Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

RNA sequencing-based transcriptome analysis of granulosa cells from follicular fluid: Genes involved in embryo quality during in vitro fertilization and embryo transfer

  • Eun Jeong Yu ,

    Contributed equally to this work with: Eun Jeong Yu, Won Yun Choi

    Roles Data curation, Writing – original draft, Writing – review & editing

    Affiliations CHA fertility Center Gangnam, CHA University, Seoul, Korea, CHA Fertility Center Seoul Station, CHA University, Seoul, Korea

  • Won Yun Choi ,

    Contributed equally to this work with: Eun Jeong Yu, Won Yun Choi

    Roles Methodology, Writing – original draft

    Affiliations CHA fertility Center Gangnam, CHA University, Seoul, Korea, CHA Fertility Center Ilsan Woman’s & Children’s General Hospital, CHA University, Gyeonggi-do, Korea

  • Mi Seon Park,

    Roles Data curation, Formal analysis, Visualization

    Affiliation CHA fertility Center Gangnam, CHA University, Seoul, Korea

  • Jin Hee Eum,

    Roles Data curation, Formal analysis

    Affiliation CHA fertility Center Gangnam, CHA University, Seoul, Korea

  • Dong Ryul Lee,

    Roles Conceptualization

    Affiliations CHA fertility Center Gangnam, CHA University, Seoul, Korea, Department of Biomedical Science, CHA University, Gyeonggi-do, Korea

  • Woo Sik Lee,

    Roles Conceptualization, Methodology

    Affiliation CHA fertility Center Gangnam, CHA University, Seoul, Korea

  • Sang Woo Lyu,

    Roles Conceptualization, Methodology, Writing – original draft, Writing – review & editing

    Affiliation CHA fertility Center Gangnam, CHA University, Seoul, Korea

  • Sook Young Yoon

    Roles Conceptualization, Data curation, Formal analysis, Visualization, Writing – review & editing

    syyoon11@cha.ac.kr

    Affiliation CHA fertility Center Gangnam, CHA University, Seoul, Korea

Abstract

Background

Granulosa cells play an important role in folliculogenesis, however, the role of RNA transcripts of granulosa cells in assessing embryo quality remains unclear. Therefore, we aims to investigate that RNA transcripts of granulosa cells be used to assess the probability of the embryonic developmental capacity.

Methods

This prospective cohort study was attempted to figure out the probability of the embryonic developmental capacity using RNA sequencing of granulosa cells. Granulosa cells were collected from 48 samples in good-quality embryo group and 79 in only poor- quality embryo group from women undergoing in vitro fertilization and embryo transfer treatment. Three samples from each group were used for RNA sequencing.

Results

226 differentially expressed genes (DEGs) were related to high developmental competence of embryos. Gene Ontology enrichment analysis indicated that these DEGs were primarily involved in biological processes, molecular functions, and cellular components. Additionally, pathway analysis revealed that these DEGs were enriched in 13 Kyoto Encyclopedia of Genes and Genomes pathways. Reverse transcription quantitative polymerase chain reaction verified the differential expression of the 13 selected DEGs. Among them,10 genes were differently expressed in the poor-quality embryo group compared to good-quality embryo group, including CSF1R, CTSH, SERPINA1, CYP27A1, ITGB2, IL1β, TNF, TAB1, BCL2A1, and CCL4.

Conclusions

RNA sequencing data provide the support or confute granulosa expressed genes as non-invasive biomarkers for identifying the embryonic developmental capacity.

Introduction

The ability to identify oocyte quality remains one of the most significant challenges in assisted reproductive technology. The quality of oocyte is an important predictor of implantation and live birth [1, 2]. The presence of a mature and high-quality oocyte plays an essential role in the development of a high-quality embryo [3]. This means that the selection of high-quality embryos begins at the time of oocyte selection.

Embryo quality is a strong indicator for the success rate of in vitro fertilization (IVF) program, as the live birth rate increases when good quality embryos are transferred [4]. The incidence of only poor-quality embryos is usual in IVF cycles of patients with advanced maternal age and in low responding patients. According to Semondade et al. [5], the incidence rate of only poor embryos is approximately 10% at the first IVF cycle, and the recurrence rate is 3%.

Morphological features and development rates are key indicators for oocyte and embryo selection during IVF. These methods are familiar for both clinician and embryologist and are used as the standard method in embryos selection step. However, the usefulness of their assessment is being questioned because of personal bias from the embryologist [6]. In recent years, techniques for embryo selection that provide chromosomal analysis to improve clinical pregnancy have recently been developed, such as preimplantation genetic testing (PGT) [7]. Although PGT is a strong predictor for implantation, it is expensive and requires invasive embryo biopsy, which involves technical expertise [8]. Therefore, there is a high demand for a non-invasive and easy-to-perform screening tools to improve the selection of the most pregnancy-competent embryo.

Good-quality and mature oocyte in IVF are important for fertilization and embryo development [9]. Oocyte maturation occurred during folliculogenesis through orchestrated cross-talk between the oocyte and granulosa cells (GCs). Therefore, it is advocated that the functions of GCs indirectly reflect oocyte developmental competence. GCs can be easily recovered in large quantities during oocyte collection. Thus, the gene expression analysis in GCs could provide a non-invasive assessment for identifying the most competent oocytes and embryos. However, the transcriptomic analysis of GC used to identify embryo quality is still controversial. While some studies have identified candidate genes expressed in GC that could be expected as biomarkers of oocyte and embryo quality [10, 11] and successful clinical outcomes [1214], others have reported that there are no significant differences in gene expression between embryos that did or did not successfully implant [15, 16].

