Obesity and metabolic comorbidities: environmental diseases?
Journal: 2013/August - Oxidative Medicine and Cellular Longevity
ISSN: 1942-0994
Abstract:
Obesity and metabolic comorbidities represent increasing health problems. Endocrine disrupting compounds (EDCs) are exogenous agents that change endocrine function and cause adverse health effects. Most EDCs are synthetic chemicals; some are natural food components as phytoestrogens. People are exposed to complex mixtures of chemicals throughout their lives. EDCs impact hormone-dependent metabolic systems and brain function. Laboratory and human studies provide compelling evidence that human chemical contamination can play a role in obesity epidemic. Chemical exposures may increase the risk of obesity by altering the differentiation of adipocytes. EDCs can alter methylation patterns and normal epigenetic programming in cells. Oxidative stress may be induced by many of these chemicals, and accumulating evidence indicates that it plays important roles in the etiology of chronic diseases. The individual sensitivity to chemicals is variable, depending on environment and ability to metabolize hazardous chemicals. A number of genes, especially those representing antioxidant and detoxification pathways, have potential application as biomarkers of risk assessment. The potential health effects of combined exposures make the risk assessment process more complex compared to the assessment of single chemicals. Techniques and methods need to be further developed to fill data gaps and increase the knowledge on harmful exposure combinations.
Relations:
Content
Citations
(15)
References
(106)
Conditions
(1)
Chemicals
(2)
Organisms
(1)
Processes
(1)
Anatomy
(1)
Affiliates
(1)
Similar articles
Articles by the same authors
Discussion board
Oxidative Medicine and Cellular Longevity. Dec/31/2012; 2013
Published online Mar/17/2013

Obesity and Metabolic Comorbidities: Environmental Diseases?

Abstract

1. Introduction

Obesity is an increasing health problem; more than half of the European population is overweight and up to 30% is obese and its prevalence worldwide doubled since 1980 (World Health Organization 2011) [1]. Similarly, increased body weights have also been reported in pets and laboratory animals over the past decades [2]. Obesity is a condition characterized by significant clinical implications, such as comorbidities and somatic fragility, which seriously affect independence, psychological wellbeing, and overall quality of life [3, 4]. Obesity is associated with type 2 diabetes mellitus (DM), dyslipidemia, cardiovascular disease, cancer, and obstructive sleep apnea [5, 6]. Medical treatments are often ineffective and bariatric surgery is the only available therapeutic modality associated with clinically significant and relatively sustained weight loss in subjects with morbid obesity [79].

Proinflammatory factors are increased in obesity and DM, and the prevalent metabolic state is defined by the term “glucolipotoxicity,” in which excess extracellular glucose and fatty acids exert various damaging effects. Obesity and DM-associated oxidative stress eventually lead to systemic inflammation and endothelial cell dysfunction, central to the development of cardiovascular diseases and metabolic syndrome [10, 11].

Excess caloric consumption and a sedentary lifestyle are the only recognized risk factors for obesity and DM but alone do not account for the current worldwide obesity epidemic. New hypotheses are emerging to explain the etiopathogenesis of these conditions, including environmental chemicals, stress, immunological alterations, micronutrient deficits, and gut microbiota [12, 13]. Genetic modifications could be involved in predisposition to obesity; however the human genome has not undergone significant modifications over the last years. On the contrary, the correlation between accumulation of synthetic chemicals and increase of obesity prevalence might not be a random event. This dramatic change in the environment has led to the hypothesis that some environmental pollutants act as Endocrine Disrupting Chemicals (EDCs), interfering with various aspects of metabolism and of energy balance [14].

2. Endocrine and Metabolic Disruption

2.1. EDCs

EDCs have been defined as exogenous substances that alter function(s) of the endocrine system and consequently cause adverse health effects in an intact organism, or its progeny, or (sub)populations. (International Programme for Chemical Safety—IPCS). EDCs can mimic the action of natural hormones or interfere with their production, release, metabolism, and elimination [15]. These substances may derive from natural, animal, human, or plant sources. Phytoestrogens are EDCs found naturally in certain plants, including foods like whole grains, leafy greens, beans, and garlic, and can mimic the action of estrogen, showing some beneficial effect on bone mineral density, insulin resistance, and cardiovascular risk factors in women after menopause [1620]. Another class of EDCs belongs to the group of heavy metals (i.e., cadmium, mercury, and arsenic). These metals may cause occupational or residential exposure [21]. Heavy metal toxicity can result in reduced mental and central nervous function and damage to blood composition, lungs, kidneys, liver, breast, and other vital organs [22, 23]. Allergies are not uncommon and repeated long-term exposure with some metals may even cause cancer [24, 25]. Major concerns, however, are currently focused on industrial products. In fact, most EDCs are synthetic chemicals designed for use in a variety of industries. Classes of these chemicals are solvents or lubricants and their byproducts (polychlorinated biphenyls (PCBs) and dioxins), plastics, plasticizers (bisphenol A (BPA)), phthalates; pesticides (methoxychlor, chlorpyrifos, DDT) fungicides (vinclozolin), herbicides (atrazine), and antibacterials (triclosan). There are several reports of pollution from PCBs and dioxins that show a direct causal relationship between a chemical and the manifestation of an endocrine, immunological, or metabolic dysfunction in humans and in wildlife [2629] but the more common event is the widespread persistent exposure to a broad mix of chemicals. Table 1 shows a brief summary of the principal incidents caused by dioxins contamination in the last fifty years.

2.2. Mechanisms of Action

Some EDCs were designed to have long half-lives and therefore are persistent contaminants, do not decay easily, may not be metabolized, or may be broken down into more toxic compounds [30]. Others, such as BPA, although not very persistent in the environment, are so widespread in their use that there is a prevalent human exposure [31]. Humans and wildlife are exposed daily to a variety of compounds, and it is thus likely that even if none reach an effective level, the combination or mixture of chemicals may become dangerous. These chemical mixtures enter the food chain and accumulate in animals up to humans. Exposure occurs also through drinking contaminated water, breathing contaminated air, or contacting contaminated surfaces. They may exert nontraditional dose-response curves, the so-called U-shaped or inverted U-shaped dose-response curve. As a consequence, any level of exposure may cause endocrine or metabolic abnormalities, particularly if the exposure occurs during a critical developmental period, and low doses may even be more potent than high doses [32, 33]. The age of exposure is important [34], since the environment to which a developing organism (fetal life, childhood) is exposed interacts with the individual's genes to determine the propensity to develop a disease later in life. The majority of environmental factors and toxicants do not alter DNA sequence or promote genetic mutations. Therefore, they may promote abnormal phenotypes or disease through modifications of factors that regulate gene expression such as DNA methylation and histone acetylation [3539].

