Vous êtes sur la page 1sur 13

The FASEB Journal Research Communication

Essential nutrient supplementation prevents heritable


metabolic disease in multigenerational intrauterine
growth-restricted rats
Danielle Goodspeed,* Maxim D. Seferovic,* William Holland, Robert A. Mcknight,
Scott A. Summers, D. Ware Branch,{ Robert H. Lane,|| and Kjersti M. Aagaard*,1
*Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas, USA;

Department of Internal Medicine, Department of Pediatrics, and {Department of Obstetrics and
Gynecology, University of Utah, Salt Lake City, Utah, USA; Program in Cardiovascular and Metabolic
Diseases, Duke-National University of Singapore Graduate Medical School, Singapore; and ||Department
of Pediatrics, Medical College of Wisconsin and Childrens Hospital of Wisconsin, Milwaukee,
Wisconsin, USA

ABSTRACT Intrauterine growth restriction (IUGR) THE GESTATIONAL ENVIRONMENT is a key arbiter of the relative
confers heritable alterations in DNA methylation, render- risk of health and disease in later life. In utero metabolic
ing risk of adult metabolic syndrome (MetS). Because CpG substrate restriction and uteroplacental insufciency lead-
methylation is coupled to intake of essential nutrients ing to intrauterine growth restriction (IUGR) is thought
along the one-carbon pathway, we reasoned that essential to bestow lifelong epigenetic changes that are malad-
nutrient supplementation (ENS) may abrogate IUGR- aptive to the postnatal environment. This phenomenon
conferred multigenerational MetS. Pregnant Sprague- is referred to as the developmental origins of health and
Dawley rats underwent bilateral uterine artery ligation disease hypothesis and was originally identied by asso-
causing IUGR in F1. Among the F2 generation, IUGR lin- ciating low birth weight neonates with increased mor-
eage rats were underweight at birth (6.7 vs. 8.0 g, P < tality from coronary heart disease much later in life (1, 2).
0.0001) and obese by adulthood (p160: 613 vs. 510 g; P < IUGR has now been associated with increased incidence
0.0001). Dual energy X-ray absorptiometry studies re- of health conditions including vascular dysfunction (3,
vealed increased central fat mass (D+40 g), accompanied 4), type II diabetes and dyslipidemia (5, 6), obesity, and
by dyslipidemic (>30% elevated, P < 0.05) serum trigly- hypertension (2, 7, 8). A symptomatic grouping of these
cerides (139 mg/dl), very-LDLs (27.8 mg/dl), and fatty IUGR health complications later in life is broadly char-
acids (632 mM). Hyperglycemic-euglycemic clamp studies acterized as metabolic syndrome (MetS), a condition
and glucose tolerance testing revealed insulin resistance. approaching global epidemic prevalence (9). The clini-
Conversely, IUGR lineage ENS-fed rats did not manifest cal criteria for MetS is evolving as it struggles to account
MetS, with signicantly lower body weight (p160: 410 g), for large differences in global populations, age, and race
>5-fold less central fat mass, normal hepatic glucose ef- (10). Currently, the consensus of clinical criteria includes
ux, and >70% reduced circulating triglycerides and very- the following: dyslipidemia, characterized by elevated
LDLs compared with IUGR control-fed F2 offspring triglycerides, ApoB lipoproteins, and reduced HDL; el-
(P < 0.01). Moreover, increased methylation of the IGF-1 evated blood pressure; and altered glucose homeostasis,
P2 transcriptional start site among IUGR lineage F2 off- with elevated central fat mass/obesity and insulin re-
spring was reversed in ENS (P < 0.04). This is an initial sistance (1012).
demonstration that supplementation along the one-carbon Maternal inuences, including poor nutrition and ute-
pathway abrogates adult morbidity and associated epi- roplacental insufciency, are associated with IUGR and
genomic modications of IGF-1 in a rodent model of adult MetS (1316). Rodent IUGR models that restrict ei-
multigenerational MetS.Goodspeed, D., Seferovic, M. D., ther protein or carbohydrates have demonstrated adult-
Holland, W., Mcknight, R. A., Summers, S. A., Branch, onset MetS associated with important epigenetic regula-
D. W., Lane, R. H., Aagaard, K. M. Essential nutrient tory changes to key metabolic pathways (1720), and our
supplementation prevents heritable metabolic disease in previous data in primates have demonstrated that a caloric-
multigenerational intrauterine growth-restricted rats. dense maternal diet can inuence epigenetic changes in
FASEB J. 29, 807819 (2015). www.fasebj.org the fetal liver (21). Although nutrient deprivation is a risk

Key Words: one-carbon pathway epigenomics methyl donors

IGF-1 obesity 1
Correspondence: Baylor College of Medicine, Division of
Maternal-Fetal Medicine, One Baylor Plaza, Jones 314, Houston,
Abbreviations: DEXA, dual energy X-ray absorptiometry; TX 77030; E-mail: aagaardt@bcm.tmc.edu
ENS, essential nutrient supplementation; HGO, hepatic glu- doi: 10.1096/fj.14-259614
cose output; IUGR, intrauterine growth restriction; MetS, This article includes supplemental data. Please visit http://
metabolic syndrome; VLDL, very-LDL www.fasebj.org to obtain this information.

0892-6638/15/0029-0807 FASEB 807


for MetS regardless of IUGR etiology (13, 2224), maternal therefore can only be derived from dietary sources (48).
deprivation is not a predominant cause of IUGR in all but Methylation of CpG islands is catalyzed by DNA meth-
the most underdeveloped countries. Rather, uteropla- yltransferases with S-adenosyl methionine as the methyl
cental insufciency is the predominant cause of fetal nu- donor, synthesized from methionine via dietary methyl
trient restriction leading to IUGR, which is independent of donors or one-carbon metabolism (49). Notably, the
maternal nutrition. Thus, maternal nutrient restrictive essential amino acid methionine is a methyl donor, itself
models inadequately replicate the broad deprivation of dependent on B12 as a methionine synthase cofactor,
metabolites and essential nutrients (including methyl which also uses a downstream product of folic acid as
donors) in placental insufciency, as well as other impor- a substrate. In an alternate liver pathway, methionine is
tant changes such as hypoxia and acidosis, waste product synthesized from betaine, which is derived from cho-
accumulation, and associated biochemical changes such as line. Further, several of the enzymes are zinc dependent
inammation, key regulatory factor alterations, and broad (46, 50). Methylation is further inuenced along the
transcriptional changes particularly associated with hyp- renal arginine-dependent nitric oxide pathway (51).
oxia. Ergo, the surgical bilateral uterine artery ligation Studies have made use of these essential nutrients to
procedure represents a more appropriate model to study study how epigenetic methylation changes relate to dis-
epigenetic changes associated with placental insufciency ease. For example, male mice have shown epigenetic
as it replicates restriction due to circulatory defects and in reprogramming of sperm that leads to birth defects in
this way leads to reduced maternal/fetal exchange. The offspring when fed a folate-decient diet (52). In another
immediate consequences are persistent in utero hypo- example, rats using essential nutrient methyl-donor dietary
insulinemia, hypoglycemia, acidosis, and hypoxia (25, 26). supplementation (ENS) have shown modications in DNA
Extensive study of animal models has uncovered some methylation and decreased hepatic triglyceride accumu-
specic epigenetic-mediated molecular mechanisms im- lation and nonalcoholic fatty liver disease, which are one of
plicated in long-term heath complications (20, 2735). the rst hepatic alterations of metabolic syndrome (53). In
IGF-1 is the primary driver of late gestational and postnatal humans, methylation changes to IGF-1 that were associ-
growth and has been implicated in altered weight and ated with nonalcoholic fatty liver disease were partially re-
development (36, 37). Decreased expression is associated versed as a result of nutritional adjustment following
with insulin resistance later in life, as well as increased in- bariatric surgery (54).
cidence of MetS (38). In a number of model systems, Given that DNA methylation is modiable by ENS of the
a perturbed in utero environment lending epigenomic diet, we hypothesized that supplementation initiated after
modications (both histone variations and differential weaning and continued through gestation and lactation of
methylation) leads to IGF-1 expression changes (3335, the F1 generation would abrogate the heritable adult
39). Of interest to our studies herein, an identical bilateral metabolic disease resulting from in utero IUGR among F2
uterine artery ligation procedure results in altered IGF-1 offspring. To study these effects, we rendered uteropla-
expression changes, which are accompanied by site- cental insufciency-induced IUGR by performing late-
specic differential methylation (33). Of the splice var- gestation bilateral uterine artery ligation on grandparental
iants, the IGF-1B was shown to be particularly variable both (P1) pregnant Sprague-Dawley rats at e19, with subsequent
in its expression and its methylation (33). delivery of the F1 pups at e21. The F1 generation was al-
Although DNA methylation is relatively stable, there are located onto either control or ENS-supplemented diet at
developmental time points, such as gamete imprinting, weaning (p21) and then allowed to breed spontaneously;
preimplantation development, and post-midgestational the F1 generation was not surgically manipulated during
development, when methylation changes are active (40). pregnancy. Regardless, the resultant IUGR lineage rats in
These windows provide intervention points where meth- the F2 generation weighed signicantly less at birth than
ylation is particularly plastic and, for imprinting and post- the sham lineage pups. By p160, a sex-specic MetS phe-
implantation events, amenable to dietary inuence. notype was apparent. IUGR lineage males fed a control
Dietary methylation supplementation is now known to lead diet exhibited obesity, increased central fat mass accumu-
to variable gene expression (41, 42). These epigenetic lation, glucose intolerance, insulin resistance, and in-
changes associated with methylation supplementation creased triglyceride, very-LDL (VLDL), and fatty acids.
continue to inuence biology after weaning (43). For ex- ENS-fed IUGR lineage males exhibited no obesogenicity,
ample, glucose tolerance in later life is associated with glucose intolerance, insulin resistance or increases in tri-
methyl deciency during gestation (44), and supplemen- glycerides, VLDL, or fatty acids, indicating a diet induced
tation of methyl donors alters hepatic promoter methyla- reversal of metabolic disease. Accompanying these notable
tion and the transcriptional prole in nonalcoholic fatty and signicant phenotypic alterations are signicant site-
liver disease (45). specic differences in DNA methylation of the IGF-1 pro-
DNA methylation changes as causative agents of dis- moter 2 region. Specically, we demonstrate that there is
ease have long been established. It was discovered that an observable and signicant corresponding increase in
methylation of our DNA is sufciently sensitive to es- methylation among F2 generation IUGR lineage rats rel-
sential dietary methyl sources and cofactors to cause ative to sham controls; a nding that reversed with ENS diet
human disease over a decade ago (46). For example, supplementation in the F1 and F2 generation. Collectively,
methyl sources related to seasonal food consumption in we observe a multigenerational transmission of the IUGR
Gambia affect DNA methylation patterns (47). This phenotype with manifestation of adult MetS and variations
sensitivity is likely caused by methyl group substrates and in site specic methylation of the P2 promoter of IGF-1,
cofactors that constitute the one-carbon metabolism which is abrogated by dietary supplementation of one-
pathway, many components of which are essential, and carbon intermediates in the F1 generation.

