Next Article in Journal
Effects of Estradiol and Methoxychlor on Leydig Cell Regeneration in the Adult Rat Testis
Next Article in Special Issue
Aryl Hydrocarbon Receptor Repressor and TiPARP (ARTD14) Use Similar, but also Distinct Mechanisms to Repress Aryl Hydrocarbon Receptor Signaling
Previous Article in Journal
Development of Drug Delivery Systems Based on Layered Hydroxides for Nanomedicine
Previous Article in Special Issue
The Relationship between Dioxin Congeners in the Breast Milk of Vietnamese Women and Sister Chromatid Exchange
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Association of Dioxin and Other Persistent Organic Pollutants (POPs) with Diabetes: Epidemiological Evidence and New Mechanisms of Beta Cell Dysfunction

Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, Scuola Medica, 56126 Pisa, Italy
Int. J. Mol. Sci. 2014, 15(5), 7787-7811; https://doi.org/10.3390/ijms15057787
Submission received: 19 February 2014 / Revised: 16 April 2014 / Accepted: 21 April 2014 / Published: 5 May 2014
(This article belongs to the Special Issue Mechanisms of Toxicity of Dioxins and Related Compounds)

Abstract

:
The worldwide explosion of the rates of diabetes and other metabolic diseases in the last few decades cannot be fully explained only by changes in the prevalence of classical lifestyle-related risk factors, such as physical inactivity and poor diet. For this reason, it has been recently proposed that other “nontraditional” risk factors could contribute to the diabetes epidemics. In particular, an increasing number of reports indicate that chronic exposure to and accumulation of a low concentration of environmental pollutants (especially the so-called persistent organic pollutants (POPs)) within the body might be associated with diabetogenesis. In this review, the epidemiological evidence suggesting a relationship between dioxin and other POPs exposure and diabetes incidence will be summarized, and some recent developments on the possible underlying mechanisms, with particular reference to dioxin, will be presented and discussed.

1. Introduction

Diabetes mellitus is one of the most common chronic diseases, and its prevalence has exploded over the last several decades. The World Health Organization and the International Diabetes Federation estimated that the prevalence of diabetes increased from 100 to 135 million affected adults worldwide in 1994–1995 to approximately 336 million in 2011, and it is expected to rise to 439 million by 2030 [13]. Since these changes occurred too rapidly to simply reflect genetic causes and rates vary widely by country, the vast majority of new cases of diabetes are likely to be caused by changes in lifestyle and/or environment during the last few decades. Therefore, the possibility that a corresponding distribution of toxic substances might account for the geographic variations in diabetes prevalence was initially suggested in 1972 [4]. In this regard, it might be worthy to mention that in the U.S., diabetes rates have increased in concordance with the national production of synthetic organic chemicals [5]. Dioxins and other persistent organic pollutants (POPs) (such as polychlorinated biphenyls, dichlorodiphenyldichloroethylene (DDE), the main degradation product of the pesticide, dichlorodiphenyltrichloroethane (DTT), trans-nonachloro, hexachlorobenzene and the hexachlorocyclohexanes) are generally considered to be the most likely candidates as potential risk factors for type 2 diabetes [6], since they may act as endocrine disruptors. Endocrine disrupting chemicals have been defined by the Environmental Protection Agency (EPA) as “exogenous agents that interfere with the production, release, transport, metabolism, binding, action, or elimination of natural hormones in the body responsible for the maintenance of homeostasis, reproduction, development, and/or behavior” [710]. Many of these pollutants (although not all; see, e.g., the case of bisphenol A) were banned in most Western countries in the 1970s and 1980s, with a consequent diminution of their levels in the environment, but they are still detected in humans, especially in those subgroups of the general population that still show an elevated body burden due to dietary habits and current and past exposure [1113].

2. Epidemiological Evidence

Several epidemiological studies contributed to establishing a clear relationship between dioxin (as well as other POPs) exposure and type 2 diabetes incidence (for recent reviews, see [14,15]. This association was firstly reported for occupationally exposed populations and subsequently investigated in the general population.
Many veterans of the Vietnam War were exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD or simply dioxin), which was a contaminant in the herbicide formulation, Agent Orange, widely used as a defoliant by the U.S. Air Force from 1961 to 1971 [16]. Even after many years, significant residues of dioxin have been found in their serum [17,18], because of the long half-life of dioxin, which is estimated to be 7.6 years [19]. The follow-up of veterans to assess the possible long-term health consequences of exposure to Agent Orange identified type 2 diabetes as one of the diseases most consistently statistically associated with dioxin exposure [2024]. Similar conclusions were reached by epidemiological studies carried out on the population exposed to high levels of dioxin as a result of an industrial accident on 10 July 1976, in the trichlorophenol production department of a chemical plant located near the town of Seveso, 25 km north of Milan, Italy [2527]. A 25-year follow up of the exposed population showed a significantly increased mortality from diabetes mellitus among females [28,29]. This last observation could be of particular relevance in relation to the recently reported sex-specific metabolic disorders induced by a diet containing low doses of TCDD and other POPs in the progeny of obese mice [30]. Other epidemiological studies, with comparable results, were performed in professionally exposed industrial workers [31], Korean Vietnam veterans, who were also exposed to Agent Orange [32], and individuals living near municipal waste incinerators [33] or hazardous waste sites [34]. Similar results were obtained in a 24-year follow-up study of 1054 Yucheng “oil-disease” victims that, during the late 70s, were accidentally exposed to rice-bran oil laced with polychlorinated biphenyls [35]. In contrast, negative results were obtained in professionally exposed workers by other authors [3641]. In particular, Kerger et al. [42], recently re-analyzing the Ranch Hand study, supported the hypothesis that diabetes progression could lead to higher dioxin levels rather than the opposite. In more recent years, the exploration of the epidemiological relationship between dioxin (as well as other correlated POPs) exposure and diabetes incidence has been extended from professionally or accidentally exposed individuals to the general population. In this regard, a preliminary note of caution is required. A main feature of POPs is the fact that these compounds are always present as chemical mixtures, and therefore, when studies involving general populations with only background exposure to POPs are evaluated, data obtained for a single compound cannot be simply attributed to the exclusive effect of that compound, but must rather be viewed as the result of the overall effect of the mixture [43]. Fierens et al. [44], in a population-based study in Belgium, firstly found that diabetic patients had significantly increased serum levels of dioxins and coplanar polychlorinated biphenyls. In 2006, using cross-sectional data from the 1999–2002 U.S. National Health and Examination Survey, Lee et al. [45,46] reported a strong correlation between serum concentration of POPs (especially organochlorine compounds) and diabetes. The association with serum levels of POPs was subsequently extended by the same group to insulin resistance [47], to the prevalence of metabolic syndrome [48] and to abdominal obesity [49] among non-diabetic adults. These results were further supported by a study of Everett et al. [50] showing a significant positive correlation between diabetes and polychlorinated dibenzo-p-dioxin, a polychlorinated biphenyl, and DDT in a group of 1830 subjects from the 1999–2002 National Health and Examination Survey. Intriguingly, Lee and co-workers [51,52] hypothesized that serum gamma-glutamyltransferase (GGT) may predict diabetes as a general marker of exposure to various environmental xenobiotics, including diabetes-related POPs. Other studies on general populations have further confirmed the association between POPs exposure and diabetes incidence [5360].
POPs are generally lipophilic, resistant to degradation and tend to bioaccumulate within the food chain. Therefore, in spite of the general decline of their environmental levels over recent decades, today, they are especially found as contaminants in food. For this reason, some recent studies on the relationship between the consumption of foods, such as fish and marine mammals, and diabetes incidence could be particularly interesting (for a review, see [61]). Jǿrgensen et al. [62] showed a significant association between plasma POP levels and impaired beta cell function among Greenland Inuit, while Sharp [63] reported that diabetes prevalence rates are 3–5 times higher among Canadian aboriginals than in the general population, and Philibert et al. [64] found a positive correlation between diabetes and serum levels of p,p′-DDE and some polychlorinated biphenyl congeners in a First Nation community. Finally, Rylander et al. [65] reported a significant correlation between plasma POP levels and type 2 diabetes in Swedish fisherman and their wives, and Turyk et al. [66] found a similarly significant association in a group of Great Lakes sport fish consumers.
A critical evaluation of the epidemiological literature, which is far from being homogeneous and still presents several gaps and inconsistences that can seriously limit the validity of some of the data reported, is beyond the scope of this review and has been recently addressed elsewhere [14], reaching the main conclusion that POPs have generated particularly strong evidence as risk factors for diabetes in humans [14,43]. New and original ways to study the association between dioxin and other POP exposure with diabetes have been recently proposed. Fujiyoshi et al. [67], in order to offer some clues about the mechanism of the diabetogenic action of dioxin in humans, performed a molecular epidemiological investigation in adipose tissue samples from U.S. Air Force Vietnam veterans who were or were not exposed to dioxin. They conducted quantitative reverse-transcribed polymerase chain reaction studies on selected marker mRNAs from these samples and found that the most sensitive and reliable molecular indicator of dioxin-induced diabetes was the ratio of mRNA of glucose transporter 4 (GLUT4) and NFkB, a marker of inflammation. Interestingly, this ratio showed significant correlations to serum dioxin levels and to fasting glucose, not only among the exposed veterans, but even in the comparison group, who have low levels of dioxin comparable to the general public. More recently, Patel et al. [68] proposed a model environmental-wide association study (EWAS) to search for environmental factors associated with type 2 diabetes, in which epidemiological data were comprehensively and systematically interpreted in a manner analogous to genome wide association studies (GWASs).

3. Mechanisms of Dioxin-Induced Beta Cell Dysfunction

We have seen so far that overall epidemiological evidence supports a positive association between dioxin and other organochlorine POPs exposure and diabetes incidence. However, association does not necessarily imply a causal link, although several biologically plausible explanations have been proposed [6]. For this reason, very recently, a panel of experts that evaluated the association between POPs and diabetes in epidemiological studies in a workshop sponsored by the National Institute of Environmental Health Sciences (NIEHS)/National Toxicology Program (NTP), the U.S. Environmental Protection Agency (EPA) and the Food and Drug Administration National Center for Toxicological Research indicated the need of additional animal and in vitro mechanistic studies to clarify the role of POPs in metabolic disease development [14]. Regrettably, the investigation of the biological mechanisms underlying the relationship between dioxin exposure and type 2 diabetes has been rather neglected until recently.

