The Relationship between Phthalates and Diabetes: A Review: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Since the beginning of their production, in the 1930s, phthalates have been widely used in the plastics industry to provide durability and elasticity to polymers that would otherwise be rigid, or as solvents in hygiene and cosmetic products. Taking into account their wide range of applications, it is easy to understand why their use has been increasing over the years, making them ubiquitous in the environment. This way, all living organisms are easily exposed to these compounds, which have already been classified as endocrine disruptor compounds (EDC), affecting hormone homeostasis. Along with this increase in phthalate-containing products, the incidence of several metabolic diseases has also been rising, namely diabetes. 

  • plasticizers
  • phthalates
  • di-(2-ethylhexyl) phthalate (DEHP)
  • butylbenzyl phthalate (BBzP)
  • Diabetes Mellitus
  • metabolic diseases

1. Gestational Diabetes Mellitus

1.1. Epidemiological Studies

In the USA, three different studies using the same cohort, the LIFECODES pregnancy cohort, presented different outcomes. With the aim of analyzing the link between exposure to phthalates and risk factors for GDM, the authors quantified the levels of phthalates in the urine of 350 pregnant women and related them to first trimester body mass index (BMI), gestational weight gain (GWG), and second trimester glucose levels. The results showed a positive association between MEP and GWG and impaired glucose tolerance, and a negative one regarding MBP, MCPP, and ∑DEHP levels, and excessive GWG, continuous blood glucose, and impaired glucose tolerance, respectively [1]. When evaluating phthalates metabolites separately and combined in the first and second trimesters of pregnancy, the same research group found that phthalates and their mixtures may be involved in maternal glucose metabolism, since in the first trimester there was a negative correlation between MBP, MCNP, and MCPP levels and GDM and impaired glucose tolerance, while a positive association was found for MiBP and MHBP levels and impaired glucose tolerance and GDM, respectively. Moreover, the mixtures of phthalates presented similar results to the individual phthalate effects [2]. On the other hand, Noor et al. found no association between maternal urinary phthalate metabolites and infants’ birth weight from mothers with higher levels of glucose during pregnancy [3].
Reporting on a different cohort, similar results were found by Shaffer and colleagues, with urinary MEP levels being associated with GDM. 705 pregnant women provided one spot urine sample in the first and third trimesters, which were compared with GDM screening (performed between gestational weeks 24 and 28). Apart from the confirmed relationship between MEP and GDM, the levels of MBP and MCOP were associated with impaired glucose tolerance, and MCPP had a negative association with GDM. Moreover, the authors also found a possible link regarding race/ethnicity [4]. This matter must be studied further, but it seems to be in accordance with the numerous hypotheses regarding population variability.
In a different perspective, James-Todd et al. performed another prospective study, this time studying 245 pregnant women who attended a fertility clinic, where urinary DEP and DiBP metabolites (MEP and MiBP) were found to be increased and decreased, respectively, in women with higher glucose levels. It is of note that the sources of exposures of these two phthalates were predominantly different, with DEP being found in personal care products while DiBP was found in food and consumer products, and this was a subfertile population, with a higher risk of glucose dysregulation during pregnancy [5]. Reporting on the same cohort, Bellavia et al. aimed to understand the link between the use of personal care products containing phthalates and the occurrence of GDM. For this, 233 women answered a questionnaire regarding the use of personal care products (concerning the previous 24 h), and blood samples were collected at the end of the second trimester. The authors found a correlation between increased levels of blood glucose and bar soap, deodorant, and lotion, which, from other statements, are related to phthalates [6].
A longitudinal cohort involving 3269 women that provided urine and serum samples at each trimester of pregnancy found that early pregnancy exposure to phthalates may be involved with an increased risk of GDM. Specifically, higher urinary concentrations of MBP, MMP, MEOHP, and MEHHP were associated with increased blood glucose in the first trimester [7]. Three different Chinese case-control studies reported an association between phthalate exposure during pregnancy and the occurrence of GDM. Comparing women with and without GDM, Liang and colleagues also found a relationship with phthalate exposure, since there were higher levels of MEHP in the GDM cases. Moreover, MMP, MEP, MiBP, MECPP, and MEOHP have also been linked to fasting blood glucose and insulin, and insulin resistance index, which are parameters related to GDM [8]. A different cohort enrolled 676 women divided in two groups, with and without GDM, for whom urine samples were collected in early pregnancy. The urinary levels of MnOP, MBzP, MEOHP, and MECPP were all significantly associated with GDM; however, MEOHP was found to be independently linked to GDM at concentrations higher than 15.6 µg/L. Considering these results, almost 25% of the participants had an increased risk for GDM due to MEOHP concentrations [9]. More recently, relying on phthalate levels quantified in the serum of 201 women (at the time of delivery), Wang and colleagues found that MBP was the most abundant metabolite in this population sample, and they also showed a significant association between MBP and MiBP levels and the 2 h blood glucose, which in turn is related to GDM [10]. Thus, this study also shows a correlation between phthalate exposure during pregnancy and the occurrence of GDM. A different study also reported an association between phthalates and GDM in early pregnancy. Serum phthalate metabolites measured during 10–17 weeks of gestation (for women with singleton male pregnancies) showed a positive relationship between MiBP and GDM, and the quantification of the MEHP and MCOP of pregnant women without GDM was related to stimulated blood glucose levels [11].
In a different approach, Martinez-Ibarra et al. reported on a Mexican population of women with and without GDM. This time, the serum levels of three of the four evaluated miRNAs related to GDM were associated with urinary concentrations of different phthalate metabolites (MBzP, MBP, MEHP, and MiBP). It is important to note that almost 100% of the urine samples were positive for phthalate levels [12] and were several times higher than the ones reported in other studies [3][4]. These differences might be due to the different populations under study, considering that that Noor et al. and Shaffer et al. investigated American women while Martinez-Ibarra and colleagues investigated a Mexican cohort, which in turn is a country that has not yet regulated the use of phthalates, so the Mexican population is much more exposed to these types of products, and consequently at greater risk [12]. In a different Mexican cohort, 618 women provided urine samples in the second and/or third trimesters of pregnancy, which were then related to metabolic biomarkers in blood samples collected 4–5 and/or 6–8 years after delivery. In addition to the connection with some lipid parameters, the results showed a positive association between MECPTP and ∑DBP and increased glucose and insulin levels, insulin resistance, and glycosylated hemoglobin (HbA1c). These are very interesting findings, since a pre-natal exposure to phthalates seems be associated with long-term adverse health effects in the mother [13]. It has already been pointed out by other researchers that GDM, which may be due to phthalate exposure, can result in maternal and offspring health problems later in life [14][15]; however, these findings suggest that the exposure to these compounds during pregnancy, without causing any disturbance during this sensitive period, appears to be associated with metabolic changes in the future.
A different investigation also found an association between MBP, MiBP, and MEHP levels and GDM, though this entry analyzed newborn exposure to phthalates in utero by quantifying their levels in meconium, and the association was only found for mothers of male fetuses [16].
Placental corticotropin-releasing hormone (pCRH), a placenta-produced neuropeptide that greatly increases during pregnancy, has been linked to hypertension in pregnancy, depression, and trauma. Bearing this in mind, and that exposure to phthalates can be even more harmful in pregnant women with pre-existing complications, a cohort of 1018 participants was gathered to find a negative association between phthalate mixtures and pCRH levels in women with GDM, particularly in the third trimester. These results suggest that phthalates affect the production of pCRH differently throughout pregnancy [17].