808 Vol. 29 March 2015 The FASEB Journal x www.fasebj.org GOODSPEED ET AL.
MATERIALS AND METHODS manufacturers instructions. Palmitate standard was made by
dissolving 74 mg palmitate into a solution of 35 ml ethanol and
IUGR rat model development 65 ml 6% Triton X-100 to make a high standard of 2.878 mM
and then serially diluted. Absorbance was assessed on a UV/Vis
microplate reader. A standard curve was generated from which
The experiments were conducted with the review and approval of
total fatty acid contents from serum samples were interpolated
the University of Utah Animal Care committee. Surgical proce-
using the average of triplicate wells.
dures were performed as previously described (55). Briey,
pregnant Sprague-Dawley rats were obtained on e17 and accli-
mated to their environment for 2 d prior to surgery. On day e19,
pregnant females were anesthetized with an intraperitoneal in- Glucose tolerance test
jection of xylazine (8 mg/kg) and ketamine (40 mg/ml). After
conrmation of anesthetization, bilateral uterine artery ligation Adult p160 rats were fasted for 6 h and then weighed. Blood
was performed to produce the uteroplacental insufciency IUGR samples were collected by tail vein as previously described. A
model, or sham surgeries were performed for controls. Pups were sample of blood was collected before administration of glucose,
born via cesarean section after 21 d of gestation, and litters were which was then administered orally to each rat (2.5 g glucose/kg
culled to 8 pups. On p21, suckling F1 offspring were weaned onto body weight). Blood samples were subsequently collected at
an ad libitum lifelong diet of either control rat chow (Harlan 15, 30, 45, 60, 90, and 120 min after glucose challenge from the
Teklad 8640; Harlan Laboratories, Indianapolis, IN, USA) or tail as described. Serum insulin levels were measured using an
a custom-formulated isocaloric Harlan Teklad 8640 rat chow ELISA kit.
supplemented with 15 mg of folic acid, 15 mg of choline, 1.5 mg of
B12, 15 g of betaine, 11 g of L-methionine, 50 g of L-arginine, and
150 mg of zinc for every kilogram of Harlan Teklad 8640, con- Hyperinsulinemic-euglycemic clamp studies
stituting the ENS diet. Mating pairs were established by p80, prior
to the development of F1 MetS. Mating pairs were shammat 3 The use of hyperinsulinemic-euglycemic clamp studies was car-
shampat, IUGRmat 3 shampat, shammat 3 IUGRpat, and IUGRmat 3 ried out to determine insulin resistance, as the most accurate and
IUGRpat for both the control diet and ENS diet, giving a total of sensitive laboratory method that is specic to hepatic insulin
8 different mating sets. Four hours after spontaneous delivery of resistance (58). Experiments were performed as described pre-
the F2 generation (N = 512), fetal weights were obtained, and viously (59). Adult p160 rats were fasted for 6 h prior to com-
litters were again culled to 8 pups (N = 32). On p21, the F2 mencement of the experiment. Following aseptic placement of
generation was weaned onto the same diet of their maternal and jugular (for infusion) and carotid (for blood sampling) catheters,
paternal lineage. Rats were weighed both at birth and at p160. animals were allowed to recover to presurgical weight (710 d).
Clamps were performed in conscious unrestrained animals using
swivels and tethers (Instech, Plymouth Meeting, PA, USA) to al-
Dual energy X-ray absorptiometry low uninterrupted movement of the animal without disruption of
infusion lines. Hyperinsulinemia was initiated by intravenous in-
Dual energy X-ray absorptiometry (DEXA) Norland XR-36 Bone fusion of insulin (4 mU/kg per min). Blood was sampled from
Densitometer (Norland Products, Inc., Cranbury Township, NJ, arterial lines in 7 min intervals and analyzed with a Beckman
USA) was used to assess rat whole body global and regional body Glucose analyzer II (Beckman Coulter, Fullerton, CA, USA).
composition. DEXA relies on X-ray technology to separate the Euglycemia was maintained by variable infusion of 20% dextrose.
lean and fat mass based on density and has been previously used Steady-state glucose (;130 mg/dl) was achieved ;90 min after
extensively for nutritional studies (56, 57). Rats were anesthetized initiating hyperinsulinemia and maintained for $45 min. Glu-
before measurements with an intraperitoneal injection of xyla- cose infusion rates were calculated as the average glucose infusion
zine (8 mg/kg) and ketamine (40 mg/ml) and then placed in rate during the steady-state period. Additional blood samples
prone position with extremities extended and stabilized with tape were taken before initiating hyperinsulinemia and at the end of
to enable full scanning of the radius and ulna. Spatial resolution the clamp for analysis of insulin. [14C]Glucose and [3H]2-deox-
was 6.0 3 6.0 mm. Scans were performed at a speed of 60 mm/s. yglucose were given as a bolus during the steady state for estimation
The scan eld width was 12 cm, and scanning time for each ani- of hepatic glucose output and 2-deoxyglucose uptake, as previously
mal was ,3 min. Following the scan, rats were immediately eu- described (60).
thanized. To determine central fat mass, a region of interest
including the torso area was extracted from the whole body scan,
and fat mass was determined. Methylation-specic PCR

