Abstract
Phthalates and bisphenols are high production volume chemicals that are used in the manufacturing of consumer and medical products. Given the ubiquity of bisphenol and phthalate chemicals in the environment, biomonitoring studies routinely detect these chemicals in 75–90% of the general population. Accumulating evidence suggests that such chemical exposures may influence human health outcomes, including cardiovascular health. These associations are particularly worrisome for sensitive populations, including fetal, infant and pediatric groups—with underdeveloped metabolic capabilities and developing organ systems. In the presented article, we aimed to review the literature on environmental and clinical exposures to bisphenols and phthalates, highlight experimental work that suggests that these chemicals may exert a negative influence on cardiovascular health, and emphasize areas of concern that relate to vulnerable pediatric groups. Gaps in our current knowledge are also discussed, so that future endeavors may resolve the relationship between chemical exposures and the impact on pediatric cardiovascular physiology.
Keywords: BPA, cardiotoxicity, DEHP, endocrine-disrupting chemical, environmental, heart
1 |. INTRODUCTION
Over a lifetime, individuals are exposed to a variety of environmental chemicals that can contribute to nonhereditary components of health and disease. Environmental toxicology research can provide an understanding of how these chemical exposures influence human health. Of all the chemical exposures that individuals experience, phthalate and bisphenol chemicals have emerged as two potential contributors to cardiovascular dysfunction (Gao & Wang, 2014; Han & Hong, 2016; Lind & Lind, 2012; Mariana, Feiteiro, Verde, & Cairrao, 2016; Posnack, 2014). Phthalates and bisphenols are high production volume chemicals that are widely used in the manufacturing of consumer and medical-grade plastics. Due to their broad use in both industrial and consumer products, >2 billion pounds of bisphenol-A (BPA) and di-2-ethylhexyl phthalate (DEHP) are produced globally each year (Halden, 2010; Shelby, 2008). BPA is a synthetic monomer that is commonly used as a building block in polycarbonate plastic products, an epoxy resin for food packaging (e.g., canned goods), or in thermal printing applications (e.g., receipt paper). BPA polymers can degrade under normal conditions of use, and this deterioration is increased when subjected to elevated temperatures or mechanical stress (Krishnan, Stathis, Permuth, Tokes, & Feldman, 1993). DEHP is a high molecular weight phthalate that is frequently employed as a plasticizer in polyvinyl chloride (PVC) products, including medical devices and food packaging. Given the rigidity of PVC materials, phthalate plasticizers impart flexibility and elasticity to PVC products by embedding in between polymer segments to disrupt complete polymerization (National Research Council (U.S.) Committee on the Health Risks of Phthalates, 2008). Since phthalate plasticizers are not covalently bound to the PVC matrix, these chemical additives are susceptible to leaching (Tereshchenko & Posnack, 2019). Given the ubiquity of bisphenol and phthalate chemicals in the environment, biomonitoring studies routinely detect these chemicals in 75–90% of the general population, likely from multiple routes of exposure (ATSDR, 2019; Calafat, Ye, Wong, Reidy, & Needham, 2008; Hoppin, Brock, Davis, & Baird, 2002; Kato et al., 2004; Koch, Drexler, & Angerer, 2003; Koch, Rossbach, Drexler, & Angerer, 2003; Silva et al., 2004; Vandenberg et al., 2010; Y. Wang, Zhu, & Kannan, 2019; Zota, Calafat, & Woodruff, 2014).
Despite the benefits of plastics, their relative abundance in both the environment and clinical setting has raised concerns pertaining to human health. BPA and DEHP are both classified as endocrine-disrupting chemicals (EDCs) due to their ability to interfere with hormone homeostasis (La Merrill et al., 2020). BPA is considered a xenoestrogen because of its interactions with estrogen receptors (Ben-Jonathan & Steinmetz, 1998; Krishnan et al., 1993), whereas DEHP is broadly considered an antiandrogen due to its inhibitory effects on androgen receptors (Borch, Metzdorff, Vinggaard, Brokken, & Dalgaard, 2006; Swan, 2008). Although, it is important to note that both chemicals exert biological effects through multiple mechanisms of action. Once inside the body, bisphenol and phthalate chemicals may interact with nuclear receptors, hormone receptors, transcription factors, ion channels and disrupt intracellular signaling pathways (Ben-Jonathan & Steinmetz, 1998; Ito, Kamijima, & Nakajima, 2019; Jaimes et al., 2019; La Merrill et al., 2020; MacKay & Abizaid, 2018; Mariana et al., 2016; Moriyama et al., 2002; Nadal et al., 2000; Rowdhwal & Chen, 2018; Singh & Li, 2012; Sohoni & Sumpter, 1998; Soriano et al., 2016; Wetherill et al., 2007; Ye et al., 2017). Many of these interactions can-result in acute and rapid consequences. Unsurprisingly, exposure to EDCs, including phthalates and bisphenols, is associated with a variety of adverse human health outcomes, including hypertension, myocardial infarction, angina, atherosclerosis, diminished heart rate variability, impaired neurodevelopment, and inflammatory conditions (Casals-Casas & Desvergne, 2011; Halden, 2010; Mallow & Fox, 2014; Mariana et al., 2016; Schug, Blawas, Gray, Heindel, & Lawler, 2015; Verstraete et al., 2016; Yi et al., 2010). These health associations are particularly worrisome for sensitive populations, including fetal, infant and pediatric groups (Beszterda & Frański, 2018; Braun & Hauser, 2011; Braun, Sathyanarayana, & Hauser, 2013). Young children have an underdeveloped metabolic system, which can prolong exposure to EDCs and secondary metabolites (Beszterda & Frański, 2018; Lu & Rosenbaum, 2014). Early life exposures can also precipitate lasting effects on sensitive organ systems that are still developing (Meeker, 2012). Further, critically ill neonatal and pediatric patients often undergo multiple medical interventions that employ plastic materials, which can increase their cumulative exposure to levels that are 10–10,000 fold higher than observed in healthy populations (ATSDR, 2019; Calafat et al., 2009; Calafat, Needham, Silva, & Lambert, 2004; FDA, 2002; Mallow & Fox, 2014). In response to these health concerns, the U.S. Food & Drug Administration has recommended minimizing phthalate exposure to newborn boys during high-risk medical procedures—but refrained from taking further action (FDA, 2002). To date, patients continue to be inundated (inadvertently) with plastic chemicals that can distribute to sensitive organs, including the heart (Chu et al., 1978; Hillman, Goodwin, & Sherman, 1975; Jarosova, Harazim, Suchy, Kratka, & Stancova, 2009; Kim et al., 2004; Oishi & Hiraga, 1982; Shin et al., 2004; VandeVoort et al., 2016). Cardiovascular and autonomic function are highly susceptible to xenobiotic toxicity, and as such, exogenous chemical exposures may contribute to adverse health outcomes (Mladěnka et al., 2018; Rahm, Lugenbiel, Schweizer, Katus, & Thomas, 2018).
In the presented article, we aimed to review the literature on environmental and/or clinical exposures to bisphenols and phthalates, with a focus on potential cardiovascular outcomes and sensitive pediatric populations (Figure 1). We summarize (a) biomonitoring studies that have quantified environmental and clinical exposures, (b) experimental evidence of adverse cardiovascular effects and potential mechanisms, (c) epidemiological associations between chemical exposure and cardiovascular health, and (d) highlight the need for future studies to discern the risk to human health.
2 |. BISPHENOL-A
2.1 |. Environmental and clinical exposure to BPA
Bisphenols, including BPA, are used in the manufacturing of polycarbonate plastics and epoxy resins, including food and drink containers, water pipes, and thermal printed paper products (Vandenberg, Hauser, Marcus, Olea, & Welshons, 2007). Due to its ubiquitous use in consumer and medical products, human exposure to BPA is nearly continuous and biomonitoring studies routinely detect BPA in >90% of the population, including both children and adults (Calafat et al., 2005, 2009, 2008; Vandenberg et al., 2010; Woodruff, Zota, & Schwartz, 2011) (Supporting Information Table 1). Since BPA is a nonpersistent chemical, urinary samples are often preferred in biomonitoring studies as the chemical concentration is higher (as compared to plasma or serum) (Koch & Calafat, 2009), and there is concern for environmental BPA contamination that can lead to erroneous results when blood samples are used for these measurements (Teeguarden et al., 2011; Teeguarden, Twaddle, Churchwell, & Doerge, 2016). Although some investigators note that assay contamination is well controlled in most laboratories, and argue that human BPA exposure is significantly understated (Taylor et al., 2011; vom Saal & Welshons, 2014). Accordingly, serum BPA levels remain actively debated, with reports ranging widely (Aris, 2014; Y. J. Lee et al., 2008; Padmanabhan et al., 2008; Schönfelder et al., 2002; Vandenberg et al., 2010; T. Zhang, Sun, & Kannan, 2013). For urinary samples, population-based studies have reported BPA concentrations that range from undetectable to 9.3 μg/L in relatively healthy adults and children (Vandenberg et al., 2007, 2010). One study suggested that high urinary and plasma concentrations may be partly explained by sublingual BPA absorption, which bypasses first-pass metabolism, while another group has refuted this suggestion using different sites of blood collection (Gayrard et al., 2013; Teeguarden et al., 2015).
It is important to note that the degree of BPA exposure can range dramatically depending on socioeconomic factors, lifestyle choices, medical status, and route of exposure. Oral exposure to BPA is considered the most prevalent, with exposure levels linked to dietary choices, including use of canned beverages with BPA liners (Bae & Hong, 2015; Cao, Corriveau, & Popovic, 2010). Skin absorption and/or inhalation can be associated with higher levels of circulating unconjugated or biologically active BPA that may persist for longer periods of time, as compared to ingestion which is subject to first-pass metabolism (Mattison, Karyakina, Goodman, & Lakind, 2014). In plasma, reports suggest that nearly 90% of BPA is bound to serum proteins and is considered inactive (Teeguarden, Waechter Jr., Clewell III, Covington, & Barton, 2005). Studies have also shown that BPA can penetrate and accumulate in the human placenta at higher levels than detected in maternal plasma, as placental enzymes may deconjugate BPA to its active form (Nishikawa et al., 2010; Schönfelder et al., 2002). Further, studies suggest that unconjugated or active BPA can readily pass across the placenta into fetal circulation (Gerona et al., 2016), and the expression of metabolizing enzymes responsible for BPA glucuronidation to its inactive form are reduced during the prenatal period (R. N. Hines, 2008). Additional studies are needed to fully understand the extent of fetal and infant exposure to bisphenols, and the potential downstream effects on pediatric health.
While environmental exposure to BPA is continuous at a relatively low dose (Koch & Calafat, 2009; Vandenberg et al., 2007, 2010)—clinical (Calafat et al., 2009; Duty et al., 2013; Gaynor et al., 2018) and occupational (C. J. Hines et al., 2018; Ribeiro, Ladeira, & Viegas, 2017) environments can result in higher BPA exposures (Table 1) (Calafat et al., 2009; Duty et al., 2013; Gaynor et al., 2018; Huygh et al., 2015). BPA has been detected in 60% of neonatal intensive care unit (ICU) supplies, and experimental studies have identified estrogenic activity in >25% of extracts from these items (Iribarne-Durán et al., 2019). Clinical exposure can result in heightened and/or prolonged exposure to BPA, particularly in neonatal and pediatric patients with an underdeveloped metabolic system (Calafat et al., 2009). Premature infants in the NICU setting had urinary BPA levels that ranged from 1.6–946 μg/L (Calafat et al., 2009). Clinical BPA exposure is associated with high-intensity medical treatment, as premature infants requiring multiple (≥4) medical devices had higher urinary concentrations (36.6 μg/L) compared with those who required fewer devices (0–3 devices, 13.9 μg/L) (Duty et al., 2013). Collectively, premature infants undergoing intensive care had BPA exposures that were 16–32-fold higher than infants and children in the general population (Calafat et al., 2009; Duty et al., 2013). For comparison, an average 2–2.6 μg/L BPA (females, males) was detected in healthy infants not requiring invasive medical interventions (Mendonca, Hauser, Calafat, Arbuckle, & Duty, 2014). Considering the numerous sources of environmental BPA exposure during pregnancy (Joe M. Braun et al., 2011; Gerona et al., 2016), additional studies are warranted to understand prenatal exposure and risk (C. J. Patel et al., 2014). Similar results have been reported in adult intensive care patients undergoing invasive procedures including hemofiltration and circulatory support, which resulted in elevated urinary (6–680 μg/L) and serum BPA concentrations (2.6–255 μg/L) (Huygh et al., 2015). The latter underscores the importance of investigating inadvertent hospital-based chemical exposures, and the potential contribution to patient health outcomes.
TABLE 1.
Description | Biological sample | Mean concentration, μg/L (range) | Reference |
---|---|---|---|
Preoperative infants (5.4 ± 3.8 days) | Urine | 9.8 (7.3–13.3) | Gaynor et al., 2018 |
Postoperative infants (5.4 ± 3.8 days) | 13.9 (10.8–18.0) | ||
NICU infants (<44 weeks) | 30.3 (1.6–946) | Calafat et al., 2009 | |
Breastfed NICU infants (27–40 weeks) | 23.3 (12.8–40.7) (median) | Duty et al., 2013 | |
Formula-fed NICU infants (27–40 weeks) | 13.1 (9.2–40.6) (median) | ||
NICU infants using >4 medical devices | 36.6 (17.2–47.3) (median) | ||
NICU infants using <4 medical devices | 13.9 (9.2–35.1) (median) | ||
Adult, ICU | 1.3 (<LOD—203.3) | Huygh et al., 2015 | |
Adult, venovenous hemofiltration | 3.9 (<LOD—17.8) | ||
Adult, ECMO | 11.2 (1.6–17) |
The Environmental Protection Agency’s reference dose for BPA is 0.05 mg kg−1day−1—which signifies an “estimate of a daily exposure to the human population (including sensitive subgroups) that is likely to be without an appreciable risk of deleterious effects during a lifetime” (U.S. Environmental Protection Agency, 1988). Although, it is important to note that this value was last updated in 1988—and additional experimental and epidemiological studies have highlighted a nonmonotonic dose response for BPA (Gore et al., 2015; Liang, Gao, Chen, Hong, & Wang, 2014; Vandenberg, 2014). Similar to other natural hormones, a nonmontonic dose response has been observed in 20–30% of experimental BPA studies (Vandenberg, 2014). These findings suggest that risk assessments are needed for both “low” and “high” dose exposures, given the potential for a U-shaped dose response curve. Further, an increasing number of studies have shown negative health outcomes associated with BPA exposure, manufacturers have begun to replace BPA with structural analogs, including bisphenol-F (BPF) and bisphenol-S (BPS) (Moon, 2019; Moreman et al., 2017; Rochester & Bolden, 2015). Reference doses are not yet available for these structural analogs, as these other bisphenols have not been studied as thoroughly. Biomonitoring studies have compared urine concentrations of these three chemicals, and found that BPA remains the most prevalent, followed by BPF and BPS (Lehmler, Liu, Gadogbe, & Bao, 2018; Y. X. Wang, Liu, et al., 2019). Additional studies are needed to understand the degree of cumulative bisphenol exposure (e.g., BPA, BPS, BPF) and the potential risk to maternal, fetal and pediatric health across a range of concentrations (Figure 1).
2.2 |. Effects of BPA on intercellular junctions
Cardiomyocytes are connected to neighboring myocytes via intercalated disks, a highly organized structure comprised of mechanical (desmosomes, adherens, and tight junctions) and electrical connections (gap junctions) (Vermij, Abriel, & van Veen, 2017). Intercalated disks integrate electrical and mechanical function across the myocardial tissue, and disruption of intercalated disk protein expression or their localization has been associated with cardiac arrhythmias (Kleber & Saffitz, 2014; Rampazzo, Calore, van Hengel, & van Roy, 2014). Accordingly, environmental chemicals that disrupt cell–cell communication can have adverse effects on intercellular signaling and stability. To the best of our knowledge, experimental work addressing the potential effects of BPA-treatment on intercalated disk and sarcomeric proteins in cardiac preparations is limited (Belcher, Gear, & Kendig, 2015; Chapalamadugu, VandeVoort, Settles, Robison, & Murdoch, 2014). However, a number of studies have examined key protein targets in alternative cell types. As an example, experimental studies often employ a “bisphenol-a model” to induce tight junction and gap junction disassembly at the blood-testis barrier and/or in Sertoli cell lines (Li, Mruk, Lee, & Cheng, 2009, 2010). Using a supraphysiological concentration of BPA (200 μM), Li et al. reported that BPA exposure caused a transient disruption of tight junction permeability and reduced the expression of key proteins in tight junctions (occludin, junctional adhesion molecule A), adherens junctions (N-cadherin, β-catenin, α-catenin), desmosomal (desmoglein-2), and gap junctions (connexin-43) using Sertoli cells (Li et al., 2009). Similarly, Fiorini et al. noted that the expression of three connexin-43 isoforms were reduced in BPA-treated Sertoli cells, with a lesser effect on occludin and N-cadherin levels (Fiorini, Tilloy-Ellul, Chevalier, Charuel, & Pointis, 2004). Lee et al. also noted a reduction in connexin-43 expression in epithelium cells, which was mediated via nongenomic mechanisms (I.-K. Lee & Rhee, 2007). in vivo BPA exposure (2.4 mg kg−1 day−1) has also been shown to reduce connexin-43 expression in neonatal male rats at two time points tested (postnatal Days 45 and 90), as well as increase the expression of N-cadherin and zonula occludens-1 in the testes (Salian, Doshi, & Vanage, 2009). In cardiac tissue, the formation of mechanical junctions is considered a prerequisite for gap junction intercellular connections in cardiomyocytes (Rampazzo et al., 2014), and a similar disruption of key structural proteins could impair electrical conduction in the heart. Of interest, postoperative intestinal barrier dysfunction is commonly observed in children with congenital heart disease requiring cardiopulmonary bypass (Typpo et al., 2015). Cardiopulmonary bypass is known to precipitate an inflammatory response that can contribute to intestinal barrier dysfunction, yet, it is still unknown whether BPA exposure from tubing circuits may contribute to these effects.
