Open Access

Downregulation of lncRNA USP2‑AS1 in the placentas of pregnant women with non‑diabetic fetal macrosomia promotes trophoblast cell proliferation

  • Authors:
    • Yiwen Lu
    • Qiuqin Tang
    • Shanshan Yang
    • Yuting Cheng
    • Mei Li
    • Dan Guo
    • Ziqiang Fu
    • Hua Jiang
    • Wei Wu
  • View Affiliations

  • Published online on: June 7, 2022     https://doi.org/10.3892/mmr.2022.12766
  • Article Number: 250
  • Copyright: © Lu 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

Macrosomia is a common perinatal complication, with a series of adverse effects on newborns and pregnant women. However, the effects of long non‑coding RNAs (lncRNAs) on non‑diabetic fetal macrosomia (NDFMS) remain unclear. The aim of the present study was to investigate whether aberrant lncRNA expression in the placenta is involved in the pathogenesis of NDFMS and to elucidate its biological mechanisms. The expression profile of lncRNAs in the placentas of pregnant women with NDFMS was investigated using an Agilent Human LncRNA Microarray. Differentially expressed lncRNAs were selected for validation using reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). Additionally, the function of lncRNA ubiquitin‑specific peptidase 2 antisense RNA 1 (USP2‑AS1) was investigated using a trophoblast cell line. The results revealed that 763 lncRNAs were upregulated and 129 lncRNAs were downregulated in the placentas of women in the NDFMS group (|FC| ≥2.0). A total of 10 lncRNAs (|FC| ≥4.0, signal value ≥50) were selected for validation using two‑stage RT‑qPCR, indicating that the expression trends of the 10 differentially expressed lncRNAs in the NDFMS group (n=8 vs. 8 and 48 vs. 48) were consistent with the microarray data. In addition, a significant downregulation in the levels of lncRNA USP2‑AS1 was observed in both the microarray data and second‑stage verification. The overexpression of lncRNA USP2‑AS1 induced G1 phase cell cycle arrest and the number of cells entering S phase was reduced. In addition, the viability of HTR‑8/SVneo cells was significantly inhibited when lncRNA USP2‑AS1 was overexpressed. Therefore, these findings demonstrated that lncRNAs were significantly differentially expressed in the placentas of pregnant women with NDFMS and that the downregulation of lncRNA USP2‑AS1 may be involved in the pathogenesis of NDFMS, by promoting trophoblast cell viability.

Introduction

Macrosomia is a common perinatal complication that has been defined as a full-term infant with a birth weight of ≥4,000 g. In recent decades, the incidence of macrosomia has been increasing, affecting 15–45% of newborns of women with gestational diabetes mellitus and 12% of newborns of women without gestational diabetes mellitus (1). Compared to normal infants, macrosomia increases the risk of childhood obesity, adult obesity, hypertension, diabetes and other age-related diseases (2,3). Diabetes is a risk factor for the development of macrosomia (1,4); however, an effective possible strategy for the prevention and treatment of non-diabetic fetal macrosomia (NDFMS) has not yet been proposed, at least to the best of our knowledge. The underlying pathogenesis of NDFMS remains unclear and further studies are required.

The placenta is composed of the amniotic membrane, leaf-shaped chorion and maternal decidua. The placenta is the interface of nutrition exchange between the mother and fetus, which is essential for the maintenance of the normal functional fetal development (5,6). Therefore, abnormal placental development and placental dysfunction adversely affect fetal growth (79). The proliferation and apoptosis of placental trophoblasts play a key role in the development and maturation of the placenta during pregnancy. Previous studies have demonstrated that the excessive proliferation and reduced apoptosis of placental trophoblasts result in the occurrence of diabetic fetal macrosomia (10,11). However, there is only limited information available regarding the molecular mechanisms of placental development in NDFMS (12,13).

Long non-coding RNAs (lncRNAs), which have a length of >200 nucleotides, play a crucial role in disease development, by regulating the mechanisms of DNA methylation, histone modification, post-transcriptional regulation, RNA interference, imprinted genes and microRNA regulation (1417). Previous studies have reported that lncRNAs may potentially participate in the pathogenesis of placental development (1822); however, the specific biological effects of lncRNAs remain largely unknown, particularly concerning the regulatory role of ubiquitin-specific peptidase 2 antisense RNA 1 (USP2-AS1) in NDFMS.

In the present study, the expression profiles of lncRNAs in the placentas of pregnant women with NDFMS group and healthy controls were examined using an Agilent Human LncRNA Microarray, containing 40,916 lncRNA probes. Subsequently, 10 lncRNAs (|FC| ≥4.0, signal value ≥50) from the microarray results were selected for validation using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Furthermore, the function of lncRNA USP2-AS1 was investigated using HTR-8/SVneo, a trophoblast cell line. The present study aimed to provide new insight into the potential pathogenesis of NDFMS by analyzing the role of lncRNAs in placental development.