To map IGF-1 promoter methylation changes, a standard PCR-


Fasting serum lipid proling based method following bisulte modication was performed as
described previously (33). Briey, primer sets were designed to
Adult p160 rats were fasted for 12 h, and then blood was collected probe 3 regions of CpG sites of the IGF-1 P2 transcription start
by nicking the end of the tail with a scalpel once. Blood ow was site. These were forward 59-GGAGGGTTTAATTTATAAAA-
maintained throughout by gentle stroking of the tail. Bleeding GATTTTAG, reverse 59-CCCAAACCACTTCCTTACCTAA; for-
was stopped after sampling by dipping the tail in alcohol and ward 59-GTTGTTGTTGTTATTGTTYGTGGTA, reverse 59-
subsequent application of direct pressure. A coronary risk and CTAAAATCTTTTATAAATTAAACCCTCC; and forward 59-TT-
lipid proling panel was run on each serum sample. Triglyceride AGGTAAGGAAGTGGTTTGGG, reverse 59-CACTATCTCAAC-
and VLDL concentrations were obtained by clinical laboratory TACCAAACCAC. PCR conditions were as follows: 95C for 10 min,
testing performed by ARUP Laboratories (Salt Lake City, UT, and then 94C for 30 s, annealing at 53C for 30 s, and 72C for
USA) using standard quantitative enzymatic assays. Free fatty 30 s for 35 cycles. Primers were designed and gures prepared
acids were measured from serum using the Roche Half-Micro with the use of University of California, Santa Cruz Genome
Test Kit (Indianapolis, IN, USA). The kit was scaled down for use Browser (61). A subset of 8 p21 animals was used for each
in a 96-well plate (one-fth suggested amount for each reagent; condition. PCR products were cloned into the vector pSC-A
10 ml serum). All preparation was performed according to the (Stratagene, Cedar Creek, TX, USA), and 6 colonies from each

PREVENTION OF HERITABLE METABOLIC DISEASE 809


cloning were inoculated on SeqPrep 96 plates (Edge Bio- lineage groups. Rats with either maternal or paternal IUGR
systems, Gaithersburg, MD, USA). The SeqPrep 96 Plasmid lineage were also mated with sham partners of the same
Prep Kit (Edge Biosystems) was used to prepare the plasmid diet. Additionally, sham males and females were paired
DNA according to manufacturers instructions, and the DNA and fed either control or ENS diets. The F1 dams were fed
was then sequenced.
their lifelong control or ENS diet throughout gestation and
suckling. At weaning, the F2 rats were placed on the same
Statistical analyses diet as their F1 parents. All experiments were performed
on the F2 generation.
Statistical analyses were performed using Prism v6.0 (GraphPad
Software Incorporated, La Jolla, CA, USA). One-way ANOVA with
Bonferronis post hoc analysis was used to compare mean Multigenerational heritable growth restriction
changes in weight. Students t test was used to compare body mass modiable by ENS in F1 and F2
composition and serum lipid prole and mean hepatic glucose
output (HGO). Means are reported 6 SEM. Signicance was de-
termined at P , 0.05.
After spontaneous delivery, fetal weights were obtained
and litters were culled (n = 8 pups per litter). Pups de-
rived from the Shammat/Shampat lineage were signi-
RESULTS cantly larger at birth than the pups derived from the
IUGRmat/IUGRpat lineage (Fig. 2A, B). Quantication
Development of a multigenerational heritable IUGR of birth weights revealed IUGRmat/IUGRpat pups from
rat model the maternal control diet to be 16% less at birth than the
Shammat/Shampat lineage (P , 0.0001; 6.7 vs. 8.0 g, re-
P1 Sprague-Dawley female rats fed a control diet un- spectively). Pups from either IUGRmat/Shampat (5.8 g)
derwent either a sham or bilateral uterine artery ligation or Shammat/IUGRpat (6.1 g) lineage also weighed 27%
surgery at day 19 after gestation (Fig. 1A). The rats spon- and 24% less at birth, respectively, than the Shammat/
taneously delivered either the F1 sham control or IUGR Shampat lineage (P , 0.0001). With maternal ENS-fed
lineage litters (Fig. 1A) and were thereafter culled (n = 8 rats, a 30% decrease in birth weight with the IUGRmat/
pups per litter). On p21, the F1 offspring were weaned IUGRpat lineage was observed, which was statistically
onto isocaloric diets of either control or ENS consisting of identical to the nonsupplemented IUGRmat/IUGRpat
the control diet with added folic acid, choline, B12, betaine, lineage rats. Similarly, Shammat/IUGRpat ENS-treated
methionine, arginine, and zinc. To establish the F2 line, F1 rats showed a marked decrease in birth weight (35%,
rats were mated at approximately day 80 of life (Fig. 1B). P , 0.0001), whereas IUGRmat/Shampat ENS-treated
IUGR females and males were either fed control or ENS rats did not show any signicant decrease in weight
diets and were paired to establish the IUGRmat/IUGRpat compared with Shammat/Shampat controls (Fig. 2B).

Figure 1. Mating scheme for heritable growth


restriction in rats. A) Pregnant female Sprague-
Dawley rats (P1) underwent either a bilateral
uterine artery ligation or sham surgery at
gestation day e19. Uterine ligation F1 rats
exhibiting IUGR or sham control rats were
then assigned to a lifelong diet of either control
chow (control) or ENS chow at weaning (p21).
B) At P80, males and females were mated in all
combinations of both maternal and paternal
IUGR lineage with and without ENS. The
resultant F2 generation was assigned the
parental diet of either ENS or control at p21,
generating 8 F2 study groups (n = 512 animals).

810 Vol. 29 March 2015 The FASEB Journal x www.fasebj.org GOODSPEED ET AL.
Figure 2. Uterine artery ligation in rats confers heritable growth restriction and adult obesity. A) F2 pups at birth from F1
IUGRpat/IUGRmat lineage and F1 Shampat/Shammat lineage show substantial birth weight differences. B) Differences in mean
birth weight of the F2 generation from the F1 lineage fed either a control or ENS diet. Signicant differences from Shampat/
Shammat control diet are indicated. C) F2 males at p160 from the IUGRpat/IUGRmat lineage fed either a control or ENS diet.
Mean p160 weights for F2 male (D) and female (E) rats, bred from the F1 lineage that were fed either a control or ENS diet.
Signicant differences from Shampat/Shammat control diet are indicated. (aP , 0.05, bP , 0.01, cP , 0.001, dP , 0.0001, 1-way
ANOVA with Bonferronis post hoc test).

These results suggest a multigenerational effect of low decreased for males only (P , 0.0001; Fig. 2D, E). Whereas
birth weight for the F2 pups, resultant from uteropla- the Shammat/Shampat showed no signicant change in
cental insufciency of the F1 generation. mean weight when fed an ENS diet, the obesity evident in
Male and female pups reassessed at p160 demonstrated the IUGRmat/IUGRpat control fed was abolished by ENS,
a signicant effect of ENS treatment. Figure 2C shows the with a more pronounced effect apparent in the males.
IUGRmat/IUGRpat males fed an ENS diet were smaller
than the IUGRmat/IUGRpat males fed a control diet, which
was true for females as well. Males derived from IUGRmat/ Multigenerational heritable obesity and dyslipidemia
IUGRpat fed a control diet weighed ;17% greater than modiable by ENS in F1 and F2
males from the Shammat/Shampat (Fig. 2D; P , 0.0001).
Similarly, the females from the IUGRmat/IUGRpat fed To determine the basis for the weight changes in IUGR, the
a control diet weighed ;19% more than the Shammat/ relative body fat and lipid prole of the animals was char-
Shampat females (Fig. 2E; P , 0.0001). Interestingly how- acterized as an indication of metabolic health. IUGRmat/
ever, both male and female IUGRmat/IUGRpat rats repor- IUGRpat rats fed a control diet were observed to have visibly
ted an average ; 20% reduction in weight gain when fed more fat mass than either IUGRmat/IUGRpat rats fed an
an ENS diet compared with the Shammat/Shampat control ENS diet or Shammat/Shampat rats (Fig. 3A). Lean mass
diet (Fig. 2D, E; P , 0.0001). For IUGRmat/Shampat rats fed and central fat mass were quantied on adult animals
a control diet, there was no statistically signicant differ- (p160) using DEXA on a clinical caliber Norland scanner,
ence in weights from the Shammat/Shampat and only for which representative scans are shown in Fig. 3B. No
a small 8% decrease for Shammat/IUGRpat, in female rats signicant difference in lean mass in either males or
only (P , 0.05; Fig. 2D, E). However, when fed an ENS diet, females was apparent (Fig. 3C, D). The IUGRmat/
the IUGRmat/Shampat males and females weighed signi- IUGRpat males fed a control diet, however, had 3 times
cantly less than the Shammat/Shampat (P , 0.0001 and more central fat mass than Shammat/Shampat control
0.01, respectively), but Shammat/IUGRpat was signicantly fed (P = 0.05), which was also signicantly greater than

