Skip to content
Publicly Available Published by De Gruyter March 19, 2016

Characterization of visceral and subcutaneous adipose tissue transcriptome in pregnant women with and without spontaneous labor at term: implication of alternative splicing in the metabolic adaptations of adipose tissue to parturition

  • Shali Mazaki-Tovi EMAIL logo , Adi L. Tarca , Edi Vaisbuch , Juan Pedro Kusanovic , Nandor Gabor Than , Tinnakorn Chaiworapongsa , Zhong Dong , Sonia S. Hassan and Roberto Romero EMAIL logo

Abstract

Objective:

The aim of this study was to determine gene expression and splicing changes associated with parturition and regions (visceral vs. subcutaneous) of the adipose tissue of pregnant women.

Study design:

The transcriptome of visceral and abdominal subcutaneous adipose tissue from pregnant women at term with (n=15) and without (n=25) spontaneous labor was profiled with the Affymetrix GeneChip Human Exon 1.0 ST array. Overall gene expression changes and the differential exon usage rate were compared between patient groups (unpaired analyses) and adipose tissue regions (paired analyses). Selected genes were tested by quantitative reverse transcription-polymerase chain reaction.

Results:

Four hundred and eighty-two genes were differentially expressed between visceral and subcutaneous fat of pregnant women with spontaneous labor at term (q-value <0.1; fold change >1.5). Biological processes enriched in this comparison included tissue and vasculature development as well as inflammatory and metabolic pathways. Differential splicing was found for 42 genes [q-value <0.1; differences in Finding Isoforms using Robust Multichip Analysis scores >2] between adipose tissue regions of women not in labor. Differential exon usage associated with parturition was found for three genes (LIMS1, HSPA5, and GSTK1) in subcutaneous tissues.

Conclusion:

We show for the first time evidence of implication of mRNA splicing and processing machinery in the subcutaneous adipose tissue of women in labor compared to those without labor.

Introduction

Parturition imposes an increased energy demand on the laboring woman. Labor is characterized by increased concentrations of nutrients including glucose [1–5], free fatty acids [3, 6], ketone bodies [7], and lactic acid [8]. There is an approximate three-fold increase in whole body glucose utilization during labor and delivery and, as expected, energy expenditure of the parturient women in the second stage of labor is 40% higher compared to the first stage [9]. Additional support for the metabolic burden of labor can also be found in the examination of myometrial glycogen storage, which is significantly increased at term [10], but almost completely depleted during labor [11]. Consistent with these findings, examination of the human myometrial transcriptome revealed that biological processes related to metabolism were among the molecular functions enriched in the differentially expressed genes between pregnant women with and without spontaneous term labor [12].

The conventional view is that the energy expenditure of labor and delivery is equivalent to that of moderate exercise [1, 9] and that similar mechanisms (e.g. insulin and non-insulin dependent glucose uptake, enhanced hepatic gluconeogenesis, and direct sympathetic nervous system stimulation) govern the metabolic adaptation to parturition [9, 13, 14]. However, whether or not adipose tissue, the major energy reservoir, is affected by labor and delivery is still unknown. Assessment of the putative role of adipose tissue in human parturition may be of special importance considering the large body of evidence indicating that this endocrinal organ is powerful [15] and exerts autocrine, paracrine and endocrine effects by the production and secretion of highly active peptides and proteins collectively termed adipokines [16]. Importantly, adipokines have been implicated in physiological adaptations of normal gestation [17–28] as well as in the pathophysiology of preeclampsia [21, 29–50], gestational diabetes mellitus [51–65], preterm birth [66–68], delivery of large-for-gestational-age (LGA) newborns [69], small-for-gestational-age (SGA) neonates [70–76], pyelonephritis [77–79], and intrauterine infection and inflammation [80–83]. Of note is the well-established association between obesity and these complications of pregnancy [84–113].

It has been suggested that the implication of adipose tissue in physiological or pathological processes should take into account the region-specific differences between fat depots. Particularly, differences in function [114–116], gene expression [115, 117–144], and metabolic effect [145–150] between the visceral and subcutaneous adipose tissue are to be considered. Indeed, regional variations of adipose tissue in specific genes were reported in non-pregnant individuals using both high throughput techniques [131–133, 136, 151] and targeted approaches [115, 116, 131, 133, 152–166]. Overall gene expression in the adipose tissue of pregnant women has been previously reported [32, 117–123, 167–178]; however, adipose tissue gene expression, biological processes, molecular functions, and pathways associated with spontaneous term parturition have not been described. Furthermore, to our knowledge, exon-level changes that can inform on alternative promoter usage, alternative splicing, and alternative transcript termination [179] between the visceral and subcutaneous regions have not been reported in either fat or other tissue of parturient women.

We undertook this study in order to characterize the transcriptome of human visceral and subcutaneous adipose tissue during normal labor at term to gain understanding of the global changes in gene expression and splicing associated with adiposity using an unbiased approach. The aims of this study were: 1) to determine differences in visceral and subcutaneous gene expression between pregnant women with and without spontaneous labor at term; 2) to determine regional variations in the transcriptome of adipose tissue of patients with spontaneous labor at term; and 3) to identify depot-specific alternative splicing alterations in the adipose tissue of women with spontaneous labor at term.

Materials and methods

Study groups

A prospective study was performed in which visceral and subcutaneous adipose tissue samples were obtained from women undergoing cesarean section at term (≥37 weeks) in the following groups: 1) not in labor (n=25) and 2) spontaneous labor (n=15).

The inclusion criteria for both groups were as follows: 1) absence of medical complications; 2) no antibiotic administration prior to the sample collection; 3) normal post-operative course; 4) absence of meconium staining of the amniotic fluid; 5) neonatal Apgar scores >7 at 1 and 5 min; 6) absence of histologic chorioamnionitis; 7) absence of obstetric complications of pregnancy; and 8) normal pregnancy outcome, including an infant who was of appropriate-weight-for-gestational-age (AGA) without congenital anomalies.

Eligible patients were enrolled at Hutzel Women’s Hospital (Detroit, MI, USA). All women provided written informed consent prior to the collection of adipose tissue samples. The collection and utilization of the samples for research purposes was approved by the Institutional Review Boards of the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD/NIH/DHHS, Bethesda, MD, USA), and the Human Investigation Committee of Wayne State University (Detroit, MI, USA). Samples obtained from pregnant women not in labor have been previously used to study the differences in transcriptome between pregnant and non-pregnant women.

Clinical definitions

Patients not in labor underwent a cesarean section secondary to a fetus in the non-cephalic presentation, previous uterine surgery, or classical cesarean section, or an elective cesarean section with no more than one previous cesarean section. Women in spontaneous labor underwent cesarean section due to a fetal malpresentation or for non-reassuring fetal status as determined by the clinical staff. Patients with clinical or histological chorioamnionitis and those undergoing induction of labor were excluded.

Labor was diagnosed in the presence of spontaneous regular uterine contractions occurring at a minimum frequency of two every 10 min with cervical changes that required hospital admission. Histologic chorioamnionitis was diagnosed using previously described criteria [180, 181]. An AGA neonate was defined by a birth weight between the 10th and 90th percentiles for the gestational age at birth [182]. Body mass index (BMI) was calculated according to the formula: weight (kg)/height2 (m2).

Sample collection

Paired visceral and subcutaneous adipose tissue samples were obtained from each participant. Subcutaneous adipose tissue samples were collected at the site of a transverse lower abdominal incision, in the middle of the Pfannenstiel incision, from the deeper strata of subcutaneous fat. Visceral samples were obtained from the most distal portion of the greater omentum [116, 183–186]. Visceral and subcutaneous adipose tissues were collected using Metzenbaum scissors and measured approximately 1.0 cm3. Tissues were snap-frozen in liquid nitrogen and stored at –80°C until use.

RNA isolation

Total RNA was isolated from snap-frozen adipose tissue using TRI Reagent® combined with the Qiagen RNeasy Lipid Tissue kit protocol (Qiagen, Valencia, CA, USA), according to the manufacturers’ recommendations. The RNA concentrations and the A260 nm/A280 nm ratio were assessed using a NanoDrop 1000 spectrophotometer (Thermo Scientific, Wilmington, DE, USA). RNA integrity numbers were determined using the Bioanalyzer 2100 (Agilent Technologies, Wilmington, DE, USA).

Microarray analysis and quantitative real-time polymerase chain reaction

The Affymetrix GeneChip Human Exon 1.0 ST array (Affymetrix Inc., Santa Clara, CA, USA) platform was used to measure the expression levels in each unpooled specimen, per manufacturer’s instructions (http://www.affymetrix.com). The array contains approximately 5.4 million 5-μm features (probes) grouped into 1.4 million probesets interrogating more than one million exon clusters [187–189]. To verify the results from microarray-based analysis, 24 genes were selected for quantitative real-time polymerase chain reaction (qRT-PCR) assays in the same set of samples used for microarrays.

Statistical analyses

Differential expression:

The raw microarray probe intensity data were background corrected, quantile normalized [190] and summarized into one expression value for each transcript using a robust multi-array average implemented in the aroma.affymetrix package [191]. A paired moderated t-test [192] was used to test for differential expression with a false discovery rate (FDR) [193] correction of P-values to obtain q-values. Gene significance was inferred using q<0.1 and fold-change in expression >1.5 [194]. Gene ontology analysis was performed with algorithms previously described [195]. Pathway analysis was performed on the Kyoto Encyclopedia of Genes and Genomes (KEGG) [196] pathway database (96 pathways with three or more genes on our microarray platform) with an overrepresentation analysis [197]. Alternatively, the Pathway Analysis with Down-weighting of Overlaping Genes (PADOG) [198] was applied on the canonical pathways collection from the MSigDB database [199] (831 pathways with at least 20 genes represented on our microarray platform). Differential expression between adipose tissue regions of the same subjects based on qRT-PCR data was performed with a paired t-test on –ΔCt values.

Differential exon usage (splicing):

To identify differential exon usage between the groups of samples, we used the method Finding Isoforms using Robust Multichip Analysis (FIRMA) [200] to quantify how far (above or below) a given exon’s expression level was compared to the expected (average) transcript level in a given sample. Criteria for inclusion of transcripts and exons are described in the supplementary material. We applied a t-test for each probeset (typically one per exon) in each transcript based on the FIRMA scores, and inferred significance when the difference in mean FIRMA scores between groups was 2.0 or more combined with a threshold of 0.1 on the FDR-adjusted P-values (q-values). This was a more stringent approach than described in another study [200] in which positive results were identified based only on the difference in mean FIRMA scores above 1.5 units. Plotting of the probe-level expression data at exon levels vs. genomic coordinates was performed using functionality provided by the GenomeGraphs package with known isoforms in the ENSEMBL database retrieved with biomaRt [200]. All microarray analyses were performed using the R language and environment and Bioconductor [200, 201].

Demographic data analysis:

The Student’s t, Mann-Whitney U, and χ2 tests were used to identify significant differences in patient demographics between women in the microarray and qRT-PCR groups. SPSS software (version 14.0; SPSS Inc, Chicago, IL, USA) was used for statistical analysis of demographic data. A probability value of <0.05 was considered statistically significant.

Results

Demographics

Table 1 displays the demographic characteristics of patients who were included in the microarray and qRT-PCR analyses.

Table 1

Demographic and clinical characteristics of the study population.

Term labor (n=15)Term not in labor (n=25)P-value
Maternal age (years)26 (24–38)27 (25–39)0.2
Gestational age at delivery (weeks)39.7 (39–40.6)39.1 (38.9–39.4)0.2
Pre-gestational BMI (kg/m2)35.3 (30.9–38.5)37.5 (26.2–40.2)0.5
BMI at sampling (kg/m2)36.9 ( 32.5–39.7)37.2 (27.8–45.4)0.8
Gravidity3 (2–3)3 (2–4)0.5
Parity2 (1–3)2 (2–3)0.2
Ethnic origin (%)1.0
 African American91.783.3
 Caucasian8.316.7
Systolic blood pressure (mm Hg)124 (117–127)121 (115–126)0.4
Diastolic blood pressure (mm Hg)75 (67–79)66 (62–77)0.3
Cervical dilatation at sampling5 (4–7)1 (1–2)<0.001
Fasting glucose (mg/dL)93 (87–98)94 (88–97)0.7
Birth weight (g)3320 (3155–3825)3275 (3105–3500)0.7

Data are presented as median and interquartile range (IQR). BMI=Body mass index.

Regional differences in the transcriptome of adipose tissue of women with and without labor

Differential expression

Microarray analysis demonstrated 485 transcripts corresponding to 482 unique genes differentially expressed between the visceral and subcutaneous adipose tissue of pregnant women in spontaneous labor at term (q-value <0.1; fold change >1.5). A total of 329 genes had decreased expression, and 153 genes had increased expression in the subcutaneous, compared to visceral, adipose tissue. A “volcano plot” shows the differential expression of all annotated probesets on the Affymetrix GeneChip Human Exon 1.0 ST array with the log (base 10) of q-values (y-axis) plotted against the log (base 2) fold changes (x-axis) between the visceral and subcutaneous adipose tissue (Figure 1). The heatmap in Figure 2 uses a color scale to show the consistency of the expression levels within each group of samples as well as the differences between the groups that led to positive test results. A list of the top 100 genes differentially expressed between visceral and subcutaneous adipose tissue of patients with and without spontaneous labor at term is presented in Table 2; the complete list of differentially expressed probes is available as supplementary material (Supplementary Table 1).

Figure 1: Differential expression of visceral versus subcutaneous adipose tissue transcripts in pregnant women in labor.Volcano plot showing differential expression evidence between subcutaneous and visceral adipose tissue of women in labor. The x-axis represents the log2 fold changes in expression with positive values representing over-expression in the subcutaneous region compared to visceral. Transcripts outside the vertical red bars have fold change >1.5. The y-axis represents the q-values (–log10 of), with values above 1.0 corresponding to q<0.1.
Figure 1:

Differential expression of visceral versus subcutaneous adipose tissue transcripts in pregnant women in labor.

Volcano plot showing differential expression evidence between subcutaneous and visceral adipose tissue of women in labor. The x-axis represents the log2 fold changes in expression with positive values representing over-expression in the subcutaneous region compared to visceral. Transcripts outside the vertical red bars have fold change >1.5. The y-axis represents the q-values (–log10 of), with values above 1.0 corresponding to q<0.1.

Figure 2: Heat map representing fat depot-specific differences in gene expression of pregnant women in labor.Heatmap showing the consistency of gene expression levels between subcutaneous and visceral regions of the adipose tissue of women in labor. Log2 transformed transcript expression values are centered and scaled row-wise.
Figure 2:

Heat map representing fat depot-specific differences in gene expression of pregnant women in labor.

Heatmap showing the consistency of gene expression levels between subcutaneous and visceral regions of the adipose tissue of women in labor. Log2 transformed transcript expression values are centered and scaled row-wise.

Table 2

A list of the top 100 differentially expressed genes between visceral and subcutaneous adipose tissue of patients with and without spontaneous labor at term.