EDCs can bind and activate multiple hormone Nuclear Receptors (NRs). Various EDCs share receptors, and thus additive or even synergistic effects may be observed [40]. Among NRs, Estrogen Receptors (ERs) regulate many aspects of metabolism, including glucose transport, glycolysis, mitochondrial activity, and lipid metabolism [41]. It is likely that ER activation modulates neural networks controlling food intake and adipose tissue [42, 43]. Male and female ER knock-out mutant mice show increased insulin resistance and impaired glucose tolerance [44]. Neonatal exposure to a low dose of the estrogenic drug diethylstilbestrol (DES) stimulated a subsequent increase in body weight and an increase in body fat in mice [45]. BPA, a breakdown product of coatings in food and beverage containers, may act as an ER agonist. In the US population, exposure is nearly ubiquitous, and BPA has been detected in fat, blood, and urine [46]. Short exposure to BPA provokes chronic hyperinsulinemia, with perturbations of glucose and insulin tolerance tests [47]. Furthermore, high- or low-dose exposure to BPA during gestation up to puberty leads to hyperlipidemia with increased body and adipose tissue weight in both sexes [48, 49]. BPA exposure has been shown to disrupt multiple metabolic mechanisms, suggesting that it may contribute to obesity in humans [5052]. Other studies have demonstrated associations between urinary BPA concentration and adult DM, cardiovascular diseases, obesity, and abnormalities in liver function [53, 54] A longitudinal study of apparently healthy adults showed an association between baseline urinary BPA concentration and later-life coronary artery disease [55].

EDCs may also modulate other hormone NRs, particularly thyroid hormone receptor (TR) and glucocorticoid receptor (GR). BPA acts as a TR antagonist in vitro, increases serum thyroxin, and alters RC3/neurogranin expression in the developing rat brain [56]. Brominated Flame Retardants (BFRs) also disrupt the TR pathway, and exposure of rats to Polybrominated Diphenyl Ethers (PBDEs) resulted in a significant increase in lipolysis and a significant decrease in glucose oxidation [57]. Organotins and PCBs can bind GR and alter 11beta-hydroxysteroid dehydrogenase type 2 activity [58].

The body is protected from the accumulation of toxic chemicals by the expression of drug-metabolizing enzymes and transporters. This adaptive response incorporates at least three NRs: pregnane X receptor (PXR), constitutive androstane receptor (CAR), and aryl hydrocarbon receptor (AhR), as well as xenobiotic metabolic and transporter systems. PXR and CAR are members of the NR super family of sensor receptors and contribute to fatty acid, lipid, and glucose metabolism, and CAR seems to be an anti-obesity NR that ameliorates DM and fatty liver accumulation [5961]. Endogenous ligands of PXR and CAR include some bile acid derivatives, pregnanes, androstane metabolites, and other metabolic products of steroids; exogenous compounds include herbal medicines pharmaceutical drugs and synthetic steroid hormones. A number of EDCs activate both PXR and CAR: nonylphenol, Di (2-Ethylhexyl) Phthalate (DEHP), Mono-(2-Ethylhexyl) Phthalate (MEHP), BPA, some PCBs perfluorooctane sulfonate (PFOS), perfluorooctanoic acid (PFOA), and the organochlorine methoxychlor [62]. AhR is a xenosensor that mediates the biological response to a wide spectrum of xenobiotics; in particular, AhR mediates the toxic effects of dioxins [63]. Endogenous molecules that bind AhR are lipoxin 4, leukotriene derivatives, biliverdin, and bilirubin. Xenobiotics that activate AhR include various dietary phytochemicals, some PCBs, and 2,3,7,8-Tetrachlorodibenzo-p-Dioxin (TCDD). The mechanisms through which AhR regulates energy metabolism are not clearly established; however, crosstalk with ER may be involved. In addition, AhR also indirectly affects adipogenesis through inhibition of Peroxisomal Proliferator-Activated Receptor (PPAR)γ expression [64].

2.3. Health Effects

All major endocrine organs are vulnerable to endocrine disruption, including the Hypothalamus-Pituitary-Adrenal axis, reproductive organs, the pancreas, and the thyroid gland [65, 66]. For example, it has been reported that EDCs, and estrogen mimicking agents among them, prolong the proliferation phase of immature Leydig cells, prior to their maturation [67], and can interfere with peritubular myoid cells [68], and it is known that estrogens are associated with Leydig cell tumorigenesis in mice [6971]. The declining level of androgen during aging, associated with an increasing level of estrogen, has been hypothesized to be important in the development of benign prostatic hyperplasia [7274], and oral exposure to low-dose BPA seems to aggravate testosterone-induced benign hyperplasia prostate in rats [75]. Several studies have suggested that estrogen exposure may increase the risk of prostate cancer [76], and various hormones such as androgens and gonadotropin-releasing hormone may play a role in prostate cancer cell growth [77, 78]. BPA and dibutyl phthalate (DBP) seem to be able to stimulate the growth of prostate cancer cells [79]. Thyroid carcinoma is the most common endocrine malignancy being about 3 times more common in women than in man [80]. Analogously, in differentiated thyroid cancer (DTC), EDCs with estrogen-like activity may be suspected to play a role in disease progression. Experimental evidence demonstrated that estrogen, but not testosterone, promotes DTC cell proliferation and that this effect could be attenuated by tamoxifen [81, 82].

In 2006, Bruce Blumberg developed the “obesogen hypothesis”, to explain the weight gain effects of certain chemicals. This hypothesis is supported by laboratory and animal research as well as epidemiological studies that shown that a variety of EDCs can influence adipogenesis and obesity [8386]. EDCs are also known to impact hormone-dependent metabolic systems and brain functionandcan be easily found in human blood and urine and epidemiological literature on associations between EDC exposure and body weightisincreasing [8789].

