Abstract
Objectives
Pregnancy-related medical complications such as gestational diabetes mellitus (GDM) are common and associated with several obstetric and neonatal problems. There is growing evidence that microRNAs (miRNAs) are essential players in the pathophysiology of GDM. This study aimed to assess how particular miRNAs and the genes they target are expressed in GDM.
Methods
A GDM cell model was created using BeWo cells cultured in hyperglycemic (HG) conditions (25 mM glucose). Low-glucose (LG) conditions (5.5 mM glucose) were used for the BeWo cells in the control group. Differentially expressed genes (DEGs) in BeWo cells were identified by high-throughput sequencing and their levels verified in placental samples from GDM patients and controls using RT-PCR. Furthermore, the target genes of the DEGs were verified using dual-luciferase reporter assays.
Results
High-throughput sequencing revealed 220 DEGs in BeWo cells. Among these, miR-3687 was significantly upregulated, while follistatin-like 3 (FSTL3) was downregulated in BeWo cells under HG conditions. The high-throughput sequencing results were corroborated by RT-PCR, which showed that placental samples from GDM patients had significantly lower levels of FSTL3 expression and substantially higher amounts of miR-3687 expression compared to control samples. FSTL3 was established as a direct target of miR-3687 as shown by dual-luciferase reporter assays.
Conclusions
The increase of miR-3687 might facilitate the onset and advancement of GDM by suppressing FSTL3. This discovery offered a new perspective on the molecular underpinnings of GDM and indicated possible targets for therapeutic intervention.
Introduction
Gestational diabetes mellitus (GDM) presents considerable dangers throughout pregnancy, impacting both maternal and neonatal health and elevating the probability of long-term consequences, including type 2 diabetes, obesity, and cardiovascular disease in both the mother and child. GDM impacts over 14 % of pregnancies worldwide, demonstrating its widespread prevalence [1]. In China, the prevalence is notably high at 14.8 % (95 % confidence interval: 12.8–16.7 %) [2]. Typically diagnosed after 24 weeks of gestation, GDM can lead to complications earlier in pregnancy, highlighting the urgency for early identification and intervention to mitigate risks. GDM diagnosis relies on oral glucose tolerance tests administered after critical stages of fetal development have passed, limiting early intervention opportunities. The lack of definitive biomarkers for early GDM detection underscores the need for novel diagnostic tools with both diagnostic and prognostic capabilities. Early identification of GDM is crucial for initiating timely interventions to manage blood glucose concentrations and reduce the risk of adverse pregnancy outcomes and other complications.
Non-coding RNAs (ncRNAs) comprise approximately 60 % of the transcribed human genome and are involved in controlling pathways, developmental processes, and pathological states [3], [4], [5], [6]. Based on their length, they can be broadly divided into two types: long ncRNAs (lncRNAs), which are more than 200 nucleotides, and small ncRNAs, including microRNAs (miRNAs), which are usually<200 nucleotides (21–25 nucleotides long). miRNAs are naturally occurring tiny ncRNAs involved in the posttranscriptional modulation of gene expression by attaching themselves to target mRNAs and adjusting their translation [7]. Mature miRNAs bind to the 3′ untranslated regions of mRNAs, influencing their translation [8], [9], [10]. Due to their stability in bodily fluids and tissue-specific abundance, miRNAs hold promise as potential biomarkers for disease. They have been implicated in various aspects of disease pathology, including their diagnostic, prognostic, and predictive roles in conditions like GDM and its associated health consequences [11], [12], [13].
In this study, we developed a cell model of GDM using BeWo cells and identified 220 significantly differentially expressed genes (DEGs) through high throughput sequencing. Under hyperglycemic (HG) conditions (25 mM glucose), we observed a marked upregulation of miR-3687 and a significant downregulation of its target gene, FSTL3, both implicated in lipid metabolism. Real-time polymerase chain reaction (RT-PCR) analysis was employed for further evaluating these findings in placenta samples from GDM patients and controls. The observed findings indicated that follistatin-like 3 (FSTL3) was lower and miR-3687 was considerably increased in GDM placenta samples compared to controls. The dual luciferase reporter test findings established that miR-3687 directly targets FSTL3. The present study advanced our understanding of the pathophysiology of GDM and could provide critical novel insights for the clinical treatment of the condition.
Materials and methods
Sample collection
This study included patients who were examined and followed till delivery at the Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Twelve patients were recruited and separated into two groups: the GDM group (six pregnant women with GDM) and the control group (six pregnant women without GDM). The inclusion criteria were pregnant women under 40 years of age, regularly attending prenatal care at our hospital, with a gestational age of 38–40 weeks, and a singleton pregnancy. The presence of GDM was assessed using the 75 g oral glucose tolerance test. Patients with pre-existing diabetes mellitus, hypertensive disorders of pregnancy, twin or multiple pregnancies, preterm birth, fetal growth restriction, or any other known endocrinopathy were excluded from the analysis. Six full-term placental samples were collected from elective pregnancy terminations and simple cesarean deliveries in each group. The placental tissue was perfused with phosphate-buffered saline to eliminate the remaining mucus and blood. The placenta samples were subsequently cryogenically frozen in liquid nitrogen and preserved at −80 °C for extended storage. The Beijing Obstetrics and Gynecology Hospital Ethics Committee, Capital Medical University approved this study. All the participants of this study provided their written informed consent.
Cell culture and HG induction
The human choriocarcinoma-derived cell line Bewo employed in the present study was procured from Procell Life Science & Technology Co. Ltd. (Wuhan, China). The BeWo cells were maintained in Ham’s F-12 K medium (Procell), which was enhanced with 10 % fetal bovine serum (Gibco), 100 U/mL penicillin (Sigma-Aldrich, Shanghai, China), and 100 μg/mL streptomycin. The cells were kept in a humidified environment with 5 % CO2 at 37 °C.
Cells were prepared for experimentation by being removed from the original culture medium, washed with PBS (Solarbio, Beijing, China), and digested for 1–3 min using 0.25 % trypsin (Gibco). After that, a pipette was used to divide the cells into single cells, and 15 μL of the cell suspension was rescued for cell counting. After 5 min of centrifuging at 1,000 rpm, the leftover cell suspension was resuspended in the culture medium. Subsequently, the cells (2×105/well) were inoculated in 12-well plates, and grown for an additional night at 37 °C with 5 % CO2.
In previous studies, varying glucose concentrations such as 10, 25, 30 or 35.5 mM have been utilized to mimic the in vitro GDM environment, we selected 25 mM glucose (HG group) to treat BeWo cells to establish a GDM cell model and 5.5 mM glucose (LG group) as the control group according to U.Weiss et al. [14]. Following 48 h of incubation in the recommended media, the cells were harvested with centrifugation at 400 g for 2 min at ambient temperature, after which the supernatant was discarded and the cells were kept at −80 °C for future RNA extraction.
