• No results found

Bisphenol A exposure increases liver fat in juvenile fructose-fed Fischer 344 rats

N/A
N/A
Protected

Academic year: 2021

Share "Bisphenol A exposure increases liver fat in juvenile fructose-fed Fischer 344 rats"

Copied!
9
0
0

Loading.... (view fulltext now)

Full text

(1)

Bisphenol A exposure increases liver fat in

juvenile fructose-fed Fischer 344 rats

Monika Rönn, Joel Kullberg, Helen Karlsson, Johan Berglund, Filio Malmberg, Jan Örberg,

Lars Lind, Håkan Ahlström and Monica P Lind

Linköping University Post Print

N.B.: When citing this work, cite the original article.

Original Publication:

Monika Rönn, Joel Kullberg, Helen Karlsson, Johan Berglund, Filio Malmberg, Jan Örberg,

Lars Lind, Håkan Ahlström and Monica P Lind, Bisphenol A exposure increases liver fat in

juvenile fructose-fed Fischer 344 rats, 2013, Toxicology, (303), 125-132.

http://dx.doi.org/10.1016/j.tox.2012.09.013

Copyright: Elsevier

http://www.elsevier.com/

Postprint available at: Linköping University Electronic Press

(2)

Contents

lists

available

at

SciVerse

ScienceDirect

Toxicology

j o u

r n

a l

h o m

e p a g e :

w w w . e l s e v i e r . c o m / l o c a t e / t o x i c o l

Bisphenol

A

exposure

increases

liver

fat

in

juvenile

fructose-fed

Fischer

344

rats

Monika

Rönn

a

, Joel

Kullberg

b

, Helen

Karlsson

c

, Johan

Berglund

b

,

Filip

Malmberg

d

,

Jan

Örberg

e

,

Lars

Lind

f

,

Håkan

Ahlström

b

,

P.

Monica

Lind

a

,

aOccupationalandEnvironmentalMedicine,UppsalaUniversity,Uppsala,Sweden

bDepartmentofRadiology,Oncology,andRadiationScience,UppsalaUniversity,Uppsala,Sweden

cOccupationalandEnvironmentalMedicine,CountyCouncilofÖstergötland,LinköpingUniversity,Linköping,Sweden dCenterforImageAnalysis,UppsalaUniversity,Uppsala,Sweden

eDepartmentofOrganismalBiology,EnvironmentalToxicology,UppsalaUniversity,Uppsala,Sweden fDepartmentofMedicalSciences,UppsalaUniversity,Uppsala,Sweden

a

r

t

i

c

l

e

i

n

f

o

Articlehistory: Received2May2012 Receivedinrevisedform 18September2012 Accepted20September2012 Available online 8 November 2012 Keywords: MRI Liverfat Rat BisphenolA Obesity

a

b

s

t

r

a

c

t

Background:PrenatalexposuretobisphenolA(BPA)hasbeenshowntoinduceobesityinrodents.To eval-uateifexposurealsolaterinlifecouldinduceobesityorliverdamageweinvestigatedthesehypothesises inanexperimentalratmodel.

Methods:Fromfivetofifteenweeksofage,femaleFischer344ratswereexposedtoBPAviadrinkingwater (0.025,0.25or2.5mgBPA/L)containing5%fructose.Twocontrolgroupsweregiveneitherwateror5% fructosesolution.Individualweightoftheratswasdeterminedonceaweek.Atterminationmagnetic resonanceimagingwasusedtoassessadiposetissueamountanddistribution,andliverfatcontent.After sacrificetheleftperirenalfatpadandtheliverweredissectedandweighed.ApolipoproteinA-Iinplasma wasanalyzedbywesternblot.

Results:Nosignificanteffectsonbodyweightortheweightofthedissectedfadpadwereseeninrats exposedtoBPA,andMRIshowednodifferencesintotalorvisceraladiposetissuevolumesbetween thegroups.However,MRIshowedthatliverfatcontentwassignificantlyhigherinBPA-exposedrats thaninfructosecontrols(p=0.04).BPAexposurealsoincreasedtheapolipoproteinA-Ilevelsinplasma (p<0.0001).

Conclusion:WefoundnoevidencethatBPAexposureaffectsfatmassinjuvenilefructose-fedrats. How-ever,thefindingthatBPAincombinationwithfructoseinducedfatinfiltrationintheliveratdosages closetothecurrenttolerabledailyintake(TDI)mightbeofconcerngiventhewidespreaduseofthis compoundinourenvironment.

© 2012 Elsevier Ireland Ltd.

Abbreviations: apoA-I,apolipoprotinA-I;BMI,bodymassindex;BPA, bisphe-nolA;HDL,highdensitylipoproteins;IL-6,interleukin-6;LCAT,lecithin-cholesterol acyltransferase;LPS,lipopolysaccharide;LSI,liversomaticindex;LT,leantissue; MRI,magneticresonanceimaging;NOAEL,noadverseeffectlevel;PPAR-␥, per-oxisomeproliferatoractivatedreceptor-gamma;SAT,subcutaneousadiposetissue; SRBI,ScavengerReceptorClassB-I;TAT,totaladiposetissue;TDI,tolerabledaily intake;TNFalpha,tumornecrosisfactor-alpha;VAT,visceraladiposetissue;VLDL, verylowdensitylipoproteins.

∗ Correspondingauthorat:OccupationalandEnvironmentalMedicine,Uppsala University,75185Uppsala,Sweden.Tel.:+46186113642;fax:+46186114806.

E-mailaddresses:Monika.Ronn@medsci.uu.se(M.Rönn), Joel.Kullberg@radiol.uu.se(J.Kullberg),Helen.M.Karlsson@liu.se (H.Karlsson),Johan.Berglund@radiol.uu.se(J.Berglund), filip@cb.uu.se(F.Malmberg),Jan.Orberg@ebc.uu.se(J.Örberg),

Lars.Lind@medsci.uu.se(L.Lind),Hakan.Ahlstrom@radiol.uu.se(H.Ahlström), Monica.Lind@medsci.uu.se(P.M.Lind).

1.

Introduction

The

prevalence

of

obesity

(BMI

>

30)

has

risen

dramatically

in

the

world

over

the

past

two

decades.

