10.02.2015 Views

The obesogenic effects of polyunsaturated fatty acids are dependent ...

The obesogenic effects of polyunsaturated fatty acids are dependent ...

The obesogenic effects of polyunsaturated fatty acids are dependent ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>The</strong> <strong>obesogenic</strong> <strong>effects</strong> <strong>of</strong> <strong>polyunsaturated</strong> <strong>fatty</strong><br />

<strong>acids</strong> <strong>are</strong> <strong>dependent</strong> on background diets<br />

Tao Ma<br />

Supervisor:<br />

Karsten Kristiansen<br />

Lise Madsen<br />

Department <strong>of</strong> Biology, University <strong>of</strong> Copenhagen, Copenhagen DK-<br />

2200, Denmark.<br />

<strong>The</strong> Graduate School <strong>of</strong> Science, Faculty <strong>of</strong> Science, University <strong>of</strong><br />

Copenhagen, Denmark.<br />

July 2011, 家


Acknowledgement<br />

I am grateful to my supervisor Karsten Kristiansen for giving me the opportunity to continue<br />

my research life. It is full <strong>of</strong> excitement, as all I can ask for. Thanks to his guidance and<br />

inspiration. I would as well give my thanks to my co-supervisor Lise Madsen for conceiving,<br />

organizing and directing my project. That is precious experiences that I can benefit from for<br />

my future work.<br />

We can only be a part <strong>of</strong> a team in order to achieve. I would like to thanks all the group<br />

members in Denmark and Norway for their helps and joys they bring along, especially Qin<br />

Hao and Rasmus Koefoed Petersen for their patience and kindness. Also, my appreciation to<br />

Alison Keenan for her great help with the language.<br />

And finally, there is nothing can be accomplished without the continuous support and love<br />

from my family.<br />

2


Table <strong>of</strong> Contents<br />

Acknowledgement ............................................................................................................................................. 2<br />

Abstract ............................................................................................................................................................. 4<br />

Abstract in Danish .............................................................................................................................................. 5<br />

Introduction ....................................................................................................................................................... 6<br />

Obesity and food intake ................................................................................................................................ 6<br />

Polyunsaturated <strong>fatty</strong> <strong>acids</strong> in the diet ......................................................................................................... 7<br />

Adipocyte biology .......................................................................................................................................... 7<br />

Adipogenesis is <strong>dependent</strong> on cAMP levels. ................................................................................................. 8<br />

Results ............................................................................................................................................................... 9<br />

Discussion, perspectives .................................................................................................................................. 11<br />

Low carbohydrate, high protein diet ........................................................................................................... 11<br />

Branched-chain amino <strong>acids</strong> and mTOR ...................................................................................................... 12<br />

Regulation <strong>of</strong> hepatic lipogenesis ................................................................................................................ 12<br />

Fructose ....................................................................................................................................................... 13<br />

Fish oil contaminants ................................................................................................................................... 14<br />

References ....................................................................................................................................................... 16<br />

Annexes ........................................................................................................................................................... 21<br />

3


Abstract<br />

Polyunsaturated n-3 <strong>fatty</strong> <strong>acids</strong> (n-3 PUFAs) <strong>are</strong> reported to protect against high fat diet-induced<br />

obesity and inflammation in adipose tissue. While it has previously been reported that the<br />

adipogenic <strong>effects</strong> <strong>of</strong> n-6 PUFAs <strong>are</strong> <strong>dependent</strong> on the macronutrient composition <strong>of</strong> the diet,<br />

whether n-3 PUFAs metabolism is subjected to similar impact by carbohydrates and proteins in the<br />

diet is not well explored. In the present thesis two studies <strong>are</strong> described. Isocaloric high fat diets<br />

enriched with protein and carbohydrate with different weight ratio or with various carbohydrate<br />

sources were fed to male C57BL/6J mice. We show that increasing the amount <strong>of</strong> sucrose at the<br />

expense <strong>of</strong> protein in the diet correlated with increased energy efficiency and fat mass irrespective<br />

<strong>of</strong> fat sources. We propose that this effect is a result <strong>of</strong> reduced thermogenesis in fat tissues,<br />

combined repression <strong>of</strong> gluconeogenesis and ureagenesis in the liver. Also, high fat diets induced<br />

glucose intolerance regardless <strong>of</strong> the adiposity <strong>of</strong> the animals. Moreover, by using carbohydrates<br />

with differing glycemic indices we provide evidence that insulin plays a central role in promoting<br />

adiposity and inflammation in fat tissues. This idea was strengthened further by exploring<br />

pharmaceutical drugs to modulate insulin secretion.<br />

In summary, the ability <strong>of</strong> background diet, namely carbohydrates and proteins, in regulating insulin<br />

secretion significantly modulates the beneficial <strong>effects</strong> <strong>of</strong> n-3 PUFAs in development <strong>of</strong> obesity,<br />

glucose intolerance and adipose tissue inflammation.<br />

4


Abstract in Danish<br />

Polyumættede n-3 fedtsyrer (n-3 PUFA) er rapporteret at beskytte mod fedme og inflammation i<br />

fedtvæv induceret af kost med højt fedtindhold. Mens det tidligere har været rapporteret, at de<br />

adipogene effekter af n-6 PUFA er afhængige af kostens sammensætning af makronæringsst<strong>of</strong>, er<br />

påvirkning af kulhydrater og protein i kosten på n-3 PUFA metabolisme ikke godt udforsket. I<br />

nærværende afhandling er to undersøgelser beskrevet. C57BL/6J hanmus blev fodret med<br />

isokaloriske dieter med højt fedtindhold beriget med protein og kulhydrat med varierende indbyrdes<br />

vægt-forhold eller med forskellige kulhydratkilder. Vi viser, at øget sukker i kosten er korreleret<br />

med øget energieffektivitet og fedtmasse uanset fedtkilder. Vores hypotese er, at disse observationer<br />

er en følge af nedsat termogenese i fedtvæv, og gluconeogenese og ureagenese i leveren. Ligeledes<br />

inducerer kost med højt fedtindhold glukose intolerance uafhængigt af dyrenes (over)vægt. Ved<br />

brug af kulhydrater med forskellige glykæmiske indeks viser vi desuden, at insulin spiller en central<br />

rolle for udviklingen af overvægt og inflammation i fedtvæv. Denne observation blev styrket<br />

yderligere i forsøg med lægemidler, der direkte modulerer insulinsekretion.<br />

Sammenfattende kan vi konkludere, at kostens indhold af kulhydrater og proteiner og evnen til at<br />

regulere insulinsekretionen påvirker de gavnlige effekter af n-3 PUFA på udvikling af fedme,<br />

glukose intolerance og inflammation i fedtvæv.<br />

5


Introduction<br />

Obesity and food intake<br />

Obesity is when excess fat is accumulated in the body, mainly in adipose tissues, to an unhealthy<br />

extent. An adult with a body mass index (BMI) between 25 and 30 kg/m 2 is considered overweight,<br />

while an adult with a BMI over 30 kg/m 2 is regarded as obese 1 ; nonetheless, this classification is<br />

debatable when applied to different races 2,3 .<br />

Utilizing the historic record provided by American obesity data 4 , we <strong>are</strong> able to track the increasing<br />

incidence <strong>of</strong> overweight and obesity in the new world. Not trailing far behind, developing countries<br />

<strong>are</strong> catching up with this trend, which is a direct consequence <strong>of</strong> overconsumption <strong>of</strong> energy-dense<br />

food and engaging in a sedentary lifestyle 5 . Obesity is one <strong>of</strong> the biggest threats to public health in<br />

the new century, as it increases the risk <strong>of</strong> developing type 2 diabetes 6 , cardiovascular disease 7 and<br />

certain types <strong>of</strong> cancer 8 .<br />

Consequently, considerable financial resources as well as social and scientific efforts have been<br />

mobilized towards the war against obesity 9 . One focus point has been to promote a reduction in<br />

dietary fat intake, as can be observed with the “light” labeled goods filling supermarket shelves.<br />

This is mainly because fat contains more energy per unit weight than carbohydrate or protein,<br />

9kcal/g, 4.5kcal/g and 4kcal/g respectively. By reducing the energy density <strong>of</strong> food, the average<br />

energy intake <strong>of</strong> the general public should therefore be reduced. Besides the increase in average<br />

energy intake over time 10 , another major alteration over the thousands <strong>of</strong> years <strong>of</strong> dietary history is<br />

the dramatic elevation <strong>of</strong> carbohydrate content in the food at the expense <strong>of</strong> protein 11 . One recent<br />

publication suggested that over a period <strong>of</strong> two years a low-carbohydrate diet had a similar weight<br />

loss effect as a low-fat diet 12 . Moreover, a body <strong>of</strong> evidence pointed to the beneficial <strong>effects</strong> <strong>of</strong> high<br />

protein diet on whole body metabolism 13 , implying that macronutrients <strong>are</strong> not simply different<br />

with regard to their caloric value, but have other impacts on weight gain and maintenance that <strong>are</strong><br />

not fully elucidated.<br />

6


Polyunsaturated <strong>fatty</strong> <strong>acids</strong> in the diet<br />

Fat comes in the form <strong>of</strong> a blend <strong>of</strong> different <strong>fatty</strong> <strong>acids</strong>, namely saturated <strong>fatty</strong> <strong>acids</strong> (SFAs),<br />

monounsaturated <strong>fatty</strong> <strong>acids</strong> (MUFAs) and <strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong> (PUFAs). Multiple studies<br />

suggest that the consumption <strong>of</strong> MUFAs in place <strong>of</strong> SFAs improves insulin sensitivity 14,15 . <strong>The</strong><br />

increased intake <strong>of</strong> n-6 PUFAs, or higher dietary n-6 to n-3 ratio, has been implicated in promoting<br />

many diseases including obesity and diabetes 16,17 . As shown in the work <strong>of</strong> Massiera et al., in mice,<br />

pups from mothers fed a diet enriched with n-6 linoleic acid became obese. However, this can be<br />

prevented by inclusion <strong>of</strong> the n-3 PUFA α-linolenic acid in the diet. <strong>The</strong> authors attributed this<br />

phenomenon to the ability <strong>of</strong> n-6 PUFAs to promote adipogenesis 18 . In line with the animal work, a<br />

similar outcome has been observed in a human study that revealed a higher n-6/n-3 PUFA ratio in<br />

the umbilical cord plasma was associated with a higher incidence <strong>of</strong> obesity 19 . Furthermore, work<br />

from our group suggests that the adipogenic nature <strong>of</strong> n-6 PUFAs is <strong>dependent</strong> on the cellular<br />

cAMP status 20 .<br />

<strong>The</strong> possible anti-obesity mechanisms <strong>of</strong> n-3 PUFAs include, but <strong>are</strong> not limited to: 1), competition<br />

with n-6 PUFAs in enzymatic pathways, which may dampen the adipogenic and pro-inflammatory<br />

<strong>effects</strong> <strong>of</strong> n-6 PUFAs 21 , as one metabolic effect from obesity is chronic low-grade inflammation.<br />

Anti-inflammatory intervention has been shown to be helpful against metabolic dysfunction 22 . 2),<br />

up regulation <strong>of</strong> β-oxidation and mitochondrial biogenesis. In both rodent and human settings<br />

addition <strong>of</strong> n-3 PUFAs, particularly <strong>of</strong> marine source, increased lipid oxidation 23,24 . 3), activation <strong>of</strong><br />

G protein-coupled receptor 120, which exerts its anti-inflammatory <strong>effects</strong> by inhibiting both Tolllike<br />

receptor (TLR) and tissue necrosis factor- α (TNF-α) signaling pathways in the residing<br />

macrophages in adipose tissue 25 .<br />

Adipocyte biology<br />

Historically, adipose tissue was seen as a static storage unit when energy is in excess and to release<br />

energy during food deprivation in the form <strong>of</strong> <strong>fatty</strong> <strong>acids</strong> and glycerol. When energy consumption<br />

persistently exceeds energy expenditure, expansion <strong>of</strong> adipose tissue will occur, contributed to by<br />

both hyperplasia and hypertrophy <strong>of</strong> adipocytes and finally culminate in obesity. Recently adipose<br />

tissue has become appreciated as an important mediator <strong>of</strong> systemic metabolism for its role <strong>of</strong><br />

secreting regulatory proteins 26 . <strong>The</strong> first adipokine being identified was adipsin (Factor D) from<br />

7


1987 27 . Over the years, more and more adipokines were discovered including leptin, TNF-α,<br />

adiponectin and so on, most <strong>of</strong> which bridge the links between obesity, inflammation and insulin<br />

resistance 28 . Furthermore, with the rediscovery <strong>of</strong> brown adipose tissue in adult humans, people<br />

have begun exploring the possible therapeutic role <strong>of</strong> this special fat depot 29 .<br />

Insulin is an important driver for adipocyte differentiation and the primary anabolic hormone<br />

promoting energy storage 30,31 . After ingesting a meal <strong>of</strong> high glycemic carbohydrate, the high<br />

glycemia will induce pancreatic insulin secretion to promote glucose uptake in adipose tissue. As<br />

shown in animal models, mice with adipose tissue-specific insulin receptor knock out (FIRKO mice)<br />

<strong>are</strong> protected against obesity and remain glucose tolerant on a high-fat diet 32 . In contrast, mice<br />

lacking the insulin receptor in muscle (MIRKO mice) maintain normal plasma glucose levels by<br />

increasing glucose utilization in white adipose tissue (WAT) and fat mass in order to compensate<br />

the loss <strong>of</strong> muscular glucose transport activity 33 . Furthermore, mice overexpressing facilitated<br />

glucose transporter, member 4 (GLUT4) 34 or insulin receptor substrate 1 (Irs1) 35 in adipose tissue<br />

<strong>are</strong> obese.<br />

Adipogenesis is <strong>dependent</strong> on cAMP levels.<br />

Our earlier works has shown the interplay <strong>of</strong> cAMP-elevating agents and arachidonic acid in<br />

mediating adipogenesis 20,36 . <strong>The</strong> inclusion <strong>of</strong> n-6 <strong>fatty</strong> acid arachidonic acid (AA) in the induction<br />

cocktail inhibited adipogenesis <strong>of</strong> 3T3-L1 preadipocytes. This inhibitory effect requires the cAMP –<br />

elevating agent 3-isobutyl-1-methylxanthine (IBMX) in the mixture and depends on protein kinase<br />

A (PKA) and cycloxygenase (COX) activity. During the initial phase <strong>of</strong> adipogenic induction, the<br />

combination <strong>of</strong> IBMX and AA dramatically increases the expression levels <strong>of</strong> both COX-1 and 2,<br />

which in turn metabolize AA towards the inhibitory prostaglandin E 2 (PGE 2 ) and F 2α (PGF 2α ) 37 .<br />

Furthermore, when AA is present, even without an increase <strong>of</strong> cellular cAMP levels, overexpression<br />

<strong>of</strong> COX-1 and 2 can still execute their anti-adipogenic power. On the other hand, in the absence <strong>of</strong><br />

IBMX, AA induces adipogenesis which was also reported previously by others, probably through<br />

the production <strong>of</strong> adipogenic prostaglandins 18 .<br />

In order to create a similar scenario in vivo, on top <strong>of</strong> high corn oil, different proportions <strong>of</strong><br />

carbohydrate and protein were included in the diets trying to manipulate the cAMP levels by<br />

changing the plasma insulin/glucagon ratio. Indeed as we reported 38 , the insulin to glucagon ratio<br />

8


was more than 3 times higher in the mice fed with high corn oil diet enriched with sucrose than with<br />

protein. With the different diets, significant changes in body weight gains were observed both ad<br />

libitum and pair-fed., and were due to the different degrees <strong>of</strong> adiposity. As expected, in multiple<br />

adipose tissues, the expression <strong>of</strong> Crem (cAMP-responsive element modulator) and Pde4b (cAMPspecific<br />

phosphodiesterase 4b) and the phosphorylation status <strong>of</strong> CREB (cAMP-responsive element<br />

-binding protein) were differently regulated because <strong>of</strong> the diets, which revealed an altered cAMP<br />

level. Accordingly, raised circulating PGE 2 and PGF 2 levels were detected in mice fed with a<br />

protein supplemented diet that correlated with increased expressions <strong>of</strong> both Cox-1 and Cox-2 in<br />

most <strong>of</strong> fat tissues.<br />

Taken together, it was a clear demonstration <strong>of</strong> the cAMP-PKA-COX-prostaglandin axis in<br />

regulating adipogenesis both in vitro and in vivo. <strong>The</strong> <strong>obesogenic</strong> effect <strong>of</strong> n-6 <strong>fatty</strong> acid is<br />

<strong>dependent</strong> on the macronutrient content <strong>of</strong> the diets.<br />

Results<br />

As shown previously, cAMP-<strong>dependent</strong> signaling controls the <strong>obesogenic</strong> effect <strong>of</strong> n-6 PUFAs in<br />

mice. We want to further investigate whether this phenomenon is restricted to n-6 PUFAs, or if the<br />

beneficial <strong>effects</strong> <strong>of</strong> n-3 PUFAs can also be affected by background diets (Annex 1). To achieve<br />

this, we prep<strong>are</strong>d isocaloric diets enriched with either fish oil or corn oil (Table 1). After 9 weeks <strong>of</strong><br />

feeding, to our surprise, mice fed with sucrose-supplemented diets became obese regardless <strong>of</strong> the<br />

fat sources (Fig.1 E-F). This was linked to the differential insulin to glucagon level in plasma that<br />

we had previously identified (Fig.1 C-D), suggesting one <strong>of</strong> the determining <strong>effects</strong> background diet<br />

has on obesity. Furthermore, the inflammation levels in the adipose tissues were correlated with<br />

adiposity instead <strong>of</strong> dietary fat types assessed by gene expression <strong>of</strong> macrophage and inflammatory<br />

markers (Fig.2A).<br />

When comp<strong>are</strong>d to mice fed a standard chow diet, all individuals on high fat diets had impaired<br />

glucose tolerance regardless they were fat or lean. But the separation <strong>of</strong> the HOMA index readout<br />

implied different mechanisms underlying the observed glucose intolerance (Fig.2B). We observed<br />

one <strong>of</strong> the recognized beneficial <strong>effects</strong> <strong>of</strong> taking fish oil in our settings. Hepatic lipid accumulation<br />

9


was considerably less in the fish oil-fed mice than in the corn oil-fed mice (Fig.3A), but it was not<br />

directly related to the lipogenic gene expression in their livers (Fig.3B).<br />

Since the mice on high fat diets consumed equivalent amounts <strong>of</strong> food, the lean ones had markedly<br />

reduced energy efficiency (Fig.3D, 4B). <strong>The</strong> following points could partly explain the differences,<br />

1). protein-fed animals could have more brownish phenotype in inguinal WAT (iWAT) judged by<br />

the up-regulation <strong>of</strong> Ucp1 expression, which dispenses energy as heat (Fig.3E, 4E); 2). elevated<br />

gluconeogenesis in mice fed protein-enriched diets even in the fed state, indicated by the enhanced<br />

glucose production during pyruvate tolerance test and hepatic gene expressions (Fig.6A and B); 3).<br />

nitrogen metabolism was another potential contributor to the energy-wasting effect as protein<br />

cannot be stored but must be processed immediately (Fig.6B).<br />

In order to evaluate the role <strong>of</strong> insulin in modulating the <strong>obesogenic</strong> <strong>effects</strong> <strong>of</strong> fish oil, isocaloric<br />

high fish oil diets with different sucrose to protein weight ratios were fed to the mice (Annex 2).<br />

Supporting our previous findings, the fat mass development (Fig.1D), energy efficiency (Fig.1B),<br />

inflammation levels in adipose tissues (Fig.1E) were all does-<strong>dependent</strong>ly correlated to sucrose<br />

content <strong>of</strong> the diets. mRNA levels <strong>of</strong> the indicators pointing to the three energy consumption<br />

processes, Ucp1 in the iWAT--- thermogenesis (Fig.1E); Pck1 in the live--- gluconeogenesis and<br />

Agxt in the liver--- ureagenesis (Fig.1F), were inversely associated as earlier suggested.<br />

By exchanging sucrose with glucose or fructose in a fish oil-supplemented diet, we further<br />

pinpointed that the insulin stimulating agent glucose moiety in sucrose facilitated the obesitypromoting<br />

effect <strong>of</strong> fish oil, as mice fed a fructose diet gained less WAT weights (Fig.2D) and had<br />

lower plasma triglycerides levels (Fig.2E). However, in the same set <strong>of</strong> mice, hepatic genes related<br />

to β-oxidation were down-regulated and the expression <strong>of</strong> lipogenic genes were increased (Fig.2F).<br />

Our theory was further supported by including starches promote different insulin responses<br />

(different glycemic index-GI). Although these animals did not differ in weight gain, the WAT<br />

masses were significantly higher in high GI diet-fed mice (Fig.4A and C), and this was<br />

accompanied by the increased expression <strong>of</strong> lipogenic genes Fasn and Scd1 (Fig.4G).<br />

Furthermore, pharmaceutical regulators <strong>of</strong> insulin secretion were introduced to the feeding regime.<br />

Inclusion <strong>of</strong> insulin secretagogue glybenclamide in the protein-enriched high fish oil diet did not<br />

result in a separation <strong>of</strong> the body weight gains (Fig.5A). A tendency <strong>of</strong> fat mass differences was<br />

observed (Fig.5B), and could be linked to observed changes in gene expression related to<br />

10


gluconeogenesis and ureagenesis (Fig.5E). On the other hand, inhibition <strong>of</strong> insulin secretion by<br />

nifedipine did achieve the expected fat mass-reducing effect (Fig.6C)<br />

.<br />

Discussion, perspectives<br />

Low carbohydrate, high protein diet<br />

It has been suggested that a modest increase in protein ingestion combined with a low glycemic<br />

index diet could be protective against obesity in children 39 . Further, as reported in a recent large<br />

European cohort study, after an initial weight loss, a diet with lower glycemic index and higher<br />

protein content is most ideal for maintaining body weight and preventing weight regain 40 . When<br />

consuming a diet very low in carbohydrates, such as in the case <strong>of</strong> our protein-enriched mouse feed<br />

with only 8% <strong>of</strong> calories from carbohydrates, the animals need to mobilize liver glycogen storage<br />

and increase gluconeogenesis in order to sustain blood glucose levels. After feeding for a long<br />

period <strong>of</strong> time, glycogen will be exhausted, and the body will have to turn to other energy sources.<br />

Even in the fed state, these protein-fed mice still kept high rates <strong>of</strong> β-oxidation, indicated by the<br />

high circulating levels <strong>of</strong> 2-hydroxybutyrate (Annex 1Fig.5B (1.5B)), suggesting the need to<br />

mobilize fat and/or protein for basal metabolism. This situation, to a certain degree, mimics what<br />

has been observed in long term fasting. During starvation, following the decrease <strong>of</strong> circulating<br />

glucose, insulin levels drop dramatically and glucagon secretion is elevated. Although<br />

gluconeogenesis contributes partially to the blood glucose supply, it will be prioritized for central<br />

nervous system and red blood cells functions.<br />

Despite the protein-fed mice kept lean as their low fat diet-fed littermates, they showed the same<br />

degree <strong>of</strong> glucose intolerance as the obese ones on sucrose-enriched diet. If our analogy to fasting is<br />

accurate, the glucose intolerance we have observed in lean protein-fed mice can be delineated. As<br />

fasting reduced insulin secretion in rats, this repression can only be rescued by refeeding with diets<br />

high in carbohydrates 41 . Although no difference was observed in the insulin tolerance test (Fig<br />

1.s1B), there <strong>are</strong> reports both in rats and humans showing that consuming low carbohydrate, high<br />

protein diet can attenuate the suppression <strong>of</strong> hepatic glucose output by insulin 42,43 . In order to<br />

11


esolve the interplay between insulin secretion and action, detailed studies <strong>are</strong> required to clarify the<br />

mechanisms behind impaired glucose tolerance in these mice.<br />

Branched-chain amino <strong>acids</strong> and mTOR<br />

Dietary proteins <strong>are</strong> important modulators <strong>of</strong> metabolism and insulin sensitivity 44 . It has been<br />

suggested that a modest increase in protein intake could potentially combat obesity 45 , owing at least<br />

in part to an increased thermic effect. We have shown these processes include but <strong>are</strong> not limited to<br />

thermogenesis in the iWAT (See annex 3), gluconeogenesis and ureagenesis in the liver. Casein has<br />

been widely used as the main protein source in rodent feed with the supplement <strong>of</strong> L-Cystine 46 .<br />

Given its relative high percentage <strong>of</strong> branched-chain amino <strong>acids</strong> (BCAAs) and low content <strong>of</strong><br />

taurine and glycine comp<strong>are</strong>d to fish or soy protein, special consideration needs to be taken into<br />

account when using high amount <strong>of</strong> casein. In a separate study, we have shown by replacing casein<br />

with salmon protein hydrolysates in the diet, rats were protected from diet-induced obesity due to<br />

raised plasma bile acid concentrations (Annex 4). Furthermore, there <strong>are</strong> links established between<br />

excess intake <strong>of</strong> dietary protein, especially BCAAs and insulin resistance 47,48 .<br />

Activation <strong>of</strong> mTOR (mammalian target <strong>of</strong> rapamycin) by BCAAs, particularly leucine, could<br />

regulate glucose uptake in skeletal muscle through the inhibitory phosphorylation <strong>of</strong> S6 kinase on<br />

insulin receptor substrate 1 (IRS-1) 49 . Furthermore Newgard et al, had shown that by 50% increase<br />

<strong>of</strong> BCAAs in a high fat diet, rats developed insulin resistance regardless <strong>of</strong> a low rate <strong>of</strong> weight gain,<br />

which was connected to chronic phosphorylation <strong>of</strong> mTOR, JNK, and IRS1 Ser307 and the<br />

accumulation <strong>of</strong> multiple acylcarnitines in muscle 50 . This observation relates to our reported results<br />

that insulin resistance can be disconnected from adiposity. In a recent multicenter cohort<br />

metabolomic study, elevated plasma BCAAs levels had been shown to correlate strongly with and<br />

could predict future diabetes 51 . If BCAAs did play a role contributing to the overall effect we have<br />

presented, protein composition in the diet needs to be thought over in the future experiment design.<br />

Regulation <strong>of</strong> hepatic lipogenesis<br />

Hepatic glucose and lipid metabolism <strong>are</strong> cross-regulated by multiple transcriptional regulators with<br />

sterol regulatory element binding protein-1c (SREBP-1c) taking the central stage 52 . In the context <strong>of</strong><br />

12


high dietary PUFAs, the modulation is less clear. PUFAs have been shown to suppress hepatic<br />

Srebp-1c transcription 53 , mRNA stability 54 and its proteolytic activation 55 through liver X receptor<br />

(LXR) 56 - and/or SREBP-1c 55 - mediated pathways. In our study, the suppression on Srebp-1c<br />

transcription was only sustained when diets were supplemented with protein but abolished when<br />

combined with sucrose (Fig1.3B, 2.7A). <strong>The</strong> most straight forward explanation would be that<br />

increased insulin levels played a role, as multiple lines <strong>of</strong> evidence suggested the importance <strong>of</strong><br />

insulin in controlling expression and activity <strong>of</strong> Srebp-1c 57 . But further in our study it was<br />

elucidated otherwise, as modulating insulin secretion by pharmaceutical drugs did not have any<br />

effect on either Srebp-1c mRNA abundance or its activity, judged by the transcription <strong>of</strong> its target<br />

genes Scd1, Fasn and Acaca (Fig2.5E 2.7A).<br />

<strong>The</strong> role <strong>of</strong> glucose in the context <strong>of</strong> this story needs also to be discussed. Besides insulin,<br />

carbohydrate ingestion can very efficiently lead to the induction <strong>of</strong> lipogenic genes 58 . However,the<br />

main sensor <strong>of</strong> glucose carbohydrate-responsive element–binding protein (ChREBP) 59 , like<br />

SREBP-1c, was also suppressed in the presence <strong>of</strong> PUFAs 60 . Mitro et al. has shown that LXRs<br />

could also work as glucose sensors to regulate cholesterol metabolism genes and in some degree<br />

lipogenic genes as well 61 . In the LXRα/β knockout model, high carbohydrate diet-induced hepatic<br />

gene expression <strong>of</strong> Scd1 and Fasn were attenuated 62 . As a whole, they suggest that in response to<br />

glucose stimuli, LXR-<strong>dependent</strong> pathway is indispensable for the full activation <strong>of</strong> Scd1 and Fasn<br />

expression, while the regulation <strong>of</strong> Acaca is mostly under the control <strong>of</strong> ChREBP 60 . So in our high<br />

fish oil feeding supplemented with different starches, the observed elevation <strong>of</strong> Scd1 and Fasn<br />

expression with high GI diet (Fig2.4G) could be explained as under the impact <strong>of</strong> fish oil, only LXR<br />

was still able to sense plasma glucose changes though mechanism that is still under debate 63,64 . Of<br />

course all speculations need supporting evidence from follow up studies. And with mTORC1 65,66<br />

and AMP-activated protein kinase (AMPK) 67,68 also in the picture now, the already intricate control<br />

<strong>of</strong> SREBP-1c and lipogenesis is only going to be more complicated.<br />

Fructose<br />

<strong>The</strong> increased world consumption <strong>of</strong> fructose or high-fructose corn syrup has been associated with<br />

the obesity epidemic 69,70 . Fructose is taken up by the liver rapidly because <strong>of</strong> the low Km <strong>of</strong><br />

fructokinase towards fructose 71 . Without the negative feedback on phosph<strong>of</strong>ructokinase as in<br />

glucose metabolism, the reactions continue to provide substrate for glucose and lipid production 71 .<br />

13


Although fructose has long been used to induce glucose intolerance in rats, in C57BL/6 mice<br />

neither hyperinsulimia nor hyperglycemia is developed. It had been shown C57BL/6 mice <strong>are</strong> lessresponsive<br />

towards fructose-induced Srebf1 activation among different strains <strong>of</strong> mice due to a<br />

single nucleotide polymorphism in the promoter region 72 . However we failed to observe this<br />

unresponsiveness. As others have shown in rats 73 , in our fructose-fed mice despite the presence <strong>of</strong><br />

fish oil in the diet, lipogenic genes were induced and β-oxidation-related targets were repressed<br />

(Fig.2.2F). But when comp<strong>are</strong>d to sucrose- and glucose-fed mice, these animals gained least body<br />

weight, had the lowest amounts <strong>of</strong> fat tissues and lowest circulating triglycerides (Fig2.2), so the<br />

extra fat has to be either stored ectopically or burnt. <strong>The</strong> increased brown adipocyte phenotype in<br />

the iWAT could, in part, assist this process. Although others have shown the development <strong>of</strong><br />

hepatic lipid accumulation in fructose-fed mice 74 , it was not the case in our hands despite elevated<br />

liver to body weight ratios (Fig2.3A). Interestingly, the authors also reported that a nonabsorbable<br />

antibiotics treatment could attenuate lipid accumulation in liver, suggesting potential involvement<br />

<strong>of</strong> gut microbes in fructose metabolism 74 .<br />

<strong>The</strong> metabolic <strong>effects</strong> <strong>of</strong> fructose do not limit to the listed above, as there have been reports<br />

showing centrally-administrated fructose led to increased food intake in rodents 75,76 by reducing<br />

hypothalamic malonyl–CoA levels 76 . Furthermore, the ability <strong>of</strong> fructose to cross the blood-brain<br />

barrier indirectly supports this observation, as the orexigenic effect relies on CNS function 77 . One<br />

interesting study, by using hyperinsulinemic-euglycemic clamps, demonstrated that sucrose feeding<br />

was not the same as with the combination <strong>of</strong> its hydrolysis products glucose and fructose in<br />

promoting hepatic insulin resistance 78 . This may suggest the involvement <strong>of</strong> gastric emptying or the<br />

release <strong>of</strong> gut-secreted hormones.<br />

Fish oil contaminants<br />

Long chain n-3 <strong>fatty</strong> <strong>acids</strong> in fish oil <strong>are</strong> not synthesized de novo by fish themselves, but by algae<br />

they consume in the wild or added to the feed in farmed fish colonies 79 . Along with the beneficial<br />

nutrients, through food chains, persistent organic pollutants (POPs) <strong>are</strong> also accumulated. Despite<br />

the effort to limit their release, POPs still persist in the environment 80 . POPs such as organochlorine<br />

pesticides, polychlorinated biphenyls (PCBs) and dioxins <strong>are</strong> hydrophobic compounds, and<br />

consequently they accumulate in <strong>fatty</strong> tissues, and therefore <strong>are</strong> inevitably retained in the process <strong>of</strong><br />

fish oil production.<br />

14


In a pair <strong>of</strong> population studies, an association was reported between serum concentrations <strong>of</strong> POPs<br />

and insulin resistance in spite <strong>of</strong> the subjects’ diabetic status 81,82 . Furthermore, different<br />

mechanisms were proposed in explaining this deteriorating effect, which include causing<br />

mitochondrial dysfunction 83 and impairment <strong>of</strong> insulin actions both in vivo and in vitro (ref 84 ,<br />

Annex 5). In order not to weigh out the beneficial effect by pollutants, purity would be a major<br />

concern when consuming very long chain marine n-3 <strong>fatty</strong> <strong>acids</strong>. This also applies to the use <strong>of</strong> fish<br />

oil in animal feeds 85 .<br />

15


References<br />

1. World Health Organization. Obesity: preventing and managing the global epidemic Report <strong>of</strong> a WHO<br />

Consultation (WHO Technical Report Series 894). (2000).<br />

2. Wang, J. et al. Comparisons for body mass index and body fat percent among Puerto Ricans, blacks, whites and<br />

Asians living in the New York City <strong>are</strong>a. Obesity research 4, 377-84(1996).<br />

3. Deurenberg, P., Yap, M. & Staveren, W.A. van Body mass index and percent body fat: a meta analysis among<br />

different ethnic groups. International journal <strong>of</strong> obesity and related metabolic disorders : journal <strong>of</strong> the<br />

International Association for the Study <strong>of</strong> Obesity 22, 1164-71(1998).<br />

4. Centers for Disease Control and Prevention U.S. Obesity Trends. at<br />

<br />

5. Hossain, P., Kawar, B. & El Nahas, M. Obesity and diabetes in the developing world--a growing challenge. <strong>The</strong><br />

New England journal <strong>of</strong> medicine 356, 213-5(2007).<br />

6. Colditz, G.A. et al. Weight gain as a risk factor for clinical diabetes mellitus in women. Annals <strong>of</strong> internal<br />

medicine 122, 481-6(1995).<br />

7. Willett, W.C. et al. Weight, weight change, and coronary heart disease in women. Risk within the “normal”<br />

weight range. JAMA : the journal <strong>of</strong> the American Medical Association 273, 461-5(1995).<br />

8. Calle, E.E. et al. Overweight, obesity, and mortality from cancer in a prospectively studied cohort <strong>of</strong> U.S. adults.<br />

<strong>The</strong> New England journal <strong>of</strong> medicine 348, 1625-38(2003).<br />

9. NIH New strategic plan for NIH obesity research seeks to curb epidemic. NIH News (2011).at<br />

<br />

10. Wright, J. et al. Trends in Intake <strong>of</strong> Energy and Macronutrients --- United States, 1971--2000. Morbidity and<br />

Mortality Weekly Report (2004).at <br />

11. Cordain, L. et al. Origins and evolution <strong>of</strong> the Western diet: health implications for the 21st century. Am J Clin<br />

Nutr 81, 341-354(2005).<br />

12. Foster, G.D. et al. Weight and metabolic outcomes after 2 years on a low-carbohydrate versus low-fat diet: a<br />

randomized trial. Annals <strong>of</strong> internal medicine 153, 147-57(2010).<br />

13. Paddon-Jones, D. et al. Protein, weight management, and satiety. Am J Clin Nutr 87, 1558S-1561(2008).<br />

14. Vessby, B. et al. Substituting dietary saturated for monounsaturated fat impairs insulin sensitivity in healthy<br />

men and women: <strong>The</strong> KANWU Study. Diabetologia 44, 312-9(2001).<br />

15. Gillingham, L.G., Harris-Janz, S. & Jones, P.J.H. Dietary Monounsaturated Fatty Acids Are Protective Against<br />

Metabolic Syndrome and Cardiovascular Disease Risk Factors. Lipids (2011).doi:10.1007/s11745-010-3524-y<br />

16. Ailhaud, G. et al. Temporal changes in dietary fats: role <strong>of</strong> n-6 <strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong> in excessive adipose<br />

tissue development and relationship to obesity. Progress in lipid research 45, 203-36(2006).<br />

17. Ailhaud, G., Guesnet, P. & Cunnane, S.C. An emerging risk factor for obesity: does disequilibrium <strong>of</strong><br />

<strong>polyunsaturated</strong> <strong>fatty</strong> acid metabolism contribute to excessive adipose tissue development <strong>The</strong> British journal<br />

<strong>of</strong> nutrition 100, 461-70(2008).<br />

16


18. Massiera, F. et al. Arachidonic acid and prostacyclin signaling promote adipose tissue development: a human<br />

health concern Journal <strong>of</strong> lipid research 44, 271-9(2003).<br />

19. Donahue, S.M. et al. Prenatal <strong>fatty</strong> acid status and child adiposity at age 3 y: results from a US pregnancy<br />

cohort. <strong>The</strong> American journal <strong>of</strong> clinical nutrition (2011).doi:10.3945/ajcn.110.005801<br />

20. Madsen, L. et al. cAMP-<strong>dependent</strong> signaling regulates the adipogenic effect <strong>of</strong> n-6 <strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong>.<br />

<strong>The</strong> Journal <strong>of</strong> biological chemistry 283, 7196-205(2008).<br />

21. Groeger, A.L. et al. Cyclooxygenase-2 generates anti-inflammatory mediators from omega-3 <strong>fatty</strong> <strong>acids</strong>. Nature<br />

chemical biology 6, 433-41(2010).<br />

22. Iyer, A. et al. Inflammatory lipid mediators in adipocyte function and obesity. Nature reviews. Endocrinology 6,<br />

71-82(2010).<br />

23. Javadi, M. et al. <strong>The</strong> effect <strong>of</strong> six different C18 <strong>fatty</strong> <strong>acids</strong> on body fat and energy metabolism in mice. <strong>The</strong><br />

British journal <strong>of</strong> nutrition 92, 391-9(2004).<br />

24. Couet, C. et al. Effect <strong>of</strong> dietary fish oil on body fat mass and basal fat oxidation in healthy adults. International<br />

journal <strong>of</strong> obesity and related metabolic disorders : journal <strong>of</strong> the International Association for the Study <strong>of</strong><br />

Obesity 21, 637-43(1997).<br />

25. Oh, D.Y. et al. GPR120 Is an Omega-3 Fatty Acid Receptor Mediating Potent Anti-inflammatory and Insulin-<br />

Sensitizing Effects. Cell 142, 687-698(2010).<br />

26. Guerre-Millo, M. Adipose tissue and adipokines: for better or worse. Diabetes & metabolism 30, 13-9(2004).<br />

27. Cook, K.S. et al. Adipsin: a circulating serine protease homolog secreted by adipose tissue and sciatic nerve.<br />

Science (New York, N.Y.) 237, 402-5(1987).<br />

28. Ouchi, N. et al. Adipokines in inflammation and metabolic disease. Nature reviews. Immunology 11, 85-<br />

97(2011).<br />

29. Nedergaard, J., Bengtsson, T. & Cannon, B. Unexpected evidence for active brown adipose tissue in adult<br />

humans. American journal <strong>of</strong> physiology. Endocrinology and metabolism 293, E444-52(2007).<br />

30. Saltiel, A.R. & Kahn, C.R. Insulin signalling and the regulation <strong>of</strong> glucose and lipid metabolism. Nature 414,<br />

799-806(2001).<br />

31. Madsen, L. & Kristiansen, K. <strong>The</strong> importance <strong>of</strong> dietary modulation <strong>of</strong> cAMP and insulin signaling in adipose<br />

tissue and the development <strong>of</strong> obesity. Annals <strong>of</strong> the New York Academy <strong>of</strong> Sciences 1190, 1-14(2010).<br />

32. Blüher, M. et al. Adipose Tissue Selective Insulin Receptor Knockout Protects against Obesity and Obesity-<br />

Related Glucose Intolerance. Developmental Cell 3, 25-38(2002).<br />

33. Kim, J.K. et al. Redistribution <strong>of</strong> substrates to adipose tissue promotes obesity in mice with selective insulin<br />

resistance in muscle. <strong>The</strong> Journal <strong>of</strong> clinical investigation 105, 1791-7(2000).<br />

34. Shepherd, P.R. et al. Adipose cell hyperplasia and enhanced glucose disposal in transgenic mice overexpressing<br />

GLUT4 selectively in adipose tissue. <strong>The</strong> Journal <strong>of</strong> biological chemistry 268, 22243-6(1993).<br />

35. Murata, Y. et al. IRS-1 transgenic mice show increased epididymal fat mass and insulin resistance. Biochemical<br />

and biophysical research communications 364, 301-7(2007).<br />

17


36. Petersen, R.K. et al. Arachidonic acid-<strong>dependent</strong> inhibition <strong>of</strong> adipocyte differentiation requires PKA activity<br />

and is associated with sustained expression <strong>of</strong> cyclooxygenases. Journal <strong>of</strong> lipid research 44, 2320-30(2003).<br />

37. Reginato, M.J. Prostaglandins Promote and Block Adipogenesis through Opposing Effects on Peroxisome<br />

Proliferator-activated Receptor gamma. Journal <strong>of</strong> Biological Chemistry 273, 1855-1858(1998).<br />

38. Madsen, L. et al. cAMP-<strong>dependent</strong> signaling regulates the adipogenic effect <strong>of</strong> n-6 <strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong>.<br />

<strong>The</strong> Journal <strong>of</strong> biological chemistry 283, 7196-205(2008).<br />

39. Papadaki, A. et al. <strong>The</strong> effect <strong>of</strong> protein and glycemic index on childrenʼs body composition: the DiOGenes<br />

randomized study. Pediatrics 126, e1143-52(2010).<br />

40. Larsen, T.M. et al. Diets with high or low protein content and glycemic index for weight-loss maintenance. <strong>The</strong><br />

New England journal <strong>of</strong> medicine 363, 2102-13(2010).<br />

41. Grey, N.J., Goldring, S. & Kipnis, D.M. <strong>The</strong> effect <strong>of</strong> fasting, diet, and actinomycin D on insulin secretion in<br />

the rat. <strong>The</strong> Journal <strong>of</strong> clinical investigation 49, 881-9(1970).<br />

42. Rossetti, L. et al. Effect <strong>of</strong> dietary protein on in vivo insulin action and liver glycogen repletion. Am J Physiol<br />

Endocrinol Metab 257, E212-219(1989).<br />

43. Linn, T. et al. Effect <strong>of</strong> long-term dietary protein intake on glucose metabolism in humans. Diabetologia 43,<br />

1257-65(2000).<br />

44. Tremblay, F. et al. Role <strong>of</strong> dietary proteins and amino <strong>acids</strong> in the pathogenesis <strong>of</strong> insulin resistance. Annual<br />

review <strong>of</strong> nutrition 27, 293-310(2007).<br />

45. Westerterp-Plantenga, M.S. et al. Dietary protein, metabolism, and body-weight regulation: dose–response<br />

<strong>effects</strong>. International Journal <strong>of</strong> Obesity 30, S16-S23(2006).<br />

46. Reeves, P.G., Nielsen, F.H. & Fahey, G.C. AIN-93 purified diets for laboratory rodents: final report <strong>of</strong> the<br />

American Institute <strong>of</strong> Nutrition ad hoc writing committee on the reformulation <strong>of</strong> the AIN-76A rodent diet. <strong>The</strong><br />

Journal <strong>of</strong> nutrition 123, 1939-51(1993).<br />

47. Patti, M.E. et al. Bidirectional modulation <strong>of</strong> insulin action by amino <strong>acids</strong>. <strong>The</strong> Journal <strong>of</strong> clinical investigation<br />

101, 1519-29(1998).<br />

48. Promintzer, M. & Krebs, M. Effects <strong>of</strong> dietary protein on glucose homeostasis. Current opinion in clinical<br />

nutrition and metabolic c<strong>are</strong> 9, 463-8(2006).<br />

49. Krebs, M. et al. <strong>The</strong> Mammalian target <strong>of</strong> rapamycin pathway regulates nutrient-sensitive glucose uptake in<br />

man. Diabetes 56, 1600-7(2007).<br />

50. Newgard, C.B. et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean<br />

humans and contributes to insulin resistance. Cell metabolism 9, 311-26(2009).<br />

51. Wang, T.J. et al. Metabolite pr<strong>of</strong>iles and the risk <strong>of</strong> developing diabetes. Nature Medicine 17, 448-453(2011).<br />

52. Ferré, P. & Foufelle, F. SREBP-1c transcription factor and lipid homeostasis: clinical perspective. Hormone<br />

research 68, 72-82(2007).<br />

53. Xu, J. Sterol Regulatory Element Binding Protein-1 Expression Is Suppressed by Dietary Polyunsaturated Fatty<br />

Acids. A MECHANISM FOR THE COORDINATE SUPPRESSION OF LIPOGENIC GENES BY<br />

POLYUNSATURATED FATS. Journal <strong>of</strong> Biological Chemistry 274, 23577-23583(1999).<br />

18


54. Xu, J. et al. Polyunsaturated <strong>fatty</strong> <strong>acids</strong> suppress hepatic sterol regulatory element-binding protein-1 expression<br />

by accelerating transcript decay. <strong>The</strong> Journal <strong>of</strong> biological chemistry 276, 9800-7(2001).<br />

55. Takeuchi, Y. et al. Polyunsaturated <strong>fatty</strong> <strong>acids</strong> selectively suppress sterol regulatory element-binding protein-1<br />

through proteolytic processing and autoloop regulatory circuit. <strong>The</strong> Journal <strong>of</strong> biological chemistry 285, 11681-<br />

91(2010).<br />

56. Ou, J. et al. Unsaturated <strong>fatty</strong> <strong>acids</strong> inhibit transcription <strong>of</strong> the sterol regulatory element-binding protein-1c<br />

(SREBP-1c) gene by antagonizing ligand-<strong>dependent</strong> activation <strong>of</strong> the LXR. Proceedings <strong>of</strong> the National<br />

Academy <strong>of</strong> Sciences <strong>of</strong> the United States <strong>of</strong> America 98, 6027-32(2001).<br />

57. Horton, J.D., Goldstein, J.L. & Brown, M.S. SREBPs: activators <strong>of</strong> the complete program <strong>of</strong> cholesterol and<br />

<strong>fatty</strong> acid synthesis in the liver. <strong>The</strong> Journal <strong>of</strong> clinical investigation 109, 1125-31(2002).<br />

58. Matsuzaka, T. et al. Insulin-In<strong>dependent</strong> Induction <strong>of</strong> Sterol Regulatory Element-Binding Protein-1c Expression<br />

in the Livers <strong>of</strong> Streptozotocin-Treated Mice. Diabetes 53, 560-569(2004).<br />

59. Kawaguchi, T. et al. Glucose and cAMP regulate the L-type pyruvate kinase gene by<br />

phosphorylation/dephosphorylation <strong>of</strong> the carbohydrate response element binding protein. Proceedings <strong>of</strong> the<br />

National Academy <strong>of</strong> Sciences <strong>of</strong> the United States <strong>of</strong> America 98, 13710-5(2001).<br />

60. Dentin, R. et al. Polyunsaturated <strong>fatty</strong> <strong>acids</strong> suppress glycolytic and lipogenic genes through the inhibition <strong>of</strong><br />

ChREBP nuclear protein translocation. <strong>The</strong> Journal <strong>of</strong> clinical investigation 115, 2843-54(2005).<br />

61. Mitro, N. et al. <strong>The</strong> nuclear receptor LXR is a glucose sensor. Nature 445, 219-23(2007).<br />

62. Denechaud, P.-D. et al. ChREBP, but not LXRs, is required for the induction <strong>of</strong> glucose-regulated genes in<br />

mouse liver. <strong>The</strong> Journal <strong>of</strong> clinical investigation 118, 956-64(2008).<br />

63. Lazar, M.A. & Willson, T.M. Sweet dreams for LXR. Cell metabolism 5, 159-61(2007).<br />

64. Anthonisen, E.H. et al. Nuclear receptor liver X receptor is O-GlcNAc-modified in response to glucose. <strong>The</strong><br />

Journal <strong>of</strong> biological chemistry 285, 1607-15(2010).<br />

65. Porstmann, T. et al. SREBP activity is regulated by mTORC1 and contributes to Akt-<strong>dependent</strong> cell growth.<br />

Cell metabolism 8, 224-36(2008).<br />

66. Li, S., Brown, M.S. & Goldstein, J.L. Bifurcation <strong>of</strong> insulin signaling pathway in rat liver: mTORC1 required<br />

for stimulation <strong>of</strong> lipogenesis, but not inhibition <strong>of</strong> gluconeogenesis. Proceedings <strong>of</strong> the National Academy <strong>of</strong><br />

Sciences <strong>of</strong> the United States <strong>of</strong> America 107, 3441-6(2010).<br />

67. Kawaguchi, T. et al. Mechanism for <strong>fatty</strong> acid “sparing” effect on glucose-induced transcription: regulation <strong>of</strong><br />

carbohydrate-responsive element-binding protein by AMP-activated protein kinase. <strong>The</strong> Journal <strong>of</strong> biological<br />

chemistry 277, 3829-35(2002).<br />

68. Li, Y. et al. AMPK Phosphorylates and Inhibits SREBP Activity to Attenuate Hepatic Steatosis and<br />

Atherosclerosis in Diet-Induced Insulin-Resistant Mice. Cell metabolism 13, 376-88(2011).<br />

69. Basciano, H., Federico, L. & Adeli, K. Fructose, insulin resistance, and metabolic dyslipidemia. Nutrition &<br />

metabolism 2, 5(2005).<br />

70. Tappy, L. & Lê, K.-A. Metabolic <strong>effects</strong> <strong>of</strong> fructose and the worldwide increase in obesity. Physiological<br />

reviews 90, 23-46(2010).<br />

19


71. Mayes, P.A. Intermediary metabolism <strong>of</strong> fructose. <strong>The</strong> American journal <strong>of</strong> clinical nutrition 58, 754S-<br />

765S(1993).<br />

72. Nagata, R. et al. Single nucleotide polymorphism (-468 Gly to A) at the promoter region <strong>of</strong> SREBP-1c<br />

associates with genetic defect <strong>of</strong> fructose-induced hepatic lipogenesis [corrected]. <strong>The</strong> Journal <strong>of</strong> biological<br />

chemistry 279, 29031-42(2004).<br />

73. Nagai, Y. et al. Amelioration <strong>of</strong> high fructose-induced metabolic derangements by activation <strong>of</strong> PPARalpha.<br />

American journal <strong>of</strong> physiology. Endocrinology and metabolism 282, E1180-90(2002).<br />

74. Bergheim, I. et al. Antibiotics protect against fructose-induced hepatic lipid accumulation in mice: role <strong>of</strong><br />

endotoxin. Journal <strong>of</strong> hepatology 48, 983-92(2008).<br />

75. Miller, C.C. et al. Intracerebroventricular Injection <strong>of</strong> Fructose Stimulates Feeding in Rats. Nutritional<br />

Neuroscience 5, 359-362(2002).<br />

76. Cha, S.H. et al. Differential <strong>effects</strong> <strong>of</strong> central fructose and glucose on hypothalamic malonyl-CoA and food<br />

intake. Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences <strong>of</strong> the United States <strong>of</strong> America 105, 16871-5(2008).<br />

77. Cha, S.H. & Lane, M.D. Central lactate metabolism suppresses food intake via the hypothalamic AMP<br />

kinase/malonyl-CoA signaling pathway. Biochemical and biophysical research communications 386, 212-<br />

6(2009).<br />

78. Thresher, J.S. et al. Comparison <strong>of</strong> the <strong>effects</strong> <strong>of</strong> sucrose and fructose on insulin action and glucose tolerance.<br />

Am J Physiol Regulatory Integrative Comp Physiol 279, R1334-1340(2000).<br />

79. Doughman, S.D., Krupanidhi, S. & Sanjeevi, C.B. Omega-3 <strong>fatty</strong> <strong>acids</strong> for nutrition and medicine: considering<br />

microalgae oil as a vegetarian source <strong>of</strong> EPA and DHA. Current diabetes reviews 3, 198-203(2007).<br />

80. <strong>The</strong> International Programme on Chemical Safety PERSISTENT ORGANIC POLLUTANTS. (1996).<br />

81. Lee, D.-H. et al. A strong dose-response relation between serum concentrations <strong>of</strong> persistent organic pollutants<br />

and diabetes: results from the National Health and Examination Survey 1999-2002. Diabetes c<strong>are</strong> 29, 1638-<br />

44(2006).<br />

82. Lee, D.-H. et al. Association between serum concentrations <strong>of</strong> persistent organic pollutants and insulin<br />

resistance among nondiabetic adults: results from the National Health and Nutrition Examination Survey 1999-<br />

2002. Diabetes c<strong>are</strong> 30, 622-8(2007).<br />

83. Lim, S. et al. Chronic exposure to the herbicide, atrazine, causes mitochondrial dysfunction and insulin<br />

resistance. PloS one 4, e5186(2009).<br />

84. Ruzzin, J. et al. Persistent organic pollutant exposure leads to insulin resistance syndrome. Environmental<br />

health perspectives 118, 465-71(2010).<br />

85. Berntssen, M.H.G. et al. Reducing persistent organic pollutants while maintaining long chain omega-3 <strong>fatty</strong><br />

acid in farmed Atlantic salmon using decontaminated fish oils for an entire production cycle. Chemosphere 81,<br />

242-52(2010).<br />

20


Annexes<br />

1. Ma T, Liaset B, Hao Q, Petersen RK, Fjære E, et al. (2011) Sucrose Counteracts the Antinflammatory<br />

Effect <strong>of</strong> Fish Oil in Adipose Tissue and Increases Obesity Development in Mice.<br />

PLoS ONE 6(6): e21647. doi:10.1371/journal.pone.0021647<br />

2. Hao Q, Lillefosse HH, Fjære E, … Ma T, et al. (2011). Carbohydrate Source and Insulin Secretion<br />

Modulate the Obesity Promoting Effect <strong>of</strong> Fish Oil. Manuscript in revision.<br />

3. Madsen L, Pedersen LM, Lillefosse HH, Fjære E, … Ma T, et al. (2010) UCP1 Induction during<br />

Recruitment <strong>of</strong> Brown Adipocytes in White Adipose Tissue Is Dependent on Cyclooxygenase<br />

