Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Mar 1.
Published in final edited form as: Placenta. 2020 Jan 22;92:17–27. doi: 10.1016/j.placenta.2020.01.008

A Review and Meta-analysis of Omics Approaches to Study Preeclampsia

Paula A Benny 1, Fadhl M Al-Akwaa 3, Ryan J Schlueter 2, Cameron B Lassiter 1, Lana X Garmire 3,#
PMCID: PMC7306500  NIHMSID: NIHMS1557368  PMID: 32056783

Abstract

Preeclampsia is a medical condition affecting 5–10% of pregnancies. It has serious effects on the health of the pregnant mother and developing fetus. While possible causes of preeclampsia are speculated, there is no consensus on its etiology. The advancement of big data and high-throughput technologies enables to study preeclampsia at the new and systematic level. In this review, we first highlight the recent progress made in the field of preeclampsia research using various omics technology platforms, including epigenetics, genome-wide association studies (GWAS), transcriptomics, proteomics and metabolomics. Next, we integrate the results in individual omic level and studies, and show that despite the lack of coherent biomarkers in all omics studies, inhibin is a potential preeclamptic biomarker supported by GWAS, transcriptomics and DNA methylation evidence. Using the network analysis on the biomarkers of all literatures reviewed here, we identify four striking sub-networks with clear biological functions supported by previous molecular-biology and clinical observations. In summary, omics integration approach offers the promise to understand molecular mechanisms in preeclampsia.

Introduction

Preeclampsia is a medical condition that affects 5 – 10% of pregnant mothers and accounts for 40% of fetal deaths worldwide [1]. It manifests with symptoms of hypertension (>140/90 mm Hg), proteinuria (>0.3g in urine) and swelling [2]. Primarily diagnosed by new onset and rapidly developing hypertension in pregnancy, preeclampsia is often found late in the third trimester, after 34 weeks, and termed late-onset preeclampsia. Early onset preeclampsia is typically diagnosed prior to 34 weeks. Several clinical symptoms such as headache, vision changes, epigastric pain and shortness of breath can accompany more severe forms of the disease [3]. Laboratory abnormalities such as increased liver enzymes, thrombocytopenia, and proteinuria with kidney damage are also common in progressive preeclampsia [4]. Clinically, the most important complications of preeclampsia to prevent are maternal seizure and cerebrovascular accident [5]. The cornerstone of therapy in preeclampsia is stabilization of the mother and delivery of the fetus as the ultimate disease treatment. Because preeclampsia is such a common and pervasive disorder, understanding how it develops and if we can predict or treat it has become the focus of numerous studies.

Though several theories exist on the cause of preeclampsia, there is no consensus on its etiology and neither is there known way to prevent it. Since this disease has complex pathologic roots in placental morphology, maternal predisposition, genetic variance and molecular signalling, combating the scientific complexity of preeclampsia requires new and unique methodologies. In recent years, omics approaches have been utilized for such purposes. Omics refers to the field in biological science that aims at the collective characterization and quantification of pools of biological molecules that translate into the structure, function, and dynamics of organisms. Taking advantage of methods such as next generation sequencing, high-resolution mass spectroscopy and chip arrays, omics platforms generate immense volumes of data in a condensed period. By comparing preeclampsia samples with control samples, one can assess the molecular differences associated with the disease state. These significant molecular differences, in the forms of mRNA, SNPs, DNA methylation sites, proteins, metabolites etc, can potentially serve as biomarkers preeclampsia. Moreover, these abnormal molecular changes can help piece together the pathogenesis determinants for preeclampsia. By targeting and reverse these molecular determinants, it is hopeful to control, manage, and ultimate treat preeclampsia.

In this review, we systematically summarize the findings from various omics research approaches to study preeclampsia through a comprehensive PubMed search. The keywords “preeclampsia” and “epigenetics”, “transcriptomics”, “GWAS”, “proteomics” and “metabolomics” were entered into the PubMed search field and all 515 results returned were analyzed for relevant validated studies between the years 2000–2019. After screening for studies not reported in English and removing duplicates, this brought the total number of studies included to 58. We enumerated them by various platforms including genetics, epigenomics, transcriptomics, proteomics and metabolomics. In order to facilitate our understanding of this multi-faceted disorder, we enlist preeclampsia-associated molecules in individual studies, as well as lacking of common ones among different omic platforms and different studies. While this observation is due to many complex issues surrounding the disease, the samples and the analysis processes, we nevertheless show that by synthesizing the knowledge gained from different omics levels, it is possible to gain systematic insights into preeclampsia. We call on the urgent need for systematic multi-omics design with clinical information documentation, which help better understand the biological mechanism associated with this heterogenous syndrome, with the ultimate goal to narrow the gap towards therapeutics interventions.

Genetics studies of Preeclampsia

Genome wide association studies (GWAS) are typically methods used to analyse effects of single-nucleotide polymorphisms (SNPs) on disease phenotypes. There is evidence that genetic factors predispose certain populations to preeclampisa. Over the years, genetics studies have evolved from the original limited capacity of analysing a handful of SNPs to high-throughput platforms, including the Illumina SNP arrays, and more recently whole exome sequencing and whole genome sequencing platforms. Put in the perspective of clinical applications, SNPs have recently been used to identify pregnancies at high risk of miscarriage [6], similarly preeclampsia-associated SNPs may have the potential to be used as a genetic diagnostic test.

We herein compiled the results from a Pubmed literature search of all the hits resulting from the search words “GWAS” and “preeclampsia”. Studies which did not perform validation experiments on their identified targets were excluded. The resulting studies are summarized in Table 1. To date, the largest preeclampsia GWAS study consisted of offspring of 4,380 cases and 310,238 controls. The authors discovered that it was the SNPs in sFlt-1 in the fetal genome that were associated with preeclampsia risk[7]. In the maternal genome, the largest preeclampsia GWAS study was the HAPO study (Hyperglycaemia and Adverse Pregnancy Outcome) where 1070 Afro-Caribbean (n=21 cases and 1049 controls), 723 Hispanic (n=62 cases and 661 controls) and 1257 European (n=50 cases and 1207 controls) mothers were enrolled in [8]. However, in this study no individual SNPs were significantly associated with preeclampsia after Bonferroni correction, despite over one million SNPs assayed. This is could be due to the overly stringent multiple hypothesis testing [8]. Or, it is likely that multiple alleles are interacting in a multifactorial disease such as preeclampsia. A polygenic risk score might be more informative in future studies.

Table 1.

List of SNPs extracted from several preeclampsia GWAS reports. Gene of interest, sample origin (mother or child), study size, gestational age and severity of preeclampsia were included.

SNPs Gene Reference Sample Origin (mother, child) Study size (Case/control) Gestational Age (preeclampsia, control in weeks) *
rs17367504a MTHFR [13] Mother 1006/816 ^
rs7579169 INHIBIN [12] Mother 538/540 ^
rs17249754a ATP2B1 [13] Mother 1006/816 ^
rs16933812a PAX5 [13] Mother 1006/816 ^
rs12946454a PLCD3 [13] Mother 1006/816 ^
rs1426409 KIAA1239 [70] Mother 177/116 ^
rs17686866 ESRRG [70] Mother 177/116 ^
rs9831647 LMCD1 [70] Mother 177/116 ^
rs10743565 IFLTD1 [70] Mother 177/116 ^
rs11617740 FGF14 [8] Mother 137/2,986 ^
rs2839440 C21orf121 [8] Mother 137/2,986 ^
rs12641856 MGC45800 [8] Mother 137/2,986 ^
rs4815879 MCM8 [8] Mother 137/2,986 ^
rs28360974 MUC21 [8] Mother 137/2,986 ^
rs1248993 BAMBI [8] Mother 137/2,986 ^
rs975369 NPVF [8] Mother 137/2,986 ^
rs1556832 ADRA1D [8] Mother 137/2,986 ^
rs11600901 SCN2B [8] Mother 137/2,986 ^
rs7322722 MYCBP2 [8] Mother 137/2,986 ^
rs10989019 INVS [8] Mother 137/2,986 ^
rs4769613 FLT1 [7] Child 4,380/310,238 <34,37##
>34,39#
rs9478812 PLEKHG1 [3] Mother 877/2004 ###
##