In

2009–2010,

35.5%

of

adult

men

and

35.8%

of

adult

women

in

the

US

were

obese

(

Flegal

et

al.,

2012

).

Obesity

causes

negative

effects

on

quality

of

life

while

also

predisposing

individuals

to

a

number

of

diseases,

including

type

2

diabetes

and

cardiovascular

diseases.

Many

researchers

consider

obesity

mainly

as

an

unfavorable

balance

between

a

high

energy

intake

and

low

energy

expendi-ture

due

to

poor

diet

and

inadequate

exercise

habits.

However,

overweight

early

in

life

is

a

risk

factor

for

overweight

and

obesity

later

in

life,

and

paradoxically

underweight

is

another

risk

fac-tor

due

to

a

“catch

up”

phenomenon.

Obviously

there

exists

some

sort

of

programming

regarding

weight

development,

at

least

in

the

earliest

stages

of

life.

Recent

research

has

suggested

that

environ-mental

contaminants

could

play

an

important

role

in

modulating

the

balance

between

energy

intake

and

expenditure,

reviewed

in

0300-483X© 2012 Elsevier Ireland Ltd. http://dx.doi.org/10.1016/j.tox.2012.09.013

Open access under CC BY-NC-ND license.

(3)

126 M.Rönnetal./Toxicology303 (2013) 125–132

(

Janesick

and

Blumberg,

2011

).

In

a

study

on

mice

it

was

found

that

prenatal

exposure

to

tributyl

tin

(TBT)

caused

obesity

later

in

life

and

the

term

“obesogens”

was

coined

(

Grun

and

Blumberg,

2006

).

This

observation

supports

the

hypothesis

of

fetal

programming

in

humans

as

a

source

of

certain

disorders,

such

as

obesity

and

dia-betes,

emerging

many

years

later

(

Barker

et

al.,

2002

).

In

addition

to

fetal

programming,

exposure

to

certain

chemicals

in

adulthood

is

also

important.

Adult

rats

given

persistent

organic

pollutants

(POPs)

via

crude

salmon

oil

become

obese

(

Ruzzin

et

al.,

2010

),

and

pharmaceuticals,

such

as

the

antidiabetic

drug

rosiglitazone

(ROSI)

acting

on

the

important

receptor

peroxisome

proliferator-activated

receptor-gamma

(PPAR-

␥)

increase

body

fat

when

administered

to

adult

humans

(

Choi

et

al.,

2010

).

Moreover,

it

was

recently

shown

that

thiazide

antihypertensive

agents

induce

visceral

obesity

when

given

to

adult

hypertensive

patients

(

Eriksson

et

al.,

2008

).

Taken

together,

these

data

indicates

that

exposure

to

chemicals

not

only

in

utero

or

early

childhood

could

be

of

importance

for

the

develop-ment

of

obesity.

Bisphenol

A

(BPA)

was

discovered

to

be

an

artificial

estrogen

as

early

as

the

1930s

(

Dodds,

1936

),

but

the

synthesis

of

another

chemical,

diethylstilbestrol

(DES),

with

more

potent

estrogenic

properties

precluded

the

use

of

BPA

as

a

pharmaceutical

agent.

Today

its

main

applications

are

as

a

hardener

in

plastic

goods

and

as

a

monomer

for

production

of

polycarbonate

plastics.

As

such,

it

is

a

high-volume

chemical

and

circulating

levels

of

this

compound

were

measureable

in

about

98%

of

all

subjects

in

a

study

of

Swedish

elderly

persons

(

Olsen

et

al.,

2012

)

confirming

the

National

Health

and

Nutrition

Examination

Survey

(NHANES)

2007–2008

where

the

urinary

concentrations

were

measurable

in

94%

of

the

subjects

(<LOD

6.1%)

(

LaKind

et

al.,

2012

).

BPA

is

almost

completely

absorbed

in

the

gastrointestinal

tract

in

humans

and

is

highly

conjugated

to

form

the

major

metabo-lite

bisphenol

A

glucuronide

by

first

pass

metabolism

in

the

liver

(

Pottenger

et

al.,

2000

).

The

glucuronide,

which

is

not

estrogenically

active,

is

then

cleared

from

blood

by

elimination

with

urine.

In

rats

the

main

route

of

elimination

of

conjugated

BPA

is

by

biliary

and

fecal

elimination

which

enables

enterohepatic

recirculation

(

Völkel

et

al.,

2002

).

These

mechanisms

indicate

that

the

metabolism

of

BPA

is

faster

and

the

conjugation

more

efficient

in

humans,

where

enterohepatic

recirculation

is

negligible,

than

in

rats.

However,

strain

differences

has

been

reported,

and

in

female

Fischer

344

(F

344)

rats

the

excretion

via

urine

was

42%,

and

twice

as

high

as

in

CD

rats

(21%)

(

Snyder

et

al.,

2000

).

The

efficient

conjugation

and

relatively

low

BPA-exposure

are

the

main

reasons

why

BPA

is

con-sidered

to

be

safe

to

humans

despite

a

notable

amount

of

animal

studies

demonstrating

effects

on

various

outcomes

and

in

various

doses.

One

mechanism

to

further

evaluate

is

the

action

of

the

␤-glucoronidase

enzyme

present

within

many

tissues,

notably

e.g.

the

placenta

of

animals

and

humans.

␤-Glucoronidase

deconju-gates

BPA

to

its

active

form

which

may

lead

to

fetal

exposure

in

the

uterus

(

Ginsberg

and

Rice,

2009

).

There

has

been

a

focus

on

BPA

as

an

endocrine

disruptor

because

of

its

estrogenicity,

while

there

also

might

be

other

mechanisms

that

explain

the

effects

of

BPA

seen

in

various

studies.

Prenatal

exposure

to

BPA

in

rodents

has

previously

been

shown

to

induce

obesity

(

Miyawaki

et

al.,

2007;

Somm

et

al.,

2009;

Wei

et

al.,

2011

),

and

the

effect

of

exposure

to

BPA

later

in

life

has

recently

been

studied

by

e.g.

Marmugi

et

al.

(2012)

.

But

there

is

an

inconsistency

regarding

BPA

exposure

and

weight

gain

since

other

studies

show

no

significant

effects

despite

exposure

over

genera-tions

in

the

environmentally

relevant

doses

(

Ema

et

al.,

2001;

Tyl

et

al.,

2008,

2002

).