RNA isolation and quality assessment
Total RNA was isolated from BeWo cells with TRIzol (Takara), adhering to the previously described protocol [15]. RNA was purified using the Ribo-off TM rRNA depletion kit (Vazyme, N406-01). Employing a Nanodrop One (Thermo Fischer, USA), the absorbance at 260/280 nm (A260/A280) was measured to determine the quality and amount of the pure RNA. If the A260/A280 values of the samples were greater than 1.7, which was necessary for the generation of RNA sequencing libraries and PCR verification, the samples were deemed appropriate for analysis.
RNA sequencing
cDNA libraries for RNA sequencing were constructed with the KAPA Stranded mRNA-Seq Kit, employing the total RNA (1 μg) of the individual samples while following the instructions provided by the manufacturer. RNA was transformed into cDNA and ligated to Diluted Roche Adaptor (KK8726). Before sequencing, the ligated products underwent amplification, purification, quantification, and storage at −80 °C. The libraries were processed for 150 nt paired-end sequencing using the Illumina NovaSeq 6,000 equipment for high-throughput sequencing. Using Tophat2 [16], clean reads were matched to the GRCh38 genome with up to four mismatches permitted. Gene read number counting and fragments per kilobase of transcript per million fragments mapped (FPKM) computations were performed using uniquely mapped reads [17].
Bioinformatics analysis
The R Bioconductor package edgeR was used for DEG identification [18]. The cut-off values for identifying DEGs were a p-value <0.01 and a fold change>1.5 or<2/3. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) analyses were used to investigate essential pathways connected to DEGs. The KOBAS 2.0 server was used for KEGG pathway analysis and GO categorization [19], 20]. To assess each term’s enrichment, the hypergeometric test and Benjamini-Hochberg FDR controlling technique were employed.
Quantitative PCR
Placental samples were treated with TRIzol (Invitrogen, Waltham, MA, USA) to extract total RNA. While employing TaqMan miRNA assay (Applied Biosystems, Waltham, MA, USA) with miRNA-specific stem-loop RT primers and a TaqMan MicroRNA Reverse Transcription kit in a 50 µL reaction mixture, miR-3687 was reverse-transcribed to cDNA following the directions provided by the manufacturer. SuperScript II Reverse Transcriptase (Life Technologies) and Oligo (dt) primers were used to create cDNA for FSTL3 reverse transcription, which involved treating 2 μg of total RNA with DNase I. SYBR Select Master Mix (Applied Biosystems) was used to perform real-time PCR on an ABI 7,300 sequence detector (Applied Biosystems,). Table 1 showed detailed primer sequence information. The expression level of small nucleolar RNU6 was used to normalize miRNA expression. After normalizing the gene expression levels to GAPDH levels, the ΔΔCt technique was utilized for calculating the relative gene expression.
Primer sequences used in reverse transcription and real-time quantitative PCR.
Stem-loop primer | |
---|---|
miR-3687 forward primer | TGCAAGCCCGGACAGGCGTTCGT |
Universal reverse primer | GTGCAGGGTCCGAGGT |
RNU6 forward primer | CGCTTCGGCAGCACATATAC |
RNU6 reverse primer | AGGGGCCATGCTAATCTTCT |
FSTL3 forward primer | GTGCCTCCGGCAACATTGA |
FSTL3 reverse primer | GCACGAATCTTTGCAGGGA |
GAPDH forward primer | TGTTGCCATCAATGACCCCTT |
GAPDH reverse primer | CTCCACGACGTACTCAGCG |
The top 10 enriched pathways in upregulated DEGs.
Term | Database | ID | Input number | Background | p-Value | Corrected p | Input |
---|---|---|---|---|---|---|---|
Transcriptional misregulation in cancer | KEGG pathway | hsa05202 | 3 | 180 | 0.0059227 | 0.290214 | CD14|HIST2H3D|HIST2H3C |
Amoebiasis | KEGG pathway | hsa05146 | 2 | 100 | 0.0185122 | 0.316059 | CD14|LAMB3 |
Taurine and hypotaurine metabolism | KEGG pathway | hsa00430 | 1 | 11 | 0.0231504 | 0.316059 | GGT1 |
Systemic lupus erythematosus | KEGG pathway | hsa05322 | 2 | 136 | 0.0328545 | 0.316059 | HIST2H3D|HIST2H3C |
Adrenergic signaling in cardiomyocytes | KEGG pathway | hsa04261 | 2 | 149 | 0.0388403 | 0.316059 | PPP2R5B|RAPGEF3 |
Glycosaminoglycan biosyenthesis-hyparan sulfate/heparin | KEGG pathway | hsa00534 | 1 | 24 | 0.0498643 | 0.316059 | HS3ST3A1 |
Alcoholism | KEGG pathway | hsa05034 | 2 | 179 | 0.0541048 | 0.316059 | HIST2H3D|HIST2H3C |
Glyoxylate and dicarboxylate metabolism | KEGG pathway | hsa00630 | 1 | 28 | 0.057946 | 0.316059 | GLYCTK |
Glycine, serine and threonine metabolism | KEGG pathway | hsa00260 | 1 | 40 | 0.081808 | 0.316059 | GLYCTK |
Vasopressin-regulated water reabsorption | KEGG pathway | hsa04962 | 1 | 44 | 0.0896357 | 0.316059 | AQP2 |
Dual-luciferase reporter gene assay
The target genes of miR-3687 were predicted using the biology site (https://cm.jefferson.edu/rna22/Interactive), and FSTL3 was verified as a direct target gene. A dual-luciferase reporter gene test further confirmed FSTL3 and miR-3687’s targeting connection. Segments of synthetic FSTL3 3′UTR (3′-untranslated regions) were included in the pMIR-reporter plasmid utilizing SpeI and Hind III endonuclease sites. Mutation sites of the complementary sequence were engineered between the seed and wild-type FSTL3 (Wt). T4 DNA ligase was employed to ligate the target segment into the pMIR reporter gene plasmid after restriction endonuclease digestion. The co-transfection of the HEK-293 T cells (CRL-1415, Shanghai Xin Yu Biotech Co., Ltd., Shanghai, China) was carried out with miR-3687 and correctly sequenced FSTL3-Wt and FSTL3 mutant (Mut) constructs.