Therefore, the present study aims to investigate and compare the GC transcriptomic obtained from subjects producing good-quality embryos and those producing only poor quality embryos in human IVF. We carried out RNA sequencing (RNA-seq) of GCs isolated from follicular fluid to identify novel gene transcription factors correlated with embryo quality among the embryos with good quality.

Materials and methods

Study population

This prospective cohort study was conducted between January 2019 and February 2021 at the CHA Fertility Center Gangnam. The study was approved by the CHA University Gangnam CHA hospital institutional review board (GCI-19-10, May 15, 2019), Republic of Korea. All women gave written informed consent to provide material for this study. All procedures followed the rules for studies with human-origin materials established by the IRB. Of 141 embryos transfer (ETs), 48 were good quality (GQ) and 79 were poor quality (PQ) ETs; 14 cases of GQ and PQ ETs cases with endometriosis, endometrial pathologies, uterine fibroids or hydrosalpinx were excluded. Couples with severe male factor infertility as defined based on severe oligozoospermia (<5 million sperm/mL) or a history of testicular biopsy were excluded.

Ovarian stimulation and embryo transfer procedures

Patients underwent controlled ovarian stimulation using either the midluteal long gonadotropin-releasing hormone (GnRH) agonist protocol or GnRH antagonist protocol. Gonadotropin doses were individualized according to patients’ age, anti-Mullerian hormone level, antral follicle count, and previous response to stimulation. Cycle monitoring with transvaginal ultrasonography and serum estradiol measurement, was continued until hCG administration. When at least two follicles reached 18 mm in diameter, 250–500 μg of recombinant hCG was administered for final oocyte maturation. Transvaginal ultrasound-guided oocyte-retrieval under conscious sedation was carried out 34–36 hours after hCG administration. The mature oocytes were inseminated using intracytoplasmic sperm insemination (ICSI). One or two embryos were transferred to each patient, and ETs occurred on day 3, 4, or 5. All patients received luteal phase support with progesterone after ET until 8–10 weeks after pregnancy.

Isolation of GCs and assessment of embryo quality

Follicular fluids were aspirated and pooled for each patient during oocyte-retrieval. Follicle aspirates, which were not clear and were contaminated with endometriosis cysts, were discarded. Granulosa cells recovered from one woman were used as one sample. The 127 samples were divided into two groups according to the patient’s embryo quality; 48 patients with good quality blastocysts having a grade of at least 3BB and 97 patients with only poor quality blastocysts.

The follicular fluid was centrifuged at 1000 x g for 20 min to separate erythrocytes, leukocytes and GCs. The cell pellets were washed in RBC lysis buffer (Roche Diagnostics, Basel, Switzerland) for RBC removal and centrifuged at 500g for 10 min and separated enzymatically with instigation at 37°C for 30 min in the enzyme solution [Hanks’ balanced salt solution (HBSS, Gibco, Grand Island, NY) containing 0.5 mg/ml collagenase (type IV; Gibco) and 0.25mg/ml dispase II (neutral protease, grade III, Roche)]. The suspension of GCs was washed and filtered through a 40μm mesh (BD, Franklin Lakes, NJ). GCs using TRIzol Reagent (Life Technologies, San Diego, CA, USA) were stored -75°C until analysis.

Embryo grading was evaluated by two embryologists and was assessed by another senior embryologist with over at least ten years of work experience before embryo transfer. For standardization, all embryologists at our center were trained in embryo grading by the same laboratory director. Embryo quality assessment was carried out according to previously described protocol [17]. Briefly, good quality embryos were identified using the following characteristics. Cleavage embryos were classified as GQ according to Cummins criteria [18, 19]. Blastocyst quality was assessed regarding to the degree of blastocoel expansion, inner cell mass (ICM) and trophectoderm (TE) morphology on day 5 or 6 [20].

RNA isolation and NGS library preparation

Total RNA was isolated from cells using TRIzol Reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions. RNA integrity was checked by using an Agilent 2100 BioAnalyzer (Agilent, CA, USA) with an RNA integrity number value greater than 6. Only qualified samples were used for RNA library constitution. The libraries were arranged for 151 bp paired-end sequencing by the TruSeq stranded mRNA sample preparation kit (Illumina, CA, USA). After the sequential process of end repair, A-tailing, and adapter ligation, cDNA libraries were amplified by Polymerase Chain Reaction (PCR). They were quantified using the KAPA library quantification kit (Kapa Biosystems, MA, USA) following the manufacturer’s quantification protocol. After cluster amplification of denatured templates, sequencing was carried out as paired-end (2×151bp) using IlluminaNovaSeq6000 (Illumina, CA, USA).

Transcriptome data analysis

DEG analysis.

Based on the estimated read counts in the previous step, DEGs were screened using the R package TCCv.1.26.0 [21]. The TCC package uses well-set normalization strategies to compare the tagcount data. Normalization factors were estimated by the iterative DESeq2 [22] /edgeR method [23]. The Q-value was assessed based on the p-value using the p.adjust function of the R package by default parameter programs. The DEGs were evaluated with a Q- value threshold of < 0.05 for fixing errors due to multiple testing.

GO analysis.

The GO database indicates a set of hierarchically controlled vocabulary classified into three categories: biological process, cellular component, and molecular function. For functional characterization of the DEGs, GO-based trend test was assessed by the R package called GOseq [24] through the Wallenius non-central hypergeometric distribution. Selected genes with P-values < 0.05 following the test were regarded as statistically significant.