1.2. Experimental Studies

Until now, there has only been one study performed in animals relating phthalates to GDM, which may be a window for the mechanistic pathways. The authors managed to induce GDM in Sprague Dawley rats with the administration of DBP and streptozotocin (STZ), a new and more relevant model for GDM. Moreover, in vitro and in vivo studies demonstrated that exposure to DBP led to FoxM1 downregulation by pSTAT1, resulting in the decreased viability and apoptosis of β-cells, culminating in GDM [18].

1.3. Possible Mechanisms

Insulin resistance and inflammatory factors have been considered as the main factors responsible for GDM pathophysiology [19]. Yet, the increasing exposure to environmental contaminants has suggested phthalates as risk factors for several diseases, including GDM, either directly or by acting on GDM triggers. For instance, it is known that TNF-α is related to GDM by inducing adipocyte lipolysis, which can lead to a decreased insulin sensitivity by peripheral tissues, thus being considered as a biomarker for insulin resistance in pregnancy [10][20]. Using a network-based approach to understand the mechanism behind the link between DEHP and GDM, Zhang and colleagues found that exposure to DEHP may increase TNF-α expression, which suppresses GLUT4 (glucose transporter protein), as well as glucose uptake, disturbing glucose homeostasis and culminating in GDM [21]. In addition, phthalates have already been described to interact with peroxisome proliferator-activated receptors (PPAR), nuclear receptors related to glucose and lipid metabolism [22][23]. Of the main isoforms of these receptors, PPARα, is the one implicated in β-cell functioning, being responsible for insulin secretion. Thus, the interaction between phthalates and PPARα may disturb blood glucose homeostasis [2]. Moreover, PPARγ, which is related to adipogenesis, is also activated by phthalates, promoting obesity, which is an important factor for the occurrence of GDM [10]. Oxidative stress has also been suggested as a possible mechanism for GDM. In addition to being related to increased reactive oxygen species (ROS), phthalates and homeostasis model assessment-estimated insulin resistance (HOMA-IR) have been associated with a biomarker for oxidative stress (malondialdehyde—MDA) [10][24][25]. As the name implies, phthalates as EDCs may disrupt the endocrine system by interfering with the action of hormones [26]. Specifically, phthalates have been described as agonists of the estrogen receptors (ER), and estrogens are linked to insulin resistance; thus, phthalates can promote insulin signaling through ERα mediated pathways, which, when sustained, may lead to excess insulin release, β-cell exhaustion, and peripheral insulin resistance [4].
Although all of these have already been described as pathways for impaired glucose metabolism and insulin resistance, it is still unclear how phthalates promote the development of GDM; so, more studies are needed to unravel the actual mechanisms.

2. Type 1 Diabetes Mellitus

2.1. Epidemiological Studies

Very few studies have linked phthalates to T1DM so far, mainly epidemiological ones, considering the modest incidence of the disease [27]. Nevertheless, one study performed in Portugal evaluated the urinary concentration of phthalates in children with new-onset and existing T1DM and controls. The authors found no significant association between phthalate levels in the T1DM cases compared to controls; however, there was a higher concentration of MiBP in children with new-onset T1DM [28]. Considering that this relied on a small population sample, it is possible to hypothesize that resorting to a larger sample could have significant results for the relationship between phthalates and T1DM.

2.2. Experimental Studies

In animal models, phthalates effects have been evaluated together with bisphenol-A (BPA). When exposing non-obese diabetic (NOD) mice to relevant human doses of BPA and a mixture of phthalates, it was found that phthalates did not accelerate the development of T1DM; in fact, phthalates seem to diminish the effects of BPA on the number and function of macrophages, but not in insulitis development. This possible hormesis effect (protective role) of phthalates may be due to the typical non-monotonic curve, in which high doses may decrease the development of diabetes [29]. A different study showed that phthalate metabolites have less capacity to affect insulin secretion and viability in the rat pancreatic β-cell line (INS-1E) than BPA [30]. Although it is important to study the effects of a mixture of EDCs, since human beings are exposed to several contaminants at the same time, these reports hamper the study of the relationship and mechanisms of action of phthalates alone in the development of T1DM.
There is a huge gap regarding EDCs’ effects on the development of T1DM. Therefore, in addition to the need for experimental studies to understand how phthalates affect β-cells, epidemiological studies with larger sample sizes are also essential to understand whether phthalates are really involved in the development of T1DM.

2.3. Possible Mechanisms

Several pathogenic mechanisms have been pointed out as possible T1DM triggers by EDCs, including effects on β-cells, immunomodulation, epigenetics, microbiota, and vitamin D [27][31]. As previously shown, phthalates were already reported to directly affect rat β-cell secretion and viability [30]. Moreover, it is known that the activation of estrogen receptors can lead to glucose-induced insulin synthesis, its secretion by β-cells, and their survival from pro-apoptotic stimuli [27][32], and so considering that phthalates can act on these receptors [33], they can also indirectly affect β-cells through the estrogen receptors. EDCs may also affect the immune system by modulating the function of immune cells and cytokine levels, which may result in T1DM [27][31]. In experimental studies, pre-natal exposure to low doses of phthalates has been linked to epigenetic changes in genes related to the immune response in the offspring, which can promote autoimmunity [27][34]. In addition, the gut microbiota is important for a healthy immune system; however, a change in its composition has been associated with the development of T1DM [35]. Considering that phthalates have been found to alter the gut microbiota in a rodent model [31][36], it is a possible mechanism for T1DM promotion. Additionally, a vitamin D deficit and decreased intracellular calcium levels have also been related to T1DM [31][37], and, in turn, phthalates have been associated with changes in these two parameters. Specifically, urinary levels of phthalate metabolites were negatively related to circulating 25-hydroxyvitamin D [31][38][39], and phthalates have been involved in alterations in calcium handling levels, and calcium channel activity [40][41][42][43][44]. Thus, phthalates may be involved in T1DM development through vitamin D and calcium channel changes. Although all of these studies provide some evidence of the association between exposure to phthalates and T1DM and the possible mechanisms involved, more studies are needed, either experimental or epidemiological, to understand the actual effects of phthalates in this autoimmune disease.