These substances target various endocrine axes and affect adipocyte physiology and more generally the regulation of energy homeostasis [90]. Several persistent organochlorine pesticides and fungicides have been implicated in obesity [91, 92]. Metabolic alterations like metabolic syndrome and type 2 DM are recognizedasobesity comorbid conditions, and EDCs exposure may be involved in their pathogenesis. Such contribution was revealed by comprehensive studies of large and well-characterized cohorts, such as the cohort used for the NHANES (National Health and Nutrition Examination Survey) project [93, 94].

Epidemiological studies show an association between dioxins exposure and type 2 DM [95]. High and low doses of dioxins affect genes linked with hepatic circadian rhythm, cholesterol biosynthesis, fatty acid synthesis, glucose metabolism, and adipocyte differentiation, in an AhR-dependent manner [96, 97]. Weight control and intermediate metabolism require a precise balance between energy input, storage, and consumption; several NRs are involved. The PPARs act as lipid sensors that act in different organs to adapt gene expression to a given metabolic status [98]. Plasticizers, surfactants, pesticides, and dioxins can modulate PPARs activity, and the phthalates are a group of well-characterized peroxisome proliferators. Mono-(2-Ethylhexyl) Phthalate (MEHP) is proadipogenic in a cell culture model, suggesting that it may act as a metabolic disruptor and may promote obesity in vivo [99, 100]. Kanayama et al. [101] showed that among 40 EDCs, organotins such as tributyltin (TBT) and bis (triphenyltin) oxide (TPTO) are activators of human PPARγ [102].

3. Oxidative Stress and Mitochondrial Dysfunction

An increase in oxidative stress-associated inflammation has been hypothesized to be a major mechanism in the pathogenesis of obesity-related diseases. Additionally, a rise in inflammatory cytokine levels might drive a further increase in oxidative stress, setting up a vicious cycle [104, 105]. When perturbed, the mitochondrial system alters the output of matter and energy and this may result in a pathological phenotype, such as that of obesity, dyslipidemia, metabolic syndrome, hypertension, and cancers. The failure of the skeletal muscle mitochondria to oxidize fat properly leads to ectopic lipid deposits. Cellular infiltration by excess triglycerides can impair cellular function and can also lead to oxidative stress through increased ceramide formation, increased lipid peroxidation, inflammatory cytokine production, and excess Reactive Oxygen Species (ROS) formation [106, 107]. When ROS production is increased, the disturbed balance results in a prooxidative condition. This oxidative stress can then damage various cellular structures and triggers an inflammatory response associated with adiposity, insulin resistance, and metabolic syndrome, suggesting that oxidative stress could be an early event in the pathology of these chronic diseases. [108, 109]. Recently, much evidence has emerged showing that environmental toxins, including Persistent Organic Pollutants (POPs), can affect mitochondrial function and subsequent insulin resistance. In this regard, many herbicides, insecticides, rodenticides, industrial products, and industrial toxic wastes might affect mitochondrial function and cause pro-oxidative conditions [110112].

4. Exposure Monitoring

Various molecules, involved in antioxidant and detoxification pathways, have potential application as biomarkers in biomonitoring and risk assessment.

The National Academy of Science, in 1987, defined a biomarker as “a xenobiotically induced variation in cellular or biochemical components or processes, structures, or function that is measurable in a biological system (body fluids, cells, or tissues)” [113]. Biomarker responses in fish are routinely used to assess exposure of anthropogenic chemicals in the aquatic environment. The use of biomarkers could complement the current methods used to determine the presence of environmental pollutants and might also help to predict human health risks [114]. Among the various types of biomarkers in ecotoxicological studies are the following: cytochrome P450 activity (an indicator of the exposure and effect of organic contaminants, such as polycyclic aromatic hydrocarbons (PAHs), PCBs, and pesticides), the inhibition of Acetyl cholinesterase (AChE) activity (a biomarker of the exposure and effect of organophosphate (OP) and carbamate (CAR), metallothionein synthesis in hepatic and other tissues (exposure to the metals Zn, Cu, Cd, Hg, and Fe and some pesticides), antioxidant enzymes such as superoxide dismutase, catalase, and glutathione transferase (exposure to ROS, free radicals, and pollutants causing oxidative stress and lipid peroxidation, such as pesticides and metals), and vitellogenin induction (estrogenic substances) [115121]. Recently, redox markers have been used to biologically define Multiple Chemical Sensitivity (MCS) [122]. The development and use of biomarkers in ecotoxicology for providing sensitive early warning signals of incipient ecological damage is motivated by the inherent instabilities of many EDCs and the chemical specificity of some biomarkers on underlying mechanisms of toxic action. However, little is known about how cocktail effects affect these biomarker responses, and chemical safety-levels are traditionally based on experiences from lab studies with single chemicals, which are unfortunate as a chemical can be more toxic when it is mixed with other chemicals, because of the cocktail effect, for example, if there is a risk for increased bioavailability of certain pollutants that can result in harmful bioaccumulation or if there is an increased risk for accumulation of toxic metabolites or if there is an increased risk for depletion of endogenous hormones. The possible involvement of receptor crosstalks, inhibition, or activation on key biotransformation enzyme and transporter proteins such and multidrug resistance-associated proteins needs to be addressed for estimation of possible adverse pharmacokinetic interactions [123].