Materials and methods

Sample collection

The placental tissues used in the present study were provided by Changzhou Maternal and Child Health Care Hospital during the period from September, 2014 to June, 2015. A total of 96 participants were enrolled in the present study, including 48 pregnant women with NDFMS (newborn weight, ≥4,000 g) and 48 women with normal pregnancies (newborn birth weight, ≥2,500 g and <4,000 g). All participants were monotocous primigravida with full-term birth (≥37 weeks and <42 weeks). The mothers in both groups were free of diabetes or other complications (placental abruption, gestational hypertension, placenta previa, and other complications) during pregnancy. Following the removal of the placental tissue fetal membranes, three sections of placental tissues were randomly collected. Subsequently, the placental tissues were immediately stored at −80°C. The present study was conducted in compliance with the Declaration of Helsinki, and the protocol was approved by the Institutional Review Board of Nanjing Medical University (approval no. FWA00001501). Written informed consent was obtained from all pregnant women prior to their participation in the present study.

RNA extraction and RT-qPCR

Total RNA was extracted from the placental tissues and cultured cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Placental tissues of appropriate size were placed in TRIzol® for RNA extraction. All procedures were performed on ice, to prevent RNA degradation. The concentration and purity of the RNA were measured using a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Inc.), and electrophoresis with 1.5% denaturing agarose gels was used to assess RNA integrity. The PrimeScript RT reagent kit (Takara Bio, Inc.) was used to reverse transcribe the RNA samples into cDNA (RR036A; Takara Bio, Inc.), and qPCR was performed using SYBR® Premix Ex Taq™ on a LightCycler 480 II real-time fluorescent quantitative PCR system (Roche Diagnostics). For reverse transcription, the thermal cycling conditions were: 37°C for 15 min, 85°C for 5 sec and 4°C to end the reaction. For qPCR, the thermal cycling conditions consisted of a step of denaturation at 95°C for 10 min, 40 cycles at 95°C for 15 sec and 60°C for 1 min. The 2−∆ΔCq method was used for the calculation of lncRNA relative expression levels. GAPDH was used as an internal control for lncRNA quantification (23). All the reactions were run in triplicate. The primer sequences for RT-qPCR are listed in Table I.

Table I.

Sequences of primers for lncRNAs used in RT-qPCR.

Table I.

Sequences of primers for lncRNAs used in RT-qPCR.

lncRNA IDGene symbolPrimersSequences (5′-3′)
ENST00000580048.1 ENSG00000264247.1Forward TCACATCCCATGGCCAGAAG
Reverse GCCACAGGTAGAGCTGACAC
ENST00000453774.1 ENSG00000228262.2Forward GGCCATGGCTTCAACTAGACT
Reverse AGAAAAGGAAGTGAGCACGGG
ENST00000604250.1 ENSG00000228262.4Forward TGCAAGAGCATGTGGGTCAA
Reverse ACTCCCAGCCACTATGCATTC
NR_002791.2EMX2OSForward ACGATCCACTCCCTGGTACA
Reverse CGGAAAAGGGTTGGTGCAAG
uc011fns.2 HLA-DQA1Forward AAGCCACCCAGCTACCTAATTC
Reverse ACATTTCTGAGCCAAAGGCAGAG
NR_034160.1 USP2-AS1Forward GGAACTCACAACACACGGGA
Reverse TTGCACAAGATGACAGGGCT
HIT000332651 HIX0040474Forward AGAGTGTGAGACCTGTGGAG
Reverse CAACAAGTTCGTGACCGTGC
LIT3502LIT3502Forward ATGAAGGTGGCCTGGGTAGA
Reverse TCCCATGTACTCTATAAGCAGCTC
TCONS_00001644 XLOC_000983Forward GAAACACGACGGGGGACTTA
Reverse AAGGTCCATCGGATTCCACA
ENST00000587085.1 ENSG00000228262.2Forward TCTAAGCCCTGGTGAATGCTG
Reverse AGTGTGTCCTGAACCCCATT
GAPDHForward GCACCGTCAAGGCTGAGAAC
Reverse GGATCTCGCTCCTGGAAGATG

[i] lncRNA, long non-coding RNA; RT-qPCR, reverse transcription-quantitative PCR.

lncRNA microarray analysis

A total of eight samples from the NDFMS group and eight samples from the control group were prepared into two merged RNA samples, one per each group, for microarray screening. RNA labeling and microarray hybridization were performed according to the manufacturer's protocol using the Agilent Human LncRNA Microarray V4.0 (Agilent Technologies, Inc.), which contains ~77,000 probes that can detect 40,916 lncRNAs. All sequence information was selected from public curated transcriptome databases [including RefSeq (https://www.ncbi.nlm.nih.gov/refseq/), UCSC known genes (http://genome.ucsc.edu) and GENCODE (https://www.gencodegenes.org)]. The datasets are available in the NCBI GEO repository database (GSE199148).

The reverse transcribed cDNA products were used as templates, and random sequence primers were used. The products were quantitatively labeled for microarray hybridization. Each dot array was hybridized with a mixed sample, using 2 dot matrices in total. The hybridized arrays were washed, fixed and scanned using the Agilent DNA Microarray Scanner (G2565CA; Agilent Technologies, Inc.). Agilent Feature Extraction software (version 11.0.1.1; Agilent Technologies, Inc.) was used to analyze the acquired array images. Quantile normalization and subsequent data processing were performed using the GeneSpring GX v12.1 software package (Agilent Technologies, Inc.). The differential expression of lncRNAs between the two groups was screened by fold change (FC) filtering (|FC| ≥2.0). Differentially expressed lncRNAs (|FC| ≥4.0, signal value ≥50) identified in the microarray were selected using RT-qPCR.