Activity. PLoS ONE 5(6): e11391. doi:10.1371/journal.pone.0011391.<br />

4. Liaset B, Hao Q, Jørgensen H, Hallenborg P, Du ZY, Ma T, et al. (2011). Nutritional regulation <strong>of</strong><br />

bile acid metabolism improves pathological characteristics <strong>of</strong> the metabolic syndrome. J Biol<br />

Chem., in press.<br />

5. Ruzzin J, Petersen RK, Meugnier E, Madsen L, … Ma T, et al. (2010). Persistent organic<br />

pollutant exposure leads to insulin resistance syndrome. Environ Health Perspect. 118(4):465-<br />

71.<br />

21


Sucrose Counteracts the Anti-Inflammatory Effect <strong>of</strong> Fish<br />

Oil in Adipose Tissue and Increases Obesity<br />

Development in Mice<br />

Tao Ma 1 , Bjørn Liaset 2 , Qin Hao 1 , Rasmus Koefoed Petersen 1 , Even Fjære 1,2 , Ha Thi Ngo 2¤ , Haldis Haukås<br />

Lillefosse 1,2 , Stine Ringholm 1 , Si Brask Sonne 1 , Jonas Thue Treebak 3 , Henriette Pilegaard 1 , Livar<br />

Frøyland 2 , Karsten Kristiansen 1 *, Lise Madsen 1,2 *<br />

1 Department <strong>of</strong> Biology, University <strong>of</strong> Copenhagen, Copenhagen, Denmark, 2 National Institute <strong>of</strong> Nutrition and Seafood Research (NIFES), Bergen, Norway,<br />

3 Department <strong>of</strong> Exercise and Sport Sciences, University <strong>of</strong> Copenhagen, Copenhagen, Denmark<br />

Abstract<br />

Background: Polyunsaturated n-3 <strong>fatty</strong> <strong>acids</strong> (n-3 PUFAs) <strong>are</strong> reported to protect against high fat diet-induced obesity and<br />

inflammation in adipose tissue. Here we aimed to investigate if the amount <strong>of</strong> sucrose in the background diet influences the<br />

ability <strong>of</strong> n-3 PUFAs to protect against diet-induced obesity, adipose tissue inflammation and glucose intolerance.<br />

Methodology/Principal Findings: We fed C57BL/6J mice a protein- (casein) or sucrose-based high fat diet supplemented<br />

with fish oil or corn oil for 9 weeks. Irrespective <strong>of</strong> the <strong>fatty</strong> acid source, mice fed diets rich in sucrose became obese<br />

whereas mice fed high protein diets remained lean. Inclusion <strong>of</strong> sucrose in the diet also counteracted the well-known antiinflammatory<br />

effect <strong>of</strong> fish oil in adipose tissue, but did not impair the ability <strong>of</strong> fish oil to prevent accumulation <strong>of</strong> fat in the<br />

liver. Calculation <strong>of</strong> HOMA-IR indicated that mice fed high levels <strong>of</strong> proteins remained insulin sensitive, whereas insulin<br />

sensitivity was reduced in the obese mice fed sucrose irrespectively <strong>of</strong> the fat source. We show that a high fat diet decreased<br />

glucose tolerance in the mice in<strong>dependent</strong>ly <strong>of</strong> both obesity and dietary levels <strong>of</strong> n-3 PUFAs and sucrose. Of note,<br />

increasing the protein:sucrose ratio in high fat diets decreased energy efficiency irrespective <strong>of</strong> fat source. This was<br />

accompanied by increased expression <strong>of</strong> Ppargc1a (peroxisome proliferator-activated receptor, gamma, coactivator 1 alpha)<br />

and increased gluconeogenesis in the fed state.<br />

Conclusions/Significance: <strong>The</strong> background diet influence the ability <strong>of</strong> n-3 PUFAs to protect against development <strong>of</strong><br />

obesity, glucose intolerance and adipose tissue inflammation. High levels <strong>of</strong> dietary sucrose counteract the antiinflammatory<br />

effect <strong>of</strong> fish oil in adipose tissue and increases obesity development in mice.<br />

Citation: Ma T, Liaset B, Hao Q, Petersen RK, Fjære E, et al. (2011) Sucrose Counteracts the Anti-Inflammatory Effect <strong>of</strong> Fish Oil in Adipose Tissue and Increases<br />

Obesity Development in Mice. PLoS ONE 6(6): e21647. doi:10.1371/journal.pone.0021647<br />

Editor: Aimin Xu, University <strong>of</strong> Hong Kong, China<br />

Received March 28, 2011; Accepted June 4, 2011; Published June 28, 2011<br />

Copyright: ß 2011 Ma et al. This is an open-access article distributed under the terms <strong>of</strong> the Creative Commons Attribution License, which permits unrestricted<br />

use, distribution, and reproduction in any medium, provided the original author and source <strong>are</strong> credited.<br />

Funding: This work was supported by the Danish Natural Science Research Council, the Novo Nordisk Foundation, the Carlsberg Foundation, and the National<br />

Institute <strong>of</strong> Nutrition and Seafood Research, Norway. Part <strong>of</strong> the work was carried out as a part <strong>of</strong> the research program <strong>of</strong> the Danish Obesity Research Centre<br />

(DanORC). DanORC is supported by the Danish Council for Strategic Research (Grant NO 2101 06 0005). <strong>The</strong> funders had no role in study design, data collection<br />

and analysis, decision to publish, or preparation <strong>of</strong> the manuscript.<br />

Competing Interests: <strong>The</strong> authors have decl<strong>are</strong>d that no competing interests exist.<br />

* E-mail: kk@bio.ku.dk (KK); lise.madsen@nifes.no (LM)<br />

¤ Current address: Department <strong>of</strong> Food Safety and Nutrition, Division <strong>of</strong> Environmental Medicine, Norwegian Institute <strong>of</strong> Public Health, Oslo, Norway<br />

Introduction<br />

Today it is recognized that the potentially harmful <strong>effects</strong> <strong>of</strong><br />

high fat diets relates to not only the amount, but also the type <strong>of</strong><br />

dietary <strong>fatty</strong> <strong>acids</strong>. Whereas a high intake <strong>of</strong> saturated and trans<br />

<strong>fatty</strong> <strong>acids</strong> has been shown to be associated with increased risk <strong>of</strong><br />

cardiovascular diseases in several studies, intake <strong>of</strong> <strong>polyunsaturated</strong><br />

<strong>fatty</strong> <strong>acids</strong> (PUFAs) has been associated with lower cardiovascular<br />

risk [1,2]. Thus, increasing the relative amount <strong>of</strong> PUFAs,<br />

both vegetable n-6 PUFAs and marine n-3 PUFA, at the expense<br />

<strong>of</strong> saturated fat is recommended. It is important to note, however,<br />

that more than 85% <strong>of</strong> the total dietary PUFA intake in Western<br />

diets today is vegetable n-6 PUFAs, mainly linoleic acid [3]. This is<br />

largely due to the high amount <strong>of</strong> linoleic acid in corn-, sunflower-,<br />

and soybean-oil used in both home-cooking and in industrially<br />

prep<strong>are</strong>d food [4]. Moreover, animal feeds <strong>are</strong> enriched with n-6<br />

PUFAs, and although meat production methods <strong>are</strong> diverse, meat<br />

<strong>fatty</strong> acid pr<strong>of</strong>iles will always reflect that <strong>of</strong> the animal feed [4].<br />

Thus, the dietary n-3:n-6 PUFA ratio has decreased [3,4]. Although<br />

the exchange <strong>of</strong> saturated fat with vegetable n-6 PUFAs<br />

may have some beneficial <strong>effects</strong> on human health, a low n-3:n-6<br />

PUFAs ratio is associated with a high risk <strong>of</strong> several lipid-related<br />

disorders [2,3]. A high intake <strong>of</strong> n-6 PUFAs has also been associated<br />

with childhood obesity, [4,5]. Animal studies have shown<br />

that feeding mice a diet containing the n-6 PUFA, linoleic acid,<br />

during the pregnancy-lactation period leads to obesity in the<br />

<strong>of</strong>fspring [6]. This effect, however, is prevented by inclusion <strong>of</strong> the<br />

n-3 PUFA a-linolenic acid in the diet [6]. <strong>The</strong>se findings <strong>are</strong> in<br />

line with several studies demonstrating that dietary n-3 PUFAs <strong>are</strong><br />

able to limit the development <strong>of</strong> diet-induced obesity [7–13].<br />

PLoS ONE | www.plosone.org 1 June 2011 | Volume 6 | Issue 6 | e21647


Fish Oil, Sucrose and Obesity<br />

Obesity may be considered as a state <strong>of</strong> chronic low-grade<br />

inflammation [14,15]. Accumulated evidence strongly suggests<br />

that low grade chronic inflammation plays a crucial role in development<br />

<strong>of</strong> obesity related insulin resistance [16]. Furthermore,<br />

it should be noted that continuous subcutaneous infusion <strong>of</strong> lipopolysaccharide<br />

(LPS) is sufficient to induce adipose tissue inflammation,<br />

insulin resistance and obesity in mice [17]. It is also well<br />

documented that n-3 PUFAs <strong>are</strong> able to limit high fat diet-induced<br />

inflammation in adipose tissue in rodents [18–20]. Both n-3<br />

PUFAs and n-6 PUFAs <strong>are</strong> substrats for cyclo- and lipoxygenases<br />

and n-3 PUFAs <strong>are</strong> traditionally assumed to act anti-innflammatory<br />

by competitive inhibition <strong>of</strong> the biosynthesis <strong>of</strong> arachidonic<br />

acid-derived pro-inflammatory prostaglandins <strong>of</strong> the 2-series and<br />

furthermore, n-3 PUFA-derived prostaglandins <strong>of</strong> the 3-series<br />

<strong>are</strong> believed to be less inflammatory [21,22]. Recent research<br />

furthermore demonstrate that n-3 PUFAs may be converted to<br />

anti-inflammatory cyclooxygenase-2 derived electrophilic oxoderivatives<br />

and resolvins [21,22]. Moreover, by activation <strong>of</strong> the <strong>fatty</strong><br />

acid receptor, GPR120, n-3 PUFAs repress LPS- and TNFamediated<br />

inflammatory signalling responses, and thereby increase<br />

insulin sensitivity by repressing macrophage-induced adipose tissue<br />

inflammation [23]. Thus, consumption <strong>of</strong> n-6 PUFAs at the<br />

expense <strong>of</strong> n-3 PUFAs may aggravate the metabolic consequences<br />

<strong>of</strong> obesity. Increasing the dietary intake <strong>of</strong> n-3 PUFAs is therefore<br />

currently recommended by several health authorities.<br />

In order to curb the increasing obesity problem, nutritionists<br />

and authorities have largely focused on reducing fat intake, as<br />

dietary fat contains more energy per gram than proteins and carbohydrates.<br />

As an alternative to low energy diets, low carbohydrate<br />

diets <strong>are</strong> becoming increasingly popular although still controversial.<br />

<strong>The</strong> mechanisms by which such diets induce weigh loss<br />

<strong>are</strong> still not fully elucidated, but it has been documented that high<br />

protein diets increase energy expenditure in part due to a thermic<br />

effect [24]. We have previously shown that the protein:sucrose<br />

ratio in the background diet determines the adipogenic potential<br />

<strong>of</strong> dietary n-6 PUFAs in mice [25]. Mice fed n-6 PUFAs in<br />

combination with sucrose became obese, and had a markedly<br />

higher feed efficiency than mice pair-fed n-6 PUFAs in combination<br />

with proteins [25]. In fact, the high-protein fed mice needed<br />

almost 7 times more energy to achieve a weight gain <strong>of</strong> 1 g than<br />

mice on the high-sucrose diet [25]. <strong>The</strong> high protein diet led to<br />

an increased glucagon/insulin ratio, concomitant with elevated<br />

cAMP-<strong>dependent</strong> signaling, induction <strong>of</strong> COX-mediated prostaglandin<br />

synthesis and increased expression <strong>of</strong> uncoupling protein-1<br />

(UCP1) in inguinal subcutaneous white fat [25]. In the present<br />

paper we aimed to investigate whether this phenomenon is<br />

restricted to n-6 PUFAs or if the <strong>effects</strong> <strong>of</strong> dietary fats, such as fish<br />

oils, which <strong>are</strong> considered beneficial to human health, also depend<br />

on the background diets. Furthermore, we aimed to examine<br />

whether the background diet exerts an influence on the ability <strong>of</strong><br />

n-3 PUFAs to protect against glucose intolerance and adipose<br />

tissue inflammation.<br />

Results<br />

Sucrose counteracts the obesity-reducing effect <strong>of</strong> fish<br />

oil in ad libitum fed mice<br />

It is a general notion that intake <strong>of</strong> fish oil rich in n-3 PUFAs<br />

limits high fat diet-induced obesity in rodents, whereas diets rich in<br />

n-6 PUFAs have been associated with an increased propensity to<br />

develop obesity [6,26]. As we have demonstrated that the<br />

<strong>obesogenic</strong> effect <strong>of</strong> n-6 PUFAs is determined by the content <strong>of</strong><br />

carbohydrates and protein in the feed [25], we speculated whether<br />

the effect <strong>of</strong> dietary fats considered health-beneficial, such as fish<br />

oil, might be modulated by different background diets. To answer<br />

this question we fed C57BL/6J male mice isocaloric high fat diets<br />

(Table 1 and 2) containing corn oil or fish oil supplemented with<br />

either protein or sucrose or a conventional low fat diet ad libitum for<br />

9 weeks. Contrasting the general notion that fish oil attenuates<br />

high fat diet-induced obesity, the mice fed the fish oil in combination<br />

with sucrose gained as much body weight as the mice fed<br />

corn oil and sucrose (Fig. 1A and B). When combined with<br />

sucrose, fish oil did not reduce the weights <strong>of</strong> neither epididymal<br />

(eWAT) nor inguinal white adipose tissue (iWAT) mass comp<strong>are</strong>d<br />

with corn oil (Fig. 1E and F). Moreover, morphological analyses<br />

demonstrated that the adipocyte size was similar in the two sucrose<br />

fed groups (Fig. 1G). Of note, weight gain in mice fed corn oil or<br />

fish oil plus protein were indistinguishable from that <strong>of</strong> mice fed<br />

the low fat diet (Fig. 1A). Comp<strong>are</strong>d with low fat fed mice, the<br />

weights and adipocytes sizes <strong>of</strong> eWAT and iWAT in mice fed both<br />

high fat diets in combination with protein tended to be smaller, but<br />

the differences did not reach statistical significance (Fig. 1E, F<br />

and G). Thus, when combined with a high intake <strong>of</strong> sucrose fish oil<br />

did not prevent obesity. However, high dietary protein content<br />

prevented weight gain and obesity when combined with either<br />

corn or fish oil.<br />

Sucrose, but not protein or fat, strongly stimulates pancreatic<br />

insulin secretion, and accordingly, plasma levels <strong>of</strong> insulin were<br />

consistently higher in mice fed the sucrose-based diets than in mice<br />

fed the protein-based diets (Fig. 1C). Conversely, the levels <strong>of</strong><br />

plasma glucagon were lower and hence, the insulin:glucagon ratio<br />

was about three times higher in mice fed high sucrose than in mice<br />

fed high protein irrespective <strong>of</strong> whether the diets contained corn<br />

oil or fish oil (Fig. 1D). Collectively, these results indicate that<br />

intake <strong>of</strong> sucrose and hence increased insulin secretion, abrogates<br />

the protective <strong>effects</strong> <strong>of</strong> fish oil in relation to adipocyte hyperplasia<br />

and hypertrophy and thereby the development obesity.<br />

Sucrose counteracts the anti-inflammatory effect <strong>of</strong> fish<br />

oil in adipose tissue<br />

<strong>The</strong> ability <strong>of</strong> n-3 PUFAs to limit high fat diet-induced<br />

inflammation in adipose tissue is well documented [18–20]. As<br />

chronic low grade inflammation in adipose tissue is a characteristic<br />

trait <strong>of</strong> obesity [14,15] and sucrose abrogates the anti-adipogenic<br />

effect <strong>of</strong> fish oil, we asked whether the background diet also<br />

attenuated the ability <strong>of</strong> n-3 PUFAs to protect against adipose<br />

tissue inflammation. Gene expression analyses <strong>of</strong> eWAT and<br />

iWAT revealed a striking correlation between macrophage- and<br />

inflammatory markers and the intake <strong>of</strong> sucrose-based diets irrespectively<br />

<strong>of</strong> the fat source (Fig. 2A). Expressions <strong>of</strong> macrophage<br />

marker genes Emr1 (EGF-like module containing, mucin-like,<br />

hormone receptor-like sequence 1 or F4/80) and Cd68, as well as<br />

markers <strong>of</strong> inflammation Serpine1 (Plasminogen activator inhibitor-<br />

1) and Ccl2 (chemokine (C-C motif) ligand 2), were significantly<br />

higher in adipose tissue from mice fed sucrose than in mice fed<br />

high protein or a low fat diets (Fig. 2A). Moreover, we noticed<br />

a significant increase in the expression <strong>of</strong> Pparg (peroxisome<br />

proliferator-activated receptor c) in eWAT in the mice fed protein<br />

supplemented with corn oil comp<strong>are</strong>d with mice fed protein<br />

supplemented with fish oil (Fig. 2A).<br />

A high fat diet impairs glucose tolerance in<strong>dependent</strong> <strong>of</strong><br />

macronutrient composition and obesity<br />

As adipose tissue inflammation is causally linked to development<br />

<strong>of</strong> insulin resistance and glucose intolerance, we subjected mice fed<br />

the different diets for 9 weeks to an intraperitoneal glucose<br />

tolerance test (GTT). Surprisingly, the GTT demonstrated that<br />

PLoS ONE | www.plosone.org 2 June 2011 | Volume 6 | Issue 6 | e21647


Fish Oil, Sucrose and Obesity<br />

Table 1. Macronutrient composition in the diets.<br />

High fish oil<br />

High corn oil<br />

Low energy Sucrose Protein Sucrose Protein<br />

Protein (g/kg) 200 200 540 200 540<br />

Casein 200 200 540 200 540<br />

L-Cysteine 3 3 3 3 3<br />

Carbohydrate (g/kg) 619.5 439.5 99.5 439.5 99.5<br />

Corn starch 529.5 9.5 9.5 9.5 9.5<br />

Sucrose 90 430 90 430 90<br />

Fat (g/kg) 70 250 250 250 250<br />

Soybean oil 70 70 70 70 70<br />

Corn oil - - - 180 180<br />

Fish oil - 180 180 - -<br />

doi:10.1371/journal.pone.0021647.t001<br />

the glucose tolerance was impaired both in the mice fed the<br />

protein-based diets and in the mice fed the sucrose-based diets<br />

(Fig. 2B). Evidently, impaired glucose tolerance was dissociated<br />

from the state <strong>of</strong> obesity, suggesting that intake <strong>of</strong> relatively high<br />

amounts <strong>of</strong> fat reduces glucose tolerance even if weight gain and<br />

expression <strong>of</strong> inflammatory markers were maintained at low levels.<br />

However, fasting glucose and insulin levels were lower in mice fed<br />

high protein than high sucrose. Thus, calculation <strong>of</strong> HOMA-IR<br />

indicated that the mice fed proteins remained insulin sensitive,<br />

whereas insulin sensitivity tended to be reduced in the obese mice<br />

fed sucrose even though the difference between sucrose and<br />

protein fed mice did not reach statistical significance (Fig. 2B).<br />

Sucrose does not reduce the ability <strong>of</strong> fish oil to prevent<br />

diet-induced accumulation <strong>of</strong> fat in the liver<br />

As the anti-inflammatory effect <strong>of</strong> n-3 PUFAs in adipose tissue is<br />

well documented, we investigated if the high level <strong>of</strong> dietary<br />

sucrose reduced uptake <strong>of</strong> n-3 PUFAs. Thus, GC-MS analyses<br />

were performed to determine the <strong>fatty</strong> acid composition in red<br />

blood cells, liver and adipose tissues. <strong>The</strong>se analyses demonstrated<br />

the expected enrichment <strong>of</strong> n-3 PUFA in lipids in red blood cells<br />

and liver (Table 3 and 4). In adipose tissue, the enrichment <strong>of</strong> n-3<br />

was actually higher in mice fed the sucrose-based fish oil diet than<br />

in the protein-based fish oil diet group (Table 4). However,<br />

inclusion <strong>of</strong> sucrose in the diet did not reduce the ability <strong>of</strong> fish oil<br />

Table 2. Fatty acid composition in the diets.<br />

High fish oil<br />

High corn oil<br />

Low energy Sucrose Protein Sucrose Protein<br />

SFA (mg/g) 9.6 53.3 50.3 31.4 31.4<br />

MUFA (mg/g) 17.0 54.3 54.0 65.9 65.9<br />

PUFA (mg/g) 36.2 100.6 99.5 120.8 121.2<br />

n-6 (mg/g) 32.7 40.6 38.9 117.3 117.8<br />

n-3 (mg/g) 3.5 60.0 60.6 3.5 3.4<br />

n-3/n-6 0.11 1.48 1.56 0.03 0.03<br />

Abbreviations: SFA, saturated <strong>fatty</strong> <strong>acids</strong>; MUFA, monounsaturated <strong>fatty</strong> <strong>acids</strong>;<br />

PUFA, <strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong>.<br />

doi:10.1371/journal.pone.0021647.t002<br />

to prevent accumulation <strong>of</strong> fat in the liver (Fig. 3A). When sucrose<br />

was included in the diet, lipid accumulation in livers from fish oil<br />

fed mice was significantly lower than in livers from mice fed corn<br />

oil (Fig. 3A). Moreover, expressions <strong>of</strong> lipogenic genes seem to be<br />

determined by the sucrose: protein ratio in<strong>dependent</strong> <strong>of</strong> fat source<br />

(Fig. 3B). Thus, the ability <strong>of</strong> fish oil, but not corn oil, to protect<br />

against diet-induced lipid accumulation in the liver did not seem to<br />

be directly related to the suppression <strong>of</strong> lipogenic gene expression.<br />

Other hallmarks <strong>of</strong> n-3 PUFA actions <strong>are</strong> their ability to<br />

increase <strong>fatty</strong> acid oxidation and to reduce plasma triacylglycerol<br />

levels [27,28]. Plasma triacylglycerol levels were significantly<br />

reduced in mice fed fish oil in combination with proteins, but<br />

inclusion <strong>of</strong> sucrose abrogated this effect (Fig. 3C). <strong>The</strong> higher<br />

plasma levels <strong>of</strong> b-hydroxybutyrate in mice fed fish oil in<br />

combination with proteins indicated that hepatic <strong>fatty</strong> acid<br />

oxidation was increased in these mice, and inclusion <strong>of</strong> sucrose<br />

attenuated this effect (Fig. 3C). Together these results demonstrate<br />

that the protein:sucrose ratio also affects the ability <strong>of</strong> fish oil to<br />

reduce plasma levels <strong>of</strong> triacylglycerol and increase <strong>fatty</strong> acid<br />

oxidation.<br />

Increasing the protein:sucrose ratio in a high fat diet<br />

decreases energy efficiency irrespective <strong>of</strong> corn or fish oil<br />

supplementation<br />

To verify that the obesity in mice fed fish oil in combination<br />

with sucrose was simply not due to increased energy-intake, feed<br />

intake was recorded and energy efficiency calculated. Obviously,<br />

energy intake was significantly higher in mice fed high fat diets<br />

than that <strong>of</strong> mice receiving the low fat diet (Fig. 3D). Energy intake<br />

tended to be higher in mice receiving the sucrose diets than in<br />

mice fed the protein-based diets, but this was not statistically<br />

significant (Fig. 3D). Thus, energy efficiency was dramatically<br />

increased in mice receiving sucrose comp<strong>are</strong>d to protein, indicating<br />

difference in energy expenditure. A simple way to detect<br />

differences in catabolic rate is to subject mice to fasting and<br />

measure the resulting weight loss. Figure 3D shows that mice on<br />

the protein-based diets lost significantly more weight during 18 h<br />

<strong>of</strong> fasting. This supports the notion that energy expenditure is<br />

higher in mice on a protein-based diet irrespective <strong>of</strong> whether the<br />

diet is supplemented with corn oil or fish oil.<br />

Expression and activation <strong>of</strong> UCP1 in brown and white adipose<br />

tissue lead to dissipation <strong>of</strong> energy in the form <strong>of</strong> heat, and may<br />

thus protect against diet induced obesity [29]. Gene expression<br />

analyses <strong>of</strong> adipose tissues demonstrated that expression <strong>of</strong> Ucp1<br />

(uncoupling protein-1), in mice fed high protein was higher in<br />

iWAT, but not in eWAT or (iBAT) (Fig. 3E). Increased expression<br />

<strong>of</strong> Ucp1 in iWAT in mice fed the protein-based diets was<br />

accompanied by increased expression <strong>of</strong> Cpt1b (carnitine palmitoyltransferase-1b),<br />

Ppargc1a (peroxisome proliferator-activated<br />

receptor gamma coactivator 1 alpha) and Dio2 (deiodinase, iodothyronine,<br />

type II), suggesting that iWAT adopted a more brown-like<br />

phenotype (Fig. 3E). Thus, the lean phenotype in mice fed the high<br />

protein diets, appears, at least in part, to result from increased<br />

uncoupled respiration in iWAT.<br />

<strong>The</strong> <strong>obesogenic</strong> effect <strong>of</strong> fish oil is determined by the<br />

macronutrient composition in pair-fed mice<br />

Since energy intake was slightly higher in mice receiving fish oil<br />

in combination with sucrose comp<strong>are</strong>d with protein, we decided to<br />

demonstrate directly that this difference was insufficient to account<br />

for the increased adipose tissue mass. Accordingly, mice were pairfed<br />

the isocaloric diets containing fish oil in combination with<br />

sucrose or protein. To achieve identical energy intake we recorded<br />

PLoS ONE | www.plosone.org 3 June 2011 | Volume 6 | Issue 6 | e21647


Fish Oil, Sucrose and Obesity<br />

Figure 1. Sucrose counteracts the obesity-reducing effect <strong>of</strong> fish oil in ad libitum fed mice. Male C57BL/6 mice (n = 8) were fed isocaloric<br />

high fish oil or high corn oil diets with different carbohydrate and protein contents ad libitum for 9 weeks. A: Body weight development <strong>of</strong> ad libitium<br />

fed mice. (B) Prior to termination the mice were photographed. C–D: Insulin and glucagon levels were measured in plasma in the fed state. E–G: <strong>The</strong><br />

weights <strong>of</strong> epididymal and inguinal white adipose tissues were recorded and sections were stained with hematoxylin and eosin. Data <strong>are</strong> presented<br />

as means 6 SEM. Different small letters denote significant differences between the groups (P,0.05).<br />

doi:10.1371/journal.pone.0021647.g001<br />

the ad libitum feed intake <strong>of</strong> mice receiving the protein-based diet,<br />

and restricted the amount <strong>of</strong> feed to mice receiving the sucrosebased<br />

feed accordingly. Figure 4A demonstrates that even under<br />

conditions <strong>of</strong> pair-feeding, the mice receiving high sucrose gained<br />

dramatically more weight than those receiving a high protein diet.<br />

Similarly, as observed in ad libitum fed mice, energy efficiency and<br />

adipose tissue mass were significantly higher when mice were fed<br />

sucrose (Fig. 4B and C). Moreover, energy content in the feces was<br />

similar in both groups and the app<strong>are</strong>nt digestibility was not<br />

increased by increased sucrose amount in the diet (Fig. 4B). Plasma<br />

levels <strong>of</strong> insulin were higher and glucagon lower in mice fed the<br />

sucrose than protein (Fig. 4D). In iWAT, but not eWAT, we<br />

observed a significant induction <strong>of</strong> Ppargc1a and Ucp1 expression<br />

indicative <strong>of</strong> the transformation <strong>of</strong> iWAT into a more brown-like<br />

depot in protein fed mice (Fig. 4E). In iBAT, expressions <strong>of</strong> Ucp1<br />

and cyt COXII, (cytochrome c oxidase, subunit II) a marker <strong>of</strong><br />

mitochondrial content, were not significantly different in mice fed<br />

protein or sucrose (Fig. 4F).<br />

Lower expression levels <strong>of</strong> inflammatory markers in eWAT and<br />

iWAT in protein fed mice were also confirmed (Fig. 4E). In<br />

addition, as observed in ad libitum fed mice, glucose tolerance was<br />

similarly affected in protein and sucrose fed mice (Fig. 5A). Fasting<br />

levels <strong>of</strong> insulin were higher in sucrose+fish oil fed mice than the<br />

two other groups, but an ITT test showed no significant difference<br />

between the groups (Supp Fig. 1). Plasma levels <strong>of</strong> triacylglycerol<br />

were lower and b-hydroxybutyrate were higher in plasma from<br />

mice fed fish oil in combination with protein than in mice fed fish<br />

oil in combination with sucrose (Fig. 5B). However, expression <strong>of</strong><br />

genes involved in <strong>fatty</strong> acid oxidation was not increased in liver<br />

(Fig. 6B) or in muscle (not shown). Actually, expression <strong>of</strong> the<br />

classical PPARa target Acox1 (acyl-CoA oxidase 1) was higher in<br />

liver <strong>of</strong> sucrose fed mice (Fig. 6B). Thus, a possible increase in <strong>fatty</strong><br />

acid oxidation in protein fed mice as indicated by the elevated<br />

levels <strong>of</strong> b-hydroxybutyrate did not appear to be due to increased<br />

expression <strong>of</strong> genes involved in <strong>fatty</strong> acid oxidation. Of note,<br />

however, higher expressions <strong>of</strong> Srebf1 (sterol regulatory element<br />

binding transcription factor 1) as well as Acaca (acetyl-Coenzyme A<br />

carboxylase alpha) and Fasn (<strong>fatty</strong> acid synthase), indicate that<br />

sucrose overrides the suppressive effect <strong>of</strong> fish oil on lipogenic gene<br />

expression.<br />

Energy expenditure is reduced in mice fed fish oil in<br />

combination with sucrose<br />

Mice fed fish oil in combination with sucrose exhibited<br />

increased weight gain and an increased feed efficiency indicative<br />

<strong>of</strong> decreased energy expenditure. Moreover, mice fed fish oil in<br />

combination with sucrose lost significantly less weight during 18 h<br />

<strong>of</strong> fasting (Fig. 3C). <strong>The</strong>refore, we examined whether energy<br />

PLoS ONE | www.plosone.org 4 June 2011 | Volume 6 | Issue 6 | e21647


Fish Oil, Sucrose and Obesity<br />

Figure 2. A high fat diet impairs glucose tolerance in<strong>dependent</strong> <strong>of</strong> macronutrient composition and obesity. A: Expressions <strong>of</strong><br />

adipogenic and inflammatory marker genes (Pparg (peroxisome proliferator activated receptor c), Adipoq (adiponectin), Serpine1 (Plasminogen<br />

activator inhibitor-1), Ccl2 (chemokine (C-C motif) ligand 2), Emr1 (EGF-like module containing, mucin-like, hormone receptor-like sequence 1 or F4/<br />

80) and Cd68 (CD68 antigen)) were measured in epididymal and inguinal white adipose tissue using RT-qPCR (n = 8). B: Intraperitoneal glucose<br />

tolerance test was performed in a separate set <strong>of</strong> mice (n = 10). Fasting glucose and insulin levels were measured to calculate HOMA-IR. Data <strong>are</strong><br />

presented as means 6 SEM. Different small letters denote significant differences between the groups, in 2A within the same tissue (P,0.05).<br />

doi:10.1371/journal.pone.0021647.g002<br />

expenditure was reduced when fish oil was combined with sucrose.<br />

Accordingly, O 2 consumption and CO 2 production were measured<br />

by indirect calorimetry. Figure 5C shows that O 2 consumption<br />

both in the light and the dark periods tended to be lower<br />

in mice fed fish oil in combination with sucrose than with protein.<br />

As expected, mice fed fish oil in combination with sucrose had a<br />

higher CO 2 production resulting in a statistically significant higher<br />

RER <strong>of</strong> about 0.9 indicating a lower rate <strong>of</strong> <strong>fatty</strong> acid oxidation<br />

(Fig. 5C).<br />

A diet enriched with fish oil and proteins increases<br />

gluconeogenesis<br />

High circulating levels <strong>of</strong> insulin combined with a low level <strong>of</strong><br />

glucagon translate into reduced cAMP signalling in the liver.<br />

Thus, the observed reduced expressions <strong>of</strong> Crem (cAMP responsive<br />

element modulator), Pde4c (phosphodiesterase 4C, cAMP specific),<br />

Ppargc1a and Pck1 (phosphoenolpyruvate carboxykinase 1, cytosolic)<br />

as well as reduced expressions <strong>of</strong> enzymes involved in amino<br />

acid degradation in the liver <strong>of</strong> sucrose fed mice were anticipated<br />

(Fig. 4D). In the liver PGC1a is induced in response to elevated<br />

levels <strong>of</strong> cAMP and plays a central role in the control <strong>of</strong> hepatic<br />

gluconeogenesis [30–32]. In keeping with the increased expressions<br />

<strong>of</strong> Ppargc1a and Pck1 in liver from mice fed fish oil in<br />

combination with protein comp<strong>are</strong>d to sucrose, we anticipated<br />

that gluconeogenesis was induced in the fed state in the protein fed<br />

mice. To measure gluconeogenesis in vivo mice fed fish oil in<br />

combination with either protein or sucrose were intraperitoneally<br />

injected with pyruvate both after overnight fasting and in the fed<br />

PLoS ONE | www.plosone.org 5 June 2011 | Volume 6 | Issue 6 | e21647


Fish Oil, Sucrose and Obesity<br />

Table 3. Fatty acid composition in red blood cells.<br />

High fish oil<br />

High corn oil<br />

Low energy Sucrose Protein Sucrose Protein<br />

Sum total (mg/g) 3.41±0.04 3.04±0.07 2.87±0.05 3.52±0.11 3.34±0.10<br />

SFA (mg/g) 1.28±0.02 1.24±0.03 1.17±0.02 1.31±0.05 1.21±0.04<br />

MUFA (mg/g) 0.49±0.01 0.34±0.01 0.35±0.01 0.41±0.01 0.40±0.01<br />

PUFA (mg/g) 1.39±0.02 1.28±0.03 1.19±0.02 1.53±0.05 1.44±0.04<br />

n-3 (mg/g) 0.2560.00 0.7960.02 0.7360.01 0.2160.01 0.1160.00<br />

n-6 (mg/g) 1.1460.02 0.4960.01 0.4760.01 1.3260.05 1.3460.04<br />

n-3/n-6 0.2260.00 1.6260.01 1.5560.28 0.1660.00 0.0860.00<br />

Data <strong>are</strong> presented as mean 6 SEM (n = 8).<br />

doi:10.1371/journal.pone.0021647.t003<br />

Table 4. Fatty acid composition in organs.<br />

High fish oil<br />

High corn oil<br />

Low energy Sucrose Protein Sucrose Protein<br />

Liver<br />

Sum total 45±4 41±1 35±2 71±10 42±3<br />

(mg/g)<br />

% 100 100 100 100 100<br />

SFA (mg/g) 15±1 15±0 12±1 21±3 13±1<br />

% 32.560.9 36.560.4 35.260.3 29.660.3 31.360.8<br />

MUFA (mg/g) 13±2 6±1 4±0 19±4 7±1<br />

% 29.161.6 14.962.3 11.960.7 25.661.9 17.261.3<br />

PUFA (mg/g) 16±1 19±1 18±1 29±3 20±1<br />

% 36.761.6 47.262.0 51.260.4 42.761.9 47.860.9<br />

n-3 (mg/g) 3.2460.27 12.9460.51 10.8360.72 3.2860.15 1.7260.11<br />

% 7.2560.45 31.4161.25 31.3460.35 4.9760.53 4.1060.28<br />

n-6 (mg/g) 1361 660 760 2663 1861<br />

% 29.461.2 15.860.8 19.960.5 37.761.4 43.760.7<br />

n-3/n-6 0.2560.01 2.0060.03 1.5860.05 0.1360.01 0.0960.01<br />

eWAT<br />

Sum total 871±14 841±17 761±78 840±27 747±43<br />

(mg/g)<br />

% 100 100 100 100 100<br />

SFA (mg/g) 198±6 250±8 208±23 155±8 120±7<br />

% 22.760.3 29.760.5 27.360.6 18.460.6 16.160.2<br />

MUFA (mg/g) 372±9 269±6 268±26 305±7 279±12<br />

% 42.760.4 32.160.7 35.461.1 36.460.6 37.560.8<br />

PUFA (mg/g) 295±2 308±8 271±29 375±14 341±24<br />

% 33.960.6 36.660.5 35.560.7 44.560.4 45.460.7<br />

n-3 (mg/g) 18.1660.47 114.8966.64 87.07613.22 8.6860.32 6.2560.54<br />

% 2.0860.04 13.6360.59 11.2060.89 1.0360.02 0.8360.04<br />

n-6 (mg/g) 27762 19063 181618 366614 322622<br />

% 31.860.6 22.660.5 23.960.6 43.560.4 44.660.7<br />

n-3/n-6 0.0760.00 0.6060.03 0.4760.05 0.0260.00 0.0260.00<br />

Data <strong>are</strong> presented as mean 6 SEM (n = 8).<br />

doi:10.1371/journal.pone.0021647.t004<br />

state, and blood glucose was measured in the following 60 minutes.<br />

In the fasted state, mice fed sucrose or protein exhibited similar<br />

excursions, indicating similar rates <strong>of</strong> gluconeogenesis (Fig. 6A). In<br />

fed mice, however, the rise <strong>of</strong> blood glucose following the injection<br />

<strong>of</strong> pyruvate was dramatically faster and reached much higher levels<br />

after 15 and 30 min in the protein fed mice than in chow fed mice<br />

(Fig. 6A). Comp<strong>are</strong>d with chow fed mice the rise in blood glucose<br />

was also increased in mice fed fish oil in combination with sucrose,<br />

but this was not statistically significant (Fig. 6A). <strong>The</strong> decline in<br />

blood glucose in chow fed mice remains to be explained, but this<br />

was observed consistently. Taken together these results strongly<br />

support the assumption that gluconeogenesis is markedly induced in<br />

mice fed the protein-based diet.<br />

Discussion<br />

It is well documented that inclusion <strong>of</strong> n-3 PUFAs in high fat<br />

diets leads to reduced development <strong>of</strong> diet-induced obesity in<br />

rodents [7–9,11–13,33]. Unfortunately, not all studies where the<br />

anti-<strong>obesogenic</strong> <strong>effects</strong> <strong>of</strong> fish oils <strong>are</strong> studied provide a detailed<br />

description <strong>of</strong> the macronutrient composition. However, in<br />

standard commercial available high fat- and very high fat diets,<br />

starch is the most abounded carbohydrate source and the amount<br />

<strong>of</strong> sucrose is low or absent. Here we show that a high amount <strong>of</strong><br />

sucrose in the diet counteracts the obesity-reducing effect <strong>of</strong> fish oil<br />

as well as the well described anti-inflammatory effect in adipose<br />

tissue [19,23,34,35]. Irrespective <strong>of</strong> the <strong>fatty</strong> acid source, mice fed<br />

high protein diets remained lean whereas mice fed diets enriched<br />

in sucrose became obese and had higher expressions <strong>of</strong> inflammatory<br />

markers in adipose tissue. Collectively, our results demonstrate<br />

that a high intake <strong>of</strong> sucrose abrogates the protective <strong>effects</strong><br />

<strong>of</strong> fish oil in development <strong>of</strong> obesity.<br />

As dietary sucrose, but not protein or fat, stimulates secretion <strong>of</strong><br />

insulin from pancreatic b-cells, an increased dietary sucrose:protein<br />

ratio will translate into an increased insulin:glucagon ratio in<br />

the fed state. In this respect the observed higher insulin:glucagon<br />

ratio in mice fed the sucrose-based diets than in mice fed the<br />

protein-based diets was expected. Increased levels <strong>of</strong> insulin in fed<br />

mice were observed irrespectively <strong>of</strong> the type <strong>of</strong> fat in the diet.<br />

Insulin is a powerful anabolic hormone that stimulates adipocyte<br />

differentiation and adipose tissue expansion [36]. Activation <strong>of</strong><br />

insulin signaling is crucial for the development <strong>of</strong> obesity [37] and<br />

insulin receptor substrate-1 (IRS-1) transgenic mice <strong>are</strong> obese [38].<br />

Increased insulin signaling and glucose uptake in adipose tissue<br />

in the fed state in sucrose fed mice may thus override the<br />

protective effect <strong>of</strong> fish oil when it comes to protection against<br />

PLoS ONE | www.plosone.org 6 June 2011 | Volume 6 | Issue 6 | e21647


Fish Oil, Sucrose and Obesity<br />

Figure 3. Fish oil prevents diet-induced accumulation <strong>of</strong> fat in the liver. A: Total lipids were extracted from liver and separated using HPTLC.<br />

B: Expressions <strong>of</strong> lipogenic genes (Srebf1 (sterol regulatory element binding transcription factor 1) and Acaca (acetyl-Coenzyme A carboxylase alpha)<br />

were measured by RT-qPCR. C: Plasma triacylglycerol and b-hydroxybutyrate were measured in the fed state. D: Energy efficiency was calculated<br />

based on energy intake and body weight gain. E: Expression levels <strong>of</strong> brown adipose tissue marker genes (Ucp1 (Uncoupling protein-1), Ppargc1a<br />

(peroxisome proliferator-activated receptor gamma, coactivator 1 alpha), Cpt1b (carnitine palmitoyltransferase-1b) and Dio2 (deiodinase,<br />

iodothyronine, type II) were measured in white adipose tissues using RT-qPCR. Data <strong>are</strong> presented as means 6 SEM (n = 8). Different small letters<br />

denote significant differences between the groups, in 3E within the same tissue (P,0.05).<br />

doi:10.1371/journal.pone.0021647.g003<br />

obesity-development. It should also be mentioned that although<br />

several studies have demonstrated a protective effect <strong>of</strong> fish oil in<br />

obesity-development, it has been reported that inclusion <strong>of</strong> fish oil<br />

increased the amount <strong>of</strong> adipose tissue mass in hyperinsulinemic<br />

ob/ob mice [19].<br />

Differences in the insulin:glucagon ratio and hence differences<br />

in cAMP-<strong>dependent</strong> signaling may at least in part orchestrate the<br />

observed differences in energy homeostasis between the sucroseand<br />

protein-based diets regardless <strong>of</strong> whether these diet <strong>are</strong><br />

supplemented with corn oil or fish oil. In the liver, Ppargc1a is<br />

induced in response to elevated levels <strong>of</strong> cAMP and plays a central<br />

role in the control <strong>of</strong> hepatic gluconeogenesis [30–32]. High<br />

circulating levels <strong>of</strong> insulin combined with a low level <strong>of</strong> glucagon<br />

translate into reduced cAMP signalling in the liver. Thus, the<br />

observed increased gluconeogenesis in the fed state in protein fed<br />

mice may result from cAMP-mediated stimulation <strong>of</strong> Ppargc1a and<br />

Pck-1 expression. Increased gluconeogenesis in the fed state may<br />

contribute to the observed lower energy efficiency in protein fed<br />

PLoS ONE | www.plosone.org 7 June 2011 | Volume 6 | Issue 6 | e21647


Fish Oil, Sucrose and Obesity<br />

Figure 4. Sucrose counteracts the obesity-reducing effect <strong>of</strong> fish oil in pair-fed mice. Male C57BL/6 mice (n = 8) were pair-fed isocaloric high<br />

fish oil diets with different carbohydrate and protein contents for 8 weeks. A: Body weight development was followed throughout the feeding<br />

regime. B: Energy efficiency was calculated based on energy intake, weight gain and app<strong>are</strong>nt digestibility. C: <strong>The</strong> weights <strong>of</strong> different adipose tissue<br />

depots were recorded. D: Insulin and glucagon levels were measured in plasma in the fed state. E: Inflammation and adipocyte marker genes (Pparg<br />

(peroxisome proliferator-activated receptor c), Adipoq (adiponectin), Serpine1 (Plasminogen activator inhibitor-1), Ccl2 (chemokine (C-C motif) ligand<br />

2), Emr1 (EGF-like module containing, mucin-like, hormone receptor-like sequence 1 or F4/80) and Cd68 (CD68 antigen) and F: thermogenesis-related<br />

genes (Ucp1 (Uncoupling protein-1) and cyt COXII, (cytochrome c oxidase, subunit II) were measured by RT-qPCR in adipose tissues. Data <strong>are</strong><br />

presented as means 6 SEM. Different small letters denote significant differences between the groups, in 4E within the same tissue (P,0.05).<br />

doi:10.1371/journal.pone.0021647.g004<br />

mice, as 6 ATP molecules <strong>are</strong> consumed per molecule <strong>of</strong> glucose<br />

synthesized from pyruvate, rendering gluconeogenesis an energyconsuming<br />

process. Moreover, concomitant increased expressions<br />

<strong>of</strong> Gpt, Got1, Agxt and Cps1 suggest that energy consuming<br />

processes such as amino acid degradation and ureagenesis <strong>are</strong><br />

higher in protein than sucrose fed mice. As mammals have no<br />

direct storage capacity for protein it needs to be metabolically<br />

processed immediately. <strong>The</strong> high cost <strong>of</strong> urea production and<br />

gluconeogenesis is actually <strong>of</strong>ten cited reasons for the higher<br />

thermic effect <strong>of</strong> protein than other macronutrients [39,40] and<br />

this may partly explain why diets higher in protein exert a larger<br />

effect on energy expenditure than diets lower in protein [24].<br />

A second mechanism by which a low sucrose:protein ratio in the<br />

diet leads to reduced energy efficiency may be related to the observed<br />

expression <strong>of</strong> Ucp1 in iWAT. Increased cAMP-signaling is known to<br />

induce adaptive thermogenesis by induction <strong>of</strong> Ppargc1a and Ucp1<br />

expression and it is well known that the UCP1 protein allows<br />

dissipation <strong>of</strong> energy in the form <strong>of</strong> heat [41]. Of note, acute or<br />

chronic upregulation <strong>of</strong> <strong>fatty</strong> acid oxidation alone, that is increased<br />

<strong>fatty</strong> acid oxidation without a concomitant uncoupling <strong>of</strong> mitochondria,<br />

has no net effect on whole-body energy expenditure or adiposity<br />

[42]. Although Ucp1 expression was unchanged in iBAT, whole body<br />

energy homeostasis may be influenced by increased expression in<br />

iWAT. In fact, increased occurrence <strong>of</strong> brown-like adipocytes within<br />

WAT depots is a feature <strong>of</strong> mouse strains resistant to dietary obesity,<br />

such as the A/J strain [43] and reduced adiposity associated with aP2-<br />

transgenic expression <strong>of</strong> Ucp1 is linked to increased energy dissipation<br />

in white, but not interscapular brown, adipose tissue [29].<br />

Conversely, inhibition <strong>of</strong> diet-induced expression <strong>of</strong> Ucp1 in iWAT<br />

in Sv129 mice by administration <strong>of</strong> a general cyclooxygenase<br />

inhibitor accentuates obesity-development [44].<br />

Our finding that inclusion <strong>of</strong> sucrose abolishes the anti-obesity<br />

effect <strong>of</strong> fish oil seems to contradict a recent study from Sato et al.<br />

[45], as these authors demonstrated that inclusion <strong>of</strong> 5% the n-3<br />

PUFA EPA (eicosapentaenoic acid) into a high fat-high sucrose<br />

diet reduced body weight gain in mice. <strong>The</strong> reason for this<br />

discrepancy is not clear, but different dietary compositions as well<br />

as doses and type <strong>of</strong> n-3 PUFAs may account for the different<br />

PLoS ONE | www.plosone.org 8 June 2011 | Volume 6 | Issue 6 | e21647


Fish Oil, Sucrose and Obesity<br />

Figure 5. Metabolic parameters in mice fed fish oil in combination with sucrose or protein. A. Intraperitoneal glucose tolerance test was<br />

performed in mice pair fed fish oil enriched diets (n = 10). B. b-hydroxybutyrate, triacylglycerol, glycerol and free <strong>fatty</strong> <strong>acids</strong> were measured in pair-fed<br />

mice in both fasted and fed state (n = 10). C. Oxygen consumption, carbon dioxide and respiratory exchange ratio were measured during a 24-h<br />

period with indirect calorimetry (n = 8). Data <strong>are</strong> presented as means 6 SEM. Different small letters denote significant differences between the<br />

groups, in 5B between fasted or fed state (P,0.05).<br />

doi:10.1371/journal.pone.0021647.g005<br />

results obtained. <strong>The</strong> amount <strong>of</strong> n-3 PUFAs used in this study is<br />

slightly higher (6% n-3 <strong>fatty</strong> <strong>acids</strong>) than the 5% EPA used by Sato<br />

et al.. However, whereas Sato et al. used EPA, the n-3 PUFAs used<br />

in our study comprise a mixture (there<strong>of</strong> 3263 g/kg and 1863 g/<br />

kg EPA and DHA (docosahexaenoic acid), respectively). Moreover,<br />

in fish oil as used in our study, the n-3 PUFAs <strong>are</strong> present in<br />

the form <strong>of</strong> triacylglycerols, whereas Sato et al. used purified EPA<br />

ethyl ester. It should also be mentioned that the main fat source in<br />

the diets used in our study is corn-oil rich in n-6 <strong>fatty</strong> <strong>acids</strong>,<br />

whereas Sato et al. used anhydrate milk fat containing more than<br />

60% saturated fat. Last, the amount <strong>of</strong> sucrose used in our study is<br />

higher than the dose used by Sato et al. It is worth noting, however,<br />

that both the study by Sato et al. and our study demonstrated<br />

that sucrose did not reduce the ability <strong>of</strong> fish oil and/or EPA to<br />

prevent diet induced accumulation <strong>of</strong> fat in the liver.<br />

A strong association between obesity and adipose tissue inflammation<br />

exists and obesity is characterized by chronic low-grade<br />

inflammation in adipose tissues [14,15]. In light <strong>of</strong> this it may not<br />

be surprising that expression <strong>of</strong> macrophage and inflammatory<br />

marker genes was elevated in obese mice comp<strong>are</strong>d to lean mice.<br />

Still, as the anti-inflammatory effect <strong>of</strong> fish oil in adipose tissue is<br />

well described [19,23,34,35], it was unexpected that the expression<br />

<strong>of</strong> inflammatory markers was similar in adipose tissue from obese<br />

corn oil and the fish oil fed groups. In our study the state <strong>of</strong> obesity<br />

rather than the n-3:n-6 PUFA ratio in both feed and adipose<br />

tissues appe<strong>are</strong>d to determine the expression levels <strong>of</strong> inflammatory<br />

markers in adipose tissue.<br />

Chronic low grade inflammation plays an important role in<br />

development <strong>of</strong> insulin resistance [46,47]. Pioneering work by<br />

Storlien et al. [48], later confirmed by several others [13,23,35,<br />

PLoS ONE | www.plosone.org 9 June 2011 | Volume 6 | Issue 6 | e21647


Fish Oil, Sucrose and Obesity<br />

Figure 6. Gluconeogenesis is increased in fed state when animals <strong>are</strong> fed fish oil supplemented with protein. A. Pyruvate tolerance<br />

tests were performed on mice in 16 h fasted (n = 7) and fed states (n = 10). B. Hepatic gene expression (Crem (cAMP responsive element modulator),<br />

Pde4c (phosphodiesterase 4C, cAMP specific), Ppargc1a (peroxisome proliferator-activated receptor gamma, coactivator 1 alpha), Pck1<br />

(phosphoenolpyruvate carboxykinase 1, cytosolic), Gpt (glutamic pyruvic transaminase), Got1 (glutamate oxaloacetate transaminase 1), Agxt<br />

(alanine-glyoxylate aminotransferase), Cps1 (carbamoyl-phosphate synthetase 1), Acox1 (acyl-CoA oxidase 1), Cpt1a (carnitine palmitoyltransferase<br />

1a), Cpt2 (carnitine palmitoyltransferase 2), Hmgcs2 (3-hydroxy-3-methylglutaryl-Coenzyme A synthase 2), Srebf1 (sterol regulatory element binding<br />

transcription factor 1), scd1 (stearoyl-Coenzyme A desaturase 1), Acaca (acetyl-Coenzyme A carboxylase alpha), Fasn (<strong>fatty</strong> acid synthase), was<br />

measured using RT-qPCR (n = 8). Data <strong>are</strong> presented as means 6 SEM. Different small letters denote significant differences between the different<br />

groups (P,0.05).<br />

doi:10.1371/journal.pone.0021647.g006<br />

49,50] has demonstrated that n-3 PUFAs can prevent development<br />

<strong>of</strong> diet-induced insulin resistance in rodents. <strong>The</strong> insulin<br />

sensitizing effect <strong>of</strong> n-3 PUFAs is generally accepted to be related<br />

to the anti-inflammatory effect, recently demonstrated to be mediated<br />

by the GPR120 receptor [23]. Calculation <strong>of</strong> HOMA-IR<br />

indicated that mice fed high levels <strong>of</strong> proteins were more<br />

insulin sensitive than mice fed sucrose, but no significant differences<br />

was observed in an ITT. Similar to expression levels <strong>of</strong><br />

inflammatory markers in adipose tissue, this was irrespective <strong>of</strong><br />

whether the diets were supplemented with corn oil or fish oil. It<br />

was therefore unexpected that the GTT demonstrated that mice<br />

fed corn oil or fish oil in combination with sucrose or protein<br />

exhibited impaired glucose tolerance irrespective <strong>of</strong> whether or not<br />

the mice remained lean. It is possible that different mechanisms<br />

underlay the impaired glucose tolerance observed in the sucrose<br />

and the protein fed mice. It is likely that impaired glucose<br />

tolerance in sucrose fed mice is related to the obese state. Of note,<br />

in fish oil and protein fed mice, glucose tolerance was impaired<br />

even if weight gain and inflammation were maintained at low<br />

levels. Further studies <strong>are</strong> required to elucidate the mechanisms<br />

underlying the impaired glucose tolerance in these mice, but the<br />

possibility that adaption to a low carbohydrate intake with concomitant<br />

high hepatic gluconeogenesis and glucose output should<br />

be considered.<br />

Seen as a whole, our study indicate that the sucrose:protein<br />

ratio, rather than the n-6:n-3 PUFA ratio in the diet determines<br />

development <strong>of</strong> obesity, adipose tissue inflammation and glucose<br />

intolerance. Activation <strong>of</strong> the NF-kB system appears to represent a<br />

link between obesity, inflammation <strong>of</strong> adipose tissue and insulin<br />

resistance [51–53]. Insulin is able to activate NF-kB by phosporylation<br />

<strong>of</strong> IkBa in different cell systems [54], thus, high levels <strong>of</strong><br />

circulating insulin may activate the NF-kB system also in adipose<br />

tissue. Whether increased insulin levels in sucrose fed mice<br />

translated into activation <strong>of</strong> the NF-kB system in adipose tissue in<br />

these mice will require further investigation.<br />

Together our results demonstrate that the background diet<br />

exerts a crucial influence on the ability <strong>of</strong> n-3 PUFAs to protect<br />

against development <strong>of</strong> obesity, glucose intolerance and adipose<br />

tissue inflammation. High levels <strong>of</strong> dietary sucrose counteract the<br />

anti-inflammatory effect <strong>of</strong> fish oil in adipose tissue and promote<br />

