Upregulation of ICAM-1 and HLA-G Genes Expression in Cumulus Cells of Infertile Women with Poor Response to Ovarian Stimulation with a Healthy Lifestyle

DOI: https://doi.org/10.21203/rs.3.rs-100360/v1

Abstract

Purpose: Diagnosis of women with poor ovarian response (POR) to exogenous gonadotropin stimulation is a challenge for reproductive specialists. This study aimed to investigate the expression profile for ICAM-1 and HLA-G genes in the cumulus cells of infertile women with POR based on their healthy lifestyle.

Methods: Eighty women under the age of 35 were enrolled and divided into two groups: 1- POR without a healthy lifestyle (n=40) and POR with a healthy lifestyle (n=40). The ICAM-1 and HLA-G genes expression was compared by the quantitative PCR between the groups. The ICAM-1 and HLA-G protein levels were analyzed by the western blotting technique, and the methylation pattern was investigated by the methylation-specific PCR.

Results: The qRT-PCR assay showed that ICAM-1 and HLA-G genes were differentially expressed between the two groups studied. ICAM-1,HLA-G genes,and proteins expressions in POR with healthy lifestyle were upregulated compared to the second group (P<0.05). ICAM-1 and HLA-G DNA methylation status in the POR with a healthy lifestyle were decreased compared to the other group (P<0.05). The oocyte quality and clinical pregnancy ratio were significantly higher in the POR with a healthy lifestyle women than in the other ones (P<0.05).

Conclusion: Theupregulation of ICAM-1 and HLA-G in POR women is probably related to lifestyle. Lifestyle may affect the methylation pattern, protein levels and alteration of gene expression profiles in the cumulus cells of POR women. Additionally, lifestyle may be considered a marker for ovulation, oocyte maturation, preimplantation and clinical pregnancy process. 

Introduction

The term poor response to ovarian stimulation (POR) typically refers to woman with a reduced ovarian reserve or poor ovarian response to exogenous gonadotropin stimulation [1]. In fact, the POR or the cycle without ovulation is a cycle in which the ovary is unable to release the oocyte. As a result, ovulation does not occur. Despite the efforts to optimize the definition of POR, unfortunately there is still limited knowledgeabout the POR pathophysiology, and the etiology hinders the practical solutions to manage the condition[2]. Indeed, the diagnosis of anovulation is not easy. Contrary to popular belief, women with anovulation have more or less a normal menstrual cycle and usually for the first time, they find their own problem when they want to become pregnant [3]. Recently, POSEIDON criteria have been used to diagnose POR, including age, Anti Müllerian Hormone (AMH) dose, Antra Follicle Count (AFC), and oocyte number [4]. The consequential complications of POR are intrinsic ovarian resistance to gonadotropin stimulation, reduced ovarian reserve, ovulation and oocyte maturation disorder [5]. Reduced ovarian reserve is defined as the impaired fertility ability leading to infertility. In fact, excessive use of plastics objects in the human life has been proposed as one of the causes of creating POR in women [6]. Indeed, excessive use of plastics to store hot food and drinks can cause various diseases. Polycarbonate is a type of plastic containing BisphenolA (BPA) [7]. Over time, when plastic is heated or exposed to acidic or alkaline substances, it breaks down and BPA attaches to food and drink, so that it can enter the body [8]. BPA, by mimicking sex hormones, can disrupt the endocrine system, hormones and hormonal signaling pathways [9]. BPA even in small amounts may interfere with the ovulation, ovarian response, oocyte maturation and pregnancy [10]. In fact, lifestyle plays a key role in regulating ovarian response, oocyte maturation and genes expression. Gene’s expression alteration in POR patients is a condition damaging oocyte developmental, resulting in reduced rates of fertilization, embryonic development, and implantation [11]. The role of gene’s expression alteration in the pathophysiology and etiology of POR is uncertain, but studies support the hypothesis that nutrition and lifestyle play a crucial role in regulating the response of intrinsic ovarian resistance to gonadotropin stimulation, reduced ovarian reserve, oocyte maturation, implantation and gene expression [12].

ICAM-1, known as CD54, plays a pivotal role during oocyte maturation, and it is a protein encoded by the ICAM1 gene. The responsibility of ICAM-1 is encoding a cell surface glycoprotein usually expressed on endothelial cells and cells of the immune system. As mentioned, ICAM-1 plays a central role in the ovulation process [13]. Indeed, the main components of this ovulation process are the disruption of the extracellular matrix (ECM) at the follicular peak and the change in follicular vessels [14]. In these events, ICAM-1 participates by secreting proteases and active inhibitors. Indeed, biochemical markers of the oocyte maturation and ovulation are highly important. Upregulation of ICAM-1 in immature oocytes as well as downregulation of ICAM-1 in mature oocytes has been observed.

Human leukocyte antigen-G (HLA-G) is regarded to play a vital role in oocyte maturation and implantation of embryos. HLA-G can be effective in controlling trophoblast invasion and maintaining a local immunosuppressive state. The expression and distribution of HLA-G is on human spermatogenic cells, primary and secondary oocytes, and preimplantation embryos. Furthermore, the HLA-G production is related to the good quality of cumulus cells (CCs) in oocytes [15].

In the present study, we aimed to assess the genes expression, proteins level and methylation status of ICAM-1 and HLA-G in the cumulus cells of infertile POR patients based on their lifestyle following ovarian stimulation with a gonadotropin releasing hormone (GnRH) antagonist protocol.

