2014 early hepatic insult in the offspring of obese maternal mice

Upload: cristianalbertosandovalvasquez

Post on 06-Jul-2018

218 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    1/38

     

    Early hepatic insult in the offspring of obese maternal mice

    Isabele Bringhenti, Fernanda Ornellas, Marcela Anjos Martins, Carlos

    Alberto Mandarim-de-Lacerda, Marcia Barbosa Aguila

    PII: S0271-5317(14)00266-8

    DOI: doi: 10.1016/j.nutres.2014.11.006

    Reference: NTR 7423

    To appear in:   Nutrition Research

    Received date: 30 August 2014

    Revised date: 25 November 2014

    Accepted date: 27 November 2014

    Please cite this article as: Bringhenti Isabele, Ornellas Fernanda, Martins MarcelaAnjos, Mandarim-de-Lacerda Carlos Alberto, Aguila Marcia Barbosa, Early hep-atic insult in the offspring of obese maternal mice,   Nutrition Research   (2014), doi:10.1016/j.nutres.2014.11.006

    This is a PDF file of an unedited manuscript that has been accepted for publication.As a service to our customers we are providing this early version of the manuscript.The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production processerrors may be discovered which could affect the content, and all legal disclaimers thatapply to the journal pertain.

    http://dx.doi.org/10.1016/j.nutres.2014.11.006http://dx.doi.org/10.1016/j.nutres.2014.11.006http://dx.doi.org/10.1016/j.nutres.2014.11.006http://dx.doi.org/10.1016/j.nutres.2014.11.006

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    2/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT1

    Early hepatic insult in the offspring of obese maternal mice

    Isabele Bringhenti,

    Fernanda Ornellas,

    Marcela Anjos Martins,

    Carlos Alberto Mandarim-de-Lacerda 1 

    Marcia Barbosa Aguila

    Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center,

    State University of Rio de Janeiro, Brazil

    1 Corresponding author. Laboratory of Morphometry, Metabolism & Cardiovascular Disease,

    Biomedical Center, State University of Rio de Janeiro, Av 28 de setembro 87 fds, Rio de

    Janeiro, RJ, 20551-030 Brazil. Phone: +55.21.2868-8316; Fax 2868-8033.

    Email: [email protected]; URL: www.lmmc.uerj.br. 

    mailto:[email protected]://www.lmmc.uerj.br/http://www.lmmc.uerj.br/mailto:[email protected]

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    3/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT2

    List of abbreviations

    AIN-93G - American Institute of Nutrition to support growth during the pregnancy, lactation,

    and post-weaning periods

    AUC –  Area under the curve

    BM –  Body Mass

    CAT - catalase

    CPT –  Carnitine Palmitoyl-CoA Transferase

    DIO –  diet-induced obesity

    FAS –  Fatty Acid Synthase

    FFA –  Free Fatty Acids

    GPx - Glutathione Peroxidase

    GRS –  Glutathione Reductase

    HF –  High-fat diet

    IR –  insulin resistance

    MnSOD - Manganese Superoxide Dismutase

     NEFA - Non-esterified fatty acids

     NAFLD –  Nonalcoholic Fatty Liver Disease

    OGTT –  Oral Glucose Tolerance Test

    PPAR –  Peroxisome Proliferator Activator Receptor

    RT-qPCR –  Real Time quantitative Polymerase Chain Reaction

    ROS - Reactive oxygen species

    SC –  Standard chow

    SOD –  superoxide dismutase

    TAG –  Triacylglycerol

    VLDL –  Very low-density lipoprotein

    TNF –  Tumor Necrosis Factor

    Vv –  Volume Density

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    4/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT3

    Abstract

    We hypothesized that the maternal obesity initiates metabolic disorders associated with

    oxidative stress in the liver of offspring since early life. Mouse's mothers were assigned into

    two groups according to the diet offered (n=10 per group): standard chow (SC) or high-fat

    diet (HF). The results revealed that HF offspring had an increase in body mass at day 10 (+25  

    %, P

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    5/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT4

    1. Introduction

    Obesity has become a serious public health problem, affecting individuals of all age ranges,

     particularly women in the reproductive stage, setting forth a deleterious maternal nutritional

    status [1, 2].

     Nonalcoholic fatty liver disease (NAFLD) is associated with numerous physiological

    events that are initiated by the increased delivery of nonesterified fatty acids from adipose

    tissue to the liver tissue [3]. This established framework has implications in the pathogenesis

    of liver steatosis because the input of lipid exceeds the capacity of fatty acids oxidation and/or

    export. Although this condition is initially asymptomatic and benign, NAFLD is the

    elementary injury for the development of severe pathology such as liver cirrhosis and

    hepatocellular carcinoma, which may be associated with increased oxidative stress [4, 5].