3.1. In Vivo Effects of Dioxin Administration

Initially, research on dioxin toxicity was mainly focused on its acutely lethal effects [6971] in highly sensitive rodents, such as guinea pigs. Subsequently, the realization that human beings are less susceptible to these acute effects shifted the attention on low level chronic effects, such as carcinogenicity [72,73] and reproductive toxicology [7476], that can be induced in animal models at concentrations close to human background exposure [12]. Among the most common symptoms seen in animal species treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, the most toxic of the dioxin-type chemicals and, thus, the most widely used in animal toxicological studies), there is body weight loss or reduced weight gain, mostly related to the loss of fat and muscle tissue (“wasting syndrome”) [7780]. Adipose tissue loss is associated with hypophagia [81], hyperlipidemia, particularly hypertriglyceridemia [82], probably linked to the TCDD-induced drastic decline in adipose lipoprotein lipase activity [83,84], and with changes in the expression levels of the adipogenesis- and lipogenesis-related proteins in the liver and adipose tissue [8588] and hypoinsulinemia [89]. The insulin resistance-like symptoms observed in wasting syndrome, such as the loss of body and adipose tissue weight, have been recently attributed to the TCDD-induced activation of TNF-α [9092] and COX-2 [93]. Since anorexia is a well-known component of TCDD-induced wasting syndrome, the effects of dioxin administration on hypothalamic regions have been also investigated, and several alterations in the pathways regulating body weight and food intake have been reported [79,80,9498]. On the basis of these observations, dioxin seems to hinder the utilization of the available nutritional elements (e.g., glucose, triglycerides, cholesterol, etc.) in the affected animals. Among the metabolic derangements induced by TCDD, those related to glucose homeostasis could play a crucial role in the induction of wasting syndrome. One of the most characteristic alterations induced by TCDD is a marked decrease in glucose uptake by adipose tissue, liver and pancreas in vivo, as well as in vitro [99103], mainly caused by the TCDD-induced decline in the titer of glucose transporters [90,104109]. In particular, it has been proposed that the reduced glucose uptake caused by the decreased GLUT2 expression in beta cells could explain the TCDD-induced lowering of insulin production and secretion by the pancreas [37,110]. Indeed, TCDD can cause hypoinsulinemia both in the rabbit [89] and in the rat [111,112]. More recently, we showed that, in the rat, a single low dose of TCDD (1 μg/kg b.w., well below the usually lethal one, estimated to be around 125 μg/kg), was able to rapidly reduce both pancreatic insulin content and glucose-stimulated insulin secretion from isolated pancreatic islets, indicating an early involvement of the endocrine pancreas in TCDD toxicity [113]. Similar results were also reported by Kurita et al. [114]. We explored also the secretory response of isolated islets to the non-glucose secretagogue, 2-ketoisocaproate (2-KIC), which is one of the few physiological substrates apart from glucose that initiates a sustained insulin release from beta cells [115117], and we found that 2-KIC-induced insulin release was better preserved in TCDD-treated rats, indirectly confirming that the impairment in the secretory performance observed in pancreatic islets isolated from TCDD-treated rats was strictly linked to the failure of glucose to induce insulin secretion in beta cells [113]. In pancreatic beta cells, glucose utilization is regulated by glucokinase, a glucose-specific high-Km enzyme that, at glucose concentrations >5.5 mM, is the rate limiting step for glucose metabolism and, therefore, for glucose-induced insulin release [118,119]. Under these conditions, glucose transport, whose capacity largely exceeds glucose phosphorylation and metabolism, does not play a major role in determining the secretory response [120]. However, when the expression or function of the glucose transporter is significantly reduced, it may limit the access of glucose to glucokinase, thus preventing a normal glucose sensing and appropriate insulin secretion [121123]. To test whether the decrease in glucose uptake could be responsible for the impairment of insulin secretion observed in rats treated with a single low-dose injection of TCDD, we measured both GLUT2 protein levels by western blotting and pancreatic glucose uptake by using its nonmetabolizable 3H-labeled analog, 2-deoxyglucose. Despite that immunoblotting did not reveal apparent differences in GLUT2 protein levels between pancreatic islets isolated from control and TCDD-treated rats, we showed a significant decrease (−20%) of glucose uptake in the pancreas of TCDD-treated rats [113]. TCDD is thought to affect the levels of glucose transporters, not directly through, e.g., its binding to the glucose transporter protein, but rather by the inhibition of the transcriptional and translational expression of GLUT genes after binding to the cytosolic aryl-hydrocarbon receptor (AhR) [108110]. However, it has been recently shown that in 3T3-L1 adipocytes, the inhibition of AhR by alpha-naphthoflavone did not prevent the inhibitory effect of TCDD on glucose uptake, suggesting that this effect could be AhR-independent [101]. The history of the search for the action mechanism of TCDD has been largely based on the paradigm of ligand-induced activation of the AhR leading to direct activation of many target genes (the “genomic” or classical pathway) [124126]. The AhR-mediated biological responses include induction of metabolic enzymes, body weight loss, immunosuppression, skin lesion and changes in cellular growth and differentiation [127]. With particular reference to glucose and lipid metabolism, it may be interesting to note that some biochemical alterations observed in the development of type 2 diabetes (e.g., the inhibition of PEPKC activity, the induction of fatty acid synthesis and the effects on IL-1β and TNF-α) have been also reported in TCDD-treated rats [85,92,128135] and in TCDD-exposed mouse hepatocytes [136,137]. Some of these effects (expression of key gluconeogenic genes, PEPKC and G6Pase) have been very recently attributed to the TCDD-induced activation of the AhR target gene, TiPARP (TCDD-inducible poly(ADP-ribose)polymerase) [138].

3.2. The Non-Genomic Pathway of Dioxin Toxicity

In the last decade, it has been observed that many of the major risk factors for type 2 diabetes (overnutrition, low dietary fiber, sedentary lifestyle, sleep deprivation and depression) can induce local or systemic low-grade inflammation. This inflammatory response is transient or milder in individuals not at risk for type 2 diabetes. By contrast, inflammation in response to lifestyle factors is more relevant and sustained in individuals at risk for type 2 diabetes and appears to occur also in the pancreatic islets (for a recent review, see [2]). The long-term consequences of prolonged low-grade inflammation, in particular in response to metabolic stress, could result in being detrimental [139] and eventually lead to overt diabetes if counter-regulatory circuits to inflammation are compromised as a consequence of a genetic and/or epigenetic predisposition [2,140,141]. In view of the above considerations, it is pertinent to highlight that one of the major actions of TCDD is to elicit inflammatory cellular responses, which can be very intense and last for long time periods [142145]. In particular, it has been shown that TCDD rapidly induces a significant rise of intracellular [Ca++], which, in turn, activates cytosolic phospholipase A2 enzyme activity, as demonstrated by the early release of arachidonic acid, followed by activation of Src kinase and induction of several inflammatory markers. This new, inflammation-inducing action pathway of TCDD is definitely distinguished from the classical action pathway on the basis of evidence clearly indicating differences in the timing of activation of the two pathways, the possibility to be selectively blocked by specific inhibitors (e.g., calcium-signaling blockers) without affecting the DRE-driven CYP1A1 induction and the non-involvement of Arnt in the new pathway [146151]. On the basis of these observations, Matsumura designated the newly delineated pathway as the “non-genomic” inflammation pathway of the action of TCDD [152].
This new intriguing theoretical framework could represent a major breakthrough to study the mechanisms of dioxin toxicity in pancreatic beta cells. As beta cell dysfunction is increasingly recognized to play a fundamental role in type 2 diabetes pathophysiology [153,154], it became particularly relevant to investigate the mechanisms that can disrupt beta cell function [155]. In the last few years, in the attempt to clarify the mechanisms underlying the diabetogenic action of dioxin, we utilized as an experimental model the INS-1 cell line, which was established from an X-ray-induced rat transplantable insulinoma and whose cells retain the ability to dose-dependently secrete insulin when stimulated with increasing concentrations of glucose. The maintenance of this functional property makes these cells a valuable and widely used beta cell model [156,157]. By using this model, we observed that 1 h of exposure to TCDD concentrations between 12.5 and 25 nM induced a sharp decline of cell survival (below 20%) [158]. Such “in vitro” concentrations could appear quite elevated when compared with the mean body burden for TCDD and other less toxic related cogeners in the general population (estimated to be around 13 ng TEQ/kg body weight), but it should be considered that in particular conditions (such as after industrial accidents), the body burden can increase to remarkably higher levels (up to 7000 ng TEQ/kg b.w.) [12,159]. Thus, at least in such conditions of high environmental exposure, pancreatic β-cells are likely to undergo relevant damage. On the other hand, for comparison, we recall that well-known β-cytotoxic agents, such as alloxan, can produce similar effects on INS-1 cell viability only upon prolonged exposure to much higher concentrations (0.5 mM for 24 h) [160]. Taking into account the “non-genomic” pathway of dioxin toxicity as proposed by Matsumura [146], the observation that in our model, cytotoxic concentrations of TCDD very rapidly (few seconds) induced a dose-dependent increase in intracellular calcium concentration and that blocking calcium entry by EGTA significantly decreased TCDD cytotoxicity is particularly relevant [158]. The early action of TCDD to cause a rapid increase in the cytosolic concentration of Ca++ has been initially demonstrated by Hanneman et al. [161] and Puga et al. [162]. Other laboratories have confirmed such an early rise in intracellular calcium by TCDD in various cell types and tissues [151,163172]. The mechanism of the TCDD-induced increase of cytoplasmic calcium concentration has not yet been clarified. A primary route for Ca2+ influx is through “store-operated channels” in the cell membrane (originally termed capacitative Ca2+ entry), probably activated by a fall in Ca2+ within the endoplasmic reticulum [173]. Our data are consistent with such a mechanism: indeed, we showed that the TCDD-induced increase in cellular calcium can be attributed both to a transient and quantitatively modest calcium release from intracellular stores and to a more relevant and sustained influx of extracellular calcium into the cell [158]. Overall, these results clearly indicate that TCDD causes calcium influx in a manner independent from the classic action pathway of DRE-driven transcriptional mechanisms and that, very likely, calcium can be viewed as the initial trigger for this nongenomic action of TCDD. It is well known that perturbation in cellular calcium homeostasis may be associated with the early development of cell injury (for a review, see, e.g., [174]). Therefore, the TCDD-induced increase of intracellular [Ca2+] might play an important role in the mechanism of dioxin toxicity in beta cells, and a major target for its negative effects could likely be represented by mitochondria.

3.3. Dioxin-Induced Mitochondrial Dysfunction

It is known that by the accumulation of calcium when its cytosolic levels are high, mitochondria can play crucial roles in coordinating the complexities of intracellular calcium-signaling pathways [175]. On the other hand, it is also known that calcium can represent a pathological stimulus, eventually leading to apoptotic cell death. To explain this apparent paradox (Ca2+ levels as both a physiological and pathological effector of mitochondrial function), it has been proposed that Ca2+ could modulate mitochondrial ROS production by several mechanisms (stimulation of the TCA cycle and oxidative phosphorylation; stimulation of nitric oxide synthase; perturbation of mitochondrial antioxidant status) [175]. Indeed, it has been shown that TCDD can increase mitochondrial ROS production [176], probably through a modification of the mitochondrial GSSG/GSH ratio [177,178]. Thus, to study the effect of TCDD on mitochondrial function, we assayed the mitochondrial membrane potential in INS-1 cells after 10 min of exposure to TCDD, and we observed a significant, dose-related mitochondrial depolarization [158]. In β-cells, mitochondria are crucial for the physiological stimulus-secretion coupling, in which the mitochondrial metabolism of pyruvate, glycolitically derived from glucose, generates ATP, which promotes the closure of ATP-sensitive K+ channels and the consequent cell depolarization, inducing Ca2+ influx through voltage-gated Ca2+ channels, increased cytosolic [Ca2+] and finally triggering insulin exocytosis [179]. Thus, it is easy to predict that the TCDD-induced damage of mitochondrial function could result in an impaired secretory capability of beta cells. Indeed, we found that a 1-h exposure of INS-1 cells to very low TCDD concentrations (0.05–1 nM) dramatically impaired glucose-stimulated insulin secretion, showing, for the first time, that dioxin, early and at very low doses, can induce significant beta cell dysfunction [158]. We also demonstrated that in TCDD-treated INS-1 cells, the KCl-stimulated insulin secretion (independent of glucose metabolism) is well preserved, strongly suggesting that mitochondrial alterations rather than the exocytotic process might play a crucial role [158]. In this regard, it is important to note that the inhibitory effect of TCDD on glucose-stimulated insulin secretion was observed with 1 nM TCDD, i.e., well below the cytotoxic concentrations. However, it should be noted that at this concentration, TCDD is still able to cause a small, although not significant, mitochondrial depolarization and that electron microscopy indicated that several cells, with an ultrastructural morphology that was otherwise normal, nevertheless contained mitochondria presenting slight pathological alterations, which result in being significantly different from controls when analyzed by quantitative morphometry [180,181]. Slight alterations of the mitochondrial inner membrane ultrastructure have been described in a number of pathological conditions and have been associated with defects of mitochondrial function [182,183]. Interestingly, we have also shown that dehydroascorbate and epigallocatechin 3-gallate (EGCG), two compounds that were both able to protect beta cells from dioxin toxicity, were also able to significantly prevent the TCDD-induced mitochondrial depolarization and the impairment of glucose-induced insulin secretion in INS-1 cells [180,181]. In this regard, it is interesting to mention that it has been recently reported that EGCG protects insulin-producing cells against pro-inflammatory cytokine-induced injuries through the mitochondrial pathway [184].
Thus, taken together, our results seem to indicate the mitochondrion as one of the most likely targets of dioxin acute toxicity in beta cells. This conclusion could be particularly relevant in view of the recently emerging evidence showing that environmental toxins, including POPs, can affect mitochondrial function and subsequently induce insulin resistance [185189]. Indeed, mitochondrial dysfunction can affect a range of functions, from metabolic to signaling pathways, that regulate hormone action. When mitochondrial function is perturbed, diseases involving energy utilization easily result: obesity and insulin resistance are prototypes of these diseases [190]. Furthermore, mitochondrial abnormalities are associated with intracellular lipid accumulation, insulin resistance and the pathophysiology of type 2 diabetes [187,191193].