3. Type 2 Diabetes Mellitus

3.1. Epidemiological Studies

In order to analyze how pre-natal exposure affects metabolic risk factors during childhood, 757 children from women that provided urine samples during pregnancy (one in each trimester) were examined for blood lipid and glucose parameters at approximately 10 years of age. The authors found an association between second and third trimester phthalate levels and lower glucose and higher triglyceride concentrations in boys, respectively [45]. These results suggest a gender-specific relationship with phthalate exposure that could be related to metabolic impairment. In an attempt to discover the connection between DEHP substitutes and insulin resistance, one spot urine sample was collected from 356 fasting adolescents (12–19 years old). In addition to finding a correlation with DEHP as expected, insulin resistance was also related to DINP concentrations [46]. On the other hand, no connection between urinary phthalates and insulin resistance was found in a population of 107 Danish children (mean age of 12 years) [47]. Nevertheless, a different study has shown that age and gender may play a role in the correlation between phthalate exposure and insulin resistance. In a young Taiwanese population, from adolescents to young adults, a link between elevated urinary levels of MEHP and incidence of insulin resistance was shown to occur in young adults (20–30 years old), but not in adolescents (12–19 years old). Moreover, in the same age range, MEHP was also related to decreased testosterone levels in males, suggesting that testosterone levels are inversely related to insulin resistance [48].
Analyzing a broader age range (12–79 years old), participants were asked to provide a one-time mid-stream urine sample for phthalate measurement, and one blood sample for insulin and glucose parameters. Associations were found between MBzP, MiBP, MCPP, MEHP, MEHHP, and ∑DEHP with HbA1c levels, and between DEHP metabolites with higher amounts of insulin, insulin resistance and fasting glucose, reduced glucose control, and β-cell function, suggesting an involvement of phthalates in pre-diabetes [49]. Considering the straight connection between diabetes mellitus and obesity, Dirinck et al. analyzed the correlation between urinary phthalate concentrations from a 24 h urine sample and glucose metabolism in an obese/overweight population (123 adults, aged between 18 and 84 years). There was an association between phthalate metabolites and several metabolic biomarkers related to insulin; specifically, there was a positive association with resistance and impaired β-cell function, and a negative one with insulin sensitivity, even after correction for BMI. However, opposite to the study conducted by Dales et al., there was no association with HbA1c levels. The results suggest phthalates as being higher risk factors for diabetes than obesity [50]. There was also a relationship between increased urinary levels of phthalate metabolites and the incidence of T2DM, when examining a much larger population sample (n = 3781), and, despite being separated by gender, no association was found between male and female results [51]. On the other hand, in a Chinese case-control study, differences among gender, age, and BMI were found. A total of 500 participants with and without T2DM provided one spot urine sample, and T2DM participants had higher and more significant levels of MEHHP, MEOHP, MEHP, MCPP, MiBP, MMP, and ∑DEHP and decreased levels of MECPP and MCMHP. When stratified, the associations between phthalate metabolites and T2DM, HbA1c levels, and fasting glucose were more prominent for participants younger than 55 years old, with BMI inferior to 25 Kg/m2, and males older than 55 years old, respectively [52]. Similarly, in a population sample of 2330 participants from Shanghai (mean age of 53 years), Dong and colleagues also found a significant association between urinary phthalate levels and T2DM in men only, specifically, MEOHP, MEHHP, and MECPP [53]. Using men only, a case-control study of 100 diabetic and 50 non-diabetic participants found higher concentrations of MEP, MEOHP, and MBP in the cases of T2DM, with MEP and MBP being related to HOMA-IR and C-peptide, which are linked to insulin resistance [54]. In accordance with these results, an Australian cohort of 1504 men (39–84 years old) also found an association between phthalate exposure and T2DM [55]. These previous studies show the importance and the need for a sex-specific assessment across all ages, considering that phthalates are known to interact with androgen and estrogen receptors.
Nevertheless, despite gender-related differences, some older epidemiological studies have also shown a relationship between exposure to phthalates and T2DM in women. Upon investigating different populations, increased urinary levels of MBP, MiBP, MBzP, MCPP, ∑DEHP, and ∑DBP were found to be related to T2DM [56][57][58]. In a different perspective, 618 women provided urine samples in the second and third trimesters of pregnancy, which were compared with metabolic parameters measured in blood several years later. There was a positive association between urinary phthalate (mainly MECPTP and DBP) levels and insulin resistance, considering the high amounts of plasma glucose, insulin, HOMA-IR, and HbA1c% [13].
In an attempt to understand the role of metabolism in the development of T2DM due to phthalate exposure, a case-control study of 60 diabetic and 60 non-diabetic participants was performed by Duan et al.. From the fasting blood samples collected, metabolites and metabolic pathways were investigated between cases and controls and compared with urinary phthalate concentrations. Overall, there was an association between phthalate metabolites and galactose, amino acid, and pyrimidine metabolism in T2DM subjects [59].