    Liver mitochondria presents maternal origin and may be an important target in the

    investigation of metabolic disorders in the offspring of obese women, corroborated by the fact

    that this organelle constitutes a critical site of energy metabolism [6]. Accordingly, liver

    mitochondrial dysfunction may be associated with reduced beta oxidation of fatty acids and

    decreases in cytochrome C and carnitine palmitoyl-CoA transferase (CPT)-1 enzyme [7].

    Additionally, mitochondrial dysfunction leads to excessive lipids in the liver and may

     promote the formation of reactive oxygen species, leading to lipid peroxidation,

    inflammation, and fibrosis [8].

    Previous data support the concept that the offspring of diet-induced obese (DIO) dams

    have adverse metabolic programming and develop overweight/obesity and dyslipidemia in

    adulthood [9, 10]. However, such offspring have disrupted liver metabolism and develop

     NAFLD prior to any notable differences in body mass (BM) or body composition [11], which

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    6/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT5

    is a phenomenon known as “liver metabolic imprinting” [12, 13]. Nevertheless, it is important

    to consider whether the insult in the liver due to maternal obesity occurs sufficiently early to

     be detected in the offspring and to allow a preventive intervention.

    Although the literature reported a different model of lipotoxity in the fetal liver using

    nonhuman primates of obese mothers [14], the field of study is considerably complex and

    merits further studies. We were particularly interested here in assessing the hepatic insult in

    the very early life of the progeny of DIO mothers, observing the association between the liver

    metabolism and oxidative stress.

    2. Methods and materials

    2.1 Animals and diet

    This study was performed in strict accordance with the recommendations of the Guide for the

    Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was

    approved by the Committee on the Ethics of Animal Experiments of the State University of

    Rio de Janeiro (Protocol CEUA 053/10), and all efforts were made to minimize suffering.

    Animals were maintained under controlled temperature conditions of 20 ± 2º C; humidity

    60 ± 10 % and 12 h/12 h dark/light cycle, and they had free access to food and water. Thus,

    four-week-old female C57BL/6 mice were randomly assigned into two groups according to

    the diet offered (n=10 per group): standard chow (SC) or high-fat diet (HF). In the SC group,

    64 % of energy was derived from carbohydrates, 19 % of energy from protein and 17 % of

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    7/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT6

    energy from lipids. In the HF group, 32 % of energy was derived from carbohydrates, 19 % of

    energy from protein and 49 % of energy from lipids. The vitamin and mineral contents of

     both diets were identical and followed the standards for rodent diets recommended by the

    American Institute of Nutrition to support growth during the pregnancy, lactation and post-

    weaning periods (AIN-93G) (Table 1) [14].

    After 8 weeks on the two diets, at twelve weeks of age, females SC and HF were mated

    with SC-fed C57BL/6 males of the same age and the females remained on their experimental

    diets throughout pregnancy and lactation. Thus, day 0 of gestation was determined by

    observing the formation of a vaginal plug. Pups were divided according to the maternal diet:

    SC pups were birthed from dams fed SC diet, whereas HF pups were birthed from dams fed

    HF diet. One male from each litter was randomly assigned to each of the experimental groups

    (the sex was assigned according to the anogenital distance) [15] and was studied at postnatal

    day 1 (the birthday) and at postnatal day 10. Figure 1 details the design of the formation of the

    groups. We adjusted the litters that were followed until day 10 randomly to seven pups to

     provide adequate and standardized nutrition in the period. As a consequence, pups were

    divided according to the maternal diets into four groups (n = 5 each group):

    a) SC offspring postnatal day 1,

     b) HF offspring postnatal day 1,

    c) SC offspring postnatal day 10, 

    d) HF offspring postnatal day 10.

    The body mass (BM) of the mothers was measured weekly, during the eight-week before

     pregnancy. The BM of the offspring was measured at day 1 and postnatal day 10.

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    8/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT7

    2.2 Euthanasia and tissue extraction

    The newborns were rapidly decapitated, and the blood was collected immediately. At day 10,

     puppies were separated from their mothers one hour prior to decapitation, then blood was

    collected and the glycemia was measured (Accu-Check Active glucometer; Roche Applied

    Science, SP, Brazil), and the liver was carefully and rapidly dissected, weighed and made to

    further analyzes. Various fragments were frozen for molecular and biochemical analyzes or

    immersed in freshly prepared fixative (1.27 mol/L formaldehyde in 0.1 M phosphate buffer,

     pH 7.2) for 48 h at room temperature.