PREVENTION OF HERITABLE METABOLIC DISEASE 811


Figure 3. ENS prevents adult obesity. A) Representative image of the central fat found in IUGR males fed either a control or ENS
diet. BF) DEXA scans for body fat analysis based on tissue density. B) Representative DEXA scans of Shampat/Shammat and
IUGRpat/IUGRmat males fed either a control or ENS diet. Mean lean mass in F2 males (C) and females (D) fed either a control or
ENS diet are indicated compared with Shampat/Shammat control. Differences in mean central fat mass of Shampat/Shammat and
IUGRpat/IUGRmat F2 males (E) and females (F) fed either a control or ENS diet compared with IUGRmat/IUGRpat control fed.
(aP , 0.05 considered signicant, independent Student t test).

either IUGRmat/IUGRpat and Shammat/Shampat males elevation (P = 0.009), which was reversed in both IUGRmat/
fed the ENS diet (P = 0.01; Fig. 3E). IUGRmat/IUGRpat IUGRpat and Shammat/Shampat ENS-fed rats (P , 0.01; Fig.
females fed a control diet also had signicantly more 4E, F). The alterations in serum lipid concentrations sug-
central fat mass than the IUGRmat/IUGRpat females fed gest that ENS supplementation reverses the IUGR-induced
an ENS diet (Fig. 3F; P , 0.01). The increased central fat increases that characterize metabolic disease.
mass observed in IUGRmat/IUGRpat males and females
suggests that ENS prevents central fat deposition.
Fasting serum lipid proles were then assessed as Multigenerational heritable glucose intolerance and
markers of dyslipidemia, which accompanies atheroscle- dysglycemia modiable by ENS in F1 and F2
rotic disease and is an indication of MetS. Triglycerides
were elevated in IUGRmat/IUGRpat control fed males and Given the strong relationship between insulin re-
females (P , 0.04), with a signicant reduction in both sistance and MetS, hyperinsulinemic-euglycemic clamp
Shammat/Shampat and the IUGRmat/IUGRpat ENS fed for studies with carbon 14-labeled glucose and tridium-
both males and females (P , 0.003; Fig. 4A, B). Similarly, labeled 2-deoxy glucose tracers were performed on
VLDL appeared elevated for both males and females. Al- adult animals. These clamp studies were used to discern
though it did not reach statistical signicance (P = 0.13 the hepatic insulin resistance, as opposed to overall or
and 0.09, respectively), VLDL was nevertheless lowered to peripheral, as a highly specic indication of metabolic
normal levels with ENS supplementation for both sexes dysregulation. HGO was signicantly higher in IUGRmat/
(P , 0.005; Fig. 4C, D). Serum free fatty acid levels also IUGRpat males than Shammat/Shampat males fed a control
exhibited the trend of lineage-induced elevation in diet (Fig. 5A; P , 0.001), indicative of impaired hepatic in-
IUGRmat/IUGRpat control diet, with males showing ;50% sulin sensitivity. When fed an ENS diet, IUGRmat/IUGRpat

812 Vol. 29 March 2015 The FASEB Journal x www.fasebj.org GOODSPEED ET AL.
Figure 4. ENS prevents adult dyslipidemia. Mean triglyceride levels in F2 males (A) and females (B) fed either a control or ENS
diet compared with IUGRmat/IUGRpat control fed. Mean VLDL levels in F2 males (C) and females (D) fed either a control or
ENS diet compared with IUGRmat/IUGRpat control fed. Mean free fatty acid levels in F2 males (E) and females (F) fed either
a control or ENS diet compared with IUGRmat/IUGRpat control fed. (P , 0.05 considered signicant and all signicant values
annotated, independent Student t test).

males exhibited enhanced insulin-mediated suppres- also the abrogation of the phenotype are considerably
sion of HGO, reversing them to Shammat/Shampat more pronounced in the male rats compared with the
control levels (Fig. 5A; P , 0.02). Similarly signicantly females.
distinct HGO proles were observed among female
offspring (Fig. 5B). IGF-1 promoter methylation in IUGR abrogated
To test for insulin responsiveness, glucose tolerance tests by ENS
were also performed. Serum insulin levels were increased
in the IUGRmat/IUGRpat rats fed a control diet compared To test whether ENS was acting to affect methylation of
with Shammat/Shampat following a glucose challenge (Fig. critical metabolic regulatory genes, we mapped the meth-
5C, D). At 50 min, the insulin levels of both male and female ylation status of the second promoter (P2) of the IGF-1
IUGRmat/IUGRpat rats failed to return to control baseline, from liver tissue of the variously treated groups. The pro-
and the levels were still severalfold elevated at 90 min moter map is depicted in Fig. 6A, where vertical lines are
compared with sham (Fig. 5C, D). For both male and fe- indicative of individual CpG dinucleotides, and amplicons
male rats, their ENS-fed insulin response was very different. from each of the areas IIII of the P2 promoter are shown.
For males, insulin was not discernibly elevated in IUGRmat/ Sequencing of bisulte-modied hepatic genomic DNA
IUGRpat rats fed the ENS diet compared with Shammat/ revealed site (CpG dinucleotide) specic differential
Shampat (Fig. 5E). For females, there appeared to be a sig- methylation. Specically, among IUGRmat/IUGRpat
nicant amelioration of the insulin response for ENS-fed control-fed F2 offspring destined to manifest adult MetS,
IUGRmat/IUGRpat rats compared with control diet; how- we observed signicantly increased P2 methylation com-
ever, insulin remained elevated above baseline at 50 min pared with Shammat/Shampat control-fed F2 offspring (P =
(Fig. 5F). Overall, the changes associated with IUGR and 0.03). Consistent with our phenotypic analysis, there was

PREVENTION OF HERITABLE METABOLIC DISEASE 813


Figure 5. ENS prevents adult insulin resistance. Mean HGO of Shampat/Shammat and IUGRpat/IUGRmat F2 males (A) and
females (B) fed either a control or ENS diet compared with IUGRpat/IUGRmat control. Representative insulin response of
Shampat/Shammat and IUGRpat/IUGRmat F2 male and female without (C, D) and with (E, F) ENS supplementation, respectively,
after glucose challenge. Insulin was measured by ELISA (P , 0.05 considered signicant and all signicant values annotated,
independent Student t test).

no apparent effect of ENS alone on the non-IUGR lineage generation exhibited excessive weight with increased
animals (Shammat/Shampat ENS fed; Fig. 6B, C). Promoter central adiposity and a highly transformed metabolism.
area II amplicons retained CpG sites where signicant Additionally, the adult F2 phenotype included dyslipi-
methylation changes occurred, whereas areas I and III demia, signicant insulin resistance, and altered glucose
showed minimal changes (Fig. 6B). When fed a diet sup- metabolism, which altogether are characteristics of MetS.
plemented with essential nutrients inclusive of the one- The observed changes were signicant in the males, who
carbon intermediates, methylation of the P2 promoter exhibited much greater heritability compared with
CpG dinucleotides were signicantly decreased in the females. Most interestingly, the phenotypes were nearly
IUGRmat/IUGRpat rats compared with control-fed off- completely reversed with ENS along the one-carbon
spring (P = 0.04; Fig. 6C). The altered methylation pat- metabolic pathway regardless of sex or amplitude. IGF-1
terning in a site (CpG dinucleotide and region-specic methylation was also shown to be reversed by ENS. These
manner) suggests that IUGR-mediated methylation results suggest that adaptive epigenomic changes are
changes in a critical metabolic organ are neither global nor modiable and sufciently plastic to reverse or prevent
indiscriminate. Further, the changes in promoter methyl- an inherited diseased epigenome.
ation appear in agreement with IGF-mediated restricted The phenotypic consequences of bilateral intrauterine
growth in F1 as previously reported (33). artery ligation to the F1 generation rats are well established
and include the MetS phenotype of insulin resistance, di-
abetes, hypertriglyceridemia, dyslipidemia, and hyperten-
DISCUSSION sion (13, 55, 62, 63). Additionally, a myriad of physiologic
and biochemical changes underlying the phenotype have
In this study, we used our well-established bilateral uterine been found. These include metabolic changes related to
artery ligation model of uteroplacental insufciency to altered islet development and dysglycemia (20), increased
demonstrate that IUGR induces MetS in the F1 generation hepatic glucocorticoid activity (27), altered GLUT1 and
(33, 34) and in the F2 generation. Our novel ndings are GLUT2 expression (28), hepatic glucose production, he-
consistent with other ndings of multigenerational effects patic one-carbon metabolism, altered hepatic fatty acid
in the human population and a single prior maternal lin- metabolism, muscle mitochondrial lipid metabolism, and
eage rodent study (15, 62). Remarkably, the adult F2 cerebral chromatin structure (2931, 64). There is evidence