3.4. POP Exposure and Obesity

POPs are lipophilic compounds that accumulate mainly in adipose tissue. It has been proposed that mitochondrial dysfunction could play an important role in chronic low-grade inflammation in adipose tissue, which is believed to represent a crucial mechanism in the development of type 2 diabetes [140,194]. Experimental animal studies suggest that chronic exposure to low-dose POP mixtures similar to the current human background exposure may cause mitochondrial dysfunction [195], probably trough GSH depletion [196], eventually leading to insulin resistance and diabetes. Moreover, POPs acting as endocrine disrupting chemicals could negatively interfere with the carefully coordinated hormonal regulation of adipocyte metabolism, promoting the development of dyslipidemia and diabetes [197200]. As a final comment, it is important to remember that although obesity represents a strong risk factor for type 2 diabetes, a subset of obese individuals termed metabolically healthy but obese (MHO) are insulin sensitive with a preserved glucose tolerance, have a less systemic low-grade inflammation and visceral adipose tissue accumulation with respect to metabolically abnormal obese (MAO) subjects and are relatively protected from the development of cardiovascular complications. Very interestingly, Gauthier et al. [201] recently reported that MHO phenotypes present significantly lower circulating levels of POPs (dioxin- and non-dioxin-like polychlorinated biphenyls) as compared to the MAO phenotype.

3.5. Dioxin-Induced Alteration of Signal Transduction Pathways

Although the largely AhR-independent effect on mitochondrial function could play a key role in dioxin toxicity, other mechanisms leading to beta cell dysfunction can be involved. In this regard, it has been proposed that the non-genomic pathway of dioxin toxicity, initially triggered by the rapid increase in the intracellular [Ca++], could be subsequently converted in more stable long-term messages by the activation of selected protein kinases [152]. Our experiments, in agreement with previously reported results obtained in other cell types [202205], confirm that in beta cells, ERK 1/2 and JNK are activated, whereas Akt and p38 are unmodified by TCDD treatment [177]. The observation that EGCG can prevent the TCDD-stimulated selective activation of ERK and JNK is of particular interest in view of the recently reported ability of selected food phytochemicals to reduce the toxic actions of TCDD in U937 macrophages [206] and of flavones and catechins to suppress the TCDD-induced phosphorylation of ERK 1/2 in mouse hepatoma Hepa-1c1c7 cells [207]. The observed inhibition of TCDD-induced MAPK phosphorylation by EGCG could be related to the reported suppression of the dissociation of hsp90 and XAP2 from AhR by the interaction of EGCG with hsp90 [208]. This mechanism is particularly attractive, because it has been reported that in MCF10A cells, the rapid activation by TCDD of c-Src kinase is mediated by the Cdc37-HSP90 complex [209].

3.6. Dioxin-Induced Activation of Autophagy

Further information on the mechanisms of dioxin toxicity can be obtained from the ultrastructural analysis of beta cells after TCDD exposure. Indeed, dioxin cytotoxicity is associated with several ultrastructural alterations, such as extensive degranulation, mitochondrial abnormalities and peripheral nuclear condensation [158]. Among the ultrastructural alterations induced by dioxin in beta cells, one of the most characteristic is the diffuse and relevant activation of autophagy [158,181], which has been confirmed by using different experimental techniques (electron microscopy, monodansylcadaverine fluorescence, immunoblotting), in accordance with the internationally acknowledged guidelines for the study of autophagy [210]. TCDD-induced activation of autophagy was recently observed also in a bovine kidney cell line [211]. These observations could acquire particular relevance in view of the recently reported finding that beta cells in human type 2 diabetes have signs of altered autophagy, which may contribute to the loss of beta cell mass [212]. Like many other cellular defense mechanisms, autophagy can be considered as a double-edged sword. On the one hand, it has been recently demonstrated that constitutive autophagy plays important roles in the maintenance of pancreatic beta cell homeostasis [213216]. On the other hand, autophagy can paradoxically have either pro-survival or pro-death functions, depending on the context [217]. In beta cells, many pieces of evidence indicate that autophagy is a cell survival pathway that can also mediate cell death under certain condition, e.g., when autophagy is overactivated as in response, e.g., to endoplasmic reticulum stress [218] or to particularly severe organelle damage, as that induced by TCDD [181].