The cells were extracted, lysed, and centrifuged for three to 5 min after transfection for 48 h. The supernatants were then collected. The working solutions for the Renilla luciferase assay were prepared by adding 100 µL of 1:100 sample buffer to the substrate and turning on the fluorometer. The luciferase assay reagent and Renilla luciferase assay buffer were taken from the Luciferase Assay Kit (RG005, Beyotime Biotechnology Co., Shanghai, China). After that, the sample (20–100 μL) from individual groups was combined with luciferase buffer (100 μL), distributed equally using a pipette, and the relative luminescence units (RLUr) were calculated using a luminometer. Renilla luciferase was an internal control, while firefly luciferase RLU was used to calculate luciferase activity.
Statistical analysis
The statistical analysis was carried out with SPSS 19.0. The information was displayed as mean ± standard deviation (SD). A one-way analysis of variance was employed to compare differences between groups, and then LSD post hoc tests were performed. Independent-sample t-tests were employed for comparing the two groups. A statistically significant result was p<0.05 (α=0.05). Statistical significance of differentially expressed miRNAs and target genes between groups was determined using p-value or fold change filtering. KEGG pathway analysis and GO keywords were employed to investigate critical pathways connected to DEGs.
Results
DEGs in BeWo cells induced by glucose
The present study employed high throughput sequencing to identify 220 crucial DEGs in BeWo cells under high glucose (HG) conditions, comprising 158 upregulated and 62 downregulated DEGs (fold change ≥1.5 or ≤2/3, p<0.01). Principal component analysis (PCA) and volcano plots illustrating the distribution and significance of DEGs were depicted in Figures 1A and B, respectively. A heatmap displaying the top 10 DEGs based on their increment order by p-value was presented in Figure 1C.

DEGs in BeWo cells after induction by HG conditions compared with LG conditions. PCA (A), volcano plot (B), and heat map (C) of all differential DEGs.
Eight miRNAs exhibited significant differential expression in BeWo cells, all consistently upregulated under HG conditions. Specifically, miR-3687-1 and miR-3687-2 showed significantly higher expression levels in the HG group, implicating their involvement in lipid metabolism (Figure 2A). A literature review established that FSTL3 was a target gene of miR-3687, had high expression in the placenta, and participates in lipid metabolism. Consistent with this, FSTL3 expression was significantly reduced in BeWo cells under HG conditions.

miR-3687-1 and miR-3687-2 (A) shows significantly higher expression levels in BeWo cells in HG group compared with LG group. A PCA diagram of differential lncRNA (B), and the differential expressions of LINC01556, lncRNAGAS6-AS2, lncRNAMIR31HG, and lncTP53TG1 (C) in Bewo cells in HG group compared with LG group. A heatmap of the differential AS in BeWo cells (D) in HG group compared with LG group.
Furthermore, 87 lncRNA transcripts were significantly differentially expressed in BeWo cells under HG conditions compared to LG conditions. Among these, 54 lncRNAs were upregulated, while 33 were downregulated. Principal component analysis of the differentially expressed lncRNAs was shown in Figure 2B. Many of these lncRNAs have poorly understood functions, but literature searches revealed associations of LINC01556, lncRNAGAS6-AS2, lncRNAMIR31HG, and lncTP53TG1 with type 2 diabetes complications, as depicted in Figure 2C.
Additionally, a total of 1,597 alternative splicing (AS) events were significantly altered in BeWo cells under HG conditions compared to LG conditions, as illustrated in the heatmap in Figure 2D.
Functional analysis of DEGs
The functions of the DEGs and their potential associations with GDM were investigated with KEGG pathway analyses. Analysis of the 158 significantly upregulated DEGs in BeWo cells under HG conditions revealed their involvement in several pathways, including transcriptional misregulation in cancer, amoebiasis, taurine and hypotaurine metabolism, systemic lupus erythematosus, adrenergic signaling in cardiomyocytes, glycosaminoglycan biosynthesis-heparan sulfate/heparin, alcoholism, vasopressin-regulated water reabsorption, serine and threonine metabolism, glycine, and glyoxylate and dicarboxylate metabolism (Table 2, Figure 3A).

The top 10 KEGG pathways of the significantly upregulated DEGs (A) and downregulated DEGs (B). Top 10 GO pathways of the significantly expressed AS.
Conversely, KEGG analysis of downregulated DEGs highlighted their significant roles in pathways such as small cell lung cancer, toxoplasmosis, mucin-type O-glycan biosynthesis, apoptosis, viral myocarditis, VEGF signaling pathway, arrhythmogenic right ventricular cardiomyopathy, pathways in cancer, ECM-receptor interaction, and hypertrophic cardiomyopathy (Table 3, Figure 3B).
Among the top 40 significantly upregulated DEGs in BeWo cells under HG conditions, ranked by their FPKM values, miR-3687-1 and miR-3687-2 exhibited the highest expression levels among these DEGs, emphasizing their involvement in lipid metabolism.
The top 10 enriched pathways in downregulated DEGs.
Term | Database | ID | Input number | Background | p-Value | Corrected p | Input |
---|---|---|---|---|---|---|---|
Small cell lung cancer | KEGG pathway | hsa05222 | 2 | 86 | 0.006199 | 0.139378 | BIRC7|LAMA2 |
Toxoplasmosis | KEGG pathway | hsa05145 | 2 | 119 | 0.011615 | 0.139378 | BIRC7|LAMA2 |
Mucin type-O-glycan biosynethesis | KEGG pathway | hsa00512 | 1 | 31 | 0.043097 | 0.222517 | GALNT8 |
Apoptosis-multiple species | KEGG pathway | hsa04215 | 1 | 33 | 0.045819 | 0.222517 | BIRC7 |
Viral myocarditis | KEGG pathway | hsa05416 | 1 | 60 | 0.081892 | 0.222517 | LAMA2 |
VEGF signaling pathway | KEGG pathway | hsa04370 | 1 | 61 | 0.083204 | 0.222517 | SH2D2A |
Arrhythmogenic right ventricular cardiomyopathy (ARVC) | KEGG pathway | hsa05412 | 1 | 74 | 0.100109 | 0.222517 | LAMA2 |
Pathways in cancer | KEGG pathway | hsa05200 | 2 | 397 | 0.105348 | 0.222517 | BIRC7|LAMA2 |
ECM-receptor interaction | KEGG pathway | hsa04512 | 1 | 82 | 0.110372 | 0.222517 | LAMA2 |
Hypertrophic cardiomyopathy , HCM | KEGG pathway | hsa05410 | 1 | 83 | 0.111648 | 0.222517 | LAMA2 |
Furthermore, the roles of 1,598 differentially expressed AS events were predicted using GO analysis. The analysis indicated that these AS events were crucial in pathways including the regulation of small GTPase-mediated signal transduction, mitotic cell cycle, DNA-dependent transcription, gene expression, retrograde transport from endosome to Golgi, regulation of Rab GTPase activity, mitotic chromosome condensation, DNA repair, heme biosynthesis, and centrosome organization (Figure 3C).