PLoS ONE | www.plosone.org 10 June 2011 | Volume 6 | Issue 6 | e21647


Fish Oil, Sucrose and Obesity<br />

obesity development in mice. As the intake <strong>of</strong> sucrose in Western<br />

societies is high and increasing dietary intake <strong>of</strong> n-3 PUFAs is<br />

recommended by several health authorities it would be <strong>of</strong> importance<br />

to investigate whether the background diet influences the<br />

effect <strong>of</strong> fish oil also in humans.<br />

Materials and Methods<br />

Ethics Statement<br />

All animal experiments were approved by National Animal<br />

Health Authorities (Norwegain approval identification: 1840 and<br />

1841). C<strong>are</strong> and handling were in accordance with local institutional<br />

recommendations and rules. Adverse events were not<br />

observed.<br />

Animals and diets<br />

Male C57BL/6JBomTac mice approximately 8 weeks <strong>of</strong> age<br />

were obtained from Taconic Europe (Ejby, Denmark) and were<br />

divided into groups (n = 6–10). <strong>The</strong> mice were kept at a 12 h light/<br />

dark cycle at 28uC. After acclimatization the animals were fed ad<br />

libitum or pair-fed experimental diets obtained from Ssniff<br />

Spezialdiäten GmbH (Soest, Germany) described in Table 1 and<br />

2 for 8–10 weeks. All diets were supplemented with 3 g/kg L-<br />

cysteine, 10 g/kg choline bitartrate, 10 g/kg Vitamin mix AIN 76<br />

A, 45 g/kg Mineral mix AIN 93 and 0.014 g/kg t-butylhydroquinone.<br />

Mice were euthanized by cardiac puncture under<br />

anesthesia with Is<strong>of</strong>lurane (Isoba-vet, Schering-Plough, Denmark)<br />

using the Univentor 400 Anaesthesia Unit (Univentor Limited,<br />

Sweeden) in the fed state and plasma prep<strong>are</strong>d from blood. Tissues<br />

were dissected out, freeze-clamped and frozen at 280uC.<br />

Indirect calorimetry<br />

<strong>The</strong> metabolic rate <strong>of</strong> mice was measured by indirect calorimetry<br />

in open circuit chambers <strong>of</strong> Labmaster system (TSE Systems<br />

GmbH, Germany). <strong>The</strong> animals were acclimated in the chambers<br />

for 24 hours and measured continuously for another 24 hours.<br />

Glucose, insulin and pyruvate tolerance testing (GTT, ITT<br />

and PTT)<br />

GTT: mice were fasted 6 hours before intraperitoneal injection<br />

<strong>of</strong> 2 g/kg glucose in saline. ITT: mice were fasted 4 hours before<br />

i.p. injection <strong>of</strong> 0.5 Unit/kg human recombinant insulin in saline.<br />

PTT: mice in the fed state or mice that were fasted overnight were<br />

injected i.p. with <strong>of</strong> 2 g/kg pyruvate in saline. Blood was collected<br />

from the lateral tail vein at indicated time points and measured<br />

with Bayer Contour glucometer (Bayer A/S, Denmark).<br />

Plasma analyses<br />

Insulin, glucagon and glucose [25] and lipid metabolites [55]<br />

were measured in plasma as earlier described.<br />

Lipid analyses<br />

Total lipids were extracted from diets, red blood cells, liver and<br />

adipose tissue samples with chlor<strong>of</strong>orm: methanol, 2:1 (v/v). Lipid<br />

classes were analyzed using an automated High-Performance Thin<br />

References<br />

1. Erkkilä A, de Mello VDF, Risérus U, Laaksonen DE (2008) Dietary <strong>fatty</strong> <strong>acids</strong><br />

and cardiovascular disease: An epidemiological approach. Progress in Lipid<br />

Research 47: 172–187.<br />

2. Harris W (2010) <strong>The</strong> Omega-3 Index: Clinical Utility for <strong>The</strong>rapeutic<br />

Intervention. Current Cardiology Reports 12: 503–508.<br />

3. Simopoulos AP (2002) <strong>The</strong> importance <strong>of</strong> the ratio <strong>of</strong> omega-6/omega-3<br />

essential <strong>fatty</strong> <strong>acids</strong>. Biomedecine & Pharmacotherapy 56: 365–379.<br />

Layer Chromatography (HPTLC) system (Camaq, Switzerland)<br />

and separated on HPTLC plates coated with silica gel 60 F [55]<br />

whereas <strong>fatty</strong> acid composition <strong>of</strong> total lipids was analyzed on a<br />

capillary gas chromatograph with flame ionization detector (Perkin<br />

Elmer, USA) [56].<br />

Histology<br />

Sections <strong>of</strong> adipose tissue were fixed, dehydrated, embedded in<br />

paraffin blocks, cut into 3 mm thick section and stained with eosin<br />

and hematoxylin as previously described [57]. Sections were visually<br />

examined using an Olympus BX 51 binocular microscope (System<br />

microscope, Japan), fitted with an Olympus DP50 3.0 camera.<br />

RT-qPCR<br />

Total RNA was purified from mouse tissue using Trizol<br />

(Invitrogen). Reverse transcription (RT) was performed and cDNA<br />

was analyzed in duplicates by qPCR using the ABI PRISM 7700<br />

Sequence Detection System (Applied Biosystems) as earlier<br />

described [58]. Primers for RT-qPCR were designed using Primer<br />

Express 2.0 (Applied Biosystems) and <strong>are</strong> available on request.<br />

Energy in faeces and diets<br />

Energy content was determined in a bomb calorimeter following<br />

the manufacturer’s instruction (Parr Instruments, Moline,<br />

IL, USA).<br />

Statistics<br />

Data represent mean 6 SEM. ANOVA, post hoc pairwise<br />

comparison: Student t-test (RT-qPCR analysis) or Tukey HSD test<br />

(GTT, ITT and organ weights). Newman-Keuls test (nonparametric<br />

due to non-homogenous variances) rest <strong>of</strong> data. Data were<br />

considered statistical significant when P,0.05).<br />

Supporting Information<br />

Figure S1 A high fish oil diet does not impair insulin tolerance.<br />

Male C57BL/6 mice (n = 7) were fed a low fat or isocaloric high<br />

fish oil diets with different carbohydrate and protein contents ad<br />

libitum for 7 weeks. A: Insulin levels were measured in the fasted<br />

state. B: Intraperitoneal insulin tolerance test was performed. Data<br />

<strong>are</strong> presented as means 6 SEM. Different small letters denote<br />

significant differences between the groups.<br />

(TIF)<br />

Acknowledgments<br />

<strong>The</strong> authors thank Åse Heltveit and Jan Idar Hjelle at NIFES for excellent<br />

assistance with animal c<strong>are</strong> and lipid analyses.<br />

Author Contributions<br />

Conceived and designed the experiments: BL KK LM. Performed the<br />

experiments: TM BL QH RKP EF HTN HHL SR SBS JTT HP LF LM.<br />

Analyzed the data: TM BL QH RKP EF HTN HHL SR SBS JTT HP LF<br />

KK LM. Wrote the paper: KK LM.<br />

4. Ailhaud G, Massiera F, Weill P, Legrand P, Alessandri JM, et al. (2006)<br />

Temporal changes in dietary fats: role <strong>of</strong> n-6 <strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong> in<br />

excessive adipose tissue development and relationship to obesity. Prog Lipid Res<br />

45: 203–236.<br />

5. Ailhaud G, Massiera F, Alessandri JM, Guesnet P (2007) Fatty acid composition<br />

as an early determinant <strong>of</strong> childhood obesity. Genes and Nutrition 2: 39–<br />

40.<br />

PLoS ONE | www.plosone.org 11 June 2011 | Volume 6 | Issue 6 | e21647


Fish Oil, Sucrose and Obesity<br />

6. Massiera F, Saint-Marc P, Seydoux J, Murata T, Kobayashi T, et al. (2003)<br />

Arachidonic acid and prostacyclin signaling promote adipose tissue development:<br />

a human health concern J Lipid Res 44: 271–279.<br />

7. Belzung F, Raclot T, Groscolas R (1993) Fish oil n-3 <strong>fatty</strong> <strong>acids</strong> selectively limit<br />

the hypertrophy <strong>of</strong> abdominal fat depots in growing rats fed high-fat diets.<br />

Am J Physiol 246: R1111–R1118.<br />

8. Wang H, Storlien LH, Huang XF (2002) Effects <strong>of</strong> dietary fat types on body<br />

fatness, leptin, and ARC leptin receptor, NPY, and AgRP mRNA expression.<br />

Am J Physiol Endocrinol Metab 282: E1352–E1359.<br />

9. Flachs P, Horakova O, Brauner P, Rossmeisl M, Pecina P, et al. (2005)<br />

Polyunsaturated <strong>fatty</strong> <strong>acids</strong> <strong>of</strong> marine origin upregulate mitochondrial biogenesis<br />

and induce beta-oxidation in white fat. Diabetologia 48: 2365–2375.<br />

10. Ruzickova J, Rossmeisl M, Prazak T, Flachs P, Sponarova J, et al. (2005)<br />

Omega-3 PUFA <strong>of</strong> marine origin limit diet-induced obesity in mice by reducing<br />

cellularity <strong>of</strong> adipose tissue (vol 39, pg 1177, 2004). Lipids 40: 115.<br />

11. Arai T, Kim HJ, Chiba H, Matsumoto A (2009) Anti-obesity effect <strong>of</strong> fish oil and fish<br />

oil-fen<strong>of</strong>ibrate combination in female KK mice. J Atheroscler Thromb 16: 675–683.<br />

12. Rokling-Andersen MH, Rustan AC, Wensaas AJ, Kaalhus O, Wergedahl H,<br />

et al. (2009) Marine omega-3 <strong>fatty</strong> <strong>acids</strong> promote size reduction <strong>of</strong> visceral<br />

adipose depots, without altering body weight and composition, in male Wistar<br />

rats fed a high-fat diet. British Journal <strong>of</strong> Nutrition 102: 995–1006.<br />

13. Samane S, Christon R, Dombrowski L, Turcotte S, Charrouf Z, et al. (2009)<br />

Fish oil and argan oil intake differently modulate insulin resistance and glucose<br />

intolerance in a rat model <strong>of</strong> dietary-induced obesity. Metabolism-Clinical and<br />

Experimental 58: 909–919.<br />

14. de Luca C, Olefsky JM (2008) Inflammation and insulin resistance. FEBS Lett<br />

582: 97–105.<br />

15. Hotamisligil GS (2006) Inflammation and metabolic disorders. Nature 444:<br />

860–867.<br />

16. Donath MY, Shoelson SE (2011) Type 2 diabetes as an inflammatory disease.<br />

Nat Rev Immunol 11: 98–107.<br />

17. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, et al. (2007) Metabolic<br />

Endotoxemia Initiates Obesity and Insulin Resistance. Diabetes 56: 1761–1772.<br />

18. Huber J, L<strong>of</strong>fler M, Bilban M, Reimers M, Kadl A, et al. (2007) Prevention <strong>of</strong><br />

high-fat diet-induced adipose tissue remodeling in obese diabetic mice by n-3<br />

<strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong>. International Journal <strong>of</strong> Obesity 31: 1004–1013.<br />

19. Todoric J, Löffler M, Huber J, Bilban M, Reimers M, et al. (2006) Adipose tissue<br />

inflammation induced by high-fat diet in obese diabetic mice is prevented by n-3<br />

<strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong>. Diabetologia 49: 2109–2119.<br />

20. Pérez-Echarri N, Pérez-Matute P, Marcos-Gómez B, Baena MJ, Marti A, et al.<br />

(2008) Differential inflammatory status in rats susceptible or resistant to dietinduced<br />

obesity: <strong>effects</strong> <strong>of</strong> EPA ethyl ester treatment. Eur J Nutr 47: 380–386.<br />

21. Calder PC (2009) Polyunsaturated <strong>fatty</strong> <strong>acids</strong> and inflammatory processes: New<br />

twists in an old tale. Biochimie 91: 791–795.<br />

22. Groeger AL, Cipollina C, Cole MP, Woodcock SR, Bonacci G, et al. (2010)<br />

Cyclooxygenase-2 generates anti-inflammatory mediators from omega-3 <strong>fatty</strong><br />

<strong>acids</strong>. Nature Chemical Biology 6: 433–441.<br />

23. Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, et al. (2010) GPR120 Is<br />

an Omega-3 Fatty Acid Receptor Mediating Potent Anti-inflammatory and<br />

Insulin-Sensitizing Effects. Cell 142: 687–698.<br />

24. Halton TL, Hu FB (2004) <strong>The</strong> Effects <strong>of</strong> High Protein Diets on <strong>The</strong>rmogenesis,<br />

Satiety and Weight Loss: A Critical Review. J Am Coll Nutr 23: 373–385.<br />

25. Madsen L, Pedersen LM, Liaset B, Ma T, Petersen RK, et al. (2008) cAMP<strong>dependent</strong><br />

Signaling Regulates the Adipogenic Effect <strong>of</strong> n-6 Polyunsaturated<br />

Fatty Acids. J Biol Chem 283: 7196–7205.<br />

26. Madsen L, Petersen RK, Kristiansen K (2005) Regulation <strong>of</strong> adipocyte<br />

differentiation and function by <strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong>. Biochimica et<br />

Biophysica Acta Molecular Basis <strong>of</strong> Disease 1740: 266–286.<br />

27. Froyland L, Madsen L, Vaagenes H, Totland GK, Auwerx J, et al. (1997)<br />

Mitochondrion is the principal target for nutritional and pharmacological<br />

control <strong>of</strong> triglyceride metabolism. J Lipid Res 38: 1851–1858.<br />

28. Madsen L, Rustan AC, Vaagenes H, Berge K, Dyroy E, et al. (1999)<br />

Eicosapentaenoic and docosahexaenoic acid affect mitochondrial and peroxisomal<br />

<strong>fatty</strong> acid oxidation in relation to substrate preference. Lipids 34:<br />

951–963.<br />

29. Kopecky J, Hodny Z, Rossmeisl M, Syrovy I, Kozak LP (1996) Reduction <strong>of</strong><br />

dietary obesity in aP2-Ucp transgenic mice: physiology and adipose tissue<br />

distribution. Am J Physiol 270: E768–E775.<br />

30. Puigserver P, Rhee J, Donovan J, Walkey CJ, Yoon JC, et al. (2003) Insulinregulated<br />

hepatic gluconeogenesis through FOXO1-PGC-1[alpha] interaction.<br />

Nature 423: 550–555.<br />

31. Yoon JC, Puigserver P, Chen G, Donovan J, Wu Z, et al. (2001) Control <strong>of</strong><br />

hepatic gluconeogenesis through the transcriptional coactivator PGC-1. Nature<br />

413: 131–138.<br />

32. Herzig S, Long F, Jhala US, Hedrick S, Quinn R, et al. (2001) CREB regulates<br />

hepatic gluconeogenesis through the coactivator PGC-1. Nature 413: 179–183.<br />

33. Ruzickova J, Rossmeisl M, Prazak T, Flachs P, Sponarova J, et al. (2005)<br />

Omega-3 PUFA <strong>of</strong> marine origin limit diet-induced obesity in mice by reducing<br />

cellularity <strong>of</strong> adipose tissue (vol 39, pg 1177, 2004). Lipids 40: 115.<br />

34. Itoh M, Suganami T, Satoh N, Tanimoto-Koyama K, Yuan X, et al. (2007)<br />

Increased adiponectin secretion by highly purified eicosapentaenoic acid in<br />

rodent models <strong>of</strong> obesity and human obese subjects. Arteriosclerosis Thrombosis<br />

and Vascular Biology 27: 1918–1925.<br />

35. Kalupahana NS, Claycombe K, Newman SJ, Stewart T, Siriwardhana N, et al.<br />

(2010) Eicosapentaenoic Acid Prevents and Reverses Insulin Resistance in High-<br />

Fat Diet-Induced Obese Mice via Modulation <strong>of</strong> Adipose Tissue Inflammation.<br />

<strong>The</strong> Journal <strong>of</strong> Nutrition 140: 1915–1922.<br />

36. Madsen L, Kristiansen K (2010) <strong>The</strong> importance <strong>of</strong> dietary modulation <strong>of</strong><br />

cAMP and insulin signaling in adipose tissue and the development <strong>of</strong> obesity.<br />

Foods for Health in the 21St Century: A Road Map for the Future 1190: 1–14.<br />

37. Bluher M, Michael MD, Peroni OD, Ueki K, Carter N, et al. (2002) Adipose<br />

Tissue Selective Insulin Receptor Knockout Protects against Obesity and<br />

Obesity-Related Glucose Intolerance. Developmental Cell 3: 25–38.<br />

38. Murata Y, Tsuruzoe K, Kawashima J, Furukawa N, Kondo T, et al. (2007) IRS-<br />

1 transgenic mice show increased epididymal fat mass and insulin resistance.<br />

Biochem Biophys Res Commun 364: 301–307.<br />

39. Mikkelsen PB, Toubro S, Astrup A (2000) Effect <strong>of</strong> fat-reduced diets on 24-h<br />

energy expenditure: comparisons between animal protein, vegetable protein,<br />

and carbohydrate. Am J Clin Nutr 72: 1135–1141.<br />

40. Robinson SM, Jaccard C, Persaud C, Jackson AA, Jequier E, et al. (1990)<br />

Protein turnover and thermogenesis in response to high-protein and highcarbohydrate<br />

feeding in men. Am J Clin Nutr 52: 72–80.<br />

41. Cannon B, Nedergaard J (2004) Brown Adipose Tissue: Function and<br />

Physiological Significance. Physiol Rev 84: 277–359.<br />

42. Hoehn KL, Turner N, Swarbrick MM, Wilks D, Preston E, et al. (2010) Acute<br />

or chronic upregulation <strong>of</strong> mitochondrial <strong>fatty</strong> acid oxidation has no net effect<br />

on whole-body energy expenditure or adiposity. Cell Metab 11: 70–76.<br />

43. Guerra C, Koza RA, Yamashita H, Walsh K, Kozak LP (1998) Emergence <strong>of</strong><br />

Brown Adipocytes in White Fat in Mice Is Under Genetic Control. Effects on<br />

Body Weight and Adiposity. J Clin Invest 102: 412–420.<br />

44. Madsen L, Pedersen LM, Lillefosse HH, Fjaere E, Bronstad I, et al. (2010)<br />

UCP1 Induction during Recruitment <strong>of</strong> Brown Adipocytes in White Adipose<br />

Tissue Is Dependent on Cyclooxygenase Activity. PLoS ONE 5.<br />

45. Sato A, Kawano H, Notsu T, Ohta M, Nakakuki M, et al. (2010) Antiobesity<br />

Effect <strong>of</strong> Eicosapentaenoic Acid in High-Fat/High-Sucrose Diet Induced<br />

Obesity Importance <strong>of</strong> Hepatic Lipogenesis. Diabetes 59: 2495–2504.<br />

46. Xu H, Barnes GT, Yang Q, Tan G, Yang D, et al. (2003) Chronic inflammation<br />

in fat plays a crucial role in the development <strong>of</strong> obesity-related insulin resistance.<br />

J Clin Invest 112: 1821–1830.<br />

47. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, et al. (2003)<br />

Obesity is associated with macrophage accumulation in adipose tissue. J Clin<br />

Invest 112: 1796–1808.<br />

48. Storlien LH, Kraegen EW, Chisholm DJ, Ford GL, Bruce DG, et al. (1987) Fish<br />

Oil Prevents Insulin Resistance Induced by High-Fat Feeding in Rats. Science<br />

237: 885–888.<br />

49. Jucker BM, Cline GW, Barucci N, Shulman GI (1999) Differential <strong>effects</strong> <strong>of</strong><br />

safflower oil versus fish oil feeding on insulin-stimulated glycogen synthesis,<br />

glycolysis, and pyruvate dehydrogenase flux in skeletal muscle - A C-13 nuclear<br />

magnetic resonance study. Diabetes 48: 134–140.<br />

50. Neschen S, Morino K, Dong JY, Wang-Fischer Y, Cline GW, et al. (2007) N-3<br />

<strong>fatty</strong> <strong>acids</strong> preserve insulin sensitivity in vivo in a peroxisonte proliferatoractivated<br />

receptor-alpha-<strong>dependent</strong> manner. Diabetes 56: 1034–1041.<br />

51. Shoelson SE, Lee J, Yuan M (2003) Inflammation and the IKKb/IkB/NF-kB<br />

axis in obesity- and diet-induced insulin resistance. Int J Obes Relat Metab<br />

Disord 27: S49–S52.<br />

52. Kim JK, Kim YJ, Fillmore JJ, Chen Y, Moore I, et al. (2001) Prevention <strong>of</strong> fatinduced<br />

insulin resistance by salicylate. J Clin Invest 108: 437–446.<br />

53. Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, et al. (2005) IKK-b links<br />

inflammation to obesity-induced insulin resistance. Nat Med 11: 191–198.<br />

54. Pandey SK, He HJ, Chesley A, Juhaszova M, Crow MT, et al. (2002)<br />

Wortmannin-Sensitive Pathway Is Required for Insulin-Stimulated Phosphorylation<br />

<strong>of</strong> InhibitorkBa. Endocrinology 143: 375–385.<br />

55. Liaset B, Madsen L, Hao Q, Criales G, Mellgren G, et al. (2009) Fish protein<br />

hydrolysate elevates plasma bile <strong>acids</strong> and reduces visceral adipose tissue mass in<br />

rats. Biochimica et Biophysica Acta 1791: 254–262.<br />

56. Arslan G, Brunborg LA, Frøyland L, Brun JG, Valen M, et al. (2002) Effects <strong>of</strong><br />

duodenal seal oil administration in patients with inflammatory bowel disease.<br />

Lipids 37: 935–940.<br />

57. Barbatelli G, Murano I, Madsen L, Hao Q, Jimenez M, et al. (2010) <strong>The</strong><br />

emergence <strong>of</strong> cold-induced brown adipocytes in mouse white fat depots is<br />

determined predominantly by white to brown adipocyte transdifferentiation.<br />

American Journal <strong>of</strong> Physiology-Endocrinology and Metabolism 298:<br />

E1244–E1253.<br />

58. Madsen L, Petersen RK, Sørensen MB, Jørgensen C, Hallenborg P, et al. (2003)<br />

Adipocyte differentiation <strong>of</strong> 3T3-L1 preadipocytes is <strong>dependent</strong> on lipoxygenase<br />

activity during the initial stages <strong>of</strong> the differentiation process. Biochem J 375:<br />

539–549.<br />

PLoS ONE | www.plosone.org 12 June 2011 | Volume 6 | Issue 6 | e21647


Figure S1.<br />

A high fish oil diet does not impair insulin tolerance. Male C57BL/6 mice (n = 7) were fed<br />

a low fat or isocaloric high fish oil diets with different carbohydrate and protein<br />

contents ad libitum for 7 weeks. A: Insulin levels were measured in the fasted state. B:<br />

Intraperitoneal insulin tolerance test was performed. Data <strong>are</strong> presented as means ± SEM.<br />

Different small letters denote significant differences between the groups.


Carbohydrate source and insulin secretion modulate the obesity promoting<br />

effect <strong>of</strong> fish oil<br />

Qin Hao 1, $ , Haldis Haukås Lillefosse 1, 2, $ , Even Fjære 1, 2, $ , Lene Secher Myrmel 1, 2 , Lisa Kolden<br />

Midtbø 1, 2 , Ragnhild Jarlsby 2 , Tao Ma 1 , Bingbing Jia 1 , Rasmus Koefoed Petersen 1 , Si Brask<br />

Sonne 1 , André Chwalibog 3 , Livar Frøyland 2 , Bjørn Liaset 2 , Karsten Kristiansen 1, * and Lise<br />

Madsen 1, 2, * .<br />

1 Department <strong>of</strong> Biology, University <strong>of</strong> Copenhagen, Denmark. 2 National Institute <strong>of</strong> Nutrition and<br />

Seafood Research, Bergen, Norway. 3 Department <strong>of</strong> Basic Animal and Veterinary Sciences,<br />

University <strong>of</strong> Copenhagen, Denmark. $ Contributed equally.<br />

*Correspondence to: Karsten Kristiansen, Department <strong>of</strong> Biology, University <strong>of</strong> Copenhagen, Ole<br />

Maaløes Vej 5, DK 2200 Copenhagen, Denmark. Fax +45 3522 2128; Phone: +45 3532 4443; E<br />

mail: kk@bio.ku.dk or Lise Madsen, National Institute <strong>of</strong> Nutrition and Seafood Research, P.O.<br />

Box 2029 Nordnes, N 5817 Bergen, Norway. Fax +47 5590 5299; Phone: +47 4147 6177; E mail:<br />

lise.madsen@nifes.no<br />

Number <strong>of</strong> figures: 7<br />

Number <strong>of</strong> tables: 2<br />

Running title: Interaction between carbohydrates and fish oil<br />

FOOTNOTES: This work was supported by the Danish Natural Science Research Council, the<br />

Novo Nordisk Foundation, the Carlsberg Foundation and NIFES. Part <strong>of</strong> the work was carried out<br />

as a part <strong>of</strong> the research program <strong>of</strong> the Danish Obesity Research Centre (DanORC). DanORC is<br />

supported by the Danish Council for Strategic Research (Grant NO 2101 06 0005). <strong>The</strong> authors<br />

have no conflicting interests.<br />

1


1<br />

2<br />

3<br />

4<br />

5<br />

6<br />

7<br />

8<br />

9<br />

10<br />

11<br />

12<br />

13<br />

14<br />

15<br />

16<br />

17<br />

18<br />

ABSTRACT<br />

Polyunsaturated n-3 <strong>fatty</strong> <strong>acids</strong> (n-3 PUFAs) <strong>are</strong> known to attenuate diet-induced obesity and<br />

adipose tissue inflammation in rodents. In this study we aimed to investigate whether inclusion <strong>of</strong><br />

different carbohydrate sources modulated the <strong>effects</strong> <strong>of</strong> n-3 PUFAs. By feeding C57BL/6J mice<br />

isocaloric high fat diets enriched with fish oil for 6 weeks, we show that increasing amounts <strong>of</strong><br />

sucrose in the diets dose-<strong>dependent</strong>ly increased energy efficiency and white adipose tissue (WAT)<br />

mass. Mice receiving fructose had about 50% less WAT mass than mice fed a high fish oil diet<br />

supplemented with either glucose or sucrose, indicating that the glucose moiety <strong>of</strong> sucrose was<br />

responsible for the obesity promoting effect <strong>of</strong> sucrose. To investigate if the <strong>obesogenic</strong> effect <strong>of</strong><br />

sucrose and glucose was related to stimulation <strong>of</strong> insulin secretion, we combined fish oil with high<br />

and low glycemic index (GI) starch. Mice receiving the high fish oil diet containing the low GI<br />

starch had significantly less WAT than mice fed high GI starch. Moreover, inhibition <strong>of</strong> insulin<br />

secretion by administration <strong>of</strong> nifedipine significantly reduced WAT mass in mice fed a high fish<br />

oil diet in combination with sucrose. Our data show that the macronutrient composition <strong>of</strong> the diet<br />

modulates the <strong>effects</strong> <strong>of</strong> fish oil. Fish oil combined with sucrose, glucose or high-glycemic index<br />

starch promotes obesity and the reported anti-inflammatory actions <strong>of</strong> fish oil <strong>are</strong> abrogated.<br />

Collectively, our data indicate that glycemic control <strong>of</strong> insulin secretion regulates the obesity<br />

promoting effect <strong>of</strong> fish oil in combination with carbohydrates.<br />

19<br />

2


20<br />

21<br />

22<br />

23<br />

24<br />

25<br />

26<br />

27<br />

28<br />

29<br />

30<br />

31<br />

32<br />

33<br />

34<br />

35<br />

36<br />

37<br />

38<br />

39<br />

40<br />

41<br />

42<br />

43<br />

Introduction<br />

A considerable number <strong>of</strong> studies has demonstrated the beneficial <strong>effects</strong> <strong>of</strong> n-3 <strong>polyunsaturated</strong><br />

<strong>fatty</strong> <strong>acids</strong> (PUFAs) on lipid metabolism and lipid-related disorders in humans (1-3) and increasing<br />

the relative amount <strong>of</strong> n-3 PUFAs is presently recommended. Moreover, diets enriched with n-3<br />

PUFAs have been demonstrated to reduce the development <strong>of</strong> insulin resistance (4-9) and to reduce<br />

the development <strong>of</strong> diet-induced obesity in rodents (7, 10-15). We have shown that the<br />

macronutrient composition <strong>of</strong> the diet determines the adipogenic potential <strong>of</strong> dietary corn oil in<br />

mice (16). Moreover, we recently demonstrated that sucrose counteracts the anti-inflammatory<br />

effect <strong>of</strong> fish oil in adipose tissue and increases obesity development in mice (17). Mice fed a fish<br />

oil enriched diet in combination with sucrose had markedly higher feed efficiency and required less<br />

than 50% <strong>of</strong> the calories to achieve the same weight gain as mice pair-fed a fish oil enriched diet in<br />

combination with protein (17).<br />

A major difference between proteins and sucrose is the ability to cause a rise in blood<br />

glucose and stimulate insulin secretion. Insulin is a powerful anabolic hormone that stimulates<br />

adipocyte differentiation and adipose tissue expansion (18), and activation <strong>of</strong> insulin signaling is<br />

crucial for the development <strong>of</strong> obesity (19). Moreover, increased insulin signaling by transgenic<br />

expression <strong>of</strong> insulin receptor substrate-1 (IRS-1) is sufficient to induce obesity (20). Thus, it is<br />

possible that increased insulin signaling and glucose uptake in adipose tissue in the fed state in<br />

sucrose fed mice may override the anti-inflammatory and anti-obesity <strong>effects</strong> <strong>of</strong> fish oil.<br />

<strong>The</strong> glucose moiety <strong>of</strong> sucrose is responsible for the rise in blood insulin upon intake<br />

<strong>of</strong> sucrose, as fructose unlike glucose, is unable to stimulate insulin secretion (21). This in part<br />

relates to the very low levels <strong>of</strong> Slc2a5 (solute carrier family 2 (facilitated glucose transporter),<br />

member 5, GLUT5) in pancreatic beta-cells (22). Furthermore, fructose does not stimulate the<br />

release <strong>of</strong> gastric inhibitory peptide which stimulates insulin secretion indirectly (23, 24). Thus, the<br />

3


44<br />

45<br />

46<br />

47<br />

48<br />

49<br />

50<br />

51<br />

52<br />

53<br />

54<br />

55<br />

56<br />

57<br />

58<br />

59<br />

60<br />

61<br />

62<br />

ability <strong>of</strong> sucrose to counteract the beneficial <strong>effects</strong> <strong>of</strong> fish oil seems to relate to a glucose<strong>dependent</strong><br />

stimulation <strong>of</strong> insulin secretion. <strong>The</strong> fructose moiety <strong>of</strong> sucrose may further modulate the<br />

effect <strong>of</strong> fish oil on the development <strong>of</strong> obesity. Thus, the increased consumption <strong>of</strong> fructose over<br />

the past decades has been linked to development <strong>of</strong> metabolic disorders (25), and fructose is<br />

routinely used to induce glucose intolerance in rats (26).<br />

As different types <strong>of</strong> starch differ in their ability to increase postprandial blood<br />

glucose and insulin secretion, different types <strong>of</strong> starch may also modulate the effect <strong>of</strong> fish oil. <strong>The</strong><br />

glycemic index (GI) is a measure <strong>of</strong> the ability <strong>of</strong> different types <strong>of</strong> carbohydrate based foods to<br />

raise blood glucose levels within 2 hours (27). <strong>The</strong> interest in low GI diets as a tool in weight<br />

management is increasing. Although reviews and meta-analyzes conclude that such diets may be<br />

effective, their effectiveness in terms <strong>of</strong> lasting weight reduction is still a matter <strong>of</strong> debate (28-32).<br />

Different types <strong>of</strong> starches with different GI <strong>are</strong> known to induce different responses in plasma<br />

glucose and insulin in rodents (33). However, it is unknown whether different types <strong>of</strong> starch<br />

modulate the <strong>effects</strong> <strong>of</strong> fish oil enriched diets. Here we have performed systematic analyses to<br />

investigate the influence <strong>of</strong> different carbohydrate sources on the reported anti-obesity and antiinflammatory<br />

effect <strong>of</strong> n-3 PUFAs in mice. By using isocaloric high fat diets enriched with n-3<br />

PUFAs, we show that the amount <strong>of</strong> sucrose dose-<strong>dependent</strong>ly increased energy efficiency and<br />

adiposity. Moreover, we show that nutritional and pharmacological control <strong>of</strong> insulin secretion play<br />

a pivotal role determining the <strong>obesogenic</strong> effect <strong>of</strong> high fish oil-high carbohydrate diets.<br />

63<br />

64<br />

65<br />

66<br />

67<br />

EXPERIMENTAL PROCEDURES<br />

Animals and diets. Male C57BL/6JBomTac mice approximately 8 weeks <strong>of</strong> age were obtained from<br />

Taconic Europe (Ejby, Denmark) and were divided into groups (n=6-10). <strong>The</strong> mice were kept at a<br />

12 h light/dark cycle at thermoneutrality. After acclimation the animals were fed ad libitum or pairfed<br />

experimental diets obtained from Ssniff (Ssniff Spezialdiäten GmbH, Soest, Germany)<br />

4


68<br />

69<br />

70<br />

71<br />

72<br />

73<br />

74<br />

75<br />

76<br />

77<br />

78<br />

79<br />

80<br />

81<br />

82<br />

83<br />

84<br />

85<br />

86<br />

87<br />

88<br />

89<br />

90<br />

described in Table 1 and 2. <strong>The</strong> fish oil enriched diets contained 62 ± 3 g/kg n-3 <strong>fatty</strong> <strong>acids</strong>, there<strong>of</strong><br />

27 ± 3 g/kg and 18 ± 3 g/kg EPA and DHA, respectively. All diets were supplemented with 3g/kg<br />

L-cysteine, 10g/kg choline bitartrate, 10g/kg vitamin mix AIN 76 A, 45 g/kg mineral mix AIN 93<br />

and 0.014 g/kg t-butylhydroquinone. As indicated Nifedipine (N7634, Sigma) was included in the<br />

diet at a dose (1g/kg) earlier demonstrated to reduce plasma insulin levels in agouti mice (34). <strong>The</strong><br />

sulfonylurea Glybenclamide (G0639, Sigma) was used as an insulin secretagogue and was<br />

administrated daily by intraperitoneal (i.p.) injection at a dose <strong>of</strong> 2 μg/g body weight..Control mice<br />

received placebo. by daily i.p. injection. Mice in the fed state were euthanized by cardiac puncture<br />

under anesthesia with is<strong>of</strong>lurane (Isoba-vet, Schering-Plough, Denmark) using the Univentor 400<br />

Anaesthesia Unit (Univentor Limited, Sweden) and plasma was prep<strong>are</strong>d from blood. Tissues were<br />

dissected out, freeze-clamped and frozen at -80 o C. All animal experiments were approved by<br />

National Animal Health Authorities (Denmark and Norway). Adverse events were not observed.<br />

Energy measurements and digestibility. Male C57BL/6JBomTac mice (n=5 for each group) were<br />

fed experimental diets ad libitum for 4 days. Faeces, urine and feed residue were collected during a<br />

24h period before respiration measurements. <strong>The</strong> nitrogen content in feed, faeces, and urine was<br />

determined using the Tecator-Kjeltec system 1026 (Tecator AB, Höganäs, Sweden). <strong>The</strong> gross<br />

energy content <strong>of</strong> feed and faeces was determined using an adiabatic bomb calorimeter (System<br />

C700, IKA Analysentechnic GmbH, Heitersheim, Germany).<br />

Glucose and insulin tolerance testing (GTT and ITT). GTT: mice were fasted 6 hours before i.p<br />

injection <strong>of</strong> 2 g/kg glucose in saline. ITT: mice were fasted 4 hours before i.p. injection <strong>of</strong> 0.5<br />

Unit/kg human recombinant insulin in saline. Blood was collected from the lateral tail vein at<br />

indicated time points and blood glucose was measured with a Bayer Contour glucometer (Bayer<br />

A/S, Denmark).<br />

5


91<br />

92<br />

93<br />

94<br />

95<br />

96<br />

97<br />

98<br />

99<br />

100<br />

101<br />

102<br />

103<br />

104<br />

105<br />

106<br />

107<br />

108<br />

109<br />

110<br />

Plasma analysis. Insulin, glucose (16) and lipid metabolites (35) were measured in plasma as earlier<br />

described.<br />

Tissue lipid analysis. Total lipids were extracted from diets and liver with chlor<strong>of</strong>orm: methanol,<br />

2:1 (v/v). Lipid classes were analyzed in liver samples using an automated High-Performance Thin<br />

Layer Chromatography (HPTLC) system (Camaq, Switzerland) and separated on HPTLC plates<br />

coated with silica gel 60 F (35). Fatty acid composition <strong>of</strong> total lipids from diets was analyzed on a<br />

capillary gas chromatograph with flame ionization detector (Perkin Elmer, USA) (36).<br />

Histology. Sections <strong>of</strong> adipose tissue were fixed, dehydrated, embedded in paraffin blocks, cut into<br />

3 μm thick sections and stained with eosin and hematoxylin as described (37). Sections were<br />

visually examined using an Olympus BX 51 binocular microscope (Olympus Corporation, Japan),<br />

fitted with an Olympus DP50 3.0 camera.<br />

RT-qPCR. Total RNA was purified from mouse tissue using Trizol (Invitrogen). Reverse<br />

transcription (RT) was performed and cDNA was analyzed in duplicates by qPCR using the ABI<br />

PRISM 7700 Sequence Detection System (Applied Biosystems) as earlier described (38). Primers<br />

for RT-qPCR were designed using Primer Express 2.0 (Applied Biosystems) and <strong>are</strong> available on<br />

request.<br />

Statistics. Data represent mean ± SEM. ANOVA, post hoc pairwise comparison: Student t-test (RTqPCR<br />

analysis) or Tukey HSD test (GTT, ITT and organ weights). Newman-Keuls test<br />

(nonparametric due to non-homogenous variances, remaining data sets. A value <strong>of</strong> P


114<br />

115<br />

116<br />

117<br />

118<br />

119<br />

120<br />

121<br />

122<br />

123<br />

124<br />

125<br />

126<br />

127<br />

128<br />

129<br />

130<br />

131<br />

132<br />

133<br />

134<br />

135<br />

136<br />

To investigate whether sucrose dose-<strong>dependent</strong>ly counteracts the obesity protective effect <strong>of</strong> fish<br />

oil, we fed for 6 weeks C57BL/6J mice high fat diets containing 25 w% fat, there<strong>of</strong> 62 ± 3 g/kg n-3<br />

<strong>fatty</strong> <strong>acids</strong>, in combination with four diets <strong>of</strong> different carbohydrate:protein ratios (Table 1). All<br />

diets contained 25.12 kJ/g. Body weight gain and white adipose tissue (WAT) mass increased in<br />

parallel with the increase in the sucrose:protein ratio in the feed (Fig 1A and D). <strong>The</strong> weight <strong>of</strong><br />

interscapular brown adipose tissue (iBAT), liver and tibialis anterior muscle were not changed (not<br />

shown). Total energy intake was not statistically significantly different in the four groups, and thus,<br />

energy efficiency increased dose-<strong>dependent</strong>ly in response to the increased amount <strong>of</strong> dietary<br />

sucrose (Fig 1B). Moreover, expressions <strong>of</strong> inflammatory markers, such as Serpine1 (serine (or<br />

cysteine) peptidase inhibitor, clade E, member 1 or plasminogen activator inhibitor-1, PAI-1), Ccl2<br />

(Ccl2 chemokine (C-C motif) ligand 2, MCP1), Cd68 (CD68 antigen) and Emr1 (EGF-like module<br />

containing, mucin-like, hormone receptor-like sequence 1, F4/80) were increased in both eWAT<br />

and iWAT in the obese mice, suggesting that sucrose attenuated the anti-inflammatory effect <strong>of</strong> fish<br />

oil (Fig 1E). <strong>The</strong> expressions <strong>of</strong> Pparg (Peroxisome proliferator-activated receptor gamma), Srebf1<br />

(sterol regulatory element binding transcription factor 1) and Fasn (Fatty acid synthase) did not<br />

change significantly (Fig 1E). Mice receiving the highest amount <strong>of</strong> sucrose also lost significantly<br />

less body weight during a 24h fast (Fig 1C) than mice receiving the lowest amount <strong>of</strong> sucrose,<br />

indicative <strong>of</strong> a lower metabolic rate in high-sucrose fed mice.<br />

Energy may be lost as heat by the uncoupling activity <strong>of</strong> UCP1 (uncoupling protein 1<br />

(mitochondrial, proton carrier)) expressed in brown adipocytes, and increased expression <strong>of</strong> Ucp1<br />

can prevent diet-induced obesity. <strong>The</strong>refore, we measured Ucp1 expression in both white and<br />

brown adipose tissue depots. In iBAT Ucp1 expression was similar in all groups, but the expression<br />

<strong>of</strong> Ucp1 was significantly reduced in iWAT in mice fed the high amount <strong>of</strong> sucrose (Fig 1C and E).<br />

7


137<br />

138<br />

139<br />

140<br />

141<br />

142<br />

143<br />

144<br />

145<br />

146<br />

This indicates that adipocytes in iWAT from mice receiving a low amount <strong>of</strong> sucrose and a high<br />

amount <strong>of</strong> protein had a more brownish phenotype.<br />

We have previously demonstrated that a high fat diet in combination with a high<br />

protein:sucrose ratio translated into a high glucagon:insulin ratio, increased cAMP-signaling and<br />

expression <strong>of</strong> Ppargc1a (peroxisome proliferative activated receptor, gamma, coactivator 1 alpha)<br />

leading to increased gluconeogenesis and amino acid degradation in liver (16, 17). Here we show<br />

that sucrose dose-<strong>dependent</strong>ly reduced expression <strong>of</strong> Ppargc1a, Pck1 (phosphoenolpyruvate<br />

carboxykinase 1, cytosolic) and Agxt (alanine-glyoxylate aminotransferase) in the liver (Fig 1F).<br />

Expression <strong>of</strong> the lipogenic gene, Fasn, was increased whereas expressions <strong>of</strong> enzymes involved in<br />

<strong>fatty</strong> acid oxidation were unchanged when the sucrose:protein ratio was increased (Fig 1F).<br />

147<br />

148<br />

149<br />

150<br />

151<br />

152<br />

153<br />

154<br />

155<br />

156<br />

157<br />

158<br />

159<br />

<strong>The</strong> glucose moiety <strong>of</strong> sucrose is responsible for the obesity promoting effect <strong>of</strong> sucrose.<br />

To investigate whether the obesity promoting effect <strong>of</strong> sucrose fed in combination with fish oil was<br />

depended on the glucose or fructose moiety <strong>of</strong> sucrose, we prep<strong>are</strong>d diets where fish oil was<br />

combined with sucrose, glucose or fructose (Table 2). C57BL/6J mice were fed these diets adlibitum.<br />

After 7 weeks mice receiving fish oil in combination with fructose had gained less weight<br />

than mice receiving fish oil in combination with sucrose or glucose (Fig 2A). Energy intake was<br />

slightly, but not statistically significantly lower in mice fed the fructose supplemented diets and<br />

hence, energy efficiency was significantly reduced (Fig 2B). Calculation <strong>of</strong> digestibility<br />

demonstrated a minor, but not significantly reduced digestibility <strong>of</strong> protein and fat in fructose fed<br />

mice (Fig 2C). Of note, the mice receiving fructose had about 50% less white adipose tissue mass,<br />

iWAT, pWAT and eWAT, than mice receiving sucrose or glucose, combined with a tendency<br />

towards a slight decrease in the weight <strong>of</strong> the tibialis anterior muscle (Fig 2D).<br />

8


160<br />

161<br />

162<br />

163<br />

164<br />

165<br />

166<br />

167<br />

168<br />

169<br />

170<br />

171<br />

172<br />

173<br />

174<br />

175<br />

176<br />

177<br />

178<br />

179<br />

180<br />

181<br />

182<br />

183<br />

We have previously provided evidence that the different <strong>obesogenic</strong> effect <strong>of</strong> corn oil<br />

fed in combination with either protein or sucrose is related to the effect <strong>of</strong> the macronutrient<br />

composition on hormonal status (16). As sucrose and glucose, unlike fructose, stimulate insulin<br />

secretion, we measured plasma levels <strong>of</strong> glucose and insulin in the fed state. As expected, plasma<br />

glucose and insulin were lower in fructose fed mice (Fig 2E). As in mice fed a high fat diet in<br />

combination with proteins, expressions <strong>of</strong> Ppargc1a and Pck1 in the liver were increased (Fig 2F).<br />

However, unlike mice fed a high protein diet, expression <strong>of</strong> genes involved in amino acid<br />

degradation was not increased in fructose fed mice (Fig 2F). Plasma levels <strong>of</strong> 2-hydroxybutyrate, a<br />

marker for <strong>fatty</strong> acid β-oxidation, were similar in all groups, but the expression <strong>of</strong> genes involved in<br />

hepatic <strong>fatty</strong> acid oxidation was reduced in livers from fructose fed mice (Fig 2F). Hepatic<br />

expression <strong>of</strong> lipogenic genes was higher in fructose fed mice (Fig 2F). Importantly, however,<br />

excess lipid accumulation was not seen in liver or tibialis anterior muscle (Fig 3). <strong>The</strong> finding that<br />

Ucp1 expression was strongly induced in iWAT, but not iBAT in fructose fed mice, indicates that<br />

energy, as observed in protein fed mice, may be dissipated in the form <strong>of</strong> heat. Indeed, a higher<br />

expression <strong>of</strong> markers for brown adipocytes such as Ppargc1a and Cox2 (cytochrome c oxidase<br />

subunit II) suggested a higher number <strong>of</strong> brown-like adipocytes in iWAT (Fig 3C).<br />

Fructose feeding is frequently used to induce glucose intolerance in rats (26, 39) and<br />

pro-inflammatory cytokines such as CCL2 produced by both adipocytes and infiltrating<br />

macrophages <strong>are</strong> causally linked to the development <strong>of</strong> glucose intolerance (40, 41). As expression<br />

<strong>of</strong> inflammatory markers was significantly higher in mice fed fish oil in combination with glucose<br />

or sucrose than fructose (Fig 3C), a glucose tolerance test was performed in mice fed the sucrose,<br />

glucose and fructose-based diets. Of note, blood glucose levels reached higher concentrations in<br />

glucose comp<strong>are</strong>d to fructose fed mice and the <strong>are</strong>a under the curve (AUC) was significantly higher<br />

in glucose than both sucrose and fructose fed mice (Fig 3D).<br />

9


184<br />

185<br />

186<br />

187<br />

188<br />

189<br />

190<br />

191<br />

192<br />

193<br />

194<br />

195<br />

196<br />

197<br />

198<br />

199<br />

200<br />

201<br />

202<br />

203<br />

204<br />

205<br />

206<br />

High glycemic index starch increases the adipogenic potential <strong>of</strong> fish oil.<br />

To further investigate whether other types <strong>of</strong> carbohydrates, such as starch with different capacity to<br />

stimulate insulin secretion <strong>are</strong> able to modulate the adipogenic potential <strong>of</strong> fish oil, we prep<strong>are</strong>d<br />

isocaloric diets (Table 2) where fish oil was combined with low or high GI starch (100%<br />

amylopectin vs 60% amylose/40% amylopectin) demonstrated to induce the expected differences in<br />

postprandial blood glucose levels in a meal tolerance test when combined with a high fat diet (33,<br />

42). After 7 weeks <strong>of</strong> feeding, body weights were similar in mice receiving fish oil in combination<br />

with either high- or low GI starch (Fig 4A). Energy intake was also similar, and no statistical<br />

significant difference in energy efficiency was found (Fig 4A). However, mice fed fish oil in<br />

combination with the low GI starch had significantly lower levels <strong>of</strong> insulin and less white adipose<br />

tissue (Fig 4 B and C). Furthermore, expressions <strong>of</strong> inflammatory markers tended to be higher in<br />

adipose tissues from mice fed high GI starch (Fig 4E). <strong>The</strong> reduced adiposity observed in mice<br />

receiving fish oil in combination with the low GI starch was not due to decreased digestibility, as<br />

digestibility <strong>of</strong> the diet containing the low GI starch was slightly higher than digestibility <strong>of</strong> the diet<br />

containing the high GI starch (Fig 4D). Expressions <strong>of</strong> genes involved in <strong>fatty</strong> acid oxidation,<br />

gluconeogenesis and amino acid degradation in the liver were similar in the two groups <strong>of</strong> mice, but<br />

expressions <strong>of</strong> the lipogenic genes Scd1 (stearoyl-Coenzyme A desaturase 1) and Fasn were<br />

reduced in mice fed low GI starch (Fig 4F). Of note, expression <strong>of</strong> Ucp1 was similar in iBAT, but<br />

significantly higher in iWAT in mice fed the low GI starch (Fig 4E and F). Altogether, results from<br />

the experiments combining fish oil with glucose or fructose as well as high or low GI starch<br />

suggested that stimulation <strong>of</strong> insulin secretion increases the <strong>obesogenic</strong> effect <strong>of</strong> a diet enriched<br />

with dietary fish oil.<br />

207<br />

10


208<br />

209<br />

210<br />

211<br />

212<br />

213<br />

214<br />

215<br />

216<br />

217<br />

218<br />

219<br />

220<br />

221<br />

222<br />

223<br />

224<br />

225<br />

226<br />

<strong>The</strong> obesity promoting effect <strong>of</strong> sucrose in combination with fish oil is associated to increased<br />

insulin secretion.<br />

To investigate if increased insulin secretion is able to increase the <strong>obesogenic</strong> effect <strong>of</strong> fish oil when<br />

the carbohydrate load is low, we fed mice a diet enriched with fish oil in combination with proteins.<br />

To increase insulin secretion, half <strong>of</strong> the mice were injected daily with the sulfonylurea class drug<br />

glybenclamide (43, 44), whereas the second half were injected with placebo. Energy intake, body<br />

weight gain (Fig 5A) and energy efficiency (not shown) were not affected by glybenclamide, but<br />

the amount <strong>of</strong> white adipose tissue mass tended to increase although not statistically significantly<br />

(Fig 5B). However, expression <strong>of</strong> Pparg was significantly increased in both eWAT and iWAT (Fig<br />

5C). As insulin levels in the fed state only tended to increase, a glucose tolerance test was<br />

performed to validate whether the dose <strong>of</strong> glybenclamide used was sufficient to increase insulin<br />

levels. As expected, glucose tolerance was improved by daily injections <strong>of</strong> glybenclamide (Fig 5D).<br />

This was not accompanied by reduced expressions <strong>of</strong> markers for adipose tissue inflammation (Fig<br />

5C). In accordance with unchanged energy efficiency, Ucp1 expression was not significantly<br />

changed. However, glybenclamide treatment reduced expression <strong>of</strong> Pck1 and genes involved in<br />

amino acid degradation and ureagenesis (Fig 5E). Together, these results demonstrate that increased<br />

insulin secretion is unable to significantly increase the <strong>obesogenic</strong> effect <strong>of</strong> fish oil when the<br />

carbohydrate load is low. It is thus likely that a certain threshold level <strong>of</strong> carbohydrates is required<br />

in order for insulin to ellicit the <strong>obesogenic</strong> potential <strong>of</strong> fish oil.<br />

227<br />

228<br />

229<br />

230<br />

231<br />

If insulin promotes the <strong>obesogenic</strong> potential <strong>of</strong> fish oil in presence <strong>of</strong> carbohydrates,<br />

inhibition <strong>of</strong> insulin secretion should attenuate the adipogenic effect <strong>of</strong> sucrose in combination with<br />

fish oil. Since insulin secretion can be reduced by treatment with nifedipine (34) we supplemented a<br />

diet enriched with fish oil and sucrose with nifedipine. Inclusion <strong>of</strong> nifedipine did not influence<br />

body weight gain (Fig 6A), or feed intake (not shown). Consequently, energy efficiency was not<br />

11


232<br />

233<br />

234<br />

235<br />

236<br />

237<br />

238<br />

239<br />

240<br />

241<br />

242<br />

243<br />

244<br />

245<br />

246<br />

different (not shown). As expected, inclusion <strong>of</strong> nifedipine in the sucrose enriched high-fish oil diet<br />

resulted in lower levels <strong>of</strong> plasma insulin in the fed state (Fig 6B). Importantly, the mice receiving<br />

the nifedipine-supplemented diet had lower adipose tissue mass and adipocytes had a normal<br />

appearance (Fig 6C). When nifedipine was added to a standard low fat diet, no effect on adipose<br />

tissue mass was observed (not shown), and expression <strong>of</strong> inflammatory markers in white adipose<br />

tissue was, with the exception <strong>of</strong> Serpine1 in iWAT, not reduced (Fig 6D). In line with unchanged<br />

energy efficiency, we did not detect any increased expression <strong>of</strong> Ucp1 in iWAT in nifedipine<br />

treated mice (Fig 6D). Moreover, nifedipine did not increase hepatic expression <strong>of</strong> Pck1 or genes<br />

involved in lipogenesis or amino acid degradation (Fig 7A). Thus, inhibition <strong>of</strong> insulin secretion<br />

was able to partly attenuate the <strong>obesogenic</strong> effect <strong>of</strong> sucrose in combination with fish oil, but did<br />

not reduce energy efficiency. Furthemore, nifedipine did not improve the reduced glucose tolerance<br />

observed after feeding fish oil in combination with sucrose, but a insulin tolerance test indicated<br />

that insulin sensitivity was improved (Fig 7B). Together, our data indicate that an inhibition <strong>of</strong><br />

insulin secretion attenuates the adipogenic effect <strong>of</strong> sucrose in combination with fish oil. However,<br />

inhibition <strong>of</strong> insulin secretion is insufficient to reduce energy efficiency.<br />

