Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Case Report
Case Series
Commentary
Editorial
Invited Article
Media & News
Original Article
Review Article
Short Communication
Special Issue on COVID-19 & ART
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Case Report
Case Series
Commentary
Editorial
Invited Article
Media & News
Original Article
Review Article
Short Communication
Special Issue on COVID-19 & ART
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Case Report
Case Series
Commentary
Editorial
Invited Article
Media & News
Original Article
Review Article
Short Communication
Special Issue on COVID-19 & ART
View/Download PDF

Translate this page into:

Invited Article
2025
:6;
20
doi:
10.25259/JRHM_29_2025

Meta-analysis of high-throughput transcriptomics uncovers aberrant endometrial microRNA-messengerRNA networks in recurrent implantation failure

School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India.
Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, West Bengal, India.
Joint first authors
Author image

*Corresponding author: Koel Chaudhury, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India. koel@smst.iitkgp.ac.in

Licence
This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, transform, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Saha B, Sahu AK, Mitra I, Paul P, Chaudhury K. Meta-analysis of high-throughput transcriptomics uncovers aberrant endometrial microRNA-messengerRNA networks in recurrent implantation failure. J Reprod Healthc Med. 2025;6:20. doi: 10.25259/JRHM_29_2025

Abstract

Objectives:

Recurrent implantation failure (RIF) poses a significant challenge in assisted reproductive technology, with a poorly understood molecular mechanism. Endometrial receptivity, central to successful embryo implantation, is governed by various factors, including intricate gene regulatory networks. Emerging evidence implicates dysregulated messenger ribonucleic acid (mRNA) and microRNA (miRNA) interactions in the pathogenesis of RIF, yet comprehensive network-level analysis remains scarce. This meta-analysis systematically integrates mRNA and miRNA expression profiles in the endometrial tissue of RIF women to elucidate the molecular basis of the disease.

Materials and Methods:

We analyzed two mRNA GEO series (GSE) datasets (GSE26787, GSE111974) and two miRNA datasets (GSE71332, GSE108966), encompassing a total of 168 endometrial tissue samples (RIF and controls). Data were pre-processed to correct for batch effects, and differential expression analysis was performed using robust statistical methods. Meta-analytic approaches were applied to identify high-confidence differentially expressed mRNAs (DE-mRNAs) and miRNAs (DE-miRNAs) across the datasets. Validated miRNA–mRNA interactions were retrieved and visualized using network analysis tools.

Results:

We identified 14 key miRNAs and 1670 dysregulated mRNAs from the datasets. Integration revealed a miRNA–mRNA interaction network comprising 2,641 validated interactions (1,086 unique mRNAs and 14 unique miRNAs), with 12 hub genes identified. Pathway enrichment analysis highlighted the phosphatidylinositol 3-kinase-protein kinase B (PI3K-Akt) signaling pathway as the most significantly enriched, underscoring its central role in RIF pathogenesis. The key miRNAs were found to be associated with angiogenesis, immune modulation, and stromal cell function.

Conclusion:

This meta-analysis provides a comprehensive, network-based perspective on the miRNA–mRNA regulatory landscape in RIF. The PI3K-AKT pathway emerged as the central node of dysregulation and warrants further investigation. This work lays the foundation for experimentally validating our findings, aiming at improving pregnancy outcomes in women with a history of RIF.

Keywords

Interaction Network
Messenger RNA
Meta-analysis
Micro RNA
Recurrent implantation failure

INTRODUCTION

Recurrent implantation failure (RIF) is defined as a condition where implantation fails to occur for three or more cycles of in vitro fertilization (IVF) despite the transfer of two or more good-quality embryos in women under 40 years of age European Society of Human Reproduction and Embryology (ESHRE), 2023).[1] Approximately 10% of the couples undergoing IVF are affected by this condition.[2] Some of the known causes leading to RIF are hormonal imbalances, disturbances in angiogenic and immunomodulatory factors, progesterone resistance, genetic polymorphisms, shifted window of receptivity, decreased integrin expression, etc.[3]

The interplay between messenger ribonucleic acid (RNA) (mRNAs) and microRNA (miRNAs) constitutes a fundamental regulatory mechanism that fine-tunes gene expression across diverse tissues and biological processes.[4] Such dynamic regulation is especially vital in the endometrium, as it directly impacts tissue remodeling, angiogenesis, immune modulation, and receptivity to implantation.[5]

The study of miRNA–mRNA interactions in various diseases, such as cancers, is increasingly gaining attention from research groups worldwide; however, such studies in the case of RIF remain markedly limited.[6] To date, only one group has explored such interactions in the context of endometrial angiogenesis in RIF and identified miR-17-5p/HIF1A and miR-29b-3p/vascular endothelial growth factor-A (VEGFA) as key regulatory axes.[7] However, the authors focused largely on mRNA expression profiles as putative biomarkers, neglecting the underlying regulatory networks and their functional consequences. Given the critical role of angiogenesis and other molecular pathways in endometrial receptivity, there is a compelling need to further investigate miRNA–mRNA interactions to advance our mechanistic understanding of the uterine environment in RIF.