early-onset preeclampsia

#

late-onset preeclampsia

###

severe preeclampsia

####

mild preeclampsia

^

not specified

*

Type of preeclampsia

Table 1 summarizes a list of SNPs, obtained through GWAS, which were claimed to be associated with preeclampsia. Gene set enrichment analysis on the genes in Table I which the risk SNPs reside in reveals that calcium signalling pathway is involved (P<0.05) with gene members APT2B, ADRA1D and PLCD3. For example, rs7579169 (T>C), located in the intergenic region near the inhibin beta B gene (INHBB) in an Australian cohort, was validated and extended in another large Asian cohort. Inhibin is a glycoprotein which lowers follicle-stimulating hormone (FSH) levels. In preeclamptic pregnancies, inhibin protein levels are magnitudes higher compared to normal pregnancies, though the reason for this observation remains unclear [9]. It is possible that these SNPs impact on promoter activity, gene expression, amino acid sequence or protein stability. Other preeclampsia associated SNPs, such as, rs12711941 (T>G), rs7576192 (A>G) and rs2681472 were also identified near the inhibin gene [1012]. Interestingly, in a Norwegian population, rs17367504 in the methylenetetrahydrofolate reductase (MTHFR) gene was associated with protective effects against preeclampsia [13]. Given the different SNPs reported by various GWAS studies, it is of great interest to investigate if the preeclampsia-associated SNPs in each individual study can indeed be replicated, by meta-analysis of all studies that make their data public.

Epigenetics studies of preeclampsia

Epigenetics studies the inheritable changes in gene expression that do not involve any changes in the DNA sequence [14]. The most widely studied epigenetic modification is DNA methylation [15, 16]. Essentially, epigenetics causes a change in phenotype without altering genotype and is influenced by several factors including environment, age, lifestyle and genetics [17]. As DNA methylation influences gene expression and subsequent protein translation, it has the potential to be a major contributor to a disease and a mediator of environmental influences. For clinical applications, various studies have been conducted, in the hope to identify DNA methylation signals for diagnosis or treatment of preeclampsia.

We therein performed a PubMed literature search for the words “epigenetics” and “preeclampsia” and only included studies which validated their hypomethylation and hypermethylation sites. Table 2 summarizes the hypermethylated and hypomethylated regions of genes and noncoding RNAs associated with preeclampsia in epigenetic studies [15, 16, 1824]. Given the large number of genes involved in DNA methylation changes, it is valuable to investigate if these genes are enriched in certain biological functions. Thus we conducted pathway analysis on this list of genes using ConsensusPathDB (CPDB)[25]. The pathway analysis reveals an enrichment of proteasome pathways, hippo signalling pathways, cancer pathways and hypoxia signalling pathways affected in preeclampsia. Among these genes, hypermethylation of Wnt2 was observed in two studies [15, 26], and was proposed to contribute to trophoblast and signalling dysfunction in the preeclamptic placenta [27, 28]. However, precise mechanistic elucidation of Wnt2 signalling in preeclampsia are lacking [26]. Differentially methylated loci identified by omic strategies should be mapped to genes for further validation, in order to avoid false positive results from these studies.

Table 2.

List of hyper- and hypo-methylated regions associated with preeclampsia. Gene of interest, region, methylation status, characteristics, coordinates, study size, gestation age were included.

Gene/Region Methylation status Reference Sample origin Analyse type Characteristic Coordinates/Accension Study size Gestational Age (preeclampsia, control in weeks) *
WNT2 Hyper [15]
[19]
 Fetal
 placenta
 Region TSS  chr7:11696
 3492–116964841
8/16 35,37 ^
SPESP1 Hyper [15]  Fetal
 placenta
 Region TSS, 5′-UTR, body  chr15:69202
 2400–69223895
8/16 35,37 ^
NOX5 Hyper [15]  Fetal
 placenta
 Region TSS, 5′-UTR, body  chr15:6922
 2400–69223895
8/16 35,37 ^
 MIR548 H4 Hyper [15]  Fetal
 placenta
 Region TSS, 5′-UTR, body  chr15:6922
 2400–69223895
8/16 35,37 ^
ALCAM Hyper [15]  Fetal
 placenta
 Region Body  chr3:10508
 7718–105088546
8/16 35,37 ^
CDH11 Hyper [16] Fetal
placenta
Site cg26624576 31/14 35,37 ^
COL5A1 Hyper [16] Fetal
placenta
Site cg14237069 31/14 35,37 ^
NCAM1 Hypo [16] Fetal
placenta
Site cg20857767 31/14 35,37 ^
INHIBIN Hypo [18] Fetal
placenta
Site 5’UTR cg11079619 20/20 31,37 ##
 BHLHE40 Hypo [18] Fetal
placenta
Site Body cg20971407 20/20 31,37 ##
SLC2A1 Hypo [18] Fetal
placenta
Site Body cg01924561 20/20 31,37 ##
ADAM12 Hypo [18] Fetal
placenta
Site Body cg02494582 20/20 31,37 ##
 mir-519a1 Hyper [20]  Chorioamniotic membrane Promoter 30/17 32,38##
mir-301a Hyper [20]  Chorioamniotic membrane Promoter 30/17 32,38##
mir-487a Hyper [20]  Chorioamniotic membrane Promoter 30/17 32,38 ##
mir-185 Hyper [20]  Chorioamniotic membrane Promoter 30/17 32,38 ##
mir-329 Hyper [20]  Chorioamniotic membrane Promoter 30/17 32,38 ##
mir-194 Hyper [20] Chorioamniotic membrane Promoter 30/17 32,38 ##
 mir-376a1 Hyper [20]  Chorioamniotic membrane Promoter 30/17 32,38 ##
mir-486 Hyper [20]  Chorioamniotic membrane Promoter 30/17 32,38 ##
mir-744 Hyper [20]  Chorioamniotic membrane Promoter 30/17 32,38 ##
GRB2 Hypo [20]  Chorioamniotic membrane Promoter 30/17 32,38 ##
ATF3 Hypo [20]  Chorioamniotic membrane Promoter 30/17 32,38 ##
NFKB2 Hypo [20]  Chorioamniotic membrane  Region Promoter  NM_00107 7493 30/17 32,38 ##
PSMA1 Hypo [20]  Chorioamniotic membrane Promoter 30/17
PMSE1 Hypo [20]  Chorioamniotic membrane Promoter 30/17 32,38 ##
PSMD1 Hypo [20]  Chorioamniotic membrane Promoter 30/17 32,38 ##
PSMD8 Hypo [20]  Chorioamniotic membrane  Region  Promoter, Body  NM_00281 2 30/17 32,38 ##
PAPPA2 Hypo [21] Maternal
Placenta
Site Not Reported cg10994126 9/24 35,37 ##
CRH Hyper [22] Placenta 19/19 31,37##
37,39#
 CYP11A 1 Hypo [22] Placenta 19/19 31,37##
37,39#
3β-HSD type 1 Hypo [22] Placenta 19/19 31,37##
37,39#
TEAD3 Hypo [22] Placenta 19/19 31,37##
37,39#
CYP19 Hypo [22] Placenta 19/19 31,37##
37,39#
CAPN2 hypo [23] Placenta Region Promoter 824 9/9 35,37 ^
EPHX2 hypo [23] Placenta Region Promoter 2053 9/9 35,37 ^
ADORA2B hyper [23] Placenta Region Promoter 136 9/9 35,37 ^
SOX7 hyper [23] Placenta Region Promoter 83595 9/9 35,37 ^
CXCL1 hyper [23] Placenta Region Promoter 2919 9/9 35,37 ^
CDX1 hyper [23] Placenta Region Promoter 1044 9/9 35,37 ^
GLI2 hypo [24] Fetal
placenta
Region Promoter GLI2_E90_Fa 4/5 30,37##
37,39#
KRT13 hypo [24] Fetal
placenta
Region Promoter KRT13_P676_Fa 4/5 30,37##
37,39#
MEST hypo [24] Fetal
placenta
Region Promoter MEST_E150_Fa 4/5 30,37##
37,39#
TIMP3 hypo [24] Fetal
placenta
Region Promoter TIMP3_P690_Ra 4/5 30,37##
37,39#
CAPG hypo [24] Fetal
placenta
Region Promoter CAPG_E228_Fa 4/5 30,37##
37,39#
SORD [90] Placenta 7,4 ^
DGKI [90] Placenta 7,4 ^
ICA1 [90] Placenta 7,4 ^
CMIP [91] Placenta 32,30 33,37 ##
##

early-onset preeclampsia

#

late-onset preeclampsia

###

severe preeclampsia

####

mild preeclampsia

^

not specified

*

Type of preeclampsia

Besides protein coding genes, differential methylation of several microRNA (miRNA)-associated promoter regions have been reported in preeclampsia, such as miR548, miR519, miR301, miR487, miR 185, miR 329, miR194, miR376, miR486 and miR744 [14, 53]. Pathway analysis reveals miR744 and miR185 as strongly correlated with promoter activation in cancers through β-catenin, c-Jun and MMP pathways. Due to the potential of miRNAs to alter the expression of target genes, these miRNA methylation alterations are particularly interesting for further validation by larger studies.