2.3 |. Effects of BPA on ion channels and electrophysiology
Ion channels play an important role in the generation of the cardiac action potential and electrical propagation, wherein disruption of ion channel localization, expression or function can precipitate cardiac dysrhythmias (Grant, 2009). Electrophysiology studies performed in HEK cells showed that BPA blocks hNav1.5 (IC50 = 25 μM), the fast voltage-gated sodium channel subtype responsible for the action potential upstroke (Supporting Information Table 2) (O’Reilly et al., 2012). Results suggest that BPA inhibits sodium current by interacting directly with a binding site that is shared by local anesthetics, including mexiletine. In cardiac tissue, such an effect on sodium channel current is expected to reduce the rate of depolarization and slow cardiac conduction velocity. BPA exposure has also been shown to inhibit sodium channel current in isolated mouse dorsal root ganglion neurons—including both TTX-sensitive (IC50 = 40 μM) and TTX-resistant (IC50 = 70 μM) sodium currents (Q. Wang et al., 2011). BPA-inhibition was rapid, but reversible, with a dose–response relationship. Notably, these effects were blocked by protein kinase C (PKC) and protein kinase A (PKA) inhibitors, suggesting that BPA may act through a protein kinase-dependent pathway. This result is in agreement with Gao et al., which found that BPA-induced effects on ryanodine receptors were mediated through an estrogen receptor beta (ERβ) and PKA signaling pathway (Gao, Liang, Chen, & Wang, 2013).
BPA exposure has also been shown to inhibit calcium channel current, which is important for nodal cell depolarization, atrioventricular conduction, and the plateau phase of the cardiac action potential (phase 2). Using an HEK cell line, Michaela et al. demonstrated that BPA inhibits calcium current via T-type channels in a concentration-dependent manner with an IC50 = 6–33 μM, depending on channel subtype (Michaela, Mária, Silvia, Ľubica, & L’ubica, 2014). Nanomolar concentrations did not affect voltage dependence or channel gating kinetics, but micro-molar concentrations accelerated the kinetics of current decay and inhibited calcium current amplitude. These findings suggest that BPA modifies channel gating and acts as a calcium channel antagonist at higher concentrations. In a separate study, Deutschmann et al. reported that BPA was a potent blocker of multiple voltage-gated calcium channels (Deutschmann, Hans, Meyer, Häberlein, & Swandulla, 2013). BPA rapidly and reversibly inhibited calcium current through native L-, N-, P/Q-, and T-type channels in rat endocrine cells, mouse dorsal root ganglion neurons or cardiac myocytes, and recombinant human R-type calcium channels expressed in HEK cells. In the latter, BPA was found to interact with the extracellular part of the channel protein without dependence on intracellular signaling pathways or voltage, and without affecting channel gating. Since channel gating was unaffected, this suggests a second mechanism by which BPA can directly interact with calcium channels externally, in its resting state. Similar inhibitory effects on L-type calcium channels have also been observed in rat aortic preparations (Feiteiro, Mariana, Glória, & Cairrao, 2018).
Estrogenic chemicals, like BPA, have also been shown to affect other ion channels, but the results are less conclusive (Soriano et al., 2016). Estrogenic agents can exert fast-acting stimulatory or inhibitory effects on big conductance calcium, L-type calcium, and voltage-activated potassium channels (Kow & Pfaff, 2016). As an example, BPA-treatment was shown to alter the gene expression of voltage-gated sodium, voltage-gated potassium, inward rectifier potassium channels and solute carrier transporters in cardiac tissue in a sex-specific manner (Belcher et al., 2015). The latter is attributed to the estrogenic effects of BPA, which can differ in males versus females due to disparate estrogen-receptor profiles (Hutson et al., 2019; Pugach, Blenck, Dragavon, Langer, & Leinwand, 2016). Asano et al. also reported a decrease in voltage-gated calcium and potassium channel activity in coronary smooth muscle cells treated with BPA (Asano, Tune, & Dick, 2010). Although, to the best of our knowledge, the direct effects of BPA on ion channel currents have not been measured in cardiomyocyte preparations.
The inhibitory effects of BPA on sodium and calcium channel currents (Soriano et al., 2016) can result in notable changes in cardiac electrophysiology, which has been observed in both ex vivo and in vivo experimental models. BPA-exposure can result in sinus bradycardia and delayed electrical conduction in rodent models, including prolonged P-wave duration, PR interval and QRS duration (Belcher et al., 2015; B. B. Patel, Raad, Sebag, & Chalifour, 2015; Posnack et al., 2014; Valokola et al., 2019). Patel et al. noted an exaggerated effect on electrical conduction in female BPA-treated animals in response to catecholamine challenge (B. B. Patel, Raad, et al., 2015). In comparison, Belcher et al. observed heart rate slowing in BPA-treated male mice only, but noted exaggerated heart rate slowing in BPA-treated female mice in response to phenylephrine (Belcher et al., 2015). Using an ex vivo isolated heart model, we observed conduction slowing in response to acute BPA exposure that followed a dose response relationship (Posnack et al., 2014). In addition to BPA-induced effects on calcium current, which can directly impair cardiac automaticity and electrical conduction, Belcher et al. proposed that BPA may also slow heart rate by altering autonomic regulation (Belcher et al., 2015). The latter was supported by differential mRNA expression of beta-adrenergic receptors in BPA-treated animals, and may also explain increased sensitivity to beta-adrenergic agonists. Using isolated ventricular myocytes from female mice, Yan et al. reported that BPA rapidly induced arrhythmogenic triggered activities, which were exacerbated in the presence of isoproterenol (beta adrenergic agonist) and estradiol (Yan et al., 2011). The authors demonstrate that these triggered activities may be calcium mediated, as BPA altered protein phosphorylation to increase the likelihood of calcium leak from the sarcoplasmic reticulum. Notably, ryanodine inhibition suppressed spontaneous after-contractions that were induced by BPA or BPA + estradiol. Ultimately, there is evidence for BPA-induced effects on cardiac electrophysiology, but additional studies are needed to understand remaining questions about sex-specificity, dose–response relationships, direct versus intracellular signaling mechanisms, and the role of BPA exposure on developing hearts.
2.4 |. Effects of BPA on calcium handling
As demonstrated in the described electrophysiology studies, a commonly reported effect of BPA-treatment is a disruption in calcium signaling (Supporting Information Table 3). Following acute BPA exposure, our laboratory reported alterations in intracellular calcium handling in neonatal rat cardiomyocytes (Ramadan et al., 2018), which can result in reduced cardiac contractility and left ventricular developed pressure in isolated, whole rat heart preparations (Posnack, Brooks, et al., 2015). The observed effects were rapid, and included smaller calcium transient amplitudes, prolonged calcium transient duration time, and an increased propensity for calcium amplitude alter-nans (Ramadan et al., 2018). Notably, the observed effects on calcium were largely reversible, similar to reports by others (Deutschmann et al., 2013), suggesting that these effects are mediated by either direct interactions with calcium channels and/or rapid posttranslational modifications of calcium-handling proteins (Gao et al., 2013).
Studies suggest that the effects of BPA on (adult) cardiomyocyte calcium handling are female-specific, due to BPA’s interaction with estrogen receptors. Rapid exposure to low-dose BPA (1 nM) increased the incidence of ventricular arrhythmias following ischemia–reperfusion injury in female rat hearts, but these effects were not observed in male cardiac preparations (Yan et al., 2013). As previously noted, physiologically-relevant BPA concentrations can promote calcium-leak from the sarcoplasmic reticulum and increase the propensity of triggered activity in adult cardiomyocytes isolated from female rats (Yan et al., 2011). The pro-arrhythmic effects of BPA were negated when cardiomyocytes were pretreated with an estrogen receptor (ER) antagonist, or when applied to cardiac preparations from an ERβ knockout model. Follow-up studies indicated that BPA-treatment exerts monotonic effects via ERβ signaling, but collectively the disruption on multiple intracellular calcium handling targets can result in a global nonmonotonic dose–response curve (Liang et al., 2014). Mechanistic studies revealed that BPA exposure alters intracellular signaling pathways, namely through PKA and calcium/calmodulin-dependent protein kinase II (Gao et al., 2013). In cardiac preparations, BPA exposure can alter the phosphorylation of ryanodine receptors and phospholamban, key regulators of calcium release and reuptake into the sarcoplasmic reticulum. In mouse hearts, BPA exposure was also shown to alter the expression of the sarcoplasmic reticulum calcium ATPase (SERCA) with decreased protein expression in both males and females, plus reduced phospholamban and sodium/calcium exchanger expression in females (B. B. Patel, Raad, Sebag, & Chalifour, 2013). It is important to note that BPA-induced cardiac effects may be mediated by ER and PKA signaling mechanisms, particularly at low exposure levels, whereas direct inhibitory effects on cardiac ion channels may occur at higher concentrations.
The combined effects of BPA on ion channels and calcium handling are particularly concerning for pediatric populations—as the immature heart can be sensitive to toxins, electrolyte disturbances, and alterations in calcium handling. Immature myocytes have rudimentary t-tubules, inefficient excitation–contraction coupling—with a greater dependence on sarcolemmal calcium flux and diffusion (Louch, Koivumäki, & Tavi, 2015). To date, the downstream effects of BPA on post-translational modifications of calcium handling proteins (e.g., phospholamban, ryanodine) or antagonism of T-type or L-type calcium in immature cardiomyocytes is largely unknown. Future studies are warranted to understand the role of BPA exposure in developing cardiomyocytes—and also to investigate potential mitigation strategies. Indeed, recent reports have highlighted progesterone pretreatment as a protective measure to block the effects of BPA on phospholamban and prevent BPA-induced arrhythmogenesis in female (adult) cardiomyocytes (Ma, Hong, & Wang, 2017).
2.5 |. Effects of BPA on inflammation and remodeling
Pathological remodeling refers to changes in the heart that occur in response to injury, and may include cardiomyocyte hypertrophy, fibrosis, and changes in the extracellular matrix (Cohn, Ferrari, Sharpe,, & Remodeling, 2000). Recent experimental studies suggest that chronic BPA exposure can also precipitate cardiovascular dysfunction by promoting cardiac remodeling and inflammation (Supporting Information Table 4). In one study, which examined pregnant mice and their progeny during chronic BPA exposure, sex-specific phenotypic changes included increased collagen extracellular matrix, increased fibrosis and modest cardiac remodeling (Belcher et al., 2015). In a follow-up study, as part of the Consortium Linking Academic and Regulatory Insights on BPA Toxicity (CLARITY-BPA) initiative, pregnant mice and offspring were chronically exposed to a full range of BPA concentrations (2.5–25,000 μg kg−1 day−1) (Gear, Kendziorski, & Belcher, 2017). BPA-exposed female offspring were at increased risk for cardiomyopathy, and both male and female offspring were prone to myocardial degeneration with signs of inflammatory cell infiltration. Rasdi et al. also reported an increase in maternal and fetal cardiac fibrosis following intrauterine BPA exposure (Rasdi et al., 2020). At higher BPA concentrations (50,000 μg kg−1 day−1), focal inflammation was observed in the cardiac tissue of pregnant mice, which coincided with slowing of cardiac electrical conduction and increased systolic and diastolic pressure relative to vehicle control (Valokola et al., 2019). The authors noted an attenuation of BPA-induced effects when an anti-inflammatory agent was administered. These long-term studies highlight the potential adverse outcomes that perinatal BPA exposure can have on both maternal and fetal health.
In other cases, BPA exposure was found to exacerbate cardiac injury. To mimic plastic chemical exposure during cardiac surgery, Shang et al. exposed mice to a cocktail of BPA and phthalates as they recovered from myocardial infarction (Shang et al., 2019). Rodents exposed to plastic chemicals had increased cardiac dilation, increased immune cell infiltration, and impaired recovery. In a separate study, the authors also found that bisphenol exposure (25 ng/ml BPA or BPS) significantly impaired cardiac healing following myocardial infarction, with an increased inflammatory response and adverse cardiac remodeling (B. B. Patel, Kasneci, et al., 2015). Of interest, the effects of BPA exposure were significantly attenuated in ERβ-knockout mice, which suggests that inflammation and cardiac remodeling may be attributed to the estrogenic activity of BPA (Kasneci et al., 2017). Additional injury studies have found that BPA exposure increases myocardial inflammation, fibrosis and elevated pro-inflammatory markers following viral infection (Bruno et al., 2019)—and that BPA exposure exacerbated the cardiotoxic effects of a chemotherapeutic agent, by increasing the production of pro-inflammatory interleukins in cardiomyoblasts (Quagliariello et al., 2019). These in vivo studies demonstrate how bisphenol exposure from medical procedures may impair patient recovery, particularly in populations susceptible to systemic inflammation from cardiopulmonary bypass or extracorporeal membrane oxygenation procedures.
2.6 |. Effects of BPA on blood pressure regulation and cardiovascular disease
Inflammation and oxidative stress can contribute to the development of hypertension, cardiac remodeling, and cardiovascular disease (Cohn et al., 2000; González, Valls, Brito, & Rodrigo, 2014). Saura et al. noted a dose-dependent increase in angiotensin II and oxidative stress markers in BPA-treated animals (Saura et al., 2014). Pharmacological manipulations revealed that BPA-treated animals had an increase in blood pressure, which was mediated by AngII/CAMKII uncoupling of endothelial nitric oxides synthase. Of interest, population-based studies have also noted an association between BPA exposure, inflammation, and oxidative stress markers (Kataria et al., 2017; Steffensen et al., 2020; Y. X. Wang, Liu, et al., 2019; Yang et al., 2009)—including elevated F2-isoprostane levels in children. Accordingly, inflammation and oxidative stress are potential mediators of elevated blood pressure and hypertension in adults and children with higher BPA exposures (Aekplakorn, Chailurkit, & Ongphiphadhanakul, 2015; Han & Hong, 2016; Jiang et al., 2020; A. Shankar & Teppala, 2012). In pregnancy, preeclamptic women were found to have significantly higher BPA concentrations in placental tissue, compared to normotensive pregnant women (Leclerc, Dubois, & Aris, 2014). In another study, Philips et al. reported an association between BPA exposure and subclinical changes in placental hemodynamics that warrant additional study, but did not find a significant link to maternal blood pressure (Philips et al., 2019). Cross-sectional studies have also reported a link between maternal BPA exposure and elevated diastolic blood pressure in children (Bae et al., 2017; Warembourg et al., 2019). Whereas, urinary BPA levels were associated with elevated blood pressure in children, with a greater effect in young boys compared with girls (Bae et al., 2017; Khalil et al., 2013; Sol et al., 2020). Accumulating evidence suggests that these associations are directly linked to BPA exposure, as demonstrated in a randomized trial (Bae & Hong, 2015). Specifically, Bae et al. found that urinary BPA concentrations increased by >1,600% in adult participants after consuming canned beverages containing BPA-liners. This increase in urinary BPA level was associated with an acute 4.5 mmHg increase in blood pressure. A follow-up study also found a positive association between urinary BPA concentrations and hypertension in elderly participants, and a negative association with heart rate variability (Bae, Kim, Lim, Park, & Hong, 2012). The latter may also be mechanistically linked to alterations in sympathetic tone and autonomic regulation, as previously described in experimental models (Belcher et al., 2015). BPA is largely considered an estrogen agonist, yet, recent work has highlighted that estrogenic activity can induce either vasodilation or vasoconstriction—depending on the product of nitric oxide synthase (nitric oxide vs. superoxide) (Fardoun et al., 2020). This is an area of research that requires additional attention—as BPA exposure has been shown to both increase (B. B. Patel, Raad, et al., 2015; Rasdi et al., 2020) and decrease (Belcher et al., 2015) blood pressure in rodent models. Although this discrepancy may be attributed to experimental design, which include different doses, treatment duration, and animals of differing age.