Materials And Methods

Patient selection

In the present study, eighty women under the age of 35 and BMI 18-25 Kg /m2, who participated in an intracytoplasmic sperm injection (ICSI) program, were selected. The patients who were diagnosed by the Poseidon group1 subgroup 1b that included; [patients <35 years old with adequate ovarian reserve parameters (AFC>5; AMH>1.2 ng/ml) and with an unexpected poor or suboptimal ovarian response (subgroup 1b: 4-9 oocytes retrieved] (4). The patients were divided into two groups: without a healthy lifestyle (n=40, infertile women with poor response (POR) to ovarian stimulation history), and with a healthy lifestyle (n=40, infertile women with poor response (POR) to ovarian stimulation history). The healthy lifestyle group included women who did not use plastic containers for hot food, hot drink and foodstuffs store. However, the group without a healthy lifestyle included women who used excessive plastic containers to store hot food, hot drink and foodstuffs. Information on a healthy lifestyle was obtained using a validated food frequency questionnaire. The main exclusion criteria were women affected by endometriosis, and women with a history of uterus and ovaries operation. Furthermore, in this study, at the request of the patients, the treatment IVF/ICSIcycle was canceled, while they had a low number of follicles or lacked growth of follicles. The Ethics Committee at Royan Institute, Iran approved this prospective case-control study (No. IR.ACECR.ROYAN.REC.1394.150). All participants gave informed consent prior to inclusion in the study. We ensured the confidentiality of the patients’ identities by data anonymization during analysis. This research did not affect the treatment of patients.

Antagonist stimulation protocol

In this study, Controlled of ovarian stimulation (COS) was controlled from the third day of the cycle. The patients received regular, daily subcutaneous (SC) injections of recombinant follicle stimulating hormone (rFSH, Gonal-F, Serono, Switzerland). The first dose of rFSH for each patient was started according to sonographic monitoring, and AFC, estradiol (E2) level, and AMH for each patient were evaluated. In the stage of growing follicles >12 mm, the patients received SC injections of a GnRH antagonist, cetrorelix (Cetrotide®, Merck Serono, Germany). The protocol consisted of daily Cetrotide® SC injections until the criteria for human chorionic gonadotropin (hCG) administration were met. When more than 3 follicles reached diameters of at least 18 mm and E2 levels of 1000-4000 pg/mL When at least three follicles reached diameters of ≥18 mm as well as E2 levels of 1000-4000 pg/mL, each patient received an intramuscular (IM) injection of 10000 IU of hCG (Pregnyl®, Organon, Netherlands) or SC injection of 250 μgOvidrel (Merck Serono, Germany).

Isolation of cumulus cells

Following follicular puncture, the cumulus-oocyte complexes (COCs) were collected and washed at least 3-5 times in G-IVFTM medium (Vitrolife, Sweden) to remove blood and excess cells. Immediately after washing, the COCs were moved to a CO2 incubator at 37˚C for 2 hours in G-IVFTM medium. Cumulus cells were denuded from COCs by 80 IU of hyaluronidase, (Sigma, USA) [16]. After oocyte denudation, a pellet of cumulus cells was washed with phosphate-buffered saline (PBS), and RNA stabilizer reagent buffer was added for further analyses. Cumulus cells were immediately transferred toliquid nitrogenfor snap freeze, and then they were stored at -80˚C. The cumulus cells were denuded from metaphase II gametes (MII). In the IVF laboratory, MII gametes were fertilized by the ICSI process within 10 minutes after denudation, and they were incubated until the embryo transfer was processed. Fertilization was evaluated (16-20 hours after ICSI). According to our IVF laboratory standards process, embryos were graded by embryologist specialists at the pronuclear (16-20 hours) and cleavage stages (48-72 hours) [17]. Then, the embryologist selected 1 or 2 embryos for transfer. For successful embryo transfer, they considered the embryo grades, age of the patients, and previous ART cycles. The quality of the embryos at the cleavage stage was classified based on these criteria: [The excellent quality of embryo should be (≥4 cells or ≥8 cellsand <10 % fragmentation), the good quality of embryo should be (≥ 4 cells or ≥8 cells and 10-20% fragmentation), and the poor quality of embryo should be (<4 cells or <8 cells and >20 % fragmentation)] [18].

RNA extraction, cDNA synthesis and qPCR

Total RNA was extracted by a Trizol (TRI; Sigma-Aldrich, US) and treated with RNase-free DNaseI according to the manufacturer’s instructions. RNA concentration and purity were quantified using a Nanodrop2000 Spectrophotometer (Thermo, USA). Reverse transcription was performed at 25ºC for 10 min, 42ºC for 1h and then 70ºC for 10 min. For this reaction, cDNA was synthesized using the Revert AidTM H-Minus First Strand cDNA synthesis (Fermentas, German) by the random Hexa nucleotides primer (0.2µg/µl) in a solution (20µl) containing 4µl 5x reaction buffer, 1µl Ribonuclease inhibitor (20U/µl), 2µl dNTP mix (10mM) and RNase-DNase free water.