In

order

to

study

effects

of

BPA

in

doses

in

the

range

of

tolerable

daily

intake

(TDI)

we

have

used

three

exposure

levels,

the

medium

dose

being

close

to

TDI

as

established

by

the

U.S.

Environmental

Protection

Agency

(EPA)

and

the

European

Food

Safety

Authority

(EFSA)

at

50

␮g/kg

and

day.

The

low

dose

was

10

times

lower

and

the

high

dose

10

times

higher

than

the

medium

dose.

The

primary

aim

of

this

study

was

to

test

the

hypothesis

that

exposure

to

BPA

in

combination

with

carbohydrates

after

the

sen-sitive

prenatal

and

perinatal

periods

also

could

affect

fat

mass

or

liver

fat

content.

Since

exposure

to

BPA

only,

later

in

life

(

Marmugi

et

al.,

2012

)

and

perinatal

exposure

to

BPA

in

combination

with

high

fat

diet

later

in

life

(

Wei

et

al.,

2011

)

have

been

reported,

this

study

will

focus

on

exposure

to

BPA

in

combination

with

a

diet

supple-mented

with

carbohydrates.

As

fructose

is

a

widely

used

sweetener

in

processed

food

and

has

been

suggested

to

contribute

to

unfavor-able

metabolic

alterations

(

Bocarsly

et

al.,

2010;

Bremer

et

al.,

2012

)

juvenile

rats

were

exposed

to

BPA

in

combination

with

a

5%

fructose

solution,

which

is

about

the

same

fructose

concentration

as

in

com-mon

soft

drinks

(9–13%

sucrose).

Effects

on

adipose

tissue

volume

and

liver

fat

content

in

the

BPA-exposed

groups

were

evaluated

by

magnetic

resonance

imaging

(MRI)

and

compared

with

a

control

group

also

given

fructose

solution.

As

a

secondary

aim,

we

inves-tigated

whether

obesity

parameters

and

the

liver

were

affected

by

fructose

feeding

alone,

using

water-fed

rats

as

a

control

group.

2. Materialandmethods

2.1. Chemicals

Bisphenol A (BPA), (80-05-7, (CH3)2C(C6H4OH)2, ≥99% purity), fructose (C6H12O6,≥99%purity),Griessmodifiedreagent,ZnSO4,andVCl3werepurchased fromSigma–Aldrich,St.Louis,MO.NaNO3waspurchasedfromMerckchemicals, Darmstadt,Germany.

2.2. Animals

TheanimalstudywasapprovedbytheUppsalaAnimalEthicalCommittee andfollowedtheguidelines laiddownbytheSwedish LegislationonAnimal Experimentation(AnimalWelfareActSFS1998:56)andEuropeanUnionLegislation (ConventionETS123andDirective86/609/EEC).

SixtyfemaleF344ratsat3weeksofagewerepurchasedfromCharlesRiver International,Salzfeld,Germany,andhoused3rats/cageatUppsalaUniversity Hos-pitalanimalfacilityinatemperature-controlledandhumidity-controlledroom witha12-hlight/darkcycle.TominimizebackgroundBPAexposurePolysulfone IVcages(EurostandardIV)andglasswaterbottleswereused.Theratswerefeda standardpelletRM1diet(adlib.)fromNOVA-SCB,Sollentuna,Sweden.RM1isa naturalingredientdietwithalowlevelofphytoestrogens(100–200␮g/g)(Jensen andRitskes-Hoitinga,2007;Odumetal.,2001).Duringthetwo-week acclimatiza-tionperiodprecedingtheten-weekinterventionallanimalsweregivenwaterto drinkandduringtheinterventionwateror5%fructosesolution(seeSection2.3).At 5weeksofagetheratswereassignedtofivegroups(12rats/group);watercontrol (W),fructosecontrol(F),lowdoseBPA(0.025mg/L),mediumdoseBPA(0.25mg/L) orhighdoseBPA(2.5mg/L).Toavoidunnecessarystressnocage-mateswere sep-arated,butthecageswereallocatedtothedifferentgroupstoachieveequalityin weightsinallgroups.Foodandliquidconsumptionineachcageandindividual weightoftheratsweredeterminedonceaweek.

BeforeMRIexam,theratswereanesthetizedwithKetalar90mg/kgbw(Pfizer, NewYork,NY)andRompun10mg/kgbw(Bayer,Leverkusen,Germany). Immedi-atelyafterthescanningtheywerekilledbyexsanguinationsfromtheabdominal aortawhilestillunderanesthesia.

2.3. Exposure

ToprepareBPAexposuresolutions(0.025,0.25and2.5mg/L),threestock solu-tionsofBPAin1%ethanol(2.5mg/L,25mg/Land250mg/L)werediluted1:100in5% fructosesolution.ThelowdosewaschosentobewellbelowtherecommendedTDI, themediumdosecorrespondingtoTDI(50␮g/kgandday),whilethehighestdose wastentimesthislevel.TheBPAwasanalyzedbyliquidchromatography–tandem massspectrometrybytheDivisionofOccupationalandEnvironmentalMedicinein Lund,Sweden.ThedivisionisaEuropeanreferencelaboratoryintheDEMOCOPHES EUproject(www.eu-hbm.info/democophes)foranalysisofBPA.TheBPA concen-trationsinanalyzedsamplesofthesolutionswere:watercontrol–0.00020mg/L; fructosecontrol–0.00011mg/L;BPA0.025mg/L–0.029mg/L;BPA0.25mg/L– 0.25mg/LandBPA2.5mg/L–2.7mg/L.

Theexposuresolutionsweregivenadlib.fortenweeksandexposurelevels arepresentedinTable1.Thewatercontrolratsandthefructosecontrolratshad freeaccesstowatercontaining1%ethanol,and5%fructosesolutioncontaining1% ethanol,respectively.Groupsgivenfructosesolutiondrankmorethanthewater controlrats,andalsoraisedtheirliquidconsumptionduringtheexperiment,butate less.Thecontrolgroupgivenwaterhadanalmostconstantfoodandliquidintake.