A few studies have also reported a relationship between BPA exposure and peripheral and/or coronary artery disease. Shankar et al. reported a positive association between BPA exposure and peripheral artery disease in a U.S. adult population (A. Shankar, Teppala, & Sabanayagam, 2012). Similarly, Melzer et al. reported that urinary BPA levels were associated with an increased risk of myocardial infarction and coronary artery disease (D. Melzer, Rice, Lewis, Henley, & Galloway, 2010). Follow-up studies highlighted that BPA exposure was also associated with severity of coronary artery stenosis and future risk of coronary artery disease in the adult population (Melzer, Osborne, et al., 2012; Melzer, Gates, et al., 2012). Although not directly applicable to pediatric populations, future longitudinal studies could investigate the link between early life exposure to BPA and cardiometabolic health later in life (Philips, Jaddoe, & Trasande, 2017).
3 |. DI-2-ETHYLHEXYL PHTHALATE
3.1 |. Environmental and clinical exposure to DEHP
Due to its low-cost of production and superior chemical properties for the manufacturing of flexible plastic products, DEHP remains the most widely used plasticizer in PVC products. Human exposure to DEHP can occur through contaminated drinking water, as widespread use can result in environmental contamination at waste sites, industrial locations, and/or landfills (ATSDR, 2019; Wowkonowicz & Kijeńska, 2017). More commonly, daily exposure to phthalates occurs through inhalation, ingestion, or dermal contact with phthalate-containing personal care products and/or food packaging (ATSDR, 2019; Guo & Kannan, 2013; Koch et al., 2013; Rowdhwal & Chen, 2018; Wormuth, Scheringer, Vollenweider, & Hungerbühler, 2006). Indeed, dietary DEHP intake closely correlates with the daily variation of DEHP metabolites in urine samples (Fromme et al., 2007) and higher urinary phthalate levels are associated with consuming food from plastic packaging (Giovanoulis et al., 2016). Accordingly, human biomonitoring studies have reported DEHP metabolites in 75–90% of biological samples collected from the general population, including both adults and children (Hoppin et al., 2002; Kato et al., 2004; Silva et al., 2004; Zota et al., 2014). Since DEHP is a nonpersistent chemical, biomonitoring studies often use urine samples for these measurements (Koch & Calafat, 2009). Although historical work focused on measuring the parent chemical (DEHP), more recent biomonitoring studies have quantified the sum of DEHP metabolites in urine or blood samples. DEHP is rapidly hydrolyzed to its monoester (MEHP, mono-ethylhexyl phthalate), which is further metabolized into secondary metabolites (MEHHP, 5OH-mono-2-ethylhexylphthalate; MEOHP, 5oxo-mono-ethylhexylphthalate; MECPP, 5carboxy-mono-2-ethylhexylphthalate[Kato et al., 2004]). This approach improves the accuracy of measurements, reduces the likelihood of environmental artifacts, and is biologically important—as studies suggest that DEHP’s metabolites are more toxic than the parent chemical (Koch & Calafat, 2009; Latini, 2005) (Supporting Information Table 5). Recently, Zhang et al. demonstrated that median urinary DEHP concentrations is 38.5 μg/L in children (<18 years old) and 43.3 μg/L in older adults (X. Zhang et al., 2020). In pregnant females, the sum of DEHP metabolites can be even higher, ranging from 78.1–93.7 μg/L (Nassan, Gunn, Hill, Williams, & Hauser, 2020; Watkins et al., 2017).
In addition to consumer goods, phthalates are frequently used in PVC-based medical products, given their low cost, flexibility, strength, and suitability for steam sterilization (FDA, 2002; Tickner, Schettler, Guidotti, McCally, & Rossi, 2001). In the final product, DEHP can contribute up to 40% of the finished weight of plastic medical products (e.g., intravenous fluid bags, blood storage bags), and up to 80% of the weight of flexible medical tubing (Halden, 2010; Jaeger & Rubin, 1973, 2010; Plonait, Nau, Maier, Wittfoht, & Obladen, 1993). Since phthalates are not covalently bound to PVC, these chemical additives are known to leach or migrate into surrounding solutions, including blood (Table 2) (Barry et al., 1989; D’Alessandro et al., 2019; Demirel et al., 2016; Gaynor et al., 2018; Green et al., 2005; Kaestner et al., 2020; Karle et al., 1997; Mallow & Fox, 2014; Münch et al., 2020; Peck et al., 1979; Rael et al., 2009; Sjöberg, Bondesson, Sedin, & Gustafsson, 1985; Sjoberg et al., 1985; Stroustrup et al., 2018; Su et al., 2012; Takatori et al., 2008; van der Meer et al., 2014). Moreover, DEHP leaching is exacerbated under conditions of prolonged use, mechanical stress, higher temperature and/or in the presence of lipid-containing fluids. As an example, Karle et al. estimated that extracorporeal membrane oxygenation circuits leach DEHP at a rate of 0.32–0.57 μg ml−1 hr−1 in nonheparinized circuits (Karle et al., 1997), which corresponds to an exposure level that is 20–70× higher than reported for other invasive procedures. In another simulation study, Loff et al. reported that DEHP leaching was temperature dependent, with 30% greater leaching observed during lipid infusion at warmer temperatures (33 vs. 27°C). The authors noted that a 24-hr lipid infusion would result in a 2-kg infant receiving a DEHP exposure of 6.5 mg/kg. Similarly, Takatori et al. reported that enteral nutrition circuits (9–32% DEHP by weight) can result in considerable phthalate exposure in neonatal patients (148 μg kg−1 day−1 DEHP, 4 μg kg−1 day−1 MEHP).(Takatori et al., 2008).
TABLE 2.
Description | Biological sample | Mean concentration, μg/L (range) | Reference | ||
---|---|---|---|---|---|
Patient samples | |||||
NICU patients RBC exchange transfusion |
Plasma | DEHP 5.8–19.6 μg/ml MEHP 5 μg/ml (maximum) |
Sjoberg, Bondesson, Sedin, & Gustafsson, 1985 | ||
NICU patients RBC exchange transfusion |
Plasma | DEHP 2.3–19.9 μg/ml (range) MEHP 1.1–15.6 μg/ml (range) |
Sjoberg et al., 1985 | ||
Infants, bypass Age range: 2 months to 2 years |
Whole blood | DEHP 1.10–5.06 μg/ml (range) MEHP 0.06–2.66 μg/ml (range) |
Barry, Labow, Keon, Tocchi, & Rock, 1989 | ||
NICU patients, ECMO | Plasma | DEHP 0–24.18 μg/ml (range) | Karle et al., 1997 | ||
NICU patients High exposure group |
Urine | MEHP 86 (median) | Green et al., 2005 | ||
NICU patients, w/o IV injections Median |
Urine | MEHP 73.3 (38.8–157) MEHHP 62.8 (18.7–833) MEOHP 49.7 (17.1–261) ∑DEHP 155 (78.0–1,203) |
Su et al., 2012 | ||
NICU patients, with IV injections Median |
MEHP 178 (48.9–1,273) MEHHP 393 (41.5–10,225) MEOHP 266 (27.1–9,268) ∑DEHP 805 (160–20,766) |
||||
NICU patients (preterm) Gestational age range: 28.9 ± 1.5 weeks |
Urine | DEHP 5.8 (0–163.7) MEHP 13 (0–1,234.3) MEOHP 239.6 (0–5,841.5) MEHHP 319.5 (0–5,068.6) |
Demirel et al., 2016 | ||
NICU patients (preterm) Feeding tube only |
MEHHP 6.9–240.5 | ||||
NICU patients (preterm) Ongoing intervention |
MEHHP 9.2–255.3 | ||||
Infants (preoperative) Age range: 5.4 ± 3.8 days |
Urine | MECPP 58.5 (29.4–116.5) MEHHP 10.7 (5.7–20.0) MEOHP 7.2 (3.9–13.2) MEHP 1.7 (0.8–3.9) |
Gaynor et al., 2018 | ||
Infants (postoperative) Same patients as preoperative |
MECPP 1166.1 (576.7–2,357.9) MEHHP 229.0 (119.6–438.4) MEOHP 148.6 (72.2–305.6) MEHP 27.2 (14.9–49.5) |
||||
NICU patients (preterm) Median |
Urine | MEHP 7.12 (0.5–72.39) MECPP 49.72 (0.2–304.95 MEHHP 11.84 (0.56–164.58) MEOHP 11.95 (55.0–148.57) |
Stroustrup et al., 2018 | ||
NICU patients (preterm) Lowest exposure group Median (25th–75th percentile) |
∑DEHP 68.4 (31.0–107.7) | ||||
NICU patients (preterm) Highest exposure group Median (25th–75th percentile) |
∑DEHP 134.0 (80.0–199.7) | ||||
Adult ICU patients on ECMO 20–72 years old Median |
Blood | DEHP 156.0 (31.5–1,009), MEHP 15.9 (<LOD to 475), |
Kaestner et al., 2020 | ||
Adult ICU patients w/ 2+ ECMO cannulas, 20–72 years old Median |
Blood | DEHP 338 (median) MEHP 114.9 (median) |
|||
Adult ICU patients w/ >1 ECMO membranes, 20–72 years old Median |
Blood | DEHP 535 (median) MEHP 114.9 (median) |
|||
Adult ICU patients on ECMO w/o urine output, 20–72 years old Median |
Blood | DEHP 823 (median) MEHP 214 (median) |
|||
Blood/other samples | |||||
Simulated neonatal exposure, Enteral nutrition |
Nutrition | DEHP 148 μg kg−1 day−1 (maximal) MEHP 3.72 μg kg−1 day−1 (maximal) |
Takatori et al., 2008 | ||
Packed RBC storage bags Day 1 | RBC supernatant | DEHP 34.3 μM (±20.0 SD) MEHP 3.7 μM (±2.8 SD) |
Rael et al., 2009 | ||
Packed RBC storage bags Day 42 | DEHP 433.2 μM (±131.2 SD) MEHP 74.0 μM (±19.1 SD) |
||||
DEHP exposure in blood products Following passage through PVC tubing |
Plasma | DEHP 6.4–29 μg/ml (packed red blood cells) DEHP 27.6–405 μg/ml (fresh frozen plasma) DEHP 34.2–61.4 μg/ml (platelets) |
Mallow & Fox, 2014 | ||
Unwashed RBC units Median storage time: 8 days |
RBC unit blood | DEHP 5.3–38.1 mg/L (range) MEHP 0.8–14.8 mg/L (range) |
Münch et al., 2020 | ||
Washed RBC units Average storage time: 31.2 ± 8.8 days |
DEHP 18.0 ± 2.0 mg/L [SD] MEHP 1.0 ± 0.7 mg/L [SD] |
In addition to tubing circuits, substantial phthalate leaching into stored blood products has also been reported (Cole, Tocchi, Wye, Villeneuve, & Rock, 1981; Luban, Rais-Bahrami, & Short, 2006; Peck et al., 1979; van der Meer et al., 2014). In red blood cell units, phthalate concentrations range from 5,300 to 38,100 μg/L for DEHP and 800 to 14,800 μg/L for its primary metabolite, mono-2-ethylhexyl phthalate (MEHP) (Münch et al., 2020). Notably, phthalate concentrations accumulate over time in stored red blood cell units—with DEHP levels increasing from 34.3 μM (±20.0 SD) on Day 1 to 433.2 μM (±131.2 SD) on Day 42, a 12.6-fold increase (Rael et al., 2009). Similarly, MEHP levels increase from 3.7 μM (±2.8 SD) on Day 1 to 74.0 μM (±19.1 SD) on Day 42, a 20.2-fold increase. Based on these measurements, Rael et al. estimated that a single red blood cell unit transfused close to expiry (42 days) would result in a circulating level of 740 nM MEHP and 4,330 nM DEHP (Rael et al., 2009). This estimate is in agreement with phthalate exposure measurements in neonates following an exchange transfusion, which resulted in 2.3–19.9 μg/ml DEHP and 1.1–15.6 μg/ml MEHP in blood samples (Sjoberg et al., 1985). With more than 3,000,000 infants admitted to the neonatal ICU each year in the United States, phthalate exposure from medical products is a considerable concern during this developmentally vulnerable period (FDA, 2002; Stroustrup et al., 2018). Neonatal and pediatric ICU patients can be exposed to multiple DEHP-containing plastic devices, which can result in a cumulative phthalate exposure of 16 mg/kg per day (Mallow & Fox, 2014). Such exposures can precipitate adverse health outcomes, including the cardiotoxic effects described in this review.
3.2 |. Effect of DEHP on heart development and intercellular coupling
As previously described, cardiomyocytes are connected to neighboring myocytes via intercalated disks, a highly organized structure comprised of mechanical and electrical connections (Vermij et al., 2017). Intercalated disks integrate electrical and mechanical function across the myocardial tissue, and as such, disruption of these intercellular junctions can increase the incidence of cardiac arrhythmias (Kleber & Saffitz, 2014; Rampazzo et al., 2014). Accordingly, environmental chemicals that disrupt cell–cell communication can have adverse effects on intercellular signaling and stability. In rodents, DEHP-treatment has been shown to induce hepatocellular carcinomas (Klaunig, Ruch, DeAngelo, & Kaylor, 1988), which may be a consequence of DEHP’s inhibition of gap junction intercellular communication —akin to tumor promotors (Isenberg et al., 2000; Kamendulis et al., 2002). Of interest, follow-up studies noted that in the liver, the loss of gap junctions was limited to mice and rats, suggesting a species-specific effect (Isenberg et al., 2000; McKee, 2000; Pugh et al., 2000). Notably, other groups have found a similar effect, with DEHP-treatment resulting in the inhibition of gap junction intercellular communication in other cell types, including lung fibroblasts (Cruciani, Mikalsen, Vasseur, & Sanner, 1997; Malcolm & Mills, 1989), Sertoli cells (Kang, Lee, Kim, & Kim, 2002) and cardiomyocytes (Gillum et al., 2009; Posnack, Idrees, et al., 2015). Loss of gap junction intercellular coupling can result in slowed electrical conduction in heart cells, which was reported in neonatal cardiomyocytes following phthalate treatment (Gillum et al., 2009). We reported that DEHP exposure slowed electrical conduction in cardiomyocyte layers in a dose-dependent manner (1–50 μg/L), with increased exposure resulting in an arrhythmogenic phenotype (Gillum et al., 2009). Additional studies are necessary to determine the role of phthalates in cardiomyocyte coupling, using in vivo models.
In addition to intercellular coupling, intrauterine DEHP exposure has been associated with cardiac malformations. Tang et al. reported that maternal DEHP exposure (250 mg/kg—1 g/kg) led to congenital heart defects and altered the expression of key cardiac transcription factors in mice (MEF2C, GATA4, CHF1) (Tang et al., 2018). These transcription factors are essential to cardiac development and the expression of sarcomere genes (Estrella & Naya, 2014; Suluba, Shuwei, Xia, & Mwanga, 2020). Of interest, Wang et al. reported that parental occupational exposure to phthalates was associated with an increased risk of congenital heart defects, including patent ductus arteriosus, atrial and ventricular septal defects (C. Wang et al., 2015). A similar result was reported by Snijder et al., which reported an association between paternal phthalate exposure and ventricular septal defects in children (Snijder et al., 2012). Little is known about the direct effects of environmental chemical exposures on cardiac development, but additional studies are warranted to investigate this link.
3.3 |. Effect of DEHP on electrophysiology and contractility
DEHP exposure has been shown to directly affect cardiac electrophysiology and contractile performance in experimental models (Supporting Information Table 6). Multiple studies have reported a rapid cardiodepressive effect following DEHP (or MEHP) exposure. Rubin et al. reported that 4 μg/ml DEHP treatment completely stopped the spontaneous beating activity of chick cardiomyocytes after 30 min, and resulted in 97–98% cell death with 24 hr treatment (Rubin & Jaeger, 1973). Importantly, this dose is within the range of clinical exposure, as DEHP levels in children on extracorporeal membrane oxygenation can reach 24 μg/ml (Karle et al., 1997). A similar cardiodepressive effect was observed by Aronson et al., in which excised, Langendorff-perfused, rat hearts were treated with a DEHP dose similar to that reported in stored blood bags (250 μM) (Aronson, Serlick, & Preti, 1978). The authors noted a dramatic decrease in heart rate, coronary flow and systolic tension—all of which increased in severity throughout the duration of study (60 min total). Further, atrioventricular conduction and repolarization time were significantly slowed in hearts perfused with DEHP-containing buffer (Aronson et al., 1978). We reported a similar effect on human cardiomyocyte automaticity, in which the spontaneous beating rate of embryonic stem cell derived cardiomyocytes declined with 72-hr DEHP treatment (50 μg/ml)—underscoring the potential applicability to humans (Posnack, Idrees, et al., 2015).