5. Concluding Remarks

EDCs clearly contribute to diverse male and female human health problems such as decreased male sperm counts, increased incidence of hypospadias and cryptorchidism, altered male: female birth sex ratios, decreased fertility, and increased incidence of breast and testicular cancers and may be responsible for neurodevelopmental deficits in children. Recently, human exposure to EDCs has been associated with the development of some of the main diseases of the industrialized world, particularly metabolic disorders like obesity, diabetes, and metabolic syndrome. POPs such as organochlorine pesticides, dioxins, and polyfluoroalkyl compounds and no persistent pollutants such as BPA and several phthalates are endowed with metabolic disruption activity. The European Union has sponsored several international research project to investigate various obesity-related effects from EDCs exposure (Table 2), regarding the diverse types of chemical compounds involved, the influence of climate changes on contaminant spreading, the age of exposition (developmental basis of adult diseases), food safety, and human bio monitoring. A greatest challenge in environmental toxicology is to understand effects of mixture toxicity (cocktail effects) in humans and in wildlife. Considering that metabolic perturbations are only one small aspect of the EDCs-related problems to be solved and that we know only the tip of the iceberg, new integrative approaches are required to understand the complexity of the cocktail effect and its consequences when exposure occurs at various life stages [124]. Procedures for risk assessment of chemical mixtures, combined, and cumulative exposures are under development, but the scientific database needs considerable expansion [114]. In particular, there is a lack of knowledge on how to monitor effects of complex exposures. As described here, solid evidence shows that endocrine disrupters can interact and even produce synergistic effects. They may act during sensitive time windows and biomonitoring their effects in epidemiological studies is a challenging task. The potential health effects of combined exposures make the risk assessment process more complex compared to the assessment of single chemicals. Techniques and methods need to be further developed to fill data gaps and increase the knowledge on harmful exposure combinations.

Table 1
Examples of dioxin contamination incidents.
YearCountryType of accidentCause
1960–1975VietnamContamination by a defoliant, agent orangeContaminated agent orange
1976Italy (Seveso)Contamination by a cloud of toxic chemicals of an area of 15 square kilometersAccident at a chemical factory
1998GermanyContamination of milkContaminated citrus pulp pellet from Brazil
1999BelgiumContamination of poultry and eggsAnimal feed contaminated with illegally disposed PCB-based waste industrial oil
2004The NetherlandsContamination of milkContaminated clay in animal feed
2007EuropeContamination of guar gum (food additive)Guar gum from India contaminated by pentachlorophenol that contains dioxins as further contamination
2008IrelandContamination of pork meat and productsContaminated animal feed
Table 2

Examples of ongoing European research projects on endocrine disrupters [103].

Project acronym and durationProject titleResearch teamFocus
CONFFIDENCE
(2008–2012)
Contaminants in food and feed: inexpensive detection for control of exposureParticipating laboratories: 17 (NL, CZ, ES, DE, DK, BE, UK, IT, FI, CH)The main aim is to further improve food safety in Europe by the development of faster and more cost-efficient methods for the detection of a wide range of chemical contaminants (persistent organic compounds, perfluorinated compounds, and heavy metals) in different food and feed commodities
ARCRISK (2009–2013)Arctic health risks: Impacts on health in the Arctic and Europe owing to climate-induced changes in contaminant cyclingParticipating laboratories: 22 (NO, SE, DK, FI, DE, UK, ES, SI, CH, CZ, RU, CA)The main aim will be to study the influence of climate change on contaminant spreading and transfer and the resultant risk to human populations in the Arctic and other areas of Europe
COPHES (2009–2012)European coordination action on human biomonitoringParticipating laboratories: 35 (BE, DE, ES, UK, FR, DK, IT, EE, SI, NL, AT, RO, LT, HR, EL, CY, PT, SE, NO, HU, CH, SK, PL, CZ, IE, FI, LU)The main goal is to develop a coherent approach to human biomonitoring in Europe, addressing the aims of Action 3 of the European Environment and Health Action Plan.
ENFIRO (2009–2012)Life cycle assessment of environment-compatible flame retardants (prototypical case study)Participating laboratories: 12 (NL, UK, SE, DE, IT)ENFIRO will offer a prototypical case study on substitution options for BFRs resulting in a comprehensive dataset on viability of production and application, environmental safety, and a complete life cycle assessment
OBELIX (2009–2013)Obesogenic endocrine disrupting chemicals: linking prenatal exposure to the development of obesity later in lifeParticipating laboratories: 7 (NL, BE, NO, FR, SK)Examination of the hypothesis that prenatal exposure to endocrine disrupting compounds in food plays a role in the development of obesity later in life