Real-Time PCR quantification

ICAM-1 and HLA-G were chosen as target genes, and 18srRNAwas utilized normalizae each sample. Primers were designed by the Primer Express 3.0 software for the real-time PCR. Table 1 presents the primer sequences. The real-time PCR was performed in a Step OnePlus™ instrument (Applied Biosystems, USA). The reaction contained 2µl cDNA, 10µl of Power SYBR Green master mix (TAKARA, Japan) and 1µl (of 5oo nM primer) of forward and reverse primers. The reactions were performed in duplicate. The thermal cycling condition for the amplification was 95°C for 10 min, followed by 40 cycles of 95°C for 15s and 60° for 10 min. The Ct data were determined using default threshold settings. The 2-ΔΔCT method was used to analyze the relative expression level of target genes (Livak&Schmittgen, 2001). Melt curve analysis was performed to determine the specificity of the real-time PCR assay. A t-test was used to investigate whether the differences between ICAM-1 and HLA-G genes expression were calculated by the 2-ΔΔCT method.

Table 1: Real time gene expression and MSP primers sequences

Primers for real time gene expression

sequences

Product size

 

F-ICAM-1

5′-GCAATGTGCAAGAAGATAGCCA-3′

105 bp

 

R-ICAM-1

5′-GGGCAAGACCTCAGGTCATGT-3′

 

 

F-HLA-G

5′-AGCTGTGGTGGTGCCTTC-3′

106 bp

 

R-HLA-G

5′-GGGCAGGGAAGACTGCTT-3′

 

 

Primers for MSP

sequences

Product size

CpG Island Info

F- ICAM1-M

CGCGATTTTTTTGGTTTTTC

169 bp

 

R- ICAM1-M

TATTTACTTAACCACCGCCTATACG

 

chr19:10,269,612-10,271,289

F- ICAM1-UM

GTTGTGTGATTTTTTTGGTTTTTT

171 bp

 

R- ICAM1-UM

TTTACTTAACCACCACCTATACATA

 

 

F- HLA G -M

CGTAGGTATATTGTTTATATTCGCG

185 bp

 

R- HLAG -M

TACCTAAAAAAACCCCAAAACG

 

 

F- HLAG -UM

TGTAGGTATATTGTTTATATTTGTGG

186 bp

chr6:29827777-29828817

R- HLAG -UM

CTACCTAAAAAAACCCCAAAACAC

 

 

 

Protein expression level

Western blot analysis

The total protein was extracted from equal amounts of cumulus cells in all samples using lysis buffer (7M urea, 2Mthiourea, 4% CHAPS [w/v], 75 mM DTT, 1% ampholyte [w/v], and 40 mMTris‐HCl), and then the protein concentration was evaluated by bicinchoninic acid assay (Thermo Scientific, Rockford). Additionally, 40 μg of protein for each sample was used onto a discontinuous 12% SDS‐polyacrylamide gel and exposed to vertical electrophoresis. Afterward, the proteins were transferred to polyvinylidenedifluoride membranes (Bio‐Rad, Hercules). Subsequently, with 3% bovine serum albumin (Sigma‐Aldrich) as well as 1% of nonfat dry milk (Amersham, GE Healthcare Life Sciences, Little Chalfont, UK), the membranes blocked process was performed. The time required for membranes blocked process is 2 hours at RT. In this stage, the membranes were incubated for overnight at 4°C with primary antibodies against human ICAM1 (G-5) (1:1,000, Cat. No.: SC‐8439; SantaCruz, Madeira), HLA-G (4H84) (1:1,000, Cat.No.: sc-21799; SantaCruz, Madeira) and β‐actin (1:1,000, Cat. No.: A2228; Sigma‐Aldrich). In the next stage, the membranes secondary antibodies were used. Indeed, for 1.5 hours in RT, the membranes were exposed to horseradish peroxidase‐conjugated secondary antibodies. The identification of ICAM1 and β‐actin proteins was gained by goat antirabbit IgG (1:5,000, Cat. No.: ab6112; Abcam, Cambridge, UK), and antimouse IgG (1:10,000, Cat.No.: 7076; Cell Signaling Technology, Danvers) immunoglobulins, respectively (Fig. 1).Chemiluminescence detection system (Amersham) was used for protein visualization. The ImageJ software version 1.50i (US National Institutes of Health, Bethesda) was used to quantify protein bands intensity on the PVDF paper. The changes in ICAM-1 and HLA-G level were normalized against β‐actin as a housekeeping protein and then calculated regarding infertile women with a poor response to ovarian stimulation without a healthy lifestyle.

Methylation-specific polymerase chain reaction (MSP)

DNA isolation and bisulfite conversion

Genomic DNA was extracted from COCs by the QIAamp DNA Micro Kit (QIAGEN, Netherlands) according to the manufacturer’s instructions. DNA quantity and quality were measured using a Nanodrop 2000 Spectrophotometer (Thermo, USA). Genomic DNA was treated by bisulfite using the EZ DNA Methylation-Gold (Zymo research, Germany) according to the manufacturer’s instructions. After conversion, DNA was eluted in buffer (Qiagen) to a final concentration of 30 ng/μl.