3.2. Experimental Studies

Despite the scarcity of experimental studies regarding the effect of prenatal exposure on the development of GDM, as previously mentioned, there is more information on the metabolic effects that this type of exposure has on offspring. Three different studies related gestational DEHP exposure to glucose parameters in adults of the F1 generation. To achieve the goals, female Wistar rats were exposed to different concentrations of DEHP (1, 10 and 100 mg/Kg/day) from gestational day (GD) 9 to GD 21 [60] and to postnatal day (PND) 21 [61][62]. In the first study, Rajesh et al. found that DEHP induced changes in the expression of genes related to insulin gene transcription and a glucose sensing mechanism, culminating in β-cell dysfunction [60]. The other two investigations also evaluated the lactation period and analyzed only the effects observed in adult male offspring. The results showed that DEHP exposure led to impaired regulation of the GLUT2 gene and insulin signal transduction, leading to decreased glucose tolerance, insulin resistance, and hyperglycemia [61][62]. All the events reported from these three studies may lead to T2DM in offspring.
In a different perspective, male Balb/c mice were exposed to three different doses of DBP for 7 weeks, in which the highest DBP dose led to decreased insulin secretion and glucose intolerance. Moreover, when using STZ and a high-fat diet to induce T2DM, the exposure to DBP worsened the affected parameters and induced insulin resistance and T2DM-related organ lesions. The T2DM mouse model also presented a decreased PI3K/AKT signaling pathway and increased pancreatic GLUT2, which may be implicated in the DBP mechanism [63].
Two studies from the same research group evaluated the effects of DEHP in adolescent (3-week-old) female [64] and male [65] ICR mice with and without T2DM. Upon the administration of four different concentrations of DEHP for 3 weeks, glucose and lipid parameters, as well as cardiovascular risk were analyzed in the different study groups. The results showed that both T2DM male and female mice were more susceptible to DEHP exposure than normal mice; however, T2DM female mice proved to be more sensitive than their male counterparts, with an increased risk of suffering from T2DM, metabolic and cardiovascular disorders, and hepatotoxicity. Moreover, it was also suggested that DEHP activates Jun-N-terminal kinase (JNK), promoting the apoptosis of hepatic cells and the inhibition of insulin sensitivity, which may lead to metabolic disorders [64][65]. The results of these studies also allowed the authors to assume the gender differences caused by the exposure to DEHP with the incidence in female mice, which are in accordance with other reports; however, the epidemiological studies relating sex-specific differences tend to show a higher incidence in men [45][48][52][53].
Upon the exposure of the pancreatic β-cell line (INS-1) to a range of concentrations (0.001–10 µM) of MEHP and MBP for 24, 48, and 72 h, there was cell viability decrease and oxidative stress increase with mRNA expression changes for genes related to pancreatic β-cell function and apoptosis. These results imply that MEHP and MBP might affect β-cell function, which may lead to insulin resistance and consequent T2DM [66].
As was previously stated, the study conducted by Weldingh and co-workers reported an inferior potency of MEHP, MBP, and MiBP compared to BPA in affecting insulin secretion in INS-1E cells. However, it is important to note that phthalates in the serum are found in much higher concentrations than BPA, and so a new approach closer to real human exposure is needed [30]. In a different study using human pancreatic β-cells (1.1B4), a 24 h exposure to low concentrations of MEP (1–1000 nM) led to increased insulin secretion, possibly involving ERα, PPARγ, and PDX-1 (pancreatic and duodenal homeobox 1), which are related to β-cell function and survival [67]. Al-Abdulla and colleagues also demonstrated that exposure to DEHP led to impaired insulin secretion in both human and murine pancreatic β-cells [68].
Several authors have been investigating the role of oxidative stress in phthalate-induced T2DM. In an in vivo study, male Swiss albino mice (8-week-old) were treated with DEP for 3 months, after which serum, liver, and epididymal adipose tissue were removed for further analysis. Besides concluding that this chronic low-level exposure to DEP induced impaired insulin signaling in both hepatocytes and adipocytes, the authors also discovered a great increase in NOX2 (NADPH oxidase 2), which is involved in the generation of ROS [69]. Differentiated human preadipocytes were used by Schaffert et al. to analyze the effects of 20 plasticizers in PPARγ. In preadipocytes, DINP and DPHP (DEHP substitutes) metabolites activated PPARγ, inducing lipid accumulation and adipogenesis, while in mature adipocytes these compounds promoted lipid storage, oxidative stress, and impaired adipokine release related to insulin resistance [70]. Two in vitro studies on the same cell line (INS-1) found that both DEHP and DBP exert their adverse effects through oxidative stress. Specifically, DEHP acts in the lysosome–mitochondrial axis pathway, increasing ROS production and leading to DNA damage and p53 and ATM activation [71]. Additionally, on the other hand, DBP altered PDX-1 and GLUT2 levels, leading to reduced insulin synthesis and secretion through the mitochondrial apoptotic pathway and oxidative stress [72].
Viswanathan and collaborators analyzed how DEHP and MEHP affected GLUT4 in a cell model of the skeletal muscle (L6 myotubes). After the incubation of the cells with 50 and 100 µM DEHP and MEHP (24 h), the authors observed changes in GLUT4 levels and translocation, as well as in insulin signaling molecules [73]. Similarly, GLUT4 was also shown to be affected by DEHP, either in in vivo or in vitro experiments. Moreover, Sprague Dawley rats exposed to DEHP exhibited liver damage, glucose, and insulin tolerance, while in a human hepatocyte cell line (L02), DEHP interacted with PPARγ, increasing ROS levels [74]. These studies emphasize the role of PPARγ and oxidative stress in the development of T2DM induced by phthalates.
Some investigations have also demonstrated a protective or reversible role of certain molecules or compounds towards damaging phthalate effects. In the study of Deng and colleagues, when a selective insulin receptor activator, demethylasterriquinone B1 (DMAQ-B1), was administered to mice, there was a decrease in the adverse effects of DBP regarding insulin deficiency and resistance [63]. Additionally, according to She et al., pyrroloquinoline quinone (PQQ)—a compound with anti-inflammatory, anti-oxidative, hepato-, and cardioprotective properties—has the capacity to protect INS-1 cells from the adverse effects promoted by DEHP [71]. In a study that combined computational analysis with in vivo experiments, after finding that the conjoint action of DEHP, DBP, and BPA led to T2DM in rats through oxidative stress and apoptosis, a protective role of a mixture of probiotics, regarding redox properties in the pancreas, was also observed [75].

3.3. Possible Mechanisms

As previously stated, some hypotheses have emerged for the phthalates’ mechanism of action, both in epidemiological and experimental studies. So far, oxidative stress has been the most studied and with more positive evidence, but inflammatory markers, impaired adiponectin, and β-cell dysfunction have also been gaining attention.
Pancreatic β-cell dysfunction is one of the main causes for T2DM development, and some studies have shown that phthalates affect these cells through different pathways [60][66][67]. Maternal exposure to DEHP was shown to promote disrupt β-cell function in the rat offspring by affecting the glucose sensing mechanism and insulin gene transcription [60]. On the other hand, through the activation of ERα, PPARγ, and PDX-1, MEP increases insulin secretion, which, as previously mentioned, with time will progress to the failure and loss of the pancreatic β-cells [67]. This is in accordance with previous mentioned studies, since estrogens are related to insulin resistance, and thus phthalates can affect insulin signaling through Erα-mediated pathways [4]. In addition, MEHP and MBP were shown to affect the expression of β-cell-related genes and promote oxidative stress [66]. In fact, oxidative stress has been suggested as one of the possible mechanisms for the development of T2DM by exposure to phthalates, either in experimental or epidemiological studies [69][71][72]. DEP and DEHP are involved in the generation of ROS by increasing NOX2 [69] and MDA levels [71]. In the mechanism proposed by She et al., DEHP promoted lysosomal disruption in INS-1 cells, decreasing mitochondrial membrane potential, and thus increasing ROS production and p53 and ATM activation, which are related to DNA damage [71].
Other molecular pathways have been implicated and suggested as mechanisms for insulin resistance and T2DM. DEHP was shown to activate JNK, affecting Bcl-2 and Bax, leading to apoptosis and the inhibition of the insulin sensitivity of mice hepatic cells [64][65]. Moreover, both DEHP and DBP inhibited the PI3K/AKT signaling pathway and led to impaired glucose transporters (GLUT2 and GLUT4), resulting in decreased glucose tolerance, insulin resistance, and hyperglycemia [61][62][63][64][72][73]. Moreover, there seems to be a sex-specific effect of DEHP, since higher risks for T2DM were demonstrated in female mice [64].
Phthalates are considered as peroxisome proliferator activators, and many of their adverse effects may occur through the PPARs [76]. In human experimental studies, the mechanism for phthalate-induced insulin resistance seems to involve the activation of PPARγ and oxidative stress. DEHP and its substitutes, DINP and DPHP, promoted the activation of PPARγ in human preadipocytes [70], while in hepatocytes only DEHP activated PPAR [74]. Moreover, in both hepatocytes and adipocytes, the compounds induced oxidative stress, thus disturbing lipid and glucose metabolism leading to insulin resistance [70][74].