References

  • 2. KlimentidisYCBeasleyTMLinHYCanaries in the coal mine: a cross-species analysis of the plurality of obesity epidemicsProceedings of the Royal Society B2011278171216261632[PubMed][Google Scholar]
  • 3. DoniniLMCuzzolaroMSperaGObesity and eating disorders. Indications for the different levels of care. An Italian expert consensus documentEating and Weight Disorders2010151-2131[PubMed][Google Scholar]
  • 4. DoniniLMBrunaniASirtoriAAssessing disability in morbidly obese individuals: the Italian society of obesity test for obesity-related disabilitiesDisability and Rehabilitation20113325-2625092518[PubMed][Google Scholar]
  • 5. KassiEPervanidouPKaltsasGChrousosGMetabolic syndrome: definitions and controversiesBMC Medicine20119article 48[PubMed][Google Scholar]
  • 6. LubranoCSaponaraMBarbaroGRelationships between body fat distribution, epicardial fat and obstructive sleep apnea in obese patients with and without metabolic syndromePLoS ONE7201210[PubMed][Google Scholar]
  • 7. DeMariaEJBariatric surgery for morbid obesityThe New England Journal of Medicine20073562121762183[PubMed][Google Scholar]
  • 8. LubranoCCornoldiAPiliMReduction of risk factors for cardiovascular diseases in morbid-obese patients following biliary-intestinal bypass: 3 years’ follow-upInternational Journal of Obesity2004281216001606[PubMed][Google Scholar]
  • 9. LubranoCMarianiSBadialiMMetabolic or bariatric surgery long-term effects of malabsorptive versus restrictive bariatric techniques on body composition and cardiometabolic risk factorsInternational Journal of Obesity201034914041414[PubMed][Google Scholar]
  • 10. Bondia-PonsIRyanLMartinezJAOxidative stress and inflammation interactions in human obesityJournal of Physiology and Biochemistry2012684701711[PubMed][Google Scholar]
  • 11. RossiFBertoneCMontanileFHDL cholesterol is a strong determinant of endothelial progenitor cells in hypercholesterolemic subjectsMicrovascular Research2010802274279[PubMed][Google Scholar]
  • 12. ThayerKAHeindelJJBucherJRGalloMARole of environmental chemicals in diabetes and obesity: a national toxicology program workshop reviewEnvironmental Health Perspectives20121206779789[PubMed][Google Scholar]
  • 13. SalvatoriLCaporuscioFDe GirolamoLMorphological and functional features of human omental and subcutaneous adipose-derived stem cells isolated from obese patientsEndocrine Reviews, Supplement 12010313p. S1351 P2-468[Google Scholar]
  • 14. Baillie-HamiltonPFChemical toxins: a hypothesis to explain the global obesity epidemicJournal of Alternative and Complementary Medicine200282185192[PubMed][Google Scholar]
  • 15. ColbornTvom SaalFSSotoAMDevelopmental effects of endocrine-disrupting chemicals in wildlife and humansEnvironmental Health Perspectives19931015378384[PubMed][Google Scholar]
  • 16. MariniHMinutoliLPolitoFEffects of the phytoestrogen genistein on bone metabolism in osteopenic postmenopausal women: a randomized trialAnnals of Internal Medicine200714612839847[PubMed][Google Scholar]
  • 17. AtteritanoMMariniHMinutoliLEffects of the phytoestrogen genistein on some predictors of cardiovascular risk in osteopenic, postmenopausal women: a two-year randomized, double-blind, placebo-controlled studyJournal of Clinical Endocrinology and Metabolism200792830683075[PubMed][Google Scholar]
  • 18. MariniHBittoAAltavillaDBreast safety and efficacy of genistein aglycone for postmenopausal bone loss: a follow-up studyJournal of Clinical Endocrinology and Metabolism2008931247874796[PubMed][Google Scholar]
  • 19. MariniHMinutoliLPolitoFOPG and sRANKL serum concentrations in osteopenic, postmenopausal women after 2-year genistein administrationJournal of Bone and Mineral Research2008235715720[PubMed][Google Scholar]
  • 20. MariniHBittoAAltavillaDEfficacy of genistein aglycone on some cardiovascular risk factors and homocysteine levels: a follow-up studyNutrition, Metabolism and Cardiovascular Diseases2010205332340[Google Scholar]
  • 21. CastelliMRossiBCorsettiFCaroliSFerreriCCadmium and Pb in blood of Italian subjects: preliminary report of current trends11. Italian-Hungarian Symposium on Spectrochemistry. New Challenges in Human Health Protection: Anthropic and Remote Areas. Venice (Italy). October 19-24, 2003. Abstract Book2003Rome, ItalyIstituto Superiore di Sanità (ISTISAN Congressi 03/C3)p. 142[Google Scholar]
  • 22. TchounwouPBYedjouCGPatlollaAKSuttonDJHeavy metal toxicity and the environmentMolecular, Clinical and Environmental Toxicology Experientia Supplementum2012101133164[Google Scholar]
  • 23. BramaMGnessiLBascianiSCadmium induces mitogenic signaling in breast cancer cell by an ERα-dependent mechanismMolecular and Cellular Endocrinology20072641-2102108[PubMed][Google Scholar]
  • 24. 2004, http://ec.europa.eu/food/food/chemicalsafety/contaminants/scoop_3-2-11_heavy_metals_report_en.pdf
  • 25. CastelliMRossiBCorsettiFLevels of cadmium and lead in blood: an application of validated methods in a group of patients with endocrine/metabolic disorders from the Rome areaMicrochemical Journal2005791-2349355[PubMed][Google Scholar]
  • 26. WrenCDCause-effect linkages between chemicals and populations of mink (Mustela vision) and otter (Lutra canadensis) in the Great Lakes basinJournal of Toxicology and Environmental Health1991334549585[PubMed][Google Scholar]
  • 27. HansenPDvon WesternhagenHRosenthalHChlorinated hydrocarbons and hatching success in Baltic herring spring spawnersMarine Environmental Research19851515976[PubMed][Google Scholar]
  • 28. LeatherlandJFColbornTClementCEndocrine and reproductive function in Great Lakes salmonChemically Induced Alterations in Sexual and Functional Development: The Human-Wildlife Connection1992Princeton, NJ, USAPrinceton Scientific Publishing129146[Google Scholar]
  • 29. PesatoriACConsonniDTironiAZocchettiCFiniABertazziPACancer in a young population in a dioxin-contaminated areaInternational Journal of Epidemiology199322610101013[PubMed][Google Scholar]
  • 30. CalafatAMNeedhamLLGoreACHuman exposures and body burdens of endocrine-disrupting chemicalsEndocrine-Disrupting Chemicals: From Basic Research to Clinical Practice2007Totowa, NJ, USAHumana Press253268[Google Scholar]