Transcriptomics studies of preeclampsia

Gene expression profiling has provided some mechanistic insight into preeclampsia, moreover, it could provide promising “biomarkers” for this disease. Recent studies have had success identifying preeclampsia-associated transcripts in circulation, which could function as potential clinical diagnostic markers [29, 30]. We performed a PubMed literature search using the keywords “transcriptomics” and “preeclampsia” and included the studies which validated their resultant targets.

Table 3 provides a list of differentially expressed genes associated with preeclampsia. Among them, Flt-1 (fms-like tyrosine kinase-1) was the most frequently upregulated gene in preeclampsia. It is a circulating antagonist for placenta growth factor (PGF) and vascular endothelial growth factor (VEGF), both of which promote vascular expansion in the placenta [31]. Pathway enrichment analysis of the genes in Table 3 shows that up regulated genes are associated with adipocyte regulation, retinoic acid regulation, hypoxia signalling, and transcription factor network pathways; down regulated genes are enriched in cytokine-associated pathways.

Table 3.

List of the RNA transcripts associated with preeclampsia. Gene symbol, transcript fold change, tissue of origin, study size, gestational age and severity of preeclampsia were included.

Gene symbol Reference Fold change Tissue Study size Gestational Age (preeclampsia, control in weeks) *
PRG2 [32] 6.4 syncytiotrophoblasts severe preeclampsia (placenta) 4/4 27,37 ###
FSTL3 [32] 3.7 syncytiotrophoblasts severe preeclampsia (placenta) 4/4 27,37 ###
FABP4 [32] 3.5 syncytiotrophoblasts severe preeclampsia (placenta) 4/4 27,37 ###
RDH13 [32] 3.1 syncytiotrophoblasts severe preeclampsia (placenta) 4/4 27,37 ###
PLAC1 [32] −3.4 syncytiotrophoblasts severe preeclampsia (placenta) 4/4 27,37 ###
SERPINI1 [32] −4.4 syncytiotrophoblasts severe preeclampsia (placenta) 4/4 27,37 ###
GSTA3 [32] −4.8 syncytiotrophoblasts severe preeclampsia (placenta) 4/4 27,37 ###
CXCL8 [32] 3.5 cytotrophoblasts Severe preeclampsia (placenta) 3/4 27,37 ###
GH2 [32] −14.2 cytotrophoblasts Severe preeclampsia (placenta) 3/4 27,37 ###
miR-210 [24] 2.452 Placenta (full thickness) 16/16 30,29##
37,38#
miR-26a [37] −164.655 Placenta (full thickness) 16/16 ^
miR-376c-3p [71] down Circulating exosomes 5/4 35,35 ^
miR-19a-3p [71] down Circulating exosomes 5/4 35,35 ^
miR-19b-3p [71] down Circulating exosomes 5/4 35,35 ^
miR-885–5p [71] up Circulating exosomes 5/4 35,35 ^
sFLT1 [72] 2.6 Placenta (full thickness) 21/21 34,39 ###
ENG [73] 1.852919237 Placenta (full thickness) 23/37 33,37 ^
INHIBIN A [73]
[74] [75]
3.0 Placenta (full thickness) 21/21 33,37 ^
SIAE [73] 1.354912795 Placenta (full thickness) 23/37 33, 37 ^
PAPPA2 [73] 2.9 Placenta (full thickness) 21/21 33, 37 ^
BRSK2 [73] 8.6 Placenta (full thickness) 21/21 33, 37 ^
FLJ90650 [73] 10.0 Placenta (full thickness) 21/21 33, 37 ^
LEP [73] 40.0 Placenta (full thickness) 21/21 33, 37 ^
LHB [73] 4.1 Placenta (full thickness) 21/21 33, 37 ^
PDGFD [73] −2.3 Placenta (full thickness) 21/21 33, 37 ^
COX17 [73] −4.3 Placenta (full thickness) 21/21 33, 37 ^
HS3ST2 [74] −1.39  Maternal Placenta (Decidual basalis) 60/65 32,39 ^
TNFRSF14 [74] −1.35 Maternal Placenta (Decidual basalis) 60/65 32,39 ^
SLC2A6 [74] −1.32  Maternal Placenta (Decidual basalis) 60/65 32,39 ^
DPP7 [74] −1.22  Maternal Placenta (Decidual basalis) 60/65 32,39 ^
CD72 [74] −1.21  Maternal Placenta (Decidual basalis) 60/65 32,39 ^
PER3 [74] −1.20  Maternal Placenta (Decidual basalis) 60/65 32,39 ^
DBP [74] −1.20  Maternal Placenta (Decidual basalis) 60/65 32,39 ^
PDK4 [74] 1.72  Maternal Placenta (Decidual basalis) 60/65 32,39 ^
HS3ST2 [74] −1.39  Maternal Placenta (Decidual basalis) 60/65 32,39 ^
##

early-onset preeclampsia

#

late-onset preeclampsia

###

severe preeclampsia

####

mild preeclampsia

^

not specified

*

Type of preeclampsia

Besides protein coding genes, differential miRNA and long non-coding RNA (lncRNA) expression has been reported in preeclampsia too [3234]. For example, lncRNA UCA-1 primarily promotes cell migration and proliferation, presumably in an attempt to remedy poor trophoblast invasion and bad uterine spiral artery remodelling [32]. Other lncRNAs reported to be associated with preeclampsia include MEG3, HOTAIR, MALAT-1 and FLT1P1 [33]. The modes of action of these lncRNAs were proposed as chromatic state regulation (HOTAIR), metastatic gene expression regulation (MALAT1), tumor suppressor (MEG3) and angiogenesis regulator (FLT1P1). Among small RNAs, miRNAs have shown to be closely relevant in preeclampsia pathogenesis [34]. The most well studied preeclampsia associated miRNA is miR-210. It is associated with hypoxic pathways and is upregulated in response to hypoxia inducible factors [35]. miR-210 promotes angiogenesis by releasing IL-1α [36]. Another miRNA linked to preeclampsia is miR-26a down-regulated in preeclampsia, which regulate the TGF-β (transforming growth factor beta) cascade pathway, also relating to angiogenesis [37].

Proteomics studies of preeclampsia

Similar to transcriptomics, proteomics also varies temporally and spatially [38]. Global proteomics has evolved by mass spectrometry. In preeclampsia, proteomics may provide candidate biomarkers for early diagnosis, or give insights on disease progression. We searched the PubMed literature using keywords “proteomics” and “preeclampsia” and compiled studies which validated their targets.

Table 4 highlights the proteomics studies of preeclampsia. Studies from plasma [3941] and serum [4244] from peripheral blood have shown increases in fibronectin, matrix-metalloprotease (MMP), immunomodulatory molecules, heat shock proteins, C1S (complement C1s subcomponent), AMBP (alpha 1 microglobulin/bikunin) and a decrease in RBP4. Fibronectin, MMP and heat shock proteins are associated with cellular inflammatory and wound healing processes. Increases in C1S and AMBP contribute to spiral artery remodelling [45]. Additionally, apolipoproteins were found to be unusually more abundant in preeclamptic placentas, suggesting dysregulated lipid-protein biosynthesis and lipid transport pathways [46]. Genes with protein level changes can be categorised into six pathways. Up regulated genes are frequently observed in hypoxia signalling, lipid regulation, chemical carcinogenesis, thyroid hormone synthesis and cysteine and methionine metabolism pathways; down regulated genes are often seen in peroxisome signalling pathways.

Table 4.