247<br />

248<br />

249<br />

250<br />

251<br />

252<br />

DISCUSSION<br />

Due to the well described beneficial <strong>effects</strong> <strong>of</strong> n-3 PUFAs on lipid metabolism and lipid-related<br />

disorders in humans, increasing the relative amount <strong>of</strong> n-3 PUFAs in diet or as supplement is<br />

presently recommended by Health Authorities. <strong>The</strong> potential use <strong>of</strong> n-3 PUFAs in weight control in<br />

humans is not fully explored, but the ability <strong>of</strong> these <strong>fatty</strong> <strong>acids</strong> to reduce the development <strong>of</strong> diet-<br />

253<br />

induced obesity rodents is well described (7, 10-15).<br />

In this study we demonstrate that the<br />

254<br />

255<br />

composition <strong>of</strong> the diet determines the adipogenic potential <strong>of</strong> fish oil. In particular, we demonstrate<br />

that a high dose <strong>of</strong> fish oil promoted obesity when combined with sucrose, glucose or high GI<br />

12


256<br />

257<br />

258<br />

259<br />

260<br />

261<br />

262<br />

263<br />

264<br />

265<br />

266<br />

267<br />

268<br />

269<br />

270<br />

271<br />

272<br />

273<br />

274<br />

275<br />

276<br />

277<br />

278<br />

starch. Thus, many if not all reported beneficial <strong>effects</strong> <strong>of</strong> fish oil intake might be diminishes or<br />

completely abrogated by a simultaneous intake <strong>of</strong> high GI carbohydrates.<br />

Insulin is the primary anabolic hormone promoting energy storage in the fed state and<br />

is an important driver for adipocyte differentiation (18). Of note, mice lacking insulin receptors in<br />

adipose tissue (FIRKO mice), <strong>are</strong> protected against obesity and remain glucose tolerant on a highfat<br />

diet (19). Further, white adipose tissue mass is dramatically reduced in Irs1 -/- Irs2 -/- doubleknockout<br />

mice (45). In contrast, mice lacking the insulin receptor in muscle (MIRKO mice) have<br />

increased glucose utilization in WAT (46), and exhibit a more than 50% increase in fat mass and<br />

adipocyte number (47). Similarly, mice overexpressing Slc2a4 (solute carrier family 2 (facilitated<br />

glucose transporter), member 4, GLUT4) (48) or Irs1 (20) in adipose tissue <strong>are</strong> obese. Finally,<br />

treating normal rats with insulin via osmotic minipumps increases Slc2a4 expression, glucose<br />

utilization and de novo <strong>fatty</strong> acid synthesis in adipose tissue, which <strong>are</strong> accompanied by weight gain<br />

and increased adipose tissue mass (49-51). <strong>The</strong> importance <strong>of</strong> insulin secretion and insulin levels in<br />

circulation is further supported by the finding that daily injections <strong>of</strong> insulin increases weight gain<br />

in transgenic mice expressing agouti in adipose tissue (52). Of note, unlike the traditional agouti<br />

mice, mice with transgenic expression <strong>of</strong> the agouti gene driven by the aP2 promoter do not get<br />

obese unless triggered by insulin (52). Transgenic mice treated with insulin gained significantly<br />

more weigh than their wild type littermates, and the authors suggested that the daily insulin<br />

treatment mimicked the hyperinsulinemia normally observed in mice expressing the agouti gene<br />

ubiquitously (52). Moreover, weight gain is a well recognized side effect <strong>of</strong> type 2 diabetic drugs<br />

that increase insulin sensitivity (53, 54). Together these observations suggest that hyperinsulinemia<br />

is a contributing factor to development <strong>of</strong> obesity and reducing hyperinsulinemia would thus<br />

possible counteract obesity development. In keeping with this view, inhibition <strong>of</strong> insulin secretion<br />

13


279<br />

280<br />

281<br />

282<br />

283<br />

284<br />

285<br />

286<br />

287<br />

288<br />

289<br />

290<br />

by administration <strong>of</strong> nifedipine attenuated the adipogenic effect <strong>of</strong> fish oil in combination with<br />

sucrose.<br />

Increasing insulin secretion alone, however, is not sufficient to promote obesity<br />

development, as mice receiving glybenclamide in combination with proteins and fish oil did not<br />

become obese. Furthermore, increased insulin secretion by glybenclamide in the presence <strong>of</strong> low<br />

levels <strong>of</strong> carbohydrates in the feed was insufficient to increase the adipogenic potential <strong>of</strong> fish oil. It<br />

has been demonstrated that a high-fat diet is unable to increase adipose tissue mass in the absence<br />

<strong>of</strong> carbohydrates (55, 56).<br />

In our study nifedipine did not reduce adipose tissue mass in low fat fed mice (not<br />

shown). However, as emphasized above, inclusion <strong>of</strong> nifedipine attenuated hyperinsulinemia and<br />

obesity induced by fish oil in combination with sucrose. <strong>The</strong>se findings <strong>are</strong> in line with the study by<br />

Kim et al (34) demonstrating that nifedipine attenuated agouti-induced hyperinsulinemia and<br />

291<br />

obesity.<br />

Hyperinsulinemia is an early event also in obese Zucker rats, and attenuation <strong>of</strong><br />

292<br />

293<br />

294<br />

295<br />

296<br />

297<br />

298<br />

299<br />

300<br />

301<br />

hyperinsulinemia using diazoxide reduces obesity development in obese- but not lean Zucker rats<br />

in<strong>dependent</strong>ly <strong>of</strong> feed intake (57). Together our results suggest that the increased insulin secretion<br />

contribute to the obesity promoting effect <strong>of</strong> fish oil when combined with sucrose.<br />

<strong>The</strong> obesity promoting effect <strong>of</strong> increased insulin secretion is further supported by our<br />

finding that glucose, but not fructose is the obesity promoting moiety <strong>of</strong> sucrose. Unlike sucrose<br />

and glucose, fructose does not stimulate insulin secretion from pancreatic beta-cells (26) and mice<br />

receiving fish oil in combination with fructose had less adipose tissue mass than mice receiving fish<br />

oil in combination with glucose or sucrose. Mice receiving fructose in combination with fish oil<br />

were also less glucose intolerant than mice fed fish oil in combination with sucrose or glucose.<br />

Fructose is commonly used to induce glucose intolerance in rats and some mice strains also develop<br />

14


302<br />

303<br />

304<br />

305<br />

306<br />

307<br />

308<br />

309<br />

310<br />

311<br />

312<br />

313<br />

314<br />

315<br />

316<br />

317<br />

318<br />

319<br />

320<br />

321<br />

322<br />

323<br />

324<br />

325<br />

metabolic syndrome in response to high fructose feeding (58). However, in C57BL/6 mice neither<br />

hyperinsulinemia nor hyperglycemia developed in response to high fructose feeding (58).<br />

<strong>The</strong> obesity promoting effect <strong>of</strong> fish oil can also be affected by different type <strong>of</strong> starch<br />

as mice receiving fish oil in combination with a high GI starch had higher adipose tissue mass than<br />

mice receiving fish oil in combination with low GI starch. <strong>The</strong>se results may also relate to the<br />

effect <strong>of</strong> the different type <strong>of</strong> starch on postprandial glucose levels and insulin secretion (33, 42,<br />

59). Low GI diets <strong>are</strong> becoming popular in weight management also in humans although their<br />

effectiveness in terms <strong>of</strong> lasting weight reduction is not commonly accepted (28-32). <strong>The</strong> lack <strong>of</strong><br />

acceptance may, however, partly be due to insufficient power <strong>of</strong> early studies as a meta-analysis has<br />

indicated that diets in which there was a reduction in the glycemic index produced moderately more<br />

weight loss than control low fat diets in humans (32). However, a recent large European study<br />

demonstrated that intake <strong>of</strong> low glycemic index carbohydrates combined with a modest increase in<br />

protein content improved maintenance <strong>of</strong> weight loss (60).<br />

Obviously, increased adipose tissue mass is related to energy-intake. However, as<br />

demonstrated here, the macronutrient composition can influence energy efficiency, and mice<br />

consuming the same amount <strong>of</strong> calories end up with quite different amounts <strong>of</strong> adipose tissue. Of<br />

note, increasing the amount <strong>of</strong> sucrose from 13 to 43% led to an approximately 5-fold higher energy<br />

efficiency. Circulating insulin levels may also indirectly influence energy efficiency. Hepatic PGC-<br />

1α is a central target <strong>of</strong> the insulin/glucagon axis regulating activation <strong>of</strong> the entire gluconeogenesis<br />

cascade in liver (61-63) and a dose-<strong>dependent</strong> decrease in Ppargc1a and Pck1 expression was<br />

observed in response to increasing intake <strong>of</strong> dietary sucrose. This might be <strong>of</strong> importance regarding<br />

energy efficiency as gluconeogenesis requires ATP and activation <strong>of</strong> gluconeogenesis reduces feed<br />

efficiency as protein is converted to glucose at a cost <strong>of</strong> 16-20 kJ/g protein (64). Moreover,<br />

increased catabolism <strong>of</strong> amino <strong>acids</strong> requires ATP for disposal <strong>of</strong> nitrogen as urea at an energy-cost<br />

15


326<br />

327<br />

328<br />

329<br />

330<br />

331<br />

332<br />

333<br />

334<br />

335<br />

336<br />

337<br />

338<br />

339<br />

340<br />

341<br />

342<br />

343<br />

344<br />

345<br />

346<br />

<strong>of</strong> ca 5.4 kJ/g urea. Consistent with this notion, we have observed that fish oil combined with a high<br />

level <strong>of</strong> protein and low levels <strong>of</strong> sucrose induces hepatic expression <strong>of</strong> Ppargc1a that was<br />

accompanied by an induction <strong>of</strong> mRNAs encoding enzymes involved in catabolism <strong>of</strong> glucogenic<br />

amino. Thus, increased energy cost by increased gluconeogenesis and ureagenesis probably<br />

contribute to the reduced feed-efficiency observed by a high protein diet.<br />

In mice fed increasing amounts <strong>of</strong> protein at the expense <strong>of</strong> sucrose, we observed a<br />

dose-<strong>dependent</strong> increase in Ucp1 expression in iWAT. Others and we have recently demonstrated<br />

that cyclooxygenase-<strong>dependent</strong> induction <strong>of</strong> UCP1 expression in WAT counteracts diet-induced<br />

obesity (65, 66). <strong>The</strong> importance <strong>of</strong> UCP1 expressing brown-like adipocyte in WAT for regulation<br />

<strong>of</strong> energy expenditure is further underscored by aP2-UCP1 transgenic mice in which endogenous<br />

Ucp1 expression and respiration were reduced in iBAT, whereas UCP1 expression, respiration and<br />

total oxidative capacity were induced in WAT, and this was sufficient to account for the observed<br />

changes <strong>of</strong> total energy balance (67). Thus, UCP1-<strong>dependent</strong> uncoupled respiration in iWAT in<br />

combination with increased energy cost from gluconeogenesis may account for the reduced energyefficiency<br />

observed when sucrose levels in the diet were low.<br />

If the background diet determines the adipogenic potential <strong>of</strong> fish oil also in humans,<br />

this is <strong>of</strong> great concern, as the intake <strong>of</strong> refined sugars from sources such as s<strong>of</strong>t-drinks has<br />

increased dramatically during the last several decades (68). Moreover, n-3 supplements <strong>are</strong> <strong>of</strong>ten<br />

taken in combination with morning meals containing high-glycemic index carbohydrates such as<br />

cereal, bread and orange juice. Thus, comprehensive studies <strong>of</strong> the interaction between dietary<br />

macronutrients and fish oil in humans seem warranted.<br />

347<br />

348<br />

349<br />

16


350<br />

351<br />

FIGURE LEGENDS<br />

352<br />

353<br />

354<br />

355<br />

356<br />

357<br />

358<br />

359<br />

360<br />

361<br />

362<br />

363<br />

364<br />

365<br />

366<br />

367<br />

368<br />

369<br />

370<br />

FIGURE 1. Macronutrient composition determines the <strong>obesogenic</strong> effect <strong>of</strong> fish oil. Male<br />

C57BL/6 mice (n=8) were pair-fed isocaloric high fat diets with different carbohydrate/protein<br />

ratios for 6 weeks. (A) Body weight development in pair-fed mice is shown as relative increase. (B)<br />

Energy efficiency was calculated as body weight gain divided by feed energy intake. (C) Weight<br />

reduction during 24h fasting was measured and Ucp1 (Uncoupling protein-1) and Dio2 (deiodinase,<br />

iodothyronine, type II) expressions were measured by RT-qPCR. (D-F) Adipose tissue and liver<br />

were dissected out and the weights were recorded. Expressions <strong>of</strong> Pparg (peroxisome proliferator<br />

activated receptor gamma), Srebf1 (sterol regulatory element binding transcription factor 1), Fasn<br />

(<strong>fatty</strong> acid synthase), Ucp1 (Uncoupling protein-1), Serpine1 (Plasminogen activator inhibitor-1),<br />

Ccl2 (chemokine (C-C motif) ligand 2), Cd68 (CD68 antigen) and Emr1 (EGF-like module<br />

containing, mucin-like, hormone receptor-like sequence 1 or F4/80) were measured in epididymal<br />

and inguinal white adipose tissue and expressions <strong>of</strong> Acox1 (acyl-CoA oxidase 1), Cpt1a (carnitine<br />

palmitoyltransferase 1a), Fasn (<strong>fatty</strong> acid synthase), Ppargc1a (peroxisome proliferator-activated<br />

receptor gamma, coactivator 1 alpha), Pck1 (phosphoenolpyruvate carboxykinase 1, cytosolic) and<br />

Agxt (alanine-glyoxylate aminotransferase) were measured in liver using RT-qPCR and normalized<br />

to Tbp (TATA-box binding protein). Data <strong>are</strong> presented as mean ± SEM. Different small letters<br />

denote significant differences (P


374<br />

375<br />

376<br />

377<br />

378<br />

379<br />

380<br />

381<br />

382<br />

383<br />

384<br />

385<br />

386<br />

387<br />

388<br />

389<br />

with different carbohydrate sources ad-libitum for 8 weeks. (A) Body weight development is shown<br />

as relative increase. (B) Energy efficiency was calculated as body weight gain divided by feed<br />

intake. (C) Digestibility <strong>of</strong> energy, nitrogen, organic material and fat were calculated in a separate<br />

set <strong>of</strong> mice (n=5). (D-F) Blood was collected, organs were dissected out and the weights were<br />

recorded. Hormones and lipid parameters were measured in plasma and expressions <strong>of</strong> Acox1 (acyl-<br />

CoA oxidase 1), Cpt1a (carnitine palmitoyltransferase 1a), Cpt2 (carnitine palmitoyltransferase 2),<br />

Hmgcs2 (3-hydroxy-3-methylglutaryl-Coenzyme A synthase 2), Srebf1 (sterol regulatory element<br />

binding transcription factor 1), Scd1 (stearoyl-Coenzyme A desaturase 1), Acaca (acetyl-Coenzyme<br />

A carboxylase alpha), Fasn (<strong>fatty</strong> acid synthase), Crem (cAMP responsive element modulator),<br />

Pde4c (phosphodiesterase 4C, cAMP specific), Ppargc1a (peroxisome proliferator-activated<br />

receptor gamma, coactivator 1 alpha), Pck1 (phosphoenolpyruvate carboxykinase 1, cytosolic),<br />

Gpt1a (glutamic pyruvic transaminase 1a), Got1 (glutamate oxaloacetate transaminase 1, soluble),<br />

Agxt (alanine-glyoxylate aminotransferase) and Cps1 (carbamoyl-phosphate synthetase 1) were<br />

measured in liver using RT-qPCR and normalized to Tbp (TATA-box binding protein). Data <strong>are</strong><br />

presented as mean ± SEM. Different small letters denote significant differences (P


398<br />

399<br />

400<br />

401<br />

402<br />

403<br />

404<br />

subunit II), Serpine1 (Plasminogen activator inhibitor-1), Ccl2 (chemokine (C-C motif) ligand 2),<br />

Cd68 (CD68 antigen) and Emr1 (EGF-like module containing, mucin-like, hormone receptor-like<br />

sequence 1 or F4/80) were measured in epididymal and inguinal white adipose tissue by RT-qPCR<br />

and normalized to Tbp. (D) In a separate set <strong>of</strong> mice (n=9), an intraperitoneal glucose tolerance and<br />

insulin tolerance test were performed. Data <strong>are</strong> presented as mean ± SEM. Different small letters<br />

denote significant differences (P


422<br />

423<br />

424<br />

425<br />

426<br />

carboxykinase 1, cytosolic), Got1 (glutamate oxaloacetate transaminase 1, soluble), Agxt (alanineglyoxylate<br />

aminotransferase) and Cps1 (carbamoyl-phosphate synthetase 1) were measured in liver<br />

using RT-qPCR and normalized to Tbp (TATA-box binding protein). Data <strong>are</strong> presented as mean ±<br />

SEM. Different small letters denote significant differences (P


446<br />

447<br />

448<br />

449<br />

450<br />

451<br />

gamma, coactivator 1 alpha), Pck1 (phosphoenolpyruvate carboxykinase 1, cytosolic), Gpt1a<br />

(glutamic pyruvic transaminase 1a), Got1 (glutamate oxaloacetate transaminase 1, soluble), Agxt<br />

(alanine-glyoxylate aminotransferase) and Cps1 (carbamoyl-phosphate synthetase 1) were measured<br />

in liver using RT-qPCR and normalized to Tbp (TATA-box binding protein). Data <strong>are</strong> presented as<br />

mean ± SEM. Different small letters denote significant differences (P


470<br />

471<br />

472<br />

473<br />

474<br />

475<br />

476<br />

477<br />

478<br />

479<br />

480<br />

481<br />

482<br />

nifedipine supplementation (1 g/kg) diet for 4 weeks. (A) Expressions <strong>of</strong> Acox1 (acyl-CoA oxidase<br />

1), Cpt1a (carnitine palmitoyltransferase 1a), Cpt2 (carnitine palmitoyltransferase 2), Hmgcs2 (3-<br />

hydroxy-3-methylglutaryl-Coenzyme A synthase 2), Srebf1 (sterol regulatory element binding<br />

transcription factor 1), Scd1 (stearoyl-Coenzyme A desaturase 1), Acaca (acetyl-Coenzyme A<br />

carboxylase alpha), Fasn (<strong>fatty</strong> acid synthase), Crem (cAMP responsive element modulator), Pde4c<br />

(phosphodiesterase 4C, cAMP specific), Ppargc1a (peroxisome proliferator-activated receptor<br />

gamma, coactivator 1 alpha), Pck1 (phosphoenolpyruvate carboxykinase 1, cytosolic), Gpt1a<br />

(glutamic pyruvic transaminase 1a), Got1 (glutamate oxaloacetate transaminase 1, soluble), Agxt<br />

(alanine-glyoxylate aminotransferase) and Cps1 (carbamoyl-phosphate synthetase 1) were measured<br />

in liver using RT-qPCR and normalized to Tbp (TATA-box binding protein). (B) Intraperitoneal<br />

glucose tolerance test and insulin glucose tests were performed. Data <strong>are</strong> presented as mean ± SEM.<br />

Different small letters denote significant differences (P


484<br />

485<br />

486<br />

TABLE 1.<br />

Macronutrient composition in the diets<br />

Data represent weight % (g /100g)<br />

High fish oil<br />

Sucrose<br />

13% 23% 33% 43%<br />

Protein 500 400 300 200<br />

Casein 500 400 300 200<br />

L-Cysteine 3 3 3 3<br />

Carbohydrate 189,5 289,5 389,5 489,5<br />

Corn starch 9,5 9,5 9,5 9,5<br />

Cellulose 50 50 50 50<br />

Sucrose 130 230 330 430<br />

Fat 250 250 250 250<br />

Soybean oil 70 70 70 70<br />

Fish oil 180 180 180 180<br />

Energy (kJ/g) 25.12 25.12 25.12 25.13<br />

487<br />

488<br />

23


489<br />

490<br />

491<br />

492<br />

TABLE 2.<br />

Macronutrient composition in the diets<br />

Data represent weight % (g /100g)<br />

493<br />

High fish oil<br />

Low<br />

energy Sucrose Fructose Glucose Low GI High GI<br />

Protein 200 200 200 200 200 200<br />

Casein 200 200 200 200 200 200<br />

L-Cysteine 3 3 3 3 3 3<br />

Carbohydrate 669,5 489,5 489,5 489,5 489,5 489,5<br />

Corn starch 529,5 9,5 9,5 9,5 9,5 9,5<br />

Cellulose 50 50 50 50 50 50<br />

Amylose 204<br />

Amylopectin 136 340<br />

Sucrose 90 430 90 90 90 90<br />

Fructose 340<br />

Glucose 340<br />

Fat 70 250 250 250 250 250<br />

Soybean oil 70 70 70 70 70 70<br />

Fish oil 180 180 180 180 180<br />

24


Energy (kJ/g) 17.16 25.12 25.12 25.12 25.12 25.12<br />

494<br />

495<br />

496<br />

497<br />

498<br />

499<br />

500<br />

501<br />

Acknowledgments:<br />

We thank Alison Keenan for the helpful comments and suggestions during preparation <strong>of</strong> the<br />

manuscript. Moreover, we thank Åse Heltveit and Jan Idar Hjelle at NIFES for their excellent<br />

assistance with animal c<strong>are</strong> and lipid analyses.<br />

502<br />

503<br />

504<br />

505<br />

506<br />

507<br />

Authors´ contribution:<br />

B. L., K. K. and L. M. designed research. Q. H., H. H. L., E. F., L. S. M., L. K. M., R. J., T. M., B.<br />

J., R. K. P., S. B. S., A. C., L. F., B. L. and L. M. conducted research and analyzed data. K. K. and<br />

L. M. had primary responsibility for the final content. All authors read and approved the final<br />

manuscript.<br />

508<br />

509<br />

510<br />

511<br />

512<br />

513<br />

514<br />

515<br />

516<br />

517<br />

518<br />

519<br />

520<br />

521<br />

Literature Cited<br />

1. Marik, V. & Varon, J. (2009) Omega-3 dietary supplements and the risk <strong>of</strong> cardiovascular<br />

events: a systematic review. Clin Cardiol 32: 365-372.<br />

2. Harris, W. S. (1997) n-3 Fatty <strong>acids</strong> and serum lipoproteins: Human studies. Am J Clin Nutr<br />

65: S1645-S1654.<br />

3. Ruxton, C. H. S., Reed, S. C., Simpson, M. J. A. & Millington, K. J. (2004) <strong>The</strong> health<br />

benefits <strong>of</strong> omega-3 <strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong>: a review <strong>of</strong> the evidence. Journal <strong>of</strong> Human<br />

Nutrition and Dietetics 17: 449-459.<br />

4. Storlien, L. H., Kraegen, E. W., Chisholm, D. J., Ford, G. L., Bruce, D. G. & Pascoe, W. S.<br />

(1987) Fish Oil Prevents Insulin Resistance Induced by High-Fat Feeding in Rats. Science<br />

237: 885-888.<br />

25


522<br />

523<br />

524<br />

525<br />

526<br />

527<br />

528<br />

529<br />

530<br />

531<br />

532<br />

533<br />

534<br />

535<br />

536<br />

537<br />

538<br />

539<br />

540<br />

541<br />

542<br />

543<br />

544<br />

545<br />

546<br />

547<br />

548<br />

549<br />

550<br />

551<br />

552<br />

553<br />

554<br />

555<br />

556<br />

557<br />

558<br />

559<br />

560<br />

5. Jucker, B. M., Cline, G. W., Barucci, N. & Shulman, G. I. (1999) Differential <strong>effects</strong> <strong>of</strong><br />

safflower oil versus fish oil feeding on insulin-stimulated glycogen synthesis, glycolysis,<br />

and pyruvate dehydrogenase flux in skeletal muscle - A C-13 nuclear magnetic resonance<br />

study. Diabetes 48: 134-140.<br />

6. Neschen, S., Morino, K., Dong, J. Y., Wang-Fischer, Y., Cline, G. W., Romanelli, A. J.,<br />

Rossbacher, J. C., Moore, I. K., Regittnig, W. et al. (2007) N-3 <strong>fatty</strong> <strong>acids</strong> preserve insulin<br />

sensitivity in vivo in a peroxisonte proliferator-activated receptor-alpha-<strong>dependent</strong> manner.<br />

Diabetes 56: 1034-1041.<br />

7. Samane, S., Christon, R., Dombrowski, L., Turcotte, S., Charrouf, Z., Lavigne, C., Levy, E.,<br />

Bachelard, H., Amarouch, H. et al. (2009) Fish oil and argan oil intake differently modulate<br />

insulin resistance and glucose intolerance in a rat model <strong>of</strong> dietary-induced obesity.<br />

Metabolism-Clinical and Experimental 58: 909-919.<br />

8. Kalupahana, N. S., Claycombe, K., Newman, S. J., Stewart, T., Siriwardhana, N., Matthan,<br />

N., Lichtenstein, A. H. & Moustaid-moussa, N. (2010) Eicosapentaenoic Acid Prevents and<br />

Reverses Insulin Resistance in High-Fat Diet-Induced Obese Mice via Modulation <strong>of</strong><br />

Adipose Tissue Inflammation. <strong>The</strong> Journal <strong>of</strong> Nutrition 140: 1915-1922.<br />

9. Oh, D. Y., Talukdar, S., Bae, E. J., Imamura, T., Morinaga, H., Fan, W. Q., Li, P. P., Lu, W.<br />

J., Watkins, S. M. & Olefsky, J. M. (2010) GPR120 Is an Omega-3 Fatty Acid Receptor<br />

Mediating Potent Anti-inflammatory and Insulin-Sensitizing Effects. Cell 142: 687-698.<br />

10. Belzung, F., Raclot, T. & Groscolas, R. (1993) Fish oil n-3 <strong>fatty</strong> <strong>acids</strong> selectively limit the<br />

hypertrophy <strong>of</strong> abdominal fat depots in growing rats fed high-fat diets. Am J Physiol. 246:<br />

R1111-R1118.<br />

11. Wang, H., Storlien, LH. & Huang, XF. (2002) Effects <strong>of</strong> dietary fat types on body fatness,<br />

leptin, and ARC leptin receptor, NPY, and AgRP mRNA expression. Am J Physiol<br />

Endocrinol Metab. 282: E1352-E1359.<br />

12. Ruzickova, J., Rossmeisl, M., Prazak, T., Flachs, P., Sponarova, J., Vecka, M., Tvrzicka, E.,<br />

Bryhn, M. & Kopecky, J. (2004) Omega-3 PUFA <strong>of</strong> marine origin limit diet-induced obesity<br />

in mice by reducing cellularity <strong>of</strong> adipose tissue. Lipids 39, pg 1177, 2004): 1177-1185.<br />

13. Flachs, P., Horakova, O., Brauner, P., Rossmeisl, M., Pecina, P., Franssen-van Hal, N.,<br />

Ruzickova, J., Sponarova, J., Drahota, Z. et al. (2005) Polyunsaturated <strong>fatty</strong> <strong>acids</strong> <strong>of</strong> marine<br />

origin upregulate mitochondrial biogenesis and induce beta-oxidation in white fat.<br />

Diabetologia 48: 2365-2375.<br />

14. Arai, T., Kim, H. J., Chiba, H. & Matsumoto, A. (2009) Anti-obesity effect <strong>of</strong> fish oil and<br />

fish oil-fen<strong>of</strong>ibrate combination in female KK mice. J Atheroscler Thromb. 16: 675-683.<br />

15. Rokling-Andersen, M. H., Rustan, A. C., Wensaas, A. J., Kaalhus, O., Wergedahl, H., Røst,<br />

T. H., Jensen, J., Graff, B. A., Caesar, R. & Drevon, C. A. (2009) Marine omega-3 <strong>fatty</strong><br />

<strong>acids</strong> promote size reduction <strong>of</strong> visceral adipose depots, without altering body weight and<br />

composition, in male Wistar rats fed a high-fat diet. British Journal <strong>of</strong> Nutrition 102: 995-<br />

1006.<br />

26


561<br />

562<br />

563<br />

564<br />

565<br />

566<br />

567<br />

568<br />

569<br />

570<br />

571<br />

572<br />

573<br />

574<br />

575<br />

576<br />

577<br />

578<br />

579<br />

580<br />

581<br />

582<br />

583<br />

584<br />

585<br />

586<br />

587<br />

588<br />

589<br />

590<br />

591<br />

592<br />

593<br />

594<br />

595<br />

596<br />

16. Madsen, L., Pedersen, L. M., Liaset, B., Ma, T., Petersen, R. K., van den Berg, S., Pan, J.,<br />

Muller-Decker, K., Dulsner, E. D. et al. (2008) cAMP-<strong>dependent</strong> Signaling Regulates the<br />

Adipogenic Effect <strong>of</strong> n-6 Polyunsaturated Fatty Acids. J. Biol. Chem. 283: 7196-7205.<br />

17. Ma, T., Liaset, B., Hao, Q., Pedersen, R. K., Fjære, E., Ngo, H. T., Lillefosse, H. H.,<br />

Jørgensem, S. R., Wojtaszewski, J. et al. (2011) Sucrose counteracts the anti-inflammatory<br />

effect <strong>of</strong> fish oil in adipose tissue and increases obesity development in mice. PLoS ONE, In<br />

press.<br />

18. Madsen, L. & Kristiansen, K. (2010) <strong>The</strong> importance <strong>of</strong> dietary modulation <strong>of</strong> cAMP and<br />

insulin signaling in adipose tissue and the development <strong>of</strong> obesity. Foods for Health in the<br />

21St Century: A Road Map for the Future 1190: 1-14.<br />

19. Bluher, M., Michael, M. D., Peroni, O. D., Ueki, K., Carter, N., Kahn, B. B. & Kahn, C. R.<br />

(2002) Adipose Tissue Selective Insulin Receptor Knockout Protects against Obesity and<br />

Obesity-Related Glucose Intolerance. Developmental Cell 3: 25-38.<br />

20. Murata, Y., Tsuruzoe, K., Kawashima, J., Furukawa, N., Kondo, T., Motoshima, H., Igata,<br />

M., Taketa, K., Sasaki, K. et al. (2007) IRS-1 transgenic mice show increased epididymal fat<br />

mass and insulin resistance. Biochem Biophys Res Commun. 364: 301-307.<br />

21. Curry, D. L. (1989) Effects <strong>of</strong> mannose and fructose on the synthesis and secretion <strong>of</strong><br />

insulin. Pancreas 4: 2-9.<br />

22. Sato, Y., Ito, T., Udaka, N., Kanisawa, M., Noguchi, Y., Cushman, S. W. & Satoh, S. (1996)<br />

Immunohistochemical localization <strong>of</strong> facilitated-diffusion glucose transporters in rat<br />

pancreatic islets. Tissue Cell. 28: 637-643.<br />

23. Ganda, O. P., Soelder, J., Gleason, R. E., Cleator, I. G. & Reynolds, C. (1979) Metabolic<br />

Effects <strong>of</strong> Glucose, Mannose, Galactose, and Fructose in Man. J Clin Endocrinol Metab 49:<br />

616-622.<br />

24. Sykes, S., Morgan, L., english, J. & Marks, V. (1980) Evidence for preferential stimulation<br />

<strong>of</strong> gastric inhibitory polypeptide secretion in the rat by actively transported carbohydrates<br />

and their analogues. J Endocrinol 85: 201-207.<br />

25. Rizkalla, S. (2010) Health implications <strong>of</strong> fructose consumption: A review <strong>of</strong> recent data.<br />

Nutrition & Metabolism 7: 82.<br />

26. Tran, L. T., Yuen, V. G. & McNeill, J. H. (2009) <strong>The</strong> fructose-fed rat: a review on the<br />

mechanisms <strong>of</strong> fructose-induced insulin resistance and hypertension. Molecular and Cellular<br />

Biochemistry 332: 145-159.<br />

27. Jenkins, D. J., Wolever, T. M., Taylor, R. H., Barker, H., Fielden, H., Baldwin, J. M.,<br />

Bowling, A. C., Newman, H. C., Jenkins, A. L. & G<strong>of</strong>f, D. V. (1981) Glycemic index <strong>of</strong><br />

foods: a physiological basis for carbohydrate exchange. <strong>The</strong> American Journal <strong>of</strong> Clinical<br />

Nutrition 34: 362-366.<br />

27


597<br />

598<br />

599<br />

600<br />

601<br />

602<br />

603<br />

604<br />

605<br />

606<br />

607<br />

608<br />

609<br />

610<br />

611<br />

612<br />

613<br />

614<br />

615<br />

616<br />

617<br />

618<br />

619<br />

620<br />

621<br />

622<br />

623<br />

624<br />

625<br />

626<br />

627<br />

628<br />

629<br />

630<br />

631<br />

632<br />

633<br />

634<br />

28. Esfahani, A., Wong, J. M. W., Mirrahimi, A., Villa, C. R. & Kendall, C. W. C. (2011) <strong>The</strong><br />

application <strong>of</strong> the glycemic index and glycemic load in weight loss: A review <strong>of</strong> the clinical<br />

evidence. IUBMB Life 63: 7-13.<br />

29. Barclay, A. W., Petocz, P., Millan-Price, J., Flood, V. M., Prvan, T., Mitchell, P. & Brand-<br />

Miller, J. C. (2008) Glycemic index, glycemic load, and chronic disease risk-a meta-analysis<br />

<strong>of</strong> observational studies. <strong>The</strong> American Journal <strong>of</strong> Clinical Nutrition 87: 627-637.<br />

30. Lara-Castro, C. & Garvey, W. T. (2004) Diet, Insulin Resistance, and Obesity: Zoning in on<br />

Data for Atkins Dieters Living in South Beach. J Clin Endocrinol Metab 89: 4197-4205.<br />

31. Astrup, A., Larsen, T. M. & Harper, A. (2004) Atkins and other low-carbohydrate diets:<br />

hoax or an effective tool for weight loss <strong>The</strong> Lancet 364: 897-899.<br />

32. Thomas, D. E., Elliott, E. J. & Baur, L. (2007) Low glycaemic index or low glycaemic load<br />

diets for overweight and obesity. Cochrane Database <strong>of</strong> Systematic Reviews.<br />

33. Pawlak, D. B., Kushner, J. A. & Ludwig, D. S. (2004) Effects <strong>of</strong> dietary glycaemic index on<br />

adiposity, glucose homoeostasis, and plasma lipids in animals. <strong>The</strong> Lancet 364: 778-785.<br />

34. Kim, J. H., Mynatt, R. L., Moore, J. W., Woychik, R. P., Moustaid, N. & Zemel, M. B.<br />

(1996) <strong>The</strong> <strong>effects</strong> <strong>of</strong> calcium channel blockade on agouti-induced obesity. FASEB J. 10:<br />

1646-1652.<br />

35. Liaset, B., Madsen, L., Hao, Q., Criales, G., Mellgren, G., Marschall, H.-U., Hallenborg, P.,<br />

Espe, M., Frøyland, L. & Kristiansen, K. (2009) Fish protein hydrolysate elevates plasma<br />

bile <strong>acids</strong> and reduces visceral adipose tissue mass in rats. Biochimica et Biophysica Acta<br />

1791: 254-262.<br />

36. Arslan, G., Brunborg, L. A., Frøyland, L., Brun, J. G., Valen, M. & Berstad, A. (2002)<br />

Effects <strong>of</strong> duodenal seal oil administration in patients with inflammatory bowel disease.<br />

Lipids 37: 935-940.<br />

37. Barbatelli, G., Murano, I., Madsen, L., Hao, Q., Jimenez, M., Kristiansen, K., Giacobino, J.<br />

P., De Matteis, R. & Cinti, S. (2010) <strong>The</strong> emergence <strong>of</strong> cold-induced brown adipocytes in<br />

mouse white fat depots is determined predominantly by white to brown adipocyte<br />

transdifferentiation. American Journal <strong>of</strong> Physiology-Endocrinology and Metabolism 298:<br />

E1244-E1253.<br />

38. Madsen, L., Petersen, R. K., Sørensen, M. B., Jørgensen, C., Hallenborg, P., Pridal, L.,<br />

Fleckner, J., Amri, E.-Z., Krieg, P. et al. (2003) Adipocyte differentiation <strong>of</strong> 3T3-L1<br />

preadipocytes is <strong>dependent</strong> on lipoxygenase activity during the initial stages <strong>of</strong> the<br />

differentiation process. Biochem. J. 375: 539-549.<br />

39. Hwang, I. S., Ho, H., H<strong>of</strong>fman, B. B. & Reaven, G. M. (1987) Fructose-Induced Insulin<br />

Resistance and Hypertension in Rats. Hypertension 10: 512-516.<br />

40. Cancello, R. & Clement, K. (2006) Is obesity an inflammatory illness Role <strong>of</strong> low-grade<br />

inflammation and macrophage infiltration in human white adipose tissue. BJOG: An<br />

International Journal <strong>of</strong> Obstetrics and Gynaecology 113: 1141-1147.<br />

28


635<br />

636<br />

637<br />

638<br />

639<br />

640<br />

641<br />

642<br />

643<br />

644<br />

645<br />

646<br />

647<br />

648<br />

649<br />

650<br />

651<br />

652<br />

653<br />

654<br />

655<br />

656<br />

657<br />

658<br />

659<br />

660<br />

661<br />

662<br />

663<br />

664<br />

665<br />

666<br />

667<br />

668<br />

669<br />

670<br />

671<br />

672<br />

41. Guilherme, A., Virbasius, J. V., Puri, V. & Czech, M. P. (2008) Adipocyte dysfunctions<br />

linking obesity to insulin resistance and type 2 diabetes. Nat Rev Mol Cell Biol 9: 367-377.<br />

42. Coate, K. C. & Huggins, K. W. (2010) Consumption <strong>of</strong> a high glycemic index diet increases<br />

abdominal adiposity but does not influence adipose tissue pro-oxidant and antioxidant gene<br />

expression in C57BL/6 mice. Nutr Res 30(2): 141-150 (abs.).<br />

43. Yamazaki, K., Yasuda, N., Inoue, T., Yamamoto, E., Sugaya, Y., Nagakura, T., Shinoda,<br />

M., Clark, R., Saeki, T. & Tanaka, I. (2007) Effects <strong>of</strong> the Combination <strong>of</strong> a Dipeptidyl<br />

Peptidase IV Inhibitor and an Insulin Secretagogue on Glucose and Insulin Levels in Mice<br />

and Rats. J Pharmacol Exp <strong>The</strong>r 320: 738-746.<br />

44. Fagerholm, V., Scheinin, M. & Haaparanta, M. (2008) alpha2A-adrenoceptor antagonism<br />

increases insulin secretion and synergistically augments the insulinotropic effect <strong>of</strong><br />

glibenclamide in mice. Br J Pharmacol 154: 1287-1296.<br />

45. Miki, H., Yamauchi, T., Suzuki, R., Komeda, K., Tsuchida, A., Kubota, N., Terauchi, Y.,<br />

Kamon, J., Kaburagi, Y. et al. (2001) Essential Role <strong>of</strong> Insulin Receptor Substrate 1 (IRS-1)<br />

and IRS-2 in Adipocyte Differentiation. Mol. Cell. Biol. 21: 2521-2532.<br />

46. Kim, J. K., Michael, M. D., Previs, S. F., Peroni, O. D., Mauvais-Jarvis, F., Neschen, S.,<br />

Kahn, B. B., Kahn, C. R. & Shulman, G. I. (2000) Redistribution <strong>of</strong> substrates to adipose<br />

tissue promotes obesity in mice with selective insulin resistance in muscle. <strong>The</strong> Journal <strong>of</strong><br />

Clinical Investigation 105: 1791-1797.<br />

47. Cariou, B., Postic, C., Boudou, P., Burcelin, R., Kahn, C. R., Girard, J., Burnol, A. F. &<br />

Mauvais-Jarvis, F. (2004) Cellular and Molecular Mechanisms <strong>of</strong> Adipose Tissue Plasticity<br />

in Muscle Insulin Receptor Knockout Mice. Endocrinology 145: 1926-1932.<br />

48. Shepherd, P. R., Gnudi, L., Tozzo, E., Yang, H., Leach, F. & Kahn, B. B. (1993) Adipose<br />

cell hyperplasia and enhanced glucose disposal in transgenic mice overexpressing GLUT4<br />

selectively in adipose tissue. J. Biol. Chem. 268: 22243-22246.<br />

49. Cusin, I., Rohner-Jeanrenaud, F., Terrettaz, J. & Jeanrenaud, B. (1992) Hyperinsulinemia<br />

and its impact on obesity and insulin resistance. Int J Obes Relat Metab Disord. 16: S1-S11.<br />

50. Cusin, I., Terrettaz, J., Rohner-Jeanrenaud, F. & Jeanrenaud, B. (1990) Metabolic<br />

consequences <strong>of</strong> hyperinsulinaemia imposed on normal rats on glucose handling by white<br />

adipose tissue, muscles and liver. Biochem J. 267: 99-103.<br />

51. Cusin, I., Terrettaz, J., Rohner-Jeanrenaud, F., Zarjevski, N., Assimacopoulos-Jeannet, F. &<br />

Jeanrenaud, B. (1990) Hyperinsulinemia increases the amount <strong>of</strong> GLUT4 mRNA in white<br />

adipose tissue and decreases that <strong>of</strong> muscles: a clue for increased fat depot and insulin<br />

resistance. Endocrinology. 127: 3246-3248.<br />

52. Mynatt, R. L., Miltenberger, R. J., Klebig, M. L., Zemel, M. B., Wilkinson, J. E., Wilkinson,<br />

W. O. & Woychik, R. P. (1997) Combined <strong>effects</strong> <strong>of</strong> insulin treatment and adipose tissuespecific<br />

agouti expression on the development <strong>of</strong> obesity. Proc Natl Acad Sci U S A. 94:<br />

919-922.<br />

29


673<br />

674<br />

675<br />

676<br />

677<br />

678<br />

679<br />

680<br />

681<br />

682<br />

683<br />

684<br />

685<br />

686<br />

687<br />

688<br />

689<br />

690<br />

691<br />

692<br />

693<br />

694<br />

695<br />

696<br />

697<br />

698<br />

699<br />

700<br />

701<br />

702<br />

703<br />

704<br />

705<br />

706<br />

707<br />

708<br />

709<br />

710<br />

53. Mitri, J. & Hamdy, O. (2009) Diabetes medications and body weight. Expert Opin Drug Saf.<br />

8: 573-584.<br />

54. Fonseca V (2003) Effect <strong>of</strong> thiazolidinediones on body weight in patients with diabetes<br />

mellitus. Am J Med. 115: 42s-48s.<br />

55. Marsset-Baglieri, A., Fromentin, G., Tome, D., Bensaid, A., Makkarios, L. & Even, P. C.<br />

(2004) Increasing the Protein Content in a Carbohydrate-Free Diet Enhances Fat Loss<br />

during 35% but Not 75% Energy Restriction in Rats. J. Nutr. 134: 2646-2652.<br />

56. Morris, K. L., Namey, T. C. & Zemel, M. B. (2003) Effects <strong>of</strong> dietary carbohydrate on the<br />

development <strong>of</strong> obesity in heterozygous Zucker rats. <strong>The</strong> Journal <strong>of</strong> Nutritional<br />

Biochemistry 14: 32-39.<br />

57. Alemzadeh, R., Jacobs, W. & Pitukcheewanont, P. (1996) Antiobesity effect <strong>of</strong> diazoxide in<br />

obese zucker rats. Metabolism 45: 334-341.<br />

58. Nagata, R., Nishio, Y., Sekine, O., Nagai, Y., Maeno, Y., Ugi, S., Maegawa, H. &<br />

Kashiwagi, A. (2004) Single Nucleotide Polymorphism (Çô468 Gly to Ala) at the<br />

Promoter Region <strong>of</strong> Sterol Regulatory Element-binding Protein-1c Associates with Genetic<br />

Defect <strong>of</strong> Fructose-induced Hepatic Lipogenesis. J. Biol. Chem. 279: 29031-29042.<br />

59. Pawlak, D. B., Bryson, J. M., Denyer, G. S. & Brand-Miller, J. C. (2001) High Glycemic<br />

Index Starch Promotes Hypersecretion <strong>of</strong> Insulin and Higher Body Fat in Rats without<br />

Affecting Insulin Sensitivity. J. Nutr. 131: 99-104.<br />

60. Larsen, T. M., Dalskov, S. M., van Baak, M., Jebb, S. A., Papadaki, A., Pfeiffer, A. F. H.,<br />

Martinez, J. A., Handjieva-Darlenska, T., Kunesova, M. et al. (2010) Diets with High or<br />

Low Protein Content and Glycemic Index for Weight-Loss Maintenance. New England<br />

Journal <strong>of</strong> Medicine 363: 2102-2113.<br />

61. Puigserver, P., Rhee, J., Donovan, J., Walkey, C. J., Yoon, J. C., Oriente, F., Kitamura, Y.,<br />

Altomonte, J., Dong, H. et al. (2003) Insulin-regulated hepatic gluconeogenesis through<br />

FOXO1-PGC-1[alpha] interaction. Nature 423: 550-555.<br />

62. Yoon, J. C., Puigserver, P., Chen, G., Donovan, J., Wu, Z., Rhee, J., Adelmant, G., Stafford,<br />

J., Kahn, C. R. et al. (2001) Control <strong>of</strong> hepatic gluconeogenesis through the transcriptional<br />

coactivator PGC-1. Nature 413: 131-138.<br />

63. Herzig, S., Long, F., Jhala, U. S., Hedrick, S., Quinn, R., Bauer, A., Rudolph, D., Schutz,<br />

G., Yoon, C. et al. (2001) CREB regulates hepatic gluconeogenesis through the coactivator<br />

PGC-1. Nature 413: 179-183.<br />

64. Fine, E. J. & Feinman, R. D. (2004) <strong>The</strong>rmodynamics <strong>of</strong> weight loss diets. Nutrition and<br />

metabolism 1: 15-23.<br />

65. Madsen, L., Pedersen, L. M., Lillefosse, H. H., Fjaere, E., Bronstad, I., Hao, Q., Petersen, R.<br />

K., Hallenborg, P., Ma, T. et al. (2010) UCP1 Induction during Recruitment <strong>of</strong> Brown<br />

Adipocytes in White Adipose Tissue Is Dependent on Cyclooxygenase Activity. PLoS ONE<br />

5.<br />

30


711<br />

712<br />

713<br />

714<br />

715<br />

716<br />

717<br />

718<br />

719<br />

720<br />

721<br />

722<br />

66. Vegiopoulos, A., Muller-Decker, K., Strzoda, D., Schmitt, I., Chichelnitskiy, E., Ostertag,<br />

A., Diaz, M. B., Rozman, J., Hrabe de Angelis, M. et al. (2010) Cyclooxygenase-2 Controls<br />

Energy Homeostasis in Mice by de Novo Recruitment <strong>of</strong> Brown Adipocytes. Science 328:<br />

1158-1161.<br />

67. Kopecky, J., Hodny, Z., Rossmeisl, M., Syrovy, I. & Kozak, L. P. (1996) Reduction <strong>of</strong><br />

dietary obesity in aP2-Ucp transgenic mice: physiology and adipose tissue distribution. Am<br />

J Physiol. 270: E768-E775.<br />

68. Cordain, L., Eaton, S. B., Sebastian, A., Mann, N., Lindeberg, S., Watkins, B. A., O'Keefe,<br />

J. H. & Brand-Miller, J. (2005) Origins and evolution <strong>of</strong> the Western diet: health<br />

implications for the 21st century. Am J Clin Nutr 81: 341-354.<br />

31


UCP1 Induction during Recruitment <strong>of</strong> Brown Adipocytes<br />

in White Adipose Tissue Is Dependent on<br />

Cyclooxygenase Activity<br />

Lise Madsen 1,2 *, Lone M. Pedersen 3 , Haldis Haukaas Lillefosse 1,2 , Even Fjære 1,2 , Ingeborg Bronstad 4 , Qin<br />

Hao 1 , Rasmus K. Petersen 1 , Philip Hallenborg 1 , Tao Ma 1 , Rita De Matteis 5 , Pedro Araujo 2 , Josep<br />

Mercader 6 , M. Luisa Bonet 6 , Jacob B. Hansen 7 , Barbara Cannon 8 , Jan Nedergaard 8 , Jun Wang 1,9 , Saverio<br />

Cinti 10 , Peter Voshol 11 , Stein Ove Døskeland 4 , Karsten Kristiansen 1,9 *<br />

1 Department <strong>of</strong> Biology, University <strong>of</strong> Copenhagen, Copenhagen, Denmark, 2 National Institute <strong>of</strong> Nutrition and Seafood Research, Bergen, Norway, 3 Department <strong>of</strong><br />

Biochemistry and Molecular Biology, University <strong>of</strong> Southern Denmark, Odense, Denmark, 4 Department <strong>of</strong> Biomedicine, University <strong>of</strong> Bergen, Bergen, Norway,<br />

5 Department <strong>of</strong> Biomolecular Sciences, University <strong>of</strong> Urbino, Urbino, Italy, 6 Laboratory <strong>of</strong> Molecular Biology, Nutrition and Biotechnology, Universitat de les Illes Balears,<br />

and CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Palma de Mallorca, Spain, 7 Department <strong>of</strong> Biomedical Sciences, University <strong>of</strong> Copenhagen,<br />

Copenhagen, Denmark, 8 <strong>The</strong> Wenner-Gren Institute, Stockholm University, Stockholm, Sweden, 9 BGI-Shenzhen, Shenzhen, China, 10 Department <strong>of</strong> Molecular<br />

Pathology and Innovative <strong>The</strong>rapies, University <strong>of</strong> Ancona, Ancona, Italy, 11 Metabolic Research Laboratories, University <strong>of</strong> Cambridge, Cambridge, United Kingdom<br />

Abstract<br />

Background: <strong>The</strong> uncoupling protein 1 (UCP1) is a hallmark <strong>of</strong> brown adipocytes and pivotal for cold- and diet-induced<br />

thermogenesis.<br />

Methodology/Principal Findings: Here we report that cyclooxygenase (COX) activity and prostaglandin E 2 (PGE 2 ) <strong>are</strong><br />

crucially involved in induction <strong>of</strong> UCP1 expression in inguinal white adipocytes, but not in classic interscapular brown<br />

adipocytes. Cold-induced expression <strong>of</strong> UCP1 in inguinal white adipocytes was repressed in COX2 knockout (KO) mice and<br />

by administration <strong>of</strong> the COX inhibitor indomethacin in wild-type mice. Indomethacin repressed b-adrenergic induction <strong>of</strong><br />

UCP1 expression in primary inguinal adipocytes. <strong>The</strong> use <strong>of</strong> PGE 2 receptor antagonists implicated EP 4 as a main PGE 2<br />

receptor, and injection <strong>of</strong> the stable PGE 2 analog (EP 3/4 agonist) 16,16 dm PGE 2 induced UCP1 expression in inguinal white<br />

adipose tissue. Inhibition <strong>of</strong> COX activity attenuated diet-induced UCP1 expression and increased energy efficiency and<br />

adipose tissue mass in obesity-resistant mice kept at thermoneutrality.<br />

Conclusions/Significance: Our findings provide evidence that induction <strong>of</strong> UCP1 expression in white adipose tissue, but not<br />

in classic interscapular brown adipose tissue is <strong>dependent</strong> on cyclooxygenase activity. Our results indicate that<br />

cyclooxygenase-<strong>dependent</strong> induction <strong>of</strong> UCP1 expression in white adipose tissues is important for diet-induced<br />

thermogenesis providing support for a surprising role <strong>of</strong> COX activity in the control <strong>of</strong> energy balance and obesity<br />

development.<br />

Citation: Madsen L, Pedersen LM, Lillefosse HH, Fjære E, Bronstad I, et al. (2010) UCP1 Induction during Recruitment <strong>of</strong> Brown Adipocytes in White Adipose Tissue<br />

Is Dependent on Cyclooxygenase Activity. PLoS ONE 5(6): e11391. doi:10.1371/journal.pone.0011391<br />

Editor: Aimin Xu, University <strong>of</strong> Hong Kong, China<br />

Received May 19, 2010; Accepted May 30, 2010; Published June 30, 2010<br />

Copyright: ß 2010 Madsen et al. This is an open-access article distributed under the terms <strong>of</strong> the Creative Commons Attribution License, which permits<br />

unrestricted use, distribution, and reproduction in any medium, provided the original author and source <strong>are</strong> credited.<br />

Funding: This work was supported by the Danish Natural Science Research Council, the Novo Nordisk Foundation and the Carlsberg Foundation. Part <strong>of</strong> the<br />

work was carried out as a part <strong>of</strong> the research program <strong>of</strong> the Danish Obesity Research Centre (DanORC). DanORC is supported by <strong>The</strong> Danish Council for Strategic<br />

Research (Grant No 2101 06 0005). CIBER de Fisiopatologia de la Obesidad y Nutrición is an initiative <strong>of</strong> the ISCIII. <strong>The</strong> funders had no role in study design, data<br />

collection and analysis, decision to publish, or preparation <strong>of</strong> the manuscript.<br />

Competing Interests: <strong>The</strong> authors have decl<strong>are</strong>d that no competing interests exist.<br />

* E-mail: kk@bio.ku.dk (KK); lise.madsen@nifes.no (LM)<br />

Introduction<br />

<strong>The</strong> two types <strong>of</strong> adipose tissues, white (WAT) and brown (BAT),<br />

have opposite functions in whole body energy homeostasis. Whereas<br />

white adipocytes store excess energy as fat, brown adipocytes<br />

contain a large number <strong>of</strong> mitochondria dedicated to convert fat<br />

into heat through uncoupled respiration. <strong>The</strong> uncoupling <strong>of</strong><br />

respiration and the resulting heat dissipation depend on the<br />

expression <strong>of</strong> the uncoupling protein 1 (UCP1). UCP1 is an integral<br />

membrane protein unique to brown adipocyte mitochondria, where<br />

it acts as a proton channel to uncouple oxidative phosphorylation by<br />

dissipating the proton gradient across the inner mitochondrial<br />

membrane [1]. In mice, an increased content <strong>of</strong> UCP1 in adipose<br />

tissue mitochondria is strongly linked to protection against dietinduced<br />

obesity. This is true whether increased UCP1 expression is<br />

induced by transgenic expression <strong>of</strong> UCP1 itself [2;3], <strong>of</strong> forkhead<br />

box 2 (FOXC2) [4], <strong>of</strong> PR domain containing 16 (PRDM16) [5] or<br />

by disruption <strong>of</strong> the RIIb subunit <strong>of</strong> protein kinase A [6;7],<br />

eukaryotic translation initiation factor E4-binding protein 1 (4E-<br />

BP1) [8], cell death inducing DFFA like effector A and C (Cidea and<br />

Cidec/Fsp27) [9], the p160 coregulator TIF2 [10] or retinoblastoma<br />

Rb [11–13].<br />

PLoS ONE | www.plosone.org 1 June 2010 | Volume 5 | Issue 6 | e11391


Cyclooxygenases and UCP1<br />

Although it has been estimated that 50 g <strong>of</strong> brown adipocytes<br />

would be sufficient to burn 20% <strong>of</strong> the daily energy intake [14],<br />

