Open Access

Placental proteome in late‑onset of fetal growth restriction

  • Authors:
    • Tomasz Gęca
    • Aleksandra Stupak
    • Robert Nawrot
    • Anna Goździcka‑Józefiak
    • Anna Kwaśniewska
    • Wojciech Kwaśniewski
  • View Affiliations

  • Published online on: October 14, 2022     https://doi.org/10.3892/mmr.2022.12872
  • Article Number: 356
  • Copyright: © Gęca et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Fetal growth restriction (FGR) occurs when the fetus does not reach its genetically programmed intrauterine potential for growth and affects ~5‑10% of pregnancies. This condition is one of the leading causes of perinatal mortality and morbidity associated with obstetric and neonatal complications. Placental dysfunction in FGR causes an impairment in the transfer of nutrients and oxygen from the mother to the developing fetus. Maternal adaptations to placental insufficiency may also play a role in the pathophysiology of FGR. The present study aimed to compare the proteome of the placentas of 18 women with the physiological course of pregnancy and eutrophic fetus [estimated fetal weight (EFW) >10th percentile; control group] and 18 women with late FGR (EFW <10th percentile) diagnosed after 32 weeks of pregnancy, according to the Delphi consensus (study group). The U. Mann‑Whitney test was used to compare two independent groups. The R. Spearman correlation coefficient significance test was used to assess the existence of a relationship between the analyzed measurable parameters. P<0.05 was considered to indicate a statistically significant difference. The tests showed the presence of 356 different proteins which were responsible for the regulation of gene transcription control, inhibiting the activity of proteolytic enzymes, regulation of trophoblast proliferation and angiogenesis and inflammatory response. In the FGR placental proteome, other detected proteins were mostly involved in response to oxidative stress, cellular oxidation and detoxication, apoptosis, hemostatic and catabolic processes, energy transduction protein interactions, cell proliferation, differentiation and intracellular signaling. The present study used chromatographic mass‑spectrometry to compare the placental proteome profiles in pregnancies complicated by late‑onset FGR and normal pregnancy. Comparative analysis of proteomes from normal and FGR placentas showed significant differences. Further research is needed to clarify maternal and fetal adaptations to FGR.

Introduction

Pregnancy is a condition that requires numerous adaptive changes to the mother, fetus and placenta. Adaptation processes at the level of cell physiology and individual systems occurring in the course of physiological and complicated pregnancy are extremely complicated. The physiological changes that occur in pregnancy are mainly caused by the placenta. However, the way in which the maternal tissues respond to the new demands of pregnancy development is hypothesized to influence the success of the pregnancy as well (1).

The proteome defines all the proteins within a cell or a tissue. The metabolome describes metabolites including small molecules, peptides, carbohydrates, lipids, nucleosides and catabolic product. They are connected by numerous aspects of cell signaling, protein degradation and generation and post-translational modification (2). In comparison to the non-pregnant state, the first trimester of pregnancy is characterized by systemic adaptation of the mother (2). How these adaptive processes are reflected in the maternal metabolome/proteome is not well characterized.

The principal findings of the study by Handelman et al (3) were that pregnant women and non-pregnant women differ in the abundance of 44% of the profiled plasma metabolites. This finding was explained by the inhibition of specific metabolic processes producing small molecules, the activation of catabolic processes consuming small molecules or expanding blood volume, leading to dilution. The metabolite differences and associated perturbed pathways reflected the physiological changes occurring in the first 16 weeks of normal pregnancy. Finally, metabolites like blood lysolipids and dipeptides were reported to change as a consequence of advancing gestation.

During the first two trimesters of pregnancy, there is a build-up of lipids in the mother's tissues. Circulating maternal metabolic products, such as triglycerides, cholesterol, free fatty acids and phospholipids, are intended to meet the energy needs of the fetus and ensure an adequate supply of mother's milk after delivery. This period is seen as an ‘anabolic phase’ characterized by an increase in maternal fat accumulation and a progressive decrease in fasting glucose levels in as the gestational age increases, which is associated with a 10% reduction in insulin sensitivity compared to the pre-pregnancy period (46). Although fasting glucose is lowered, hepatic glucose production (via gluconeogenesis and glycogenolysis) is increased, leading to an increase in fasting insulin. As a consequence, decreased maternal liver sensitivity to insulin leads to increased hepatic glucose production (4). During the third trimester of pregnancy, the maternal metabolic state is characterized by a ‘catabolic phase’ in which peritoneal insulin sensitivity is further reduced and fat storage in the peritoneum and subcutaneous tissue is impaired, which is a source of calories for the mother and fetus (4,5). Insulin resistance in pregnant women increases significantly with gestational age in normal pregnancy to ensure adequate glucose transfer to the fetus and maternal inositols positively correlate with crown-rump length (CRL) (7). They are correlated with insulin sensitivity and may be mechanistically related to glucose homeostasis (8). These events demonstrate active changes in energy requirements during pregnancy. Other age-related carbohydrate metabolic changes include placental polyol pathways which are very active in the first trimester of pregnancy (9). One hypothesis regarding elevated levels of polyols in early pregnancy is that they are an early source of carbohydrates for the placenta and embryo. In addition, the polyol pathway may facilitate the re-oxidation of pyridine nucleotides under low oxygen conditions, helping to regulate intracellular pH during periods of high glycolysis (10).

The placenta serves a key role in regulating the metabolic environment in pregnancy. The human placenta is adapted from the initial hypoxic environment in the first trimester to increased oxygenation in the second trimester of pregnancy when the spiral arteries remodel (11,12). The oxygen concentration in the interstitial junctions increases from 2–3% in the 8th week of pregnancy to 8.5% in the 12th week of pregnancy (13). These changes are accompanied by increased oxidative stress and, as a consequence, an increase in placental antioxidant factors maintaining redox homeostasis. These changes are particularly evident in the metabolism of hexadecanoic acid, erythritol and 2-deoxyribose (14). Placental cholesterol has also been found to be elevated in correlation with CRL. Higher cholesterol levels may be the result of increased levels of progestogen hormones. Maternal cholesterol is a precursor to both progesterone and estrogen (10).

Placental dysfunction is the main culprit in fetal growth restriction (FGR) causing an impairment in the transfer of nutrients and oxygen from the mother to the developing fetus. Maternal adaptations to placental insufficiency may also play a role in the pathophysiology of FGR (15).

FGR affects ~5–10% of pregnancies and is the leading cause of perinatal mortality and morbidity (16). This condition occurs due to placental dysfunction when the fetus does not reach its genetically programmed intrauterine potential for growth. FGR is associated with obstetric and neonatal complications and the development of cardiometabolic diseases at an older age (17,18). The etiology of FGR is complex and related to fetus (genetic and chromosomal abnormalities and congenital metabolic disorders), placenta (impaired placentation and implantation) and pregnant woman (placental dysfunction of vascular origin) (16,18). Risk factors of developing FGR include: Smoking, hypertension, severe chronic anemia, pregestational diabetes mellitus, autoimmune diseases, congenital malformations and infections, chromosomal abnormalities. unfortunately, the presence of these risk factors is only found in 30% of cases (19).

In clinical practice FGR must be distinguished from small for gestational age (SGA) fetuses, which represent constitutionally smaller fetuses. In contrast to SGA fetuses, FGR is pathological condition associated with insufficiency of placenta and less supply of oxygen and nutrients to the developing fetus. The current standard of recognition FGR is based on ultrasound examinations. FGR can be divided as early or late onset according to gestational age at onset/recognition (32 weeks). Early-onset FGR is associated with gestational hypertension and/or pre-eclampsia in ≤70% and represents ~20–30% of cases of FGR whereas late-onset FGR represents 70–80% of all FGR and to a lesser extent is associated with the development of hypertensive disorders of pregnancy (20). According to the DELPHI consensus early-onset FGR is diagnosed before 32 weeks of gestation when estimated fetal weight (EFW) or abdominal circumference(AC) is <3rd centile or there is absent end-diastolic flow in the umbilical artery (UA) or AC or EFW is <10th centile combined with a pulsatility index (PI) >95th centile in either the UA or uterine artery (21,22). For late FGR (≥32 weeks), the following criteria must be fulfilled: AC or EFW <3rd centile and four contributory parameters (EFW or AC <10th) centile, AC or EFW crossing centiles by > two quartiles on growth charts and cerebroplacental ratio <5th centile or UA-PI >95th centile (21,22).

There is no effective antenatal therapy for FGR. Hence, delivery of the newborn remains the only option to avoid stillbirth. When this occurs preterm, the further risk of morbidity and mortality is introduced (23,24). In current clinical practice for monitoring the growth restricted fetus cardiotocography and Doppler sonography are used. In the management of late-onset FGR the cerebroplacental ratio (CPR) is important because it not only allows a more precise diagnosis of late-onset perinatal outcomes but also helps to predict unfavorable perinatal outcomes (2527). The two subtypes of FGR show different pathogenic and clinical features. Defective placentation, due to a poor trophoblastic invasion of the maternal spiral arteries, is hypothesized to play a central role in the pathogenesis of early-onset preeclampsia and FGR (28).

One of the promising diagnostic methods is comparative proteomics based on the analysis of the protein profile in normal and abnormal tissues (29). Reviews have highlighted the proteomic approaches that have been used to explore pre-eclampsia (PE), FGR and preterm birth (3034).

The aim of the present study was to compare the proteome of the placentas in women with fetal growth restriction and with a physiological course of pregnancy.

Materials and methods

Patients between 32–36 weeks' gestation with singleton pregnancy with late onset FGR who were hospitalized in the Department of Obstetrics and Pathology of Pregnancy at the Medical University of Lublin between 2019 and 2021 were recruited for the present study. The criteria for excluding patients from the analysis were: Multiple pregnancy, presence of any antenatal infections, positive TORCH test result, treatment with antibiotics during pregnancy, any form of hypertension in pregnancy, pre-pregnancy and gestational diabetes, nephropathy, thyroid dysfunction and any other general diseases before pregnancy, using any drugs or stimulants, nicotinism and fetuses with birth defects and chromosomal abnormalities.

After obtaining informed written consent, 36 pregnant women who delivered by cesarean section were involved in the study, including 18 women with physiological pregnancy and eutrophic fetus (EFW >10th percentile; control group) and 18 women with late FGR (EFW <10th percentile; study group) diagnosed after 32 weeks of pregnancy, according to Delphi consensus (21). The age range of the control group was 18–38 years whereas the age range of the study group was 19–37 years old. To estimate the weight of the fetus during an ultrasound scan, a regression equation was used considering the biparietal diameter, the length of the femur and the head and abdominal circumferences, as proposed by Hadlock et al (35). Doppler measurements of the umbilical artery free loop were obtained within 1 week of delivery using a Voluson E9 with RA4B 3D 4–8 MHz curvilinear probe (Cytiva). The PI, RI and CPR were then calculated. PI=(S-D)/A and RI=(S-D)/S, where S is the systolic peak, D is the end diastolic flow and A is the temporal average frequency. Whereas cerebroplacental ratio (CPR) is the ratio between the PI of middle cerebral and umbilical artery (PI MCA/PI UA) and reflect the distribution of cardiac output in favor of cerebral blood flow. It is one of the parameters that has the best accuracy in predicting perinatal outcomes (36). In response to intrauterine hypoxia, redistribution of fetal blood flow to the brain occurs and the value of CPR decreases <1. In cases of late-onset FGR, the tolerance to hypoxia is lower than in early-onset FGR (37).

Clinical information on mothers was obtained from standardized medical records and patient interviews, including smoking, age, weight and body mass index (BMI) at the start of the first trimester, pregnancy weight gain and TORCH. BMI was calculated as body weight (kg)/height (m)2. The following data on infants were also obtained from medical records: Gestational age at delivery, sex and birth weight of the newborn, placental weight, body length, head circumference and neonatal complications. Gestational age was determined based on the date of the last menstruation and the first-trimester ultrasound scan (based on CRL). Birth weight and placenta weight, body length and head circumference were measured immediately after birth using appropriate measuring tools.