List of proteins associated with preeclampsia. Proteins, fold changes, sample origin, sample size of studies, gestational age and severity of preeclampsia were also included.

Protein Fold change Reference Sample origin Sample size Gestational Age (preeclampsia, control in weeks) *
apolipoproteins A-I ↑ 2

[76] Plasma^ 25/25^ 38,39^
[77] Placenta 5/5 35,37 ^
[78] Amniotic fluid 18/16 36,36^
apolipoproteins and A-II ↑ 2 [76] plasma 25/25 38,39^
haptoglobin ↓ −1.8 [76] plasma 25/25 38,39^
hemopexin ↓ −1.8 [76] plasma 25/25 38,39^
Fibronectin ↑ 2.1 [44] serum 30/58 35,40###
39,40####
RBP4 [42] serum 10/10 36,38 ###
Hsp 27
[79] Placenta
Placenta
5/5
10/10
35,37^
Hsp 70 [79] placenta 5/5 35,37^
Laminin ↑ 0.18 [80] placenta trophoblastic cells 1/1 ^
HSPE1 ↑ 5.86 [81] placenta 4/4 36,37^
PRDX3 ↑ 5.01 [81] placenta 4/4 36,37^
TFRC ↑ 3.9 [81] placenta 4/4 36,37^
Chorionic somatomammotropin hormone [43] serum 5/5 36,38^
Transthyretin [77]
[43]
 Placenta Serum 5/5
5/5
35,37 ^
 Plasma retinol-binding protein 4 [43] serum 5/5 36,38^
Alpha-2-HS-glycoprotein [43] serum 5/5 36,38^
pappalysin-2 ↓ −1 [44] serum 30/58 35,40###
39,40####
choriogonadotropin-beta ↓ −7.6 [44] serum 30/58 35,40###
39,40####
apolipoprotein C-III ↑ 2.1 [44] serum 30/58 35,40###
39,40####
cystatin-C ↑ 6.5 [44] serum 30/58 35,40###
39,40####
 vascular endothelial growth factor receptor-1 ↓ −8.4 [44] serum 30/58 35,40###
39,40####
endoglin ↓ −1 [44] serum 30/58 35,40###
39,40####
 chloride intracellular channel 3 [77] placenta 5/5 35,37 ^
protein disulphide isomerase [77] placenta 5/5 35,37 ^
peroxiredoxin 2 [77] placenta 5/5 35,37 ^
peroxiredoxin 3 [77] placenta 5/5 35,37 ^
Hsc 70 [77] placenta 5/5 35,37 ^
SOD-1 [77] placenta 5/5 35,37 ^
actin gamma 1 propeptide [77] placenta 5/5 35,37 ^
chain A of enoyl-coenzyme A hydratase [77] placenta 5/5 35,37 ^
HSP gp96 [77] placenta 5/5 35,37 ^
fibrinogen gamma chain 6.4 [41] plasma 39/57 38,40^
SBBI42 [78] amniotic fluid 18/16 36,36^
 Chaperonin (heatshock protein 60 ↑ 223 [82] placenta 8/8 33,36^
Glutathione S-transferase ↑ 177 [82] placenta 8/8 33,36^
 Voltage-dependent anion channel ↑ 185 [82] placenta 8/8 33,36^
ER-60protease ↑ 179 [82] placenta 8/8 33,36^
 Chain H, Cathepsin D at pH 7.5 ↑ 245 [82] placenta 8/8 33,36^
Catalase [83] placenta 3/3 29,38^
glucose-regulated protein [83] placenta 3/3 29,38^
Endothelial monocyte- activating polypeptide [83] placenta 3/3 29,38^
ENG ↑ 3.21 [84] placenta 10/10 35,39^
VIM ↓ −1.58 [84] placenta 10/10 35,39^
MMP-7 ↑ 1.4 [85] plasma 76/90 38,39###
38,39####
PlGF ↓ −1.1 [85] plasma 76/90 38,39###
38,39####
SOD1 ↑ 222.39 [86] placenta 20/20 ^
SMS ↑ 323.31 [86] placenta 20/20 ^
GSTK1 ↑ 90.64 [86] placenta 20/20 ^
GPX1 ↑ 25.18 [86] placenta 20/20 ^
GPX3 ↑ 5.80 [86] placenta 20/20 ^
GCLM ↑ 0.66 [86] placenta 20/20 ^
GGCT ↑ 1.26 [86] placenta 20/20 ^
GSTT2B ↑ 0.83 [86] placenta 20/20 ^
NFKB1 ↑ 67.84 [86] placenta 20/20 ^
Q9GZM7 ↓ 1.6 [87] placenta 25/25 ^
Protein AMBP (P02760) ↑ 1.8 [88]  cerebrospinal fluid 43/55 30,38^
 Complement C1s subcomponent [39] plasma 13/13 ^
P01009 [89] urine 114/92 36,33###
36,33####
P02768 [89] urine 114/92 36,33###
36,33####
Glycoprotein ormonesαchain ↑1.7 [93] Placenta 20,20 37,37^
Apolipoprotein A‐I ↑1.3 [93] Placenta 20,20 37,37^
Solute carrier organic anion ↑1.3 [93] Placenta 20,20 37,37^
transporter family member ↓0.79 [93] Placenta 20,20 37,37^
Platelet‐basic protein ↓0.77 [93] Placenta 20,20 37,37^
MMP - 7 ↑1.9 [94] Plasma 90,33 31,39##
Transferrin ↑3 [95] Urine 7,7 ^
Complement factor B ↑2 [95] Urine 7,7 ^
pregnancy-specific beta-1-glycoprotein 2 ↓2 [7] Serum 5,7 33,38^
##

early-onset preeclampsia

#

late-onset preeclampsia

###

severe preeclampsia

####

mild preeclampsia

^

not specified

*

Type of preeclampsia

Although a promising platform, thus far there are no clinically applicable protein biomarkers existing for preeclampsia, due to small sample sizes of the studies and lack of sufficient sensitivity and specificity required of clinical diagnostics. However, proteomics could be a useful tool for monitoring the disease or identifying at risk populations [47, 48].

Metabolomics studies of preeclampsia

Metabolomics has recently taken the spotlight in providing biomarker candidates, representing a more holistic approach to metabolites within a cell, tissue or organism [49]. It takes the platforms of mass spectrometry and NMR spectroscopy platforms. In preeclampsia, metabolomics are typically measured in blood, placenta and urine. We performed a PubMed literature search for the keywords “metabolomics” and “preeclampsia” and compiled the resulting studies.

Table 5 summarizes the differential metabolite profiles associated with preeclampsia in the literature [5056]. Serum samples [5053] are the most commonly used materials. In those samples, higher lipid, 3-hydroxybutyrate, arginine, phenylalanine, alanine and leucine levels and lower valine levels were detected in preeclampsia cases [50, 52, 53, 55]. Aberrant valine, leucine, arginine and phenylalanine levels are indicative of decreased amino acid biosynthesis. In urine, decreases in hippurate and increases in creatinine levels have been observed [57], suggesting aberrant kidney functions especially in kidney’s filtration processes. Aberrant metabolite profiles could be indicative of the effects of preeclampsia due to the high blood pressure associated with the disease. Metabolite profiling of placenta tissue has also been carried out by several groups [5860]. In the preeclamptic placenta tissue, the urea cycle is affected, evident by increased glutamate and glutamine levels. Therefore, a panel of several metabolite aberrations could serve as a diagnostic biomarker set for preeclampsia identification or monitoring of preeclampsia progression. Mapping these metabolites in Table 5 to their functional pathways would provide additional insight into the biochemical processes involved in preeclampsia progression [6163]. Metabolomic pathway enrichment analysis shows that lipid transport, neurotransmitter regulation and protein biosynthesis pathways are highly correlated to preeclampsia.

Table 5.

List of metabolites associated with preeclampsia. Metabolites, fold changes sample origin, study size, gestational age and severity of preeclampsia were also included.