BAT has traditionally been considered to be virtually absent and<br />

<strong>of</strong> no physiological relevance in adult humans. This view has<br />

recently changed dramatically with the demonstration <strong>of</strong> functional<br />

BAT in adult humans [15–19] adding to the observation <strong>of</strong><br />

brown-like multilocular adipocytes expressing UCP1 interspersed<br />

within human WAT [20–22]. Actually, UCP1 mRNA has been<br />

detected in all adipose tissues in adult humans, and it has been<br />

estimated that 1 in 100–200 adipocytes in human intraperitoneal<br />

adipose tissue expresses UCP1 [23].<br />

Classic interscapular brown adipocytes and brown-like adipocytes<br />

found in WAT depots appear to originate from distinct<br />

lineages. Brown pre-adipocytes derived from the interscapular<br />

region (iBAT) demonstrate myogenic gene expression [24] and<br />

classic brown adipocytes arise from Myf5 expressing progenitors<br />

[25]. In contrast, brown-like adipocytes appearing in white<br />

adipose tissue by b-adrenergic stimulation (‘‘brite adipocytes’’)<br />

appear to originate from another lineage, much closer to white<br />

adipocytes [26–29] and display different molecular markers [30].<br />

Several lines <strong>of</strong> evidence suggest that the number <strong>of</strong> brown-like<br />

adipocytes in WAT depots might influence whole body energy<br />

balance. Increased occurrence <strong>of</strong> brown-like adipocytes within<br />

WAT depots is a feature <strong>of</strong> mouse strains resistant to dietary<br />

obesity, such as the A/J strain [31;32], and reduced adiposity<br />

associated with aP2-transgenic expression <strong>of</strong> UCP1 is linked to<br />

increased energy dissipation in white, but not interscapular brown,<br />

adipose tissue [33]. Human obesity is associated with a reduced<br />

expression <strong>of</strong> UCP1 and other thermogenesis related genes in<br />

WAT depots [34;35]. Thus, identification <strong>of</strong> factors controlling<br />

induction <strong>of</strong> UCP1 expression and an increase in the number <strong>of</strong><br />

brown-like adipocytes in white depots obviously deserves further<br />

attention.<br />

It is intriguing that the cold-induced occurrence <strong>of</strong> brown-like<br />

adipocytes and UCP1 requires the presence <strong>of</strong> the b 3 -adrenoceptor<br />

in previously white adipose tissue, but not in interscapular<br />

brown adipose tissue [36]. Furthermore, the presence <strong>of</strong> the b 3 -<br />

adrenoceptor is required for full stimulation <strong>of</strong> energy expenditure<br />

and oxygen consumption in white adipose tissue [37].<br />

Adipocytes from lean rats have higher isoprenalin-stimulated<br />

prostaglandin E 2 (PGE 2 ) synthesis, than adipocytes from obese<br />

Zucker rats [38]. We therefore hypothesized that prostaglandins<br />

or related products synthesized by cyclooxygenases (COXs)<br />

might be involved in the recruitment <strong>of</strong> brown adipocytes in<br />

white depots. <strong>The</strong> COXs have previously been implicated in<br />

adipogenesis [39–41], but no specific role has been assigned. Here,<br />

we demonstrate that COX activity is crucially involved in the<br />

induction <strong>of</strong> UCP1 expression in WAT providing further evidence<br />

for a role <strong>of</strong> COXs in the control <strong>of</strong> energy balance and obesity<br />

development. In view <strong>of</strong> the worldwide epidemic <strong>of</strong> obesity and<br />

associated metabolic disorders it is obviously <strong>of</strong> importance to<br />

identify pathways that can be manipulated genetically or<br />

pharmacologically and regulate the induction <strong>of</strong> UCP1 expression<br />

and recruitment <strong>of</strong> brown-like adipocytes in white adipose tissues.<br />

Results<br />

COX1 and COX2 protein expression is upregulated in<br />

iWAT during cold treatment<br />

When mice <strong>are</strong> kept at 28uC, close to thermoneutrality, the<br />

majority <strong>of</strong> the adipocytes in the inguinal white adipose depot<br />

(iWAT) – the major subcutaneous depot in the mouse – appear as<br />

UCP1-negative, spherical unilocular adipocytes [42]. In iWAT<br />

from warm-acclimated mice, only endothelial cells and macrophages<br />

stained positive for COX1 and COX2, respectively<br />

(Figure 1A and B). However, when mice were transferred to a<br />

cold environment, the emerging multilocular adipocytes stained<br />

positive for COX1 and COX2. In particular, cells that appe<strong>are</strong>d<br />

to be in a transition state between uni- and multilocular cells<br />

stained strongly (Figure 1A and B). Western blotting demonstrated<br />

increased expression <strong>of</strong> COX1 and COX2 in iWAT, and also in<br />

iBAT, after cold exposure (Figure 1C). Real time qPCR analysis<br />

verified that genes preferentially expressed in brown vs. white<br />

adipose tissue, such as UCP1, peroxisome proliferator activated<br />

receptor gamma coactivator 1a (PGC1a), type II thyroxine<br />

deiodinase (Dio2), cytochrome C oxidase subunit 8b (Cox8b),<br />

epithelial V like antigen 1 (Eva1) and Cidea were all highly<br />

induced in iWAT upon cold exposure, whereas expression <strong>of</strong> 4E-<br />

BP1 and <strong>of</strong> nuclear receptor interacting protein 140 (Nrip1/<br />

RIP140) was reduced (Figure 1D). Immunohistochemical staining<br />

<strong>of</strong> iBAT from cold-treated mice demonstrated that adipocytes<br />

stained positive for COX2, whereas only endothelial cells stained<br />

positive for COX1 (Figure S1A). <strong>The</strong> lack <strong>of</strong> COX1 and COX2<br />

expression in adipocytes from iBAT in warm-acclimated mice was<br />

verified by analysis <strong>of</strong> protein and RNA isolated from fractionated<br />

adipose tissue, in which COX1 and COX2 was detected solely in<br />

the stromal vascular fraction (Figure S1B).<br />

Inhibition <strong>of</strong> COX activity represses induction <strong>of</strong> UCP1<br />

expression<br />

Differentiated mouse embryo fibroblasts (MEFs) lacking the<br />

retinoblastioma (Rb) gene, resemble brown or brown-like<br />

adipocytes in demonstrating b-adrenergic induction <strong>of</strong> UCP1<br />

expression [43]. To achieve a robust induction <strong>of</strong> UCP1<br />

expression, differentiated Rb 2/2 MEFs were treated with a<br />

combination <strong>of</strong> isoproterenol and 9-cis retinoic acid [44]. Just as<br />

cold exposure increased COX1 and COX2 mRNA and protein<br />

levels in brown-like adipocytes (Figure 1), isoproterenol/9-cis<br />

retinoic acid treatment increased COX1 and COX2 mRNA and<br />

protein expression (Figure 2A and B) in this model system.<br />

Upregulation <strong>of</strong> COX1 and COX2 expression in Rb 2/2<br />

adipocytes was accompanied by increased production <strong>of</strong> PGE 2 ,<br />

the primary prostaglandin produced by mature adipocytes<br />

[45;46], but not <strong>of</strong> PGF 2a and 6-keto-PGF 1a (Figure 2C). This<br />

indicates that Rb 2/2 adipocytes resemble mature adipocytes in<br />

producing PGE 2 as the major prostaglandin species.<br />

To investigate the importance <strong>of</strong> COX activity for induction <strong>of</strong><br />

UCP1 expression, we treated differentiated Rb 2/2 adipocytes<br />

with isoproterenol/9-cis retinoic acid in the absence or presence <strong>of</strong><br />

the general COX inhibitor indomethacin. Indomethacin prevented<br />

induction <strong>of</strong> UCP1 mRNA and protein expression (Figure 2D<br />

and E), thus suggesting the intriguing possibility that COX activity<br />

is required for induction <strong>of</strong> UCP1.<br />

To examine if COX activity was required also in primary<br />

adipocytes, we induced cells from the stromal vascular fraction <strong>of</strong><br />

iBAT and iWAT to differentiate and then treated the mature<br />

adipocytes with the b-adrenergic agonist isoproterenol in the<br />

absence and presence <strong>of</strong> indomethacin. Interestingly, indomethacin<br />

inhibited isoproterenol-induced UCP1 expression in cells<br />

derived from iWAT but not from iBAT (Figure 3A), indicating<br />

that COX activity is required for b-adrenergic induction <strong>of</strong> UCP1<br />

expression in adipocytes from iWAT, but not in iBAT adipocytes.<br />

In keeping with this notion, indomethacin only marginally<br />

attenuated induction <strong>of</strong> UCP1 expression in the WT-1 cell model<br />

representing interscapular brown adipocytes (Text S1, Figure S2)<br />

[47].<br />

To investigate the role <strong>of</strong> COX activity during induction <strong>of</strong><br />

UCP1 expression in iWAT and iBAT in vivo, we treated warm-<br />

PLoS ONE | www.plosone.org 2 June 2010 | Volume 5 | Issue 6 | e11391


Cyclooxygenases and UCP1<br />

Figure 1. Cold exposure induces COX1 and COX2 expression in iWAT and iBAT. Sv129 mice were warm-acclimated at 28–30uC for 6 days<br />

and then transferred to 4–6uC. Samples for cryosections, RNA and protein extractions were prep<strong>are</strong>d from iBAT and iWAT after 2, 4 and 6 days at 4–<br />

6uC. A–B. Representative COX1 (A) and COX2 (B) immunoreactivity in iWAT from mice kept at 28–30uC for 6 days and after 6 days <strong>of</strong> cold exposure. C.<br />

Proteins were isolated from warm-acclimated mice (lane 1) and after 2, 4 and 6 days <strong>of</strong> cold exposure. COX1 and COX2 expression were determined<br />

by Western blotting. D. RNA was harvested from iBAT and iWAT from individual mice (n = 4 in each group) that were warm-acclimated or coldexposed<br />

for 6 days. Expressions <strong>of</strong> genes were measured by RT-qPCR in duplicates and normalized to TBP (TATA box binding protein). <strong>The</strong> bars<br />

represent mean 6 standard error. * indicates statistical difference (p,0.05) comp<strong>are</strong>d to expression in warm acclimated mice.<br />

doi:10.1371/journal.pone.0011391.g001<br />

acclimated mice with the COX inhibitor indomethacin and<br />

transferred the mice to 4uC. Measurements <strong>of</strong> rectal temperature<br />

revealed that mice treated with indomethacin had slightly, but<br />

significantly lower body temperature (Figure 3B). As expected,<br />

UCP1 expression was induced in both iBAT and iWAT in<br />

vehicle-treated mice (Figure 3C and D). While indomethacin<br />

treatment only slightly attenuated cold-induced UCP1 expression<br />

in iBAT, it almost completely prevented the induction <strong>of</strong> UCP1<br />

expression in iWAT (Figure 3C and D). Thus, COX activity<br />

appe<strong>are</strong>d to be necessary for cold-induced UCP1 expression in<br />

iWAT, but not in iBAT. In addition, indomethacin treatment<br />

attenuated cold-induced enhancement <strong>of</strong> PGC1a, Dio2,Cox8b,<br />

Eva1 and Cidea expression in iWAT, while preventing coldinduced<br />

repression <strong>of</strong> RIP140 and 4E-BP1 expression in iWAT<br />

(Figure 3D).<br />

Forced expression <strong>of</strong> COX2 induces UCP1 expression in<br />

Rb 2/2 adipocytes<br />

Since indomethacin attenuated b-adrenergically stimulated<br />

UCP1 expression in Rb 2/2 adipocytes and primary inguinal<br />

PLoS ONE | www.plosone.org 3 June 2010 | Volume 5 | Issue 6 | e11391


Cyclooxygenases and UCP1<br />

Figure 2. Indomethacin prevents isoproterenol-induced UCP1 expression in Rb-negative adipocytes. Rb-negative mouse embryo<br />

fibroblasts were induced to differentiate as described in experimental procedures. Differentiated cells were treated with vehicle or isoproterenol<br />

(100 nM) and 9-cis-retinoic acid (1 mM) for 24 h. Indomethacin (1 mM) was included when indicated in the figure. A and D. RNA was isolated and<br />

expressions <strong>of</strong> genes were measured by RT-qPCR in duplicates and normalized to TBP. C. <strong>The</strong> levels <strong>of</strong> prostaglandin E 2 ,F 2a and 6-keto-prostaglandin<br />

F 1a were determined in cell medium using ELISA kits after 24 h. B and E. Proteins were harvested and expressions <strong>of</strong> COX1, COX2 and UCP1 were<br />

measured by Western blotting. <strong>The</strong> bars represent mean 6 standard error. <strong>The</strong> experiments were performed in triplicates and performed 3–5 times.<br />

* indicates statistical significant difference (p,0.05).<br />

doi:10.1371/journal.pone.0011391.g002<br />

adipocytes, but not in WT-1 cells and primary interscapular<br />

brown adipocytes, we again used Rb 2/2 adipocytes as a model<br />

system for ‘‘brite’’ adipocytes. To investigate the relative<br />

importance <strong>of</strong> COX1 and COX2 activities in mediating induction<br />

<strong>of</strong> UCP1 expression in such cells, we treated Rb 2/2 adipocytes<br />

with isoproterenol/9-cis retinoic acid in the absence and presence<br />

<strong>of</strong> selective COX1 and COX2 inhibitors. As shown in Figure 4A,<br />

selective inhibition <strong>of</strong> COX1 and COX2 with SC560 or NS398,<br />

respectively, partially prevented UCP1 induction, whereas a<br />

combination <strong>of</strong> these inhibitors or treatment with the non-selective<br />

inhibitor indomethacin fully prevented UCP1 induction. Accordingly,<br />

activities <strong>of</strong> both COX1 and COX2 seem necessary for full<br />

UCP1 induction.<br />

To further examine the relative importance <strong>of</strong> COX1 and<br />

COX2 for prostaglandin synthesis and UCP1 expression, these<br />

enzymes were retrovirally expressed both singly and in combination<br />

in Rb 2/2 MEFs (Figure 4B). <strong>The</strong> cells were induced to<br />

differentiate, and on day 8, the medium was replaced by fresh<br />

medium, which was harvested 24 h later and analyzed for PGE 2<br />

content. <strong>The</strong> level <strong>of</strong> PGE 2 was higher when the cells were<br />

transduced with COX2 alone or in combination with COX1,<br />

than with COX1 alone (Figure 4C). <strong>The</strong>se results, together with<br />

the fact that PGE 2 formation in adipose tissue in COX2 KO<br />

mice is significantly lower than in COX1 KO mice [48], point to<br />

COX2 expression as being <strong>of</strong> major importance for PGE 2<br />

production. In accordance with this, forced expression <strong>of</strong> COX1<br />

alone was unable to induce UCP1 expression (Figure 4D).<br />

However, UCP1 expression was significantly induced by forced<br />

expression <strong>of</strong> COX2 alone or in combination with COX1<br />

(Figure 4D). Increased expression <strong>of</strong> UCP1 was accompanied<br />

with increased expression <strong>of</strong> PGC1a, Dio2, Cox8b, Eva1 and<br />

Cidea, as well as reduced expression <strong>of</strong> RIP140, but not 4E-BP1<br />

(Figure 4D).<br />

Cold-induced UCP1 expression is attenuated in iWAT in<br />

COX2 KO mice<br />

To confirm the importance <strong>of</strong> COX2 for UCP1 induction in<br />

iWAT, wild-type and COX2 KO mice were challenged with a<br />

cold environment after warm acclimation. <strong>The</strong> wild-type mice<br />

defended their body temperature better than the COX2 KO mice<br />

(Figure 5A). <strong>The</strong> COX2 KO mice develop severe nephropathy<br />

and <strong>are</strong> susceptible to peritonitis in early life [49]; therefore, KO<br />

and wild-type littermates 6 weeks <strong>of</strong> age were used in this<br />

experiment. Unfortunately, we were unable to collect sufficient<br />

amounts <strong>of</strong> iWAT from these young mice to detect UCP1 or COX<br />

by Western blotting. However, as expected, cold-induced UCP1<br />

mRNA expression was attenuated in iWAT in COX2 KO mice<br />

(Figure 5B). Cold-induced expression <strong>of</strong> Dio2 and Cidea was also<br />

attenuated in iWAT in the COX2 KO mice and PGC1a<br />

expression also tended to be attenuated (Figure 5B). Moreover,<br />

the cold-induced reduction <strong>of</strong> RIP140 expression was prevented in<br />

the COX2 KO mice (Figure 5B). Expression <strong>of</strong> Cox8b, Eva1 and<br />

4E BP1 was, however, not significantly different in iWAT from<br />

wild-type and COX2 KO mice, suggesting that inhibition <strong>of</strong> both<br />

COX1 and COX2 might be necessary to attenuate cold-induced<br />

changes in the expression <strong>of</strong> these genes. As expected, we observed<br />

no differences in UCP1 expression in iBAT in COX2 KO and<br />

wild-type mice, and surprisingly, cold-treated COX2 KO mice<br />

had significantly higher expression <strong>of</strong> PGC1a in iBAT than did<br />

wild-type mice (Figure 5B).<br />

PGE 2 induces UCP1 expression via activation <strong>of</strong> the EP 3 /<br />

EP 4 receptors<br />

PGE 2 is reported to mediate its action by interacting with four<br />

subtypes <strong>of</strong> PGE receptors, the EP 1 ,EP 2 ,EP 3 and EP 4 receptors<br />

[50], but may also bind to the prostaglandin F (FP) receptor with an<br />

PLoS ONE | www.plosone.org 4 June 2010 | Volume 5 | Issue 6 | e11391


Cyclooxygenases and UCP1<br />

Figure 3. Indomethacin prevents cold-induced UCP1 expression in iWAT. A. Stromal vascular fractions (SVFs) were isolated from mouse<br />

iWAT and iBAT, cultured and induced to differentiate as described in experimental procedures. Differentiated adipocytes were treated with vehicle or<br />

isoproterenol (100 nM) in the absence or presence <strong>of</strong> indomethacin (1 mM) for 24 h. Expression <strong>of</strong> UCP1 was measured by RT-qPCR in duplicates and<br />

normalized to PPARc. <strong>The</strong> bars represent mean 6 standard deviation. <strong>The</strong> experiment was performed in triplicates and repeated 3 times. B–D. C57Bl/<br />

6 mice were warm-acclimated at 28–30uC for 10 days and then transferred to 4–6uC for 48 h. Mice were injected with vehicle or indomethacin<br />

(2.5 mg/kg) 2 h prior transfer to 4uC and thereafter every 12 h. Rectal temperature was measured before the mice were transferred and after 24 and<br />

48 h (B). Protein and RNA extractions were isolated after 48 h. UCP1 expression was measured by Western blotting (C) and expressions <strong>of</strong> genes were<br />

measured by RT-qPCR and normalized to TBP (D). <strong>The</strong> bars represent mean 6 error (n = 5–6). * indicates statistical significant difference (p,0.05).<br />

doi:10.1371/journal.pone.0011391.g003<br />

affinity that is only 10–30 fold lower than that <strong>of</strong> PGF 2a [51]. In<br />

order to probe the relative importance <strong>of</strong> these receptors in<br />

mediating the possible effect <strong>of</strong> PGE 2 on induction <strong>of</strong> UCP1,<br />

expression <strong>of</strong> the EP and FP receptors was measured in adipose<br />

tissue and in Rb 2/2 adipocytes. All receptors were expressed in<br />

both white and brown adipose tissue, whereas no expression <strong>of</strong> the<br />

EP 3 receptor could be detected in Rb 2/2 adipocytes (Figure 6A),<br />

implying a minor if any role <strong>of</strong> this receptor in mediating the PGE 2<br />

response in those cells. Thus, Rb 2/2 adipocytes were treated with<br />

isoproterenol and 9-cis retinoic in the absence or presence <strong>of</strong><br />

AL8810, AH6809, or AH23848, that <strong>are</strong> FP-, EP 1 /EP 2 and EP 4<br />

receptor antagonists, respectively. Isoproterenol-stimulated UCP1<br />

expression was not affected by the FP receptor antagonist, but<br />

slightly attenuated by the EP 1 /EP 2 receptor antagonist and strongly<br />

attenuated by the EP 4 antagonist (Figure 6B). Reduced expression<br />

<strong>of</strong> UCP1 was accompanied by reduced expression <strong>of</strong> PGC1a and<br />

Cidea (Figure 6B). To verify the importance <strong>of</strong> PGE 2 signaling via<br />

the EP 4 receptor with a possible minor contribution by the EP 3<br />

receptor, mice were injected with an EP 3 /EP 4 receptor agonist [52],<br />

the stable PGE 2 analogue 16,16-dimethyl-PGE 2 As predicted,<br />

qPCR analysis revealed that UCP1 expression was induced in<br />

iWAT, but not in iBAT (Figure 6C). Together, the in vitro and in vivo<br />

results suggest that PGE 2 -induced UCP1 expression at least in part<br />

is mediated via the EP 3 /EP 4 receptors with EP 4 being the<br />

predominant receptor involved.<br />

Inhibition <strong>of</strong> COX activity increases adiposity and energy<br />

efficiency in obesity resistant Sv129 mice<br />

Diet-induced thermogenesis protects several mouse strains<br />

against obesity [53–55]. Since it appears that the protection<br />

against diet-induced obesity is related to increased occurrence <strong>of</strong><br />

brown-like adipocytes in white depots [56;57], we aimed to<br />

investigate the hypothesis that indomethacin could also attenuate<br />

diet-induced UCP1 expression and thereby increase the propensity<br />

for diet-induced obesity in Sv129 mice. Since it was recently<br />

demonstrated that UCP1-deficient mice become obese when<br />

housed at thermoneutrality [58], we predicted that the most<br />

pronounced effect <strong>of</strong> COX inhibition would be observed for mice<br />

kept under thermoneutral conditions. Accordingly, we fed Sv129<br />

mice a very high-fat diet with or without indomethacin<br />

PLoS ONE | www.plosone.org 5 June 2010 | Volume 5 | Issue 6 | e11391


Cyclooxygenases and UCP1<br />

Figure 4. UCP1 expression is induced by forced expression <strong>of</strong> COX2 alone or in combination with COX1 in cultured cells. A. Rb 2/2<br />

MEFs were induced to differentiate as described in experimental procedures. Differentiated adipocytes were treated with vehicle or isoproterenol<br />

(100 nM) and 9-cis-retinoic acid (1 mM) in the absence and presence <strong>of</strong> the COX1 inhibitor SC560 (50 nM) or the COX2 inhibitor NS398 (5 mM), alone<br />

or in combination, or with indomethacin (1 mM), for 24 h. Expression <strong>of</strong> UCP1 was measured by RT-qPCR. <strong>The</strong> bars represent mean 6 standard error.<br />

<strong>The</strong> experiment was performed in triplicates and repeated 2 times. B–D. Rb 2/2 MEFs were retrovirally transduced with empty vector, vector<br />

encoding COX1 or COX2, or both. <strong>The</strong> transduced cells were selected and induced to differentiate and analyzed for COX1 and COX2 expression by<br />

Western blotting (B). PGE 2 levels were measured in cell media (C). RNA was isolated on day 8 and expressions <strong>of</strong> genes were measured by RT-qPCR<br />

(D). <strong>The</strong> bars represent mean 6 standard error. Different letters indicate statistically significant difference (p,0.05). <strong>The</strong> experiments were performed<br />

in triplicates.<br />

doi:10.1371/journal.pone.0011391.g004<br />

PLoS ONE | www.plosone.org 6 June 2010 | Volume 5 | Issue 6 | e11391


Cyclooxygenases and UCP1<br />

Figure 5. Cold-induced UCP1 expression is attenuated in iWAT in COX2 KO mice. A B COX2 KO mice and wild-type littermates were warmacclimated<br />

at 28–30uC for 10 days and then transferred to 4–6uC for 48 h. Rectal temperature was measured before transfer and after 24 and 48 h (A).<br />

RNA was extracted and expressions <strong>of</strong> genes were measured by RT-qPCR (B). <strong>The</strong> bars represent mean 6 standard error (n = 5–6). * indicates<br />

statistical significant difference between wild-type and KO mice (p,0.05).<br />

doi:10.1371/journal.pone.0011391.g005<br />

supplementation for 4 weeks while keeping the mice at 28–30uC.<br />

As demonstrated in Figure 7A, indomethacin supplementation led<br />

to a higher weight gain. Energy intake was slightly, but not<br />

statistically significantly lower (data not shown). However, energy<br />

intake relative to body weight gain was significantly lower in mice<br />

that received the indomethacin-supplemented high-fat diet<br />

(Figure 7A). Mice fed the diet supplemented with indomethacin<br />

also had more WAT in different depots, but not iBAT (Figure 7B).<br />

Histological analysis revealed that the adipocytes in both iWAT<br />

and iBAT appe<strong>are</strong>d normal, but adipocytes in iWAT in mice fed<br />

the high-fat diet supplemented with indomethacin were slightly<br />

larger (Figure 7C and D). As expected, feeding mice a high-fat diet<br />

lead to augmented expression <strong>of</strong> UCP1 in both iBAT and iWAT<br />

in vehicle-treated mice (Figure 7E). Importantly, diet-induced<br />

UCP1 expression was prevented in iWAT, but not in iBAT in the<br />

indomethacin-treated mice (Figure 7E). Reduced expression <strong>of</strong><br />

UCP1 in iWAT in mice fed a high-fat diet supplemented with<br />

indomethacin was accompanied with reduced expression <strong>of</strong><br />

Cox8b. Expression <strong>of</strong> PGC1a, Dio2, Eva1, Cidea, 4E-BP1 and<br />

RIP140 was not affected by inclusion <strong>of</strong> indomethacin with the<br />

high-fat diet (Figure 7E). Collectively, these results underscore the<br />

notion that inhibition <strong>of</strong> COX activity attenuates the acquisition <strong>of</strong><br />

‘‘brite’’ adipocytes in white adipose depots with an accompanying<br />

increase in feed efficiency leading to accumulation <strong>of</strong> more adipose<br />

tissue. Obviously, other mechanisms may contribute to the<br />

increase in feed efficiency, but the lack <strong>of</strong> ‘‘brite’’ adipocyte<br />

recruitment seems a key player.<br />

Discussion<br />

<strong>The</strong> unique energy-dissipating ability <strong>of</strong> UCP1 makes control <strong>of</strong><br />

its expression and activation potential targets for the development<br />

<strong>of</strong> novel drugs for the treatment <strong>of</strong> obesity and obesity-associated<br />

diseases. Here we present evidence that COX activity and COXderived<br />

PGE 2 <strong>are</strong> intimately linked to induction <strong>of</strong> UCP1<br />

expression in iWAT, but not in iBAT. Thus, cold-induced<br />

expression <strong>of</strong> UCP1 in iWAT was repressed in mice treated with<br />

the general COX inhibitor indomethacin, and in COX2 KO<br />

PLoS ONE | www.plosone.org 7 June 2010 | Volume 5 | Issue 6 | e11391


Cyclooxygenases and UCP1<br />

PLoS ONE | www.plosone.org 8 June 2010 | Volume 5 | Issue 6 | e11391


Cyclooxygenases and UCP1<br />

Figure 6. UCP1 expression is attenuated by an EP 4 receptor antagonist in Rb 2/2 adipocytes and induced by the EP4 receptor<br />

agonist 16,16dmPGE 2 in vivo. A.Expressions <strong>of</strong> EP 1 ,EP 2 ,EP 3 ,EP 4 and FP receptors were measured by RT-qPCR in iBAT and iWAT isolated from<br />

warm- and cold-acclimated mice, and in Rb 2/2 adipocytes treated with vehicle or isoproterenol (100 nM) and 9-cis-retinoic acid (1 mM). B. Rb 2/2<br />

adipocytes were treated with vehicle or isoproterenol (100 nM) and 9-cis-retinoic acid (1 mM) by RT-qPCR in absence and presence <strong>of</strong> AL8810,<br />

AH6809, or AH23848, which <strong>are</strong> FP, EP 1 /EP 2 and EP 4 receptor antagonists, respectively. Expressions <strong>of</strong> genes were measured by RT-qPCR. <strong>The</strong> bars<br />

represent mean 6 standard error. Different letters indicate statistically significant differences (p,0.05). C. C57BL/6J mice were subcutaneously<br />

injected with vehicle or 16,16dmPGE 2 (50 mM/kg) every 12 h for 48 h. Expressions <strong>of</strong> genes were measured by RT-qPCR. <strong>The</strong> bars represent mean 6<br />

standard error (n = 5). * indicates statistical significant difference between vehicle and 16,16 dmPGE 2 treated mice (p,0.05).<br />

doi:10.1371/journal.pone.0011391.g006<br />

mice. Also, injection <strong>of</strong> a stable analog <strong>of</strong> the COX2 downstream<br />

product PGE 2 , 16,16-dimethyl-PGE 2 , induced UCP1 expression<br />

in iWAT. Forced expression <strong>of</strong> COX2, alone or in combination<br />

with COX1, induced UCP1 expression in a cell model resembling<br />

inguinal adipocytes. Finally, the inhibition <strong>of</strong> COX activity not<br />

only attenuated diet-induced UCP1 expression in iWAT, but also<br />

increased weight gain in Sv129 mice kept at thermoneutrality.<br />

<strong>The</strong> association between diet-induced thermogenesis and the<br />

recruitment <strong>of</strong> brown adipose tissue was first noted more than 30<br />

years ago and believed to involve the classical brown adipose tissue<br />

located in the interscapular region [59]. <strong>The</strong> anti-obesity role <strong>of</strong><br />

UCP1 was challenged by the finding that UCP1 KO mice were<br />

not obese [60]. However, the recent demonstration that UCP1<br />

ablation per se induced obesity when the mice were kept at<br />

thermoneutrality [61] clearly indicates that UCP1 is important in<br />

diet-induced energy dissipation at thermoneutrality. Our data<br />

indicate that inhibition <strong>of</strong> COX activity increased weight gain and<br />

concomitantly attenuated diet-induced UCP1 expression in<br />

iWAT, but not iBAT in Sv129 mice kept at thermoneutrality,<br />

pointing to a novel role <strong>of</strong> COX activity in the control <strong>of</strong> energy<br />

balance and the development <strong>of</strong> obesity. <strong>The</strong>se results <strong>are</strong> in line<br />

with our earlier observation that enhanced cAMP signaling in<br />

response to an increased glucagon/insulin ratio led to an increased<br />

COX-mediated PGE 2 production. This was accompanied by<br />

increased expression <strong>of</strong> UCP1 in iWAT, but not in iBAT, and<br />

decreased feed efficiency [62]. Although neither COX1 KO nor<br />

COX2 KO mice <strong>are</strong> obese, COX2 +/2 KO mice have more<br />

adipose tissue than wild-type littermates when fed an <strong>obesogenic</strong><br />

diet [48]. <strong>The</strong> reason why COX2 +/2 , but not COX2 KO mice<br />

were reported to be prone to obesity is not clear. However, these<br />

studies were not performed at thermoneutrality [48]. A similar<br />

phenomenon is actually seen in GLUT4 KO mice, where the<br />

majority <strong>of</strong> GLUT4 2/+ , but not GLUT4 2/2 mice develops<br />

diabetes [63]. Moreover, release <strong>of</strong> PGE 2 from adipose tissue in<br />

COX2 +/2 mice was reported to be reduced comp<strong>are</strong>d to adipose<br />

tissue from wild-type mice [48] and adipose tissue cultures<br />

obtained from obese rats have lower PGE 2 release rates than<br />

cultures from lean rats [38]. In addition, microsomal prostaglandin<br />

α<br />

Figure 7. Indomethacin prevents high-fat diet-induced UCP1 expression in iWAT but not iBAT in the obesity-resistant Sv129 mouse<br />

strain. Mice were fed a very high-fat diet (VHF) with or without indomethacin supplementation (16 ppm) for 4 weeks at a temperature <strong>of</strong> 28–30uC.<br />

One group <strong>of</strong> mice was killed before the experiment started. A. Body weight gain and energy intake relative to body weight gain. B. Different<br />

adipose tissue depots were dissected and weighed. C and D. Representative paraffin-embedded representative sections from iWAT and iBAT were<br />

stained with hematoxylin and eosin. <strong>The</strong> scale bars represent 50 mM. E. Expressions <strong>of</strong> genes in iWAT and iBAT were measured by RT-qPCR. <strong>The</strong> bars<br />

represent mean 6 standard error (n = 6). * indicates statistical significant difference (p,0.05) between different groups.<br />

doi:10.1371/journal.pone.0011391.g007<br />

PLoS ONE | www.plosone.org 9 June 2010 | Volume 5 | Issue 6 | e11391


Cyclooxygenases and UCP1<br />

E synthase1 (mPGES1) expression is reported to be downregulated<br />

in iWAT and eWAT in obese mice [64], suggesting a<br />

dysregulation <strong>of</strong> prostaglandin synthesis in obesity.<br />

PGE 2 mediates its action by interacting with four subtypes <strong>of</strong><br />

PGE 2 receptors, the EP 1 ,EP 2 ,EP 3 and EP 4 receptors [50]. Using<br />

the Rb 2/2 brown-like adipocytes, we show that b-adrenergic<br />

stimulation <strong>of</strong> UCP1 expression is attenuated by an EP 4 receptor<br />

antagonist. This combined with our finding that injection <strong>of</strong> the<br />

EP 3 /EP 4 receptor agonist 16,16-dimethyl-PGE 2 increased expression<br />

<strong>of</strong> UCP1 in iWAT indicates that the action <strong>of</strong> PGE 2 is<br />

predominantly mediated via the EP 4 receptor with a possible minor<br />

contribution by the EP 3 receptor. Most EP 4 KO mice die shortly<br />

after birth, and no adipose tissue phenotype has been reported for<br />

the few surviving pups [65]. Similarly, to our knowledge, no adipose<br />

tissue phenotype has been reported for the EP 3 KO mice.<br />

Collectively, our findings strongly indicate that both cold- and<br />

diet-induced expression <strong>of</strong> UCP1 in iWAT, but not in iBAT,<br />

requires COX activity and most likely PGE 2 formation.<br />

Furthermore, our results point to differential roles <strong>of</strong> induction<br />

<strong>of</strong> UCP1 expression in iWAT and iBAT in the context <strong>of</strong> diet- and<br />

cold-induced thermogenesis. We suggest that whereas UCP1 in<br />

iWAT plays an important role in protection against obesity, UCP1<br />

in iBAT is essential for temperature adaptation. Upon cold<br />

challenge, body temperatures were only slightly lower in wild-type<br />

mice treated with indomethacin and in COX2 KO mice<br />

comp<strong>are</strong>d to non-treated wild-type mice. This is in line with the<br />

earlier finding that UCP1 expression is blunted in iWAT, but not<br />

in iBAT, in cold-adapted b 3 adrenoreceptor KO mice [66].<br />

<strong>The</strong> importance <strong>of</strong> COX in diet-induced expression <strong>of</strong> UCP1 in<br />

iWAT and diet-induced thermogenesis is underscored by our<br />

demonstration that inclusion <strong>of</strong> the general COX inhibitor<br />

indomethacin in the diet augmented high-fat diet-induced obesity<br />

in Sv129 mice kept at thermoneutrality irrespectively <strong>of</strong> UCP1<br />

expression in iBAT. Increased expression <strong>of</strong> UCP1 in WAT with<br />

accompanying increased thermogenic activity coupled with unchanged<br />

or even reduced BAT activity has been observed in several<br />

genetically modified lean mouse models such as RIP140 [67],<br />

Caveolin 1 [68], Fsp27 [69], hormone sensitive lipase [70] and<br />

vitamin D receptor [71] KO mice. Further, in RIIb mice [72], pRbdeficient<br />

mice [11;73] and in mice overexpressing FOXC2 [74],<br />

protection against diet-induced obesity is accompanied by an<br />

increased RIa/RIIb ratio, rendering PKA somewhat more sensitive<br />

to cAMP, which is accompanied by an increased occurrence <strong>of</strong><br />

brown adipocytes in WAT. Last, it should be recalled that in aP2-<br />

UCP1 transgenic mice, both endogenous UCP1 expression and<br />

respiration <strong>are</strong> actually reduced in iBAT [75]. UCP1 expression,<br />

respiration and total oxidative capacity <strong>are</strong>, however, strongly<br />

induced in WAT and the oxidative capacity <strong>of</strong> WAT is sufficient for<br />

the changes <strong>of</strong> total energy balance induced by the transgene [76].<br />

In keeping with the earlier notion that i) mouse strains that have<br />

more UCP1-expressing adipocytes in their WAT depots <strong>are</strong><br />

protected against diet-induced obesity [77;78] and ii) brown-like<br />

multilocular adipocytes expressing UCP1 <strong>are</strong> detected interspersed<br />

within white adipose tissue in humans [20;21;79], we suggest that<br />

factors influencing UCP1 expression in white adipose tissue <strong>are</strong> <strong>of</strong><br />

particular importance for the regulation <strong>of</strong> energy balance and the<br />

development <strong>of</strong> obesity also in humans.<br />

Materials and Methods<br />

Ethics Statement<br />

<strong>The</strong> animal experiments were approved by the Norwegian<br />

Animal Health Authorities, ID 819 and 888. C<strong>are</strong> and handling<br />

were in accordance with local institutional recommendations.<br />

Cell culture, transduction and differentiation<br />

Mouse embryo fibroblasts (MEFs) were prep<strong>are</strong>d from wild-type<br />

and Rb 2/2 embryos [80]. <strong>The</strong> cells were grown and differentiated<br />

in AmnioMax Medium as described earlier [81]. Retrovira<br />

expressing pLXSN-hygro, pBabe-puro, pLXSN-COX1 or<br />

pBabe-COX2 were harvested from Phoenix–Eco cells, plated at<br />

30–40% confluence in DMEM supplemented with 10% fetal<br />

bovine serum, and transductions performed as described [82].<br />

Isolation <strong>of</strong> the stromal vascular fraction and adipocytes<br />

from mice<br />

<strong>The</strong> stromal vascular fraction and adipocytes were obtained<br />

from iWAT and iBAT dissected from 8-week old C57BL/6J mice<br />

as earlier described [83]. Contaminating erythrocytes were<br />

eliminated from the stromal-vascular fraction by a wash with<br />

sterile distilled water. Cells were plated and induced to<br />

differentiate as described [83].<br />

Cold acclimation experiments<br />

Groups (n = 5–8) <strong>of</strong> 10-week old male mice were acclimated at a<br />

temperature <strong>of</strong> 28–30uC for at least 1 week and transferred to 4uC<br />

for 1, 2, 3 or 6 days. Where relevant, mice were injected with<br />

indomethacin (2.5 mg/kg) 2 h prior transfer to 4uC. <strong>The</strong> mice<br />

received a dose <strong>of</strong> indomethacin every 12 h. Injections were<br />

performed subcutaneously from a 0.75 mg/ml solution. Final dose<br />

was 5 mg/kg/day. Control mice received vehicle. Animals were<br />

housed individually with a 12 h light/dark cycle and free access to<br />

pellet food and water. Mice used for immunohistochemical<br />

analyses were immediately perfused intracardially with 4%<br />

paraformaldehyde. iBAT, iWAT, lung and skin were dissected<br />

and frozen for immunohistochemistry on cryosections. For<br />

morphology experiments, the mice were immediately perfused<br />

with 4% paraformaldehyde in 0.1 M phosphate buffer, pH 7.4 for<br />

5 min. COX2 KO mice (B6;129P2 Ptgs2 tm1Unc) and corresponding<br />

wild-type littermates were obtained from Taconic. C57BL/6J<br />

used in indomethacin experiments were obtained from Møllegård<br />

breeding laboratories.<br />

16,16dmPGE 2 injections<br />

Male, C57BL/6J approx 10-weeks old from Møllegård<br />

breeding laboratories, Denmark were divided into two groups<br />

(n = 5). <strong>The</strong> mice received a dose <strong>of</strong> 50 mg/kg 16,16dmPGE 2 every<br />

12 h for 48 h. Injections were performed subcutaneously and the<br />

total dose was 0.1 mg/kg/day. Control mice received vehicle.<br />

Animals were housed individually with a 12 h light/dark cycle and<br />

free access to pellet food and water.<br />

High-fat feeding<br />

Male Sv129 mice, 11 weeks old, were obtained from Taconic.<br />

<strong>The</strong> mice were acclimated for 1 week at a temperature <strong>of</strong> 28–30uC<br />

and divided into three groups (n = 6 in each). One group <strong>of</strong> mice<br />

was sacrificed before dietary intervention while the remaining<br />

mice were fed a very high-fat diet (ssniff EF R/M acc D12492)<br />

with or without indomethacin supplementation (16 ppm) for 4<br />

weeks at a temperature <strong>of</strong> 28–30uC. Body weight and feed intake<br />

were recorded twice a week. Mice were anesthetized using<br />

is<strong>of</strong>lurane, cardiac puncture was performed and mice were killed<br />

by cervical dissociation. Tissues were immediately frozen in liquid<br />

N 2 .<br />

Real time qPCR<br />

Total RNA was extracted from cultured cells or mouse tissue<br />

using TRIzol (Invitrogen). Reverse transcription and qPCR were<br />

PLoS ONE | www.plosone.org 10 June 2010 | Volume 5 | Issue 6 | e11391


Cyclooxygenases and UCP1<br />

performed in duplicates as described earlier [83]. Primer<br />

sequences <strong>are</strong> available on request.<br />

Western blotting<br />

Preparation <strong>of</strong> extracts from mouse tissues or whole cell dishes,<br />

electrophoresis, blotting, visualization and stripping <strong>of</strong> membranes<br />

were performed as described [84]. Primary antibodies used were<br />

goat anti-COX1, goat anti-COX2, rabbit anti-UCP1 and rabbit<br />

anti-TFIIB antibodies (Santa Cruz Biotechnology). Secondary<br />

antibodies were horseradish peroxidase-conjugated anti-mouse,<br />

anti-goat or anti-rabbit antibodies obtained from DAKO.<br />

Immunohistochemistry<br />

COX1 (M-20; sc 1754) and COX2 (M-19; sc 1747) antibodies<br />

were obtained from Santa Cruz Biotechnology, diluted 1:300 on<br />

cryosections and 1:100 on paraffin-embedded sections (for<br />

COX1). Lung [85] and skin [86] were used as positive control<br />

for both COX1 and COX2 antibodies.<br />

Histological analyses<br />

Parts <strong>of</strong> adipose tissue were fixed in 4% formaldehyde in PB<br />

buffer for 24 h, washed in PB, dehydrated in ethanol, embedded<br />

in paraffin after 2610 min xylen treatment. Sections (8 mm thick)<br />

<strong>of</strong> the embedded tissue sections were subjected to standard<br />

hematoxylin and eosin staining.<br />

Supporting Information<br />

Text S1<br />

Experimental.<br />

Found at: doi:10.1371/journal.pone.0011391.s001 (0.04 MB<br />

DOC)<br />

Figure S1 COX1 and COX2 <strong>are</strong> mainly expressed in the<br />

stromal-vascular fraction <strong>of</strong> iBAT in warm-acclimated mice. A.<br />

Sv129 mice were warm-acclimated at 28–30uC for 6 days and<br />

then transferred to 4–6uC. Samples for cryosections were prep<strong>are</strong>d<br />

from iBAT and iWAT after four days <strong>of</strong> cold exposure.<br />

Representative COX2 immunoblots in iBAT B. iBAT from<br />

warm-acclimated mice was fractionated into SVF and adipocyte<br />

fractions, respectively. Expression levels <strong>of</strong> COX1 and COX2<br />

were determined by RT-qPCR and Western blotting.<br />

Found at: doi:10.1371/journal.pone.0011391.s002 (15.01 MB<br />

EPS)<br />

Figure S2 Inhibition <strong>of</strong> COX does not prevent isoproterenol/9-<br />

cis-retinoic acid-induced UCP1 expression in WT-1 cells.<br />

Differentiated WT-1 cells were treated with a combination <strong>of</strong><br />

isoproterenol (100 nM) and 9-cis-retinoic acid (1 mM) for 24 h.<br />

When included, indomethacin (1 mM) were added 2 h before<br />

isoproterenol and 9-cis retinoic acid. UCP1 and PGE1a<br />

expressions were determined by RT-qPCR.<br />

Found at: doi:10.1371/journal.pone.0011391.s003 (2.44 MB EPS)<br />

Author Contributions<br />

Conceived and designed the experiments: LM KK. Performed the<br />

experiments: LM LMP HHL EF IB QH RKP PH TM RDM PA JM<br />

MLB JBH BC JN JW SC PV SOD. Analyzed the data: LM LMP HHL EF<br />

RKP PH TM PA JM MLB JBH BC JN JW SC PV SOD KK. Wrote the<br />

paper: LM LMP BC JN SOD KK.<br />

References<br />

1. Cannon B, Nedergaard J (2004) Brown Adipose Tissue: Function and<br />

Physiological Significance. Physiol Rev 84: 277–359.<br />

2. Kopecky J, Hodny Z, Rossmeisl M, Syrovy I, Kozak LP (1996) Reduction <strong>of</strong><br />

dietary obesity in aP2-Ucp transgenic mice: physiology and adipose tissue<br />

distribution. Am J Physiol 270: E768–E775.<br />

3. Kopecky J, Rossmeisl M, Hodny Z, Syrovy I, Horakova M, et al. (1996)<br />

Reduction <strong>of</strong> dietary obesity in aP2-Ucp transgenic mice: mechanism and<br />

adipose tissue morphology. Am J Physiol Endocrinol Metab 270: E776–E786.<br />

4. Cederberg A, Gronning LM, Ahren B, Tasken K, Carlsson P, et al. (2001)<br />

FOXC2 Is a Winged Helix Gene that Counteracts Obesity, Hypertriglyceridemia,<br />

and Diet-Induced Insulin Resistance. Cell 106: 563–573.<br />

5. Seale P, Kajimura S, Yang W, Chin S, Rohas LM, et al. (2007) Transcriptional<br />

control <strong>of</strong> brown fat determination by PRDM16. Cell Metab 6: 38–54.<br />

6. Cummings DE, Brandon EP, Planas JV, Motamed K, Idzerda RL, et al. (1996)<br />

Genetically lean mice result from targeted disruption <strong>of</strong> the RII[beta] subunit <strong>of</strong><br />

protein kinase A. Nature 382: 622–626.<br />

7. Nolan MA, Sikorski MA, McKnight GS (2004) <strong>The</strong> role <strong>of</strong> uncoupling protein 1<br />

in the metabolism and adiposity <strong>of</strong> RII beta-protein kinase A-deficient mice. Mol<br />

Endocrinol 18: 2302–2311.<br />

8. Tsukiyama-Kohara K, Poulin F, Kohara M, DeMaria CT, Cheng A, et al.<br />

(2001) Adipose tissue reduction in mice lacking the translational inhibitor 4E-<br />

BP1. Nat Med 7: 1128–1132.<br />

9. Zhou Z, Yon Toh S, Chen Z, Guo K, Peng Ng C, et al. (2003) Cidea-deficient<br />

mice have lean phenotype and <strong>are</strong> resistant to obesity. Nat Genet 35: 49–56.<br />

10. Picard F, Gehin M, Annicotte JS, Rocchi S, Champy MF, et al. (2002) SRC-1<br />

and TIF2 Control Energy Balance between White and Brown Adipose Tissues.<br />

Cell 111: 931–941.<br />

11. Dali-Youcef N, Mataki C, Coste A, Messaddeq N, Giroud S, et al. (2007)<br />

Adipose tissue-specific inactivation <strong>of</strong> the retinoblastoma protein protects against<br />

diabesity because <strong>of</strong> increased energy expenditure. Proc Natl Acad Sci U S A<br />

104: 10703–10708.<br />

12. Mercader J, Ribot J, Murano I, Feddersen S, Cinti S, et al. (2009)<br />

Haploinsufficiency <strong>of</strong> the retinoblastoma protein gene reduces diet-induced<br />

obesity, insulin resistance and hepatosteatosis in mice. Am J Physiol 297:<br />

E184–E193.<br />

13. Hansen JB, Jorgensen C, Petersen RK, Hallenborg P, De Matteis R, et al. (2004)<br />

Retinoblastoma protein functions as a molecular switch determining white<br />

versus brown adipocyte differentiation. Proc Natl Acad Sci U S A 101:<br />

4112–4117.<br />

14. Cannon B, Nedergaard J (2004) Brown Adipose Tissue: Function and<br />

Physiological Significance. Physiol Rev 84: 277–359.<br />

15. Cypess AM, Lehman S, Williams G, Tal I, Rodman D, et al. (2009)<br />

Identification and Importance <strong>of</strong> Brown Adipose Tissue in Adult Humans.<br />

N Engl J Med 360: 1509–1517.<br />

16. van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, Drossaerts JMAF,<br />

Kemerink GJ, et al. (2009) Cold-Activated Brown Adipose Tissue in Healthy Men.<br />

N Engl J Med 360: 1500–1508.<br />

17. Virtanen KA, Lidell ME, Orava J, Heglind M, Westergren R, et al. (2009)<br />

Functional Brown Adipose Tissue in Healthy Adults. N Engl J Med 360:<br />

1518–1525.<br />

18. Zing<strong>are</strong>tti MC, Crosta F, Vitali A, Guerrieri M, Frontini A, et al. (2009) <strong>The</strong><br />

presence <strong>of</strong> UCP1 demonstrates that metabolically active adipose tissue in the<br />

neck <strong>of</strong> adult humans truly represents brown adipose tissue. FASEB J 23:<br />

3113–3120.<br />

19. Saito M, Okamatsu-Ogura Y, Matsushita M, Watanabe K, Yoneshiro T, et al.<br />

(2009) High incidence <strong>of</strong> metabolically active brown adipose tissue in healthy<br />

adult humans: <strong>effects</strong> <strong>of</strong> cold exposure and adiposity. Diabetes 58: 1526–<br />

1531.<br />

20. Garruti G, Ricquier D (1992) Analysis <strong>of</strong> uncoupling protein and its mRNA in<br />

adipose tissue deposits <strong>of</strong> adult humans. Int J Obes Relat Metab Disord 16:<br />

383–390.<br />

21. Lean ME, James WP, Jennings G, Trayhurn P (1986) Brown adipose tissue in<br />

patients with phaeochromocytoma. Int J Obes 10: 219–227.<br />

22. Oberk<strong>of</strong>ler H, Dallinger G, Liu YM, Hell E, Krempler F, et al. (1997)<br />

Uncoupling protein gene: quantification <strong>of</strong> expression levels in adipose tissues <strong>of</strong><br />

obese and non-obese humans. J Lipid Res 38: 2125–2133.<br />

23. Oberk<strong>of</strong>ler H, Dallinger G, Liu YM, Hell E, Krempler F, et al. (1997)<br />

Uncoupling protein gene: quantification <strong>of</strong> expression levels in adipose tissues <strong>of</strong><br />

obese and non-obese humans. J Lipid Res 38: 2125–2133.<br />

24. Timmons JA, Wennmalm K, Larsson O, Walden TB, Lassmann T, et al. (2007)<br />

Myogenic gene expression signature establishes that brown and white adipocytes<br />

originate from distinct cell lineages. PNAS 104: 4401–4406.<br />

25. Seale P, Bjork B, Yang W, Kajimura S, Chin S, et al. (2008) PRDM16 controls a<br />

brown fat/skeletal muscle switch. Nature 454: 961–967.<br />

26. Himms-Hagen J, Melnyk A, Zing<strong>are</strong>tti MC, Ceresi E, Barbatelli G, et al. (2000)<br />

Multilocular fat cells in WAT <strong>of</strong> CL-316243-treated rats derive directly from<br />

white adipocytes. Am J Physiol Cell Physiol 279: C670–C681.<br />

27. Granneman JG, Burnazi M, Zhu Z, Schwamb LA (2003) White adipose tissue<br />

contributes to UCP1-in<strong>dependent</strong> thermogenesis. Am J Physiol Endocrinol<br />

Metab 285: E1230–E1236.<br />

28. Cinti S (2009) Transdifferentiation properties <strong>of</strong> adipocytes in the Adipose<br />

Organ. Am J Physiol Endocrinol Metab: 00183.<br />

PLoS ONE | www.plosone.org 11 June 2010 | Volume 5 | Issue 6 | e11391


Cyclooxygenases and UCP1<br />

29. Granneman JG, Li P, Zhu Z, Lu Y (2005) Metabolic and cellular plasticity in<br />

white adipose tissue I: <strong>effects</strong> <strong>of</strong> beta3-adrenergic receptor activation.<br />

Am J Physiol Endocrinol Metab 289: E608–E616.<br />

30. Petrovic N, Walden TB, Shabalina IG, Timmons JA, Cannon B, et al. (2009)<br />

Chronic PPARc Activation <strong>of</strong> Epididymally Derived White Adipocyte Cultures<br />

Reveals a Population <strong>of</strong> <strong>The</strong>rmogenically Competent, UCP1-containing<br />

Adipocytes Molecularly Distinct From Classical Brown Adipocytes. J Biol Chem<br />

Dec 22. [Epub ahead <strong>of</strong> print].<br />

31. Guerra C, Koza RA, Yamashita H, Walsh K, Kozak LP (1998) Emergence <strong>of</strong><br />

Brown Adipocytes in White Fat in Mice Is Under Genetic Control. Effects on<br />

Body Weight and Adiposity. J Clin Invest 102: 412–420.<br />

32. Watson PM, Commins SP, Beiler RJ, Hatcher HC, Gettys TW (2000)<br />

Differential regulation <strong>of</strong> leptin expression and function in A/J vs. C57BL/6J<br />

mice during diet-induced obesity. Am J Physiol Endocrinol Metab 279:<br />

E356–E365.<br />

33. Kopecky J, Rossmeisl M, Hodny Z, Syrovy I, Horakova M, et al. (1996)<br />

Reduction <strong>of</strong> dietary obesity in aP2-Ucp transgenic mice: mechanism and<br />

adipose tissue morphology. Am J Physiol Endocrinol Metab 270: E776–E786.<br />

34. Oberk<strong>of</strong>ler H, Dallinger G, Liu YM, Hell E, Krempler F, et al. (1997)<br />

Uncoupling protein gene: quantification <strong>of</strong> expression levels in adipose tissues <strong>of</strong><br />

obese and non-obese humans. J Lipid Res 38: 2125–2133.<br />

35. Semple RK, Crowley VC, Sewter CP, Laudes M, Christodoulides C, et al.<br />

(2003) Expression <strong>of</strong> the thermogenic nuclear hormone receptor coactivator<br />

