Distruptores Metabolicos (Articulo Referencial Para Examen)

download Distruptores Metabolicos (Articulo Referencial Para Examen)

of 31

Transcript of Distruptores Metabolicos (Articulo Referencial Para Examen)

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    1/31

    Endocrine Disruptors:From Endocrine to Metabolic DisruptionCristina Casals-Casas and Beatrice DesvergneCenter for Integrative Genomics, Faculty of Biology and Medicine, University of LauLausanne, 1015 Switzerland; email: [email protected]

    Annu. Rev. Physiol. 2011. 73:13562

    First published online as a Review in Advance onNovember 5, 2010

    The Annual Review of Physiologyis online atphysiol.annualreviews.org

    This articles doi:10.1146/annurev-physiol-012110-142200

    Copyright c 2011 by Annual Reviews. All rights reserved

    0066-4278/11/0315-0135$20.00

    Keywordspersistent organic pollutants, phthalates, bisphenol A, obesity,diabetes, nuclear receptors

    Abstract Synthetic chemicals currently used in a variety of industrial and cultural applications are leading to widespread contamination oenvironment. Even though the intended uses of pesticides, plasters, antimicrobials, and ame retardants are benecial, effects onman health are a global concern. These so-called endocrine-disrupchemicals (EDCs) can disrupt hormonal balance and result in devemental and reproductive abnormalities. New in vitro, in vivo, anddemiological studies link human EDCexposure with obesity, metabsyndrome, and type 2 diabetes. Here we review the main chemical cpounds that may contribute to metabolic disruption. We then prestheir demonstrated or suggested mechanisms of action with respenuclear receptor signaling. Finally, we discuss the difculties of assessing the risks linked to EDC exposure, including developmexposure, problems of high- and low-dose exposure, and the comity of current chemical environments.

    135

    Click here for quick links toAnnual Reviews content online,including:

    Other articles in this volume Top cited articles Top downloaded articles Our comprehensive search

    FurtherANNUALREVIEWS

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    2/31

    EDCs: endocrine-disrupting chemicals

    Metabolic syndrome:a combination of disorders includingimpaired glucosetolerance or insulinresistance,dyslipidemia, highblood pressure, andobesity

    NRs: nuclearreceptors

    1. INTRODUCTION

    1.1. Endocrine Disruption

    Endocrine disruptors are exogenous com-pounds with the potential to disturb hormonalregulation and the normal endocrine system,consequently affecting healthand reproduction

    in animals and humans (1). Endocrine disrup-tors can interfere with the production, release,metabolism, and elimination of or can mimicthe occurrence of natural hormones (2). En-docrine disruptors may also be derived fromnatural animal, human, or plant (phytoestro-gen) sources; however, for the most part in-ternational concern is currently focused onsynthetic chemicals and endocrine-disruptingchemicals (EDCs). This concern is further am-pliedby twofactors, theexpansioninchemical

    production, which hasnowreached 400milliontons globally, and the increased pollution fromthese chemicals. As such, the impact on humanhealth through known or unknown effects of these chemicals on hormonal systems is great.

    The term endocrine disruptors was rstcoined by Ana Soto and collaborators, whoidentied a number of developmental effectsof EDCs in wildlife and humans (3). AlthoughEDCs can target various hormone systems, anumber of observations concerning reproduc-

    tive development and sex differentiation, to-gether with early embryonic development andpuberty, have focused on EDC interference with sex steroid hormones.

    1.2. Metabolic Disruption: A Subdivision of Endocrine Disruption In addition to the developmental and repro-ductive effects, there is also a growing con-cern that metabolic disorders may be linked with EDCs. Globalobesityrates have risen dra-matically over the past three decades in adults,children, and adolescents, especially in devel-oped countries.Obesityis frequently associated with metabolic disorders (including type 2 dia-betes, metabolic syndrome, cardiovascular andpulmonary complications, and liver disease) as well as other health issues such as psycholog-

    ical/social problems, reproductive defects, ansome forms of cancer.

    A combination of genetic, lifestyle, and e vironmental factors likely account for the rapand signicant increase in obesity rates. Athough genetic factors may explain a portioof obesity predisposition, they alone are unab

    to account for the sudden appearance and progression of the current worldwide obesity epdemic. Modern lifestyles that include excessienergy intake, lack of physical activity, sledeprivation, and more stable home temperatures appear to be major contributing factorof obesity. However, the increased incidencof metabolic diseases also correlates with sustantial changes in the chemical environmenresulting from new industrial and agriculturprocedures initiatedoverthe past 40years.Thichange in the environment has led to the hypothesis that some of the numerous environmental pollutants are EDCs, interfering with various aspects of metabolism and adding aother risk factor for obesity (4, 5). This hypothesis is supported by laboratory and anmal research as well as epidemiological studthat have shown that a variety of environmetal EDCs can inuence adipogenesis and obsity (reviewed in References 510). Such EDChave been referred to as environmental obesogens (11). However, because adverse effects EDCs may also lead to other metabolic diseasesuchasmetabolicsyndromeandtype2diabetesthis subclass of EDCs would be better referreto as metabolic disruptors (12).

    1.3. A Common Molecular Mechanism for Endocrine Disruption and Metabolic Disruption Hormones function mainly through interac-tions with their cognate receptors, which canbeclassied into two large groups: (a) membranebound receptors, which respond primarilto peptide hormones such as insulin, an(b) nuclear receptors (NRs), which are activateby interaction with small lipophilic hormonesuch as sex steroid hormones. EDCs may posess multiple mechanisms of action; howeve

    136 Casals-Casas Desvergne

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    3/31

    Estrogen receptor(ERs): ER and are members of thnuclear receptorsuperfamily. Theyform homodimerbind to DNA Retinoid X recept(RXR): a membethe nuclear receptsuperfamily and amajor partner of onuclear receptorsas PPAR, PXR, aCAR, with whichforms heterodime

    Persistent organicpollutants: chem

    that persist in theenvironment andbioaccumulate wirisks of adverse eto human health aenvironment

    OCPs:organochlorinepesticides

    because many EDCs are small lipophilic com-pounds, one privileged route is through theirdirect interaction with a given NR, which pre-sumably perturbs or modulates downstreamgene expression. For example, most EDC-associated reproductive and developmental de-fects are thought to result from EDCs interfer-

    ing with the function of the estrogen receptor(ER) and/or androgen receptor (AR), thereby disrupting the normal activity of estrogens andandrogens ligands.

    In humans, the NR superfamily encom-passes 48 members that share a commonstructure and, once activated, bind as dimers tospecic response elements located near targetgene promoters. These dimers may be homo-dimers or heterodimers with retinoid X recep-tor (RXR), another member of the NR super-family. In addition to the sex steroid receptors,the NR superfamily includes transcription fac-tors that play pivotal roles in the integration of the complexities of metabolic homeostasis anddevelopment. The ability of EDCs to interact with these NRs is supported by, and explains,the wide range of metabolic perturbations re-ported in both experimental and epidemiolog-ical studies. It also reinforces the concept of as-sociating endocrine and metabolic disruption.

    The present review focuses on metabolicdisruptors and is organized into three sections. The rst section discusses the chemical com-pounds that are presently considered to be ma- jor potential endocrine/metabolic disruptors. Also summarized is the impact of these chem-icals on human health and metabolism on thebasis of available epidemiological studies. Thesecond section highlights recent advances inestablished or proposed mechanisms of EDC-mediated metabolicdisruption.The last sectionhighlights the main challenges that scientistsand regulators face in this eld.

    2. A MYRIAD OF ENDOCRINE-DISRUPTINGCHEMICALSEDCs encompass a variety of chemical classes,including pesticides, compounds used in the

    plastic industry and in consumer products, andother industrial by-products and pollutants. They are often widely dispersed in the envi-ronment and, if persistent, can be transportedlong distances; EDCs are found in virtually allregions of theworld (1317). Persistent organicpollutants are prevalent among environmen-

    tal contaminants because they are resistant tocommonmodesofchemical,biological, or pho-tolytic degradation.Moreover, many EDCs canbe stored for years in animal and human fatmass. However, other EDCs that are rapidly degraded in the environment or the humanbody, or that may be present for only short pe-riods of time, can also have serious deleteriouseffects if exposure occurs during critical devel-opmental periods.

    EDCs can be categorized according to theirintended use (e.g., pesticides) or their struc-tural properties (e.g., dioxins). The main cate-goriesof chemicals with suspected metabolism-disrupting activity are presented below (see Tables 1 and 2). For more detailed informa-tion, the interestedreadermayrefer to in-depthreviews focusedon specic chemical categories,as discussed below.

    2.1. PesticidesPesticides are any substance or mixture of sub-stances intended forpreventing, destroying, re-pelling, or mitigating any pest (1, 18). Severalhundreds, if not thousands, of different chemi-cals are used as pesticides, and human exposureto these pesticides is widespread. Prominentchemical families include organochlorine pes-ticides (OCPs), organophosphates, carbamates,triazines, and pyrethroids. All OCPs are per-sistent. Even though OCPs such as the insec-ticide dichlorodiphenyltrichloroethane (DDT)are currently banned in most developed coun-tries and were subsequently replaced in 1975by organophosphates and carbamates, DDTcontamination still exists. OCPs are detectedin human breast milk and adipose tissueand may exhibit estrogenic, antiestrogenic,or antiandrogenic activity. Their association with breast cancer is suspected but not yet

    www.annualreviews.org Pollutants and Metabolic Disruption 137

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    4/31

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    5/31

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    6/31

    NHANES

    The National Health and Nutrition Examination Survey (NHANES) is the American food consumption database pro-gram conducted by the National Center for Health Statistics,Centers for Disease Control and Prevention. This program was

    designed to collect data and to assess the health and nutritionalstatus of adults and children in the United States. The programbegan in 1960, but in 1999 NHANES was changed to include thetesting of blood and urine for an extensive number of chemicalsand to monitor roughly 5,000 to 10,000 nationally representativepersons per year. All NHANES surveys are cross-sectional andcontain a core set of physical examinations, clinical and labora-tory tests, andpersonal interviews. Information about diseaseandhealthstatus,diet, sociodemographics, occupation, andeducationis collected. In addition, tests are conducted for a varietyof mate-rials, such as micronutrients, disease markers, and environmentalpollutants. NHANESs partnership with the EnvironmentalProtection Agency has also allowed for the implementation of longitudinal studies of many important environmental inu-ences on health [NHANES I Epidemiologic Follow-Up Study (NHEFS)]. NHANES and NHEFS data are public and availablefor researchers at http://www.cdc.gov/nchs/nhanes.htm .