Metabolite dysregulation Fold change Reference Sample origin Case/controls Gestational Age (preeclampsia, control in weeks) *
Valine ↓ −1.27 [50] maternal serum 59/115 #
Leucine ↑ 1.16 [50] maternal serum 59/115 #
Hippurate [51] urine 26/561 38,40^
Creatinine [51] urine 26/561 38,40^
Stearoylcarnitine ↑ 0.829 [52] maternal serum 167/500 31,40##
37,40#
Arginine ↑ 1.02 [53] maternal serum 50/108 ##
2-hydroxybutyrate ↑ 1.08 [53] maternal serum 50/108 ##
Citrate ↑ 0.06 [54] maternal plasma 30/60 ##
Glycerol <0.001 [54] maternal plasma 30/60 ##
Hydroxyisovalerate ↑ 0.01 [54] maternal plasma 30/60 ##
Methionine ↑ 0.024 [54] maternal plasma 30/60 ##
Hydroxyhexanoylcarnitine ↑ 0.78 [55] maternal serum 41/41 35,39^
Alanine ↑ 0.78 [55] maternal serum 41/41 35,39^
Phenylalanine ↑ 0.80 [55] maternal serum 41/41 35,39^
Glutamate ↑ 0.79 [55] maternal serum 41/41 35,39^
↑ 0.76 [56] placenta 6/6 ^
Glutamine ↑ 0.42 [56] placenta 6/6 ^
Cysteine ↑0.64 [97] Placenta 10,10 ##,#
eicosapentaenoic acid ↑2 [98] Plasma 10,10 37,38^
docosahexaenoic acid ↑1.4 [98] Plasma 10,10 37,38^
4-Hydroxyglutamate ↑3 [99] Serum 194,325 35,40##
39,40#
##

early-onset preeclampsia

#

late-onset preeclampsia

###

severe preeclampsia

####

mild preeclampsia

^

not specified

*

Type of preeclampsia

Although lots of metabolomics markers have been investigated, so far none has been selected as either predictive or diagnostic biomarkers for preeclampsia [64]. Like other omics studies mentioned earlier (proteomics and gene expression), metabolomics studies of preeclampsia also are limited by sample sizes and the heterogeneity of this disease. In fact, even in relatively well-studied field such as breast cancer, proposed metabolomics biomarkers have very little consistency across populations [65]. Knowing such challenge, future directions may focus on multi-omics studies and biological-knowledge based integration approach, in order to identify robust biomarkers for preeclampsia.

Integrative omics studies of preeclampsia

Multi-omics studies would significantly increase the robustness of discoveries in preeclampsia research. While there are several studies and repositories dedicated toward integrative multi-omic cancer research, such as The Cancer Genome Atlas (TCGA), none currently has been reported for preeclampsia. Rarely studies measured and then integrated multi-omics data from the same samples, except for two, one which validated preeclampsia biomarkers by integrating placental mRNA expression (GEO) and proteomics profiling [66] and the other which integrated epigenetics and transcriptomics of the same preeclampsia samples [100]. Clearly, much more work needs to be done, in order to obtain coherent biomarker signatures of preeclampsia at the multi-omics level.

Nevertheless, combining biomarkers from different studies and different omics levels may still yield interesting insights, despite many issues such as that the samples in different omics studies are not on the same subjects, same samples (eg. placenta, blood), same gestational ages as well as lacking many other unknown clinical confounders. The rationale here is that all these different omics, although assayed in different technological platforms and different samples, are all to some degrees reflecting the changes in the common basic biological units - genes. Thus we integrated at the gene level the GWAS, epigenetics, transcriptomics, proteomics and metabolomics biomarker candidates, from the above reviewed reports (Figure 1). Preeclampsia-associated markers identified within each omic platform were assigned to their study (S) to create a grouping system for multi-omic level analysis of all five platforms, despite that each original study only investigated one omic type. We then combined the grouping information with individual biomarkers to generate the heterogenous graph, in which multiple types of nodes are present. One type of node (no-color) is study ID shown as the reference index, and the other types of nodes represent biomarkers identified in one or more omics types. No single gene is associated with preeclampsia across all five omics platforms. The lack of coherence could be due to many reasons, such as the heterogeneity among different populations in these studies, the multi-factorial nature of preeclampsia, and limitations of sample sizes in each study. However, one gene, inhibin, showed fairly consistent changes in GWAS, DNA differential methylation and transcriptome levels. Inhibin levels generally rise in the first trimester, fall during the second trimester and rise again in the third trimester [67]. In preeclamptic pregnancies, inhibin levels are magnitudes higher compared to normal pregnancies [9]. Consistently, hypomethylation of inhibin was observed in preeclamptic placenta [18]. Further clinical evaluation on inhibin as biomarker is certainly of great interest.

Figure 1.

Figure 1.

Bipartite graph merging all omics platforms used to study preeclampsia. A bipartite group is composed of two types of nodes. One type of nodes depicts the biomarkers found in different omic-type studies. Omic-type is annotated by node color as indicated in the plot: Grey = GWAS, orange = Methylation, pink = Transcriptomics, blue = Proteomics, bright green = metabolomics. The other type of nodes, labelled as S, signifies the study information.

Although different genes, proteins or metabolites may show up as biomarkers in individual omics level, the biological functions that they are involved in may be shared. For example, previous we have found that pathways, a type of biological functional unit, are good surrogate biomarker features to predict breast cancer status, based upon metabolomics data set and applied to TCGA RNA-Seq validation dataset [103]. With this consideration, we examined these biomarker candidates on the background of a Protein-Protein interaction network next (Figure 2). Using Cytoscape (STRING App), we mapped the top targets within each omic platform to reveal protein-protein interaction networks which are dominant in preeclampsia. We used enzymes as the surrogates of metabolites, in order to integrate with the protein-protein interaction network. While it is also possible to use transport proteins as a surrogate for metabolites, we chose enzymes for this review study. Network analysis reveals four striking sub-networks with clear biological functions, supported by previous molecular-biology and clinical observations. The green network is composed of proteins associated with oxidant-antioxidant pathways (PRDX1, PRDX3, SOD, HSPE1, INVS, NOX5). This likely corresponds to the events facilitating the hypoxic microenvironment in preeclampsia. The blue network is composed of protein-protein interactions between proteasome pathways, cytokine release and inflammation (PSMA1, PSMA8, NFKB1, NFKB2, IL8). This sub-network explains the activated inflammatory response in preeclampsia. The yellow sub-network includes genes involved in protein transport, protein regulation and extracellular matrix remodelling pathways (TIMP3, TTR, SERPINA1, APO3, RBP4, AMBP, CST3, CXCL, VIM). This sub-network could be especially important in explaining the pathology observed in the preeclamptic placenta. The purple sub-network is comprised of protein-protein interactions associated with the angiogenesis and cell proliferation pathway (NCAM, FLT1, PGF, GRB2, PDGF, FGF14). Reduced spiral artery formation and reduced angiogenesis, hallmarks of preeclampsia, are likely manifested by these genes. Despite such emerging patterns, we should point out that one caveat of the network analysis here is that we have combined studies in placenta and blood together, thus the PPI based on blood biomarkers is inferred.

Figure 2.

Figure 2.

Protein-protein interaction network (STRING database) generated by combining all biomarkers of different omic types, reviewed earlier in the report. The network is done using Cytoscape visualization. Four dominant sub-networks associated with preeclampsia are shown (blue, green, yellow, purple), each enriched with biological functions. Antioxidant pathway involving PRDX2, PRDX3, HSPE1, GCLM and SOD1 (green). Proteasome and cytokine signalling pathway involving PSMA1, PSMD8, NFKB1, NFKB2 (blue). Protein regulation and transport pathways involving C1s, TFRC, CXCL1, TTR, TIMP3 (yellow). Angiogenesis signalling pathway involving FLT1, PGF, FGF, ENG (purple). The other red targets are still associated with preeclampsia but not within the four dominant sub-networks.

Therefore, even though the biomarkers were identified by different studies and at different omic level, integrative analysis on the protein-protein interaction networks of these biomarkers reveals coherent and coordinated biological functions that are otherwise much less clear in the individual omic studies.

Limitations and perspectives

Large amounts of omics data related to preeclampsia are accruing with the advancement of technologies. There is strong evidence that preeclampsia is a complex disease that involves programming changes at DNA, epigenetic, transcriptomic, protein and metabolite levels. Clinically, the application of understanding how each of these elements affects the process of the disease state may be able to provide insights into treatment and prevention. Omics approaches can open the door for new studies and research with focus on therapeutic approaches in the comparison of patients with and without preeclampsia [101, 102]. For instance, we already know that low-dose aspirin prevents early onset preeclampsia, but exactly how and why this prevention takes place is postulated, leaving room for omics studies to find out at what level the medication could be working. This is only one example of how future studies could determine and then lead to advances in how we treat and prevent preeclampsia by understanding the disease in a more stepwise molecular state.