Clinical characteristics of patients
Based on the findings, targets were identified for further investigation to confirm the relationship between miR-3687 and its target gene, FSTL3, in GDM. The present research included 12 pregnant women divided into two groups: GDM (n=6) and control. The clinical parameters of the two groups, such as maternal age, gravida, parity, and gestational age at delivery, were not significantly different. However, women with GDM had significantly higher mean cholesterol levels, serum triglyceride, and body mass index than the control group.
Expression of miR-3687 and FSTL3 in the placenta samples of GDM and control groups
After identifying potential functions of differentially expressed miR-3687 and its target gene FSTL3, the focus shifted to investigating their roles in GDM development. The levels of miR-3687 and FSTL3 expression in placental tissues from GDM patients and controls were measured using RT-PCR analysis. The findings showed that GDM placenta samples had considerably higher expression of miR-3687 than control samples (Figure 4A). On the other hand, FSTL3 expression in GDM placental tissues was markedly less than in controls (Figure 4B). These findings suggested that miR-3687 and FSTL3 contributed to the pathogenesis of GDM.

miR-3687 and FSTL3 relative expression level were verified in the placenta samples from the GDM group and the normal women by qPCR (A and B). The luciferase reporter assay of miR-3687 and FSTL3 (C).
Direct binding of miR-3687 to FSTL3 in placenta samples
The experiment aimed to confirm if miR-3687 directly targets FSTL3 using a dual luciferase reporter assay. In the FSTL3-MT group, cells transfected with miR-3687 mimics showed substantially decreased luciferase activity than the negative control group, suggesting a direct interaction between miR-3687 and FSTL3 (Figure 4C).
Discussion
GDM represents a prevalent medical complication during pregnancy, characterized by normal glucose metabolism before conception followed by altered glucose metabolism, inadequate insulin secretion, and sustained production of anti-insulin-like hormones throughout pregnancy. Currently, screening and diagnosis of GDM typically occur in the mid to late stages of pregnancy, limiting the window for blood glucose intervention and management. Poor glycemic control can result in severe maternal and newborn problems, emphasizing the vital importance of early detection and diagnosis of GDM. Despite the importance of early detection, definitive biomarkers for early GDM identification are lacking. Consequently, researchers actively investigate molecular biomarkers to facilitate early diagnosis and intervention. Previous studies have demonstrated that BeWo trophoblast cells exhibit functional, metabolic, and morphological similarities to primary human trophoblast cultures [21], 22]. And varying glucose concentrations such as 10, 25, 30, or 35.5 mM have been utilized to mimic the in vitro GDM environment [23], [24], [25], [26]. In other reasearches, human trophoblastic cell line HTR8/SVneo were also cultured with different doses of glucose to simulate GDM [27], 28]. However, there was currently no standardized glucose concentration and cell line for GDM models, and different cell line and glucose conditions may influence the experimental outcomes. We selected 25 mM glucose (HG group) to treat BeWo cells to establish a GDM cell model and 5.5 mM glucose (LG group) as the control group according to U.Weiss et al. [14]. We identified genes potentially implicated in GDM-related glucose metabolism through high-throughput sequencing and bioinformatics analyses. This approach provided insights into the molecular mechanisms underlying GDM pathophysiology and offered potential targets for future therapeutic interventions and biomarker development.
Initially, we conducted comprehensive RNA-sequencing of BeWo cells, revealing significant upregulation of 158 genes and downregulation of 62 genes under HG conditions. Furthermore, eight miRNAs exhibited substantial differential expression in the HG group relative to the LG group, all continuously elevated. Notably, miR-3687-1 and miR-3687-2 exhibited substantial upregulation, as depicted in Figure 2. Prior research had associated miR-3687 with multiple cancers, including prostate cancer and bladder cancer, wherein it regulated genes pertinent to cell adhesion, angiogenesis, cell cycle control, JAK-STAT signaling, MAPK signaling, nitric oxide signaling, and VEGF signaling [29], [30], [31]. However, its role in GDM remains unexplored.
FSTL3 is a downstream target gene of miR-3687, highly expressed in the placenta and involved in lipid metabolism. FSTL3, a circulating glycoprotein, is pivotal in glucose metabolism and obesity [32]. FSTL3, an external regulatory protein, interacts with activin A, a member of the transforming growth factor β superfamily, consequently affecting glucose metabolism [33], 34]. Thadhani et al. reported reduced first-trimester circulating levels of FSTL3 in pregnant women who subsequently developed GDM [35]. Similarly, Hu et al. demonstrated significantly lower maternal and placental FSTL3 concentrations in GDM women during the third trimester, suggesting its potential involvement in GDM pathogenesis [36].
In our study, FSTL3 was notably downregulated in BeWo cells under HG conditions, contrasting with the upregulation of miR-3687. Database and literature evidence further supported that miR-3687 regulated genes involved in glucose and lipid metabolism by suppressing FSTL3 expression. Studies also suggested that miR-3687 expression in human adipocytes can be induced by metformin, underscoring their potential roles in GDM onset [37].
After full-term delivery in clinical practice, placental tissues were obtained from six GDM and six control patients to confirm the roles of miR-3687 and FSTL3 in the etiology of GDM. PCR analysis confirmed significant overexpression of miR-3687 and simultaneous underexpression of FSTL3 in the GDM full-term placenta compared to controls, consistent with our previous experiments. Furthermore, dual luciferase reporter gene assays verified the direct targeting of FSTL3 by miR-3687. Although there were no notable variations in age, gestational age, or medical history across the patient groups, individuals with GDM demonstrated substantially higher body mass index, blood triglyceride, and cholesterol levels than the control group. The data indicated that miR-3687 likely modulates glycolipid metabolism by suppressing FSTL3 expression, therefore playing a role in the etiology of GDM.
lncRNAs are emerging as potential new molecular biomarkers of GDM. Unlike microRNAs, lncRNAs are heterogeneous in size and function, localized in poorly conserved genomic regions [38]. Lu et al. examined lncRNA expression profiles throughout early gestation. Five lncRNAs (RP11-160H22.5, LOC149086, PCBP1-AS1, RP11-230G5.2, and XLOC_014172) correlated with macrosomia in pregnancy were discovered, with RP11-230G5.2 and XLOC_014172 demonstrating moderate predictive capability for macrosomia in GDM pregnancies within larger cohorts [39]. Several lncRNAs had drawn attention in various aspects of diabetes and related complications: LINC01556 in diabetic retinopathy, GAS6-AS2 in the PI3K/AKT signaling pathway, MIR31HG in lipid metabolism, and LINC00324 in diabetes wound repair [40], [41], [42], [43]. Our study identified 87 lncRNAs with significant expression changes in BeWo cells under HG conditions, including 54 upregulated and 33 downregulated lncRNAs. However, their expression levels were generally low, necessitating further research to elucidate their specific roles in GDM.