This entry is adapted from the peer-reviewed paper 10.3390/metabo13060746

References

  1. James-Todd, T.M.; Meeker, J.D.; Huang, T.; Hauser, R.; Ferguson, K.K.; Rich-Edwards, J.W.; McElrath, T.F.; Seely, E.W. Pregnancy urinary phthalate metabolite concentrations and gestational diabetes risk factors. Environ. Int. 2016, 96, 118–126.
  2. James-Todd, T.; Ponzano, M.; Bellavia, A.; Williams, P.L.; Cantonwine, D.E.; Calafat, A.M.; Hauser, R.; Quinn, M.R.; Seely, E.W.; McElrath, T.F. Urinary phthalate and DINCH metabolite concentrations and gradations of maternal glucose intolerance. Environ. Int. 2022, 161, 107099.
  3. Noor, N.; Ferguson, K.K.; Meeker, J.D.; Seely, E.W.; Hauser, R.; James-Todd, T.; McElrath, T.F. Pregnancy phthalate metabolite concentrations and infant birth weight by gradations of maternal glucose tolerance. Int. J. Hyg. Environ. Health 2019, 222, 395–401.
  4. Shaffer, R.M.; Ferguson, K.K.; Sheppard, L.; James-Todd, T.; Butts, S.; Chandrasekaran, S.; Swan, S.H.; Barrett, E.S.; Nguyen, R.; Bush, N.; et al. Maternal urinary phthalate metabolites in relation to gestational diabetes and glucose intolerance during pregnancy. Environ. Int. 2019, 123, 588–596.
  5. James-Todd, T.M.; Chiu, Y.H.; Messerlian, C.; Minguez-Alarcon, L.; Ford, J.B.; Keller, M.; Petrozza, J.; Williams, P.L.; Ye, X.; Calafat, A.M.; et al. Trimester-specific phthalate concentrations and glucose levels among women from a fertility clinic. Environ. Health 2018, 17, 55.
  6. Bellavia, A.; Minguez-Alarcon, L.; Ford, J.B.; Keller, M.; Petrozza, J.; Williams, P.L.; Hauser, R.; James-Todd, T.; Team, E.S. Association of self-reported personal care product use with blood glucose levels measured during pregnancy among women from a fertility clinic. Sci. Total Environ. 2019, 695, 133855.
  7. Gao, H.; Zhu, B.B.; Huang, K.; Zhu, Y.D.; Yan, S.Q.; Wu, X.Y.; Han, Y.; Sheng, J.; Cao, H.; Zhu, P.; et al. Effects of single and combined gestational phthalate exposure on blood pressure, blood glucose and gestational weight gain: A longitudinal analysis. Environ. Int. 2021, 155, 106677.
  8. Liang, Q.X.; Lin, Y.; Fang, X.M.; Gao, Y.H.; Li, F. Association Between Phthalate Exposure in Pregnancy and Gestational Diabetes: A Chinese Cross-Sectional Study. Int. J. Gen. Med. 2022, 15, 179–189.
  9. Chen, W.; He, C.; Liu, X.; An, S.; Wang, X.; Tao, L.; Zhang, H.; Tian, Y.; Wu, N.; Xu, P.; et al. Effects of exposure to phthalate during early pregnancy on gestational diabetes mellitus: A nested case-control study with propensity score matching. Environ. Sci. Pollut. Res. Int. 2022, 30, 33555–33566.
  10. Wang, H.; Chen, R.; Gao, Y.; Qu, J.; Zhang, Y.; Jin, H.; Zhao, M.; Bai, X. Serum concentrations of phthalate metabolites in pregnant women and their association with gestational diabetes mellitus and blood glucose levels. Sci. Total Environ. 2023, 857, 159570.
  11. Fisher, B.G.; Frederiksen, H.; Andersson, A.M.; Juul, A.; Thankamony, A.; Ong, K.K.; Dunger, D.B.; Hughes, I.A.; Acerini, C.L. Serum Phthalate and Triclosan Levels Have Opposing Associations With Risk Factors for Gestational Diabetes Mellitus. Front. Endocrinol. Lausanne 2018, 9, 99.
  12. Martinez-Ibarra, A.; Martinez-Razo, L.D.; Vazquez-Martinez, E.R.; Martinez-Cruz, N.; Flores-Ramirez, R.; Garcia-Gomez, E.; Lopez-Lopez, M.; Ortega-Gonzalez, C.; Camacho-Arroyo, I.; Cerbon, M. Unhealthy Levels of Phthalates and Bisphenol A in Mexican Pregnant Women with Gestational Diabetes and Its Association to Altered Expression of miRNAs Involved with Metabolic Disease. Int. J. Mol. Sci. 2019, 20, 3343.
  13. Wu, H.; Just, A.C.; Colicino, E.; Calafat, A.M.; Oken, E.; Braun, J.M.; McRae, N.; Cantoral, A.; Pantic, I.; Pizano-Zarate, M.L.; et al. The associations of phthalate biomarkers during pregnancy with later glycemia and lipid profiles. Environ. Int. 2021, 155, 106612.
  14. Hill, M.; Parizek, A.; Simjak, P.; Koucky, M.; Anderlova, K.; Krejci, H.; Vejrazkova, D.; Ondrejikova, L.; Cerny, A.; Kancheva, R. Steroids, steroid associated substances and gestational diabetes mellitus. Physiol. Res. 2021, 70, S617–S634.
  15. Filardi, T.; Panimolle, F.; Lenzi, A.; Morano, S. Bisphenol A and Phthalates in Diet: An Emerging Link with Pregnancy Complications. Nutrients 2020, 12, 525.
  16. Guo, J.; Wu, M.; Gao, X.; Chen, J.; Li, S.; Chen, B.; Dong, R. Meconium Exposure to Phthalates, Sex and Thyroid Hormones, Birth Size and Pregnancy Outcomes in 251 Mother-Infant Pairs from Shanghai. Int. J. Environ. Res. Public Health 2020, 17, 7711.
  17. Barrett, E.S.; Corsetti, M.; Day, D.; Thurston, S.W.; Loftus, C.T.; Karr, C.J.; Kannan, K.; LeWinn, K.Z.; Smith, A.K.; Smith, R.; et al. Prenatal phthalate exposure in relation to placental corticotropin releasing hormone (pCRH) in the CANDLE cohort. Environ. Int. 2022, 160, 107078.