  • 31. Casals-CasasCFeigeJNDesvergneBInterference of pollutants with PPARs: endocrine disruption meets metabolismInternational Journal of Obesity200832supplement 6S53S61[PubMed][Google Scholar]
  • 32. SheehanDMWillinghamEGaylorDBergeronJMCrewsDNo threshold dose for estradiol-induced sex reversal of turtle embryos: how little is too much?Environmental Health Perspectives19991072155159[PubMed][Google Scholar]
  • 33. vom SaalFSAkingbemiBTBelcherSMChapel Hill bisphenol A expert panel consensus statement: integration of mechanisms, effects in animals and potential to impact human health at current levels of exposureReproductive Toxicology2007242131138[PubMed][Google Scholar]
  • 34. BarkerDJPThe developmental origins of adult diseaseEuropean Journal of Epidemiology200318733736[PubMed][Google Scholar]
  • 35. SkinnerMKManikkamMGuerrero-BosagnaCEpigenetic transgenerational actions of environmental factors in disease etiologyTrends in Endocrinology and Metabolism2010214214222[PubMed][Google Scholar]
  • 36. HansonMAGluckmanPDDevelopmental origins of health and disease: new insightsBasic and Clinical Pharmacology and Toxicology200810229093[PubMed][Google Scholar]
  • 37. MorganDKWhitelawEThe role of epigenetics in mediating environmental effects on phenotypeNestle Nutrition Workshop Series: Pediatric Program200963109119[PubMed][Google Scholar]
  • 38. PetrangeliELubranoCRavennaLGene methylation of oestrogen and epidermal growth factor receptors in neoplastic and perineoplastic breast tissuesThe British Journal of Cancer1995724973975[PubMed][Google Scholar]
  • 39. PetrangeliELubranoCOrtolaniFEstrogen receptors: new perspectives in breast cancer managementJournal of Steroid Biochemistry and Molecular Biology1994494–6327331[PubMed][Google Scholar]
  • 40. Diamanti-KandarakisEBourguignonJPGiudiceLCEndocrine-disrupting chemicals: an endocrine society scientific statementEndocrine Reviews2009304293342[PubMed][Google Scholar]
  • 41. ChenJQBrownTRRussoJRegulation of energy metabolism pathways by estrogens and estrogenic chemicals and potential implications in obesity associated with increased exposure to endocrine disruptorsBiochimica et Biophysica Acta20091793711281143[PubMed][Google Scholar]
  • 42. BrownLMGentLDavisKCleggDJMetabolic impact of sex hormones on obesityBrain Research201013507785[PubMed][Google Scholar]
  • 43. CookePSNaazARole of estrogens in adipocyte development and functionExperimental Biology and Medicine20042291111271135[PubMed][Google Scholar]
  • 44. HeinePATaylorJAIwamotoGALubahnDBCookePSIncreased adipose tissue in male and female estrogen receptor-α knockout miceProceedings of the National Academy of Sciences of the United States of America200097231272912734[PubMed][Google Scholar]
  • 45. NewboldRRPrenatal exposure to diethylstilbestrol (DES)Fertility and Sterility2008892e55e56[PubMed][Google Scholar]
  • 46. SchecterAMalikNHaffnerDBisphenol A (BPA) in U.S. foodEnvironmental Science and Technology2010442494259430[PubMed][Google Scholar]
  • 47. Alonso-MagdalenaPRoperoABCarreraMPPancreatic insulin content regulation by the Estrogen receptor ERαPLoS ONE200834[PubMed][Google Scholar]
  • 48. MiyawakiJSakayamaKKatoHYamamotoHMasunoHPerinatal and postnatal exposure to bisphenol A increase adipose tissue mass and serum cholesterol level in miceJournal of Atherosclerosis and Thrombosis2007145245252[PubMed][Google Scholar]
  • 49. HeindelJJvom SaalFSRole of nutrition and environmental endocrine disrupting chemicals during the perinatal period on the aetiology of obesityMolecular and Cellular Endocrinology20093041-29096[PubMed][Google Scholar]
  • 50. SakuraiKKawazumaMAdachiTBisphenol A affects glucose transport in mouse 3T3-F442A adipocytesThe British Journal of Pharmacology20041412209214[PubMed][Google Scholar]
  • 51. vom SaalFSNagelSCCoeBLAngleBMTaylorJAThe estrogenic endocrine disrupting chemical bisphenol A, (BPA) and obesityMolecular and Cellular Endocrinology20123541-27484[PubMed][Google Scholar]
  • 52. WilsonNKChuangJCMorganMKLordoRASheldonLSAn observational study of the potential exposures of preschool children to pentachlorophenol, bisphenol-A, and nonylphenol at home and daycareEnvironmental Research20071031920[PubMed][Google Scholar]
  • 53. CarwileJLMichelsKBUrinary bisphenol A and obesity: NHANES 2003–2006Environmental Research20111116825830[PubMed][Google Scholar]
  • 54. LangIAGallowayTSScarlettAAssociation of urinary bisphenol A concentration with medical disorders and laboratory abnormalities in adultsThe Journal of the American Medical Association20083001113031310[PubMed][Google Scholar]
  • 55. MelzerDOsborneNJHenleyWEUrinary bisphenol A concentration and risk of future coronary artery disease in apparently healthy men and womenCirculation20121251214821490[PubMed][Google Scholar]
  • 56. ZoellerRTBansalRParrisCBisphenol-A, an environmental contaminant that acts as a thyroid hormone receptor antagonist in vitro, increases serum thyroxine, and alters RC3/neurogranin expression in the developing rat brainEndocrinology20051462607612[PubMed][Google Scholar]
  • 57. LeglerJNew insights into the endocrine disrupting effects of brominated flame retardantsChemosphere2008732216222[PubMed][Google Scholar]
  • 58. SargisRMJohnsonDNChoudhuryRABradyMJEnvironmental endocrine disruptors promote adipogenesis in the 3T3-L1 cell line through glucocorticoid receptor activationObesity201018712831288[PubMed][Google Scholar]
  • 59. NakamuraKMooreRNegishiMSueyoshiTNuclear pregnane X receptor cross-talk with FoxA2 to mediate drug-induced regulation of lipid metabolism in fasting mouse liverThe Journal of Biological Chemistry20072821397689776[PubMed][Google Scholar]
  • 60. GaoJHeJZhaiYWadaTXieWThe constitutive androstane receptor is an antiobesity nuclear receptor that improves insulin sensitivityThe Journal of Biological Chemistry2009284382598425992[PubMed][Google Scholar]
  • 61. MoreauAVilaremMJMaurelPPascussiJMXenoreceptors CAR and PXR activation and consequences on lipid metabolism, glucose homeostasis, and inflammatory responseMolecular Pharmaceutics2008513541[PubMed][Google Scholar]