The material for proteomic research was fragments of normal placentas constituting control and fragments of placentas collected from women diagnosed with FGR. All samples were collected by trained personnel as follows: In aseptic conditions, during the cesarean section, immediately after the delivery of the child, the placenta was placed in sterile containers containing ice. Specimen collection personnel were wearing a sterile protective apron, face masks and sterile gloves to ensure sterility throughout the sampling process. The placentae were weighed and collected. A total of four placenta biopsies of 1.0×1.0×1.5 cm from each placenta was obtained. The collection site was ~3-4 cm from the umbilical cord insertion site peripherally from four different placental quadrants. Only sections from the inner part of the placenta were collected for examination to eliminate possible infections during cesarean section (risk of contamination). Sections from 18 placentae from women with impaired fetal growth and 18 control placenta were qualified for the study. Each placenta sample was placed in a sterile, labeled cryovial, then frozen in liquid nitrogen and stored at −80°C until DNA extraction and further analysis.

Written informed consent was obtained from all subjects included and the study was performed in accordance with the principles of the Helsinki Declaration The research was issued by the Bioethics Committee at the Medical University of Lublin (approval no. KE-0254/87/2020). Derived data supporting the findings of this study are available from the corresponding author on request.

Identification of proteins
Protein isolation

Each test was performed in duplicate. Proteins isolated from the tested material and control (18 samples FGR and 18 control) were analyzed in a polyacrylamide (PAA) gel, then pooled and analyzed using mass spectrometry (MS). The obtained results were searched using the Mascot algorithm (MASCOT 2.4.1; http://www.matrixscience.com/) against the Uniprot database 2019_02 (559228 sequences; 200905869 residues, http://www.uniprot.org/) with a filter searching for human proteins. In order to prepare samples for further analyses, protein isolation was performed. 100 mg of each tissue was crushed in a mortar with liquid nitrogen and subsequently 500 µl of isolation buffer (0.1% Tris-Cl, 10% glycerol) was added and gently mixed. The mixture was then transferred into 1.5 ml Eppendorf tubes and centrifuged at 10,000 × g for 10 min at 4°C. Finally, the supernatant was transferred to new tubes and frozen at −20°C for further study.

SDS-PAGE analysis

In order to verify the protein composition of protein samples, sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) was conducted in a slab mini-gel apparatus according to Laemmli (38) using 10% polyacrylamide as the separating gel and 5% polyacrylamide as the stacking gel. The proteins were denatured by heating them to 100°C in the presence of 2-mercaptoethanol for 5 min. Then, 50 µg of each sample was put into the gel (two technical replicates). After electrophoresis the resulting gels were fixed and stained using sensitive Coomassie Blue Staining (0.02% Coomassie Brilliant Blue G-250, 12 h staining at room temperature) (39). Separating gel at 10% was chosen in order to separate the whole protein spectrum of a sample, including proteins with high as well as low molecular weighs.

MS analysis

Stained protein bands were excised from the gel and analyzed by liquid chromatography (LC) coupled to mass spectrometer in the Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland. Tryptic peptide mixtures were analyzed by LC-ESI-MS/MS using nanoflow HPLC and the LTQ-Orbitrap XL (Thermo Fisher Scientific, Inc.) as mass analyzer with two technical replicates. Spectrometer parameters were as follows: capillary voltage: 2.5 kV, cone: 40 V, N2 gas flow: 0 and m/z range 300–2,000, with positive ionization mode. Excised gel fragments were placed in 1.5 ml Eppendorf tubes filled with 10% methanol and 2% acetic acid. The proteins were digested using trypsin. The generated peptides were concentrated, desalted on an RP-C18 precolumn (LC Packings) and further separated by UltiMate nano-HPLC (LC Packings) using water containing 0.1% TFA as a mobile phase with a linear acetonitrile gradient (10–30%) over 50 min with the flow rate of 150 nl/min. The column was directly coupled to a nanospray ion source operating in a data-dependent MS to MS/MS switch mode. Proteins were identified by tandem mass spectrometry (MS/MS) via information-dependent acquisition of fragmentation spectra of multiple-charged peptides.

Protein identification algorithm

The spectral data were analyzed by MASCOT 2.4.1 (Matrix Science; www.matrixscience.com) and searched against the Uniprot 2019_02 (559228 sequences; 200905869 residues) database with Homo sapiens (human; 20492 sequences) filter. Mascot search criteria were as follows: protein scores >31 indicated identity or extensive homology (P<0.05); with carbamidomethyl (C) and oxidation (M) variable modifications; peptide mass tolerance ±50 ppm and fragment mass tolerance ±0.8 Da. Protein identifications were accepted when at least two peptide fragments per protein were identified.

Quantitative analysis

To achieve a non-label quantitative comparison of proteins between analyzed samples, the Exponentially Modified Protein Abundance Index (emPAI) was employed (40). The number of peptides per protein normalized by the theoretical number of peptides is called the protein abundance index (PAI). To determine protein abundance from the nano-LC-MS/MS experiments a modified form of PAI is used: the exponential form of PAI minus one (emPAI=10PAI-1). The emPAI value is proportional to protein abundance in a protein mixture. Resultant protein and peptide lists were saved in Excel files (Microsoft Corporation).

Statistical analysis

The obtained results were analyzed statistically. The values of the analyzed parameters measured on the nominal or ordinal scale were characterized by the number and percentage, while those measured on the interval scale by the arithmetic mean, standard deviation, median, 25 and 75th percentiles and the range of variation. Due to the skewed distribution of the measured parameters assessed using the W. Shapiro-Wilk test or the heterogeneity of variance assessed using the F-Fischer test, non-parametric tests were used to analyze the existence of differences between the studied subgroups. The U. Mann-Whitney test was used to compare two independent groups. The Spearman correlation coefficient significance test was used to assess the existence of a relationship between the analyzed measurable parameters. Statistical analyzes were performed based on the Statistica v. 10.0 software (StatSoft). P<0.05 was considered to indicate a statistically significant difference.

Results

The results of the study were obtained from 18 placentas taken from women with fetal growth disorders and from 18 control placentas. The clinical characteristics along with anthropometric measurements of mothers and their newborns are presented in Table I.

Table I.

Clinical characteristics and anthropometric measurements of mothers and newborns.

Table I.

Clinical characteristics and anthropometric measurements of mothers and newborns.

VariablesControl group n=Study group (FGR) n=P-value
Baseline characteristics
  Age (years)30.2±6.528.2±5.60.466
  Height (m)   1.7±0.06   1.67±0.080.373
  Actual Weight (kg)80.3±11.379.5±70.854
  Weight before the pregnancy (kg)63.6±11.366.7±6.30.458
  BMI before the pregnancy (kg/m2)   22±3.523.9±2.10.154
  Weight gain (kg)14 (1228)12.5 (1115)0.032
  Weight of the placenta (kg)515±46328±53<0.001
  Parity2 (14)  1 (13)0.504
  Gestation2 (14)1.5 (14)0.699
Perinatal outcomes
  Gestational age at the delivery (weeks)39 (3841)37 (35.4-40)0.002
  Fetal weight at birth (g)3,540 (2,910–3,890)2,300 (1,385–2,570)<0.001
  Neonatal length (cm)  54 (4757)  48 (3551)0.001
  APGAR 1 min (points)  9 (810)8 (69)0.002
  APGAR 5 min (points)10 (910)10 (610)0.597
Feto-placental Doppler before delivery
  UA PI0.77 (0.72-0.91)1.11 (0.98-1.9)<0.001
  MCA PI1.44±0.211.31±0.220.191
  UtA PI0.79±0.050.93±0.170.025
  CPR1.703 (1.48-2.444)0.995 (0.737-1.687)<0.001

[i] Values are shown as median [interquartile range]; statistical analysis was performed using a Mann-Whitney U test. Variables following a normal distribution are reported as mean ± standard deviation, statistical test for them was the standard t-test. BMI, body mass index; PI, pulsatility index; UA, umbilical artery; MCA, middle cerebral artery; UtA, uterine artery; CPR, cerebro-placental ratio; APGAR, Appearance, Pulse, Grimace, Activity, Respirations.

There were no statistically significant differences between the study groups in terms of age, height, fertility, BMI before pregnancy, body weight before pregnancy and that measured at delivery. The control group was characterized by statistically significantly higher weight gain in pregnancy compared to the study group (P=0.032) and differences in placenta weight (P=0.00000005). As a result of the ultrasound analysis of vascular flows using the color Doppler technique, a statistically significantly higher mean pulsation index (PI) in the gestational uterine arteries was found in the study group compared to the control group (P=0.025), as well as a higher PI in the artery umbilical cord of fetuses with FGR compared to eutrophic ones (P=0.0001). The cerebro-placental ratio (CPR) was statistically significantly higher in the control group compared to the study group (P=0.0005). Pregnant women in the test group gave birth statistically significantly earlier than in the control group (P=0.002). Newborns with FGR were characterized by lower birth weight (P=0.0001) and shorter body length (P=0.001) as well as lower Apgar score in the first minute of life (P=0.002) compared to neonates from the control group.

The tests performed showed the presence of 356 different proteins. From this group, those proteins that were identified in comparable amounts in both samples, both in the test and control groups, were eliminated. Only those proteins that were not present in the control group were analyzed. Proteins were ranked according to their emPAI, which is proportional to the protein content of the given sample. Tables II and III show the results of the studies for control placenta (Table II) and placenta from women with FGR (Table III).

Table II.

Human placental proteins identified by LC-ESI-MS/MS from control samples.

Table II.

Human placental proteins identified by LC-ESI-MS/MS from control samples.

AccessionaIdentified proteinbScorecMolecular mass (Da)dMatched PeptideseSequence coverage (%)fExponentially Modified Protein Abundance Indexg
Q5QNW6Histone H2B type 2-F OS=Homo sapiens OX=9606 GN=HIST2H2BF PE=1 SV=32,17913,9125053.218.58
P01009Alpha-1-antitrypsin OS=Homo sapiens OX=9606 GN=SERPINA1 PE=1 SV=33,14146,8458167.715.21
P07355Annexin A2 OS=Homo sapiens OX=9606 GN=ANXA2 PE=1 SV=21,97238,7644272.314.05
P08758Annexin A5 OS=Homo sapiens OX=9606 GN=ANXA5 PE=1 SV=21,94235,9604174.113.66
P68032Actin, alpha cardiac muscle 1 OS=Homo sapiens OX=9606 GN=ACTC1 PE=1 SV=12,59142,268845712.27
P62736Actin, aortic smooth muscle OS=Homo sapiens OX=9606 GN=ACTA2 PE=1 SV=12,45642,304735712.27
P06576ATP synthase subunit beta, mitochondrial OS=Homo sapiens OX=9606 GN=ATP5F1B PE=1 SV=33,38456,5256364.89.88
P0DML2Chorionic somatomammotropin hormone 1 OS=Homo sapiens OX=9606 GN=CSH1 PE=1 SV=11,94825,2344364.19.19
P04083Annexin A1 OS=Homo sapiens OX=9606 GN=ANXA1 PE=1 SV=22,48938,8744460.77.7
O43707Alpha-actinin-4 OS=Homo sapiens OX=9606 GN=ACTN4 PE=1 SV=23,227105,1568561.76.78
P18206Vinculin OS=Homo sapiens OX=9606 GN=VCL PE=1 SV=42,047124,1825748.23.33
Q05707Collagen alpha-1(XIV) chain OS=Homo sapiens OX=9606 GN=COL14A1 PE=1 SV=32,929194,2687938.52.32
P35579Myosin-9 OS=Homo sapiens OX=9606 GN=MYH9 PE=1 SV=42,649227,4036232.91.79
P21333Filamin-A OS=Homo sapiens OX=9606 GN=FLNA PE=1 SV=42,658282,7716729.41.21
Q9Y490Talin-1 OS=Homo sapiens OX=9606 GN=TLN1 PE=1 SV=32,191271,3475630.31.05
Q09666Neuroblast differentiation-associated protein AHNAK OS=Homo sapiens OX=9606 GN=AHNAK PE=1 SV=22,362629,1137414.70.52

a Database accession numbers according to: Uniprot 2019_02 (559228 sequences; 200905869 residues) database with Homo sapiens (human) (20492 sequences) filter.

b Identified homologous proteins.

c Mascot Search Probability Based Mowse Score. Ions score is-10×Log(P), where P is the probability that the observed match is a random event. Protein scores >31 indicate identity or extensive homology (P<0.05).

d Theoretical mass (Da) of identified proteins. The values were retrieved from the protein database.

e Number of matched peptides with Mascot search data (www.matrixscience.com).

f Amino acid sequence coverage for the identified proteins.

g Exponentially modified protein abundance index of identified protein according to Mascot Search data.