This meta-analysis aims to systematically integrate available datasets to establish comprehensive regulatory networks, which will facilitate the identification of potential therapeutic targets and support future translational research in RIF. We have used two mRNA datasets (GSE26787, GSE111974) and two miRNA datasets (GSE71332, GSE108966) to perform differential expression analysis. Each dataset is derived from endometrial tissue samples obtained from women with a history of RIF, with each sample analyzed on a distinct sequencing platform.

MATERIALS AND METHODS

Data acquisition

The Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo) was systematically interrogated to identify mRNA and miRNA expression datasets pertinent to RIF. The search strategy utilized the following key terms: “Recurrent implantation failure,” “Homo sapiens [porgn: txid9609],” and the methodology filters “Expression profiling by array,” “Non-coding RNA profiling by array,” and “Non-coding RNA profiling by high throughput sequencing.”

Two mRNA expression datasets, GSE111974 and GSE26787, were obtained from the GEO database. The GSE111974 dataset, comprising 48 endometrial tissue samples (24 RIF and 24 controls), was generated using Agilent-039494 SurePrint G3 Human GE v2 8x60K Microarray. The second dataset (GSE26787) was generated using Affymetrix Human Genome U133 Plus 2.0 Array and included 10 endometrial tissue samples (5 RIF and 5 controls).

For miRNA, two datasets (GSE71332 and GSE108966) were considered. The GSE71332 dataset was generated on 12 endometrial tissue samples (7 RIF and 5 controls) with Agilent-046064 Unrestricted_Human_miRNA_V19.0_ Microarray. The second miRNA dataset, GSE108966, was generated on Illumina HiSeq 2500 (miRNA V21.0) using 110 endometrium tissue samples (36 RIF and 74 controls). The summary of the datasets is presented in Table 1.

Table 1: Details of the datasets used for this study.
Accession Platform Sample type Sample number
mRNA
  GSE111974 Agilent-039494 SurePrint G3 Human GE v2 8×60K Microarray 039381 Endometrium RIF: 24
Controls: 24
  GSE26787 Affymetrix Human Genome U133 Plus 2.0 Array Endometrium RIF: 5
Controls: 5
miRNA
  GSE71332 Agilent-046064 Unrestricted_Human_miRNA_V19.0_Microarray Endometrium RIF: 7
Controls: 5
  GSE108966 Illumina HiSeq 2500 (Homo sapiens) Endometrium RIF: 36
Controls: 74

mRNA: Messenger RNA, miRNA: MicroRNA, RIF: Recurrent implantation failure

RNA sequencing data

The R statistical programming language (version 4.5.0, 2025-04-11; Bioconductor version 3.2.1) was used for data processing and integration. For mRNA analysis, expression data from GSE111974 and GSE26787 were log2-transformed and quantile-normalized using the limma package.[8] Probes were aggregated to gene-level expression through median values based on platform-specific annotation files.[9] Genes with variance below the 25th percentile across samples were filtered out to reduce noise.[10,11] Batch effects (non-biological differences) were corrected using the ComBat function from the surrogate variable analysis package.[12]

Pre-processing of the GSE71332 miRNA dataset was performed similarly to the mRNA datasets. For the second miRNA dataset (GSE108966), raw count data were retrieved, and DESeqDataSet (a central data structure of the DESeq2 package) was created. The ComBat-Seq function was used for batch correction.[13]

To identify differentially expressed genes (DEGs) between RIF and controls, differential gene expression analysis was performed separately for each dataset using limma and DESeq2, as applicable.

Meta-analysis of differentially expressed RNAs

mRNA expression

A meta-analysis was performed on the common DEGs of the GSE111974 and GSE26787 datasets. For each DEG, logFC values were combined using (i) a fixed-effects model and (ii) integrating p-values with degrees of freedom adjusted for study count by Fisher’s method.[14,15] False discovery rate (FDR) < 0.05 [(i) and (ii) methods], consistent logFC direction, and effect ratio (min|max[logFC]|) > 0.5 were set for high-confidence genes.[14,16]

miRNA expression

Integrative miRNA analysis was performed combining logFC values through (i) fixed-effects model and integrating P-values using (ii) Fisher’s and (iii) weighted Stouffer’s methods.[17] High-confidence miRNAs exhibited FDR < 0.05 across all three methods (i, ii, and iii), consistent effect direction, effect ratio >0.5, and leave-one-out sensitivity (P < 0.05 in both datasets).

Weighted gene co-expression network analysis (WGCNA)

WGCNA, version 1.73 (https://rdocumentation.org/packages/WGCNA/versions/1.73) package in the R language was used to analyze gene modules and genes that correlated highly with RIF. The combined, batch-corrected mRNA expression matrix from GSE111974 and GSE26787 was used for WGCNA. Quality control was performed using the good Samples Genes function from the WGCNA package. A soft-thresholding power was selected based on scale-free topology fit (R2 > 0.85), with a power of 9.[18] An unsigned adjacency matrix was computed, followed by the topological overlap matrix calculation. Hierarchical clustering with dynamic tree cutting identified initial modules, which were merged based on eigengene dissimilarity (cutHeight = 0.25). Module– trait correlations were calculated for RIF and control traits, and the top 10% most connected genes per module were identified.[19]

miRNA–mRNA target integration and network construction

Validated miRNA–mRNA interactions were retrieved using the multiMiR package, restricting targets to high-confidence mRNAs and miRNAs.[20] Only interactions with consistent directionality (negative correlation between miRNA and mRNA logFC) were retained. A network of these miRNA–mRNA interactions was visualized in Cytoscape (version 3.10.2) (https://cytoscape.org/). Further, the overlapping hub genes from WGCNA modules and the miRNA–mRNA interactions were taken into consideration.[18]

Gene ontology (GO) analysis

GO enrichment analysis was performed using the org.Hs.eg. db annotation package from Bioconductor. GO terms were mapped based on the GO database (release: February 6, 2025) and Entrez Gene annotations (sourced: February 22, 2025). The GO file was obtained from the GO consortium (http://current.geneontology.org/ontology/go-basic.obo). All annotations and mappings were performed using R version 4.5.0 and Bioconductor version 3.2.1.

Pathway enrichment analysis

Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment was performed in R using clusterProfiler (v4.16.0), converting gene symbols to ENTREZ IDs through org.Hs.eg.db (v3.21.0).[21] The enrichKEGG() function was applied with organism= “has,” P-value cutoff = 0.05, and q-value cutoff = 0.2, utilizing the most current KEGG database (2024) accessed through the KEGG REST API.[22]

Figure 1 represents summary of the study (1a) and the detailed workflow (1b).

(a): Graphical abstract of the present study. Created in BioRender. Paul, P. (2025) https://BioRender.com/vam5ze8
Figure 1
(a): Graphical abstract of the present study. Created in BioRender. Paul, P. (2025) https://BioRender.com/vam5ze8
(b): Overall workflow of the study. (mRNA: messenger RNA, miRNA: microRNA, RMA: Robust Multi-array Average, DE: Differentially expressed, FDR: False discovery rate, WGCNA: Weighted Gene Co-expression Network Analysis, KEGG: Kyoto Encyclopedia of Genes and Genomes).
Figure 1
(b): Overall workflow of the study. (mRNA: messenger RNA, miRNA: microRNA, RMA: Robust Multi-array Average, DE: Differentially expressed, FDR: False discovery rate, WGCNA: Weighted Gene Co-expression Network Analysis, KEGG: Kyoto Encyclopedia of Genes and Genomes).

RESULTS

We identified 1,670 high-confidence DEGs and 14 high-confidence DEmiRNAs [Figures 2 and 3]. A total of 1,256 validated targets overlapping with the total DEGs were identified. Among these, based on logFC values, 1,086 DEGs exhibited a negative correlation with miRNA–mRNA expression.

Heatmaps of high-confidence DEGs. (a) GSE111974 and (b) GSE26787 show expression levels for RIF (Red) and controls (Blue), with dendrogram clustering. Color intensity indicates the extent of expression (red: High, yellow: Low). DEGs: Differentially expressed genes, RIF: Recurrent implantation failure.
Figure 2:
Heatmaps of high-confidence DEGs. (a) GSE111974 and (b) GSE26787 show expression levels for RIF (Red) and controls (Blue), with dendrogram clustering. Color intensity indicates the extent of expression (red: High, yellow: Low). DEGs: Differentially expressed genes, RIF: Recurrent implantation failure.
Heatmaps of high-confidence DEmiRNAs. (a) GSE71332 and (b) GSE108966 show expression levels for recurrent implantation failure (Red) and controls (Blue), with dendrogram clustering. Color intensity indicates the extent of expression (red: High, yellow: Low). DEmiRNAs: Differentially expressed microRNAs.
Figure 3:
Heatmaps of high-confidence DEmiRNAs. (a) GSE71332 and (b) GSE108966 show expression levels for recurrent implantation failure (Red) and controls (Blue), with dendrogram clustering. Color intensity indicates the extent of expression (red: High, yellow: Low). DEmiRNAs: Differentially expressed microRNAs.

Modules and genes screened by WGCNA

A total of 7,118 highly correlated genes were grouped into eight distinct modules using WGCNA. Each module was represented by a unique color: black, blue, brown, dark gray, dark red, gray, light green, and purple. Among these, four modules (black, brown, dark gray, and light green) were identified as significant, with adjusted P > 0.05. A gene dendrogram illustrating the clustering of modules is shown in Figure 4.

WGCNA results. (a) Cluster dendrogram with dynamic tree cut and merged dynamic modules, showing gene clustering. (b) Module–trait relationships heatmap with BH-adjusted P-values, indicating correlations between modules (e.g., MEblack, MEblue) and recurrent implantation failure/controls traits (red: Positive, blue: Negative). WGCNA: Weighted gene co-expression network analysis, ME: Module Eigengene.
Figure 4:
WGCNA results. (a) Cluster dendrogram with dynamic tree cut and merged dynamic modules, showing gene clustering. (b) Module–trait relationships heatmap with BH-adjusted P-values, indicating correlations between modules (e.g., MEblack, MEblue) and recurrent implantation failure/controls traits (red: Positive, blue: Negative). WGCNA: Weighted gene co-expression network analysis, ME: Module Eigengene.

miRNA–mRNA network construction

A total of 2,641 interactions could be observed in the miRNAs-target network (14 miRNAs and 1,086 targets/ DEGs). Next, we selected the top 500 unique targets based on a combination of score-based sorting and constructed a representative subnetwork of 728 interactions [Figure 5]. Twelve hub genes (LNX2, DUOX1, MFSD6, RANBP17, PEA15, CD38, SLC7A7, UBAP1L, TYROBP, LAPTM5, SRGN, and GPR65) were identified from the intersection between WGCNA-derived genes (n = 70) and 1,086 unique miRNA targets. The complete list of miRNAs and hub genes is provided in Tables 2 and 3. As visualized from the network [Figure 5a and b], hsa-miR-101-3p, hsa-miR-27b-3p, hsa-miR-30e-5p, hsa-miR-301a-3p, hsa-miR-19b-3p, hsa-miR-455-3p, hsa-miR-27a-3p, hsa-miR-449a, hsa-miR-424-5p, hsa-miR-362-3p, hsa-miR-143-3p, hsa-miR-141-3p, hsa-miR-15a-5p, and hsa-miR-6724-5p were found to be associated with GPR65, CD38, SLC7A7 (upregulated hub genes) and LNX2, DUOX1, MFSD6, RANBP17, PEA15 (downregulated hub genes). However, according to the combination score-derived ranking, the remaining four genes (UBAP1L, TYROBP, LAPTM5, and SRGN) do not appear within those 500 interactions.

miRNA-mRNA interaction network. (a) Down-regulated miRNAs (red diamonds) interacting with up regulated mRNAs (yellow ovals), along with the overlapping WGCNA hub genes/mRNA (blue arrowheads). (b) miRNA-mRNA interaction network. Up-regulated miRNAs (red diamonds) interacting with down regulated mRNAs (yellow ovals), along with the overlapping WGCNA hub genes/mRNA (green arrowheads). mRNA: Messenger RNA, miRNA: MicroRNA.
Figure 5:
miRNA-mRNA interaction network. (a) Down-regulated miRNAs (red diamonds) interacting with up regulated mRNAs (yellow ovals), along with the overlapping WGCNA hub genes/mRNA (blue arrowheads). (b) miRNA-mRNA interaction network. Up-regulated miRNAs (red diamonds) interacting with down regulated mRNAs (yellow ovals), along with the overlapping WGCNA hub genes/mRNA (green arrowheads). mRNA: Messenger RNA, miRNA: MicroRNA.
Table 2: Complete list of differentially expressed microRNAs.
Differentially expressed miRNAs (DE-miRNAs)
1. hsa-miR-101-3p Up-regulated
2. hsa-miR-27b-3p Up-regulated
3. hsa-miR-30e-5p Up-regulated
4. hsa-miR-301a-3p Up-regulated
5. hsa-miR-19b-3p Up-regulated
6. hsa-miR-455-3p Up-regulated
7. hsa-miR-27a-3p Up-regulated
8. hsa-miR-449a Up-regulated
9. hsa-miR-424-5p Up-regulated
10. hsa-miR-362-3p Up-regulated
11. hsa-miR-143-3p Up-regulated
12. hsa-miR-141-3p Up-regulated
13. hsa-miR-15a-5p Down-regulated
14. hsa-miR-6724-5p Down-regulated

RNA: Ribonucleic acid

Table 3: Complete list of the key hub genes identified.
Key hub genes identified from meta-analysis
1. DUOX1 Dual oxidase 1 Up-regulated
2. PEA15 Phosphoprotein enriched in astrocytes-15 Up-regulated
3. LAPTM5 Lysosomal protein transmembrane 5 Down-regulated
4. MFSD6 Major facilitator superfamily domain containing 6 Up-regulated
5. LNX2 Ligand of numb-protein X 2 Up-regulated
6. UBAP1L Ubiquitin associated protein 1 like Down-regulated
7. CD38 Cluster of differentiation 38 Down-regulated
8. GPR65 G protein-coupled receptor 65 Down-regulated
9. TYROBP TYRO protein tyrosine kinase-binding protein Down-regulated
10. SRGN Serglycin Down-regulated
11. RANBP17 RAN binding protein 17 Up-regulated
12. SLC7A7 Solute carrier family 7 Down-regulated

GO analysis

GO enrichment analysis of the DEGs revealed significant clustering in cellular components such as the early endosome, apical part of the cell, cell cortex, and various synaptic and membrane-associated structures, suggesting roles in intracellular trafficking and cell polarity [Figure 6a]. For molecular function, cell adhesion and cell–cell adhesion mediator activity were notably enriched, highlighting the importance of adhesion mechanisms in endometrial biology [Figure 6b]. In terms of biological processes, the most prominent enrichments included wound healing, cell-substrate adhesion, regulation of actin filament-based processes, and several coagulation-related pathways, collectively indicating that disruptions in cellular organization, adhesion, and tissue repair may contribute to the molecular pathology of RIF [Figure 6c].

GO enrichment associated with the DEGs. (a) Cellular components (b) Molecular functions (c) Biological processes (d) KEGG pathway; pathways with gene ratio (number of input genes associated with the term/total input genes) and p.adjust; circle size represents gene count, color intensity reflects p.adjust values (red: Lower, blue: Higher). GO: Gene ontology, KEGG: Kyoto encyclopedia of genes and genomes, TNF: Tumor Necrosis Factor, TOR: Target of Rapamycin, GTP: Guanosine triphosphate.
Figure 6:
GO enrichment associated with the DEGs. (a) Cellular components (b) Molecular functions (c) Biological processes (d) KEGG pathway; pathways with gene ratio (number of input genes associated with the term/total input genes) and p.adjust; circle size represents gene count, color intensity reflects p.adjust values (red: Lower, blue: Higher). GO: Gene ontology, KEGG: Kyoto encyclopedia of genes and genomes, TNF: Tumor Necrosis Factor, TOR: Target of Rapamycin, GTP: Guanosine triphosphate.

Identification of pathways significantly associated with differentially expressed mRNAs

KEGG pathway enrichment analysis revealed considerable involvement of key signaling pathways [Figure 6d]. The analysis showed significant enrichment of 15 pathways (adjusted P < 0.05), with the PI3K-Akt signaling pathway emerging as the most prominently enriched pathway (GeneRatio = 0.07, adjusted P < 0.01, gene count = 35). Other significantly enriched pathways included human papillomavirus infection, apelin signaling, focal adhesion, insulin resistance, Ras, and mTOR signaling. Notably, leukocyte transendothelial migration and TNF signaling pathways, directly relevant to endometrial biology and implantation, could be identified. It is well established that these pathways are associated with immune modulation and inflammatory responses critical for embryo-endometrial interactions. The Hippo signaling pathway and focal adhesion pathway were also found to be enriched, suggesting involvement in cell adhesion and tissue remodeling processes, essential for endometrial receptivity. These results collectively highlight the central role of cell survival, proliferation, and vascularization pathways in the pathogenesis of RIF, with the PI3K-Akt pathway representing a key regulatory node in RIF-associated molecular dysfunction.

DISCUSSION

This study provides novel insights into the molecular underpinnings of RIF and underscores the importance of miRNA–mRNA interactions in endometrial biology. Meta-analysis of endometrial transcriptomic datasets on women with RIF identified a distinct set of dysregulated mRNAs and miRNAs, highlighting their possible role in endometrial receptivity. Further, integration of high-throughput mRNA and miRNA expression profiles revealed a comprehensive interaction network involving 14 key miRNAs and 1,086 unique mRNAs, with 12 overlapping hub genes (LNX2, DUOX1, MFSD6, RANBP17, PEA15, CD38, SLC7A7, UBAP1L, TYROBP, LAPTM5, SRGN, and GPR65) at the core of these regulatory relationships.

Out of the 192 miRNAs identified, 14 miRNAs (hsa-miR-101-3p, hsa-miR-27b-3p, hsa-miR-30e-5p, hsa-miR-301a-3p, hsa-miR-19b-3p, hsa-miR-455-3p, hsa-miR-27a-3p, hsa-miR-449a, hsa-miR-424-5p, hsa-miR-362-3p, hsa-miR-143-3p, hsa-miR-141-3p, hsa-miR-15a-5p, and hsa-miR-6724-5p) were differentially expressed in the endometrium of RIF patients. These miRNAs are implicated in various biological processes relevant to implantation, including angiogenesis, immune modulation, cell proliferation, and apoptosis. For instance, the role of miR-301a-3p is evidenced in proliferation, epithelial-mesenchymal transition (EMT), and cell cycle progression through the PTEN and H19/GAS1 axis.[23] While miR-101-3p modulates inflammation and tissue remodeling through COX-2 and MMPs, miR-19b-3p converges on PTEN and PI3K/AKT signaling, impairing trophoblast function and immune balance.[24,25] The miR-362-3p controls proliferation and migration by regulating the FOXO3/AKT pathway.[26] The two miRNAs, miR-455-3p and miR-424-5p, are known to support proliferation, differentiation, angiogenesis, and apoptosis.[27,28] In addition, miR-143-3p and miR-141-3p influence EMT and stromal cell function; both processes are critical for successful embryo attachment.[29,30] Further, miR-27a-3p and miR-27b-3p are reported to drive decidualization, angiogenesis, and remodeling of the endometrium.[31,32] While miR-30e-5p also influences EMT, angiogenesis, and cell migration, miR-449a regulates stromal apoptosis and formation of pinopodes.[33,34] In addition, miR-15a-5p exerts tumor-suppressive effects, and hsa-miR-6724-5p is linked to oxidative stress responses and metabolic regulation.[35,36] Table 4 summarizes the 14 dysregulated miRNAs, their potential targets, and the relevant biological processes involved.

Table 4: Details of the 14 dysregulated microRNAs, their potential targets, and the relevant biological processes involved.
Sl. No. miRNAs Targets (genes/proteins/pathways) Relevant biological processes References
1 miR-101-3p COX-2 (PTGS2), EZH2, MMPs, DUSP1 Inflammation, Epithelial-mesenchymal transition, tissue remodeling, trophoblast invasion Harati et al.[24]
2 miR-27b-3p IGFBP1, EFNA1, HIF1A, ANGPTL2, MMP13 Angiogenesis, endometrial remodeling, decidualization Reed et al.[32]
3 miR-30e-5p SNAI2, VEGFA, HGF, GRP78, SOX9, CDH1 EMT, angiogenesis, endometrial receptivity, cell migration Zhang et al.[33]
4 miR-301a-3p PTEN, BTG1, TP63, H19/GAS1 axis Proliferation, EMT, endometrial invasiveness, cell cycle, endometrial receptivity Zhang and Liu[23]
5 miR-19b-3p PTEN, PIK3R1, AKT1, TNFAIP3, SOCS3 Immune modulation, spermatogenesis, embryo quality, cell proliferation Wang et al.[25]
6 miR-455-3p SMAD2/3, RUNX2, SOCS3, HDAC2 Proliferation, differentiation, chromatin regulation, tissue remodeling Zhan et al.[27]
7 miR-27a-3p IGFBP1, DKK1, BTG1, LIF, NCOA3 Decidualization, endometrial remodeling, receptivity, cell adhesion Di Pietro et al.[31]
8 miR-449a HDAC1, CDK6, E2F3, Bcl-2, Cdk2 Stromal cell apoptosis, pinopode formation, receptivity, cell cycle An et al.[34]
9 miR-424-5p SPP1, SGK2, ANG, FGFR1, CDC25A Cell adhesion, angiogenesis, apoptosis, migration Xuan et al.[28]
10 miR-362-3p E2F1, AKT, SERBP1, FOXO3, CDK6 Proliferation, FOXO3/Protein Kinase B (PKB) pathway, migration, cell cycle progression Zhu et al.[26]
11 miR-143-3p SOX5, HOXA10, MAP3K7, SERPINE1 EMT, proliferation, immune modulation, apoptosis Yang et al.[29]
12 miR-141-3p Keap1, Nrf2 Autophagy regulation, tumor-stroma interactions Liang et al.[30]
13 miR-15a-5p CXCL10, LIN28a, p-ERK Tumor suppression Weissman et al.[35]
14 miR-6724-5p - Bladder cancer progression Doyle et al.[36]

Interestingly, a direct association between the hub genes and the dysregulated miRNAs could be established. The miR-101-3p directly regulates immune-related hub genes such as CD38, which is a calcium-signaling molecule implicated in immune regulation and endometrial motility.[37] It also targets the SRGN/LAPTM5 axis, which participates in extracellular matrix remodeling.[38] The expression of miR-15a-5p could be associated with a number of hub genes identified in the present study. This miRNA is known to regulate DUOX1, which is linked to redox homeostasis.[39] Further, it targets PEA15, underlining its role in apoptosis and integrin signaling. Furthermore, the association of miR-15a-5p with the hub genes MFSD6, LNX2, RANBP17, and UBAP1L highlights its potential to influence transcriptional regulation and protein turnover. Two miRNAs, miR-19b-3p and miR-301a-3p, converged on GPR65, a pH-sensing receptor associated with immune cell activity and inflammatory responses.[40] Interestingly, both these processes are considered essential for successful implantation. TYROBP, a hub gene with a major role in NK-cell-mediated signaling, was found to be targeted by miR-424-5p, miR-449a, along with the miR-27 family (miR-27a-3p, miR-27b-3p).[41] The interactions reported here are supported by experimental validation from diverse global and targeted studies, curated in the Tarbase database.

Pathway enrichment analysis indicated the PI3K-Akt signaling pathway to be most significantly enriched, followed by pathways related to cell survival, proliferation, immune modulation, and vascularization. Enrichment of these pathways aligns with the known importance of cell survival, angiogenesis, and immune regulation in making the endometrium receptive and facilitating successful implantation. Recent studies have shown that dysregulated miRNA–mRNA networks are associated with impaired endometrial receptivity and embryo implantation in women with various uterine disorders.[42] Figure 7 illustrates the key findings of this work.

Overall findings of the present study. (ECM; Extracellular matrix, EMT; Epithelialmesenchymal transition)” Created in BioRender. Paul, P. (2025) https://BioRender.com/mdcz1js.
Figure 7:
Overall findings of the present study. (ECM; Extracellular matrix, EMT; Epithelialmesenchymal transition)” Created in BioRender. Paul, P. (2025) https://BioRender.com/mdcz1js.

In the context of advancing transcriptomic technologies, recent literature has highlighted the value of single-cell RNA sequencing (scRNA-seq) in understanding uterine pathologies. An excellent review underscores how scRNA-seq studies deepen our understanding of cellular interactions and disease mechanisms in uterine disorders such as endometriosis, adenomyosis, fibroids, and endometrial cancer, offering potential for novel treatment strategies.[43] Similarly, Kirschen and co-workers provide a comprehensive overview of how transcriptomic approaches, including bulk RNA sequencing and multi-omics, elucidate gene regulation, non-coding RNAs, and disease-specific signatures across the menstrual cycle and various uterine pathologies, including RIF.[44]

The dysregulated miRNAs and mRNAs identified in the present study represent potential biomarkers of RIF and highlight the need to conduct further mechanistic studies to validate their specific roles in endometrial receptivity. The integration of transcriptomic data from multiple datasets and the construction of a robust interaction network provide a foundation for future research aimed at elucidating the molecular mechanisms underlying RIF and developing targeted therapeutic strategies.

Limitations

This study is associated with a few limitations. First, identification of potential biomarkers is primarily driven by statistical analysis; validation in a patient cohort could not be performed. Second, the number of relevant studies publicly available is limited, and the sample size within these studies is relatively small. This constraint could have affected the statistical power and generalizability of our findings. These limitations underline the importance of future studies with a large, well-matched patient population to better elucidate the complex molecular mechanisms underlying RIF.

CONCLUSION

In summary, this meta-analysis demonstrates that RIF is associated with altered expression of specific endometrial mRNAs and miRNAs, with their pathways largely related to cell survival, proliferation, immune modulation, and vascularization. This study also suggests that miRNA–mRNA regulatory networks could serve as a useful framework for understanding RIF. The identified hub genes and miRNAs hold significant potential as candidates for further research, opening up the possibility of more accurate diagnostic and therapeutic strategies for RIF.

Acknowledgments:

The authors gratefully acknowledge ICMR for providing financial assistance (Project sanction number: Dev/ SG-00349/2024) and IIT Kharagpur for providing resources for carrying out this work.

Ethical approval:

Institutional Review Board approval is not required.

Declaration of patient consent:

Patient’s consent not required as there are no patients in this study.

Conflicts of interest:

There are no conflicts of interest.

Use of artificial intelligence (AI)-assisted technology for manuscript preparation:

The authors confirm that they have used artificial intelligence (AI) assisted technology for assisting in rephrasing, grammar correction in the manuscript only.

Financial support and sponsorship: Nil.

References

  1. , , , , , , et al. ESHRE good practice recommendations on recurrent implantation failure. Hum Reprod Open. 2023;2023:hoad023.
    [CrossRef] [PubMed] [Google Scholar]
  2. , , . Recurrent implantation failure: A comprehensive summary from etiology to treatment. Front Endocrinol (Lausanne). 2023;13:1061766.
    [CrossRef] [PubMed] [Google Scholar]
  3. , , . Biomolecular markers of recurrent implantation failure-A review. Int J Mol Sci. 2021;22:10082.
    [CrossRef] [PubMed] [Google Scholar]
  4. , , , , , . miRNA interplay: Mechanisms and therapeutic interventions in cancer. MedComm Oncol. 2024;3:e93.
    [CrossRef] [Google Scholar]
  5. . The role of microRNAs in regulatory T cells and in the immune response. Immune Netw. 2011;11:11.
    [CrossRef] [PubMed] [Google Scholar]
  6. , , , , , , et al. Comprehensive analysis of miRNA-mRNA regulatory pairs associated with colorectal cancer and the role in tumor immunity. BMC Genomics. 2023;24:724.
    [CrossRef] [PubMed] [Google Scholar]
  7. , . Comprehensive analysis of circRNAmiRNA-mRNA network related to angiogenesis in recurrent implantation failure. BMC Med Genomics. 2024;17:193.
    [CrossRef] [PubMed] [Google Scholar]
  8. , , , , , , et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.
    [CrossRef] [PubMed] [Google Scholar]
  9. , , , , , , et al. Strategies for aggregating gene expression data: The collapseRows R function. BMC Bioinformatics. 2011;12:322.
    [CrossRef] [PubMed] [Google Scholar]
  10. , , , , , , et al. Variance of gene expression identifies altered network constraints in neurological disease. PLoS Genet. 2011;7:e1002207.
    [CrossRef] [PubMed] [Google Scholar]
  11. , . Filtering for increased power for microarray data analysis. BMC Bioinformatics. 2009;10:11.
    [CrossRef] [PubMed] [Google Scholar]
  12. , , , , . The SVA package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28:882-3.
    [CrossRef] [PubMed] [Google Scholar]
  13. , , . ComBat-seq: Batch effect adjustment for RNA-seq count data. NAR Genom Bioinform. 2020;2:lqaa078.
    [CrossRef] [PubMed] [Google Scholar]
  14. , , , , , , et al. An R package suite for microarray meta-analysis in quality control, differentially expressed gene analysis and pathway enrichment detection. Bioinformatics. 2012;28:2534-6.
    [CrossRef] [PubMed] [Google Scholar]
  15. . Combining probability from independent tests: The weighted Z-method is superior to Fisher's approach. J Evol Biol. 2005;18:1368-73.
    [CrossRef] [PubMed] [Google Scholar]
  16. , , . Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.
    [CrossRef] [PubMed] [Google Scholar]
  17. . Optimally weighted Z-test is a powerful method for combining probabilities in meta-analysis. J Evol Biol. 2011;24:1836-41.
    [CrossRef] [PubMed] [Google Scholar]
  18. , . WGCNA: An R package for weighted correlation network analysis. BMC Bioinformatics. 2008;9:559.
    [CrossRef] [PubMed] [Google Scholar]
  19. , , . Defining clusters from a hierarchical cluster tree: The dynamic tree cut package for R. Bioinformatics. 2008;24:719-20.
    [CrossRef] [PubMed] [Google Scholar]
  20. , , , , , , et al. The multiMiR R package and database: Integration of microRNA-target interactions along with their disease and drug associations. Nucleic Acids Res. 2014;42:e133.
    [CrossRef] [PubMed] [Google Scholar]
  21. , , , , , , et al. ClusterProfiler 4.0 A universal enrichment tool for interpreting omics data. Innovation (Camb). 2021;2:100141.
    [CrossRef] [PubMed] [Google Scholar]
  22. , , , , . KEGG for taxonomy-based analysis of pathways and genomes. Nucleic Acids Res. 2023;51:D587-92.
    [CrossRef] [PubMed] [Google Scholar]
  23. , . MicroRNA (MiR)-301a-3p regulates the proliferation of esophageal squamous cells via targeting PTEN. Bioengineered. 2020;11:972-83.
    [CrossRef] [PubMed] [Google Scholar]
  24. , , , , . Loss of miR-101-3p promotes transmigration of metastatic breast cancer cells through the brain endothelium by inducing COX-2/MMP1 signaling. Pharmaceuticals. 2020;13:144.
    [CrossRef] [PubMed] [Google Scholar]
  25. , , . miR-19-3p targets PTEN to regulate cervical cancer cell proliferation, invasion, and autophagy. Genet Res (Camb). 2023;2023:4784500.
    [CrossRef] [PubMed] [Google Scholar]
  26. , , , , . SP1/AKT/FOXO3 signaling is involved in miR-362-3p-mediated inhibition of cell-cycle pathway and EMT progression in renal cell carcinoma. Front Cell Dev Biol. 2020;8:297.
    [CrossRef] [PubMed] [Google Scholar]
  27. , , , , , , et al. miR-455-3p functions as a tumor suppressor by restraining Wnt/β-Catenin signaling via TAZ in pancreatic cancer. Cancer Manag Res. 2020;12:1483-92.
    [CrossRef] [PubMed] [Google Scholar]
  28. , , , . Sequence requirements for miR-424-5p regulating and function in cancers. Int J Mol Sci. 2022;23:4037.
    [CrossRef] [PubMed] [Google Scholar]
  29. , , , , . miR-143-3p inhibits endometriotic stromal cell proliferation and invasion by inactivating autophagy in endometriosis. Mol Med Rep. 2021;23:356.
    [CrossRef] [PubMed] [Google Scholar]
  30. , , . Role of microRNAs in embryo implantation. Reprod Biol Endocrinol. 2017;15:90.
    [CrossRef] [PubMed] [Google Scholar]
  31. , , , , , , et al. miR-27a-3p and miR-124-3p, upregulated in endometrium and serum from women affected by chronic endometritis, are new potential molecular markers of endometrial receptivity. Am J Reprod Immunol. 2018;80:e12958.
    [CrossRef] [PubMed] [Google Scholar]
  32. , , , , , . Estrogen-regulated miRNA-27b is altered by bisphenol A in human endometrial stromal cells. Reproduction. 2018;156:559-67.
    [CrossRef] [PubMed] [Google Scholar]
  33. , , , , , , et al. miR-30e-5p represses angiogenesis and metastasis by directly targeting AEG-1 in squamous cell carcinoma of the head and neck. Cancer Sci. 2020;111:356-68.
    [CrossRef] [PubMed] [Google Scholar]
  34. , , , , , , et al. miR-449a regulates caprine endometrial stromal cell apoptosis and endometrial receptivity. Sci Rep. 2017;7:12248.
    [CrossRef] [PubMed] [Google Scholar]
  35. , , , , , , et al. MicroRNA-15a-5p acts as a tumor suppressor in histiocytosis by mediating CXCL10-ERK-LIN28a-let-7 axis. Leukemia. 2022;36:1139-49.
    [CrossRef] [PubMed] [Google Scholar]
  36. , , , , , , et al. The TGFβ induced microRNAome of the trabecular meshwork. Cells. 2024;13:1060.
    [CrossRef] [PubMed] [Google Scholar]
  37. , , , , . miR-101: An important regulator of gene expression and tumor ecosystem. Cancers (Basel). 2022;14:5861.
    [CrossRef] [PubMed] [Google Scholar]
  38. , , , , , , et al. Identification of miR-101-3p targets and functional features based on bioinformatics, meta-analysis and experimental verification in hepatocellular carcinoma. Am J Transl Res. 2017;9:2088-105.
    [Google Scholar]
  39. , , . DUOX1 in mammalian disease pathophysiology. J Mol Med. 2021;99:743-54.
    [CrossRef] [PubMed] [Google Scholar]
  40. , , , , , , et al. Tissue macrophages: Origin, heterogeneity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther. 2025;10:93.
    [CrossRef] [PubMed] [Google Scholar]
  41. , , , , , , et al. MicroRNAs in cancer immunology: Master regulators of the tumor microenvironment and immune evasion, with therapeutic potential. Cancers (Basel). 2025;17:2172.
    [CrossRef] [PubMed] [Google Scholar]
  42. , , , , , , et al. Uterine fluid microRNAs are dysregulated in women with recurrent implantation failure. Hum Reprod. 2022;37:734-46.
    [CrossRef] [PubMed] [Google Scholar]
  43. , , , . Mapping human uterine disorders through single-cell transcriptomics. Cells. 2025;14:156.
    [CrossRef] [PubMed] [Google Scholar]
  44. , , , , , . Uterine transcriptome: Understanding physiology and disease processes. Biology (Basel). 2023;12:634.
    [CrossRef] [PubMed] [Google Scholar]
Show Sections