    PVC: polyvinylchloride

    demonstrated by epidemiological studies (19). A well-documented case study in the UnitedKingdom listed 127 pesticides identied ashaving endocrine-disrupting properties (16).Despite confounding issues stemming fromthe multifactorial causes of disease and thechallenges in monitoring pesticide exposure,this study underscores the link between med-ical problems and pesticide exposure (16). With respect to metabolic disorders, a largenumber of epidemiological studies have alsolinked pesticide exposure with obesity, dia-betes, insulin resistance, and metabolic syn-drome (9, 20). For example, an association wasdiscovered between prenatal exposure to theDDT breakdown product dichlorodiphenyl-dichloroethylene (DDE) and increased body mass index in adult women (21). Similarly,cord blood levels of the OCP hexachloroben-zene correlated with a two- to threefold-higherrisk of an elevated body mass index and obe-sity in children (22). Another study used the

    National Health and Nutrition ExaminationSurvey (NHANES) database and carried oua cross-sectional analysis of 1,721 adults (sNHANES and Observational Studies sidebars); the study reported a positive assocition between diabetes and the levels of 19 dferent persistent pollutants (including OCPs

    measured in serum (21). Other epidemiological studies nd a signicant association btween pesticide exposure [mostly OCPs sucas heptachlore epoxide, oxychlordane, or hexachlorocyclohexane ( -HCH)] and higherincidences of metabolic syndrome, insulin rsistance, and diabetes (23, 24). A higher prevlence of diabetes is also associated with DDexposure (25).

    2.2. DioxinsDioxins consist of a group of organochlrines that include the polychlorinated dibenzodioxins (PCDDs), the polychlorinated dibenzofurans (PCDFs), and the polychlorinatebiphenyls (PCBs) and other related compounds. Dioxins can be produced from natural sources such as volcanoes and foreres but are created mostly by human ativity as by-products in organochloride production, in incineration of chlorine-containingsubstances such as polyvinyl chloride (PVCand in bleached paper production. The PCDD2,3,7,8-tetrachlorodibenzo- p-dioxin (TCDD)is the most toxic of all dioxins. This dioxin wa major contaminant in the Seveso catastrophand in the Vietnam War (Agent Orange); it wasalso used as a poison in the attempted assasnation of Viktor Yushchenko (17).

    Dioxins are fat soluble and readily climb tfood chain via their bioaccumulation in fat tisues. They are neither readily metabolized noexcreted, and TCDD has a half-life of approximately 8 years in humans. Several epidemilogical studies have evaluated the toxic effects TCDDandothersdioxins on thegeneral popu-lation as well as heavilyexposed subgroups suas Vietnam War veterans (reviewed in Refeences 17 and 26). These studies demonstrate cause and effect between dioxin exposure, w

    140 Casals-Casas Desvergne

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    7/31

    PFCs: polyuorocompounds

    an increase in cancers, nervous system degen-eration, immune damage, thyroid disease, andreproductiveandsexualdevelopmentdisorders. With respect to metabolism, exploration of theNHANES database indicated that PCDDs andPCDFs are weakly associated with metabolicdisorders, whereas PCBs are strongly associ-

    ated with type 2 diabetes (27). Several cross-sectional investigations further supported thesecorrelations (reviewed in Reference 25).

    2.3. OrganotinsOrganotins, including tributyltin chloride(TBT) andbis(triphenyltin) oxide (TPTO), arepersistent organic pollutants that have been widelyused as agricultural fungicides, as rodentrepellents, as molluscicides, and in antifoulingpaints for ships and shing nets. Organotincompounds such as PVC are also used to stabi-lize plastics.

    TBT and TPTO provide one of the clear-est examples of environmental endocrine dis-ruption: Exposure of marine gastropods to very lowconcentrationsof thesecompounds inducesan irreversible sexual abnormality in femalestermed imposex, resulting in impaired repro-ductive tness and possibly sterility (28). Con-cerns over the toxicity of these compounds ledto a worldwide restriction and a ban on marineuses. Currently, human exposure may comefrom dietary sources, such as sh and shell-sh, or through contaminated drinking waterandfood.However,noepidemiologicaldata areavailableconcerninghuman exposure, although TBT has been reported to have modest adverseeffects on mammalian male and female repro-ductive tracts (29, 30). Nonetheless, recent ex-perimental studies revealed proadipogenic ac-tivity of TBT and TPTO (11, 31).

    2.4. PlasticsOwing to their variety, robustness, and ex-tremely low costs, plastics are fundamental inmodernlife,publichealth, andmedicine; plasticproduction exceeded 300 million tons in 2010(32). After more than ve decadesof debate and

    OBSERVATIONAL STUDIES

    Different types of observational studies are detailed http://www.vetmed.wsu.edu/courses-jmgay/glossclinstudy.htm . A summary is presented below.

    Clinical studies aimed at evaluating the effects of EDC ex

    sure rely mainly on observational studies, which interpret expiments of nature and are exposed to large confounding biasHowever, such studies provide preliminary evidence to be uas a basis for hypotheses.

    Cross-sectional studies examine the relationship between deases and other factors (such as EDC levels) at one point in tin a denedpopulation. Cross-sectional studies lack anyinformtionon timingof exposure andinclude only prevalentcases.Thcan suggest a link between a disease and the factor of interes

    In longitudinal studies, or cohort studies, a panel of indivials is interviewed repeatedly over a period of time. This allow

    prospective and analytical study of a group in which somedividuals have had, currently have, or will have the exposof interest to determine the association between that exposuandan outcome. Cohort studiesare stronger than cross-sectionaand case-control studies when well executed, but they are mexpensive.

    Case-control studies are a retrospective comparison of tproportion of cases with a potential risk factor to the propotion of controls (individuals without the disease) with the sarisk factor. Due to the potential for many forms of bias in tstudy type, case-control studies provide relatively weak empirevidence, even when properly executed.

    research, controversy still surrounds the risksthat plastics may cause in humans, particularly with respect to endocrine-disrupting proper-ties. Adverse effects can stem from the variouscomponents of plastics, the additives used, ora combination of both. Laboratory animal andepidemiological studies have studied the effectsof several of these substances on human health. A few examples are provided below.

    2.4.1. Polyuoroalkyl compounds. Polyu-oroalkyl compounds (PFCs) are synthetic u-orinated organic compounds used in a widerange of industrial applications and consumerproducts, including paper, leather, textile coat-ings, and re-ghting foam, and in the polymer

    www.annualreviews.org Pollutants and Metabolic Disruption 141

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    8/31

    BFRs: brominatedame retardants

    industry. Among them, peruorooctane sul-fonate (PFOS) and peruorooctanoate (PFOA)are widely detected in the environment. PFCsare classied as persistent organic pollutants,even though they are not stored in fat tissue butinstead form chemical adducts with liver andserum proteins. Laboratory rodents exposed to

    PFCs exhibit developmental effects such as re-duced birthweightandincreased neonatalmor-tality (3335). In addition to hormonal pertur-bations with decreased testosterone levels andincreased estradiol levels in adult rats (36), re-ductions in serum cholesterol and/or triglyc-eride in mice and rats exposed to high doses of PFOS and PFOA (37) suggest that these chem-icals disturb normal lipid metabolism.

    Inverse relationships were observed be-tween PFOS and PFOA concentrations in cordblood and birth weight, ponderal index, andhead circumference in children (38; reviewedin Reference 15). Numerous studies have eval-uated the possible associations of blood PFClevelswith metabolicparameters; themost con-sistent result is the positive association betweenPFC exposure and increased cholesterol, par-ticularly high-density lipoprotein (HDL) (3944). The same reports discounted any associa-tion with metabolic diseases such as type 2 dia-betes andmetabolic syndrome (3944) andsug-gested a complex positive correlation of PFClevels with hyperglycemia but an inverse corre-lation with metabolic syndrome in adolescents(45). Most of these studies are cross-sectionaland as such fail to provide a causal link butrather draw attention to the potential effects of PFCs on human physiology.

    2.4.2. Brominated ame retardants. Bromi-nated ame retardants (BFRs), particularly polybrominated diphenyl ethers (PBDEs), areadditives used as ame retardants in a greatnumber of consumer products such as houseelectronic equipment, clothing, and furniture. Although these compounds have decreased reincidences, they are highly prevalent, ubiq-uitous, and persistent pollutants. The mainsources of human exposure come from indoorenvironments, diet, and occupational exposure

    (13, 46, 47). Despite their benecial effectthese chemicals are also thought to adverseaffect human health through endocrine disruption and developmental neurotoxicity (48). Iaddition, increased incidences of hepatocellular carcinoma and thyroid adenoma have beeobserved in rodents, albeit with relatively hig

    exposure doses (49). Taken together, these observations have led to the recent ban of PBDEin several American states and in the EuropeaUnion,where a ban onall BFR use isbeing contemplated. Meanwhile, PBDEs are still presenin environmental samples and are detected imilk, serum, and adipose tissue in animals anhumans.

    Published studies addressing the consequences of human BRFexposure remainscarceand the effects of BFR exposure on sex sterohormone systems are still poorly understoodOne study demonstrated a correlation between PBDEs in breast milk and congenitacryptorchidism, although confounding factormay have been present (50). Only a few epdemiological studies have addressed the posible impacts of these persistent pollutants ometabolic parameters in humans. The mostelling study revealed that, among six dferent BFRs, PBDE-153 and the polybrominated biphenyl PBB-153 showed an inverteU-shaped association with type 2 diabetes anmetabolic syndrome (51). Clearly, more studies are needed to clarify the possible extent BFR-related damage.

    2.4.3. Alkylphenols. Alkylphenols such as n-nonylphenol and 4-n-octylphenol are surfactants widely used in detergents, emulsierantistatic agents, demulsiers, and solubilizeand are found commonly in wastewater (52 They are also used as additives to plastics suas PVC and polystyrene, from which they caleach. Alkylphenols are capable of initiatiproliferation in breast tumor cells in the laboratory (53), consistent with their capacities foestrogenic and antiandrogenic activity (54, 5andreferencestherein).However,giventhelowenvironmental concentrations and the currenregulation of alkylphenol use in the Europea

    142 Casals-Casas Desvergne

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    9/31

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    10/31

    Diethylhexylphthalate (DEHP):the most-usedphthalate; itsmetabolite, MEHP[mono(2-ethylhexyl)phthalate],interferes with severaltypes of nuclearreceptors

    leads to the urinary excretion of the conjugatedmetabolites.

    Among all the phthalates, diethylhexylphthalate (DEHP) elicits the most concern, with more than two million tons producedannually. This compound is widely used inmedical devices and in a variety of food

    products. DEHP causes animal toxicity inmany physiological systems; however, many of the abnormalities that have been characterizedsince the 1940s have occurred at high DEHPdoses (32). In addition, DEHP promotes livertumor development in rodent models throughsevere peroxisomal proliferation. However,peroxisome proliferation has not been ob-served in humans, and according to a decisionof the International Agency for Research onCancer, DEHP cannot be classied as a humancarcinogen.