Table III.

Human placental proteins identified by liquid chromatography electrospray ionization tandem mass spectrometry from the group of fetal growth restriction cases.

Table III.

Human placental proteins identified by liquid chromatography electrospray ionization tandem mass spectrometry from the group of fetal growth restriction cases.

AccessionaIdentified proteinbScorecMolecular mass (Da)dMatched PeptideseSequence coverage (%)fExponentially Modified Protein Abundance Indexg
P02647Apolipoprotein A-I OS=Homo sapiens OX=9606 GN=APOA1 PE=1 SV=148630,7591433.33.48
P32119Peroxiredoxin-2 OS=Homo sapiens OX=9606 GN=PRDX2 PE=1 SV=530022,016725.82.12
P6310414-3-3 protein zeta/delta OS=Homo sapiens OX=9606 GN=YWHAZ PE=1 SV=123727,866725.31.86
P01009Alpha-1-antitrypsin OS=Homo sapiens OX=9606 GN=SERPINA1 PE=1 SV=349746,8451223.91.46
P60174Triosephosphate isomerase OS=Homo sapiens OX=9606 GN=TPI1 PE=1 SV=336331,002920.61.25
P07195L-lactate dehydrogenase B chain OS=Homo sapiens OX=9606 GN=LDHB PE=1 SV=224436,845515.30.77
P04040Catalase OS=Homo sapiens OX=9606 GN=CAT PE=1 SV=346859,9031016.90.76
P02790Hemopexin OS=Homo sapiens OX=9606 GN=HPX PE=1 SV=235452,2411511.30.76
P27797Calreticulin OS=Homo sapiens OX=9606 GN=CALR PE=1 SV=140148,250920.40.69
P07237Protein disulfide-isomerase OS=Homo sapiens OX=9606 GN=P4HB PE=1 SV=331657,403716.90.67
P26038Moesin OS=Homo sapiens OX=9606 GN=MSN PE=1 SV=348767,8701110.70.64
P14618Pyruvate kinase PKM OS=Homo sapiens OX=9606 GN=PKM PE=1 SV=424858,36078.90.43
P29401Transketolase OS=Homo sapiens OX=9606 GN=TKT PE=1 SV=327268,38777.70.36
O43707Alpha-actinin-4 OS=Homo sapiens OX=9606 GN=ACTN4 PE=1 SV=2253105,15687.20.33
P00450Ceruloplasmin OS=Homo sapiens OX=9606 GN=CP PE=1 SV=1344122,81776.70.19
Q9BVA1Tubulin beta-2B chain OS=Homo sapiens OX=9606 GN=TUBB2B PE=1 SV=123850,28947.20.18

a Database accession numbers according to: Uniprot 2019_02 (559228 sequences; 200905869 residues) database with Homo sapiens (human) (20492 sequences) filter.

b Identified homologous proteins.

c Mascot Search Probability Based Mowse Score. Ions score is-10×Log(P), where P is the probability that the observed match is a random event. Protein scores >31 indicate identity or extensive homology (P<0.05).

d Theoretical mass (Da) of identified proteins. The values were retrieved from the protein database.

e Number of matched peptides with Mascot search data (www.matrixscience.com).

f Amino acid sequence coverage for the identified proteins.

g Exponentially modified protein abundance index of identified protein according to Mascot Search data.

The function of the proteins identified in the control sample is described in Table IV and those in the test sample in Table V.

Table IV.

Functions of proteins identified in control placenta.

Table IV.

Functions of proteins identified in control placenta.

First author, yearProteinsFunctions(Refs.)
Arimura Y, 2018Histone H2B type 2-FCore component of nucleosome serves a central role in transcription regulation, DNA repair, DNA replication and chromosomal stability(44)
Pater D, 2021 Alpha-1-antitrypsin α1-antitrypsin is a protein belonging to the serpin superfamily(45)
Xi Y, 2020Annexin A2Placental anticoagulant protein IV(46)
Monceau V, 2004Annexin A5Is a cellular protein with presumed function of plasma membrane repair and hemostasis(123)
Li A, 2021Actin, alpha cardiac muscle 1The alpha actins are found in muscle tissues and are a major constituent of the contractile apparatus.(124)
Yuan SM, 2018Actin, aortic smooth muscleα-2 actin is found in smooth muscle cells, a family of globular multi-functional proteins that form microfilaments(125)
Jonckheere A, 2012ATP synthase subunit beta, mitochondrial Mitochondrial membrane ATP synthase [F(1)F(0) ATP synthase or Complex V] produces ATP from ADP in the presence of a proton gradient across the membrane(126)
Männik J, 2012Chorionic somatomammotropin hormone 1Produced only during pregnancy and is involved in stimulating lactation, fetal growth and metabolism.(127)
D'Acquisto F, 2008Annexin A1Plays important roles in the innate immune response as effector of glucocorticoid-mediated responses and regulator of the inflammatory process.(47)
Peng W, 2021 Alpha-actinin-4Alpha actinin is an actin-binding protein with multiple roles in different cell types.(48)
Bays JL, 2017VinculinVinculin is a cytoplasmic actin-binding protein enriched in focal adhesions and adherens junctions that is essential for embryonic development.(128)
Patino MG, 2002Collagen alpha-1(XIV) chainIt likely serves a role in collagen binding and cell-cell adhesion.(129)
Sudo H, 2013Neuroblast differentiation-associated proteinProtein may play a role in such diverse processes as blood-brain barrier formation, cell structure and migration, cardiac calcium channel regulation and tumor metastasis(130)
Sun H, 2020Myosin-9Serves a role in cytokinesis, cell shape and specialized functions such as secretion and capping. During cell spreading, serves an important role in cytoskeleton reorganization, focal contact formation(131)
Su W, 2012Filamin AFilamins play multiple cellular roles, serving as organizers of cell structure (e.g., cytoskeleton) and function, regulating cell signaling, transcription, cell adhesion, focal adhesion assembly, cell apoptosis and organ development.(132)
Burrudge K, 2018TalinProbably involved in connections of major cytoskeletal structures to the plasma membrane.(133)

Table V.

Functions of proteins identified in fetal growth restriction.

Table V.

Functions of proteins identified in fetal growth restriction.

First author, yearProteinsFunctions(Refs.)
Mangaraj M, 2016Apolipoprotein A-IComponent and a major structural protein of high-density lipoprotein, serves a vital role in reverse cholesterol transport and cellular cholesterol homeostasis. Its multifunctional role in immunity, inflammation, apoptosis, viral, bacterial infection(49)
Duan T, 2016 Peroxiredoxin-2Thiol-specific peroxidase that catalyzes the reduction of hydrogen peroxide and organic hydroperoxides to water and alcohols, respectively.(134)
Pennington KL, 201814-3-3 protein zeta/deltaAdapter protein implicated in the regulation of a large spectrum of both general and specialized signaling pathways. Binds to a large number of partners, usually by recognition of a phosphoserine or phosphothreonine motif. Binding generally results in the modulation of the activity of the binding partner.(50)
Stockley RA, 2015 Alpha-1-antitrypsinProtein produced in the liver that protects the body's tissues from being damaged by infection-fighting agents released by its immune system(135)
Wierenga RK, 2010Triosephosphate isomerase (TIP)Enzyme which very fast interconverts dihydroxyacetone phosphate and D:-glyceraldehyde-3-phosphate.(51)
Chen Y, 2019L-lactate dehydrogenase B chainLDH catalyzes the conversion of lactate to pyruvate and back, as it converts NAD+ to NADH and back; transfers hydride from one molecule to another.(88)
Fu W, 2014CatalaseCatalyzes the reaction by which hydrogen peroxide is decomposed to water and oxygen.(52)
Poillerat V, 2020HemopexinIs the plasma protein with the highest binding affinity to heme. It is mainly expressed in liver and belongs to acute phase reactants, the synthesis of which is induced after inflammation.(53)
Varricchio L, 2017CalreticulinIs a chaperone protein which resides primarily in the endoplasmic reticulum and is involved in a variety of cellular processes, among them, cell adhesion. Additionally, it functions in protein folding quality control and calcium homeostasis. Calreticulin is also found in the nucleus, suggesting that it may have a role in transcription regulation.(54)
Khan HA, 2014Protein disulfide-isomeraseIs a prototypic thiol isomerase that catalyzes the formation and cleavage of thiol-disulfide bonds during protein folding in the endoplasmic reticulum (ER. PDI is induced during endoplasmic reticulum (ER) stress and it serves as a vital cellular defense against general protein misfolding via its chaperone activity. It is also responsible for the isomerization, formation and rearrangement of protein disulfide bonds(55)
Karvar S, 2020MoesinIs particularly important in immunity acting on both T and B-cells homeostasis and self-tolerance, regulating lymphocyte egress from lymphoid organs. Ezrin-radixin-moesin (ERM) family protein that connects the actin cytoskeleton to the plasma membrane and thereby regulates the structure and function of specific domains of the cell cortex.(56)
Sizemore ST, 2018Pyruvate kinase PKMCatalyse the conversion of phosphoenol-pyruvate (PEP) to pyruvate at the final step of glycolysis.(57)
Wilkinson HC, 2020TransketolaseIs an important enzyme in the non-oxidative branch of the pentose phosphate pathway (PPP), a pathway responsible for generating reducing equivalents, which is essential for energy transduction and for generating ribose for nucleic acid synthesis. Transketolase also links the PPP to glycolysis, allowing a cell to adapt to a variety of energy needs, depending on its environment(58)
Feng D, 2018 Alpha-actinin-4Is a widely expressed homodimeric protein that bundles and cross-links filamentous actin.(136)
Zanardi A, 2018CeruloplasminBinds to copper and carries it throughout your body. If you have low ceruloplasmin, it can point to a genetic condition called Wilson disease, a copper deficiency or other medical conditions(137)
Lehmann SG, 2017Tubulin beta-2B chainIs the major constituent of microtubules. It binds two moles of GTP, one at an exchangeable site on the beta chain and one at a non-exchangeable site on the alpha chain (By similarity). TUBB2B is implicated in neuronal migration.(138)

Discussion

The development of the human placenta is complex and not well described. For the study of placental changes such methods as cell culture technologies, advanced imaging techniques, omics technologies, biomarkers and proteomics are used (4143).

The results of the present study with the use of a comparative analysis of proteomes from normal and FGR placentas showed in the cells of the normal placenta the presence of proteins responsible for the regulation of gene transcription control (histon, annexin) and proteins inhibiting the activity of proteolytic enzymes (alpha 1-antitrypsin), the deficiency of which may lead to oxidative stress and damage to the placenta. This inhibitor also serves an important role in the regulation of trophoblast proliferation and angiogenesis. In the normal placenta, the present study also identified actins that serve an important role in determination of cell proliferation and invasion, (mainly F-actin), rearrangement of the action cytoskeleton, regulation membrane dynamics and inflammatory response (annexin) (4448). In the FGR placental proteome other detected proteins are mostly involved in response to stress (peroxiredoxin-2), cellular oxidation and detoxication (catalase), apoptosis (apolipoprotein), hemostatic and catabolic processes, energy transduction (pyruvate kinase, transketolase), protein folding (calreticulin, protein disulfide-isomerase) and interactions (protein disulfide-isomerase 14-3-3 protein zeta/delta), immunity (moesin) and inflammation (hemopexin). In FGR the expression of peroxiredoxin-2 antioxidant protein was also observed, the role of which is to neutralize reactive oxygen species (ROS) and regulate multiple cellular functions such as cell proliferation, differentiation and intracellular signaling. ROS accumulation is also limited by catalase (present in FGR) which inactivates H2O2. The 14-3-3 protein zeta/delta present in FGR integrates and controls multiple signaling pathways. The detected triosephosphate isomerase (TIP) during gluconeogenesis, provides the two substrates required for aldolase to generate fructose-1,6-bisphosphate, which is converted to fructose-6-phosphate and glucose-6-phosphate, important precursors for cell wall components and nucleic acids. Lactate dehydrogenase enzyme, which is important in maintaining a high level of pyruvate and is highly expressed in tissues with high-energy demands, was also present in the study group (4958).