ENTREZSymbolNameFold changeq-Value
364AQP7Aquaporin 71.60.007
355FASFas (TNF receptor superfamily, member 6)–1.90.007
100293763AQP7P1Aquaporin 7 pseudogene 11.80.007
9871*SEC24DSEC24 family, member D (Saccharomyces cerevisiae)–1.70.007
83666*PARP9Poly(ADP-ribose) polymerase family, member 9–1.60.008
729085*CCBP2Chemokine binding protein 2–1.60.008
6285S100BS100 calcium binding protein B1.60.008
10555AGPAT21-Acylglycerol-3-phosphate O-acyltransferase 2 (lysophosphatidic acid acyltransferase, beta)1.60.008
6574SLC20A1Solute carrier family 20 (phosphate transporter), member 1–1.90.008
54566EPB41L4BErythrocyte membrane protein band 4.1 like 4B1.60.008
58477*SRPRBSignal recognition particle receptor, B subunit–1.80.008
54988*ACSM5Acyl-CoA synthetase medium-chain family member 51.60.008
9180OSMROncostatin M receptor–1.90.008
284221FAM38B2Family with sequence similarity 38, member B2–2.00.008
2819GPD1Glycerol-3-phosphate dehydrogenase 1 (soluble)1.70.008
9052GPRC5AG protein-coupled receptor, family C, group 5, member A–1.70.008
10973*ASCC3Activating signal cointegrator 1 complex subunit 3–1.50.008
6517SLC2A4Solute carrier family 2 (facilitated glucose transporter), member 41.60.008
6713SQLESqualene epoxidase–1.60.008
83716CRISPLD2Cysteine-rich secretory protein LCCL domain containing 2–1.90.008
10249GLYATGlycine-N-acyltransferase2.40.008
23555TSPAN15Tetraspanin 151.60.008
8908GYG2Glycogenin 21.50.008
5578PRKCAProtein kinase C, alpha–1.50.008
54884RETSATRetinol saturase (all-trans-retinol 13,14-reductase)1.60.008
5055SERPINB2Serpin peptidase inhibitor, clade B (ovalbumin), member 2–4.90.008
57568*SIPA1L2Signal-induced proliferation-associated 1 like 2–1.50.008
5207PFKFB16-Phosphofructo-2-kinase/fructose-2,6-biphosphatase 11.90.008
60559*SPCS3Signal peptidase complex subunit 3 homolog (S. cerevisiae)–1.60.008
9197*SLC33A1Solute carrier family 33 (acetyl-CoA transporter), member 1–1.50.008
3991LIPELipase, hormone-sensitive1.60.008
26064*RAI14Retinoic acid induced 14–1.60.008
8542*APOL1Apolipoprotein L, 1–1.60.008
374969CCDC23Coiled-coil domain containing 231.50.008
8639AOC3Amine oxidase, copper containing 3 (vascular adhesion protein 1)1.60.008
11098PRSS23Protease, serine, 23–1.70.008
2822GPLD1Glycosylphosphatidylinositol specific phospholipase D11.70.008
8659ALDH4A1Aldehyde dehydrogenase 4 family, member A11.80.008
4718THRSPThyroid hormone responsive (SPOT14 homolog, rat)1.80.008
55024*BANK1B-cell scaffold protein with ankyrin repeats 11.60.008
6782*HSPA13Heat shock protein 70 kDa family, member 13–1.80.008
65983GRAMD3GRAM domain containing 3–1.80.008
4137MAPTMicrotubule-associated protein tau1.60.008
1009CDH11Cadherin 11, type 2, OB-cadherin (osteoblast)–2.00.008
10130*PDIA6Protein disulfide isomerase family A, member 6–1.50.008
51602*NOP58NOP58 ribonucleoprotein homolog (yeast)–1.60.008
81539SLC38A1Solute carrier family 38, member 1–2.40.008
51716CES1Carboxylesterase 1 (monocyte/macrophage serine esterase 1)2.20.008
9643*MORF4L2Mortality factor 4 like 2–1.50.008
666BOKBCL2-related ovarian killer1.60.008
9945GFPT2Glutamine-fructose-6-phosphate transaminase 2–2.40.008
55254*TMEM39ATransmembrane protein 39A–1.50.008
22915MMRN1Multimerin 1–3.80.008
84293C10orf58Chromosome 10 open reading frame 581.60.008
64757MOSC1MOCO sulphurase C-terminal domain containing 11.60.008
64805*P2RY12Purinergic receptor P2Y, G-protein coupled, 121.60.008
91607SLFN13Schlafen family member 13–1.50.009
220ALDH1A3Aldehyde dehydrogenase 1 family, member A3–2.30.009
212*ALAS2Aminolevulinate, delta-, synthase 21.70.009
10962MLLT11Myeloid/lymphoid or mixed-lineage leukemia (trithorax homolog, Drosophila); translocated to, 11–2.20.009
358*AQP1Aquaporin 1 (Colton blood group)–1.50.009
23612*PHLDA3Pleckstrin homology-like domain, family A, member 31.50.009
6578SLCO2A1Solute carrier organic anion transporter family, member 2A1–1.70.009
286753*TUSC5Tumor suppressor candidate 51.50.009
5271*SERPINB8Serpin peptidase inhibitor, clade B (ovalbumin), member 8–1.70.009
63924CIDECCell death-inducing DFFA-like effector c1.80.009
4189*DNAJB9DnaJ (Hsp40) homolog, subfamily B, member 9–1.70.009
56265CPXM1Carboxypeptidase X (M14 family), member 1–1.70.009
58528*RRAGDRas-related GTP binding D1.50.009
262*AMD1Adenosylmethionine decarboxylase 1–1.50.009
222166C7orf41Chromosome 7 open reading frame 411.60.009
338APOBApolipoprotein B [including Ag(x) antigen]2.40.009
158295MGC24103Hypothetical MGC24103–1.50.009
1805*DPTDermatopontin1.50.009
10237*SLC35B1Solute carrier family 35, member B1–1.50.009
623BDKRB1Bradykinin receptor B1–3.00.009
5740PTGISProstaglandin I2 (prostacyclin) synthase–2.00.009
6272SORT1Sortilin 11.70.009
1645AKR1C2Aldo-keto reductase family 1, member C2 (dihydrodiol dehydrogenase 2; bile acid binding protein; 3-a hydroxysteroid dehydrogenase, type III)2.10.009
5649RELNReelin–1.60.009
6446SGK1Serum/glucocorticoid regulated kinase 1–1.90.009
51330TNFRSF12ATumor necrosis factor receptor superfamily, member 12A–1.80.009
388403*YPEL2Yippee-like 2 (Drosophila)1.70.009
80704SLC19A3Solute carrier family 19, member 31.50.009
5140PDE3BPhosphodiesterase 3B, cGMP-inhibited1.70.009
3036HAS1Hyaluronan synthase 1–1.70.009
1646AKR1C1Aldo-keto reductase family 1, member C1 (dihydrodiol dehydrogenase 1; 20-a (3-a)-hydroxysteroid dehydrogenase)2.30.009
1962EHHADHEnoyl-Coenzyme A, hydratase/3-hydroxyacyl Coenzyme A dehydrogenase1.50.009
90355*C5orf30Chromosome 5 open reading frame 301.60.009
283383GPR133G protein-coupled receptor 133–2.10.009
4199ME1Malic enzyme 1, NADP(+)-dependent, cytosolic1.70.009
6366CCL21Chemokine (C-C motif) ligand 21–3.50.009
4023LPLLipoprotein lipase1.60.009
6385SDC4Syndecan 4–2.50.009
84649DGAT2Diacylglycerol O-acyltransferase homolog 2 (mouse)1.60.009
80339*PNPLA3Patatin-like phospholipase domain containing 31.60.009
783CACNB2Calcium channel, voltage-dependent, beta 2 subunit–1.70.009
7086TKTTransketolase1.50.009
63895*FAM38BFamily with sequence similarity 38, member B–1.60.009
4883NPR3Natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C)1.80.009

Among the 482 genes differentially expressed between visceral and subcutaneous adipose tissue in patients with spontaneous labor at term, 91 were not part of the 632 genes differentially expressed in the not in labor group (ENTREZ IDs suffixed by a * in Table 2 and Supplementary Table 1).

In order to gain further insight into the biology of the differential gene expression, Gene Ontology enrichment analysis was employed. A total of 94 biological processes were associated with regional differences in the spontaneous term labor group (q<0.05) (Table 3). Pathway analysis performed using an over-representation on the KEGG database resulted in seven significant pathways in this comparison (q<0.05): complement and coagulation cascades, cytokine-cytokine receptor interaction, focal adhesion, steroid hormone biosynthesis, ECM-receptor interaction, African trypanosomiasis, and protein digestion and absorption.

Table 3

Bological processes associated with regional differences in the spontaneous term labor group.

Biological processTerm sizeDE genesOdds ratioq-Value
Response to external stimulus951742.7<0.001
Retinal metabolic process98216.8<0.001
Circulatory system development739612.6<0.001
Regulation of complement activation18927.1<0.001
Blood vessel morphogenesis347353.2<0.001
Multicellular organismal process48702321.7<0.001
Positive regulation of cellular component movement283303.3<0.001
Anatomical structure formation involved in morphogenesis846612.2<0.001
Regulation of cell motility498422.6<0.001
Terpenoid metabolic process63137.2<0.001
Retinol metabolic process17718.9<0.001
Phototransduction, visible light70136.2<0.001
Vasculature development393342.70.001
Triglyceride catabolic process25812.70.001
Positive regulation of signal transduction905602.10.001
Glomerular filtration19715.80.001
Neutral lipid catabolic process29810.30.002
Regulation of inflammatory response123164.20.002
Cell motility461352.50.002
Positive regulation of cell-substrate adhesion60116.10.002
Detection of light stimulus86134.90.003
Acute inflammatory response75125.20.003
Reproductive system development321282.60.004
Striated muscle cell differentiation164183.40.005
Regulation of hormone levels122153.90.005
Tissue morphogenesis498372.20.005
Death1510841.70.006
Glycerolipid catabolic process3687.70.007
Cellular response to jasmonic acid stimulus33Inf0.008
Positive regulation of phosphate metabolic process776501.90.008
Receptor-mediated endocytosis173183.20.008
Cellular developmental process28841401.50.008
Protein activation cascade4996.10.009
Tube development407312.30.010
Urogenital system development211202.90.011
Positive regulation vascular endothelial growth factor production21610.80.011
Cell junction assembly177183.10.011
Positive regulation of angiogenesis102134.00.011
Response to lipid607412.00.012
Positive regulation of macrophage derived foam cell differentiation14514.90.012
Cell chemotaxis181183.00.013
Single-organism process634292.70.013
Response to oxygen-containing compound948561.80.013
Terpenoid biosynthetic process8426.80.013
Embryonic limb morphogenesis106133.80.014
Regulation of response to stress847521.80.014
Negative regulation of protein processing234212.70.014
Positive regulation of epithelial cell proliferation123143.50.016
Regulation of behavior155163.10.017
Regulation of multicellular organismal development1167661.70.017
Response to wounding168163.10.017
Regulation of phosphorylation1016591.70.019
Complement activation, alternative pathway9421.50.019
Negative regulation of cardiac muscle tissue development16512.20.019
Epithelium development624402.00.020
Muscle cell migration4685.70.021
Oxoacid metabolic process849511.80.022
Regulation of transport1300711.60.023
Regulation of leukocyte chemotaxis72104.40.023
Positive regulation of focal adhesion assembly17511.20.023
Regulation of protein metabolic process152153.10.024
Small molecule metabolic process1919971.50.024
Endothelial cell morphogenesis10417.90.025
Negative regulation of heart growth10417.90.025
Response to acid chemical233202.60.026
Negative regulation of endopeptidase activity150153.00.028
Establishment of localization34641581.40.028
Cellular response to tumor necrosis factor90113.80.031
Endodermal cell differentiation3975.90.034
Retinoic acid biosynthetic process5340.30.034
Protein secretion352262.20.035
Cellular lipid metabolic process480322.00.035
Cellular response to endogenous stimulus836491.70.035
Regulation of cell adhesion418292.10.035
Positive regulation of locomotion193172.70.035
Appendage morphogenesis123133.20.035
Negative regulation of muscle tissue development2967.00.035
Positive regulation of cell proliferation487322.00.036
Regulation of striated muscle tissue development79103.90.036
Lung development110123.40.038
Triglyceride biosynthetic process5384.80.039
Positive regulation of mesenchymal cell proliferation3066.70.040
Negative regulation of muscle organ development3066.70.040
Positive regulation of leukocyte migration81103.80.042
Neutral lipid biosynthetic process5484.70.042
Peptide transport248202.40.043
Peptide hormone secretion195172.60.045
Inflammatory response227182.50.047
Cell adhesion493312.00.047
Response to toxic substance129133.00.047
Positive regulation of MAPK cascade236192.40.047
Regulation of cell-matrix adhesion6994.10.047
Daunorubicin metabolic process6326.80.049
Doxorubicin metabolic process6326.80.049

qRT-PCR analysis

The results of qRT-PCR confirmed the differential expression of nine of 29 genes found to be significant on the microarray analysis: lipoprotein lipase (LPL), retinol binding protein 4 (RBP4), leptin (LEP), complement component 4B (Chido blood group) (C4B), insulin-like growth factor binding protein 2 (IGFBP2), monoglyceride lipase (MGLL), annexin A8 (ANXA8), klotho beta (KLB), and prolactin (PRL).

Differential splicing

Using the Affymetrix GeneChip Human Exon 1.0 ST array that probes individual exons of known genes, we compared the exon usage (inclusion) rates between adipose tissue regions. Significant differences in exon usage were found for 42 genes between visceral and subcutaneous adipose tissue of pregnant women not in labor (Table 4) but not in the labor group.

Table 4

A list of the alternative splicing events associated with the regional differences of the adipose tissue of pregnant women not in labor.

ENTREZSYMBOLNameExon IDaDiff. FIRMAbP-valueq-Value
5376PMP22Peripheral myelin protein 228874245.2<0.001<0.001
6711SPTBNSpectrin, beta, non-erythrocytic 1103031–5.1<0.001<0.001
25818KLK5Kallikrein-related peptidase 5960481–4.6<0.001<0.001
85442KNDC1Kinase non-catalytic C-lobe domain (KIND) containing 1596822–3.5<0.001<0.001
388610TRNP1TMF1-regulated nuclear protein 17232–3.1<0.001<0.001
1612DAPK1Death-associated protein kinase 15381103.1<0.001<0.001
9201DCLK1Doublecortin-like kinase 1742860–3.0<0.001<0.001
9214FAIM3Fas apoptotic inhibitory molecule 384252–2.9<0.001<0.001
25891PAMR1Peptidase domain containing associated with muscle regeneration 16565162.9<0.001<0.001
3983ABLIM1Actin binding LIM protein 16190432.9<0.001<0.001
286204CRB2Crumbs homolog 2 (Drosophila)544486–2.8<0.001<0.001
11343MGLLMonoglyceride lipase236116–2.7<0.0010.0017
25891PAMR1Peptidase domain containing associated with muscle regeneration 16565162.7<0.001<0.001
1674DESDesmin131889–2.7<0.0010.0068
10231RCAN2Regulator of calcineurin 23990762.6<0.001<0.001
23524SRRM2Serine/arginine repetitive matrix 2826444–2.6<0.001<0.001
23524SRRM2Serine/arginine repetitive matrix 2826444–2.5<0.001<0.001
157506RDH10Retinol dehydrogenase 10 (all-trans)4912732.5<0.0010.0017
85442KNDC1Kinase non-catalytic C-lobe domain (KIND) containing 1596819–2.5<0.001<0.001
79804HAND2Heart and neural crest derivatives expressed 2298919–2.4<0.001<0.001
65108MARCKSL155081–2.4<0.001<0.001
4071TM4SF1Transmembrane 4 L six family member 1240134–2.4<0.0010.006
4837NNMTNicotinamide N-methyltransferase644508–2.3<0.0010.0099
51090PLLPPlasma membrane proteolipid (plasmolipin)8551892.3<0.001<0.001
1674DESDesmin131895–2.3<0.0010.0028
152ADRA2CAdrenergic, a-2C-, receptor249900–2.3<0.0010.0037
81539SLC38A1Solute carrier family 38, member 17072062.3<0.001<0.001
57121LPAR5Lysophosphatidic acid receptor 5701054–2.3<0.0010.001
56920SEMA3GSema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3G224606–2.3<0.001<0.001
9945GFPT2Glutamine-fructose-6-phosphate transaminase 23591712.2<0.001<0.001
2627GATA6GATA binding protein 6908232–2.2<0.001<0.001
4824NKX3-1NK3 homeobox 1506935–2.1<0.001<0.001
3036HAS1Hyaluronan synthase 1960742–2.1<0.001<0.001
5420PODXLPodocalyxin-like472183–2.1<0.001<0.001
5420PODXLPodocalyxin-like4722132.1<0.001<0.001
3339HSPG2Heparan sulfate proteoglycan 252523–2.1<0.0010.0746
23555TSPAN15Tetraspanin 15582734–2.1<0.001<0.001
23428SLC7A8Solute carrier family 7 (cationic amino acid transporter, y+ system), member 8772193–2.1<0.001<0.001
3913LAMB2Laminin, beta 2 (laminin S)223487–2.0<0.001<0.001
23428SLC7A8Solute carrier family 7 (cationic amino acid transporter, y+ system), member 8772200–2.0<0.001<0.001
89932PAPLNPapilin, proteoglycan-like sulfated glycoprotein763903–2.0<0.001<0.001
255743NPNTNephronectin2638822.0<0.001<0.001

aExon Identifier based on annotation provided HuEx-1_0-st-v2.na30.hg19.probeset.csv file from www.affymetrix.com. bFIRMA scores are a measure of the exon abundance relative to the overall gene level in a given sample. Positive differences in FIRMA scores represent a higher exon usage rate in subcutaneous compared to visceral adipose tissue of women not in labor.

Patients with spontaneous term labor versus pregnant women not in labor

Differential expression

We did not find significant differences in gene expression in either visceral or subcutaneous adipose tissue of pregnant women with and without spontaneous labor using our predefined gene selection criteria. However, when applying PADOG pathway analysis, four KEGG pathways (spliceosome, snare interactions in vesicular transport, pathogenic Escherichia coli infection, DNA replication) and three Reactome database [202] pathways (processing of capped intron containing pre-mRNA, mRNA processing, mRNA splicing) were found to be significantly perturbed in the presence of labor in the subcutaneous region of the adipose tissue (see enrichment plots for two of these pathways in Figure 3). Unlike the over-representation approach requiring gene selection as a first step, PADOG determines whether the differential expression t-scores of a given pathway are higher (in absolute value) than those of all genes profiled on the array and, hence, detects potentially smaller but systematic differential expression in a given pathway compared to all genes on the array (Figure 3). When comparing the visceral region of the women in labor to those without labor, the PADOG identified the Reactome asparagine N-linked glycosylation pathway to be associated with parturition (see Figure S1).

Figure 3: Pathway perturbation associated with parturition in subcutaneous tissue.PADOG pathway enrichment plots showing evidence of pathway perturbation associated with parturition in subcutaneous tissue. The distribution of moderated t-scores of genes in KEGG spliceosome and reactome mRNA splicing is superimposed on the distribution of all genes on the array, and shows more differential expression in these pathways than in the pool of all genes.
Figure 3:

Pathway perturbation associated with parturition in subcutaneous tissue.

PADOG pathway enrichment plots showing evidence of pathway perturbation associated with parturition in subcutaneous tissue. The distribution of moderated t-scores of genes in KEGG spliceosome and reactome mRNA splicing is superimposed on the distribution of all genes on the array, and shows more differential expression in these pathways than in the pool of all genes.

Differential splicing

Significant differences in exon usage were found between subcutaneous adipose tissue of pregnant women with and without spontaneous labor at term for three genes: Glutathione S-transferase kappa 1 (GSTK1), heat shock 70 kDa protein 5 (glucose-regulated protein, 78 kDa) (HSPA5), and LIM and senescent cell antigen-like-containing domain protein 1 (LIMS1). None of the three genes were differentially expressed between visceral and subcutaneous adipose tissue of parturient women as the change in mRNA abundance was present only for one exon of each gene (Figures 4 and 5 illustrate the differential exon usage for LIMS1 and GSTK1). For all three genes, the exon showing differential usage had lower expression in the group of women in labor compared to the not-in-labor group. These three genes were not among the 42 genes with differential exon usage between visceral and subcutaneous adipose tissue of pregnant women not in labor (Table 4).

Figure 4: Differential exon usage for LIMS1 gene in subcutaneous adipose tissue of women with and without labor.The top panel shows the log2 expression of probes targeting 12 exonic regions of the LIMS1 gene (separated by vertical gray lines). There are 1–4 probes per probeset. Each line corresponds to a sample, with colors blue and gray denoting one patient with and without labor, respectively. The second exon from the 5′ end targeted by Affymetrix probeset ID 2499062 (see red rectangles), shows systematically lower expression in women in labor, while the expression level for all other exons is very similar between groups, hence resulting in significantly lower FIRMA scores for this probeset between groups. The middle panel shows the genomic region and the gene model with each exon represented by one olive-colored rectangle. ENSEMBL transcripts that do or do not include the exon with differential usage are represented in blue with their corresponding identifiers.
Figure 4:

Differential exon usage for LIMS1 gene in subcutaneous adipose tissue of women with and without labor.