Methylation-specific PCR (MSP)

The DNA methylation status of ICAM-1 and HLA-G genes in COCs samples was evaluated by the methylation-specific PCR (MSP). For MSP, we had to use specific primer pairs for both methylated and unmethylated promoter sequences. The primers were designed using the Meth primer and GeneRunner software. Each MSP reaction was performed in a total volume of 25 μL. One microliter of sodium bisulfite converted DNA was added into a 24 μL reaction mixture containing 0.5 U of hot start Gold Taq Polymerase ( Promega, USA), 5 μ L of the 10 × PCR buffer, 2.0 μ L of MgCl2 (50 mmol/L), 0.5 μ L of dNTP (10 mmol/L; Fermentas) and 1 μ L of the corresponding forward and reverse primers (10 μ mol/L dH2O up to final volume of 25 μ L). Sodium bisulfite treated DNA was amplified in two separate MSP reactions, one with a set of primers specific for methylated, and the one for unmethylated ICAM-1 and HLA-G promoters sequences (Table 1). For MSP positive control, we used fully methylated DNA (100%) and unmethylated DNA (0%), which the fully methylated DNA was made of MsssImethylase (NEB, Ipswich, MA, USA) using human placental genomic DNA (gDNA; Sigma Aldrich), and the unmethylated DNA was made of human placental genomic DNA (gDNA; Sigma Aldrich). To check the MSP method sensitivity, we used the fully methylated and unmethylated DNA with different serial dilutions: the ratio 1% to 99%, 5% to 95%, 10% to 90%, 20% to 80%, 35% to 65%, 60% to 40%, and 100% to 0% Methylated DNA was combined with unmethylated DNA, respectively. Thermo cycling conditions were as follows: 95 °C for 5 min followed by 40 cycles of 95 °C for 30 s, 63 °C for 60 s and 72 °C for 60 s, with a final extension of 72 °C for 4 min. MSP products for methylated and unmethylated ICAM-1 and HLA-G promoters were run on 2% agarose gels containing 40 mMTris-acetate/1.0 mM EDTA (pH = 8) and were visualized by ethidium bromide staining (Fig2). Methylation genes pattern visualization was performed using the enhanced chemiluminescence detection system (Amersham). The intensity of methylation genes bands on the agarose gel was quantified using the ImageJ software version 1.50i (US National Institutes of Health, Bethesda).

Data analysis

Relative gene expressions were calculated by the 2-ΔΔCt method. The normalized ΔCt value of each sample was calculated using reference genes. In this study, categorical variables were presented as number (%) and continuous variables as mean ± SD. The independent t-test was used to assess the mean differences in demographic and clinical characteristics between the groups. Chi-square analysis was used for qualitative data. Univariate and backward multiple linear regression, including all variables, were used to evaluate the association between ICAM-1 and HLA-G genes, proteins, methylation and some demographic and clinical variables. Statistical analyses were conducted using the IBM SPSS Statistics for Windows, Version 22.0 (IBM Crop., Armonk, NY, USA). All sta­tistical tests were 2-tailed, and a P<0.05 was considered statistically significant.

Results

Table 2 presents the participants’ demographic and clinical characteristics. The mean (SD) number of oocytes was statistically significantly higher in the healthy lifestyle group (Group1) than in the group without a healthy lifestyle (Group 2), (3.83 ± 0.99 vs. 3.27 ± 0.64, p = 0.004). The number of the MII retrieved oocyte was statistically significantly larger in Group 1 than in Group 2 (2.83 ± 0.99 vs. 2.35 ± 0.66, p = 0.013).

Table 2

Clinical parameters for withouthealthy lifestyleand withhealthy lifestyleof POR patients

(n = 80)

Without healthy lifestyle

With healthy lifestyle

P-value

Age (year)

29.60 ± 3.77

29.66 ± 4.26

0.948

Marriage duration (year)

4.15 ± 0.89

4.39 ± 1.24

0.322

Infertility duration (year)

3.23 ± 0.80

3.59 ± 1.16

0.109

AMH

1.30 ± 0.22

1.39 ± 0.31

0.123

AFC

5.95 ± 1.04

6.49 ± 1.38

0.051

no .Oocyte

3.27 ± 0.64

3.83 ± 0.99

0.004*

no. MII

2.35 ± 0.66

2.83 ± 0.99

0.013*

Oocyte quality

2.50 ± 0.93

3.51 ± 1.05

0.000*

Total embryo

1.30 ± 0.56

1.93 ± 026

0.000*

no.clinical Pregnancy

1.80 ± 0.41

1.70 ± 0.46

0.040

ICAM1 Gene (Fold change)

1.14 ± 0.65

14.1 ± 4.52

0.000*

HLA-G Gene (Fold change)

1.17 ± 0.63

14.11 ± 4.52

0.000*

ICAM1 DNA methylation (Fold change)

1.20 ± 0.57

15.13 ± 4.28

0.000*

HLA-G DNA methylation (Fold change)

1.25 ± 0.53

15.21 ± 4.33

0.000*

ICAM1 Protein (Fold change)

0.98 ± 0.07

1.22 ± 0.12

0.000*

HLA-G Protein (Fold change)

0.98 ± 0.07

1.22 ± 0.12

0.000*

Continuous variables are presented as mean ± SD. *P < 0.05 was considered significant.
AFC: antral follicular count AMH: anti Müllerianhormone ,MII: Retrieved oocytes were classified into metaphase II

The clinical pregnancy rate (fetal heart detection by ultrasound) was significantly different between the two groups. Indeed, the clinical pregnancy rate was higher in the healthy lifestyle group than in Group 2 (1.80 ± 0.41 vs. 1.70 ± 0.46, 0.040). There was a statistically significant difference in the clinical variables of total embryo number and oocyte quality between the groups. The number of total embryo and oocyte quality was statistically significantly higher in Group 1 than in Group 2 (oocyte quality: 3.51 ± 1.05 vs. 2.50 ± 0.93, p-0.000; total embryo: 1.93 ± 026 vs. 1.30 ± 0.56, p = 0.000). In addition, a marginally significant difference was observed in the AFC level between the two groups (p = 0.051). There was no statistically significant difference in age, marriage duration, infertility duration, and AMH level.