The results of the present study showed that the highest emPAI coefficient, which proves the proportionally high protein content in the FGR samples, concerns apolipoprotein 1A (Apo1A). Apo1A is one of the proteins responsible primarily for cholesterol transport, but it also has anti-inflammatory properties by influencing lipid peroxidation and the immune system. The anti-inflammatory effect of Apo 1A is based on the inhibition of the migration in the endothelium of cells of the immune system as a result of a decrease in the expression of integrins, inhibition of the activation of monocytes and the synthesis of cytokines (49). The presence of these apolipoproteins in the FGR placentas suggested disturbed cholesterol homeostasis and inflammation processes. This will result in abnormal lipids metabolisms, vascular damage, disturbances in the function of cellular molecules and incorrect folding of proteins, which would in turn be indicative of the presence of chaperones (e.g., 14-3-3 protein zeta/delta). In the studied (FGR) group no structural proteins were found in opposition to the group of proteins from the normal placentas.

The abnormal lipid metabolism during pregnancy with intrauterine growth restriction of the SGA type was also noted by Bernard et al (59). Another study shows that in the antenatal period, the ratio of fatty acids in the mother's blood serum compared to its concentration in the newborn decreases in FGR, which suggests increased energy and metabolic demand of the fetus (60).

Statistically significant differences in the synthesis of cholesterol in fetuses with FGR compared to eutrophic ones were also shown, e.g., the concentration of cholesterol during pregnancy in fetuses with FGR was slightly increased (2.48 times), while in the serum of correctly developing fetuses the concentration of cholesterol increased 6.54 times (61). Also in the urine of newborns from pregnancies with FGR, an increased level of myo-inositol is found, which correlates with the negative regulation of the release of free fatty acids from adipose tissue (62). In turn, the results of Bahado-Singh et al in addition to identifying potential FGR biomarkers, provides information on the dysregulation of placental biochemistry in FGR (41). The univariate analysis of metabolites showed a global decrease in phosphatidylcholine in FGR. The decrease in lipid metabolites can be explained by the decreased level of placental energy substrates as a result of hypoxia leading to significant changes in lipid metabolism, as pointed out by Raff et al (63).

Paules et al analyzed gene ontology and revealed the pathways and biological processes involved in late-onset FGR which were mostly related to the efflux of cholesterol and phospholipids (33). The lipoproteins Apolipoprotein C2, Apolipoprotein C3 and Apolipoprotein E with parallel pathway of LXR/RXR activation, are fundamental in the balance of cholesterol levels and known protective function against dysregulated fetoplacental lipid homeostasis. Those lipoproteins are also involved in atherosclerosis and IL-12 signaling in inflammation, lipid dysregulation and endothelial cell dysfunction. The existing oxidative stress, inflammation and placental thrombosis interrupt the placental ability to transfer nutrients and oxygen to the fetus and therefore implicate the normal fetal growth.

In contrast to the present study, Bahado-Singh et al (41) describe 3-hydroxybutyric acid as the most important metabolite for distinguishing between FGR and control placental tissue. 3-hydroxybutyric acid is synthesized by the liver and is a source of energy for the brain when glucose levels are low. Moreover, it is the end product of fatty acid oxidation by ketogenesis and a substrate for lipid synthesis in the biological cascade of ketolysis. Placental trihydroxybutyric acid deficiency could potentially explain the low lipid levels.

Research to date suggests that both the placenta and vascular endothelium of a pregnant woman are tissues that use oxygen to produce energy through mitochondrial-mediated oxidative phosphorylation. This highly energetic process is also supported by the formation of ROS that regulate intracellular signaling and tissue adaptation (64,65). ROS are more and more often recognized as signaling molecules that regulate physiological processes, while oxidative stress is a state that disrupts signaling pathways in the cell (65,66).

The present study implied that there was a general disruption of fetal energy substrates and metabolism in the FGR. This was evidenced by the high result obtained for peroxiredoxin-2 (Prxs). It belongs to the family of antioxidant proteins involved in the fight against free radicals. This protein causes the production of inflammatory cytokines. In addition, it performs several other functions related to the regulation of cell proliferation, differentiation and protection against oxidative stress (67,68). During inflammation, high levels of peroxides are produced by phagocytes and the cytoprotective antioxidant role of Prxs in inflammation cannot be overestimated (67,68). Peroxides also serve to regulate inflammatory signaling pathways and Prxs are known to be a critical modulator of signaling peroxides (69). Hemopexin is also associated with antioxidant stress. Hemopexin is an intracellular glycoprotein responsible for maintaining blood homeostasis by regulating free heme, which eliminates the harmful pro-oxidative and pro-infusion potential of heme (70). Hpx serves a neuroprotective role after ischemic injuries. According to Li et al (71) local expression of Hpx by neurons contributes to protection against free heme through induction of HO isoenzyme, or HO1. The upregulation of HO1 in ischemic astrocytes and fibroblasts has a protective effect against ischemia by reducing oxidation, stress and apoptosis. In Hpx−/− mice following ischemic stroke, there is evidence of increased oxidative damage, infarct volume and general neurological deficiency (71,72).

Calreticulin (CARL) in physiological, normal cells acts as a chaperone to help protein fold properly in the endoplasmic reticulum. CALR supports Ca2+ dependent processes such as adhesion and signaling integrin and ensures correct antigen presentation on MHC class I molecules as well as participation in Ca2+ transport, an essential component for placental and fetal development (54). During physiological pregnancy, 30 g Ca2+ migrates from mother to fetus across the placenta to facilitate the development of the fetal skeletal system (73). It has been shown that CALR, as a molecular chaperone of the placenta, is necessary for the proper development of the trophoblast and placenta (7476). However, extracellular CALR release is unusual and, since CALR is a stress response protein, stress may be involved in extracellular CALR release (77). Studies have shown an increase in CALR mRNA and protein levels in maternal blood and placenta of patients with pre-eclampsia (78,79). Iwahashi et al (80) provide evidence that induction of stress in the endoplasmic reticulum leads to the extracellular release of CALR, which may contribute to placental dysfunction by inhibiting cytotrophoblast syncytialysis. In addition, tumor cells undergo immunogenic cell death (ICD) by exposing CALRs on their surface, which promotes the uptake of tumor cells by phagocytes and ultimately supports the initiation of anti-tumor immunity. In this way, loss of function CALR mutations promotes oncogenesis not only because they disrupt cell homeostasis in healthy cells, but also because they threaten natural and therapy-controlled immune surveillance (80).

Protein disulfide-isomerase (PDIA3) is a chaperone that modulates the folding of newly synthesized glycoproteins, exhibits isomerase and redox activity and is involved in the pathogenesis of numerous diseases (81). However, the role of PDIA3 in pregnancy-related diseases remains to be elucidated. Mo et al (81) reveal a key role of PDIA3 in the biology of placental trophoblasts in women with PE. Immunohistochemistry and western blot analysis showed that PDIA3 expression was decreased in villi trophoblasts from women with PE compared to pregnancies with normal blood pressure. Furthermore, using the Cell Counting Kit-8 assay, flow cytometry and 5-ethynyl-2′-deoxyuridine (EdU) staining, it was found that siRNA-mediated PDIA3 knockdown significantly promotes apoptosis and inhibits proliferation in the HTR8/SVneo cell line, while overexpression PDIA3 reversed these effects. In addition, RNA sequencing and western blot analysis showed that PDIA3 knockdown inhibited MDM2 protein expression in HTR8 cells, concomitantly with a marked increase in p53 and p21 expression. Conversely, PDIA3 overexpression had the opposite effect. Moreover, immunohistochemistry and western blotting showed that MDM2 protein expression was decreased and p21 was increased in the trophoblasts of women with PE compared to women with pregnancies with normal blood pressure. PDIA3 expression is decreased in the trophoblasts of women with PE and decreased PDIA3 induces trophoblast apoptosis and inhibits trophoblast proliferation by regulating the MDM2/p53/p21 pathway (81).

In FGR, the 14-3-3 zeta/delta proteins were also identified described as specific proteins of the brain tissue; their first described function was to activate the synthesis of neurotransmitters (82).

Currently, ~200 different cellular proteins have been identified as binding partners for 14-3-3 proteins and they are involved in almost every cellular process, including signal transduction, cell cycle control, apoptosis, transcription regulation, cytoskeleton rearrangements, cell adhesion, chromosome maintenance, protein localization, protein transport, protein degradation, exocytosis, endocytosis, development and stress response (83,84). The 14-3-3 proteins play a key role in subcellular localization. Injured central nervous system (CNS) neurons, unfortunately, have a poor ability to regenerate spontaneously, resulting in permanent functional deficits following hypoxic injury (84). Kaplan et al (85) show that the 14-3-3 adapters are central proteins that are attractive targets for manipulating cell signaling. Researchers demonstrate a positive role for 14-3-3s in axon growth and regulation of phosphorylation and function of 14-3-3s. They showed that fusicoccin-A (FC-A), a small molecule stabilizer of protein-protein interactions 14-3-3, stimulates axon growth in vitro and regeneration in vivo (85).

Moesin (MSN) is a member of the ezrin-radixin-moesin (ERM) family of proteins, which binds plasma membrane proteins to actin fibers in the cell cortex and is essential for vascular endothelial function. They are found in cell surface structures such as microvilli, filopodia, beauty, wrinkle membranes, retraction fibers and cell adhesion sites where actin fibers are associated with plasma membranes. Ezrin-radixin-binding protein-50-kDa (EBP50) is a protein that serves an important role in cancer development. Embryo and tumor growth are similar. Embryo implantation is a key process for a successful pregnancy although the mechanism of embryo implantation is not fully understood. Lipopolysaccharides can stimulate endothelial cells to secrete MSN (86). Additionally, MSN is required for induced endothelial cell hyperpermeability and inflammatory responses and high levels of MSN in the blood are detected in mice and human patients with sepsis (87). Hence, MSN is involved in the pathogenesis of sepsis and MSN may be a potential biomarker for assessing the severity of endothelial damage during sepsis (88). MSN deficiency in mice significantly affects lymphocyte homeostasis; the number of NK cells in peripheral blood and bone marrow decreases, but it increases in the spleen. MSN-deficient NK cells show increased cell death and impaired signaling in response to IL-15, suggesting that MSN regulates NK cell survival through IL-15-mediated signaling. It can therefore be that MSN can be regarded as a regulator of NK cell homeostasis in vivo (89).

Noteworthy is the presence of such proteins in FGR as lactate dehydrogenase (LDH). LDH intracellular enzyme is important in energy production in nearly all cells in the body. Its highest concentrations are in the heart, liver, muscles, kidneys and lungs. The total concentration of lactate dehydrogenase is made up of five different enzyme variants (isoenzymes), which are produced by different tissues. Only a small fraction of LDH can be found in the blood because the enzyme is released into it when cells die or damage. Therefore, serum lactate dehydrogenase is a nonspecific marker of tissue damage in the body, while an increase in placenta with FGR is indicative of placental insufficiency (88).

Pyruvate kinase (PKM) serves a key role in regulating cellular metabolism. The conversion of phosphoenolpyruvate (PEP) to pyruvate, which is catalyzed by pyruvate kinase, is the final rate-limiting step in glycolysis. There are four isomeric, tissue-specific forms of pyruvate kinase found in mammals: PKL, PKR, PKM1 and PKM2. PKM1a and PKM2 are formed by a single mRNA transcript of the PKM gene by alternative inclusion. The PKM2 dimer regulates the rate of the glycolysis step that shifts glucose metabolism from the normal respiratory chain to lactate production in cancer cells. In addition to being a regulator of metabolism, it also acts as a protein kinase that contributes to oncogenesis. It is mainly described in neoplastic processes (57). Hasan et al (90 demonstrate the expression of PKM2 in normoxic states (20% O2) and hypoxic states (0.1% O2) in two prostate cancer cell lines, PC3 and LNCaP. The authors show that hypoxia significantly increases the expression of PKM2 mRNA in both cell lines (46). This suggests that under hypoxic conditions, PKM2 expression is further promoted by HIF-1α activation (90). Tumor angiogenesis is initiated by PKM2 dimer in the blood, thereby increasing endothelial cell proliferation, migration and cell-ECM adhesion, leading to tumor growth (91,92). Most important is the activation of IGF-IR, a PKM2-mediated tumor angiogenesis event, by disrupting the NF-κB/miR-148a/152t feedback loop, promoting tumor growth and angiogenesis (93). Under hypoxia, IGF-1/IGF-IR mediates the interaction of HIF-1α with the NF-κB p65/RelA subunit and the PKM2 promoter and PKM2 expression is also enhanced by the repression of miR-148a and miR-152 (94). The binding leads to nuclear translocation of PKM2, where it acts as a protein kinase and interacts with other molecules to control the expression of VEGF, thus promoting tumor angiogenesis.