    Experimental studies at low doses of DEHPexposure, which appear to be most pertinent tohuman health, have demonstrated subtle repro-ductive toxicity in male rodents (64, 65). Otherreproductive outcomes include testicular dys-genesis together with permanent feminizationand demasculinization, resulting in a reducedanogenital distance (66).

    Some epidemiological studies reportedan association between cord blood levelsof mono(2-ethylhexyl)phthalate (MEHP), aDEHP metabolite, and shorter gestational ageof delivery. Indirect evidence also suggeststhat diethyl phthalate and dibutyl phthalatemay impart antiandrogenic effects in theperinatal period (reviewed in Reference 67). Maternal urine levels of metabolites of DEHP(benzylbutyl phthalate, diethyl phthalate, anddibutyl phthalate) are associated with a higherrisk of incomplete testicular descent for malehuman infants and are inversely correlated with the anogenital distance (68, 69). Otherdevelopmental effects of phthalate exposuremay cause damage to the pulmonary systemand may result in asthma (70).

    More recently, several studies have demon-strated a correlation between phthalates andmetabolic disorders. In short- and long-termrodent studies, dose-related deregulation of

    levels of serum insulin, blood glucose, livglycogen, T3, T4, thyroid-stimulating hor-mone, and cortisol was observed (71, 72). humans, the log-transformed concentrations ofseveral phthalate metabolites positively corrlated with abdominal obesity and insulin resitance in adult males (73). These analyses su

    port the concept of environmental obesogenbut await further conrmation by longitudinastudies.

    At rst glance, this presentation of so mantypes of chemicals suspected of generatimetabolic disruptors may seem alarming (se Tables 1 and 2). However, because many studies discussed here are cross-sectional, a dentivecausal link between metabolicdisordersanEDCexposure is still hypothetical. Forthat reason, parallel studies aimed at identifying thmolecularmechanisms of EDCactivity withregard to metabolism should provide greater insights into the real health risks posed by thecompounds.

    3. METABOLIC DISRUPTION: MECHANISTIC APPROACHESIn the context of endocrine disruptionmetabolic disruption may result from thremain types of activity. First, hormones in general andsexsteroidhormones in particular contribute to general body homeostasis througdiverse metabolic regulations. Thus, a certainumber of metabolic perturbations are simplthe result of hormonal disruption. Second, direct EDC activity through receptors responding to xenobiotics and regulating xenobiotmetabolism may also contribute to a metabolphenotype. Third, EDC interactions with spe-cialized metabolic receptors may serve as a pmarymechanismformetabolicdisruption.Thiarticle presents and discusses experimental oservations linking EDCs with metabolic diruption along these three types of activity (sFigure 1 ).

    3.1. Metabolic Disruption Through Hormone ReceptorsHormone receptors belong to a class of clasic hormone receptors that recognize only on

    144 Casals-Casas Desvergne

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    11/31

    or a few molecules with high afnity. Thyroidhormone (TH), mineralocorticoid, glucocorti-coid, retinoic acid, estrogen, vitamin D, pro-gesterone, and androgen receptors belong tothis class. Initial studies identied ER and ARas the targets of many EDCs, which resulted indevelopmental and reproductive effects, as well

    as metabolic alterations.3.1.1. Metabolic disruption mediated by in-appropriate activation of the estrogen re-ceptor. ER and ER are the main media-tors of the biological effects of estrogens. Uponestrogen binding, they form homodimers thatbind to the promoters of estrogen-responsivegenes. These molecules share a similar struc-ture and bind to the same response elementbut have varying relative binding afnities forsome steroid hormones. In addition to their well-established roles in reproduction, ERandER areinvolved inbraindevelopmentandfunction of many other organs, such as skin,bone, and liver.

    Several lines of evidence link ERs tometabolism. For example, in postmenopausal women and ovarectomized rodents in whichestrogen is low, one observes an increase in white adipose tissue; estrogen replacementtherapy reverses these effects. ER but notER appears to mediate these effects, asinferred from studies using mice in which ERis knocked out: Both male and female mutantmice show increased insulin resistance andimpaired glucose tolerance (74, 75). Althoughthe underlying mechanisms remain unclearfor these observed results, it seems likely thatER activation modulates neural networkscontrolling food intake as well as acts directly in adipose tissue (reviewed in Reference 76). Ata cellular level, preadipocytes also express ERand ER , and during development, estrogenscontribute to an increase in adipocyte number, with subsequent effects on adipocyte function(77). At the molecular level, ERs and estrogensregulate many aspects of metabolism, includingglucose transport, glycolysis, mitochondrialstructure and activity, and fatty acid oxidation(reviewed in Reference 8).

    Bisphenol A

    Adipogenesis Insulin levelsBody weight

    Phthalates Organotins Dioxins

    ARNTAhR

    PFC

    ?

    CAR/PXR

    RXRPPAR RXR PPAR RGRGR ER ER RXRTR

    Figure 1Endocrine-disrupting chemicals (EDCs) interact with aryl hydrocarbonreceptor (AhR) and with diverse members of the nuclear receptor (NR)superfamily, which convey EDC-mediated metabolic disruption. Sensorreceptors like peroxisome proliferatoractivated receptors (PPARs) play prominent role: PPAR activated by phthalates or organotins inducesadipogenesis in vitro, and this effect is inhibited by dioxins through AhRactivation. PPAR , which has a major role in fatty acid oxidation, limitsadipogenic activity and is activated in vivo by phthalates and polyuoroacompounds (PFCs). Estrogenic EDCs such as bisphenol A have also beeinvolved in metabolic disruption through complex interaction with hormoreceptors such as the estrogen receptors (ERs), thyroid hormone receptor(TR), and glucocorticoid receptor (GR). These receptors are important focontrol of adipogenesis, weight gain, and insulin levels, although theunderlying mechanisms are not yet well understood (dashed lines ). Finally,though the mechanisms have yet to be described (dashed lines and questionmark), the xenosensors pregnane X receptor (PXR), constitutive androstareceptor (CAR), and AhR also play a crucial role in regulating metabolichomeostasis via direct or indirect interaction with the metabolic pathwayOther abbreviations used: ARNT, aryl hydrocarbon receptor nucleartranslocator; RXR, retinoid X receptor.

    Experimental evidence showing the effectsof estrogen-mimicking EDCs such as BPA on metabolism remains scarce and has beenrestricted to cultured cell line models. Studiesusing 3T3-L1 cells suggested that early BPA exposure may enhance adipocyte differenti-ation in a dose-dependent manner and may permanently disrupt adipocyte-specic geneexpression and leptin synthesis (78, 79). Forinstance, the estrogenic surfactant octylphenolelevates adipocyte production of resistinthrough activation of the ER and extracellularsignalregulated kinase pathways in 3T3-L1

    www.annualreviews.org Pollutants and Metabolic Disruption 145

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    12/31

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    13/31

    Pregnane X recep(PXR): a nuclearreceptor known a xenosensor and mregulator of detoxicationpathways; knownsteroid X receptohumans

    Constitutiveandrostane recept(CAR): a nucleareceptor known a xenosensor and mregulator of detoxicationpathways

    AhR: aryl

    hydrocarbon receCytochrome P450(CYP) family: a and diverse groupenzymes playing important role in detoxicationpathways. Theirsubstrates includemetabolicintermediates and xenobiotic substa

    loss. The exact opposite results are observed with low TH levels.

    In contrast to TR, GR forms homodimersand resides in the cytosol, forming complexes with molecular chaperones. Ligand bindingreleases the chaperones, triggers GR nucleartranslocation, and inuences gene expression.

    Glucocorticoids acting through GRs allow anorganism to adequately respond to physical oremotional stresses by promoting gluconeogen-esis, increasing blood glucose levels, and mo-bilizing the oxidation of fatty acids. The phar-macological uses of glucocorticoids, chiey inthe context of controlling chronic inamma-tion, have serious metabolic side effects such asdiabetes, muscle wasting, and growth retarda-tion in children.

    EDCs also interact with these TR and GRreceptors. For instance, in differentiating 3T3-L1 cells, BPA and dicyclohexyl phthalate stim-ulate GR-mediated lipid accumulation andsyn-ergize with a weak GR agonist to increaseexpression of adipocyte-specic markers (91).BPA may also act as an antagonist of the TRpathway by enhancing recruitment of the core-pressor NCoR to TR (92). In parallel, perinatalexposure of BPA increases levels of thyroxine(T4) (93). Given the important role of TH inenergy homeostasis, BPA effects on TR dur-ing development may be important in long-term body weight increase. BFRs also disruptthe TH pathway, and daily exposure of rats toPBDE over four weeks resulted in a signicantincrease in lipolysis and a signicant decreasein glucose oxidation, characteristics associated with obesity, insulin resistance, and type 2 di-abetes, although such exposure had no effecton body weight and adipocyte size. Althoughthe underlying molecular mechanisms remainto be experimentally addressed, these physio-logical effects are consistent with a change inER and TR pathways (94, 95).

    3.2. Metabolic Disruption Through Xenosensors The body is protected from the accumulationof toxic chemicals by a complex strategy that

    in part takes place in the liver, regulating theexpression of drug-metabolizing enzymes andtransporters. This adaptive response incorpo-rates at least three xenosensors: pregnane X re-ceptor (PXR), constitutive androstane receptor(CAR),and aryl hydrocarbonreceptor (AhR),as well as xenobiotic metabolism and transporter

    systems.3.2.1. Pregnane X receptor and constitutiveandrostane receptor. PXR and CAR aremembers of the NR superfamily of sensorreceptors, and although they were originally dened as xenosensors involved in regulatingthe metabolism of xenobiotics, their contribu-tion to fatty acid, lipid, and glucose metabolismhas been only recently appreciated (96, 97).

    PXR and CAR regulate gene expression by forming heterodimers with RXR that bind to xenobiotic response sequences present in thepromotersof their target genes.However, theirmechanisms of activation differ. PXR is locatedprimarily in the nucleus and is strongly acti- vated upon ligand binding. In contrast, in theabsence of ligand, CAR is retained in the cy-toplasm through association with the cytoplas-mic CAR-retention protein (CCRP) and heat-shock protein 90 (HSP90). In the presence of activators, CAR dissociates from its two chap-erones and translocates into the nucleus, whereit forms heterodimers with RXR (reviewed inReference 98).

    PXR and CAR are highly expressed in theliver, where they act as master regulators of detoxication pathways through induction of phase I to phase III enzymes. In the rst phase,a polar group is added to hydrophobic sub-strates by hydroxylation and oxidation via thecytochrome P450 (CYP) mono-oxygenase sys-tem. CYP3A is responsible for the metabolismof up to 60% of the drugs presently on themarket (reviewed in Reference 94) and is amajor target gene of PXR, whereasphenobarbital-induced activation of CARtriggers the expression of CYP2B. Phase IIenzymes increase hydrophilicity of the com-pounds through various conjugation reactions,and phase III involves transporters that allow

    www.annualreviews.org Pollutants and Metabolic Disruption 147

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    14/31

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    15/31

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    16/31

    CYPexpression,altering ER transcriptional ac-tivity via coactivator squelching, or promot-ing DNA-binding competition (121, 122) (seeFigure 2 ). In addition, AhR also indirectly affects adipogenesis through inhibition of PPAR expression (123).