    2.3 Liver structure and biochemistry

    The fixed liver fragments were then embedded in Paraplast Plus (Sigma-Aldrich Co., St.

    Louis, Mo., USA), sectioned at a nominal thickness of 5 µm and stained with hematoxylin

    and eosin. At least five microscopic fields per animal were analyzed in a blinded way at

    random in digital images (Nikon Eclipse 90i microscope with DS-Ri1 camera, Tokyo, Japan).

    We used a test-system made up of 36 test points (PT) produced with the STEPanizer web-

     based system (www.stepanizer.com) put over the images for the analysis. Briefly, we

    estimated the volume density of liver steatosis using the point-counting technique: Vv[liver,

    steatosis] = Pp[steatosis]/PT[liver] (Pp is the number of points that hit the fat drops into the

    hepatocyte) [16, 17].

    The hepatic triacylglycerol (TAG) levels were measured in 50 mg of frozen liver tissue

     placed in an ultrasonic processor with 1 ml of isopropanol. The homogenate was centrifuged

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    9/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT8

    at 2000 g, and 5 µl of the supernatant was used to measure TAG with a kit and an automated

    spectrophotometer (Bioclin System II, Quibasa Ltda., Belo Horizonte, Brazil).

    2.4 Antioxidants enzymes activity

    Catalase (CAT), glutathione peroxidase (GPx) and manganese superoxide dismutase

    (MnSOD) activities were measured in the liver homogenate of the 10 days old offspring. CAT

    activity was measured in terms of the rate of decrease in hydrogen peroxide concentration at

    240 nm [18]. GPx activity was measured by monitoring the oxidation of NADPH at 340 nm

    in the presence of hydrogen peroxide [19]. MnSOD activity was assayed by measuring the

    inhibition of adrenaline auto-oxidation at 480 nm [20]. The activity of antioxidant enzymes

    was expressed in U/mg protein.

    2.5 RT-qPCR.

    Total RNA was extracted from approximately 50 mg of liver tissue using Trizol reagent

    (Invitrogen, CA, USA). RNA amount was determined using Nanovue (GE Life Sciences)

    spectroscopy, and 1 µg of RNA was treated with DNAse I (Invitrogen). Synthesis of the first

    strand cDNA was performed using Oligo (dT) primers for mRNA and Superscript III reverse

    transcriptase (both Invitrogen). Quantitative real-time PCR (RT-qPCR) was performed using

    a Biorad CFX96 cycler and the SYBR Green mix (Invitrogen). Table 2 shows the primers for

    qPCR as they were designed using the Primer3 online software.

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    10/38

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    11/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT10

    3. Results

    3.1 Maternal body mass

    The BM did not show a significant difference comparing the SC dams with the HF dams

    when the study started (SC, 10.46 ± 0.43 g vs. HF, 10.39 ± 0.34 g). However, after eight

    weeks in the experimental diets (before pregnancy), we found a greater BM in the HF dams

    than in the SC dams (SC, 22.88 ± 0.48 g vs. HF, 29.13 ± 0.22 g; +27 %, P

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    12/38

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    13/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT12

    3.4 Antioxidant enzymes activity

    CAT activity: there was no difference between the SC offspring and the HF offspring in the

     postnatal day 10 (SC, 0.037 ± 0.002 U/mg ptn vs. HF, 0.049 ± 0.008 U/mg ptn; P >0.05).

    GPx-1 activity: there was no difference between the SC offspring and the HF offspring in

    the postnatal day 10 (SC, 9.04 ± 1.20 U/mg ptn vs. HF, 9.24 ± 0.90 U/mg ptn; P >0.05).

    MnSOD activity: in postnatal day 10, the MnSOD activity was lower in the HF offspring

    than in the SC offspring (HF, 17.34 ± 4.78 U/mg ptn vs.SC, 51.32 ± 13.21 U/mg ptn,

     P

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    14/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT13

    the pups accounted for 11.73 % of the variance of the PPAR-gamma expression ( P

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    15/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT14

    ( P

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    16/38

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    17/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT16

    reporting that a developing fetus is highly susceptible to excess lipids, and this may increase

    the risk of NAFLD [31].

    A recent study of the 8-week-old offspring of obese dams showed that the offspring were

    hyperinsulinemic with significantly increased hepatic oxidative damage markers and with

    reduced levels of the antioxidant enzyme GPx-1. Additionally, mitochondrial complex I and

    II activities were elevated, whereas the levels of mitochondrial cytochrome c were

    significantly reduced, and glutamate dehydrogenase was increased, suggesting mitochondrial

    dysfunction. Offspring of obese dams also had significantly greater hepatic lipid content,

    associated with increased levels of PPAR-gamma and reduced triglyceride lipase. Liver

    glycogen and protein content were concomitantly reduced in the offspring of obese dams [11].