Transketolase (TKT) in the non-oxidative branch of the pentose phosphate pathway (PPP); it regulates the level of ribose-5-phosphate (R5P) and de novo nucleotide biosynthesis (95). Maintaining genome integrity is essential because genomic information regulates cell proliferation, growth arrest and important metabolic processes in cells (96). Genomes are constantly exposed to endogenous and environmental DNA-damaging agents such as oxidizing agents, nitrosamines and polycyclic aromatic hydrocarbons. Altered cellular metabolism, which is intertwined with DNA damage and repair pathways, leads to genomic instability, while accumulation of genome instability results in metabolic abnormalities (97,98). A number of different metabolic pathways are involved in de novo synthesis of nucleotides (99,100). R5P, an intermediate product of PPP, is an important precursor in the biosynthesis of both DNA and RNA. PPP is one of the branches of glycolysis and serves a key role in meeting the cellular requirements of biosynthesis and antioxidant defense (101). Moreover, PPP is essential for repairing double-strand breaks after DNA damage has occurred in mammalian cells (101). TKT and transaldolase are the two major enzymes that mediate reversible reactions in non-oxidative PPP. The main purpose of PPP is the production of R5P and NADPH. R5P is the major backbone of RNA and is critical for nucleotide synthesis. NADPH is the major antioxidant that keeps the two major redox molecules, glutathione and thioredoxin, in a reduced state. Thus, NADPH counteracts ROS, allowing cells to survive oxidative stress.

Alpha-actinin (ACTN) members maintain the structures of the cytoskeleton and modulate cell mobility (102). Among the four members of the ACTN family in humans, ACTN2 and ACTN3 are specific to muscle cells while ACTN1 and ACTN4 are ubiquitous (103,104). ACTN4 is present at the leading edge of moving cells, suggesting that ACTN4 may be involved in cell migration (103,104). Moreover, ACTN4 signaling connects integrins with the actin cytoskeleton and enhances the invasion of trophoblasts in the placenta (105). ACTN4 deficiency dramatically reduces the proliferation and invasion of various neoplastic cells (106). Accumulating evidence strongly suggests that ACTN4 may be involved in trophoblast proliferation and invasion. However, the true functions of ACTN4 in trophoblast and placenta development remain to be elucidated.

Peng et al (48) suggest that ACTN4 expression is essential for normal trophoblast proliferation and differentiation in early pregnancy. Downregulation of ACTN4 may result in insufficient proliferation, invasion and migration of the trophoblast via the AKT/GSK3β/Snail pathway which may lead to pre-eclampsia. Proper development of the placenta and its component, pedigree in the early stages, is crucial for a successful pregnancy. Dysregulation of extravillous trophoblasts (EVT) disrupts the normal invasion of trophoblasts into the uterus, which in turn leads to incomplete remodeling of the spiral arteries and placental hypoperfusion (107). In Peng et al (48), ACTN4 was mainly expressed in cytotrophoblasts (CTBs) and EVT of the normal placenta but was barely detected in these cells from placental severe pre-eclampsia. Decreased ACTN4 levels reduce villi, trophoblast proliferation and ex vivo explants overgrowth. Moreover, the deficiency of ACTN4 results in significant inhibition of cell invasion and motility. Such attenuated proliferation, invasion and migration are a result of ACTN4 mediated by inactivation of the AKT/GSK3β/Snail pathway. CTBs are the so-called placental epithelial stem cells that, depending on the received signals, can maintain a balance between their differentiation into both ST and EVT. In addition, isolated CTBs without proliferation capacity may spontaneously differentiate into STBs after 24-h cultivation, suggesting that the self-renewal potential of CTBs is necessary to maintain proliferation and differentiation capacity (48).

Ceruloplazmin is an acute-phase protein, both pro-oxidative and antioxidant, synthesized by hepatocytes and involved in angiogenesis, coagulation and nitric oxide (NO) homeostasis (108,109). The main role of ceruloplasmin in the turnover of iron is the oxidation of Fe2+ to Fe3+, a process necessary for the binding of iron to transferrin (the main iron transporting protein) and ferritin (the main iron storing protein) (109). Increased serum levels of ceruloplasmin have been associated with an increased risk of cardiovascular disorders and serve as a predictor of adverse clinical outcomes in patients with acute coronary syndromes or myocardial infarction (110,111). Significantly higher concentrations of ceruloplasmin are found in patients with pre-eclampsia in whom the placental expression of ceruloplasmin, most likely derived from syncytiotrophoblasts, is high (112). Bellos et al (113) show that serum ceruloplasmin may be a useful screening and control tool for assessing pregnant women with a history of developing preeclampsia.

In 2022, the work of Surekh et al (114) was published, assessing the effect of maternal iron deficiency anemia (IDA) in 200 pregnant women on the expression of the iron transporter, although not ceruloplasmin but cyclopen in the term placenta. Placental cyclopen expression was investigated by mRNA analysis and protein immunohistochemistry. The cyclopene mRNA and the protein expression in the placenta showed a statistically significant increase with increasing the severity of anemia. The immunohistochemical expression of the cyclopen protein showed a statistically significant increase with the increase in the severity of the anemia. Similarly, placental cyclopen mRNA expression was higher in anemic mothers compared to non-anemic mothers. Surekh et al (114) showed for the first time a marked increase in cyclopene expression at both protein and mRNA levels in the term placenta in maternal IDA. This study helped determine how placental iron transport proteins could be regulated in response to maternal iron status and newborns and broadened our knowledge on the relationship between the iron state in the mother and the newborn and the mechanisms of modifying the placental iron transport concerning these parameters.

A prospective cohort analysis of 107 single pregnancies who underwent amniocentesis at 16–22 weeks according to standard genetic indications showed glucose, alkaline phosphatase (ALP), LDH, ceruloplasmin, ferritin, highly sensitive C-reactive protein and IL-6 in the mother's blood and amniotic fluid (115). The median concentration of ferritin in the amniotic fluid and IL-6 and the mean concentration of ALP in the amniotic fluid were higher in the group of premature infants, but this difference did not reach statistical significance. The maternal mean levels of ALP and LDH were slightly higher. Only the median maternal ferritin concentration in the intrauterine growth restriction group was higher than in the patients corresponding to the gestational age (P=0.03). In conclusion, low levels of glucose in the amniotic fluid are associated with the risk of preterm labor, while high levels of ferritin in the mother's blood increase the risk of FGR.

A similar study investigated the levels of zinc, copper, iron and magnesium ions and certain binding proteins in the amniotic fluid under FGR conditions (116). FGR showed a decrease in the content of zinc, iron and magnesium ions and an increase in the content of copper in the amniotic fluid in the second and third trimesters of pregnancy. In these trimesters, levels of ceruloplasmin, ferritin and Ca2+, Mg2+ and ATPase were lower in FGR, while levels of zinc-a-2-glycoprotein were higher than during the same periods of normal pregnancy. Changes in the parameters tested in the amniotic fluid were associated with disturbances in the development of newborns.

Tubulin beta-2B chain (TUBB2B) is a cytoskeleton component that serves a key role in CNS corticogenesis, mediating mitosis and cell translocation and the formation of synaptic connections (117,118). Heterozygous de novo missense variants in tubulin genes are associated with a heterogeneous group of disorders characterized by cortical malformations cerebral dysplasia, is known as ‘tubulinopathies’. Affected patients most often show a range of neurodevelopmental disorders, including cognitive and motor impairment, abnormal muscle tone and epilepsy (119121).

Alpha-1 antitrypsin (AAT) is a serum protein synthesized in the liver and secreted into the blood. AAT deficiency is associated with various clinical symptoms of the neonatal period (45). The most common symptom is ‘neonatal hepatitis syndrome’, occasionally referred to as ‘neonatal cholestasis’ or ‘cholestatic hepatitis’. The increase in antitrypsin-1 in FGR indicates the activation of compensatory mechanisms that protect the fetus.

Proteomic analysis in iron-treated mice compared to control mice showed 66 differentially expressed hippocampal proteins (30 upregulated and 36 downregulated) (122). Bioinformatics analysis showed that the deregulated proteins included, but was not limited to, mitochondrial-associated proteins (e.g., ADP/ATP translocator 1 and zeta/delta 14-3-3 protein) cytoskeleton proteins (TUBB2B and tubulin alpha-4A chain). Research suggests that dysregulation of synaptic, mitochondrial and cytoskeleton proteins may be involved in memory impairment induced by molecular mechanisms of iron neurotoxicity (122). In the future, it would be worthwhile to perform validation of the detected proteomic differences.

The limitations of the present study included increased risk of false positive results due to relatively small sample size and large number of tested targets that might cause random detection of statistical significance when margin for alpha error is set as <0.05).

Current screening tools used in pregnancy e.g., clinical factors, ultrasound scan and placental biomarkers are unable to identify the risk of growth impairment of fetuses. Comparative analysis of proteomes from normal and FGR placentas show significant differences. The changes detected in the FGR placenta proteome are complex and mainly concern proteins involved in the stress response, cellular oxidation and detoxification, apoptosis, catabolic processes, energy transduction and inflammation. The data from the present study about proteome and late FGR presents notable findings for understanding the disease pathophysiology. Elucidating proteomic changes in the placenta may help to uncover the underlying mechanism of the FGR and identify novel targets for therapeutics. Future work in validating these proteomic differences may also enable identification of early diagnosis for FGR. Additionally, the proteomics results can serve as a screening tool where proteins identified as significantly changed will need to be confirmed by traditional validation methods (for example western blotting).

Acknowledgements

Not applicable.

Funding

The present study was supported by Medical University of Lublin (grant no. DS 128).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

TG participated in conceptualization, data curation, formal analysis and writing the draft. AS conceived and designed the study, collected data, visualization and writing the original draft. RN participated in formal analysis, investigation, methodology. AGJ performed experiments and writing the original draft. AK participated in analysis and interpretation of data, data curation, funding acquisition, supervision and original writing. WK analyzed and interpretation of data, the project administration, supervision and review and editing. TG, AS confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Bioethics Committee at the Medical University of Lublin (approval no. KE-0254/87/2020). Written informed consent was obtained from all subjects included.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Zhang S, Lin H, Kong S, Wang S, Wang H, Wang H and Armant DR: Physiological and molecular determinants of embryo implantation. Mol Aspects Med. 34:939–980. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Zhan X, Long Y and Lu M: Exploration of variations in proteome and metabolome for predictive diagnostics and personalized treatment algorithms: Innovative approach and examples for potential clinical application. J Proteomics. 188:30–40. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Handelman SK, Romero R, Tarca AL, Pacora P, Ingram B, Maymon E, Chaiworapongsa T, Hassan SS and Erez O: The plasma metabolome of women in early pregnancy differs from that of non-pregnant women. PLoS One. 14:e02246822019. View Article : Google Scholar : PubMed/NCBI

4 

Lain KY and Catalano PM: Metabolic changes in pregnancy. Clin Obstet Gynecol. 50:938–948. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Zeng Z, Liu F and Li S: Metabolic adaptations in pregnancy: A review. Ann Nutr Metab. 70:59–65. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Herrera E and Desoye G: Maternal and fetal lipid metabolism under normal and gestational diabetic conditions. Horm Mol Biol Clin Investig. 26:109–127. 2016.PubMed/NCBI

7 

McLachlan KA, O'Neal D, Jenkins A and Alford FP: Do adiponectin, TNFα, leptin and CRP relate to insulin resistance in pregnancy? Studies in women with and without gestational diabetes, during and after pregnancy. Diabet Metab Res Rev. 22:131–138. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Catalano PM, Roman-Drago NM, Amini SB and Sims EAH: Longitudinal changes in body composition and energy balance in lean women with normal and abnormal glucose tolerance during pregnancy. Am J Obstetr Gynecol. 179:156. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Jauniaux E, Hempstock J, Teng C, Battaglia FC and Burton GJ: Polyol concentrations in the fluid compartments of the human concentrations in the fluid compartments of the human conceptus during the first trimester of pregnancy: Maintenance of redox potential in a low oxygen environment. J Clin Endocrinol Metab. 90:1171–1175. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Murgia F, Iuculano A, Peddes C, Santoru ML, Tronci L, Deiana M, Atzori L and Monni G: Metabolic fingerprinting of chorionic villous samples in normal pregnancy and chromosomal disorders. Prenat Diagn. 39:848–858. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Jauniaux E, Cindrova-Davies T, Johns J, Dunster C, Hempstock J, Kelly FJ and Burton GJ: Distribution and transfer pathways of antioxidant molecules inside the first trimester human gestational sac. J Clin Endocrinol Metab. 89:1452–1458. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Burton GJ, Watson AL, Hempstock J, Skepper JN and Jauniaux E: Uterine glands provide histiotrophic nutrition for the human fetus during the first trimester of pregnancy. J Clin Endocrinol Metab. 87:2954–2959. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Jauniaux E, Watson AL, Hempstock J, Bao YP, Skepper JN and Burton GJ: Onset of maternal arterial blood flow and placental oxidative stress: A possible factor in human early pregnancy failure. Am J Pathol. 157:2111–2122. 2000. View Article : Google Scholar : PubMed/NCBI

14 

Burton GJ and Jauniaux E: Placental oxidative stress: From miscarriage to preeclampsia. Reprod Sci. 11:342–352. 2004.