As Fig [3, (a,b)] shows, the relative expression of ICAM1 and HLA-G genes and proteins was significantly increased in Group 1 (p = 0.000) than in Group 2.

A notable increase in ICAM1 andHLA-G DNA methylation was observed in Group 2 and in Group 1 (p = 0.000), (Fig. 3, c).

Based on the univariate analysis (Table 3, 4), oocyte number (p = 0.03), and oocyte quality, total embryo, and type of group (p = 0.000) in the groups studied (women with and without a healthy lifestyle) were significantly associated with ICAM1 andHLA-G gene expression and DNA methylation. Variables of oocyte number, and oocyte quality, total embryo, MII number and type of group were significantly associated (Table2) with ICAM1 and HLA-G proteins expression in the groups studied.

Table 3

Univariate regression analysis for factors associated with ICAM1and HLA-G Genes,proteins and DNA methylation

 

ICAM1 Gene

HLA-G Gene

ICAM1 Protein

HLA-G Protein

B

SE

P value

B

SE

P value

B

SE

P value

B

SE

P value

Age

-0.105

0.204

0.609

0.108

0.204

0.598

-0.005

0.004

0.277

-0.004

0.004

0.322

AMH

3.11

3.014

0.305

3.09

3.01

0.307

0.50

0.065

0.444

0.044

0.065

0.499

AFC

1.15

0.65

0.079

1.14

0.64

0.081

0.024

0.014

0.088

0.022

0.014

0.108

no .Oocyte

1.99

0.903

0.030

1.98

0.901

0.031

0.058

0.019

0.003

0.056

0.019

0.004

no. MII

1.66

0.91

0.073

1.66

0.91

0.073

0.051

0.019

0.009

0.049

0.019

0.013

Oocyte quality

2.62

0.67

0.000

2.62

0.67

0.000

0.052

0.015

0.001

0.050

0.015

0.001

Total embryo

7.34

1.28

0.000

7.32

1.28

0.000

0.149

0.028

0.000

0.147

0.028

0.000

no .Pregnancy

-0.91

1.89

0.631

-0.903

1.88

0.632

-0.022

0.040

0.589

-0.017

0.o40

0.674

Group

(Without healthy lifestylevs. With healthy lifestyle)

-12.96

0.722

0.000

-12.96

0.721

0.000

-0.242

0.022

0.000

-0.240

0.022

0.000

B; Unstandardized coefficient, SE; Standard error, AFC: antral follicular count, AMH: Anti Müllerian hormone

Table 4

Multivariate regression analysis for factors associated with ICAM1and HLA-G Genes and proteins

 

ICAM1 Gene

HLA-G Gene

ICAM1 Protein

HLA-G Protein

B

SE

P value

B

SE

P value

B

SE

P value

B

SE

P value

AMH

           

-0.148

0.061

0.018

-0.149

0.062

0.018

no. MII

           

0.048

0.019

0.016

0.046

0.020

0.021

Group

(Without healthy lifestylevs. With healthy lifestyle)

-12.96

0.722

0.000

-12.96

0.721

0.000

-0.233

0.022

0.000

-0.231

0.022

0.000

B; Unstandardized coefficient, SE; Standard error, AMH: anti Müllerian hormone

After adjusting all variables, based on the multiple linear regression model results, the AMH level had negatively significant association with the ICAM1 and HLA-G proteins expression (p = 0.018); for each unit increase in the AMH level, the expected oocyte quality was decreased by 0.14. The multiple linear regression model indicated that the number of MII was positively associated with the ICAM1 and HLA-G proteins expression (p = 0.021); for each unit increase in AMH, the expected ICAM1 and HLA-G proteins expression was increased by 0.04. This multivariate model revealed in groups 1 and 2 was negatively associated with ICAM1 and HLA-G genes. Although the ICAM1 and HLA-G protein expression in women without a healthy lifestyle was decreased by nearly 13 times than in women with a healthy lifestyle (p = 0.000), the expected ICAM1 and HLA-G protein expression was decreased by nearly 0.23 times in women without a healthy lifestyle than in women with a healthy lifestyle (p = 0.000) (Table 4). The rest of variables included in the univariate model (Table 3) were not significantly associated with oocyte quality in multiple models.