  • 62. RenHAleksunesLMWoodCCharacterization of peroxisome proliferator-activated receptor α-independent effects of PPARα activators in the rodent liver: di-(2-ethylhexyl) phthalate also activates the constitutive-activated receptorToxicological Sciences200911314559[PubMed][Google Scholar]
  • 63. BockKWKöhleCAh receptor: dioxin-mediated toxic responses as hints to deregulated physiologic functionsBiochemical Pharmacology2006724393404[PubMed][Google Scholar]
  • 64. CimafrancaMAHanlonPRJefcoateCRTCDD administration after the pro-adipogenic differentiation stimulus inhibits PPARγ through a MEK-dependent process but less effectively suppresses adipogenesisToxicology and Applied Pharmacology20041961156168[PubMed][Google Scholar]
  • 66. SilvestroniLRossiFMagnantiMLubranoCSantiemmaVPalleschiSA novel aspect of lindane testicular toxicity: in vitro effects on peritubular myoid cellsReproductive Toxicology1999136431441[PubMed][Google Scholar]
  • 67. HengKAnand-IvellRTeerdsKIvellRThe endocrine disruptors dibutyl phthalate (DBP) and diethylstilbestrol (DES) influence Leydig cell regeneration following ethane dimethane sulphonate treatment of adult male ratsInternational Journal of Andrology2012353353363[PubMed][Google Scholar]
  • 68. SilvestroniLRossiFMagnantiMLubranoCSantiemmaVPalleschiSA novel aspect of lindane testicular toxicity: in vitro effects on peritubular myoid cellsReproductive Toxicology1999136431441[PubMed][Google Scholar]
  • 69. BascianiSBramaMMarianiSImatinib mesylate inhibits Leydig cell tumor growth: evidence for in vitro and in vivo activityCancer Research200565518971903[PubMed][Google Scholar]
  • 70. BascianiSMarianiSSperaGGnessiLRole of platelet-derived growth factors in the testisEndocrine Reviews2010316916939[PubMed][Google Scholar]
  • 71. MarianiSBascianiSArizziMSperaGGnessiLPDGF and the testisTrends in Endocrinology and Metabolism20021311117[PubMed][Google Scholar]
  • 72. LubranoCSciarraFSperaGImmunoreactive EGF in human benign prostatic hyperplasia: relationships with androgen and estrogen receptorsJournal of Steroid Biochemistry and Molecular Biology1992413–8683687[PubMed][Google Scholar]
  • 73. PetrangeliESciarraFDi SilverioFEffects of two different medical treatments on dihydrotestosterone content and androgen receptors in human benign prostatic hyperplasiaJournal of Steroid Biochemistry1988301–6395399[PubMed][Google Scholar]
  • 74. LubranoCToscanoVPetrangeliERelationship between epidermal growth factor and its receptor in human benign prostatic hyperplasiaJournal of Steroid Biochemistry and Molecular Biology1993464463468[PubMed][Google Scholar]
  • 75. WuJHJiangXRLiuGMLiuXYHeGLSunZYOral exposure to low-dose bisphenol A aggravates testosterone-induced benign hyperplasia prostate in ratsToxicology and Industrial Health2011279810819[PubMed][Google Scholar]
  • 76. MargelDFleshnerNEOral contraceptive use is associated with prostate cancer: an ecological studyThe British Medical Journal201112[PubMed][Google Scholar]
  • 77. RavennaLSalvatoriLMorroneSEffects of triptorelin, a gonadotropin-releasing hormone agonist, on the human prostatic cell lines PC3 and LNCaPJournal of Andrology2000214549557[PubMed][Google Scholar]
  • 78. RavennaLLubranoCDi SilverioFAndrogenic and antiandrogenic control on epidermal growth factor, epidermal growth factor receptor, and androgen receptor expression in human prostate cancer cell line LNCaPProstate1995266290298[PubMed][Google Scholar]
  • 79. KimKLeeHChoiKBisphenol A and phthalate stimulated the growth of human prostate cancer cells and altered downstream target genes of ras signaling pathwayEndocrine Abstracts201229article P751[Google Scholar]
  • 80. TrimboliPUlisseSGrazianoFMTrend in thyroid carcinoma size, age at diagnosis, and histology in a retrospective study of 500 cases diagnosed over 20 yearsThyroid2006161111511155[PubMed][Google Scholar]
  • 81. GoldenSHBrownACauleyJAHealth disparities in endocrine disorders: biological, clinical, and nonclinical factors—an endocrine society statementJournal of Clinical Endocrinology and Metabolism2012979E1579E1639[PubMed][Google Scholar]
  • 82. LeeMLChenGGVlantisACTseGMKLeungBCHVan HasseltCAInduction of thyroid papillary carcinoma cell proliferation by estrogen is associated with an altered expression of Bcl-xLCancer Journal2005112113121[PubMed][Google Scholar]
  • 83. GrünFBlumbergBEnvironmental obesogens: organotins and endocrine disruption via nuclear receptor signalingEndocrinology20061476S50S55[PubMed][Google Scholar]
  • 84. Turrio BaldassarriLLa RoccaCIacovellaNDetermination of polycyclic aromatic hydrocarbons and polychlorobiphenyls in whole human blood by SFE-GC-MSOrganohalogen Compounds199940285288[Google Scholar]
  • 85. LubranoCObesità e inquinamentoSicurezza Sanitaria2008983845[Google Scholar]
  • 86. LubranoCBottaccioliFInterferenti endocrini e obesitàGeni e comportamenti. Scienza e arte della vita2009Milan, ItalyRed Edizioni219226[Google Scholar]
  • 87. ECTowards the establishment of a priority list of substances for further evaluation of their role in endocrine disruption—preparation of a candidate list of substances as a basis for priority settingFinal Report by BKH Consulting Engineers, Delft, The Netherlands TNO Nutrition and Food Research, Zeist, The Netherlands, for the European Commission, EC DG ENV M0355008/1786Q/10/11/00, 2000
  • 88. Turrio-BaldassarriLLa RoccaCIacovellaNDetermination of polycyclic aromatic hydrocarbons and polychlorobiphenyls in whole human blood by SFE EGC-MSOrganohalogen Compounds199940285288[Google Scholar]
  • 89. HatchEENelsonJWStahlhutRWWebsterTFAssociation of endocrine disruptors and obesity: perspectives from epidemiological studiesInternational Journal of Andrology2010332324332[PubMed][Google Scholar]
  • 91. KarmausWOsuchJREneliIMaternal levels of dichlorodiphenyl-dichloroethylene (DDE) may increase weight and body mass index in adult female offspringOccupational and Environmental Medicine2009663143149[PubMed][Google Scholar]
  • 92. SminkARibas-FitoNGarciaRExposure to hexachlorobenzene during pregnancy increases the risk of overweight in children aged 6 yearsActa Paediatrica, International Journal of Paediatrics2008971014651469[PubMed][Google Scholar]