15 

King VJ, Bennet L, Stone PR, Clark A, Gunn AJ and Dhillon SK: Fetal growth restriction and stillbirth: Biomarkers for identifying at risk fetuses. Front Physiol. 13:9597502022. View Article : Google Scholar : PubMed/NCBI

16 

Nardozza LM, Caetano AC, Zamarian AC, Mazzola JB, Silva CP, Marçal VM, Lobo TF, Peixoto AB and Araujo Júnior E: Fetal growth restriction: Current knowledge. Arch Gynecol Obstet. 295:1061–1077. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Visentin S, Grumolato F, Nardelli GB, Di Camillo B, Grisan E and Cosmi E: Early origins of adult disease: Low birth weight and vascular remodeling. Atherosclerosis. 237:391–399. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Yzydorczyk C, Armengaud JB, Peyter AC, Chehade H, Cachat F, Juvet C, Siddeek B, Simoncini S, Sabatier F, Dignat-George F, et al: Endothelial dysfunction in individuals born after fetal growth restriction: Cardiovascular and renal consequences and preventive approaches. J Dev Orig Health Dis. 8:448–464. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Kalanithi LE, Illuzzi JL, Nossov VB, Frisbaek Y, Abdel-Razeq S, Copel JA and Norwitz ER: Intrauterine growth restriction and placental location. J Ultrasound Med. 26:1481–1489. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Unterscheider J, Daly S, Geary MP, Kennelly MM, McAuliffe FM, O'Donoghue K, Hunter A, Morrison JJ, Burke G, Dicker P, et al: Optimizing the definition of intrauterine growth restriction: The multicenter prospective PORTO Study. Am J Obstet Gynecol. 208:290.e1–e6. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Gordijn SJ, Beune IM, Thilaganathan B, Papageorghiou A, Baschat AA, Baker PN, Silver RM, Wynia K and Ganzevoort W: Consensus definition of fetal growth restriction: A Delphi procedure. Ultrasound Obstet Gynecol. 48:333–339. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Baschat AA: Late onset FGR is generally linked with milder placental insufficiency than early-onset FGR, but the risk of stillbirth is high due to wors fetal hemodynamic adaptation. Planning management and delivery of the growth-restricted fetus. Best Pract Res Clin Obstet Gynaecol. 49:53–65. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Zeitlin J, Ancel PY, Saurel-Cubizolles MJ and Papiernik E: The relationship between intrauterine growth restriction and preterm delivery: An empirical approach using data from a European case-control study. BJOG. 107:750–758. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Parker SE and Werler MM: Epidemiology of ischemic placental disease: A focus on preterm gestations. Semin Perinatol. 38:133–138. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Figueras F and Gratacós E: Update on the diagnosis and classification of fetal growth restriction and proposal of a stage-based management protocol. Fetal Diagn Ther. 36:86–98. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Monteith C, Flood K, Pinnamaneni R, Levine TA, Alderdice FA, Unterscheider J, McAuliffe FM, Dicker P, Tully EC, Malone FD and Foran A: An abnormal cerebroplacental ratio (CPR) is predictive of early childhood delayed neurodevelopment in the setting of fetal growth restriction. Am J Obstet Gynecol. 221:273.e1–273.e9. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Khalil A, Morales-Roselló J, Townsend R, Morlando M, Papageorghiou A, Bhide A and Thilaganathan B: Value of third-trimester cerebroplacental ratio and uterine artery Doppler indices as predictors of stillbirth and perinatal loss. Ultrasound Obstet Gynecol. 47:74–80. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Mecacci F, Avagliano L, Lisi S, Clemenza S, Serena C, Vannuccini S, Rambaldi MP, Simeone S, Ottanelli S and Petragli F: Fetal growth restriction: Does an integrated maternal hemodynamic-placental model fit better? Rep Sci. 28:2422–2435. 2021. View Article : Google Scholar : PubMed/NCBI

29 

Leite DFB, Morillon AC, Melo Júnior EF, Souza RT, McCarthy FP, Khashan A, Baker P, Kenny LC and Cecatti JG: Examining the predictive accuracy of metabolomics for small-for-gestational-age babies: A systematic review. BMJ Open. 9:e0312382019. View Article : Google Scholar : PubMed/NCBI

30 

Law KP, Han TL, Tong C and Baker PN: Mass spectrometry-based proteomics for pre-eclampsia and preterm birth. Int J Mol Sci. 16:10952–10985. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Nguyen TPH, Patrick CJ, Parry LJ and Familari M: Using proteomics to advance the search for potential biomarkers for preeclampsia: A systematic review and meta-analysis. PLoS One. 14:e02146712019. View Article : Google Scholar : PubMed/NCBI

32 

Dahabiyeh LA: The discovery of protein biomarkers in pre-eclampsia: The promising role of mass spectrometry. Biomarkers. 23:609–621. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Paules C, Youssef L, Miranda J, Crovetto F, Estanyol JM, Fernandez G, Crispi F and Gratacós E: Maternal proteomic profiling reveals alterations in lipid metabolism in late-onset fetal growth restriction. Sci Rep. 10:210332020. View Article : Google Scholar : PubMed/NCBI

34 

Conrad MS, Gardner ML, Miguel C, Freitas MA, Rood KM and Ma'ayeh M: Proteomic analysis of the umbilical cord in fetal growth restriction and preeclampsia. PLoS One. 17:e02620412022. View Article : Google Scholar : PubMed/NCBI

35 

Hadlock FP, Harrist RB, Sharman RS, Deter RL and Park SK: Estimation of fetal weight with the use of head, body and femur measurements-a prospective study. Am J Obstet Gynecol. 151:333–337. 1985. View Article : Google Scholar : PubMed/NCBI

36 

Ebbing C, Rasmussen S and Kiserud T: Middle cerebral artery blood flow velocities and pulsatility index and the cerebroplacental pulsatility ratio: Longitudinal reference ranges and terms for serial measurements. Ultrasound Obstet Gynecol. 30:287–296. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Jugović D, Tumbri J, Medić M, Jukić MK, Kurjak A, Arbeille P and Salihagić-Kadić A: New Doppler index for prediction of perinatal brain damage in growth-restricted and hypoxic fetuses. Ultrasound Obstet Gynecol. 30:303–311. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Laemmli UK: Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature. 227:680–685. 1970. View Article : Google Scholar : PubMed/NCBI

39 

Neuhoff V, Arold N, Taube D and Ehrhardt W: Improved staining of proteins in polyacrylamide gels including isoelectric focusing gels with a clear background at nanogram sensitivity using Coomassie Brilliant Blue G-250 and R250. Electrophoresis. 9:255–262. 1988. View Article : Google Scholar : PubMed/NCBI

40 

Ishihama Y, Oda Y, Tabata T, Sato T, Nagasu T, Rappsilber J and Mann M: Exponentially modified protein abundance index (emPAI) for estimation of absolute protein amount in proteomics by the number of sequenced peptides per protein. Mol Cell Proteomics. 4:1265–1272. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Bahado-Singh RO, Turkoglu O, Yilmaz A, Kumar P, Zeb A, Konda S, Sherman E, Kirma J, Allos M, Odibo A, et al: Metabolomic identification of placental alterations in fetal growth restriction. J Mater Fetal Neonatal Med. 35:447–456. 2022. View Article : Google Scholar : PubMed/NCBI

42 

Youssef L, Simões RV, Miranda J, García-Martín ML, Paules C, Crovetto F, Amigó N, Cañellas N, Gratacos E and Crispi F: Paired maternal and fetal metabolomics reveal a differential fingerprint in preeclampsia versus fetal growth restriction. Sci Rep. 11:144222021. View Article : Google Scholar : PubMed/NCBI

43 

Heazell AE, Brown M, Dunn WB, Worton SA, Crocker IP, Baker PN and Kell DB: Analysis of the metabolic footprint and tissue metabolome of placental villous explants cultured at different oxygen tensions reveals novel redox biomarkers. Placenta. 29:691–698. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Arimura Y, Ikura M, Fujita R, Noda M, Kobayashi W, Horikoshi N, Sun J, Shi L and Kusakabe M: Cancer-associated mutations of histones H2B, H3.1 and H2A.Z.1 affect the structure and stability of the nucleosome. Nucleic Acids Res. 46:10007–10018. 2018.PubMed/NCBI

45 

Patel D, McAllister SL and Teckman JH: Alpha-1 antitrypsin deficiency liver disease. Transl Gastroenterol Hepatol. 6:232021. View Article : Google Scholar : PubMed/NCBI

46 

Xi Y, Rong Y and Wang Y: Roles of Annexin A protein family in autophagy regulation and therapy. Biomed Pharmacother. 130:1105912020. View Article : Google Scholar : PubMed/NCBI

47 

D'Acquisto F, Perretti M and Flower RJ: Annexin-A1: A pivotal regulator of the innate and adaptive immune systems. J Pharmacol. 155:152–169. 2008.

48 

Peng W, Liu Y, Qi H and Li Q: Alpha-actinin-4 is essential for maintaining normal trophoblast proliferation and differentiation during early pregnancy. Rep Biol Endoc. 19:482021. View Article : Google Scholar

49 

Mangaraj M, Nanda R and Panda S: Apolipoprotein A-I: A molecule of diverse function. Indian J Clin Biochem. 31:253–259. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Pennington KL, Chan TY, Torres MP and Andersen IJ: The dynamic and stress-adaptive signaling hub of 14-3-3: Emerging mechanisms of regulation and context-dependent protein-protein interactions. Oncogene. 37:5587–5604. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Wierenga RK, Kapetaniou EG and Venkatesan R: Triosephosphate isomerase: A highly evolved biocatalyst. Cell Mol Life Sci. 67:3961–3982. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Fu W, Wang W, Hao J, Zhu X and Sun M: Purification and characterization of catalase from marine bacterium Acinetobacter sp. YS0810. Biomed Res Int. 2014:4096262014. View Article : Google Scholar : PubMed/NCBI

53 

Poillerat V, Gentinetta T, Leon J, Wassmer A, Edler M, Torset C, Luo D, Tuffin G and Roumenina LT: Hemopexin as an inhibitor of hemolysis-induced complement activation. Front Immunol. 11:16842020. View Article : Google Scholar : PubMed/NCBI

54 

Varricchio L, Falchi M, Dall'Ora M, De Benediyyis C, Ruggeri A, Uversky VN and Migliaccio AR: Calreticulin: Challenges posed by the intrinsically disordered nature of calreticulin to the study of its function. Front Cell Dev Biol. 6:962017. View Article : Google Scholar : PubMed/NCBI

55 

Khan HA and Mutus B: Protein disulfide isomerase a multifunctional protein with multiple physiological roles. Front Chem. 2:702014.PubMed/NCBI

56 

Karvar S, Ansa-Addo SA, Suda J, Singh S, Zhu L, Li Z and Dony DC: Moesin, an Ezrin/Radixin/Moesin family member, regulates hepatic fibrosis. Hepatology. 72:1073–1084. 2020. View Article : Google Scholar : PubMed/NCBI