The top panel shows the log2 expression of probes targeting 12 exonic regions of the LIMS1 gene (separated by vertical gray lines). There are 1–4 probes per probeset. Each line corresponds to a sample, with colors blue and gray denoting one patient with and without labor, respectively. The second exon from the 5′ end targeted by Affymetrix probeset ID 2499062 (see red rectangles), shows systematically lower expression in women in labor, while the expression level for all other exons is very similar between groups, hence resulting in significantly lower FIRMA scores for this probeset between groups. The middle panel shows the genomic region and the gene model with each exon represented by one olive-colored rectangle. ENSEMBL transcripts that do or do not include the exon with differential usage are represented in blue with their corresponding identifiers.

Figure 5: Differential exon usage for the GSTK1 gene in subcutaneous adipose tissue of women with and without labor.See Figure 3 legend for layout details. Affymetrix probeset ID 3028993 (see red rectangles), shows systematically lower expression in women in labor, while the expression level for all other exons is very similar between groups, hence resulting in significantly lower FIRMA scores for this probeset between groups. The only ENSEMBL transcript that includes the exonic region with differential usage between groups is ENST00000479303, and an imbalance of this isoform with respect to the other isoforms can explain the observed differences.
Figure 5:

Differential exon usage for the GSTK1 gene in subcutaneous adipose tissue of women with and without labor.

See Figure 3 legend for layout details. Affymetrix probeset ID 3028993 (see red rectangles), shows systematically lower expression in women in labor, while the expression level for all other exons is very similar between groups, hence resulting in significantly lower FIRMA scores for this probeset between groups. The only ENSEMBL transcript that includes the exonic region with differential usage between groups is ENST00000479303, and an imbalance of this isoform with respect to the other isoforms can explain the observed differences.

Discussion

The principal findings of this study include the following: 1) Visceral and subcutaneous adipose tissue transcriptome of pregnant women with spontaneous labor at term were different: i) 482 genes were differentially expressed between the two fat depots; ii) Gene Ontology analysis indicated specific biological processes (e.g. cell adhesion, vasculature development, and circulatory system development); iii) the KEGG pathways enriched in differentially expressed genes were: complement and coagulation cascades, cytokine-cytokine receptor interaction, focal adhesion, steroid hormone biosynthesis, ECM-receptor interaction, African trypanosomiasis, and protein digestion and absorption. 2) Significant differences in alternative spliced genes were found between the subcutaneous adipose tissue of pregnant women with and without spontaneous labor at term; three genes affected by alternative splicing were LIM and senescent cell antigen-like-containing domain protein 1 (LIMS1), heat shock 70 kDa protein 5 (glucose-regulated protein, 78 kDa) (HSPA5), and Glutathione S-transferase kappa 1 (GSTK1); and 3) visceral and subcutaneous adipose tissue transcriptome of pregnant women with and without spontaneous labor at term did not differ significantly.

Visceral versus subcutaneous adipose tissue in pregnant women with spontaneous labor at term

This study describes, for the first time, the transcriptome of visceral and subcutaneous adipose tissue of pregnant women with spontaneous labor at term. High throughput technology has been employed in obstetrics [203–208]. Specifically, the transcriptome of the uterine cervix [209–217], myometrium [12, 218–224], chorioamniotic membranes [225, 226], amniotic fluid [227–236], maternal blood [237], and umbilical cord blood [238] have been reported. Region-specific differences were extensively investigated in non-pregnant individuals using both targeted and high-dimensional biology techniques [124–142, 145, 148–151, 153, 239–242]. In contrast, previous reports concerning gene expression in adipose tissue of pregnant women have used only the targeted approach [32, 117–123, 167–176] with two exceptions [178]. Resi et al. investigated the transcriptome of subcutaneous adipose tissue obtained from the gluteal depot. Participants in that study included healthy non-obese women and healthy women not in labor [178]. This is the first report to use either a high-dimensional biological technique or a targeted approach in the investigation of fat depots during normal human labor.

Bashiri et al. [243] have determined alterations in genome-wide transcription expression in visceral and abdominal subcutaneous fat depots in obese and lean pregnant women (four in each group) using the Affymetrix Human Exon 1.0 ST platform. The authors reported that global alteration in gene expression was identified in pregnancy complicated by obesity and the identification of indolethylamine N-methyltransferase, tissue factor pathway inhibitor-2, and ephrin type-B receptor 6 that were not previously associated with fat metabolism during pregnancy. In addition, subcutaneous fat of obese pregnant women demonstrated increased coding protein transcripts associated with apoptosis as compared to lean pregnant women. Of note, all participants in Bashiri et al. [243] were not in labor.

Comparison between the transcriptome of visceral and subcutaneous adipose tissue in pregnant women with and without spontaneous labor at term: evidence for an active role of adipose tissue response in the metabolic adaptation to parturition

An additional novel finding reported herein is the implication of alternative splicing in subcutaneous adipose tissue of pregnant women in spontaneous labor at term. Alternative splicing is a major biological process by which a relatively limited number of genes can be expended into elaborate proteomes [244]. It has been estimated that approximately two-thirds to three-quarters of all human genes undergo alternative splicing [201, 244–246]. This process allows cells to include or exclude different selective sections of pre-mRNA during RNA processing [247]. The altered transcripts result in closely related proteins expressed from a single locus [247]. The splicing process may affect function, localization, binding properties, and stability of the encoded proteins [244, 248] as well as degradation of the transcript [244, 249, 250]. It is an important regulatory mechanism that has been shown to be involved in several molecular pathways including angiogenesis and differentiation [247, 251]. To our knowledge, this is the first report implicating alternative splicing in parturition-related differences of subcutaneous adipose tissue or any other tissue.

While we did not find significant differences in gene expression between either visceral or subcutaneous adipose tissue of pregnant women with and without spontaneous labor at term, we identified three genes affected by alternative splicing: Glutathione S-transferase kappa 1 (GSTK1), heat shock 70 kDa protein 5 (glucose-regulated protein, 78 kDa) (HSPA5), and LIM and senescent cell antigen-like-containing domain protein 1 (LIMS1). The Kappa class of glutathione S-transferases (GSTK) was first identified in the mitochondrial matrix from rat liver [252]. The human glutathione S-transferase kappa 1 (GSTK1) gene and protein were first characterized less than a decade ago [253]. Further studies of human GSTK1-1 have confirmed its presence in mitochondria and peroxisomes [253–255]. GSTK1-1 is highly expressed in adipose tissue, and its expression level was negatively correlated with obesity in humans and mice [256]. Importantly, GSTK1-1 plays a critical and selective role in regulating adiponectin biosynthesis. Specifically, suppression of GSTK1-1 inhibits adiponectin multimerization, probably by functioning as protein disulfide isomerase that regulates adiponectin disulfide bond formation, which is essential for multimerization.

Adiponectin, identified independently by four groups [257–260], is the most abundant gene (AMP1) product of adipose tissue; it circulates at a relatively high concentration [261]. Adiponectin has an important role in the pathophysiology of insulin resistance and diabetes [262], atherosclerosis [263], hypertension [264], dyslipidemia [265], and angiogenesis [266]. A solid body of evidence supports the role of adiponectin in normal gestation and pregnancy complications: 1) circulating maternal adiponectin correlates with insulin resistance indices during pregnancy [267]; 2) patients with gestational diabetes mellitus (GDM) have a lower concentration of adiponectin compared to those without GDM [51, 53, 268, 269]; 4) overweight pregnant patients have a lower adiponectin concentration than pregnant women of normal weight; and 5) preeclampsia is associated with altered maternal adiponectin concentrations [21, 29, 32–34, 36, 38, 45, 46]. Collectively, these findings suggest that adiponectin may play a regulatory role in metabolic and vascular complications of pregnancy. Adiponectin circulates in human plasma in distinct forms: 1) low-molecular-weight (LMW) trimers; 2) medium-molecular-weight (MMW) hexamers; and 3) high-molecular-weight (HMW) oligomers (12–18 subunits) [270]. These adiponectin multimers can exert distinct biological effects [270], activate different single transduction pathways [271, 272], and may have different affinities to the adiponectin receptors [273]. Consistent with these findings, the ratio of HMW to total adiponectin [270] has a better correlation with insulin resistance [270], obesity [274], cardiovascular diseases [275], and other impaired metabolic states [276, 277] than total adiponectin. Alterations in the relative distribution of adiponectin have been reported in normal gestation [17, 22, 26, 278, 279] as well as in preeclampsia [31, 280], gestational diabetes [52, 281], and delivery of SGA neonates [17, 71, 72, 278–281]. We have previously determined concentrations of circulating maternal adiponectin multimers in women with normal pregnancy and in those with preterm labor, with and without intra-amniotic inflammation/infection [66]. We have found that labor, per se, regardless of the presence of infection/inflammation, is associated with significant quantitative and qualitative alterations in adiponectin multimers. Taken together, the results of our previous and present studies suggest that the differences in the expression of GSTK1 in the subcutaneous adipose tissue between pregnant women with and without labor may provide a molecular mechanism for the altered regulation of adiponectin and adiponectin multimers associated with labor. This, in turn, may be important for the regulation of energy expenditure associated with parturition.

Heat shock 70 kDa protein 5 (HSPA5), also known as 78 kD glucose-regulated protein (GRP78) or immunoglobulin heavy chain-binding protein (BiP) [282, 283], is an ER-resident multifunctional molecular chaperone [284] belonging to the Hsp70 family of heat shock proteins [285]. HSPA5 is a key component of the unfolded protein response (UPR) signaling pathway that plays an important role in ER homeostasis [286]. HSPA5 increases the ER protein folding capacity by forming multiprotein complexes with other ER chaperones and regulates the activity of the ER-transmembrane sensor proteins PERK, IRE1, and ATF6 by sequestering them in inactive complexes [287, 288]. Recently, several studies proposed that increased endoplasmic reticulum stress may represent the proximal cause of the association between obesity and adipocyte insulin resistance [289–291]. Moreover, studies examining human adipose tissue have indicated that there is an increase in the ER stress transcript HSPA5 as a function of increased BMI [292, 293]. Thus, it can be hypothesized that parturition imposes increased metabolic demands and results in ER stress which, in turn, is attenuated by overproduction of HSPA5 in subcutaneous adipose tissue. Further studies are needed to test this hypothesis.

The additional gene affected by alternative splicing in subcutaneous adipose tissue of pregnant women with spontaneous labor at term is LIM and senescent cell antigen-like-containing domain protein 1 (LIMS1). LIM domain proteins contain at least one double zinc-finger motif, and they express mainly in mammalian hearts, particularly in cardiomyocytes [294]. These proteins contain between one and five LIM domains and have been implicated in the development of the heart and heart disorders. There are two members in the five-domain LIM family: LIMS1 and LIMS2. They act as adaptor proteins forming ternary complexes and participate in cell-cell, cell-matrix adhesion, migration, growth, and cell survival [295, 296]. LIMS1 and LIMS2 also function as stress sensors that enable the heart to detect mechanical stretch and respond by increasing contractile force. Other members in this large family have been implicated in the development of the heart [297–302], kidney [303–305], and liver [306] as well as in cancer [307–313] and in neurodegenerative disease [312, 314]. Interestingly, a member of the LIM family, four and a half LIM domains (FHL1), was found to be differentially expressed between visceral and omental adipose tissue in humans. To our knowledge, this report represents the first evidence that LIMS1 is expressed in human adipose tissue. Based on previous reports concerning the physiological role of this gene in other organs, it is tempting to postulate that LIMS1 is involved in the remodeling of the subcutaneous adipose tissue.

Strengths and limitations of the study

The major strengths of this study include the novel findings reported herein: 1) the implementation of a high throughput technique in the investigation of different adipose tissue depots, 2) the evaluation of paired specimens, 3) the inclusion of well-matched controls, and 4) the relatively large sample size. Our results include the first description of the transcriptome of adipose tissue – visceral and subcutaneous – in parturient women. Significant differences in alternative spliced genes were found in the subcutaneous adipose tissue between pregnant women with and without labor, implicating that alternative splicing in labor may be associated with differences in subcutaneous adipose tissue for the first time. We have identified the LIMS1 gene, previously unrecognized, to be expressed in subcutaneous adipose tissue. Several limitations of our study should also be acknowledged. The cross-sectional nature of this study does not allow us to determine either a temporal or a causal relationship between labor and alterations in adipose tissue region-specific gene expression. In addition, as most of the participants in the study were African American, the generalization of our findings to pregnant women of different ethnic origins will require future investigation.

Conclusion

We provide evidence for the association between labor and changes in gene expression in adipose tissue. Specifically, alternative splicing has been implicated in human parturition for the first time, providing a putative molecular mechanism by which regulation of adipose tissue metabolic adaptations to the increased energy demand associated with labor occurs. In addition, we provide evidence that human parturition is characterized by a unique pattern of adipose tissue region-specific alterations in gene expression. Collectively, our data indicate that adipose tissue may play a role in the metabolic regulation of human parturition.


Corresponding authors: Shali Mazaki-Tovi, MD, Department of Obstetrics and Gynecology, Sheba Medical Center, Tel Hashomer, 52621 Israel, Tel.: (+972) 3-530-2169, Fax: (+972) 3-5302922, E-mail: ; and Tel Aviv University, Tel Aviv, Israel; and Roberto Romero, MD, D(Med)Sci, Perinatology Research Branch, NICHD/NIH/ DHHS, Hutzel Women’s Hospital, Box No. 4, 3990 John R, Detroit, MI 48201, USA, Tel.: (+313) 993-2700, Fax: (+313) 993-2694, E-mail: ; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA; Department of Biostatistics and Epidemiology, Michigan State University, East Lansing, MI, USA; and Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA

Acknowledgments

This project was supported, in part, by the Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services.

References

[1] Kashyap ML, Sivasamboo R, Sothy SP, Cheah JS, Gartside PS. Carbohydrate and lipid metabolism during human labor: free fatty acids, glucose, insulin, and lactic acid metabolism during normal and oxytocin-induced labor for postmaturity. Metabolism. 1976;25:865–75.10.1016/0026-0495(76)90119-0Search in Google Scholar

[2] Katz M, Kroll D, Shapiro Y, Cristal N, Meizner I. Energy expenditure in normal labor. Isr J Med Sci. 1990;26:254–7.Search in Google Scholar

[3] Whaley WH, Zuspan FP, Nelson GH, Ahlquist RP. Alterations of plasma free fatty acids and glucose during labor. Am J Obstet Gynecol. 1967;97:875–80.10.1016/0002-9378(67)90510-8Search in Google Scholar

[4] Raivio KO, Teramo K. Blood glucose of the human fetus prior to and during labor. Acta Paediatr Scand. 1968;57:512–6.10.1111/j.1651-2227.1968.tb06971.xSearch in Google Scholar

[5] Paterson P, Phillips L, Wood C. Relationship between maternal and fetal blood glucose during labor. Am J Obstet Gynecol. 1967;98:938–45.10.1016/0002-9378(67)90080-4Search in Google Scholar

[6] Gurson CT, Etili L, Soyak S. Relation between endogenous lipoprotein lipase activity, free fatty acids, and glucose in plasma of women in labour and of their newborns. Arch Dis Child. 1968;43:679–83.10.1136/adc.43.232.679Search in Google Scholar

[7] Felig P, Lynch V. [Starvation in human pregnancy: hypoglycemia, hypoinsulinemia, and hyperketonemia.]. Science. 1970;170:990–2.10.1097/00006254-197106000-00002Search in Google Scholar

[8] Low JA, Pancham SR, Worthington D, Boston RW. Acid-base, lactate, and pyruvate characteristics of the normal obstetric patient and fetus during the intrapartum period. Am J Obstet Gynecol. 1974;120:862–7.10.1016/0002-9378(74)90331-7Search in Google Scholar

[9] Banerjee B, Khew KS, Saha N, Ratnam SS. Energy cost and blood sugar level during different stages of labour and duration of labour in Asiatic women. J Obstet Gynaecol Br Commonw. 1971;78:927–9.10.1111/j.1471-0528.1971.tb00206.xSearch in Google Scholar PubMed

[10] Milwidsky A, Gutman A. Glycogen metabolism of normal human myometrium and leiomyoma – possible hormonal control. Gynecol Obstet Invest. 1983;15:147–52.10.1159/000299405Search in Google Scholar PubMed

[11] Laudanski T. [Energy metabolism of the myometrium in pregnancy and labor]. Zentralbl Gynakol. 1985;107:568–73.Search in Google Scholar

[12] Mittal P, Romero R, Tarca AL, Gonzalez J, Draghici S, Xu Y, et al. Characterization of the myometrial transcriptome and biological pathways of spontaneous human labor at term. J Perinat Med. 2010;38:617–43.10.1515/jpm.2010.097Search in Google Scholar PubMed PubMed Central

[13] Maheux PC, Bonin B, Dizazo A, Guimond P, Monier D, Bourque J, et al. Glucose homeostasis during spontaneous labor in normal human pregnancy. J Clin Endocrinol Metab. 1996;81:209–15.Search in Google Scholar

[14] Scheepers HC, de Jong PA, Essed GG, Kanhai HH. Fetal and maternal energy metabolism during labor in relation to the available caloric substrate. J Perinat Med. 2001;29:457–64.10.1515/JPM.2001.064Search in Google Scholar PubMed

[15] Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444:860–7.10.1038/nature05485Search in Google Scholar PubMed

[16] Rosen ED, Spiegelman BM. Adipocytes as regulators of energy balance and glucose homeostasis. Nature. 2006;444:847–53.10.1038/nature05483Search in Google Scholar PubMed PubMed Central

[17] Catalano PM, Hoegh M, Minium J, Huston-Presley L, Bernard S, Kalhan S, et al. Adiponectin in human pregnancy: implications for regulation of glucose and lipid metabolism. Diabetologia. 2006;49:1677–85.10.1007/s00125-006-0264-xSearch in Google Scholar PubMed