Event-related genes provide new insights for predicting and diagnosing GDM. Alongside significantly differentially expressed ncRNAs, our analysis highlighted 1597 AS events under HG conditions, a noteworthy finding in this study. For predicting the functional consequences of these differentially expressed AS events, GO analysis was used. Notably, four genes were identified as credible AS events based on their p values: Nuclear receptor coactivator 6 (NCOA6), associated with insulin secretion and glucose metabolism in pancreatic β-cells [44], RIPK3 and RNF216, associated with weight-loss regain in obese subjects, G protein-coupled receptor 78 (GPR78), a key target for anti-diabetic compounds, and PQBP1, involved in lipid metabolism [44], [45], [46], [47], [48]. These differentially expressed AS events likely contribute to the pathogenesis of GDM, warranting further investigation.
Other DEGs in BeWo cells under HG conditions might also play roles in GDM pathogenesis. ZnHIT2, a zinc finger protein critical in energy metabolism, was significantly upregulated in HG-cultured BeWo cells. SNORD3A, associated with ER stress, showed significant upregulation [49], 50]. However, in HG-cultured BeWo cells, RGS2 and IGFBP2 were substantially downregulated. IGFBP2 increases insulin sensitivity, whereas RGS2 increases insulin resistance [51], 52]. These findings suggested that gene regulation under HG conditions involved multiple genes with potentially conflicting roles in metabolic pathways. This complexity underscored the need for further cellular and in vivo experiments to elucidate each gene’s expression and regulatory changes in GDM research.
So far there were no relevant literature reports on miR-3687 in GDM. We detected and firstly reported the differential expression of miR-3687 as well as other key differential genes between GDM and healthy individuals, trying to provide a better understanding of the development and progression of GDM. Although some preliminary findings have been made in this study and the expression of differential miR-3687 and FSTL3 has been preliminarily verified by tissue verification, limitations for this study should be outlined. The main study limitation is the relatively small sample size. Notably, comprehensive management of GDM throughout pregnancy is often a key factor influencing maternal and fetal outcomes. However, in clinical practice, the adherence of most patients is poor, leading to fluctuations in blood sugar levels and the occurrence of fetal complications. Based on enough informed consent, we included six normal pregnant women as control and another six patients with GDM as experiment group. The inclusion and exclusion criteria of subject recruitment was very strict and the clinical parameters of the two groups, such as maternal age, gravida, parity, and gestational age at delivery were not significantly different. Moreover, the placental samples used in this study were obtained from women with well-controlled glucose levels. Although the small sample size may limit the generalizability of this study’s findings, it ensures the authenticity of the clinical data from this group of patients and their good adherence. We hope that in the future, more pregnant women will be willing to participate in such clinical research in the future, allowing us to further expand our sample size and make more discoveries that can aid in the prevention and treatment of diseases. Secondly, only one experimental validation method was used in this study, the lack of further functional verification and mechanism research may limit the accurate explanation of the biological significance of differential genes. Ideally, these results should be verified by multiple validation methods such as animal experiments. We hope that the discussion of these limitations will attract more researcher’ attention and promote the in-depth exploration of this field. We believe that through further investigations we can more comprehensively reveal the molecular mechanisms of GDM and provide more in-depth theoretical support for the diagnosis and prevention of GDM.
Our study revealed differential expression of various ncRNAs, especially miRNAs and lncRNAs, in placental and related cells under HG conditions. Notably, miR-3687 emerged as a significant regulator, influencing glucose and lipid metabolism in patients with GDM by targeting the downstream gene FSTL3. The results obtained imply that miR-3687 was essential to the pathophysiology of GDM. Therefore, despite the mentioned limitations, our study offered new insights into the molecular pathways causing GDM through ncRNAs.
Funding source: Beijing Municipal Administration of Hospitals Clinical Medicine Development
Award Identifier / Grant number: ZYLX201830
Funding source: Beijing Hospitals Authority Ascent Plan
Award Identifier / Grant number: DFL20191401
Acknowledgments
We acknowledge all patients who participated in this study.
-
Research ethics: The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board of Beijing Obstetrics and Gynecology Hospital on July 2022 (Document ID: 2022-KY-074-02. All experimental protocols were approved by the Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Informed consent was obtained from all subjects and/or their legal guardian(s).
-
Informed consent: Informed consent was obtained from all individuals included in this study.
-
Author contributions: Y J proposed the study design and drafted the original manuscript under the guidance of XK Y; Y J and XH Z performed the experimental work and collected patients’ information; Y J and Y F were responsible for data collection and formal analysis. All authors reviewed the manuscript and approved the final version of this article.
-
Use of Large Language Models, AI and Machine Learning Tools: None declared.
-
Conflict of interest: The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.
-
Research funding: This study was financially supported by Beijing Municipal Administration of Hospitals Clinical Medicine Development (grant numbers ZYLX201830) and Beijing Hospitals Authority Ascent Plan (grant numbers DFL20191401).
-
Data availability: The datasets used and analysed during the current study available from the corresponding author on reasonable request.