    Additional experimental and epidemiologi-

    cal studies are still required to assess whether AhR-mediated responses affect metabolism inaddition to the well-known roles of Ahr in im-munity, development, and cancer.

    3.3. Metabolic Disruption Through Peroxisome ProliferatorActivatedReceptors Metabolic homeostasis requires a controlledbalance between energy storage and consump-tion; several NRs and their coregulators are in-strumental in these processes.Amongthese, thePPARs act as lipid sensors that cooperate indifferent organs to adapt gene expression to agivenmetabolicstatus. PPARsaresensorrecep-tors with a rather large ligand-binding domain, which can accommodate a variety of ligands,primarily lipid derivatives. In the presence of ligand, PPARs heterodimerize with RXR andbind to the PPAR response elements localizedin the promoter regions of their target genes(124).

    The PPAR family is composed of threeisotypes: PPAR , - / , and - . PPAR is ex-pressed predominantly in tissues characterizedby a high rate of fatty acid catabolism such asliver, kidney, heart, and muscle. PPAR wasrst identied as the protein responsible forthe induction of peroxisome proliferation inrodents exposed to a variety of compoundscollectively termed peroxisome proliferators.However, humans do not undergo peroxisomeproliferation and are thereby protected fromthe consequent liver tumors observed insensitive species. PPAR plays a major role infatty acid oxidation in all species, controllinglipoprotein metabolism and limiting inamma-tion. PPAR is ubiquitously expressed, sharespartially overlapping functions with PPAR ,and also plays a role in cell differentiation and

    survival (125, 126). Finally, PPAR functionin adipogenesis, lipid storage, and the controof insulin sensitivity; it also participates inammatory responses (127).

    Plasticizers, surfactants, pesticides, andioxins can modulate PPAR activity, althougfairly little is known about the molecular mech

    anisms and the physiological outputs involve The specicity of this PPAR-mediated rsponse is highlighted in a study in whi200 pesticides were systematically screened ftheir peroxisome proliferation activity. Onlthree compounds were identied as havinPPAR transcriptional activity, and none possessed PPAR transcriptional activity (127 Among these pesticides, diclofop-methyl anpyrethrins induced PPAR target gene expression at levels similar to those induced by clasagonists in mice (128, 129).

    The phthalates are another group of wellcharacterized peroxisome proliferators (130In vitro transactivation assays and intact celllar systems were used to reveal that phthalatandtheirmetabolitesbindandactivatethethreePPARs, among other NRs (131134). Thesstudies also determined the range of potencand efcacy of phthalate monoesters, showing differences between isotypes and specie Modeling the DEHP metabolite MEHP in thePPAR ligand-binding pocket indicates tha MEHP may contact residues similar to thosdened for the classic PPAR agonist rosiglitazone (135). MEHP induces adipogenesis inPPAR -dependent manner, albeit with loweefciency than rosiglitazone in 3T3-L1 cel(134). Accordingly, gene expression microarraanalyses indicate that 70% of the genes are regulated by either ligand, some of them to diffeing degrees. However, 30% of the genes are exclusively regulated by rosiglitazone and not b MEHP, suggesting that MEHP acts as a selective modulator of PPAR rather than as a fulagonist. This differential activity results fromthe different abilities of MEHP and rosiglitazone to induce the release of corepressors sucas NCoR and the recruitment of coactivatorsuch as p300 or PGC1 (133). Taken togetherthese in vitro data demonstrate that MEHP i

    150 Casals-Casas Desvergne

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    17/31

    proadipogenic in a cell culture model, suggest-ing that it may act as a metabolic disruptor andmay promote obesity in vivo.

    Paradoxically, in vivo experiments partially contradict these results (136). Adult micetreated with high or low doses of DEHP areprotected from weight gain, gaining 30% less

    weight than controls. These mice possess a re-duced fat mass and a metabolic improvement, withlower levels of triglycerides in the liverandthe blood, smaller adipocytes, and enhancedglucose tolerance. These effects were not ob-served in PPAR -null mice, conrming thatin vivo DEHP activity is mediated primarily by PPAR in the liver, leading to increasedfatty acid catabolism and induced expressionof PPAR target genes such as that encodingbroblast growth factor 21 (FGF21) (137) orgenes controlling fatty acid -oxidation. Sur-prisingly, this phenotype is also completely abolished in PPAR -humanized mice (mice in which themouse PPAR allelesare replaced by the human PPAR gene). These mice, whenexposed to a DEHP-containing diet, tend tobe more sensitive to diet-induced obesity thanare untreated controls (136). These observa-tions point to the possibility of species-specicEDC activity, due at least in part to evolution-ary differences in the receptors interacting withthem.

    Studies of phthalate exposure in utero have yielded dissimilar results. Male and female off-spring of rats exposed to diisobutyl phthalateand butylparaben exhibit reduced plasma lep-tin and insulin levels, similar to the modi-cations observed upon in utero exposure torosiglitazone (138). In contrast, a study evalu-ating the impact of in utero exposure to DEHPcould not identify parameters indicating adultmetabolic disorders (6). These differences may be attributable to the compounds tested as wellas the specic experimental protocols. In any case, these studies highlight the necessity to in- vestigate the risks engendered by fetal exposureto phthalates.

    The PFCs, particularly PFOA and PFOS,can also activate mouse and human PPARs intransactivation assays (139), although the in

    vivo consequences of such activity remain quitecontroversial. Adult mice exposedto high dosesof PFOA exhibit weight loss, which is abro-gated in PPAR -null mice (140). The pro-posed mechanism involves PPAR -dependentanorexigenic activity in the hypothalamus of adult rodents (141). In contrast, Hines et al.

    (142) reported that PFOA has noeffect onbody weight gain when exposure occurs at the adultstage.However, developmentalexposure to lowPFC levels results in increased body weightand increased serum insulin and leptin levelsat midlife (142).Again, species-specic PPARactivity was proposed because low doses of PFOA signicantly activate the function of PPAR in wild-type mice but not in PPAR -humanized mice. Human PPAR may there-fore be less responsive to PFOA, increasingthe possibility of species-specic EDC activ-ity. More specically, the extent to which thesePPAR activators inuence metabolic home-ostasis in humans deserves more study (143).

    Several EDCs also specically targetPPAR . Using a high-throughput method,Kanayama et al. (31) showed that among 40EDCs, organotins such as TBT and TPTOare activators of human PPAR and RXR. TBT binds to and activates the three humansubtypes of RXR as well as many permissiveheterodimeric partners such as liver X receptor(LXR), nuclear receptorrelated 1 protein(NURR1), PPAR , and PPAR , but notPPAR . Organotins bind and activate, pri-marily through RXR and not through PPAR ,the PPAR :RXR heterodimer at nanomolarconcentrations. The crystal structure of theRXR ligand-binding domain bound to TBTindicates that TBT binds with high afnity toRXR, even though TBT is structurally distinctfrom above-described ligands and only partly occupies the RXR ligand-binding pocket(144). Consistent with the critical role playedby PPAR :RXR signaling in mammalianadipogenesis, TBT promotes adipogenesisin 3T3-L1 cells by direct transcriptionaleffects on the PPAR target genes. In uteroexposure to TBT in rodents led to alter-ations in fat structure and metabolism, with a

    www.annualreviews.org Pollutants and Metabolic Disruption 151

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    18/31

    disorganization of hepatic and gonadal archi-tecture, steatosis in the liver, and an increase inlipid accumulation and mature adipocytes. Thefat massbut not the total body weightof inutero TBT-treated mice signicantly increasesin adulthood, supporting the conclusion thatembryonic and chronic lifetime organotin

    exposure may contribute to the incidence of obesity through disruption of the PPAR :RXRpathway (28).

    Altogether, these many examples of EDCinteraction with receptors highlight the factthat a given compound can interfere withdifferent NRs and different pathways (see Table 1 ). For example, depending on the com-pound, BFRs interface with AR, ER, and pro-gesterone receptor to elicit both agonist- andantagonist-like effects (94). PBDEs bind but donot activate AhR (145); in contrast, they in-duce the expression of various CYP enzymes,in part through the activation of PXR (146,147). PBDEs are also active in TH regula-tion by disrupting peripheral TH transport andmetabolism/deactivation or by binding and ac-tivating TRs(148,149).The nal consequencesof EDCs exposure are thus due to cross-talk be-tween these pathways, rather than to a linearcausation chain (96, 114, 122, 123, 150), andare much more complex to decipher in vivo.

    4. METABOLIC DISRUPTORS: TOWARD MANY CHALLENGES This review emphasizes the remarkable emer-gence of EDC-related research, which hasshifted focus from endocrine disruption tometabolic disruption. Epidemiological studiesthat underscore the parallels between EDC ex-posure and obesity incidence, as well as animallaboratory studies that demonstrate the ability of EDCs to act on metabolic transcription fac-tors, are lines of investigation that cannot becasually discounted. However, there are many difculties to overcome beforeonecanfairlyas-sess the risks that past, present, and future envi-ronmental chemicals engender on human and wildlife health. In this last section, we highlight

    some of the importantquestions that remainforresearchers and regulators.

    4.1. Monitoring Exposure Levels Ambient monitoring is performed by samplinair, dust, water, etc., and by measuring the levels of the pollutants of interest in these sample This method is often reasonably easy and reable. However, ambient monitoring provides value valid only at the time of sampling, whimaynotreectlevelsofchronicexposure;italsdoes not take into account the efciency wi which living organisms breathe, ingest, or asorb these compounds. In that respect, biomon-itoring is a more appropriate evaluation of thpresence of compounds or their metabolites ibiological samples, particularly in blood an

    urine, and ideally in tissue samples. Biomontoring does not identify the source of the contamination but provides the individual exposure level at a given time point. Biomonitorinassays also reect past exposure to persistepollutants. However, biomonitoring is invasivand costly and cannot be proposed as a standarroutine evaluation, except for occupational exposure. Alternatively, biomonitoring of wildlifsamples can be more easily performed and mserve as a good indicator of exposure to som

    pollutants widely found in the environment.

    4.2. Identifying the MetabolicEffective DoseOne current issue in identifying the metabolieffective dose concerns the so-called U-shapeor inverted U-shaped dose-response curve. dose-response curve of this form is reportefor BPA: Effects are observed at very low dos(from 10 12 M) and at high doses (10 8 M), buno effects are observed at intermediate dos(10 9 M). These U-shaped curves suggest thexistence of two independent mechanisms folow doses and high doses (151, 152). Howevmechanistic studies are still lacking, and evdence for low-dose activity is not availableepidemiological studies.