RT-qPCR

Approximately 1 μg RNA from each sample was reverse transcribed to cDNA using a SensiFAST™ cDNA Synthesis Kit (Meridian Bioscience, Tennessee, USA) following the manufacturer’s instructions. Thirteen genes were selected for validation from the list of DEGs. Primers were designed using the Primer3 primer-design program (version 0.4.0; http://bioinfo.ut.ee/primer3-0.4.0/) (S1 Table). The qPCR reactions were performed on the CFX Connect Real-Time PCR detection System (Bio-Rad Laboratories, Inc., USA) using iQ™ SYBR® Green Supermix (Bio-Rad Laboratories, Inc., USA). Relative quantification analyses were carried out using the comparative CT method, and relative gene expression levels were calculated using the 2−ΔΔCT method.

Clinical outcome and statistics

The primary outcomes of the study were clinical pregnancy outcomes, including clinical pregnancy rate, and ongoing pregnancy rate. Clinical pregnancy was defined as the presence of at least one gestational sac with a fetal heartbeat on ultrasonography. Ongoing pregnancy was defined when a positive heartbeat was at 12 weeks or more of gestation on ultrasonography. All statistical analyses were conducted using SPSS (version 25.0; Chicago, IL, USA) software. Categorical variables were performed using the chi‐square and Fisher’s exact tests. Continuous variables were assessed using Student’s t test. A probability (p) value <0.05 was considered to determine statistical significance. A p value below 0.05 was considered statistically significant.

Results

Study population

There were 141 women undergoing IVF and embryo transfers (ETs), which included 48 women who had good-quality (GQ) ETs, 79 with only poor-quality (PQ) ETs, and 14 with either GQ or PQ that were excluded from this study (Fig 1). Most patients (n = 83) underwent double ETs, 59 samples came from a single ET, and 3 from triple ETs. ETs for 91 samples occurred on day 3 or 4, and for 50 on day 5, no significant difference between the GQ and PQ groups according to the day of ET.

Table 1 presented the characteristics of patients between the GQ and the PQ groups. There were no significant differences in age, body mass index, infertility duration, and anti-Mullerian hormone level. However, there were more ETs in the PQ group than in the GQ group. The clinical pregnancy rate was higher in the GQ (52.0%) than in the PQ embryo transfer group (40.5%); however, the difference was statistically insignificant (p  =  0.09), and the ongoing pregnancy rate of the GQ group was significantly higher than that of the PQ group (45.8% vs. 24%, respectively, p  =  0.02).

thumbnail
Table 1. Patient characteristics for the analysis of granulosa cell gene expression associated with embryo quality.

https://doi.org/10.1371/journal.pone.0280495.t001

Identification of differentially expressed genes (DEGs): samples clustered according to embryo quality

Three GCs samples from each group were selected and used for RNA-seq analysis. Six hundred and forty genes were differentially expressed in our cohort. We analyzed DEGs between the two groups. Compared with that in the GQ embryo group, 18 genes were upregulated, whereas 208 genes were downregulated in the PQ embryo group (p < 0.05; FC of log2-transformed fragments per kilobase of transcript per million (FPKM) > 1) (Fig 2A). As shown in Fig 2B, we developed unsupervised hierarchical clustering of DEGs. A detailed list of 226 DEGs that were upregulated and downregulated in each group is presented in S2 Table.

thumbnail
Fig 2. Hierarchical clustering and Differential Expression (DE) analysis of granulosa cells from good-quality (GQ) embryo transfer group compared with granulosa cells from poor-quality (PQ) embryo transfer group.

(A) A Venn diagram depicting the distribution of significant differentially expressed genes (DEGs) (fold change > 2.5, q <0.05) in our study. Numbers represent the number of different genes showing upregulated or downregulated expression in PQ embryo samples compared with GQ embryo samples with red representing upregulated expression and blue indicating downregulated expression. Numbers in parenthesis represent the number of DEGs in only PQ or GQ group. (B) Heatmap of the DEGs between the GQ and PQ embryo groups. The color key from blue to red represents the relative gene expression level from low to high, respectively. (C) Volcano plot showing DEG analysis between GQ embryo and PQ embryo groups using DESeq2; 640 genes were differentially expressed (210 downregulated (FC < -2 and FDR < 0.05) and 25 upregulated (FC > 2 and FDR < 0.05). The upregulated genes are represented using red dots; downregulated genes are denoted using blue dots; and the black dots indicate genes with no significant changes.

https://doi.org/10.1371/journal.pone.0280495.g002

Gene ontology (GO) enrichment analysis: Differential expression revealed significant differences in gene expression regarding to the degree of embryo quality

To further extend the molecular properties of the 226 DEGs, we performed a GO analysis of the up- or down-regulated groups (Fig 3). The DEGs were divided into three categories: biological processes, molecular functions, and cellular components. In the GO category biological process, DEGs were enriched in the response to stimulus, metabolic process, biological regulation, immune system process, binding and metabolic process, cellular process, single-organism process, cell and cell part, developmental process, cellular component organization or biogenesis, and the reproductive process. The top 20 genes displayed in the different embryo qualities are presented in Tables 2 and 3. KEGG pathway enrichment analysis showed that the DEGs in our study participated in 13 pathways (Fig 4). Several of these are related to reproduction, such as the chemokine signaling pathway, phagosome, cytokine-cytokine receptor interaction, cell adhesion molecules, and NF-kB signaling pathway.

thumbnail
Fig 3. Histogram of the Gene Ontology(GO) analysis of the differentially expressed genes (DEGs).