    We observed a decrease in mRNA expression of CPT-1 in both ages in this study. CPT-1

    is a regulatory enzyme and a target gene of PPAR-alpha in the mitochondria that transfers

    fatty acids from the cytosol to the mitochondria prior to beta-oxidation [32, 33].

    Consequently, we found the mRNA of FAS to be elevated in both ages of the HF offspring.

    FAS is an intermediate enzyme that utilizes acetyl-CoA and malonyl-CoA to form palmitic

    acid (C16:0) in the process of fatty acid synthesis. Therefore, FAS plays a role in energy

    metabolism and shows augmented levels in association with NAFLD [34, 35].

    The disruption in the mRNA expression of antioxidant enzymes may be associated with

    hepatic damage [36]. Studies have found that oxidative stress may occur because of increased

    reactive oxygen species (ROS) production and/or failure of the antioxidant system. The

    antioxidant system is composed of nonenzymatic molecules and antioxidant enzymes such as

    MnSOD, CAT and GPx1 that react with and inactivate ROS [37]. Notably, a redox-centered

     pathogenesis is associated with NAFLD, whereby the redox state/ROS may modulate lipid

    metabolism and insulin sensitivity [38]. Indeed, oxidative stress has been observed to precede

    insulin resistance [39].

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    18/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT17

    It is important to highlight that epigenetic mechanisms comprise several levels of

    interconnected and interdependent codes, including methylation of DNA, covalent

    modifications of histones, and the gene-regulating and chromatin-organizing activities of non-

    coding RNAs [40]. Such processes change the binding of transcription activators and

    repressors to specific gene promoters, and/or alter the large-scale conformation and function

    of chromatin itself, which modulates gene expression [41]. Additionally, studies have

    associated obesity with DNA hypermethylation of enzymes involved in mitochondrial fatty

    acid oxidation, gluconeogenesis and lipogenesis in the liver, causing silencing of their

    expression and contributing to obesity-induced liver insulin resistance [42, 43].

    Although we did not evaluate mitochondrial dysfunction, a recent study reported an

    increase in mitochondrial complex I and II activities, resulting in an increase in free radical

    generation. The mitochondrial dysfunction associated with reduced antioxidant capacity,

    together with reduced GPx1 expression, contributes to the pro-oxidative environment in the

    offspring of DIO mothers later in life. Furthermore, increased mitochondrial metabolism is

    associated with generated ROS with PPAR-gamma activity and adipocyte differentiation [44].

    Our findings demonstrated a decrease in mRNA expression of catalase, MnSOD and GPx1

    from the day of birth to day 10 after birth. In addition, the analysis of antioxidant enzyme

    activities in the 10 days offspring showed a reduction in the SOD activity, while the activities

    of the CAT and GPx were preserved, despite the reduced mRNA level observed to both of the

    enzymes. An impairment of the hepatocyte antioxidant defense mechanisms may have

    contributed to steatosis development in the offspring since the protective capacity of the

    hepatocytes to inhibit degradation induced by free radicals was reduced [36].

    In conclusion, the message of the current study is that of DIO in maternal mice may induce

    an early oxidative dysfunction coupled with hepatic steatosis in the offspring that is

    measurable since the first days of life. Therefore, this time may represent an interventional

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    19/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT18

    opportunity to prevent liver damage progression and the development of metabolic disorders

    later in life, such as insulin resistance.

    Acknowledgment

    The authors would like to thank Aline Penna and Michele Moura for technical assistance.

    Financial support

    The research was supported by the National council for scientific research (CNPq, Brazil), the

    Rio de Janeiro Foundation for research (Faperj), and the Coordination of Improvement of

    Higher Education Personnel (CAPES). The funding sources had no role in study design; in

    the collection, analysis and interpretation of data; in the writing of the report; and in the

    decision to submit the manuscript for publication.

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    20/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT19

    References

    [1].  Flegal KM, Carroll MD, Kit BK, Ogden CL. Prevalence of obesity and trends in the

    distribution of body mass index among US adults, 1999-2010. JAMA. 2012;307:491-7.

    [2].  Borengasser SJ, Kang P, Faske J, Gomez-Acevedo H, Blackburn ML, Badger TM, et al.

    High fat diet and in utero exposure to maternal obesity disrupts circadian rhythm and

    leads to metabolic programming of liver in rat offspring. PLoS One. 2014;9:e84209.