    152 Casals-Casas Desvergne

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    19/31

    A second complication is exposure to a mix-ture of EDCs rather than to a single EDC. Hu-mans and wildlife are exposed daily to a variety of compounds, and it is thus likely that even if none of the compounds reach an effective level,the combination or mixture of chemicals may become effective. This scenario is supported by

    the observation that various EDCs share re-ceptors, and thus additive effects should be ob-served (see Figure 1 ; Tables 1 and 2). Fewexperimental studies addressing this issue ex-ist, and given the vast number of EDCs, it isunclear how to monitor exposure and how touse or develop assays that capture the effects of these mixtures. At a practical level it is also un-clear how regulatory decisions will be amendedto account for exposure to mixtures rather thanto single compounds.

    4.3. Establishing the Link Between Exposure and Metabolic Effects Metabolic alterations such as metabolic syn-drome and type 2 diabetes are complex andmultifactorial. Elevated body mass index andobesity are not diseases but rather contributeto various alterations that lead to the diseasedstate. EDC exposure is one more factor thatincreases an already long list of predisposingfactors that act in combination to increase the

    risk of obesity or other metabolic alterations.Such contributions can be revealed only by comprehensive studies of large and well-characterized cohorts, such as the cohort usedfor the NHANES project (see sidebar entitledNHANES). However, these studies are cross-sectional (see sidebar entitled ObservationalStudies), due to the difculties inherent in theevaluation of exposure, and may be subjectto unidentied confounding factors. At best,these studies can demonstrate correlations but

    not causal links between exposure and effects.

    4.4. Experimental Exploration of the Metabolic Disruption Properties of Endocrine-Disrupting ChemicalsHow can we reach the most appropriate un-derstanding of EDC biology that will help to

    dene appropriate actions? Although cellularmodels are exible and allow large numbersof conditions and doses to be tested, they arelimited and unable to factor in bioavailabil-ity of the compounds and their metabolites orthe route of exposure. Reductionist molecu-lar and cellular studies may not take into ac-

    count the knowledge that several EDCs inter-fere with a variety of NRs, not all of whichare expressed in the same tissue or at the sametime. Additionally, EDCs can affect several or-gans in the body withdifferent intensities, lead-ing to a global response that may be oppositeof that observed in cell cultures. Experimen-tation in animals is therefore unavoidable, asanimals are threatened by environmental con-taminants and provide a reasonable approxi-mation of human metabolism for most com-pounds. However, two main issues must beconsidered. First, EDC-interacting receptorsdisplay species-specic activity that is well de-scribed for PPAR , PXR, and CAR (98, 136). The development of humanized mice, in whichthe gene of the receptorof interest is exchangedfor the human allele, provides new tools for thistype of investigation (136, 143). Second, issuesof dose and exposure time complicate exper-imental design (see Table 2 ). Mice may livefor two years, and a three-month exposure canbe considered chronic. Does this experimentalsetup accurately reect the decades over whichhumans might be exposed?

    4.5. Exposure During Critical Periodsof Development The dramatic effect of DESexposure on femalebabies illustrates the critical issue of exposureduring development (see Section 3.1.1). Con-sistent with the well-known functions of clas-sic hormones in developing reproductive or-gans, the effects are easily understood whenthese hormones are disrupted or mimicked.Evendisregarding transgenerational effects, themetabolic consequences of prenatal exposurethat manifest in adulthood are difcult to as-sess and to understand. As discussed above (seeSection 3.1.1), controversy surrounds many of

    www.annualreviews.org Pollutants and Metabolic Disruption 153

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    20/31

    these consequences, andonly systematicanimalstudies may lead to a fair risk evaluation. Futureefforts should be aimed at elucidating whetherand how epigenetic imprinting is involved inthese pathologies (153).

    4.6. How Should New ChemicalsBe Regulated? Asdescribed in Table 1 , several chemical prod-ucts are either totally banned or authorized foruseunderstrictconditions.Banning a persistentorganic pollutant will immediately limit expo-sure, but a number of years will pass before thechemical is entirely suppressed, and such sup-pression will occur only if large transnationalterritories ban the chemical of interest (see Table 2 ). Inevitably, banned products are re-placed by others that bring other unwanted ef-fects. How do we establish the most pertinentand effective modes of risk evaluation not only for the present but also for any future com-pound released to consumers?

    Fromtheresearchpointofview,recentyearshave witnessed many efforts to set up new tech-nologies for EDC detection in human tissues,including scenarios of low doses, nonpersistentEDCs, and the developmental period (27, 56,67). Newintegrative approaches combiningge-nomics, proteomics, metabolomics, systems bi-ology, andcomputational modelingshouldhelpto understand the complexity of the cocktail ef-fect andits consequenceswhen exposure occursat various life stages (101, 150, 154). In addi-tion, these approaches should help to evaluate

    the global effects of EDC interference with different NRs and the activation of a complex bological network. Before reaching this level understanding, these integrative strategies mahelp dene a signature: a composite signal no explanatory value but reecting exposure metabolicdisruptors. Such a signaturemayhel

    inexperimentally establishinga predictivevaluto new compounds.From the regulatory point of view, govern

    ments face the challenge of making appropratedecisionsby balancingpotentialrisksagaindemonstrated advantages. Help for future decisions will come from two complementary prcesses. The rst process is illustrated by thEuropean Registration, Evaluation, Authorisation, and Restriction of Chemicals (REACHprogram, aimed at protection of human healthand the environment through systematic registration, assessment, and annotation of an exhaustive list of industry compounds. The secondprocess encourages research through gransubsidiaries, as illustrated by the recent naning of BPA research by the National Institutes of Health(NIH).These initiatives includeincentives for transparency and collaborationboth at the level of government and betweescientists. Metabolic perturbations areonly onsmall aspect of the EDC-related problems tbe solved, but what we know now may be onthe tip of the iceberg. In the present context oendemic metabolic disorders, with severe ecnomic, social, and professional consequenceevery actionrst to understand andthen to con-trol risk factors is benecial on all counts.

    SUMMARY POINTS

    1. Increasing human exposure to endocrine-disrupting chemicals (EDCs) has been asso-ciated with the development of some of the main ailments of the industrialized world,particularly metabolic disorders like obesity, diabetes, and metabolic syndrome.

    2. Amongdifferentmechanisms of action, lipophilic EDCs compoundscanbind specically to nuclear receptors andcan displace the corresponding endogenous ligands to modulatehormone-responsive pathways.

    3. Persistent organic pollutants such as organochlorine pesticides, dioxins, and polyuo-roalkyl compounds and nonpersistent pollutants such as bisphenol A and several phtha-lates are suspected of metabolic disruption activity.

    154 Casals-Casas Desvergne

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    21/31

    4. A major mechanism of EDC-mediated metabolic disruption is through EDC interaction with nuclear receptors, including (a) sex steroid hormone receptors, (b) receptors actingas xenobiotic sensors, and (c ) receptors specialized in metabolic regulations.

    5. Thiseldislitteredbycontroversies,inpartduetothedifcultiesinprovingordisprovingEDC activity. The major issues are the monitoring of exposure levels, the identicationof the metabolic effective dose, and the establishment of a link between (a) either expo-sure during critical periods of development or chronic exposure at very low doses and(b) metabolic effects. Finally, this area of research would benet tremendously if com-mon methodologies of experimental EDC exposure were established. All these issuesneed further work to create a common and effective regulatory policy for environmentalchemical pollutants.

    DISCLOSURE STATEMENT The authors are not aware of any afliations, memberships, funding, or nancial holdings thatmight be perceived as affecting the objectivity of this review.

    LITERATURE CITED1. U.S. Environ. Prot. Agency. 2000. Endocrine Disruptor Screening and Testing Advisory Com-

    mittee ( EDSTAC ). Washington, DC: U.S. Environ. Prot. Agency. http://www.epa.gov/scipoly/ oscpendo/edspoverview/edstac.htm (accessed September 14, 2006)

    2. Tabb M, Blumberg B. 2006. New modes of action for endocrine-disrupting chemicals.Mol. Endocrinol.20:47582

    3. Colborn T, vom Saal FS, Soto AM. 1993. Developmental effects of endocrine-disrupting chemicals in wildlife and humans.Environ. Health Perspect.101:37884

    4. McAllister EJ, Dhurandhar NV, Keith SW, Aronne LJ, Barger J, et al. 2009. Ten putative contributorsto the obesity epidemic. Crit. Rev. Food Sci. Nutr.49:868913

    5. Baillie-Hamilton PF. 2002. Chemical toxins: a hypothesis to explain the global obesity epidemic.

    J. Altern. Complement Med.8:185926. Casals-Casas C, Feige JN, Desvergne B. 2008. Interference of pollutants with PPARs: Endocrine dis-

    ruption meets metabolism. Int. J. Obes.32(Suppl. 6):53617. ElobeidMA,Allison DB.2008.Putativeenvironmental-endocrinedisruptorsandobesity:a review.Curr.

    Opin. Endocrinol. Diabetes Obes.15:40388. Chen JQ, Brown TR, Russo J. 2009. Regulation of energy metabolism pathways by estrogens and

    estrogenicchemicals andpotentialimplicationsin obesityassociatedwith increasedexposureto endocrinedisruptors. Biochim. Biophys. Acta1793:112843

    9. Hatch EE, Nelson JW, Stahlhut RW, Webster TF. 2010. Association of endocrine disruptors andobesity: perspectives from epidemiological studies.Int. J. Androl.33:32432

    10. Gr un F, Blumberg B. 2009. Endocrine disrupters as obesogens.Mol. Cell. Endocrinol.304:192911. Gr un F, Blumberg B. 2006. Environmental obesogens: organotins and endocrine disruption via nuclear

    receptor signaling. Endocrinology147:S505512. Ben-Jonathan N, Hugo ER, Brandebourg TD. 2009. Effects of bisphenol A on adipokine release from

    human adipose tissue: implications for the metabolic syndrome.Mol. Cell. Endocrinol.304:495413. CostaLG,GiordanoG, Tagliaferri S,Caglieri A, MuttiA. 2008. Polybrominateddiphenyl ether (PBDE)

    ameretardants: environmental contamination,humanbodyburden and potentialadverse health effects. Acta Biomed.79:17283

    14. Jensen AA, LeffersH. 2008.Emerging endocrinedisrupters: peruoroalkylated substances. Int. J. Androl.31:16169

    www.annualreviews.org Pollutants and Metabolic Disruption 155

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    22/31

    15. Kovarova J, Svobodova Z. 2008. Peruorinated compounds: occurrence and risk prole.Neuro. Endocrinol. Lett.29:599608

    16. McKinlay R, Plant JA, Bell JN, Voulvoulis N. 2008. Endocrine disrupting pesticides: implications risk assessment.Environ. Int. 34:16883