Genes were classified into three GO domains: biological process, cellular component, and molecular function. The left y-axis shows the gene count in each category. The solid columns indicate DEGs.

https://doi.org/10.1371/journal.pone.0280495.g003

thumbnail
Fig 4. Validation of RNAseq results using RT-qPCR of 13 targets normalized to GAPDH in duplicate.

Fold change was calculated using the 2ΔΔCt method between the good-quality (GQ) and poor-quality (PQ) embryos. *p <0.05. (A) Six genes associated with embryo quality both in our and previous studies. (B) Seven genes randomly selected from the NF-kB pathway.

https://doi.org/10.1371/journal.pone.0280495.g004

thumbnail
Table 2. Top 20 up-regulated genes in the poor-quality embryo transfer group compared with the good-quality embryo transfer group.

https://doi.org/10.1371/journal.pone.0280495.t002

thumbnail
Table 3. Top 20 downregulated genes in the poor-quality embryo transfer group compared with the good-quality embryo transfer group.

https://doi.org/10.1371/journal.pone.0280495.t003

Our findings enhanced the available literature exploring processes associated with embryo quality

When comparing our DEGs to those previously reported in the literature, Table 4 summarizes the findings of the detailed list of 68 genes that show genes positively associated with embryo quality. The GC gene biomarker studies have varied depending on the endpoints chosen, including ovarian development, follicular development, folliculogenesis, oocyte and embryo quality.

Validation of DEGs using RT-qPCR analysis

To examine the reliability of the RNA-seq data, we selected 13 genes to verify their expression in GC using RT-qPCR (Fig 4). When comparing our DEGs to those reported in previous studies, we found six genes that were related to embryo quality both in our study and previous published literature, including colony stimulating factor receptor 1 (CSFR1) [38], cathepsin H (CTSH) [37], cadherin 13 (CDH13) [35], serpin family A member 1 (SERPINA1) [39], cytochrome P450 family 27 subfamily A member 1 (CYP27A1) [40], and integrin subunit beta 2 (ITGB2) [16]. Among the significantly enriched pathways, we randomly selected seven genes from the NF-kB pathway: four genes that have refuting findings between different studies (interleukin 1 beta (IL1β), tumor necrosis factor (TNF), BCL2 related protein A1 (BCL2A1), C-C motif chemokine ligand 4 (CCL4)), and three genes that did not support an association with pregnancy or embryo quality (MYD88 innate immune signal transduction adaptor(MYD88), mitogen-activated protein kinase kinase kinase 7(MAP3K7), and TGF-beta activated kinase 1 binding protein 1(TAB1)).

Thirteen candidate genes, comprising eleven downregulated genes in GCs from only PQ embryo groups, CSF1R, CTSH, IL1β, TNF, BCL2A1, CCL4, SERPINA1, CYP27A1, ITGB2, MYD88, MAP3K7, and two upregulated genes in GC from PQ embryo groups, TAB1, and CDH13, were selected and analyzed using RT-qPCR. The RT-qPCR results were consistent with the RNA-seq data, which means that the RNA-seq results were dependable and that it could be used to perform accurate differential expression analysis of mRNA.

Discussion

In the present study, we analyzed GC expression of CSF1R, CTSH, IL1β, TNF, BCL2A1, CCL4, SERPINA1, CYP27A1, ITGB2 from GCs collected during IVF from oocytes that developed into GQ embryos. Selected genes were shown to be well differentiated between immature MI and mature MII oocytes. In recent years, many studies have been performed to analyze GC expression in association with various endpoints: oocyte quality, embryo development and pregnancy (Table 4). Since GC is an easily accessible material that is normally discarded during the IVF cycle, it represents a good biological material for research and diagnostic purposes.

Several signaling pathways are important in mediating or modifying the proliferative response of GCs to FSH stimulation, the primary mediator of GC proliferation during folliculogenesis [41]. Our RNA-Seq results showed that some differential pathways might verify oocyte development, such as the chemokine signaling pathway, cytokine-cytokine receptor interaction, phagosome, cell adhesion molecules, and NF-kB signaling pathway. Thus, we could infer that the PQ embryos were mainly related to the expression levels of cytokine reaction, apoptosis, and the adherent junction pathway.

In our study, the differential expression of transcripts in the NF-kB signaling pathway was related to embryo quality (Fig 4B). This pathway participate in apoptosis and cell growth, as well as in immune, inflammatory and acute phase responses [42]. The downregulation of immune and inflammation-related genes in the PQ embryo group was in agreement with the hypothesis that ovulatory process is an acute inflammatory reaction [43]. Consistent with previous studies [44, 45], various ovulation-associated factors such as PDE2A (3′5’-cyclic nucleotide phosphodiesterase 2A), RGS1 and RGS16 (a regulator of G-protein signaling 1 and 16), ADAMTS1 (a disintegrin-like and metalloprotease with thrombospondin type 1 motif, 1), and PTGS1 (prostaglandin-endoperoxide synthetase 1), were all upregulated in GCs from the PQ embryo group.

CSF-1R is activated in a similar way by homodimeric growth factors colony-stimulating factor-1 (CSF-1) and interleukin (IL)-34 [46]. CSF-1 may be essential in regulating the response of GCs to gonadotropin and may promote in the early embryonic development [38]. The biological activity of CSF-1 depends on its binding to CSF-1R in target cells [47].