4. Conclusions

In the last few decades, a considerable body of epidemiological evidence has been accumulated, whose conclusions strongly suggest that exposure to dioxin and other POPs can be considered as a new risk factor for diabetes in humans in addition to the traditional lifestyle-related factors, such as excess of energy intake and a lack of exercise [14,43]. Consequently, the association between environmental pollutants and diabetes is now regarded as an emerging topic in the field of environmental health sciences and could be rightly considered as one of the “paradoxes of progress” [5]. On the other hand, an increasing number of experimental evidence clearly indicates that pancreatic beta cells can be considered a relevant and sensitive target of dioxin cytotoxicity, throwing some light on the underlying biological mechanisms (Figure 1). The obtained results have turned the attention towards new interesting pathogenetic perspectives, showing the possibility of a new, non-genomic pathway and indicating the mitochondrion as one of the main targets of the acute toxicity of dioxin. However, many basic questions are still to be addressed, and a further experimental effort will be necessary in the years to come to better understand how dioxin exposure could contribute to the worldwide rising prevalence of type 2 diabetes.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wild, S.; Roglic, G.; Green, A.; Sicree, R.; King, H. Global prevalence of diabetes: Estimates for the year 2000 and projections for 2030. Diabetes Care 2004, 27, 1047–1053. [Google Scholar]
  2. Kolb, H.; Mandrup-Poulsen, T. The global diabetes epidemics as a consequence of lifestyle-induced low-grade inflammation. Diabetologia 2010, 53, 10–20. [Google Scholar]
  3. Shaw, J.E.; Sicree, R.A.; Zimmet, P.Z. Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes Res. Clin. Pract 2010, 87, 4–14. [Google Scholar]
  4. Longnecker, M.P.; Daniels, J.L. Environmental contaminants as etiologic factors for diabetes. Environ. Health Perspect 2001, 109, 871–876. [Google Scholar]
  5. Neel, B.A.; Sargis, R.M. The paradox of progress: Environmental disruption of metabolism and the diabetes epidemic. Diabetes 2011, 60, 1838–1848. [Google Scholar]
  6. Remillard, R.B.; Bunce, N.J. Linking dioxins to diabetes: Epidemiology and biologic plausibility. Environ. Health Perspect 2002, 110, 853–858. [Google Scholar]
  7. Environmental Protection Agency, Special Report on Environmental Endocrine Disruption: An Effects Assessment and Analysis; U.S Environmental Protection Agency: Washington, DC, USA, 1997.
  8. Müllerová, D.; Kopecký, J. White adipose tissue: Storage and effector site for environmental pollutants. Physiol. Res 2007, 56, 375–381. [Google Scholar]
  9. Schug, T.T.; Janesick, A.; Blumberg, B.; Heindel, J.J. Endocrine disrupting chemicals and disease susceptibility. J. Steroid Biochem. Mol. Biol 2011, 127, 204–215. [Google Scholar]
  10. Alonso-Magdalena, P.; Quesada, I.; Nadal, A. Endocrine disruptors in the etiology of type 2 diabetes mellitus. Nat. Rev. Endocrinol 2011, 7, 346–353. [Google Scholar]
  11. Schafer, K.S.; Kegley, S.E. Persistent toxic chemicals in the US food supply. J. Epidemiol. Commun. Health 2002, 56, 813–817. [Google Scholar]
  12. Arisawa, K.; Takeda, H.; Mikasa, H. Background exposure to PCDDs/PCDFs/PCBs and its potential health effects: A review of epidemiologic studies. J. Med. Investig 2005, 52, 10–21. [Google Scholar]
  13. Lee, D.-H.; Jacobs, D.R.; Porta, M. Editorial: Could low-level background exposure to persistent organic pollutants contribute to the social burden of type 2 diabetes? J. Epidemiol. Commun. Health 2008, 60, 1006–1008. [Google Scholar]
  14. Taylor, K.W.; Novak, R.F.; Anderson, H.A.; Birnbaum, L.S.; Blystone, C.; Devito, M.; Jacobs, D.; Köhrle, J.; Lee, D.H.; Rylander, L.; et al. Evaluation of the association between persistent organic pollutants (POPs) and diabetes in epidemiological studies: A national toxicology program workshop review. Environ. Health Perspect 2013, 121, 774–783. [Google Scholar]
  15. Magliano, D.J.; Loh, V.H.; Harding, J.L.; Botton, J.; Shaw, J.E. Persistent organic pollutants and diabetes: A review of the epidemiological evidence. Diabetes Metab 2014, 40, 1–14. [Google Scholar]
  16. Booker, S.M. Dioxin in Vietnam: Fighting a legacy of war. Environ. Health Perspect 2001, 109, A116–A117. [Google Scholar]
  17. Longnecker, M.P.; Michalek, J.E. Serum dioxin level in relation to diabetes mellitus a mong Air Force veterans with background levels of exposure. Epidemiology 2000, 11, 44–48. [Google Scholar]
  18. Pavuk, M.; Patterson, D.G., Jr.; Turner, W.E.; Needham, L.L.; Ketchum, N.S. Polychlorinated dibenzo-p-dioxins (PCDDs), polychlorinated dibenzofurans (PCDFs), and dioxin-like polychlorinated biphenyls (PCBs) in the serum of US Air Force veterans in 2002. Chemosphere 2007, 68, 62–68. [Google Scholar]
  19. Michalek, J.E.; Tripathi, R.C. Pharmacokinetics of TCDD in veterans of Operation Ranch Hand: 15-year follow-up. J. Toxicol. Environ. Health A 1999, 57, 369–378. [Google Scholar]
  20. Henriksen, G.L.; Ketchum, N.S.; Michalek, J.E.; Swaby, J.A. Serum dioxin and diabetes mellitus in veterans of Operation Ranch Hand. Epidemiology 1997, 8, 252–258. [Google Scholar]
  21. Michalek, J.E.; Akhtar, F.Z.; Kiel, J.L. Serum dioxin, insulin, fasting glucose, and sex hormone-binding globulin in veterans of Operation Ranch Hand. J. Clin. Endocrinol. Metab 1999, 84, 1540–1543. [Google Scholar]
  22. Michalek, J.E.; Ketchum, N.S.; Tripathi, R.C. Diabetes mellitus and 2,3,7,8-tetrachlorodibenzo-p-dioxin elimination in veterans of Operation Ranch Hand. J. Toxicol. Environ. Health A 2003, 66, 211–221. [Google Scholar]
  23. Kang, H.K.; Dalager, N.A.; Needham, L.L.; Patterson, D.G., Jr.; Lees, P.S.; Yates, K.; Matanoski, G.M. Health status of Army Chemical Corps Vietnam veterans who sprayed defoliant in Vietnam. Am. J. Ind. Med 2006, 49, 875–884. [Google Scholar]
  24. Michalek, J.E.; Pavuk, M. Diabetes and cancer in veterans of Operation Ranch Hand after adjustment for calendar period, days of spraying, and time spent in Southeast Asia. J. Occup. Environ. Med 2008, 50, 330–340. [Google Scholar]
  25. Bertazzi, P.A.; Bernucci, I.; Brambilla, G.; Consonni, D.; Pesatori, A.C. The Seveso studies on early and long-term effects of dioxin exposure: A review. Environ. Health Perspect 1998, 106, 625–633. [Google Scholar]
  26. Bertazzi, P.A.; di Domenico, A. Health consequences of the Seveso, Italy, accident. In Dioxin and Health, 2nd ed; Schecter, A., Gasiewicz, T.A., Eds.; Wiley: Hoboken, NJ, USA, 2003; pp. 827–853. [Google Scholar]
  27. Pesatori, A.C. Dioxin contamination in Seveso: The social tragedy and the scientific challenge. Med. Lav 1995, 86, 111–124. [Google Scholar]
  28. Consonni, D.; Pesatori, A.C.; Zocchetti, C.; Sindaco, R.; D’Oro, L.C.; Rubagotti, M.; Bertazzi, P.A. Mortality in a population exposed to dioxin after the Seveso, Italy, accident in 1976: 25 years of follow-up. Am. J. Epidemiol 2008, 167, 847–858. [Google Scholar]
  29. Warner, M.; Mocarelli, P.; Brambilla, P.; Wesselink, A.; Samuels, S.; Signorini, S.; Eskenazi, B. Diabetes, metabolic syndrome, and obesity in relation to serum dioxin concentrations: The Seveso women’s health study. Environ. Health Perspect 2013, 121, 906–911. [Google Scholar]
  30. Naville, D.; Pinteur, C.; Vega, N.; Menade, Y.; Vigier, M.; le Bourdais, A.; Labaronne, E.; Debard, C.; Luquain-Costaz, C.; Bégeot, M.; et al. Low-dose food contaminants trigger sex-specific, hepatic metabolic changes in the progeny of obese mice. FASEB J 2013, 27, 3860–3870. [Google Scholar]
  31. Vena, J.; Boffetta, P.; Becher, H.; Benn, T.; Bueno-de-Mesquita, H.B.; Coggon, D.; Colin, D.; Flesch-Janys, D.; Green, L.; Kauppinen, T.; et al. Exposure to dioxin and nonneoplastic mortality in the expanded IARC international cohort study of phenoxy herbicide and chlorophenol production workers and sprayers. Environ. Health Perspect 1998, 106, 645–653. [Google Scholar]
  32. Kim, J.S.; Lim, H.S.; Cho, S.I.; Cheong, H.K.; Lim, M.K. Impact of Agent Orange exposure among Korean Vietnam veterans. Ind. Health 2003, 41, 149–157. [Google Scholar]
  33. Chen, H.L.; Su, H.J.; Guo, Y.L.; Liao, P.C.; Hung, C.F.; Lee, C.C. Biochemistry examinations and health disorder evaluation of Taiwanese living near incinerators and with low serum PCDD/Fs levels. Sci. Total Environ 2006, 366, 538–548. [Google Scholar]
  34. Kouznetsova, M.; Huang, X.; Ma, J.; Lessner, L.; Carpenter, D.O. Increased rate of hospitalization for diabetes and residential proximity of hazardous waste sites. Environ. Health Perspect 2007, 115, 75–79. [Google Scholar]
  35. Wang, S.L.; Tsai, P.C.; Yang, C.Y.; Leon Guo, Y. Increased risk of diabetes and polychlorinated biphenyls and dioxins: A 24-year follow-up study of the Yucheng cohort. Diabetes Care 2008, 31, 1574–1579. [Google Scholar]
  36. Steenland, K.; Piacitelli, L.; Deddens, J.; Fingerhut, M.; Chang, L.I. Cancer, heart disease, and diabetes in workers exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin. J. Natl. Cancer Inst 1999, 91, 779–786. [Google Scholar]
  37. Calvert, G.M.; Sweeney, M.H.; Deddens, J.; Wall, D.K. Evaluation of diabetes mellitus, serum glucose, and thyroid function among United States workers exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Occup. Environ. Med 1999, 56, 270–276. [Google Scholar]
  38. Steenland, K.; Calvert, G.; Ketchum, N.; Michalek, J. Dioxin and diabetes mellitus: An analysis of the combined NIOSH and Ranch Hand data. Occup. Environ. Med 2001, 58, 641–648. [Google Scholar]
  39. Karouna-Renier, N.K.; Rao, K.R.; Lanza, J.J.; Davis, D.A.; Wilson, P.A. Serum profiles of PCDDs and PCDFs, in individuals near the Escambia Wood Treating Company Superfund site in Pensacola, FL. Chemosphere 2007, 69, 1312–1319. [Google Scholar]
  40. Collins, J.J.; Bodner, K.; Aylward, L.L.; Wilken, M.; Bodnar, C.M. Mortality rates among trichlorophenol workers with exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Am. J. Epidemiol 2009, 170, 501–506. [Google Scholar]
  41. Collins, J.J.; Bodner, K.; Aylward, L.L.; Wilken, M.; Swaen, G.; Budinsky, R.; Rowlands, C.; Bodnar, C.M. Mortality rates among workers exposed to dioxins in the manufacture of pentachlorophenol. J. Occup. Environ. Med 2009, 51, 1212–1219. [Google Scholar]
  42. Kerger, B.D.; Scott, P.K.; Pavuk, M.; Gough, M.; Paustenbach, D.J. Re-analysis of Ranch Hand study supports reverse causation hypothesis between dioxin and diabetes. Crit. Rev. Toxicol 2012, 42, 669–687. [Google Scholar]
  43. Lee, D.H.; Porta, M.; Jacobs, D.R., Jr.; Vandenberg, L.N. Chlorinated persistent organic pollutants, obesity, and type 2 diabetes. Endocr. Rev 2014, in press. [Google Scholar]
