Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 May 1.
Published in final edited form as: Placenta. 2009 Apr 11;30(5):411–417. doi: 10.1016/j.placenta.2009.03.002

Placental Gene Expression Responses to Maternal Protein Restriction in the Mouse

Ciprian P Gheorghe 1, Ravi Goyal 1, Joshua D Holweger 1, Lawrence D Longo 1
PMCID: PMC2674533  NIHMSID: NIHMS104534  PMID: 19362366

Abstract

OBJECTIVE

Maternal protein restriction has been shown to have deleterious effects on placental development, and has long-term consequences for the progeny. We tested the hypothesis that, by the use of microarray technology, we could identify specific genes and cellular pathways in the developing placenta that are responsive to maternal protein deprivation, and propose a potential mechanism for observed gene expression changes.

METHODS

We fed pregnant FVB/NJ mice from day post coitum 10.5 (DPC10.5) to DPC17.5, an isocaloric diet containing 50% less protein than normal chow. We used the Affymetrix Mouse 430A_2.0 array to measure gene expression changes in the placenta. We functionally annotated the regulated genes, and examined over-represented functional categories and performed pathway analysis. For selected genes, we confirmed the microarray results by use of qPCR.

RESULTS

We observed 244 probe sets, corresponding to 235 genes, regulated by protein restriction (p < 0.001), with ninety-one genes being up-regulated, and 153 down-regulated. Up-regulated genes included those involved in the p53 pathway, apoptosis, negative regulators of cell growth, negative regulators of cell metabolism and genes related to epigenetic control. Down-regulated genes included those involved in nucleotide metabolism.

CONCLUSIONS

Microarray analysis has allowed us to describe the genetic response to maternal protein deprivation in the mouse placenta. We observed that negative regulators of cell growth and metabolism in conjunction with genes involved in epigenesis were up-regulated, suggesting that protein deprivation may contribute to growth restriction and long-term epigenetic changes in stressed tissues and organs. The challenge will be to understand the cellular and molecular mechanisms of these gene expression responses.

Introduction

Successful placental development is crucial for optimal growth, maturation, and survival of the embryo/fetus. The placenta, a fetomaternal organ joining mother and offspring during pregnancy in mammals, serves as an endocrine organ in the “maternal-placental-fetal” complex, in addition to its role in the exchange of respiratory gases, exchange of nutrients, an immunologic barrier, and other functions. As has been recognized for many years, deviation in the normal gene expression pattern may lead to altered placental phenotype, as well as a modified phenotype of the conceptus. Previously, we have examined developmental gene expression patterns in the developing murine placenta, and reported numerous placental genes are up- or down-regulated to a significant degree, and that specific functional groups of genes are regulated at the different developmental ages [1] and with maternal hypoxia [2]. However, a number of stressors during gestation can lead to altered placental and fetal growth and development. One of the important stressors is maternal malnutrition, which during pregnancy may have deleterious consequences for the progeny. Historical data point to these effects in human populations. For instance, during WWII, the people of both Holland and Russia were subjected to severe dietary restrictions due to interdiction of food supplies by the German army [3]. The children born under these conditions not only were small for gestational age, but they also developed significant health problems later in life [4, 5]. Several major sequelae have been described including those of the cardiovascular system, type II diabetes, and mood and personality disorders [6].

Nutritional deprivation influences not only placental growth and morphology, but also alters the hormonal milieu of the developing fetus, and causes subsequent cardiovascular, hormonal and behavioral consequences in the adult [7, 8]. These epidemiologic observations have led to speculation regarding the mechanism of changes in the placenta, and their effects on the developing fetus. The observations made in human subjects have been confirmed in several animal models. An important question, is the extent to which these observed effects result from an overall caloric restriction, as opposed to a qualitative component in the diet that triggers the responses. Evidence from several animal models points to protein deprivation as a major factor in these defects [9]. For example, in the rat the growth reducing effects of a low calorie diet can only be reversed by a dietary increase in protein levels; vitamin supplements, and caloric increases, while carbohydrates failed to reverse the observed effects [10]. Other studies have revealed that dietary amino acid balance is a key mediator of some of the cardiovascular and metabolic effects observed in response to protein deprivation [9]. However, no studies have examined the global changes in the placental gene expression with maternal protein restriction. We thus tested the hypothesis that, by the use of microarray technology, we could identify specific genes and cellular pathways in the developing placenta that are responsive to maternal protein deprivation, and propose a potential mechanism for phenotypic changes that have been observed.

Materials and Methods

Animals

Eight-week old FVB/NJ male and female mice were obtained from the Jackson Laboratories (Bar Harbor, ME) and housed at the Animal Research Facility, Loma Linda University, Loma Linda, CA under conditions of 14 h light, 10 h darkness, ambient temperature of 20°C, and relative humidity of 30-60%. All experimental protocols were in compliance with the Animal Welfare Act, the National Institutes of Health Guide for the Care and Use of Animals, and were approved by the Institutional Animal Care and Use Committee of Loma Linda University.

Breeding and tissue collection

Mice were bred by overnight monogamous pairing of virgin females with a male, the male was removed in the morning, and that day was considered 0.5 day post coitum (0.5 dpc). Mice were weighed daily and pregnancy was confirmed by examining vaginal plugs on day 0.5 and weight gain by 10.5 dpc. At 17.5 dpc the pregnant females were euthanized. The uterus was removed rapidly and placed in a petri dish containing RNA Later solution (Ambion, Austin, TX). Entire placentae were isolated under a dissection microscope and maternal deciduas and endometrial tissues were removed. The isolated and cleaned placentae were snap frozen in liquid nitrogen, and stored at -80 °C for later analysis. RNA was isolated from the entire placentae using the TRIZOL reagent kit (Life Technologies, Rockville, MD), and was stored at -80 °C until further analysis. We confirmed the developmental stages of the embryos by visual inspection according to a modified Theiler staging system [11]. Details of the staging system are available online at http://genex.hgu.mrc.ac.uk/Databases/Anatomy/MAstaging.html.

Protein restriction

The mice were initially fed a normal mouse chow (20% protein content by weight, diet # TD91352). At 0.5 dpc the pregnant mice were divided into two groups, one group (n=3) were continued on normal mouse chow (control) and another group (n=3) were switched to a custom protein diet (10% protein by weight, diet # TD92208) (Teklad, Indianapolis, IA). The 50% protein deprivation was continued from 10.5 dpc to 17.5 dpc (total 7 days.). Studies in several species suggest that severe protein reduction leads to fetal programming of adulthood diseases in the offspring such as hypertension, schizophrenia, behavioral abnormalities etc [12-14]. Studies also indicate that maternal protein deprivation causes altered gene expression in different organs during different time points in the offspring lifespan and lead to these disorders. However, changes in the placental gene expression with this degree of protein deprivation are unknown, and were the focus of present study. The timing of the protein restriction was chosen in order to avoid interfering with fertilization and implantation of the embryo. We also sought to focus on the mature placenta, as in the mouse the allantoic fusion does not occur until 8 dpc and the placenta is not fully formed until 10.5 dpc. The diets were designed to ensure that mice would receive the same amount of calories and nutrients, but a reduced amount of protein. Maternal food intake and maternal weights were measured daily in order to assure isocaloric food intake.