CTSH expression is increased in human GCs during follicular maturation in vivo [37]. Our study demonstrated that the expression of CTSH was higher in GCs from GQ embryos than that from PQ embryos with statistical significance, suggesting embryo development failure was caused by poor follicular maturation and GC dysfunction.

SERPINA1 is the gene for an acute-phase protein that increases in response to inflammation [48]. This gene, implicated in the inflammatory response, reduces the levels of proinflammatory mediators, and increases anti-inflammatory cytokines [49]. In our study, increased SERPINA1 expression levels were observed in GQ embryo. Overexpression of SERPINA1 in GCs from the GQ embryo group indicates decreased proinflammatory cytokine levels, which leads to increased developmental competence. This finding suggests that an impaired follicular fluid microenvironment characterized by elevated pro-inflammatory cytokines may cause poor quality of oocyte.

Changes in the cellular structure of the corpus luteum (CL) on natural luteolysis may contribute to elevated concentrations of CYP27A1 mRNA. For example, macrophages invade the CL during luteolysis and express CYP27A1 [50]. In addition, increased CYP27A1 protein concentrations in mitochondria cause elevated conversion of cholesterol into 27OH, which decreases progesterone secretion in human luteinized GC [40].

ITGB2 is an integrin subunit that is a pivotal mediator of uterine receptivity and embryonic development [51]. Overexpression of this gene is associated with GCs of healthy follicles and decreased with atresia [52].

Reorganization of cellular composition for ovulation includes follicular wall degradation, and oocyte expulsion during the advanced stages of follicular development [53]. IL1β, TNF-α, and CCL4 are pro-inflammatory cytokines that act locally on ovarian follicular cells and are involved in the ovulation process [54]. During the periovulatory phase, ovarian macrophages collect in the ovary and secret these pro-inflammatory cytokines [5557]. They regulate the secretion of steroid hormones for follicle growth and are important for ovulation process and the development and regression of the CL [58, 59]. For instance, IL-1β has been shown to induce ovulation by promoting follicular rupture [60]. In the present study, IL1β, TNF-α, and CCL4 were significantly upregulated in GCs from oocytes that yielded GQ embryos. This finding suggests that these cytokines may modulate oocyte quality and embryo development.

TAB1 may represents the initiation of inflammatory functions and may serve as a connector of the IL-1 induced signaling pathway leading to activation of c-Jun N-terminal kinase (JNK) and NF-κB signaling pathways [61, 62]. B-cell lymphoma-2 (BCL2) family proteins are pivotal regulators of apoptosis [63]. The ovaries of Bcl2 knockout mice were revealed to have fewer primordial follicles [64]. However, this loss may be due to GC apoptosis, which indirectly affects follicle development. BCL2 family members, such as BCL2A1, are expressed during embryonic genome activation and maintained until the blastocyst stage [33], suggesting that these are required throughout early embryonic development.

The strength of our study is that it was prospective and sampled a large group of patients who were followed up clinically. We also performed sorting and enrichment analysis of RNA-sequencing data, the most unbiased approach that can be used to explore transcriptomic signatures [65]. Furthermore, by incorporating information from DEGs, pathway analysis, and related studies, the present study provides a list of candidate genes with functions and expression levels closely related to embryo quality in IVF. In this study, RNA-sequencing data of whole GCs collection revealed a difference between the group that produced good-quality embryos and the group that produced only poor-quality embryos. This difference could be used for therapeutic purposes to improve embryo quality in the future.

The present study, however, has some limitations. First, GCs obtained from the follicular fluid were GCs in all follicles and cannot accurately reflect the quality of single oocyte. In clinical practice, it may be more difficult to collect GCs from a single follicle than cumulus cells. Second, we reported expression levels of the genes, but did not analyze expression ot corresponding proteins and the role of post-translational modifications.

Conclusions

We found that the expression profiles of specific genes in GCs was associated with embryo quality during IVF and suggested the need to develop treatment strategies that can compensate for the poor quality due to the deficiency of specific genes during follicular development. Furthermore, we reported the published data that support or confute granulosa expressed genes as biomarkers for identifying oocyte or embryo quality. This study improves our understanding of reproductive function of GCs, which could be helpful for more targeted studies aiming to improve oocyte and embryo competence in the future.

Supporting information

S1 Table. Primer sequences used for real-time qPCR.

https://doi.org/10.1371/journal.pone.0280495.s001

(DOCX)

S2 Table. A detailed list of 226 DEGs that were upregulated and downregulated in good-quality and poor-quality embryo group.

https://doi.org/10.1371/journal.pone.0280495.s002

(XLSX)

Acknowledgments

We sincerely thank the patients and their cohabitants for their willingness to participate in the study.