814 Vol. 29 March 2015 The FASEB Journal x www.fasebj.org GOODSPEED ET AL.
Figure 6. IGF-1 promoter methylation mediated by IUGR and ENS. A) The 3 analyzed regions of promoter 2 of IGF-1 are
depicted together with the amplied areas sequenced. CpG dinucleotides are represented as vertical lines. In total, among the 3
indicated regions, the methylation status of 11 CpG dinucleotides was determined in bisulte-treated hepatic tissue. B)
Methylation patterns of CpG islands on the P2 transcription start site. Open circles are unmethylated CpGs, whereas closed
circles indicate methylated. Promoter site in indicated on the horizontal axis. C) Quantitation of overall methylation changes at
the 11 CpG sites for IUGR and Sham linage mice with and without supplementation. Differences from IUGRmat/IUGRpat are
indicated (independent Student t test, SEM).

of compromised kidney development and function and promoter was altered specically and signicantly in the
an altered intestinal morphology (32, 65, 66). Further, liver with inclusion of one-carbon metabolism inter-
important molecules related to development and to mediates as dietary supplements. Of note, these mod-
a MetS phenotype, such as IGF-1, PGC-1, CPTI, and ications occurred only among animals arising from the
PDX1, have been shown to be epigenetically altered (33, IUGR lineage and thus accompany reversal of the adult
34, 67, 68). MetS phenotype. Although at rst glance it might be
In these extensive and multigenerational experiments, expected that ENS would lead to diffuse methylation
we were able to enrich our deep phenotyping data across effects in multiple organ and tissue types, it is particularly
both the F1 and F2 generations with initial interrogations signicant that IGF-1 (a central anabolic and devel-
into key regulatory genes, such as IGF-1 (33, 34, 67, 68). We opmental regulator with systemic endocrine function) is
were guided in our decision of which genes and promoters altered by ENS; this is consistent with prior ndings in
to interrogate by the established biology, as well as the a single generation model of uterine artery ligation in the
known role of methylation in regulating alternate splice rat (33).
sites for transcription of the IGF-1 P2 promoter (33). We Hepatic expression of IGF-1 has diverse and systemic
show that CpG site-specic methylation of the IGF-1 P2 roles in development, growth, and glucose homeostasis,

PREVENTION OF HERITABLE METABOLIC DISEASE 815


and it is known to be adaptively regulated in IUGR (6971). synthesis. Higher levels of choline have been shown to be
Increased promoter methylation in the F1 IUGR rat liver protective of metabolic decits, potentially decreasing the
dampens IGF-1 expression in what is also thought to be an effects of heritable epigenetic aberrations in females (78, 79).
additional epigenetically mediated adaptive mechanism We nd the effectiveness of ENS supplementation re-
(33). It is well established that the IGF-1 gene sequence has markable in nearly reversing IUGR-associated adult meta-
variable epigenetic regulation, and accompanying tran- bolic morbidities. We intentionally initiated the ENS diet
scriptional changes have been shown repeatedly and by intervention in the F1 at the time of weaning and prior to
multiple investigators (33, 35, 39, 54). Despite this, the the onset of adult MetS, in an effort to both introduce
mechanisms for multigenerational inheritance observed supplementation prior to adolescence, as well as prior to
here are thus far unclear. Aside from its role in growth and evidence of metabolic dysfunction. We demonstrated the
development, low levels of IGF-1, like those seen accom- development of the multigenerational phenotype and
panying increased methylation following placental in- the adult MetS phenotypic reversal (but not multigenera-
sufciency (33), is critical to later glucose homeostasis and tional IUGR per se) is abrogated with an early in life ENS
insulin resistance (37, 72, 73). Our ndings herein signif- intervention.
icantly contribute to the role of epigenomic modications Although the ndings here present an interesting
in both regulating IUGR-mediated adult MetS and an ac- model of the principle of nongenomic coded inheritance
companying variation in IGF-1 expression and demon- and its implications for human health, there are never-
strate a favorable impact of one-carbon intermediate theless limitations. In humans, as in mice, a variety of fac-
supplementation. tors, including a nonstandard environment, variable
Although the metabolic effects to those born of IUGR parental care, and unique nonepigenetically determined
pregnancies are well documented, fewer studies have biology of the mother, can inuence the development of
reported on the multigenerational metabolic effects of the fetus and confer health altering changes that are not
IUGR. Recently, a study focusing on the F2 generation genetically heritable (80). Likewise, it must also be con-
uncovered that rats with low maternal birth weight due to sidered that the F2 germ line was exposed to relative re-
late-gestation (e19) uteroplacental insufciency exhibited striction and rescuing ENS supplementation, whereas their
a sex-specic reduced insulin response and altered pan- F1 mothers were undergoing growth restriction in utero.
creatic morphology (55). In another study, female F2 rats There are examples of germ-line inherited changes that
exhibited increased hepatic weight with aberrant glucose disappear in F3 (81). It is conceivable that the F2 effects
and insulin metabolism (74). Natural experiments and and reversal thereof came about through epigenetic
comparisons to human populations have borne out the modications in meiosis. Nevertheless, this does not di-
validity of the rodent IUGR models, nding similarities in minish the signicance that epigenetically programmed
epigenetic metabolic regulation (23, 33, 55, 65). Of par- changes regardless of when they occurred are subject to
ticular interest, the Dutch famine studies have identied intervention and mitigation with appropriate diet. It has
transgenerational epigenetic modications and a pro- been suggested that supplementation along the one-
pensity for increased adiposity and poor health in F2 as carbon pathway alters the biochemical balance of reac-
a result of F1 in utero metabolic restriction (23, 24). tions for carbohydrate metabolism. As with most experi-
Therefore, although some aspects of metabolic dysfunc- ments of this kind, it is not possible to fully isolate the
tion have been demonstrated, it was previously unclear that desired readouts resultant from genetic regulatory
MetS can be passed down from F1. changes from the direct effects of treatment on metabo-
The heritable MetS found in our IUGR model of ute- lism. Although it is suggested here that maternal supple-
roplacental insufciency indicates there are sex-specic mentation of ENS diet may stave off later metabolic disease,
phenotypes for heritability. For instance, males are more this conclusion is tempered by the use of an animal model.
severely affected than females in our model, which is Further, our model of uteroplacental insufciency is
highlighted by IUGRmat/IUGRpat lineage males de- a particularly severe one.
veloping the entire spectrum of MetS by adulthood. To our knowledge, we are the rst group to show a near-
However, we are able to ameliorate the metabolic syn- complete reversal of heritable MetS through dietary sup-
drome apparent in the IUGRmat/IUGRpat lineage male plementation, as well as a multigenerational transmission
rats with feeding of an ENS diet. Conversely, although down both maternal and paternal lineages. Using robust
IUGRmat/IUGRpat lineage females show a trend toward metrics of central fat mass, glucose tolerance, hepatic glu-
MetS, the distinction between IUGRmat/IUGRpat com- cose efux, serum lipid, and free fatty acids, we show that
pared with Shammat/Shampat was most evident with re- supplementation with essential nutrients along the one-
spect to adult weight. Similarly, the metabolic end points of carbon pathway prevents adult obesity, dyslipidemia, and
insulin dyslipidemia and insulin resistance were consider- insulin resistance, indicative of metabolic disease. The
ably more robust for males. Nevertheless, there was both novel demonstration of a paternal inheritance of adult
evidence of signicant multigenerational inheritance of MetS has substantial translational implications, as it
the adult MetS phenotype in IUGR lineage offspring, as underlines the role of paternal, as well as maternal, factors
well as a statistical amelioration when fed the ENS diet, as important to noncoded heritability of MetS. Identi-
regardless of sex. These results are consistent with reports cations of the mechanisms and genetic targets of herita-
from others that show sex-specic differences in IUGR (27, ble epigenetic-mediated metabolic changes and the
62, 7577). A suggested mechanism to account for the molecular mechanisms of phenotypic reversal through
protected phenotype in females is thought to be through ENS supplementation will reveal insights into the causes
estrogen signaling, where estrogen regulates levels of of the developmental origins of health and disease as
choline, a critical downstream substrate of methionine they relate to IUGR. Supplementation of IUGR offspring,