(4)

Table1

ExposureofbisphenolA,liquidandfoodconsumption(RM1adlib.)andenergyintakeduringtheten-weekstudyofjuvenilefemaleFischer344ratsgiveneitherwaterora 5%fructosesolutionorbisphenolA(BPA)–0.025,0.25or2.5mg/L–dissolvedina5%fructosesolution.Foodandliquidconsumptionaremeasuredpercage(4cages/group) andallvaluesaregivenasthecalculatedmean/rat.N=12/group,w=week.

Control:water Control:5% fructosesolution BPA0.025mg/L+5% fructosesolution BPA0.25mg/L+5% fructosesolution BPA2.5mg/L+5% fructosesolution BPAexposure,meanw1–10(␮g/kg/day) 0 0 5.1 54.3 487.3

BPAexposurehighest(␮g/kg/day) 0 0 5.6(w2) 61.6(w3) 595.3(w2) BPAexposurelowest(␮g/kg/day) 0 0 4.6(w9) 46.3(w6) 400.3(w9) Liquid,meanw1–10(g/ratandday)d 11.5 28.3 28.1 30.1 24.7

Liquidw1(g/rat/day) 11.6 20.8 20.4 21.8 19.8

Liquidw10(g/rat/day) 10.8 32.0 33.0 36.8 29.4

Food,meanw1–10(g/ratandday)d 10.2 8.7 8.5 8.3 8.7

Foodw1(g/rat/day) 10.8 10.1 10.0 9.6 9.3

Foodw10(g/rat/day) 10.0 8.3 7.9 7.8 8.3

Fructoseenergymeanw1–10(kcal/rat/day) 0 5.7 5.6 6.0 4.9 Foodenergymeanw1–10(kcal/rat/day) 28.9 24.6 24.1 23.5 24.6 Energytotmeanw1–10(kcal/rat/day) 28.9 30.3 29.7 29.5 29.5

Differenceinmeancaloricintakewaslessthan5%betweenthegroupswithhighest andlowestcaloricintake.

2.4. Magneticresonanceimagingandpostprocessing

TheMRimagingwasperformedona1.5TclinicalMRsystem(Achieva;Philips Healthcare,Best,Netherlands)usingaquadraturekneecoil.Theratslayinprone position.MRcompatiblepadswereusedtopositiontheanimalinthecoil cen-ter.Twobottlesofwarmtapwaterwerepositionednexttotheratstohelpthem maintaintheirbodytemperature.

TwodifferentMRprotocolswereused.Awhole-bodysingleechowater–fat imaging protocolwas usedto analyze adiposetissue distribution.A32-echo water–fatimagingprotocolcoveringmostoftheliverwasusedtoanalyzeliver fatcontentandtherelaxationparameterR2*usingmodel-basedfittingtotime domaindata.Thismodel-baseddeterminationoffatcontentandR2*issimilarto quantificationofresonancepeakheightsandwidths,respectively,fromthe cor-respondingMRspectrum.Theimagedataandtheanalysisusedareillustratedin

Fig.2.

The whole-body imaging was performed using a volume of interest (100mm×100mm×150mm,sagittal×coronal×axial) positionedtocoverthe volumefrom necktotail, seeFig.1a. Aspoiled3Dsinglegradient-echo pro-tocolwithimagingparametersrepetitiontime8ms,echotime3.2ms,andflip angle12◦wasused.Theacquiredvoxelsizewas0.5mm×0.5mm×1.0mm.The reconstructedvoxelsizewas0.45mm×0.45mm×1.0mm.Fold-overdirectionwas anterior–posterior.Totalimagingtime,usingonesignalaveragewas4min17s. Waterfatshiftwas0.486pixels.Noparallelimagingwasused.

Waterandfatimageswerereconstructedfromthecomplexsingleechoimage datausingapreviouslypresentedmodel-basedmethod(Berglundetal.,2010).The possibilitytoseparatewaterandfatsignalfromasingleechoacquisitioncanbe ratherintuitivelyrealized.Theechotimeusedinthecurrentprotocolgivesan approximatephaseshiftof270◦betweenwaterandfat.Hence,aftercorrectionfor B0inhomogeneity,thewaterandfatsignalvectorsarealignedalongthepositive realaxisandnegativeimaginaryaxis,respectively.Inbrief,thealgorithm deter-minedthewaterandfatcontentineachvoxelusingthreeassumptions.First,the majorityofvoxelswereassumedtohaveoneoftwodifferentwater:fatsignalratios. Theassumedratioswere100:0,formusclesandorgans,and0:100,foradipose tis-sue.Second,thestaticmagneticfielddistributionwasassumedtobesmooth.Third, voxelswithanequalamountoffatandwaterwerelocatedoninterfacesbetween water-dominantregionsandadiposetissue.Thefirstassumptionlefttwopossible alternativesforthestaticmagneticfieldineachvoxel.Usingthesecondassumption, therightalternativecouldbeselectedusingoptimization.Inthisstudyamulti-scale beliefpropagationapproachwasused(FelzenszwalbandHuttenlocher,2006).To allowacontinuousspectrumofwater:fatratios,thephasemapwasfilteredusingan averagingfilter.Thedeterminationofthestaticmagneticfielddistributionallowed directcalculationofthewaterandfatcomponents.Methodfeasibilityhas previ-ouslybeendemonstratedinwhole-bodyscansofahumansubjectatboth1.5Tand 3.0T(Berglundetal.,2010).

Volumesoftotal,visceral,subcutaneousadiposetissue,andleantissue(TAT, VAT,SAT,andLT,respectively)werequantifiedusingasemi-automatedapproach. Fatfractionimages,definedbyfat/(fat+water),werecalculatedandadiposetissue andleantissuewereseparatedbythresholdingat50%fatfraction.

Toreducetheeffectoffatfractionsoriginatingfrombackgroundandlow sig-nalregionsintheanalysis,thetissueoftheratswasseparatedfrombackground byclustering.Thewaterandfatimageswereclusteredintothreeclasses(adipose tissue,leantissue,andbackground)usingaversionofFuzzyC-meansthat incorpo-ratesspatialcontinuity(Liewetal.,2005).Fatfractionsoriginatingfromnoiseinlow signalregionsweresuppressedbymultiplyingbythebackgroundclusterinverse.