Discussion

In the present study, our finding showed that ICAM-1 and HLA-G transcripts and proteins in POR with healthy lifestyle were upregulated compared to the POR without a healthy lifestyle. In addition, the ICAM-1 and HLA-G DNA methylation status in the POR with a healthy lifestyle was hypomethylated compared to the POR without a healthy lifestyle. In fact, lifestyle habit can affect the transcript and proteins profile alterations in cumulus cells from POR patients. Furthermore, it may affect the methylation status in the cumulus cells of POR patients. Moreover, based on our data, the groups without a healthy lifestyle showed significantly lower numbers of good quality oocytes and clinical pregnancy rate compared to the group with a healthy lifestyle. As a result, lifestyle habit can affect the ovulation, oocyte maturation and clinical pregnancy rate in POR patients. Oocyte quality is one of the major limiting factors for the success of the ART cycle in POR patients [19]. Oocyte maturation is achieved during folliculogenesis. Folliculogenesis is the maturation of the ovarian follicle. Folliculogenesis needs communication between the oocyte and surrounding somatic cells [20]. One of the kinds of somatic cells is cumulus cells. Cumulus cells play a potential role in achieving oocyte development [21]. Based on a recent study, some genes are expressed in the cumulus cells that can be considered genetic markers for the association between oocyte and embryo quality. Probably, developmental signals from cumulus cells are transferred to the oocyte via gap junctions and pathways, which can affect the oocyte maturation [22]. The present study aimed to determine the novel biochemical marker to evaluate the oocyte quality and clinical pregnancy rate in the ART treatment of POR patients, and our data showed that lifestyle might be considered a biochemical marker for oocyte maturation increasing the clinical pregnancy rate. One of the consequent complications of POR is oocyte maturation disorder [23]. Nowadays, overuse of plastic objects to store hot food and hot drinks has caused numerous health problems in humans like reproduction disorders, including POR [24]. In fact, plastic objects made of polycarbonate and polycarbonates contain BPA [25]. According to the recent study, BPA may affect the infertility related gene expression that is closely associated with reproductive, but this association interaction mechanism is unclear. Owing to heat, acidic or alkaline substances, polycarbonate will break and BPA can attach to food and drink, and it can enter the body [26]. BPA can act like sex hormones, so that it can disrupt the endocrine system and hormonal signaling pathways. BPA in small amounts may interfere in the reproduction related gene expression, and it can interfere in the function of ovary, uterus and other reproductive organs [27]. Pednekar et al. [28] reported that infertile women showed significantly higher plasma concentrations of BPA. As a result, lifestyle habit plays a vital role in the regulating of ovarian response, oocyte maturation, reproductive process and genes expression. In this study, we evaluated the expression profile for ICAM-1 and HLA-G genes in the cumulus cells of infertile women with poor response to ovarian stimulation (POR) based on their healthy lifestyle. The probable effect of plastic objects usage was observed on the genes, proteins, DNA methylation pattern, oocyte quality and clinical pregnancy rate in these women. According to other studies, altered gene expression of CCs may play a role in women with POR pathogenesis [29, 30]. Therefore, assessment of genes expression is valuable to understand the etiology of infertile women with POR patients, since oocyte maturation dysfunction is one of the disorders in POR patients [31]. Recently, it has been shown that alternation of ICAM1 and HLA-G genes can affect the quality and quantity of oocytes [32]. Our findings revealed that a major contributor to oocyte maturity disorder and infertility in the POR patients without a healthy lifestyle can be a reduction in ICAM1 and HLA-G transcript and proteins expression as well as hypermethylation of DNA in ICAM1 and HLA-G. According to recent studies, comparison of POR patients to healthy, normal ovulatory fertile women with a history of male infertility history has shown differential expression of ICAM-1 genes [33].

The ICAM-1 gene expression pattern indicated that it is important in oocyte maturity and grading. It appeared that dysregulation of ICAM-1 expression could be associated with defective oocyte maturation and grading [34].The studies demonstrated that ICAM-1 was a probable key effector of the oocyte maturation. Although the sICAM-1 release was very high in immature oocytes, it was decreased in mature oocytes. They regarded sICAM-1 as a biochemical marker for oocyte maturation and grading [35]. The ICAM-1 appears to play a critical role in oocyte development, while ICAM-1 levels play the role of a predictive marker in oocyte maturation and quality. Borgtti and Rizzo [36] proposed sICAM-1 as a biochemical marker for oocyte maturation and grading. Their findings supported a significant correlation between ICAM-1 gene and protein expression and oocyte maturation. A recent study has shown that ICAM-1 is expressed on leukocytes, epithelial and endothelial cells [37]. Furthermore, HLA-G plays a vital role in the oocyte maturation and preimplantation of embryos. Jurisicova et al. detected HLA-G mRNA expression by preimplantation human embryos [38]. They demonstrated the presence of HLA-G mRNA and protein in the blastocysts tested [39]. Yao et al. [40] revealed the expression of the HLA-G protein by human preimplantation embryos with an increasing ratio of positive embryos with developmental stage. Kaneko [41] indicated that the low expression of HLA-G in cumulus cells was associated with lower oocyte quality, poor fertilization, and reduced embryo development [42]. Rooij van reported the low level of HLA-G in the cumulus cells of POR patients [43]. The gene expression alternation in the cumulus cells of women with POR after dehydroepiandrosterone (DHEA) supplementation can affect the oocyte maturation [44]. According to recent study, oocyte quality decreases with increasing age [45]. Our findings indicated that age was directly associated with oocyte quality and aging. Furthermore, another study showed that younger POR patients’ quality of oocyte and grading were better than older patients’ [46]. Our study also highlighted the role of age as one of the components in oocyte maturation. The present study emphasized the association of lifestyle with the ICAM1 and HLA-G genes expression alteration and oocyte quality as an opinion for the deeper understanding of the etiology of infertile women with POR patients.