816 Vol. 29 March 2015 The FASEB Journal x www.fasebj.org GOODSPEED ET AL.
as rst identied at birth, may be a viable intervention to maternal health and second generation fetal growth. J. Physiol.
mitigate the risks conferred by multigenerational in- 590, 617630
17. Sohi, G., Marchand, K., Revesz, A., Arany, E., and Hardy, D. B.
heritance of metabolic-related diseases. (2011) Maternal protein restriction elevates cholesterol in adult
rat offspring due to repressive changes in histone modications
at the cholesterol 7alpha-hydroxylase promoter. Mol. Endocrinol.
The authors thank Xing Yu for the technical contributions 25, 785798
made during this project. Funding for this project was 18. Gong, L., Pan, Y.-X., and Chen, H. (2010) Gestational low pro-
provided by U.S. National Institutes of Health (NIH) Grant tein diet in the rat mediates Igf2 gene expression in male off-
DP21DP2OD001500-01 (Mapping the Fetal Primate Epige- spring via altered hepatic DNA methylation. Epigenetics 5,
nome and Metabolome), and the U.S. NIH Eunice Kennedy 619626
Shriver National Institute of Child Health and Human 19. Zheng, S., Rollet, M., and Pan, Y.-X. (2011) Maternal protein
Development Reproductive Scientist Development Program restriction during pregnancy induces CCAAT/enhancer-binding
Grant 5K12HD00849 (Transgenerational Growth Restriction protein (C/EBPb) expression through the regulation of histone
through Altered Placental Epigenetics) (to K.A.). modication at its promoter region in female offspring rat
skeletal muscle. Epigenetics 6, 161170
20. Schwitzgebel, V. M., Somm, E., and Klee, P. (2009) Modeling
intrauterine growth retardation in rodents: Impact on pancreas
REFERENCES development and glucose homeostasis. Mol. Cell. Endocrinol. 304,
7883
21. Suter, M., Bocock, P., Showalter, L., Hu, M., Shope, C.,
1. Barker, D. J., Winter, P. D., Osmond, C., Margetts, B., and McKnight, R., Grove, K., Lane, R., and Aagaard-Tillery, K. (2011)
Simmonds, S. J. (1989) Weight in infancy and death from Epigenomics: maternal high-fat diet exposure in utero disrupts
ischaemic heart disease. Lancet 2, 577580 peripheral circadian gene expression in nonhuman primates.
2. Barker, D. J., Osmond, C., Golding, J., Kuh, D., and Wadsworth, FASEB J. 25, 714726
M. E. (1989) Growth in utero, blood pressure in childhood and
22. Hales, C. N., Barker, D. J., Clark, P. M., Cox, L. J., Fall, C.,
adult life, and mortality from cardiovascular disease. BMJ 298,
Osmond, C., and Winter, P. D. (1991) Fetal and infant growth
564567
and impaired glucose tolerance at age 64. BMJ 303, 10191022
3. Skilton, M. R., Evans, N., Grifths, K. A., Harmer, J. A., and
23. Heijmans, B. T., Tobi, E. W., Stein, A. D., Putter, H., Blauw, G.
Celermajer, D. S. (2005) Aortic wall thickness in newborns with
J., Susser, E. S., Slagboom, P. E., and Lumey, L. H. (2008)
intrauterine growth restriction. Lancet 365, 14841486
Persistent epigenetic differences associated with prenatal ex-
4. Koklu, E., Ozturk, M. A., Gunes, T., Akcakus, M., and Kurtoglu, S.
posure to famine in humans. Proc. Natl. Acad. Sci. USA 105,
(2007) Is increased intima-media thickness associated with pre-
1704617049
atherosclerotic changes in intrauterine growth restricted new-
24. Painter, R. C., Osmond, C., Gluckman, P., Hanson, M., Phillips,
borns? Acta Paediatr. 96, 1858
D. I., and Roseboom, T. J. (2008) Transgenerational effects of
5. Barker, D. J. P., Hales, C. N., Fall, C. H., Osmond, C., Phipps, K.,
prenatal exposure to the Dutch famine on neonatal adiposity
and Clark, P. M. (1993) Type 2 (non-insulin-dependent) diabetes
and health in later life. BJOG 115, 12431249
mellitus, hypertension and hyperlipidaemia (syndrome X): re-
25. Ogata, E. S., Bussey, M. E., and Finley, S. (1986) Altered gas
lation to reduced fetal growth. Diabetologia 36, 6267
exchange, limited glucose and branched chain amino acids, and
6. Barker, D. J., Martyn, C. N., Osmond, C., Hales, C. N., and Fall,
hypoinsulinism retard fetal growth in the rat. Metabolism 35,
C. H. (1993) Growth in utero and serum cholesterol concen-
970977
trations in adult life. BMJ 307, 15241527
26. Ogata, E. S., Bussey, M. E., LaBarbera, A., and Finley, S. (1985)
7. Curhan, G. C., Chertow, G. M., Willett, W. C., Spiegelman, D.,
Altered growth, hypoglycemia, hypoalaninemia, and ketonemia
Colditz, G. A., Manson, J. E., Speizer, F. E., and Stampfer, M. J.
in the young rat: postnatal consequences of intrauterine growth
(1996) Birth weight and adult hypertension and obesity in
retardation. Pediatr. Res. 19, 3237
women. Circulation 94, 13101315
8. Curhan, G. C., Willett, W. C., Rimm, E. B., Spiegelman, D., 27. Baserga, M., Hale, M. A., McKnight, R. A., Yu, X., Callaway, C. W.,
Ascherio, A. L., and Stampfer, M. J. (1996) Birth weight and and Lane, R. H. (2005) Uteroplacental insufciency alters he-
adult hypertension, diabetes mellitus, and obesity in US men. patic expression, phosphorylation, and activity of the glucocor-
Circulation 94, 32463250 ticoid receptor in fetal IUGR rats. Am. J. Physiol. Regul. Integr.
9. Ervin, R. B. (2009) Prevalence of metabolic syndrome among Comp. Physiol. 289, R1348R1353
adults 20 years of age and over, by sex, age, race and ethnicity, 28. Lane, R. H., Crawford, S. E., Flozak, A. S., and Simmons, R. A.
and body mass index: United States, 2003-2006. National Health (1999) Localization and quantication of glucose transporters in
Statistics Report 13, 17 liver of growth-retarded fetal and neonatal rats. Am. J. Physiol.
10. Kassi, E., Pervanidou, P., Kaltsas, G., and Chrousos, G. (2011) 276, E135E142
Metabolic syndrome: denitions and controversies. BMC Med. 9, 48 29. Lane, R. H., Kelley, D. E., Ritov, V. H., Tsirka, A. E., and
11. Cornier, M.-A., Dabelea, D., Hernandez, T. L., Lindstrom, R. C., Gruetzmacher, E. M. (2001) Altered expression and function of
Steig, A. J., Stob, N. R., Van Pelt, R. E., Wang, H., and Eckel, R. H. mitochondrial beta-oxidation enzymes in juvenile intrauterine-
(2008) The metabolic syndrome. Endocr. Rev. 29, 777822 growth-retarded rat skeletal muscle. Pediatr. Res. 50, 8390
12. Eckel, R. H., Alberti, K. G., Grundy, S. M., and Zimmet, P. Z. 30. Lane, R. H., MacLennan, N. K., Hsu, J. L., Janke, S. M., and
(2010) The metabolic syndrome. Lancet 375, 181183 Pham, T. D. (2002) Increased hepatic peroxisome proliferator-
13. Simmons, R. A., Templeton, L. J., and Gertz, S. J. (2001) In- activated receptor-gamma coactivator-1 gene expression in a rat
trauterine growth retardation leads to the development of type 2 model of intrauterine growth retardation and subsequent insulin
diabetes in the rat. Diabetes 50, 22792286 resistance. Endocrinology 143, 24862490
14. Benyshek, D. C., Johnston, C. S., and Martin, J. F. (2006) Glucose 31. Ke, X., Lei, Q., James, S. J., Kelleher, S. L., Melnyk, S., Jernigan,
metabolism is altered in the adequately-nourished grand- S., Yu, X., Wang, L., Callaway, C. W., Gill, G., Chan, G. M.,
offspring (F3 generation) of rats malnourished during gesta- Albertine, K. H., McKnight, R. A., and Lane, R. H. (2006) Ute-
tion and perinatal life. Diabetologia 49, 11171119 roplacental insufciency affects epigenetic determinants of
15. Jimenez-Chillaron, J. C., Isganaitis, E., Charalambous, M., Gesta, chromatin structure in brains of neonatal and juvenile IUGR
S., Pentinat-Pelegrin, T., Faucette, R. R., Otis, J. P., Chow, A., rats. Physiol. Genomics 25, 1628
Diaz, R., Ferguson-Smith, A., and Patti, M. E. (2009) In- 32. Baserga, M., Hale, M. A., Ke, X., Wang, Z. M., Yu, X., Callaway,
tergenerational transmission of glucose intolerance and obesity C. W., McKnight, R. A., and Lane, R. H. (2006) Uteroplacental
by in utero undernutrition in mice. Diabetes 58, 460468 insufciency increases p53 phosphorylation without triggering
16. Gallo, L. A., Tran, M., Moritz, K. M., Mazzuca, M. Q., Parry, L. J., the p53-MDM2 functional circuit response in the IUGR rat kid-
Westcott, K. T., Jefferies, A. J., Cullen-McEwen, L. A., and ney. Am. J. Physiol. requl. Integr. Comp. Physiol. 291, R412R418
Wlodek, M. E. (2012) Cardio-renal and metabolic adaptations 33. Fu, Q., Yu, X., Callaway, C. W., Lane, R. H., and McKnight, R. A.
during pregnancy in female rats born small: implications for (2009) Epigenetics: intrauterine growth retardation (IUGR)