Cells and cell culture

HTR-8/SVneo cells were generously provided by Professor Yanling Wang (Institute of Zoology, Chinese Academy of Sciences, Beijing, China). The HTR-8/SVneo cell line (https://web.expasy.org/cellosaurus/CVCL_7162) was initially developed by Graham et al (24). The HTR-8/SVneo cell line was generated using freshly isolated evCTB from a first-trimester placenta, which was transfected with a plasmid containing the simian virus 40 large T antigen (SV40). A recent study demonstrated that this cell line contains two distinct populations, one of epithelial origin and one of mesenchymal origin (25). The trophoblasts were cultured in RPMI-1640 medium (Shanghai Basal Media Technologies Co., Ltd.) supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 U/ml streptomycin (Biosharp Life Sciences) in humidified air at 37°C with 5% CO2. Fresh medium was replaced every 2 days, depending on the cell status.

Vector construction and cell transfection

The lncRNA USP2-AS1 overexpression lentiviral vector plasmid (Shanghai OBiO Technology Corp., Ltd.) was constructed to overexpress full-length lncRNA USP2-AS1. The pLenti-EF1a-EGFP-F2A-Puro-CMV-USP2-AS1 overexpression plasmid (4 µg) was transfected into the HTR-8/SVneo cells using Lipofectamine 2000® (Invitrogen, Thermo Fisher Scientific, Inc.). The control was an empty vector. Cells were collected to determine infection efficiency following 48 h incubation at 37°C. The infection efficiency were verified by analyzing the relative expression of lncRNA USP2-AS1.

Cell viability assay

The Cell Counting Kit-8 (CCK-8; Vazyme Biotech Co., Ltd.) assay was used to determine cell viability. After the transfected cells were cultured at 37°C for 24 h, they were counted and transferred to a 96-well orifice plate to ensure that the number of cells in each sample was the same. After 12 h, the serum-free RPMI-1640 medium (PM150110; Procell Life Science & Technology Co., Ltd.) without penicillin and streptomycin was replaced, and the cells were cultured at 37°C for an additional 4 h. CCK-8 (10 µl) was then added to each well, and the ultraviolet absorbance value was measured at a wavelength of 450 nm using an enzyme standard instrument (Infinite M200 Pro; Tecan Group, Ltd.) after 10 and 30 min.

Cell apoptosis assay

The cells were seeded in 6-well plates (1×103 cells/well). Following a 24-h transfection, the treated cells were washed twice with cold PBS. Cell suspensions (5×106 cells in 400 µl of combination solution) were stained with FITC-labeled Annexin V (C1062S; Annexin V-FITC apoptosis detection kit; Beyotime Institute of Biotechnology) and PI (P-CA-201; Procell Life Science & Technology Co., Ltd.) for 15 min at room temperature in the dark. Binding buffer (400 µl; C1062S; Beyotime Institute of Biotechnology) was then added, and the cells were analyzed using flow cytometry (BD FACSCalibur and CellQuest Pro, v6.0; BD Biosciences). Annexin V-positive and PI-positive cells were considered apoptotic cells.

Cell cycle assay

Following a 24-h transfection, the cells were washed twice with PBS. The supernatant was discarded, and 1 ml pre-cooled 75% ethanol was added to the cell pellet. The cells were mixed and incubated at 4°C for >12 h for fixation. The cells were then washed twice with PBS (Gibco; Thermo Fisher Scientific, Inc.) and centrifuged at 111 × g at 4°C for 5 min. Cells were resuspended in 100 µl PBS (Gibco; Thermo Fisher Scientific, Inc.) and 50–100 µl of PI (P-CA-201; Procell Life Science & Technology Co., Ltd.) were then added, followed by incubation at room temperature in the dark for 30 min. The cell cycle phase was determined using flow cytometry (BD CellQuest Pro, Version 6.0; BD Biosciences).

Statistical analysis

Data are presented as the mean ± SD. If data were in normal distribution, the Student's t-test was used to compare two groups. Otherwise, the Mann-Whitney U-test was used. The maternal age, gestational age, pre-pregnant BMI, gravidity, pregnant weight gain, placental weight, birth weight between two groups were analyzed using Student's t-test. The infant sex was analyzed using Mann-Whitney U test. Statistical analyses were performed using GraphPad Prism version 5.01 (GraphPad Software Inc.). In all cases, P<0.05 was considered to indicate a statistically significant difference.

Results

Patient clinical characteristics

The maternal and infant clinical characteristics of the study cohorts are summarized in Table II. The median maternal age of NDFMS and control group were 26. The age range (minimum-maximum) of NDFMS and control group were 21–26 and 21–34, respectively. Birth weight, placental weight and weight gain during pregnancy were significantly higher in the NDFMS group than in the control group (P<0.05). A significant difference was also found between the NDFMS group and the control group regarding the sex of the infants (P<0.05). However, maternal age, gestational age, pre-pregnancy body mass index and gravidity did not differ significantly between the two groups (P>0.05).

Table II.

Clinical characteristics of the study population.

Table II.

Clinical characteristics of the study population.