TheVATvolumewassegmentedfromthefatfractionimageusingapreviously describedsemi-automatedmethod(Malmbergetal.,2009).Theoperatormanually

placedforegroundseedsintheVATdepotandbackgroundseedsinSAT, mus-cles,organs,andinthebackground.Thealgorithmthendeterminedtheboundary betweenVATandothertissues.Theoperatorinteractivelyadded/removedseedsin athree-planeviewuntilthesegmentationwasvisuallydeterminedtobeaccurate. TwooperatorssegmentedtheVATdepotinallanimals.ThemeanVATvolumewas used(meanCVwas1.40%).Thesubcutaneousadiposetissuevolumewascalculated asthedifferencebetweentheTATandVATvolumes.

The 32-echo water–fat liverimaging was performedusing a 3D spoiled gradient echoacquisition with the following scanparameters: Fieldof view, 95mm×95mm×15.6mm(sagittal×coronal×axial),acquiredandreconstructed voxelsize,1.19mmisotropic,repetitiontime,55ms,firstechotime,1.628ms,inter echospacing,1.274ms,flipangle,35◦.Imagingtime1min46s.Thewater–fatimage reconstructionwasperformedusingapreviouslydescribedmethodthatemploys awhole-imageoptimizationapproach(BerglundandKullberg,2012).Amulti-peak triglyceridespectrummodelderivedfromliverMRspectroscopy(Hamiltonetal., 2010)andacommonR2*forallpeakswereusedinthemodeling.TheR2*parameter canbethoughtofasthepeakwidthinfrequencydomainandcanbeusedtodetect liverirondeposition(positivecorrelation).Inthepresentstudy,theR2*parameter wasusedasanadditionalbiomarkerofliverstatus.TheliverfatcontentandR2* fromtheentireliverwasanalyzedbymanualidentificationofthevolumeof inter-estandbyfittingofaGaussianfunctiontotheliverfatfractionandR2*histograms (seeFig.1fandg).ThecenteroftheGaussianfunctionwasusedtosamplerobust estimatesofliverfatcontentandR2*.

2.5. Tissuesampling

AtterminationbloodwascollectedfromtheabdominalaortainEDTA-treated tubes(Greinerbio-one,Frickenhausen,Germany)andcentrifugedfor10minto prepareplasma.Aliquoteswerestoredat−70◦Cpendingbiochemicalanalysesof thefollowingcirculatingmarkers:triglycerides,cholesterol,andapolipoprotein A-I(apoA-I).Theliverandtheleftperirenalfatpad(seeFig.2)weredissectedand weighed.Theliverweightwasusedtocalculatetheliversomaticindex(LSI,liver weight×100/bodyweight).

2.6. Biochemistry

Theanalysisofcholesterolandtriglycerideswasastandardlaboratorytechnique andwasperformedonanArchitectC8000analyzer(AbbottLaboratories,Abbott Park,IL,USA)andreportedusingSIunits.AnalysisofproteinapoA-I:Priortowestern blot1␮lofplasmafromratsofallgroups(W;n=12,F;n=12,BPA0.025mg/L;n=11, BPA0.25mg/L;n=8andBPA2.5mg/L;n=9)wereseparatedonSDS-polyacrylamide gradientgels(T=5–20%,C=1.5%)withstackinggels(T=5%,C=1.5%)for1h(180V, 60mA)inelectrodebuffer(0.15%(w/v)Tris,0.72%(w/v)glycine,0.05%(w/v)SDS) usingaMiniProteanIIelectrophoresiscell(BioRad).Samplesweredilutedinsample cocktail(4%(w/v)SDS,200mMDTT,20%(w/v)sucrose)andboiledfor3min.Plasma proteinsseparatedbySDSPAGEweretransferredtoaPVDFmembrane.After block-ing1h(5%milkinTBS)andincubationovernightwithprimaryantibodies1:1000 (2%milkinTTBS)againstapoA-I(rabbitantiratapoA-I,polyclonal,Ab20453,Abcam, UK),themembranewasincubatedfor1hwithgoatanti-rabbitHRP-conjugated secondaryantibodies1:40000(2%milkinTTBS).Proteinswerevisualizedusing anECLpluswesternblottingdetectionsystem.Gelimageswereevaluatedusing ImageLab3.0.1(BioRad,Hercules,CA)andapoA-Ilevelsweredeterminedas intensity/mm2.

2.7. Statisticalanalysis

DifferencesbetweenthefructosecontrolgroupandthethreeBPAplusfructose exposedgroupswereevaluatedbyfactorialANOVA.WhenthethreeBPAgroups

(5)

128 M.Rönnetal./Toxicology303 (2013) 125–132

Fig.1.IllustrationoftheMRimagedataandpostprocessing.In(a)onecoronalslicefromawholebodyfatimageisshownwitharedoverlayofthesegmentedVATdepot. In(b–e)oneaxialslicefromtheliverscanisshown,where(b)showsthereconstructedwaterimage,(c)thefatimage,(d)thesignalfatfractionimage,and(e)theR2*image. Themanuallysegmentedregionoftheliverisillustratedbytheyellowdelineation.Images(f)and(g)showthedistributions,andthefittedGaussianfunctions,ofthefat signalfractionandR2*data,respectively,fromthedelineatedlivervolume.

Fig.2. Illustrationofthelocationandshapeoftheleftfatpad(seearrow).

wereanalyzedvsthefructosecontrolgrouponebyone,aBonferroniadjustment for3testswasusedandp<0.0167consideredsignificant(p=0.05/3=0.0167).

Inthesecondaryanalysis,whenthewatercontrolgroupwascomparedwith thefructosecontrolgroupp<0.05wasconsideredassignificant.

StatView(SASInc,USA)wasusedforcalculations.

3.

Results

3.1.

Primary

aim

No

differences

between

the

four

fructose-fed

groups

were

seen

regarding

the

initial

body

weight

recorded

prior

to

the

intervention

(p

=

0.83,

Table

2

).

Neither

did

the

weight

at

the

time

of

termination

of

the

experiment

(p

=

0.84),

nor

the

weight

gain

during

the

inter-vention

(p

=

0.68),

differ

between

the

four

groups.

No

differences

were

found

between

the

four

groups

regarding

the

weight

of

the

fat

pad

(p

=

0.32),

and

MRI

showed

no

differences

in

total

or

vis-ceral

adipose

tissue

volumes

between

the

four

groups

(see

Table

2

for

details).