To further characterize the effect of phthalates on cardiac electrophysiology, our laboratory selected a dose of MEHP (60 μM) comparable to patient plasma levels after exchange transfusion for isolated, Langendorff-perfused heart studies (Jaimes et al., 2019; Sjoberg et al., 1985). In this study, we observed that MEHP-treatment slowed sinus node recovery time, increased atrioventricular conduction time, and slowed epicardial conduction velocity (Jaimes et al., 2019). Effects on conduction time may be partly attributed to inhibitory effects on voltage-gated sodium channels, as voltage-clamp studies in HEK 293 cells revealed a reduction in fast and late sodium current, albeit at relatively high MEHP concentrations (IC50 874 and 231 μmol/L, respectively). Slowed conduction velocity may also be attributed to impaired gap junction intercellular coupling and reduced connexin-43 expression, which has been reported in neonatal cardiomyocytes following phthalate treatment (Gillum et al., 2009). Indeed, we reported that DEHP exposure slowed cardiomyocyte conduction velocity in a dose-dependent manner (1–50 μg/L), starting 24 hr after exposure (Gillum et al., 2009). After prolonged treatment (72 hr), cardiomyocyte uncoupling resulted in an arrhythmogenic phenotype, which may be attributed to decreased connexin expression and/or localization.
Relatively few studies have examined the mechanistic underpinnings of phthalate chemicals on cardiac ion channels and calcium handling that may explain the observed changes in cardiac electrophysiology and contractility. Using microarray analysis, our laboratory reported that DEHP-treatment alters the mRNA expression of ion channel and calcium-handling genes in neonatal cardiomyocytes (Posnack, Lee, Brown, & Sarvazyan, 2011). Specifically, DEHP treatment decreased the expression of calcium handling genes, including calponin, troponin C, and calsequestrin 2—and increased the expression of ryanodine receptor 2, cardiac calcium transporting ATPase, triadin, voltage-dependent L-type calcium channel, phospholamban, sodium/potassium transporting ATPase. Such changes in global gene expression may explain some of the phenotypic observations in both cardiomyocytes and heart tissue, as both nodal cell automaticity and myocardial contractility are sensitive to changes in calcium ion homeostasis. As an example, our group reported that human embryonic stem cell-derived cardiomyocytes are sensitive to DEHP-treatment, which reduced the spontaneous beating rate, decreased the calcium transient amplitude and altered calcium transient kinetics (Posnack, Idrees, et al., 2015). DEHP-treated cells also displayed triggered activity following external pacing, which can be calcium-mediated. These results align with those of Barry et al., which reported decreased contractility in human atrial trabeculae in response to MEHP exposure (Barry, Labow, Keon, & Tocchi, 1990). The authors suggested that MEHP may act on cholinergic receptors, as atropine blocked its cardiodepressive effects. Alterations in calcium current have also been reported in other cell types. Mariana et al. found that DEHP-treatment inhibited L-type calcium channel current in vascular smooth muscle cells (Mariana, Feiteiro, & Cairrao, 2017). Future studies are warranted to fully elucidate the mechanistic effect of phthalates on calcium flux, cardiac electrophysiology, and contractility—including the relative timing required for these effects.
Finally, phthalate exposure has been shown to inhibit potassium ion channels that are important for establishing cardiac repolarization (Phase 3) and the resting membrane potential (Phase 4, cardiac action potential). Wu et al. reported that DEHP-treatment suppressed the amplitude of ether-a-go–go-related-gene potassium current (IK[erg]) in a pituitary cell line, in a concentration-dependent manner (IC50 = 16.3 μM) (Wu, Yang, Yeh, & Huang, 2012). It is important to note that the human ether-a-go-go-related gene (hERG) has been implicated in cardiac arrhythmias, as hERG channel inhibition delays repolarization (Phase 3) and presents as QT prolongation on electrocardiograms (Sanguinetti & Tristani-Firouz, 2006). DEHP exposure has also been shown to alter potassium current in hippo-campal cells. Ran et al. reported that high phthalate concentrations (100–300 μM DEHP) reduced the frequency of mini excitatory postsynaptic currents, increased the firing threshold needed to induce neuronal action potentials, and reduced the outward current through voltage-gated potassium channels (Kv current) (Ran, Luo, Gan, Liu, & Yang, 2019). As previously mentioned, cardiac repolarization is largely determined by outward potassium current. To the best of our knowledge, the direct effect of phthalates on potassium current have not been measured in cardiomyocyte preparations—however, our group did observe a dose–response relationship between increased DEHP exposure (1–50 μg/ml) and decreased potassium channel gene expression (Posnack et al., 2011).
3.4 |. Effects of DEHP on metabolism
EDCs, including DEHP, have been shown to interact with nuclear receptors to modulate gene expression (Casals-Casas & Desvergne, 2011). Our laboratory previously reported that DEHP exposure upregulates PPARα (peroxisome proliferator-activated receptor alpha) and its coactivator, PGC1α (peroxisome proliferator-activated receptor gamma, coactivator 1alpha) to increase the utilization of fatty acid substrates in cardiomyocytes (Posnack, Swift, Kay, Lee, & Sarvazyan, 2012). Importantly, both PPARα and PGC1α act as sensors to modulate metabolic activity (Lopaschuk, Ussher, Folmes, Jaswal, & Stanley, 2010). We also reported that myocardial reliance on fatty acid oxidation resulted in an inhibition of glucose oxidation and an accumulation of lactate. Using an ex vivo Langendorff-perfused heart model, Aronson et al. reported a 400% increase in lactate levels, 50% decrease in ATP production and depressed contractile function following DEHP exposure (Aronson et al., 1978). Similarly, Martinelli et al. reported an accumulation in the skeletal muscle of DEHP-treated animals (Martinelli, Mocchiutti, & Bernal, 2006), which further supports the notion that DEHP disrupts glucose metabolism. Notably, PPARα overexpression in mice leads to an unexpected phenotype with increased plasma glucose/insulin levels and glucose/insulin intolerance (B. Finck et al., 2005). Cardiac-specific PPARα overexpression in mice leads to lipid accumulation, reduced glucose utilization, insulin resistance, and signs of cardiomyopathy (B. N. Finck et al., 2002).
Given its interaction with PPAR nuclear receptors, this is one potential mechanism by which DEHP exposure can disrupt energy metabolism and contribute to metabolic disorders. Indeed, Amara et al. reported that DEHP exposure (5–200 mg/kg) altered the serum lipid profile of mice by increasing total cholesterol, triglycerides, low and high density lipoproteins (Amara et al., 2019). The authors also reported an increase in inflammatory and oxidative stress markers, which coincided with increased myocardial injury in DEHP-treated animals. Similar findings have been observed in children with increased MEHP exposure, which was associated with obesity, elevated triglycerides and increased blood pressure in children age 6–18 years old (Amin et al., 2018). Most recently, a meta-analysis study by Golestanzadeh et al. concluded that phthalate exposure was associated with increased cardiometabolic risk factors in children (Golestanzadeh, Riahi, & Kelishadi, 2019).
3.5 |. Effects of DEHP on blood pressure regulation
Early life exposure to phthalates can lead to vascular adaptations that increase the risk of cardiovascular disease later in life (Sol et al., 2020; Trasande et al., 2013). Cross-sectional studies have found an association between higher urinary phthalate levels and a propensity for elevated systolic blood pressure in children (Trasande et al., 2013; Trasande & Attina, 2015) (Supporting Information Table 7). Specifically, the work of Trasande et al. has highlighted a link between phthalate exposure and increased blood pressure in children (aged 8–19 years), while an observational cohort study by Jenkins et al. found a similar association in premature infants subjected to clinical phthalate exposure (Jenkins et al., 2019; Trasande et al., 2013). Experimental work indicates that the mechanisms responsible for phthalate-induced prehypertension are multifactorial, but may be related to increased cortisol: cortisone levels and mineralcorticoid receptor-mediated sodium retention (Jenkins et al., 2019; Zhao et al., 2010). Other studies suggest that DEHP-induced oxidative stress may be responsible for increased systolic blood pressure (K. Ferguson, Loch-Caruso, & Meeker, 2011; Hong et al., 2009). Human urinary DEHP-metabolite levels have been associated with increased serum C-reactive protein, an inflammatory marker (K. Ferguson et al., 2011), and the oxidative stress markers gamma glutamyltransferase (K. Ferguson et al., 2011) and malondialdehyde (Hong et al., 2009; Kambia et al., 2011). The latter is a biomarker of free radical production, which may alter arterial tone and increase blood pressure (Kambia et al., 2011). Additional studies have found an association between maternal urinary phthalate levels and 8-isoprostane, a biomarker of oxidative stress, in children (14 years of age) (Tran et al., 2017) and women with preterm birth (K. K. Ferguson, McElrath, Chen, Mukherjee, & Meeker, 2015). Increased oxidative stress during the vulnerable period of development could alter fetal programming and increase the risk of cardiovascular disease later in life (Giussani et al., 2012; Warembourg et al., 2019).
The downstream effects of maternal DEHP exposure on the developing fetus remain less clear. Valvi et al. and Vafeiadi et al. have reported that maternal DEHP exposure is associated with a decrease in systolic blood pressure in children aged <7 years (Vafeiadi et al., 2018; Valvi et al., 2015). Similarly, Sol et al. reported that maternal phthalate exposure was associated with a decrease in systolic and diastolic blood pressure in girls, but not boys (mean age 9.7 years) (Sol et al., 2020). It has been suggested that the endocrine-disrupting properties of DEHP results in impaired adrenal function, which is a potential mechanism for this decrease in blood pressure. Indeed, some experimental studies have reported a decrease in blood pressure following maternal DEHP-treatment (100–300 mg kg−1 day−1), which may be related to altered adrenal function that decreases aldosterone production and angiotensin II expression (Martinez-Arguelles et al., 2014, 2013; Martinez-Arguelles, Guichard, Culty, Zirkin, & Papadopoulos, 2011). Yet, others have reported that maternal DEHP exposure (0.25–30 mg/kg) impairs renal function and decreases endothelial nitric oxide synthase activity, resulting in increased blood pressure in rodent offspring (K.-I. Lee et al., 2016; Wei et al., 2012). Differing conclusions between studies may be partly attributed to variable dosing regimens, which can lead to significant differences in circulating phthalate levels. Additional mechanistic studies are needed to fully understand the effects of phthalate exposure during prenatal and postnatal development, and the potential impact on cardiovascular health later in life.
4 |. CONCLUSION
We aimed to review the literature on environmental and clinical exposures to BPA and DEHP—two high production volume chemicals that have emerged as potential contributors to cardiovascular dysfunction. Our current understanding of BPA and DEHP cardiovascular toxicity is a product of epidemiological and experimental studies. Epidemiological studies are useful in identifying the extent of chemical exposures and their association with adverse health conditions, whereas experimental studies are necessary to identify causation and pinpoint potential mechanisms. Collaborative efforts are needed between investigators with expertise in epidemiology, clinical, and basic science to fully address cardiovascular health concerns related to “plastic” exposure. Future areas of interest include: (a) epidemiological or clinical research with additional cardiovascular endpoints—including electrocardiogram or echocardiogram measurements; (b) clinical and experimental work that focuses on vulnerable patient populations—including those with congenital heart defects, cardiomyopathies, or cardiac arrhythmias, as chemical exposures may contribute to cardiovascular dysfunction; (c) clinical and experimental work focused on neonatal, infant and pediatric populations—as cardiac, autonomic and metabolic systems continue to develop and mature after birth; (d) use of (nonrodent) experimental models that better replicate human electrophysiology; (e) studies that explore the effects of bisphenol and phthalate mixtures (or studies that take into account a pediatric patient’s “exposome”); and finally, (f) comparative studies that examine the cardiac safety profile of alternative chemicals. The latter is particularly important as we move toward adopting BPA and DEHP substitutes, without a thorough understanding of their safety or impact on cardiovascular health (M. Ferguson, Lorenzen-Schmidt, & Pyle, 2019; Gao, Ma, Chen, & Wang, 2015; Lozano & Cid, 2013; Trasande, 2017; van der Meer et al., 2014). Through comprehensive examination of bisphenol and phthalate toxicity in vulnerable populations, protective measures or mitigation strategies may be revealed and implemented.
Supplementary Material
ACKNOWLEDGMENTS
The authors gratefully acknowledge Devon Guerrelli, MS for helpful discussions on cell junctions and editorial assistance. This work was supported by the National Institutes of Health (R01HL139472 to N. G. P.), Sheikh Zayed Institute for Pediatric Surgical Innovation, and the Children’s National Heart Institute.
Funding information
Children’s National Heart Institute; Sheikh Zayed Institute for Pediatric Surgical Innovation; National Institutes of Health, Grant/Award Number: R01HL139472
Footnotes
CONFLICT OF INTEREST
Nothing to disclose.
DATA AVAILABILITY STATEMENT
Data sharing is not applicable to this article as no new data were created or analyzed in this study.
SUPPORTING INFORMATION
Additional supporting information may be found online in the Supporting Information section at the end of this article.