References

  1. 1. Ahlstrom A, Westin C, Reismer E, Wikland M, Hardarson T. Trophectoderm morphology: an important parameter for predicting live birth after single blastocyst transfer. Hum Reprod 2011; 26(12); 3289–96, pmid:21972253
  2. 2. della Ragione T, Verheyen G, Papanikolaou EG, Van Landuyt L, Devroey P, Van Steirteghem A. Developmental stage on day-5 and fragmentation rate on day-3 can influence the implantation potential of top-quality blastocysts in IVF cycles with single embryo transfer. Reprod Biol Endocrinol 2007; 5(2, pmid:17257401
  3. 3. Sirard MA, Richard F, Blondin P, Robert C. Contribution of the oocyte to embryo quality. Theriogenology 2006; 65(1); 126–36, pmid:16256189
  4. 4. Ziebe S, Petersen K, Lindenberg S, Andersen AG, Gabrielsen A, Andersen AN. Embryo morphology or cleavage stage: how to select the best embryos for transfer after in-vitro fertilization. Hum Reprod 1997; 12(7); 1545–9, pmid:9262293
  5. 5. Sermondade N, Delarouziere V, Ravel C, Berthaut I, Verstraete L, Mathieu E, et al. Characterization of a recurrent poor-quality embryo morphology phenotype and zygote transfer as a rescue strategy. Reprod Biomed Online 2012; 24(4); 403–9, pmid:22377150
  6. 6. Alfarawati S, Fragouli E, Colls P, Stevens J, Gutierrez-Mateo C, Schoolcraft WB, et al. The relationship between blastocyst morphology, chromosomal abnormality, and embryo gender. Fertil Steril 2011; 95(2); 520–4, pmid:20537630
  7. 7. Sigalos G, Triantafyllidou O, Vlahos NF. Novel embryo selection techniques to increase embryo implantation in IVF attempts. Arch Gynecol Obstet 2016; 294(6); 1117–24, pmid:27628754
  8. 8. Cimadomo D, Capalbo A, Ubaldi FM, Scarica C, Palagiano A, Canipari R, et al. The Impact of Biopsy on Human Embryo Developmental Potential during Preimplantation Genetic Diagnosis. Biomed Res Int 2016; 2016(7193075, pmid:26942198
  9. 9. Gilchrist RB, Lane M, Thompson JG. Oocyte-secreted factors: regulators of cumulus cell function and oocyte quality. Hum Reprod Update 2008; 14(2); 159–77, pmid:18175787
  10. 10. McKenzie LJ, Pangas SA, Carson SA, Kovanci E, Cisneros P, Buster JE, et al. Human cumulus granulosa cell gene expression: a predictor of fertilization and embryo selection in women undergoing IVF. Hum Reprod 2004; 19(12); 2869–74, pmid:15471935
  11. 11. van Montfoort AP, Geraedts JP, Dumoulin JC, Stassen AP, Evers JL, Ayoubi TA. Differential gene expression in cumulus cells as a prognostic indicator of embryo viability: a microarray analysis. Mol Hum Reprod 2008; 14(3); 157–68, pmid:18204071
  12. 12. Hamel M, Dufort I, Robert C, Gravel C, Leveille MC, Leader A, et al. Identification of differentially expressed markers in human follicular cells associated with competent oocytes. Hum Reprod 2008; 23(5); 1118–27, pmid:18310048
  13. 13. Hamel M, Dufort I, Robert C, Leveille MC, Leader A, Sirard MA. Genomic assessment of follicular marker genes as pregnancy predictors for human IVF. Mol Hum Reprod 2010; 16(2); 87–96, pmid:19778949
  14. 14. Hamel M, Dufort I, Robert C, Leveille MC, Leader A, Sirard MA. Identification of follicular marker genes as pregnancy predictors for human IVF: new evidence for the involvement of luteinization process. Mol Hum Reprod 2010; 16(8); 548–56, pmid:20610614
  15. 15. Burnik Papler T, Vrtacnik Bokal E, Lovrecic L, Kopitar AN, Maver A. No specific gene expression signature in human granulosa and cumulus cells for prediction of oocyte fertilisation and embryo implantation. PLoS One 2015; 10(3); e0115865, pmid:25769026
  16. 16. Devjak R, Fon Tacer K, Juvan P, Virant Klun I, Rozman D, Vrtacnik Bokal E. Cumulus cells gene expression profiling in terms of oocyte maturity in controlled ovarian hyperstimulation using GnRH agonist or GnRH antagonist. PLoS One 2012; 7(10); e47106, pmid:23082142
  17. 17. Kim MK, Park JK, Jeon Y, Choe SA, Lee HJ, Kim J, et al. Correlation between Morphologic Grading and Euploidy Rates of Blastocysts, and Clinical Outcomes in In Vitro Fertilization Preimplantation Genetic Screening. J Korean Med Sci 2019; 34(4); e27, pmid:30686949
  18. 18. Cummins JM, Breen TM, Harrison KL, Shaw JM, Wilson LM, Hennessey JF. A formula for scoring human embryo growth rates in in vitro fertilization: its value in predicting pregnancy and in comparison with visual estimates of embryo quality. J In Vitro Fert Embryo Transf 1986; 3(5); 284–95, pmid:3783014
  19. 19. Reinblatt SL, Ishai L, Shehata F, Son WY, Tulandi T, Almog B. Effects of ovarian endometrioma on embryo quality. Fertil Steril 2011; 95(8); 2700–2, pmid:21444070
  20. 20. Gardner DK, Surrey E, Minjarez D, Leitz A, Stevens J, Schoolcraft WB. Single blastocyst transfer: a prospective randomized trial. Fertil Steril 2004; 81(3); 551–5, pmid:15037401
  21. 21. Sun J, Nishiyama T, Shimizu K, Kadota K. TCC: an R package for comparing tag count data with robust normalization strategies. BMC Bioinformatics 2013; 14(219, pmid:23837715
  22. 22. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014; 15(12); 550, pmid:25516281
  23. 23. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2010; 26(1); 139–40, pmid:19910308
  24. 24. Young MD, Wakefield MJ, Smyth GK, Oshlack A. Gene ontology analysis for RNA-seq: accounting for selection bias. Genome Biol 2010; 11(2); R14, pmid:20132535
  25. 25. Sasson R, Rimon E, Dantes A, Cohen T, Shinder V, Land-Bracha A, et al. Gonadotrophin-induced gene regulation in human granulosa cells obtained from IVF patients. Modulation of steroidogenic genes, cytoskeletal genes and genes coding for apoptotic signalling and protein kinases. Mol Hum Reprod 2004; 10(5); 299–311, pmid:15026540
  26. 26. Liu C, Su K, Chen L, Zhao Z, Wang X, Yuan C, et al. Prediction of oocyte quality using mRNA transcripts screened by RNA sequencing of human granulosa cells. Reprod Biomed Online 2021; 43(3); 413–20, pmid:34400084
  27. 27. Conti M. Specificity of the cyclic adenosine 3’,5’-monophosphate signal in granulosa cell function. Biol Reprod 2002; 67(6); 1653–61, pmid:12444038
  28. 28. Seger R, Hanoch T, Rosenberg R, Dantes A, Merz WE, Strauss JF, 3rd, et al. The ERK signaling cascade inhibits gonadotropin-stimulated steroidogenesis. J Biol Chem 2001; 276(17); 13957–64, pmid:11278318
  29. 29. Koks S, Velthut A, Sarapik A, Altmae S, Reinmaa E, Schalkwyk LC, et al. The differential transcriptome and ontology profiles of floating and cumulus granulosa cells in stimulated human antral follicles. Mol Hum Reprod 2010; 16(4); 229–40, pmid:19933312
  30. 30. Duffy DM, Seachord CL, Dozier BL. An ovulatory gonadotropin stimulus increases cytosolic phospholipase A2 expression and activity in granulosa cells of primate periovulatory follicles. J Clin Endocrinol Metab 2005; 90(10); 5858–65, pmid:15972573
  31. 31. Kawano Y, Fukuda J, Nasu K, Nishida M, Narahara H, Miyakawa I. Production of macrophage inflammatory protein-3alpha in human follicular fluid and cultured granulosa cells. Fertil Steril 2004; 82 Suppl 3(1206–11, pmid:15474097
  32. 32. Lu CL, Yan ZQ, Song XL, Xu YY, Zheng XY, Li R, et al. Effect of exogenous gonadotropin on the transcriptome of human granulosa cells and follicular fluid hormone profiles. Reprod Biol Endocrinol 2019; 17(1); 49, pmid:31234873
  33. 33. Metcalfe AD, Hunter HR, Bloor DJ, Lieberman BA, Picton HM, Leese HJ, et al. Expression of 11 members of the BCL-2 family of apoptosis regulatory molecules during human preimplantation embryo development and fragmentation. Mol Reprod Dev 2004; 68(1); 35–50, pmid:15039946
  34. 34. Yamamoto Y, Kuwahara A, Taniguchi Y, Yamasaki M, Tanaka Y, Mukai Y, et al. Tumor necrosis factor alpha inhibits ovulation and induces granulosa cell death in rat ovaries. Reprod Med Biol 2015; 14(3); 107–15, pmid:26161038
  35. 35. Makrigiannakis A, Coukos G, Christofidou-Solomidou M, Gour BJ, Radice GL, Blaschuk O, et al. N-cadherin-mediated human granulosa cell adhesion prevents apoptosis: a role in follicular atresia and luteolysis? Am J Pathol 1999; 154(5); 1391–406, pmid:10329592
  36. 36. Yerushalmi GM, Salmon-Divon M, Yung Y, Maman E, Kedem A, Ophir L, et al. Characterization of the human cumulus cell transcriptome during final follicular maturation and ovulation. Mol Hum Reprod 2014; 20(8); 719–35, pmid:24770949
  37. 37. Oksjoki S, Soderstrom M, Vuorio E, Anttila L. Differential expression patterns of cathepsins B, H, K, L and S in the mouse ovary. Mol Hum Reprod 2001; 7(1); 27–34, pmid:11134357
  38. 38. Wei Z, Song X, Zhifen Z. Molecular mechanism and functional role of macrophage colonystimulating factor in follicular granulosa cells. Mol Med Rep 2017; 16(3); 2875–80, pmid:28656272
  39. 39. Burnik Papler T, Vrtacnik Bokal E, Maver A, Kopitar AN, Lovrecic L. Transcriptomic Analysis and Meta-Analysis of Human Granulosa and Cumulus Cells. PLoS One 2015; 10(8); e0136473, pmid:26313571
  40. 40. Xu Y, Hutchison SM, Hernandez-Ledezma JJ, Bogan RL. Increased 27-hydroxycholesterol production during luteolysis may mediate the progressive decline in progesterone secretion. Mol Hum Reprod 2018; 24(1); 2–13, pmid:29177442
  41. 41. Richards JS. Hormonal control of gene expression in the ovary. Endocr Rev 1994; 15(6); 725–51, pmid:7705279
  42. 42. Kucharczak J, Simmons MJ, Fan Y, Gelinas C. To be, or not to be: NF-kappaB is the answer—role of Rel/NF-kappaB in the regulation of apoptosis. Oncogene 2003; 22(56); 8961–82, pmid:14663476