  18. Chen, M.; Zhao, S.; Guo, W.H.; Zhu, Y.P.; Pan, L.; Xie, Z.W.; Sun, W.L.; Jiang, J.T. Maternal exposure to Di-n-butyl phthalate (DBP) aggravate gestational diabetes mellitus via FoxM1 suppression by pSTAT1 signalling. Ecotoxicol. Environ. Saf. 2020, 205, 111154.
  19. John, C.M.; Mohamed Yusof, N.I.S.; Abdul Aziz, S.H.; Mohd Fauzi, F. Maternal Cognitive Impairment Associated with Gestational Diabetes Mellitus-A Review of Potential Contributing Mechanisms. Int. J. Mol. Sci. 2018, 19, 3894.
  20. Friedman, J.E.; Kirwan, J.P.; Jing, M.; Presley, L.; Catalano, P.M. Increased skeletal muscle tumor necrosis factor-alpha and impaired insulin signaling persist in obese women with gestational diabetes mellitus 1 year postpartum. Diabetes 2008, 57, 606–613.
  21. Zhang, T.; Wang, S.; Li, L.; Zhu, A.; Wang, Q. Associating diethylhexyl phthalate to gestational diabetes mellitus via adverse outcome pathways using a network-based approach. Sci. Total Environ. 2022, 824, 153932.
  22. Sarath Josh, M.K.; Pradeep, S.; Vijayalekshmi Amma, K.S.; Balachandran, S.; Abdul Jaleel, U.C.; Doble, M.; Spener, F.; Benjamin, S. Phthalates efficiently bind to human peroxisome proliferator activated receptor and retinoid X receptor alpha, beta, gamma subtypes: An in silico approach. J. Appl. Toxicol. 2014, 34, 754–765.
  23. Desvergne, B.; Feige, J.N.; Casals-Casas, C. PPAR-mediated activity of phthalates: A link to the obesity epidemic? Mol. Cell Endocrinol. 2009, 304, 43–48.
  24. Kim, J.H.; Park, H.Y.; Bae, S.; Lim, Y.H.; Hong, Y.C. Diethylhexyl phthalates is associated with insulin resistance via oxidative stress in the elderly: A panel study. PLoS ONE 2013, 8, e71392.
  25. Cho, Y.J.; Park, S.B.; Han, M. Di-(2-ethylhexyl)-phthalate induces oxidative stress in human endometrial stromal cells in vitro. Mol. Cell Endocrinol. 2015, 407, 9–17.
  26. Casals-Casas, C.; Desvergne, B. Endocrine disruptors: From endocrine to metabolic disruption. Annu. Rev. Physiol. 2011, 73, 135–162.
  27. Bodin, J.; Stene, L.C.; Nygaard, U.C. Can exposure to environmental chemicals increase the risk of diabetes type 1 development? Biomed. Res. Int. 2015, 2015, 208947.
  28. Castro-Correia, C.; Correia-Sa, L.; Norberto, S.; Delerue-Matos, C.; Domingues, V.; Costa-Santos, C.; Fontoura, M.; Calhau, C. Phthalates and type 1 diabetes: Is there any link? Environ. Sci. Pollut. Res. Int. 2018, 25, 17915–17919.
  29. Bodin, J.; Kocbach Bolling, A.; Wendt, A.; Eliasson, L.; Becher, R.; Kuper, F.; Lovik, M.; Nygaard, U.C. Exposure to bisphenol A, but not phthalates, increases spontaneous diabetes type 1 development in NOD mice. Toxicol. Rep. 2015, 2, 99–110.
  30. Weldingh, N.M.; Jorgensen-Kaur, L.; Becher, R.; Holme, J.A.; Bodin, J.; Nygaard, U.C.; Bolling, A.K. Bisphenol A Is More Potent than Phthalate Metabolites in Reducing Pancreatic beta-Cell Function. Biomed. Res. Int. 2017, 2017, 4614379.
  31. Predieri, B.; Bruzzi, P.; Bigi, E.; Ciancia, S.; Madeo, S.F.; Lucaccioni, L.; Iughetti, L. Endocrine Disrupting Chemicals and Type 1 Diabetes. Int. J. Mol. Sci. 2020, 21, 2937.
  32. Tiano, J.; Mauvais-Jarvis, F. Selective estrogen receptor modulation in pancreatic beta-cells and the prevention of type 2 diabetes. Islets 2012, 4, 173–176.
  33. Engel, A.; Buhrke, T.; Imber, F.; Jessel, S.; Seidel, A.; Volkel, W.; Lampen, A. Agonistic and antagonistic effects of phthalates and their urinary metabolites on the steroid hormone receptors ERalpha, ERbeta, and AR. Toxicol. Lett. 2017, 277, 54–63.
  34. Martinez-Arguelles, D.B.; Papadopoulos, V. Identification of hot spots of DNA methylation in the adult male adrenal in response to in utero exposure to the ubiquitous endocrine disruptor plasticizer di-(2-ethylhexyl) phthalate. Endocrinology 2015, 156, 124–133.
  35. Zhou, H.; Sun, L.; Zhang, S.; Zhao, X.; Gang, X.; Wang, G. Evaluating the Causal Role of Gut Microbiota in Type 1 Diabetes and Its Possible Pathogenic Mechanisms. Front. Endocrinol. Lausanne 2020, 11, 125.
  36. Hu, J.; Raikhel, V.; Gopalakrishnan, K.; Fernandez-Hernandez, H.; Lambertini, L.; Manservisi, F.; Falcioni, L.; Bua, L.; Belpoggi, F.; Teitelbaum, S.L.; et al. Effect of postnatal low-dose exposure to environmental chemicals on the gut microbiome in a rodent model. Microbiome 2016, 4, 26.
  37. Ahn, C.; Kang, J.H.; Jeung, E.B. Calcium homeostasis in diabetes mellitus. J. Vet. Sci. 2017, 18, 261–266.
  38. Johns, L.E.; Ferguson, K.K.; Meeker, J.D. Relationships Between Urinary Phthalate Metabolite and Bisphenol A Concentrations and Vitamin D Levels in U.S. Adults: National Health and Nutrition Examination Survey (NHANES), 2005–2010. J. Clin. Endocrinol. Metab. 2016, 101, 4062–4069.