57 

Sizemore ST, Zhang M, Cho JH, Sizemore GM, Hurwitz B, Kaur B, Lehman NL, Ostrowski MC, Robe PA, Miao W, et al: Pyruvate kinase M2 regulates homologous recombinationmediated DNA double-strand break repair. Cell Res. 28:1090–1102. 2018. View Article : Google Scholar : PubMed/NCBI

58 

Wilkinson HC and Dalby PA: The Two-species model of transketolase explains donor substrate-binding, inhibition and heat-activation. Sci Rep. 10:41482020. View Article : Google Scholar : PubMed/NCBI

59 

Bernard JY, Tint MT, Aris IM, Chenc LW, Quaha PL, Tand KH, Yeo GS, Fortier MV, Yap F, Shek L, et al: Maternal plasma phosphatidylcholine polyunsaturated fatty acids during pregnancy and offspring growth and adiposity. Prostaglandins Leukot Essent Fatty Acids. 121:21–29. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Visentin S, Crotti S, Donazzolo E, D'Aronco S, Nitti D, Cosmi E and Agostini M: Medium chain fatty acids in intrauterine growth restricted and small for gestational age pregnancies. Metabolomics. 13:542017. View Article : Google Scholar

61 

Clinton CM, Bain JR, Muehlbauer MJ, Li YY, Li L, O'Neal SK and Ferguson KK: Non-targeted urinary metabolomics in pregnancy and associations with fetal growth restriction. Sci Rep. 10:53072020. View Article : Google Scholar : PubMed/NCBI

62 

Dessì A, Atzori L, Noto A, Visser GH, Gazzolo D, Zanardo V, Barberini L, Puddu M, Ottonello G, Atzei A, et al: Metabolomics in newborns with intrauterine growth retardation (IUGR): Urine reveals markers of metabolic syndrome. J Mater Fetal Neonat Med. 24 (Suppl 2):S35–S39. 2011. View Article : Google Scholar

63 

Raff H, Bruder ED, Jankowski BM and Goodfriend TL: Neonatal hypoxic hyperlipidemia in the rat: Effects on aldosterone and corticosterone synthesis in vitro. Am J Physiol Regul Integr Comp Physiol. 278:R663–R668. 2000. View Article : Google Scholar : PubMed/NCBI

64 

Jiang F, Zhang Y and Dusting GJ: NADPH oxidase-mediated redox signaling: Roles in cellular stress response, stress tolerance and tissue repair. Pharmacol Rev. 63:218–242. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Sena LA and Chandel NS: Physiological roles of mitochondrial reactive oxygen species. Mol Cell. 48:158–167. 2012. View Article : Google Scholar : PubMed/NCBI

66 

Schieber M and Chandel NS: ROS function in redox signaling and oxidative stress. Curr Biol. 24:R453–R462. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Gretes MC, Poole LB and Karplus PA: Peroxiredoxins in parasites. Antioxid Redox Signal. 17:608–633. 2012. View Article : Google Scholar : PubMed/NCBI

68 

Sun HN, Kim SU, Huang SM, Kim JM, Park YH, Kim SH, Yang HY, Chung KJ, Lee TH, Choi HS, et al: Microglial peroxiredoxin V acts as an inducible anti-inflammatory antioxidant through cooperation with redox signaling cascades. J Neurochem. 114:39–50. 2010.PubMed/NCBI

69 

Diet A, Abbas K, Bouton C, Guillon B, Tomasello F, Fourquet S, Toledano MB and Drapier JC: Regulation of peroxiredoxins by nitric oxide in immunostimulated macrophages. J Biol Chem. 282:36199–36205. 2007. View Article : Google Scholar : PubMed/NCBI

70 

Vinchi F, Costa da Silva M, Ingoglia G, Petrillo S, Brinkman N, Zuercher A, Cerwenka A, Tolosano E and Muckenthaler MU: Hemopexin therapy reverts heme-induced pro-inflammatory phenotypic switching of macrophages in a mouse model of sickle cell disease. Blood. 127:473–486. 2016. View Article : Google Scholar : PubMed/NCBI

71 

Li R, Saleem S, Zhen G, Cao W, Zhuang H, Lee J, Smith A, Altruda F, Tolosano E and Doré S: Heme-hemopexin complex attenuates neuronal cell death and stroke damage. J Cereb Blood Flow Metab. 29:953–964. 2009. View Article : Google Scholar : PubMed/NCBI

72 

Yang Y, Dong B, Lu J, Wang G and Yu Y: Hemopexin reduces blood-brain barrier injury and protects synaptic plasticity in cerebral ischemic rats by promoting EPCs through the HO-1 pathway. Brain Res. 1699:177–185. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Belkacemi L, Bédard I, Simoneau L and Lafond J: Calcium channels, transporters and exchangers in placenta: A review. Cell Calcium. 37:1–8. 2005. View Article : Google Scholar : PubMed/NCBI

74 

Michalak M, Groenendyk J, Szabo E, Gold LI and Opas M: Calreticulin, a multi-process calcium-buffering chaperone of the endoplasmic reticulum. Biochem J. 417:651–666. 2009. View Article : Google Scholar : PubMed/NCBI

75 

Yamamoto M, Ikezaki M, Toujima S, Iwahashi N, Mizoguchi M, Nanjo S, Minami S, Ihara Y and Ino K: Calreticulin is involved in invasion of human extravillous trophoblasts through functional regulation of integrin beta1. Endocrinology. 158:3874–3889. 2017. View Article : Google Scholar : PubMed/NCBI

76 

Iwahashi N, Ikezaki M, Matsuzaki I, Yamamoto M, Toujima S, Murata SI, Ihara Y and Ino K: Calreticulin regulates syncytialization through control of the synthesis and transportation of E-cadherin in BeWo cells. Endocrinology. 160:359–374. 2019.PubMed/NCBI

77 

Gold LI, Eggleton P, Sweetwyne MT, Van Duyn LB, Greives MR, Naylor SM and Murphy-Ullrich JE: Calreticulin: Non-endoplasmic reticulum functions in physiology and disease. FASEB J. 24:665–683. 2010. View Article : Google Scholar : PubMed/NCBI

78 

Shi Z, Hou W, Hua X, Zhang X, Liu X and Wang X and Wang X: Overexpression of calreticulin in pre-eclamptic placentas: Effect on apoptosis, cell invasion and severity of pre-eclampsia. Cell Biochem Biophys. 63:183–189. 2012. View Article : Google Scholar : PubMed/NCBI

79 

Gu VY, Wong MH, Stevenson JL, Crawford KE, Brennecke SP and Gude NM: Calreticulin in human pregnancy and pre-eclampsia. Mol Hum Reprod. 14:309–315. 2008. View Article : Google Scholar : PubMed/NCBI

80 

Iwahashi N, Ikezaki M, Nishitsuji K, Yamamoto M, Matsuzaki I, Kato N, Takaoka N, Taniguchi M, Murata SI, Ino K and Ihara Y: Extracellularly released calreticulin induced by endoplasmic reticulum stress impairs syncytialization of cytotrophoblast model BeWo cells. Cells. 10:13052021. View Article : Google Scholar : PubMed/NCBI

81 

Mo HQ, Tian FJ, Ma XL, Zhang YC, Zhang CX, Zeng WH, Zhang Y and Lin Y: PDIA3 regulates trophoblast apoptosis and proliferation in preeclampsia via the MDM2/p53 pathway. Reproduction. 160:293–305. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Foote M and Zhou Y: 14-3-3 proteins in neurological disorders. Int J Biochem Mol Biol. 3:152–164. 2012.PubMed/NCBI

83 

Muslin AJ and Xing H: 14-3-3 proteins: Regulation of subcellular localization by molecular interference. Cell Signal. 12:703–709. 2000. View Article : Google Scholar : PubMed/NCBI

84 

Mackintosh C: Dynamic interactions between 14-3-3 proteins and phosphoproteins regulate diverse cellular processes. Biochem J. 381:329–342. 2004. View Article : Google Scholar : PubMed/NCBI

85 

Kaplan A, Morquette B, Kroner A, Leong SY, Madwar C, Sanz R, Benerjee SL, Antel J, Bisson N, David S and Fournier AE: Small-molecule stabilization of 14-3-3 Protein-protein interactions stimulates axon regeneration. Neuron. 93:1082–1093.e5. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Kwon OK, Lee W, Kim SJ, Lee YM, Lee JY, Kim JY, Bae JS and Lee S: In-depth proteomics approach of secretome to identify novel biomarker for sepsis in LPS-stimulated endothelial cells. Electrophoresis. 36:2851–2858. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Lee W, Kwon OK, Han MS, Lee YM, Kim SW, Kim KM, Lee T, Lee S and Bae JS: Role of moesin in HMGB1-stimulated severe inflammatory responses. Thromb Haemost. 114:350–363. 2015. View Article : Google Scholar : PubMed/NCBI

88 

Chen Y, Wang J, Zhang L, Zhu J, Zeng Y and Huang JA: Moesin is a novel biomarker of endothelial injury in sepsis. J Immunol Res. 2021:66956792021. View Article : Google Scholar : PubMed/NCBI

89 

Satooka H, Matsui M, Ichioka S, Nakamura Y and Hirata T: The ERM protein moesin regulates natural killer cell homeostasis in vivo. Cell Immunol. 371:1044562022. View Article : Google Scholar : PubMed/NCBI

90 

Hasan D, Gamen E, Tarboush NA, Ismail Y, Pak O and Azab B: PKM2 and HIF1α regulation in prostate cancer cell lines. PLoS One. 13:e02037452018. View Article : Google Scholar : PubMed/NCBI

91 

Li Z, Yang P and Li Z: The multifaceted regulation and functions of PKM2 in tumor progression. Biochim Biophys Acta Rev Cancer. 1846:285–296. 2014. View Article : Google Scholar : PubMed/NCBI

92 

Li L, Zhang Y, Qiao J, Yang JJ and Liu ZR: Pyruvate kinase M2 in blood circulation facilitates tumor growth by promoting angiogenesis. J Biol Chem. 289:25812–25821. 2014. View Article : Google Scholar : PubMed/NCBI

93 

Xu Q, Liu LZ, Yin Y, He J, Li Q, Qian X, You Y, Lu Z, Peiper SC, Shu Y and Jiang BH: Regulatory circuit of PKM2/NFκB/miR-148a/152-modulated tumor angiogenesis and cancer progression. Oncogene. 34:5482–5493. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Azoitei N, Becher A, Steinestel K, Rouhi A, Diepold K, Genze F, Simmet T and Seufferlein T: PKM2 promotes tumor angiogenesis by regulating HIF-1α through NF-κB activation. Mol Cancer. 15:32016. View Article : Google Scholar : PubMed/NCBI

95 

Chen Y, Zhang T, Zeng S, Xu R, Jin K, Coorey NJ, Wang Y, Wang K, Lee SR, Yam M, et al: Transketolase in human Müller cells is critical to resist light stress through the pentose phosphate and NRF2 pathways. Redox Biol. 54:1023792022. View Article : Google Scholar : PubMed/NCBI

96 

Jiang P, Du W and Yang X: A critical role of glucose-6-phosphate dehydrogenase in TAp73-mediated cell proliferation. Cell Cycle. 12:3720–3726. 2013. View Article : Google Scholar : PubMed/NCBI

97 

Krockenberger M, Engel JB, Schmidt M, Kohrenhagen N, Häusler SF, Dombrowski Y, Kapp M, Dietl J and Honig A: Expression of transketolase-like 1 protein (TKTL1) in human endometrial cancer. Anticancer Res. 30:1653–1659. 2010.PubMed/NCBI

98 

Staiger WI, Coy JF, Grobholz R, Hofheinz RD, Lukan N, Post S, Schwarzbach MH and Willeke F: Expression of the mutated transketolase TKTL1, a molecular marker in gastric cancer. Oncol Rep. 16:657–661. 2006.PubMed/NCBI

99 

Hertl M and Cosimi AB: Liver transplantation for malignancy. Oncologist. 10:269–281. 2005. View Article : Google Scholar : PubMed/NCBI

100 

Sun J, Hoshino H, Takaku K, Nakajima O, Muto A, Suzuki H, Tashiro S, Takahashi S, Shibahara S, Alam J, et al: Hemoprotein Bach1 regulates enhancer availability of heme oxygenase-1 gene. EMBO J. 21:5216–5224. 2002. View Article : Google Scholar : PubMed/NCBI