  43. 43. Duffy DM, Ko C, Jo M, Brannstrom M, Curry TE. Ovulation: Parallels With Inflammatory Processes. Endocr Rev 2019; 40(2); 369–416, pmid:30496379
  44. 44. Wyse BA, Fuchs Weizman N, Kadish S, Balakier H, Sangaralingam M, Librach CL. Transcriptomics of cumulus cells—a window into oocyte maturation in humans. J Ovarian Res 2020; 13(1); 93, pmid:32787963
  45. 45. Wissing ML, Kristensen SG, Andersen CY, Mikkelsen AL, Host T, Borup R, et al. Identification of new ovulation-related genes in humans by comparing the transcriptome of granulosa cells before and after ovulation triggering in the same controlled ovarian stimulation cycle. Hum Reprod 2014; 29(5); 997–1010, pmid:24510971
  46. 46. Stanley ER, Chitu V. CSF-1 receptor signaling in myeloid cells. Cold Spring Harb Perspect Biol 2014; 6(6), pmid:24890514
  47. 47. Chitu V, Stanley ER. Regulation of Embryonic and Postnatal Development by the CSF-1 Receptor. Curr Top Dev Biol 2017; 123(229–75, pmid:28236968
  48. 48. Badola S, Spurling H, Robison K, Fedyk ER, Silverman GA, Strayle J, et al. Correlation of serpin-protease expression by comparative analysis of real-time PCR profiling data. Genomics 2006; 88(2); 173–84, pmid:16713170
  49. 49. Boots CE, Jungheim ES. Inflammation and Human Ovarian Follicular Dynamics. Semin Reprod Med 2015; 33(4); 270–5, pmid:26132931
  50. 50. Fu X, Menke JG, Chen Y, Zhou G, MacNaul KL, Wright SD, et al. 27-hydroxycholesterol is an endogenous ligand for liver X receptor in cholesterol-loaded cells. J Biol Chem 2001; 276(42); 38378–87, pmid:11504730
  51. 51. Monniaux D, Huet-Calderwood C, Le Bellego F, Fabre S, Monget P, Calderwood DA. Integrins in the ovary. Semin Reprod Med 2006; 24(4); 251–61, pmid:16944422
  52. 52. Le Bellego F, Pisselet C, Huet C, Monget P, Monniaux D. Laminin-alpha6beta1 integrin interaction enhances survival and proliferation and modulates steroidogenesis of ovine granulosa cells. J Endocrinol 2002; 172(1); 45–59, pmid:11786373
  53. 53. Abedel-Majed MA, Romereim SM, Davis JS, Cupp AS. Perturbations in Lineage Specification of Granulosa and Theca Cells May Alter Corpus Luteum Formation and Function. Front Endocrinol (Lausanne) 2019; 10(832, pmid:31849844
  54. 54. Sarapik A, Velthut A, Haller-Kikkatalo K, Faure GC, Bene MC, de Carvalho Bittencourt M, et al. Follicular proinflammatory cytokines and chemokines as markers of IVF success. Clin Dev Immunol 2012; 2012(606459, pmid:22007253
  55. 55. Cohen PE, Nishimura K, Zhu L, Pollard JW. Macrophages: important accessory cells for reproductive function. J Leukoc Biol 1999; 66(5); 765–72, pmid:10577508
  56. 56. Wu R, Van der Hoek KH, Ryan NK, Norman RJ, Robker RL. Macrophage contributions to ovarian function. Hum Reprod Update 2004; 10(2); 119–33, pmid:15073142
  57. 57. Turner EC, Hughes J, Wilson H, Clay M, Mylonas KJ, Kipari T, et al. Conditional ablation of macrophages disrupts ovarian vasculature. Reproduction 2011; 141(6); 821–31, pmid:21393340
  58. 58. Jabbour HN, Sales KJ, Catalano RD, Norman JE. Inflammatory pathways in female reproductive health and disease. Reproduction 2009; 138(6); 903–19, pmid:19793840
  59. 59. Greenfeld CR, Roby KF, Pepling ME, Babus JK, Terranova PF, Flaws JA. Tumor necrosis factor (TNF) receptor type 2 is an important mediator of TNF alpha function in the mouse ovary. Biol Reprod 2007; 76(2); 224–31, pmid:17065602
  60. 60. Vassiliadis S, Relakis K, Papageorgiou A, Athanassakis I. Endometriosis and infertility: a multi-cytokine imbalance versus ovulation, fertilization and early embryo development. Clin Dev Immunol 2005; 12(2); 125–9, pmid:16050143
  61. 61. Takaesu G, Kishida S, Hiyama A, Yamaguchi K, Shibuya H, Irie K, et al. TAB2, a novel adaptor protein, mediates activation of TAK1 MAPKKK by linking TAK1 to TRAF6 in the IL-1 signal transduction pathway. Mol Cell 2000; 5(4); 649–58, pmid:10882101
  62. 62. Owerbach D, Pina L, Gabbay KH. A 212-kb region on chromosome 6q25 containing the TAB2 gene is associated with susceptibility to type 1 diabetes. Diabetes 2004; 53(7); 1890–3, pmid:15220215
  63. 63. Aouacheria A, Brunet F, Gouy M. Phylogenomics of life-or-death switches in multicellular animals: Bcl-2, BH3-Only, and BNip families of apoptotic regulators. Mol Biol Evol 2005; 22(12); 2395–416, pmid:16093567
  64. 64. Ratts VS, Flaws JA, Kolp R, Sorenson CM, Tilly JL. Ablation of bcl-2 gene expression decreases the numbers of oocytes and primordial follicles established in the post-natal female mouse gonad. Endocrinology 1995; 136(8); 3665–8, pmid:7628407
  65. 65. Wang Z, Gerstein M, Snyder M. RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet 2009; 10(1); 57–63, pmid:19015660