik O., Parvez F., Rahaman R., Roy S., Paul-Brutus R., Segers S., Slavkovich V., Islam T., Levy D., Mey J.L., van Geen A., Graziano J.H., Ahsan H., Chen Y. Association between arsenic exposure from drinking water and plasma levels of cardiovascular markers. Am. J. Epidemiol. 2012;175:1252–1261. doi: 10.1093/aje/kwr464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Wu F., Molinaro P., Chen Y. Arsenic exposure and subclinical endpoints of cardiovascular diseases. Curr. Environ. Health Rep. 2014;1:148–162. doi: 10.1007/s40572-014-0011-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Xia Y., Niu Y., Cai J., Lin Z., Liu C., Li H., Chen C., Song W., Zhao Z., Chen R., Kan H. Effects of personal short-term exposure to ambient ozone on blood pressure and vascular endothelial function: a mechanistic study based on DNA methylation and metabolomics. Environ. Sci. Technol. 2018;52:12774–12782. doi: 10.1021/acs.est.8b03044. [DOI] [PubMed] [Google Scholar]
  • 212.Xiao L., Zhang Z., Luo X. Roles of xenobiotic receptors in vascular pathophysiology. Circ. J. 2014;78:1520–1530. doi: 10.1253/circj.cj-14-0343. [DOI] [PubMed] [Google Scholar]
  • 213.Yang D., Yang Q., Fu N., Li S., Han B., Liu Y., Tang Y., Guo X., Lv Z., Zhang Z. Hexavalent chromium induced heart dysfunction via Sesn2-mediated impairment of mitochondrial function and energy supply. Chemosphere. 2021;264 doi: 10.1016/j.chemosphere.2020.128547. [DOI] [PubMed] [Google Scholar]
  • 214.Yi T., Wang J., Zhu K., Tang Y., Huang S., Shui X., Ding Y., Chen C., Lei W. Aryl hydrocarbon receptor: a new player of pathogenesis and therapy in cardiovascular diseases. BioMed. Res. Int. 2018;2018 doi: 10.1155/2018/6058784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Yildiz A., Karaca M., Biceroglu S., Nalbantcilar M.T., Coskun U., Arik F., Aliyev F., Yiginer O., Turkoglu C. Effect of chronic arsenic exposure from drinking waters on the QT interval and transmural dispersion of repolarization. J. Int. Med. Res. 2008;36:471–478. doi: 10.1177/147323000803600311. [DOI] [PubMed] [Google Scholar]
  • 216.Zhu K., Meng Q., Zhang Z., Yi T., He Y., Zheng J., Lei W. Aryl hydrocarbon receptor pathway: role, regulation and intervention in atherosclerosis therapy (Review) Mol. Med. Rep. 2019;20:4763–4773. doi: 10.3892/mmr.2019.10748. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Toxicology Reports are provided here courtesy of Elsevier

RESOURCES