  44. Fierens, S.; Mairesse, H.; Heilier, J.F.; de Burbure, C.; Focant, J.F.; Eppe, G.; de Pauw, E.; Bernard, A. Dioxin/polychlorinated biphenyl body burden, diabetes and endometriosis: findings in a population-based study in Belgium. Biomarkers 2003, 8, 529–534. [Google Scholar]
  45. Lee, D.H.; Lee, I.K.; Song, K.; Steffes, M.; Toscano, W.; Baker, B.A.; Jacobs, D.R., Jr. A strong dose-response relation between serum concentrations of persistent organic pollutants and diabetes: Results from the National Health and Examination Survey 1999–2002. Diabetes Care 2006, 29, 1638–1644. [Google Scholar]
  46. Lee, D.H.; Lee, I.K.; Steffes, M.; Jacobs, D.R., Jr. Extended analyses of the association between serum concentrations of persistent organic pollutants and diabetes. Diabetes Care 2007, 30, 1596–1598. [Google Scholar]
  47. Lee, D.H.; Lee, I.K.; Jin, S.H.; Steffes, M.; Jacobs, D.R., Jr. Association between serum concentrations of persistent organic pollutants and insulin resistance among nondiabetic adults: Results from the National Health and Nutrition Examination Survey 1999–2002. Diabetes Care 2007, 30, 622–628. [Google Scholar]
  48. Lee, D.H.; Lee, I.K.; Porta, M.; Steffes, M.; Jacobs, D.R., Jr. Relationship between serum concentrations of persistent organic pollutants and the prevalence of metabolic syndrome among non-diabetic adults: Results from the National Health and Nutrition Examination Survey 1999–2002. Diabetologia 2007, 50, 1841–1851. [Google Scholar]
  49. Lee, D.H.; Lind, L.; Jacobs, D.R., Jr.; Salihovic, S.; van Bavel, B.; Lind, P.M. Associations of persistent organic pollutants with abdominal obesity in the elderly: The Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study. Environ. Int 2012, 40, 170–178. [Google Scholar]
  50. Everett, C.J.; Frithsen, I.L.; Diaz, V.A.; Koopman, R.J.; Simpson, W.M., Jr.; Mainous, A.G., 3rd. Association of a polychlorinated dibenzo-p-dioxin, a polychlorinated biphenyl, and DDT with diabetes in the 1999–2002 National Health and Nutrition Examination Survey. Environ. Res 2007, 103, 413–418. [Google Scholar]
  51. Lee, D.H.; Jacobs, D.R., Jr.; Gross, M.; Kiefe, C.I.; Roseman, J.; Lewis, C.E.; Steffes, M. Gamma-glutamyltransferase is a predictor of incident diabetes and hypertension: The Coronary Artery Risk Development in Young Adults (CARDIA) Study. Clin. Chem 2003, 49, 1358–1366. [Google Scholar]
  52. Lee, D.H.; Jacobs, D.R., Jr. Association between serum concentrations of persistent organic pollutants and gamma glutamyltransferase: Results from the National Health and Examination Survey 1999–2002. Clin. Chem 2006, 52, 1825–1827. [Google Scholar]
  53. Rignell-Hydbom, A.; Rylander, L.; Hagmar, L. Exposure to persistent organochlorine pollutants and type 2 diabetes mellitus. Hum. Exp. Toxicol 2007, 26, 447–452. [Google Scholar]
  54. Uemura, H.; Arisawa, K.; Hiyoshi, M.; Satoh, H.; Sumiyoshi, Y.; Morinaga, K.; Kodama, K.; Suzuki, T.; Nagai, M.; Suzuki, T. Associations of environmental exposure to dioxins with prevalent diabetes among general inhabitants in Japan. Environ. Res 2008, 108, 63–68. [Google Scholar]
  55. Rignell-Hydbom, A.; Lidfeldt, J.; Kiviranta, H.; Rantakokko, P.; Samsioe, G.; Agardh, C.D.; Rylander, L. Exposure to p,p′-DDE: A risk factor for type 2 diabetes. PLoS One 2009, 4, e7503. [Google Scholar]
  56. Lee, D.H.; Steffes, M.W.; Sjödin, A.; Jones, R.S.; Needham, L.L.; Jacobs, D.R., Jr. Low dose of some persistent organic pollutants predicts type 2 diabetes: A nested case-control study. Environ. Health Perspect 2010, 118, 1235–1242. [Google Scholar]
  57. Tanaka, T.; Morita, A.; Kato, M.; Hirai, T.; Mizoue, T.; Terauchi, Y.; Watanabe, S.; Noda, M. SCOP Study Group. Congener-specific polychlorinated biphenyls and the prevalence of diabetes in the Saku Control Obesity Program (SCOP). Endocr. J 2011, 58, 589–596. [Google Scholar]
  58. Lee, D.H.; Lind, P.M.; Jacobs, D.R., Jr.; Salihovic, S.; van Bavel, B.; Lind, L. Polychlorinated biphenyls and organochlorine pesticides in plasma predict development of type 2 diabetes in the elderly: The prospective investigation of the vasculature in Uppsala Seniors (PIVUS) study. Diabetes Care 2011, 34, 1778–1784. [Google Scholar]
  59. Everett, C.J.; Thompson, O.M. Associations of dioxins, furans and dioxin-like PCBs with diabetes and pre-diabetes: Is the toxic equivalency approach useful? Environ. Res 2012, 118, 107–111. [Google Scholar]
  60. Nakamoto, M.; Arisawa, K.; Uemura, H.; Katsuura, S.; Takami, H.; Sawachika, F.; Yamaguchi, M.; Juta, T.; Sakai, T.; Toda, E.; et al. Association between blood levels of PCDDs/PCDFs/dioxin-like PCBs and history of allergic and other diseases in the Japanese population. Int. Arch. Occup. Environ. Health 2013, 86, 849–859. [Google Scholar]
  61. Bonefeld-Jorgensen, E. Biomonitoring in Greenland: Human biomarkers of exposure and effects—A short review. Rural Remote Health 2010, 10, 1362. [Google Scholar]
  62. Jørgensen, M.E.; Borch-Johnsen, K.; Bjerregaard, P. A cross-sectional study of the association between persistent organic pollutants and glucose intolerance among Greenland Inuit. Diabetologia 2008, 51, 1416–1422. [Google Scholar] [Green Version]
  63. Sharp, D. Environmental toxins, a potential risk factor for diabetes among Canadian Aboriginals. Int. J. Circumpolar Health 2009, 68, 316–326. [Google Scholar]
  64. Philibert, A.; Schwartz, H.; Mergler, D. An exploratory study of diabetes in a First Nation community with respect to serum concentrations of p,p′-DDE and PCBs and fish consumption. Int. J. Environ. Res. Public Health 2009, 6, 3179–3189. [Google Scholar]
  65. Rylander, L.; Rignell-Hydbom, A.; Hagmar, L. A cross-sectional study of the association between persistent organochlorine pollutants and diabetes. Environ. Health 2005, 4, 1–6. [Google Scholar]
  66. Turyk, M.; Anderson, H.A.; Knobeloch, L.; Imm, P.; Persky, V.W. Prevalence of diabetes and body burdens of polychlorinated biphenyls, polybrominated diphenyl ethers, and p,p′-diphenyldichloroethene in Great Lakes sport fish consumers. Chemosphere 2009, 75, 674–679. [Google Scholar]
  67. Fujiyoshi, P.T.; Michalek, J.E.; Matsumura, F. Molecular epidemiologic evidence for diabetogenic effects of dioxin exposure in U.S. Air force veterans of the Vietnam war. Environ. Health Perspect 2006, 114, 1677–1683. [Google Scholar]
  68. Patel, C.J.; Bhattacharya, J.; Butte, A.J. An Environment-Wide Association study (EWAS) on type 2 diabetes mellitus. PLoS One 2010, 5, e10746. [Google Scholar]
  69. Higginbotham, G.R.; Huang, A.; Firestone, D.; Verrett, J.; Ress, J.; Campbell, A.D. Chemical and toxicological evaluations of isolated and synthetic chloro derivatives of dibenzo-p-dioxin. Nature 1968, 220, 702–703. [Google Scholar]
  70. Kimbrough, R.D. Toxicity of chlorinated hydrocarbons and related compounds. A review including chlorinated dibenzodioxins and chlorinated dibenzofurans. Arch. Environ. Health 1972, 25, 125–131. [Google Scholar]
  71. Schwetz, B.A.; Norris, J.M.; Sparschu, G.L.; Rowe, U.K.; Gehring, P.J.; Emerson, J.L.; Gerbig, C.G. Toxicology of chlorinated dibenzo-p-dioxins. Environ. Health Perspect 1973, 5, 87–99. [Google Scholar]
  72. Dragan, Y.P.; Schrenk, D. Animal studies addressing the carcinogenicity of TCDD (or related compounds) with an emphasis on tumour promotion. Food Addit. Contam 2000, 17, 289–302. [Google Scholar]
  73. Hernández, L.G.; van Steeg, H.; Luijten, M.; van Benthem, J. Mechanisms of non-genotoxic carcinogens and importance of a weight of evidence approach. Mutat. Res 2009, 682, 94–109. [Google Scholar]
  74. Fischer, B. Receptor-mediated effects of chlorinated hydrocarbons. Andrologia 2000, 32, 279–283. [Google Scholar]
  75. Yonemoto, J. The effects of dioxin on reproduction and development. Ind. Health 2000, 38, 259–268. [Google Scholar]
  76. Petersen, S.L.; Krishnan, S.; Hudgens, E.D. The aryl hydrocarbon receptor pathway and sexual differentiation of neuroendocrine functions. Endocrinology 2006, 147, S33–S42. [Google Scholar]
  77. McConnell, E.E.; Moore, J.A.; Dalgard, D.W. Toxicity of 2,3,7,8 tetrachlorodibenzo-p-dioxin in rhesus monkeys (Macaca mulatta) following a single oral dose. Toxicol. Appl. Pharmacol 1978, 43, 175–187. [Google Scholar]
  78. Olson, J.R.; Holscher, M.A.; Neal, R.A. Toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in the golden Syrian hamster. Toxicol. Appl. Pharmacol 1980, 55, 67–78. [Google Scholar]
  79. Tuomisto, J.T.; Pohjanvirta, R.; Unkila, M.; Tuomisto, J. 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced anorexia and wasting syndrome in rats: Aggravation after ventromedial hypothalamic lesion. Eur. J. Pharmacol 1995, 293, 309–317. [Google Scholar]
  80. Lindén, J.; Lensu, S.; Tuomisto, J.; Pohjanvirta, R. Dioxins, the aryl hydrocarbon receptor and the central regulation of energy balance. Front. Neuroendocrinol 2010, 31, 452–478. [Google Scholar]
  81. Seefeld, M.D.; Corbett, S.W.; Keesey, R.E.; Peterson, R.E. Characterization of the wasting syndrome in rats treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol. Appl. Pharmacol 1984, 73, 311–322. [Google Scholar]
  82. Swift, L.L.; Gasiewicz, T.A.; Dunn, G.D.; Soulé, P.D.; Neal, R.A. Characterization of the hyperlipidemia in guinea pigs induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol. Appl. Pharmacol 1981, 59, 489–499. [Google Scholar]
  83. Brewster, D.W.; Matsumura, F. TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) reduces lipoprotein lipase activity in the adipose tissue of the guinea pig. Biochem. Biophys. Res. Commun 1984, 122, 810–817. [Google Scholar]
  84. Olsen, H.; Enan, E.; Matsumura, F. 2,3,7,8-Tetrachlorodibenzo-p-dioxin mechanism of action to reduce lipoprotein lipase activity in the 3T3-L1 preadipocyte cell line. J. Biochem. Mol. Toxicol 1998, 12, 29–39. [Google Scholar]
  85. Nishiumi, S.; Yabushita, Y.; Furuyashiki, T.; Fukuda, I.; Ashida, H. Involvement of SREBPs in 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced disruption of lipid metabolism in male guinea pig. Toxicol. Appl. Pharmacol 2008, 229, 281–289. [Google Scholar]
  86. Lo, R.; Celius, T.; Forgacs, A.L.; Dere, E.; MacPherson, L.; Harper, P.; Zacharewski, T.; Matthews, J. Identification of aryl hydrocarbon receptor binding targets in mouse hepatic tissue treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol. Appl. Pharmacol 2011, 257, 38–47. [Google Scholar]
  87. Forgacs, A.L.; Kent, M.N.; Makley, M.K.; Mets, B.; DelRaso, N.; Jahns, G.L.; Burgoon, L.D.; Zacharewski, T.R.; Reo, N.V. Comparative metabolomic and genomic analyses of TCDD-elicited metabolic disruption in mouse and rat liver. Toxicol. Sci 2012, 125, 41–55. [Google Scholar]
  88. Angrish, M.M.; Dominici, C.Y.; Zacharewski, T.R. TCDD-elicited effects on liver, serum, and adipose lipid composition in C57BL/6 mice. Toxicol. Sci 2013, 131, 108–115. [Google Scholar]
  89. Ebner, K.; Brewster, D.W.; Matsumura, F. Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on serum insulin and glucose levels in the rabbit. J. Environ. Sci. Health B 1988, 23, 427–438. [Google Scholar]