References
1. Guariguata, L, Linnenkamp, U, Beagley, J, Whiting, DR, Cho, NH. Global estimates of the prevalence of hyperglycaemia in pregnancy. Diabetes Res Clin Pract 2014;103:176–85. https://doi.org/10.1016/j.diabres.2013.11.003.Search in Google Scholar PubMed
2. Gao, C, Sun, X, Lu, L, Liu, F, Yuan, J. Prevalence of gestational diabetes mellitus in mainland China: a systematic review and meta-analysis. J Diabetes Investig 2019;10:154–62. https://doi.org/10.1111/jdi.12854.Search in Google Scholar PubMed PubMed Central
3. Mattick, JS, Makunin, IV. Non-coding RNA. Hum Mol Genet 2006;15:R17–29. https://doi.org/10.1093/hmg/ddl046.Search in Google Scholar PubMed
4. Djebali, S, Davis, CA, Merkel, A, Dobin, A, Lassmann, T, Mortazavi, A, et al.. Landscape of transcription in human cells. Nature 2012;489:101–8. https://doi.org/10.1038/nature11233.Search in Google Scholar PubMed PubMed Central
5. National Human Genome Research Institute. The ENCODE (ENCyclopedia of DNA elements) project. Science 2004;306:636–40. https://doi.org/10.1126/science.1105136.Search in Google Scholar PubMed
6. Anastasiadou, E, Jacob, LS, Slack, FJ. Non-coding RNA networks in cancer. Nat Rev Cancer 2018;18:5–18. https://doi.org/10.1038/nrc.2017.99.Search in Google Scholar PubMed PubMed Central
7. Mononen, N, Lyytikainen, LP, Seppala, I, Mishra, PP, Juonala, M, Waldenberger, M, et al.. Whole blood microRNA levels associate with glycemic status and correlate with target mRNAs in pathways important to type 2 diabetes. Sci Rep 2019;9:8887. https://doi.org/10.1038/s41598-019-43793-4.Search in Google Scholar PubMed PubMed Central
8. Bartel, DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281–97. https://doi.org/10.1016/s0092-8674(04)00045-5.Search in Google Scholar PubMed
9. Finnegan, EF, Pasquinelli, AE. MicroRNA biogenesis: regulating the regulators. Crit Rev Biochem Mol Biol 2013;48:51–68. https://doi.org/10.3109/10409238.2012.738643.Search in Google Scholar PubMed PubMed Central
10. Lee, Y, Jeon, K, Lee, JT, Kim, S, Kim, VN. MicroRNA maturation: stepwise processing and subcellular localization. EMBO J 2002;21:4663–70. https://doi.org/10.1093/emboj/cdf476.Search in Google Scholar PubMed PubMed Central
11. Wander, PL, Boyko, EJ, Hevner, K, Parikh, VJ, Tadesse, MG, Sorensen, TK, et al.. Circulating early- and mid-pregnancy microRNAs and risk of gestational diabetes. Diabetes Res Clin Pract 2017;132:1–09. https://doi.org/10.1016/j.diabres.2017.07.024.Search in Google Scholar PubMed PubMed Central
12. Sebastiani, G, Guarino, E, Grieco, GE, Formichi, C, Delli, PC, Ceccarelli, E, et al.. Circulating microRNA (miRNA) expression profiling in plasma of patients with gestational diabetes mellitus reveals upregulation of miRNA mir-330-3p. Front Endocrinol (Lausanne) 2017;8:345. https://doi.org/10.3389/fendo.2017.00345.Search in Google Scholar PubMed PubMed Central
13. Pheiffer, C, Dias, S, Rheeder, P, Adam, S. Decreased expression of circulating mir-20a-5p in south african women with gestational diabetes mellitus. Mol Diagn Ther 2018;22:345–52. https://doi.org/10.1007/s40291-018-0325-0.Search in Google Scholar PubMed
14. Weiss, U, Cervar, M, Puerstner, P, Schmut, O, Haas, J, Mauschitz, R, et al.. Hyperglycaemia in vitro alters the proliferation and mitochondrial activity of the choriocarcinoma cell lines BeWo, JAR and JEG-3 as models for human first-trimester trophoblast. Diabetologia 2001;44:209–19. https://doi.org/10.1007/s001250051601.Search in Google Scholar PubMed
15. Klohonatz, KM, Coleman, SJ, Islas-Trejo, AD, Medrano, JF, Hess, AM, Kalbfleisch, T, et al.. Coding RNA sequencing of equine endometrium during maternal recognition of pregnancy. Genes (Basel) 2019;10. https://doi.org/10.3390/genes10100749.Search in Google Scholar PubMed PubMed Central
16. Kim, D, Pertea, G, Trapnell, C, Pimentel, H, Kelley, R, Salzberg, SL. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol 2013;14:R36. https://doi.org/10.1186/gb-2013-14-4-r36.Search in Google Scholar PubMed PubMed Central
17. Agricola, E, Bove, T, Oppizzi, M, Marino, G, Zangrillo, A, Margonato, A, et al.. Ultrasound comet-tail images”: a marker of pulmonary edema: a comparative study with wedge pressure and extravascular lung water. Chest 2005;127:1690–5. https://doi.org/10.1378/chest.127.5.1690.Search in Google Scholar PubMed
18. Robinson, MD, McCarthy, DJ, Smyth, GK. Edger: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2010;26:139–40. https://doi.org/10.1093/bioinformatics/btp616.Search in Google Scholar PubMed PubMed Central
19. Ashburner, M, Ball, CA, Blake, JA, Botstein, D, Butler, H, Cherry, JM, et al.. Gene ontology: tool for the unification of biology. The gene ontology consortium. Nat Genet 2000;25:25–9. https://doi.org/10.1038/75556.Search in Google Scholar PubMed PubMed Central
20. Altermann, E, Klaenhammer, TR. PathwayVoyager: pathway mapping using the kyoto encyclopedia of genes and genomes (KEGG) database. BMC Genom 2005;6:60. https://doi.org/10.1186/1471-2164-6-60.Search in Google Scholar PubMed PubMed Central
21. Easton, Z, Delhaes, F, Mathers, K, Zhao, L, Vanderboor, C, Regnault, T. Syncytialization and prolonged exposure to palmitate impacts BeWo respiration. Reproduction 2021;161:73–88. https://doi.org/10.1530/rep-19-0433.Search in Google Scholar PubMed PubMed Central
22. Tobin, KA, Johnsen, GM, Staff, AC, Duttaroy, AK. Long-chain polyunsaturated fatty acid transport across human placental choriocarcinoma (BeWo) cells. Placenta 2009;30:41–7. https://doi.org/10.1016/j.placenta.2008.10.007.Search in Google Scholar PubMed
23. Wang, S, Ning, J, Huai, J, Yang, H. Hyperglycemia in pregnancy-associated oxidative stress augments altered placental glucose transporter 1 trafficking via AMPKalpha/p38MAPK signaling cascade. Int J Mol Sci 2022;23. https://doi.org/10.3390/ijms23158572.Search in Google Scholar PubMed PubMed Central