However,

MRI

revealed

a

greater

fat

infiltration

in

the

liver

of

BPA-exposed

rats

than

in

the

fructose-fed

control

rats.

In

the

medium-dose

and

the

high-dose

group

of

BPA

exposed

rats

the

liver

fat

content

was

higher

when

compared

with

the

fructose

control

group

(p

=

0.011,

medium

dose;

p

=

0.012,

high

dose).

The

lowest

dose

of

BPA

did

not

significantly

influence

liver

fat

content

(

Fig.

3

).

Also

the

MRI

liver

R2*

analysis

showed

an

effect

on

the

liver

by

BPA,

being

significant

in

all

three

groups

when

compared

one

by

one

to

the

fructose

control

group

(low-dose;

p

=

0.0008,

middle-dose;

p

<

0.0001,

high-dose;

p

=

0.0161,

Table

2

).

A

similar

picture

emerged,

although

not

as

pronounced

as

for

the

R2*

signal,

when

the

liver

somatic

index

(LSI)

was

investigated.

(6)

Table2

Detailsofinitialbodyweightandresultsofweightgain,adiposeandleantissuevolumes,liverweightandcirculatingmarkersinjuvenilefemaleFischer344ratsinastudy withtwocontrolgroupsgiveneitherwaterora5%fructosesolutionandthreeexposedgroupsgivenbisphenolA(BPA)–0.025,0.25or2.5mg/L–dissolvedin5%fructose solutionfortenweeks.Numbersofobservationsare12ifnototherwisestated.Thep-valuesrepresenttheANOVAp-valueforadifferencebetweenthefourfructose-fed groups(controlgroupwithonlyfructoseandthethreegroupsgivenfructoseplusBPA).Allvaluesaregivenasmean±SD.

Control:water Control:5%fructose solution BPA 0.025mg/L+5% fructosesolution BPA 0.25mg/L+5% fructosesolution BPA2.5mg/L+5% fructosesolution ANOVA p-value

Initialbodyweight(g) 92.6±10.5 87.8±13.6 84.7±11.1 88.7±10.3 88.3±11.7 0.83 Weightgain,week1–10(g) 80.1±9.0 86.2±14.4 88.3±7.7 82.6±8.9 86.4±13.9 0.68 Bodyweightatsacrifice 172.3a±5.4 171.7±7.2 173.7±9.9 172.9±11.8 175.1±8.2 0.84

Estimatedbodyweight,MRI(g) 148.4±4.7 149.1±5.9 151.5±9.2 149.6±9.5 153.1±8.0 0.63

Totaladiposetissue(cm3) 28.2±3.4 29.8±5.7 29.6±4.9 28.9±5.2 31.7±5.0 0.60

Visceraladiposetissue(cm3) 13.9±1.5 14.3±3.0 13.9±2.4 13.8±2.6 15.3±2.8 0.57

Subcutaneousadiposetissue(cm3) 14.4±2.0 15.4±2.8 15.6±2.6 15.1±2.9 16.4±2.3 0.65

LeanTissue(cm3) 95.5±2.9 95.2±4.3 96.6±5.0 95.4±6.0 96.1±6.5 0.92

Liverfat(%) 5.5±1.0 5.6a±0.86 6.4±1.2 7.0±1.7* 6.9±0.73* 0.037

LiverR2*(1/s) 45.7±3.3 46.5±3.6 51.6±3.8*** 53.6±4.1*** 50.1±2.2* <0.001

Fatpad(g) 0.74±0.15 0.86±0.21 0.77±0.14 0.76±0.14 0.88±0.25 0.32

Fatpad/bodyweightratio(%) 0.43±0.08 0.50±0.11 0.44±0.07 0.44±0.08 0.50±0.13 0.28

Liver(g) 4.8±0.29 5.1±0.20 5.6±0.86 5.6±0.68 5.4±0.43 0.19

Liversomaticindex(LSI) 2.8±0.16 3.0±0.10** 3.2±0.32* 3.2±0.34* 3.1±0.17 0.08

Cholesterol(mmol/L) 3.0±0.17 3.1±0.23 3.2±0.16 3.1a±0.20 3.3±0.20 0.24

Triglycerides(mmol/L) 0.88±0.22 1.3±0.36* 1.7±0.62 1.7a±0.79 1.6±0.83 0.48

ApoA-I(intensity/mm2) 5648±1249 5967±1714 7622a±2468 11271b±3049*** 10524c±2023*** <0.001

Glucoseweek9(mmol/L) 4.6±0.79 4.7±0.70 4.5±0.45 4.4±0.36 4.5±0.53 0.49

ASAT 0.95±0.052 0.96±0.22 1.24±1.3 1.32a±1.29 1.02±0.22 0.75 ALAT 0.82±0.097 0.73±0.087 0.82±0.41 0.80a±0.19 0.76±0.16 0.79 an=11. bn=8. c n=9. *p<0.05. **p<0.01. ***p<0.001.

Whengivenaftervaluesinthefructosecontrolgroupthisindicatesadifferencevsthewatercontrolgroup.WhengivenaftervaluesinanyofthethreeBPAgroupsthis indicatesadifferencevsthefructosecontrolgroup.

LSI

was

increased

in

the

low-dose

(p

=

0.043,

not

significant

follow-ing

Bonferroni

adjustment)

and

middle-dose

group

(p

=

0.018,

not

significant

following

Bonferroni

adjustment),

but

not

significantly

so

in

the

high-dose

group

when

compared

with

the

fructose-fed

control

rats

(

Table

2

).

Both

the

medium-dose

and

high-dose

of

BPA

groups

showed

sig-nificantly

higher

levels

of

plasma

apo

A-I,

when

compared

with

the

fructose

control

group

(p

<

0.0001,

medium

dose;

p

<

0.0001

high

Fig.3. Liverfat content(%)(mean±SEM, water; n=12,fructose;n=11,BPA 0.025mg/L;n=12,BPA0.25mg/L;n=12andBPA2.5mg/L;n=12)injuvenilefemale Fischer344ratsgivenwater,5%fructosesolutionorbisphenolA(0.025,0.25or 2.5mg/L)dissolvedin5%fructosesolutionfortenweeks.

dose).

The

lowest

dose

of

BPA

did

not

cause

any

significant

differ-ence

in

apo

A-I

(

Fig.