  90. Vogel, C.F.; Zhao, Y.; Wong, P.; Young, N.F.; Matsumura, F. The use of c-src knockout mice for the identification of the main toxic signaling pathway of TCDD to induce wasting syndrome. J. Biochem. Mol. Toxicol 2003, 17, 305–315. [Google Scholar]
  91. Kern, P.A.; Dicker-Brown, A.; Said, S.T.; Kennedy, R.; Fonseca, V.A. The stimulation of tumor necrosis factor and inhibition of glucose transport and lipoprotein lipase in adipose cells by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Metabolism 2002, 51, 65–68. [Google Scholar]
  92. Nishiumi, S.; Yoshida, M.; Azuma, T.; Yoshida, K.; Ashida, H. 2,3,7,8-tetrachlorodibenzo-p-dioxin impairs an insulin signaling pathway through the induction of tumor necrosis factor-alpha in adipocytes. Toxicol. Sci 2010, 115, 482–491. [Google Scholar]
  93. Li, W.; Vogel, C.F.; Matsumura, F. Studies on the cell treatment conditions to elicit lipolytic responses from 3T3-L1 adipocytes to TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin. J. Cell Biochem 2007, 102, 389–402. [Google Scholar]
  94. Fetissov, S.O.; Huang, P.; Zhang, Q.; Mimura, J.; Fujii-Kuriyama, Y.; Rannug, A.; Hökfelt, T.; Ceccatelli, S. Expression of hypothalamic neuropeptides after acute TCDD treatment and distribution of Ah receptor repressor. Regul. Pept 2004, 119, 113–124. [Google Scholar]
  95. Korkalainen, M.; Lindén, J.; Tuomisto, J.; Pohjanvirta, R. Effect of TCDD on mRNA expression of genes encoding bHLH/PAS proteins in rat hypothalamus. Toxicology 2005, 208, 1–11. [Google Scholar]
  96. Lindén, J.; Korkalainen, M.; Lensu, S.; Tuomisto, J.; Pohjanvirta, R. Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and leptin on hypothalamic mRNA expression of factors participating in food intake regulation in a TCDD-sensitive and a TCDD-resistant rat strain. J. Biochem. Mol. Toxicol 2005, 19, 139–148. [Google Scholar]
  97. Lensu, S.; Miettinen, R.; Pohjanvirta, R.; Lindén, J.; Tuomisto, J. Assessment by c-Fos immunostaining of changes in brain neural activity induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and leptin in rats. Basic Clin. Pharmacol. Toxicol 2006, 98, 363–371. [Google Scholar]
  98. Moon, B.H.; Hong, C.G.; Kim, S.Y.; Kim, H.J.; Shin, S.K.; Kang, S.; Lee, K.J.; Kim, Y.K.; Lee, M.S.; Shin, K.H. A single administration of 2,3,7,8-tetrachlorodibenzo-p-dioxin that produces reduced food and water intake induces long-lasting expression of corticotropin-releasing factor, arginine vasopressin, and proopiomelanocortin in rat brain. Toxicol. Appl. Pharmacol 2008, 233, 314–322. [Google Scholar]
  99. Enan, E.; Liu, P.C.; Matsumura, F. 2,3,7,8-Tetrachlorodibenzo-p-dioxin causes reduction of glucose transporting activities in the plasma membranes of adipose tissue and pancreas from the guinea pig. J. Biol. Chem 1992, 267, 19785–19791. [Google Scholar]
  100. Enan, E.; Liu, P.C.; Matsumura, F. TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) causes reduction in glucose uptake through glucose transporters on the plasma membrane of the guinea pig adipocyte. J. Environ. Sci. Health B 1992, 27, 495–510. [Google Scholar]
  101. Hsu, H.F.; Tsou, T.C.; Chao, H.R.; Kuo, Y.T.; Tsai, F.Y.; Yeh, S.C. Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on adipogenic differentiation and insulin-induced glucose uptake in 3T3-L1 cells. J. Hazard. Mater 2010, 182, 649–655. [Google Scholar]
  102. Olsen, H.; Enan, E.; Matsumura, F. Regulation of glucose transport in the NIH 3T3 L1 preadipocyte cell line by TCDD. Environ. Health Perspect 1994, 102, 454–458. [Google Scholar]
  103. Enan, E.; Lasley, B.; Stewart, D.; Overstreet, J.; Vandevoort, C.A. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) modulates function of human luteinizing granulosa cells via cAMP signaling and early reduction of glucose transporting activity. Reprod. Toxicol 1996, 10, 191–198. [Google Scholar]
  104. Enan, E.; Matsumura, F. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD)-induced changes in glucose transporting activity in guinea pigs, mice, and rats in vivo and in vitro. J. Biochem. Toxicol 1994, 9, 97–106. [Google Scholar]
  105. Liu, P.C.; Matsumura, F. Differential effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on the “adipose-type” and “brain-type” glucose transporters in mice. Mol. Pharmacol 1995, 47, 65–73. [Google Scholar]
  106. Nagashima, H.; Matsumura, F. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD)-induced down-regulation of glucose transporting activities in mouse 3T3-L1 preadipocyte. J. Environ. Sci. Health B 2002, 37, 1–14. [Google Scholar]
  107. Ishida, T.; Kan-o, S.; Mutoh, J.; Takeda, S.; Ishii, Y.; Hashiguchi, I.; Akamine, A.; Yamada, H. 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced change in intestinal function and pathology: Evidence for the involvement of arylhydrocarbon receptor-mediated alteration of glucose transportation. Toxicol. Appl. Pharmacol 2005, 205, 89–97. [Google Scholar]
  108. Liu, P.C.; Matsumura, F. TCDD suppresses insulin-responsive glucose transporter (GLUT-4) gene expression through C/EBP nuclear transcription factors in 3T3-L1 adipocytes. J. Biochem. Mol. Toxicol 2006, 20, 79–87. [Google Scholar]
  109. Tonack, S.; Kind, K.; Thompson, J.G.; Wobus, A.M.; Fischer, B.; Santos, A.N. Dioxin affects glucose transport via the arylhydrocarbon receptor signal cascade in pluripotent embryonic carcinoma cells. Endocrinology 2007, 148, 5902–5912. [Google Scholar]
  110. Matsumura, F. Mechanism of action of dioxin-type chemicals, pesticides, and other xenobiotics affecting nutritional indexes. Am. J. Clin. Nutr 1995, 61, 695S–701S. [Google Scholar]
  111. Gorski, J.R.; Rozman, K. Dose-response and time course of hypothyroxinemia and hypoinsulinemia and characterization of insulin hypersensitivity in 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-treated rats. Toxicology 1987, 44, 297–307. [Google Scholar]
  112. Gorski, J.R.; Muzi, G.; Weber, L.W.; Pereira, D.W.; Arceo, R.J.; Iatropoulos, M.J.; Rozman, K. Some endocrine and morphological aspects of the acute toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Toxicol. Pathol 1988, 16, 313–320. [Google Scholar]
  113. Novelli, M.; Piaggi, S.; de Tata, V. 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced impairment of glucose-stimulated insulin secretion in isolated rat pancreatic islets. Toxicol. Lett 2005, 156, 307–314. [Google Scholar]
  114. Kurita, H.; Yoshioka, W.; Nishimura, N.; Kubota, N.; Kadowaki, T.; Tohyama, C. Aryl hydrocarbon receptor-mediated effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on glucose-stimulated insulin secretion in mice. J. Appl. Toxicol 2009, 29, 689–694. [Google Scholar]
  115. Panten, U.; Kriegstein, E.; Poser, W.; Schönborn, J.; Hasselblatt, A. Effects of l-leucine and alpha-ketoisocaproic acid upon insulin secretion and metabolism of isolated pancreatic islets. FEBS Lett 1972, 20, 225–228. [Google Scholar]
  116. Lenzen, S.; Formanek, H.; Panten, U. Signal function of metabolism of neutral amino acids and 2-keto acids for initiation of insulin secretion. J. Biol. Chem 1982, 257, 6631–6633. [Google Scholar]
  117. Lembert, N.; Idahl, L.A. Alpha-ketoisocaproate is not a true substrate for ATP production by pancreatic beta-cell mitochondria. Diabetes 1998, 47, 339–344. [Google Scholar]
  118. Matschinsky, F.M. Glucokinase as glucose sensor and metabolic signal generator in pancreatic beta-cells and hepatocytes. Diabetes 1990, 39, 647–652. [Google Scholar]
  119. Malaisse, W.J.; Malaisse-Lagae, F.; Rasschaert, J.; Zähner, D.; Sener, A.; Davies, D.R.; van Schaftingen, E. The fuel concept for insulin release: Regulation of glucose phosphorylation in pancreatic islets. Biochem. Soc. Trans 1990, 18, 107–108. [Google Scholar]
  120. Efrat, S.; Tal, M.; Lodish, H.F. The pancreatic beta-cell glucose sensor. Trends Biochem. Sci 1994, 19, 535–538. [Google Scholar]
  121. Orci, L.; Ravazzola, M.; Baetens, D.; Inman, L.; Amherdt, M.; Peterson, R.G.; Newgard, C.B.; Johnson, J.H.; Unger, R.H. Evidence that down-regulation of beta-cell glucose transporters in non-insulin-dependent diabetes may be the cause of diabetic hyperglycemia. Proc. Natl. Acad. Sci. USA 1990, 87, 9953–9957. [Google Scholar]
  122. Thorens, B.; Weir, G.C.; Leahy, J.L.; Lodish, H.F.; Bonner-Weir, S. Reduced expression of the liver/beta-cell glucose transporter isoform in glucose-insensitive pancreatic beta cells of diabetic rats. Proc. Natl. Acad. Sci. USA 1990, 87, 6492–6496. [Google Scholar]
  123. Valera, A.; Solanes, G.; Fernández-Alvarez, J.; Pujol, A.; Ferrer, J.; Asins, G.; Gomis, R.; Bosch, F. Expression of GLUT-2 antisense RNA in beta cells of transgenic mice leads to diabetes. J. Biol. Chem 1994, 269, 28543–28546. [Google Scholar]
  124. Fisher, J.M.; Jones, K.W.; Whitlock, J.P., Jr. Activation of transcription as a general mechanism of 2,3,7,8-tetrachlorodibenzo-p-dioxin action. Mol. Carcinog 1989, 1, 216–221. [Google Scholar]
  125. Silbergeld, E.K.; Gasiewicz, T.A. Dioxins and the Ah receptor. Am. J. Ind. Med 1989, 16, 455–474. [Google Scholar]
  126. Bock, K.W.; Köhle, C. Ah receptor: Dioxin-mediated toxic responses as hints to deregulated physiologic functions. Biochem. Pharmacol 2006, 72, 393–404. [Google Scholar]
  127. Hankinson, O. The aryl hydrocarbon receptor complex. Annu. Rev. Pharmacol. Toxicol 1995, 35, 307–340. [Google Scholar]
  128. Gorski, J.R.; Weber, L.W.; Rozman, K. Tissue-specific alterations of de novo fatty acid synthesis in 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-treated rats. Arch. Toxicol 1988, 62, 146–151. [Google Scholar]
  129. Weber, L.W.; Lebofsky, M.; Greim, H.; Rozman, K. Key enzymes of gluconeogenesis are dose-dependently reduced in 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-treated rats. Arch. Toxicol 1991, 65, 119–123. [Google Scholar]
  130. Weber, L.W.; Lebofsky, M.; Stahl, B.U.; Kettrup, A.; Rozman, K. Comparative toxicity of four chlorinated dibenzo-p-dioxins (CDDs) and their mixture. Part II: Structure-activity relationships with inhibition of hepatic phosphoenolpyruvate carboxykinase, pyruvate carboxylase, and gamma-glutamyl transpeptidase activities. Arch. Toxicol 1992, 66, 478–483. [Google Scholar]
  131. Stahl, B.U.; Beer, D.G.; Weber, L.W.; Rozman, K. Reduction of hepatic phosphoenolpyruvate carboxykinase (PEPCK) activity by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is due to decreased mRNA levels. Toxicology 1993, 79, 81–95. [Google Scholar]
  132. Fan, F.; Yan, B.; Wood, G.; Viluksela, M.; Rozman, K.K. Cytokines (IL-1beta and TNFalpha) in relation to biochemical and immunological effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in rats. Toxicology 1997, 116, 9–16. [Google Scholar]
  133. Croutch, C.R.; Lebofsky, M.; Schramm, K.W.; Terranova, P.F.; Rozman, K.K. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and 1,2,3,4,7,8-hexachlorodibenzo-p-dioxin (HxCDD) alter body weight by decreasing insulin-like growth factor I (IGF-I) signaling. Toxicol. Sci 2005, 85, 560–571. [Google Scholar]