[18] Mazaki-Tovi S, Kanety H, Sivan E. Adiponectin and human pregnancy. Curr Diab Rep. 2005;5:278–81.10.1007/s11892-005-0023-2Search in Google Scholar PubMed

[19] Kajantie E, Hytinantti T, Hovi P, Andersson S. Cord plasma adiponectin: a 20-fold rise between 24 weeks gestation and term. J Clin Endocrinol Metab. 2004;89:4031–6.10.1210/jc.2004-0018Search in Google Scholar PubMed

[20] Mazaki-Tovi S, Kanety H, Pariente C, Hemi R, Wiser A, Schiff E, et al. Maternal serum adiponectin levels during human pregnancy. J Perinatol. 2007;27:77–81.10.1038/sj.jp.7211639Search in Google Scholar PubMed

[21] Cortelazzi D, Corbetta S, Ronzoni S, Pelle F, Marconi A, Cozzi V, et al. Maternal and foetal resistin and adiponectin concentrations in normal and complicated pregnancies. Clin Endocrinol (Oxf). 2007;66:447–53.10.1111/j.1365-2265.2007.02761.xSearch in Google Scholar PubMed

[22] Mazaki-Tovi S, Romero R, Kusanovic JP, Erez O, Vaisbuch E, Gotsch F, et al. Adiponectin multimers in maternal plasma. J Matern Fetal Neonatal Med. 2008;21:796–815.10.1080/14767050802266881Search in Google Scholar PubMed PubMed Central

[23] Nien JK, Mazaki-Tovi S, Romero R, Erez O, Kusanovic JP, Gotsch F, et al. Plasma adiponectin concentrations in non-pregnant, normal and overweight pregnant women. J Perinat Med. 2007;35:522–31.10.1515/JPM.2007.123Search in Google Scholar PubMed PubMed Central

[24] Mazaki-Tovi S, Romero R, Kusanovic JP, Vaisbuch E, Erez O, Than NG, et al. Maternal visfatin concentration in normal pregnancy. J Perinat Med. 2009;37:206–17.10.1515/JPM.2009.054Search in Google Scholar PubMed PubMed Central

[25] Haugen F, Drevon CA. Activation of nuclear factor-kappaB by high molecular weight and globular adiponectin. Endocrinology. 2007;148:5478–86.10.1210/en.2007-0370Search in Google Scholar PubMed

[26] Mazaki-Tovi S, Kanety H, Pariente C, Hemi R, Yissachar E, Schiff E, et al. Insulin sensitivity in late gestation and early postpartum period: the role of circulating maternal adipokines. Gynecol Endocrinol. 2011;27:725–31.10.3109/09513590.2010.500426Search in Google Scholar PubMed

[27] Nien JK, Mazaki-Tovi S, Romero R, Kusanovic JP, Erez O, Gotsch F, et al. Resistin: a hormone which induces insulin resistance is increased in normal pregnancy. J Perinat Med. 2007;35:513–21.10.1515/JPM.2007.122Search in Google Scholar PubMed PubMed Central

[28] Kasher-Meron M, Mazaki-Tovi S, Barhod E, Hemi R, Haas J, Gat I, et al. Chemerin concentrations in maternal and fetal compartments: implications for metabolic adaptations to normal human pregnancy. J Perinat Med. 2014;42:371–8.10.1515/jpm-2013-0166Search in Google Scholar PubMed

[29] D’Anna R, Baviera G, Corrado F, Giordano D, Di Benedetto A, Jasonni VM. Plasma adiponectin concentration in early pregnancy and subsequent risk of hypertensive disorders. Obstet Gynecol 2005;106:340–4.10.1097/01.AOG.0000168441.79050.03Search in Google Scholar PubMed

[30] D’Anna R, Baviera G, Corrado F, Giordano D, De VA, Nicocia G, et al. Adiponectin and insulin resistance in early- and late-onset pre-eclampsia. BJOG. 2006;113:1264–9.10.1111/j.1471-0528.2006.01078.xSearch in Google Scholar PubMed

[31] Mazaki-Tovi S, Romero R, Vaisbuch E, Kusanovic JP, Erez O, Gotsch F, et al. Maternal serum adiponectin multimers in preeclampsia. J Perinat Med. 2009;37:349–63.10.1515/JPM.2009.085Search in Google Scholar PubMed PubMed Central

[32] Haugen F, Ranheim T, Harsem NK, Lips E, Staff AC, Drevon CA. Increased plasma levels of adipokines in preeclampsia: relationship to placenta and adipose tissue gene expression. Am J Physiol Endocrinol Metab. 2006;290:E326–33.10.1152/ajpendo.00020.2005Search in Google Scholar PubMed

[33] Kajantie E, Kaaja R, Ylikorkala O, Andersson S, Laivuori H. Adiponectin concentrations in maternal serum: elevated in preeclampsia but unrelated to insulin sensitivity. J Soc Gynecol Investig. 2005;12:433–9.10.1016/j.jsgi.2005.04.006Search in Google Scholar PubMed

[34] Lu D, Yang X, Wu Y, Wang H, Huang H, Dong M. Serum adiponectin, leptin and soluble leptin receptor in pre-eclampsia. Int J Gynaecol Obstet. 2006;95:121–6.10.1016/j.ijgo.2006.06.015Search in Google Scholar PubMed

[35] Mazaki-Tovi S, Romero R, Kim SK, Vaisbuch E, Kusanovic JP, Erez O, et al. Could alterations in maternal plasma visfatin concentration participate in the phenotype definition of preeclampsia and SGA? J Matern Fetal Neonatal Med. 2010;23:857–68.10.3109/14767050903301017Search in Google Scholar PubMed PubMed Central

[36] Naruse K, Yamasaki M, Umekage H, Sado T, Sakamoto Y, Morikawa H. Peripheral blood concentrations of adiponectin, an adipocyte-specific plasma protein, in normal pregnancy and preeclampsia. J Reprod Immunol. 2005;65:65–75.10.1016/j.jri.2004.09.004Search in Google Scholar PubMed

[37] Nien JK, Mazaki-Tovi S, Romero R, Erez O, Kusanovic JP, Gotsch F, et al. Adiponectin in severe preeclampsia. J Perinat Med. 2007;35:503–12.10.1515/JPM.2007.121Search in Google Scholar PubMed PubMed Central

[38] Ramsay JE, Jamieson N, Greer IA, Sattar N. Paradoxical elevation in adiponectin concentrations in women with preeclampsia. Hypertension. 2003;42:891–4.10.1161/01.HYP.0000095981.92542.F6Search in Google Scholar PubMed

[39] Vaisbuch E, Romero R, Mazaki-Tovi S, Erez O, Kim SK, Chaiworapongsa T, et al. Retinol binding protein 4 – a novel association with early-onset preeclampsia. J Perinat Med. 2010;38:129–39.10.1515/jpm.2009.140Search in Google Scholar PubMed PubMed Central

[40] Chen D, Dong M, Fang Q, He J, Wang Z, Yang X. Alterations of serum resistin in normal pregnancy and pre-eclampsia. Clin Sci (Lond). 2005;108:81–4.10.1042/CS20040225Search in Google Scholar PubMed

[41] Fasshauer M, Waldeyer T, Seeger J, Schrey S, Ebert T, Kratzsch J, et al. Circulating high-molecular-weight adiponectin is upregulated in preeclampsia and is related to insulin sensitivity and renal function. Eur J Endocrinol. 2008;158:197–201.10.1530/EJE-07-0454Search in Google Scholar PubMed

[42] Fasshauer M, Waldeyer T, Seeger J, Schrey S, Ebert T, Kratzsch J, et al. Serum levels of the adipokine visfatin are increased in pre-eclampsia. Clin Endocrinol (Oxf). 2008;69:69–73.10.1111/j.1365-2265.2007.03147.xSearch in Google Scholar PubMed

[43] Seol HJ, Kim JW, Kim HJ. Retinol-binding protein-4 is decreased in patients with preeclampsia in comparison with normal pregnant women. J Perinat Med. 2011;39:287–9.10.1515/jpm.2011.016Search in Google Scholar PubMed

[44] Stepan H, Philipp A, Roth I, Kralisch S, Jank A, Schaarschmidt W, et al. Serum levels of the adipokine chemerin are increased in preeclampsia during and 6 months after pregnancy. Regul Pept. 2011;168:69–72.10.1016/j.regpep.2011.03.005Search in Google Scholar

[45] Suwaki N, Masuyama H, Nakatsukasa H, Masumoto A, Sumida Y, Takamoto N, et al. Hypoadiponectinemia and circulating angiogenic factors in overweight patients complicated with pre-eclampsia. Am J Obstet Gynecol. 2006;195:1687–92.10.1016/j.ajog.2006.04.003Search in Google Scholar

[46] Hendler I, Blackwell SC, Mehta SH, Whitty JE, Russell E, Sorokin Y, et al. The levels of leptin, adiponectin, and resistin in normal weight, overweight, and obese pregnant women with and without preeclampsia. Am J Obstet Gynecol. 2005;193(3 Pt 2):979–83.10.1016/j.ajog.2005.06.041Search in Google Scholar

[47] Briana DD, Malamitsi-Puchner A. Adipocytokines in Normal and Complicated Pregnancies. Reprod Sci. 2009;16:921–37.10.1177/1933719109336614Search in Google Scholar

[48] Fasshauer M, Bluher M, Stumvoll M, Tonessen P, Faber R, Stepan H. Differential regulation of visfatin and adiponectin in pregnancies with normal and abnormal placental function. Clin Endocrinol (Oxf). 2007;66:434–9.10.1111/j.1365-2265.2007.02751.xSearch in Google Scholar

[49] McCarthy JF, Misra DN, Roberts JM. Maternal plasma leptin is increased in preeclampsia and positively correlates with fetal cord concentration. Am J Obstet Gynecol. 1999;180(3 Pt 1):731–6.10.1016/S0002-9378(99)70280-2Search in Google Scholar

[50] Schiff E, Friedman SA, Baumann P, Sibai BM, Romero R. Tumor necrosis factor-alpha in pregnancies associated with preeclampsia or small-for-gestational-age newborns. Am J Obstet Gynecol. 1994;170(5 Pt 1):1224–9.10.1016/S0002-9378(94)70130-XSearch in Google Scholar

[51] Kinalski M, Telejko B, Kuzmicki M, Kretowski A, Kinalska I. Tumor necrosis factor alpha system and plasma adiponectin concentration in women with gestational diabetes. Horm Metab Res 2005;37:450–4.10.1055/s-2005-870238Search in Google Scholar PubMed

[52] Mazaki-Tovi S, Romero R, Vaisbuch E, Erez O, Mittal P, Chaiwaropongsa T, et al. Maternal Serum Adiponectin Multimers in Gestational Diabetes. J Perinat Med. 2009;37:637–50.10.1515/JPM.2009.101Search in Google Scholar PubMed PubMed Central

[53] Ranheim T, Haugen F, Staff AC, Braekke K, Harsem NK, Drevon CA. Adiponectin is reduced in gestational diabetes mellitus in normal weight women. Acta Obstet Gynecol Scand. 2004;83:341–7.10.1111/j.0001-6349.2004.00413.xSearch in Google Scholar PubMed

[54] Worda C, Leipold H, Gruber C, Kautzky-Willer A, Knofler M, Bancher-Todesca D. Decreased plasma adiponectin concentrations in women with gestational diabetes mellitus. Am J Obstet Gynecol. 2004;191:2120–4.10.1016/j.ajog.2004.04.038Search in Google Scholar PubMed

[55] Chan TF, Chen YL, Lee CH, Chou FH, Wu LC, Jong SB, et al. Decreased plasma visfatin concentrations in women with gestational diabetes mellitus. J Soc Gynecol Investig. 2006;13:364–7.10.1016/j.jsgi.2006.04.007Search in Google Scholar PubMed

[56] Chan TF, Chen HS, Chen YC, Lee CH, Chou FH, Chen IJ, et al. Increased serum retinol-binding protein 4 concentrations in women with gestational diabetes mellitus. Reprod Sci. 2007;14:169–74.10.1177/1933719106298407Search in Google Scholar PubMed

[57] Chen D, Fang Q, Chai Y, Wang H, Huang H, Dong M. Serum resistin in gestational diabetes mellitus and early postpartum. Clin Endocrinol (Oxf). 2007;67:208–11.10.1111/j.1365-2265.2007.02862.xSearch in Google Scholar PubMed

[58] Mazaki-Tovi S, Romero R, Kusanovic JP, Vaisbuch E, Erez O, Than NG, et al. Visfatin in human pregnancy: maternal gestational diabetes vis-a-vis neonatal birthweight. J Perinat Med. 2009;37:218–31.10.1515/JPM.2009.053Search in Google Scholar PubMed PubMed Central

[59] Haider DG, Handisurya A, Storka A, Vojtassakova E, Luger A, Pacini G, et al. Visfatin response to glucose is reduced in women with gestational diabetes mellitus. Diabetes Care. 2007;30:1889–91.10.2337/dc07-0013Search in Google Scholar PubMed

[60] Krzyzanowska K, Krugluger W, Mittermayer F, Rahman R, Haider D, Shnawa N, et al. Increased visfatin concentrations in women with gestational diabetes mellitus. Clin Sci (Lond). 2006;110:605–9.10.1042/CS20050363Search in Google Scholar PubMed

[61] Krzyzanowska K, Zemany L, Krugluger W, Schernthaner GH, Mittermayer F, Schnack C, et al. Serum concentrations of retinol-binding protein 4 in women with and without gestational diabetes. Diabetologia. 2008;51:1115–22.10.1007/s00125-008-1009-9Search in Google Scholar PubMed PubMed Central

[62] Kuzmicki M, Telejko B, Szamatowicz J, Zonenberg A, Nikolajuk A, Kretowski A, et al. High resistin and interleukin-6 levels are associated with gestational diabetes mellitus. Gynecol Endocrinol. 2009;25:258–63.10.1080/09513590802653825Search in Google Scholar PubMed

[63] Lewandowski KC, Stojanovic N, Press M, Tuck SM, Szosland K, Bienkiewicz M, et al. Elevated serum levels of visfatin in gestational diabetes: a comparative study across various degrees of glucose tolerance. Diabetologia. 2007;50:1033–7.10.1007/s00125-007-0610-7Search in Google Scholar PubMed

[64] Lewandowski KC, Stojanovic N, Bienkiewicz M, Tan BK, Prelevic GM, Press M, et al. Elevated concentrations of retinol-binding protein-4 (RBP-4) in gestational diabetes mellitus: negative correlation with soluble vascular cell adhesion molecule-1 (sVCAM-1). Gynecol Endocrinol. 2008;24:300–5.10.1080/09513590802141052Search in Google Scholar PubMed

[65] Szamatowicz J, Kuzmicki M, Telejko B, Zonenberg A, Nikolajuk A, Kretowski A, et al. Serum visfatin concentration is elevated in pregnant women irrespectively of the presence of gestational diabetes. Ginekol Pol. 2009;80:14–8.Search in Google Scholar

[66] Mazaki-Tovi S, Romero R., Vaisbuch E, Erez O, Mittal P, Chaiwaropongsa T, et al. Dysregulation of maternal serum adiponectin in preterm labor. J Matern Fetal Neonatal Med. 2009;22:887–904.10.1080/14767050902994655Search in Google Scholar PubMed PubMed Central

[67] Mazaki-Tovi S, Romero R., Vaisbuch E, Erez O, Chaiwaropongsa T, Mittal P, et al. Maternal Plasma Visfatin in Preterm Labor. J Matern Fetal Neonatal Med. 2009;22:693–704.10.1080/14767050902994788Search in Google Scholar PubMed PubMed Central

[68] Mazaki-Tovi S, Romero R, Vaisbuch E, Kim SK, Kusanovic JP, Chaiworapongsa T, et al. Evidence for differential regulation of the adipokine visfatin in the maternal and fetal compartments in normal spontaneous labor at term. J Perinat Med. 2010;38:281–8.10.1515/jpm.2010.045Search in Google Scholar PubMed PubMed Central

[69] Mazaki-Tovi S, Romero R, Vaisbuch E, Kusanovic JP, Chaiworapongsa T, Kim SK, et al. Retinol-binding protein 4: a novel adipokine implicated in the genesis of LGA in the absence of gestational diabetes mellitus. J Perinat Med. 2010;38:147–55.10.1515/jpm.2010.044Search in Google Scholar PubMed PubMed Central

[70] Briana DD, Boutsikou M, Baka S, Gourgiotis D, Marmarinos A, Hassiakos D, et al. Perinatal changes of plasma resistin concentrations in pregnancies with normal and restricted fetal growth. Neonatology. 2008;93:153–7.10.1159/000108412Search in Google Scholar PubMed

[71] Mazaki-Tovi S, Romero R., Vaisbuch E, Erez O, Mittal P, Chaiwaropongsa T, et al. Maternal Serum Adiponectin Multimers in Patients with a Small-For-Gestational-Age Newborn. J Perinat Med. 2009;37:623–35.10.1515/JPM.2009.128Search in Google Scholar PubMed PubMed Central

[72] Catov JM, Patrick TE, Powers RW, Ness RB, Harger G, Roberts JM. Maternal leptin across pregnancy in women with small-for-gestational-age infants. Am J Obstet Gynecol. 2007;196:558.10.1016/j.ajog.2007.01.032Search in Google Scholar PubMed

[73] Mazaki-Tovi S, Vaisbuch E, Romero R, Kusanovic JP, Chaiworapongsa T, Kim SK, et al. Maternal and neonatal circulating visfatin concentrations in patients with pre-eclampsia and a small-for-gestational age neonate. J Matern Fetal Neonatal Med. 2010;23:1119–28.10.3109/14767050903572190Search in Google Scholar PubMed PubMed Central

[74] Briana DD, Malamitsi-Puchner A. The role of adipocytokines in fetal growth. Ann N Y Acad Sci. 2010;1205:82–7.10.1111/j.1749-6632.2010.05650.xSearch in Google Scholar PubMed