  39. Johns, L.E.; Ferguson, K.K.; Cantonwine, D.E.; McElrath, T.F.; Mukherjee, B.; Meeker, J.D. Urinary BPA and Phthalate Metabolite Concentrations and Plasma Vitamin D Levels in Pregnant Women: A Repeated Measures Analysis. Environ. Health Perspect. 2017, 125, 087026.
  40. Mariana, M.; Feiteiro, J.; Cairrao, E. Cardiovascular Response of Rat Aorta to Di-(2-ethylhexyl) Phthalate (DEHP) Exposure. Cardiovasc. Toxicol. 2018, 18, 356–364.
  41. Batista-Silva, H.; Dambros, B.F.; Rodrigues, K.; Cesconetto, P.A.; Zamoner, A.; Sousa de Moura, K.R.; Gomes Castro, A.J.; Van Der Kraak, G.; Mena Barreto Silva, F.R. Acute exposure to bis(2-ethylhexyl)phthalate disrupts calcium homeostasis, energy metabolism and induces oxidative stress in the testis of Danio rerio. Biochimie 2020, 175, 23–33.
  42. Liu, P.S.; Chen, Y.Y. Butyl benzyl phthalate blocks Ca2+ signaling coupled with purinoceptor in rat PC12 cells. Toxicol. Appl. Pharmacol. 2006, 210, 136–141.
  43. Nakamura, R.; Teshima, R.; Sawada, J. Effect of dialkyl phthalates on the degranulation and Ca2+ response of RBL-2H3 mast cells. Immunol. Lett. 2002, 80, 119–124.
  44. Posnack, N.G.; Idrees, R.; Ding, H.; Jaimes, R., 3rd; Stybayeva, G.; Karabekian, Z.; Laflamme, M.A.; Sarvazyan, N. Exposure to phthalates affects calcium handling and intercellular connectivity of human stem cell-derived cardiomyocytes. PLoS ONE 2015, 10, e0121927.
  45. Sol, C.M.; Santos, S.; Duijts, L.; Asimakopoulos, A.G.; Martinez-Moral, M.P.; Kannan, K.; Jaddoe, V.W.V.; Trasande, L. Fetal phthalates and bisphenols and childhood lipid and glucose metabolism. A population-based prospective cohort study. Environ. Int. 2020, 144, 106063.
  46. Attina, T.M.; Trasande, L. Association of Exposure to Di-2-Ethylhexylphthalate Replacements With Increased Insulin Resistance in Adolescents From NHANES 2009–2012. J. Clin. Endocrinol. Metab. 2015, 100, 2640–2650.
  47. Carlsson, A.; Sorensen, K.; Andersson, A.M.; Frederiksen, H.; Juul, A. Bisphenol A, phthalate metabolites and glucose homeostasis in healthy normal-weight children. Endocr. Connect. 2018, 7, 232–238.
  48. Chen, S.Y.; Hwang, J.S.; Sung, F.C.; Lin, C.Y.; Hsieh, C.J.; Chen, P.C.; Su, T.C. Mono-2-ethylhexyl phthalate associated with insulin resistance and lower testosterone levels in a young population. Environ. Pollut. 2017, 225, 112–117.
  49. Dales, R.E.; Kauri, L.M.; Cakmak, S. The associations between phthalate exposure and insulin resistance, beta-cell function and blood glucose control in a population-based sample. Sci. Total Environ. 2018, 612, 1287–1292.
  50. Dirinck, E.; Dirtu, A.C.; Geens, T.; Covaci, A.; Van Gaal, L.; Jorens, P.G. Urinary phthalate metabolites are associated with insulin resistance in obese subjects. Environ. Res. 2015, 137, 419–423.
  51. Nam, D.J.; Kim, Y.; Yang, E.H.; Lee, H.C.; Ryoo, J.H. Relationship between urinary phthalate metabolites and diabetes: Korean National Environmental Health Survey (KoNEHS) cycle 3 (2015–2017). Ann. Occup. Environ. Med. 2020, 32, e34.
  52. Duan, Y.; Sun, H.; Han, L.; Chen, L. Association between phthalate exposure and glycosylated hemoglobin, fasting glucose, and type 2 diabetes mellitus: A case-control study in China. Sci. Total Environ. 2019, 670, 41–49.
  53. Dong, R.; Zhao, S.; Zhang, H.; Chen, J.; Zhang, M.; Wang, M.; Wu, M.; Li, S.; Chen, B. Sex Differences in the Association of Urinary Concentrations of Phthalates Metabolites with Self-Reported Diabetes and Cardiovascular Diseases in Shanghai Adults. Int. J. Environ. Res. Public Health 2017, 14, 598.
  54. Al-Bazi, M.M.; Kumosani, T.A.; Al-Malki, A.L.; Kurunthachalam, K.; Moselhy, S.S. Screening the incidence of diabetogensis with urinary phthalate in Saudi subjects. Environ. Sci. Pollut. Res. Int. 2022, 29, 28743–28748.
  55. Bai, P.Y.; Wittert, G.; Taylor, A.W.; Martin, S.A.; Milne, R.W.; Jenkins, A.J.; Januszewski, A.S.; Shi, Z. The association between total phthalate concentration and non-communicable diseases and chronic inflammation in South Australian urban dwelling men. Environ. Res. 2017, 158, 366–372.
  56. Svensson, K.; Hernandez-Ramirez, R.U.; Burguete-Garcia, A.; Cebrian, M.E.; Calafat, A.M.; Needham, L.L.; Claudio, L.; Lopez-Carrillo, L. Phthalate exposure associated with self-reported diabetes among Mexican women. Environ. Res. 2011, 111, 792–796.
  57. James-Todd, T.; Stahlhut, R.; Meeker, J.D.; Powell, S.G.; Hauser, R.; Huang, T.; Rich-Edwards, J. Urinary phthalate metabolite concentrations and diabetes among women in the National Health and Nutrition Examination Survey (NHANES) 2001–2008. Environ. Health Perspect. 2012, 120, 1307–1313.
  58. Sun, Q.; Cornelis, M.C.; Townsend, M.K.; Tobias, D.K.; Eliassen, A.H.; Franke, A.A.; Hauser, R.; Hu, F.B. Association of urinary concentrations of bisphenol A and phthalate metabolites with risk of type 2 diabetes: A prospective investigation in the Nurses′ Health Study (NHS) and NHSII cohorts. Environ. Health Perspect. 2014, 122, 616–623.