4

).

Plasma

cholesterol

and

plasma

triglycerides

were

not

significantly

altered

by

the

BPA

exposure.

Neither

was

blood

glucose

at

week

9,

or

ASAT

and

ALAT

altered

by

BPA

exposure.

3.2.

Secondary

aim

Of

all

variables

studied

(see

Table

2

),

only

plasma

triglycerides

and

LSI

were

significantly

increased

by

fructose

feeding

alone

when

compared

to

the

water-fed

control

p

=

0.0011

and

p

=

0.0031,

respectively.

Fig.4.ApolipoproteinA-I(INT/mm2)(mean±SEM,water;n=12,fructose;n=12, BPA0.025mg/L;n=11,BPA0.25mg/L;n=8andBPA2.5mg/L;n=9)injuvenile femaleFischer344ratsgivenwater,5%fructosesolutionorbisphenolA(0.025, 0.25or2.5mg/L)dissolvedin5%fructosesolutionfortenweeks.

(7)

130 M.Rönnetal./Toxicology303 (2013) 125–132

4.

Discussion

4.1.

BPA,

fructose

and

lipid

metabolism

The

present

study

disclosed

no

evidence

that

BPA

exposure

in

juvenile

female

fructose-fed

F

344

rats

would

increase

fat

mass,

despite

the

use

of

both

weights

and

MR

imaging

based

detailed

quantification

of

different

adipose

tissue

compartments.

However,

the

observed

increase

in

liver

fat

infiltration,

detected

by

MRI

in

parallel

with

increase

in

LSI,

although

in

the

latter

case

not

sig-nificant

following

strict

Bonferroni

correction

for

multiple

testing,

even

at

dosages

close

to

TDI,

is

a

finding

that

warrants

further

investigations.

Interestingly,

an

increase

in

liver

fat

infiltration

appeared

at

the

middle

dose,

but

was

not

further

increased

at

the

highest

BPA

dose.

This

finding

confirms

a

previous

in

vivo

study

on

mice

by

Marmugi

et

al.,

using

the

same

dose

range

of

BPA

as

in

the

present

study,

but

without

fructose.

The

Marmugi

study

showed

an

impact

on

the

hepatic

transcriptome,

particularly

on

genes

involved

in

lipid

synthesis

and

that

various

transcription

factors

followed

a

non

monotonic

dose–response

curve

(

Marmugi

et

al.,

2012

).

In

addi-tion,

also

in

line

with

the

Marmugi

study,

the

most

significant

effects

were

observed

within

one

magnitude

around

the

current

TDI.

However,

Marmugi

et

al.

used

mice

and

did

not

combine

BPA

with

fructose,

so

our

study

is

not

entirely

comparable

with

theirs.

Low-dose

effects

of

BPA

are

currently

highlighted

and

under

dis-cussion

worldwide

(

Rhomberg

and

Goodman,

2012;

Richter

et

al.,

2007;

Ryan

et

al.,

2010;

Vandenberg

et

al.,

2012

)

and

therefore

three

dosages

were

used,

of

which

the

medium

dose

corresponded

to

the

defined

human

TDI,

as

established

by

the

U.S.

Environmental

Protection

Agency

(EPA)

and

the

European

Food

Safety

Authority

(EFSA)

at

50

␮g/kg

and

day.

TDI

is

equal

to

NOAEL

(5000

␮g/kg

and

day,

this

is

the

highest

dose

which

did

not

induce

any

adverse

effect

in

animal

testing),

divided

by

a

factor

of

100

to

compen-sate

for

possible

species

differences

in

sensitivity.

The

current

TDI

is

assumed

to

be

considerably

higher

than

the

calculated

human

exposure.

However,

in

the

present

study

and

others,

effects

are

seen

in

rats

and

mice

at

doses

close

to

the

current

TDI

and

even

at

lower

doses

(

Richter

et

al.,

2007

).

Low

dose

effects

of

environ-mental

contaminants

have

previously

been

suggested

based

on

epidemiological

studies,

as

well

as

in

experimental

settings

using

BPA

(

Lee

et

al.,

2011;

Marmugi

et

al.,

2012;

Rubin

et

al.,

2001;

Soriano

et

al.,

2012

).

Also

non

monotonic

relationships

are

sug-gested

in

e.g.

a

study

by

Wei

et

al.

where

pregnant

Wistar

rats

were

exposed

to

BPA

(50,

250

or

1250

␮g/kg

bw

and

day)

and

their

offspring

given

normal

or

high

fat

diet

after

weaning.

Only

the

lowest

dose

(50

␮g/kg

and

day)

resulted

in

such

effects

as

increased

body

weight,

elevated

serum

insulin

and

impaired

glu-cose

tolerance

in

adult

offspring.

In

the

rats

fed

a

high

fat

diet

the

effects

were

exacerbated

and

included

metabolic

syndrome

(obesity,

dyslipidemia,

hyperleptindemia,

hyperglycemia,

hyper-insulinemia

and

glucose

intolerance).

Rats

perinatally

exposed

to

the

higher

doses

did

not

show

any

of

the

adverse

effects

regard-less

of

diet

(

Wei

et

al.,

2011

).

A

similar

study

has

been

performed

with

CD-1

mice

by

Ryan

et

al.

but

with

a

different

conclusion.

In

this

experiment

the

mice

exposed

to

BPA

(approximately

0.25

␮g/kg

bw

and

day

via

the

diet)

during

gestation

and

lactation

had

heavier

and

longer

pups

at

weaning

than

pups

from

the

control

groups,

but

the

differences

did

not

persist

until

adulthood,

regardless

of

a

high

fat

or

low

fat

diet

given

from

9

weeks

of

age.

As

in

our

study

MRI

was

used

to

determine

body

composition

and

no

increase

in

body

fat

was

seen

in

the

BPA

exposed

rats

(

Ryan

et

al.,

2010

).

However,

these

studies

are

not

fully

comparable

due

to

differences

regarding

doses

and

species

and

the

time

point

when

the

modi-fied

diet

was

introduced.

Further

Ryan

et

al.

housed

their

mice

in

cages

made

of

polycarbonate

and

used

water

bottles

also

made

of

polycarbonate,

which

might

have

been

sources

of

BPA

contamina-tion

in

the

control

groups

masking

subtle

effects,

though

it

was

otherwise

a

very

sound

study.