Probe preparation, microarray hybridization, and data analysis

The RNA was processed for use on the Affymetrix Mouse 430A_2.0 array (Affymetrix, Santa Clara, CA) according to the manufacturer’s instructions. Briefly, 5 μg of total RNA was reverse transcribed to double stranded cDNA (Superscript II kit, Life Technologies). The double stranded cDNA was used in an in-vitro transcription reaction to generate biotynilated cRNA probes. The cRNA probes were purified, fragmented, and hybridized to the Affymetrix chip. Washes and staining were performed in an Affymetrix Gene Chip Fluidics station 400. The Affymetrix arrays were scanned using a Gene Array Scanner (Hewlett Packard, Austin, TX), and processed at the Microarray Facility, University of California Irvine, (Irvine, CA). The hybridizations were performed in triplicate for control and protein restricted conditions. All the placentas obtained from one mouse were pooled, and the total RNA isolated was considered as one RNA sample. Six such RNA samples, three each from protein restricted and control mice dams were used for microarray hybridization. Analyses were performed using BRB ArrayTools developed by Dr. Richard Simon and Amy Peng Lam (http://linus.nci.nih.gov/BRBArrayTools.html). We analyzed the data using the random variance method at a significance of p < 0.001 [15]. The genes were assigned to functional classes based on the GO database (http://www.geneontology.org/GO.annotation.html ), and significantly over-represented GO categories in the gene sets were analyzed using the Gene Ontology Tree Machine (http://genereg.ornl.gov/gotm/). We also manually functionally annotated genes using Pubmed searches.

Real Time PCR

In an effort to validate the results of the microarray analysis, we chose several genes that were shown to be regulated by gestational protein restriction for analysis using real time PCR. RNA was isolated from mice different than the ones used for the microarray (n=5). Exon spanning primers were designed using the Universal Probe Library Assay Design Center (Roche, Indianapolis, IN). The primers were synthesized by Integrated DNA technologies (Coralville, CA). The primer sequences selected are shown in Table 1. Total RNA (1 μg per reaction) was reverse transcribed using random hexamers and the SuperScript II reverse transcriptase kit (Invitrogen, Carlsbad, CA). Relative expression was normalized to 18S RNA and fold changes were calculated using the ΔΔCt method. Samples were analyzed on the Roche LightCycler 1.5 (Roche, Indianapolis, IN).

Table 1.

Primers used for qPCR to verify expression of selected genes

Gene Name Accession Number Forward Primer (Position) Reverse Primer (Position) Amplicon Length
Rai 17 NM_183208 gagacaagttcacccccaag (753-772) ggccaagttcttcacacca (794-812) 60
p53 NM_001127233 gcccatgctacagaggagtc (1206-1225) agactggcccttcttggtct (1257 - 1276) 72
Cebpa NM_007678 ccttcaacgacgagttcctg (331 - 350) tggccttctcctgctgtc (373 - 390) 60
Jmy NM_021310 aagggctatgaagaggtgctt (1341 - 1361) ctttctatagtcttgtgcttgtcca (1393 - 1417) 77
Hipk2 NM_010433 cagcagtgacaccgatgaag (2806 - 2825) tctttgcttggagactgtgc (2853 - 2872) 67
18S NR_003278.1 ctcaacacgggaaacctcac (1247-1266) cgctccaccaactaagaacg (1337-1356) 110

Results

In response to protein deprivation the placental weights remained unchanged while pup weights were significantly reduced (p< 0.05) as shown in Figure 1.

Figure 1.

Figure 1

Pup and placental weights at 17.5 dpc after 50% protein restriction (* p < 0.05).

To evaluate the genetic response to protein deprivation we used the Affymetrix Mouse 430A_2.0 oligonucleotide array to compare gene expression levels between normal placentae at 17.5 dpc, and those from pregnancies in which the mothers were exposed to seven days of protein deprivation. Of 22,690 genes examined by on the microarray, using the random variance model [15], we observed 244 probe sets, corresponding to 235 genes, that were influenced by protein restriction (p < 0.001; some probe sets hybridize to different areas of the same gene. This is a design of the Affymetrix chip which serves as an internal control). As a consequence of maternal protein deprivation, 91 of these probe sets were up-regulated, while 153 were down-regulated. As noted in Table 2, among the gene ontology classes most over-represented in the up-regulated group, were regulators of apoptosis (Bcl2-like 2, p53, endophilin, Fas-activated serine/threonine kinase), negative regulators of cell growth (farnesyltransferase CAAX box beta, cadherin 5, CCAAT/enhancer binding protein (C/EBP) alpha, inositol polyphosphate-5-phosphatase D, p53), and negative regulators of cellular metabolism (nuclear receptor co-repressor 2, histone deacetylase 7A, SPEN homolog, transcriptional regulator). A number of genes involved in the p53 pathway were up-regulated. The genes rai17 and hipk2 were up-regulated, both of which are activators of p53. Rai17 induces the expression of p53 and is a cofactor of p53-mediated gene regulation [16]. Hipk2 is a kinase that phosphorylates Serine 46 on the p53 protein and activates its pro-apoptotic effects [17]. We also noted up-regulation of the gene jmy, a co-factor of p53. Jmy is up-regulated in response to DNA damage and binds to p53 in a protein complex that enhances its activity [18]. In addition we noted the up-regulation of two genes, Cebpa and Inpp5d, which are induced by p53. Cebpa is a leucine zipper transcription factor involved in the terminal differentiation of several cell types. It is up-regulated in response to UV radiation, and serves as a DNA damage induced G1 checkpoint in the cell [19]. Inpp5d is a phosphatase involved in inositol-mediated signaling, and has a potential anti-survival effect on the cell. It has been identified as a p53 transcriptional target [20].

Table 2.