As an effort to demonstrate the known and unknowns in preeclampsia omics field, we conducted a comprehensive literature review on the various studies at different omic levels. This review has presented the challenges in identifying robust biomarkers in preeclampsia. Many reasons may contribute to the observed lack of consistency in various omics studies. Besides sample size, the heterogeneity and the lack of detailed understanding of the heterogeneity of this syndrome, both at the clinical and molecular levels, likely play a part. The differing results obtained from experiments shows the importance of rigor in study design, on selecting sample type, sample site, type of omic platform and stage in disease to break through current limitations in understanding the etiology and effect of heterogeneous medical conditions such as preeclampsia. Performing matched mother-baby dyad sampling and multi-omics studies will increase useful information, relevance and perspective obtained from these omics approaches. Finally, we would like to call on community effort to share detailed description on clinical outcomes (eg. severity of preeclampsia, availability of medical record data) and the omics data at the raw data level. Harmonization of data, data collection, data sharing and development of public databases would equip investigators with advanced tools to improve the feasibility of multi-omic approaches toward more rigorous diagnosis and treatment of preeclampsia. Previously, only a very few databases have been created to archive significant findings from “Big Data” for preeclampsia studies. dbPEC collected and curated thousands of articles from PubMed, to generate a list of genes associated with preeclampsia [68]. PESNPdb is another database focused exclusively on SNPs associated with preeclampsia [69]. The vast amount of information from other earlier omics studies awaits to be re-collected, re-synthesized and integrated to understand preeclampsia. All of these aspects, once implemented, will enable better powered and more accurate biomarker discoveries at the subtype level.

Nevertheless, we showed that a systematic and multi-omic analysis approach, even at the post-statistics level and among very different populations of limited sizes, has the potential merit to unravel the general molecular mechanisms of preeclampsia coherently. These pathways are candidate targets for future therapeutic interventions for treating and controlling preeclampsia. Thus, our review and meta-study demonstrates the proof-of-principle to gain systematic molecular information on preeclampsia, by orchestrating multiple dimensions of omics data.

Highlights.

We review recent progress in preeclampisa using various omics technology platforms.

We show that in general coherent preecalmpsia biomarkers are lacking in all omics studies.

Using network systems biology approach, we identify four sub-networks related to preeclampisa.

Acknowledgements

Dr. Lana X Garmire’s research is supported by grants K01ES025434 awarded by NIEHS through funds provided by the trans-NIH Big Data to Knowledge (BD2K) initiative (http://datascience.nih.gov/bd2k), P20 COBRE GM103457 awarded by NIH/NIGMS, R01 LM012373 and LM 012907 awarded by NLM, and R01 HD084633 awarded by NICHD to LX Garmire. Funding was also provided in part by the Department of Obstetrics and Gynecology, University of Hawaii. The authors thank all members of the Garmire Group for their helpful discussions.

Footnotes

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

Conflict of Interest

The authors declare no competing interests exist.

Conflict of Interest Statement

The authors declares no conflict of interest.