[75] Mazaki-Tovi S, Kasher-Meron M, Hemi R, Haas J, Gat I, Lantsberg D, et al. Chemerin is present in human cord blood and is positively correlated with birthweight. Am J Obstet Gynecol. 2012;207:412.e1–10.10.1016/j.ajog.2012.08.008Search in Google Scholar PubMed

[76] Mazaki-Tovi S, Kanety H, Pariente C, Hemi R, Kuint J, Yinon Y, et al. Cord blood adiponectin and infant growth at one year. J Pediatr Endocrinol Metab. 2011;24:411–8.10.1515/jpem.2011.181Search in Google Scholar PubMed

[77] Mazaki-Tovi S, Romero R., Vaisbuch E, Chaiworapongsa T, Erez O, Mittal P, et al. Low circulating maternal adiponectin in patients with pyelonephritis: adiponectin at the crossroads of pregnancy and infection. J Perinat Med. 2009;38:9–17.10.1515/jpm.2009.134Search in Google Scholar PubMed PubMed Central

[78] Mazaki-Tovi S, Vaisbuch E, Romero R, Kusanovic JP, Chaiworapongsa T, Kim SK, et al. Maternal plasma concentration of the pro-inflammatory adipokine pre-B-cell-enhancing factor (PBEF)/visfatin is elevated in pregnant patients with acute pyelonephritis. Am J Reprod Immunol. 2010;63:252–62.10.1111/j.1600-0897.2009.00804.xSearch in Google Scholar PubMed PubMed Central

[79] Vaisbuch E, Romero R, Mazaki-Tovi S, Kusanovic JP, Chaiworapongsa T, Dong Z, et al. Maternal plasma retinol binding protein 4 in acute pyelonephritis during pregnancy. J Perinat Med. 2010;38:359–66.10.1515/jpm.2010.066Search in Google Scholar PubMed PubMed Central

[80] Mazaki-Tovi S, Romero R., Vaisbuch E, Kusanovic JP, Erez O, Mittal P, et al. Adiponectin in amniotic fluid in normal pregnancy, spontaneous labor at term, and preterm labor: a novel association with subclinical intrauterine infection/inflammation. J Matern Fetal Neonatal Med. 2009;23:120–30.10.3109/14767050903026481Search in Google Scholar PubMed PubMed Central

[81] Vaisbuch E, Mazaki-Tovi S, Kusanovic JP, Erez O, Than GN, Kim SK, et al. Retinol binding protein 4: an adipokine associated with intra-amniotic infection/inflammation. J Matern Fetal Neonatal Med. 2010;23:111–9.10.3109/14767050902994739Search in Google Scholar PubMed PubMed Central

[82] Mazaki-Tovi S, Romero R, Kusanovic JP, Erez O, Gotsch F, Mittal P, et al. Visfatin/Pre-B cell colony-enhancing factor in amniotic fluid in normal pregnancy, spontaneous labor at term, preterm labor and prelabor rupture of membranes: an association with subclinical intrauterine infection in preterm parturition. J Perinat Med. 2008;36:485–96.10.1515/JPM.2008.084Search in Google Scholar PubMed PubMed Central

[83] Mazaki-Tovi S, Vaisbuch E, Romero R, Kusanovic JP, Chaiworapongsa T, Kim SK, et al. Hyperresistinemia-a novel feature in systemic infection during human pregnancy. Am J Reprod Immunol. 2010;63:358–69.10.1111/j.1600-0897.2010.00809.xSearch in Google Scholar PubMed PubMed Central

[84] Karatas A, Ozlu T, Erdem A. Maternal metformin, obesity, and metabolic syndrome: the contribution of autonomic nervous system function. Am J Obstet Gynecol. 2014;210:282.10.1016/j.ajog.2013.09.035Search in Google Scholar PubMed

[85] Marrs CC, Moussa HN, Sibai BM, Blackwell SC. The relationship between primary cesarean delivery skin incision type and wound complications in women with morbid obesity. Am J Obstet Gynecol. 2014;210:319–4.10.1016/j.ajog.2014.01.018Search in Google Scholar PubMed

[86] Subramaniam A, Jauk VC, Goss AR, Alvarez MD, Reese C, Edwards RK. Mode of delivery in women with class III obesity: planned cesarean compared with induction of labor. Am J Obstet Gynecol. 2014;211:700–9.10.1016/j.ajog.2014.06.045Search in Google Scholar PubMed

[87] Mackeen AD, Schuster M, Berghella V. Suture versus staples for skin closure after cesarean: a metaanalysis. Am J Obstet Gynecol. 2015;212:621.10.1016/j.ajog.2014.12.020Search in Google Scholar PubMed

[88] Mei-Dan E, Ray JG, Vigod SN. Perinatal outcomes among women with bipolar disorder: a population-based cohort study. Am J Obstet Gynecol. 2015;212:367–8.10.1016/j.ajog.2014.10.020Search in Google Scholar PubMed

[89] Sugerman HJ. Effect of obesity on incidence of preeclampsia. Am J Obstet Gynecol. 2014;210:375.10.1016/j.ajog.2013.10.014Search in Google Scholar PubMed

[90] Zera CA, Seely EW, Wilkins-Haug LE, Lim KH, Parry SI, McElrath TF. The association of body mass index with serum angiogenic markers in normal and abnormal pregnancies. Am J Obstet Gynecol. 2014;211:247.10.1016/j.ajog.2014.03.020Search in Google Scholar PubMed

[91] Kessous R, Shoham-Vardi I, Pariente G, Sergienko R, Holcberg G, Sheiner E. Recurrent pregnancy loss: a risk factor for long-term maternal atherosclerotic morbidity? Am J Obstet Gynecol. 2014;211:414.e1–11.10.1016/j.ajog.2014.05.050Search in Google Scholar PubMed

[92] Bigelow CA, Pereira GA, Warmsley A, Cohen J, Getrajdman C, Moshier E, et al. Risk factors for new-onset late postpartum preeclampsia in women without a history of preeclampsia. Am J Obstet Gynecol. 2014;210:338.10.1016/j.ajog.2013.11.004Search in Google Scholar PubMed

[93] Yao R, Ananth CV, Park BY, Pereira L, Plante LA. Obesity and the risk of stillbirth: a population-based cohort study. Am J Obstet Gynecol. 2014;210:457–9.10.1016/j.ajog.2014.01.044Search in Google Scholar PubMed

[94] Harper LM, Jauk VC, Owen J, Biggio JR. The utility of ultrasound surveillance of fluid and growth in obese women. Am J Obstet Gynecol. 2014;211:524–8.10.1016/j.ajog.2014.04.028Search in Google Scholar PubMed PubMed Central

[95] Lesseur C, Armstrong DA, Paquette AG, Li Z, Padbury JF, Marsit CJ. Maternal obesity and gestational diabetes are associated with placental leptin DNA methylation. Am J Obstet Gynecol. 2014;211:654–9.10.1016/j.ajog.2014.06.037Search in Google Scholar PubMed PubMed Central

[96] Ahmadzia HK, Thomas SM, Dude AM, Grotegut CA, Boyd BK. Prediction of birthweight from third-trimester ultrasound in morbidly obese women. Am J Obstet Gynecol. 2014;211:431–7.10.1016/j.ajog.2014.06.041Search in Google Scholar PubMed

[97] Hermann M, Le RC, Blondel B, Goffinet F, Zeitlin J. The risk of prelabor and intrapartum cesarean delivery among overweight and obese women: possible preventive actions. Am J Obstet Gynecol. 2015;212:241–9.10.1016/j.ajog.2014.08.002Search in Google Scholar PubMed

[98] Acosta O, Ramirez VI, Lager S, Gaccioli F, Dudley DJ, Powell TL, et al. Increased glucose and placental GLUT-1 in large infants of obese nondiabetic mothers. Am J Obstet Gynecol. 2015;212:227.10.1016/j.ajog.2014.08.009Search in Google Scholar PubMed

[99] Daly N, Stapleton M, O’Kelly R, Kinsley B, Daly S, Turner MJ. The role of preanalytical glycolysis in the diagnosis of gestational diabetes mellitus in obese women. Am J Obstet Gynecol. 2015;213:84.e1–5.10.1016/j.ajog.2015.03.022Search in Google Scholar PubMed

[100] Poole AT, Vincent KL, Olson GL, Patrikeev I, Saade GR, Stuebe A, et al. Effect of lactation on maternal postpartum cardiac function and adiposity: a murine model. Am J Obstet Gynecol. 2014;211:424–7.10.1016/j.ajog.2014.06.004Search in Google Scholar PubMed PubMed Central

[101] Sween LK, Althouse AD, Roberts JM. Early-pregnancy percent body fat in relation to preeclampsia risk in obese women. Am J Obstet Gynecol. 2015;212:84–7.10.1016/j.ajog.2014.07.055Search in Google Scholar PubMed

[102] Ceyhan ST, Safer U, Cintosun U. Bioelectric impedance analysis in pregnant women. Am J Obstet Gynecol. 2015;212:120.10.1016/j.ajog.2014.09.033Search in Google Scholar PubMed

[103] Karachaliou M, Georgiou V, Roumeliotaki T, Chalkiadaki G, Daraki V, Koinaki S, et al. Association of trimester-specific gestational weight gain with fetal growth, offspring obesity, and cardiometabolic traits in early childhood. Am J Obstet Gynecol. 2015;212:502.10.1016/j.ajog.2014.12.038Search in Google Scholar PubMed PubMed Central

[104] Hancke K, Gundelach T, Hay B, Sander S, Reister F, Weiss JM. Pre-pregnancy obesity compromises obstetric and neonatal outcomes. J Perinat Med. 2015;43:141–6.10.1515/jpm-2014-0069Search in Google Scholar PubMed

[105] Farren M, Daly N, O’Higgins AC, McKeating A, Maguire PJ, Turner MJ. The interplay between maternal obesity and gestational diabetes mellitus. J Perinat Med. 2015;43:311–7.10.1515/jpm-2014-0272Search in Google Scholar PubMed

[106] O’Higgins AC, Doolan A, Mullaney L, Daly N, McCartney D, Turner MJ. The relationship between gestational weight gain and fetal growth: time to take stock? J Perinat Med. 2014;42:409–15.10.1515/jpm-2013-0209Search in Google Scholar PubMed

[107] Kevane B, Donnelly J, D’Alton M, Cooley S, Preston RJ, Ni AF. Risk factors for pregnancy-associated venous thromboembolism: a review. J Perinat Med. 2014;42:417–25.10.1515/jpm-2013-0207Search in Google Scholar PubMed

[108] Zollner U, Dietl J. Perinatal risks after IVF and ICSI. J Perinat Med. 2013;41:17–22.10.1515/jpm-2012-0097Search in Google Scholar PubMed

[109] Gubler T, Krahenmann F, Roos M, Zimmermann R, Ochsenbein-Kolble N. Determinants of successful breastfeeding initiation in healthy term singletons: a Swiss university hospital observational study. J Perinat Med. 2013;41:331–9.10.1515/jpm-2012-0102Search in Google Scholar PubMed

[110] Willis K, Sheiner E. Bariatric surgery and pregnancy: the magical solution? J Perinat Med. 2013;41:133–40.10.1515/jpm-2012-0237Search in Google Scholar PubMed

[111] Galjaard S, Devlieger R, Van Assche FA. Fetal growth and developmental programming. J Perinat Med. 2013;41:101–5.10.1515/jpm-2012-0020Search in Google Scholar PubMed

[112] Horosz E, Bomba-Opon DA, Szymanska M, Wielgos M. Maternal weight gain in women with gestational diabetes mellitus. J Perinat Med. 2013;41:523–8.10.1515/jpm-2012-0254Search in Google Scholar PubMed

[113] Soni S, Chivan N, Cohen WR. Effect of maternal body mass index on oxytocin treatment for arrest of dilatation. J Perinat Med. 2013;41:517–21.10.1515/jpm-2013-0024Search in Google Scholar PubMed

[114] Tchernof A, Belanger C, Morisset AS, Richard C, Mailloux J, Laberge P, et al. Regional differences in adipose tissue metabolism in women: minor effect of obesity and body fat distribution. Diabetes. 2006;55:1353–60.10.2337/db05-1439Search in Google Scholar PubMed

[115] Dicker A, Astrom G, Wahlen K, Hoffstedt J, Naslund E, Wiren M, et al. Primary differences in lipolysis between human omental and subcutaneous adipose tissue observed using in vitro differentiated adipocytes. Horm Metab Res. 2009;41:350–5.10.1055/s-0028-1112135Search in Google Scholar PubMed

[116] Drolet R, Belanger C, Fortier M, Huot C, Mailloux J, Legare D, et al. Fat depot-specific impact of visceral obesity on adipocyte adiponectin release in women. Obesity (Silver Spring). 2009;17:424–30.10.1038/oby.2008.555Search in Google Scholar PubMed

[117] Kuzmicki M, Telejko B, Wawrusiewicz-Kurylonek N, Kalejta K, Lemancewicz A, Zdrodowski M, et al. The expression of transcription factor 7-like 2 (TCF7L2) in fat and placental tissue from women with gestational diabetes. Diabetes Res Clin Pract. 2011;94:e43–6.10.1016/j.diabres.2011.07.033Search in Google Scholar PubMed

[118] Kuzmicki M, Telejko B, Wawrusiewicz-Kurylonek N, Nikolajuk A, Zwierz-Gugala D, Jelski W, et al. Retinol-binding protein 4 in adipose and placental tissue of women with gestational diabetes. Gynecol Endocrinol. 2011;27:1065–9.10.3109/09513590.2011.579651Search in Google Scholar PubMed

[119] Ma Y, Cheng Y, Wang J, Cheng H, Zhou S, Li X. The changes of visfatin in serum and its expression in fat and placental tissue in pregnant women with gestational diabetes. Diabetes Res Clin Pract. 2010;90:60–5.10.1016/j.diabres.2010.06.010Search in Google Scholar PubMed

[120] Lappas M, Mitton A, Permezel M. In response to oxidative stress, the expression of inflammatory cytokines and antioxidant enzymes are impaired in placenta, but not adipose tissue, of women with gestational diabetes. J Endocrinol. 2010;204:75–84.10.1677/JOE-09-0321Search in Google Scholar PubMed

[121] Kleiblova P, Dostalova I, Bartlova M, Lacinova Z, Ticha I, Krejci V, et al. Expression of adipokines and estrogen receptors in adipose tissue and placenta of patients with gestational diabetes mellitus. Mol Cell Endocrinol. 2010;314:150–6.10.1016/j.mce.2009.08.002Search in Google Scholar PubMed

[122] Morgan SA, Bringolf JB, Seidel ER. Visfatin expression is elevated in normal human pregnancy. Peptides. 2008;29:1382–9.10.1016/j.peptides.2008.04.010Search in Google Scholar PubMed

[123] Barker G, Lim R, Georgiou HM, Lappas M. Omentin-1 is decreased in maternal plasma, placenta and adipose tissue of women with pre-existing obesity. PLoS One. 2012;7:e42943.10.1371/journal.pone.0042943Search in Google Scholar PubMed PubMed Central

[124] Samaras K, Botelho NK, Chisholm DJ, Lord RV. Subcutaneous and visceral adipose tissue gene expression of serum adipokines that predict type 2 diabetes. Obesity (Silver Spring). 2010;18:884–9.10.1038/oby.2009.443Search in Google Scholar PubMed

[125] Walker GE, Marzullo P, Verti B, Guzzaloni G, Maestrini S, Zurleni F, et al. Subcutaneous abdominal adipose tissue subcompartments: potential role in rosiglitazone effects. Obesity (Silver Spring). 2008;16:1983–91.10.1038/oby.2008.326Search in Google Scholar PubMed

[126] Dolinkova M, Dostalova I, Lacinova Z, Michalsky D, Haluzikova D, Mraz M, et al. The endocrine profile of subcutaneous and visceral adipose tissue of obese patients. Mol Cell Endocrinol. 2008;291:63–70.10.1016/j.mce.2008.05.001Search in Google Scholar PubMed

[127] Insenser M, Montes-Nieto R, Vilarrasa N, Lecube A, Simo R, Vendrell J, et al. A nontargeted proteomic approach to the study of visceral and subcutaneous adipose tissue in human obesity. Mol Cell Endocrinol. 2012;363:10–9.10.1016/j.mce.2012.07.001Search in Google Scholar PubMed

[128] Avram MM, Avram AS, James WD. Subcutaneous fat in normal and diseased states 3. Adipogenesis: from stem cell to fat cell. J Am Acad Dermatol. 2007;56:472–92.10.1016/j.jaad.2006.06.022Search in Google Scholar PubMed

[129] Avram AS, Avram MM, James WD. Subcutaneous fat in normal and diseased states: 2. Anatomy and physiology of white and brown adipose tissue. J Am Acad Dermatol. 2005;53:671–83.10.1016/j.jaad.2005.05.015Search in Google Scholar PubMed

[130] Avram MM, Avram AS, James WD. Subcutaneous fat in normal and diseased states: 1. Introduction. J Am Acad Dermatol. 2005;53:663–70.10.1016/j.jaad.2005.05.014Search in Google Scholar PubMed

[131] Gealekman O, Guseva N, Hartigan C, Apotheker S, Gorgoglione M, Gurav K, et al. Depot-specific differences and insufficient subcutaneous adipose tissue angiogenesis in human obesity. Circulation 2011;123:186–94.10.1161/CIRCULATIONAHA.110.970145Search in Google Scholar PubMed PubMed Central

[132] Kloting N, Berthold S, Kovacs P, Schon MR, Fasshauer M, Ruschke K, et al. MicroRNA expression in human omental and subcutaneous adipose tissue. PLoS One. 2009;4:e4699.10.1371/journal.pone.0004699Search in Google Scholar PubMed PubMed Central

[133] Linder K, Arner P, Flores-Morales A, Tollet-Egnell P, Norstedt G. Differentially expressed genes in visceral or subcutaneous adipose tissue of obese men and women. J Lipid Res. 2004;45:148–54.10.1194/jlr.M300256-JLR200Search in Google Scholar PubMed

[134] Mathur SK, Jain P, Mathur P. Microarray evidences the role of pathologic adipose tissue in insulin resistance and their clinical implications. J Obes. 2011;2011:587495.10.1155/2011/587495Search in Google Scholar PubMed PubMed Central

[135] Mutch DM, Tordjman J, Pelloux V, Hanczar B, Henegar C, Poitou C, et al. Needle and surgical biopsy techniques differentially affect adipose tissue gene expression profiles. Am J Clin Nutr. 2009;89:51–7.10.3945/ajcn.2008.26802Search in Google Scholar PubMed

[136] Vohl MC, Sladek R, Robitaille J, Gurd S, Marceau P, Richard D, et al. A survey of genes differentially expressed in subcutaneous and visceral adipose tissue in men. Obes Res. 2004;12:1217–22.10.1038/oby.2004.153Search in Google Scholar PubMed

[137] Zha JM, Di WJ, Zhu T, Xie Y, Yu J, Liu J, et al. Comparison of gene transcription between subcutaneous and visceral adipose tissue in Chinese adults. Endocr J 2009;56:935–44.10.1507/endocrj.K09E-091Search in Google Scholar

[138] de Souza Batista CM, Yang RZ, Lee MJ, Glynn NM, Yu DZ, Pray J, et al. Omentin plasma levels and gene expression are decreased in obesity. Diabetes. 2007;56:1655–61.10.2337/db06-1506Search in Google Scholar PubMed

[139] Van H, V, Dicker A, Ryden M, Hauner H, Lonnqvist F, Naslund E, et al. Increased lipolysis and decreased leptin production by human omental as compared with subcutaneous preadipocytes. Diabetes. 2002;51:2029–36.10.2337/diabetes.51.7.2029Search in Google Scholar PubMed

[140] Guan H, Arany E, van Beek JP, Chamson-Reig A, Thyssen S, Hill DJ, et al. Adipose tissue gene expression profiling reveals distinct molecular pathways that define visceral adiposity in offspring of maternal protein-restricted rats. Am J Physiol Endocrinol Metab. 2005;288:E663–73.10.1152/ajpendo.00461.2004Search in Google Scholar PubMed

[141] Lee YS. The role of genes in the current obesity epidemic. Ann Acad Med Singapore. 2009;38:45–3.10.47102/annals-acadmedsg.V38N1p45Search in Google Scholar

[142] Copland JA, Davies PJ, Shipley GL, Wood CG, Luxon BA, Urban RJ. The use of DNA microarrays to assess clinical samples: the transition from bedside to bench to bedside. Recent Prog Horm Res. 2003;58:25–53.10.1210/rp.58.1.25Search in Google Scholar PubMed

[143] Einstein FH, Atzmon G, Yang XM, Ma XH, Rincon M, Rudin E, et al. Differential responses of visceral and subcutaneous fat depots to nutrients. Diabetes. 2005;54:672–8.10.2337/diabetes.54.3.672Search in Google Scholar PubMed

[144] Einstein FH, Fishman S, Muzumdar RH, Yang XM, Atzmon G, Barzilai N. Accretion of visceral fat and hepatic insulin resistance in pregnant rats. Am J Physiol Endocrinol Metab. 2008;294:E451–5.10.1152/ajpendo.00570.2007Search in Google Scholar PubMed

[145] Despres JP, Lemieux I. Abdominal obesity and metabolic syndrome. Nature. 2006;444:881–7.10.1007/978-0-387-32164-6_8Search in Google Scholar

[146] Van Gaal LF, Mertens IL, De Block CE. Mechanisms linking obesity with cardiovascular disease. Nature. 2006;444:875–80.10.1038/nature05487Search in Google Scholar PubMed

[147] Russell AW, McIntyre HD, Whitehead JP, Prins JB. Adipose tissue from pregnant women with and without gestational diabetes mellitus: insulin-sensitive but resistant to hyperosomolarity. Am J Obstet Gynecol. 2005;193:2017–23.10.1016/j.ajog.2005.04.058Search in Google Scholar PubMed

[148] Wronska A, Kmiec Z. Structural and biochemical characteristics of various white adipose tissue depots. Acta Physiol (Oxf). 2012;205:194–208.10.1111/j.1748-1716.2012.02409.xSearch in Google Scholar PubMed

[149] Wajchenberg BL. Subcutaneous and visceral adipose tissue: their relation to the metabolic syndrome. Endocr Rev. 2000;21:697–738.10.1210/edrv.21.6.0415Search in Google Scholar PubMed

[150] Fox CS, Massaro JM, Hoffmann U, Pou KM, Maurovich-Horvat P, Liu CY, et al. Abdominal visceral and subcutaneous adipose tissue compartments: association with metabolic risk factors in the Framingham Heart Study. Circulation. 2007;116:39–48.10.1161/CIRCULATIONAHA.106.675355Search in Google Scholar PubMed

[151] von Eyben FE, Kroustrup JP, Larsen JF, Celis J. Comparison of gene expression in intra-abdominal and subcutaneous fat: a study of men with morbid obesity and nonobese men using microarray and proteomics. Ann NY Acad Sci. 2004;1030:508–36.10.1196/annals.1329.063Search in Google Scholar PubMed

[152] Langin D, Holm C, Lafontan M. Adipocyte hormone-sensitive lipase: a major regulator of lipid metabolism. Proc Nutr Soc. 1996;55:93–109.10.1079/PNS19960013Search in Google Scholar

[153] Giusti V, Suter M, Verdumo C, Gaillard RC, Burckhardt P, Pralong FP. Molecular determinants of human adipose tissue: differences between visceral and subcutaneous compartments in obese women. J Clin Endocrinol Metab. 2004;89:1379–84.10.1210/jc.2003-031507Search in Google Scholar PubMed

[154] Vidal H. Gene expression in visceral and subcutaneous adipose tissues. Ann Med. 2001;33:547–55.10.3109/07853890108995965Search in Google Scholar PubMed

[155] Lihn AS, Bruun JM, He G, Pedersen SB, Jensen PF, Richelsen B. Lower expression of adiponectin mRNA in visceral adipose tissue in lean and obese subjects. Mol Cell Endocrinol. 2004;219:9–15.10.1016/j.mce.2004.03.002Search in Google Scholar PubMed

[156] Ryden M, Elizalde M, Van H, V, Ohlund A, Hoffstedt J, Bringman S, et al. Increased expression of eNOS protein in omental versus subcutaneous adipose tissue in obese human subjects. Int J Obes Relat Metab Disord. 2001;25:811–5.10.1038/sj.ijo.0801625Search in Google Scholar

[157] Livingston JN, Lerea KM, Bolinder J, Kager L, Backman L, Arner P. Binding and molecular weight properties of the insulin receptor from omental and subcutaneous adipocytes in human obesity. Diabetologia. 1984;27:447–53.10.1007/BF00273909Search in Google Scholar

[158] Dusserre E, Moulin P, Vidal H. Differences in mRNA expression of the proteins secreted by the adipocytes in human subcutaneous and visceral adipose tissues. Biochim Biophys Acta. 2000;1500:88–96.10.1016/S0925-4439(99)00091-5Search in Google Scholar

[159] Marette A, Mauriege P, Marcotte B, Atgie C, Bouchard C, Theriault G, et al. Regional variation in adipose tissue insulin action and GLUT4 glucose transporter expression in severely obese premenopausal women. Diabetologia. 1997;40:590–8.10.1007/s001250050720Search in Google Scholar PubMed

[160] Atzmon G, Yang XM, Muzumdar R, Ma XH, Gabriely I, Barzilai N. Differential gene expression between visceral and subcutaneous fat depots. Horm Metab Res. 2002;34:622–8.10.1055/s-2002-38250Search in Google Scholar PubMed

[161] Lefebvre AM, Laville M, Vega N, Riou JP, van GL, Auwerx J, et al. Depot-specific differences in adipose tissue gene expression in lean and obese subjects. Diabetes. 1998;47:98–103.10.2337/diab.47.1.98Search in Google Scholar PubMed

[162] Montague CT, Prins JB, Sanders L, Zhang J, Sewter CP, Digby J, et al. Depot-related gene expression in human subcutaneous and omental adipocytes. Diabetes. 1998;47:1384–91.10.2337/diabetes.47.9.1384Search in Google Scholar PubMed

[163] Lee MJ, Gong DW, Burkey BF, Fried SK. Pathways regulated by glucocorticoids in omental and subcutaneous human adipose tissues: a microarray study. Am J Physiol Endocrinol Metab. 2011;300:E571–80.10.1152/ajpendo.00231.2010Search in Google Scholar PubMed PubMed Central

[164] Van Harmelen V, Reynisdottir S, Eriksson P, Thorne A, Hoffstedt J, Lonnqvist F, et al. Leptin secretion from subcutaneous and visceral adipose tissue in women. Diabetes. 1998;47:913–7.10.2337/diabetes.47.6.913Search in Google Scholar PubMed

[165] Masuzaki H, Ogawa Y, Isse N, Satoh N, Okazaki T, Shigemoto M, et al. Human obese gene expression. Adipocyte-specific expression and regional differences in the adipose tissue. Diabetes. 1995;44:855–8.10.2337/diab.44.7.855Search in Google Scholar PubMed

[166] Hube F, Lietz U, Igel M, Jensen PB, Tornqvist H, Joost HG, et al. Difference in leptin mRNA levels between omental and subcutaneous abdominal adipose tissue from obese humans. Horm Metab Res. 1996;28:690–3.10.1055/s-2007-979879Search in Google Scholar PubMed

[167] Hojbjerre L, Alibegovic AC, Sonne MP, Dela F, Vaag A, Bruun JM, et al. Increased lipolysis but diminished gene expression of lipases in subcutaneous adipose tissue of healthy young males with intrauterine growth retardation. J Appl Physiol. 2011;111:1863–70.10.1152/japplphysiol.00960.2011Search in Google Scholar PubMed

[168] Liu Y, Zhu L, Pan Y, Sun L, Chen D, Li X. Adiponectin levels in circulation and breast milk and mRNA expression in adipose tissue of preeclampsia women. Hypertens Pregnancy. 2012;31:40–9.10.3109/10641955.2010.525273Search in Google Scholar PubMed

[169] Telejko B, Kuzmicki M, Zonenberg A, Modzelewska A, Niedziolko-Bagniuk K, Ponurkiewicz A, et al. Ghrelin in gestational diabetes: serum level and mRNA expression in fat and placental tissue. Exp Clin Endocrinol Diabetes. 2010;118:87–92.10.1055/s-0029-1238313Search in Google Scholar PubMed

[170] Colomiere M, Permezel M, Lappas M. Diabetes and obesity during pregnancy alter insulin signalling and glucose transporter expression in maternal skeletal muscle and subcutaneous adipose tissue. J Mol Endocrinol. 2010;44:213–23.10.1677/JME-09-0091Search in Google Scholar PubMed

[171] Telejko B, Kuzmicki M, Wawrusiewicz-Kurylonek N, Szamatowicz J, Nikolajuk A, Zonenberg A, et al. Plasma apelin levels and apelin/APJ mRNA expression in patients with gestational diabetes mellitus. Diabetes Res Clin Pract. 2010;87:176–83.10.1016/j.diabres.2009.10.018Search in Google Scholar PubMed

[172] Telejko B, Kuzmicki M, Zonenberg A, Szamatowicz J, Wawrusiewicz-Kurylonek N, Nikolajuk A, et al. Visfatin in gestational diabetes: serum level and mRNA expression in fat and placental tissue. Diabetes Res Clin Pract. 2009;84:68–75.10.1016/j.diabres.2008.12.017Search in Google Scholar PubMed

[173] Orcy RB, Schroeder S, Martins-Costa SH, Ramos JG, Schechinger W, Klein H, et al. Signalization of Akt/PKB in the placenta, skeletal muscle and adipose tissue of preeclampsia patients. Gynecol Obstet Invest. 2008;66:231–6.10.1159/000147169Search in Google Scholar PubMed

[174] Zhou Y, Zhang M, Guo W, Yu M, Xue K, Huang S, et al. Expression of resistin protein in normal human subcutaneous adipose tissue and pregnant women subcutaneous adipose tissue and placenta. J Huazhong Univ Sci Technolog Med Sci. 2006;26:288–91.10.1007/BF02829553Search in Google Scholar PubMed

[175] Okuno S, Akazawa S, Yasuhi I, Kawasaki E, Matsumoto K, Yamasaki H, et al. Decreased expression of the GLUT4 glucose transporter protein in adipose tissue during pregnancy. Horm Metab Res. 1995;27:231–4.10.1055/s-2007-979946Search in Google Scholar PubMed

[176] Donker RB, Molema G, Faas MM, Kallenberg CG, van Pampus MG, Timmer A, et al. Absence of in vivo generalized pro-inflammatory endothelial activation in severe, early-onset preeclampsia. J Soc Gynecol Investig. 2005;12:518–28.10.1016/j.jsgi.2005.06.007Search in Google Scholar

[177] Mackay VA, Huda SS, Stewart FM, Tham K, McKenna LA, Martin I, et al. Preeclampsia is associated with compromised maternal synthesis of long-chain polyunsaturated fatty acids, leading to offspring deficiency. Hypertension. 2012;60:1078–85.10.1161/HYPERTENSIONAHA.112.197897Search in Google Scholar

[178] Resi V, Basu S, Haghiac M, Presley L, Minium J, Kaufman B, et al. Molecular inflammation and adipose tissue matrix remodeling precede physiological adaptations to pregnancy. Am J Physiol Endocrinol Metab. 2012;303:E832–40.10.1152/ajpendo.00002.2012Search in Google Scholar

[179] Bemmo A, Benovoy D, Kwan T, Gaffney DJ, Jensen RV, Majewski J. Gene expression and isoform variation analysis using Affymetrix Exon Arrays. BMC Genomics. 2008;9:529.10.1186/1471-2164-9-529Search in Google Scholar

[180] Pacora P, Chaiworapongsa T, Maymon E, Kim YM, Gomez R, Yoon BH, et al. Funisitis and chorionic vasculitis: the histological counterpart of the fetal inflammatory response syndrome. J Matern Fetal Med. 2002;11:18–25.10.1080/jmf.11.1.18.25Search in Google Scholar

[181] Redline RW. Inflammatory responses in the placenta and umbilical cord. Semin Fetal Neonatal Med. 2006;11:296–301.10.1016/j.siny.2006.02.011Search in Google Scholar

[182] Alexander GR, Himes JH, Kaufman RB, Mor J, Kogan M. A United States national reference for fetal growth. Obstet Gynecol. 1996;87:163–8.10.1016/0029-7844(95)00386-XSearch in Google Scholar

[183] Bashan N, Dorfman K, Tarnovscki T, Harman-Boehm I, Liberty IF, Bluher M, et al. Mitogen-activated protein kinases, inhibitory-kappaB kinase, and insulin signaling in human omental versus subcutaneous adipose tissue in obesity. Endocrinology. 2007;148:2955–62.10.1210/en.2006-1369Search in Google Scholar PubMed

[184] Harman-Boehm I, Bluher M, Redel H, Sion-Vardy N, Ovadia S, Avinoach E, et al. Macrophage infiltration into omental versus subcutaneous fat across different populations: effect of regional adiposity and the comorbidities of obesity. J Clin Endocrinol Metab. 2007;92:2240–7.10.1210/jc.2006-1811Search in Google Scholar PubMed

[185] Berndt J, Kloting N, Kralisch S, Kovacs P, Fasshauer M, Schon MR, et al. Plasma visfatin concentrations and fat depot-specific mRNA expression in humans. Diabetes. 2005;54:2911–6.10.2337/diabetes.54.10.2911Search in Google Scholar PubMed

[186] Bluher M, Bashan N, Shai I, Harman-Boehm I, Tarnovscki T, Avinaoch E, et al. Activated Ask1-MKK4-p38MAPK/JNK stress signaling pathway in human omental fat tissue may link macrophage infiltration to whole-body Insulin sensitivity. J Clin Endocrinol Metab. 2009;94:2507–15.10.1210/jc.2009-0002Search in Google Scholar

[187] Gardina PJ, Clark TA, Shimada B, Staples MK, Yang Q, Veitch J, et al. Alternative splicing and differential gene expression in colon cancer detected by a whole genome exon array. BMC Genomics. 2006;7:325.10.1186/1471-2164-7-325Search in Google Scholar

[188] Kurokawa K, Kuwano Y, Tominaga K, Kawai T, Katsuura S, Yamagishi N, et al. Brief naturalistic stress induces an alternative splice variant of SMG-1 lacking exon 63 in peripheral leukocytes. Neurosci Lett. 2010;484:128–32.10.1016/j.neulet.2010.08.031Search in Google Scholar

[189] Zhang W, Duan S, Bleibel WK, Wisel SA, Huang RS, Wu X, et al. Identification of common genetic variants that account for transcript isoform variation between human populations. Hum Genet. 2009;125:81–93.10.1007/s00439-008-0601-xSearch in Google Scholar

[190] Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003;4:249–64.10.1093/biostatistics/4.2.249Search in Google Scholar

[191] Bengtsson H, Simpson K, Bullard J, Hansen K. Aroma.affymetrix: a generic framework in R for analyzing small to very large affymetrix data sets in bounded memory. Tech Report # 745 of the Department of Statistics, University of California, Berkeley, 2008.Search in Google Scholar

[192] Smyth GK. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004;3:Article3.10.2202/1544-6115.1027Search in Google Scholar

[193] Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I. Controlling the false discovery rate in behavior genetics research. Behav Brain Res. 2001;125:279–84.10.1016/S0166-4328(01)00297-2Search in Google Scholar

[194] Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA. 2001;98:5116–21.10.1073/pnas.091062498Search in Google Scholar PubMed PubMed Central

[195] Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5:R80.10.1186/gb-2004-5-10-r80Search in Google Scholar PubMed PubMed Central

[196] Draghici S, Khatri P, Tarca AL, Amin K, Done A, Voichita C, et al. A systems biology approach for pathway level analysis. Genome Res. 2007;17:1537–45.10.1101/gr.6202607Search in Google Scholar PubMed PubMed Central

[197] Khatri P, Draghici S, Ostermeier GC, Krawetz SA. Profiling gene expression using onto-express. Genomics. 2002;79:266–70.10.1006/geno.2002.6698Search in Google Scholar PubMed

[198] Tarca AL, Draghici S, Bhatti G, Romero R. Down-weighting overlapping genes improves gene set analysis. BMC Bioinformatics. 2012;13:136.10.1186/1471-2105-13-136Search in Google Scholar PubMed PubMed Central

[199] Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–50.10.1073/pnas.0506580102Search in Google Scholar PubMed PubMed Central

[200] Purdom E, Simpson KM, Robinson MD, Conboy JG, Lapuk AV, Speed TP. FIRMA: a method for detection of alternative splicing from exon array data. Bioinformatics. 2008;24:1707–14.10.1093/bioinformatics/btn284Search in Google Scholar PubMed PubMed Central

[201] Clark TA, Schweitzer AC, Chen TX, Staples MK, Lu G, Wang H, et al. Discovery of tissue-specific exons using comprehensive human exon microarrays. Genome Biol. 2007;8:R64.10.1186/gb-2007-8-4-r64Search in Google Scholar PubMed PubMed Central

[202] Joshi-Tope G, Gillespie M, Vastrik I, D’Eustachio P, Schmidt E, de BB, et al. Reactome: a knowledgebase of biological pathways. Nucleic Acids Res. 2005;33(Database issue):D428–32.10.1093/nar/gki072Search in Google Scholar PubMed PubMed Central

[203] Khoury MJ, Romero R. The integration of genomics into obstetrics and gynecology: a HuGE challenge. Am J Obstet Gynecol. 2006;195:1503–5.10.1016/j.ajog.2006.10.883Search in Google Scholar PubMed PubMed Central

[204] Kolialexi A, Mavrou A, Spyrou G, Tsangaris GT. Mass spectrometry-based proteomics in reproductive medicine. Mass Spectrom Rev. 2008;27:624–34.10.1002/mas.20181Search in Google Scholar PubMed

[205] Romero R, Espinoza J, Gotsch F, Kusanovic JP, Friel LA, Erez O, et al. The use of high-dimensional biology (genomics, transcriptomics, proteomics, and metabolomics) to understand the preterm parturition syndrome. BJOG. 2006;113(Suppl 3):118–35.10.1111/j.1471-0528.2006.01150.xSearch in Google Scholar PubMed PubMed Central

[206] Romero R, Tromp G. High-dimensional biology in obstetrics and gynecology: functional genomics in microarray studies. Am J Obstet Gynecol. 2006;195:360–3.10.1016/j.ajog.2006.06.077Search in Google Scholar PubMed

[207] Romero R, Kusanovic JP, Gotsch F, Erez O, Vaisbuch E, Mazaki-Tovi S, et al. Isobaric labeling and tandem mass spectrometry: a novel approach for profiling and quantifying proteins differentially expressed in amniotic fluid in preterm labor with and without intra-amniotic infection/inflammation. J Matern Fetal Neonatal Med. 2010;261:261–80.10.3109/14767050903067386Search in Google Scholar PubMed

[208] Romero R, Mazaki-Tovi S, Vaisbuch E, Kusanovic JP, Chaiworapongsa T, Gomez R, et al. Metabolomics in premature labor: a novel approach to identify patients at risk for preterm delivery. J Matern Fetal Neonatal Med. 2010;23:1344–59.10.3109/14767058.2010.482618Search in Google Scholar PubMed PubMed Central

[209] Hassan SS, Romero R, Haddad R, Hendler I, Khalek N, Tromp G, et al. The transcriptome of the uterine cervix before and after spontaneous term parturition. Am J Obstet Gynecol. 2006;195:778–86.10.1016/j.ajog.2006.06.021Search in Google Scholar PubMed

[210] Hassan SS, Romero R, Tarca AL, Draghici S, Pineles B, Bugrim A, et al. Signature pathways identified from gene expression profiles in the human uterine cervix before and after spontaneous term parturition. Am J Obstet Gynecol. 2007;197:250–7.10.1016/j.ajog.2007.07.008Search in Google Scholar PubMed PubMed Central

[211] Hassan SS, Romero R, Tarca AL, Nhan-Chang CL, Vaisbuch E, Erez O, et al. The transcriptome of cervical ripening in human pregnancy before the onset of labor at term: identification of novel molecular functions involved in this process. J Matern Fetal Neonatal Med. 2009;22:1183–93.10.3109/14767050903353216Search in Google Scholar PubMed PubMed Central

[212] Hassan SS, Romero R, Tarca AL, Nhan-Chang CL, Mittal P, Vaisbuch E, et al. The molecular basis for sonographic cervical shortening at term: identification of differentially expressed genes and the epithelial-mesenchymal transition as a function of cervical length. Am J Obstet Gynecol. 2010;203:472.10.1016/j.ajog.2010.06.076Search in Google Scholar PubMed

[213] Hassan SS, Romero R, Pineles B, Tarca AL, Montenegro D, Erez O, et al. MicroRNA expression profiling of the human uterine cervix after term labor and delivery. Am J Obstet Gynecol. 2010;202:80–8.10.1016/j.ajog.2009.08.016Search in Google Scholar PubMed

[214] Huber A, Hudelist G, Czerwenka K, Husslein P, Kubista E, Singer CF. Gene expression profiling of cervical tissue during physiological cervical effacement. Obstet Gynecol. 2005;105:91–8.10.1097/01.AOG.0000146636.61611.e3Search in Google Scholar PubMed

[215] Mowa CN, Li T, Jesmin S, Folkesson HG, Usip SE, Papka RE, et al. Delineation of VEGF-regulated genes and functions in the cervix of pregnant rodents by DNA microarray analysis. Reprod Biol Endocrinol. 2008;6:64.10.1186/1477-7827-6-64Search in Google Scholar PubMed PubMed Central

[216] Read CP, Word RA, Ruscheinsky MA, Timmons BC, Mahendroo MS. Cervical remodeling during pregnancy and parturition: molecular characterization of the softening phase in mice. Reproduction. 2007;134:327–40.10.1530/REP-07-0032Search in Google Scholar PubMed

[217] Wang H, Stjernholm Y, Ekman G, Eriksson H, Sahlin L. Different regulation of oestrogen receptors alpha and beta in the human cervix at term pregnancy. Mol Hum Reprod. 2001;7:293–300.10.1093/molehr/7.3.293Search in Google Scholar PubMed

[218] Bethin KE, Nagai Y, Sladek R, Asada M, Sadovsky Y, Hudson TJ, et al. Microarray analysis of uterine gene expression in mouse and human pregnancy. Mol Endocrinol. 2003;17:1454–69.10.1210/me.2003-0007Search in Google Scholar PubMed

[219] Bukowski R, Hankins GD, Saade GR, Anderson GD, Thornton S. Labor-associated gene expression in the human uterine fundus, lower segment, and cervix. PLoS Med. 2006;3:e169.10.1371/journal.pmed.0030169Search in Google Scholar PubMed PubMed Central

[220] Chan EC, Fraser S, Yin S, Yeo G, Kwek K, Fairclough RJ, et al. Human myometrial genes are differentially expressed in labor: a suppression subtractive hybridization study. J Clin Endocrinol Metab. 2002;87:2435–41.10.1210/jcem.87.6.8439Search in Google Scholar PubMed

[221] Esplin MS, Fausett MB, Peltier MR, Hamblin S, Silver RM, Branch DW, et al. The use of cDNA microarray to identify differentially expressed labor-associated genes within the human myometrium during labor. Am J Obstet Gynecol. 2005;193:404–13.10.1016/j.ajog.2004.12.021Search in Google Scholar PubMed

[222] Havelock JC, Keller P, Muleba N, Mayhew BA, Casey BM, Rainey WE, et al. Human myometrial gene expression before and during parturition. Biol Reprod. 2005;72:707–19.10.1095/biolreprod.104.032979Search in Google Scholar PubMed

[223] O’Brien M, Morrison JJ, Smith TJ. Upregulation of PSCDBP, TLR2, TWIST1, FLJ35382, EDNRB, and RGS12 gene expression in human myometrium at labor. Reprod Sci. 2008;15:382–93.10.1177/1933719108316179Search in Google Scholar PubMed

[224] Chaemsaithong P, Madan I, Romero R, Than NG, Tarca AL, Draghici S, et al. Characterization of the myometrial transcriptome in women with an arrest of dilatation during labor. J Perinat Med. 2013;41:665–81.10.1515/jpm-2013-0086Search in Google Scholar PubMed PubMed Central

[225] Haddad R, Tromp G, Kuivaniemi H, Chaiworapongsa T, Kim YM, Mazor M, et al. Human spontaneous labor without histologic chorioamnionitis is characterized by an acute inflammation gene expression signature. Am J Obstet Gynecol. 2006;195:394.e1–24.10.1016/j.ajog.2005.08.057Search in Google Scholar PubMed PubMed Central

[226] Nhan-Chang CL, Romero R, Tarca AL, Mittal P, Kusanovic JP, Erez O, et al. Characterization of the transcriptome of chorioamniotic membranes at the site of rupture in spontaneous labor at term. Am J Obstet Gynecol. 2010;202:462–41.10.1016/j.ajog.2010.02.045Search in Google Scholar PubMed PubMed Central

[227] Bujold E, Romero R, Kusanovic JP, Erez O, Gotsch F, Chaiworapongsa T, et al. Proteomic profiling of amniotic fluid in preterm labor using two-dimensional liquid separation and mass spectrometry. J Matern Fetal Neonatal Med. 2008;21:697–713.10.1080/14767050802053289Search in Google Scholar PubMed PubMed Central

[228] Cho CK, Shan SJ, Winsor EJ, Diamandis EP. Proteomics analysis of human amniotic fluid. Mol Cell Proteomics. 2007;6:1406–15.10.1074/mcp.M700090-MCP200Search in Google Scholar PubMed

[229] Cobo T, Palacio M, Navarro-Sastre A, Ribes A, Bosch J, Filella X, et al. Predictive value of combined amniotic fluid proteomic biomarkers and interleukin-6 in preterm labor with intact membranes. Am J Obstet Gynecol. 2009;200:499–6.10.1016/j.ajog.2008.12.036Search in Google Scholar PubMed

[230] Gravett MG, Novy MJ, Rosenfeld RG, Reddy AP, Jacob T, Turner M, et al. Diagnosis of intra-amniotic infection by proteomic profiling and identification of novel biomarkers. J Am Med Assoc. 2004;292:462–9.10.1001/jama.292.4.462Search in Google Scholar PubMed

[231] Michaels JE, Dasari S, Pereira L, Reddy AP, Lapidus JA, Lu X, et al. Comprehensive proteomic analysis of the human amniotic fluid proteome: gestational age-dependent changes. J Proteome Res. 2007;6:1277–85.10.1021/pr060543tSearch in Google Scholar PubMed

[232] Park JS, Oh KJ, Norwitz ER, Han JS, Choi HJ, Seong HS, et al. Identification of proteomic biomarkers of preeclampsia in amniotic fluid using SELDI-TOF mass spectrometry. Reprod Sci. 2008;15:457–68.10.1177/1933719108316909Search in Google Scholar PubMed

[233] Park SJ, Yoon WG, Song JS, Jung HS, Kim CJ, Oh SY, et al. Proteome analysis of human amnion and amniotic fluid by two-dimensional electrophoresis and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Proteomics. 2006;6:349–63.10.1002/pmic.200500084Search in Google Scholar PubMed

[234] Romero R, Espinoza J, Rogers WT, Moser A, Nien JK, Kusanovic JP, et al. Proteomic analysis of amniotic fluid to identify women with preterm labor and intra-amniotic inflammation/infection: the use of a novel computational method to analyze mass spectrometric profiling. J Matern Fetal Neonatal Med. 2008;21:367–88.10.1080/14767050802045848Search in Google Scholar PubMed PubMed Central

[235] Ruetschi U, Rosen A, Karlsson G, Zetterberg H, Rymo L, Hagberg H, et al. Proteomic analysis using protein chips to detect biomarkers in cervical and amniotic fluid in women with intra-amniotic inflammation. J Proteome Res. 2005;4:2236–42.10.1021/pr050139eSearch in Google Scholar PubMed

[236] Vuadens F, Benay C, Crettaz D, Gallot D, Sapin V, Schneider P, et al. Identification of biologic markers of the premature rupture of fetal membranes: proteomic approach. Proteomics. 2003;3:1521–5.10.1002/pmic.200300455Search in Google Scholar PubMed

[237] Madan I, Than NG, Romero R, Chaemsaithong P, Miranda J, Tarca AL, et al. The peripheral whole-blood transcriptome of acute pyelonephritis in human pregnancya. J Perinat Med. 2014;42:31–53.10.1515/jpm-2013-0085Search in Google Scholar PubMed PubMed Central

[238] Madsen-Bouterse SA, Romero R, Tarca AL, Kusanovic JP, Espinoza J, Kim CJ, et al. The transcriptome of the fetal inflammatory response syndrome. Am J Reprod Immunol. 2010;63:73–92.10.1111/j.1600-0897.2009.00791.xSearch in Google Scholar PubMed PubMed Central

[239] Alvehus M, Buren J, Sjostrom M, Goedecke J, Olsson T. The human visceral fat depot has a unique inflammatory profile. Obesity (Silver Spring). 2010;18:879–83.10.1038/oby.2010.22Search in Google Scholar PubMed

[240] Miranda M, Escote X, Ceperuelo-Mallafre V, Alcaide MJ, Simon I, Vilarrasa N, et al. Paired subcutaneous and visceral adipose tissue aquaporin-7 expression in human obesity and type 2 diabetes: differences and similarities between depots. J Clin Endocrinol Metab. 2010;95:3470–9.10.1210/jc.2009-2655Search in Google Scholar PubMed

[241] Ibrahim MM. Subcutaneous and visceral adipose tissue: structural and functional differences. Obes Rev. 2010;11:11–8.10.1111/j.1467-789X.2009.00623.xSearch in Google Scholar PubMed

[242] varez-Llamas G, Szalowska E, de Vries MP, Weening D, Landman K, Hoek A, et al. Characterization of the human visceral adipose tissue secretome. Mol Cell Proteomics. 2007;6:589–600.10.1074/mcp.M600265-MCP200Search in Google Scholar PubMed

[243] Bashiri A, Heo HJ, Ben-Avraham D, Mazor M, Budagov T, Einstein FH, et al. Pregnancy complicated by obesity induces global transcript expression alterations in visceral and subcutaneous fat. Mol Genet Genomics. 2014;289:695–705.10.1007/s00438-014-0841-5Search in Google Scholar PubMed PubMed Central

[244] Thorsen K, Sorensen KD, Brems-Eskildsen AS, Modin C, Gaustadnes M, Hein AM, et al. Alternative splicing in colon, bladder, and prostate cancer identified by exon array analysis. Mol Cell Proteomics. 2008;7:1214–24.10.1074/mcp.M700590-MCP200Search in Google Scholar PubMed

[245] Johnson JM, Castle J, Garrett-Engele P, Kan Z, Loerch PM, Armour CD, et al. Genome-wide survey of human alternative pre-mRNA splicing with exon junction microarrays. Science. 2003;302:2141–4.10.1126/science.1090100Search in Google Scholar PubMed

[246] Kampa D, Cheng J, Kapranov P, Yamanaka M, Brubaker S, Cawley S, et al. Novel RNAs identified from an in-depth analysis of the transcriptome of human chromosomes 21 and 22. Genome Res. 2004;14:331–42.10.1101/gr.2094104Search in Google Scholar PubMed PubMed Central

[247] Moller-Levet CS, Betts GN, Harris AL, Homer JJ, West CM, Miller CJ. Exon array analysis of head and neck cancers identifies a hypoxia related splice variant of LAMA3 associated with a poor prognosis. PLoS Comput Biol. 2009;5:e1000571.10.1371/journal.pcbi.1000571Search in Google Scholar PubMed PubMed Central

[248] Stamm S, Ben-Ari S, Rafalska I, Tang Y, Zhang Z, Toiber D, et al. Function of alternative splicing. Gene. 2005;344:1–20.10.1016/j.gene.2004.10.022Search in Google Scholar PubMed

[249] Lareau LF, Inada M, Green RE, Wengrod JC, Brenner SE. Unproductive splicing of SR genes associated with highly conserved and ultraconserved DNA elements. Nature. 2007;446:926–9.10.1038/nature05676Search in Google Scholar PubMed

[250] Ni JZ, Grate L, Donohue JP, Preston C, Nobida N, O’Brien G, et al. Ultraconserved elements are associated with homeostatic control of splicing regulators by alternative splicing and nonsense-mediated decay. Genes Dev. 2007;21:708–18.10.1101/gad.1525507Search in Google Scholar PubMed PubMed Central

[251] Venables JP. Unbalanced alternative splicing and its significance in cancer. Bioessays. 2006;28:378–86.10.1002/bies.20390Search in Google Scholar PubMed

[252] Harris JM, Meyer DJ, Coles B, Ketterer B. A novel glutathione transferase [13] isolated from the matrix of rat liver mitochondria having structural similarity to class theta enzymes. Biochem J. 1991;278(Pt 1):137–41.10.1042/bj2780137Search in Google Scholar PubMed PubMed Central

[253] Morel F, Rauch C, Petit E, Piton A, Theret N, Coles B, et al. Gene and protein characterization of the human glutathione S-transferase kappa and evidence for a peroxisomal localization. J Biol Chem. 2004;279:16246–53.10.1074/jbc.M313357200Search in Google Scholar PubMed

[254] Thomson RE, Bigley AL, Foster JR, Jowsey IR, Elcombe CR, Orton TC, et al. Tissue-specific expression and subcellular distribution of murine glutathione S-transferase class kappa. J Histochem Cytochem. 2004;52:653–62.10.1177/002215540405200509Search in Google Scholar PubMed

[255] Jowsey IR, Thomson RE, Orton TC, Elcombe CR, Hayes JD. Biochemical and genetic characterization of a murine class Kappa glutathione S-transferase. Biochem J. 2003;373(Pt 2):559–69.10.1042/bj20030415Search in Google Scholar