Conclusion

The current study was the first single center lifestyle program of POR patients in Iran. We found that upregulation of ICAM1 and HLA-G in POR appeared to be related to nutrition and lifestyle. The nutrition and lifestyle may be effective on the methylation pattern, expression profile and proteins level in cumulus cells from POR. Moreover, nutrition and lifestyle may act as a biochemical marker for ovulation and the oocyte maturation process.

Abbreviations

POR

Poor Ovarian Response

AMH

Anti Müllerian Hormone

AFC

Antra Follicle Count

BPA

BisphenolA

ECM

Extracellular Matrix

HLA-G

Human Leukocyte Antigen-G

CCs

Cumulus Cells

GnRH

Gonadotropin Releasing Hormone

ICSI

Intracytoplasmic Sperm Injection

COS

Controlled Of Ovarian Stimulation

SC

Subcutaneous

RFSH

Recombinant Follicle Stimulating Hormone

E2

Estradiol

HCG

Human Chorionic Gonadotropin

IM

Intramuscular

CoCs

Cumulus-Oocyte Complexes

PBS

Phosphate-Buffered Saline

MII

Metaphase II Gametes

MSP

Methylation-Specific Polymerase Chain Reaction

DHEA

Dehydroepiandrosterone

Declarations

Funding 

Not available

Conflicts of interest/Competing interests 

Authors have NO conflict of interest

Availability of data and material 

They are available on the request

Code availability 

None

Authors' contributions 

ZN and AM: conceptionalization of project, ZN and SA: data analyses, SA: collection and laboratory work, MK: data collection

Acknowledgments

This study was funded by Royan Institute (code number: 94000163). The authors wish to thank ForoughAzamSayahpourandMaryam Jahangiri. The authors also would like to thank the patients for their cooperation. The author(s) declared no potential conflicts of interest with respect to the research, authorship, and publication of this article.