Genes Up-regulated by protein deprivation

Fold Change Probe Set Gene Description
1.86 1451895_a_at 24-dehydrocholesterol reductase
1.776 1427385_s_at actinin, alpha 1
1.435 1433477_at active BCR-related gene
1.648 1419140_at activin receptor IIB
1.378 1423973_a_at ADP-ribosylation factor 3
1.728 1438501_at ribosomal protein S17
1.9 1423556_at aldo-keto reductase family 1, member B7
1.745 1424956_at AT hook, DNA binding motif, containing 1
1.463 1423526_at AT rich interactive domain 3B (Bright like)
1.532 1425227_a_at ATPase, H+ transporting, lysosomal V0 subunit a isoform 1
1.411 1427565_a_at ATP-binding cassette, sub-family C (CFTR/MRP), member 5
1.537 1430453_a_at Bcl2-like 2
1.723 1423816_at CAAX box 1 homolog B (human)
2.539 1422047_at cadherin 5
1.628 1451896_a_at calcium homeostasis endoplasmic reticulum protein
1.504 1417175_at casein kinase 1, epsilon
1.603 1422450_at catenin (cadherin associated protein), delta 1
1.538 1418982_at CCAAT/enhancer binding protein (CEB/P), alpha
2.01 1451359_at cDNA sequence BC005662
1.421 1424726_at cDNA sequence BC014685
1.635 1460034_at cDNA sequence BC042901
1.467 1419833_s_at centaurin, delta 3
1.433 1448274_at complement component 1, q subcomponent binding protein
1.52 1425204_s_at DEAD (Asp-Glu-Ala-Asp) box polypeptide 19a
1.743 1421143_at diaphanous homolog 1 (Drosophila)
1.637 1450475_at distal-less homeobox 3
1.376 1419502_at DNA segment, Chr 11, Lothar Hennighausen 1, expressed
1.416 1421032_a_at DnaJ (Hsp40) homolog, subfamily B, member 12
1.893 1452100_at Dullard homolog (Xenopus laevis)
1.497 1418648_at EGL nine homolog 3 (C. elegans)
1.461 1425788_a_at enoyl Coenzyme A hydratase domain containing 2
1.64 1427039_at epsin 1
1.454 1452273_at expressed sequence AA409316
1.64 1424428_at expressed sequence AI225782
1.631 1420602_a_at extraembryonic, spermatogenesis, homeobox 1
1.937 1434309_at farnesyltransferase, CAAX box, beta
1.439 1460635_at Fas-activated serine/threonine kinase
1.553 1449849_a_at F-box and leucine-rich repeat protein 6
1.395 1423442_a_at F-box and WD-40 domain protein 2
1.496 1418396_at G-protein signalling modulator 3 (AGS3-like, C. elegans)
1.457 1424101_at heterogeneous nuclear ribonucleoprotein L
1.728 1417637_a_at high mobility group 20 B
1.618 1420813_at histone deacetylase 7A
1.825 1422799_at HLA-B associated transcript 2
2.829 1429566_a_at homeodomain interacting protein kinase 2
1.576 1424195_a_at inositol polyphosphate-5-phosphatase D
2.023 1418265_s_at interferon regulatory factor 2
1.378 1448759_at interleukin 2 receptor, beta chain
1.879 1448668_a_at interleukin-1 receptor-associated kinase 1
1.572 1452327_at IQ motif and Sec7 domain 1
1.515 1426873_s_at junction plakoglobin
1.497 1420639_at junction-mediating and regulatory protein
1.548 1428881_at kinesin 2
1.422 1451066_at leukocyte receptor cluster (LRC) member 4
1.551 1426648_at MAP kinase-activated protein kinase 2
1.625 1456618_at MAP/microtubule affinity-regulating kinase 4
1.493 1437226_x_at MARCKS-like 1
1.519 1456702_x_at methionine adenosyltransferase II, alpha
1.392 1417857_at methylmalonic aciduria (cobalamin deficiency) type A
1.56 1417472_at myosin, heavy polypeptide 9, non-muscle
1.438 1452254_at myotubularin related protein 9
1.446 1428367_at N-deacetylase/N-sulfotransferase (heparan glucosaminyl) 1
1.963 1448893_at nuclear receptor co-repressor 2
2.067 1424952_at OCIA domain containing 1
2.138 1423967_at paralemmin
2.745 1427550_at paternally expressed 10
1.918 1449851_at period homolog 1 (Drosophila)
1.58 1438938_x_at prohibitin 2
1.662 1418078_at proteaseome (prosome, macropain) 28 subunit, 3
1.658 1418341_at RAB4A, member RAS oncogene family
1.707 1450903_at RAD23b homolog (S. cerevisiae)
2.93 1418535_at ral guanine nucleotide dissociation stimulator,-like 1
1.585 1425266_a_at RAP1, GTP-GDP dissociation stimulator 1
5.137 1455984_at retinoic acid induced 17
1.888 1438507_x_at ribosomal protein L14
1.613 1416033_at RIKEN cDNA 1110006I15 gene
1.909 1431213_a_at RIKEN cDNA 1300007C21 gene
2.844 1460609_at RIKEN cDNA 2810013E07 gene
1.922 1431753_x_at RIKEN cDNA 2900073H19 gene
1.677 1427979_at RIKEN cDNA 4732418C07 gene
1.786 1425000_s_at RIKEN cDNA 5430407P10 gene
1.863 1437180_at RIKEN cDNA 6530403A03 gene
1.411 1448251_at RIKEN cDNA 9030425E11 gene
1.775 1452351_at RIKEN cDNA C030027K23 gene
1.378 1436906_at ring finger protein 166
2.115 1450918_s_at Rous sarcoma oncogene
1.717 1434438_at SAM domain and HD domain, 1
1.422 1416294_at secretory carrier membrane protein 3
1.902 1422167_at semaphoring 5A
1.555 1418011_a_at SH3-domain GRB2-like B1 (endophilin)
1.891 1449005_at solute carrier family 16 (monocarboxylic acid transporters), member 3
1.954 1452139_at solute carrier family 35, member C1
1.872 1418326_at solute carrier family 7 (cationic amino acid transporter, y+system), member 5
1.713 1419329_at sorbin and SH3 domain containing 3
1.768 1420397_a_at SPEN homolog, transcriptional regulator (Drosophila)
1.808 1425787_a_at synaptotagmin-like 3
1.871 1423616_at TAR (HIV) RNA binding protein 2
1.603 1420593_a_at TEA domain family member 3
1.612 1451586_at testis enhanced gene transcript
1.57 1452768_at testis expressed gene 261
1.475 1419954_s_at testis expressed gene 27
1.828 1421148_a_at Tial1 cytotoxic granule-associated RNA binding protein-like 1
2.282 1460743_at tigger transposable element derived 5
1.371 1426691_at tight junction associated protein 1
1.471 1426610_a_at transcription termination factor 1
1.649 1426538_a_at transformation related protein 53
1.759 1448412_a_at TSC22 domain family 4
1.991 1451771_at two pore channel 1
1.658 1448370_at Unc-51 like kinase 1 (C. elegans)
2.773 1423726_at vesicle amine transport protein 1 homolog (T californica)
1.878 1420834_at vesicle-associated membrane protein 2
1.722 1448121_at WW domain binding protein 2
1.631 1423750_a_at Zinc finger protein 162, mRNA (cDNA clone MGC:7095 IMAGE:3157495)
2.533 1418670_s_at Unknown

Overall, the present study shows that the major pathways up-regulated with maternal protein deprivation are the p53 pathway, regulators of apoptosis, negative regulators of cell growth and metabolism and certain epigenetic regulators such as histone deacetylases, methionine adenosyl-transferase II alpha. In contrast, as noted in Table 3, among down-regulated genes, particularly striking were those genes related to nucleotide metabolism, and certain epigenetic regulators such as histone 2, Mcm6 and telomeric repeat binding factor 1. The major placental gene pathways up- or down- regulated by maternal protein deprivation. We verified the expression of Cebpa, p53, Rai17, Jmy, Hipk2 and Inpp5d by the use of real-time qPCR (Table 4). The expression of these genes were altered to similar extent as observed during our Microarray analysis.