24. Nanobashvili, K, Jack-Roberts, C, Bretter, R, Jones, N, Axen, K, Saxena, A, et al.. Maternal choline and betaine supplementation modifies the placental response to hyperglycemia in mice and human trophoblasts. Nutrients 2018;10. https://doi.org/10.3390/nu10101507.Search in Google Scholar PubMed PubMed Central
25. Guo, J, Zhou, M, Zhao, M, Li, S, Fang, Z, Li, A, et al.. TIGAR deficiency induces caspase-1-dependent trophoblasts pyroptosis through NLRP3-ASC inflammasome. Front Immunol 2023;14:1114620. https://doi.org/10.3389/fimmu.2023.1114620.Search in Google Scholar PubMed PubMed Central
26. Yung, H, Alnaes-Katjavivi, P, Jones, CJP, El-Bacha, T, Golic, M, Staff, A, et al.. Placental endoplasmic reticulum stress in gestational diabetes: the potential for therapeutic intervention with chemical chaperones and antioxidants. Diabetologia 2016;59:2240–50. https://doi.org/10.1007/s00125-016-4040-2.Search in Google Scholar PubMed PubMed Central
27. Peng, H, Li, M, Li, H. High glucose suppresses the viability and proliferation of HTR-8/SVneo cells through regulation of the mir-137/PRKAA1/IL-6 axis. Int J Mol Med 2018;42:799–810. https://doi.org/10.3892/ijmm.2018.3686.Search in Google Scholar PubMed PubMed Central
28. Ji, Y, Zhang, W, Yang, J, Li, C. MiR-193b inhibits autophagy and apoptosis by targeting IGFBP5 in high glucose-induced trophoblasts. Placenta 2020;101:185–93. https://doi.org/10.1016/j.placenta.2020.09.015.Search in Google Scholar PubMed
29. Ronnau, C, Fussek, S, Smit, FP, Aalders, TW, van Hooij, O, Pinto, P, et al.. Upregulation of mir-3195, mir-3687 and mir-4417 is associated with castration-resistant prostate cancer. World J Urol 2021;39:3789–97. https://doi.org/10.1007/s00345-021-03723-4.Search in Google Scholar PubMed PubMed Central
30. Xie, Q, Chen, C, Li, H, Xu, J, Wu, L, Yu, Y, et al.. Mir-3687 overexpression promotes bladder cancer cell growth by inhibiting the negative effect of FOXP1 on cyclin e2 transcription. Mol Ther 2019;27:1028–38. https://doi.org/10.1016/j.ymthe.2019.03.006.Search in Google Scholar PubMed PubMed Central
31. Kasiviswanathan, D, Chinnasamy, PR, Bhuvaneswari, S, Kumar, P, Sundaresan, L, Philip, M, et al.. Interactome of miRNAs and transcriptome of human umbilical cord endothelial cells exposed to short-term simulated microgravity. NPJ Microgravity 2020;6:18. https://doi.org/10.1038/s41526-020-00108-6.Search in Google Scholar PubMed PubMed Central
32. Perakakis, N, Kokkinos, A, Peradze, N, Tentolouris, N, Ghaly, W, Tsilingiris, D, et al.. Follistatins in glucose regulation in healthy and obese individuals. Diabetes Obes Metabol 2019;21:683–90. https://doi.org/10.1111/dom.13572.Search in Google Scholar PubMed PubMed Central
33. Tsuchida, K, Arai, KY, Kuramoto, Y, Yamakawa, N, Hasegawa, Y, Sugino, H. Identification and characterization of a novel follistatin-like protein as a binding protein for the TGF-beta family. J Biol Chem 2000;275:40788–96. https://doi.org/10.1074/jbc.m006114200.Search in Google Scholar PubMed
34. Bartholin, L, Maguer-Satta, V, Hayette, S, Martel, S, Gadoux, M, Corbo, L, et al.. Transcription activation of FLRG and follistatin by activin a, through smad proteins, participates in a negative feedback loop to modulate activin a function. Oncogene 2002;21:2227–35. https://doi.org/10.1038/sj.onc.1205294.Search in Google Scholar PubMed
35. Thadhani, R, Powe, CE, Tjoa, ML, Khankin, E, Ye, J, Ecker, J, et al.. First-trimester follistatin-like-3 levels in pregnancies complicated by subsequent gestational diabetes mellitus. Diabetes Care 2010;33:664–9. https://doi.org/10.2337/dc09-1745.Search in Google Scholar PubMed PubMed Central
36. Hu, D, Tian, T, Guo, J, Wang, H, Chen, D, Dong, M. Decreased maternal and placental concentrations of follistatin-like 3 in gestational diabetes. Clin Chim Acta 2012;413:533–6. https://doi.org/10.1016/j.cca.2011.10.029.Search in Google Scholar PubMed
37. Fujita, K, Iwama, H, Oura, K, Tadokoro, T, Hirose, K, Watanabe, M, et al.. Metformin-suppressed differentiation of human visceral preadipocytes: involvement of microRNAs. Int J Mol Med 2016;38:1135–40. https://doi.org/10.3892/ijmm.2016.2729.Search in Google Scholar PubMed PubMed Central
38. Anfossi, S, Babayan, A, Pantel, K, Calin, GA. Clinical utility of circulating non-coding RNAs - an update. Nat Rev Clin Oncol 2018;15:541–63. https://doi.org/10.1038/s41571-018-0035-x.Search in Google Scholar PubMed
39. Lu, J, Wu, J, Zhao, Z, Wang, J, Chen, Z. Circulating LncRNA serve as fingerprint for gestational diabetes mellitus associated with risk of macrosomia. Cell Physiol Biochem 2018;48:1012–8. https://doi.org/10.1159/000491969.Search in Google Scholar PubMed
40. Han, L, Chen, C, Lu, X, Song, Y, Zhang, Z, Zeng, C, et al.. Alterations of 5-hydroxymethylcytosines in circulating cell-free DNA reflect retinopathy in type 2 diabetes. Genomics 2021;113:79–87. https://doi.org/10.1016/j.ygeno.2020.11.014.Search in Google Scholar PubMed
41. Nagai, K, Matsubara, T, Mima, A, Sumi, E, Kanamori, H, Iehara, N, et al.. Gas6 induces akt/mTOR-mediated mesangial hypertrophy in diabetic nephropathy. Kidney Int 2005;68:552–61. https://doi.org/10.1111/j.1523-1755.2005.00433.x.Search in Google Scholar PubMed
42. Huang, Y, Jin, C, Zheng, Y, Li, X, Zhang, S, Zhang, Y, et al.. Knockdown of lncRNA MIR31HG inhibits adipocyte differentiation of human adipose-derived stem cells via histone modification of FABP4. Sci Rep 2017;7:8080. https://doi.org/10.1038/s41598-017-08131-6.Search in Google Scholar PubMed PubMed Central
43. Ji, Z, Wang, J, Yang, S, Tao, S, Shen, C, Wei, H, et al.. Graphene oxide accelerates diabetic wound repair by inhibiting apoptosis of ad-MSCs via linc00324/mir-7977/STK4 pathway. FASEB J 2022;36:e22623. https://doi.org/10.1096/fj.202201079rr.Search in Google Scholar
44. Yoon, J, Lee, KJ, Oh, GS, Kim, GH, Kim, SW. Regulation of nampt expression by transcriptional coactivator NCOA6 in pancreatic beta-cells. Biochem Biophys Res Commun 2017;487:600–6. https://doi.org/10.1016/j.bbrc.2017.04.098.Search in Google Scholar PubMed
45. Goyenechea, E, Crujeiras, AB, Abete, I, Martinez, JA. Expression of two inflammation-related genes (RIPK3 and RNF216) in mononuclear cells is associated with weight-loss regain in obese subjects. J Nutrigenet Nutrigenomics 2009;2:78–84. https://doi.org/10.1159/000210452.Search in Google Scholar PubMed
46. Oh, DY, Olefsky, JM. G protein-coupled receptors as targets for anti-diabetic therapeutics. Nat Rev Drug Discov 2016;15:161–72. https://doi.org/10.1038/nrd.2015.4.Search in Google Scholar PubMed
47. Barella, LF, Jain, S, Kimura, T, Pydi, SP. Metabolic roles of g protein-coupled receptor signaling in obesity and type 2 diabetes. FEBS J 2021;288:2622–44. https://doi.org/10.1111/febs.15800.Search in Google Scholar PubMed
48. Takahashi, K, Yoshina, S, Masashi, M, Ito, W, Inoue, T, Shiwaku, H, et al.. Nematode homologue of PQBP1, a mental retardation causative gene, is involved in lipid metabolism. PLoS One 2009;4:e4104. https://doi.org/10.1371/journal.pone.0004104.Search in Google Scholar PubMed PubMed Central
49. Cloutier, P, Poitras, C, Durand, M, Hekmat, O, Fiola-Masson, E, Bouchard, A, et al.. R2TP/prefoldin-like component RUVBL1/RUVBL2 directly interacts with ZNHIT2 to regulate assembly of u5 small nuclear ribonucleoprotein. Nat Commun 2017;8:15615. https://doi.org/10.1038/ncomms15615.Search in Google Scholar PubMed PubMed Central
50. Cohen, E, Avrahami, D, Frid, K, Canello, T, Levy, LE, Zeligson, S, et al.. Snord 3a: a molecular marker and modulator of prion disease progression. PLoS One 2013;8:e54433. https://doi.org/10.1371/journal.pone.0054433.Search in Google Scholar PubMed PubMed Central
51. Vazquez-Jimenez, JG, Corpus-Navarro, MS, Rodriguez-Chavez, JM, Jaramillo-Ramirez, HJ, Hernandez-Aranda, J, Galindo-Hernandez, O, et al.. The increased expression of regulator of g-protein signaling 2 (RGS2) inhibits insulin-induced akt phosphorylation and is associated with uncontrolled glycemia in patients with type 2 diabetes. Metabolites 2021;11. https://doi.org/10.3390/metabo11020091.Search in Google Scholar PubMed PubMed Central
52. Hedbacker, K, Birsoy, K, Wysocki, RW, Asilmaz, E, Ahima, RS, Farooqi, IS, et al.. Antidiabetic effects of IGFBP2, a leptin-regulated gene. Cell Metab 2010;11:11–22. https://doi.org/10.1016/j.cmet.2009.11.007.Search in Google Scholar PubMed
© 2025 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Reviews
- Pharmacologic thromboprophylaxis following cesarean delivery-what is the evidence? A critical reappraisal
- Fetal cardiac diagnostics in Indonesia: a study of screening and echocardiography
- Original Articles – Obstetrics
- Comparative analysis of antidiuretic effects of oxytocin and carbetocin in postpartum hemorrhage prophylaxis: a retrospective cohort study
- Severe thrombocytopenia in pregnancy: a cross-sectional analysis of perinatal and neonatal outcomes across different platelet count categories
- Association of urinary misfolded protein quantification with preeclampsia and adverse pregnancy outcomes: a retrospective case study
- Differentially expressed genes in the placentas with pre-eclampsia and fetal growth restriction using RNA sequencing and verification
- Upregulation of microRNA-3687 promotes gestational diabetes mellitus by inhibiting follistatin-like 3
- Placental elasticity in trisomy 21: prenatal assessment with shear-wave elastography
- Penicillin allergies and selection of intrapartum antibiotic prophylaxis against group B Streptococcus at a safety-net institution
- Assessing high-risk perinatal complications as risk factors for postpartum mood disorders
- Original Articles – Fetus
- Assessment of fetal thymus size in pregnancies of underweight women
- Normal fetal echocardiography ratios - a multicenter cross-sectional retrospective study
- Original Articles – Neonates
- Evaluation of the relationship of fetal lung elastography values with the development of postpartum respiratory distress in late preterm labor cases
- Short Communication
- Radiographic thoracic area in newborn infants with Down’s syndrome
- Letter to the Editor
- Teaching prospective parents basic newborn life support (BNLS) for unplanned out-of-hospital births
Articles in the same Issue
- Frontmatter
- Reviews
- Pharmacologic thromboprophylaxis following cesarean delivery-what is the evidence? A critical reappraisal
- Fetal cardiac diagnostics in Indonesia: a study of screening and echocardiography
- Original Articles – Obstetrics
- Comparative analysis of antidiuretic effects of oxytocin and carbetocin in postpartum hemorrhage prophylaxis: a retrospective cohort study
- Severe thrombocytopenia in pregnancy: a cross-sectional analysis of perinatal and neonatal outcomes across different platelet count categories
- Association of urinary misfolded protein quantification with preeclampsia and adverse pregnancy outcomes: a retrospective case study
- Differentially expressed genes in the placentas with pre-eclampsia and fetal growth restriction using RNA sequencing and verification
- Upregulation of microRNA-3687 promotes gestational diabetes mellitus by inhibiting follistatin-like 3
- Placental elasticity in trisomy 21: prenatal assessment with shear-wave elastography
- Penicillin allergies and selection of intrapartum antibiotic prophylaxis against group B Streptococcus at a safety-net institution
- Assessing high-risk perinatal complications as risk factors for postpartum mood disorders
- Original Articles – Fetus
- Assessment of fetal thymus size in pregnancies of underweight women
- Normal fetal echocardiography ratios - a multicenter cross-sectional retrospective study
- Original Articles – Neonates
- Evaluation of the relationship of fetal lung elastography values with the development of postpartum respiratory distress in late preterm labor cases
- Short Communication
- Radiographic thoracic area in newborn infants with Down’s syndrome
- Letter to the Editor
- Teaching prospective parents basic newborn life support (BNLS) for unplanned out-of-hospital births