[256] Shield AJ, Murray TP, Cappello JY, Coggan M, Board PG. Polymorphisms in the human glutathione transferase Kappa (GSTK1) promoter alter gene expression. Genomics. 2010;95:299–305.10.1016/j.ygeno.2010.02.007Search in Google Scholar PubMed

[257] Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose-specific gene dysregulated in obesity. J Biol Chem. 1996;271:10697–703.10.1074/jbc.271.18.10697Search in Google Scholar PubMed

[258] Maeda K, Okubo K, Shimomura I, Funahashi T, Matsuzawa Y, Matsubara K. cDNA cloning and expression of a novel adipose specific collagen-like factor, apM1 (AdiPose Most abundant Gene transcript 1). Biochem Biophys Res Commun. 1996;221:286–9.10.1006/bbrc.1996.0587Search in Google Scholar PubMed

[259] Nakano Y, Tobe T, Choi-Miura NH, Mazda T, Tomita M. Isolation and characterization of GBP28, a novel gelatin-binding protein purified from human plasma. J Biochem (Tokyo). 1996;120:803–12.10.1093/oxfordjournals.jbchem.a021483Search in Google Scholar PubMed

[260] Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem. 1995;270:26746–9.10.1074/jbc.270.45.26746Search in Google Scholar PubMed

[261] Yokota T, Oritani K, Takahashi I, Ishikawa J, Matsuyama A, Ouchi N, et al. Adiponectin, a new member of the family of soluble defense collagens, negatively regulates the growth of myelomonocytic progenitors and the functions of macrophages. Blood. 2000;96:1723–32.10.1182/blood.V96.5.1723Search in Google Scholar

[262] Berg AH, Combs TP, Du X, Brownlee M, Scherer PE. The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nat Med. 2001;7:947–53.10.1038/90992Search in Google Scholar PubMed

[263] Okamoto Y, Arita Y, Nishida M, Muraguchi M, Ouchi N, Takahashi M, et al. An adipocyte-derived plasma protein, adiponectin, adheres to injured vascular walls. Horm Metab Res. 2000;32:47–50.10.1055/s-2007-978586Search in Google Scholar PubMed

[264] Iwashima Y, Katsuya T, Ishikawa K, Ouchi N, Ohishi M, Sugimoto K, et al. Hypoadiponectinemia is an independent risk factor for hypertension. Hypertension. 2004;43:1318–23.10.1161/01.HYP.0000129281.03801.4bSearch in Google Scholar PubMed

[265] Matsubara M, Maruoka S, Katayose S. Decreased plasma adiponectin concentrations in women with dyslipidemia. J Clin Endocrinol Metab. 2002;87:2764–9.10.1210/jcem.87.6.8550Search in Google Scholar PubMed

[266] Ouchi N, Kobayashi H, Kihara S, Kumada M, Sato K, Inoue T, et al. Adiponectin stimulates angiogenesis by promoting cross-talk between AMP-activated protein kinase and Akt signaling in endothelial cells. J Biol Chem. 2004;279:1304–9.10.1074/jbc.M310389200Search in Google Scholar PubMed PubMed Central

[267] McLachlan KA, O’Neal D, Jenkins A, Alford FP. Do adiponectin, TNFalpha, leptin and CRP relate to insulin resistance in pregnancy? Studies in women with and without gestational diabetes, during and after pregnancy. Diabetes Metab Res Rev. 2006;22:131–8.10.1002/dmrr.591Search in Google Scholar PubMed

[268] Ategbo JM, Grissa O, Yessoufou A, Hichami A, Dramane KL, Moutairou K, et al. Modulation of adipokines and cytokines in gestational diabetes and macrosomia. J Clin Endocrinol Metab. 2006;91:4137–43.10.1210/jc.2006-0980Search in Google Scholar PubMed

[269] Thyfault JP, Hedberg EM, Anchan RM, Thorne OP, Isler CM, Newton ER, et al. Gestational diabetes is associated with depressed adiponectin levels. J Soc Gynecol Investig. 2005;12:41–5.10.1016/j.jsgi.2004.07.003Search in Google Scholar PubMed

[270] Pajvani UB, Hawkins M, Combs TP, Rajala MW, Doebber T, Berger JP, et al. Complex distribution, not absolute amount of adiponectin, correlates with thiazolidinedione-mediated improvement in insulin sensitivity. J Biol Chem. 2004;279:12152–62.10.1074/jbc.M311113200Search in Google Scholar PubMed

[271] Waki H, Yamauchi T, Kamon J, Ito Y, Uchida S, Kita S, et al. Impaired multimerization of human adiponectin mutants associated with diabetes. Molecular structure and multimer formation of adiponectin. J Biol Chem. 2003;278:40352–63.10.1074/jbc.M300365200Search in Google Scholar PubMed

[272] Tsao TS, Tomas E, Murrey HE, Hug C, Lee DH, Ruderman NB, et al. Role of disulfide bonds in Acrp30/adiponectin structure and signaling specificity. Different oligomers activate different signal transduction pathways. J Biol Chem. 2003;278:50810–7.10.1074/jbc.M309469200Search in Google Scholar PubMed

[273] Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, et al. Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature. 2003;423:762–9.10.1038/nature01705Search in Google Scholar PubMed

[274] Araki S, Dobashi K, Kubo K, Asayama K, Shirahata A. High molecular weight, rather than total, adiponectin levels better reflect metabolic abnormalities associated with childhood obesity. J Clin Endocrinol Metab. 2006;91:5113–6.10.1210/jc.2006-1051Search in Google Scholar PubMed

[275] Kobayashi H, Ouchi N, Kihara S, Walsh K, Kumada M, Abe Y, et al. Selective suppression of endothelial cell apoptosis by the high molecular weight form of adiponectin. Circ Res. 2004;94:e27–e31.10.1161/01.RES.0000119921.86460.37Search in Google Scholar PubMed PubMed Central

[276] Aroda V, Ciaraldi TP, Chang SA, Dahan MH, Chang RJ, Henry RR. Circulating and cellular adiponectin in polycystic ovary syndrome: relationship to glucose tolerance and insulin action. Fertil Steril. 2008;89:1200–8.10.1016/j.fertnstert.2007.04.046Search in Google Scholar PubMed PubMed Central

[277] Modan-Moses D, Stein D, Pariente C, Yaroslavsky A, Ram A, Faigin M, et al. Modulation of adiponectin and leptin during refeeding of female anorexia nervosa patients. J Clin Endocrinol Metab. 2007;92:1843–7.10.1210/jc.2006-1683Search in Google Scholar PubMed

[278] Ong GK, Hamilton JK, Sermer M, Connelly PW, Maguire G, Zinman B, et al. Maternal serum adiponectin and infant birthweight: the role of adiponectin isoform distribution. Clin Endocrinol (Oxf). 2007;67:108–14.10.1111/j.1365-2265.2007.02846.xSearch in Google Scholar PubMed

[279] Retnakaran R, Hanley AJ, Connelly PW, Maguire G, Sermer M, Zinman B. Low serum levels of high-molecular weight adiponectin in Indo-Asian women during pregnancy: evidence of ethnic variation in adiponectin isoform distribution. Diabetes Care. 2006;29:1377–9.10.2337/dc06-0413Search in Google Scholar PubMed

[280] Takemura Y, Osuga Y, Koga K, Tajima T, Hirota Y, Hirata T, et al. Selective increase in high molecular weight adiponectin concentration in serum of women with preeclampsia. J Reprod Immunol. 2007;73:60–5.10.1016/j.jri.2006.05.004Search in Google Scholar PubMed

[281] Retnakaran R, Connelly PW, Maguire G, Sermer M, Zinman B, Hanley AJ. Decreased high-molecular-weight adiponectin in gestational diabetes: implications for the pathophysiology of Type 2 diabetes. Diabet Med. 2007;24:245–52.10.1111/j.1464-5491.2007.02077.xSearch in Google Scholar PubMed

[282] Qian Y, Zheng Y, Taylor R, Tiffany-Castiglioni E. Involvement of the molecular chaperone Hspa5 in copper homeostasis in astrocytes. Brain Res. 2012;1447:9–19.10.1016/j.brainres.2012.01.065Search in Google Scholar PubMed

[283] Qian Y, Meng B, Zhang X, Zheng Y, Taylor R, Tiffany-Castiglioni E. HSPA5 Forms Specific Complexes with Copper. Neurochem Res. 2013;38:321–9.10.1007/s11064-012-0923-xSearch in Google Scholar PubMed

[284] Uckun FM, Qazi S, Ozer Z, Garner AL, Pitt J, Ma H, et al. Inducing apoptosis in chemotherapy-resistant B-lineage acute lymphoblastic leukaemia cells by targeting HSPA5, a master regulator of the anti-apoptotic unfolded protein response signalling network. Br J Haematol. 2011;153:741–52.10.1111/j.1365-2141.2011.08671.xSearch in Google Scholar PubMed

[285] Wisniewska M, Karlberg T, Lehtio L, Johansson I, Kotenyova T, Moche M, et al. Crystal structures of the ATPase domains of four human Hsp70 isoforms: HSPA1L/Hsp70-hom, HSPA2/Hsp70-2, HSPA6/Hsp70B’, and HSPA5/BiP/GRP78. PLoS One. 2010;5:e8625.10.1371/journal.pone.0008625Search in Google Scholar PubMed PubMed Central

[286] Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell. 2010;140:900–17.10.1016/j.cell.2010.02.034Search in Google Scholar PubMed PubMed Central

[287] Todd DJ, Lee AH, Glimcher LH. The endoplasmic reticulum stress response in immunity and autoimmunity. Nat Rev Immunol. 2008;8:663–74.10.1038/nri2359Search in Google Scholar PubMed

[288] Pfaffenbach KT, Lee AS. The critical role of GRP78 in physiologic and pathologic stress. Curr Opin Cell Biol. 2011;23:150–6.10.1016/j.ceb.2010.09.007Search in Google Scholar PubMed PubMed Central

[289] Sharma NK, Das SK, Mondal AK, Hackney OG, Chu WS, Kern PA, et al. Endoplasmic reticulum stress markers are associated with obesity in nondiabetic subjects. J Clin Endocrinol Metab. 2008;93:4532–41.10.1210/jc.2008-1001Search in Google Scholar PubMed PubMed Central

[290] Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science. 2004;306:457–61.10.1126/science.1103160Search in Google Scholar PubMed

[291] Nakatani Y, Kaneto H, Kawamori D, Yoshiuchi K, Hatazaki M, Matsuoka TA, et al. Involvement of endoplasmic reticulum stress in insulin resistance and diabetes. J Biol Chem. 2005;280:847–51.10.1074/jbc.M411860200Search in Google Scholar PubMed

[292] Das SK, Chu WS, Mondal AK, Sharma NK, Kern PA, Rasouli N, et al. Effect of pioglitazone treatment on endoplasmic reticulum stress response in human adipose and in palmitate-induced stress in human liver and adipose cell lines. Am J Physiol Endocrinol Metab. 2008;295:E393–E400.10.1152/ajpendo.90355.2008Search in Google Scholar PubMed PubMed Central

[293] Mondal AK, Das SK, Varma V, Nolen GT, McGehee RE, Elbein SC, et al. Effect of endoplasmic reticulum stress on inflammation and adiponectin regulation in human adipocytes. Metab Syndr Relat Disord. 2012;10:297–306.10.1089/met.2012.0002Search in Google Scholar PubMed PubMed Central

[294] Li A, Ponten F, dos Remedios CG. The interactome of LIM domain proteins: the contributions of LIM domain proteins to heart failure and heart development. Proteomics. 2012;12:203–25.10.1002/pmic.201100492Search in Google Scholar PubMed

[295] Kovalevich J, Tracy B, Langford D. PINCH: more than just an adaptor protein in cellular response. J Cell Physiol. 2011;226:940–7.10.1002/jcp.22437Search in Google Scholar PubMed PubMed Central

[296] Wu C. PINCH, N(i)ck and the ILK: network wiring at cell-matrix adhesions. Trends Cell Biol. 2005;15:460–6.10.1016/j.tcb.2005.07.002Search in Google Scholar PubMed

[297] Chen H, Huang XN, Yan W, Chen K, Guo L, Tummalapali L, et al. Role of the integrin-linked kinase/PINCH1/alpha-parvin complex in cardiac myocyte hypertrophy. Lab Invest. 2005;85:1342–56.10.1038/labinvest.3700345Search in Google Scholar PubMed

[298] Hannigan GE, Coles JG, Dedhar S. Integrin-linked kinase at the heart of cardiac contractility, repair, and disease. Circ Res. 2007;100:1408–14.10.1161/01.RES.0000265233.40455.62Search in Google Scholar PubMed

[299] Liang X, Zhou Q, Li X, Sun Y, Lu M, Dalton N, et al. PINCH1 plays an essential role in early murine embryonic development but is dispensable in ventricular cardiomyocytes. Mol Cell Biol. 2005;25:3056–62.10.1128/MCB.25.8.3056-3062.2005Search in Google Scholar PubMed PubMed Central

[300] Liang X, Sun Y, Schneider J, Ding JH, Cheng H, Ye M, et al. Pinch1 is required for normal development of cranial and cardiac neural crest-derived structures. Circ Res. 2007;100:527–35.10.1161/01.RES.0000259041.37059.8cSearch in Google Scholar PubMed PubMed Central

[301] Liang X, Sun Y, Ye M, Scimia MC, Cheng H, Martin J, et al. Targeted ablation of PINCH1 and PINCH2 from murine myocardium results in dilated cardiomyopathy and early postnatal lethality. Circulation. 2009;120:568–76.10.1161/CIRCULATIONAHA.109.864686Search in Google Scholar PubMed PubMed Central

[302] Liang X, Sun Y, Chen J. Particularly interesting cysteine- and histidine-rich protein in cardiac development and remodeling. J Investig Med. 2009;57:842–8.10.2310/JIM.0b013e3181c5e31dSearch in Google Scholar PubMed PubMed Central

[303] Jung KY, Chen K, Kretzler M, Wu C. TGF-beta1 regulates the PINCH-1-integrin-linked kinase-alpha-parvin complex in glomerular cells. J Am Soc Nephrol. 2007;18:66–73.10.1681/ASN.2006050421Search in Google Scholar PubMed

[304] Shi X, Qu H, Kretzler M, Wu C. Roles of PINCH-2 in regulation of glomerular cell shape change and fibronectin matrix deposition. Am J Physiol Renal Physiol. 2008;295:F253–63.10.1152/ajprenal.00070.2008Search in Google Scholar PubMed PubMed Central

[305] Yang Y, Guo L, Blattner SM, Mundel P, Kretzler M, Wu C. Formation and phosphorylation of the PINCH-1-integrin linked kinase-alpha-parvin complex are important for regulation of renal glomerular podocyte adhesion, architecture, and survival. J Am Soc Nephrol. 2005;16:1966–76.10.1681/ASN.2004121112Search in Google Scholar PubMed

[306] Gkretsi V, Mars WM, Bowen WC, Barua L, Yang Y, Guo L, et al. Loss of integrin linked kinase from mouse hepatocytes in vitro and in vivo results in apoptosis and hepatitis. Hepatology. 2007;45:1025–34.10.1002/hep.21540Search in Google Scholar PubMed

[307] Tu Y, Li F, Goicoechea S, Wu C. The LIM-only protein PINCH directly interacts with integrin-linked kinase and is recruited to integrin-rich sites in spreading cells. Mol Cell Biol. 1999;19:2425–34.10.1128/MCB.19.3.2425Search in Google Scholar PubMed PubMed Central

[308] Wang-Rodriguez J, Dreilinger AD, Alsharabi GM, Rearden A. The signaling adapter protein PINCH is up-regulated in the stroma of common cancers, notably at invasive edges. Cancer. 2002;95:1387–95.10.1002/cncr.10878Search in Google Scholar PubMed

[309] Zhang JT, Li QX, Wang D, Zhu ZL, Yang YH, Cui DS, et al. Up-regulation of PINCH in the stroma of oral squamous cell carcinoma predicts nodal metastasis. Oncol Rep.;14:1519–22.10.3892/or.14.6.1519Search in Google Scholar

[310] Zhu Z, Yang Y, Zhang Y, Wang Z, Cui D, Zhang J, et al. PINCH expression and its significance in esophageal squamous cell carcinoma. Dis Markers. 2008;25:75–80.10.1155/2008/473860Search in Google Scholar PubMed PubMed Central

[311] Kim SK, Jang HR, Kim JH, Noh SM, Song KS, Kim MR, et al. The epigenetic silencing of LIMS2 in gastric cancer and its inhibitory effect on cell migration. Biochem Biophys Res Commun. 2006;349:1032–40.10.1016/j.bbrc.2006.08.128Search in Google Scholar PubMed

[312] Wang MW, Gu P, Zhang ZY, Zhu ZL, Li YM, Zhao HX, et al. Expression of PINCH protein in gliomas and its clinicopathological significance. Oncology. 2007;72:343–6.10.1159/000113064Search in Google Scholar PubMed

[313] Hehlgans S, Haase M, Cordes N. Signalling via integrins: implications for cell survival and anticancer strategies. Biochim Biophys Acta. 2007;1775:163–80.10.1016/j.bbcan.2006.09.001Search in Google Scholar PubMed

[314] Chen K, Tu Y, Zhang Y, Blair HC, Zhang L, Wu C. PINCH-1 regulates the ERK-Bim pathway and contributes to apoptosis resistance in cancer cells. J Biol Chem 2008;283:2508–17.10.1074/jbc.M707307200Search in Google Scholar PubMed


Supplemental Material:

The online version of this article (DOI: 10.1515/jpm-2015-0259) offers supplementary material, available to authorized users.


  1. The authors stated that there are no conflicts of interest regarding the publication of this article.

Received: 2015-7-27
Accepted: 2015-10-26
Published Online: 2016-3-19
Published in Print: 2016-10-1

©2016 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 19.4.2024 from https://www.degruyter.com/document/doi/10.1515/jpm-2015-0259/html
Scroll to top button