  134. Fletcher, N.; Wahlström, D.; Lundberg, R.; Nilsson, C.B.; Nilsson, K.C.; Stockling, K.; Hellmold, H.; Håkansson, H. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) alters the mRNA expression of critical genes associated with cholesterol metabolism, bile acid biosynthesis, and bile transport in rat liver: a microarray study. Toxicol. Appl. Pharmacol 2005, 207, 1–24. [Google Scholar]
  135. Sato, S.; Shirakawa, H.; Tomita, S.; Ohsaki, Y.; Haketa, K.; Tooi, O.; Santo, N.; Tohkin, M.; Furukawa, Y.; Gonzalez, F.J.; et al. Low-dose dioxins alter gene expression related to cholesterol biosynthesis, lipogenesis, and glucose metabolism through the aryl hydrocarbon receptor-mediated pathway in mouse liver. Toxicol. Appl. Pharmacol 2008, 229, 10–19. [Google Scholar]
  136. Zhang, W.; Sargis, R.M.; Volden, P.A.; Carmean, C.M.; Sun, X.J.; Brady, M.J. PCB 126 and other dioxin-like PCBs specifically suppress hepatic PEPCK expression via the aryl hydrocarbon receptor. PLoS One 2012, 7, e37103. [Google Scholar]
  137. Puga, A.; Maier, A.; Medvedovic, M. The transcriptional signature of dioxin in human hepatoma HepG2 cells. Biochem. Pharmacol 2000, 60, 1129–1142. [Google Scholar]
  138. Diani-Moore, S.; Ram, P.; Li, X.; Mondal, P.; Youn, D.Y.; Sauve, A.A.; Rifkind, A.B. Identification of the aryl hydrocarbon receptor target gene tiparp as a mediator of suppression of hepatic gluconeogenesis by 2,3,7,8-tetrachlorodibenzo-p-dioxin and of nicotinamide as a corrective agent for this effect. J. Biol. Chem 2010, 285, 38801–38810. [Google Scholar]
  139. Donath, M.Y.; Böni-Schnetzler, M.; Ellingsgaard, H.; Halban, P.A.; Ehses, J.A. Cytokine production by islets in health and diabetes: cellular origin, regulation and function. Trends Endocrinol. Metab 2010, 21, 261–267. [Google Scholar]
  140. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar]
  141. Donath, M.Y.; Schumann, D.M.; Faulenbach, M.; Ellingsgaard, H.; Perren, A.; Ehses, J.A. Islet inflammation in type 2 diabetes: From metabolic stress to therapy. Diabetes Care 2008, 31, S161–S164. [Google Scholar]
  142. Matsumura, F. On the significance of the role of cellular stress response reactions in the toxic actions of dioxin. Biochem. Pharmacol 2003, 66, 527–540. [Google Scholar]
  143. Matsumura, F.; Vogel, C.F. Evidence supporting the hypothesis that one of the main functions of the aryl hydrocarbon receptor is mediation of cell stress responses. Biol. Chem 2006, 387, 1189–1194. [Google Scholar]
  144. Kim, M.J.; Pelloux, V.; Guyot, E.; Tordjman, J.; Bui, L.C.; Chevallier, A.; Forest, C.; Benelli, C.; Clément, K.; Barouki, R. Inflammatory pathway genes belong to major targets of persistent organic pollutants in adipose cells. Environ. Health Perspect 2012, 120, 508–514. [Google Scholar]
  145. Vogel, C.F.; Kahn, E.M.; Leung, P.S.; Gershwin, M.E.; Chang, W.L.; Wu, D.; Haarmann-Stemmann, T.; Hoffmann, A.; Denison, M.S. Cross-talk between Aryl Hydrocarbon Receptor and the inflammatory response: A Role for NF-κB. J. Biol. Chem 2014, 289, 1866–1875. [Google Scholar]
  146. Dong, B.; Matsumura, F. Roles of cytosolic phospholipase A2 and Src kinase in the early action of 2,3,7,8-tetrachlorodibenzo-p-dioxin through a nongenomic pathway in MCF10A cells. Mol. Pharmacol 2008, 74, 255–263. [Google Scholar]
  147. Dong, B.; Matsumura, F. The conversion of rapid TCCD nongenomic signals to persistent inflammatory effects via select protein kinases in MCF10A cells. Mol. Endocrinol 2009, 23, 549–558. [Google Scholar]
  148. Li, W.; Matsumura, F. Significance of the nongenomic, inflammatory pathway in mediating the toxic action of TCDD to induce rapid and long-term cellular responses in 3T3-L1 adipocytes. Biochemistry 2008, 47, 13997–14008. [Google Scholar]
  149. Sciullo, E.M.; Dong, B.; Vogel, C.F.; Matsumura, F. Characterization of the pattern of the nongenomic signaling pathway through which TCDD-induces early inflammatory responses in U937 human macrophages. Chemosphere 2009, 74, 1531–1537. [Google Scholar]
  150. Dong, B.; Nishimura, N.; Vogel, C.F.; Tohyama, C.; Matsumura, F. TCDD-induced cyclooxygenase-2 expression is mediated by the nongenomic pathway in mouse MMDD1 macula densa cells and kidneys. Biochem. Pharmacol 2010, 79, 487–497. [Google Scholar]
  151. Xu, G.; Li, Y.; Yoshimoto, K.; Chen, G.; Wan, C.; Iwata, T.; Mizusawa, N.; Duan, Z.; Liu, J.; Jiang, J. 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced inflammatory activation is mediated by intracellular free calcium in microglial cells. Toxicology 2013, 308, 158–167. [Google Scholar]
  152. Matsumura, F. The significance of the nongenomic pathway in mediating inflammatory signaling of the dioxin-activated Ah receptor to cause toxic effects. Biochem. Pharmacol 2009, 77, 608–626. [Google Scholar]
  153. Prentki, M.; Nolan, C.J. Islet beta cell failure in type 2 diabetes. J. Clin. Investig 2006, 116, 1802–1812. [Google Scholar]
  154. Ashcroft, F.M.; Rorsman, P. Diabetes mellitus and the β cell: The last ten years. Cell 2012, 148, 1160–1171. [Google Scholar]
  155. Hectors, T.L.; Vanparys, C.; van der Ven, K.; Martens, G.A.; Jorens, P.G.; van Gaal, L.F.; Covaci, A.; de Coen, W.; Blust, R. Environmental pollutants and type 2 diabetes: A review of mechanisms that can disrupt beta cell function. Diabetologia 2011, 54, 1273–1290. [Google Scholar]
  156. Asfari, M.; Janjic, D.; Meda, P.; Li, G.; Halban, P.A.; Wollheim, C.B. Establishment of 2-mercaptoethanol-dependent differentiated insulin-secreting cell lines. Endocrinology 1992, 130, 167–178. [Google Scholar]
  157. Merglen, A.; Theander, S.; Rubi, B.; Chaffard, G.; Wollheim, C.B.; Maechler, P. Glucose sensitivity and metabolism-secretion coupling studied during two-year continuous culture in INS-1E insulinoma cells. Endocrinology 2004, 145, 667–678. [Google Scholar]
  158. Piaggi, S.; Novelli, M.; Martino, L.; Masini, M.; Raggi, C.; Orciuolo, E.; Masiello, P.; Casini, A.; de Tata, V. Cell death and impairment of glucose-stimulated insulin secretion induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the beta-cell line INS-1E. Toxicol. Appl. Pharmacol 2007, 220, 333–340. [Google Scholar]
  159. Mitrou, P.I.; Dimitriadis, G.; Raptis, S.A. Toxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin and related compounds. Eur. J. Intern. Med 2001, 12, 406–411. [Google Scholar]
  160. Sakurai, K.; Katoh, M.; Someno, K.; Fujimoto, Y. Apoptosis and mitochondrial damage in INS-1 cells treated with alloxan. Biol. Pharm. Bull 2001, 24, 876–882. [Google Scholar]
  161. Hanneman, W.H.; Legare, M.E.; Barhoumi, R.; Burghardt, R.C.; Safe, S.; Tiffany-Castiglioni, E. Stimulation of calcium uptake in cultured rat hippocampal neurons by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicology 1996, 112, 19–28. [Google Scholar]
  162. Puga, A.; Hoffer, A.; Zhou, S.; Bohm, J.M.; Leikauf, G.D.; Shertzer, H.G. Sustained increase in intracellular free calcium and activation of cyclooxigenase-2 expression in mouse hepatoma cells treated with dioxin. Biochem. Pharmacol 1997, 54, 1287–1296. [Google Scholar]
  163. Tannheimer, S.L.; Barton, S.L.; Ethier, S.P.; Burchiel, S.W. Carcinogenic polycyclic aromatic hydrocarbons increase intracellular Ca2+ and cell proliferation in primary human mammary epithelial cells. Carcinogenesis 1997, 18, 1177–1182. [Google Scholar]
  164. N’Diaye, M.; le Ferrec, E.; Lagadic-Gossmann, D.; Corre, S.; Gilot, D.; Lecureur, V.; Monteiro, P.; Rauch, C.; Galibert, M.D.; Fardel, O. Aryl hydrocarbon receptor- and calcium-dependent induction of the chemokine CCL1 by the environmental contaminant benzo[a]pyrene. J. Biol. Chem 2006, 281, 19906–19915. [Google Scholar]
  165. Dale, Y.R.; Eltom, S.E. Calpain mediates the dioxin-induced activation and down-regulation of the aryl hydrocarbon receptor. Mol. Pharmacol 2006, 70, 1481–1487. [Google Scholar]
  166. Xie, A.; Walker, N.J.; Wang, D. Dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin) enhances triggered afterdepolarizations in rat ventricular myocytes. Cardiovasc. Toxicol 2006, 6, 99–110. [Google Scholar]
  167. Kim, S.Y.; Lee, H.G.; Choi, E.J.; Park, K.Y.; Yang, J.H. TCDD alters PKC signaling pathways in developing neuronal cells in culture. Chemosphere 2007, 67, S421–S427. [Google Scholar]
  168. Monteiro, P.; Gilot, D.; le Ferrec, E.; Rauch, C.; Lagadic-Gossmann, D.; Fardel, O. Dioxin-mediated up-regulation of aryl hydrocarbon receptor target genes is dependent on the calcium/calmodulin/CaMKIalpha pathway. Mol. Pharmacol 2008, 73, 769–777. [Google Scholar]
  169. Sul, D.; Kim, H.S.; Cho, E.K.; Lee, M.; Kim, H.S.; Jung, W.W.; Hwang, K.W.; Park, S.Y. 2,3,7,8-TCDD neurotoxicity in neuroblastoma cells is caused by increased oxidative stress, intracellular calcium levels, and tau phosphorylation. Toxicology 2009, 255, 65–71. [Google Scholar]
  170. Kobayashi, D.; Ahmed, S.; Ishida, M.; Kasai, S.; Kikuchi, H. Calcium/calmodulin signaling elicits release of cytochrome c during 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced apoptosis in the human lymphoblastic T-cell line, L-MAT. Toxicology 2009, 258, 25–32. [Google Scholar]
  171. Kim, Y.H.; Shim, Y.J.; Shin, Y.J.; Sul, D.; Lee, E.; Min, B.H. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces calcium influx through T-type calcium channel and enhances lysosomal exocytosis and insulin secretion in INS-1 cells. Int. J. Toxicol 2009, 28, 151–161. [Google Scholar]
  172. Morales-Hernández, A.; Sánchez-Martín, F.J.; Hortigón-Vinagre, M.P.; Henao, F.; Merino, J.M. 2,3,7,8-Tetrachlorodibenzo-p-dioxin induces apoptosis by disruption of intracellular calcium homeostasis in human neuronal cell line SHSY5Y. Apoptosis 2012, 17, 1170–1181. [Google Scholar]
  173. Parekh, A.B.; Putney, J.W., Jr. Store-operated calcium channels. Physiol. Rev 2005, 85, 757–810. [Google Scholar]
  174. Orrenius, S.; Zhivotovsky, B.; Nicotera, P. Regulation of cell death: The calcium-apoptosis link. Nat. Rev. Mol. Cell Biol 2003, 4, 552–565. [Google Scholar]
  175. Brookes, P.S.; Yoon, Y.; Robotham, J.L.; Anders, M.W.; Sheu, S.-S.; Calcium, A.T.P. ROS: A mitochondrial love-hate triangle. Am. J. Physiol. Cell Physiol 2004, 287, C817–C833. [Google Scholar]
  176. Senft, A.P.; Dalton, T.P.; Nebert, D.W.; Genter, M.B.; Hutchinson, R.J.; Shertzer, H.G. Dioxin increases reactive oxygen production in mouse liver mitochondria. Toxicol. Appl. Pharmacol 2002, 185, 74–75. [Google Scholar]
  177. Shen, D.; Dalton, T.P.; Nerbert, D.W.; Shertzer, H.G. Glutathione redox state regulates mitochondrial reactive oxygen production. J. Biol. Chem 2005, 280, 25305–25312. [Google Scholar]
  178. Shertzer, H.G.; Genter, M.B.; Shen, D.; Nebert, D.W.; Chen, Y.; Dalton, T.P. TCDD decreases ATP levels and increases reactive oxygen production through changes in mitochondrial F0F1-ATP synthase and ubiquinone. Toxicol. Appl. Pharmacol 2006, 217, 363–374. [Google Scholar]