    17. Schecter A, Birnbaum L, Ryan JJ, Constable JD. 2006. Dioxins: an overview.Environ. Res.101:4192818. U.S. Environ. Prot. Agency. 2007. What is a pesticide?http://www.epa.gov/pesticides/about/ . Re

    trieved on September 15, 200719. Salehi F, Turner MC, Phillips KP, Wigle DT, Krewski D, Aronson KJ. 2008. Review of the etiolog

    of breast cancer with special attention to organochlorines as potential endocrine disruptors.J. Toxico Environ. Health B11:276300

    20. Porta M, Lee DH, Puigdomenech E. 2009. Transgenerational inheritance of environmental obesogensOccup. Environ. Med.66:14142

    21. Karmaus W, Osuch JR, Eneli I, Mudd LM, Zhang J, et al. 2009. Maternal levels of dichlorodiphendichloroethylene (DDE) may increase weight and body mass index in adult female offspring.Occu Environ. Med.66:14349

    22. Smink A, Ribas-Fito N, Garcia R, Torrent M, Mendez MA, et al. 2008. Exposure to hexachlorobenzeduring pregnancy increases the risk of overweight in children aged 6 years.Acta Paediatr.97:146569

    23. Lee DH, Lee IK, Porta M, Steffes M, Jacobs DR Jr. 2007. Relationship between serum concentratioof persistent organic pollutants and the prevalence of metabolic syndrome among nondiabetic adulresults fromtheNationalHealth andNutritionExamination Survey 19992002. Diabetologia50:18415

    24. Park SK,Son HK,Lee SK,Kang JH, Chang YS, et al. 2010. Relationshipbetween serum concentrationof organochlorine pesticides and metabolic syndrome among non-diabetic adults.J. Prev. Med. Publ Health 43:18

    25. Turyk M, Anderson H, Knobeloch L, Imm P, Persky V. 2009. Organochlorine exposure and incidencof diabetes in a cohort of Great Lakes sport sh consumers.Environ. Health Perspect.117:107682

    26. Pelclova D, Urban P,Preiss J, Lukas E, Fenclova Z,et al. 2006. Adversehealth effects in humans exposeto 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Rev. Environ. Health21:11938

    27. Lee DH, Lee IK, Steffes M, Jacobs DR Jr. 2007. Extended analyses of the association between serconcentrations of persistent organic pollutants and diabetes.Diabetes Care30:159698

    28. Gr un F, Watanabe H, Zamanian Z, Maeda L, Arima K, et al. 2006. Endocrine-disrupting organoticompounds are potent inducers of adipogenesis in vertebrates.Mol. Endocrinol.20:214155

    29. Omura M, Ogata R, Kubo K, Shimasaki Y, Aou S, et al. 2001. Two-generation reproductive toxicstudy of tributyltin chloride in male rats.Toxicol. Sci.64:2243230. Ogata R, Omura M, Shimasaki Y, Kubo K, Oshima Y, et al. 2001. Two-generation reproductive toxic

    study of tributyltin chloride in female rats.J. Toxicol. Environ. Health A63:1274431. Kanayama T, Kobayashi N, MamiyaS, NakanishiT, NishikawaJ. 2005. Organotincompoundspromote

    adipocyte differentiation as agonists of the peroxisome proliferator-activated receptor gamma/retinoi X receptor pathway. Mol. Pharmacol.67:76674

    32. Halden RU. 2010. Plastics and health risks.Annu. Rev. Public Health31:1799433. Fuentes S, Colomina MT, Rodriguez J, Vicens P, Domingo JL. 2006. Interactions in development

    toxicology: concurrentexposureto peruorooctanesulfonate(PFOS)andstress inpregnantmice. Toxico Lett. 164:8189

    34. Lau C, Butenhoff JL, Rogers JM. 2004. The developmental toxicity of peruoroalkyl acids and th

    derivatives.Toxicol. Appl. Pharmacol.198:2314135. Luebker DJ, York RG, Hansen KJ, Moore JA, Butenhoff JL. 2005. Neonatal mortality from in uter

    exposure to peruorooctanesulfonate (PFOS) in Sprague-Dawley rats: dose-response, and biochemicand pharamacokinetic parameters. Toxicology215:14969

    36. Shi Z, Zhang H, Liu Y, Xu M, Dai J. 2007. Alterations in gene expression and testosterone synthesisthe testes of male rats exposed to peruorododecanoic acid.Toxicol. Sci.98:20615

    37. Thibodeaux JR, Hanson RG, Rogers JM, Grey BE, Barbee BD, et al. 2003. Exposure to peruorooctasulfonateduring pregnancyin ratandmouse. I. Maternal andprenatalevaluations. Toxicol. Sci.74:3698

    156 Casals-Casas Desvergne

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    23/31

    38. Apelberg BJ,Witter FR,Herbstman JB,CalafatAM, Halden RU,et al.2007. Cord serumconcentrationsof peruorooctane sulfonate (PFOS) and peruorooctanoate (PFOA) in relation to weight and size atbirth. Environ. Health Perspect.115:167076

    39. Gilliland FD, Mandel JS. 1996. Serum peruorooctanoic acid and hepatic enzymes, lipoproteins, andcholesterol: a study of occupationally exposed men.Am. J. Ind. Med.29:56068

    40. Nelson JW, Hatch EE, Webster TF. 2010. Exposure to polyuoroalkyl chemicals and cholesterol, body weight, and insulin resistance in the general U.S. population.Environ. Health Perspect.118:197202

    41. Costa G, Sartori S, Consonni D. 2009. Thirty years of medical surveillance in peruooctanoic acid

    production workers. J. Occup. Environ. Med.51:3647242. MacNeil J, Steenland NK, Shankar A, Ducatman A. 2009. A cross-sectional analysis of type II diabetes

    in a community with exposure to peruorooctanoic acid (PFOA).Environ. Res.109:997100343. Olsen GW, Zobel LR. 2007. Assessment of lipid, hepatic, and thyroid parameters with serum peru-

    orooctanoate (PFOA) concentrations in uorochemical production workers. Int. Arch. Occup. Environ. Health 81:23146

    44. LinCY,LinLY,ChiangCK,WangWJ,SuYN,etal.2010.Investigationoftheassociationsbetweenlow-dose serumperuorinatedchemicalsandliverenzymes inUSadults. Am.J. Gastroenterol.105(6):135463

    45. Lin CY, Chen PC, Lin YC, Lin LY. 2009. Association among serum peruoroalkyl chemicals, glucosehomeostasis, and metabolic syndrome in adolescents and adults.Diabetes Care32:7027

    46. Sjodin A, Hagmar L, Klasson-Wehler E, Kronholm-Diab K, Jakobsson E, Bergman A. 1999. Flameretardant exposure: polybrominated diphenyl ethers in blood from Swedish workers.Environ. Health

    Perspect.107:6434847. JakobssonK, ThuressonK, Rylander L, SjodinA, HagmarL, Bergman A. 2002. Exposure to polybromi-

    nated diphenyl ethers and tetrabromobisphenol A among computer technicians.Chemosphere46:7091648. Hamers T, Kamstra JH, Sonneveld E, Murk AJ, Kester MH, et al. 2006. In vitro proling of the

    endocrine-disrupting potency of brominated ame retardants. Toxicol. Sci.92:1577349. Darnerud PO. 2003. Toxic effects of brominated ame retardants in man and in wildlife.Environ. Int.

    29:8415350. Main KM, Kiviranta H, Virtanen HE, Sundqvist E, Tuomisto JT, et al. 2007. Flame retardants in

    placenta and breast milk and cryptorchidism in newborn boys.Environ. Health Perspect.115:15192651. Lim JS, Lee DH, Jacobs DR Jr. 2008. Association of brominated ame retardants with diabetes and

    metabolic syndrome in the U.S. population, 20032004.Diabetes Care31:1802752. Soares A, Guieysse B, Jefferson B, Cartmell E, Lester JN. 2008. Nonylphenol in the environment: a

    critical review on occurrence, fate, toxicity and treatment in wastewaters.Environ. Int. 34:10334953. Soto AM, Justicia H, Wray JW, Sonnenschein C. 1991.p-Nonyl-phenol: an estrogenic xenobiotic re-

    leased from modied polystyrene.Environ. Health Perspect.92:1677354. Lee HJ, Chattopadhyay S, Gong EY, Ahn RS, Lee K. 2003. Antiandrogenic effects of bisphenol A and

    nonylphenol on the function of androgen receptor. Toxicol. Sci.75:404655. Bonefeld-Jorgensen EC, Long M, Hofmeister MV, Vinggaard AM. 2007. Endocrine-disrupting poten-

    tial of bisphenolA, bisphenolA dimethacrylate,4-n-nonylphenol,and 4- n-octylphenol in vitro:newdataand a brief review.Environ. Health Perspect.115(Suppl. 1):6976

    56. Lopez-Espinosa MJ,FreireC, Arrebola JP,Navea N, Taouki J, et al. 2009. Nonylphenol andoctylphe-nol in adipose tissue of women in Southern Spain.Chemosphere76:84752

    57. Calafat AM, Kuklenyik Z, Reidy JA, Caudill SP, Ekong J, Needham LL. 2005. Urinary concentrationsof bisphenolA and4-nonylphenol in a human referencepopulation. Environ. Health Perspect.113:39195

    58. Dodds EC. 1936. The pharmacological action and clinical use of drugs with a camphor- and coramine-like action. Proc. R. Soc. Med.29:65557

    59. Richter CA,Birnbaum LS,Farabollini F, Newbold RR,Rubin BS,et al. 2007. In vivo effects of bisphenol A in laboratory rodent studies.Reprod. Toxicol.24:199224

    60. Lang IA,GallowayTS, ScarlettA, HenleyWE, DepledgeM, etal.2008.Associationof urinary bisphenol A concentration with medical disorders and laboratory abnormalities in adults.JAMA 300:130310

    61. Sugiura-Ogasawara M, Ozaki Y, Sonta S, Makino T, Suzumori K. 2005. Exposure to bisphenol A isassociated with recurrent miscarriage.Hum. Reprod.20:232529

    www.annualreviews.org Pollutants and Metabolic Disruption 157

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    24/31

    62. Takeuchi T, Tsutsumi O, IkezukiY, Takai Y, Taketani Y. 2004. Positive relationship between androgenandthe endocrinedisruptor, bisphenolA, innormalwomenand womenwith ovarian dysfunction. Endoc J. 51:16569

    63. Frederiksen H, Skakkebaek N, Andersson A. 2007. Metabolism of phthalates in humans.Mol. Nutr. Foo Res.51:899911

    64. Arcadi F, Costa C, Imperatore C, Marchese A, Rapisarda A, et al. 1998. Oral toxicity of bis(2-ethylhephthalate during pregnancy and suckling in the Long-Evans rat.Food Chem. Toxicol.36:96370