  • 93. Casals-CasasCDesvergneBEndocrine disruptors: from endocrine to metabolic disruptionAnnual Review of Physiology201173135162[PubMed][Google Scholar]
  • 94. ElobeidMAPadillaMABrockDWRudenDMAllisonDBEndocrine disruptors and obesity: an examination of selected persistent organic pollutants in the NHANES 1999–2002 dataInternational Journal of Environmental Research and Public Health20107729883005[PubMed][Google Scholar]
  • 95. FierensSMairesseHHeilierJFDioxin/polychlorinated biphenyl body burden, diabetes and endometriosis: findings in a population-based study in BelgiumBiomarkers200386529534[PubMed][Google Scholar]
  • 96. SatoSShirakawaHTomitaSLow-dose dioxins alter gene expression related to cholesterol biosynthesis, lipogenesis, and glucose metabolism through the aryl hydrocarbon receptor-mediated pathway in mouse liverToxicology and Applied Pharmacology200822911019[PubMed][Google Scholar]
  • 97. MatthewsJGustafssonJAEstrogen receptor and aryl hydrocarbon receptor signaling pathwaysNuclear Receptor Signaling20064article e016[Google Scholar]
  • 98. AuwerxJPPARγ, the ultimate thrifty geneDiabetologia199942910331049[PubMed][Google Scholar]
  • 99. FeigeJNGelmanLRossiDActivation of PPARα and PPARγ by environmental phthalate monoestersToxicological Sciences200774297308[Google Scholar]
  • 100. ZoeteVGrosdidierAMichielinOPeroxisome proliferator-activated receptor structures: ligand specificity, molecular switch and interactions with regulatorsBiochimica et Biophysica Acta200717718915925[PubMed][Google Scholar]
  • 101. KanayamaTKobayashiNMamiyaSNakanishiTNishikawaJIOrganotin compounds promote adipocyte differentiation as agonists of the peroxisome proliferator-activated receptor γ/retinoid X receptor pathwayMolecular Pharmacology2005673766774[PubMed][Google Scholar]
  • 102. GrunFWatanabeHZamanianZEndocrine-disrupting organotin compounds are potent inducers of adipogenesis in vertebratesMolecular Endocrinology200520921412155[Google Scholar]
  • 104. NunnAVWBellJDGuyGWLifestyle-induced metabolic inflexibility and accelerated ageing syndrome: insulin resistance, friend or foe?Nutrition and Metabolism20096article 16[PubMed][Google Scholar]
  • 105. HotamisligilGSInflammation and metabolic disordersNature20064447121860867[PubMed][Google Scholar]
  • 106. LowellBBShulmanGIMitochondrial dysfunction and type 2 diabetesScience20053075708384387[PubMed][Google Scholar]
  • 107. RoggeMMThe role of impaired mitochondrial lipid oxidation in obesityBiological Research For Nursing2009104356373[PubMed][Google Scholar]
  • 108. MeigsJBLarsonMGFoxCSKeaneyJFVasanRSBenjaminEJAssociation of oxidative stress, insulin resistance, and diabetes risk phenotypes: the framingham offspring studyDiabetes Care2007301025292535[PubMed][Google Scholar]
  • 109. Krzystek-KorpackaMPatrynEBoehmDBerdowskaIZielinskiBNoczynskaAAdvanced oxidation protein products (AOPPs) in juvenile overweight and obesity prior to and following weight reductionClinical Biochemistry20084112943949[PubMed][Google Scholar]
  • 110. LimSChoYMParkKSLeeHKPersistent organic pollutants, mitochondrial dysfunction, and metabolic syndromeAnnals of the New York Academy of Sciences20101201166176[PubMed][Google Scholar]
  • 111. WuMXuHShenYQiuWYangMOxidative stress in zebrafish embryos induced by short-term exposure to bisphenol A, nonylphenol, and their mixtureEnvironmental Toxicology and Chemistry2011301023352341[PubMed][Google Scholar]
  • 112. Yang YjHongYCOhSYBisphenol A exposure is associated with oxidative stress and inflammation in postmenopausal womenEnvironmental Research20091096797801[PubMed][Google Scholar]
  • 113. HuggettRJStegemanJJPageDSParkerKRWoodinBBrownJSBiomarkers in fish from Prince William Sound and the Gulf of Alaska: 1999-2000Environmental Science and Technology2003371840434051[PubMed][Google Scholar]
  • 114. SilinsIHögbergJCombined toxic exposures and human health: biomarkers of exposure and effectInternational Journal of Environmental Research and Public Health201183629647[PubMed][Google Scholar]
  • 115. ValavanidisAVlahogianniTDassenakisMScoullosMMolecular biomarkers of oxidative stress in aquatic organisms in relation to toxic environmental pollutantsEcotoxicology and Environmental Safety2006642178189[PubMed][Google Scholar]
  • 116. FarahatFMEllisonCABonnerMRBiomarkers of chlorpyrifos exposure and effect in Egyptian cotton field workersEnvironmental Health Perspectives20111196801806[PubMed][Google Scholar]
  • 117. AlbersJWGarabrantDHBerentSRichardsonRJParaoxonase status and plasma butyrylcholinesterase activity in chlorpyrifos manufacturing workersJournal of Exposure Science and Environmental Epidemiology20102017989[PubMed][Google Scholar]
  • 118. PanemangaloreMDowlaHAByersMEOccupational exposure to agricultural chemicals: effect on the activities of some enzymes in the blood of farm workersInternational Archives of Occupational and Environmental Health19997228488[PubMed][Google Scholar]
  • 119. LivingstoneDRContaminant-stimulated reactive oxygen species production and oxidative damage in aquatic organismsMarine Pollution Bulletin2001428656666[PubMed][Google Scholar]
  • 120. García-ClosasMMalatsNSilvermanDNAT2 slow acetylation, GSTM1 null genotype, and risk of bladder cancer: results from the Spanish bladder cancer study and meta-analysesThe Lancet20053669486649659[PubMed][Google Scholar]
  • 121. AbhishekSKaurNKaurSLataMSharmaJKSharmaAAssociation of GSTM1 and GSTT1 gene deletions with susceptibility to DNA damage in the pesticide-exposed workers of PunjabRejuvenation Research2010132-3281284[PubMed][Google Scholar]
  • 122. de LucaCScordoMGCesareoEBiological definition of multiple chemical sensitivity from redox state and cytokine profiling and not from polymorphisms of xenobiotic-metabolizing enzymesToxicology and Applied Pharmacology20102483285292[PubMed][Google Scholar]
  • 123. CelanderMCCocktail effects on biomarker responses in fishAquatic Toxicology20111053-4, supplement7277[PubMed][Google Scholar]
  • 124. EveillardALasserreFde TayracMIdentification of potential mechanisms of toxicity after di-(2-ethylhexyl)-phthalate (DEHP) adult exposure in the liver using a systems biology approachToxicology and Applied Pharmacology20092363282292[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.