CharacteristicControl (n=48)NDFMS (n=48)P-values
Maternal age (years)26.33±2.6826.88±3.270.69
Gestational age (weeks)39.66±1.0139.74±0.950.63
Pre-pregnant BMI (kg/m2)20.06±2.6421.21±3.900.08
Gravidity1.25±0.601.34±0.730.51
Pregnant weight gain (kg)16.84±4.1119.83±4.890.002
Placental weight (g)593.33±102.31760.73±124.14<0.001
Birth weight (g)3322.29±309.604238.04±235.86<0.001
Infant sex, n (%) 0.008
  Male21 (43.75)34 (70.83)
  Female27 (56.25)14 (29.17)

[i] Values are the mean ± SD. NDFMS, non-diabetic fetal macrosomia; BMI, body mass index.

lncRNA microarray analysis

To investigate the potential role of lncRNAs in NDFMS, an Agilent Human LncRNA Microarray (potential to detect 40,916 lncRNAs) was used to analyze the lncRNA expression profiles in the NDFMS group and control group. In total, 892 (2.18%, 892/40,916) differentially expressed lncRNAs were identified with 763 (85.54%, 763/892) significantly upregulated lncRNAs (FC≥2) and 129 (14.46%, 129/892) significantly downregulated lncRNAs (FC ≤-2) in the placentas of women in the NDFMS group in comparison with the control placentas. A scatter plot was drawn to demonstrate the changes in lncRNA expression between the two groups (Fig. 1A), and cluster analysis revealed the clustering association of differentially expressed lncRNAs between the two groups (Fig. 1B). A total of eight samples from the NDFMS group and eight samples from the control group were prepared into two merged RNA samples. Therefore, it was not possible to calculate a P-value for the statistics. The lncRNA with the largest FC, among the upregulated lncRNAs, was HIT000075832 (FC=9.19), and the lncRNA with the lowest FC among the downregulated lncRNAs was ENST00000513672.1 (FC=−11.48). The top 10 upregulated and downregulated lncRNAs in the placentas of women in the NDFMS group are presented in Table III. Subsequently, the lncRNA microarray data were further screened, and the genomic locations of differentially expressed lncRNAs were classified and analyzed (Fig. 1C). The distribution of differentially expressed lncRNAs in gene sites may imply the potential role of lncRNAs. According to their positions in the genome, lncRNAs can be divided into five categories, as follows: bidirectional, antisense, sense, intergenic and intronic. Among the 892 differentially expressed lncRNAs between the two groups, a total of 364 out of 892 (40.81%) lncRNAs were classified into these five categories. Intergenic lncRNAs accounted for the largest proportion in the classification of differentially expressed lncRNAs. Intergenic lncRNAs have a higher evolutionary conservatism and tissue specificity, and they demonstrate active transcriptional activities. lncRNAs in other genomic locations may also play a variety of important potential biological roles, including gene regulation, cell differentiation and chromatin remodeling (26). Additionally, the chromosome distribution of upregulated and downregulated lncRNAs is demonstrated in Fig. 1D.

Table III.

The top 10 upregulated and downregulated lncRNAs in NDFMS, compared with the control.

Table III.

The top 10 upregulated and downregulated lncRNAs in NDFMS, compared with the control.

lncRNA IDGene symbolFCaRegulationChromosome
HIT000075832 HIX01147339.19 Up    7
ENST00000567862.1 ENSG00000261310.17.31 Up    16
TCONS_00007242 XLOC_0032236.64 Up    3
HIT000067251 HIX00301096.32 Up    2
ENST00000430463.1 ENSG00000215498.44.87 Up    22
uc011fns.2 HLA-DQA14.80 Up    6
TCONS_00006281 XLOC_0028824.80 Up    3
ENST00000503357.1 ENSG00000249290.14.78 Up    3
ENST00000433249.1 ENSG00000236556.14.60 Up    10
uc021vkt.1abParts4.54 Up    2
ENST00000513672.1 ENSG00000248322.1−11.48Down1
ENST00000607437.1 ENSG00000228262.4−10.41Down2
ENST00000415714.1 ENSG00000235142.2−9.81Down6
ENST00000610239.1 ENSG00000272727.1−9.00Down4
ENST00000594455.1 ENSG00000230768.2−8.94Down1
ENST00000564381.1 ENSG00000261045.1−8.17Down16
ENST00000598737.1 ENSG00000228486.3−7.87Down2
TCONS_00001693 XLOC_001052−7.02Down1
ENST00000433965.1 ENSG00000235142.2−6.87Down6
ENST00000580048.1 ENSG00000264247.1−6.58Down18

a FC, fold change; positive numbers represent upregulation and negative numbers represent downregulation. lncRNAs, long non-coding RNAs; NDFMS, non-diabetic fetal macrosomia.

lncRNA RT-qPCR verification

In general, when the signal value of the microarray was >50, the detection result was reliable. A total of 12 differentially expressed lncRNAs (|FC| ≥4.0, signal value ≥50) are depicted in Table IV. The two lncRNA (RNA147334|p0438_imsncRNA843 and LIT3501) could not be amplified with RT-qPCR using the designed primers. The remaining 10 differentially expressed lncRNAs were selected for verification. RT-qPCR verification was divided into two stages. Firstly, samples analyzed using an lncRNA microarray (n=8 vs. 8) were used for phase I RT-qPCR verification (Table V). Subsequently, more placental tissue samples from the NDFMS and control group (n=48 vs. 48) were used for phase II lncRNA expression verification (Fig. 2). The two-stage RT-qPCR verification results shared a consistent similar trend as compared with the microarray expression data results (the expression trend of 10 lncRNAs was consistent with the microarray data). Among these, ubiquitin-specific peptidase 2 antisense RNA 1 (USP2-AS1) demonstrated a significantly decreased expression in both microarray and two-stage RT-qPCR verification results. Therefore, USP2-AS1 was selected as the candidate lncRNA for the subsequent experiments.

Table IV.

List of differentially expressed lncRNAs in the Agilent Human LncRNA Microarray results.

Table IV.

List of differentially expressed lncRNAs in the Agilent Human LncRNA Microarray results.

lncRNA IDaGene symbolFCbRegulation
ENST00000580048.1 ENSG00000264247.1−6.58Down
RNA147334|p0438_imsncRNA843Null−6.41Down
ENST00000453774.1 ENSG00000228262.2−5.36Down
ENST00000604250.1 ENSG00000228262.4−5.21Down
NR_002791.2EMX2OS−4.96Down
uc011fns.2 HLA-DQA14.80 Up    
NR_034160.1 USP2-AS1−4.73Down
LIT3501LIT3501−4.71Down
HIT000332651 HIX0040474−4.68Down
LIT3502LIT3502−4.60Down
TCONS_00001644 XLOC_000983−4.34Down
ENST00000587085.1 ENSG00000228262.2−4.22Down

a Signal value is >50.

b FC, Fold change; positive numbers represent upregulation and negative numbers represent downregulation; their absolute values are >4. lncRNAs, long non-coding RNAs; NDFMS, non-diabetic fetal macrosomia.

Table V.

Expression of lncRNAs in 8 placental tissues of women in the NDFMS and control group.

Table V.

Expression of lncRNAs in 8 placental tissues of women in the NDFMS and control group.

Gene symbolControl (n=8)NDFMS (n=8)P-value
ENSG00000264247.10.104±0.0740.031±0.0080.172
ENSG00000228262.20.115±0.0540.141±0.0260.142
ENSG00000228262.40.155±0.0700.110±0.0290.208
EMX2OS0.115±0.0450.087±0.0570.600
HLA-DQA10.002±0.0020.034±0.0110.0008
USP2-AS10.344±0.4160.087±0.0180.002
HIX0040474 0.00006±0.00002 0.00009±0.000040.075
LIT35020.0007±0.001 0.00007±0.000030.916
XLOC_0009830.109±0.570.064±0.0160.093
ENSG00000228262.20.122±0.0230.105±0.0240.093

[i] Values are the mean ± SD. lncRNAs, long non-coding RNAs; NDFMS, non-diabetic fetal macrosomia.

Effects of lncRNA USP2-AS1 on human chorionic trophoblasts

The proliferation and apoptosis of trophoblasts are fundamental for the development of the placenta and the pathogenesis of NDFMS. In the present study, HTR-8/SVneo cells were used to elucidate the role of lncRNA USP2-AS1 in placental development. The transfection efficiency of lncRNA USP2-AS1 was first examined. Compared with the control group, the USP2-AS1 expression levels in HTR-8/SVneo cells in the USP2-AS1 overexpression group were significantly increased following transfection (Fig. 3A). The overexpression of lncRNA USP2-AS1 arrested the cells in the G1 phase and reduced the number of cells entering the S phase (Fig. 3B). The overexpression of lncRNA USP2-AS1 also significantly decreased cell viability (Fig. 3C). However, the overexpression of lncRNA USP2-AS1 significantly decreased cell apoptosis compared with the control group (Fig. 3D).

Discussion

The occurrence of macrosomia is dependent on various factors. For environmental factors, including the occurrence of diabetes in pregnant women, the probability of producing macrosomia is ~26%, and the probability of producing macrosomia of pregnant women without diabetes is limited to 5–8% (1). Pregnant women with excessive nutrition, obese pregnant women and overweight pregnant women have been reported to also be susceptible to macrosomia (2729). Normal placental function exists in only a few overdue pregnancies, and the fetal weight increases with the period of pregnancy. The incidence of a large amount of amniotic fluid in pregnant women is high. Genetic factors also have a certain effect on the weight of the fetus (30). Usually, the incidence of fetal macrosomia is high in tall parents (31). Among these confounding factors, the policy of encouraging one child in family planning implemented by the Chinese government and the corresponding inclusion and exclusion criteria of the present study were effectively controlled. However, after controlling the aforementioned factors, there is still a certain risk for macrosomia: the weight of the placenta is a variable exerting marked influence on the occurrence of macrosomia (32). A comparison of the data suggested that the weight of the placenta was associated with fetal birth weight. The size of the placenta has been demonstrated to affect the birth weight of the fetus (33).

All the nutrients required for the growth of the fetus are supplied by the mother through the placenta. However, the purpose of the placenta is not merely for material exchange; it also has a number of other functions, as follows: i) defense function: it functions as a barrier against a number of bacteria, pathogens and drugs (34); ii) cooperative function: chorionic gonadotropin, placental lactogen, estrogen, progesterone, oxytocin enzyme, thermostable alkaline phosphatase, cytokines and growth factors are secreted (34); iii) storage function: a large number of nutrients (protein, glycogen, calcium and iron) are stored in placental cells for fetal growth requirements (35); and iv) metabolic regulation function: the placenta may regulate the metabolism of the body similar to that of the liver (36). Previous research results by the authors revealed that the placental weights of the macrosomia group were significantly higher than those of the normal group (12). Thus, it was considered worthy of investigation to define which factors lead to the overdevelopment of placentas and the occurrence of macrosomia.

lncRNAs are non-coding RNAs with a length of >200 nucleotides, and are related to numerous pregnancy complications, including gestational diabetes (37). In the present study, the expression profiles of lncRNAs in the placentas of pregnant women with NDFMS were investigated using an Agilent Human LncRNA Microarray V4.0. In the placentas of the women in the NDFMS group, 763 lncRNAs were upregulated and 129 lncRNAs were downregulated. Subsequently, 10 differentially expressed lncRNAs were selected to validate the preliminary results, and the two-stage RT-qPCR verifications were consistent with the microarray results. In addition, lncRNA USP2-AS1 exhibited a significant downregulation in both the microarray data and second-stage RT-qPCR verification. Therefore, lncRNA USP2-AS1 was the most prominent candidate lncRNA, and was used for subsequent analysis.

lncRNA USP2-AS1, located on the human chromosome 11q23.3, is a lncRNA with a length of 2,486 nucleotides. A previous study revealed that USP2-AS1 promotes the growth and metastasis of colon adenocarcinoma cells and may play a carcinogenic effect in colon adenocarcinoma (38). In addition, lncRNA USP2-AS1 has been demonstrated to be upregulated in ovarian cancer. Mechanistic analysis have revealed that USP2-AS1 promotes ovarian cancer progression via the miR-520d-3p/KIAA1522 axis (39). lncRNAs have also been revealed to play vital biological regulatory effects in the development of the placenta (1822); however, the role and mechanisms of action of USP2-AS1 in NDFMS remain unclear.

The growth patterns of placental cells bear similarities to those of tumor cells, which are often referred to as ‘pseudotumors’. Therefore, the present study focused on the key molecules that regulate the biological function of placental cells. Firstly, a cell model in which the target lncRNA was overexpressed in a trophoblast cell line was generated, and the viability and apoptosis of the cells was evaluated. Following USP2-AS1 overexpression, the cells were blocked in the G1 phase, and the cell viability and apoptotic rates were decreased. It was hypothesized that the decrease in the apoptosis of HTR-8/SVneo cells overexpressing USP2-AS1 may have been a compensation effect. These results suggested that USP2-AS1 mainly promotes placental development by affecting the proliferative activity of placental cells, which may lead to NDFMS. However, further studies are required for the elucidation of the precise molecular mechanisms of USP2-AS1 in the placentas of pregnant women with macrosomia. The combination of basic and clinical research will provide a breakthrough point for the research of non-diabetic macrosomia and a theoretical basis for the prevention and treatment of clinical non-diabetic macrosomia.

The present study had several limitations. Firstly, the subjects were women who resided in the vicinity of Jiangsu Province, resulting in regional limitations. Secondly, the present study did not predict the lncRNA target genes or explore their functions in NDFMS. Finally, the function of aberrantly expressed lncRNA USP-AS1 was not verified further with the use of an animal model. In a follow-up project by the authors, the biological functions of lncRNA USP-AS1 in macrosomia will be further explored in vivo and also by applying molecular mechanism research.

In conclusion, the present study identified the expression profile of lncRNAs in the placentas of women with NDFMS and revealed for the first time, to the best of our knowledge, that lncRNA USP2-AS1 participates in the pathogenesis of NDFMS by regulating cell function. The present study provides new insight into exploring the post-transcriptional regulatory mechanisms of NDFMS, suggesting potential biological targets for future clinical treatment of NDFMS.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant nos. 81771597, 81971405), Major Project of University Natural Science Research Project of Jiangsu Province (grant no. 20KJA330001), Medical Scientific Research Project of Jiangsu Provincial Health Commission (grant no. Z2019010), and the Priority Academic Program for the Development of Jiangsu Higher Education Institutions (Public Health and Preventive Medicine).

Availability of data and materials

The datasets generated and/or analyzed during the current study are available in the NCBI GEO repository database (GSE199148), [https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE199148].

Authors' contributions

WW and HJ were involved in the conceptualization and design of the study. DG and QT were involved in the acquisition of data. YL and YC were involved in data analysis. YL, ML, YC, WW and HJ were involved in the interpretation of the data. YL, SY, DG and ZF performed the experiments. WW, QT and HJ contributed materials/analysis tools. YL and YC were involved in the preparation of the original draft. WW and HJ were involved in the reviewing and editing of the manuscript. YL, ZF and QT confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was supervised and approved by the Institutional Review Board of Nanjing Medical University (FWA00001501). Written informed consent was obtained from all pregnant women prior to their participation in the present study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kc K, Shakya S and Zhang H: Gestational diabetes mellitus and macrosomia: A literature review. Ann Nutr Metab. 66 (Suppl 2):S14–S20. 2015. View Article : Google Scholar

2 

Linder N, Lahat Y, Kogan A, Fridman E, Kouadio F, Melamed N, Yogev Y and Klinger G: Macrosomic newborns of non-diabetic mothers: Anthropometric measurements and neonatal complications. Arch Dis Child Fetal Neonatal Ed. 99:F353–F358. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Koyanagi A, Zhang J, Dagvadorj A, Hirayama F, Shibuya K, Souza JP and Gülmezoglu AM: Macrosomia in 23 developing countries: An analysis of a multicountry, facility-based, cross-sectional survey. Lancet. 381:476–483. 2013. View Article : Google Scholar

4 

Valvi D, Oulhote Y, Weihe P, Dalgård C, Bjerve KS, Steuerwald U and Grandjean P: Gestational diabetes and offspring birth size at elevated environmental pollutant exposures. Environ Int. 107:205–215. 2017. View Article : Google Scholar

5 

Jansson T and Powell TL: Role of placental nutrient sensing in developmental programming. Clin Obstet Gynecol. 56:591–601. 2013. View Article : Google Scholar

6 

Jansson T and Powell TL: Role of the placenta in fetal programming: Underlying mechanisms and potential interventional approaches. Clin Sci (Lond). 113:1–13. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Tian FY, Wang XM, Xie C, Zhao B, Niu Z, Fan L, Hivert MF and Chen WQ: Placental surface area mediates the association between FGFR2 methylation in placenta and full-term low birth weight in girls. Clin Epigenetics. 10:392018. View Article : Google Scholar : PubMed/NCBI

8 

Ilekis JV, Tsilou E, Fisher S, Abrahams VM, Soares MJ, Cross JC, Zamudio S, Illsley NP, Myatt L, Colvis C, et al: Placental origins of adverse pregnancy outcomes: Potential molecular targets: An executive workshop summary of the eunice Kennedy Shriver National institute of child health and human development. Am J Obstet Gynecol. 215 (Suppl 1):S1–S46. 2016. View Article : Google Scholar

9 

Longtine MS and Nelson DM: Placental dysfunction and fetal programming: The importance of placental size, shape, histopathology, and molecular composition. Semin Reprod Med. 29:187–196. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Lunghi L, Ferretti ME, Medici S, Biondi C and Vesce F: Control of human trophoblast function. Reprod Biol Endocrinol. 5:62007. View Article : Google Scholar

11 

Ishihara N, Matsuo H, Murakoshi H, Laoag-Fernandez J, Samoto T and Maruo T: Changes in proliferative potential, apoptosis and Bcl-2 protein expression in cytotrophoblasts and syncytiotrophoblast in human placenta over the course of pregnancy. Endocr J. 47:317–327. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Guo D, Jiang H, Chen Y, Yang J, Fu Z, Li J, Han X, Wu X, Xia Y, Wang X, et al: Elevated microRNA-141-3p in placenta of non-diabetic macrosomia regulate trophoblast proliferation. EBioMedicine. 38:154–161. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Li J, Fu Z, Jiang H, Chen L, Wu X, Ding H, Xia Y, Wang X, Tang Q and Wu W: IGF2-derived miR-483-3p contributes to macrosomia through regulating trophoblast proliferation by targeting RB1CC1. Mol Hum Reprod. 24:444–452. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Lai J, Chen B, Zhang G, Li X, Mok H and Liao N: Molecular characterization of breast cancer: A potential novel immune-related lncRNAs signature. J Transl Med. 18:4162020. View Article : Google Scholar : PubMed/NCBI

15 

Sun QM, Hu B, Fu PY, Tang WG, Zhang X, Zhan H, Sun C, He YF, Song K, Xiao YS, et al: Long non-coding RNA 00607 as a tumor suppressor by modulating NF-κB p65/p53 signaling axis in hepatocellular carcinoma. Carcinogenesis. 39:1438–1446. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Liang H, Yu T, Han Y, Jiang H, Wang C, You T, Zhao X, Shan H, Yang R, Yang L, et al: LncRNA PTAR promotes EMT and invasion-metastasis in serous ovarian cancer by competitively binding miR-101-3p to regulate ZEB1 expression. Mol Cancer. 17:1192018. View Article : Google Scholar : PubMed/NCBI

17 

Knauss JL, Miao N, Kim SN, Nie Y, Shi Y, Wu T, Pinto HB, Donohoe ME and Sun T: Long noncoding RNA Sox2ot and transcription factor YY1 co-regulate the differentiation of cortical neural progenitors by repressing Sox2. Cell Death Dis. 9:7992018. View Article : Google Scholar : PubMed/NCBI

18 

Jiang S, Chen Q, Liu H, Gao Y and Yang X, Ren Z, Gao Y, Xiao L, Zhong M, Yu Y and Yang X: Preeclampsia-Associated lncRNA INHBA-AS1 regulates the proliferation, invasion, and migration of placental trophoblast cells. Mol Ther Nucleic Acids. 22:684–695. 2020. View Article : Google Scholar : PubMed/NCBI

19 

Chen Q, Jiang S, Liu H, Gao Y, Yang X, Ren Z, Gao Y, Xiao L, Hu H, Yu Y, et al: Association of lncRNA SH3PXD2A-AS1 with preeclampsia and its function in invasion and migration of placental trophoblast cells. Cell Death Dis. 11:5832020. View Article : Google Scholar : PubMed/NCBI

20 

Ma Y, Liang X, Wu H, Zhang C and Ma Y: Long noncoding RNA NR_002794 is upregulated in pre-eclampsia and regulates the proliferation, apoptosis and invasion of trophoblast cells. Mol Med Rep. 20:4567–4575. 2019.PubMed/NCBI

21 

Pengjie Z, Xionghui C, Yueming Z, Ting X, Na L, Jianying T and Zhice X: LncRNA uc003fir promotes CCL5 expression and negatively affects proliferation and migration of trophoblast cells in preeclampsia. Pregnancy Hypertens. 14:90–96. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Zhang Y, Jin F, Li XC, Shen FJ, Ma XL, Wu F, Zhang SM, Zeng WH, Liu XR, Fan JX, et al: The YY1-HOTAIR-MMP2 signaling axis controls trophoblast invasion at the maternal-fetal interface. Mol Ther. 25:2394–2403. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Graham CH, Hawley TS, Hawley RG, MacDougall JR, Kerbel RS, Khoo N and Lala PK: Establishment and characterization of first trimester human trophoblast cells with extended lifespan. Exp Cell Res. 206:204–211. 1993. View Article : Google Scholar : PubMed/NCBI

25 

Abou-Kheir W, Barrak J, Hadadeh O and Daoud G: HTR-8/SVneo cell line contains a mixed population of cells. Placenta. 50:1–7. 2017. View Article : Google Scholar

26 

Dhanoa JK, Sethi RS, Verma R, Arora JS and Mukhopadhyay CS: Long non-coding RNA: Its evolutionary relics and biological implications in mammals: A review. J Anim Sci Technol. 60:252018. View Article : Google Scholar

27 

Du MC, Ouyang YQ, Nie XF, Huang Y and Redding SR: Effects of physical exercise during pregnancy on maternal and infant outcomes in overweight and obese pregnant women: A meta-analysis. Birth. 46:211–221. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Poblete JA and Olmos P: Obesity and Gestational diabetes in pregnant care and clinical practice. Curr Vasc Pharmacol. 19:154–164. 2021. View Article : Google Scholar

29 

Yang W, Han F, Gao X, Chen Y, Ji L and Cai X: Relationship between gestational weight gain and pregnancy complications or delivery outcome. Sci Rep. 7:125312017. View Article : Google Scholar : PubMed/NCBI

30 

Lucarini N, Bottini FG, Borgiani P, Amante A, Gerlini G and Bottini E: Genetic and non genetic factors in the outcome of diabetic pregnancy. J Perinat Med. 22:379–385. 1994. View Article : Google Scholar : PubMed/NCBI

31 

Wilcox MA, Newton CS and Johnson IR: Paternal influences on birthweight. Acta Obstet Gynecol Scand. 74:15–18. 1995. View Article : Google Scholar : PubMed/NCBI

32 

Tanaka K, Matsushima M, Izawa T, Furukawa S, Kobayashi Y and Iwashita M: Influence of maternal obesity on fetal growth at different periods of pregnancies with normal glucose tolerance. J Obstet Gynaecol Res. 44:691–696. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Wallace JM, Horgan GW and Bhattacharya S: Placental weight and efficiency in relation to maternal body mass index and the risk of pregnancy complications in women delivering singleton babies. Placenta. 33:611–618. 2012. View Article : Google Scholar

34 

Burton GJ and Jauniaux E: What is the placenta? Am J Obstet Gynecol. 213 (Suppl 4):S6 e1S6-82015. View Article : Google Scholar

35 

Gude NM, Roberts CT, Kalionis B and King RG: Growth and function of the normal human placenta. Thromb Res. 114:397–407. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Burton GJ and Fowden AL: The placenta: A multifaceted, transient organ. Philos Trans R Soc Lond B Biol Sci. 370:201400662015. View Article : Google Scholar

37 

Li T, Hu D and Gong Y: Identification of potential lncRNAs and co-expressed mRNAs in gestational diabetes mellitus by RNA sequencing. J Matern Fetal Neonatal Med. Feb 22–2021.(Epub ahead of print). View Article : Google Scholar

38 

Li D, Bao J, Yao J and Li J: lncRNA USP2-AS1 promotes colon cancer progression by modulating Hippo/YAP1 signaling. Am J Transl Res. 12:5670–5682. 2020.PubMed/NCBI

39 

Guo B, Yu L, Sun Y, Yao N and Ma L: Long Non-Coding RNA USP2-AS1 accelerates cell proliferation and migration in ovarian cancer by sponging miR-520d-3p and Up-Regulating KIAA1522. Cancer Manag Res. 12:10541–10550. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2022
Volume 26 Issue 2

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
Lu Y, Tang Q, Yang S, Cheng Y, Li M, Guo D, Fu Z, Jiang H and Wu W: Downregulation of lncRNA <em>USP2‑AS1</em> in the placentas of pregnant women with non‑diabetic fetal macrosomia promotes trophoblast cell proliferation. Mol Med Rep 26: 250, 2022
APA
Lu, Y., Tang, Q., Yang, S., Cheng, Y., Li, M., Guo, D. ... Wu, W. (2022). Downregulation of lncRNA <em>USP2‑AS1</em> in the placentas of pregnant women with non‑diabetic fetal macrosomia promotes trophoblast cell proliferation. Molecular Medicine Reports, 26, 250. https://doi.org/10.3892/mmr.2022.12766
MLA
Lu, Y., Tang, Q., Yang, S., Cheng, Y., Li, M., Guo, D., Fu, Z., Jiang, H., Wu, W."Downregulation of lncRNA <em>USP2‑AS1</em> in the placentas of pregnant women with non‑diabetic fetal macrosomia promotes trophoblast cell proliferation". Molecular Medicine Reports 26.2 (2022): 250.
Chicago
Lu, Y., Tang, Q., Yang, S., Cheng, Y., Li, M., Guo, D., Fu, Z., Jiang, H., Wu, W."Downregulation of lncRNA <em>USP2‑AS1</em> in the placentas of pregnant women with non‑diabetic fetal macrosomia promotes trophoblast cell proliferation". Molecular Medicine Reports 26, no. 2 (2022): 250. https://doi.org/10.3892/mmr.2022.12766