The

above

mentioned

studies

were

carried

out

with

rodents

which

are

said

to

be

poor

models

for

BPA

in

humans

due

to

dif-ferent

toxicokinetics.

According

to

a

study

by

Tominaga

et

al.

using

nonhuman

primates;

chimpanzees

(Pantroglodytes

verus)

and

cynomolgus

monkeys

(Macaca

fascicularis),

there

are

differ-ences

also

among

different

primate

species.

In

rodents

the

BPA

is

longer,

primarily

explained

by

enterohepatic

recirculation

in

rodents

but

not

in

primates.

The

conjugation

rate

in

the

liver

is

faster

in

rodents

than

in

primates,

primarily

explained

by

a

higher

hepatic

blood

flow-rate

in

rodents

(

Tominaga

et

al.,

2006

).

However,

there

seem

to

be

no

differences

in

the

metabolites

formed

e.g.

it

is

a

question

of

rate

and

time

and

not

in

the

fate

of

BPA.

The

calculated

mean

exposure

in

humans

is

well

below

the

TDI,

but

there

are

still

uncertainties

about

the

exact

sources

of

exposure.

Further,

based

on

the

WHO

report:

“Joint

FAO/WHO

Expert

Meeting

to

Review

Toxicological

and

Health

Aspects

of

Bisphenol

A

Summary

Report”

(

http://www.who.int/foodsafety/

chem/chemicals/BPA

Summary2010.pdf

),

the

most

sensitive

indi-viduals

– newborn

babies

– are

also

the

ones

with

highest

exposure.

According

to

this

report

the

highest

estimated

exposure

occurs

in

infants

0–6

months

of

age

who

are

fed

with

liquid

formula

out

of

PC

bottles:

2.4

␮g/kg

bw

per

day

(mean)

and

4.5

␮g/kg

bw

per

day

(95th

percentile),

which

is

very

close

to

the

lowest

dose

used

in

the

present

study.

In

children,

teenagers

and

adults

the

mean

exposure

was

<0.01–0.40

␮g/kg

bw

per

day.

Prenatal

exposure

to

BPA

has

been

shown

to

increase

expression

of

lipogenic

genes

and

adipocyte

size

in

rodents

(

Marmugi

et

al.,

2012;

Somm

et

al.,

2009

).

Studies

on

isolated

cells

have

shown

BPA

to

induce

production

of

proinflammatory

cytokines,

such

as

IL-6

and

TNF-alpha

(

Yamashita

et

al.,

2005

),

and

to

induce

expres-sion

of

adipogenic

transcription

factors

(

Phrakonkham

et

al.,

2008

),

including

PPAR-gamma

activation

(

Kwintkiewicz

et

al.,

2010

).

How

these

in

vitro

findings

relate

to

the

present

finding

of

an

increase

in

liver

fat

infiltration

in

combined

exposure

to

fructose

and

BPA

is

not

understood.

The

above-mentioned

study

by

Marmugi

et

al.

further

suggests

that

exposure

to

low

BPA

doses

may

influence

de

novo

fatty

acid

synthesis

and

thereby

contributing

to

hepatic

steatosis

in

mice

(

Marmugi

et

al.,

2012

).

Interestingly,

fructose

has

also

been

pointed

out

as

a

possible

contributor

to

similar

effects

on

the

liver

by

its

interaction

with

the

Glut5

receptor

(

Lustig,

2010

).

In

line

with

Lustig,

it

has

been

suggested

by

others,

summarized

in

a

review

by

Yilmaz

(2012)

that

high

fructose

diet

(60–70%

of

total

energy

intake)

may

contribute

to

non-alcohol

fatty

liver

disease,

metabolic

syndrome,

formation

of

advanced

glycated

end

products

as

well

as

direct

dysmetabolic

effects

on

liver

enzymes.

In

the

present

study,

it

can

be

concluded

that

5%

fructose

alone

or

in

combination

with

BPA

results

in

unfavorable

metabolic

alterations.

There

are

three

possible

sources

of

increases

in

liver

fat;

de

novo

lipid

synthesis,

decreased

degradation

or

increased

transport

of

cholesteryl

esters

to

the

liver.

According

to

our

data

the

most

likely

mechanisms

behind

the

lipid

accumulation

in

the

liver

are

a

combination

of

de

novo

lipid

synthesis

and

increased

reversed

transport

(also

Section

4.2

).

The

individual

contribution

from

fructose

and

BPA

can

only

be

postulated,

but

according

to

the

liver

fat

accumulation

in

the

fructose

group

and

further

increase

accompanied

by

the

increase

of

plasma

apo

A-I

(

Fig.

4

)

after

BPA

exposure,

we

suggest

that

fructose

is

the

main

contributor

to

the

de

novo

lipid

synthesis

while

BPA

is

the

main

contributor

to

the

increased

reverse

transport.

The

decrease

in

plasma

apo

A-I

and

thereby

LSI

at

the

highest

BPA

dose

may

be

a

negative

feedback

response

on

apo

A-I

synthesis

but

has

to

be

further

inves-tigated.

References

Related documents

There was no difference in weight gain or body composition changes between women that reported the highest quarter of total training time/week (low intensity, cardiovascular and

LudoWCilsVWes eulos fponté feriet, fi, quod de annis paulo ante pro-. ad

High gestational weight gain (GWG) is primarily linked to morbidity associated with high fetal birth weight (FBW) (1) but women with excessive GWG are also more likely to have

Conclusions: The antenatal lifestyle intervention reduced mean GWG and short-term PPWR but no long-term effects on maternal weight retention or offspring obesity were

SilviScan is an instrument for measurement on the same sample of radial variations in growth and properties of wood (density and stiffness [modulus of elasticity; MOE]), fibres

body size and BMI early in life and increased risk of atrial fibrillation and heart failure, along with increased risk of heart failure, AMI, and premature death among patients

Thereto, we sought to estimate the impact of body size, body weight and obesity on the risk of early CVD and mortality in young women and in obese patients with and without

Compared to the Grow Up 1974 birth cohort, boys in the 1990 cohort had a higher prevalence of overweight and obesity and a lower prevalence of underweight, when the WHO 2007