References

  • 1.Sibai B, Dekker G, and Kupferminc M, Pre-eclampsia. Lancet, 2005. 365(9461): p. 785–99. [DOI] [PubMed] [Google Scholar]
  • 2.Mustafa R, et al. , A comprehensive review of hypertension in pregnancy. J Pregnancy, 2012. 2012: p. 105918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Villanueva Egan LA, Bohorquez Barragan ME, and Alanis Lopez P, [Factors related to the diagnosis of HELLP syndrome in patients with severe preeclampsia]. Ginecol Obstet Mex, 2004. 72: p. 394–9. [PubMed] [Google Scholar]
  • 4.Roberts JM, et al. , Vitamins C and E to prevent complications of pregnancy-associated hypertension. N Engl J Med, 2010. 362(14): p. 1282–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Zamorski MA and Green LA, Preeclampsia and hypertensive disorders of pregnancy. Am Fam Physician, 1996. 53(5): p. 1595–610. [PubMed] [Google Scholar]
  • 6.Wang Y, et al. , Clinical application of SNP array analysis in first-trimester pregnancy loss: a prospective study. Clin Genet, 2017. 91(6): p. 849–858. [DOI] [PubMed] [Google Scholar]
  • 7.McGinnis R, et al. , Variants in the fetal genome near FLT1 are associated with risk of preeclampsia. Nat Genet, 2017. 49(8): p. 1255–1260. [DOI] [PubMed] [Google Scholar]
  • 8.Zhao L, Bracken MB, and DeWan AT, Genome-wide association study of pre-eclampsia detects novel maternal single nucleotide polymorphisms and copy-number variants in subsets of the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) study cohort. Ann Hum Genet, 2013. 77(4): p. 277–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ree PH, et al. , Early detection of preeclampsia using inhibin a and other second-trimester serum markers. Fetal Diagn Ther, 2011. 29(4): p. 280–6. [DOI] [PubMed] [Google Scholar]
  • 10.Wang Q, et al. , Single nucleotide polymorphisms near the inhibin beta B gene on 2q14 are associated with pre-eclampsia in Han Chinese women. Eur J Obstet Gynecol Reprod Biol, 2015. 193: p. 127–31. [DOI] [PubMed] [Google Scholar]
  • 11.Wan JP, et al. , The common single-nucleotide polymorphism rs2681472 is associated with early-onset preeclampsia in Northern Han Chinese women. Reprod Sci, 2014. 21(11): p. 1423–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Johnson MP, et al. , Genome-wide association scan identifies a risk locus for preeclampsia on 2q14, near the inhibin, beta B gene. PLoS One, 2012. 7(3): p. e33666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Thomsen LC, et al. , The antihypertensive MTHFR gene polymorphism rs17367504-G is a possible novel protective locus for preeclampsia. J Hypertens, 2017. 35(1): p. 132–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Dupont C, Armant DR, and Brenner CA, Epigenetics: definition, mechanisms and clinical perspective. Semin Reprod Med, 2009. 27(5): p. 351–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Yeung KR, et al. , DNA methylation profiles in preeclampsia and healthy control placentas. Am J Physiol Heart Circ Physiol, 2016. 310(10): p. H1295–303. [DOI] [PubMed] [Google Scholar]
  • 16.Anton L, et al. , Differential methylation of genes associated with cell adhesion in preeclamptic placentas. PLoS One, 2014. 9(6): p. e100148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Feil R and Fraga MF, Epigenetics and the environment: emerging patterns and implications. Nat Rev Genet, 2012. 13(2): p. 97–109. [DOI] [PubMed] [Google Scholar]
  • 18.Blair JD, et al. , Widespread DNA hypomethylation at gene enhancer regions in placentas associated with early-onset pre-eclampsia. Mol Hum Reprod, 2013. 19(10): p. 697–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ferreira JC, et al. , WNT2 promoter methylation in human placenta is associated with low birthweight percentile in the neonate. Epigenetics, 2011. 6(4): p. 440–9. [DOI] [PubMed] [Google Scholar]
  • 20.Ching T, et al. , Genome-wide hypermethylation coupled with promoter hypomethylation in the chorioamniotic membranes of early onset pre-eclampsia. Mol Hum Reprod, 2014. 20(9): p. 885–904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chu T, et al. , Comprehensive analysis of preeclampsia-associated DNA methylation in the placenta. PLoS One, 2014. 9(9): p. e107318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hogg K, et al. , Early onset pre-eclampsia is associated with altered DNA methylation of cortisol-signalling and steroidogenic genes in the placenta. PLoS One, 2013. 8(5): p. e62969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Jia RZ, et al. , Screening for differential methylation status in human placenta in preeclampsia using a CpG island plus promoter microarray. Int J Mol Med, 2012. 30(1): p. 133–41. [DOI] [PubMed] [Google Scholar]
  • 24.Yuen RK, et al. , DNA methylation profiling of human placentas reveals promoter hypomethylation of multiple genes in early-onset preeclampsia. Eur J Hum Genet, 2010. 18(9): p. 1006–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kamburov A, et al. , The ConsensusPathDB interaction database: 2013 update. Nucleic acids research, 2013. 41(Database issue): p. D793–D800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Liu Y and Ma Y, Promoter Methylation Status of WNT2 in Placenta from Patients with Preeclampsia. Med Sci Monit, 2017. 23: p. 5294–5301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zhang Z, et al. , Association of Wnt2 and sFRP4 expression in the third trimester placenta in women with severe preeclampsia. Reprod Sci, 2013. 20(8): p. 981–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.MacDonald BT, Tamai K, and He X, Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell, 2009. 17(1): p. 9–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Struwe E, et al. , Microarray analysis of placental tissue in intrauterine growth restriction. Clin Endocrinol (Oxf), 2010. 72(2): p. 241–7. [DOI] [PubMed] [Google Scholar]
  • 30.Whitehead CL, et al. , Placental specific mRNA in the maternal circulation are globally dysregulated in pregnancies complicated by fetal growth restriction. J Clin Endocrinol Metab, 2013. 98(3): p. E429–36. [DOI] [PubMed] [Google Scholar]
  • 31.Kendall RL and Thomas KA, Inhibition of vascular endothelial cell growth factor activity by an endogenously encoded soluble receptor. Proc Natl Acad Sci U S A, 1993. 90(22): p. 10705–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gormley M, et al. , Preeclampsia: novel insights from global RNA profiling of trophoblast subpopulations. Am J Obstet Gynecol, 2017. [DOI] [PubMed]
  • 33.Song X, et al. , Dysregulation of LncRNAs in placenta and pathogenesis of preeclampsia. Curr Drug Targets, 2017. [DOI] [PubMed]
  • 34.Morales-Prieto DM, et al. , Elsevier Trophoblast Research Award Lecture: origin, evolution and future of placenta miRNAs. Placenta, 2014. 35 Suppl: p. S39–45. [DOI] [PubMed] [Google Scholar]
  • 35.Biro O, Nagy B, and Rigo J Jr., Identifying miRNA regulatory mechanisms in preeclampsia by systems biology approaches. Hypertens Pregnancy, 2017. 36(1): p. 90–99. [DOI] [PubMed] [Google Scholar]
  • 36.Luo R, et al. , Hypoxia-inducible miR-210 contributes to preeclampsia via targeting thrombospondin type I domain containing 7A. Sci Rep, 2016. 6: p. 19588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Brooks SA, et al. , miRNAs as common regulators of the transforming growth factor (TGF)-beta pathway in the preeclamptic placenta and cadmium-treated trophoblasts: Links between the environment, the epigenome and preeclampsia. Food Chem Toxicol, 2016. 98(Pt A): p. 50–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Graves PR and Haystead TA, Molecular biologist’s guide to proteomics. Microbiol Mol Biol Rev, 2002. 66(1): p. 39–63; table of contents. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kim SM, et al. , Expression changes of proteins associated with the development of preeclampsia in maternal plasma: A case-control study. Proteomics, 2016. 16(10): p. 1581–9. [DOI] [PubMed] [Google Scholar]
  • 40.Kolla V, et al. , Quantitative proteomic (iTRAQ) analysis of 1st trimester maternal plasma samples in pregnancies at risk for preeclampsia. J Biomed Biotechnol, 2012. 2012: p. 305964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Blumenstein M, et al. , A proteomic approach identifies early pregnancy biomarkers for preeclampsia: novel linkages between a predisposition to preeclampsia and cardiovascular disease. Proteomics, 2009. 9(11): p. 2929–45. [DOI] [PubMed] [Google Scholar]
  • 42.Lu Q, et al. , Serum markers of pre-eclampsia identified on proteomics. J Obstet Gynaecol Res, 2016. 42(9): p. 1111–8. [DOI] [PubMed] [Google Scholar]
  • 43.Liu C, et al. , Proteomic analysis of human serum for finding pathogenic factors and potential biomarkers in preeclampsia. Placenta, 2011. 32(2): p. 168–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Rasanen J, et al. , Comprehensive maternal serum proteomic profiles of preclinical and clinical preeclampsia. J Proteome Res, 2010. 9(8): p. 4274–81. [DOI] [PubMed] [Google Scholar]
  • 45.Goldberg M, et al. , Synthesis of complement proteins in the human chorion is differentially regulated by cytokines. Mol Immunol, 2007. 44(7): p. 1737–42. [DOI] [PubMed] [Google Scholar]
  • 46.Jarvie E, et al. , Lipotoxicity in obese pregnancy and its potential role in adverse pregnancy outcome and obesity in the offspring. Clin Sci (Lond), 2010. 119(3): p. 123–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Horgan RP and Kenny LC, ‘Omic’ technologies: genomics, transcriptomics, proteomics and metabolomics. The Obstetrician & Gynaecologist, 2011. 13(3): p. 189–195. [Google Scholar]
  • 48.Mischak H, et al. , Epidemiologic design and analysis for proteomic studies: a primer on -omic technologies. Am J Epidemiol, 2015. 181(9): p. 635–47. [DOI] [PubMed] [Google Scholar]
  • 49.Fanos V, et al. , Metabolomics application in maternal-fetal medicine. Biomed Res Int, 2013. 2013: p. 720514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Bahado-Singh RO, et al. , Metabolomic determination of pathogenesis of late-onset preeclampsia. J Matern Fetal Neonatal Med, 2017. 30(6): p. 658–664. [DOI] [PubMed] [Google Scholar]
  • 51.Austdal M, et al. , First Trimester Urine and Serum Metabolomics for Prediction of Preeclampsia and Gestational Hypertension: A Prospective Screening Study. Int J Mol Sci, 2015. 16(9): p. 21520–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Koster MP, et al. , First-Trimester Serum Acylcarnitine Levels to Predict Preeclampsia: A Metabolomics Approach. Dis Markers, 2015. 2015: p. 857108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Bahado-Singh RO, et al. , Validation of metabolomic models for prediction of early-onset preeclampsia. Am J Obstet Gynecol, 2015. 213(4): p. 530 e1–530 e10. [DOI] [PubMed] [Google Scholar]