    [3].  Hughes AN, Oxford JT. A lipid-rich gestational diet predisposes offspring to

    nonalcoholic fatty liver disease: a potential sequence of events. Hepat Med. 2014;6:15-

    23.

    [4].  Paredes AH, Torres DM, Harrison SA. Nonalcoholic fatty liver disease. Clin Liver Dis.

    2012;16:397-419.

    [5].  Shibahara J, Ando S, Sakamoto Y, Kokudo N, Fukayama M. Hepatocellular carcinoma

    with steatohepatitic features: a clinicopathological study of Japanese patients.

    Histopathology. 2014;64:951-62.

    [6].  Bruce KD, Cagampang FR, Argenton M, Zhang J, Ethirajan PL, Burdge GC, et al.

    Maternal high-fat feeding primes steatohepatitis in adult mice offspring, involving

    mitochondrial dysfunction and altered lipogenesis gene expression. Hepatology.

    2009;50:1796-808.

    [7].  Sanyal AJ, Campbell-Sargent C, Mirshahi F, Rizzo WB, Contos MJ, Sterling RK, et al.

     Nonalcoholic steatohepatitis: association of insulin resistance and mitochondrial

    abnormalities. Gastroenterology. 2001;120:1183-92.

    [8].  Serviddio G, Bellanti F, Giudetti AM, Gnoni GV, Capitanio N, Tamborra R, et al.

    Mitochondrial oxidative stress and respiratory chain dysfunction account for liver

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    21/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT20

    toxicity during amiodarone but not dronedarone administration. Free Radic Biol Med.

    2011;51:2234-42.

    [9].  Ornellas F, Mello VS, Mandarim-de-Lacerda CA, Aguila MB. Sexual dimorphism in fat

    distribution and metabolic profile in mice offspring from diet-induced obese mothers.

    Life Sci. 2013;93:454-63.

    [10]. Oben JA, Mouralidarane A, Samuelsson AM, Matthews PJ, Morgan ML, McKee C, et

    al. Maternal obesity during pregnancy and lactation programs the development of

    offspring non-alcoholic fatty liver disease in mice. J Hepatol. 2010;52:913-20.

    [11]. Alfaradhi MZ, Fernandez-Twinn DS, Martin-Gronert MS, Musial B, Fowden A,

    Ozanne SE. Oxidative stress and altered lipid homeostasis in the programming of

    offspring fatty liver by maternal obesity. Am J Physiol Regul Integr Comp Physiol.

    2014;307:R26-34.

    [12]. Burgueno AL, Cabrerizo R, Gonzales Mansilla N, Sookoian S, Pirola CJ. Maternal

    high-fat intake during pregnancy programs metabolic-syndrome-related phenotypes

    through liver mitochondrial DNA copy number and transcriptional activity of liver

    PPAR-GC1A. J Nutr Biochem. 2013;24:6-13.

    [13]. Bayol SA, Simbi BH, Fowkes RC, Stickland NC. A maternal "junk food" diet in

     pregnancy and lactation promotes nonalcoholic Fatty liver disease in rat offspring.

    Endocrinology. 2010;151:1451-61.

    [14]. Reeves PG, Nielsen FH, Fahey GC, Jr. AIN-93 purified diets for laboratory rodents:

    final report of the American Institute of Nutrition ad hoc writing committee on the

    reformulation of the AIN-76A rodent diet. J Nutr. 1993;123:1939-51.

    [15]. Hotchkiss AK, Vandenbergh JG. The anogenital distance index of mice ( Mus musculus

    domesticus): an analysis. Contemp Top Lab Anim Sci. 2005;44:46-8.

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    22/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT21

    [16]. Aguila MB, Pinheiro AR, Parente LB, Mandarim-de-Lacerda CA. Dietary effect of

    different high-fat diet on rat liver stereology. Liver Int. 2003;23:363-70.

    [17]. Catta-Preta M, Mendonca LS, Fraulob-Aquino J, Aguila MB, Mandarim-de-Lacerda

    CA. A critical analysis of three quantitative methods of assessment of hepatic steatosis

    in liver biopsies. Virchows Arch. 2011;459:477-85.

    [18]. Aebi H. Catalase in vitro. Methods Enzymol. 1984;105:121-6.

    [19]. Paglia DE, Valentine WN. Studies on the quantitative and qualitative characterization of

    erythrocyte glutathione peroxidase. J Lab Clin Med. 1967;70:158-69.

    [20]. Bannister JV, Calabrese L. Assays for superoxide dismutase. Methods Biochem Anal.

    1987;32:279-312.

    [21]. Magliano DC, Bargut TC, de Carvalho SN, Aguila MB, Mandarim-de-Lacerda CA,

    Souza-Mello V. Peroxisome proliferator-activated receptors-alpha and gamma are

    targets to treat offspring from maternal diet-induced obesity in mice. PLoS One.

    2013;8:e64258.

    [22]. Sullivan EL, Smith MS, Grove KL. Perinatal exposure to high-fat diet programs energy

     balance, metabolism and behavior in adulthood. Neuroendocrinology. 2011;93:1-8.

    [23]. Srinivasan M, Katewa SD, Palaniyappan A, Pandya JD, Patel MS. Maternal high-fat

    diet consumption results in fetal malprogramming predisposing to the onset of

    metabolic syndrome-like phenotype in adulthood. Am J Physiol Endocrinol Metab.

    2006;291:E792-9.

    [24]. Brion MJ, Ness AR, Rogers I, Emmett P, Cribb V, Davey Smith G, et al. Maternal

    macronutrient and energy intakes in pregnancy and offspring intake at 10 y: exploring

     parental comparisons and prenatal effects. Am J Clin Nutr. 2010;91:748-56.

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    23/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT22

    [25]. Arentson-Lantz EJ, Buhman KK, Ajuwon K, Donkin SS. Excess pregnancy weight gain

    leads to early indications of metabolic syndrome in a swine model of fetal

     programming. Nutr Res. 2014;34:241-9.

    [26]. Gregorio BM, Souza-Mello V, Carvalho JJ, Mandarim-de-Lacerda CA, Aguila MB.

    Maternal high-fat intake predisposes nonalcoholic fatty liver disease in C57BL/6

    offspring. Am J Obstet Gynecol. 2010;203:495 e1-8.

    [27]. Cederroth CR, Nef S. Fetal programming of adult glucose homeostasis in mice. PLoS

    ONE. 2009;4:e7281.

    [28]. Cerf ME, Louw J. High fat programming induces glucose intolerance in weanling

    Wistar rats. Horm Metab Res. 2010;42:307-10.

    [29]. Okamura M, Inagaki T, Tanaka T, Sakai J. Role of histone methylation and

    demethylation in adipogenesis and obesity. Organogenesis. 2010;6:24-32.

    [30]. Shankar K, Kang P, Harrell A, Zhong Y, Marecki JC, Ronis MJ, et al. Maternal

    overweight programs insulin and adiponectin signaling in the offspring. Endocrinology.

    2010;151:2577-89.

    [31]. McCurdy CE, Bishop JM, Williams SM, Grayson BE, Smith MS, Friedman JE, et al.

    Maternal high-fat diet triggers lipotoxicity in the fetal livers of nonhuman primates. J

    Clin Invest. 2009;119:323-35.

    [32]. Huang YY, Gusdon AM, Qu S. Nonalcoholic fatty liver disease: molecular pathways

    and therapeutic strategies. Lipids Health Dis. 2013;12:171.

    [33]. Lee HJ, Choi SS, Park MK, An YJ, Seo SY, Kim MC, et al. Fenofibrate lowers

    abdominal and skeletal adiposity and improves insulin sensitivity in OLETF rats.

    Biochem Biophys Res Commun. 2002;296:293-9.

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    24/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT23

    [34]. Kohjima M, Enjoji M, Higuchi N, Kato M, Kotoh K, Yoshimoto T, et al. Re-evaluation

    of fatty acid metabolism-related gene expression in nonalcoholic fatty liver disease. Int

    J Mol Med. 2007;20:351-8.

    [35]. Morgan K, Uyuni A, Nandgiri G, Mao L, Castaneda L, Kathirvel E, et al. Altered

    expression of transcription factors and genes regulating lipogenesis in liver and adipose

    tissue of mice with high fat diet-induced obesity and nonalcoholic fatty liver disease.

    Eur J Gastroenterol Hepatol. 2008;20:843-54.

    [36]. Rolo AP, Teodoro JS, Palmeira CM. Role of oxidative stress in the pathogenesis of

    nonalcoholic steatohepatitis. Free Radic Biol Med. 2012;52:59-69.

    [37]. Roberts CK, Sindhu KK. Oxidative stress and metabolic syndrome. Life Sci.

    2009;84:705-12.

    [38]. Serviddio G, Bellanti F, Vendemiale G. Free radical biology for medicine: learning

    from nonalcoholic fatty liver disease. Free Radic Biol Med. 2013;65:952-68.

    [39]. Matsuzawa-Nagata N, Takamura T, Ando H, Nakamura S, Kurita S, Misu H, et al.

    Increased oxidative stress precedes the onset of high-fat diet-induced insulin resistance

    and obesity. Metabolism. 2008;57:1071-7.