PGC-1[alpha] is reduced in the adipose tissue <strong>of</strong> morbidly obese subjects.<br />

Int J Obes Relat Metab Disord 28: 176–179.<br />

36. Jimenez M, Barbatelli G, Allevi R, Cinti S, Seydoux J, et al. (2003) b 3 -<br />

Adrenoceptor knockout in C57BL/6J mice depresses the occurrence <strong>of</strong> brown<br />

adipocytes in white. European Journal <strong>of</strong> Biochemistry 270: 699–705.<br />

37. Grujic D, Susulic VS, Harper ME, Himms-Hagen J, Cunningham BA, et al.<br />

(1997) beta 3-Adrenergic Receptors on White and Brown Adipocytes Mediate<br />

beta 3-Selective Agonist-induced Effects on Energy Expenditure, Insulin<br />

Secretion, and Food Intake. J Biol Chem 272: 17686–17693.<br />

38. Gaskins HD, Hausman DB, Martin RJ, Hausman GJ (1989) Evidence for<br />

abnormal prostaglandin synthesis in obese Zucker rat adipose cell cultures. J Nutr<br />

119: 458–462.<br />

39. Madsen L, Pedersen LM, Liaset B, Ma T, Petersen RK, et al. (2008) cAMP<strong>dependent</strong><br />

Signaling Regulates the Adipogenic Effect <strong>of</strong> n-6 Polyunsaturated<br />

Fatty Acids. J Biol Chem 283: 7196–7205.<br />

40. Petersen RK, Jorgensen C, Rustan AC, Froyland L, Muller-Decker K, et al.<br />

(2003) Arachidonic acid-<strong>dependent</strong> inhibition <strong>of</strong> adipocyte differentiation<br />

requires PKA activity and is associated with sustained expression <strong>of</strong><br />

cyclooxygenases. J Lipid Res 44: 2320–2330.<br />

41. Vassaux G, Gaillard D, Darimont C, Ailhaud G, Negrel R (1992) Differential<br />

response <strong>of</strong> preadipocytes and adipocytes to prostacyclin and prostaglandin E2:<br />

physiological implications. Endocrinology 131: 2393–2398.<br />

42. Cinti S (2009) Transdifferentiation properties <strong>of</strong> adipocytes in the Adipose<br />

Organ. Am J Physiol Endocrinol Metab: 00183.<br />

43. Hansen JB, Jorgensen C, Petersen RK, Hallenborg P, De Matteis R, et al. (2004)<br />

Retinoblastoma protein functions as a molecular switch determining white<br />

versus brown adipocyte differentiation. Proc Natl Acad Sci U S A 101:<br />

4112–4117.<br />

44. Hansen JB, Jorgensen C, Petersen RK, Hallenborg P, De Matteis R, et al. (2004)<br />

Retinoblastoma protein functions as a molecular switch determining white<br />

versus brown adipocyte differentiation. Proc Natl Acad Sci U S A 101:<br />

4112–4117.<br />

45. Hyman BT, Stoll LL, Spector AA (1982) Prostaglandin production by 3T3-L1<br />

cells in culture. Biochim Biophys Acta 713: 375–385.<br />

46. Madsen L, Pedersen LM, Liaset B, Ma T, Petersen RK, et al. (2008) cAMP<strong>dependent</strong><br />

Signaling Regulates the Adipogenic Effect <strong>of</strong> n-6 Polyunsaturated<br />

Fatty Acids. J Biol Chem 283: 7196–7205.<br />

47. Tseng YH, Kokkotou E, Schulz TJ, Huang TL, Winnay JN, et al. (2008) New<br />

role <strong>of</strong> bone morphogenetic protein 7 in brown adipogenesis and energy<br />

expenditure. Nature 454: 1000–1004.<br />

48. Fain JN, Ballou LR, Bahouth SW (2001) Obesity is induced in mice<br />

heterozygous for cyclooxygenase-2. Prostaglandins Other Lipid Mediat 65:<br />

199–209.<br />

49. Morham SG, Langenbach R, L<strong>of</strong>tin CD, Tiano HF, Vouloumanos N, et al.<br />

(1995) Prostaglandin synthase 2 gene disruption causes severe renal pathology in<br />

the mouse. Cell 83: 473–482.<br />

50. Ushikubi F, Hirata M, Narumiya S (1995) Molecular biology <strong>of</strong> prostanoid<br />

receptors: an overview. J Lipin Mediat 12: 343–359.<br />

51. Abramovitz M, Adam M, Boie Y, Carrière MC, Denis D, et al. (2000) <strong>The</strong><br />

utilization <strong>of</strong> recombinant prostanoid receptors to determine the affinities and<br />

selectivities <strong>of</strong> prostaglandins and related analogs. Biochimica et Biophysica Acta<br />

1483: 285–293.<br />

52. Kunikata T, Tanaka A, Miyazawa T, Kato S, Takeuchi K (2002) 16,16-<br />

Dimethyl Prostaglandin E2 Inhibits Indomethacin-Induced Small Intestinal<br />

Lesions Through EP3 and EP4 Receptors. Digestive Diseases and Sciences 47:<br />

894–904.<br />

53. Koza RA, Hohmann SM, Guerra C, Rossmeisl M, Kozak LP (2000) Synergistic<br />

Gene Interactions Control the Induction <strong>of</strong> the Mitochondrial Uncoupling<br />

Protein (Ucp1) Gene in White Fat Tissue. J Biol Chem 275: 34486–34492.<br />

54. Guerra C, Koza RA, Yamashita H, Walsh K, Kozak LP (1998) Emergence <strong>of</strong><br />

Brown Adipocytes in White Fat in Mice Is Under Genetic Control. Effects on<br />

Body Weight and Adiposity. J Clin Invest 102: 412–420.<br />

55. Collins S, Daniel KW, Petro AE, Surwit RS (1997) Strain-Specific Response to<br />

b3-Adrenergic Receptor Agonist Treatment <strong>of</strong> Diet-Induced Obesity in Mice.<br />

Endocrinology 138: 405–413.<br />

56. Guerra C, Koza RA, Yamashita H, Walsh K, Kozak LP (1998) Emergence <strong>of</strong><br />

Brown Adipocytes in White Fat in Mice Is Under Genetic Control. Effects on<br />

Body Weight and Adiposity. J Clin Invest 102: 412–420.<br />

57. Watson PM, Commins SP, Beiler RJ, Hatcher HC, Gettys TW (2000)<br />

Differential regulation <strong>of</strong> leptin expression and function in A/J vs. C57BL/6J<br />

mice during diet-induced obesity. Am J Physiol Endocrinol Metab 279:<br />

E356–E365.<br />

58. Feldmann HM, Golozoubova V, Cannon B, Nedergaard J (2009) UCP1<br />

Ablation Induces Obesity and Abolishes Diet-Induced <strong>The</strong>rmogenesis in Mice<br />

Exempt from <strong>The</strong>rmal Stress by Living at <strong>The</strong>rmoneutrality. Cell Metabolism 9:<br />

203–209.<br />

59. Rothwell NJ, Stock MJ (1979) A role for brown adipose tissue in diet-induced<br />

thermogenesis. Nature 281: 31–35.<br />

60. Enerbäck S, Jacobsson A, Simpson EM, Guerra C, Yamashita H, et al. (1997)<br />

Mice lacking mitochondrial uncoupling protein <strong>are</strong> cold-sensitive but not obese.<br />

Nature 387: 90–94.<br />

61. Feldmann HM, Golozoubova V, Cannon B, Nedergaard J (2009) UCP1<br />

Ablation Induces Obesity and Abolishes Diet-Induced <strong>The</strong>rmogenesis in Mice<br />

Exempt from <strong>The</strong>rmal Stress by Living at <strong>The</strong>rmoneutrality. Cell Metabolism 9:<br />

203–209.<br />

62. Madsen L, Pedersen LM, Liaset B, Ma T, Petersen RK, et al. (2008) cAMP<strong>dependent</strong><br />

Signaling Regulates the Adipogenic Effect <strong>of</strong> n-6 Polyunsaturated<br />

Fatty Acids. J Biol Chem 283: 7196–7205.<br />

63. Stenbit AE, Tsao TS, Li J, Burcelin R, Geenen DL, et al. (1997) GLUT4<br />

heterozygous knockout mice develop muscle insulin resistance and diabetes. Nat<br />

Med 3: 1096–1101.<br />

64. Hetu PO, Riendeau D (2007) Down-regulation <strong>of</strong> Microsomal Prostaglandin E2<br />

Synthase-1 in Adipose Tissue by High-fat Feeding. Obesity Res 15: 60–68.<br />

65. Sakuma Y, Tanaka K, Suda M, Komatsu Y, Yasoda A, et al. (2000) Impaired<br />

Bone Resorption by Lipopolysaccharide In Vivo in Mice Deficient in the<br />

Prostaglandin E Receptor EP4 Subtype. Infect Immun 68: 6819–6825.<br />

66. Jimenez M, Barbatelli G, Allevi R, Cinti S, Seydoux J, et al. (2003) b 3 -<br />

Adrenoceptor knockout in C57BL/6J mice depresses the occurrence <strong>of</strong> brown<br />

adipocytes in white. European Journal <strong>of</strong> Biochemistry 270: 699–705.<br />

67. Leonardsson G, Steel JH, Christian M, Pocock V, Milligan S, et al. (2004)<br />

Nuclear receptor corepressor RIP140 regulates fat accumulation. Proc Natl<br />

Acad Sci U S A 101: 8437–8442.<br />

68. Razani B, Combs TP, Wang XB, Frank PG, Park DS, et al. (2002) Caveolin-1-<br />

deficient mice <strong>are</strong> lean, resistant to diet-induced obesity, and show hypertriglyceridemia<br />

with adipocyte abnormalities. J Biol Chem 277: 8635–8647.<br />

69. Toh SY, Gong J, Du G, Li JZ, Yang S, et al. (2008) Up-Regulation <strong>of</strong><br />

Mitochondrial Activity and Acquirement <strong>of</strong> Brown Adipose Tissue-Like<br />

Property in the White Adipose Tissue <strong>of</strong> Fsp27 Deficient Mice. PLoS ONE 3:<br />

e2890.<br />

70. Ström K, Hansson O, Lucas S, Nevsten P, Fernandez C, et al. (2008)<br />

Attainment <strong>of</strong> Brown Adipocyte Features in White Adipocytes <strong>of</strong> Hormone-<br />

Sensitive Lipase Null Mice. PLoS ONE 3: e1793.<br />

71. Narvaez CJ, Matthews D, Broun E, Chan M, Welsh J (2009) Lean Phenotype<br />

and Resistance to Diet-Induced Obesity in Vitamin D Receptor Knockout Mice<br />

Correlates with Induction <strong>of</strong> Uncoupling Protein-1 in White Adipose Tissue.<br />

Endocrinology 150: 651–661.<br />

72. Cummings DE, Brandon EP, Planas JV, Motamed K, Idzerda RL, et al. (1996)<br />

Genetically lean mice result from targeted disruption <strong>of</strong> the RII[beta] subunit <strong>of</strong><br />

protein kinase A. Nature 382: 622–626.<br />

73. Hansen JB, Jorgensen C, Petersen RK, Hallenborg P, De Matteis R, et al. (2004)<br />

Retinoblastoma protein functions as a molecular switch determining white<br />

versus brown adipocyte differentiation. Proc Natl Acad Sci U S A 101:<br />

4112–4117.<br />

74. Cederberg A, Gronning LM, Ahren B, Tasken K, Carlsson P, et al. (2001)<br />

FOXC2 Is a Winged Helix Gene that Counteracts Obesity, Hypertriglyceridemia,<br />

and Diet-Induced Insulin Resistance. Cell 106: 563–573.<br />

75. Kopecky J, Rossmeisl M, Hodny Z, Syrovy I, Horakova M, et al. (1996)<br />

Reduction <strong>of</strong> dietary obesity in aP2-Ucp transgenic mice: mechanism and<br />

adipose tissue morphology. Am J Physiol Endocrinol Metab 270: E776–E786.<br />

76. Kopecky J, Rossmeisl M, Hodny Z, Syrovy I, Horakova M, et al. (1996)<br />

Reduction <strong>of</strong> dietary obesity in aP2-Ucp transgenic mice: mechanism and<br />

adipose tissue morphology. Am J Physiol Endocrinol Metab 270: E776–E786.<br />

77. Guerra C, Koza RA, Yamashita H, Walsh K, Kozak LP (1998) Emergence <strong>of</strong><br />

Brown Adipocytes in White Fat in Mice Is Under Genetic Control. Effects on<br />

Body Weight and Adiposity. J Clin Invest 102: 412–420.<br />

78. Watson PM, Commins SP, Beiler RJ, Hatcher HC, Gettys TW (2000)<br />

Differential regulation <strong>of</strong> leptin expression and function in A/J vs. C57BL/6J<br />

mice during diet-induced obesity. Am J Physiol Endocrinol Metab 279:<br />

E356–E365.<br />

79. Oberk<strong>of</strong>ler H, Dallinger G, Liu YM, Hell E, Krempler F, et al. (1997)<br />

Uncoupling protein gene: quantification <strong>of</strong> expression levels in adipose tissues <strong>of</strong><br />

obese and non-obese humans. J Lipid Res 38: 2125–2133.<br />

PLoS ONE | www.plosone.org 12 June 2010 | Volume 5 | Issue 6 | e11391


Cyclooxygenases and UCP1<br />

80. Hansen JB, Jorgensen C, Petersen RK, Hallenborg P, De Matteis R, et al. (2004)<br />

Retinoblastoma protein functions as a molecular switch determining white<br />

versus brown adipocyte differentiation. Proc Natl Acad Sci U S A 101:<br />

4112–4117.<br />

81. Hansen JB, Jorgensen C, Petersen RK, Hallenborg P, De Matteis R, et al. (2004)<br />

Retinoblastoma protein functions as a molecular switch determining white<br />

versus brown adipocyte differentiation. Proc Natl Acad Sci U S A 101:<br />

4112–4117.<br />

82. Madsen L, Pedersen LM, Liaset B, Ma T, Petersen RK, et al. (2008) cAMP<strong>dependent</strong><br />

Signaling Regulates the Adipogenic Effect <strong>of</strong> n-6 Polyunsaturated<br />

Fatty Acids. J Biol Chem 283: 7196–7205.<br />

83. Madsen L, Petersen RK, Sørensen MB, Jørgensen C, Hallenborg P, et al. (2003)<br />

Adipocyte differentiation <strong>of</strong> 3T3-L1 preadipocytes is <strong>dependent</strong> on lipoxygenase<br />

activity during the initial stages <strong>of</strong> the differentiation process. Biochem J 375:<br />

539–549.<br />

84. Hansen JB, Jorgensen C, Petersen RK, Hallenborg P, De Matteis R, et al. (2004)<br />

Retinoblastoma protein functions as a molecular switch determining white<br />

versus brown adipocyte differentiation. Proc Natl Acad Sci U S A 101:<br />

4112–4117.<br />

85. Bauer AK, Dwyer-Nield LD, Malkinson AM (2000) High cyclooxygenase 1<br />

(COX-1) and cyclooxygenase 2 (COX-2) contents in mouse lung tumors.<br />

Carcinogenesis 21: 543–550.<br />

86. Muller-Decker K, Hirschner W, Marks F, Furstenberger G (2002) <strong>The</strong> Effects <strong>of</strong><br />

Cyclooxygenase Isozyme Inhibition onIncisional Wound Healing in Mouse<br />

Skin. 119: 1189–1195.<br />

PLoS ONE | www.plosone.org 13 June 2010 | Volume 5 | Issue 6 | e11391


JBC Papers in Press. Published on June 16, 2011 as Manuscript M111.234732<br />

<strong>The</strong> latest version is at http://www.jbc.org/cgi/doi/10.1074/jbc.M111.234732<br />

Nutritional Regulation <strong>of</strong> Bile Acid Metabolism is Associated with<br />

Improved Pathological Characteristics <strong>of</strong> the Metabolic Syndrome<br />

Bjørn Liaset 1* , Qin Hao 2 , Henry Jørgensen 3 , Philip Hallenborg 4 , Zhen-Yu Du 1 , Tao Ma 2 , Hanns-<br />

Ulrich Marschall 5 , Mogens Kruhøffer 6 , Ruiqiang Li 7 , Qibin Li 7 , Christian Clement Yde 3 ,<br />

Gabriel Criales 1 , Hanne C. Bertram 8 , Gunnar Mellgren 9, 10 , Erik Snorre Øfjord 11 , Erik-Jan<br />

Lock 1 , Marit Espe 1 , Livar Frøyland 1 , Lise Madsen 1,2 , Karsten Kristiansen 2*<br />

1 National Institute <strong>of</strong> Nutrition and Seafood Research, Bergen, Norway, 2 Department <strong>of</strong> Biology,<br />

University <strong>of</strong> Copenhagen, Denmark, 3 Department <strong>of</strong> Animal Health, Welf<strong>are</strong> and Nutrition, Aarhus<br />

University , Denmark, 4 Department <strong>of</strong> Biochemistry and Molecular Biology, University <strong>of</strong> Southern<br />

Denmark, Denmark, 5 Institute <strong>of</strong> Medicine, Sahlgrenska Academy, University <strong>of</strong> Gothenburg,<br />

Sweden, 6 AROS Applied Biotechnology, Aarhus, Denmark, 7 Beijing Genomic Institute, Shenzhen,<br />

Peoples Republic <strong>of</strong> China, 8 Department <strong>of</strong> Food Science, Aarhus University, Denmark, 9 Institute <strong>of</strong><br />

Medicine, University <strong>of</strong> Bergen, Norway, 10 Hormone Laboratory, Haukeland University Hospital,<br />

Norway, 11 Center for Clinical Trials, Bergen, Norway.<br />

*Correspondence to: Bjørn Liaset, National Institute <strong>of</strong> Nutrition and Seafood Research, P.O.<br />

Box 2029 Nordnes, N-5817 Bergen, Norway. Fax: +47 55 90 52 99; Phone: +47 46 81 12 97;<br />

E-mail: bli@nifes.no<br />

or<br />

Karsten Kristiansen, Department <strong>of</strong> Biology, University <strong>of</strong> Copenhagen, Ole Maaløes Vej 5, DK-2200<br />

Copenhagen, Denmark. Fax +45 3522 2128; Phone +45 6011 2408; E mail: kk@bio.ku.dk<br />

Running title: Bile acid metabolism is regulated by dietary protein source<br />

Bile <strong>acids</strong> (BAs) <strong>are</strong> powerful<br />

regulators <strong>of</strong> metabolism, and mice treated<br />

orally with cholic acid <strong>are</strong> protected from<br />

diet-induced obesity, hepatic lipid<br />

accumulation, and increased plasma triacyl<br />

glycerol (TAG) and glucose levels. Here, we<br />

show that plasma BA concentration in rats<br />

was elevated by exchanging the dietary<br />

protein source from casein to salmon<br />

protein hydrolysate (SPH). Importantly, the<br />

SPH-treated rats were resistant to dietinduced<br />

obesity. SPH-treated rats had<br />

reduced fed state plasma glucose and TAG<br />

levels, and lower TAG in liver. <strong>The</strong> elevated<br />

plasma BA concentration was associated<br />

with induction <strong>of</strong> genes involved in energy<br />

metabolism and uncoupling, Dio2, Pgc-1α<br />

and Ucp1, in interscapular brown adipose<br />

tissue. Interestingly, the same<br />

transcriptional pattern was found in white<br />

adipose tissue depots <strong>of</strong> both abdominal and<br />

1<br />

subcutaneous origin. Accordingly, rats fed<br />

SPH-based diet exhibited increased wholebody<br />

energy expenditure and heat<br />

dissipation. In skeletal muscle, expressions<br />

<strong>of</strong> the PPARβ/δ target genes (Cpt-1b,<br />

Angptl4, Adrp, and Ucp3) were induced.<br />

Pharmacological removal <strong>of</strong> BAs by<br />

inclusion <strong>of</strong> 0.5wt% cholestyramine to the<br />

high-fat SPH diet attenuated the reduction<br />

in abdominal obesity, the reduction in liver<br />

TAG and the decrease in non-fasted plasma<br />

TAG and glucose levels. Induction <strong>of</strong> Ucp3<br />

gene expression in muscle by SPH treatment<br />

was completely abolished by cholestyramine<br />

inclusion. Taken together, our data provide<br />

evidence that bile acid metabolism can be<br />

modulated by diet, and that such<br />

modulation may prevent/ameliorate<br />

characteristic features <strong>of</strong> the metabolic<br />

syndrome.<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

Copyright 2011 by <strong>The</strong> American Society for Biochemistry and Molecular Biology, Inc.


Bile <strong>acids</strong> (BAs) <strong>are</strong> synthesized in the<br />

liver from cholesterol. After their synthesis,<br />

they <strong>are</strong> conjugated to the amino <strong>acids</strong> taurine<br />

or glycine in a species-<strong>dependent</strong> manner (1).<br />

Conjugation <strong>of</strong> bile <strong>acids</strong> increases their<br />

solubility and facilitates their secretion into<br />

bile (2). In the intestine, the bile <strong>acids</strong> aid in<br />

the absorption <strong>of</strong> lipophilic nutrients (3).<br />

Normally, the bile <strong>acids</strong> <strong>are</strong> efficiently taken<br />

up by the enterocytes <strong>of</strong> the small intestine,<br />

transported back to the liver, and <strong>are</strong> thus<br />

conserved through the enterohepatic<br />

circulation (4). Bile <strong>acids</strong> <strong>are</strong> activators <strong>of</strong> the<br />

nuclear receptor farnesoid X receptor α<br />

(Fxr/Nr1h4) (5-7). <strong>The</strong> main physiological role<br />

<strong>of</strong> Fxr is to maintain bile acid homeostasis and<br />

to regulate genes encoding enzymes involved<br />

in bile acid synthesis and transport.<br />

In addition to bile acid homeostasis,<br />

Fxr is important for normal lipid metabolism.<br />

Fxr -/- mice display higher triacylglycerol<br />

(TAG) concentrations in serum and liver (8,9)<br />

and have an increased synthesis <strong>of</strong><br />

apolipoprotein (Apo) B-containing lipoproteins<br />

(9). In line with these findings, bile acid<br />

treatment has been reported, in a Fxr<strong>dependent</strong><br />

manner, to induce liver very-lowdensity<br />

lipoprotein receptor (Vldlr)<br />

transcription (10) and to prevent hepatic TAG<br />

accumulation, VLDL secretion and elevated<br />

serum TAG concentration in mice (11). Thus,<br />

bile <strong>acids</strong> and Fxr play central roles in VLDL<br />

lipoprotein metabolism and in the control <strong>of</strong><br />

TAG levels.<br />

Glucose metabolism is also regulated<br />

by bile <strong>acids</strong> and mice treated with cholic acid<br />

<strong>are</strong> protected from diet-induced hyperglycemia<br />

(12) and have down-regulated liver expression<br />

<strong>of</strong> the gluconeogenic phosphoenolpyruvate<br />

carboxykinase (Pck1) gene (13,14).<br />

Furthermore, Fxr -/- mice <strong>are</strong> glucose intolerant<br />

and insulin resistant, and insulin signaling is<br />

blunted in several tissues (15-17). Activation<br />

<strong>of</strong> Fxr with a synthetic ligand, GW4064, or<br />

hepatic over-expression <strong>of</strong> constitutively active<br />

Fxr lowered blood glucose in both diabetic<br />

db/db and wild-type mice (15). It is therefore<br />

evident that bile acid signaling and Fxr<br />

function <strong>are</strong> essential for normal glucose<br />

regulation.<br />

Bile <strong>acids</strong> <strong>are</strong> also endogenous<br />

activators <strong>of</strong> the G-protein coupled receptor<br />

Tgr5 (also referred to as GB37/ M-BAR/<br />

Gpbar1) (18,19). Stimulation <strong>of</strong> Tgr5 by bile<br />

2<br />

<strong>acids</strong> has been shown to increase energy<br />

expenditure and to protect mice from dietinduced<br />

obesity and glucose intolerance (20).<br />

Consistent with the role <strong>of</strong> Tgr5 in the control<br />

<strong>of</strong> energy expenditure, female Tgr5 -/- mice<br />

show increased adiposity when challenged<br />

with a high fat diet (21). Also, Tgr5 activation<br />

by bile <strong>acids</strong> has been reported to promote<br />

production and release <strong>of</strong> glucagon-like<br />

peptide 1 (GLP-1) in enteroendocrine cell lines<br />

(22) and in mice (23), adding another aspect <strong>of</strong><br />

bile acid treatment for the prevention <strong>of</strong> dietinduced<br />

glucose intolerance and insulin<br />

resistance. Thus, bile acid signaling through<br />

Tgr5 might be important for the maintenance<br />

<strong>of</strong> normal energy homeostasis and insulin<br />

sensitivity.<br />

<strong>The</strong> benefits <strong>of</strong> activating Tgr5 or Fxr<br />

for the prevention <strong>of</strong> the metabolic syndrome<br />

have stimulated the development <strong>of</strong> synthetic<br />

ligands for these receptors (24,25). Another<br />

strategy to increase Tgr5 and/or Fxr signaling<br />

would be to alter endogenous BA metabolism.<br />

As hepatic bile acid conjugation is important<br />

for secretion <strong>of</strong> BAs into bile (2), and rats<br />

conjugate BAs to both taurine and glycine with<br />

high efficiency (26), the dietary levels <strong>of</strong> these<br />

amino <strong>acids</strong> might be crucial for BA<br />

conjugation and secretion (27). Feeding rats<br />

casein-based diets supplemented with either<br />

glycine or taurine led to reduced plasma<br />

cholesterol and liver TAG concentrations,<br />

whereas only taurine supplementation reduced<br />

plasma TAG levels (28). Furthermore, taurine<br />

administration has been reported to induce<br />

hepatic protein expression <strong>of</strong> the canalicular<br />

transporter proteins ATP-binding-cassette b11,<br />

Abcb11 (also called Bsep) and Abcc2 (also<br />

called Mrp2). Concomitantly, bile flow and<br />

taurocholate excretion was induced in<br />

experiments with perfused rat livers (29). In<br />

rats fed low-fat diets, we have previously<br />

shown that BA metabolism can be modulated<br />

by dietary proteins with different endogenous<br />

glycine and taurine contents (30).<br />

Thus, evidence is accumulating that<br />

BA metabolism can be modulated by dietary<br />

levels <strong>of</strong> glycine and taurine in normal-energy<br />

diets. However, development <strong>of</strong> the metabolic<br />

syndrome is tightly associated with intake <strong>of</strong><br />

energy-dense diets. <strong>The</strong>refore, the present<br />

study was undertaken in order to test the<br />

hypothesisis that rats treated with a taurineand<br />

glycine-rich protein source, salmon protein<br />

Downloaded from www.jbc.org by guest, on July 7, 2011


hydrolysate (SPH), would be protected from<br />

developing high-fat diet-induced pathological<br />

characteristics <strong>of</strong> the metabolic syndrome. We<br />

demonstrate that bile acid metabolism can be<br />

modulated by the protein source in high-fat fed<br />

rats, and that such a modulation may protect<br />

against development <strong>of</strong> the metabolic<br />

syndrome.<br />

EXPERIMENTAL PROCEDURES<br />

Indirect calorimetric measurements – A<br />

separate set <strong>of</strong> rats (n=6/ group) was fed<br />

experimental diets for 17 days. Heat<br />

production was calculated from gas exchange<br />

measurements, as previously described (32).<br />

Gas exchange was determined twice, each time<br />

for 22 hours. From the average gas exchange<br />

measurements, heat production was calculated<br />

by the respiratory quotient method, and<br />

reported per 24 h.<br />

Animals - Male Wistar Hannover GALAS<br />

(HanTac:WH) were obtained from Taconic<br />

Europe (Ejby, Denmark) and divided into<br />

experimental groups (n=6). <strong>The</strong> rats were kept<br />

at a 12-h light/ dark cycle in a temperaturecontrolled<br />

room at 22 °C. After<br />

acclimatization, the animals were fed<br />

experimental diets with either SPH or casein as<br />

the sole protein source (Supplemental Table 1).<br />

<strong>The</strong> composition <strong>of</strong> the protein sources has<br />

been described elsewhere (31). Feed intake<br />

and body weight were recorded throughout the<br />

experiments and faeces were collected the last<br />

5 days. <strong>The</strong> rats were killed by cardiac<br />

puncture under anaesthesia (0.23 mg/ kg BW<br />

Fentanyl (Janssen) and 0.45 mg/ kg BW<br />

Dormitor Vet (Orion Pharma)). Heparinplasma<br />

and EDTA-plasma containing aprotinin<br />

were prep<strong>are</strong>d from blood. Tissues were<br />

dissected out and weighted. A portion <strong>of</strong> the<br />

liver was used for sub-cellular fractionation<br />

and measurement <strong>of</strong> mitochondrial carnitine<br />

palmitoyl transferase-1 capacity, and portions<br />

<strong>of</strong> interscapular brown adipose tissue (iBAT)<br />

and epididymal white adipose tissue (eWAT)<br />

were homogenized and used for determination<br />

<strong>of</strong> palmitoyl-Coenzyme A oxidation capacity.<br />

<strong>The</strong> rest <strong>of</strong> the tissues were freeze-clamped<br />

and frozen at -80 o C. All animal experiments<br />

were approved by the National State Board <strong>of</strong><br />

Biological Experiments with Living Animals<br />

(Norway and Denmark).<br />

Whole body composition – After 40 days <strong>of</strong><br />

feeding, rats were anaesthetized (0.4 mg<br />

Dormitor Vet (Medetomidin Hydrochlorid)/ kg<br />

BW rat (Orion Pharma, Espoo, Finland), and<br />

whole body composition was determined by a<br />

dual X-ray absorptiometry scanner equipped<br />

with a small animal option (Discovery QDR<br />

Series, Hologic, Bedford, MA, USA).<br />

3<br />

Plasma metabolomics - A separate set <strong>of</strong> rats<br />

(n=5/ group) was fed experimental diets for 25<br />

days. After termination in the fed state, 200 µl<br />

heparin plasma <strong>of</strong> each sample was mixed with<br />

a solution <strong>of</strong> 400 µl 0.9% saline and 20% D 2 O.<br />

Measurements were performed at 310 K on a<br />

on a Bruker Avance III 600 spectrometer,<br />

operating at a 1 H frequency <strong>of</strong> 600.13 MHz,<br />

and equipped with a 5-mm 1 H TXI probe<br />

(Bruker BioSpin, Rheinstetten, Germany). 1 H<br />

NMR spectra were acquired using the Carr-<br />

Purcell-Meiboom-Gill (CPMG) spin-echo<br />

pulse sequence with water suppression. All<br />

spectra were referenced to the lactate doublet<br />

signal at 1.33 ppm. <strong>The</strong> spectra were<br />

segmented into 0.013 ppm bins and each <strong>of</strong> the<br />

bins was integrated. <strong>The</strong> reduced spectra<br />

excluding the residual water signal were<br />

normalized to the whole spectrum. Principal<br />

component analysis (PCA) and partial least<br />

squ<strong>are</strong>s regression discriminate analysis (PLS-<br />

DA) was performed using the Unscrambler<br />

s<strong>of</strong>tw<strong>are</strong> version 9.8 (Camo, Oslo, Norway) to<br />

elucidate biochemical differences between predefined<br />

classes. Martens' uncertainty test was<br />

applied to find significant variables on the full<br />

cross-validated data (33,34).<br />

Bile acid measurements - For total bile acid<br />

measurements in feces, bile <strong>acids</strong> were<br />

extracted according to Suckling et al (35).<br />

Amounts <strong>of</strong> total bile <strong>acids</strong> in fecal extracts<br />

and in non-fasted EDTA-plasma were<br />

determined enzymatically by the 3-αhydroxysteroid<br />

dehydrogenase reaction<br />

(Dialab, Vienna, Austria). Bile <strong>acids</strong> in liver<br />

samples were extracted and analyzed using<br />

gas-chromatography-mass spectrometry and<br />

electrospray-mass spectrometry as previously<br />

described in detail (36).<br />

Real time RT-qPCR - Total RNA was purified<br />

tissues using Trizol, and cDNA was<br />

Downloaded from www.jbc.org by guest, on July 7, 2011


synthesized from individual rats. Gene<br />

expression was determined in individual<br />

samples by real-time qPCR using ABI PRISM<br />

7700 Sequence Detection System (Applied<br />

Biosystems) carried out in 96-well plates and<br />

in duplicate as earlier described (37). Primers<br />

for real-time PCR (Supplemental Table 2)<br />

were designed using Primer Express 2.0<br />

(Applied Biosystems).<br />

Liver microarray analysis- <strong>The</strong> following<br />

procedures were all performed according to<br />

Affymetrix standard procedures. Briefly, equal<br />

amounts <strong>of</strong> RNA isolated from livers were<br />

pooled (n=6) and 5 µg <strong>of</strong> total RNA was used<br />

as starting material for the target preparation.<br />

First and second strand cDNA synthesis were<br />

performed using the SuperScript II System<br />

(Invitrogen) according to the manufacturers’<br />

instructions except using an oligo-dT primer<br />

containing a T7 RNA polymerase promoter<br />

site. Labeled aRNA was prep<strong>are</strong>d using the<br />

BioArray High Yield RNA Transcript<br />

Labeling Kit (Enzo) using Biotin labeled CTP<br />

and UTP (Enzo) in the reaction together with<br />

unlabeled NTP´s. Unincorporated nucleotides<br />

were removed using RNeasy columns<br />

(Qiagen). Fifteen µg <strong>of</strong> cRNA were<br />

fragmented, loading onto the Affymetrix Rat<br />

Genome RAE 230 2.0 probe array cartridge<br />

and hybridized for 16h. <strong>The</strong> arrays were<br />

washed and stained in the Affymetrix Fluidics<br />

Station and scanned using a confocal laserscanning<br />

microscope (GeneChip® Scanner<br />

3000 System with Workstation and<br />

AutoLoader). <strong>The</strong> raw images files from the<br />

quantitative scanning were analyzed by the<br />

Affymetrix Gene Expression Analysis<br />

S<strong>of</strong>tw<strong>are</strong> (MAS 5.0) resulting in cell files<br />

containing background corrected probe values.<br />

Liver microarray KEGG pathway analysis –<br />

Liver microarray data was used to identify<br />

metabolic pathways regulated by treatments,<br />

using the KEGG resource (38,39), release 43.<br />

Genes with at least two-fold change in<br />

expression level between the two treatment<br />

groups were taken as differentially expressed<br />

genes (DEGs). <strong>The</strong> enrichment <strong>of</strong> these DEGs<br />

among KEGG pathways was measured by twotailed<br />

Fisher’s exact test. <strong>The</strong> pathways with P-<br />

value less than 0.05 were considered as<br />

statistically significant and were further<br />

studied.<br />

Histological analyses – Lipids in cryosections<br />

<strong>of</strong> frozen liver samples were stained by the<br />

standard Oil Red-O method. Parts <strong>of</strong> the<br />

epididymal white adipose tissue (eWAT) were<br />

subjected to standard hematoxylin and eosin<br />

staining as previously described (40).<br />

Plasma measurements - Lipids, metabolites<br />

and enzyme activities in plasma were<br />

determined by commercially available<br />

enzymatic kits: alanine aminotransferase<br />

(ALAT), total cholesterol, HDL-cholesterol,<br />

LDL-cholesterol (Dialab, Vienna, Austria),<br />

glucose, TAG (MaxMat, Montpellier, France),<br />

OH-butyrate (Randox, Crumlin, United<br />

Kingdom). Hormones were measured in EDTA<br />

plasma containing aprotonin: insulin by an<br />

ELISA kit (DRG Diagnostics, Germany) and<br />

glucagon by a Radioimmunoassay (RIA) Kit<br />

(LINCO Research, ST. Charles, MO, USA).<br />

Liver glycogen content – Glycogen in liver<br />

samples was converted to glucose by treatment<br />

with amyloglucosidase as described elsewhere<br />

(41). Glucose content after the treatment was<br />

determined using a commercial glucoses<br />

quantification kit (Dialab, Vienna, Austria).<br />

Liver lipid analyses - Total lipids were<br />

extracted from liver samples with chlor<strong>of</strong>orm:<br />

methanol, 2:1 (v/v). <strong>The</strong> lipid classes (TAG,<br />

cholesterol and sterol esters) were analyzed on<br />

an automated Camaq HPTLC system and<br />

separated on HPTLC silica gel 60 F plates as<br />

previously described (42).<br />

Liver mitochondrial preparation and CPT I<br />

assay – Liver mitochondria-enriched fractions<br />

were prep<strong>are</strong>d as previously described (30).<br />

Freshly prep<strong>are</strong>d mitochondrial enriched<br />

fractions were used for determination <strong>of</strong><br />

carnitine-palmitoyl transferase capacity, as<br />

previously described (43), with or without<br />

addition <strong>of</strong> the CPTI inhibitor malonyl-CoA<br />

(5µM).<br />

Liver cytosolic preparation and total<br />

glutathione determination – After removal <strong>of</strong><br />

the mitochondrial enriched fraction, the<br />

homogenates were further centrifuged at<br />

100 000g av for 90 minutes at 4 o C. <strong>The</strong><br />

supernatant collected after this centrifugation<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

4


was used as the liver cytosolic fraction and<br />

total glutathione (GSH) content was<br />

determined by a colorimetric assay, Bioxytech<br />

GSH-400 (OXISResarch TM , Portland, OR,<br />

USA).<br />

Adipose tissue <strong>fatty</strong> acid oxidation capacity –<br />

samples from eWAT and iBAT was<br />

homogenized in a glycine-glycine buffer as<br />

previously described (44). <strong>The</strong> homogenates<br />

were centrifuged at 1000 g for 5 minutes and<br />

the supernatant was collected (post-nuclear<br />

fraction). Fatty acid oxidation capacity was<br />

measured in the post-nuclear fractions by the<br />

acid-soluble product method using radiolabeled<br />

palmitoyl-Coenzyme A as the<br />

substrate, as previously described in detail<br />

(30).<br />

In vitro adipocyte differentiation and <strong>fatty</strong> acid<br />

oxidation – <strong>The</strong> stromal vascular fraction was<br />

isolated from eWAT dissected out from rats as<br />

earlier described (37). Contaminating<br />

erythrocytes were eliminated from the stromalvascular<br />

fraction by a wash with sterile<br />

distilled water. Cells were plated and induced<br />

to differentiate as described (37).<br />

Differentiated cells were treated with<br />

increasing concentrations <strong>of</strong> taurocholic acid in<br />

the medium for 24h. Afterwards, the cells were<br />

scraped and homogenized in a buffer<br />

containing 0.25 M sucrose, 2 mM EGTA and<br />

10 mM Tris/HCl, pH 7.4. Fatty acid oxidation<br />

capacity was evaluated by the total amount <strong>of</strong><br />

acid-soluble products using radio-labeled<br />

palmitoyl-Coenzyme A as the substrate, as<br />

previously described in detail (30).<br />

Energy in faeces and diets - <strong>The</strong> gross energy<br />

content was determined in a bomb calorimeter<br />

following the manufacturer’s instruction (Parr<br />

Instruments, Moline, IL, USA).<br />

Statistical analysis – Data <strong>are</strong> presented as<br />

mean+S.E. Analysis <strong>of</strong> variance was<br />

performed by post-hoc pairwise comparison<br />

Student’s t-test or Tukey’s HSD test. Data that<br />

failed to show homogeneity in variance by<br />

Levenes’ test were tested by the nonparametric<br />

tests Mann-Whitney U test or<br />

Kruskal-Wallis test. Data were considered<br />

statistically different at P


the SPH+c’am treated groups (Fig 1I).<br />

However, the lean body mass including bones<br />

was borderline reduced in the SPH-treated<br />

(231±4 g, P = 0.052) but was significantly<br />

reduced in the SPH+cholestyramine-treated<br />

(223±1 g, P = 0.005), relative to the casein-fed<br />

rats (259±7 g).<br />

Since bile <strong>acids</strong> <strong>are</strong> known to increase<br />

energy expenditure and energy dissipation in<br />

the form <strong>of</strong> heat (20), we considered it likely<br />

that the reduced fat content in the SPH-treated<br />

rats was accompanied by higher energy<br />

dissipation. To demonstrate this we determined<br />

energy expenditure in the rats by indirect<br />

calorimetry. Indeed, SPH-treated rats had<br />

significantly higher O 2 consumption and heat<br />

production, as well as CO 2 elimination relative<br />

to the casein treated rats (Fig 1K-M).<br />

Importantly, and providing further evidence for<br />

a role <strong>of</strong> the increased plasma BA<br />

concentration with regard to the increased<br />

energy expenditure, pharmacological lowering<br />

<strong>of</strong> plasma BAs attenuated heat production, and<br />

led to higher body fat content at an equal<br />

energy intake (Fig 1J-M). <strong>The</strong> higher CO 2<br />

production in the SPH-treated rats was<br />

in<strong>dependent</strong> <strong>of</strong> the reduced bodyweight in<br />

these animals, as total CO 2 production<br />

(Liter/24h) was higher in the SPH, relative to<br />

the casein fed rats, 8.69 ± 0.15 and 8.09 ±<br />

0.11, respectively (P = 0.03). <strong>The</strong><br />

corresponding value for the SPH+c’am treated<br />

rats was 8.31 ± 0.18, which was not<br />

statistically different from neither the SPH nor<br />

the casein treated rats.<br />

Nutritional regulation <strong>of</strong> endogenous<br />

BA metabolism modulates fat oxidation<br />

capacity in brown and white adipose tissues.<br />

Mice ingesting cholic acid supplemented diets<br />

have increased fat oxidation and energy<br />

dissipation as heat in iBAT, which is important<br />

to prevent high-fat diet-induced adiposity (20).<br />

To elucidate whether nutritional modulation <strong>of</strong><br />

plasma BAs could induce fat oxidation in<br />

iBAT, we measured ex vivo <strong>fatty</strong> acid<br />

oxidation capacity in iBAT. In agreement with<br />

the increased whole-body heat production,<br />

<strong>fatty</strong> acid oxidation capacity was significantly<br />

higher in iBAT from SPH fed rats comp<strong>are</strong>d<br />

with rats fed casein (Fig 2A). <strong>The</strong> metabolic<br />

effect <strong>of</strong> bile <strong>acids</strong> on energy expenditure is<br />

critically <strong>dependent</strong> on the cAMP-inducible<br />

thyroid activating enzyme type 2<br />

6<br />

iodothyronine deiodinase (Dio2) and is lost in<br />

Dio2 -/- mice (20). Expression <strong>of</strong> Dio2 has been<br />

linked to expression <strong>of</strong> the thyroid-responsive<br />

gene, uncoupling protein 1 (Ucp1) (45).<br />

Exogenously added bile <strong>acids</strong> have also been<br />

shown to increase the expression <strong>of</strong><br />

peroxisome proliferator-activated receptor γ<br />

coactivator 1α (Pgc-1α) in brown adipose<br />

tissue (20), a key regulator <strong>of</strong> mitochondrial<br />

biogenesis and energy expenditure (46,47).<br />

Here we show that the higher heat production<br />

and <strong>fatty</strong> acid oxidation capacity in iBAT was<br />

accompanied by induction <strong>of</strong> genes encoding<br />

Ucp1 and Dio2 whereas Pgc-1α expression<br />

was not significantly increased (Fig. 2B-D).<br />

Even though brown adipose tissue is<br />

considered the major adipose tissue to dissipate<br />

chemical energy in the form <strong>of</strong> heat (48), white<br />

adipose tissue is plastic and under certain<br />

conditions the expression <strong>of</strong> Pgc-1α and Ucp1<br />

can be induced (49-51). Under conditions with<br />

increased intracellular cAMP signaling, studies<br />

in mice suggest that induced expression <strong>of</strong><br />

Ucp1 in white adipose tissues is associated<br />

with a lean and healthy phenotype (40,52-54).<br />

As BAs <strong>are</strong> known to increase intracellular<br />

cAMP levels, we speculated whether the<br />

increased plasma BA concentrations could also<br />

alter the phenotype <strong>of</strong> WAT. Indeed, the SPHtreated<br />

rats displayed higher expressions <strong>of</strong><br />

Ucp1, Dio2 and Pgc-1α in white adipose<br />

tissues <strong>of</strong> subcutaneous origin (inguinal,<br />

iWAT) and <strong>of</strong> abdominal origin, eWAT and<br />

MeWAT (Fig. 2E-G). <strong>The</strong>se data supported<br />

the idea that not only iBAT, but also white<br />

adipose tissues contributed to the lean<br />

phenotype in the SPH-treated rats.<br />

To further corroborate the hypothesis<br />

that nutritional regulation <strong>of</strong> plasma BA levels<br />

was <strong>of</strong> importance for the WAT phenotype, we<br />

investigated WAT from the rats treated with<br />

SPH and SPH+c’am. Pharmacological removal<br />

<strong>of</strong> BAs from circulation attenuated the<br />

reduction in abdominal WAT mass (eWAT +<br />

MeWAT + perirenal/retroperitoneal WAT)<br />

(Fig. 2H). Furthermore, ex vivo fat oxidation<br />

capacity and expression <strong>of</strong> Ucp1, Dio2 and<br />

Pgc-1α were lower in eWAT from SPH+c’am,<br />

relative to the SPH treated rats, even though<br />

the difference did not reach significant levels<br />

(Fig. 2I, J). <strong>The</strong> lower plasma BA<br />

concentrations were also accompanied by<br />

larger adipocyte cell sizes in eWAT (Fig. 2K).<br />

Downloaded from www.jbc.org by guest, on July 7, 2011


In order to demonstrate that bile <strong>acids</strong> could<br />

induce fat oxidation in white adipocytes, we<br />

isolated preadipocytes from the eWAT depot,<br />

and treated differentiated adipocytes with<br />

increasing concentrations <strong>of</strong> taurocholic acid<br />

for 24hrs. Comp<strong>are</strong>d to untreated cells, a<br />

physiologic relevant dose <strong>of</strong> 25 µM<br />

taurocholic acid significantly raised fat<br />

oxidation capacity in the adipocytes (Fig 2L).<br />

Nutritional regulation <strong>of</strong> endogenous<br />

BA metabolism alters TAG concentrations in<br />

liver and plasma. Oral bile acid treatment<br />

lowers TAG concentrations in mouse liver (11)<br />

and blood (11,55). Moreover, BA treatment<br />

has been reported to down-regulate <strong>fatty</strong> acid<br />

de novo synthesis via repression <strong>of</strong> Sterol<br />

regulatory-element binding protein 1c (Srebp-<br />

1c) and its downstream lipogenic target genes<br />

in mouse primary hepatocytes and liver<br />

(11,56). We found no significant differences in<br />

hepatic gene-expressions <strong>of</strong> Srebp-1c (24±4 vs<br />

34±4, P=0.18), Acetyl-CoA carboxylase 1<br />

(Acc1) (36±2 vs 43±2, P=0.10), <strong>fatty</strong> acid<br />

synthase (Fas) (3.8±0.8 vs 5.7±1.3, P=0.10)<br />

between the high-fat SPH and high-fat casein<br />

treated rats, respectively. Others have reported<br />

that in HepG2 cells, BA treatment represses<br />

transcription <strong>of</strong> microsomal triglyceride<br />

transfer protein (MTP) and APO B, both<br />

important for hepatic secretion <strong>of</strong> VLDL (57).<br />

However, the high-fat SPH-treated rats had<br />

higher liver expression <strong>of</strong> ApoB (6.3±0.6 vs<br />

4.1±0.2, P=0.009) and Mtp (6.6±0.5 vs<br />

5.0±0.2, P=0.14), comp<strong>are</strong>d to high-fat casein<br />

treated rats, respectively. Our results obtained<br />

using rat therefore deviate from the reported<br />

findings in mice and HepG2 cells.<br />

To gain further information on the<br />

mechanisms by which nutritional regulation <strong>of</strong><br />

BA metabolism modulates hepatic TAG<br />

metabolism we used a microarray approach.<br />

To identify functional differences between<br />

treatment groups, we performed a Kegg<br />

pathway analysis. Six Kegg metabolic<br />

pathways were significantly altered, and four<br />

tended (P


Accordingly, when we measured<br />

mitochondrial CPT-1 capacity in the presence<br />

<strong>of</strong> 5 µM malonyl-CoA, we observed a stronger<br />

inhibition in SPH-treated rats than in casein<br />

treated rats, even though the difference not was<br />

significant (Fig. 4G). Furthermore, the fasting<br />

plasma levels <strong>of</strong> the ketone-body β-<br />

hydroxybutyrate, a marker <strong>of</strong> liver<br />

mitochondrial <strong>fatty</strong> acid oxidation was<br />

significantly elevated (Fig. 4H) and liver TAG<br />

deposition was lower in the SPH-treated rats<br />

(Fig. 4I, J), further supporting that the SPHtreated<br />

rats had higher liver fat catabolism.<br />

Exogenous bile acid supplementation (11), as<br />

well as the activation <strong>of</strong> the PPAR β/δ receptor<br />

(61) prevents increased plasma TAG through<br />

inhibition <strong>of</strong> hepatic VLDL secretion. Higher<br />

liver Vldlr expression might be one underlying<br />

mechanism to the lower plasma VLDL<br />

amounts, as both BA-treatment (10) and<br />

expression and activation <strong>of</strong> the<br />

PPARβ/δ receptor (62) might induce Vldlr<br />

transcription. As the rats given the high-fat<br />

SPH diet had both elevated plasma BA<br />

concentrations and induction <strong>of</strong> Pparβ/δ gene<br />

expression in liver, we hypothesized that<br />

plasma TAG would be decreased in these<br />

animals. As predicted, the plasma TAG<br />

concentrations were reduced in the non-fasted<br />

state (Fig. 4K), and this decrease was<br />

accompanied by a pronounced elevation in<br />

liver Vldlr expression (Fig. 4L). To verify that<br />

plasma VLDL concentration actually was<br />

reduced, we analyzed plasma samples by<br />

nuclear magnetic resonance (NMR), which<br />

confirmed that SPH-treated rats had decreased<br />

VLDL amounts (Fig. 4M). <strong>The</strong> NMR analysis<br />

also suggested that plasma LDL- or HDLcholesterol<br />

concentrations were elevated in the<br />

SPH-treated rats, but no significant differences<br />

were observed in plasma cholesterol levels by<br />

standard clinical chemistry methods (Fig. N,<br />

O).<br />

Our data supported the hypothesis that<br />

nutritional regulation <strong>of</strong> endogenous BA<br />

metabolism could modulate hepatic <strong>fatty</strong> acid<br />

oxidation capacity, liver TAG storage and<br />

plasma non-fasted TAG-rich VLDL<br />

concentrations. To investigate further the<br />

potential role <strong>of</strong> BAs in relation to the<br />

observed <strong>effects</strong> on TAG metabolism, we<br />

examined the rats treated with<br />

SPH+cholestyramine. Indeed, pharmacological<br />

removal <strong>of</strong> BAs by the cholestyramine<br />

treatment attenuated the reduction in liver and<br />

non-fasted plasma TAG concentrations (Fig.<br />

4P, Q), and attenuated the elevation in liver<br />

mitochondrial fat oxidation capacity and nonfasted<br />

plasma β-hydroxybutyrate (Fig. 4R, S).<br />

We conclude that nutritional regulation <strong>of</strong><br />

endogenous BA metabolism contributes<br />

significantly to the observed modulation <strong>of</strong><br />

lipid metabolism introduced by SPH treatment.<br />

Nutritional regulation <strong>of</strong> endogenous<br />

BA metabolism modulates skeletal muscle<br />

Ucp3 expression. PPARβ/δ is abundantly<br />

expressed in skeletal muscle, a peripheral<br />

tissue that accounts for approximately 40% <strong>of</strong><br />

total body mass. Activation <strong>of</strong> PPARβ/δ leads<br />

to increased expression <strong>of</strong> genes involved in<br />

lipid catabolism and energy uncoupling in<br />

skeletal muscle (63-65). One <strong>of</strong> the genes<br />

robustly induced by PPARβ/δ activation is<br />

Ucp3, and mice over-expressing the human<br />

UCP3 in skeletal muscle <strong>are</strong> protected from<br />

diet-induced adiposity (66). In keeping with<br />

the strong hepatic Pparβ/δ induction<br />

combined with the lean phenotype <strong>of</strong> the SPH<br />

treated rats, we hypothesized that<br />

PPARβ/δ signaling was altered also in skeletal<br />

muscle <strong>of</strong> the SPH treated rats. As in liver,<br />

skeletal muscle expressions <strong>of</strong> Pparα and its<br />

target genes Acox1 were down-regulated in the<br />

SPH treated rats (Fig. 5A). In contrast,<br />

expression <strong>of</strong> Pparβ/δ , and its downstream<br />

target genes Adrp and angiopoietin-like 4<br />

(Angptl4, also called fasting-induced adipose<br />

factor, Fiaf) were significantly induced (Fig.<br />

5B). Concomitantly, Cpt-1b, Ucp2 and in<br />

particular Ucp3 expressions were elevated<br />

(Fig. 5C). To further corroborate a regulatory<br />

role for BAs on skeletal muscle Ucp3<br />

expression, we measured the Ucp3 expression<br />

in the SPH+c’am treated rats. Of note, skeletal<br />

muscle Ucp3 induction by SPH treatment was<br />

completely abolished by reducing plasma BA<br />

concentrations with cholestyramine (Fig. 5D).<br />

Our data indicate that besides the known<br />

regulatory <strong>effects</strong> <strong>of</strong> BAs on liver and adipose<br />

tissue metabolism, skeletal muscle <strong>fatty</strong> acid<br />

oxidation and uncoupling may also be<br />

regulated through altered bile acid metabolism.<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