  59. Duan, Y.; Sun, H.; Yao, Y.; Han, L.; Chen, L. Perturbation of serum metabolome in relation to type 2 diabetes mellitus and urinary levels of phthalate metabolites and bisphenols. Environ. Int. 2021, 155, 106609.
  60. Rajesh, P.; Balasubramanian, K. Gestational exposure to di(2-ethylhexyl) phthalate (DEHP) impairs pancreatic beta-cell function in F1 rat offspring. Toxicol. Lett. 2015, 232, 46–57.
  61. Rajagopal, G.; Bhaskaran, R.S.; Karundevi, B. Maternal di-(2-ethylhexyl) phthalate exposure alters hepatic insulin signal transduction and glucoregulatory events in rat F(1) male offspring. J. Appl. Toxicol. 2019, 39, 751–763.
  62. Rajagopal, G.; Bhaskaran, R.S.; Karundevi, B. Developmental exposure to DEHP alters hepatic glucose uptake and transcriptional regulation of GLUT2 in rat male offspring. Toxicology 2019, 413, 56–64.
  63. Deng, T.; Zhang, Y.; Wu, Y.; Ma, P.; Duan, J.; Qin, W.; Yang, X.; Chen, M. Dibutyl phthalate exposure aggravates type 2 diabetes by disrupting the insulin-mediated PI3K/AKT signaling pathway. Toxicol. Lett. 2018, 290, 1–9.
  64. Ding, Y.; Xu, T.; Mao, G.; Chen, Y.; Qiu, X.; Yang, L.; Zhao, T.; Xu, X.; Feng, W.; Wu, X. Di-(2-ethylhexyl) phthalate-induced hepatotoxicity exacerbated type 2 diabetes mellitus (T2DM) in female pubertal T2DM mice. Food Chem. Toxicol. 2021, 149, 112003.
  65. Ding, Y.; Gao, K.; Liu, Y.; Mao, G.; Chen, K.; Qiu, X.; Zhao, T.; Yang, L.; Feng, W.; Wu, X. Transcriptome analysis revealed the mechanism of the metabolic toxicity and susceptibility of di-(2-ethylhexyl)phthalate on adolescent male ICR mice with type 2 diabetes mellitus. Arch. Toxicol. 2019, 93, 3183–3206.
  66. Karabulut, G.; Barlas, N. The possible effects of mono butyl phthalate (MBP) and mono (2-ethylhexyl) phthalate (MEHP) on INS-1 pancreatic beta cells. Toxicol. Res. Camb 2021, 10, 601–612.
  67. Guven, C.; Dal, F.; Aydogan Ahbab, M.; Taskin, E.; Ahbab, S.; Adin Cinar, S.; Sirma Ekmekci, S.; Gulec, C.; Abaci, N.; Akcakaya, H. Low dose monoethyl phthalate (MEP) exposure triggers proliferation by activating PDX-1 at 1.1B4 human pancreatic beta cells. Food Chem. Toxicol. 2016, 93, 41–50.
  68. Al-Abdulla, R.; Ferrero, H.; Soriano, S.; Boronat-Belda, T.; Alonso-Magdalena, P. Screening of Relevant Metabolism-Disrupting Chemicals on Pancreatic beta-Cells: Evaluation of Murine and Human In Vitro Models. Int. J. Mol. Sci. 2022, 23, 4182.
  69. Mondal, S.; Mukherjee, S. Long-term dietary administration of diethyl phthalate triggers loss of insulin sensitivity in two key insulin target tissues of mice. Hum. Exp. Toxicol. 2020, 39, 984–993.
  70. Schaffert, A.; Karkossa, I.; Ueberham, E.; Schlichting, R.; Walter, K.; Arnold, J.; Bluher, M.; Heiker, J.T.; Lehmann, J.; Wabitsch, M.; et al. Di-(2-ethylhexyl) phthalate substitutes accelerate human adipogenesis through PPARgamma activation and cause oxidative stress and impaired metabolic homeostasis in mature adipocytes. Environ. Int. 2022, 164, 107279.
  71. She, Y.; Jiang, L.; Zheng, L.; Zuo, H.; Chen, M.; Sun, X.; Li, Q.; Geng, C.; Yang, G.; Jiang, L.; et al. The role of oxidative stress in DNA damage in pancreatic beta cells induced by di-(2-ethylhexyl) phthalate. Chem. Biol. Interact. 2017, 265, 8–15.
  72. Yang, R.; Zheng, J.; Qin, J.; Liu, S.; Liu, X.; Gu, Y.; Yang, S.; Du, J.; Li, S.; Chen, B.; et al. Dibutyl phthalate affects insulin synthesis and secretion by regulating the mitochondrial apoptotic pathway and oxidative stress in rat insulinoma cells. Ecotoxicol. Environ. Saf. 2023, 249, 114396.
  73. Viswanathan, M.P.; Mullainadhan, V.; Chinnaiyan, M.; Karundevi, B. Effects of DEHP and its metabolite MEHP on insulin signalling and proteins involved in GLUT4 translocation in cultured L6 myotubes. Toxicology 2017, 386, 60–71.
  74. Zhang, W.; Shen, X.Y.; Zhang, W.W.; Chen, H.; Xu, W.P.; Wei, W. Di-(2-ethylhexyl) phthalate could disrupt the insulin signaling pathway in liver of SD rats and L02 cells via PPARgamma. Toxicol. Appl. Pharmacol. 2017, 316, 17–26.
  75. Baralic, K.; Zivancevic, K.; Jorgovanovic, D.; Javorac, D.; Radovanovic, J.; Gojkovic, T.; Buha Djordjevic, A.; Curcic, M.; Mandinic, Z.; Bulat, Z.; et al. Probiotic reduced the impact of phthalates and bisphenol A mixture on type 2 diabetes mellitus development: Merging bioinformatics with in vivo analysis. Food Chem. Toxicol. 2021, 154, 112325.
  76. Ito, Y.; Kamijima, M.; Nakajima, T. Di(2-ethylhexyl) phthalate-induced toxicity and peroxisome proliferator-activated receptor alpha: A review. Environ. Health Prev. Med. 2019, 24, 47.
More
This entry is offline, you can click here to edit this entry!
Video Production Service