  • 54.Bahado-Singh RO, et al. , Metabolomics and first-trimester prediction of early-onset preeclampsia. J Matern Fetal Neonatal Med, 2012. 25(10): p. 1840–7. [DOI] [PubMed] [Google Scholar]
  • 55.Odibo AO, et al. , First-trimester prediction of preeclampsia using metabolomic biomarkers: a discovery phase study. Prenat Diagn, 2011. 31(10): p. 990–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Dunn WB, et al. , Changes in the metabolic footprint of placental explant-conditioned culture medium identifies metabolic disturbances related to hypoxia and pre-eclampsia. Placenta, 2009. 30(11): p. 974–80. [DOI] [PubMed] [Google Scholar]
  • 57.Kenny LC, et al. , Robust early pregnancy prediction of later preeclampsia using metabolomic biomarkers. Hypertension, 2010. 56(4): p. 741–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Austdal M, et al. , Metabolic profiles of placenta in preeclampsia using HR-MAS MRS metabolomics. Placenta, 2015. 36(12): p. 1455–62. [DOI] [PubMed] [Google Scholar]
  • 59.Walejko JM, et al. , Global Metabolomics of the Placenta Reveals Distinct Metabolic Profiles between Maternal and Fetal Placental Tissues Following Delivery in Non-Labored Women. Metabolites, 2018. 8(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bahado-Singh R, et al. , Integrated Proteomic and Metabolomic prediction of Term Preeclampsia. Sci Rep, 2017. 7(1): p. 16189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Huang S, et al. , Novel personalized pathway-based metabolomics models reveal key metabolic pathways for breast cancer diagnosis. Genome Med, 2016. 8(1): p. 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Alakwaa FM, Chaudhary K, and Garmire LX, Deep Learning Accurately Predicts Estrogen Receptor Status in Breast Cancer Metabolomics Data. J Proteome Res, 2018. 17(1): p. 337–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Alakwaa F, Huang S, and Garmire L, Lilikoi: an R package for personalized pathway-based classification modeling using metabolomics data. bioRxiv, 2018. [DOI] [PMC free article] [PubMed]
  • 64.Nobakht MGBF, Application of metabolomics to preeclampsia diagnosis. Syst Biol Reprod Med, 2018: p. 1–16. [DOI] [PubMed]
  • 65.Huang S, et al. , Novel personalized pathway-based metabolomics models reveal key metabolic pathways for breast cancer diagnosis. Genome Medicine, 2016. 8(1): p. 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Liu LY, et al. , Integrating multiple ‘omics’ analyses identifies serological protein biomarkers for preeclampsia. BMC Med, 2013. 11: p. 236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Muttukrishna S, et al. , Measurement of serum concentrations of inhibin-A (alpha-beta A dimer) during human pregnancy. Clin Endocrinol (Oxf), 1995. 42(4): p. 391–7. [DOI] [PubMed] [Google Scholar]
  • 68.Triche EW, et al. , Bioinformatic approach to the genetics of preeclampsia. Obstet Gynecol, 2014. 123(6): p. 1155–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Tuteja G, et al. , PESNPdb: a comprehensive database of SNPs studied in association with pre-eclampsia. Placenta, 2012. 33(12): p. 1055–7. [DOI] [PubMed] [Google Scholar]
  • 70.Zhao L, et al. , Genome-wide association study identifies a maternal copy-number deletion in PSG11 enriched among preeclampsia patients. BMC Pregnancy Childbirth, 2012. 12: p. 61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Sandrim VC, et al. , Circulating microRNA expression profiles in pre-eclampsia: evidence of increased miR-885–5p levels. BJOG, 2016. 123(13): p. 2120–2128. [DOI] [PubMed] [Google Scholar]
  • 72.Sitras V, et al. , Differential placental gene expression in severe preeclampsia. Placenta, 2009. 30(5): p. 424–33. [DOI] [PubMed] [Google Scholar]
  • 73.Tsai S, et al. , Transcriptional profiling of human placentas from pregnancies complicated by preeclampsia reveals disregulation of sialic acid acetylesterase and immune signalling pathways. Placenta, 2011. 32(2): p. 175–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yong HE, et al. , Genome-wide transcriptome directed pathway analysis of maternal pre-eclampsia susceptibility genes. PLoS One, 2015. 10(5): p. e0128230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Brew O, Sullivan MH, and Woodman A, Comparison of Normal and Pre-Eclamptic Placental Gene Expression: A Systematic Review with Meta-Analysis. PLoS One, 2016. 11(8): p. e0161504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Mary S, et al. , Placental Proteomics Provides Insights into Pathophysiology of Pre-Eclampsia and Predicts Possible Markers in Plasma. J Proteome Res, 2017. 16(2): p. 1050–1060. [DOI] [PubMed] [Google Scholar]
  • 77.Gharesi-Fard B, Zolghadri J, and Kamali-Sarvestani E, Proteome differences of placenta between pre-eclampsia and normal pregnancy. Placenta, 2010. 31(2): p. 121–5. [DOI] [PubMed] [Google Scholar]
  • 78.Park JS, et al. , Identification of proteomic biomarkers of preeclampsia in amniotic fluid using SELDI-TOF mass spectrometry. Reprod Sci, 2008. 15(5): p. 457–68. [DOI] [PubMed] [Google Scholar]
  • 79.Yang JI, et al. , The Proteomic Analysis of Human Placenta with Pre-eclampsia and Normal Pregnancy. J Korean Med Sci, 2015. 30(6): p. 770–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ma K, et al. , A proteomic analysis of placental trophoblastic cells in preeclampsia-eclampsia. Cell Biochem Biophys, 2014. 69(2): p. 247–58. [DOI] [PubMed] [Google Scholar]
  • 81.Shi Z, et al. , Comparative proteomics analysis suggests that placental mitochondria are involved in the development of pre-eclampsia. PLoS One, 2013. 8(5): p. e64351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Kim YN, et al. , Toward a better understanding of preeclampsia: Comparative proteomic analysis of preeclamptic placentas. Proteomics Clin Appl, 2007. 1(12): p. 1625–36. [DOI] [PubMed] [Google Scholar]
  • 83.Webster RP, et al. , Differences in the proteome profile in placenta from normal term and preeclamptic preterm pregnancies. Proteomics Clin Appl, 2007. 1(5): p. 446–56. [DOI] [PubMed] [Google Scholar]
  • 84.Zhang W, et al. , Detergent-Insoluble Proteome Analysis Revealed Aberrantly Aggregated Proteins in Human Preeclampsia Placentas. J Proteome Res, 2017. 16(12): p. 4468–4480. [DOI] [PubMed] [Google Scholar]
  • 85.Erez O, et al. , The prediction of late-onset preeclampsia: Results from a longitudinal proteomics study. PLoS One, 2017. 12(7): p. e0181468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Jin X, et al. , Proteomics analysis of human placenta reveals glutathione metabolism dysfunction as the underlying pathogenesis for preeclampsia. Biochim Biophys Acta, 2017. 1865(9): p. 1207–1214. [DOI] [PubMed] [Google Scholar]
  • 87.Mary S, et al. , Tubulointerstitial nephritis antigen-like 1 protein is downregulated in the placenta of pre-eclamptic women. Clin Proteomics, 2017. 14: p. 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.van den Berg CB, et al. , Elevated levels of protein AMBP in cerebrospinal fluid of women with preeclampsia compared to normotensive pregnant women. Proteomics Clin Appl, 2017. 11(1–2). [DOI] [PubMed] [Google Scholar]
  • 89.Buhimschi IA, et al. , Proteomic profiling of urine identifies specific fragments of SERPINA1 and albumin as biomarkers of preeclampsia. Am J Obstet Gynecol, 2008. 199(5): p. 551 e1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ariff A, et al. , Analysis of the Epigenome in Multiplex Pre-eclampsia Families Identifies SORD, DGKI, and ICA1 as Novel Candidate Risk Genes. Front Genet. 2019. 10:227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Wang T, et al. , Epigenome-wide association data implicate fetal/maternal adaptations contributing to clinical outcomes in preeclampsia. Epigenomics. 2019. 11(9):1003–1019. [DOI] [PubMed] [Google Scholar]
  • 92.Gray KJ, et al. , Gene-Centric Analysis of Preeclampsia Identifies Maternal Association at PLEKHG1. Hypertension. 2018. 72(2):408–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Feng Y, et al. , Application of iTRAQ proteomics in identification of the differentially expressed proteins of placenta of pregnancy with preeclampsia. J Cell Biochem. 2019. 120(4):5409–5416. [DOI] [PubMed] [Google Scholar]
  • 94.Tarca AL, et al. , The prediction of early preeclampsia: Results from a longitudinal proteomics study. PLoS One. 2019. 14(6):e0217273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Ding W, et al. , Isobaric tag for relative and absolute quantitation based quantitative proteomics reveals unique urinary protein profiles in patients with preeclampsia. J Cell Mol Med. 2019. 23(8):5822–5826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Ling Y, et al. , Screening of serum biomarkers of preeclampsia by proteomics combination with bioinformatics. Hypertens Pregnancy. 2019. 38(3):184–192. [DOI] [PubMed] [Google Scholar]
  • 97.Kawasaki K, et al. , Metabolomic Profiles of Placenta in Preeclampsia. Hypertension. 2019. 73(3):671–679. [DOI] [PubMed] [Google Scholar]
  • 98.Liu Y, et al. , Metabolomics revealed decreased level of omega-3 PUFA-derived protective eicosanoids in pregnant women with pre-eclampsia. Clin Exp Pharmacol Physiol. 2019. 46(8):705–710 [DOI] [PubMed] [Google Scholar]
  • 99.Sovio U, et al. , 4-Hydroxyglutamate is a novel predictor of pre-eclampsia. Int J Epidemiol. 2019. pii: dyz098 10.1093/ije/dyz098. [DOI] [PMC free article] [PubMed]
  • 100.Leavey K, et al. , Epigenetic regulation of placental gene expression in transcriptional subtypes of preeclampsia. Clin Epigenet 10, 28 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Leavey K, et al. , Unsupervised Placental Gene Expression Profiling Identifies Clinically Relevant Subclasses of Human Preeclampsia. Hypertension. 2016. 68(1):137–47. [DOI] [PubMed] [Google Scholar]
  • 102.Leavey K, et al. , Large scale aggregate microarray analysis reveals three distinct molecular subclasses of human preeclampsia. PLoS One. 2015. 10(2):e0116508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Huang S, Chong N, Lewis NE, Jia W, Xie G, Garmire LX. Novel personalized pathway-based metabolomics models reveal key metabolic pathways for breast cancer diagnosis. Genome Med. 2016. 8(1):34. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES