Skip to main content
Journal of Translational Medicine logoLink to Journal of Translational Medicine
. 2014 Aug 8;12:204. doi: 10.1186/1479-5876-12-204

Placenta-derived exosomes continuously increase in maternal circulation over the first trimester of pregnancy

Suchismita Sarker 1, Katherin Scholz-Romero 1, Alejandra Perez 2, Sebastian E Illanes 1,2,3, Murray D Mitchell 1, Gregory E Rice 1,2, Carlos Salomon 1,2,
PMCID: PMC4283151  PMID: 25104112

Abstract

Background

Human placenta releases specific nanovesicles (i.e. exosomes) into the maternal circulation during pregnancy, however, the presence of placenta-derived exosomes in maternal blood during early pregnancy remains to be established. The aim of this study was to characterise gestational age related changes in the concentration of placenta-derived exosomes during the first trimester of pregnancy (i.e. from 6 to 12 weeks) in plasma from women with normal pregnancies.

Methods

A time-series experimental design was used to establish pregnancy-associated changes in maternal plasma exosome concentrations during the first trimester. A series of plasma were collected from normal healthy women (10 patients) at 6, 7, 8, 9, 10, 11 and 12 weeks of gestation (n = 70). We measured the stability of these vesicles by quantifying and observing their protein and miRNA contents after the freeze/thawing processes. Exosomes were isolated by differential and buoyant density centrifugation using a sucrose continuous gradient and characterised by their size distribution and morphology using the nanoparticles tracking analysis (NTA; Nanosight™) and electron microscopy (EM), respectively. The total number of exosomes and placenta-derived exosomes were determined by quantifying the immunoreactive exosomal marker, CD63 and a placenta-specific marker (Placental Alkaline Phosphatase PLAP).

Results

These nanoparticles are extraordinarily stable. There is no significant decline in their yield with the freeze/thawing processes or change in their EM morphology. NTA identified the presence of 50–150 nm spherical vesicles in maternal plasma as early as 6 weeks of pregnancy. The number of exosomes in maternal circulation increased significantly (ANOVA, p = 0.002) with the progression of pregnancy (from 6 to 12 weeks). The concentration of placenta-derived exosomes in maternal plasma (i.e. PLAP+) increased progressively with gestational age, from 6 weeks 70.6 ± 5.7 pg/ml to 12 weeks 117.5 ± 13.4 pg/ml. Regression analysis showed that weeks is a factor that explains for >70% of the observed variation in plasma exosomal PLAP concentration while the total exosome number only explains 20%.

Conclusions

During normal healthy pregnancy, the number of exosomes present in the maternal plasma increased significantly with gestational age across the first trimester of pregnancy. This study is a baseline that provides an ideal starting point for developing early detection method for women who subsequently develop pregnancy complications, clinically detected during the second trimester. Early detection of women at risk of pregnancy complications would provide an opportunity to develop and evaluate appropriate intervention strategies to limit acute adverse sequel.

Keywords: Exosomes, Pregnancy, Placenta, Fetal-maternal exchange

Background

The placenta plays a pivotal role in mediating maternal adaptation to pregnancy as well as regulating fetal growth and development. Pregnancy-induced changes are affected by the release of soluble autacoids as early as 6 to 8 weeks of gestation [1, 2] and the invasion of placental cells into the maternal tissues to modify maternal immune, cardiovascular and metabolic activities. Recently, we and others [37] have identified an additional pathway by which the placenta communicates with the maternal system to induce changes during pregnancy-placental exosomal signalling.

Exosomes are bilipid membrane-bound nanovesicles (50–120 nm diameter) that are actively released (via exocytosis) from cells into the extracellular space and body fluids under physiological and pathophysiological conditions [8]. Their molecular cargo of proteins, microRNAs, mRNAs and lipids appear to be selectively packaged by the late endosomal system to regulate the phenotype of target cells [3, 4, 6]. Recent studies have highlighted the putative utility of tissue-specific nanovesicles (e.g. exosomes) in the diagnosis of disease onset and treatment monitoring [4, 9, 10].

Previously, we have established that placental cells release exosomes in response to changes in the extracellular milieu (including oxygen tension and glucose concentration) and that placental cell-derived exosomes regulate target cell migration and invasion [3, 4]. In addition, we have identified placental-derived exosomes in maternal blood and reported that the concentration of placental exosomes in the maternal blood increases during normal, healthy pregnancy [7]. During early placentation, the cytotrophoblast cells form a highly invasive extravillous trophoblast that can migrate into the decidua and invade the first third of the myometrium, inducing remodelling of spiral arterioles to produce low-resistance vascular system, essential for fetal development [11]. The relative reduction of utero-placental flow caused by abnormal placentation triggers the development of placental originated diseases such as preeclampsia. Available data suggest that the concentrations of placental-derived exosomes in the maternal blood could be a potential marker of abnormal placentation [12, 13].

Early detection of disease risk and onset is the first step in implementing efficacious treatment and improving patient outcome. To date, the concentration profile of placenta-derived exosomes in the maternal blood during first trimester has not been established. Until this profile is defined, the utility of placental exosomes as an early biomarker for placental dysfunction will remain equivocal. In this study, therefore, a time-series experimental design was used to test the hypothesis that the concentration of placental exosomes in the maternal plasma of normal healthy women changes during the early pregnancy state (i.e. 6–12 weeks).

Methods

Patient selection and sample collection

A time-series experimental design was used to establish the variation in plasma exosome characteristics during normal pregnancy. All experimental procedures were conducted within an ISO17025 accredited (National Association of Testing Authorities, Australia) research facility. All data were recorded within a 21 CERF part 11 compliant electronic laboratory notebook (Iris note, Redwood City, CA, USA). Plasma samples were collected from 10 women during their first trimester of pregnancy. All patients were enrolled with informed consent and underwent routine obstetrical care at the Hospital Parroquial de San Bernardo (Santiago, Chile). Estimation of gestational age was made based on the first day of their last menstrual period and confirmed by transvaginal ultrasound at the recruitment (i.e. 6 weeks). Each patient, gave consent to have weekly blood sample collection between 6 and 12 weeks of gestation (n = 70, 10 patients with weekly blood collection at 6, 7, 8, 9, 10, 11 and 12 weeks of pregnancy). The protocol of the study was approved by the Institutional Review Board of the Universidad de los Andes (Santiago, Chile). Obstetrical history and physical findings were recorded regarding previous spontaneous abortions, course of previous pregnancies, hypertension, gestational diabetes and preeclampsia. Peripheral venous blood samples were collected in EDTA treated tubes (BD Vacutainer® Plus plastic plasma tube) from which plasma samples were obtained by centrifugation at 2000 × g at 4°C for 10 min. The plasma samples were stored in aliquots at −80°C until analysed (not more than three months).

Exosome isolation

Exosomes were isolated as previously described [3, 4, 7, 14]. Briefly, plasma from each patient was utilised to isolate exosomes. Plasma (2.5 ml) was diluted with equal volume of PBS (pH 7.4) and exosomes were isolated through differential centrifugation, microfiltration and buoyant density ultracentrifugation. Centrifugation was initially performed at 2,000 × g at 4°C for 30 min (Thermo Fisher Scientific Ins., Asheville, NC, USA, Sorvall®, high speed microcentrifuge, fixed rotor angle: 90°) followed by 12,000 × g at 4°C for 45 min to sediment cell nuclei, mitochondria and debris. The supernatant fluid (~5 ml) was transferred to an ultracentrifuge tube (Ultracrimp tubes, Thermo Fisher Scientific Ins., Asheville, NC, USA) and was centrifuged at 200,000 × g at 4°C for 2 h (Thermo Fisher Scientific Ins., Asheville, NC, USA, Sorvall®, T-8100, fixed angle ultracentrifuge rotor). The pellet was suspended in PBS (5 ml) and filtered through a 0.22 μm filter (SteritopTM, Millipore, Billerica, MA, USA). The filtrate was centrifuged at 200,000 × g at 4°C for 70 min (Thermo Fisher Scientific Ins., Asheville, NC, USA, Sorvall®, T-8100, fixed angle ultracentrifuge rotor) and the pellet resuspended in 2.5 M sucrose (4 ml).

Purification of exosomes using a continuous sucrose gradient

The resuspended 200,000 g pellet in 2.5 M sucrose was added at the bottom of an ultracentrifuge tube. A continuous sucrose gradient (26 ml; 0.25-2.5 M) was made above 4 ml of exosome suspension using a Hoefer SG30 gradient maker (GE Healthcare, NSW, Australia) and centrifuged at 110,000 g for 20 h (Sorvall, SureSpin™ 630/360, Swinging-Bucket ultracentrifuge rotor). Fractions (10 in total, 3 ml each) were collected automatically using a Pulse-Free Flow Peristaltic Pump with a flow rate range of 3 ml per min (GILSON Miniplus® model 3) and the Fraction Collector (GILSON FC 203B model). The density of each fraction was determined using the refraction index with OPTi digital refractometer (Bellingham + Stanley Inc., Lawrenceville, GA, USA). The coefficient of variation (CV) was less than 8% for the density of each fraction. Fractions (3 ml each) were diluted in PBS (60 ml) and then centrifuged at 200,000 × g for 70 min. The 200,000 g pellet was resuspended in 50 μl PBS and stored at −80°C. Exosomal protein concentrations were determined by a colorimetric assay (DC™ Protein Assay, Bio-Rad Laboratories, Hercules, CA, USA) [4].