  179. Maechler, P.; Carobbio, S.; Rubi, B. In beta-cells, mitochondria integrate and generate metabolic signals controlling insulin secretion. Int. J. Biochem. Cell Biol 2006, 38, 696–709. [Google Scholar]
  180. Martino, L.; Novelli, M.; Masini, M.; Chimenti, D.; Piaggi, S.; Masiello, P.; de Tata, V. Dehydroascorbate protection against dioxin-induced toxicity in the beta-cell line INS-1E. Toxicol. Lett 2009, 189, 27–34. [Google Scholar]
  181. Martino, L.; Masini, M.; Novelli, M.; Giacopelli, D.; Beffy, P.; Masiello, P.; de Tata, V. The aryl receptor inhibitor epigallocatechin-3-gallate protects INS-1E beta-cell line against acute dioxin toxicity. Chemosphere 2013, 93, 1447–1455. [Google Scholar]
  182. Mannella, C.A. The relevance of mitochondrial membrane topology to mitochondrial function. Biochim. Biophys. Acta 2006, 1762, 140–147. [Google Scholar]
  183. Kroemer, G.; Galluzzi, L.; Brenner, C. Mitochondrial membrane permeabilization in cell death. Physiol. Rev 2007, 87, 99–163. [Google Scholar]
  184. Zhang, Z.; Ding, Y.; Dai, X.; Wang, J.; Li, Y. Epigallocatechin-3-gallate protects pro-inflammatory cytokine induced injuries in insulin-producing cells through the mitochondrial pathway. Eur. J. Pharmacol 2011, 670, 311–316. [Google Scholar]
  185. Lim, S.; Cho, Y.M.; Park, K.S.; Lee, H.K. Persistent organic pollutants, mitochondrial dysfunction, and metabolic syndrome. Ann. N. Y. Acad. Sci 2010, 1201, 166–176. [Google Scholar]
  186. Lee, H.K. Mitochondrial dysfunction and insulin resistance: The contribution of dioxin-like substances. Diabetes Metab. J 2011, 35, 207–215. [Google Scholar]
  187. Park, W.H.; Jun, D.W.; Kim, J.T.; Jeong, J.H.; Park, H.; Chang, Y.S.; Park, K.S.; Lee, H.K.; Pak, Y.K. Novel cell-based assay reveals associations of circulating serum AhR-ligands with metabolic syndrome and mitochondrial dysfunction. Biofactors 2013, 39, 494–504. [Google Scholar]
  188. Kennedy, L.H.; Sutter, C.H.; Leon Carrion, S.; Tran, Q.T.; Bodreddigari, S.; Kensicki, E.; Mohney, R.P.; Sutter, T.R. 2,3,7,8-Tetrachlorodibenzo-p-dioxin-mediated production of reactive oxygen species is an essential step in the mechanism of action to accelerate human keratinocyte differentiation. Toxicol. Sci 2013, 132, 235–249. [Google Scholar]
  189. Pereira, S.P.; Pereira, G.C.; Pereira, C.V.; Carvalho, F.S.; Cordeiro, M.H.; Mota, P.C.; Ramalho-Santos, J.; Moreno, A.J.; Oliveira, P.J. Dioxin-induced acute cardiac mitochondrial oxidative damage and increased activity of ATP-sensitive potassium channels in Wistar rats. Environ. Pollut 2013, 180, 281–290. [Google Scholar] [Green Version]
  190. Fröjdö, S.; Vidal, H.; Pirola, L. Alterations of insulin signaling in type 2 diabetes: A review of the current evidence from humans. Biochim. Biophys. Acta 2009, 1792, 83–92. [Google Scholar]
  191. Lowell, B.B.; Shulman, G.I. Mitochondrial dysfunction and type 2 diabetes. Science 2005, 307, 384–387. [Google Scholar]
  192. Abdul-Ghani, M.A.; DeFronzo, R.A. Mitochondrial dysfunction, insulin resistance, and type 2 diabetes mellitus. Curr. Diabetes Rep 2008, 8, 173–178. [Google Scholar]
  193. Cheng, Z.; Tseng, Y.; White, M.F. Insulin signaling meets mitochondria in metabolism. Trends Endocrinol. Metab 2010, 21, 589–598. [Google Scholar]
  194. López-Armada, M.J.; Riveiro-Naveira, R.R.; Vaamonde-García, C.; Valcárcel-Ares, M.N. Mitochondrial dysfunction and the inflammatory response. Mitochondrion 2013, 13, 106–118. [Google Scholar]
  195. Ruzzin, J.; Petersen, R.; Meugnier, E.; Madsen, L.; Lock, E.J.; Lillefosse, H.; Ma, T.; Pesenti, S.; Sonne, S.B.; Marstrand, T.T.; et al. Persistent organic pollutant exposure leads to insulin resistance syndrome. Environ. Health Perspect 2010, 118, 465–471. [Google Scholar]
  196. Slezak, B.P.; Hatch, G.E.; DeVito, M.J.; Diliberto, J.J.; Slade, R.; Crissman, K.; Hassoun, E.; Birnbaum, L.S. Oxidative stress in female B6C3F1 mice following acute and subchronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Toxicol. Sci 2000, 54, 390–398. [Google Scholar]
  197. Regnier, S.M.; Sargis, R.M. Adipocytes under assault: Environmental disruption of adipose physiology. Biochim. Biophys. Acta 2014, 1842, 520–533. [Google Scholar]
  198. Baillie-Hamilton, P.F. Chemical toxins: A hypothesis to explain the global obesity epidemic. J. Altern. Complement. Med 2002, 8, 185–192. [Google Scholar]
  199. Arsenescu, V.; Arsenescu, R.I.; King, V.; Swanson, H.; Cassis, L.A. Polychlorinated biphenyl-77 induces adipocyte differentiation and proinflammatory adipokines and promotes obesity and atherosclerosis. Environ. Health Perspect 2008, 116, 761–768. [Google Scholar]
  200. Ibrahim, M.M.; Fjære, E.; Lock, E.J.; Naville, D.; Amlund, H.; Meugnier, E.; Frøyland, L.; Le Magueresse Battistoni, B.; Madsen, L.; Jessen, N.; et al. Chronic consumption of farmed salmon containing persistent organic pollutants causes insulin resistance and obesity in mice. PLoS One 2011, 6, e25170. [Google Scholar]
  201. Gauthier, M.S.; Rabasa-Lhoret, R.; Prud’homme, D.; Karelis, A.D.; Geng, D.; van Bavel, B.; Ruzzin, J. The metabolically healthy but obese phenotype is associated with lower plasma levels of persistent organic pollutants as compared to the metabolically abnormal obese phenotype. J. Clin. Endocrinol. Metab 2014. [Google Scholar] [CrossRef]
  202. Tan, Z.; Chang, X.; Puga, A.; Xia, Y. Activation of mitogen-activated protein kinases (MAPKs) by aromatic hydrocarbons: Role in the regulation of aryl hydrocarbon receptor (AHR) function. Biochem. Pharmacol 2002, 64, 771–780. [Google Scholar]
  203. Kwon, M.J.; Jeong, K.S.; Choi, E.J.; Lee, B.H. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD)-induced activation of mitogen-activated protein kinase signaling pathway in Jurkat T cells. Pharmacol. Toxicol 2003, 93, 186–190. [Google Scholar]
  204. Weiss, C.; Faust, D.; Dürk, H.; Kolluri, S.K.; Pelzer, A.; Schneider, S.; Dietrich, C.; Oesch, F.; Göttlicher, M. TCDD induces c-jun expression via a novel Ah (dioxin) receptor-mediated p38-MAPK-dependent pathway. Oncogene 2005, 24, 4975–4983. [Google Scholar]
  205. Park, S.J.; Yoon, W.K.; Kim, H.J.; Son, H.Y.; Cho, S.W.; Jeong, K.S.; Kim, T.H.; Kim, S.H.; Kim, S.R.; Ryu, S.Y. 2,3,7,8-Tetrachlorodibenzo-p-dioxin activates ERK and p38 mitogen-activated protein kinases in RAW 264.7 cells. Anticancer Res 2005, 25, 2831–2836. [Google Scholar]
  206. Sciullo, E.M.; Vogel, C.F.; Wu, D.; Murakami, A.; Ohigashi, H.; Matsumura, F. Effects of selected food phytochemicals in reducing the toxic actions of TCDD and p,p′-DDT in U937 macrophages. Arch. Toxicol 2010, 84, 957–966. [Google Scholar]
  207. Mukai, R.; Shirai, Y.; Saito, N.; Fukuda, I.; Nishiumi, S.; Yoshida, K.; Ashida, H. Suppression mechanisms of flavonoids on aryl hydrocarbon receptor-mediated signal transduction. Arch. Biochem. Biophys 2010, 501, 134–141. [Google Scholar]
  208. Palermo, C.M.; Westlake, C.A.; Gasiewicz, T.A. Epigallocatechin gallate inhibits aryl hydrocarbon receptor gene transcription through an indirect mechanism involving binding to a 90 kDa heat shock protein. Biochemistry 2005, 44, 5041–5052. [Google Scholar]
  209. Park, S.; Dong, B.; Matsumura, F. Rapid activation of c-Src kinase by dioxin is mediated by the Cdc37-HSP90 complex as part of Ah receptor signaling in MCF10A cells. Biochemistry 2007, 46, 899–908. [Google Scholar]
  210. Klionsky, D.J.; Abdalla, F.C.; Abeliovich, H.; Abraham, R.T.; Acevedo-Arozena, A.; Adeli, K.; Aqostinis, P.; Aquirre-Ghiso, J.A.; Ait-Mohamed, O.; Ait-Si-Ali, S.; et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 2012, 8, 445–544. [Google Scholar]
  211. Fiorito, F.; Ciarcia, R.; Granato, G.E.; Marfe, G.; Iovane, V.; Florio, S.; de Martino, L.; Pagnini, U. 2,3,7,8-tetrachlorodibenzo-p-dioxin induced autophagy in a bovine kidney cell line. Toxicology 2011, 290, 258–270. [Google Scholar]
  212. Masini, M.; Bugliani, M.; Lupi, R.; del Guerra, S.; Boggi, U.; Filipponi, F.; Marselli, L.; Masiello, P.; Marchetti, P. Autophagy in human type 2 diabetes pancreatic beta cells. Diabetologia 2009, 52, 1083–1086. [Google Scholar]
  213. Ebato, C.; Uchida, T.; Arakawa, M.; Komatsu, M.; Ueno, T.; Komiya, K.; Azuma, K.; Hirose, T.; Tanaka, K.; Kominami, E.; et al. Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. Cell Metab 2008, 8, 325–332. [Google Scholar]
  214. Jung, H.S.; Chung, K.W.; Won Kim, J.; Kim, J.; Komatsu, M.; Tanaka, K.; Nguyen, Y.H.; Kang, T.M.; Yoon, K.H.; Kim, J.W.; et al. Loss of autophagy diminishes pancreatic beta cell mass and function with resultant hyperglycemia. Cell Metab 2008, 8, 318–324. [Google Scholar]
  215. Meijer, A.J.; Codogno, P. Autophagy: A sweet process in diabetes. Cell Metab 2008, 8, 275–276. [Google Scholar]
  216. Fujitani, Y.; Ueno, T.; Watada, H. Autophagy in health and disease. 4. The role of pancreatic beta-cell autophagy in health and diabetes. Am. J. Physiol. Cell Physiol 2010, 299, C1–C6. [Google Scholar]
  217. Levine, B.; Kroemer, G. Autophagy in the pathogenesis of disease. Cell 2008, 132, 27–42. [Google Scholar]
  218. Martino, L.; Masini, M.; Novelli, M.; Beffy, P.; Bugliani, M.; Marselli, L.; Masiello, P.; Marchetti, P.; de Tata, V. Palmitate activates autophagy in INS-1E β-cells and in isolated rat and human pancreatic islets. PLoS One 2012, 7, e36188. [Google Scholar]
Figure 1. Schematic representation of the proposed pathways involved in the acute dioxin toxicity in pancreatic beta cells. (TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin; AhR, aryl hydrocarbon receptor; ARNT, aryl hydrocarbon receptor nuclear translocator; MAPKs, mitogen-activated protein kinases).
Figure 1. Schematic representation of the proposed pathways involved in the acute dioxin toxicity in pancreatic beta cells. (TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin; AhR, aryl hydrocarbon receptor; ARNT, aryl hydrocarbon receptor nuclear translocator; MAPKs, mitogen-activated protein kinases).
Ijms 15 07787f1

Share and Cite

MDPI and ACS Style

De Tata, V. Association of Dioxin and Other Persistent Organic Pollutants (POPs) with Diabetes: Epidemiological Evidence and New Mechanisms of Beta Cell Dysfunction. Int. J. Mol. Sci. 2014, 15, 7787-7811. https://doi.org/10.3390/ijms15057787

AMA Style

De Tata V. Association of Dioxin and Other Persistent Organic Pollutants (POPs) with Diabetes: Epidemiological Evidence and New Mechanisms of Beta Cell Dysfunction. International Journal of Molecular Sciences. 2014; 15(5):7787-7811. https://doi.org/10.3390/ijms15057787

Chicago/Turabian Style

De Tata, Vincenzo. 2014. "Association of Dioxin and Other Persistent Organic Pollutants (POPs) with Diabetes: Epidemiological Evidence and New Mechanisms of Beta Cell Dysfunction" International Journal of Molecular Sciences 15, no. 5: 7787-7811. https://doi.org/10.3390/ijms15057787

Article Metrics

Back to TopTop