PREVENTION OF HERITABLE METABOLIC DISEASE 817


modies the histone code along the rat hepatic IGF-1 gene. 52. Lambrot, R., Xu, C., Saint-Phar, S., Chountalos, G., Cohen, T.,
FASEB J. 23, 24382449 Paquet, M., Suderman, M., Hallett, M., and Kimmins, S. (2013)
34. Park, J. H., Stoffers, D. A., Nicholls, R. D., and Simmons, R. A. Low paternal dietary folate alters the mouse sperm epigenome
(2008) Development of type 2 diabetes following intrauterine and is associated with negative pregnancy outcomes. Nat. Com-
growth retardation in rats is associated with progressive epige- mun. 4, 2889
netic silencing of Pdx1. J. Clin. Invest. 118, 23162324 53. Cordero, P., Gomez-Uriz, A. M., Campion, J., Milagro, F. I., and
35. Tosh, D. N., Fu, Q., Callaway, C. W., McKnight, R. A., McMillen, Martinez, J. A. (2013) Dietary supplementation with methyl
I. C., Ross, M. G., Lane, R. H., and Desai, M. (2010) Epigenetics donors reduces fatty liver and modies the fatty acid synthase
of programmed obesity: alteration in IUGR rat hepatic IGF1 DNA methylation prole in rats fed an obesogenic diet. Genes
mRNA expression and histone structure in rapid vs. delayed Nutr. 8, 105113
postnatal catch-up growth. Am. J. Physiol. Gastrointest. Liver Physiol. 54. Ahrens, M., Ammerpohl, O., von Schonfels, W., Kolarova, J.,
299, G1023G1029 Bens, S., Itzel, T., Teufel, A., Herrmann, A., Brosch, M.,
36. Woods, K. A., Camacho-Hubner, C., Savage, M. O., and Clark, Hinrichsen, H., Erhart, W., Eqberts, J., Sipos, B., Schreiber, S.,
A. J. (1996) Intrauterine growth retardation and postnatal Hasler.R., Stickel, F., Becker, T., Krawczak, M., Rocken,
growth failure associated with deletion of the insulin-like growth C., Siebert, R., Schafmayer, C., and Hampe, J. (2013) DNA
factor I gene. N. Engl. J. Med. 335, 13631367 methylation analysis in nonalcoholic fatty liver disease suggests
37. Netchine, I., Azzi, S., Houang, M., Seurin, D., Perin, L., Ricort, distinct disease-specic and remodeling signatures after bariatric
J. M., Daubas, C., Legay, C., Mester, J., Herich, R., Godeau, F., surgery. Cell Metab. 18, 296302
and Le Bouc, Y. (2009) Partial primary deciency of insulin-like 55. Lane, R. H., Kelley, D. E., Gruetzmacher, E. M., and Devaskar,
growth factor (IGF)-I activity associated with IGF1 mutation S. U. (2001) Uteroplacental insufciency alters hepatic fatty acid-
demonstrates its critical role in growth and brain development. metabolizing enzymes in juvenile and adult rats. Am. J. Physiol.
J. Clin. Endocrinol. Metab. 94, 39133921 Regul. Integr. Comp. Physiol. 280, R183R190
38. Woods, K. A., Camacho-Hubner, C., Bergman, R. N., Barter, D., 56. Lukaski, H. C. (1993) Soft tissue composition and bone mineral
Clark, A. J., and Savage, M. O. (2000) Effects of insulin-like status: evaluation by dual-energy X-ray absorptiometry. J. Nutr.
growth factor I (IGF-I) therapy on body composition and insulin 123(2, Suppl), 438443
resistance in IGF-I gene deletion. J. Clin. Endocrinol. Metab. 85, 57. Howie, G. J., Sloboda, D. M., Kamal, T., and Vickers, M. H.
14071411 (2009) Maternal nutritional history predicts obesity in adult off-
39. Tan, Y., Liu, J., Deng, Y., Cao, H., Xu, D., Cu, F., Lei, Y., spring independent of postnatal diet. J. Physiol. 587, 905915
Magdalou, J., Wu, M., Chen, L., and Wang, H. (2012) Caffeine- 58. Kim, J. K. (2009) Hyperinsulinemic-euglycemic clamp to assess
induced fetal rat over-exposure to maternal glucocorticoid and insulin sensitivity in vivo. Methods Mol. Biol. 560, 221238
histone methylation of liver IGF-1 might cause skeletal growth 59. Holland, W. L., Brozinick, J. T., Wang, L. P., Hawkins, E. D.,
retardation. Toxicol. Lett. 214, 279287 Sargent, K. M., Liu, Y., Narra, K., Hoehn, K. L., Knotts, T. A.,
40. Morgan, H. D., Santos, F., Green, K., Dean, W., and Reik, W. Siesky, A., Nelson, D. H., Karathansis, S. K., Fonternot, G. K.,
(2005) Epigenetic reprogramming in mammals. Hum. Mol. Genet. Birnhaum, M. J. and Summers, S. A. (2007) Inhibition of
14, R47R58 ceramide synthesis ameliorates glucocorticoid-, saturated-fat-,
41. Waterland, R. A., and Jirtle, R. L. (2003) Transposable elements: and obesity-induced insulin resistance. Cell Metab. 5, 167179
targets for early nutritional effects on epigenetic gene regulation. 60. Ye, J. M., Doyle, P. J., Iglesias, M. A., Watson, D. G., Cooney, G. J.,
Mol. Cell. Biol. 23, 52935300 and Kraegen, E. W. (2001) Peroxisome proliferator-activated
42. Waterland, R. A., and Jirtle, R. L. (2004) Early nutrition, epige- receptor (PPAR)-alpha activation lowers muscle lipids and
netic changes at transposons and imprinted genes, and en- improves insulin sensitivity in high fat-fed rats: comparison with
hanced susceptibility to adult chronic diseases. Nutrition 20, PPAR-gamma activation. Diabetes 50, 411417
6368 61. Karolchik, D., Hinrichs, A. S., Furey, T. S., Roskin, K. M., Sugnet,
43. Waterland, R. A., Lin, J. R., Smith, C. A., and Jirtle, R. L. C. W., Haussler, D., and Kent, W. J. (2004) The UCSC Table
(2006) Post-weaning diet affects genomic imprinting at the Browser data retrieval tool. Nucleic Acids Res. 32, D493D496
insulin-like growth factor 2 (Igf2) locus. Hum. Mol. Genet. 15, 62. Tran, M., Gallo, L. A., Jefferies, A. J., Moritz, K. M., and Wlodek,
705716 M. E. (2013) Transgenerational metabolic outcomes associated
44. Jahangir, E., Vita, J. A., Handy, D., Holbrook, M., Palmisano, J., with uteroplacental insufciency. J. Endocrinol. 217, 105118
Beal, R., Loscalzo, J., and Eberhardt, R. T. (2009) The effect of L- 63. Baserga, M., Bares, A. L., Hale, M. A., Callaway, C. W., McKnight,
arginine and creatine on vascular function and homocysteine R. A., Lane, P. H., and Lane, R. H. (2009) Uteroplacental in-
metabolism. Vasc. Med. 14, 239248 sufciency affects kidney VEGF expression in a model of IUGR
45. Seki, Y., Williams, L., Vuguin, P. M., and Charron, M. J. (2012) with compensatory glomerular hypertrophy and hypertension.
Minireview: Epigenetic programming of diabetes and obesity: Early Hum. Dev. 85, 361367
animal models. Endocrinology 153, 10311038 64. MacLennan, N. K., James, S. J., Melnyk, S., Piroozi, A., Jernigan,
46. Van den Veyver, I. B. (2002) Genetic effects of methylation diets. S., Hsu, J. L., Janke, S. M., Pham, T. D., and Lane, R. H. (2004)
Annu. Rev. Nutr. 22, 255282 Uteroplacental insufciency alters DNA methylation, one-carbon
47. Dominguez-Salas, P., Moore, S. E., Cole, D., da Costa, K. A., Cox, metabolism, and histone acetylation in IUGR rats. Physiol.
S. E., Dyer, R. A., Fulford, A. J., Innis, S. M., Waterland, R. A., Genomics 18, 4350
Zeisel, S. H., Prentice, A. M., and Hennig, B. J. (2013) DNA 65. Fu, Q., McKnight, R. A., Yu, X., Wang, L., Callaway, C. W., and
methylation potential: dietary intake and blood concentrations Lane, R. H. (2004) Uteroplacental insufciency induces site-
of one-carbon metabolites and cofactors in rural African women. specic changes in histone H3 covalent modications and affects
Am. J. Clin. Nutr. 97, 12171227 DNA-histone H3 positioning in day 0 IUGR rat liver. Physiol.
48. Cordero, P., Campion, J., Milagro, F. I., and Martinez, J. A. Genomics 20, 108116
(2013) Transcriptomic and epigenetic changes in early liver 66. Baserga, M., Bertolotto, C., Maclennan, N. K., Hsu, J. L., Pham,
steatosis associated to obesity: effect of dietary methyl donor T., Laksana, G. S., and Lane, R. H. (2004) Uteroplacental in-
supplementation. Mol. Genet. Metab. 110, 388395 sufciency decreases small intestine growth and alters apoptotic
49. Niculescu, M. D., and Zeisel, S. H. (2002) Diet, methyl donors homeostasis in term intrauterine growth retarded rats. Early
and DNA methylation: interactions between dietary folate, me- Hum. Dev. 79, 93105
thionine and choline. J. Nutr. 132(8, Suppl)2333S2335S 67. Thompson, R. F., Fazzari, M. J., Niu, H., Barzilai, N., Simmons,
50. Anderson, O. S., Sant, K. E., and Dolinoy, D. C. (2012) Nutrition R. A., and Greally, J. M. (2010) Experimental intrauterine growth
and epigenetics: an interplay of dietary methyl donors, one- restriction induces alterations in DNA methylation and gene
carbon metabolism and DNA methylation. J. Nutr. Biochem. 23, expression in pancreatic islets of rats. J. Biol. Chem. 285,
853859 1511115118
51. Dominguez-Salas, P., Cox, S. E., Prentice, A. M., Hennig, B. J., 68. Ganu, R. S., Harris, R. A., Collins, K., and Aagaard, K. M. (2012)
and Moore, S. E. (2012) Maternal nutritional status, C(1) me- Early origins of adult disease: approaches for investigating the
tabolism and offspring DNA methylation: a review of current programmable epigenome in humans, nonhuman primates, and
evidence in human subjects. Proc. Nutr. Soc. 71, 154165 rodents. ILAR J. 53, 306321