    [40]. Goldberg AD, Allis CD, Bernstein E. Epigenetics: a landscape takes shape. Cell.

    2007;128:635-8.

    [41]. Gluckman PD, Hanson MA, Buklijas T, Low FM, Beedle AS. Epigenetic mechanisms

    that underpin metabolic and cardiovascular diseases. Nat Rev Endocrinol. 2009;5:401-

    8.

    [42].  Jiang M, Zhang Y, Liu M, Lan MS, Fei J, Fan W, et al. Hypermethylation of hepatic

    glucokinase and L-type pyruvate kinase promoters in high-fat diet-induced obese rats.

    Endocrinology. 2011;152:1284-9.

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    25/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT24

    [43]. Sookoian S, Rosselli MS, Gemma C, Burgueno AL, Fernandez Gianotti T, Castano GO,

    et al. Epigenetic regulation of insulin resistance in nonalcoholic fatty liver disease:

    impact of liver methylation of the peroxisome proliferator-activated receptor gamma

    coactivator 1alpha promoter. Hepatology. 2010;52:1992-2000.

    [44]. Tormos KV, Anso E, Hamanaka RB, Eisenbart J, Joseph J, Kalyanaraman B, et al.

    Mitochondrial complex III ROS regulate adipocyte differentiation. Cell Metab.

    2011;14:537-44.

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    26/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT25

    Figure legends

    Figure 1. Sampling design for the study. For the composition of experimental groups, one

    male pup of each litter was considered in this study, totaling a sample of five animals per

    group. 

    Figure 2. Body mass.

    Data are presented as means and standard error of the means (n = 5 per group). Significant

    differences are indicated (one-way ANOVA and post-hoc Holm-Sydak tests).

    Abbreviations: SC, standard chow group; HF, high-fat diet group.

    Symbols: †, significantly different from SC counterpart; ‡, different from day 1. 

    Figure 3. Blood glucose levels.

    Data are presented as means and standard error of the means (n = 5 per group). Significant

    differences are indicated (one-way ANOVA and post-hoc Holm-Sydak tests).

    Abbreviations: SC, standard chow group; HF, high-fat diet group.

    Symbols: †, significantly different from SC counterpart; ‡, significantly different from day 1.

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    27/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT26

    Figure 4. Photomicrographs of liver tissue (hematoxylin-eosin stain, same magnification for

    all photomicrographs): (A) SC day 1; (B) HF day 1; (C) SC day 10; (D) HF day 10; (E)

    triacylglycerol content; and (F) volume density of steatosis.

    Data are presented as means and standard error of the means (n = 5 per group). Significant

    differences are indicated (one-way ANOVA and post-hoc Holm-Sydak tests).

    Abbreviations: SC, standard chow group; HF, high-fat diet group.

    Symbols: †, significantly different from SC counterpart.

    Figure 5. Correlation of liver steatosis (variable y) and liver triacylglycerol levels (variable x).

    The coefficient of correlation of Pearson, r, and P  values are indicated.

    Abbreviations: SC, standard chow group; HF, high-fat diet group. 

    Figure 6. Relative mRNA expression of FAS, PPAR-gamma, CPT1 and PPAR-alpha.

    Data are presented as means and standard error of the means (n = 5 per group). Significant

    differences are indicated (one-way ANOVA and post-hoc Holm-Sydak tests). (A) FAS

    relative expression; (B) PPAR-gamma relative expression; (C) CPT-1 relative expression; (D)

    PPAR-alpha relative expression. An endogenous control (beta-actin) was used to normalize

    the expression of the selected genes.

    Abbreviations: SC, standard chow group; HF, high-fat diet group. FAS, fatty acid synthase;

    PPAR, peroxisome-proliferator activator receptor; and CPT, carnitine palmitoyltransferase.

    Symbols: †, significantly different from SC counterpart; ‡, significantly different from day 1. 

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    28/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT27

    Figure 7. Relative mRNA expression of catalase, MnSOD, GPx-1 and GRS An endogenous

    control (beta-actin) was used to normalize the expression of the selected genes.

    Data are presented as means and standard error of the means (n = 5 per group). Significant

    differences are indicated (one-way ANOVA and post-hoc Holm-Sydak tests). (A) Catalase

    relative expression; (B) MnSOD relative expression; (C) GPx-1 relative expression; (D) GRS

    relative expression.

    Abbreviations: SC, standard chow group; HF, high-fat diet group; MnSOD, manganese

    superoxide dismutase; GPx, glutathione peroxidase; GRS, glutathione reductase.