    65. Li L, Jester WJ, Orth J. 1998. Effects of relatively low levels of mono-(2-ethylhexyl) phthalate

    cocultured Sertoli cells and gonocytes from neonatal rats.Toxicol. Appl. Pharmacol.153:2586566. FosterP.2006. Disruption of reproductivedevelopmentin male ratoffspring followingin utero exposure

    to phthalate esters. Int. J. Androl.29:1404767. Meeker J, Sathyanarayana S, Swan S. 2009. Phthalates and other additives in plastics: human expo

    and associated health outcomes.Philos. Trans. R. Soc. Lond. Ser. B364:209711368. Swan S, Main K, Liu F, Stewart S, Kruse R, et al. 2005. Decrease in anogenital distance among m

    infants with prenatal phthalate exposure.Environ. Health Perspect.113:10566169. Sharpe R. 2005. Phthalate exposure during pregnancy and lower anogenital index in boys: wider im

    cations for the general population? Environ. Health Perspect.113:A504570. Bornehag C, Sundell J, Weschler C, Sigsgaard T, Lundgren B, et al. 2004. The association betwee

    asthma and allergic symptoms in children and phthalates in house dust: a nested case-control stud Environ. Health Perspect.112:139397

    71. Gayathri N, Dhanya C, Indu A, Kurup P. 2004. Changes in some hormones by low doses of di (2-ethhexyl) phthalate (DEHP), a commonly used plasticizer in PVC blood storage bags & medical tubin Indian J. Med. Res.119:13944

    72. Heudorf U, Mersch-Sundermann V, Angerer J. 2007. Phthalates: toxicology and exposure.Int. J. Hyg Environ. Health210:62334

    73. Stahlhut R, van Wijngaarden E, Dye T, Cook S, Swan S. 2007. Concentrations of urinary phthalametabolites are associated with increased waist circumference and insulin resistance in adult U.S. ma Environ. Health Perspect.115:87682

    74. Tchernof A, Calles-Escandon J, Sites CK, Poehlman ET. 1998. Menopause, central body fatness, aninsulin resistance: effects of hormone-replacement therapy.Coron. Artery Dis.9:50311

    75. Heine PA, Taylor JA, Iwamoto GA, Lubahn DB, Cooke PS. 2000. Increased adipose tissue in male afemale estrogen receptor- knockout mice. Proc. Natl. Acad. Sci. USA97:1272934

    76. Brown LM, Gent L, Davis K, Clegg DJ. 2010. Metabolic impact of sex hormones on obesity.Brain Re1350:7785

    77. Cooke PS, Naaz A. 2004. Role of estrogens in adipocyte development and function.Exp. Biol. Me229:112735

    78. Wada K, Sakamoto H, Nishikawa K, Sakuma S, Nakajima A, et al. 2007. Lifestyle-related diseases ofdigestive system: Endocrine disruptors stimulate lipid accumulation in target cells related to metabolsyndrome. J. Pharmacol. Sci.105:13337

    79. Phrakonkham P, Viengchareun S, Belloir C, Lombes M, Artur Y, Canivenc-Lavier MC. 2008. Dieta xenoestrogens differentially impair 3T3-L1 preadipocyte differentiation and persistently affect leptsynthesis. J. Steroid Biochem. Mol. Biol.110:95103

    80. Lee MJ, Lin H, Liu CW, Wu MH, Liao WJ, et al. 2008.Octylphenol stimulates resistin gene expressionin 3T3-L1 adipocytes via the estrogen receptor and extracellular signal-regulated kinase pathways.Am

    J. Physiol. Cell Physiol.294:15425181. Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, et al. 2001. The hormone resistin links obe

    to diabetes. Nature 409:3071282. Alonso-Magdalena P, Ropero AB, Carrera MP, Cederroth CR, Baquie M, et al. 2008. Pancreatic insul

    content regulation by the estrogen receptor ER . PLoS ONE 3:e206983. Martensson UE, Salehi SA, Windahl S, Gomez MF, Sward K, et al. 2009. Deletion of the G protein

    coupled receptor 30 impairs glucose tolerance, reduces bone growth, increases blood pressure, aneliminates estradiol-stimulated insulin release in female mice.Endocrinology150:68798

    158 Casals-Casas Desvergne

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    25/31

    84. Thomas P, Dong J. 2006. Binding and activation of the seven-transmembrane estrogen receptor GPR30by environmental estrogens: a potential novel mechanism of endocrine disruption.J. Steroid Biochem. Mol. Biol.102:17579

    85. Rubin MM. 2007. Antenatal exposure to DES: lessons learned. . . future concerns. Obstet. Gynecol. Surv.62:54855

    86. Newbold RR, Padilla-Banks E, Snyder RJ, Jefferson WN. 2007. Perinatal exposure to environmentalestrogens and the development of obesity. Mol. Nutr. Food Res.51:91217

    87. Rubin BS, Soto AM. 2009. Bisphenol A: perinatal exposure and body weight.Mol. Cell. Endocrinol.304:5562

    88. Somm E, Schwitzgebel VM, Toulotte A, Cederroth CR, Combescure C, et al. 2009. Perinatal exposureto bisphenol A alters early adipogenesis in the rat.Environ. Health Perspect.117:154955

    89. Miyawaki J, Sakayama K, Kato H, Yamamoto H, Masuno H. 2007. Perinatal and postnatal exposure tobisphenol A increases adipose tissue mass and serum cholesterol level in mice.J. Atheroscler. Thromb.14:24552

    90. Heindel JJ, vom Saal FS. 2009. Role of nutrition and environmental endocrine disrupting chemicalsduring the perinatal period on the aetiology of obesity. Mol. Cell. Endocrinol.304:9096

    91. SargisRM,JohnsonDN,ChoudhuryRA, BradyMJ.2010.Environmentalendocrinedisruptors promoteadipogenesis in the 3T3-L1 cell line through glucocorticoid receptor activation.Obesity18:128388

    92. Moriyama K, Tagami T, Akamizu T, Usui T, Saijo M, et al. 2002. Thyroid hormone action is disruptedby bisphenol A as an antagonist.J. Clin. Endocrinol. Metab.87:518590

    93. Zoeller RT, Bansal R, Parris C. 2005. Bisphenol-A, an environmental contaminant that acts as a thyroidhormone receptor antagonist in vitro, increases serumthyroxine,and alters RC3/neurograninexpressionin the developing rat brain. Endocrinology146:60712

    94. Legler J. 2008. New insights into the endocrine disrupting effects of brominated ame retardants.Chemosphere73:21622

    95. Hoppe AA, Carey GB. 2007. Polybrominated diphenyl ethers as endocrine disruptors of adipocytemetabolism. Obesity15:294250

    96. Moreau A, Vilarem MJ, Maurel P, Pascussi JM. 2008. Xenoreceptors CAR and PXR activation andconsequences on lipid metabolism, glucose homeostasis, and inammatory response.Mol. Pharm.5:3541

    97. Wada T, Gao J, Xie W. 2009. PXR and CAR in energy metabolism.Trends Endocrinol. Metab.20:27379

    98. Kretschmer XC, Baldwin WS. 2005. CAR and PXR: xenosensors of endocrine disrupters?Chem. Biol. Interact.155:1112899. Ren H, Aleksunes LM, Wood C, Vallanat B, George MH, et al. 2010. Characterization of peroxisome

    proliferator-activated receptor independent effects of PPAR activators in the rodent liver: Di-(2-ethylhexyl) phthalate also activates the constitutive-activated receptor.Toxicol. Sci.113:4559

    100. Eveillard A, Mselli-Lakhal L, Mogha A, Lasserre F, Polizzi A, et al. 2009. Di-(2-ethylhexyl)-phthalate(DEHP) activates the constitutive androstane receptor (CAR): a novel signaling pathway sensitive tophthalates. Biochem. Pharmacol.77:173546

    101. Eveillard A, Lasserre F, de Tayrac M, Polizzi A, Claus S, et al. 2009. Identication of potential mech-anisms of toxicity after di-(2-ethylhexyl)-phthalate (DEHP) adult exposure in the liver using a systemsbiology approach. Toxicol. Appl. Pharmacol.236:28292

    102. Hernandez JP, Huang W, Chapman LM, Chua S, Moore DD, Baldwin WS. 2007. The environmental

    estrogen, nonylphenol, activates the constitutive androstane receptor. Toxicol. Sci.98:41626103. MorereP, NouvetG, Stain JP,Paillot B, Metayer J, Hemet J. 1975. Information obtainedby liver biopsy

    in 100 tuberculous patients. Sem. Hop.51:2095102 (In French)104. CalandreEP,Rodriquez-LopezC,BlazquezA,CanoD.1991.Serum lipids, lipoproteins andapolipopro-

    teinsA andB inepilepticpatientstreatedwith valproicacid,carbamazepineor phenobarbital. Acta Neurol.Scand.83:25053

    105. Lahtela JT,ArrantoAJ,SotaniemiEA.1985. Enzymeinducers improve insulin sensitivity in noninsulin-dependent diabetic subjects. Diabetes 34:91116

    www.annualreviews.org Pollutants and Metabolic Disruption 159

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    26/31

    106. Piriou A, Warnet JM, Jacqueson A, Claude JR, Truhaut R. 1979. Fatty liver induced by high doses rifampicin in the rat: possible relation with an inhibition of RNA polymerases in eukariotic cells.ArchToxicol. Suppl.1979:33337

    107. Nakamura K, Moore R, Negishi M, Sueyoshi T. 2007. Nuclear pregnane X receptor cross-talk wiFoxA2 to mediate drug-induced regulation of lipid metabolism in fasting mouse liver.J. Biol. Chem282:976876

    108. Zhou J, Zhai Y, Mu Y, Gong H, Uppal H, et al. 2006. A novel pregnane X receptor-mediated and sterregulatory element-binding protein-independent lipogenic pathway. J. Biol. Chem.281:1501320

    109. Argaud D, Halimi S, Catelloni F, Leverve XM. 1991. Inhibition of gluconeogenesis in isolated rhepatocytes after chronic treatment with phenobarbital. Biochem. J.280(Pt. 3):66369

    110. Kodama S, Koike C, Negishi M, Yamamoto Y. 2004. Nuclear receptors CAR and PXR cross talk wFOXO1 to regulate genes that encode drug-metabolizing and gluconeogenic enzymes.Mol. Cell. Bi24:793140

    111. Bhalla S, Ozalp C, Fang S, Xiang L, Kemper JK. 2004. Ligand-activated pregnane X receptor interfe with HNF-4 signaling by targeting a common coactivator PGC-1 . Functional implications in hepaticholesterol and glucose metabolism.J. Biol. Chem.279:4513947

    112. Gao J, He J, Zhai Y, Wada T, Xie W. 2009. The constitutive androstane receptor is an antiobesitynuclear receptor that improves insulin sensitivity.J. Biol. Chem.284:2598492