818 Vol. 29 March 2015 The FASEB Journal x www.fasebj.org GOODSPEED ET AL.
69. Seferovic, M. D., Ali, R., Kamei, H., Liu, S., Khosravi, J. M., 75. Maloney, C. A., Hay, S. M., Young, L. E., Sinclair, K. D., and Rees,
Nazarian, S., Han, V. K., Duan, C., and Gupta, M. B. (2009) W. D. (2011) A methyl-decient diet fed to rat dams during the
Hypoxia and leucine deprivation induce human insulin-like peri-conception period programs glucose homeostasis in adult
growth factor binding protein-1 hyperphosphorylation and in- male but not female offspring. J. Nutr. 141, 95100
crease its biological activity. Endocrinology 150, 220231 76. Fu, Q., McKnight, R. A., Yu, X., Callaway, C. W., and Lane, R. H.
70. Abu Shehab, M., Inoue, S., Han, V. K. M., and Gupta, M. B. (2006) Growth retardation alters the epigenetic characteristics of
(2009) Site specic phosphorylation of insulin-like growth factor hepatic dual specicity phosphatase 5. FASEB J. 20, 21272129
binding protein-1 (IGFBP-1) for evaluating clinical relevancy in 77. Lane, R. H., Maclennan, N. K., Daood, M. J., Hsu, J. L., Janke,
fetal growth restriction. J. Proteome Res. 8, 53255335 S. M., Pham, T. D., Puri, A. R., and Watchko, J. F. (2003) IUGR
71. Watson, C. S., Bialek, P., Anzo, M., Khosravi, J., Yee, S. P., and alters postnatal rat skeletal muscle peroxisome proliferator-
Han, V. K. (2006) Elevated circulating insulin-like growth factor activated receptor-gamma coactivator-1 gene expression in a -
binding protein-1 is sufcient to cause fetal growth restriction. ber specic manner. Pediatr. Res. 53, 9941000
Endocrinology 147, 11751186 78. Fischer, L. M., Costa, K., Kwock, L., Galanko, J., and Zeisel, S. H.
72. Sjogren, K., Wallenius, K., Liu, J. L., Bohlooly-Y, M., Pacini, G., (2010) Dietary choline requirements of women: effects of es-
Svensson, L., Tornell, J., Isaksson, O. G., Ahren, B., Jansson, J. O., trogen and genetic variation. Am. J. Clin. Nutr. 92, 11131119
and Ohlsson, C. (2001) Liver-derived IGF-I is of importance for 79. Resseguie, M., Song, J., Niculescu, M. D., da Costa, K. A., Randall,
normal carbohydrate and lipid metabolism. Diabetes 50, T. A., and Zeisel, S. H. (2007) Phosphatidylethanolamine N-
15391545 methyltransferase (PEMT) gene expression is induced by estro-
73. Haluzik, M., Yakar, S., Gavrilova, O., Setser, J., Boisclair, Y., and gen in human and mouse primary hepatocytes. FASEB J. 21,
LeRoith, D. (2003) Insulin resistance in the liver-specic IGF-1 26222632
gene-deleted mouse is abrogated by deletion of the acid-labile 80. Grossniklaus, U., Kelly, W. G., Ferguson-Smith, A. C., Pembrey,
subunit of the IGF-binding protein-3 complex: relative roles of M., and Lindquist, S. (2013) Transgenerational epigenetic in-
growth hormone and IGF-1 in insulin resistance. Diabetes 52, heritance: how important is it? Nat. Rev. Genet. 14, 228235
24832489 81. Waterland, R. A., Travisano, M., and Tahiliani, K. G. (2007) Diet-
74. Thamotharan, M., Garg, M., Oak, S., Rogers, L. M., Pan, G., induced hypermethylation at agouti viable yellow is not inherited
Sangiorgi, F., Lee, P. W., and Devaskar, S. U. (2007) Trans- transgenerationally through the female. FASEB J. 21, 33803385
generational inheritance of the insulin-resistant phenotype
in embryo-transferred intrauterine growth-restricted adult fe-
male rat offspring. Am. J. Physiol. Endocrinol. Metab. 292, E1270 Received for publication July 23, 2014.
E1279 Accepted for publication October 14, 2014.

PREVENTION OF HERITABLE METABOLIC DISEASE 819

Vous aimerez peut-être aussi