Table 3.

Genes down-regulated by protein deprivation

Fold Change Probe Set Gene Description
0.463 1422716_a_at acid phosphatase 1, soluble
0.697 1438170_x_at adhesion regulating molecule 1
0.637 1423781_at amyloid beta precursor protein binding protein 1
0.655 1437688_x_at ATPase, H+ transporting, lysosomal accessory protein 2
0.724 1451223_a_at basic transcription factor 3-like 4
0.728 1450732_a_at bicaudal D homolog 2 (Drosophila)
0.636 AFFX-BioB-M_at Biotin synthase /// biotin synthesis, sulfur insertion?
0.585 1436885_a_at calcium homeostasis endoplasmic reticulum protein
0.663 1437670_x_at CD151 antigen
0.508 1451232_at CD151 antigen
0.584 1425646_at cDNA sequence BC016495
0.65 1419403_at cDNA sequence BC017612
0.724 1423682_a_at cell division cycle associated 4
0.642 1436390_a_at chloride channel CLIC-like 1
0.577 1433718_a_at chromobox homolog 1 (Drosophila HP1 beta)
0.498 1436838_x_at coactosin-like 1 (Dictyostelium)
0.51 1454781_x_at COMM domain containing 9
0.398 1437982_x_at COX15 homolog, cytochrome c oxidase assembly protein (yeast)
0.717 1434705_at C-terminal binding protein 2, mRNA (cDNA clone MGC:27651 IMAGE:4511826)
0.655 1454149_a_at cyclin L2
0.605 1438371_x_at DEAD (Asp-Glu-Ala-Asp) box polypeptide 5
0.673 AFFX-r2-Ec-bioD-3_at dethiobiotin synthetase
0.631 1419915_at DNA segment, Chr 10, ERATO Doi 438, expressed, mRNA (cDNA clone MGC:7199 IMAGE:3482163)
0.637 1449339_at DNA segment, Chr 10, ERATO Doi 641, expressed
0.638 1429411_a_at enhancer of yellow 2 homolog (Drosophila)
0.566 1416236_a_at epithelial V-like antigen 1
0.652 1424013_at eukaryotic translation termination factor 1
0.657 1439411_a_at exportin 7
0.596 1455912_x_at Expressed sequence AW538196 (AW538196), mRNA
0.679 1434108_at F-box protein 11
0.656 1452247_at fragile X mental retardation gene 1, autosomal homolog
0.628 1439150_x_at GH regulated TBC protein 1, mRNA (cDNA clone MGC:27905 IMAGE:3500563)
0.641 1419072_at glutathione S-transferase, mu 7
0.697 1424030_at grainyhead-like 1 (Drosophila)
0.483 1416855_at growth arrest specific 1
0.531 1418106_at hairy/enhancer-of-split related with YRPW motif 2
0.522 1434047_x_at heterogeneous nuclear ribonucleoprotein A2/B1
0.36 1437099_x_at Heterogeneous nuclear ribonucleoprotein F, mRNA
0.611 1422155_at (cDNA clone MGC:36543 IMAGE:4950131) histone 2, H3c2
0.618 1455777_x_at hydroxysteroid (17-beta) dehydrogenase 4
0.438 1455930_at hypothetical LOC433566 /// hypothetical LOC434498
0.486 1423033_at intergral membrane protein 1
0.555 1449099_at LPS-responsive beige-like anchor
0.712 1416343_a_at lysosomal membrane glycoprotein 2
0.44 1455978_a_at matrilin 2
0.686 1434378_a_at Max dimerization protein 4, mRNA (cDNA clone MGC:19425 IMAGE:3490469)
0.651 1422498_at melanoma antigen, family H, 1
0.597 1438852_x_at minichromosome maintenance deficient 6 (MIS5 homolog, S. pombe) (S. cerevisiae)
0.565 1455787_x_at multiple inositol polyphosphate histidine phosphatase 1
0.596 1456381_x_at myeloid cell leukemia sequence 1
0.553 1456028_x_at Myristoylated alanine rich protein kinase C substrate (Marcks), mRNA
0.341 1438955_x_at peptidylprolyl isomerase F (cyclophilin F)
0.637 1455860_at phosphatidylinositol glycan, class H
0.356 1420132_s_at Pituitary tumor-transforming 1 interacting protein, mRNA (cDNA clone MGC:38220 IMAGE:5323397)
0.658 1425721_at pleckstrin homology domain interacting protein
0.658 1451740_at polyadenylate binding protein-interacting protein 1
0.51 1456270_s_at preferentially expressed antigen in melanoma like 6
0.629 1437845_x_at protein O-fucosyltransferase 2
0.665 1451225_at protein tyrosine phosphatase, non-receptor type 11
0.678 1455105_at protein tyrosine phosphatase, non-receptor type 12
0.728 1419069_at RAB guanine nucleotide exchange factor (GEF) 1
0.471 1419946_s_at RAB2, member RAS oncogene family (Rab2), mRNA
0.506 1455809_x_at resistance to inhibitors of cholinesterase 8 homolog (C. elegans)
0.483 1426604_at ribonuclease L (2′, 5′-oligoisoadenylate synthetase-dependent)
0.489 1418337_at ribose 5-phosphate isomerase A
0.665 1437246_x_at ribosomal protein S6 /// similar to 40S ribosomal protein S6
0.644 1417222_a_at RIKEN cDNA 2310075C12 gene
0.636 1452167_at RIKEN cDNA 2810407C02 gene
0.657 1426986_at RIKEN cDNA 2810485I05 gene
0.645 1418173_at RIKEN cDNA 4631426H08 gene
0.315 1418997_at RIKEN cDNA 4930469P12 gene
0.488 1456582_x_at RIKEN cDNA 5230400G24 gene
0.711 1435240_at RIKEN cDNA 5830435C13 gene
0.647 1440831_at RIKEN cDNA 6230421P05 gene
0.474 1436213_a_at RIKEN cDNA C430010P07 gene
0.656 1448434_at ring finger protein 103
0.711 1423740_a_at RNA binding motif protein 10
0.687 1419977_s_at RNA binding motif protein 35b
0.48 1420982_at RNA-binding region (RNP1, RRM) containing 2
0.695 1437461_s_at RNA-binding region (RNP1, RRM) containing 3
0.63 1437995_x_at septin 7
0.605 1418422_at serine (or cysteine) peptidase inhibitor, clade B, member 9g
0.489 1419913_at Serine/threonine kinase receptor associated protein (Strap), mRNA
0.659 1416041_at serum/glucocorticoid regulated kinase
0.676 1416862_at signal transducing adaptor molecule (SH3 domain and ITAM motif) 1
0.666 1439433_a_at solute carrier family 35 (UDP-galactose transporter), member 2
0.66 1452281_at Son of sevenless homolog 2 (Drosophila)
0.67 1454794_at spastin
0.585 1417300_at sphingomyelin phosphodiesterase, acid-like 3B
0.727 1436809_a_at spindlin
0.564 1455899_x_at suppressor of cytokine signaling 3
0.485 1420175_at Tax1 (human T-cell leukemia virus type I) binding protein 1, mRNA (cDNA clone MGC:11692 IMAGE:3962810)
0.612 1431332_a_at telomeric repeat binding factor 1
0.626 1415908_at testis-specific protein, Y-encoded-like 1
0.536 1437454_a_at thioredoxin domain containing 14 /// similar to thioredoxin-related transmembrane protein 2
0.366 1420042_at THO complex 1
0.462 1422781_at toll-like receptor 3
0.625 1437729_at Transcribed locus, strongly similar to XP_484309.1 PREDICTED: similar to ribosomal protein L27A [Mus musculus]
0.59 1436392_s_at transcription factor AP-2, gamma
0.372 1449671_at Transmembrane 7 superfamily member 1 (Tm7sf1), mRNA
0.454 1419918_at transmembrane emp24 protein transport domain containing 7
0.535 1439444_x_at transmembrane emp24-like trafficking protein 10 (yeast)
0.513 1435064_a_at transmembrane protein 27
0.693 1428586_at transmembrane protein 41B
0.6 1424275_s_at tripartite motif-containing 41
0.562 1425562_s_at tRNA nucleotidyl transferase, CCA-adding, 1
0.709 1438855_x_at tumor necrosis factor, alpha-induced protein 2
0.518 1439005_x_at Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide (Ywhaz), mRNA
0.682 1437714_x_at Ubiquitin specific peptidase 14, mRNA (cDNA clone MGC:7106 IMAGE:3157723)
0.522 1452011_a_at UDP-glucuronate decarboxylase 1
0.615 1421050_at vacuolar protein sorting 25 (yeast)
0.708 1457285_at zinc finger protein 187
0.613 1419181_at zinc finger protein 326
0.513 1427590_at zinc finger protein 39
0.426 1449691_at Zinc finger protein 644, mRNA (cDNA clone IMAGE:4037170)
0.511 1451518_at zinc finger protein 709
0.586 AFFX-r2-Bs-phe-3_at Unknown
0.585 AFFX-ThrX-M_at Unknown
0.575 AFFX-DapX-M_at Unknown
0.571 1419997_at Unknown
0.512 1424746_at Unknown
0.471 1447977_x_at Unknown