101 

Mitsuishi Y, Taguchi K, Kawatani Y, Shibata T, Nukiwa T, Aburatani H, Yamamoto M and Motohashi H: Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell. 22:66–79. 2012. View Article : Google Scholar : PubMed/NCBI

102 

Sjoblom B, Salmazo A and Djinovic-Carugo K: Alpha-actinin structure and regulation. Cell Mol Life Sci. 65:2688–2701. 2008. View Article : Google Scholar : PubMed/NCBI

103 

Mills M, Yang N, Weinberger R, Vander Woude DL, Beggs AH, Easteal S and North K: Differential expression of the actin-binding proteins, alpha-actinin-2 and −3, in different species: Implications for the evolution of functional redundancy. Hum Mol Genet. 10:1335–1346. 2001. View Article : Google Scholar : PubMed/NCBI

104 

Hamill KJ, Hopkinson SB, Skalli O and Jones JC: Actinin-4 in keratinocytes regulates motility via an effect on lamellipodia stability and matrix adhesions. FASEB J. 27:546–556. 2013. View Article : Google Scholar : PubMed/NCBI

105 

Bridger PS, Haupt S, Leiser R, Johnson GA, Burghardt RC, Tinneberg HR and Pfarrer C: Integrin activation in bovine placentomes and in caruncular epithelial cells isolated from pregnant cows. Biol Reprod. 79:274–282. 2008. View Article : Google Scholar : PubMed/NCBI

106 

Zhang YY, Tabataba H, Liu XY, Wang JY, Yan XG, Farrelly M, Jiang CC, Guo ST, Liu T, Kao HY, et al: ACTN4 regulates the stability of RIPK1 in melanoma. Oncogene. 37:4033–4045. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Pollheimer J, Vondra S, Baltayeva J, Beristain AG and Knöfler M: Regulation of placental extravillous trophoblasts by the maternal uterine environment. Front Immunol. 9:25972018. View Article : Google Scholar : PubMed/NCBI

108 

Kennedy DJ, Fan Y, Wu Y, Pepoy M, Hazen SL and Tang WH: Plasma ceruloplasmin, a regulator of nitric oxide activity and incident cardiovascular risk in patients with CKD. Clin J Am Soc Nephrol. 9:462–467. 2014. View Article : Google Scholar : PubMed/NCBI

109 

Göçmen AY, Sahin E, Semiz E and Gümuşlü S: Is elevated serum ceruloplasmin level associated with increased risk of coronary artery disease? Can J Cardiol. 24:209–212. 2008. View Article : Google Scholar : PubMed/NCBI

110 

Ziakas A, Gavrilidis S, Souliou E, Giannoglou G, Stiliadis I, Karvounis H, Efthimiadis G, Mochlas S, Vayona MA, Hatzitolios A, et al: Ceruloplasmin is a better predictor of the long-term prognosis compared with fibrinogen, CRP, and IL-6 in patients with severe unstable angina. Angiology. 60:50–59. 2009. View Article : Google Scholar : PubMed/NCBI

111 

Hammadah M, Fan Y, Wu Y, Hazen SL and Tang WH: Prognostic value of elevated serum ceruloplasmin levels in patients with heart failure. J Card Fail. 20:946–952. 2014. View Article : Google Scholar : PubMed/NCBI

112 

Guller S, Buhimschi CS, Ma YY, Huang ST, Yang L, Kuczynski E, Zambrano E, Lockwood CJ and Buhimschi IA: Placental expression of ceruloplasmin in pregnancies complicated by severe preeclampsia. Lab Invest. 88:1057–1067. 2008. View Article : Google Scholar : PubMed/NCBI

113 

Bellos I, Papantoniou N and Pergialiotis V: Serum ceruloplasmin levels in preeclampsia: A meta-analysis. J Matern Fetal Neonatal Med. 31:2342–2348. 2018. View Article : Google Scholar : PubMed/NCBI

114 

Surekha MV, Sujatha T, Gadhiraju S, Kumar PU, Kotturu SK, Sharada K and Bhaskar V: Impact of maternal iron deficiency anaemia on the expression of the newly discovered multi-copper ferroxidase, Zyklopen, in term placentas. J Obstet Gynaecol. 42:74–82. 2022. View Article : Google Scholar : PubMed/NCBI

115 

Ozgu-Erdinc AS, Cavkaytar S, Aktulay A, Buyukkagnici U, Erkaya S and Danisman N: Mid-trimester maternal serum and amniotic fluid biomarkers for the prediction of preterm delivery and intrauterine growth retardation. J Obstet Gynaecol Res. 40:1540–1546. 2014. View Article : Google Scholar : PubMed/NCBI

116 

Pogorelova TN, Linde VA, Gunko VO and Selyutina SN: The imbalance of metal-containing proteins and free metal ions in the amniotic fluid during fetal growth. Biomed Khim. 62:69–72. 2016.(In Russian). View Article : Google Scholar : PubMed/NCBI

117 

Ayala R, Shu T and Tsai LH: Trekking across the brain: The journey of neuronal migration. Cell. 128:29–43. 2007. View Article : Google Scholar : PubMed/NCBI

118 

Tischfield MA and Engle EC: Distinct alpha- and beta-tubulin isotypes are required for the positioning, differentiation and survival of neurons: New support for the ‘multi-tubulin’ hypothesis. Biosci Rep. 30:319–330. 2010. View Article : Google Scholar : PubMed/NCBI

119 

Bahi-Buisson N, Poirier K, Fourniol F, Saillour Y, Valence S, Lebrun N, Hully M, Bianco CF, Boddaert N, Elie C, et al: The wide spectrum of tubulinopathies: What are the key features for the diagnosis? Brain. 137:1676–1700. 2014. View Article : Google Scholar : PubMed/NCBI

120 

Hebebrand M, Hüffmeier U, Trollmann R, Hehr U, Uebe S, Ekici AB, Kraus C, Krumbiegel M, Reis A, Thiel CT and Popp B: The mutational and phenotypic spectrum of TUBA1A-associated tubulinopathy. Orphanet J Rare Dis. 14:382019. View Article : Google Scholar : PubMed/NCBI

121 

Romaniello R, Zucca C, Arrigoni F, Bonanni P, Panzeri E, Bassi MT and Borgatti R: Epilepsy in tubulinopathy: Personal series and literature review. Cells. 8:6692019. View Article : Google Scholar : PubMed/NCBI

122 

Wang X, Zhang J, Zhou L, Xu B, Ren X, He K, Nie L, Li X, Liu J, Yang X and Yuan J: Long-term iron exposure causes widespread molecular alterations associated with memory impairment in mice. Food Chem Toxicol. 130:242–252. 2019. View Article : Google Scholar : PubMed/NCBI

123 

Monceau V, Belikova Y, Kratassiouk G, Charue D, Camors E, Communal C, Trouvé P, Russo-Marie F and Charlemagne D: Externalization of endogenous annexin A5 participates in apoptosis of rat cardiomyocytes. Cardiovass Res. 64:496–506. 2004. View Article : Google Scholar : PubMed/NCBI

124 

Li A, Su X, Tian Y, Song G, Zan L and Wang H: Effect of actin alpha cardiac Muscle 1 on the proliferation and differentiation of bovine myoblasts and preadipocytes. Animals (Basel). 11:34682021. View Article : Google Scholar : PubMed/NCBI

125 

Yuan SM and Wu N: Aortic α-smooth muscle actin expressions in aortic disorders and coronary artery disease: An immunohistochemical study. Anatol J Cardiol. 19:11–16. 2018.PubMed/NCBI

126 

Jonckheere A, Smeitink JM and Rodenburg RJT: Mitochondrial ATP synthase: Architecture, function and pathology. J Inherit Metab Dis. 35:211–225. 2012. View Article : Google Scholar : PubMed/NCBI

127 

Männik J, Vaas P, Rull K, Teesalu P and Laan M: Differential placental expression profile of human Growth Hormone/Chorionic Somatomammotropin genes in pregnancies with pre-eclampsia and gestational diabetes mellitus. Mol Cell Endocrinol. 355:180–187. 2012. View Article : Google Scholar : PubMed/NCBI

128 

Bays JL and DeMali KA: Vinculin in cell-cell and cell-matrix adhesions. Cell Mol Life Sci. 74:2999–3009. 2017. View Article : Google Scholar : PubMed/NCBI

129 

Patino MG, Neiders MS, Mirdza E, Sebastiano A, Noble B and Cohen RE: Collagen: An overview. Implant Dent. 11:280–285. 2002. View Article : Google Scholar : PubMed/NCBI

130 

Sudo H, Tsuji AB, Sugyo A, Abe M, Hino O and Saga T: AHNAK is highly expressed and plays a key role in cell migration and invasion in mesothelioma. Inter J Oncol. 20:530–538. 2013.

131 

Sun H, Zhao A, Li M, Dong H, Sun Y, Zhang X, Zhu Q, Bukhari AA, Cao CH, Su D, et al: Interaction of calcium binding protein S100A16 with myosin-9 promotes cytoskeleton reorganization in renal tubulointerstitial fibrosis. Cell Death Dis. 11:1462020. View Article : Google Scholar : PubMed/NCBI

132 

Su W, Mruk DD and Cheng CY: Filamin A: A regulator of blood-testis barrier assembly during post-natal development. Spermatogenesis. 2:73–78. 2012. View Article : Google Scholar : PubMed/NCBI

133 

Burrudge K: Talin: A protein designed for mechanotransduction. Emerg Top Life Sci. 2:673–675. 2018. View Article : Google Scholar : PubMed/NCBI

134 

Duan T, Fan K, Chen S, Yao O, Zeng R, Hong Z, Peng L, Shao Y and Yao B: Role of peroxiredoxin 2 in H2O2 induced oxidative stress of primary Leydig cells. Molec Med Rep. 13:4807–4813. 2016. View Article : Google Scholar : PubMed/NCBI

135 

Stockley RA: The multiple facets of alpha-1-antitrypsin. Ann Transl Med. 3:1302015.PubMed/NCBI

136 

Feng D, Notbohm J, Benjamin A, He S, Wang M, Ang LH, Bantawa M, Bouzid M, Del Gado E, Krishnan R and Pollak MR: Disease-causing mutation in α-actinin-4 promotes podocyte detachment through maladaptation to periodic stretch. Proc Natl Acad Sci USA. 115:1517–1522. 2018. View Article : Google Scholar : PubMed/NCBI

137 

Zanardi A, Conti MA, Cremones P, D'Adamo E, Gilberti P, Apostoli C, Cannistraci A, Piperno S, David S and Alessio M: Ceruloplasmin replacement therapy ameliorates neurological symptoms in a preclinical model of aceruloplasminemia. EMBO Mol Med. 10:91–106. 2018. View Article : Google Scholar : PubMed/NCBI

138 

Lehmann SG, Bourgoin-Voillard S, Seve M and Rachidi W: Tubulin beta-3 chain as a new candidate protein biomarker of human skin aging: A preliminary study. Oxid Med Cell Longev. 2017:51403602017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2022
Volume 26 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gęca T, Stupak A, Nawrot R, Goździcka‑Józefiak A, Kwaśniewska A and Kwaśniewski W: Placental proteome in late‑onset of fetal growth restriction. Mol Med Rep 26: 356, 2022
APA
Gęca, T., Stupak, A., Nawrot, R., Goździcka‑Józefiak, A., Kwaśniewska, A., & Kwaśniewski, W. (2022). Placental proteome in late‑onset of fetal growth restriction. Molecular Medicine Reports, 26, 356. https://doi.org/10.3892/mmr.2022.12872
MLA
Gęca, T., Stupak, A., Nawrot, R., Goździcka‑Józefiak, A., Kwaśniewska, A., Kwaśniewski, W."Placental proteome in late‑onset of fetal growth restriction". Molecular Medicine Reports 26.6 (2022): 356.
Chicago
Gęca, T., Stupak, A., Nawrot, R., Goździcka‑Józefiak, A., Kwaśniewska, A., Kwaśniewski, W."Placental proteome in late‑onset of fetal growth restriction". Molecular Medicine Reports 26, no. 6 (2022): 356. https://doi.org/10.3892/mmr.2022.12872