REFERENCES
- Aekplakorn W, Chailurkit L-O, & Ongphiphadhanakul B (2015). Association of serum bisphenol A with hypertension in Thai population. International Journal of Hypertension, 2015, 594189. 10.1155/2015/594189 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amara I, Timoumi R, Annabi E, Neffati F, Najjar MF, Bouaziz C, & Abid-Essefi S (2019). Di (2-ethylhexyl) phthalate induces cardiac disorders in BALB/c mice. Environmental Science and Pollution Research International, 26(8), 7540–7549. 10.1007/s11356-019-04219-w [DOI] [PubMed] [Google Scholar]
- Amin M, Ebrahimpour K, Parastar S, Shoshtari-Yeganeh B, Hashemi M, Mansourian M, … Kelishadi R (2018). Association of Urinary Concentrations of phthalate metabolites with cardiometabolic risk factors and obesity in children and adolescents. Chemosphere, 211, 547–556. 10.1016/J.CHEMOSPHERE.2018.07.172 [DOI] [PubMed] [Google Scholar]
- Aris A (2014). Estimation of bisphenol A (BPA) concentrations in pregnant women, fetuses and nonpregnant women in eastern townships of Canada. Reproductive Toxicology, 45, 8–13. 10.1016/j.reprotox.2013.12.006 [DOI] [PubMed] [Google Scholar]
- Aronson CE, Serlick ER, & Preti G (1978). Effects of di-2-ethylhexyl phthalate on the isolated perfused rat heart. Toxicology and Applied Pharmacology, 44(1), 155–169. 10.1016/0041-008X(78)90295-8 [DOI] [PubMed] [Google Scholar]
- Asano S, Tune JD, & Dick GM (2010). Bisphenol A activates Maxi-K (K[Ca]1.1) channels in coronary smooth muscle. British Journal of Pharmacology, 160(1), 160–170. 10.1111/j.1476-5381.2010.00687.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- ATSDR. (2019). Toxicological profile for di(2-ethylhexyl)phthalate (DEHP). Atlanta, GA: Agency for Toxic Substances and Disease Registry. https://www.atsdr.cdc.gov/toxprofiles/tp9.pdf. [PubMed] [Google Scholar]
- Bae S, & Hong Y-C (2015). Exposure to bisphenol A from drinking canned beverages increases blood pressure: Randomized crossover trial. Hypertension, 65(2), 313–319. 10.1161/HYPERTENSIONAHA.114.04261 [DOI] [PubMed] [Google Scholar]
- Bae S, Kim JH, Lim Y-H, Park HY, & Hong Y-C (2012). Associations of bisphenol A exposure with heart rate variability and blood pressure. Hypertension, 60(3), 786–793. 10.1161/HYPERTENSIONAHA.112.197715 [DOI] [PubMed] [Google Scholar]
- Bae S, Lim Y-H, Lee YA, Shin CH, Oh S-Y, & Hong Y-C (2017). Maternal urinary bisphenol A concentration during midterm pregnancy and children’s blood pressure at age 4. Hypertension (Dallas, Tex.: 1979), 69(2), 367–374. 10.1161/HYPERTENSIONAHA.116.08281 [DOI] [PubMed] [Google Scholar]
- Barry YA, Labow RS, Keon WJ, Tocchi M, & Rock G (1989). Perioperative exposure to plasticizers in patients undergoing cardiopulmonary bypass. The Journal of Thoracic and Cardiovascular Surgery, 97(6), 900–905. [PubMed] [Google Scholar]
- Barry YA, Labow RS, Keon WJ, & Tocchi M (1990). Atropine inhibition of the cardiodepressive effect of mono (2-ethylhexyl)phthalate on human myocardium. Toxicology and Applied Pharmacology, 106(1), 48–52. 10.1016/0041-008X(90)90104-3 [DOI] [PubMed] [Google Scholar]
- Belcher SM, Gear RB, & Kendig EL (2015). Bisphenol A alters autonomic tone and extracellular matrix structure and induces sex-specific effects on cardiovascular function in male and female CD-1 mice. Endocrinology, 156(3), 882–895. 10.1210/en.2014-1847 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ben-Jonathan N, & Steinmetz R (1998). Xenoestrogens: The emerging story of bisphenol A. Trends in Endocrinology and Metabolism, 9(3), 124–128. 10.1016/S1043-2760(98)00029-0 [DOI] [PubMed] [Google Scholar]
- Beszterda M, & Frański R (2018). Endocrine disruptor compounds in environment: As a danger for children health. Pediatric Endocrinology, Diabetes, and Metabolism, 24(2), 88–95. 10.18544/PEDM-24.02.0107 [DOI] [PubMed] [Google Scholar]
- Borch J, Metzdorff SB, Vinggaard AM, Brokken L, & Dalgaard M (2006). Mechanisms underlying the anti-androgenic effects of diethylhexyl phthalate in fetal rat testis. Toxicology, 223 (1–2), 144–155. 10.1016/j.tox.2006.03.015 [DOI] [PubMed] [Google Scholar]
- Braun JM, & Hauser R (2011). Bisphenol A and children’s health. Current Opinion in Pediatrics, 23, 233–239. 10.1097/MOP.0b013e3283445675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Braun JM, Kalkbrenner AE, Calafat AM, Bernert JT, Ye X, Silva MJ, … Lanphear BP (2011). Variability and predictors of urinary bisphenol A concentrations during pregnancy. Environmental Health Perspectives, 119(1), 131–137. 10.1289/ehp.1002366 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Braun JM, Sathyanarayana S, & Hauser R (2013). Phthalate exposure and children’s health. Current Opinion in Pediatrics, 25(2), 247–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bruno KA, Mathews JE, Yang AL, Frisancho JA, Scott AJ, Greyner HD, … Fairweather D (2019). BPA alters estrogen receptor expression in the heart after viral infection activating cardiac mast cells and T cells leading to perimyocarditis and fibrosis. Frontiers in Endocrinology, 10, 598. 10.3389/fendo.2019.00598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calafat AM, Kuklenyik Z, Reidy JA, Caudill SP, Ekong J, & Needham LL (2005). Urinary concentrations of bisphenol A and 4-Nonylphenol in a human reference population. Environmental Health Perspectives, 113(4), 391–395. 10.1289/ehp.7534 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calafat AM, Needham LL, Silva MJ, & Lambert G (2004). Exposure to di-(2-ethylhexyl) phthalate among premature neonates in a neonatal intensive care unit. Pediatrics, 113(5), e429–e434. 10.1126/science.170.3956.460 [DOI] [PubMed] [Google Scholar]
- Calafat AM, Weuve J, Ye X, Jia LT, Hu H, Ringer S, … Hauser R (2009). Exposure to bisphenol A and other phenols in neonatal intensive care unit premature infants. Environmental Health Perspectives, 117(4), 639–644. 10.1289/ehp.0800265 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calafat AM, Ye X, Wong LY, Reidy JA, & Needham LL (2008). Exposure of the U.S. population to bisphenol A and 4-tertiary-octylphenol: 2003–2004. Environmental Health Perspectives, 116(1), 39–44. 10.1289/ehp.10753 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao X-L, Corriveau J, & Popovic S (2010). Sources of low concentrations of bisphenol A in canned beverage products. Journal of Food Protection, 73(8), 1548–1551. 10.4315/0362-028x-73.8.1548 [DOI] [PubMed] [Google Scholar]
- Casals-Casas C, & Desvergne B (2011). Endocrine disruptors: From endocrine to metabolic disruption. Annual Review of Physiology, 73, 135–162. 10.1146/annurevphysiol-012110-142200 [DOI] [PubMed] [Google Scholar]
- Chapalamadugu KC, VandeVoort CA, Settles ML, Robison BD, & Murdoch GK (2014). Maternal bisphenol A exposure impacts the fetal heart transcriptome. PLoS One, 9(2), e89096. 10.1371/journal.pone.0089096 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu I, Villeneuve DC, Secours V, Franklin C, Rock G, & Viau A (1978). Metabolism and tissue distribution of mono-2-ethylhexyl phthalate in the rat. Drug Metabolism and Disposition: The Biological Fate of Chemicals, 6(2), 146–149. [PubMed] [Google Scholar]
- Cohn JN, Ferrari R, Sharpe N, & International Forum on Cardiac Remodeling. (2000). Cardiac remodeling—Concepts and clinical implications: A consensus paper from an international forum on cardiac remodeling. Journal of the American College of Cardiology, 35(3), 569–582. 10.1016/S0735-1097(99)00630-0 [DOI] [PubMed] [Google Scholar]
- Cole RS, Tocchi M, Wye E, Villeneuve DC, & Rock G (1981). Contamination of commercial blood products by di-2-ethylhexyl phthalate and mono-2-ethylhexyl phthalate. Vox Sanguinis, 40(5), 317–322. 10.1111/j.1423-0410.1981.tb00715.x [DOI] [PubMed] [Google Scholar]
- Cruciani V, Mikalsen S, Vasseur P, & Sanner T (1997). Effects of peroxisome proliferators and 12-O-tetradecanoyl phorbol-13-acetate on intercellular communication and connexin43 in two hamster fibroblast systems. International Journal of Cancer, 73(2), 240–248. [DOI] [PubMed] [Google Scholar]
- D’Alessandro A, Reisz JA, Zhang Y, Gehrke S, Alexander K, Kanias T, … Gladwin MT (2019). Effects of aged stored autologous red blood cells on human plasma metabolome. Blood Advances, 3(6), 884–896. 10.1182/bloodadvances.2018029629 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Demirel A, Çoban A, Yıldırım S¸., Doğan C, Sancı R, & İnce Z (2016). Hidden toxicity in neonatal intensive care units: Phthalate exposure in very low birth weight infants. Journal of Clinical Research in Pediatric Endocrinology, 8(3), 298–304. 10.4274/jcrpe.3027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deutschmann A, Hans M, Meyer R, Häberlein H, & Swandulla D (2013). Bisphenol A inhibits voltage-activated Ca(2+) channels in vitro: Mechanisms and structural requirements. Molecular Pharmacology, 83(2), 501–511. 10.1124/mol.112.081372 [DOI] [PubMed] [Google Scholar]
- Duty SM, Mendonca K, Hauser R, Calafat AM, Ye X, Meeker JD, … Ringer S (2013). Potential sources of bisphenol A in the neonatal intensive care unit. Pediatrics, 131 (3), 483–489. 10.1542/peds.2012-1380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Estrella NL, & Naya FJ (2014). Transcriptional networks regulating the costamere, sarcomere, and other cytoskeletal structures in striated muscle. Cellular and Molecular Life Sciences: CMLS, 71(9), 1641–1656. 10.1007/S00018-013-1512-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fardoun M, Dehaini H, Shaito A, Mesmar J, El-Yazbi A, Badran A, … Eid AH (2020). The hypertensive potential of estrogen: An untold story. Vascular Pharmacology, 124, 106600. 10.1016/J.VPH.2019.106600 [DOI] [PubMed] [Google Scholar]
- FDA. (2002). Safety assessment of Di(2-ethylhexyl)phthalate (DEHP) released from PVC medical devices. ICOMM. Retrieved from https://noharm-uscanada.org/sites/default/files/documents-files/116/Safety_Assessment_of_DEHP.pdf [Google Scholar]
- Feiteiro J, Mariana M, Glória S, & Cairrao E (2018). Inhibition of L-type calcium channels by bisphenol A in rat aorta smooth muscle. Journal of Toxicological Sciences, 43(10), 579–586. 10.2131/jts.43.579 [DOI] [PubMed] [Google Scholar]
- Ferguson K, Loch-Caruso R, & Meeker J (2011). Urinary phthalate metabolites in relation to biomarkers of inflammation and oxidative stress: NHANES 1999–2006. Environmental Research, 111(5), 718–726. 10.1016/J.ENVRES.2011.02.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferguson KK, McElrath TF, Chen Y-H, Mukherjee B, & Meeker JD (2015). Urinary phthalate metabolites and biomarkers of oxidative stress in pregnant women: A repeated measures analysis. Environmental Health Perspectives, 123(3), 210–216. 10.1289/ehp.1307996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferguson M, Lorenzen-Schmidt I, & Pyle WG (2019). Bisphenol S rapidly depresses heart function through estrogen receptor-β and decreases phospholamban phosphorylation in a sex-dependent manner. Scientific Reports, 9(1), 15948. 10.1038/s41598-019-52350-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Finck B, Bernal-Mizrachi C, Han D, Coleman T, Sambandam N, LaRiviere L, … Kelly D (2005). A potential link between muscle peroxisome proliferator-activated receptor-alpha signaling and obesity-related diabetes. Cell Metabolism, 1 (2), 133–144. 10.1016/J.CMET.2005.01.006 [DOI] [PubMed] [Google Scholar]
- Finck BN, Lehman JJ, Leone TC, Welch MJ, Bennett MJ, Kovacs A, … Kelly DP (2002). The cardiac phenotype induced by PPARalpha overexpression mimics that caused by diabetes mellitus. The Journal of Clinical Investigation, 109(1), 121–130. 10.1172/JCI14080 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fiorini C, Tilloy-Ellul A, Chevalier S, Charuel C, & Pointis G (2004). Sertoli cell junctional proteins as early targets for different classes of reproductive toxicants. Reproductive Toxicology, 18(3), 413–421. 10.1016/j.reprotox.2004.01.002 [DOI] [PubMed] [Google Scholar]
- Fromme H, Bolte G, Koch HM, Angerer J, Boehmer S, Drexler H, … Liebl B (2007). Occurrence and daily variation of phthalate metabolites in the urine of an adult population. International Journal of Hygiene and Environmental Health, 210(1), 21–33. 10.1016/j.ijheh.2006.09.005 [DOI] [PubMed] [Google Scholar]
- Gao X, Liang Q, Chen Y, & Wang H-SS (2013). Molecular mechanisms underlying the rapid arrhythmogenic action of bisphenol A in female rat hearts. Endocrinology, 154(12), 4607–4617. 10.1210/en.2013-1737 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao X, Ma J, Chen Y, & Wang H-S (2015). Rapid responses and mechanism of action for low-dose bisphenol S on ex vivo rat hearts and isolated myocytes: Evidence of female-specific proarrhythmic effects. Environmental Health Perspectives, 123(6), 571–578. 10.1289/ehp.1408679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao X, & Wang H-S (2014). Impact of bisphenol A on the cardiovascular system—Epidemiological and experimental evidence and molecular mechanisms. International Journal of Environmental Research and Public Health, 11(8), 8399–8413. 10.3390/ijerph110808399 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaynor JW, Ittenbach RF, Calafat AM, Burnham NB, Bradman A, Bellinger DC, … Spray TL (2018). Perioperative exposure to suspect neurotoxicants from medical devices in newborns with congenital heart defects. The Annals of Thoracic Surgery, 107(2), 567–572. 10.1016/j.athoracsur.2018.06.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gayrard V, Lacroix MZ, Collet SH, Viguié C, Bousquet-Melou A, Toutain PL, & Picard-Hagen N (2013). High bio-availability of bisphenol A from sublingual exposure. Environmental Health Perspectives, 121(8), 951–956. 10.1289/ehp.1206339 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gear R, Kendziorski JA, & Belcher SM (2017). Effects of bisphenol A on incidence and severity of cardiac lesions in the NCTR-Sprague-Dawley rat: A CLARITY-BPA study. Toxicology Letters, 275, 123–135. 10.1016/j.toxlet.2017.05.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gerona RR, Pan J, Zota AR, Schwartz JM, Friesen M, Taylor JA, … Woodruff TJ (2016). Direct measurement of bisphenol A (BPA), BPA glucuronide and BPA sulfate in a diverse and low-income population of pregnant women reveals high exposure, with potential implications for previous exposure estimates: A cross-sectional study. Environmental Health: A Global Access Science Source, 15(1), 15. 10.1186/s12940-016-0131-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gillum N, Karabekian Z, Swift LM, Brown RP, Kay MW, & Sarvazyan N (2009). Clinically relevant concentrations of di (2-ethylhexyl) phthalate (DEHP) uncouple cardiac syncytium. Toxicology and Applied Pharmacology, 236(1), 25–38. 10.1016/j.taap.2008.12.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giovanoulis G, Alves A, Papadopoulou E, Cousins AP, Schütze A, Koch HM, … Voorspoels S (2016). Evaluation of exposure to phthalate esters and DINCH in urine and nails from a Norwegian study population. Environmental Research, 151, 80–90. 10.1016/j.envres.2016.07.025 [DOI] [PubMed] [Google Scholar]
- Giussani DA, Camm EJ, Niu Y, Richter HG, Blanco CE, Gottschalk R, … Herrera EA (2012). Developmental programming of cardiovascular dysfunction by prenatal hypoxia and oxidative stress. PLoS One, 7(2), e31017. 10.1371/journal.pone.0031017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Golestanzadeh M, Riahi R, & Kelishadi R (2019). Association of exposure to phthalates with cardiometabolic risk factors in children and adolescents: A systematic review and meta-analysis. Environmental Science and Pollution Research International, 26 (35), 35670–35686. 10.1007/s11356-019-06589-7 [DOI] [PubMed] [Google Scholar]
- González J, Valls N, Brito R, & Rodrigo R (2014). Essential hypertension and oxidative stress: New insights. World Journal of Cardiology, 6(6), 353–366. 10.4330/wjc.v6.i6.353 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, … Zoeller RT (2015). EDC-2: The Endocrine Society’s second scientific statement on endocrine-disrupting chemicals. Endocrine Reviews, 36(6), E1–E150. 10.1210/er.2015-1010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grant AO (2009). Cardiac ion channels. Circulation. Arrhythmia and Electrophysiology, 2(2), 185–194. 10.1161/CIRCEP.108.789081 [DOI] [PubMed] [Google Scholar]
- Green R, Hauser R, Calafat AM, Weuve J, Schettler T, Ringer S, … Hu H (2005). Use of di(2-ethylhexyl) phthalate-containing medical products and urinary levels of mono (2-ethylhexyl) phthalate in neonatal intensive care unit infants. Environmental Health Perspectives, 113(9), 1222–1225. 10.1289/ehp.7932 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo Y, & Kannan K (2013). A survey of phthalates and parabens in personal care products from the United States and its implications for human exposure. Environmental Science & Technology, 47(24), 14442–14449. 10.1021/es4042034 [DOI] [PubMed] [Google Scholar]