Table 4.

qPCR validation of selected genes

Gene Name Fold Change by Microarray Avg ddCT Mean Fold Change by qPCR (SEM)
Cebpa 1.6 -0.93 1.9 (0.21)
p53 1.5 -1.14 2.1 (0.33)
Rai17 5.1 -1.135 2.1 (0.41)
Jmy 1.5 -1.15 2.2 (0.27)
Hipk2 2.8 -1.13 2.1 (0.22)
Inpp5d 1.6 -0.71 1.6 (0.18)

Discussion

Microarray analysis is an invaluable tool to examine genetic mechanisms of cancer growth, development, responses to stress and, other processes. Numerous studies have been conducted utilizing this powerful tool of cDNA and oligo microarray, to elucidate the gene expression patterns in various physiological and pathological conditions. Previously, by the use of microarray analysis we have reported the changes in placental gene expression with fetal development [1] and acute maternal hypoxia [2]. In the present study we report the alterations in the placental gene expression with the maternal protein deprivation. Maternal protein restriction may play an important role in several disorders. Epidemiologic data in humans and studies in laboratory animals provide useful lessons on the effects of caloric restriction/malnutrition on fetal development and disease prevalence in adulthood [4, 5, 12, 21-26]. Those fetuses exposed to maternal caloric restriction in mid-gestation had a much greater incidence of bronchitis and other pulmonary disease [27] and renal disease as evidenced by microalbuminuria [28]. Females conceived during the famine also had a much higher prevalence of obesity as adults [4]. The cellular/molecular mechanisms of these in utero “programming” effects are unknown.

Studies in ruminants also have demonstrated that under-nutrition can have profound consequences for the fetus. In sheep, restricted maternal nutrition in early to mid-gestation was associated with an increase in placental weight, an increase in crown-rump length, and lower fetal to placental weight ratios [29]. Maternal under-nutrition also caused an alteration of cardiovascular homeostatic regulation by the renin-angiotensin system, and exposed the lambs to higher levels of glucocorticoids [30]. These hormonal effects also were associated with hypertension in the lambs [31]. Protein restriction in bovines also caused an increase in placental weight and an altered placental morphology [32].

Studies in rats have shown similar effects. Maternal protein restriction in rats triggers hypertension in the pups in adulthood. These effects appear to be mediated through a suppression of the renin-angiotensin system in the pups [33]. An alteration of placental glucocorticoid metabolism also was observed in placentae of rats fed a protein restricted diet. The activity of 11β-hydoxysteroid dehydrogenase, an enzyme present in the placenta, which normally protects the pups from maternal glucocorticoid excess, was reduced in protein restricted rats [34]. Another hormonal alteration in nutritionally deprived rats was an increase in somatostatin expression in the periventricular nucleus of the pups. This led to much lower levels of growth hormone, and had deleterious effects on the growth of the pups post-partum [35]. Fetal undernourishment also led to neuronal sequelae. The facial motor nucleus in pups was under-developed, and led to a functional decrease in the ability of pups to suckle and chew.

A study somewhat similar to ours was conducted in the rat, revealing an increase in genes involved in apoptosis, and p53 [36]. A direct comparison of the results between the studies is difficult, however, because of intra-species differences and the different timing of the protein restriction. Nonetheless, it is of interest to note similar themes emerging. The earliest large-scale studies on caloric restriction were related to the slowdown of aging process in mice skeletal muscles [37]. In the present study, maternal protein restriction showed up-regulation of the genes responsible for the negative regulation of cell growth and metabolism in the placenta. It is of interest to observe that caloric or protein restriction in different tissues and at different ages effect similar groups of gene responsible for the decrease in cellular growth and metabolism. However, further studies are needed to examine the biologic mechanisms by which protein/caloric restriction produces up-regulation of this particular pathway.