    113. Dong B, Saha PK, Huang W, Chen W, Abu-Elheiga LA, et al. 2009. Activation of nuclear receptor CARameliorates diabetes and fatty liver disease.Proc. Natl. Acad. Sci. USA106:1883136

    114. Pascussi JM,Gerbal-ChaloinS, DuretC, Daujat-Chavanieu M,Vilarem MJ,MaurelP. 2008. Thetangleof nuclear receptors that controls xenobiotic metabolism and transport: crosstalk and consequence Annu. Rev. Pharmacol. Toxicol.48:132

    115. Deleted in proof 116. Bock KW, Kohle C. 2006. Ah receptor: dioxin-mediated toxic responses as hints to deregulated phy

    ologic functions. Biochem. Pharmacol.72:393404117. Kohle C, Bock KW. 2009. Coordinate regulation of human drug-metabolizing enzymes, and conjuga

    transporters by the Ah receptor, pregnane X receptor and constitutive androstane receptor. Biochem Pharmacol.77:68999

    118. Barnes-Ellerbe S, Knudsen KE, Puga A. 2004. 2,3,7,8-Tetrachlorodibenzo- p-dioxin blocks androgendependent cell proliferation of LNCaP cells through modulation of pRB phosphorylation. Mol. Pharmacol.66:50211

    119. Sato S, Shirakawa H, Tomita S, Ohsaki Y, Haketa K, et al. 2008. Low-dose dioxins alter gene expressrelated to cholesterol biosynthesis, lipogenesis, and glucose metabolism through the aryl hydrocarboreceptor-mediated pathway in mouse liver. Toxicol. Appl. Pharmacol.229:1019

    120. Arsenescu V, Arsenescu RI, King V, Swanson H, Cassis LA. 2008. Polychlorinated biphenyl-77 induadipocyte differentiation and proinammatory adipokines and promotes obesity and atherosclerosi Environ. Health Perspect.116:76168

    121. Matthews J, Gustafsson JA. 2006. Estrogen receptor and aryl hydrocarbon receptor signaling pathwa Nucl. Recept. Signal.4:e016

    122. Swedenborg E, Ruegg J, Makela S, Pongratz I. 2009. Endocrine disruptive chemicals: mechanismsaction and involvement in metabolic disorders.J. Mol. Endocrinol.43:110

    123. Cimafranca MA, Hanlon PR, Jefcoate CR. 2004. TCDD administration after the proadipogenic diferentiation stimulus inhibits PPAR through a MEK-dependent process but less effectively suppress

    adipogenesis.Toxicol. Appl. Pharmacol.196:15668124. Feige JN, Gelman L, Michalik L, Desvergne B, Wahli W. 2006. From molecular action to physiologic

    outputs:Peroxisome proliferator-activated receptorsarenuclear receptorsat thecrossroadsof keycellulafunctions. Prog. Lipid. Res.45:12059

    125. Michalik L, Wahli W. 2007. Peroxisome proliferator-activated receptors (PPARs) in skin health, repaand disease.Biochim. Biophys. Acta1771:99198

    126. Barish GD, Narkar VA, Evans RM. 2006. PPAR : a dagger in the heart of the metabolic syndrom J. Clin. Investig.116:59097

    160 Casals-Casas Desvergne

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    27/31

    127. Auwerx J. 1999. PPAR , the ultimate thrifty gene. Diabetologia42:103349128. Takeuchi S, MatsudaT, KobayashiS, Takahashi T, KojimaH. 2006. In vitro screening of 200 pesticides

    for agonistic activity via mouse peroxisome proliferator-activated receptor (PPAR) and PPAR andquantitative analysis of in vivo induction pathway.Toxicol. Appl. Pharmacol.217:23544

    129. Palut D, Ludwicki JK, Kostka G, Kopec-Szlezak J, Wiadrowska B, Lembowicz K. 2001. Studies of early hepatocellular proliferation and peroxisomal proliferation in Wistar rats treated with herbicide diclofop.Toxicology158:11926

    130. WardJM,Peters JM,PerellaCM,GonzalezFJ. 1998.Receptorand nonreceptor-mediated organ-specic

    toxicity of di(2-ethylhexyl)phthalate (DEHP) in peroxisome proliferator-activated receptor -null mice.Toxicol. Pathol.26:24046

    131. BilityMT, ThompsonJT,McKeeRH, DavidRM,Butala JH,etal.2004.Activation ofmouseandhumanperoxisome proliferator-activated receptors (PPARs) by phthalate monoesters.Toxicol. Sci.82:17082

    132. Hurst CH, Waxman DJ. 2003. Activation of PPAR and PPAR by environmental phthalate mo-noesters. Toxicol. Sci.74:297308

    133. Feige JN, Gelman L, Rossi D, Zoete V, Metivier R, et al. 2007. The endocrine disruptor monoethyl-hexyl-phthalate is a selective peroxisome proliferator-activated receptor modulator that promotesadipogenesis.J. Biol. Chem.282:1915266

    134. LapinskasPJ,BrownS, Leesnitzer LM, BlanchardS, Swanson C,etal.2005.Role ofPPAR inmediatingthe effects of phthalates and metabolites in the liver.Toxicology207:14963

    135. Zoete V, Grosdidier A, Michielin O. 2007. Peroxisome proliferator-activated receptor structures: ligand

    specicity, molecular switch and interactions with regulators.Biochim. Biophys. Acta1771:91525136. Feige JN, Gerber A, Casals-Casas C, Yang Q, Winkler C, et al. 2010. The pollutant diethylhexyl

    phthalate regulates hepatic energy metabolism via species-specic PPAR -dependent mechanisms. En-viron. Health Perspect.118:23441

    137. Inagaki T, Dutchak P, Zhao G, Ding X, Gautron L, et al. 2007. Endocrine regulation of the fastingresponse by PPAR -mediated induction of broblast growth factor 21.Cell Metab.5:41525

    138. Boberg J, Metzdorff S, Wortziger R, Axelstad M, Brokken L, et al. 2008. Impact of diisobutyl phthalateand other PPAR agonists on steroidogenesis and plasma insulin and leptin levels in fetal rats.Toxicology250:7581

    139. Takacs ML, Abbott BD. 2007. Activation of mouse and human peroxisome proliferator-activated recep-tors ( , / , ) by peruorooctanoic acid and peruorooctane sulfonate.Toxicol. Sci.95:10817

    140. XieY, Yang Q, NelsonBD, DePierre JW.2003. Therelationship between liver peroxisome proliferation

    andadipose tissueatrophy induced byperoxisome proliferatorexposure andwithdrawal in mice.Biochem. Pharmacol.66:74956

    141. Asakawa A,ToyoshimaM, HaradaKH, Fujimiya M,InoueK,KoizumiA.2008. Theubiquitousenviron-mental pollutant peruorooctanoic acid inhibits feeding behavior via peroxisome proliferator-activatedreceptor- . Int. J. Mol. Med.21:43945

    142. Hines EP, White SS, Stanko JP, Gibbs-Flournoy EA, Lau C, Fenton SE. 2009. Phenotypic dichotomy following developmental exposure to peruorooctanoic acid (PFOA) in female CD-1 mice: Low dosesinduce elevated serum leptin and insulin, and overweight in mid-life.Mol. Cell. Endocrinol.304:97105

    143. Nakamura T, Ito Y, Yanagiba Y, Ramdhan DH, Kono Y, et al. 2009. Microgram-order ammoniumperuorooctanoate may activate mouse peroxisome proliferator-activated receptor , but not humanPPAR . Toxicology265:2733

    144. le Maire A, Grimaldi M, Roecklin D, Dagnino S, Vivat-HannahV, et al. 2009. Activation of RXR-PPAR

    heterodimers by organotin environmental endocrine disruptors. EMBO Rep.10:36773145. Peters AK, Nijmeijer S, Gradin K, Backlund M, Bergman A, et al. 2006. Interactions of polybrominated

    diphenyl ethers with the aryl hydrocarbon receptor pathway. Toxicol. Sci.92:13342146. Sanders JM, Burka LT, Smith CS, Black W, James R, Cunningham ML. 2005. Differential expression

    of CYP1A, 2B, and 3A genes in the F344 rat following exposure to a polybrominated diphenyl ethermixture or individual components. Toxicol. Sci.88:12733

    147. Pacyniak EK, Cheng X, Cunningham ML, Crofton K, Klaassen CD, Guo GL. 2007. The ame retar-dants, polybrominated diphenyl ethers, are pregnane X receptor activators.Toxicol. Sci.97:94102

    www.annualreviews.org Pollutants and Metabolic Disruption 161

  • 8/6/2019 Distruptores Metabolicos (Articulo Referencial Para Examen)

    28/31

    148. DarnerudPO.2008.Brominated ameretardantsas possibleendocrinedisrupters. Int. J. Androl.31:15260

    149. Costa LG, Giordano G. 2007. Developmental neurotoxicity of polybrominated diphenyl ether (PBDEame retardants. Neurotoxicology28:104767

    150. Soto AM, Rubin BS, Sonnenschein C. 2009. Interpreting endocrine disruption from an integrativbiology perspective.Mol. Cell. Endocrinol.304:37

    151. Vanderberg JP. 2009. Reections on early malaria vaccine studies, the rst successful human mala vaccination, and beyond.Vaccine27:29

    152. vom Saal FS, Akingbemi BT, Belcher SM, Birnbaum LS, Crain DA, et al. 2007. Chapel Hill bisphenoexpert panel consensus statement: integration of mechanisms, effects in animals and potential to impahuman health at current levels of exposure.Reprod. Toxicol.24:13138

    153. Crews D, McLachlan JA. 2006. Epigenetics, evolution, endocrine disruption, health, and diseas Endocrinology147:S410

    154. Vineis P, Khan AE, Vlaanderen J, Vermeulen R. 2009. The impact of new research technologies on ounderstanding of environmental causes of disease: the concept of clinical vulnerability.Environ. Healt8:54

    155. Lin HK, Altuwaijri S, Lin WJ, Kan PY, Collins LL, Chang C. 2002. Proteasome activity is required androgen receptor transcriptional activity via regulation of androgen receptor nuclear translocation aninteraction with coregulators in prostate cancer cells.J. Biol. Chem.277:3657076

    156. Hugo ER, Brandebourg TD, Woo JG, Loftus J, Alexander JW, Ben-Jonathan N. 2008. Bisphenol

    at environmentally relevant doses inhibits adiponectin release from human adipose tissue explants aadipocytes. Environ. Health Perspect.116:164247

    157. Moreno-AliagaMJ,MatsumuraF.2002.Effectsof 1,1,1-trichloro-2,2-bis( p-chlorophenyl)-ethane( p, pDDT) on 3T3-L1 and 3T3-F442A adipocyte differentiation. B