- Halden RU (2010). Plastics and health risks. Annual Review of Public Health, 31, 179–194. 10.1146/annurev.publhealth.012809.103714 [DOI] [PubMed] [Google Scholar]
- Han C, & Hong Y-C (2016). Bisphenol A, hypertension, and cardiovascular diseases: Epidemiological, laboratory, and clinical trial evidence. Current Hypertension Reports, 18(2), 11. 10.1007/s11906-015-0617-2 [DOI] [PubMed] [Google Scholar]
- Hillman LS, Goodwin SL, & Sherman WR (1975). Identification and measurement of plasticizer in neonatal tissues after umbilical catheters and blood products. The New England Journal of Medicine, 292(8), 381–386. 10.1056/NEJM197502202920801 [DOI] [PubMed] [Google Scholar]
- Hines CJ, Christianson AL, Jackson MV, Ye X, Pretty JR, Arnold JE, & Calafat AM (2018). An evaluation of the relationship among urine, air, and hand measures of exposure to bisphenol A (BPA) in US manufacturing workers. Annals of Work Exposures and Health, 62(7), 840–851. 10.1093/annweh/wxy042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hines RN (2008). The ontogeny of drug metabolism enzymes and implications for adverse drug events. Pharmacology & Therapeutics, 118(2), 250–267. 10.1016/j.pharmthera.2008.02.005 [DOI] [PubMed] [Google Scholar]
- Hong Y, Park E, Park M, Ko J, Oh S, Kim H, … Ha E (2009). Community level exposure to chemicals and oxidative stress in adult population. Toxicology Letters, 184(2), 139–144. 10.1016/J.TOXLET.2008.11.001 [DOI] [PubMed] [Google Scholar]
- Hoppin JA, Brock JW, Davis BJ, & Baird DD (2002). Reproducibility of urinary phthalate metabolites in first morning urine samples. Environmental Health Perspectives, 110(5), 515–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hutson DD, Gurrala R, Ogola BO, Zimmerman MA, Mostany R, Satou R, & Lindsey SH (2019). Estrogen receptor profiles across tissues from male and female Rattus norvegicus. Biology of Sex Differences, 10(1), 4. 10.1186/s13293-019-0219-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huygh J, Clotman K, Malarvannan G, Covaci A, Schepens T, Verbrugghe W, … Jorens PG (2015). Considerable exposure to the endocrine disrupting chemicals phthalates and bisphenol-A in intensive care unit (ICU) patients. Environment International, 81, 64–72. 10.1016/j.envint.2015.04.008 [DOI] [PubMed] [Google Scholar]
- Iribarne-Durán LM, Artacho-Cordón F, Peña-Caballero M, Molina-Molina JM, Jiménez-Díaz I, Vela-Soria F, … Olea N (2019). Presence of bisphenol A and parabens in a neonatal intensive care unit: An exploratory study of potential sources of exposure. Environmental Health Perspectives, 127(11), 117004. 10.1289/EHP5564 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Isenberg JS, Kamendulis LM, Smith JH, Ackley DC, Pugh G, Lington AW, & Klaunig JE (2000). Effects of di-2-ethylhexyl phthalate (DEHP) on gap-junctional intercellular communication (GJIC), DNA synthesis, and peroxisomal beta oxidation (PBOX) in rat, mouse, and hamster liver. Toxicological Sciences, 56(1), 73–85. 10.1093/toxsci/56.1.73 [DOI] [PubMed] [Google Scholar]
- Ito Y, Kamijima M, & Nakajima T (2019). Di(2-ethylhexyl) phthalate-induced toxicity and peroxisome proliferator-activated receptor alpha: A review. Environmental Health and Preventive Medicine, 24(1), 47. 10.1186/s12199-019-0802-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaeger RJ, & Rubin RJ (1973). Extraction, localization, and metabolism of di-2-ethylhexyl phthalate from PVC plastic medical devices. Environmental Health Perspectives, 3, 95–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaeger RJ, & Rubin RJ (2010). Migration of a phthalate ester plasticizer from polyvinyl chloride blood bags into stored human blood and its localization in human tissues. The New England Journal of Medicine, 287(22), 1114–1118. 10.1056/NEJM197211302872203 [DOI] [PubMed] [Google Scholar]
- Jaimes R, McCullough D, Siegel B, Swift L, McInerney D, Hiebert J, … Posnack N (2019). Plasticizer interaction with the heart: Chemicals used in plastic medical devices can interfere with cardiac electrophysiology. Circulation. Arrhythmia and Electrophysiology, 12, e007294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jarosova A, Harazim J, Suchy P, Kratka L, & Stancova V (2009). The distribution and accumulation of phthalates in the organs and tissues of chicks after the administration of feedstuffs with different phthalate concentrations. Veterinární Medicína, 54(9), 427–434. 10.17221/2/2009-VETMED [DOI] [Google Scholar]
- Jenkins R, Tackitt S, Gievers L, Iragorri S, Sage K, Cornwall T, … Rozansky D (2019). Phthalate-associated hypertension in premature infants: A prospective mechanistic cohort study. Pediatric Nephrology, 34(8), 1413–1424. 10.1007/s00467-019-04244-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang S, Liu H, Zhou S, Zhang X, Peng C, Zhou H, … Lu Q (2020). Association of bisphenol A and its alternatives bisphenol S and F exposure with hypertension and blood pressure: A cross-sectional study in China. Environmental Pollution (Barking, Essex: 1987), 257, 113639. 10.1016/j.envpol.2019.113639 [DOI] [PubMed] [Google Scholar]
- Kaestner F, Seiler F, Rapp D, Eckert E, Müller J, Metz C, … Göen T (2020). Exposure of patients to di(2-ethylhexy)phthalate (DEHP) and its metabolite MEHP during extracorporeal membrane oxygenation (ECMO) therapy. PLoS One, 15(1), e0224931. 10.1371/journal.pone.0224931 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kambia N, Dine T, Gressier B, Frimat B, Cazin J-L, Luyckx M, … Gottrand F (2011). Correlation between exposure to phthalates and concentrations of malondialdehyde in infants and children undergoing cyclic parenteral nutrition. Journal of Parenteral and Enteral Nutrition, 35(3), 395–401. 10.1177/0148607110381769 [DOI] [PubMed] [Google Scholar]
- Kamendulis LM, Isenberg JS, Smith JH, Pugh G, Lington AW, & Klaunig JE (2002). Comparative effects of phthalate monoesters on gap junctional intercellular communication and peroxisome proliferation in rodent and primate hepatocytes. Journal of Toxicology and Environmental Health. Part A, 65 (8), 569–588. 10.1080/152873902317349736 [DOI] [PubMed] [Google Scholar]
- Kang K, Lee Y, Kim H, & Kim S (2002). Di-(2-ethylhexyl) phthalate-induced cell proliferation is involved in the inhibition of gap junctional intercellular communication and blockage of apoptosis in mouse sertoli cells. Journal of Toxicology and Environmental Health. Part A, 65(5–6), 447–459. 10.1080/15287390252808109 [DOI] [PubMed] [Google Scholar]
- Karle VA, Short BL, Martin GR, Bulas DI, Getson PR, Luban NL, … Rubin RJ (1997). Extracorporeal membrane oxygenation exposes infants to the plasticizer, di(2-ethylhexyl) phthalate. Critical Care Medicine, 25(4), 696–703. [DOI] [PubMed] [Google Scholar]
- Kasneci A, Lee JS, Yun TJ, Shang J, Lampen S, Gomolin T, … Chalifour LE (2017). From the cover: Lifelong exposure of C57bl/6n male mice to bisphenol A or bisphenol S reduces recovery from a myocardial infarction. Toxicological Sciences, 159(1), 189–202. 10.1093/toxsci/kfx133 [DOI] [PubMed] [Google Scholar]
- Kataria A, Levine D, Wertenteil S, Vento S, Xue J, Rajendiran K, … Trachtman H (2017). Exposure to bisphenols and phthalates and association with oxidant stress, insulin resistance, and endothelial dysfunction in children. Pediatric Research, 81(6), 857–864. 10.1038/pr.2017.16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kato K, Silva MJ, Reidy JA, Hurtz D, Malek NA, Needham LL, … Calafat AM (2004). Mono(2-ethyl-5-hydroxyhexyl) phthalate and mono-(2-ethyl-5-oxohexyl) phthalate as biomarkers for human exposure assessment to di-(2-ethylhexyl) phthalate. Environmental Health Perspectives, 112(3), 327–330. 10.1289/ehp.6663 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khalil N, Ebert JR, Wang L, Belcher S, Lee M, Czerwinski SA, & Kannan K (2013). Bisphenol A and cardiometabolic risk factors in obese children. The Science of the Total Environment, 470–471C, 726–732. 10.1016/j.scitotenv.2013.09.088 [DOI] [PubMed] [Google Scholar]
- Kim CS, Sapienza PP, Ross IA, Johnson W, Luu HMD, & Hutter JC (2004). Distribution of bisphenol A in the neuroendocrine organs of female rats. Toxicology and Industrial Health, 20(1–5), 41–50. 10.1191/0748233704th186oa [DOI] [PubMed] [Google Scholar]
- Klaunig JE, Ruch RJ, DeAngelo AB, & Kaylor WH (1988). Inhibition of mouse hepatocyte intercellular communication by phthalate monoesters. Cancer Letters, 43(1–2), 65–71. 10.1016/0304-3835(88)90215-7 [DOI] [PubMed] [Google Scholar]
- Kleber AG, & Saffitz JE (2014). Role of the intercalated disc in cardiac propagation and arrhythmogenesis. Frontiers in Physiology, 5, 404. 10.3389/fphys.2014.00404 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koch HM, & Calafat AM (2009). Human body burdens of chemicals used in plastic manufacture. Philosophical Transactions of the Royal Society B: Biological Sciences, 364(1526), 2063–2078. 10.1098/rstb.2008.0208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koch HM, Drexler H, & Angerer J (2003). An estimation of the daily intake of di(2-ethylhexyl)phthalate (DEHP) and other phthalates in the general population. International Journal of Hygiene and Environmental Health, 206(2), 77–83. 10.1078/1438-4639-00205 [DOI] [PubMed] [Google Scholar]
- Koch HM, Lorber M, Christensen KLY, Pälmke C, Koslitz S, & Brüning T (2013). Identifying sources of phthalate exposure with human biomonitoring: Results of a 48h fasting study with urine collection and personal activity patterns. International Journal of Hygiene and Environmental Health, 216(6), 672–681. 10.1016/j.ijheh.2012.12.002 [DOI] [PubMed] [Google Scholar]
- Koch HM, Rossbach B, Drexler H, & Angerer J (2003). Internal exposure of the general population to DEHP and other phthalates—Determination of secondary and primary phthalate monoester metabolites in urine. Environmental Research, 93 (2), 177–185. 10.1016/S0013-9351(03)00083-5 [DOI] [PubMed] [Google Scholar]
- Kow L-M, & Pfaff DW (2016). Rapid estrogen actions on ion channels: A survey in search for mechanisms. Steroids, 111, 46–53. 10.1016/j.steroids.2016.02.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krishnan AV, Stathis P, Permuth SF, Tokes L, & Feldman D (1993). Bisphenol-A: An estrogenic substance is released from polycarbonate flasks during autoclaving. Endocrinology, 132(6), 2279–2286. 10.1210/endo.132.6.8504731 [DOI] [PubMed] [Google Scholar]
- La Merrill MA, Vandenberg LN, Smith MT, Goodson W, Browne P, Patisaul HB, … Zoeller RT (2020). Consensus on the key characteristics of endocrine-disrupting chemicals as a basis for hazard identification. Nature Reviews Endocrinology, 16(1), 45–57. 10.1038/s41574-019-0273-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Latini G (2005). Monitoring phthalate exposure in humans. Clinica Chimica Acta, 361(1–2), 20–29. 10.1016/J.CCCN.2005.05.003 [DOI] [PubMed] [Google Scholar]
- Leclerc F, Dubois M-F, & Aris A (2014). Maternal, placental and fetal exposure to bisphenol A in women with and without preeclampsia. Hypertension in Pregnancy, 33(3), 341–348. 10.3109/10641955.2014.892607 [DOI] [PubMed] [Google Scholar]
- Lee I-K, & Rhee S-K (2007). Inhibitory effect of bisphenol A on gap junctional intercellular communication in an epithelial cell line of rat mammary tissue. Archives of Pharmacal Research, 30(3), 337–343. 10.1007/BF02977615 [DOI] [PubMed] [Google Scholar]
- Lee K-I, Chiang C-W, Lin H-C, Zhao J-F, Li C-T, Shyue S-K, & Lee T-S (2016). Maternal exposure to di-(2-ethylhexyl) phthalate exposure deregulates blood pressure, adiposity, cholesterol metabolism and social interaction in mouse offspring. Archives of Toxicology, 90(5), 1211–1224. 10.1007/s00204-015-1539-0 [DOI] [PubMed] [Google Scholar]
- Lee YJ, Ryu HY, Kim HK, Min CS, Lee JH, Kim E, … Yoon HS (2008). Maternal and fetal exposure to bisphenol A in Korea. Reproductive Toxicology, 25(4), 413–419. 10.1016/j.reprotox.2008.05.058 [DOI] [PubMed] [Google Scholar]
- Lehmler HJ, Liu B, Gadogbe M, & Bao W (2018). Exposure to bisphenol A, bisphenol F, and bisphenol S in U.S. adults and children: The National Health and nutrition examination survey 2013–2014. ACS Omega, 3(6), 6523–6532. 10.1021/acsomega.8b00824 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li MWM, Mruk DD, Lee WM, & Cheng CY (2009). Disruption of the blood-testis barrier integrity by bisphenol A in vitro: Is this a suitable model for studying blood-testis barrier dynamics? The International Journal of Biochemistry & Cell Biology, 41(11), 2302–2314. 10.1016/j.biocel.2009.05.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li MWM, Mruk DD, Lee WM, & Cheng CY (2010). Connexin 43 is critical to maintain the homeostasis of the blood-testis barrier via its effects on tight junction reassembly. Proceedings of the National Academy of Sciences of the United States of America, 107(42), 17998–18003. 10.1073/pnas.1007047107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liang Q, Gao X, Chen Y, Hong K, & Wang H-SS (2014). Cellular mechanism of the nonmonotonic dose response of bisphenol A in rat cardiac myocytes. Environmental Health Perspectives, 122(6), 601–608. 10.1289/ehp.1307491 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lind L, & Lind PM (2012). Can persistent organic pollutants and plastic-associated chemicals cause cardiovascular disease? Journal of Internal Medicine, 271(6), 537–553. 10.1111/j.1365-2796.2012.02536.x [DOI] [PubMed] [Google Scholar]
- Lopaschuk GD, Ussher JR, Folmes CD, Jaswal JS, & Stanley WC (2010). Myocardial fatty acid metabolism in health and disease. Physiological Reviews, 90(1), 207–258. 10.1152/physrev.00015.2009 [DOI] [PubMed] [Google Scholar]
- Louch WE, Koivumäki JT, & Tavi P (2015). Calcium signalling in developing cardiomyocytes: Implications for model systems and disease. The Journal of Physiology, 593(5), 1047–1063. 10.1113/jphysiol.2014.274712 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lozano M, & Cid J (2013). DEHP plasticizer and blood bags: Challenges ahead. ISBT Science Series, 8(1), 127–130. 10.1111/voxs.12027 [DOI] [Google Scholar]
- Lu H, & Rosenbaum S (2014). Developmental pharmacokinetics in pediatric populations. The Journal of Pediatric Pharmacology and Therapeutics, 19(4), 262–276. 10.5863/1551-6776-19.4.262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luban N, Rais-Bahrami K, & Short B (2006). I want to say one word to you—just one word—“plastics”. Transfusion, 46(4), 503–506. 10.1111/j.1537-2995.2006.00766.x [DOI] [PubMed] [Google Scholar]
- Ma J, Hong K, & Wang H-S (2017). Progesterone protects against bisphenol A-induced arrhythmias in female rat cardiac myocytes via rapid signaling. Endocrinology, 158(4), 778–790. 10.1210/en.2016-1702 [DOI] [PMC free article] [PubMed] [Google Scholar]
- MacKay H, & Abizaid A (2018). A plurality of molecular targets: The receptor ecosystem for bisphenol-A (BPA). Hormones and Behavior, 101, 59–67. 10.1016/J.YHBEH.2017.11.001 [DOI] [PubMed] [Google Scholar]
- Malcolm AR, & Mills LJ (1989). Inhibition of gap-junctional intercellular communication between Chinese hamster lung fibroblasts by di(2-ethylhexyl) phthalate (DEHP) and trisodium nitrilotriacetate monohydrate (NTA). Cell Biology and Toxicology, 5(2), 145–153. 10.1007/bf00122649 [DOI] [PubMed] [Google Scholar]
- Mallow EB, & Fox MA (2014). Phthalates and critically ill neonates: Device-related exposures and non-endocrine toxic risks. Journal of Perinatology, 34(12), 892–897. 10.1038/jp.2014.157 [DOI] [PubMed] [Google Scholar]
- Mariana M, Feiteiro J, & Cairrao E (2017). Cardiovascular response of rat aorta to di-(2-ethylhexyl) phthalate (DEHP) exposure. Cardiovascular Toxicology, 18, 356–364. 10.1007/s12012-017-9439-6 [DOI] [PubMed] [Google Scholar]
- Mariana M, Feiteiro J, Verde I, & Cairrao E (2016). The effects of phthalates in the cardiovascular and reproductive systems: A review. Environment International, 94, 758–776. [DOI] [PubMed] [Google Scholar]
- Martinez-Arguelles DB, Campioli E, Lienhart C, Fan J, Culty M, Zirkin BR, & Papadopoulos V (2014). In utero exposure to the endocrine disruptor di-(2-ethylhexyl) phthalate induces long-term changes in gene expression in the adult male adrenal gland. Endocrinology, 155(5), 1667–1678. 10.1210/en.2013-1921 [DOI] [PubMed] [Google Scholar]
- Martinez-Arguelles DB, McIntosh M, Rohlicek CV, Culty M, Zirkin BR, & Papadopoulos V (2013). Maternal in utero exposure to the endocrine disruptor di-(2-ethylhexyl) phthalate affects the blood pressure of adult male offspring. Toxicology and Applied Pharmacology, 266(1), 95–100. 10.1016/j.taap.2012.10.027 [DOI] [PubMed] [Google Scholar]