In a previous study, we used the Affymetrix oligonucleotide array to define developmental changes in gene expression from 10.5 dpc to 17.5 dpc in the mouse placenta [1]. In addition, we have reported on significant changes in mouse placental gene expression in response to maternal hypoxia for 48 hours, from 15.5 dpc to 17.5 dpc [2]. In the present study of placental gene regulation in response to maternal dietary protein restriction, we demonstrate a profound down-regulation of cell growth and proliferation and an up-regulation of genes coding for apoptotic proteins (Tables 2 and 3). Of particular interest, p53 along with rai17, Hipk2, jmy, Cebpa and Inpp5d (proteins that either activate, or are cofactors of, or are induced by p53), an important regulator of cell growth and proliferation were up-regulated. This pathway serves as a G1 checkpoint, and arrests growth and/or induces apoptosis in response to cellular damage. Mutations in the p53 gene have been implicated in a number of cancers and other pathological processes [38]. Hipk2, an upstream regulator of p53, activates its transcriptional activity and pro-apoptotic activities through phosporylation at Ser 46 [17]. Cebpa, a transcription factor induced by p53, mediates some of the downstream effects of p53 activation [19]. Several studies on the effects of nutritional deprivation have demonstrated that the p53 pathway is a crucial mediator of the observed biological effects. Mice deficient in p53 (p53 -/-) are more susceptible to cancer, but caloric restriction partially reversed that effect [39].

The present study has demonstrated a significant up-regulation of genes responsible for apoptosis regulation such as Bcl2-like 2, p53, endophilin, Fas-activated serine/threonine kinase. Apoptosis and its associated regulatory mechanisms are physiological events crucial to the maintenance of homeostasis in the placenta and other organs. Imbalance of these processes may cause various pathological conditions, may compromise placental function and, consequently, pregnancy success. Increased apoptosis occurs in the placentas of pregnant women with several developmental abnormalities, while increased Bcl-2 expression is generally associated with pregnancy-associated tumors and decreased expression is associated with placentas of the diabetic women [40]. Another important finding of the study was upregulation of Fas-activated serine/threonine phosphoprotein (FAST). FAST is a survival protein, bound to the outer mitochondrial membrane and mediates alternative and constitutive splicing, which may affect the expression of several other genes [41].

A potentially important finding of the present study, is that protein deprivation altered the expression of several genes involved in DNA methylation and histone acetylation, which are involved in epigenetic regulation of gene expression. The expression levels of histone deacetylase 7A and methionine adenosyltransferase II alpha, were significantly elevated. Histone actetylation triggers changes in chromatin structure, and regulates transcriptional availability of genes. In turn, histone deacetylation increases histone affinity for DNA, thereby repressing transcription . Methionine adenosyltransferase II alpha synthesizes AdoMet, the direct precursor used for DNA methylation by methyltransferases. Histone 2 (h3c2) is down-regulated, along with Mcm6 and telomeric repeat binding factor 1. These proteins contribute to DNA replication, stability, and structure [42, 43]. Recent studies in the human, have demonstrated in small for gestational age pregnancies an altered DNA methylation pattern in imprinted regions of the genome, and that imprinted genes are expressed in an unbalanced manner in pregnancies affected by intrauterine growth retardation [44, 45].

In several animal models, in addition to potential deleterious effects, a positive aspect of nutritional deprivation in the adult is that of prolonged lifespan and reduced cancer rates. A proposed mechanism for these benefits is that nutritional restriction in the absence of malnutrition inhibits cellular proliferation and induces apoptosis. This effect has been shown in mice lacking p53, in which -/- and +/- mutants have lowered spontaneous cancer rates when fed a complete, but calorically reduced diet [39]. In the adult and aging animal, nutritional restriction has been shown to have beneficial effects that increased life span [46]. In contrast, a different picture has emerged in the fetus. As discussed above, caloric and protein deprivation have been shown to trigger fetal programming of adult disease, and lead to an increased prevalence of metabolic disorders in adulthood. In the developing fetus, numerous animal studies have shown the negative long-term effects of caloric and protein deprivation on the cardiovascular, renal, nervous system and metabolism (for review see [47]). Both fetal and placental growths are essential for the long-term well-being of the individual. Thus, one would anticipate that profound inhibition of cellular growth at key time points during development would have grave long-term consequences for the embryo/fetus. This suggests that the timing of the treatment is a key determinant in the effect on the organism.

Perspective/Conclusions

The present data support the hypothesis that maternal protein restriction triggers an up-regulation of apoptosis-related genes, an increase in the p53 pathway, a change in epigenetic modulators, and an overall down-regulation of cellular proliferation and growth associated genes. The upregulation of inhibitory transcription factors, and other key negative cellular regulators altered by protein deprivation, offers a picture of profound and global down-regulation of the entire cellular proliferative machinery. These results suggest numerous avenues for future research, and raise a number of fundamental questions regarding energy/protein balance and cellular growth. A critical challenge will be to understand the cellular and molecular mechanisms of these epigenetic responses.

Acknowledgement

We thank Brenda Kreutzer for her assistance in the preparation of this manuscript and JD Heck of the DNA Array Core, University of California Irvine, Irvine, CA for technical assistance. This work was supported, in part by USPHS grant HD-03807 to LDL.

References

  • [1].Gheorghe C, Mohan S, Longo LD. Gene expression patterns in the developing murine placenta. J Soc Gynecol Investig. 2006;13:256–262. doi: 10.1016/j.jsgi.2006.02.007. [DOI] [PubMed] [Google Scholar]
  • [2].Gheorghe CP, Mohan S, Oberg KC, Longo LD. Gene expression patterns in the hypoxic murine placenta: a role in epigenesis? Reprod Sci. 2007;14:223–233. doi: 10.1177/1933719107302860. [DOI] [PubMed] [Google Scholar]
  • [3].Ravelli GP, Stein ZA, Susser MW. Obesity in young men after famine exposure in utero and early infancy. N Engl J Med. 1976;295:349–353. doi: 10.1056/NEJM197608122950701. [DOI] [PubMed] [Google Scholar]
  • [4].Roseboom TJ, van der Meulen JH, Ravelli AC, Osmond C, Barker DJ, Bleker OP. Effects of prenatal exposure to the Dutch famine on adult disease in later life: an overview. Twin Res. 2001;4:293–298. doi: 10.1375/1369052012605. [DOI] [PubMed] [Google Scholar]
  • [5].Neugebauer R, Hoek HW, Susser E. Prenatal exposure to wartime famine and development of antisocial personality disorder in early adulthood. JAMA. 1999;282:455–462. doi: 10.1001/jama.282.5.455. [DOI] [PubMed] [Google Scholar]
  • [6].Godfrey KM. Maternal regulation of fetal development and health in adult life. Eur J Obstet Gynecol Reprod Biol. 1998;78:141–150. doi: 10.1016/s0301-2115(98)00060-8. [DOI] [PubMed] [Google Scholar]
  • [7].Barker DJ, Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. Lancet. 1986;1:1077–1081. doi: 10.1016/s0140-6736(86)91340-1. [DOI] [PubMed] [Google Scholar]
  • [8].Barker DJ, Winter PD, Osmond C, Margetts B, Simmonds SJ. Weight in infancy and death from ischaemic heart disease. Lancet. 1989;2:577–580. doi: 10.1016/s0140-6736(89)90710-1. [DOI] [PubMed] [Google Scholar]
  • [9].Boujendar S, Arany E, Hill D, Remacle C, Reusens B. Taurine supplementation of a low protein diet fed to rat dams normalizes the vascularization of the fetal endocrine pancreas. J Nutr. 2003;133:2820–2825. doi: 10.1093/jn/133.9.2820. [DOI] [PubMed] [Google Scholar]
  • [10].Hsueh AM, Agustin CE, Chow BF. Growth of young rats after differential manipulation of maternal diet. J Nutr. 1967;91:195–200. doi: 10.1093/jn/91.2.195. [DOI] [PubMed] [Google Scholar]
  • [11].Downs KM, Davies T. Staging of gastrulating mouse embryos by morphological landmarks in the dissecting microscope. Development. 1993;118:1255–1266. doi: 10.1242/dev.118.4.1255. [DOI] [PubMed] [Google Scholar]
  • [12].Hoek HW, Susser E, Buck KA, Lumey LH, Lin SP, Gorman JM. Schizoid personality disorder after prenatal exposure to famine. Am J Psychiatry. 1996;153:1637–1639. doi: 10.1176/ajp.153.12.1637. [DOI] [PubMed] [Google Scholar]
  • [13].Susser E, Neugebauer R, Hoek HW, Brown AS, Lin S, Labovitz D, Gorman JM. Schizophrenia after prenatal famine. Further evidence. Arch Gen. Psychiatry. 1996;53:25–31. doi: 10.1001/archpsyc.1996.01830010027005. [DOI] [PubMed] [Google Scholar]
  • [14].Vehaskari VM, Woods LL. Prenatal programming of hypertension: lessons from experimental models. J Am Soc Nephrol. 2005;16:2545–2556. doi: 10.1681/ASN.2005030300. [DOI] [PubMed] [Google Scholar]
  • [15].Wright GW, Simon RM. A random variance model for detection of differential gene expression in small microarray experiments. Bioinformatics. 2003;19:2448–2455. doi: 10.1093/bioinformatics/btg345. [DOI] [PubMed] [Google Scholar]
  • [16].Lee J, Beliakoff J, Sun Z. The novel PIAS-like protein hZimp10 is a transcriptional co-activator of the p53 tumor suppressor. Nucleic Acids Res. 2007;35:4523–4534. doi: 10.1093/nar/gkm476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Hofmann TG, Möller A, Sirma H, Zentgraf H, Taya Y, Dröge W, Will H, Schmitz ML. Regulation of p53 activity by its interaction with homeodomain-interacting protein kinase-2. Nat Cell Biol. 2002;4:1–10. doi: 10.1038/ncb715. [DOI] [PubMed] [Google Scholar]
  • [18].Hershko T, Chaussepied M, Oren M, Ginsberg D. Novel link between E2F and p53: proapoptotic cofactors of p53 are transcriptionally upregulated by E2F. Cell Death Differ. 2005;12:377–383. doi: 10.1038/sj.cdd.4401575. [DOI] [PubMed] [Google Scholar]
  • [19].Yoon K, Smart RC. C/EBPalpha is a DNA damage-inducible p53-regulated mediator of the G1 checkpoint in keratinocytes. Mol Cell Biol. 2004;24:10650–10660. doi: 10.1128/MCB.24.24.10650-10660.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Kerley-Hamilton JS, Pike AM, Li N, DiRenzo J, Spinella MJ. A p53-dominant transcriptional response to cisplatin in testicular germ cell tumor-derived human embryonal carcinoma. Oncogene. 2005;24:6090–6100. doi: 10.1038/sj.onc.1208755. [DOI] [PubMed] [Google Scholar]
  • [21].Stein Z, Susser M. The Dutch famine, 1944-1945, and the reproductive process. II. Interrelations of caloric rations and six indices at birth. Pediatr Res. 1975;9:76–83. doi: 10.1203/00006450-197502000-00004. [DOI] [PubMed] [Google Scholar]
  • [22].Lumey LH, Van Poppel FW. The Dutch famine of 1944-45: mortality and morbidity in past and present generations. Soc Hist Med. 1994;7:229–246. doi: 10.1093/shm/7.2.229. [DOI] [PubMed] [Google Scholar]
  • [23].Painter RC, De Rooij SR, Bossuyt PM, Osmond C, Barker DJ, Bleker OP, Roseboom TJ. A possible link between prenatal exposure to famine and breast cancer: a preliminary study. Am J Hum Biol. 2006;18:853–856. doi: 10.1002/ajhb.20564. [DOI] [PubMed] [Google Scholar]
  • [24].Painter RC, de Rooij SR, Bossuyt PM, Simmers TA, Osmond C, Barker DJ, Bleker OP, Roseboom TJ. Early onset of coronary artery disease after prenatal exposure to the Dutch famine. Am J Clin Nutr. 2006;84:322–327. doi: 10.1093/ajcn/84.1.322. [DOI] [PubMed] [Google Scholar]
  • [25].Roseboom TJ, van der Meulen JH, Ravelli AC, van Montfrans GA, Osmond C, Barker DJ, Bleker OP. Blood pressure in adults after prenatal exposure to famine. J Hypertens. 1999;17:325–330. doi: 10.1097/00004872-199917030-00004. [DOI] [PubMed] [Google Scholar]
  • [26].Roseboom TJ, van der Meulen JH, van Montfrans GA, Ravelli AC, Osmond C, Barker DJ, Bleker OP. Maternal nutrition during gestation and blood pressure in later life. J Hypertens. 2001;19:29–34. doi: 10.1097/00004872-200101000-00004. [DOI] [PubMed] [Google Scholar]
  • [27].Lopuhaa CE, Roseboom TJ, Osmond C, Barker DJ, Ravelli AC, Bleker OP, van der Zee JS, van der Meulen JH. Atopy, lung function, and obstructive airways disease after prenatal exposure to famine. Thorax. 2000;55:555–561. doi: 10.1136/thorax.55.7.555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Painter RC, Roseboom TJ, van Montfrans GA, Bossuyt PM, Krediet RT, Osmond C, Barker DJ, Bleker OP. Microalbuminuria in adults after prenatal exposure to the Dutch famine. J Am Soc Nephrol. 2005;16:189–194. doi: 10.1681/ASN.2004060474. [DOI] [PubMed] [Google Scholar]
  • [29].Heasman L, Clarke L, Firth K, Stephenson T, Symonds ME. Influence of restricted maternal nutrition in early to mid gestation on placental and fetal development at term in sheep. Pediatr Res. 1998;44:546–551. doi: 10.1203/00006450-199810000-00013. [DOI] [PubMed] [Google Scholar]
  • [30].Edwards LJ, Simonetta G, Owens JA, Robinson JS, McMillen IC. Restriction of placental and fetal growth in sheep alters fetal blood pressure responses to angiotensin II and captopril. J Physiol. 1999;515:897–904. doi: 10.1111/j.1469-7793.1999.897ab.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Dodic M, Baird R, Hantzis V, Koukoulas I, Moritz K, Peers A, Wintour EM. Organs/systems potentially involved in one model of programmed hypertension in sheep. Clin Exp Pharmacol Physiol. 2001;28:952–956. doi: 10.1046/j.1440-1681.2001.03556.x. [DOI] [PubMed] [Google Scholar]
  • [32].Perry VE, Norman ST, Owen JA, Daniel RC, Phillips N. Low dietary protein during early pregnancy alters bovine placental development. Anim Reprod Sci. 1999;55:13–21. doi: 10.1016/s0378-4320(98)00157-2. [DOI] [PubMed] [Google Scholar]
  • [33].Langley SC, Jackson AA. Increased systolic blood pressure in adult rats induced by fetal exposure to maternal low protein diets. Clin Sci (Lond) 1994;86:217–22. doi: 10.1042/cs0860217. discussion 121. [DOI] [PubMed] [Google Scholar]
  • [34].Langley-Evans SC, Phillips GJ, Benediktsson R, Gardner DS, Edwards CR, Jackson AA, Seckl JR. Protein intake in pregnancy, placental glucocorticoid metabolism and the programming of hypertension in the rat. Placenta. 1996;17:169–172. doi: 10.1016/s0143-4004(96)80010-5. [DOI] [PubMed] [Google Scholar]
  • [35].Huizinga CT, Oudejans CB, Steiner RA, Clifton DK, Delemarre-van de Waal HA. Effects of intrauterine and early postnatal growth restriction on hypothalamic somatostatin gene expression in the rat. Pediatr Res. 2000;48:815–820. doi: 10.1203/00006450-200012000-00019. [DOI] [PubMed] [Google Scholar]
  • [36].Buffat C, Mondon F, Rigourd V, Boubred F, Bessières B, Fayol L, Feuerstein JM, Gamerre M, Jammes H, Rebourcet R, Miralles F, Courbières B, Basire A, Dignat-Georges F, Carbonne B, Simeoni U, Vaiman D. A hierarchical analysis of transcriptome alterations in intrauterine growth restriction (IUGR) reveals common pathophysiological pathways in mammals. J Pathol. 2007;213:337–346. doi: 10.1002/path.2233. [DOI] [PubMed] [Google Scholar]
  • [37].Lee CK, Klopp RG, Weindruch R, Prolla TA. Gene expression profile of aging and its retardation by caloric restriction. Science. 1999;285:1390–1393. doi: 10.1126/science.285.5432.1390. [DOI] [PubMed] [Google Scholar]
  • [38].Ryan KM, Phillips AC, Vousden KH. Regulation and function of the p53 tumor suppressor protein. Curr Opin Cell Biol. 2001;13:332–337. doi: 10.1016/s0955-0674(00)00216-7. [DOI] [PubMed] [Google Scholar]
  • [39].Hursting SD, Lavigne JA, Berrigan D, Donehower LA, Davis BJ, Phang JM, Barrett JC, Perkins SN. Diet-gene interactions in p53-deficient mice: insulin-like growth factor-1 as a mechanistic target. J Nutr. 2004;134:2482S–2486S. doi: 10.1093/jn/134.9.2482S. [DOI] [PubMed] [Google Scholar]
  • [40].Sgarbosa F, Barbisan LF, Brasil MA, Costa E, Calderon IM, Gonçalves CR, Bevilacqua E, Rudge MV. Changes in apoptosis and Bcl-2 expression in human hyperglycemic, term placental trophoblast. Diabetes Res Clin Pract. 2006;73:143–149. doi: 10.1016/j.diabres.2005.12.014. [DOI] [PubMed] [Google Scholar]
  • [41].Simarro M, Mauger D, Rhee K, Pujana MA, Kedersha NL, Yamasaki S, Cusick ME, Vidal M, Garcia-Blanco MA, Anderson P. Fas-activated serine/threonine phosphoprotein (FAST) is a regulator of alternative splicing. Proc Natl Acad Sci USA. 2007;104:11370–11375. doi: 10.1073/pnas.0704964104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].O’Connor MS, Safari A, Liu D, Qin J, Songyang Z. The human Rap1 protein complex and modulation of telomere length. J Biol Chem. 2004;279:28585–28591. doi: 10.1074/jbc.M312913200. [DOI] [PubMed] [Google Scholar]
  • [43].Yu Z, Feng D, Liang C. Pairwise interactions of the six human MCM protein subunits. J Mol Biol. 2004;340:1197–1206. doi: 10.1016/j.jmb.2004.05.024. [DOI] [PubMed] [Google Scholar]
  • 44.Guo L, Choufani S, Ferreira J, Smith A, Chitayat D, Shuman C, Uxa R, Keating S, Kingdom J, Weksberg R. Altered gene expression and methylation of the human chromosome 11 imprinted region in small for gestational age (SGA) placentae. Dev Biol. 2008;320:79–91. doi: 10.1016/j.ydbio.2008.04.025. [DOI] [PubMed] [Google Scholar]
  • 45.McMinn J, Wei M, Schupf N, Cusmai J, Johnson EB, Smith AC, Weksberg R, Thaker HM, Tycko B. Unbalanced placental expression of imprinted genes in human intrauterine growth restriction. Placenta. 2006;27:540–549. doi: 10.1016/j.placenta.2005.07.004. [DOI] [PubMed] [Google Scholar]
  • 46.Nikolich-Zugich J, Messaoudi I. Mice and flies and monkeys too: caloric restriction rejuvenates the aging immune system of non-human primates. Exp Gerontol. 2005;40:884–893. doi: 10.1016/j.exger.2005.06.007. [DOI] [PubMed] [Google Scholar]
  • 47.McMillen IC, Robinson JS. Developmental origins of the metabolic syndrome: prediction, plasticity, and programming. Physiol Rev. 2005;85:571–633. doi: 10.1152/physrev.00053.2003. [DOI] [PubMed] [Google Scholar]

RESOURCES