Identification of nanoparticles by nanoparticle tracking analysis (NTA)

NTA measurements were performed using a NanoSight NS500 instrument (NanoSight NTA 2.3 Nanoparticle Tracking and Analysis Release Version Build 0033) following the manufacturer’s instructions. The NanoSight NS500 instrument measured the rate of Brownian motion of nanoparticles in a light scattering system that provides a reproducible platform for specific and general nanoparticle characterization (NanoSight Ltd., Amesbury, United Kingdom). Samples were processed in duplicates and diluted with PBS over a range of concentrations to obtain between 10 and 100 particles per image (optimal ~50 particles x image) before analysing with NTA system. The samples were mixed before introducting into the chamber (temperature: 25°C and viscosity: 0.89 cP) and the camera level set to obtain image that has sufficient contrast to clearly identify particles while minimizing background noise a video recording (camera level: 10 and capture duration: 60 s). The captured videos (2 videos per sample) were then processed and analysed. A combination of high shutter speed (450) and gain (250) followed by manual focusing enabled optimum visualization of a maximum number of vesicles. We included a minimum of 200 tracks completed per video in duplicates. NTA post acquisition settings were optimized and kept constant between samples (Frames Processed: 1496 of 1496, Frames per Second: 30, camera shutter: 20 ms; Calibration: 139 nm/pixel, Blur: 3×3; Detection Threshold: 10; Min Track Length: Auto; Min Expected Size: Auto), and each video was then analyzed to give the mean, mode, and median particles size together with an estimate of the number of particles. An Excel spreadsheet (Microsoft Corp., Redmond, Washington) was also automatically generated, showing the concentration at each particle size.

Transmission electron microscopy (TEM)

For the TEM analysis, exosome pellets (as described above, 30 μg protein) were fixed in 3% (w/v) glutaraldehyde and 2% paraformaldehyde in cacodylate buffer, pH 7.3. Exosome samples were then applied to a continuous carbon grid and negatively stained with 2% uranyl acetate. The samples were examined in an FEI Tecnai 12 transmission electron microscope (FEI™, Hillsboro, Oregon, USA) in the Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology (QUT) (see Acknowledgements).

Quantification of placental cell-derived exosome

The concentration of exosomes in maternal circulation was expressed as the total immunoreactive exosomal CD63 (ExoELISA™, System Biosciences, Mountain View, CA). Briefly, 10 μg of exosomal protein was immobilised in 96-well microtiter plates and incubated overnight (binding step). Plates were washed three times for 5 min using a wash buffer solution and then incubated with exosome specific primary antibody (CD63) at room temperature (RT) for 1 h under agitation. Plates were washed and incubated with secondary antibody (1:5000) at RT 1 h under agitation. Plates were washed and incubated with Super-sensitive TMB ELISA substrate at RT for 45 min under agitation. The reaction was terminated using Stop Buffer solution. Absorbance was measured at 450 nm. The number of exosomes/ml, (ExoELISA™ kit) was obtained using an exosomal CD63 standard curve calibrated against nanoparticle tracking data (i.e. number of exosomes, NanoSight™).

For placental cell-derived exosomes, the concentration of exosomal PLAP was quantified using a commercial ELISA kit (MYBioSource MBS701995, San Diego, CA, USA) according to manufacturer’s instructions (detection range: 84–2000 pg/ml; sensitivity: 34 pg/ml; intra-assay precision within an assay: CV% < 10%; inter-assay between assays: CV% < 15%) Briefly, 10 μg of exosomal protein was added to each well of a 96-well microtitre plate and incubated at 37°C for 30 min. Plates were washed three times while shaking for 20 s and 50 μl of HRP-conjugate was added to each well and incubated at 37°C for 20 min. Plates were washed and incubated with 50 μl of substrate A and 50 μl of substrate B at 37°C for 15 min. The incubation was terminated using 50 μl of stop solution at RT for 2 min under agitation. Absorbance was measured at 450 nm. Exosomal PLAP was expressed as pg PLAP /ml plasma.

Stability of the exosomal quantification

To determine the stability of the exosomes during freeze-thaw cycles, fresh plasma (5.0 ml) from healthy women were obtained and divided into two 2.5 ml samples (A and B). Exosomes were immediately isolated from the first aliquot (A: fresh plasma) by differential and buoyant density centrifugation and then characterised by the number of exosome particles using an ELISA kit (ExoELISA™, System Biosciences, Mountain View, CA), morphologically by electron microscope, microRNA content by real time PCR and protein profiling by mass-spectrometry. Sample B plasma was stored at −80°C for 2 months (B: frozen plasma), prior to exosome isolation and characterisation. miRNA isolation: miRNA were isolated from exosome particles as we have previously described [14]. Ambion mirVana PARIS Kit (Invitrogen, USA) was used to extract exosomal total RNA from fresh and frozen plasma by following the manufacturer’s procedure. Exosomes were first lysed by adding cell disruption buffer and vortexed or pipetted vigorously. Denaturing solution was added to samples and incubated on ice for 5 min. The first two steps stabilize RNA and inactivate RNases. The lysate is then subjected to Acid-Phenol:Chloroform extraction by adding Acid-Phenol:Chloroform, vortexed and centrifuged at 10,000 × g for 5 min. Recovery of the aqueous phase obtains semi-pure RNA samples, removing most of the other cellular components. 100% ethanol was mixed and passed through a filter cartilage. The filter was washed three times and the RNA was eluted with nuclease-free water. Real-time PCR: Reverse transcription was performed using the miScript Reverse Transcription Kit (QIAGEN, Valencia, CA, USA) in a total volume of 20 μl. cDNA was synthesised from the maximum volume of exosomal RNA (12 μl) using the BIO-RAD T100™ Thermal Cycler (USA) running for 60 min at 37°C, 5 min for 95°C and 60 min for 37°C. As the control, RNase-free water was added as the RNA template. Real-time PCR was performed with miScript SYBR Green Kit (QIAGEN, Valencia, CA, USA). Forward primers (miScript primer assays, QIAGEN, Valencia, CA, USA) designed to detect the housekeeping gene, human RNU6-2 (RNU6B) was used. The reactions were performed in triplicate using the BIO-RAD iQ™5 Multicolor Real-Time PCR Detection System (USA) with the following conditions: 94°C for 3 min, 35 amplification cycles of 94°C for 45 s, 55°C for 30 s and 72°C for 30 s, 72°C for 10 min, 12°C for ∞ min. Proteomic analysis of exosomes by mass spectrometry (MS): We utilised a Liquid Chromatography (LC) and Mass Spectrometry (MS) LC/MS/MS instrumentation available within the University of Queensland Centre for Clinical Research (5500qTRAP and 5600 Triple TOF) to undertake in depth quantitative proteomic analysis of the exosome samples (isolated from fresh and frozen plasma) to determine the proteome of exosomes as we have previously published [4]. Briefly, exosomes were adjusted to 8 M urea in 50 mM ammonium bicarbonate, pH 8.5, and reduced with tris (2-carboxyethyl) phosphine (5 mM) at room temperature for 1 h. Proteins were then alkylated in 10 mM IAA for 1 h in the dark. The sample was diluted 1:10 with 50 mM ammonium bicarbonate and then digested with trypsin (20 μg) at 37°C for 18 h. The samples were dried by centrifugal evaporation to remove the acetonitrile and then redissolved in Solvent A. The digested protein samples were analysed using a 5600 Triple TOF mass spectrometer (ABSciex) to obtain initial high mass accuracy survey MS/MS data, identifing the peptides present in the samples. The in depth proteomic analysis was performed using the Information Dependent Acquisition (IDA) experiments on the 5600 Triple TOF MS and utilized an enhanced MS survey scan (m/z350–1500) followed by 50 data-dependent product ion scans of the 50 most intense precursor ions. The MS data was analysed with the Markerview software package using Principal Components Analysis (PCA) or PCA-Discriminate Analysis (PCA-DA) which compares data across multiple samples, groupings the data sets, and graphically showing the groups in a Scores plot. The Loadings plot provides valuable insight into variables that lead to sample clustering and illustrates which biomarkers are up- or down-regulated. All mass spectra were analysed using the Mascot and Protein Pilot search engines against the Swissprot-swissprot database with the species set as human (scores greater than 30). False discovery rate (FDR) was estimated using a reversed sequence database. Finally, proteins identified were submitted to bioinformatic pathway analysis (Ingenuity Pathway Analysis [IPA]; Ingenuity Systems, Mountain View, CA; http://www.ingenuity.com).

Statistical analysis

Data are presented as mean ± SEM, with n = 10 different patients per group (i.e. 6, 7, 8, 9, 10, 11, 12 weeks). The effect of gestational age on number of exosome particles and placental-derived exosomes were assessed by two-way ANOVA, with variance partitioned between gestational age and subject. Statistical difference between group means was assessed by Dunn’s test to compare each treatment to the control group where the data distribution approximates normality and by Mann–Whitney U-test for distribution independent data analysis. Two group means were statistically assessed by Student’s t-test. Statistical significance was defined as p < 0.05.

Results

Exosome characterisation

Maternal plasma exosomes isolated by differential and sucrose density gradient centrifugation were characterised by a buoyant density of 1.122 to 1.197 g/ml (fractions 4 to 7) (Figure 1A-D). Nanoparticle tracking analysis showed a particle size distribution of 200,000 × g pellet (Figure 1A) ranging from 30 to 300 nm in diameter corresponding to microsomal fraction (including exosomes particles) with an average of 147 ± 71 nm (mean ± SD) (Figure 1B). After the sucrose continuous gradient, we mixed the enriched exosomal fractions (1.122 to 1.197 g/ml) (Figure 1C) and obtained a particle size distribution ranged from 50 to 140 nm in diameter, with an average of 98 ± 39 nm (mean ± SD) (Figure 1D). Electron microscopy revealed the presence of spherical vesicles, with a typical cup-shape and diameters ranging from 30 to 120 nm (Figure 1D, insert).

Figure 1.

Figure 1

Characterisation of exosome from maternal circulation. Exosome were isolated from women uncomplicated pregnancies during first trimester by differential and buoyant density centrifugation (see Methods). (A) Flow chart for the exosome purification procedure based on differential ultracentrifugation. (B) Representative particles size distribution of microsomal fraction. (C) Flow chart for the exosome purification procedure based on sucrose continuous gradient (exosome enriched fractions in yellow 4–7). (D) Representative particles size distribution of enriched exosomal fractions (fraction 4–7 were mixed). Insert: Representative electron micrograph exosome fractions (pooled enriched exosome population from fractions 4 to 7), Scale bar 200 nm.

The stability of exosomes after a freeze and thaw cycle was evaluated using fresh and frozen plasma. No significant difference was observed using fresh or frozen plasma in exosome quantification, exosomal marker expression, microRNA expression or protein content (Figure 2A-D, Table 1).

Figure 2.

Figure 2

Characteristics of exosomes isolated from plasma immediately after phlebotomy (○) and after 30 days stored at −80°C (●). (A) Number of exosome particles. (B) Exosomes characterization. b1: electron microscope (scale bar 100 nm) and b2: Western blot for CD63 (exosomal marker); lane 1: Fresh and lane 2: stored. (C) Expression of miRNA RNU6B in exosomes. (D) Venn diagram of proteins identified in fresh and stored exosomes.

Table 1.

Common proteins identified in exosomes isolated from fresh plasma and after freeze/thawing cycles

Protein ID Symbol Entrez gene name Location Type(s)
A2MG_HUMAN A2M alpha-2-macroglobulin Extracellular Space transporter
A2ML1_HUMAN A2ML1 alpha-2-macroglobulin-like 1 Cytoplasm other
ACACA_HUMAN ACACA acetyl-CoA carboxylase alpha Cytoplasm enzyme
ACTN3_HUMAN ACTN3 actinin, alpha 3 Plasma Membrane other
ADAL_HUMAN ADAL adenosine deaminase-like Cytoplasm enzyme
ATS16_HUMAN ADAMTS16 ADAM metallopeptidase with thrombospondin type 1 motif, 16 Extracellular Space other
ATS9_HUMAN ADAMTS9 ADAM metallopeptidase with thrombospondin type 1 motif, 9 Extracellular Space peptidase
DSRAD_HUMAN ADAR adenosine deaminase, RNA-specific Nucleus enzyme
ADCY7_HUMAN ADCY7 adenylate cyclase 7 Plasma Membrane enzyme
KFA_HUMAN AFMID arylformamidase Nucleus enzyme
ANGT_HUMAN AGT angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Extracellular Space growth factor
ALBU_HUMAN ALB albumin Extracellular Space transporter
AMZ1_HUMAN AMZ1 archaelysin family metallopeptidase 1 Other peptidase
ANK2_HUMAN ANK2 ankyrin 2, neuronal Plasma Membrane other
ANKAR_HUMAN ANKAR ankyrin and armadillo repeat containing Nucleus transcription regulator
AKD1B_HUMAN ANKDD1B ankyrin repeat and death domain containing 1B Other other
ANKL1_HUMAN ANKLE1 ankyrin repeat and LEM domain containing 1 Other other
ANR12_HUMAN ANKRD12 ankyrin repeat domain 12 Nucleus other
ANR26_HUMAN ANKRD26 ankyrin repeat domain 26 Nucleus transcription regulator
ANKUB_HUMAN ANKUB1 ankyrin repeat and ubiquitin domain containing 1 Other other
APOA1_HUMAN APOA1 apolipoprotein A-I Extracellular Space transporter
APOB_HUMAN APOB apolipoprotein B Extracellular Space transporter
APOL1_HUMAN APOL1 apolipoprotein L, 1 Extracellular Space transporter
APOP1_HUMAN APOPT1 apoptogenic 1, mitochondrial Cytoplasm other
DP13B_HUMAN APPL2 adaptor protein, phosphotyrosine interaction, PH domain and leucine zipper containing 2 Cytoplasm other
RHG15_HUMAN ARHGAP15 Rho GTPase activating protein 15 Cytoplasm other
RHG08_HUMAN ARHGAP8/PRR5-ARHGAP8 Rho GTPase activating protein 8 Cytoplasm other
ARHGB_HUMAN ARHGEF11 Rho guanine nucleotide exchange factor (GEF) 11 Cytoplasm other
ASPM_HUMAN ASPM asp (abnormal spindle) homolog, microcephaly associated (Drosophila) Nucleus other
ATG2B_HUMAN ATG2B autophagy related 2B Other other
AT2A3_HUMAN ATP2A3 ATPase, Ca++ transporting, ubiquitous Cytoplasm transporter
RENR_HUMAN ATP6AP2 ATPase, H + transporting, lysosomal accessory protein 2 Cytoplasm transporter
ATR_HUMAN ATR ataxia telangiectasia and Rad3 related Nucleus kinase
B4GT7_HUMAN B4GALT7 xylosylprotein beta 1,4-galactosyltransferase, polypeptide 7 Cytoplasm enzyme
BEND4_HUMAN BEND4 BEN domain containing 4 Other other
OSTCN_HUMAN BGLAP bone gamma-carboxyglutamate (gla) protein Extracellular Space other
BLM_HUMAN BLM Bloom syndrome, RecQ helicase-like Nucleus enzyme
BRCA2_HUMAN BRCA2 breast cancer 2, early onset Nucleus transcription regulator
BRPF1_HUMAN BRPF1 bromodomain and PHD finger containing, 1 Nucleus transporter
CS068_HUMAN C19orf68 chromosome 19 open reading frame 68 Other other
CA174_HUMAN C1orf174 chromosome 1 open reading frame 174 Nucleus other
CA228_HUMAN C1orf228 chromosome 1 open reading frame 228 Other other
C1QC_HUMAN C1QC complement component 1, q subcomponent, C chain Extracellular Space other
CO3_HUMAN C3 complement component 3 Extracellular Space peptidase
CO4A_HUMAN C4A/C4B complement component 4B (Chido blood group) Extracellular Space other
C4BPA_HUMAN C4BPA complement component 4 binding protein, alpha Extracellular Space other
CI078_HUMAN C9orf78 chromosome 9 open reading frame 78 Other other
CAH3_HUMAN CA3 carbonic anhydrase III, muscle specific Cytoplasm enzyme
CABIN_HUMAN CABIN1 calcineurin binding protein 1 Nucleus other
CAND1_HUMAN CAND1 cullin-associated and neddylation-dissociated 1 Cytoplasm transcription regulator
CAN1_HUMAN CAPN1 calpain 1, (mu/I) large subunit Cytoplasm peptidase
CAN2_HUMAN CAPN2 calpain 2, (m/II) large subunit Cytoplasm peptidase
CASC5_HUMAN CASC5 cancer susceptibility candidate 5 Nucleus other
C8AP2_HUMAN CASP8AP2 caspase 8 associated protein 2 Nucleus transcription regulator
CC154_HUMAN CCDC154 coiled-coil domain containing 154 Other other
CC171_HUMAN CCDC171 coiled-coil domain containing 171 Other other
CCD30_HUMAN CCDC30 coiled-coil domain containing 30 Other other
CCD37_HUMAN CCDC37 coiled-coil domain containing 37 Other other
CCD80_HUMAN CCDC80 coiled-coil domain containing 80 Nucleus other
CCHCR_HUMAN CCHCR1 coiled-coil alpha-helical rod protein 1 Cytoplasm other
CENPH_HUMAN CENPH centromere protein H Nucleus other
CP135_HUMAN CEP135 centrosomal protein 135 kDa Cytoplasm other
CFAH_HUMAN CFH complement factor H Extracellular Space other
CHD4_HUMAN CHD4 chromodomain helicase DNA binding protein 4 Nucleus enzyme
CHD9_HUMAN CHD9 chromodomain helicase DNA binding protein 9 Cytoplasm other
ACHG_HUMAN CHRNG cholinergic receptor, nicotinic, gamma (muscle) Plasma Membrane transmembrane receptor
CHSTB_HUMAN CHST11 carbohydrate (chondroitin 4) sulfotransferase 11 Cytoplasm enzyme
CHSS3_HUMAN CHSY3 chondroitin sulfate synthase 3 Cytoplasm enzyme
CILP1_HUMAN CILP cartilage intermediate layer protein, nucleotide pyrophosphohydrolase Extracellular Space phosphatase
CLNK_HUMAN CLNK cytokine-dependent hematopoietic cell linker Cytoplasm other
CLUS_HUMAN CLU clusterin Cytoplasm other
CMBL_HUMAN CMBL carboxymethylenebutenolidase homolog (Pseudomonas) Cytoplasm enzyme
CNO6L_HUMAN CNOT6L CCR4-NOT transcription complex, subunit 6-like Cytoplasm enzyme
COPA1_HUMAN COL25A1 collagen, type XXV, alpha 1 Cytoplasm other
CROCC_HUMAN CROCC ciliary rootlet coiled-coil, rootletin Plasma Membrane other
CSRN1_HUMAN CSRNP1 cysteine-serine-rich nuclear protein 1 Nucleus transcription regulator
DIAC_HUMAN CTBS chitobiase, di-N-acetyl- Cytoplasm enzyme
CUL9_HUMAN CUL9 cullin 9 Cytoplasm other
CWC25_HUMAN CWC25 CWC25 spliceosome-associated protein homolog (S. cerevisiae) Other other
CP1A2_HUMAN CYP1A2 cytochrome P450, family 1, subfamily A, polypeptide 2 Cytoplasm enzyme
CP51A_HUMAN CYP51A1 cytochrome P450, family 51, subfamily A, polypeptide 1 Cytoplasm enzyme
DAPL1_HUMAN DAPL1 death associated protein-like 1 Other other
DCAF6_HUMAN DCAF6 DDB1 and CUL4 associated factor 6 Nucleus transcription regulator
DCR1B_HUMAN DCLRE1B DNA cross-link repair 1B Nucleus enzyme
DCSTP_HUMAN DCSTAMP dendrocyte expressed seven transmembrane protein Plasma Membrane other
DCX_HUMAN DCX doublecortin Cytoplasm other
DDX51_HUMAN DDX51 DEAD (Asp-Glu-Ala-Asp) box polypeptide 51 Other enzyme
DEN2D_HUMAN DENND2D DENN/MADD domain containing 2D Cytoplasm other
DESM_HUMAN DES desmin Cytoplasm other
DGAT1_HUMAN DGAT1 diacylglycerol O-acyltransferase 1 Cytoplasm enzyme
DGC14_HUMAN DGCR14 DiGeorge syndrome critical region gene 14 Nucleus other
DHX30_HUMAN DHX30 DEAH (Asp-Glu-Ala-His) box helicase 30 Nucleus enzyme
DIP2B_HUMAN DIP2B DIP2 disco-interacting protein 2 homolog B (Drosophila) Cytoplasm other
DMXL1_HUMAN DMXL1 Dmx-like 1 Extracellular Space other
DYH17_HUMAN DNAH17 dynein, axonemal, heavy chain 17 Cytoplasm other
DYH2_HUMAN DNAH2 dynein, axonemal, heavy chain 2 Other other
DYH3_HUMAN DNAH3 dynein, axonemal, heavy chain 3 Extracellular Space enzyme
DYH5_HUMAN DNAH5 dynein, axonemal, heavy chain 5 Cytoplasm enzyme
DNJC7_HUMAN DNAJC7 DnaJ (Hsp40) homolog, subfamily C, member 7 Cytoplasm other
DOP1_HUMAN DOPEY1 dopey family member 1 Cytoplasm other
DSCAM_HUMAN DSCAM Down syndrome cell adhesion molecule Plasma Membrane other
DUS3L_HUMAN DUS3L dihydrouridine synthase 3-like (S. cerevisiae) Other other
DYHC2_HUMAN DYNC2H1 dynein, cytoplasmic 2, heavy chain 1 Cytoplasm other
COE2_HUMAN EBF2 early B-cell factor 2 Nucleus other
EBP_HUMAN EBP emopamil binding protein (sterol isomerase) Cytoplasm enzyme
EIF3C_HUMAN EIF3C eukaryotic translation initiation factor 3, subunit C Other translation regulator
ENPP1_HUMAN ENPP1 ectonucleotide pyrophosphatase/phosphodiesterase 1 Plasma Membrane enzyme
ENPP5_HUMAN ENPP5 ectonucleotide pyrophosphatase/phosphodiesterase 5 (putative) Extracellular Space enzyme
PERE_HUMAN EPX eosinophil peroxidase Cytoplasm enzyme
EXOS1_HUMAN EXOSC1 exosome component 1 Nucleus enzyme
F150A_HUMAN FAM150A family with sequence similarity 150, member A Other other
F196B_HUMAN FAM196B family with sequence similarity 196, member B Other other
F208B_HUMAN FAM208B family with sequence similarity 208, member B Other other
YV021_HUMAN FAM230B family with sequence similarity 230, member B (non-protein coding) Extracellular Space other
FA78B_HUMAN FAM78B family with sequence similarity 78, member B Other other
FBF1_HUMAN FBF1 Fas (TNFRSF6) binding factor 1 Nucleus other
FIBA_HUMAN FGA fibrinogen alpha chain Extracellular Space other
FIBB_HUMAN FGB fibrinogen beta chain Extracellular Space other
FR1OP_HUMAN FGFR1OP FGFR1 oncogene partner Cytoplasm kinase
FGRL1_HUMAN FGFRL1 fibroblast growth factor receptor-like 1 Plasma Membrane transmembrane receptor
FIBG_HUMAN FGG fibrinogen gamma chain Extracellular Space other
FHAD1_HUMAN FHAD1 forkhead-associated (FHA) phosphopeptide binding domain 1 Other other
FIGL2_HUMAN FIGNL2 fidgetin-like 2 Other other
FLNB_HUMAN FLNB filamin B, beta Cytoplasm other
FINC_HUMAN FN1 fibronectin 1 Extracellular Space enzyme
FRMD3_HUMAN FRMD3 FERM domain containing 3 Other other
G6PC2_HUMAN G6PC2 glucose-6-phosphatase, catalytic, 2 Cytoplasm phosphatase
GAK_HUMAN GAK cyclin G associated kinase Nucleus kinase
GSH0_HUMAN GCLM glutamate-cysteine ligase, modifier subunit Cytoplasm enzyme
GCN1L_HUMAN GCN1L1 GCN1 general control of amino-acid synthesis 1-like 1 (yeast) Cytoplasm translation regulator
CXB1_HUMAN GJB1 gap junction protein, beta 1, 32 kDa Plasma Membrane transporter
GLRA2_HUMAN GLRA2 glycine receptor, alpha 2 Plasma Membrane ion channel
GMEB1_HUMAN GMEB1 glucocorticoid modulatory element binding protein 1 Nucleus transcription regulator
GOGA3_HUMAN GOLGA3 golgin A3 Cytoplasm transporter
AATC_HUMAN GOT1 glutamic-oxaloacetic transaminase 1, soluble Cytoplasm enzyme
GRID2_HUMAN GRID2 glutamate receptor, ionotropic, delta 2 Plasma Membrane ion channel
GSAP_HUMAN GSAP gamma-secretase activating protein Cytoplasm peptidase
GSAS1_HUMAN GSN-AS1 GSN antisense RNA 1 Other other
GSHB_HUMAN GSS glutathione synthetase Cytoplasm enzyme
HERC1_HUMAN HERC1 HECT and RLD domain containing E3 ubiquitin protein ligase family member 1 Cytoplasm other
HES1_HUMAN HES1 hes family bHLH transcription factor 1 Nucleus transcription regulator
HILS1_HUMAN HILS1 histone linker H1 domain, spermatid-specific 1, pseudogene Nucleus other
HIP1_HUMAN HIP1 huntingtin interacting protein 1 Cytoplasm other
HJURP_HUMAN HJURP Holliday junction recognition protein Nucleus other
HPTR_HUMAN HPR haptoglobin-related protein Extracellular Space peptidase
5HT2A_HUMAN HTR2A 5-hydroxytryptamine (serotonin) receptor 2A, G protein-coupled Plasma Membrane G-protein coupled receptor
I23O2_HUMAN IDO2 indoleamine 2,3-dioxygenase 2 Cytoplasm enzyme
GILT_HUMAN IFI30 interferon, gamma-inducible protein 30 Cytoplasm enzyme
IGHA1_HUMAN IGHA1 immunoglobulin heavy constant alpha 1 Extracellular Space other
IGHG1_HUMAN IGHG1 immunoglobulin heavy constant gamma 1 (G1m marker) Extracellular Space other
IGHM_HUMAN IGHM immunoglobulin heavy constant mu Plasma Membrane transmembrane receptor
IGJ_HUMAN IGJ immunoglobulin J polypeptide, linker protein for immunoglobulin alpha and mu polypeptides Extracellular Space other
IGKC_HUMAN IGKC immunoglobulin kappa constant Extracellular Space other
KV401_HUMAN IGKV4-1 immunoglobulin kappa variable 4-1 Extracellular Space other
LAC1_HUMAN IGLC1 immunoglobulin lambda constant 1 (Mcg marker) Cytoplasm other
LAC2_HUMAN IGLC2 immunoglobulin lambda constant 2 (Kern-Oz- marker) Extracellular Space other
IHH_HUMAN IHH indian hedgehog Extracellular Space enzyme
RED_HUMAN IK IK cytokine, down-regulator of HLA II Extracellular Space cytokine
IL1AP_HUMAN IL1RAP interleukin 1 receptor accessory protein Plasma Membrane transmembrane receptor
IRPL2_HUMAN IL1RAPL2 interleukin 1 receptor accessory protein-like 2 Plasma Membrane transmembrane receptor
IL26_HUMAN IL26 interleukin 26 Extracellular Space cytokine
INCE_HUMAN INCENP inner centromere protein antigens 135/155 kDa Nucleus other
IQCF6_HUMAN IQCF6 IQ motif containing F6 Other other
JARD2_HUMAN JARID2 jumonji, AT rich interactive domain 2 Nucleus transcription regulator
KTNB1_HUMAN KATNB1 katanin p80 (WD repeat containing) subunit B 1 Cytoplasm enzyme
KCND2_HUMAN KCND2 potassium voltage-gated channel, Shal-related subfamily, member 2 Plasma Membrane ion channel
KCNQ5_HUMAN KCNQ5 potassium voltage-gated channel, KQT-like subfamily, member 5 Plasma Membrane ion channel
KDM2B_HUMAN KDM2B lysine (K)-specific demethylase 2B Nucleus other
KDM5A_HUMAN KDM5A lysine (K)-specific demethylase 5A Nucleus transcription regulator
TALD3_HUMAN KIAA0586 KIAA0586 Cytoplasm other
K1161_HUMAN KIAA1161 KIAA1161 Nucleus other
KI13A_HUMAN KIF13A kinesin family member 13A Cytoplasm transporter
KIF19_HUMAN KIF19 kinesin family member 19 Extracellular Space enzyme
KIRR1_HUMAN KIRREL kin of IRRE like (Drosophila) Plasma Membrane other
KLC2_HUMAN KLC2 kinesin light chain 2 Cytoplasm other
KLRF1_HUMAN KLRF1 killer cell lectin-like receptor subfamily F, member 1 Plasma Membrane transmembrane receptor
LDB1_HUMAN LDB1 LIM domain binding 1 Nucleus transcription regulator
LHPL3_HUMAN LHFPL3 lipoma HMGIC fusion partner-like 3 Other other
LIPC_HUMAN LIPC lipase, hepatic Extracellular Space enzyme
YP023_HUMAN LOC100128265 uncharacterized LOC100128265 Other other
LRP1B_HUMAN LRP1B low density lipoprotein receptor-related protein 1B Plasma Membrane transmembrane receptor
LTBP2_HUMAN LTBP2 latent transforming growth factor beta binding protein 2 Extracellular Space other
LY75_HUMAN LY75 lymphocyte antigen 75 Plasma Membrane transmembrane receptor
MACD1_HUMAN MACROD1 MACRO domain containing 1 Cytoplasm enzyme
MANF_HUMAN MANF mesencephalic astrocyte-derived neurotrophic factor Extracellular Space other
MLP3A_HUMAN MAP1LC3A microtubule-associated protein 1 light chain 3 alpha Cytoplasm other
MAP4_HUMAN MAP4 microtubule-associated protein 4 Cytoplasm other
MA7D3_HUMAN MAP7D3 MAP7 domain containing 3 Cytoplasm other
MBD5_HUMAN MBD5 methyl-CpG binding domain protein 5 Nucleus other
MDN1_HUMAN MDN1 MDN1, midasin homolog (yeast) Nucleus other
MEX3B_HUMAN MEX3B mex-3 RNA binding family member B Other kinase
MFNG_HUMAN MFNG MFNG O-fucosylpeptide 3-beta-N-acetylglucosaminyltransferase Cytoplasm enzyme
MKL1_HUMAN MKL1 megakaryoblastic leukemia (translocation) 1 Nucleus transcription regulator
MRE11_HUMAN MRE11A MRE11 meiotic recombination 11 homolog A (S. cerevisiae) Nucleus enzyme
RM32_HUMAN MRPL32 mitochondrial ribosomal protein L32 Cytoplasm translation regulator
MYBA_HUMAN MYBL1 v-myb avian myeloblastosis viral oncogene homolog-like 1 Nucleus transcription regulator
MYO15_HUMAN MYO15A myosin XVA Cytoplasm other
MYO3A_HUMAN MYO3A myosin IIIA Cytoplasm kinase
MYO6_HUMAN MYO6 myosin VI Cytoplasm other
ULA1_HUMAN NAE1 NEDD8 activating enzyme E1 subunit 1 Cytoplasm enzyme
NUCL_HUMAN NCL nucleolin Nucleus other
NCOA2_HUMAN NCOA2 nuclear receptor coactivator 2 Nucleus transcription regulator
NEBU_HUMAN NEB nebulin Cytoplasm other
NEDD4_HUMAN NEDD4 neural precursor cell expressed, developmentally down-regulated 4, E3 ubiquitin protein ligase Cytoplasm enzyme
NHS_HUMAN NHS Nance-Horan syndrome (congenital cataracts and dental anomalies) Nucleus other
NOA1_HUMAN NOA1 nitric oxide associated 1 Cytoplasm other
NRX3A_HUMAN NRXN3 neurexin 3 Other transporter
NSD1_HUMAN NSD1 nuclear receptor binding SET domain protein 1 Nucleus transcription regulator
NSN5C_HUMAN NSUN5P2 NOP2/Sun domain family, member 5 pseudogene 2 Other other
NET5_HUMAN NTN5 netrin 5 Other other
NUD15_HUMAN NUDT15 nudix (nucleoside diphosphate linked moiety X)-type motif 15 Cytoplasm phosphatase
OBSCN_HUMAN OBSCN obscurin, cytoskeletal calmodulin and titin-interacting RhoGEF Cytoplasm kinase
OCEL1_HUMAN OCEL1 occludin/ELL domain containing 1 Other other
ODFP2_HUMAN ODF2 outer dense fiber of sperm tails 2 Cytoplasm other
NOE2_HUMAN OLFM2 olfactomedin 2 Cytoplasm other
OPN4_HUMAN OPN4 opsin 4 Plasma Membrane G-protein coupled receptor
OR4K1_HUMAN OR4K1 olfactory receptor, family 4, subfamily K, member 1 Plasma Membrane G-protein coupled receptor
PALB2_HUMAN PALB2 partner and localizer of BRCA2 Nucleus other
PAR3L_HUMAN PARD3B par-3 family cell polarity regulator beta Plasma Membrane other
PARP4_HUMAN PARP4 poly (ADP-ribose) polymerase family, member 4 Cytoplasm enzyme
PCDH8_HUMAN PCDH8 protocadherin 8 Plasma Membrane other
PCLO_HUMAN PCLO piccolo presynaptic cytomatrix protein Cytoplasm transporter
PEAK1_HUMAN PEAK1 pseudopodium-enriched atypical kinase 1 Plasma Membrane kinase
PEG10_HUMAN PEG10 paternally expressed 10 Nucleus other
PER3_HUMAN PER3 period circadian clock 3 Nucleus other
PFD6_HUMAN PFDN6 prefoldin subunit 6 Cytoplasm other
PIGS_HUMAN PIGS phosphatidylinositol glycan anchor biosynthesis, class S Cytoplasm enzyme
P3C2A_HUMAN PIK3C2A phosphatidylinositol-4-phosphate 3-kinase, catalytic subunit type 2 alpha Cytoplasm kinase
SOX_HUMAN PIPOX pipecolic acid oxidase Cytoplasm enzyme
PLCD3_HUMAN PLCD3 phospholipase C, delta 3 Cytoplasm enzyme
PLXA4_HUMAN PLXNA4 plexin A4 Plasma Membrane transmembrane receptor
PNKD_HUMAN PNKD paroxysmal nonkinesigenic dyskinesia Nucleus other
PNKP_HUMAN PNKP polynucleotide kinase 3'-phosphatase Nucleus kinase
DPOLQ_HUMAN POLQ polymerase (DNA directed), theta Nucleus enzyme
PMGT1_HUMAN POMGNT1 protein O-linked mannose N-acetylglucosaminyltransferase 1 (beta 1,2-) Cytoplasm enzyme
PPIG_HUMAN PPIG peptidylprolyl isomerase G (cyclophilin G) Nucleus enzyme
PP12C_HUMAN PPP1R12C protein phosphatase 1, regulatory subunit 12C Cytoplasm phosphatase
PPT2_HUMAN PPT2 palmitoyl-protein thioesterase 2 Cytoplasm enzyme
PREB_HUMAN PREB prolactin regulatory element binding Nucleus transcription regulator
PPCEL_HUMAN PREPL prolyl endopeptidase-like Other peptidase
PRG4_HUMAN PRG4 proteoglycan 4 Extracellular Space other
PRP31_HUMAN PRPF31 pre-mRNA processing factor 31 Nucleus other
PRC2A_HUMAN PRRC2A proline-rich coiled-coil 2A Cytoplasm other
PSB3_HUMAN PSMB3 proteasome (prosome, macropain) subunit, beta type, 3 Cytoplasm peptidase
PRS7_HUMAN PSMC2 proteasome (prosome, macropain) 26S subunit, ATPase, 2 Nucleus peptidase
PTPRM_HUMAN PTPRM protein tyrosine phosphatase, receptor type, M Plasma Membrane phosphatase
PTTG3_HUMAN PTTG3P pituitary tumor-transforming 3, pseudogene Other other
PZP_HUMAN PZP pregnancy-zone protein Extracellular Space other
RAB10_HUMAN RAB10 RAB10, member RAS oncogene family Cytoplasm enzyme
RB3GP_HUMAN RAB3GAP1 RAB3 GTPase activating protein subunit 1 (catalytic) Cytoplasm other
RAB6A_HUMAN RAB6A RAB6A, member RAS oncogene family Cytoplasm enzyme
RAB8B_HUMAN RAB8B RAB8B, member RAS oncogene family Cytoplasm enzyme
RGPA2_HUMAN RALGAPA2 Ral GTPase activating protein, alpha subunit 2 (catalytic) Cytoplasm other
RBM23_HUMAN RBM23 RNA binding motif protein 23 Nucleus other
REG1A_HUMAN REG1A regenerating islet-derived 1 alpha Extracellular Space growth factor
RELN_HUMAN RELN reelin Extracellular Space peptidase
RFC4_HUMAN RFC4 replication factor C (activator 1) 4, 37 kDa Nucleus other
RFX8_HUMAN RFX8 RFX family member 8, lacking RFX DNA binding domain Other other
RMND1_HUMAN RMND1 required for meiotic nuclear division 1 homolog (S. cerevisiae) Cytoplasm other
RNF17_HUMAN RNF17 ring finger protein 17 Cytoplasm other
RN213_HUMAN RNF213 ring finger protein 213 Cytoplasm enzyme
RN219_HUMAN RNF219 ring finger protein 219 Other other
FTM_HUMAN RPGRIP1L RPGRIP1-like Cytoplasm other
RL29_HUMAN RPL29 ribosomal protein L29 Cytoplasm other
RL37_HUMAN RPL37 ribosomal protein L37 Cytoplasm other
KS6A4_HUMAN RPS6KA4 ribosomal protein S6 kinase, 90 kDa, polypeptide 4 Cytoplasm kinase
RTKN_HUMAN RTKN rhotekin Cytoplasm other
RYR2_HUMAN RYR2 ryanodine receptor 2 (cardiac) Plasma Membrane ion channel
SAMD8_HUMAN SAMD8 sterile alpha motif domain containing 8 Cytoplasm other
SASH1_HUMAN SASH1 SAM and SH3 domain containing 1 Extracellular Space other
UTER_HUMAN SCGB1A1 secretoglobin, family 1A, member 1 (uteroglobin) Extracellular Space cytokine
SCUB3_HUMAN SCUBE3 signal peptide, CUB domain, EGF-like 3 Plasma Membrane other
SPB9_HUMAN SERPINB9 serpin peptidase inhibitor, clade B (ovalbumin), member 9 Cytoplasm other
SET1A_HUMAN SETD1A SET domain containing 1A Nucleus ion channel
SHAN1_HUMAN SHANK1 SH3 and multiple ankyrin repeat domains 1 Cytoplasm other
SHAN3_HUMAN SHANK3 SH3 and multiple ankyrin repeat domains 3 Plasma Membrane other
CTL1_HUMAN SLC44A1 solute carrier family 44 (choline transporter), member 1 Plasma Membrane transporter
SNTAN_HUMAN SNTN sentan, cilia apical structure protein Other other
SOLH1_HUMAN SOHLH1 spermatogenesis and oogenesis specific basic helix-loop-helix 1 Cytoplasm transcription regulator
SPAG7_HUMAN SPAG7 sperm associated antigen 7 Nucleus other
SPA2L_HUMAN SPATA2L spermatogenesis associated 2-like Other other
CYTSB_HUMAN SPECC1 sperm antigen with calponin homology and coiled-coil domains 1 Nucleus other
SPO11_HUMAN SPO11 SPO11 meiotic protein covalently bound to DSB Nucleus enzyme
SPTN5_HUMAN SPTBN5 spectrin, beta, non-erythrocytic 5 Plasma Membrane other
SRGP2_HUMAN SRGAP2 SLIT-ROBO Rho GTPase activating protein 2 Cytoplasm other
SRG2C_HUMAN SRGAP2C SLIT-ROBO Rho GTPase activating protein 2C Other other
SIA7B_HUMAN ST6GALNAC2 ST6 (alpha-N-acetyl-neuraminyl-2,3-beta-galactosyl-1,3)-N-acetylgalactosaminide alpha-2,6-sialyltransferase 2 Cytoplasm enzyme
STXB1_HUMAN STXBP1 syntaxin binding protein 1 Cytoplasm transporter
SP20H_HUMAN SUPT20H suppressor of Ty 20 homolog (S. cerevisiae) Nucleus other
SPT6H_HUMAN SUPT6H suppressor of Ty 6 homolog (S. cerevisiae) Nucleus transcription regulator
SVEP1_HUMAN SVEP1 sushi, von Willebrand factor type A, EGF and pentraxin domain containing 1 Cytoplasm other
SYNJ1_HUMAN SYNJ1 synaptojanin 1 Cytoplasm phosphatase
TADA3_HUMAN TADA3 transcriptional adaptor 3 Nucleus transcription regulator
TBX20_HUMAN TBX20 T-box 20 Nucleus transcription regulator
TDRD1_HUMAN TDRD1 tudor domain containing 1 Cytoplasm other
TET1_HUMAN TET1 tet methylcytosine dioxygenase 1 Nucleus other
THMS1_HUMAN THEMIS thymocyte selection associated Cytoplasm other
TLK2_HUMAN TLK2 tousled-like kinase 2 Cytoplasm kinase
TM131_HUMAN TMEM131 transmembrane protein 131 Extracellular Space other
T132C_HUMAN TMEM132C transmembrane protein 132C Other other
T151A_HUMAN TMEM151A transmembrane protein 151A Other other
TM232_HUMAN TMEM232 transmembrane protein 232 Other other
TNFA_HUMAN TNF tumor necrosis factor Extracellular Space cytokine
TPD54_HUMAN TPD52L2 tumor protein D52-like 2 Cytoplasm other
TRML4_HUMAN TREML4 triggering receptor expressed on myeloid cells-like 4 Other other
TRI32_HUMAN TRIM32 tripartite motif containing 32 Nucleus transcription regulator
TRI65_HUMAN TRIM65 tripartite motif containing 65 Other other
TARA_HUMAN TRIOBP TRIO and F-actin binding protein Nucleus other
TRIPB_HUMAN TRIP11 thyroid hormone receptor interactor 11 Cytoplasm transcription regulator
TROAP_HUMAN TROAP trophinin associated protein Cytoplasm peptidase
TRPC5_HUMAN TRPC5 transient receptor potential cation channel, subfamily C, member 5 Plasma Membrane ion channel
TSG13_HUMAN TSGA13 testis specific, 13 Other other
TTC12_HUMAN TTC12 tetratricopeptide repeat domain 12 Other other
TITIN_HUMAN TTN titin Cytoplasm kinase
GCP6_HUMAN TUBGCP6 tubulin, gamma complex associated protein 6 Cytoplasm other
TRXR3_HUMAN TXNRD3 thioredoxin reductase 3 Cytoplasm enzyme
UBQLN_HUMAN UBQLNL ubiquilin-like Other other
UCKL1_HUMAN UCKL1 uridine-cytidine kinase 1-like 1 Cytoplasm kinase
UGDH_HUMAN UGDH UDP-glucose 6-dehydrogenase Nucleus enzyme
USP9X_HUMAN USP9X ubiquitin specific peptidase 9, X-linked Plasma Membrane peptidase
UTRO_HUMAN UTRN utrophin Plasma Membrane transmembrane receptor
VP13C_HUMAN VPS13C vacuolar protein sorting 13 homolog C (S. cerevisiae) Cytoplasm other
WAC_HUMAN WAC WW domain containing adaptor with coiled-coil Nucleus other
WDR1_HUMAN WDR1 WD repeat domain 1 Extracellular Space other
WDR35_HUMAN WDR35 WD repeat domain 35 Cytoplasm other
WDR43_HUMAN WDR43 WD repeat domain 43 Nucleus other
WFDC3_HUMAN WFDC3 WAP four-disulfide core domain 3 Extracellular Space other
YIPF1_HUMAN YIPF1 Yip1 domain family, member 1 Cytoplasm other
NIPA_HUMAN ZC3HC1 zinc finger, C3HC-type containing 1 Nucleus other
ZFHX4_HUMAN ZFHX4 zinc finger homeobox 4 Extracellular Space other
ZF64B_HUMAN ZFP64 ZFP64 zinc finger protein Nucleus other
ZN132_HUMAN ZNF132 zinc finger protein 132 Nucleus other
ZNF14_HUMAN ZNF14 zinc finger protein 14 Nucleus transcription regulator
ZN215_HUMAN ZNF215 zinc finger protein 215 Nucleus transcription regulator
Z286B_HUMAN ZNF286B zinc finger protein 286B Other other
ZN345_HUMAN ZNF345 zinc finger protein 345 Nucleus transcription regulator
ZN532_HUMAN ZNF532 zinc finger protein 532 Other other
ZN561_HUMAN ZNF561 zinc finger protein 561 Nucleus other
ZN624_HUMAN ZNF624 zinc finger protein 624 Nucleus other
ZNF74_HUMAN ZNF74 zinc finger protein 74 Nucleus other

List of common exosomal proteins are presented as Protein ID, Symbol, Entrez Gene Name, Location and type. No significant differences were observed en exosomal protein content from fresh or frozen plasma (coefficient of variation < 5%) after different freeze thawing cycle from the same sample.

Placenta-derived exosome increased during first trimester in normal pregnancy

Pooled exosome-containing fractions (i.e. fractions 4 to 7) were further characterised by determining the number of exosome (NEP) and exosomal PLAP concentration in the serial samples of maternal plasma obtained during first trimester of pregnancy (i.e. 6–12 weeks).

The gestational age variation in plasma exosome number was analysed by two-way ANOVA with the variance partitioned between gestational age and subject. A significantly effect of gestational age was identified (n = 69, one missing value, p < 0.005). A post-hoc multiple range test was used to identify statistically significant (p <0.05) differences between pairwise comparisons (Figure 3A). In addition, a significant effect of subject was identified (n = 69, one missing value, p < 0.05) (Figure 3B). In addition, NEP and gestational age (i.e. 6–12 weeks) displayed a significant positive linear relationship (r2 = 0.202, p < 0.001, n = 69, one missing value).

Figure 3.

Figure 3

Exosome profiling across first trimester pregnancy. Enriched exosomal population (i.e. number of exosome particles) and placenta-derived exosomes (i.e. exosomal PLAP) were quantified in in peripheral plasma of women in the first trimester of pregnancy by ELISA. (A) exosomes as particles per ml plasma. (B) individual variation in exosome number for each week (C) exosomal PLAP during first trimester of pregnancy (i.e. 6–12 weeks). (D) individual variation in exosomal PLAP for each week. Data are presented as aligned dot plot and values are mean ± SEM. In A, two-way ANOVA **p = 0.0048, Dunn’s post-hoc test analysis = *p < 0.05 6 vs. 7 weeks and p < 0.005: 6 vs. 12 weeks. In C, two-way ANOVA ***p < 0.0001, Dunn’s post-hoc test analysis = *p < 0.05 6 vs. 9 and 10 weeks, p < 0.005: 6 vs. 11 and 12 weeks, and p < 0.005: 8 vs. 11 and 12 weeks.

To assess gestational variation in placenta-derived exosomes, exosomal immunoreative (IR) PLAP was quantified using a commercial ELISA kit (see Methods). IR exosomal PLAP concentrations were analysed by two-way ANOVA with the variance partitioned between gestational age and subject. A significant effect of gestational age was identified (p < 0.0001, n = 69, one missing value) (Figure 3C). A post-hoc multiple range test was used to identify statistically significant (p <0.05) differences between pairwise comparisons (Figure 3D). No significant effect of patient on exosomal PLAP concentration was identified (p = 0.123). Immunoreactive exosomal PLAP concentration and gestational age displayed a significant positive linear linear relationship (r2 = 0.711, p < 0.001, n = 69, one missing value).

Specific placental-derived exosomes

Exosomal PLAP concentration and exosome number were subjected to linear regression analysis. The fitted linear model was described by the following equation: plasma exosomal PLAP pg/ml = 85.6 + 5.47 × 10−11 × exosome number/ml (p < 0.006, n = 69, one missing pair). The coefficient of determination (r2) was 10.8 (Figure 4A).

Figure 4.

Figure 4

Contribution of placental-derived exosomes into maternal circulation. (A) Relationship between exosomal PLAP and exosomes (particles per ml plasma) across first trimester of pregnancy (i.e. 6–12 weeks represented by colours). (B) Ratio of specific placental exosome and exosomes. In A, values are mean ± SEM, Linear correlation (−). In B, Data are presented as aligned dot plot and values are mean ± SEM, two-way ANOVA p > 0.05.

To estimate changes in the relative contribution of placental exosomes within the total exosomes present in maternal plasma and identify changes over the gestational age, the apparent PLAP content per 109 exosome (PLAP ratio) was determined. Overall PLAP ratio averaged 2.01 ± 0.33 × 10−9 exosomal PLAP (pg) per exosome. The effects of gestational age on PLAP ratio were assessed by Kruskal-Wallis one-way ANOVA. No significant effect of gestational age on PLAP ratio was identified (p = 0.06) (Figure 4B).

Discussion

Currently, there are no proven means of identifying presymptomatic women who subsequently develop complications of pregnancy during early pregnancy. Most women who are triaged into high-risk clinical units based on previous poor obstetric history ultimately have uncomplicated pregnancies. Available evidence supports the hypothesis that the aetiology of pregnancy complications begins during 1st trimester [15, 16]. If this is the case, profile of placenta-derived biomarkers during early pregnancy may be common between women with risk of developing pregnancy complications. Identification of such characteristics would provide opportunity to develop clinically useful early pregnancy screening tests.

Previously we have established that normal pregnancy is associated with the increase of exosomes into maternal plasma and the concentration of placenta-derived exosomes increases by 6-fold in uncomplicated healthy pregnancy during the first to third trimester [7] , however, the exosome profile in early pregnancy (i.e. from 6 to 12 weeks) remained to be established. The aim of this study was to characterise placenta-derived exosomes in maternal plasma over the first trimester of pregnancy and observe inter-subject variations in the exosome concentration. Weekly collected blood samples (from 6 to 12 weeks) were collected from normal healthy women to isolate and characterise the exosomes. The presence of exosomes were confirmed by: size (50–120 nM), and buoyant density (1.122- 1.197 g/ml). Endosomal (CD63) and placental (PLAP) antigens were identified in maternal plasma from as early as sixth week of pregnancy. The number of exosomes present in the maternal plasma increased progressively during the first trimester, as well as the exosomal PLAP concentration.

We isolated exosomes from the maternal plasma by differential and buoyant density centrifugation using a sucrose continuous gradient [7, 17]. The purification of exosomes from plasma and other biological fluids is not trivial, however, the use of an automatic system for fraction collection after the sucrose continuous gradient enable a high-reproducibility density, and decreasing the coefficient of variation between samples. In addition, using purification method based on the density of exosomes discards vesicles with the same size of exosomes with no endosomal origin, increasing the purity of exosome samples.

Previous studies have established that extracellular vesicles, including exosomes are released under physiological and pathophysiological conditions as well as during gestation [18]. The release of these vesicles is increased during pregnancy in response to different pathological conditions, presumably due to exosomal secretion from the placental trophoblast cells to the maternal peripheral circulation [19, 20] . In this study, we have established that exosomes are very stable when stored at −80°C. We obtained similar exosome yield from fresh and stored samples (i.e. plasma) and were able to identify gestational age differences in plasma exosome number in samples stored in long term. The isolation of exosomes from stored biofluids is the normal rather than the exception. These results are consistent with those of other studies [21, 22] suggesting that the exosomal content is protected inside these vesicles, highlighting the potential use of exosomes as biomarker for their high stability under different conditions.

As exosomes carry different kinds of protein, mRNA and miRNA [23], engaging in cell-to-cell communication, it is likely that they play an important role in modifying the maternal physiological state to maintain a successful pregnancy [24]. Interestingly, in this study we found that placental-derived exosomes increased systematically during the first trimester as early as sixth week of pregnancy when the intervillous circulation is not fully established. However, it has been observed that communication between placental and fetal circulation occurs at the beginning of the fourth week post conception [25]. Moreover, the lacunar spaces are formed in the trophoblast from as early as nine days post-ovulation and maternal blood flows into the trophoblast lacunae between ten and eleven days after fecundation. In addition, it has been reported that the intervillous blood flow is present in an early stage (i.e. < seventh week) [26] and increases gradually from fourth week during the first trimester of pregnancy [27].

Trophoblast plugs occlude the spiral arteries to prevent the contact of maternal blood flow into the intervillous space, however, at the same time trophoblast plug are in contact with the maternal blood, and could releases soluble proteins (e.g. human chorionic gonadotropin, hCG) and vesicles (e.g. nanovesicles) into maternal circulation. Interesting to highlight that hCG can be measured in maternal plasma as early as 4 weeks of gestation, confirming the presence of molecules released from the trophoblast in early pregnancy. Moreover, β-hCG and pregnancy-associated plasma protein A (PAPP-A) have been measured in maternal plasma as early as 6 weeks of gestation [28].

Specific placental-derived exosomes were quantified in the maternal circulation using the immunoreactive placental protein PLAP. Recent studies have demonstrated the presence of exosomes-PLAP+ive only in peripheral circulation of pregnant women [7, 29]. PLAP is an integral membrane protein (enzyme) unique to the placenta (it has also been observed in some gynaecologic cancers), produced mainly by syncytiotrophoblast [30, 31]. Nevertheless, PLAP expression has been found in primary trophoblast cytotrophoblast cells [7] and ED27 trophoblast-like cells, both isolated from first trimester chorionic villi, and also in JEG-3 cells (a extravillous trophoblast model) [32]. In addition, using immunohistochemistry stain for PLAP, the majority of chorionic trophoblastic cells were positive for PLAP [33]. During the first trimester of pregnancy, the release of placental exosomes into the maternal blood may result from extravillous trophoblast and/or syncytiotrophoblast cells; however, while a definitive answer awaits further investigation, it is of relevance to note that fetal cells are present in maternal blood from 4 weeks of pregnancy and that trophoblast cells invade the decidua and myometrium from the time of implantation. Thus, a cellular and exosomal pathway exists for delivery into the maternal circulation.

Recently, several attempts and techniques were undertaken to determine and characterize the exosomal content in different biological fluids including normal human blood plasma [3436]. As, the content of these released exosomes are placenta- specific [37], studying these nanovesicles is excellent method to understand the different processes occurring during embryo/fetal development and the feto-maternal interaction. Exosome analysis provides diagnostic and therapeutic potential, and biomarker opportunities for the early detection of diseases [3840]. To date, several research studies have been performed to identify the morphologic and proteomic characteristics of exosomes released from the placental extravilous trophoblast cells and expression profile of these exosomal contents relates to common pregnancy conditions [8, 41, 42]. However, all these studies considered the late second or third trimester of pregnancy plasma samples for analysis.

Conclusions

In conclusion, this study present longitudinal data on placental-derived exosomes in the first trimester of pregnancy, starting from as early as 6 weeks after implantation. Early detection of women at risk of complications of pregnancy would provide opportunity to evaluate appropriate intervention strategies to limit acute adverse squeal. The rationale for developing early pregnancy screening tests is not only for the management of the contemporaneous pregnancy but also to optimise lifelong and intergenerational health. If this can be achieved, it will provide an opportunity for early assignment of risk and the implementation of an alternative clinical management strategy to improve outcome for both the mother and baby.

Acknowledgements

We acknowledge the assistance of Dr. Jamie Riches and Dra. Rachel Hancock of the Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology (QUT) for the electron microscope analyses. This project was supported, in part by funding from Therapeutics Innovation Australia.

Footnotes

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

SS, KSR, and CS contributed in generating experimental data. CS, MDM and GER contributed in discussion and reviewed/edited manuscript. AP and SEI contributed obtaining clinical samples and management of patients. SS, CS and GER wrote the manuscript and drew the figures. All authors read and approved the final manuscript.

Contributor Information

Suchismita Sarker, Email: s.sarker@uq.edu.au.

Katherin Scholz-Romero, Email: k.scholz@uq.edu.au.

Alejandra Perez, Email: aleperezs@gmail.com.

Sebastian E Illanes, Email: sillanes@uandes.cl.

Murray D Mitchell, Email: murray.mitchell@uq.edu.au.

Gregory E Rice, Email: g.rice@uq.edu.au.

Carlos Salomon, Email: c.salomongallo@uq.edu.au.

References

  • 1.Liou JD, Pao CC, Hor JJ, Kao SM. Fetal cells in the maternal circulation during first trimester in pregnancies. Hum Genet. 1993;92:309–311. doi: 10.1007/BF00244479. [DOI] [PubMed] [Google Scholar]
  • 2.Wataganara T, Chen AY, LeShane ES, Sullivan LM, Borgatta L, Bianchi DW, Johnson KL. Cell-free fetal DNA levels in maternal plasma after elective first-trimester termination of pregnancy. Fertil Steril. 2004;81:638–644. doi: 10.1016/j.fertnstert.2003.07.028. [DOI] [PubMed] [Google Scholar]
  • 3.Salomon C, Kobayashi M, Ashman K, Sobrevia L, Mitchell MD, Rice GE. Hypoxia-induced changes in the bioactivity of cytotrophoblast-derived exosomes. PLoS One. 2013;8:e79636. doi: 10.1371/journal.pone.0079636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Salomon C, Ryan J, Sobrevia L, Kobayashi M, Ashman K, Mitchell M, Rice GE. Exosomal Signaling during Hypoxia Mediates Microvascular Endothelial Cell Migration and Vasculogenesis. PLoS One. 2013;8:e68451. doi: 10.1371/journal.pone.0068451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Tolosa JM, Schjenken JE, Clifton VL, Vargas A, Barbeau B, Lowry P, Maiti K, Smith R. The endogenous retroviral envelope protein syncytin-1 inhibits LPS/PHA-stimulated cytokine responses in human blood and is sorted into placental exosomes. Placenta. 2012;33:933–941. doi: 10.1016/j.placenta.2012.08.004. [DOI] [PubMed] [Google Scholar]
  • 6.Delorme-Axford E, Donker RB, Mouillet JF, Chu T, Bayer A, Ouyang Y, Wang T, Stolz DB, Sarkar SN, Morelli AE, Sadovsky Y, Coyne CB. Human placental trophoblasts confer viral resistance to recipient cells. Proc Natl Acad Sci U S A. 2013;110:12048–12053. doi: 10.1073/pnas.1304718110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Salomon C, Torres MJ, Kobayashi M, Scholz-Romero K, Sobrevia L, Dobierzewska A, Illanes SE, Mitchell MD, Rice GE. A gestational profile of placental exosomes in maternal plasma and their effects on endothelial cell migration. PLoS One. 2014;9:e98667. doi: 10.1371/journal.pone.0098667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Atay S, Gercel-Taylor C, Kesimer M, Taylor DD. Morphologic and proteomic characterization of exosomes released by cultured extravillous trophoblast cells. Exp Cell Res. 2011;317:1192–1202. doi: 10.1016/j.yexcr.2011.01.014. [DOI] [PubMed] [Google Scholar]
  • 9.Théry C, Zitvogel L, Amigorena S. Exosomes: Composition, biogenesis and function. Nat Rev Immunol. 2002;2:569–579. doi: 10.1038/nri855. [DOI] [PubMed] [Google Scholar]
  • 10.Sahoo S, Losordo DW. Exosomes and cardiac repair after myocardial infarction. Circ Res. 2014;114(2):333–344. doi: 10.1161/CIRCRESAHA.114.300639. [DOI] [PubMed] [Google Scholar]
  • 11.Aplin JD. Implantation, trophoblast differentiation and haemochorial placentation: mechanistic evidence in vivo and in vitro. J Cell Sci. 1991;99(Pt 4):681–692. doi: 10.1242/jcs.99.4.681. [DOI] [PubMed] [Google Scholar]
  • 12.Mincheva-Nilsson L. Placental exosome-mediated immune protection of the fetus: feeling groovy in a cloud of exosomes. Expert Rev Obstetrics Gynaecol. 2010;5:619–634. doi: 10.1586/eog.10.43. [DOI] [Google Scholar]
  • 13.Kshirsagar S, Alam S, Jasti S, Hodes H, Nauser T, Gilliam M, Billstrand C, Hunt J, Petroff M. Immunomodulatory molecules are released from the first trimester and term placenta via exosomes. Placenta. 2012;33:982–990. doi: 10.1016/j.placenta.2012.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kobayashi M, Salomon C, Tapia J, Illanes SE, Mitchell MD, Rice GE. Ovarian cancer cell invasiveness is associated with discordant exosomal sequestration of Let-7 miRNA and miR-200. J Transl Med. 2014;12:4. doi: 10.1186/1479-5876-12-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gagnon R. Placental insufficiency and its consequences. Eur J Obstet Gynecol Reprod Biol. 2003;110(Suppl 1):S99–S107. doi: 10.1016/S0301-2115(03)00179-9. [DOI] [PubMed] [Google Scholar]
  • 16.Masoura S, Kalogiannidis IA, Gitas G, Goutsioulis A, Koiou E, Athanasiadis A, Vavatsi N. Biomarkers in pre-eclampsia: a novel approach to early detection of the disease. J Obstet Gynaecol J Inst Obstet Gynaecol. 2012;32:609–616. doi: 10.3109/01443615.2012.709290. [DOI] [PubMed] [Google Scholar]
  • 17.Thery C, Amigorena S, Raposo G, Clayton A. Current protocols in cell biology / editorial board, Juan S Bonifacino [et al.] 2006. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. [DOI] [PubMed] [Google Scholar]
  • 18.Pap E, Pállinger E, Falus A, Kiss AA, Kittel A, Kovács P, Buzás EI. T Lymphocytes are Targets for Platelet- and Trophoblast-Derived Microvesicles During Pregnancy. Placenta. 2008;29:826–832. doi: 10.1016/j.placenta.2008.06.006. [DOI] [PubMed] [Google Scholar]
  • 19.Dragovic RA, Southcombe JH, Tannetta DS, Redman CW, Sargent IL. Multicolor flow cytometry and nanoparticle tracking analysis of extracellular vesicles in the plasma of normal pregnant and pre-eclamptic women. Biol Reprod. 2013;89:1–12. doi: 10.1095/biolreprod.113.113266. [DOI] [PubMed] [Google Scholar]
  • 20.Salomon C, Sobrevia L, Ashman K, Illanes S, Mitchell MD, Rice GE. The role of placental exosomes in gestational diabetes mellitus. In: Sobrevia L, editor. Gestational Diabetes-Causes, Diagnosis and Treatment. 2013. [Google Scholar]
  • 21.Ge Q, Zhou Y, Lu J, Bai Y, Xie X, Lu Z. miRNA in plasma exosome is stable under different storage conditions. Molecules. 2014;19:1568–1575. doi: 10.3390/molecules19021568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Taylor DD, Gercel-Taylor C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol. 2008;110:13–21. doi: 10.1016/j.ygyno.2008.04.033. [DOI] [PubMed] [Google Scholar]
  • 23.van der Pol E, Böing AN, Harrison P, Sturk A, Nieuwland R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol Rev. 2012;64:676–705. doi: 10.1124/pr.112.005983. [DOI] [PubMed] [Google Scholar]
  • 24.Mincheva-Nilsson L, Baranov V. The Role of Placental Exosomes in Reproduction. Am J Reprod Immunol. 2010;63:520–533. doi: 10.1111/j.1600-0897.2010.00822.x. [DOI] [PubMed] [Google Scholar]
  • 25.Jauniaux E, Jurkovic D, Campbell S. Current topic: in vivo investigation of the placental circulations by Doppler echography. Placenta. 1995;16:323–331. doi: 10.1016/0143-4004(95)90089-6. [DOI] [PubMed] [Google Scholar]
  • 26.Valentin L, Sladkevicius P, Laurini R, Soderberg H, Marsal K. Uteroplacental and luteal circulation in normal first-trimester pregnancies: Doppler ultrasonographic and morphologic study. Am J Obstet Gynecol. 1996;174:768–775. doi: 10.1016/S0002-9378(96)70462-3. [DOI] [PubMed] [Google Scholar]
  • 27.Merce LT, Barco MJ, Alcazar JL, Sabatel R, Troyano J. Intervillous and uteroplacental circulation in normal early pregnancy and early pregnancy loss assessed by 3-dimensional power Doppler angiography. Am J Obstet Gynecol. 2009;200:311–318. doi: 10.1016/j.ajog.2008.10.020. [DOI] [PubMed] [Google Scholar]
  • 28.Wortelboer EJ, Koster MP, Kuc S, Eijkemans MJ, Bilardo CM, Schielen PC, Visser GH. Longitudinal trends in fetoplacental biochemical markers, uterine artery pulsatility index and maternal blood pressure during the first trimester of pregnancy. Ultrasound Obstet Gynecol Off J Int Soc Ultrasound Obstet Gynecol. 2011;38:383–388. doi: 10.1002/uog.9029. [DOI] [PubMed] [Google Scholar]
  • 29.Sabapatha A, Gercel-Taylor C, Taylor DD. Specific isolation of placenta-derived exosomes from the circulation of pregnant women and their immunoregulatory consequences. Am J Reprod Immunol. 2006;56:345–355. doi: 10.1111/j.1600-0897.2006.00435.x. [DOI] [PubMed] [Google Scholar]
  • 30.Vongthavaravat V, Nurnberger MM, Balodimos N, Blanchette H, Koff RS. Isolated elevation of serum alkaline phosphatase level in an uncomplicated pregnancy: a case report. Am J Obstet Gynecol. 2000;183:505–506. doi: 10.1067/mob.2000.105841. [DOI] [PubMed] [Google Scholar]
  • 31.Leitner K, Szlauer R, Ellinger I, Ellinger A, Zimmer KP, Fuchs R. Placental alkaline phosphatase expression at the apical and basal plasma membrane in term villous trophoblasts. J Histochem Cytochem Off J Histochem Soc. 2001;49:1155–1164. doi: 10.1177/002215540104900909. [DOI] [PubMed] [Google Scholar]
  • 32.Kniss DA, Xie Y, Li Y, Kumar S, Linton EA, Cohen P, Fan-Havard P, Redman CW, Sargent IL. ED(27) trophoblast-like cells isolated from first-trimester chorionic villi are genetically identical to HeLa cells yet exhibit a distinct phenotype. Placenta. 2002;23:32–43. doi: 10.1053/plac.2001.0749. [DOI] [PubMed] [Google Scholar]
  • 33.Bashiri A, Katz O, Maor E, Sheiner E, Pack I, Mazor M. Positive placental staining for alkaline phosphatase corresponding with extreme elevation of serum alkaline phosphatase during pregnancy. Arch Gynecol Obstet. 2007;275:211–214. doi: 10.1007/s00404-006-0212-5. [DOI] [PubMed] [Google Scholar]
  • 34.Hina K, Christopher GA, Michael L, Ching Seng A, Adam M, Richard JS, Mark DH, Suresh M. Comparative proteomics evaluation of plasma exosome isolation techniques and assessment of the stability of exosomes in normal human blood plasma. PROTEOMICS. 2013;13(22):3354–3364. doi: 10.1002/pmic.201300282. [DOI] [PubMed] [Google Scholar]
  • 35.Gallo A, Tandon M, Alevizos I, Illei GG. The Majority of MicroRNAs Detectable in Serum and Saliva Is Concentrated in Exosomes. PLoS One. 2012;7:e30679. doi: 10.1371/journal.pone.0030679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tauro BJ, Greening DW, Mathias RA, Ji H, Mathivanan S, Scott AM, Simpson RJ. Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods. 2012;56:293–304. doi: 10.1016/j.ymeth.2012.01.002. [DOI] [PubMed] [Google Scholar]
  • 37.Luo S-S, Ishibashi O, Ishikawa G, Ishikawa T, Katayama A, Mishima T, Takizawa T, Shigihara T, Goto T, Izumi A, Ohkuchi A, Matsubara S, Takeshita T, Takizawa T. Human Villous Trophoblasts Express and Secrete Placenta-Specific MicroRNAs into Maternal Circulation via Exosomes. Biol Reprod. 2009;81:717–729. doi: 10.1095/biolreprod.108.075481. [DOI] [PubMed] [Google Scholar]
  • 38.Simpson RJ, Lim JW, Moritz RL, Mathivanan S. Exosomes: proteomic insights and diagnostic potential. Expert Rev Proteomics. 2009;6:267–283. doi: 10.1586/epr.09.17. [DOI] [PubMed] [Google Scholar]
  • 39.Pant S, Hilton H, Burczynski ME. The multifaceted exosome: Biogenesis, role in normal and aberrant cellular function, and frontiers for pharmacological and biomarker opportunities. Biochem Pharmacol. 2012;83:1484–1494. doi: 10.1016/j.bcp.2011.12.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Michelle EM, Joshua NL. FedExosomes: Engineering Therapeutic Biological Nanoparticles that Truly Deliver. Pharmaceuticals. 2013;6:659–680. doi: 10.3390/ph6050659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Donker RB, Mouillet JF, Chu T, Hubel CA, Stolz DB, Morelli AE, Sadovsky Y. The expression profile of C19MC microRNAs in primary human trophoblast cells and exosomes. Mol Hum Reprod. 2012;18:417–424. doi: 10.1093/molehr/gas013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Tannetta DS, Dragovic RA, Gardiner C, Redman CW, Sargent IL. Characterisation of Syncytiotrophoblast Vesicles in Normal Pregnancy and Pre-Eclampsia: Expression of Flt-1 and Endoglin. PLoS One. 2013;8(56764):1–12. doi: 10.1371/journal.pone.0056754. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Translational Medicine are provided here courtesy of BMC

RESOURCES