8


Nutritional regulation <strong>of</strong> endogenous<br />

BA metabolism modulates high-fat dietinduced<br />

hyperglycemia. Oral BA treatment<br />

reduces hyperglycemia in mice (12), possibly<br />

through reduced hepatic glucose output by<br />

repression <strong>of</strong> the Pck1 gene expression (13,14).<br />

In addition, PPARβ/δ activation reduces<br />

hepatic glucose output and improves peripheral<br />

glucose disposal (67). In the present study, the<br />

SPH-treated rats both had elevated plasma BA<br />

levels and induction <strong>of</strong> Pparβ/δ target gene<br />

expression in liver and skeletal muscle.<br />

<strong>The</strong>refore, we decided to examine whether<br />

glucose metabolism was improved in these<br />

animals. <strong>The</strong> non-fasted plasma insulin and<br />

liver Pck1 expression were lower in the SPHtreated<br />

rats (Fig. 6A, B). Hepatic glycogen<br />

concentrations were significantly higher (Fig. 6<br />

C), whereas non-fasted glucose was<br />

significantly lower in the SPH-treated rats<br />

(Fig. 6D). No difference was observed in<br />

fasting plasma glucose levels (Fig. 6D). <strong>The</strong><br />

lower fed-state plasma insulin and glucose<br />

concentrations, as well as the reduced hepatic<br />

Pck-1 expression in the SPH fed rats might<br />

indicate that these animals were more insulinsensitive,<br />

relative to those fed casein. This<br />

notion is in agreement with a recent report in<br />

which rats fed a high-fat SPH-diet had<br />

increased whole-body insulin-sensitivity<br />

determined by the hyperinsulinemiceuglycemic<br />

clamp technique (68). In the<br />

present study, pharmacological removal <strong>of</strong><br />

BAs attenuated the reduction in non-fasted<br />

blood glucose (Fig. 6E). <strong>The</strong>refore, our data<br />

support the hypothesis that nutritional<br />

modulation <strong>of</strong> endogenous BA metabolism<br />

may regulate blood glucose concentrations.<br />

Possible determinants <strong>of</strong> the elevated<br />

plasma BA levels in the SPH treated rats. In<br />

the present study, the SPH treated rats had<br />

elevated plasma BA levels. Increased<br />

circulating BAs might be due to cholestasis or<br />

increased bile acid synthesis. <strong>The</strong> plasma<br />

levels <strong>of</strong> alanine aminotransferase, although<br />

slightly elevated, did not indicate<br />

hepatocellular damage in the SPH fed rats (Fig.<br />

7A). Furthermore, liver Ucp2 gene expression,<br />

recently shown to be up-regulated in bile duct<br />

obstructed rats (69), was borderline (P=0.053)<br />

down-regulated in the SPH fed rats (Fig. 7B).<br />

Hence, it is unlikely that the elevated plasma<br />

BA level was due to cholestasis in the present<br />

9<br />

study.<br />

Supplementing taurine (70,71) or<br />

glycine (70) at doses <strong>of</strong> 50g/ kg diet to caseinbased,<br />

atherogenic diets (containing 1wt%<br />

cholesterol and 0.25 wt% cholic acid) have<br />

previously been shown to increase fecal BA<br />

excretion in rats. Furthermore, the rats treated<br />

with the taurine-supplemented diet had higher<br />

liver GSH content, decreased liver cholesterol<br />

concentration and higher cholesterol-7a<br />

hydroxylase (Cyp7a1) mRNA levels and<br />

enzyme activity (71). In the present study, the<br />

SPH diet provided taurine (1.9g/kg diet),<br />

whereas the casein diet was free <strong>of</strong> taurine.<br />

Also, the glycine level differed and was 23 and<br />

4 g/ kg diet in the SPH and casein diets,<br />

respectively (Supplemental Fig. 1). <strong>The</strong> SPHtreated<br />

rats had significantly reduced liver<br />

cholesterol and sterol ester concentrations (Fig.<br />

7C). However, the SPH fed rats had lower liver<br />

expression <strong>of</strong> genes involved in the de novo<br />

bile acid synthesis, Cyp7a1, and in bile acid<br />

conjugation, Bile acid Coenzyme A: amino<br />

acid N-acyltransferase, (Baat) (Fig. 7D).<br />

<strong>The</strong>refore, our data does not support that a<br />

higher BA synthesis was the underlying factor<br />

for the elevated plasma BA concentrations.<br />

Secretion <strong>of</strong> bile <strong>acids</strong> into bile is<br />

partly <strong>dependent</strong> on bile acid availability and<br />

transport, and partly determined by the<br />

availability and transport <strong>of</strong> phospholipids,<br />

cholesterol, glutathione and bicarbonate, as<br />

reviewed in (72). In the present study, the liver<br />

expression <strong>of</strong> genes involved in canalicular<br />

bile flow generation was either increased (Atpbinding<br />

cassette b4, Abcb4, also called Mdr2),<br />

tended (P=0.09) to be increased (Abcb11/<br />

Bsep), or was statistically unaltered<br />

(Abcb1b/Mdr1 and Abcc2/ Mrp2) in the SPHtreated<br />

rats (Fig. 7E). Furthermore, the liver<br />

gene expression <strong>of</strong> Glutathione synthase (Gss),<br />

as well as the liver concentration <strong>of</strong> glutathione<br />

(GSH), a primary osmotic driving force in<br />

hepatic bile formation (73), was increased by<br />

SPH treatment (Fig. 7F, G). However, fecal<br />

BA excretion measured over 5 days did not<br />

differ between the treatments (Fig. 7H). Thus,<br />

our liver data indicate that the SPH fed rats had<br />

increased biliary BA secretion, yet the fecal<br />

BA excretion was unaltered. If this was the<br />

case, the SPH-fed rats must have had an<br />

efficient intestinal BA re-uptake. In humans<br />

with a normal hepatic function, the major<br />

determinant <strong>of</strong> circulating bile acid<br />

Downloaded from www.jbc.org by guest, on July 7, 2011


concentration is their rate <strong>of</strong> intestinal<br />

absorption (74). In the present study, the<br />

hepatic amount <strong>of</strong> α-muricholic acid, a bile<br />

acid <strong>of</strong> intestinal origin (75) was significantly<br />

higher in the SPH-fed rats (Fig. 1B). Based on<br />

the higher hepatic presence <strong>of</strong> intestinalderived<br />

BA species, as well as the fact that the<br />

SPH + cholestyramine treated rats exhibited<br />

reduced plasma BA levels, we conclude that<br />

the increased plasma BA level in the SPH-fed<br />

rats was likely due to higher intestinal influx.<br />

DISCUSSION<br />

Bile <strong>acids</strong> strongly affect metabolism<br />

and energy-expenditure in mice (11,12,20).<br />

From studies in rats, it is known that<br />

supplementing low-fat, casein-based diets with<br />

rather high doses <strong>of</strong> glycine and/or taurine<br />

affects bile acid metabolism (29,70,71).<br />

Previously, we have reported that exchanging<br />

casein with a glycine and taurine rich proteinsource<br />

modulated BA metabolism in low-fat<br />

fed rats (30). Here we show that choice <strong>of</strong><br />

dietary protein source is sufficient to increase<br />

plasma BA levels also in high-fat fed rats.<br />

Importantly, the elevated plasma BA level was<br />

accompanied with attenuated diet-induced<br />

obesity and ameliorated characteristics <strong>of</strong> the<br />

metabolic syndrome.<br />

Little is known about the role(s) that<br />

endogenous BA metabolism may play in the<br />

development <strong>of</strong> the metabolic syndrome.<br />

However, a few recent publications support<br />

such a role for endogenous bile acid<br />

metabolism. Bile secretion is impaired in both<br />

the Zucker (fa/fa) rat (76) and in the ob/ob<br />

mice (77), two animal models extensively<br />

studied as they both develop metabolic features<br />

resembling the metabolic syndrome.<br />

Furthermore, in lean and fat mouse lines<br />

developed from the same founder population<br />

by long-term divergent selection for low- or<br />

high body fat %, the lean mice exhibited<br />

higher hepatic Cyp8b1 and Abcb11 gene<br />

expressions, and had increased blood BA<br />

concentrations, relative to the obese mice (78).<br />

Even though the underlying mechanisms need<br />

to be further elucidated, our data strongly<br />

indicate a regulatory role for endogenous bile<br />

acid metabolism on the development <strong>of</strong> the<br />

metabolic syndrome.<br />

We used diets in which the casein was<br />

fully exchanged with SPH. Obviously,<br />

10<br />

differences in dietary amino <strong>acids</strong> other than<br />

taurine and glycine, such as the level <strong>of</strong> the<br />

branched-chain amino <strong>acids</strong> might have<br />

affected the outcome in the present study<br />

(Supplemetal Fig. 1). Moreover, we recognize<br />

that taurine has metabolic <strong>effects</strong> beyond bile<br />

acid metabolism, such as anti-oxidant capacity<br />

(79). However, the taurine tissue level in the<br />

rats (Supplemetal Fig 1) excludes the<br />

possibility that the observed differences were<br />

due to increased tissue taurine concentrations.<br />

Since addition <strong>of</strong> cholestyramine reduced<br />

plasma BA levels and attenuated the beneficial<br />

<strong>effects</strong> on adiposity, TAG metabolism and<br />

hyperglycemia, we conclude that parts <strong>of</strong> the<br />

beneficial <strong>effects</strong> found by SPH treatment was<br />

caused by nutritional regulation <strong>of</strong> endogenous<br />

BA metabolism.<br />

Our results provide evidence that<br />

nutritional regulation <strong>of</strong> bile acid metabolism<br />

may attenuate characteristics <strong>of</strong> the metabolic<br />

syndrome in high-fat fed rats. <strong>The</strong> relevance <strong>of</strong><br />

our findings for man remains to be elucidated.<br />

However, it has been reported that obese<br />

subjects had a lower post-prandial bile-acid<br />

response, relative to normal weight subjects<br />

(80). Furthermore, subjects that had previously<br />

undergone gastric bypass, showed higher<br />

circulating bile acid levels, relative to both<br />

obese and severely obese objects. Also, total<br />

bile <strong>acids</strong> were inversely correlated with 2hr<br />

post meal-glucose and fasting TAG levels (81).<br />

In another study with objects subjected to<br />

bariatric surgery, circulating bile acid and<br />

GLP-1 levels increased post surgically, relative<br />

to pre-surgical baseline levels (82). Finally, it<br />

was reported in a human intervention study<br />

with cross-over design that subjects on a high<br />

fat, high protein diet had increased fasting<br />

plasma bile acid concentrations as comp<strong>are</strong>d to<br />

subjects on a high-fat diet alone. <strong>The</strong> elevation<br />

in plasma BA levels was accompanied by<br />

reduced hepatic lipids and increased fasting<br />

plasma concentrations <strong>of</strong> β-hydroxybutyrate<br />

(83). Thus, nutritional regulation <strong>of</strong><br />

endogenous BA metabolism may also be<br />

related to development <strong>of</strong> the metabolic<br />

syndrome in man.<br />

In conclusion, we provide compelling<br />

evidence that plasma bile acid levels can be<br />

modulated by the dietary protein source in<br />

high-fat treated rats. Increased levels <strong>of</strong> plasma<br />

BAs were associated with a significant<br />

reduction in diet-induced obesity and resulted<br />

Downloaded from www.jbc.org by guest, on July 7, 2011


in increased whole body energy expenditure<br />

and dissipation <strong>of</strong> energy in the form <strong>of</strong> heat.<br />

Concomitantly, fed-state plasma glucose and<br />

TAG concentrations were reduced. Thus,<br />

protein source <strong>dependent</strong> increase in plasma<br />

BA levels appears to have the potential to<br />

attenuate pathological characteristics <strong>of</strong> the<br />

metabolic syndrome in the high-fat fed rats.<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

11


REFERENCES<br />

1. Shonsey EM, Sfakianos M, Johnson M, et al. (2005) Meth. Enzymol. 400: 374-394<br />

2. Vessey DA, Whitney J, and Gollan JL. (1983) Biochem. J. 214: 923-927.<br />

3. H<strong>of</strong>mann AF. (1999) News Physiol. Sci. 14: 24-29.<br />

4. Pauli-Magnus C, Stieger B, Meier Y, Kullak-Ublick GA, and Meier PJ. (2005) J. Hepatol. 43:<br />

342-357.<br />

5. Parks DJ, Blanchard SG, Bledsoe RK, et al. (1999) Science 284: 1365-1368.<br />

6. Makishima M, Okamoto AY, Repa JJ, et al. (1999) Science 284: 1362-1365.<br />

7. Wang H, Chen J, Hollister K, Sowers LC, and Forman BM. (1999) Mol. Cell 3: 543-553.<br />

8. Sinal CJ, Tohkin M, Miyata M, Ward JM, Lambert G, and Gonzalez FJ. (2000) Cell 102: 731-<br />

744.<br />

9. Lambert G, Amar MJA, Guo G, Brewer HB, Gonzalez FJ, and Sinal CJ. (2003) J. Biol. Chem. 278:<br />

2563-2570.<br />

10. Sirvent A, Claudel T, Martin G, et al. (2004) Febs Letters 566: 173-177.<br />

11. Watanabe M, Houten SM, Wang L, et al. (2004) J. Clin. Invest. 113: 1408-1418.<br />

12. Ikemoto S, Takahashi M, Tsunoda N, et al. (1997) Am. J. Physiol. Endocrinol. Metab. 36: E37-<br />

E45.<br />

13. De Fabiani E, Mitro N, Gilardi F, Caruso D, Galli G, and Crestani M. (2003) J. Biol. Chem. 278:<br />

39124-39132.<br />

14. Cao RS, Cronk ZX, Zha WB, et al. (2010) J. Lipid Res. 51: 2234-2244.<br />

15. Zhang YQ, Lee FY, Barrera G, et al. (2006) Proc. Natl. Acad. Sci. USA 103: 1006-1011.<br />

16. Ma K, Saha PK, Chan L, and Moore DD. (2006) J. Clin. Invest. 116: 1102-1109.<br />

17. Cariou B, van Harmelen K, Duran-Sandoval D, et al. (2006) J. Biol. Chem. 281: 11039-11049.<br />

18. Maruyama T, Miyamoto Y, Nakamura T, et al. (2002) Biochem. Biophys. Res. Commun. 298:<br />

714-719.<br />

19. Kawamata Y, Fujii R, Hosoya M, et al. (2003) J. Biol. Chem. 278: 9435-9440.<br />

20. Watanabe M, Houten SM, Mataki C, et al. (2006) Nature 439: 484-489.<br />

21. Maruyama T, Tanaka K, Suzuki J, et al. (2006) J. Endocrinol. 191: 197-205.<br />

22. Katsuma S, Hirasawa A, and Tsujimoto G. (2005) Biochem. Biophys. Res. Commun. 329: 386-<br />

390.<br />

23. Thomas C, Gioiello A, Noriega L, et al. (2009) Cell Metab. 10: 167-177.<br />

24. Sato H, Macchiarulo A, Thomas C, et al. (2008) J. Med. Chem. 51: 1831-1841.<br />

25. Evans MJ, Mahaney PE, Borges-Marcucci L, et al. (2009) Am. J. Physiol. Gastrointest. Liver<br />

Physiol. 296: G543-G552.<br />

26. Zhang RW, Barnes S, and Diasio RB. (1992) Am. J. Physiol. 262: G351-G358.<br />

27. Hardison WGM. (1978) Gastroenterology 75: 71-75.<br />

28. Park T, Oh J, and Lee K. (1999) Nutr. Res. 19: 1777-1789.<br />

29. Mühlfeld A, Kubitz R, Dransfeld O, Haussinger D, and Wettstein M. (2003) Arch. Biochem.<br />

Biophys. 413: 32-40.<br />

30. Liaset B, Madsen L, Hao Q, et al. (2009) Biochim. Biophys. Acta (BBA) - Mol. Cell Biol. Lip.<br />

1791: 254-262.<br />

31. Liaset B, and Espe M. (2008) Proc. Biochem. 43: 42-48.<br />

32. Lauridsen C, Yong C, Halekoh U, et al. (2008) J. Sci. Food Agric. 88: 720-732.<br />

33. Martens HA, and Dardenne P. (1998) Chemometr. Intelligent Lab. Syst. 44: 99-121.<br />

34. Martens H, and Martens M. (2000) Food Qual. Pref. 11: 5-16.<br />

35. Suckling KE, Benson GM, Bond B, et al. (1991) Atherosclerosis 89: 183-190.<br />

36. Marschall H-U, Wagner M, Bodin K, et al. (2006) J. Lipid Res. 47: 582-592.<br />

37. Madsen L, Petersen RK, Sorensen MB, et al. (2003) Biochem. J. 375: 539-549.<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

12


38. Kanehisa M, and Goto S. (2000) Nucl. Acids Res. 28: 27-30.<br />

39. Kanehisa M, Goto S, Hattori M, et al. (2006) Nucl. Acids Res. 34: D354-D357.<br />

40. Madsen L, Pedersen LM, Lillefosse HH, et al. (2010) PLoS ONE 5: e11391<br />

41. Bär A, Lina BAR, de Groot DMG, de Bie B, and Appel MJ. (1999) Regul. Toxicol. Pharmacol. 29:<br />

S11-S28.<br />

42. Liaset B, Julshamn K, and Espe M. (2003). Proc. Biochem. 38: 1747-1759.<br />

43. Madsen L, and Berge RK. (1999) Lipids 34: 447-456.<br />

44. Bas S, and Giacobino JP. (1983) Arch. Biochem. Biophys. 222: 416-423.<br />

45. Xue BZ, Coulter A, Rim JS, Koza RA, and Kozak LP. (2005) Mol. Cell. Biol. 25: 8311-8322.<br />

46. Wu Z, Puigserver P, Andersson U, et al. (1999) Cell 98: 115-124.<br />

47. Lin J, Handschin C, and Spiegelman BM. (2005) Cell Metab. 1: 361-370.<br />

48. Cannon B, and Nedergaard J. (2004) Physiol. Rev. 84: 277-359.<br />

49. Hansen JB, Jorgensen C, Petersen RK, et al. (2004) Proc. Natl. Acad. Sci. USA 101: 4112-4117.<br />

50. Frontini A, and Cinti S. (2010) Cell Metab. 11: 253-256.<br />

51. Barbatelli G, Murano I, Madsen L, et al. (2010) Am. J. Physiol. Endocrinol. Metab. 298: E1244-<br />

E1253.<br />

52. Ghorbani M, Claus TH, and Himms-Hagen J. (1997) Biochem. Pharmacol. 54: 121-131.<br />

53. Cederberg A, Gronning LM, Ahren B, Tasken K, Carlsson P, and Enerback S. (2001) Cell 106:<br />

563-573.<br />

54. Madsen L, Pedersen LM, Liaset B, et al. (2008) J. Biol. Chem. 283: 7196-7205.<br />

55. Kast HR, Nguyen CM, Sinal CJ, et al. (2001) Mol. Endocrinol. 15: 1720-1728.<br />

56. Zhang Y, Castellani LW, Sinal CJ, Gonzalez FJ, and Edwards PA. (2004) Genes Dev. 18: 157-169.<br />

57. Hirokane H, Nakahara M, Tachibana S, Shimizu M, and Sato R. (2004) J. Biol. Chem. 279:<br />

45685-45692.<br />

58. Pineda Torra I, Claudel T, Duval C, Kosykh V, Fruchart J-C, and Staels B. (2003) Mol.<br />

Endocrinol. 17: 259-272.<br />

59. Sinal CJ, Yoon M, and Gonzalez FJ. (2001) J. Biol. Chem. 276: 47154-47162.<br />

60. Wang Y-X, Lee C-H, Tiep S, et al. (2003) Cell 113: 159-170.<br />

61. Akiyama TE, Lambert G, Nicol CJ, et al. (2004) J. Biol. Chem. 279: 20874-20881.<br />

62. Sanderson LM, Boekschoten MV, Desvergne B, Muller M, and Kersten S. (2010) Physiol.<br />

Genom. 41: 42-52.<br />

63. Tanaka T, Yamamoto J, Iwasaki S, et al. (2003) Proc. Natl. Acad. Sci. USA 100: 15924-15929.<br />

64. Dressel U, Allen TL, Pippal JB, Rohde PR, Lau P, and Muscat GEO. (2003) Mol. Endocrinol. 17:<br />

2477-2493.<br />

65. Kleiner S, Nguyen-Tran V, B<strong>are</strong> O, Huang XM, Spiegelman B, and Wu ZD. (2009) J. Biol. Chem.<br />

284: 18624-18633.<br />

66. Clapham JC, Arch JRS, Chapman H, et al. (2000) Nature 406: 415-418.<br />

67. Lee C-H, Olson P, Hevener A, et al. (2006) Proc. Natl. Acad. Sci. USA 103: 3444-3449.<br />

68. Pilon G, Ruzzin J, Rioux L-E, et al. (2011) Metab. Clin. Exp. doi:10.1016/j.metabol.2010.12.005<br />

69. Enochsson L, Isaksson B, Strömmer L, Erlanson-Albertsson C, and Permert J. (2010) Nutrition<br />

26: 405-410.<br />

70. Sugiyama K, Ohishi A, Ohnuma Y, and Muramatsu K. (1989) Agricult. Biol. Chem. 53: 1647-<br />

1652.<br />

71. Yokogoshi H, Mochizuki H, Nanami K, Hida Y, Miyachi F, and Oda H. (1999) J. Nutr. 129: 1705-<br />

1712.<br />

72. Esteller A. (2008) World J. Gastroenterol. 14: 5641-5649.<br />

73. Ballatori N, and Truong AT. (1992) Am. J. Physiol. Gastrointest. Liver Physiol. 263: G617-624.<br />

74. Larusso NF, H<strong>of</strong>fman NE, Korman MG, H<strong>of</strong>mann AF, and Cowen AE. (1978) Am. J. Digest. Dis.<br />

23: 385-391.<br />

75. H<strong>of</strong>mann AF, and Hagey LR. (2008) Cell. Mol. Life Sci. 65: 2461-2483.<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

13


76. Pizarro M, Balasubramaniyan N, Solis N, et al. (2004) Gut 53: 1837-1843.<br />

77. Martin IV, Schmitt J, Minkenberg A, et al. (2010) Biol. Chem. 391: 1441-1449.<br />

78. Simoncic M, Rezen T, Juvan P, et al. (2011) BMC Genomics 12: 96.<br />

79. Bouckenooghe T, Remacle C, and Reusens B. (2006) Curr. Opin. Clin. Nutr. Metab. C<strong>are</strong> 9:<br />

728-733.<br />

80. Glicksman C, Pournaras DJ, Wright M, et al. (2010) Annals Clin. Biochem. 47: 482-484.<br />

81. Patti M-E, Houten SM, Bianco AC, et al. (2009) Obesity 17: 1671-1677.<br />

82. Nakatani H, Kasama K, Oshiro T, Watanabe M, Hirose H, and Itoh H. (2009) Metab. Clin. Exp.<br />

58: 1400-1407.<br />

83. Bortolotti M, Kreis R, Debard C, et al. (2009) Am. J. Clin. Nutr. 90: 1002-1010.<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

14


FOOTNOTES<br />

This work was carried out as a part <strong>of</strong> the ‘DOCMAR’ research program funded by Innovation<br />

Norway and RUBIN. This work was supported by the Danish Natural Science Research Council, <strong>The</strong><br />

Novo Nordisk Foundation, <strong>The</strong> Carlsberg Foundation and performed as part <strong>of</strong> the research program<br />

<strong>of</strong> the Danish Obesity Research Centre, supported by the Danish Council for Strategic Research, Grant<br />

2101-06-0005. Financial support was also received from the University <strong>of</strong> Bergen, Program<br />

Committee on Nutrition, from the Eckbo Foundation and from the Norwegian Research Council,<br />

Grant 200515-I30.<br />

ACKNOWLEDGMENTS<br />

We thank Aase Heltveit and Lars Erik Pindard for help during the animal studies. We also thank Guro<br />

K. Christensen and Jacop Wessels for valuable technical assistance.<br />

FIGURE LEGENDS<br />

FIGURE 1. Elevated plasma bile acid concentration is an underlying factor for the increased<br />

energy expenditure and decreased adiposity elicited by salmon protein hydrolysate (SPH)<br />

feeding.(A-F) Male Wistar rats (n=6) were fed high-fat diets (45 kcal% fat) ad libitum for 46 days<br />

with either SPH or casein as the sole protein source. (A-B) SPH-fed rats had elevated plasma bile<br />

<strong>acids</strong> (BAs), whereas total liver BAs were unchanged. (C-E) <strong>The</strong> SPH-fed rats showed reduced body<br />

weight gain. Energy efficiency, calculated as bodyweight gain per energy intake, and white adipose<br />

tissue masses were reduced by SPH treatment. (F) Energy digestibility was equal in SPH and casein<br />

fed rats. (G-M) Three groups <strong>of</strong> rats (n=6) were pair-fed the SPH-diet, the casein diet and the SPHdiet<br />

with 0.5wt% cholestyramine (c’am). (G-I) Inclusion <strong>of</strong> cholestyramine to the SPH-diet attenuated<br />

the increase in plasma BA concentrations, without modulating liver total BAs or growth. (J) Inclusion<br />

<strong>of</strong> cholestyramine attenuated the reduction in body fat mass determined by dual-X ray absorptiometry<br />

(DEXA). (K-L) Three groups <strong>of</strong> rats (n=6) were pair-fed the SPH-diet, the casein diet and the SPHdiet<br />

with 0.5wt% cholestyramine (c’am) and energy expenditure was calculated by indirect<br />

calorimetry. Cholestyramine treatment attenuated the increase in O2 consumption, CO2 elimination and<br />

heat production. Data <strong>are</strong> presented as mean + S.E. Significant differences from casein-fed rats <strong>are</strong><br />

denoted by * (P


FIGURE 3. Liver microarray analysis. Male Wistar rats (n=6) were fed high-fat diets (45 kcal% fat)<br />

ad libitum for 46 days with either SPH or casein as the sole protein source. RNA from individual rats<br />

was pooled within experimental groups, and the pooled samples were analyzed by Affymetrix arrays.<br />

(A) Data were analyzed and significantly altered KEGG pathways (P


fed the same diets, plus the SPH-diet with 0.5wt% cholestyramine (c’am) for 46 days.<br />

Pharmacological removal <strong>of</strong> bile <strong>acids</strong> by 0.5 wt% cholestyramine attenuated the glucose lowering<br />

effect <strong>of</strong> SPH. Pck1 mRNA levels <strong>are</strong> normalized to General transcription factor IIB (Gtf2b). Data <strong>are</strong><br />

presented as mean + S.E. Significant difference from casein-fed rats <strong>are</strong> denoted by * (P


Figure 1<br />

A<br />

30<br />

Plasma BA<br />

**<br />

B<br />

1,5<br />

Liver BAs<br />

SPH<br />

Casein<br />

C<br />

300<br />

Growth chart<br />

µmol/ L<br />

20<br />

10<br />

0<br />

SPH<br />

Casein<br />

µmol/ tissue<br />

1,0<br />

0,5<br />

0,0<br />

**<br />

Gram BW<br />

200<br />

100<br />

0<br />

0 10 20 30 40 50<br />

Days <strong>of</strong> feeding<br />

Casein<br />

SPH<br />

D<br />

Energy efficiency<br />

E<br />

Adipose tissue masses<br />

F<br />

Energy digestibility<br />

Gram BW/ MJ intake<br />

21<br />

14<br />

7<br />

0<br />

**<br />

SPH<br />

Casein<br />

g tissue<br />

6<br />

4<br />

2<br />

0<br />

**<br />

**<br />

eWAT MeWAT iBAT<br />

SPH<br />

Casein<br />

% <strong>of</strong> ingested energy<br />

96<br />

64<br />

32<br />

0<br />

SPH<br />

Casein<br />

G<br />

µmol/ L<br />

J<br />

Gram<br />

54<br />

36<br />

18<br />

0<br />

90<br />

60<br />

30<br />

0<br />

Plasma BA<br />

**<br />

SPH SPH +<br />

c'am<br />

Body fat<br />

**<br />

SPH SPH +<br />

c'am<br />

Casein<br />

Casein<br />

H<br />

µmol/ tissue<br />

K<br />

Liter O2/ kg BW/ 24h<br />

3<br />

2<br />

1<br />

0<br />

60<br />

50<br />

40<br />

Liver total BAs<br />

SPH SPH +<br />

c'am<br />

O 2 consumed<br />

**<br />

*<br />

SPH SPH +<br />

c'am<br />

Casein<br />

Casein<br />

I<br />

Gram BW<br />

L<br />

Liter CO2/ kg BW/ 24h<br />

350<br />

300<br />

250<br />

200<br />

150<br />

55<br />

45<br />

35<br />

Growth chart<br />

0 10 20 30 40 50<br />

CO 2 eliminated<br />

**<br />

*<br />

SPH SPH +<br />

c'am<br />

Days <strong>of</strong> feeding<br />

Casein<br />

M<br />

kJ/ 24h/ kg BW 0.75<br />

780<br />

720<br />

660<br />

600<br />

SPH SPH +<br />

c'am<br />

Casein<br />

SPH<br />

SPH + c'am<br />

Heat production<br />

**<br />

*<br />

Casein<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

18


Figure 2<br />

A<br />

nmol/ min/ mg prot<br />

2,4<br />

1,6<br />

0,8<br />

iBAT Palm-CoA ox<br />

*<br />

B<br />

Ucp1/ Gtf2b<br />

360<br />

240<br />

120<br />

iBAT Ucp1<br />

**<br />

C<br />

Dio2/ Gtf2b<br />

6<br />

4<br />

2<br />

iBAT Dio2<br />

*<br />

D<br />

Pgc-1α/ Gtf2b<br />

3,9<br />

2,6<br />

1,3<br />

iBAT Pgc-1α<br />

0,0<br />

SPH<br />

Casein<br />

0<br />

SPH<br />

Casein<br />

0<br />

SPH<br />

Casein<br />

0,0<br />

SPH<br />

Casein<br />

E<br />

Relative to casein<br />

3,6<br />

2,4<br />

1,2<br />

0<br />

**<br />

WAT Ucp1<br />

iWAT eWAT MeWAT<br />

F<br />

Relative to casein<br />

7,5<br />

5,0<br />

2,5<br />

0,0<br />

WAT Dio2<br />

*<br />

*<br />

*<br />

iWAT eWAT MeWAT<br />

G<br />

Relative to casein<br />

2,4<br />

1,6<br />

0,8<br />

0<br />

WAT Pgc-1α<br />

* *<br />

iWAT eWAT MeWAT<br />

SPH<br />

Casein<br />

SPH<br />

Casein<br />

SPH<br />

Casein<br />

H<br />

Gram<br />

K<br />

30<br />

20<br />

10<br />

0<br />

Abdominal fat<br />

**<br />

SPH SPH +<br />

c'am<br />

Casein<br />

I<br />

Relative to casein<br />

eWAT Palm-CoA ox<br />

1,4<br />

1,2<br />

1,0<br />

0,8<br />

SPH SPH +<br />

c'am<br />

eWAT adipocyte cell size<br />

SPH SPH + c'am Casein<br />

Casein<br />

J<br />

Rel expression<br />

9<br />

6<br />

3<br />

0<br />

L<br />

eWAT gene-expression<br />

Ucp1 Dio2 Pgc-1a<br />

Relative to control<br />

1,8<br />

1,2<br />

0,6<br />

0<br />

In vitro adipocyte Palm-CoA<br />

oxidation<br />

SPH<br />

SPH + c'am<br />

Casein<br />

* ** *<br />

0 25 75 200<br />

Taurocholic acid, µmol/L<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

19


Figure 3<br />

A<br />

KEGG Pathway<br />

>2 fold Changed<br />

in this pathway<br />

Total expressed<br />

genes in this<br />

pathway<br />

Fisher P-<br />

value<br />

Genes induced by SPH treatment<br />

Type I diabetes mellitus 6 19 0,015 RT1-Aw2, RT1-A3, RT1-T24-1, -<br />

RT1-CE2, RT1-CE15, RT1-CE10<br />

Genes repressed by SPH<br />

treatment<br />

PPAR signaling pathway 10 49 0,024 Cd36, Fabp5, Fabp2, Cpt1a, Angptl4, Cyp7a1, Apoa2, Scd1<br />

Fads2, Me1<br />

Cell adhesion molecules (CAMs) 10 50 0,027 RT1-Aw2, RT1-A3, RT1-T24-1, RT1-CE2, Ptprm, Igsf4a, Jam3<br />

RT1-CE15, RT1-CE10, Cldn1<br />

Nicotinate and nicotinamide metabolism 4 11 0,031 - Enpp3, Enpp2, Pbef1, Aox1<br />

Bile acid biosynthesis 5 18 0,038 Hadhb Adh7, Adh1, Srd5a1, Cyp7a1<br />

Sulfur metabolism 3 7 0,044 - Smp2a, Sult2a1, Sult2a2_predicted<br />

Antigen processing and presentation 7 35 0,058 RT1-Aw2,RT1-A3,RT1-T24-1,RT1-CE2, Klrd1<br />

RT1-CE15,RT1-CE10<br />

Terpenoid biosynthesis 2 3 0,058 Fdps, Fdft1 -<br />

Jak-STAT signaling pathway 8 47 0,085 Akt2, Socs2, Stat3, Stat5b, Stam2, Ghr Il6ra, RGD1560373_predicted<br />

Hematopoietic cell lineage 5 25 0,100 Itga5, Itga5_mapped, Cd36, Thpo Il6ra, Cd1d1<br />

B<br />

Signal log ratio < -2 -2,0 -1,5 -1,0 -0,5 0.5 1.0 1,5 2,0 > 2<br />

Function<br />

Gene<br />

Cellular lipid import<br />

Fatty acid binding<br />

cd36 antigen<br />

lipase, hepatic<br />

low density lipoprotein receptor<br />

low density lipoprotein receptor-related protein 3<br />

low density lipoprotein receptor-related protein 4<br />

low density lipoprotein receptor-related protein associated protein 1<br />

very low density lipoprotein receptor<br />

<strong>fatty</strong> acid binding protein 2, intestinal<br />

<strong>fatty</strong> acid binding protein 4, adipocyte<br />

<strong>fatty</strong> acid binding protein 5, epidermal<br />

<strong>fatty</strong> acid binding protein 6, ileal (gastrotropin)<br />

Fatty acid activation acyl-CoA synthetase long-chain family member 3<br />

acyl-CoA synthetase long-chain family member 4<br />

acyl-CoA synthetase long-chain family member 5<br />

acyl-CoA synthetase long-chain family member 6<br />

Mitochondrial <strong>fatty</strong> acid oxidation<br />

carnitine palmitoyltransferase 1, liver<br />

carnitine palmitoyltransferase 1b<br />

solute carrier family 25 (carnitine/acylcarnitine translocase), member 20<br />

carnitine palmitoyltransferase 2<br />

acyl-Coenzyme A dehydrogenase, short/branched chain<br />

acyl-Coenzyme A dehydrogenase, very long chain<br />

enoyl coenzyme A hydratase 1, peroxisomal/mitochondrial<br />

HADH (trifunctional protein), alpha subunit<br />

HADH (trifunctional protein), beta subunit<br />

mitochondrial acyl-CoA thioesterase 1<br />

dodecenoyl-coenzyme A delta isomerase<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

Krebs/ TCA-cycle<br />

citrate synthase<br />

aconitase 2, mitochondrial<br />

isocitrate dehydrogenase 3, gamma<br />

malate dehydrogenase, mitochondrial<br />

pyruvate dehydrogenase kinase, isoenzyme 2<br />

pyruvate dehydrogenase kinase, isoenzyme 4<br />

Ketone body metabolism acetyl-coenzyme A acetyltransferase 1<br />

3-hydroxy-3-methylglutaryl CoA lyase<br />

20


Figure 4<br />

A<br />

6<br />

Liver Pparα<br />

B<br />

21<br />

Liver mRNA levels<br />

C<br />

2,1<br />

Liver Ppar β/δ<br />

**<br />

D<br />

33<br />

Liver Adrp<br />

**<br />

Ppar α/ Gtf2b<br />

4<br />

2<br />

*<br />

Rel expression<br />

14<br />

7<br />

*<br />

SPH<br />

Casein<br />

Ppar β/δ/ Gtf2b<br />

1,4<br />

0,7<br />

Adrp/ Gtf2b<br />

22<br />

11<br />

0<br />

SPH<br />

Casein<br />

0<br />

Cpt-1a<br />

Acox1<br />

0,0<br />

SPH<br />

Casein<br />

0<br />

SPH<br />

Casein<br />

E<br />

nmol/ mg prot/ min<br />

Liver mit CPT-1<br />

5,1<br />

3,4<br />

1,7<br />

0,0<br />

*<br />

SPH<br />

Casein<br />

F<br />

Acc2/ Gtf2b<br />

0,9<br />

0,6<br />

0,3<br />

0,0<br />

Liver Acc2<br />

**<br />

SPH<br />

Casein<br />

G<br />

% inhibition<br />

CPT-1 inhibition by<br />

malonyl-CoA<br />

0<br />

-15<br />

-30<br />

-45<br />

SPH<br />

Casein<br />

H<br />

mmol/ L<br />

Plasma OH-butyrate<br />

*<br />

1,5<br />

1,0<br />

0,5<br />

0,0<br />

Fasted<br />

Fed<br />

SPH<br />

Casein<br />

I<br />

mg/ tissue<br />

M<br />

Regression coefficient<br />

270<br />

180<br />

90<br />

0<br />

40<br />

30<br />

20<br />

10<br />

0<br />

-10<br />

Liver TAG<br />

**<br />

SPH<br />

(R 2 =0.92)<br />

Casein<br />

J<br />

Liver Oil red-O<br />

staining<br />

SPH<br />

Casein<br />

Fed plasma metabolomic by NMR<br />

Unassigned peaks<br />

(not in casein)<br />

Tyr<br />

Lipid (unsaturated)<br />

Choline<br />

Creatine<br />

Pyruvate or oxalacetate<br />

K<br />

mmol/ L<br />

3<br />

2<br />

1<br />

0<br />

Plasma TAG<br />

Fasted<br />

*<br />

Lipid (mainly HDL or LDL)<br />

Fed<br />

SPH<br />

Casein<br />

L<br />

Vldlr/ Gtf2b<br />

N<br />

mmol/ L<br />

O<br />

36<br />

24<br />

12<br />

2,4<br />

1,6<br />

0,8<br />

0,0<br />

0,9<br />

0<br />

Liver Vldlr<br />

*<br />

SPH<br />

Fed plasma<br />

cholesterol<br />

Total<br />

HDL<br />

Casein<br />

Fed plasma<br />

cholesterol<br />

SPH<br />

Casein<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

-20<br />

-30<br />

Glycoprotein<br />

Lipid (mainly VLDL)<br />

mmol/ L<br />

0,6<br />

0,3<br />

SPH<br />

Casein<br />

-40<br />

9 8 7 6 5 4 3 2 1<br />

Chemical shift (ppm)<br />

0,0<br />

LDL<br />

HDL/total<br />

P<br />

mg/ tissue<br />

240<br />

160<br />

80<br />

0<br />

Liver TAG<br />

SPH SPH +<br />

c'am<br />

Casein<br />

Q<br />

mmol/ L<br />

Fed plasma TAG<br />

4,8<br />

3,2<br />

1,6<br />

0,0<br />

**<br />

SPH SPH +<br />

c'am<br />

Casein<br />

R<br />

nmol/ mg prot/ min<br />

Liver mit Palm-CoA<br />

oxidation<br />

2,1<br />

1,9<br />

1,7<br />

1,5<br />

SPH SPH +<br />

c'am<br />

Casein<br />

S<br />

mmol/ L<br />

Fed plasma OHbutyrate<br />

0,51<br />

0,34<br />

0,17<br />

0,00<br />

SPH SPH +<br />

c'am<br />

Casein<br />

21


Figure 5<br />

A<br />

Ppar α related<br />

B<br />

Ppar β/δ related<br />

Relative expression<br />

1,8<br />

1,2<br />

0,6<br />

0<br />

* *<br />

Ppar a Acox Mcad<br />

SPH<br />

Casein<br />

Relative expression<br />

2,7<br />

1,8<br />

0,9<br />

0<br />

** *<br />

**<br />

Ppar b/d Adrp Angptl4<br />

SPH<br />

Casein<br />

C<br />

Relative expression<br />

5,1<br />

3,4<br />

1,7<br />

0,0<br />

Fatty acid oxidation and uncoupling<br />

*<br />

*<br />

**<br />

Cpt-1b Ucp2 Ucp3<br />

SPH<br />

Casein<br />

D<br />

Relative expression<br />

2,4<br />

1,6<br />

0,8<br />

0<br />

Ppar β/δ and Ucp3<br />

*<br />

##<br />

Ppar b/d<br />

Ucp3<br />

SPH<br />

SPH+c'am<br />

Casein<br />

Figure 6<br />

A<br />

pmol/ L<br />

D<br />

mmol/ L<br />

81<br />

54<br />

27<br />

0<br />

18<br />

12<br />

6<br />

Fed state hormones<br />

*<br />

Insulin Glucagon<br />

Plasma glucose<br />

**<br />

SPH<br />

Casein<br />

SPH<br />

Casein<br />

B<br />

E<br />

Pck1/ Gtf2b<br />

mmol/ L<br />

1,5<br />

1,0<br />

0,5<br />

0,0<br />

12,6<br />

8,4<br />

4,2<br />

Liver Pck1<br />

*<br />

C<br />

mg/ g liver<br />

Liver glycogen<br />

105<br />

70<br />

35<br />

**<br />

0<br />

SPH Casein<br />

SPH Casein<br />

Fed plasma glucose<br />

*<br />

SPH<br />

SPH + c'am<br />

Casein<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

0<br />

Fasted<br />

Fed<br />

0,0<br />

SPH<br />

SPH + c'am Casein<br />

22


Figure 7<br />

A<br />

Plasma ALAT<br />

B<br />

Liver Ucp2<br />

C<br />

Liver cholesterol<br />

U/ L<br />

39<br />

26<br />

13<br />

0<br />

**<br />

SPH<br />

Casein<br />

Relative expression<br />

1,2<br />

0,8<br />

0,4<br />

0<br />

SPH<br />

Casein<br />

mg/ tissue<br />

36<br />

24<br />

12<br />

0<br />

**<br />

Cholesterol<br />

**<br />

Sterol esters<br />

SPH<br />

Casein<br />

D<br />

Relative expression<br />

F<br />

Relative expression<br />

1,2<br />

0,8<br />

0,4<br />

0<br />

1,8<br />

1,2<br />

0,6<br />

0,0<br />

Bile acid synthesis<br />

Cyp7a1<br />

**<br />

Baat<br />

SPH<br />

Casein<br />

Liver expression GSH<br />

synthezising genes<br />

**<br />

Gclc Gclm Gss<br />

SPH<br />

E<br />

Relative expression<br />

Casein<br />

2,7<br />

1,8<br />

0,9<br />

0<br />

G<br />

µmol/ mg protein<br />

Apical hepatocyte transporters<br />

Abcb11/<br />

Bsep<br />

0,18<br />

0,12<br />

0,06<br />

0,00<br />

Abcc2 /<br />

Mrp2<br />

Liver GSH<br />

*<br />

SPH<br />

**<br />

Abcb4/<br />

Mdr 2<br />

Casein<br />

Abcb1b/<br />

Mdr1<br />

H<br />

µmol/ 5 days<br />

51<br />

34<br />

17<br />

0<br />

SPH<br />

Casein<br />

Fecal bile <strong>acids</strong><br />

SPH<br />

Casein<br />

Downloaded from www.jbc.org by guest, on July 7, 2011<br />

23


Research<br />

Persistent Organic Pollutant Exposure Leads to Insulin Resistance Syndrome<br />

Jérôme Ruzzin, 1 Rasmus Petersen, 2,3 Emmanuelle Meugnier, 4 Lise Madsen, 1,3 Erik-Jan Lock, 1 Haldis Lillefosse, 1<br />

Tao Ma, 2,3 Sandra Pesenti, 4 Si Brask Sonne, 3 Troels Torben Marstrand, 5 Marian Kjellevold Malde, 1 Zhen-Yu Du, 1<br />

Carine Chavey, 6 Lluis Fajas, 6 Anne-Katrine Lundebye, 1 Christian Lehn Brand, 7 Hubert Vidal, 4 Karsten Kristiansen, 3<br />

and Livar Frøyland 1<br />

1 National Institute <strong>of</strong> Nutrition and Seafood Research (NIFES), Bergen, Norway; 2 Department <strong>of</strong> Biochemistry and Molecular Biology,<br />

University <strong>of</strong> Southern Denmark, Odense, Denmark; 3 Department <strong>of</strong> Biology, University <strong>of</strong> Copenhagen, Copenhagen, Denmark;<br />

4 INSERM U-870, INRA (Institut national de la recherche agronomique) U-1235, Lyon 1 University, Institut National des Sciences<br />

Appliquées de Lyon and and Hospices Civils de Lyon, Oullins, France; 5 <strong>The</strong> Bioinformatics Centre, Department <strong>of</strong> Biology and Biotech<br />

Research and Innovation Centre, University <strong>of</strong> Copenhagen, Copenhagen, Denmark; 6 Institut de Recherche en Cancérologie de<br />

Montpellier, INSERM U896 Metabolism and Cancer Laboratory, Montpellier, France; 7 Department <strong>of</strong> Insulin Pharmacology, Histology<br />

and Delivery, Novo Nordisk A/S, Maaloev, Denmark<br />

Bac k g r o u n d: <strong>The</strong> incidence <strong>of</strong> the insulin resistance syndrome has increased at an alarming rate<br />

worldwide, creating a serious challenge to public health c<strong>are</strong> in the 21st century. Recently, epidemiological<br />

studies have associated the prevalence <strong>of</strong> type 2 diabetes with elevated body burdens <strong>of</strong><br />

persistent organic pollutants (POPs). However, experimental evidence demonstrating a causal link<br />

between POPs and the development <strong>of</strong> insulin resistance is lacking.<br />

Objective: We investigated whether exposure to POPs contributes to insulin resistance and metabolic<br />

disorders.<br />

Met h o d s: Sprague-Dawley rats were exposed for 28 days to lipophilic POPs through the consumption<br />

<strong>of</strong> a high-fat diet containing either refined or crude fish oil obtained from farmed Atlantic<br />

salmon. In addition, differentiated adipocytes were exposed to several POP mixtures that mimicked<br />

the relative abundance <strong>of</strong> organic pollutants present in crude salmon oil. We measured body weight,<br />

whole-body insulin sensitivity, POP accumulation, lipid and glucose homeostasis, and gene expression<br />

and we performed micro array analysis.<br />

Res u l t s: Adult male rats exposed to crude, but not refined, salmon oil developed insulin resistance,<br />

abdominal obesity, and hepatosteatosis. <strong>The</strong> contribution <strong>of</strong> POPs to insulin resistance was<br />

confirmed in cultured adipocytes where POPs, especially organochlorine pesticides, led to robust<br />

inhibition <strong>of</strong> insulin action. Moreover, POPs induced down-regulation <strong>of</strong> insulin-induced gene-1<br />

(Insig-1) and Lpin1, two master regulators <strong>of</strong> lipid homeostasis.<br />

Con c l u s i o n: Our findings provide evidence that exposure to POPs commonly present in food<br />

chains leads to insulin resistance and associated metabolic disorders.<br />

Key w o r d s : contaminants, farmed salmon, metabolic syndrome, nonalcoholic <strong>fatty</strong> liver,<br />

obesity, pollution, public health, type 2 diabetes. Environ Health Perspect 118:465–471<br />

(2010). doi:10.1289/ehp.0901321 [Online 19 November 2009]<br />

factors can only partly explain the worldwide<br />

explosive prevalence <strong>of</strong> insulin resistance–<br />

associated metabolic diseases. We therefore<br />

sought to elucidate whether the exposure to<br />

POPs present in a food matrix could contribute<br />

to insulin resistance and metabolic<br />

disorders.<br />

POPs accumulate in the lipid fraction <strong>of</strong><br />

fish, and fish consumption represents a source<br />

<strong>of</strong> POP exposure to humans (Dougherty et al.<br />

2000; Hites et al. 2004; Schafer and Kegley<br />

2002). <strong>The</strong>refore, certain European countries<br />

have dietary recommendations to limit the<br />

consumption <strong>of</strong> <strong>fatty</strong> fish per week (Scientific<br />

Advisory Committee on Nutrition 2004).<br />

On the other hand, n-3 <strong>polyunsaturated</strong> <strong>fatty</strong><br />

<strong>acids</strong> present in fish oil have a wide range <strong>of</strong><br />

beneficial <strong>effects</strong> (Jump 2002), including protection<br />

against high-fat (HF) diet– induced<br />

insulin resistance (Storlien et al. 1987).<br />

Accordingly, we fed rats an HF diet containing<br />

either crude (HFC) or refined (HFR) fish<br />

oil obtained from farmed Atlantic salmon and<br />

Despite international agreements intended to<br />

limit the release <strong>of</strong> persistent organic pollutants<br />

(POPs) such as organochlorine pesticides,<br />

polychlorinated biphenyls (PCBs),<br />

polychlorinated dibenzo-p-dioxins (PCDDs),<br />

and polychlorinated dibenz<strong>of</strong>urans (PCDFs),<br />

POPs still persist in the environment and food<br />

chains (Atlas and Giam 1981; Dougherty et al.<br />

2000; Fisher 1999; Jorgenson 2001; Schafer<br />

and Kegley 2002; Van den Berg 2009). Most<br />

human populations <strong>are</strong> exposed to POPs<br />

through consumption <strong>of</strong> fat-containing food<br />

such as fish, dairy products, and meat (Fisher<br />

1999). Humans bioaccumulate these lipophilic<br />

and hydrophobic pollutants in <strong>fatty</strong> tissues<br />

for many years because POPs <strong>are</strong> highly<br />

resistant to metabolic degradation (Fisher<br />

1999; Kiviranta et al. 2005). <strong>The</strong> physiological<br />

impact associated with chronic exposure<br />

to low doses <strong>of</strong> different mixtures <strong>of</strong> POPs is<br />

poorly understood, but epidemiological studies<br />

have reported that Americans, Europeans, and<br />

Asian patients with type 2 diabetes accumulated<br />

greater body burdens <strong>of</strong> POPs, including<br />

2,3,7,8‐tetrachlorodibenzo-p-dioxin (TCDD),<br />

2,2´,4,4´,5,5´-hexachlorobiphenyl (PCB153),<br />

coplanar PCBs (PCB congeners 77, 81,<br />

126, and 169), p,p´-diphenyldichloroethene<br />

(DDE), oxychlordane, and trans-nonachlor<br />

(Fierens et al. 2003; Henriksen et al. 1997;<br />

Lee et al. 2006; Rignell-Hydbom et al. 2007;<br />

Turyk et al. 2009; Wang et al. 2008).<br />

<strong>The</strong> incidences <strong>of</strong> type 2 diabetes and the<br />

insulin resistance syndrome have increased at<br />

a globally alarming rate, and > 25% <strong>of</strong> adults<br />

in the United States have been estimated to<br />

be affected by metabolic abnormalities associated<br />

with insulin resistance (Ford et al. 2004).<br />

Impaired insulin action is a central dysfunction<br />

<strong>of</strong> the insulin resistance syndrome characterized<br />

by abdominal obesity and defects in<br />

both lipid and glucose homeostasis, increasing<br />

the risk for developing type 2 diabetes,<br />

cardio vascular diseases, non alcoholic <strong>fatty</strong><br />

liver disease, polycystic ovarian disease, and<br />

certain types <strong>of</strong> cancer (Biddinger and Kahn<br />

2006; Reaven 2005). Although a sedentary<br />

lifestyle and consumption <strong>of</strong> high-fat food<br />

<strong>are</strong> considered major contributors to insulin<br />

resistance and obesity, these conventional risk<br />

Address correspondence to J. Ruzzin, National<br />

Institute <strong>of</strong> Nutrition and Seafood Research, N-5817<br />

Bergen, Norway. Telephone: (0047) 41450448. Fax:<br />

(0047) 559052 99. E-mail: jerome.ruzzin@nifes.no<br />

Supplemental Material is available online<br />

(doi:10.1289/ehp.0901321 via http://dx.doi.org/).<br />

We thank J. Burén for technical assistance on primary<br />

adipocyte investigations, Å. Heltveit for animal<br />

c<strong>are</strong>, J.I. Hjelle for analysis <strong>of</strong> hepatic lipids,<br />

J. Wessel for assistance on tissue harvest, K. Heggstad<br />

for his expertise on contaminant analysis, J. Rieusset<br />

for advice on histology investigation, and C. Debard<br />

for real-time PCR analysis. We thank T. Fenchel,<br />

P. Grandjean, and E. Lund for discussions and comments<br />

on the manuscript.<br />

This work was supported by grants from the<br />

European Research Council, the Research Council<br />

<strong>of</strong> Norway, the Danish Natural Science Research<br />

Council, <strong>The</strong> Diabetes Association, and the Novo<br />

Nordisk Foundation.<br />

C.L.B., employed at Novo Nordisk (a leading<br />

manu facturer <strong>of</strong> insulin analogs and diabetes management<br />

c<strong>are</strong> products), holds sh<strong>are</strong>s in the company,<br />

and contributed in<strong>dependent</strong>ly with the clamp studies.<br />

<strong>The</strong> Novo Nordisk Foundation supports basic science<br />

in<strong>dependent</strong>ly <strong>of</strong> the company interests <strong>of</strong> Novo<br />

Nordisk A/S. <strong>The</strong> remaining authors decl<strong>are</strong> they have<br />

no competing financial interest.<br />

Received 11 August 2009; accepted 19 November<br />

2009.<br />

Environmental Health Perspectives • v o l u m e 118 | number 4 | April 2010 465


Ruzzin et al.<br />

investigated the metabolic impacts <strong>of</strong> POPs<br />

and their ability to interfere with n-3 <strong>polyunsaturated</strong><br />

<strong>fatty</strong> <strong>acids</strong>.<br />

Materials and Methods<br />

Tissue RNA from liver <strong>of</strong> rats fed HFC and<br />

HFR was extracted using Trizol, and microarray<br />

analysis was performed using the Operon<br />

Rat Oparray. Levels <strong>of</strong> specific mRNA were<br />

quantified using real-time polymerase chain<br />

reaction (PCR) as described previously (Rome<br />

et al. 2008). 3T3-L1 cells were exposed to<br />

different POP mixtures, and we measured<br />

insulin-stimulated glucose uptake and mRNA<br />

expression <strong>of</strong> target genes. Details <strong>of</strong> the<br />

methods <strong>are</strong> available in the Supplemental<br />

Material (doi:10.1289/ehp.0901321).<br />

Animals. All experimental protocols<br />

described below were approved by the<br />

Norwegian State Board <strong>of</strong> Biological<br />

Experiments with Living Animals, and the<br />

animals were treated humanely and with<br />

regard for alleviation <strong>of</strong> suffering. Male<br />

Sprague-Dawley rats (Taconic, Ry, Denmark)<br />

weighing 200–250 g were housed with a<br />

12-hr light/dark cycle and with free access<br />

to food and tap water. Animals were fed a<br />

standard diet (chow; 17% fat-derived calories,<br />

3.4 kcal/g) or an HF diet (65% fatderived<br />

calories, 5.5 kcal/g) for 28 days<br />

(Lavigne et al. 2001). Two additional HF<br />

diets were made by substituting corn oil (20%<br />

wt/wt) with either crude or refined salmon<br />

oil. Crude salmon oil was obtained by heating<br />

the rest raw material <strong>of</strong> farmed Atlantic<br />

salmon to 92°C and separating oil from water<br />

and solid material. Refined salmon oil was<br />

obtained by bleaching, carbon filtering, and<br />

deodorizing the crude oil. HF, HFC, and<br />

HFR diets were supplemented with cellulose<br />

(50 g/kg), choline bitartrate (2 g/kg),<br />

American Institute <strong>of</strong> Nutrition (AIN) vitamin<br />

mixture 76 (14 g/kg), and AIN mineral<br />

mixture 76 (67 g/kg) (MP Biochemicals,<br />

Inc, Illrich, France) to meet the daily nutrient<br />

requirement levels <strong>of</strong> adult rats (Reeves et al.<br />

1993). Fatty acid composition <strong>of</strong> HF, HFC,<br />

and HFR diets was analyzed as previously<br />

described (Jordal et al. 2007).<br />

Hepatic lipids. We determined levels <strong>of</strong><br />

triacylglycerol, diacyl glycerol, and total cholesterol<br />

in frozen liver samples <strong>of</strong> overnight-fasted<br />

rats using high-performance thin-layer chromatography<br />

as described previously (Berntssen<br />

et al. 2005). Frozen (O.C.T. compound;<br />

Sakura Finetek Europe, Zoeterwoude, the<br />

Netherlands) and fixed (paraffin-embedded)<br />

liver sections were stained with Oil red O and<br />

hematoxylin and eosin (H&E), respectively.<br />

Determination <strong>of</strong> POP levels. We measured<br />

levels <strong>of</strong> POPs as described previously<br />

(Berntssen et al. 2005; Julshamn et al. 2004).<br />

Determination <strong>of</strong> insulin action in peripheral<br />

tissues. We used soleus muscles and<br />

epididymal fat <strong>of</strong> overnight-fasted animals<br />

to assess insulin-stimulated glucose uptake<br />

as described previously (Buren et al. 2002;<br />

Ruzzin et al. 2005).<br />

Hyperinsulinemic–euglycemic clamps.<br />

Animals were catheterized, and hyperinsulinemic–euglycemic<br />

clamps were performed<br />

7 days later (Brand et al. 2003; Ruzzin<br />

et al. 2007). After a 6-hr fasting period,<br />

conscious unrestrained catheterized animals<br />

were infused with a prime (6 µCi) continuous<br />

(0.1 µCi/min for basal; 0.17 µCi/min for<br />

clamp) infusion <strong>of</strong> [3- 3 H]glucose from –90 to<br />

120 min for assessment <strong>of</strong> whole-body glucose<br />

disappearance (R d ) and appearance (R a ) using<br />

Steele’s non–steady-state equations (Steele<br />

et al. 1956). <strong>The</strong> hyperinsulinemic–euglycemic<br />

clamp was performed (0–120 min) by a<br />

continuous infusion <strong>of</strong> human insulin (3 mU/<br />

kg/min) (Actrapid, Novo Nordisk, Bagsvaerd,<br />

Denmark), and euglycemia (~ 115 mg/dL)<br />

was maintained by variable infusion rates <strong>of</strong><br />

a 20% non labeled glucose solution [glucose<br />

infusion rate (GIR)]. At the end <strong>of</strong> the clamp,<br />

rats were given a lethal dose <strong>of</strong> pento barbital<br />

sodium; liver, epididymal fat, and gastrocnemius<br />

muscles were removed, frozen in liquid<br />

nitrogen, and stored at –80°C for determination<br />

<strong>of</strong> POP levels. Plasma glucose and insulin<br />

levels were analyzed by the glucose oxidase<br />

method (YSI 2300 STAT Plus glucose analyzer;<br />

YSI Incorporated, Yellow Spring, OH,<br />

USA) and an enzyme-linked immunosorbent<br />

assay kit (DRG Instruments, Marburg,<br />

Germany), respectively. To determine plasma<br />

[3- 3 H]glucose, plasma was deproteinized,<br />

dried to remove tritiated water, resuspended<br />

in water, and counted in biodegradable scintillation<br />

fluid (Nerliens Meszansky, Oslo,<br />

Norway) on a beta scintillation counter (Tri-<br />

Carb 1900TR; Packard, Meriden, CT, USA).<br />

All samples were run in duplicate. Hepatic<br />

glucose production (HGP) was calculated as<br />

tracer-determined R a minus GIR.<br />

Insulin resistance was further assessed by<br />

the homeostasis model assessment <strong>of</strong> insulin<br />

resistance (HOMA-IR) index as described by<br />

Lee et al. (2008).<br />

Cultured adipocyte studies. We used cultured<br />

and differentiated 3T3-L1 cells (Petersen<br />

et al. 2008) to assess insulin-stimulated glucose<br />

uptake and mRNA expression <strong>of</strong> target genes.<br />

On day 8 <strong>of</strong> the differentiation program, cells<br />

were exposed to vehicle (dimethyl sulfoxide)<br />

or POP mixtures for 48 hr, and glucose uptake<br />

was assessed.<br />

Cytotoxicity. Membrane integrity <strong>of</strong><br />

POP-treated adipocytes was determined<br />

by the release <strong>of</strong> lactate dehydrogenase into<br />

cell medium by a Tox7 kit (Sigma-Aldrich,<br />

Leirdal, Norway).<br />

Statistical analysis. We examined differences<br />

between groups for statistical significance<br />

using analysis <strong>of</strong> variance (ANOVA)<br />

with the least-squ<strong>are</strong> difference post hoc<br />

test. We used one-class statistical analysis <strong>of</strong><br />

microarray to identify differentially expressed<br />

genes (Tusher et al. 2001) between HFC- and<br />

HFR-fed rats. We determined statistical significance<br />

<strong>of</strong> the real-time PCR results using<br />

the Student’s t-test, and the threshold for significance<br />

was set at p ≤ 0.05.<br />

Results<br />

Characteristics <strong>of</strong> animals exposed to POPs.<br />

As we expected, concentrations <strong>of</strong> POPs were<br />

consistently much higher in the HFC diet<br />

than in the HFR diet [Supplemental Material,<br />

Table 1 (doi:10.1289/ehp.0901321)], whereas<br />

the contents <strong>of</strong> n-3 <strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong><br />

and other <strong>fatty</strong> <strong>acids</strong> were similar in the two<br />

diets because both the crude and the refined<br />

fish oils were obtained from the same batch<br />

<strong>of</strong> farmed salmon (Supplemental Material,<br />

Table 2 (doi:10.1289/ehp.0901321).<br />

After 28 days, rats fed the HFC diet<br />

appe<strong>are</strong>d normal, although they tended to<br />

gain more weight than rats fed the HFR<br />

diet despite similar daily energy intake<br />

(Figure 1A,B). Intake <strong>of</strong> the HFC diet, but not<br />

HFR diet, enhanced the accumulation <strong>of</strong> visceral<br />

adipose tissue induced by HF consumption<br />

(Figure 1C,D). Pr<strong>of</strong>ound dys regulation<br />

in lipid homeo stasis was further observed in<br />

livers <strong>of</strong> HFC-fed rats, which exhibited elevated<br />

levels <strong>of</strong> triacyl glycerol, diacyl glycerol,<br />

and total cholesterol comp<strong>are</strong>d with HF-fed<br />

rats; livers <strong>of</strong> HFR-fed rats tended to exhibit<br />

a reduced lipid accumulation (Figure 1E–G).<br />

Histological examinations highlighted the<br />

development <strong>of</strong> severe hepatosteatosis in rats<br />

fed HFC (Figure 1H) and confirmed that the<br />

presence <strong>of</strong> POPs in salmon oil provokes significant<br />

impairment <strong>of</strong> lipid metabolism.<br />

To gain further insight into the phenotypical<br />

changes <strong>of</strong> animals exposed to POPs,<br />

we performed a comparison <strong>of</strong> gene expression<br />

pr<strong>of</strong>iles in the liver <strong>of</strong> rats fed the HFC<br />

and HFR diets, using oligonucleo tide microarrays.<br />

<strong>The</strong> expression <strong>of</strong> genes involved in<br />

drug metabo lism was affected, indicating<br />

dietary POP exposure [Supplemental Material,<br />

Table 3 (doi:10.1289/ehp.0901321)]. We<br />

also observed major differences for genes<br />

involved in lipid metabo lism and for several<br />

genes linked to lipid deposition (Supplemental<br />

Material, Table 3). Of interest, POPs induced<br />

robust down-regulation <strong>of</strong> insulin-induced<br />

gene-1 (Insig-1) and Lpin1, two master regulators<br />

<strong>of</strong> lipo genesis and synthesis <strong>of</strong> triglyceride<br />

and cholesterol (Croce et al. 2007;<br />

Engelking et al. 2004; Finck et al. 2006; Lee<br />

and Ye 2004). Real-time PCR analysis confirmed<br />

the strong repression <strong>of</strong> Lpin 1 and<br />

Insig-1 genes in the liver <strong>of</strong> rats consuming the<br />

HFC diet (Table 1). Similarly, in adipose tissue<br />

<strong>of</strong> HFC-fed rats, expression <strong>of</strong> Lpin1 and<br />

Insig-1 genes was repressed comp<strong>are</strong>d with<br />

466 v o l u m e 118 | number 4 | April 2010 • Environmental Health Perspectives


POP exposure provokes insulin resistance<br />

HFR-fed animals [mean ± SE, 78 ± 8 vs. 55 ±<br />

5 (n = 9, p = 0.02) for Insig-1 and 98 ± 11 vs.<br />

64 ± 8 (n = 9, p = 0.03) for Lpin1 for HFRand<br />

HFC-fed rats, respectively]. Furthermore,<br />

POPs induced a significant increase in the<br />

expression level <strong>of</strong> SREBP1C (sterol regulatory<br />

element-binding protein 1C), the master<br />

regulator <strong>of</strong> the lipogenic pathway, and<br />

FAS (<strong>fatty</strong> acid synthase), a well-known target<br />

gene <strong>of</strong> SREBP1C (Table 1). Interestingly, the<br />

hepatic expression <strong>of</strong> LXRα (liver X receptor<br />

alpha) was not affected, suggesting that the<br />

oxy sterol pathway was not modified by POP<br />

exposure (Table 1). Altogether, these results<br />

demonstrate that POP exposure signifi cantly<br />

affects the expression <strong>of</strong> critical genes involved<br />

in the regulation <strong>of</strong> lipid homeostasis. Gene<br />

set enrichment analysis further revealed significant<br />

<strong>effects</strong> on several biological pathways<br />

[Supplemental Material, Table 4 (doi:10.1289/<br />

ehp.0901321)]. This analysis demonstrated<br />

a highly significant up-regulation <strong>of</strong> pathways<br />

designated “pathogenic Escherichia coli<br />

infection” (EPEC/EHEC). <strong>The</strong> core genes<br />

up-regulated in the pathways include TLR5,<br />

ROCK2, CD14, and YWHAZ, a gene encoding<br />

a member <strong>of</strong> the 14-3-3 family <strong>of</strong> proteins<br />

reported to interact with insulin receptor substrate-1<br />

and thereby regulating insulin signaling.<br />

Similarly, the roles <strong>of</strong> toll-like receptors,<br />

Body weight gain (g)<br />

200<br />

160<br />

120<br />

80<br />

40<br />

0<br />

*<br />

Chow<br />

HF<br />

HFR<br />

HFC<br />

Energy intake<br />

(kcal/day)<br />

200<br />

160<br />

120<br />

80<br />

40<br />

0<br />

*<br />

* *<br />

Chow<br />

HF<br />

Visceral fat (g)<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

*<br />

*<br />

**<br />

Triacylglycerol<br />

(mg/g)<br />

120<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

*<br />

*<br />

**<br />

Diacylglycerol (mg/g)<br />

1.2<br />

0.8<br />

0.4<br />

0<br />

*<br />

*<br />

Total cholesterol<br />

(mg/g)<br />

**<br />

20<br />

15<br />

**<br />

10<br />

5<br />

0<br />

* *<br />

HFR<br />

HFC<br />

Chow<br />

HF HFR HFC<br />

Oil red O<br />

100 µM<br />

100 µM 100 µM 100 µM<br />

H&E<br />

200 µM<br />

200 µM 200 µM 200 µM<br />

50 µM 50 µM 50 µM 50 µM<br />

Figure 1. Characteristics <strong>of</strong> rats fed salmon oil containing POPs. Body weight gain (A) and daily energy intake (B) in rats fed chow or the HF, HFR, or HFC diets<br />

over a 4-week period. (C) Exposed ventral view <strong>of</strong> a representative rat from each diet group showing increased visceral adipose tissue after consumption <strong>of</strong><br />

the HFC diet. (D) Quantification <strong>of</strong> visceral fat (epididymal and perirenal fat pads). (E–G) Levels <strong>of</strong> hepatic triacylglycerol (E), diacyl glycerol (F), and total cholesterol<br />

(G). (H) Representative histological sections <strong>of</strong> liver stained with Oil red O (top) or H&E at low (middle) and high (bottom) magnifications; the three sections<br />

for each treatment group <strong>are</strong> from the same liver sample. All data <strong>are</strong> shown as mean ± SE; n = 8–9.<br />

*p < 0.02 comp<strong>are</strong>d with control. **p < 0.04 comp<strong>are</strong>d with HF.<br />

Environmental Health Perspectives • v o l u m e 118 | number 4 | April 2010 467


Ruzzin et al.<br />

CD14, and rho kinases in regulating insulin<br />

signaling and establishment <strong>of</strong> insulin resistance<br />

in response to chronic low-grade inflammation<br />

<strong>are</strong> well documented (Begum et al.<br />

2002; Cani et al. 2007; Furukawa et al. 2005;<br />

Petersen et al. 2008; Tzivion et al. 2001).<br />

Effects <strong>of</strong> POPs on insulin action in vivo.<br />

Next, we assessed the impacts <strong>of</strong> POPs on<br />

whole-body insulin action. In the basal state,<br />

intake <strong>of</strong> the HFC diet exacerbated the hyperinsulinemia<br />

induced by HF consumption,<br />

whereas animals fed HFR and control diets<br />

had similar plasma insulin levels (Figure 2A).<br />

Basal plasma glucose levels were similar in<br />

all groups (Figure 2B), but the HOMA-IR<br />

index was significantly increased in rats fed<br />

Table 1. Real-time PCR determination <strong>of</strong> mRNA expression <strong>of</strong> a set <strong>of</strong> relevant genes in the liver <strong>of</strong> rats<br />

fed HFR or HFC diets (n = 9 per group).<br />

HFR HFC p-Value<br />

Genes related to mitochondrial function<br />

PGC1α 0.73 ± 0.3 0.05 ± 0.02 0.043<br />

PPARα (peroxisome proliferator-activated receptor α) 76 ± 7 75 ± 18 0.988<br />

CS (citrate synthase) 316 ± 19 214 ± 10 0.002<br />

SDHA (succinate dehydrogenase) 74 ± 2 63 ± 4 0.038<br />

MCAD (medium chain acyl CoA dehydrogenase) 332 ± 30 170 ± 18 0.003<br />

Genes related to lipogenesis<br />

SREBP1C 3.0 ± 0.3 4.6 ± 0.6 0.021<br />

LXRα 50 ± 3 51 ± 7 0.932<br />

FAS 1.1 ± 0.1 1.9 ± 0.2 0.01<br />

Lpin 1 96 ± 17 22 ± 10 0.0017<br />

Insig-1 123 ± 23 43 ± 12 0.0071<br />

Plasma insulin<br />

(ng/mL)<br />

GIR<br />

(mg/kg/min)<br />

Clamp HGP<br />

(mg/kg/min)<br />

Glucose uptake<br />

(mmol/kg ww/30 min)<br />

3<br />

2<br />

1<br />

0<br />

15<br />

10<br />

5<br />

0<br />

15<br />

10<br />

5<br />

0<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

*<br />

#<br />

#<br />

**<br />

##<br />

0 2,000<br />

#<br />

Insulin (µU/mL)<br />

#<br />

Chow<br />

HF<br />

HFR<br />

HFC<br />

#<br />

Plasma glucose<br />

(mg/dL)<br />

Basal HGP<br />

(mg/kg/min)<br />

Clamp R d<br />

(mg/kg/min)<br />

Glucose uptake<br />

(fmoL/1,000 cells/min)<br />

Figure 2. Effects <strong>of</strong> salmon oil and POPs on insulin action and glucose metabolism evaluated by hyperinsulinemic–euglycemic<br />

clamps performed in rats fed chow or HF, HFR, or HFC diets over a 4-week period.<br />

(A) Basal insulinemia. (B) Basal glycemia. (C) GIR. (D) Basal HGP. (E) HGP during the clamps. (F) Glucose<br />

disposal rate (R d ). (G) Insulin-stimulated glucose uptake in soleus muscles. (H) Insulin-stimulated glucose<br />

uptake in primary adipocytes. All data <strong>are</strong> shown as mean ± SE; n = 6–9.<br />

*p < 0.04 comp<strong>are</strong>d with chow control. **p < 0.04 comp<strong>are</strong>d with HF. # p < 0.05 comp<strong>are</strong>d with HFR. ## p < 0.03 comp<strong>are</strong>d<br />

with HF.<br />

160<br />

120<br />

80<br />

40<br />

0<br />

15<br />

10<br />

5<br />

0<br />

15<br />

10<br />

5<br />

0<br />

8<br />

6<br />

4<br />

2<br />

0<br />

0<br />

#<br />

Insulin (µU/mL)<br />

2,000<br />

##<br />

the HFC diet (7.1 for control rats, 11.2 for<br />

rats fed HF, 8.4 for rats fed HFR, and 15.5<br />

for rats fed HFC; p < 0.04).<br />

<strong>The</strong> performance <strong>of</strong> hyperinsulinemic–​<br />

euglycemic clamp, the gold standard for<br />

investigating and quantifying insulin resistance<br />

(Kraegen et al. 1983), revealed that the<br />

consumption <strong>of</strong> the HFC diet aggravated<br />

HF-induced reduced GIR, whereas HFR-fed<br />

rats showed no impairment <strong>of</strong> insulin action<br />

comp<strong>are</strong>d with control rats (Figure 2C).<br />

Reduced GIR reflects decreased insulinmediated<br />

suppression <strong>of</strong> HGP, reduced insulinstimulated<br />

peripheral glucose disposal rates,<br />

or both. Analysis <strong>of</strong> these parameters revealed<br />

that basal HGP was similar in all groups<br />

(Figure 2D), whereas suppression <strong>of</strong> HGP<br />

by insulin was impaired in animals consuming<br />

both HFC and HF diets (Figure 2E).<br />

Moreover, intake <strong>of</strong> HFC led to impaired<br />

insulin-mediated glucose disposal in peripheral<br />

tissues, which mainly include skeletal muscles<br />

and adipose tissue (Figure 2F). To investigate<br />

this further, we determined the rates <strong>of</strong> glucose<br />

uptake in isolated soleus muscles and primary<br />

adipocytes. We found that insulin-stimulated<br />

glucose uptake was reduced to a similar extent<br />

in skeletal muscle <strong>of</strong> animals fed HFC and<br />

HF diets (Figure 2G). In contrast, rats fed<br />

the HFR diet were protected against muscle<br />

insulin resistance (Figure 2G). In adipose tissue,<br />

the ability <strong>of</strong> insulin to stimulate glucose<br />

uptake was impaired in both the HFR and HF<br />

groups, and this metabolic defect was worsened<br />

by the consumption <strong>of</strong> the HFC diet<br />

(Figure 2H). Thus, exposure to POPs present<br />

in HFC exacerbated the deleterious metabolic<br />

<strong>effects</strong> <strong>of</strong> HF and attenuated the protective<br />

<strong>effects</strong> <strong>of</strong> n-3 <strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong>,<br />

which indicates that the presence <strong>of</strong> environmental<br />

organic contaminants may influence<br />

the outcomes <strong>of</strong> food and dietary products.<br />

<strong>The</strong>re is growing evidence that mitochondrial<br />

dysfunction contributes to insulin resistance<br />

(Lowell and Shulman 2005). To assess<br />

the impact <strong>of</strong> POPs on hepatic mitochondrial<br />

content, we measured mitochondrial<br />

DNA levels by quantitative polymerase chain<br />

reaction (qPCR), using primers specific for<br />

the COXII gene, and determined the ratio<br />

between mitochondrial DNA and nuclear<br />

DNA as previously validated (Bonnard et al.<br />

2008). We found no app<strong>are</strong>nt modification<br />

<strong>of</strong> the amount <strong>of</strong> mitochondrial DNA in the<br />

liver <strong>of</strong> the animals fed HFC (ratio COXII/<br />

PPIA, 1.1 ± 0.2 (mean ± SE) for rats fed HFR<br />

and 0.9 ± 0.1 for rats fed HFC, p = 0.189).<br />

However, despite this app<strong>are</strong>nt lack <strong>of</strong> change<br />

in mitochondrial content, we observed significant<br />

reduction in the expression <strong>of</strong> several<br />

genes related to mitochondrial function,<br />

such as PGC1α (peroxisome proliferatoractivated<br />

receptor gamma-coactivator-1<br />

alpha), citrate synthase, medium-chain acyl<br />

468 v o l u m e 118 | number 4 | April 2010 • Environmental Health Perspectives


POP exposure provokes insulin resistance<br />

CoA dehydrogenase, and SDHA (succinate<br />

dehydrogenase) (Table 1), indicating the presence<br />

<strong>of</strong> alterations in mitochondrial function<br />

and oxidative capacities in the liver <strong>of</strong> the rats<br />

exposed to POPs.<br />

Analysis <strong>of</strong> POPs distribution in these<br />

animals revealed that whereas both liver and<br />

adipose tissue stored organochlorine pesticides,<br />

indicator PCBs, mono-ortho-substituted<br />

PCBs, and non–ortho-substituted PCBs,<br />

the liver preferentially retained PCDDs or<br />

PCDFs [Supplemental Material, Table 5<br />

(doi:10.1289/ehp.0901321)].<br />

Effects <strong>of</strong> POPs on insulin action in vitro.<br />

To further demonstrate the contribution <strong>of</strong><br />

lipophilic POPs to the development <strong>of</strong> insulin<br />

resistance–associated metabolic disturbances,<br />

we exposed differentiated adipocytes<br />

to a POP mixture that mimicked the relative<br />

abundance <strong>of</strong> organic contaminants found<br />

in crude salmon oil. Incubation <strong>of</strong> adipocytes<br />

with this POP mixture impaired the<br />

ability <strong>of</strong> insulin to stimulate glucose uptake<br />

(Figure 3A), which is in agreement with the<br />

reduced insulin–stimulated glucose uptake<br />

observed in adipose tissue <strong>of</strong> rats fed the<br />

HFC diet (Figure 2H). We then determined<br />

whether POP exposure, as observed in rats<br />

fed the HFC diet, could affect the expression<br />

<strong>of</strong> Lpin1 and Insig-1 mRNA in cultured<br />

adipo cytes. After 48-hr treatment with the<br />

POP mixture, Lpin1 and Insig-1 mRNA levels<br />

were dose-<strong>dependent</strong>ly reduced in adipocytes<br />

[Supplemental Material, Figure 1<br />

(doi:10.1289/ehp.0901321)], which confirms<br />

the ability <strong>of</strong> POPs to interfere with key<br />

regulators <strong>of</strong> lipid metabolism. Importantly,<br />

the metabolic defects observed in adipocytes<br />

exposed to POPs were in<strong>dependent</strong> <strong>of</strong> cytotoxicity,<br />

as demonstrated by the absence <strong>of</strong><br />

an increased release <strong>of</strong> lactate dehydrogenase<br />

into the cell culture media (Supplemental<br />

Material, Figure 2). Altogether, these findings<br />

clearly establish the capacity <strong>of</strong> POPs to<br />

impair insulin action and associated metabolic<br />

abnormalities in a cell-autonomous manner.<br />

Humans and other organisms <strong>are</strong> chronically<br />

exposed to a variety <strong>of</strong> organic pollutants.<br />

To investigate which POPs contributed significantly<br />

to the impairment <strong>of</strong> insulin action,<br />

we incubated adipocytes with different POP<br />

mixtures. Although adipocytes exposed to a<br />

PCDD or PCDF mixture showed normal<br />

insulin action (Figure 3B,C), those exposed<br />

to non-ortho- substituted and mono-orthosubstituted<br />

PCB mixtures had reduced insulin<br />

action (Figure 3D,E). Impaired insulin action<br />

was in<strong>dependent</strong> <strong>of</strong> the total toxic equivalent<br />

(TEQ) concentration (Van den Berg et al.<br />

2006) <strong>of</strong> the mixtures; up to 6.027ng WHO<br />

2005 TEQ/mL for the PCDF mixture comp<strong>are</strong>d<br />

with 0.0016ng WHO 2005 TEQ/mL<br />

for the mono-ortho-PCB mixture. <strong>The</strong>se findings<br />

demon strate that risk assessment based<br />

on TEQ assigned to dioxins and dioxin-like<br />

PCBs (Van den Berg et al. 2006) is unlikely<br />

to reflect the risk <strong>of</strong> insulin resistance. Further<br />

investigations showed that insulin-stimulated<br />

glucose uptake was dramatically reduced in<br />

adipocytes treated with both the mixture <strong>of</strong><br />

organochlorine pesticides (Figure 3F) and<br />

dichloro diphenyl trichloroethanes (DDTs)<br />

(Figure 3G), whereas the mixture <strong>of</strong> indicator<br />

PCBs had less inhibitory <strong>effects</strong> on insulin<br />

action (Figure 3H).<br />

Relative glucose uptake<br />

(fold increase)<br />

Relative glucose uptake<br />

(fold increase)<br />

Relative glucose uptake<br />

(fold increase)<br />

Relative glucose uptake<br />

(fold increase)<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

POPs<br />

PCDFs<br />

Vehicle<br />

1 nM<br />

10 nM<br />

100 nM<br />

* *<br />

TEQ (ng WHO<br />

2005 TEQ/mL)<br />

0.060<br />

0.603<br />

6.027<br />

Mono-ortho-PCBs<br />

TEQ (ng WHO<br />

2005 TEQ/mL)<br />

0.0002<br />

0.0016<br />

1.0158<br />

* *<br />

* * *<br />

* * *<br />

*<br />

* *<br />

* *<br />

*<br />

*<br />

Discussion<br />

In this study, we demonstrate for the first time<br />

a causal relationship between POPs and insulin<br />

resistance in rats. In vivo, chronic exposure to<br />

low doses <strong>of</strong> POPs commonly found in food<br />

chains induced severe impairment <strong>of</strong> wholebody<br />

insulin action and contributed to the<br />

development <strong>of</strong> abdominal obesity and hepatosteatosis.<br />

Treatment in vitro <strong>of</strong> differentiated<br />

adipocytes with nano molar concentrations <strong>of</strong><br />

POP mixtures mimicking those found in crude<br />

7<br />

6<br />

5<br />

PCDDs<br />

TEQ (ng WHO<br />

2005 TEQ/mL)<br />

0.004<br />

0.043<br />

* * *<br />

4 0.425<br />

3<br />

2<br />

1<br />

0<br />

7 Non-ortho-PCBs<br />

TEQ (ng WHO<br />

6<br />

*<br />

2005 TEQ/mL)<br />

5 0.010<br />

0.103<br />

4 1.025<br />

3<br />

* * *<br />

2<br />

*<br />

*<br />

1<br />

* *<br />

* *<br />

0<br />

Relative glucose uptake<br />

(fold increase)<br />

Relative glucose uptake<br />

(fold increase)<br />

Relative glucose uptake<br />

(fold increase)<br />

DDTs<br />

7<br />

PCBs<br />

Vehicle<br />

Vehicle<br />

1 nM<br />

6<br />

10 nM<br />

10 nM<br />

5<br />

100 nM<br />

100 nM<br />

1 µM<br />

4<br />

3<br />

*<br />

*<br />

*<br />

*<br />

*<br />

2<br />

*<br />

*<br />

*<br />

* *<br />

* 1<br />

*<br />

* * ** * *<br />

0<br />

0 1 3 10 30<br />

0 1 3 10 30<br />

Insulin (nM)<br />

*<br />

* * *<br />

*<br />

*<br />

*<br />

Relative glucose uptake<br />

(fold increase)<br />

Figure 3. Effects <strong>of</strong> POPs on insulin action in adipocytes shown as the ability <strong>of</strong> differentiated 3T3-L1 adipocytes to<br />

take up radioactive-labeled glucose in response to insulin measured after 48 hr exposure to several POP mixtures<br />

found in crude oil from farmed Atlantic salmon. (A) POP mixture, (B) PCDD mixture, (C) PCDF mixture, (D) non–<br />

ortho-substituted PCB mixture, (E) mono–ortho-substituted PCB mixture, (F) Pesticide mixture, (G) DDT mixture,<br />

or (H) PCB mixture. Concentrations <strong>of</strong> POP mixtures <strong>are</strong> shown according to the highest contaminant compound<br />

present in the mixture, as well as the World Health Organization (WHO) 2005 TEQ for dioxins and dioxin-like PCBs<br />

(Van den Berg et al. 2006). Glucose uptake was determined in eight parallel wells for each mixture and for each<br />

concentration. Data <strong>are</strong> expressed as relative glucose uptake and presented as mean ± SE.<br />

*p < 0.05 comp<strong>are</strong>d with vehicle (dimethyl sulfoxide)-treated cells.<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

Organochlorine pesticides<br />

Vehicle<br />

1 nM<br />

10 nM<br />

100 nM<br />

* * * *<br />

* *<br />

Insulin (nM)<br />

* * *<br />

*<br />

* *<br />

*<br />

* *<br />

*<br />

Environmental Health Perspectives • v o l u m e 118 | number 4 | April 2010 469


Ruzzin et al.<br />

salmon oil induced a significant inhibition <strong>of</strong><br />

insulin-<strong>dependent</strong> glucose uptake. <strong>The</strong>se data<br />

provide compelling evidence that exposure to<br />

POPs increases the risk <strong>of</strong> developing insulin<br />

resistance and metabolic disorders.<br />

Despite intense investigations and establishment<br />

<strong>of</strong> both preventive and therapeutic<br />

strategies, insulin resistance–associated meta bolic<br />

diseases such as type 2 diabetes, obesity, and<br />

non alcoholic <strong>fatty</strong> liver disease have reached<br />

alarming proportions worldwide (Angulo<br />

2002; Ford et al. 2004; Zimmet et al. 2001).<br />

By 2015, the World Health Organization<br />

(WHO) estimates that > 1.5 billion people<br />

will be overweight and that 338 million people<br />

will die from chronic diseases such as diabetes<br />

and heart disease (WHO 2005). Although<br />

physical inactivity and regular intake <strong>of</strong> highenergy<br />

diets <strong>are</strong> recognized contributors (Hill<br />

and Peters 1998; Roberts and Barnard 2005),<br />

these lifestyle factors can only partially explain<br />

the explosive and uncontrolled global increase<br />

in metabolic diseases. Recently, the development<br />

<strong>of</strong> insulin resistance and inflammation<br />

was found to be exacerbated in humans and<br />

animals exposed to air pollution (Kelishadi<br />

et al. 2009; Sun et al. 2009). Furthermore,<br />

the widespread environmental contaminant<br />

bisphenol A was reported to impair pancreatic<br />

beta cells and trigger insulin resistance (Alonso-<br />

Magdalena et al. 2006). Our data, together<br />

with the finding that type 2 diabetics accumulate<br />

significant body burdens <strong>of</strong> POPs (Lee<br />

et al. 2006), provide additional evidence that<br />

global environmental pollution contributes to<br />

the epidemic <strong>of</strong> insulin resistance–associated<br />

metabolic diseases.<br />

Although rats chronically fed the HFC<br />

diet for 28 days were exposed to a relatively<br />

high intake <strong>of</strong> organic pollutants, the<br />

POPs<br />

Nuclear<br />

receptors<br />

(AhR, CAR, PXR, )<br />

TLR5, ROCK2,<br />

CD14, and YWHAZ<br />

PGC1α<br />

Insig-1<br />

Lpin1<br />

SREBP1c<br />

FAS<br />

concentrations <strong>of</strong> PCDDs/PCDFs and indicator<br />

PCBs in adipose tissue <strong>of</strong> these animals<br />

did not exceed those observed in Northern<br />

Europeans 40–50 years <strong>of</strong> age (Kiviranta et al.<br />

2005), thereby indicating that doses <strong>of</strong> POP<br />

exposure sufficient to induce detrimental<br />

health <strong>effects</strong> were not excessive. Whether the<br />

exposure to lower levels <strong>of</strong> POPs would induce<br />

similar detrimental <strong>effects</strong> as those observed in<br />

the present study remains to be investigated.<br />

Dietary interventions <strong>are</strong> current strategies<br />

to prevent or treat metabolic diseases,<br />

and nutritional guidelines <strong>are</strong> usually based<br />

on energy density and glycemic index <strong>of</strong> the<br />

diet; however, the levels <strong>of</strong> POPs present in<br />

food has received less attention. Given that<br />

POPs <strong>are</strong> ubiquitous in food chains (Fisher<br />

1999), such under estimation may interfere<br />

with the expected beneficial <strong>effects</strong> <strong>of</strong> some<br />

dietary recommendations and lead to poor<br />

outcomes. For instance, the presence <strong>of</strong> POPs<br />

in food products may, to some extent, explain<br />

the conflicting results regarding the protective<br />

<strong>effects</strong> <strong>of</strong> n-3 <strong>polyunsaturated</strong> <strong>fatty</strong> <strong>acids</strong><br />

against the incidence <strong>of</strong> myocardial infarction<br />

(Guallar et al. 1999; Rissanen et al. 2000).<br />

Overall, better understanding <strong>of</strong> the interactions<br />

between POPs and nutrients will help<br />

improve nutritional education <strong>of</strong> patients with<br />

insulin resistance syndrome.<br />

To protect consumer health, the presence<br />

<strong>of</strong> contaminants in food is internationally<br />

regulated. In the European Union legislation,<br />

certain POPs including dioxins and dioxin-like<br />

PCBs <strong>are</strong> regulated in foodstuffs (European<br />

Union 2006). Risk assessment <strong>of</strong> these organic<br />

pollutants is based on the ability <strong>of</strong> individual<br />

compounds to produce hetero geneous toxic<br />

and biological <strong>effects</strong> through the binding <strong>of</strong><br />

the aryl hydrocarbon receptor. Interestingly,<br />

Chronic low-grade<br />

inflammation<br />

Mitochondrial function<br />

Fatty acid oxidation<br />

Lipogenesis<br />

Insulin resistance<br />

syndrome<br />

Figure 4. Schematic representation <strong>of</strong> the possible mechanisms behind the development <strong>of</strong> the insulin<br />

resistance syndrome induced by POP exposure. POPs may activate nuclear receptors including aryl<br />

hydrocarbon receptor (AhR), pregnane X receptor (PXR), constitutive androstane receptor (CAR), or yet<br />

unknown receptors. POP exposure may induce the regulation <strong>of</strong> genes involved in the inflammatory pathway,<br />

mitochondrial function, lipid oxidation, and lipogenesis, thereby contributing to the development <strong>of</strong><br />

the insulin resistance syndrome.<br />

we found that cultured adipo cytes exposed to a<br />

PCDF or PCDD mixture have normal insulin<br />

action, even though the TEQ <strong>of</strong> these mixtures<br />

could be up to 3,500 times higher than the<br />

TEQ <strong>of</strong> the non-ortho-substituted and monoortho-substituted<br />

PCB mixtures that impaired<br />

insulin action. <strong>The</strong>se findings demonstrate that<br />

risk assessment based on WHO TEQs assigned<br />

to dioxins and dioxin-like PCBs is unlikely to<br />

reflect the risk <strong>of</strong> insulin resistance and the possible<br />

development <strong>of</strong> metabolic disorders.<br />

Although the production <strong>of</strong> organochlorine<br />

pesticides has been restricted since<br />

the 1970s, the global production and use <strong>of</strong><br />

pesticides <strong>are</strong> poorly controlled (Jorgenson<br />

2001; Nweke and Sanders 2009), and the<br />

presence <strong>of</strong> these environmental chemicals in<br />

seafood still remains unregulated in European<br />

countries (European Union 2008). Of the<br />

POP mixtures tested in vitro, organochlorine<br />

pesticides were the most potent disruptors <strong>of</strong><br />

insulin action. This powerful inhibitory effect<br />

<strong>of</strong> pesticides on insulin action likely explains<br />

the common finding emerging from several<br />

in<strong>dependent</strong> cross-sectional studies reporting<br />

an association between type 2 diabetes and<br />

the body burdens <strong>of</strong> p,p´-DDE, oxychlordane,<br />

or trans-nonachlor (Lee et al. 2006; Rignell-<br />

Hydbom et al. 2007; Turyk et al. 2009).<br />

<strong>The</strong>refore, widespread pesticide exposure to<br />

humans appears to be <strong>of</strong> particular global concern<br />

in relation to public health.<br />

We draw two main conclusions from<br />

these observations. First, exposure to POPs<br />

present in the environment and food chains<br />

<strong>are</strong> capable <strong>of</strong> causing insulin resistance and<br />

impair both lipid and glucose metabolism,<br />

thus supporting the notion that these chemicals<br />

<strong>are</strong> potential contributors to the rise in<br />

prevalence <strong>of</strong> insulin resistance and associated<br />

disorders (Figure 4). Second, although beneficial,<br />

the presence <strong>of</strong> n-3 <strong>polyunsaturated</strong> <strong>fatty</strong><br />

<strong>acids</strong> in crude salmon oil (in the HFC diet)<br />

could not counteract the deleterious metabolic<br />

<strong>effects</strong> induced by POP exposure. Altogether,<br />

our data provide novel insights regarding the<br />

ability <strong>of</strong> POPs to mediate insulin resistance–<br />

associated metabolic abnormalities and provide<br />

solid evidence reinforcing the importance<br />

<strong>of</strong> international agreements to limit the release<br />

<strong>of</strong> POPs to minimize public health risks.<br />

References<br />

Alonso-Magdalena P, Morimoto S, Ripoll C, Fuentes E, Nadal A.<br />

2006. <strong>The</strong> estrogenic effect <strong>of</strong> bisphenol A disrupts pancreatic<br />

β-cell function in vivo and induces insulin resistance.<br />

Environ Health Perspect 114:106–112.<br />

Angulo P. 2002. Nonalcoholic <strong>fatty</strong> liver disease. N Engl J Med<br />

346(16):1221–1231.<br />

Atlas E, Giam CS. 1981. Global transport <strong>of</strong> organic pollutants:<br />

ambient concentrations in the remote marine atmosphere.<br />

Science 211(4478):163–165.<br />

Begum N, Sandu OA, Ito M, Lohmann SM, Smolenski A. 2002.<br />

Active Rho kinase (ROK-α) associates with insulin receptor<br />

substrate-1 and inhibits insulin signaling in vascular smooth<br />

muscle cells. J Biol Chem 277(8):6214–6222.<br />

470 v o l u m e 118 | number 4 | April 2010 • Environmental Health Perspectives


POP exposure provokes insulin resistance<br />

Berntssen MHG, Lundebye AK, Torstensen BE. 2005. Reducing<br />

the levels <strong>of</strong> dioxins and dioxin-like PCBs in farmed<br />

Atlantic salmon by substitution <strong>of</strong> fish oil with vegetable oil<br />

in the feed. Aquac Nutr 11(3):219–231.<br />

Biddinger SB, Kahn CR. 2006. From mice to men: insights into the<br />

insulin resistance syndromes. Annu Rev Physiol 68:123–158.<br />

Bonnard C, Durand A, Peyrol S, Chanseaume E, Chauvin MA,<br />

Morio B, et al. 2008. Mitochondrial dysfunction results<br />

from oxidative stress in the skeletal muscle <strong>of</strong> diet-induced<br />

insulin-resistant mice. J Clin Invest 118(2):789–800.<br />

Brand CL, Sturis J, Gotfredsen CF, Fleckner J, Fledelius C,<br />

Hansen BF, et al. 2003. Dual PPARalpha/gamma activation<br />

provides enhanced improvement <strong>of</strong> insulin sensitivity and<br />

glycemic control in ZDF rats. Am J Physiol 284(4):E841–854.<br />

Buren J, Liu HX, Jensen J, Eriksson JW. 2002. Dexamethasone<br />

impairs insulin signalling and glucose transport by depletion<br />

<strong>of</strong> insulin receptor substrate-1, phosphatidylinositol<br />

3-kinase and protein kinase B in primary cultured rat adipocytes.<br />

Eur J Endocrinol 146(3):419–429.<br />

Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D,<br />

et al. 2007. Metabolic endotoxemia initiates obesity and<br />

insulin resistance. Diabetes 56(7):1761–1772.<br />

Croce MA, Eagon JC, LaRiviere LL, Korenblat KM, Klein S,<br />

Finck BN. 2007. Hepatic lipin 1β expression is diminished<br />

in insulin-resistant obese subjects and is reactivated by<br />

marked weight loss. Diabetes 56(9):2395–2399.<br />

Dougherty CP, Holtz SH, Reinert JC, Panyacosit L, Axelrad DA,<br />

Woodruff TJ. 2000. Dietary exposures to food contaminants<br />

across the United States. Environ Res 84(2):170–185.<br />

Engelking LJ, Kuriyama H, Hammer RE, Horton JD, Brown MS,<br />

Goldstein JL, et al. 2004. Overexpression <strong>of</strong> Insig-1 in<br />

the livers <strong>of</strong> transgenic mice inhibits SREBP processing<br />

and reduces insulin-stimulated lipogenesis. J Clin Invest<br />

113(8):1168–1175.<br />

European Union. 2006. Amending regulation (EC) No 466/2001<br />

setting maximum levels for certain contaminants in foodstuffs<br />

as regards dioxins and dioxin-like PCBs. Commission<br />

Regulation (EC) No 199/2006. Official Journal <strong>of</strong> the<br />

European Union L32:34–38. Available: http://www.health.<br />

gov.mt/fsc/fsc_euleg_files/RegEC199_2006e.pdf [accessed<br />

19 February 2010].<br />

European Union. 2008. Amending regulation (EC) No 396/2005<br />

on maximum residue levels <strong>of</strong> pesticides in or on food<br />

and feed <strong>of</strong> plant and animal origin, as regards the implementing<br />

powers conferred on the Commission. Regulation<br />

(EC) No 299/2008. Official Journal <strong>of</strong> the European Union<br />

L97:37–71. Available: http://eur-lex.europa.eu/LexUriServ/<br />

LexUriServ.douri=OJ:L:2008:097:0067:0071:EN:PDF<br />

[accessed 19 February 2010].<br />

Fierens S, Mairesse H, Heilier JF, De Burbure C, Focant JF,<br />

Eppe G, et al. 2003. Dioxin/polychlorinated biphenyl body burden,<br />

diabetes and endometriosis: findings in a populationbased<br />

study in Belgium. Biomarkers 8(6):529–534.<br />

Finck BN, Gropler MC, Chen Z, Leone TC, Croce MA, Harris TE,<br />

et al. 2006. Lipin 1 is an inducible amplifier <strong>of</strong> the hepatic<br />

PGC-1alpha/PPARalpha regulatory pathway. Cell Metab<br />

4(3):199–210.<br />

Fisher BE. 1999. Most unwanted. Environ Health Perspect<br />

107:A18–23.<br />

Ford ES, Giles WH, Mokdad AH. 2004. Increasing prevalence <strong>of</strong><br />

the metabolic syndrome among U.S adults. Diabetes C<strong>are</strong><br />

27(10):2444–2449.<br />

Furukawa N, Ongusaha P, Jahng WJ, Araki K, Choi CS, Kim HJ,<br />

et al. 2005. Role <strong>of</strong> Rho-kinase in regulation <strong>of</strong> insulin<br />

action and glucose homeostasis. Cell Metab 2(2):119–129.<br />

Guallar E, Aro A, Jimenez FJ, Martin-Moreno JM, Salminen I,<br />

van’t Veer P, et al. 1999. Omega-3 <strong>fatty</strong> <strong>acids</strong> in adipose<br />

tissue and risk <strong>of</strong> myocardial infarction—<strong>The</strong> EURAMIC<br />

study. Arterioscler Thromb 19(4):1111–1118.<br />

Henriksen GL, Ketchum NS, Michalek JE, Swaby JA. 1997.<br />

Serum dioxin and diabetes mellitus in veterans <strong>of</strong> Operation<br />

Ranch Hand. Epidemiology 8(3):252–258.<br />

Hill JO, Peters JC. 1998. Environmental contributions to the<br />

obesity epidemic. Science 280(5368):1371–1374.<br />

Hites RA, Foran JA, Carpenter DO, Hamilton MC, Knuth BA,<br />

Schwager SJ. 2004. Global assessment <strong>of</strong> organic contaminants<br />

in farmed salmon. Science 303(5655):226–229.<br />

Jordal AEO, Lie O, Torstensen BE. 2007. Complete replacement<br />

<strong>of</strong> dietary fish oil with a vegetable oil blend affect liver lipid<br />

and plasma lipoprotein levels in Atlantic salmon (Salmo<br />

salar L.). Aquac Nutr 13(2):114–130.<br />

Jorgenson JL. 2001. Aldrin and dieldrin: a review <strong>of</strong> research on<br />

their production, environmental deposition and fate, bioaccumulation,<br />

toxicology and epidemiology in the United<br />

States. Environ Health Perspect 109(suppl 1):113–139.<br />

Julshamn K, Lundebye AK, Heggstad K, Berntssen MHG,<br />

Boe B. 2004. Norwegian monitoring programme on the<br />

inorganic and organic contaminants in fish caught in the<br />

B<strong>are</strong>nts Sea, Norwegian Sea and North Sea, 1994–2001.<br />

Food Addit Contam 21(4):365–376.<br />

Jump DB. 2002. <strong>The</strong> biochemistry <strong>of</strong> n-3 <strong>polyunsaturated</strong> <strong>fatty</strong><br />

<strong>acids</strong>. J Biol Chem 277(11):8755–8758.<br />

Kelishadi R, Mirghaffari N, Poursafa P, Gidding SS. 2009. Lifestyle<br />

and environmental factors associated with inflammation,<br />

oxidative stress and insulin resistance in children.<br />

Atherosclerosis 203(1):311–319.<br />

Kiviranta H, Tuomisto JT, Tuomisto J, Tukiainen E, Vartiainen T.<br />

2005. Polychlorinated dibenzo-p-dioxins, dibenz<strong>of</strong>urans, and<br />

biphenyls in the general population in Finland. Chemosphere<br />

60(7):854–869.<br />

Kraegen EW, James DE, Bennett SP, Chisholm DJ. 1983. In vivo<br />

insulin sensitivity in the rat determined by euglycemic<br />

clamp. Am J Physiol 245(1):E1–E7.<br />

Lavigne C, Tremblay F, Asselin G, Jacques H, M<strong>are</strong>tte A. 2001.<br />

Prevention <strong>of</strong> skeletal muscle insulin resistance by dietary<br />

cod protein in high fat-fed rats. Am J Physiol 281(1):E62–E71.<br />

Lee DH, Lee IK, Song K, Steffes M, Toscano W, Baker BA, et al.<br />

2006. A strong dose-response relation between serum concentrations<br />

<strong>of</strong> persistent organic pollutants and diabetes:<br />

results from the National Health and Examination Survey<br />

1999–2002. Diabetes C<strong>are</strong> 29(7):1638–1644.<br />

Lee JN, Ye J. 2004. Proteolytic activation <strong>of</strong> sterol regulatory<br />

element-binding protein induced by cellular stress through<br />

depletion <strong>of</strong> Insig-1. J Biol Chem 279(43):45257–45265.<br />

Lee S, Muniyappa R, Yan X, Chen H, Yue LQ, Hong EG, et al. 2008.<br />

Comparison between surrogate indexes <strong>of</strong> insulin sensitivity<br />

and resistance and hyperinsulinemic euglycemic clamp<br />

estimates in mice. Am J Physiol 294(2):261–270.<br />

Lowell BB, Shulman GI. 2005. Mitochondrial dysfunction and<br />

type 2 diabetes. Science 307(5708):384–387.<br />

Nweke OC, Sanders WH III. 2009. Modern environmental health<br />

hazards: a public health issue <strong>of</strong> increasing significance in<br />

Africa. Environ Health Perspect 117:863–870.<br />

Petersen RK, Madsen L, Pedersen LM, Hallenborg P, Hagland H,<br />

Viste K, et al. 2008. Cyclic AMP (cAMP)-mediated stimulation<br />

<strong>of</strong> adipocyte differentiation requires the synergistic<br />

action <strong>of</strong> Epac- and cAMP-<strong>dependent</strong> protein kinase-<strong>dependent</strong><br />

processes. Mol Cell Biol 28(11):3804–3816.<br />

Reaven GM. 2005. Why Syndrome X From Harold Himsworth<br />

to the insulin resistance syndrome. Cell Metab 1(1):9–14.<br />

Reeves PG, Nielsen FH, Fahey GC Jr. 1993. AIN-93 purified diets<br />

for laboratory rodents: final report <strong>of</strong> the American Institute<br />

<strong>of</strong> Nutrition ad hoc writing committee on the reformulation<br />

<strong>of</strong> the AIN-76A rodent diet. J Nutr 123(11):1939–1951.<br />

Rignell-Hydbom A, Rylander L, Hagmar L. 2007. Exposure to<br />

persistent organochlorine pollutants and type 2 diabetes<br />

mellitus. Hum Exp Toxicol 26(5):447–452.<br />

Rissanen T, Voutilainen S, Nyyssonen K, Lakka TA, Salonen JT.<br />

2000. Fish oil-derived <strong>fatty</strong> <strong>acids</strong>, docosahexaenoic acid<br />

and docosapentaenoic acid, and the risk <strong>of</strong> acute coronary<br />

events—the Kuopio Ischaemic Heart Disease Risk Factor<br />

Study. Circulation 102(22):2677–2679.<br />

Roberts CK, Barnard RJ. 2005. Effects <strong>of</strong> exercise and diet on<br />

chronic disease. J Appl Physiol 98(1):3–30.<br />

Rome S, Lecomte V, Meugnier E, Rieusset J, Debard C, Euthine<br />

V, et al. 2008. Microarray analyses <strong>of</strong> SREBP-1a and<br />

SREBP-1c target genes identify new regulatory pathways<br />

in muscle. Physiol Genomics 34(3):327–337.<br />

Ruzzin J, Turcotte S, Leblanc V, Lavigne C, Froyland L,<br />

Remman TR, et al. 2007. Dietary salmon protein improves<br />

insulin sensitivity in high-fat fed obese rats [Abstract]. Ann<br />

Nutr Metab 51:347.<br />

Ruzzin J, Wagman AS, Jensen J. 2005. Glucocorticoid-induced<br />

insulin resistance in skeletal muscles: defects in insulin<br />

signalling and the <strong>effects</strong> <strong>of</strong> a selective glycogen synthase<br />

kinase-3 inhibitor. Diabetologia 48(10):2119–2130.<br />

Schafer KS, Kegley SE. 2002. Persistent toxic chemicals in the US<br />

food supply. J Epidemiol Community Health 56(11):813–817.<br />

Scientific Advisory Committee on Nutrition. 2004. Advice on Fish<br />

Consumption: Benefits and Risks. Available: http://cot.food.<br />

gov.uk/pdfs/fishreport2004full.pdf [accessed 19 February<br />

2010].<br />

Steele R, Wall JS, De Bodo RC, Altszuler N. 1956. Measurement<br />

<strong>of</strong> size and turnover rate <strong>of</strong> body glucose pool by the isotope<br />

dilution method. Am J Physiol 187(1):15–24.<br />

Storlien LH, Kraegen E.W, Chisholm DJ, Ford GL, Bruce DG,<br />

Pascoe WS. 1987. Fish oil prevents insulin resistance induced<br />

by high-fat feeding in rats. Science 237(4817):885–888.<br />

Sun Q, Yue P, Deiuliis JA, Lumeng CN, Kampfrath T, Mikolaj MB,<br />

et al. 2009. Ambient air pollution exaggerates adipose<br />

inflammation and insulin resistance in a mouse model <strong>of</strong><br />

diet-induced obesity. Circulation 119(4):538–546.<br />

Turyk M, Anderson HA, Knobeloch L, Imm P, Persky VW.<br />

2009. Prevalence <strong>of</strong> diabetes and body burdens <strong>of</strong> polychlorinated<br />

biphenyls, polybrominated diphenyl ethers,<br />

and p,p´-diphenyldichloroethene in Great Lakes sport fish<br />

consumers. Chemosphere 75(5):674–679.<br />

Tusher VG, Tibshirani R, Chu G. 2001. Significance analysis <strong>of</strong><br />

microarrays applied to the ionizing radiation response.<br />

Proc Natl Acad Sci USA 98(9):5116–5121.<br />

Tzivion G, Shen YH, Zhu J. 2001. 14-3-3 Proteins; bringing new<br />

definitions to scaffolding. Oncogene 20(44):6331–6338.<br />

Van den Berg H. 2009. Global status <strong>of</strong> DDT and its alternatives<br />

for use in vector control to prevent disease. Environ Health<br />

Perspect 17:1656–1663.<br />

Van den Berg M, Birnbaum LS, Denison M, De Vito M, Farland W,<br />

Feeley M, et al. 2006. <strong>The</strong> 2005 World Health Organization<br />

reevaluation <strong>of</strong> human and mammalian toxic equivalency<br />

factors for dioxins and dioxin-like compounds. Toxicol Sci<br />

93(2):223–241.<br />

Wang SL, Tsai PC, Yang CY, Guo YL. 2008. Increased risk <strong>of</strong><br />

diabetes and polychlorinated biphenyls and dioxins—a<br />

24-year follow-up study <strong>of</strong> the Yucheng cohort. Diabetes<br />

C<strong>are</strong> 31(8):1574–1579.<br />

WHO. 2005. Chronic Diseases: a Vital Investment. Geneva:World<br />

Health Organization. Available: http://www.who.int/chp/<br />

chronic_disease_report/contents/en/index.html [accessed<br />

19 February 2010].<br />

Zimmet P, Alberti KG, Shaw J. 2001. Global and societal implications<br />

<strong>of</strong> the diabetes epidemic. Nature 414(6865):782–787.<br />

Environmental Health Perspectives • v o l u m e 118 | number 4 | April 2010 471

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!