- Martinez-Arguelles DB, Guichard T, Culty M, Zirkin BR, & Papadopoulos V (2011). In utero exposure to the antiandrogen di-(2-ethylhexyl) phthalate decreases adrenal aldosterone production in the adult rat. Biology of Reproduction, 85(1), 51–61. 10.1095/biolreprod.110.089920 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mattison DR, Karyakina N, Goodman M, & Lakind JS (2014). Pharmaco-and toxicokinetics of selected exogenous and endogenous estrogens: A review of the data and identification of knowledge gaps. Critical Reviews in Toxicology, 44, 696–724. 10.3109/10408444.2014.930813 [DOI] [PubMed] [Google Scholar]
- McKee RH (2000). The role of inhibition of gap junctional intercellular communication in rodent liver tumor induction by phthalates: Review of data on selected phthalates and the potential relevance to man. Regulatory Toxicology and Pharmacology: RTP, 32(1), 51–55. 10.1006/rtph.2000.1407 [DOI] [PubMed] [Google Scholar]
- Meeker JD (2012). Exposure to environmental endocrine disruptors and child development. Archives of Pediatrics & Adolescent Medicine, 166(6), E1. 10.1001/ARCHPEDIATRICS.2012.241 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melzer D, Osborne NJ, Henley WE, Cipelli R, Young A, Money C, … Galloway TS (2012). Urinary bisphenol A concentration and risk of future coronary artery disease in apparently healthy men and women. Circulation, 125(12), 1482–1490. 10.1161/CIRCULATIONAHA.111.069153 [DOI] [PubMed] [Google Scholar]
- Melzer D, Rice NE, Lewis C, Henley WE, & Galloway TS (2010). Association of urinary bisphenol A concentration with heart disease: Evidence from NHANES 2003/06. PLoS One, 5(1), e8673. 10.1371/journal.pone.0008673 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melzer D, Gates P, Osborne NJ, Osborn NJ, Henley WE, Cipelli R, … Galloway TS (2012). Urinary bisphenol A concentration and angiography-defined coronary artery stenosis. PLoS One, 7(8), e43378. 10.1371/journal.pone.0043378 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mendonca K, Hauser R, Calafat AM, Arbuckle TE, & Duty SM (2014). Bisphenol A concentrations in maternal breast milk and infant urine. International Archives of Occupational and Environmental Health, 87(1), 13–20. 10.1007/s00420-012-0834-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martinelli MI, Mocchiutti NO, & Bernal CA (2006). Dietary di(2-ethylhexyl)phthalate-impaired glucose metabolism in experimental animals. Human & Experimental Toxicology, 25 (9), 531–538. 10.1191/0960327106HET651OA [DOI] [PubMed] [Google Scholar]
- Michaela P, Mária K, Silvia H, Ľubica L, & L’ubica L (2014). Bisphenol A differently inhibits CaV3.1, Ca V3.2 and Ca V3.3 calcium channels. Naunyn Schmiedebergs Archives of Pharmacology, 387(2), 153–163. 10.1007/s00210-013-0932-6 [DOI] [PubMed] [Google Scholar]
- Mladěnka P, Applová L, Patočka J, Costa VM, Remiao F, Pourová J, … TOX-OER and CARDIOTOX Hradec Králové Researchers and Collaborators. (2018). Comprehensive review of cardiovascular toxicity of drugs and related agents. Medicinal Research Reviews, 38(4), 1332–1403. 10.1002/med.21476 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moon MK (2019). Concern about the safety of bisphenol A substitutes. Diabetes and Metabolism Journal, 43, 46–48. 10.4093/dmj.2019.0027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moreman J, Lee O, Trznadel M, David A, Kudoh T, & Tyler CR (2017). Acute toxicity, teratogenic, and estrogenic effects of bisphenol A and its alternative replacements bisphenol S, bisphenol F, and bisphenol AF in zebrafish embryo-larvae. Environmental Science & Technology, 51(21), 12796–12805. 10.1021/ACS.EST.7B03283 [DOI] [PubMed] [Google Scholar]
- Moriyama K, Tagami T, Akamizu T, Usui T, Saijo M, Kanamoto N, … Nakao K (2002). Thyroid hormone action is disrupted by bisphenol A as an antagonist. The Journal of Clinical Endocrinology and Metabolism, 87(11), 5185–5190. 10.1210/jc.2002-020209 [DOI] [PubMed] [Google Scholar]
- Münch F, Göen T, Zimmermann R, Adler W, Purbojo A, Höllerer C, … Rüffer A (2020). Reduction of exposure to plasticizers in stored red blood cell units. Perfusion, 35(1), 32–38. 10.1177/0267659119851403 [DOI] [PubMed] [Google Scholar]
- Nadal A, Ropero AB, Laribi O, Maillet M, Fuentes E, & Soria B (2000). Nongenomic actions of estrogens and xenoestrogens by binding at a plasma membrane receptor unrelated to estrogen receptor α and estrogen receptor β. Proceedings of the National Academy of Sciences of the United States of America, 97(21), 11603–11608. 10.1073/pnas.97.21.11603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nassan FL, Gunn JA, Hill MM, Williams PL, & Hauser R (2020). Association of urinary concentrations of phthalate metabolites with quinolinic acid among women: A potential link to neurological disorders. Environment International, 138, 105643. 10.1016/j.envint.2020.105643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- National Research Council (U.S.) Committee on the Health Risks of Phthalates. (2008). Phthalates and cumulative risk assessment: The tasks ahead. Washington, DC: National Academies Press. [PubMed] [Google Scholar]
- Nishikawa M, Iwano H, Yanagisawa R, Koike N, Inoue H, & Yokota H (2010). Placental transfer of conjugated bisphenol A and subsequent reactivation in the rat fetus. Environmental Health Perspectives, 118(9), 1196–1203. 10.1289/ehp.0901575 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oishi S, & Hiraga K (1982). Distribution and elimination of di-2-ethylhexyl phthalate (DEHP) and mono-2-ethylhexyl phthalate (MEHP) after a single oral administration of DEHP in rats. Archives of Toxicology, 51(2), 149–155. 10.1007/BF00302755 [DOI] [Google Scholar]
- O’Reilly AO, Eberhardt E, Weidner C, Alzheimer C, Wallace BA, & Lampert A (2012). Bisphenol A binds to the local anesthetic receptor site to block the human cardiac sodium channel. PLoS One, 7(7), e41667. 10.1371/journal.pone.0041667 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Padmanabhan V, Siefert K, Ransom S, Johnson T, Pinkerton J, Anderson L, … Kannan K (2008). Maternal bisphenol-A levels at delivery: A looming problem? Journal of Perinatology, 28(4), 258–263. 10.1038/sj.jp.7211913 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patel BB, Kasneci A, Bolt AM, Di Lalla V, Di Iorio MR, Raad M, … Chalifour LE (2015). Chronic exposure to bisphenol A reduces successful cardiac remodeling after an experimental myocardial infarction in male C57bl/6n mice. Toxicological Sciences, 146(1), 101–115. 10.1093/toxsci/kfv073 [DOI] [PubMed] [Google Scholar]
- Patel BB, Raad M, Sebag IA, & Chalifour LE (2013). Lifelong exposure to bisphenol A alters cardiac structure/function, protein expression, and DNA methylation in adult mice. Toxicological Sciences, 133(1), 174–185. 10.1093/toxsci/kft026 [DOI] [PubMed] [Google Scholar]
- Patel BB, Raad M, Sebag IA, & Chalifour LE (2015). Sex-specific cardiovascular responses to control or high fat diet feeding in C57bl/6 mice chronically exposed to bisphenol A. Toxicology Reports, 2, 1310–1318. 10.1016/j.toxrep.2015.09.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patel CJ, Yang T, Hu Z, Wen Q, Sung J, El-Sayed YY, … Butte AJ (2014). Investigation of maternal environmental exposures in association with self-reported preterm birth. Reproductive Toxicology, 45, 1–7. 10.1016/j.reprotox.2013.12.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peck CC, Odom DG, Friedman HI, Albro PW, Hass JR, Brady JT, & Jess DA (1979). Di-2-ethylhexyl phthalate (DEHP) and mono-2-ethylexyl phthalate (MEHP) accumulation in whole blood and red cell concentrates. Transfusion, 19(2), 137–146. [DOI] [PubMed] [Google Scholar]
- Philips EM, Jaddoe VWV, & Trasande L (2017). Effects of early exposure to phthalates and bisphenols on cardiometabolic outcomes in pregnancy and childhood. Reproductive Toxicology (Elmsford, NY), 68, 105–118. 10.1016/j.reprotox.2016.08.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Philips EM, Trasande L, Kahn LG, Gaillard R, Steegers EAP, & Jaddoe VWV (2019). Early pregnancy bisphenol and phthalate metabolite levels, maternal hemodynamics and gestational hypertensive disorders. Human Reproduction (Oxford, England), 34(2), 365–373. 10.1093/humrep/dey364 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Plonait SL, Nau H, Maier RF, Wittfoht W, & Obladen M (1993). Exposure of newborn infants to di-(2-ethylhexyl)-phthalate and 2-ethylhexanoic acid following exchange transfusion with polyvinylchloride catheters. Transfusion, 33(7), 598–605. [DOI] [PubMed] [Google Scholar]
- Posnack NG, Lee NH, Brown R, & Sarvazyan N (2011). Gene expression profiling of DEHP-treated cardiomyocytes reveals potential causes of phthalate arrhythmogenicity. Toxicology, 279(1–3), 54–64. 10.1016/j.tox.2010.09.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Posnack NG, Idrees R, Ding H, Jaimes Iii R, Stybayeva G, Karabekian Z, … Sarvazyan N (2015). Exposure to phthalates affects calcium handling and intercellular connectivity of human stem cell-derived cardiomyocytes. PLoS One, 10(3), e0121927. 10.1371/journal.pone.0121927 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Posnack NG (2014). The adverse cardiac effects of di(2-ethylhexyl)phthalate and bisphenol A. Cardiovascular Toxicology, 14(4), 339–357. 10.1007/s12012-014-9258-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Posnack NG, Brooks D, Chandra A, Jaimes R, Sarvazyan N, & Kay MW (2015). Physiological response of cardiac tissue to bisphenol A: Alterations in ventricular pressure and contractility. American Journal of Physiology. Heart and Circulatory Physiology, 309(2), H267–H275. 10.1152/ajpheart.00272.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Posnack NG, Jaimes R, Asfour H, Swift LM, Wengrowski AM, Sarvazyan N, … Kay MW (2014). Bisphenol A exposure and cardiac electrical conduction in excised rat hearts. Environmental Health Perspectives, 122(4), 384–390. 10.1289/ehp.1206157 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Posnack NG, Swift LM, Kay MW, Lee NH, & Sarvazyan N (2012). Phthalate exposure changes the metabolic profile of cardiac muscle cells. Environmental Health Perspectives, 120(9), 1243–1251. 10.1289/ehp.1205056 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pugach EK, Blenck CL, Dragavon JM, Langer SJ, & Leinwand LA (2016). Estrogen receptor profiling and activity in cardiac myocytes. Molecular and Cellular Endocrinology, 431, 62–70. 10.1016/j.mce.2016.05.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pugh G, Isenberg J, Kamendulis L, Ackley D, Clare L, Brown R, … Klaunig J (2000). Effects of di-isononyl phthalate, di-2-ethylhexyl phthalate, and clofibrate in Cynomolgus monkeys. Toxicological Sciences: An Official Journal of the Society of Toxicology, 56(1), 181–188. 10.1093/TOXSCI/56.1.181 [DOI] [PubMed] [Google Scholar]
- Quagliariello V, Coppola C, Mita DG, Piscopo G, Iaffaioli RV, Botti G, & Maurea N (2019). Low doses of bisphenol A have pro-inflammatory and pro-oxidant effects, stimulate lipid peroxidation and increase the cardiotoxicity of doxorubicin in cardiomyoblasts. Environmental Toxicology and Pharmacology, 69, 1–8. 10.1016/j.etap.2019.03.006 [DOI] [PubMed] [Google Scholar]
- Rael LT, Bar-Or R, Ambruso DR, Mains CW, Slone DS, Craun ML, & Bar-Or D (2009). Phthalate esters used as plasticizers in packed red blood cell storage bags may lead to progressive toxin exposure and the release of pro-inflammatory cytokines. Oxidative Medicine and Cellular Longevity, 2(3), 166–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rahm AK, Lugenbiel P, Schweizer PA, Katus HA, & Thomas D (2018). Role of ion channels in heart failure and channelopathies. Biophysical Reviews, 10(4), 1097–1106. 10.1007/s12551-018-0442-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramadan M, Sherman M, Jaimes R, Chaluvadi A, Swift L, & Posnack NG (2018). Disruption of neonatal cardiomyocyte physiology following exposure to bisphenol-A. Scientific Reports, 8(1), 7356. 10.1038/s41598-018-25719-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rampazzo A, Calore M, van Hengel J, & van Roy F (2014). Intercalated discs and arrhythmogenic cardiomyopathy. Circulation: Cardiovascular Genetics, 7(6), 930–940. 10.1161/CIRCGENETICS.114.000645 [DOI] [PubMed] [Google Scholar]
- Ran D, Luo Y, Gan Z, Liu J, & Yang J (2019). Neural mechanisms underlying the deficit of learning and memory by exposure to di(2-ethylhexyl) phthalate in rats. Ecotoxicology and Environmental Safety, 174, 58–65. 10.1016/J.ECOENV.2019.02.043 [DOI] [PubMed] [Google Scholar]
- Rasdi Z, Kamaludin R, Ab. Rahim S Syed Ahmad Fuad SB Othman MHD, Siran R, … Sheikh Abdul Kadir SH (2020). The impacts of intrauterine bisphenol A exposure on pregnancy and expression of miRNAs related to heart development and diseases in animal model. Scientific Reports, 10(1), 5882. 10.1038/s41598-020-62420-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ribeiro E, Ladeira C, & Viegas S (2017). Occupational exposure to bisphenol A (BPA): A reality that still needs to be unveiled. Toxics, 5(3), 22. 10.3390/toxics5030022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rochester JR, & Bolden AL (2015). Bisphenol S and F: A systematic review and comparison of the hormonal activity of bisphenol A substitutes. Environmental Health Perspectives, 123(7), 643–650. 10.1289/ehp.1408989 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rowdhwal SSS, & Chen J (2018). Toxic effects of di-2-ethylhexyl phthalate: An overview. BioMed Research International, 2018, 1750368. 10.1155/2018/1750368 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rubin RJ, & Jaeger RJ (1973). Some pharmacologic and toxicologic effects of di-2-ethylhexyl phthalate (DEHP) and other plasticizers. Environmental Health Perspectives, 3, 53–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salian S, Doshi T, & Vanage G (2009). Neonatal exposure of male rats to bisphenol A impairs fertility and expression of sertoli cell junctional proteins in the testis. Toxicology, 265, 56–67. 10.1016/j.tox.2009.09.012 [DOI] [PubMed] [Google Scholar]
- Sanguinetti M, & Tristani-Firouz M (2006). hERG potassium channels and cardiac arrhythmia. Nature, 440(7083), 463–469. 10.1038/NATURE04710 [DOI] [PubMed] [Google Scholar]
- Saura M, Marquez S, Reventun P, Olea-Herrero N, Arenas MI, Moreno-Gómez-Toledano R, … Bosch RJ (2014). Oral administration of bisphenol A induces high blood pressure through angiotensin II/CaMKII-dependent uncoupling of eNOS. FASEB Journal, 28(11), 4719–4728. 10.1096/fj.14-252460 [DOI] [PubMed] [Google Scholar]
- Schönfelder G, Wittfoht W, Hopp H, Talsness CE, Paul M, & Chahoud I (2002). Parent bisphenol A accumulation in the human maternal-fetal-placental unit. Environmental Health Perspectives, 110(11), A703–A707. 10.1289/ehp.110-1241091 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schug TT, Blawas AM, Gray K, Heindel JJ, & Lawler CP (2015). Elucidating the links between endocrine disruptors and neurodevelopment. Endocrinology, 156(6), 1941–1951. 10.1210/en.2014-1734 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shang J, Corriveau J, Champoux-Jenane A, Gagnon J, Moss E, Dumas P, … Chalifour LE (2019). Recovery from a myocardial infarction is impaired in male C57bl/6 N mice acutely exposed to the bisphenols and phthalates that escape from medical devices used in cardiac surgery. Toxicological Sciences, 168(1), 78–94. 10.1093/toxsci/kfy276 [DOI] [PubMed] [Google Scholar]
- Shankar A, Teppala S, & Sabanayagam C (2012). Bisphenol A and peripheral arterial disease: Results from the NHANES. Environmental Health Perspectives, 120, 1297–1300. 10.1289/ehp.1104114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shankar A, & Teppala S (2012). Urinary bisphenol A and hypertension in a multiethnic sample of US adults. Journal of Environmental and Public Health, 2012, 481641. 10.1155/2012/481641 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shelby MD (2008). NTP-CERHR monograph on the potential human reproductive and developmental effects of bisphenol A. NTP CERHR MON, 22, v vii–ix, 1–64 passim. [PubMed] [Google Scholar]
- Shin BS, Kim CH, Jun YS, Kim DH, Lee BM, Yoon CH, … Yoo SD (2004). Physiologically based pharmacokinetics of bisphenol A. Journal of Toxicology and Environmental Health, Part A, 67(23–24), 1971–1985. 10.1080/15287390490514615 [DOI] [PubMed] [Google Scholar]