    Symbols: †, significantly different from SC counterpart; ‡, significantly different from day 1. 

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    29/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT28

    Figure 1

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    30/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT29

    Figure 2

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    31/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT30

    Figure 3

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    32/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT31

    Figure 4

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    33/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT32

    Figure 5

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    34/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT33

    Figure 6

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    35/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT34

    Figure 7

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    36/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT35

    Table 1. Ingredient and proximate analysis of diets fed to mice

    Abbreviations: SC: standard chow; HF: high-fat diet.

    Nutrients (g/kg) SC HF

    Casein 190.00 230.00

    Corn starch 539.50 299.50

    Sucrose 100.00 100.00

    Soybean oil 70.00 70.00

    Lard - 200.00

    Fiber 50.00 50.00

    Vitamin Mixture (mg) 10.00 10.00

    Mineral Mixture (mg) 35.00 35.00

    Cysteine 3.00 3.00

    Choline 2.50 2.50

    Antioxidants 0.01 0.01

    Total mass (g) 1.00 1.00

    Energy (kcal/kg) 3.95 4.95

    Carbohydrates (%) 64.00 32.00

    Protein (%) 19.00 19.00

    Lipids (%) 17.00 49.00

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    37/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT36

    Table 2. RT-qPCR primers and respective sequences

    Abbreviations: Carnitine palmitoyltransferase I (CPT-1); Catalase (CAT); Fatty acid synthase

    (FAS); Glutathione peroxidase (GPx1); Glutathione reductase (GSR); Manganese Superoxide

    Dismutase (MnSOD); Peroxisome proliferator activator receptor (PPAR  ).

    Name 5-3’  Primers

    FAS FW CACCCACTGGAAGCTGGTAT

    FAS RV TCGAGGAAGGCACTACACCT

    CPT-1 FW AAGGAATGCAGGTCCACATC

    CPT-1 RV CCAGGCTACAGTGGGACATT

    PPAR alpha  FW TCGAGGAAGGCACTACACCT

    PPAR alpha  RV TCTTCCCAAAGCTCCTTCAA

    PPAR  gamma2  FW ACGATCTGCCTGAGGTCTGT

    PPAR  gamma2  RV CATCGAGGACATCCAAGACA

    MnSOD FW GGTCTCCAACATGCCTCTCT

    MnSOD RV AACCATCCACTTCGAGCAGA

    GPx1

    GPx1

    FW

    RV

    TCTGCAGATCGTTCATCTCG

    GTCCACCGTGTATGCCTTCT

    CAT

    CAT

    FW

    RV

    CAAGTTTTTGATGCCCTGGGT

    ACATGGTCTGGGACTTCTGG

    GSR

    GSR

    FW

    RV

    CAGCATAGACGCCTTTGACA

    CACGACCATGATTCCAGATG

    Beta-actin FW CTCCGGCATGTGCAA

    Beta-actin RV CCCACCATCACACCCT

  • 8/17/2019 2014 Early Hepatic Insult in the Offspring of Obese Maternal Mice

    38/38

       A   C   C    E    P

        T    E    D

         M   A    N    U    S   C    R    I    P    T

    ACCEPTED MANUSCRIPT37

    Table 3. Two-way ANOVA analyzing high-fat diet and age.

    Abbreviations: CAT (catalase), CPT (carnitine palmitoyl transferase), FAS (fatty acid

    synthase), GPx (glutathione peroxidase), GRS (glutathione reductase), MnSOD (manganese

    superoxide dismutase), PPAR (peroxisome-proliferator activator receptor), TAG

    (triacylglycerol). 

    Two-Way ANOVA 

    % of the variation and significance test 

    Interaction Diet Age

    %  P≤   %  P≤   %  P≤  

    Body Mass 1.63 0.0001 2.25 0.0001 95.65 0.0001

    CAT 1.24 NS 67.76 0.0001 1.38 NS

    CPT-1 0.38 NS 62.94 0.0001 2.30 NS

    FAS  4.29 NS 52.22 0.0001 10.91 0.05

    Glycemia 2.59 0.01 4.81 0.001 88.77 0.0001

    GPx-1 22.87 0.0001 22.87 0.0001 39.68 0.0001

    GRS 12.36 NS 34.81 0.01 0.97 NS

    Hepatic TAG 0.29 NS 69.66 0.0001 0.44 NS

    Liver steatosis  0.87 NS 79.23 0.0001 0.13 NS

    MnSOD 4.86 NS 62.47 0.0001 9.54 0.05

    PPAR-alpha 4.69 NS 55.04 0.001 2.53 NS

    PPAR-gamma 0.057 NS 61.51 0.0001 11.73 0.05