- Silva MJ, Barr DB, Reidy JA, Malek NA, Hodge CC, Caudill SP, … Calafat AM (2004). Urinary levels of seven phthalate metabolites in the U.S. population from the National Health and Nutrition Examination Survey (NHANES) 1999–2000. Environmental Health Perspectives, 112(3), 331–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singh S, & Li SS-L (2012). Epigenetic effects of environmental chemicals bisphenol A and phthalates. International Journal of Molecular Sciences, 13(8), 10143–10153. 10.3390/ijms130810143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sjöberg P, Bondesson U, Sedin G, & Gustafsson J (1985). Dispositions of di- and mono-(2-ethylhexyl) phthalate in newborn infants subjected to exchange transfusions. European Journal of Clinical Investigation, 15(6), 430–436. [DOI] [PubMed] [Google Scholar]
- Sjoberg PO, Bondesson UG, Sedin EG, & Gustafsson JP (1985). Exposure of newborn infants to plasticizers. Plasma levels of di-(2-ethylhexyl) phthalate and mono-(2-ethylhexyl) phthalate during exchange transfusion. Transfusion, 25(5), 424–428. 10.1046/j.1537-2995.1985.25586020115.x [DOI] [PubMed] [Google Scholar]
- Snijder CA, Vlot IJ, Burdorf A, Obermann-Borst SA, Helbing WA, Wildhagen MF, … Steegers-Theunissen RPM (2012). Congenital heart defects and parental occupational exposure to chemicals. Human Reproduction, 27(5), 1510–1517. 10.1093/humrep/des043 [DOI] [PubMed] [Google Scholar]
- Sohoni P, & Sumpter JP (1998). Several environmental oestrogens are also anti-androgens. The Journal of Endocrinology, 158(3), 327–339. 10.1677/joe.0.1580327 [DOI] [PubMed] [Google Scholar]
- Sol CM, Santos S, Asimakopoulos AG, Martinez-Moral M-P, Duijts L, Kannan K, … Jaddoe VWV (2020). Associations of maternal phthalate and bisphenol urine concentrations during pregnancy with childhood blood pressure in a population-based prospective cohort study. Environment International, 138, 105677. 10.1016/j.envint.2020.105677 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soriano S, Ripoll C, Alonso-Magdalena P, Fuentes E, Quesada I, Nadal A, & Martinez-Pinna J (2016). Effects of bisphenol A on ion channels: Experimental evidence and molecular mechanisms. Steroids, 111, 12–20. 10.1016/j.steroids.2016.02.020 [DOI] [PubMed] [Google Scholar]
- Steffensen I-L, Dirven H, Couderq S, David A, D’Cruz SC, Fernández MF, … Hofer T (2020). Bisphenols and oxidative stress biomarkers-associations found in human studies, evaluation of methods used, and strengths and weaknesses of the biomarkers. International Journal of Environmental Research and Public Health, 17(10), 3609. 10.3390/ijerph17103609 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stroustrup A, Bragg JB, Andra SS, Curtin PC, Spear EA, Sison DB, … Gennings C (2018). Neonatal intensive care unit phthalate exposure and preterm infant neurobehavioral performance. PLoS One, 13(3), e0193835. 10.1371/journal.pone.0193835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Su P-H, Chang Y-Z, Chang H-P, Wang S-L, Haung H-I, Huang P-C, & Chen J-Y (2012). Exposure to di(2-ethylhexyl) phthalate in premature neonates in a neonatal intensive care unit in Taiwan. Pediatric Critical Care Medicine, 13(6), 671–677. 10.1097/PCC.0b013e3182455558 [DOI] [PubMed] [Google Scholar]
- Suluba E, Shuwei L, Xia Q, & Mwanga A (2020). Congenital heart diseases: Genetics, non-inherited risk factors, and signaling pathways. Egyptian Journal of Medical Human Genetics, 21 (1), 11. 10.1186/s43042-020-0050-1 [DOI] [Google Scholar]
- Swan SH (2008). Environmental phthalate exposure in relation to reproductive outcomes and other health endpoints in humans. Environmental Research, 108(2), 177–184. 10.1016/j.envres.2008.08.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takatori S, Okamoto Y, Kitagawa Y, Hori S, Izumi S-I, Makino T, & Nakazawa H (2008). Simulated neonatal exposure to DEHP and MEHP from PVC enteral nutrition products. International Journal of Pharmaceutics, 352(1–2), 139–145. 10.1016/j.ijpharm.2007.10.020 [DOI] [PubMed] [Google Scholar]
- Tang C, Deng Y, Duan H, Zhang Y, Li Y, Qiu D, … Wang C (2018). The effect of maternal exposure to di-(2-ethylhexyl)-phthalate on fetal cardiac development in mice. Journal of Applied Toxicology, 38, 834–842. 10.1002/jat.3591 [DOI] [PubMed] [Google Scholar]
- Taylor JA, Vom Saal FS, Welshons WV, Drury B, Rottinghaus G, Hunt PA, … VandeVoort CA (2011). Similarity of bisphenol A pharmacokinetics in rhesus monkeys and mice: Relevance for human exposure. Environmental Health Perspectives, 119(4), 422–430. 10.1289/ehp.1002514 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teeguarden JG, Calafat AM, Ye X, Doerge DR, Churchwell MI, Gunawan R, & Graham MK (2011). Twenty-four hour human urine and serum profiles of bisphenol A during high-dietary exposure. Toxicological Sciences, 123(1), 48–57. 10.1093/toxsci/kfr160 [DOI] [PubMed] [Google Scholar]
- Teeguarden JG, Twaddle NC, Churchwell MI, & Doerge DR (2016). Urine and serum biomonitoring of exposure to environmental estrogens I: Bisphenol A in pregnant women. Food and Chemical Toxicology, 92, 129–142. 10.1016/j.fct.2016.03.023 [DOI] [PubMed] [Google Scholar]
- Teeguarden JG, Twaddle NC, Churchwell MI, Yang X, Fisher JW, Seryak LM, & Doerge DR (2015). 24-Hour human urine and serum profiles of bisphenol A: Evidence against sublingual absorption following ingestion in soup. Toxicology and Applied Pharmacology, 288(2), 131–142. 10.1016/j.taap.2015.01.009 [DOI] [PubMed] [Google Scholar]
- Teeguarden JG, Waechter JM Jr., Clewell HJ III, Covington TR, & Barton HA (2005). Evaluation of oral and intravenous route pharmacokinetics, plasma protein binding, and uterine tissue dose metrics of bisphenol A: A physiologically based pharmacokinetic approach. Toxicological Sciences, 85(2), 823–838. 10.1093/toxsci/kfi135 [DOI] [PubMed] [Google Scholar]
- Tereshchenko LG, & Posnack NG (2019). Does plastic chemical exposure contribute to sudden death of patients on dialysis? Heart Rhythm, 16(2), 312–317. 10.1016/j.hrthm.2018.08.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tickner JA, Schettler T, Guidotti T, McCally M, & Rossi M (2001). Health risks posed by use of di-2-ethylhexyl phthalate (DEHP) in PVC medical devices: A critical review. American Journal of Industrial Medicine, 39(1), 100–111 JOUR. [DOI] [PubMed] [Google Scholar]
- Tran V, Tindula G, Huen K, Bradman A, Harley K, Kogut K, … Holland N (2017). Prenatal phthalate exposure and 8-isoprostane among Mexican–American children with high prevalence of obesity. Journal of Developmental Origins of Health and Disease, 8(2), 196–205. 10.1017/S2040174416000763 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trasande L (2017). Exploring regrettable substitution: Replacements for bisphenol A. The Lancet Planetary Health, 1(3), e88–e89. 10.1016/S2542-5196(17)30046-3 [DOI] [PubMed] [Google Scholar]
- Trasande L, & Attina TM (2015). Association of exposure to di-2-ethylhexylphthalate replacements with increased blood pressure in children and adolescents. Hypertension, 66(2), 301–308. 10.1161/HYPERTENSIONAHA.115.05603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trasande L, Sathyanarayana S, Spanier AJ, Trachtman H, Attina TM, & Urbina EM (2013). Urinary phthalates are associated with higher blood pressure in childhood. Journal of Pediatrics, 163(3), 747–753.e1. 10.1016/j.jpeds.2013.03.072 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Typpo KV, Larmonier CB, Deschenes J, Redford D, Kiela PR, & Ghishan FK (2015). Clinical characteristics associated with postoperative intestinal epithelial barrier dysfunction in children with congenital heart disease. Pediatric Critical Care Medicine, 16(1), 37–44. 10.1097/PCC.0000000000000256 [DOI] [PMC free article] [PubMed] [Google Scholar]
- U.S. Environmental Protection Agency. (1988). Bisphenol A: Integrated risk information system, Washington, DC: U.S. Environmental Protection Agency. https://cfpub.epa.gov/ncea/iris/iris_documents/documents/subst/0356_summary.pdf. [Google Scholar]
- Vafeiadi M, Myridakis A, Roumeliotaki T, Margetaki K, Chalkiadaki G, Dermitzaki E, … L C (2018). Association of early life exposure to phthalates with obesity and cardiometabolic traits in childhood: Sex specific associations. Frontiers in Public Health, 6, 327. 10.3389/FPUBH.2018.00327 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Valokola MG, Karimi G, Razavi BM, Kianfar M, Jafarian AH, Jaafari MR, & Imenshahidi M (2019). The protective activity of nanomicelle curcumin in bisphenol A-induced cardiotoxicity following subacute exposure in rats. Environmental Toxicology, 34(3), 319–329. 10.1002/tox.22687 [DOI] [PubMed] [Google Scholar]
- Valvi D, Casas M, Romaguera D, Monfort N, Ventura R, Martinez D, … Vrijheid M (2015). Prenatal phthalate exposure and childhood growth and blood pressure: Evidence from the Spanish INMA-Sabadell birth cohort study. Environmental Health Perspectives, 123(10), 1022–1029. 10.1289/ehp.1408887 [DOI] [PMC free article] [PubMed] [Google Scholar]
- van der Meer PF, Reesink HW, Panzer S, Wong J, Ismay S, Keller A, … Benjamin R (2014). Should DEHP be eliminated in blood bags? Vox Sanguinis, 106(2), 176–195. 10.1111/vox.12099 [DOI] [PubMed] [Google Scholar]
- Vandenberg LN (2014). Non-monotonic dose responses in studies of endocrine disrupting chemicals: Bisphenol A as a case study. Dose-Response, 12(2), 259–276. 10.2203/dose-response.13-020.Vandenberg [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vandenberg LN, Chahoud I, Heindel JJ, Padmanabhan V, Paumgartten FJR, & Schoenfelder G (2010). Urinary, circulating, and tissue biomonitoring studies indicate widespread exposure to bisphenol A. Environmental Health Perspectives, 118(8), 1055–1070. 10.1289/ehp.0901716 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vandenberg LN, Hauser R, Marcus M, Olea N, & Welshons WV (2007). Human exposure to bisphenol A (BPA). Reproductive Toxicology, 24(2), 139–177. 10.1016/j.reprotox.2007.07.010 [DOI] [PubMed] [Google Scholar]
- VandeVoort CA, Gerona RR, vom Saal FS, Tarantal AF, Hunt PA, Hillenweck A, & Zalko D (2016). Maternal and fetal pharmacokinetics of oral radiolabeled and authentic bisphenol A in the rhesus monkey. PLoS One, 11(12), e0165410. 10.1371/journal.pone.0165410 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vermij SH, Abriel H, & van Veen TAB (2017). Refining the molecular organization of the cardiac intercalated disc. Cardiovascular Research, 113(3), cvw259. 10.1093/cvr/cvw259 [DOI] [PubMed] [Google Scholar]
- Verstraete S, Vanhorebeek I, Covaci A, Güiza F, Malarvannan G, Jorens PG, & Van den Berghe G (2016). Circulating phthalates during critical illness in children are associated with long-term attention deficit: A study of a development and a validation cohort. Intensive Care Medicine, 42(3), 379–392. 10.1007/s00134-015-4159-5 [DOI] [PubMed] [Google Scholar]
- vom Saal FS, & Welshons WV (2014). Evidence that bisphenol A (BPA) can be accurately measured without contamination in human serum and urine, and that BPA causes numerous hazards from multiple routes of exposure. Molecular and Cellular Endocrinology, 398(1–2), 101–113. 10.1016/j.mce.2014.09.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C, Zhan Y, Wang F, Li H, Xie L, Liu B, … Hua Y (2015). Parental occupational exposures to endocrine disruptors and the risk of simple isolated congenital heart defects. Pediatric Cardiology, 36(5), 1024–1037. 10.1007/s00246-015-1116-6 [DOI] [PubMed] [Google Scholar]
- Wang Q, Cao J, Zhu Q, Luan C, Chen X, Yi X, … Xiao H (2011). Inhibition of voltage-gated sodium channels by bisphenol A in mouse dorsal root ganglion neurons. Brain Research, 1378, 1–8. 10.1016/j.brainres.2011.01.022 [DOI] [PubMed] [Google Scholar]
- Wang Y, Zhu H, & Kannan K (2019). A review of biomonitoring of phthalate exposures. Toxics, 7(2), 21. 10.3390/toxics7020021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang YX, Liu C, Shen Y, Wang Q, Pan A, Yang P, … Zeng Q (2019). Urinary levels of bisphenol A, F and S and markers of oxidative stress among healthy adult men: Variability and association analysis. Environment International, 123, 301–309. 10.1016/j.envint.2018.11.071 [DOI] [PubMed] [Google Scholar]
- Warembourg C, Maitre L, Tamayo-Uria I, Fossati S, Roumeliotaki T, Aasvang GM, … Basagaña X (2019). Early-life environmental exposures and blood pressure in children. Journal of the American College of Cardiology, 74(10), 1317–1328. 10.1016/j.jacc.2019.06.069 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watkins DJ, Sánchez BN, Téllez-Rojo MM, Lee JM, Mercado-García A, Blank-Goldenberg C, … Meeker JD (2017). Phthalate and bisphenol A exposure during in utero windows of susceptibility in relation to reproductive hormones and pubertal development in girls. Environmental Research, 159, 143–151. 10.1016/j.envres.2017.07.051 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei Z, Song L, Wei J, Chen T, Chen J, Lin Y, … Xu S (2012). Maternal exposure to di-(2-ethylhexyl)phthalate alters kidney development through the renin-angiotensin system in offspring. Toxicology Letters, 212(2), 212–221. 10.1016/j.toxlet.2012.05.023 [DOI] [PubMed] [Google Scholar]
- Wetherill YB, Akingbemi BT, Kanno J, McLachlan JA, Nadal A, Sonnenschein C, … Belcher SM (2007). In vitro molecular mechanisms of bisphenol A action. Reproductive Toxicology (Elmsford, NY), 24(2), 178–198. 10.1016/j.reprotox.2007.05.010 [DOI] [PubMed] [Google Scholar]
- Woodruff TJ, Zota AR, & Schwartz JM (2011). Environmental chemicals in pregnant women in the United States: NHANES 2003–2004. Environmental Health Perspectives, 119(6), 878–885. 10.1289/ehp.1002727 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wormuth M, Scheringer M, Vollenweider M, & Hungerbühler K (2006). What are the sources of exposure to eight frequently used phthalic acid esters in Europeans? Risk Analysis, 26(3), 803–824. 10.1111/j.1539-6924.2006.00770.x [DOI] [PubMed] [Google Scholar]
- Wowkonowicz P, & Kijeńska M (2017). Phthalate release in leachate from municipal landfills of Central Poland. PLoS One, 12(3), e0174986. 10.1371/journal.pone.0174986 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu S, Yang W, Yeh C, & Huang H (2012). The inhibition by di(2-ethylhexyl)-phthalate of Erg-mediated K+ current in pituitary tumor (GH3) cells. Archives of Toxicology, 86(5), 713–723. 10.1007/S00204-012-0805-7 [DOI] [PubMed] [Google Scholar]
- Yan S, Chen Y, Dong M, Song W, Belcher SM, & Wang HS (2011). Bisphenol A and 17β-estradiol promote arrhythmia in the female heart via alteration of calcium handling. PLoS One, 6(9), e25455. 10.1371/journal.pone.0025455 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan S, Song W, Chen Y, Hong K, Rubinstein J, & Wang H-S (2013). Low-dose bisphenol A and estrogen increase ventricular arrhythmias following ischemia-reperfusion in female rat hearts. Food and Chemical Toxicology, 56, 75–80. 10.1016/j.fct.2013.02.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang YJ, Hong Y-C, Oh S-Y, Park M-S, Kim H, Leem J-H, & Ha E-H (2009). Bisphenol A exposure is associated with oxidative stress and inflammation in postmenopausal women. Environmental Research, 109(6), 797–801. 10.1016/j.envres.2009.04.014 [DOI] [PubMed] [Google Scholar]
- Ye H, Ha M, Yang M, Yue P, Xie Z, & Liu C (2017). Di2-ethylhexyl phthalate disrupts thyroid hormone homeostasis through activating the Ras/Akt/TRHr pathway and inducing hepatic enzymes. Scientific Reports, 7(1), 40153. 10.1038/srep40153 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yi H, Xue L, Guo M-X, Ma J, Zeng Y, Wang W, … Li W-X (2010). Gene expression atlas for human embryogenesis. FASEB Journal, 24(9), 3341–3350. 10.1096/fj.10-158782 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang T, Sun H, & Kannan K (2013). Blood and urinary bisphenol A concentrations in children, adults, and pregnant women from China: Partitioning between blood and urine and maternal and fetal cord blood. Environmental Science and Technology, 47(9), 4686–4694. 10.1021/es303808b [DOI] [PubMed] [Google Scholar]
- Zhang X, Tang S, Qiu T, Hu X, Lu Y, Du P, … Giesy JP (2020). Investigation of phthalate metabolites in urine and daily phthalate intakes among three age groups in Beijing, China. Environmental Pollution (Barking, Essex: 1987), 260, 114005. 10.1016/j.envpol.2020.114005 [DOI] [PubMed] [Google Scholar]
- Zhao B, Chu Y, Huang Y, Hardy DO, Lin S, & Ge R-S (2010). Structure-dependent inhibition of human and rat 11β-hydroxysteroid dehydrogenase 2 activities by phthalates. Chemico-Biological Interactions, 183(1), 79–84. 10.1016/j.cbi.2009.09.014 [DOI] [PubMed] [Google Scholar]
- Zota AR, Calafat AM, & Woodruff TJ (2014). Temporal trends in phthalate exposures: Findings from the National Health and Nutrition Examination Survey, 2001–2010. Environmental Health Perspectives, 122, 235–241. 10.1289/EHP.1306681 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.