References

  1. Ferraretti A-P, Marca A, Fauser B-C, Tarlatzis B, Nargund G, Gianaroli L. ESHRE working group on Poor Ovarian Response Definition. ESHRE consensus on the definition of ‘poor response’ to ovarian stimulation forin vitro fertilization: the Bologna criteria. Hum Reprod. 2011. 26:1616–1624.Nagels HE. Androgens (dehydroepiandrosterone or testosterone) for women undergoing assisted reproduction.Cochrane Database Syst Rev. 2015. 26(11): CD009749.
  2. Pandian Z, McTavish A-R, Aucott L, Hamilton P-L, Bhattacharya S. View authors' declarations of interest Interventions for ‘poor responders’ to controlled ovarian hyper stimulation (COH) in in-vitro fertilisation (IVF). Cochrane Database Syst Rev.2010. 20(1): CD004379.
  3. Humaidan P, Alviggi C, Fischer R, Esteves S-C. The novel POSEIDON stratification of ‘Low prognosis patients in Assisted Reproductive Technology’ and its proposed marker of successful outcome. F1000 Research. 2016.5:2911.
  4. Patrizio P, Vaiarelli A, Setti P-E-L, Tobler K, Shoham G, Leong M, Shooham Z. How to define, diagnose and treat poor responders? Responses from a worldwide survey of IVF clinics.Reprod Biomed Online. 2015. 30(6): 581–592.
  5. Xiaona H, Dan C-h, Yonghua H-e, Wenting Z, Wei Z, Jun Z. Bisphenol-A and Female Infertility: A Possible Role of Gene-Environment Interactions. Int J Environ Res Public Health. 2015;12:11101–16.
  6. Liu F, Liu W-N, Guo ZhaoQ-X, Zhuang W, Jia R. X-D. Study on environmental and psychological risk factors for female infertility. Chinese Journal of Industrial Hygiene Occupational Diseases. 2013;31:922–3.
  7. Nishikawa M, Iwano H, Yanagisawa R. Placental transfer of conjugated bisphenol A and subsequent reactivation in the rat fetus. Environ. Health Perspect.2010. 118, 1196–1203.
  8. Dang V-H, Choi K-C, Jeung E-B. Tetra bromo diphenyl ether (BDE 47) evokes estrogenicity and calbindin-D9k expression through an estrogen receptor-mediated pathway in the uterus of immature rats. Toxicol Sci. 2007;97:504–11. 5.
  9. Choi K-C, Jeung E-B. The biomarker and endocrine disruptors in mammals. J Reprod Dev. 2003;49:337–45.
  10. Borgatti MRizzoR, Dal Canto M-B, Fumagalli D,Renzini M-M, Fadini R, Stignani M, Baricordi O-R. Gambari R.Release of sICAM-1 in Oocytes andIn VitroFertilizedHuman Embryos. PLoS ON December 2008. 3: 12, e3970.
  11. Rebmann V, Switala M, Eueb I, Schwah E, Merzenich M, Wilde H-G. Rapid evaluation of soluble HLA-G levels in supernatants of in vitrofertilized embryos. Human Immunol.2007. 68: 251–258.
  12. Uzzi B, Rizzo R, Ivo Noci, Melchiorri L, Scarselli B, Bencini E, Menicucci A. BaricordiO-R.HLA-Gexpression in early embryos is a fundamental prerequisite for the obtainment ofpregnancy. European J Immunol. 2002;32:311–5.
  13. OciI,Fuzzi B, Rizzo R, Melchiorri L, Criscuoli L, Dabizzi S, Biagiotti R, Pellegrini S, Menicucci A, Baricordi O-R. Embryonicsoluble HLA-G as a marker of developmental potential in embryos. HumanReprod.2005. 20: 138–146.
  14. Borgatti M, Rizzo R, Dal Canto M-B, Fumagalli D,Renzini M-M, Fadini R, Stignani M, Baricordi O-R, Gambari R. Release of sICAM-1 in Oocytes andIn VitroFertilizedHuman Embryos. PLoS ON December 2008. 3: 12, e3970.
  15. Wissing M-L, Sonne S-B, Westergaard D, Nguyen K-h, Belling K, Høst T-H, Mikkelsen A-L. The transcriptome of corona radiata cells from individual MІІ oocytes that after ICSI developed to embryos selected for transfer: PCOS women compared to healthy women. J Ovarian Res. 2014;7:110.
  16. Wissing M-L, Sonne S-B, Westergaard D, Nguyen K-h, Belling K, Høst T-H, Mikkelsen A-L. The transcriptome of corona radiata cells from individual MІІ oocytes that after ICSI developed to embryos selected for transfer: PCOS women compared to healthy women. J Ovarian Res. 2014;7:110.
  17. Valojerdi M-R, Karimian L, Yazdi P-E, Sadighi Gilani M-A, Madani T, Baghestani A-R. Efficacy of a human embryo transfer medium: a prospective, randomized clinical trial study. J Assist Reprod Genet. 2006;23(5):207–12.
  18. Einstein A, Podolsky B, Rosen N. Impact of insulin resistance onthe developmental potential of immature oocytes retrieved from human chorionic gonadotropin primed women with polycystic ovary syndrome undergoing in vitro maturation. FertilSteril. 2009;91(3):957–9.
  19. Knight PG, Glister C. Local roles of TGF-beta superfamily members in the control of ovarian follicle development. AnimReprod Sci. 2003;78(3–4):165–83.
  20. Cecconi S, Ciccare C, Barberi M, Macchiare G, Caniparic R. Granulosa cell-oocyte interactions. Eur J ObstetGynecolReprod Biol. 2004;115(Suppl 1):19–22.
  21. Salehi EAflatoonian R, Moeini A, Yamini N, Asadi E, Khosravizadeh Z, Tarzjani M-D, Naghibi harat Z, Abolhassani F. Apoptotic biomarkers in cumulus cells in relation to embryo quality in polycystic ovary syndrome. Gynecol Obstet. 2017;296:1219–27.
  22. Patrizio P, Vaiarelli A, Setti P-E-L, Tobler K, Shoham G, Leong M, Shooham Z. How to define, diagnose and treat poor responders? Responses from a worldwide survey of IVF clinics.Reprod Biomed Online. 2015. 30(6): 581–592.
  23. Ziv-Gal A, Flaws J-A. Evidence for bisphenol A-induced female infertility. Fertility and Sterility. 2016–2017. 106: 4:827–856.
  24. Liu F, Liu W-N, Zhao Q-X, Guo W, Zhuang R, Jia X-D. Study on environmental and psychological risk factors for female infertility. Chinese Journal of Industrial Hygiene Occupational Diseases. 2013;31:922–3.
  25. Xiaona H, Dan C-h, Yonghua H-e, Wenting Z, Wei Z, Jun Z. Bisphenol-A and Female Infertility: A Possible Role of Gene-Environment Interactions. Int J Environ Res Public Health. 2015;12:11101–16.
  26. Ziv-Gal A, Flaws J-A. Evidence for bisphenol A-induced female infertility. Fertility and Sterility. 2016–2017. 106: 4:827–856.
  27. Borgatti M, Rizzo R, Dal Canto M-B, Fumagalli D, Renzini M-M, Fadini R, Stignani M, Baricordi O-R, Gambari R. Release of sICAM-1 in Oocytes andIn VitroFertilizedHuman Embryos. PLoS ON December 2008. 3: 12, e3970.
  28. Tsuia K, Lin L, Horng H-C, Shian C-B, Chenga J-T, Wang P-H. Gene expression of cumulus cells in women with poor ovarian response after dehydroepiandrosterone supplementation. Taiwanese Journal of Obstetrics Gynecology. 2014;53(4):559–65.
  29. Van Rooij, Bancsi F-J-M-L, Broekmans F, Looman C-W-N, Habbema D, Velde E-R. Women older than 40 years of age and those with elevated follicle-stimulating hormone levels differ in poor response rate and embryo quality in in vitro fertilization. Fertility sterility. 2003;79:3.
  30. Borgatti M, Rizzo R, Dal Canto M-B, Fumagalli D, Renzini M-M, Fadini R, Stignani M, Baricordi O-R, Gambari R. Release of sICAM-1 in Oocytes andIn VitroFertilizedHuman Embryos. PLoS ON December 2008. 3: 12, e3970.
  31. Thomson A, Greer M-R, Young A, Boswell F, Telfer J-F, Cameron I-T, Norman J-E, Campbell S. Expression of intercellular adhesion molecules ICAM-1 and ICAM-2in human endometrium: regulation by interferon-g. Mol Hum Reprod. 1999;5:1:64–70.
  32. Oci I, Fuzzi B, Rizzo R, Melchiorri L, Criscuoli L, Dabizzi S, Biagiotti R, Pellegrini S, Menicucci A, Baricordi O-R. Embryonicsoluble HLA-G as a marker of developmental potential in embryos. HumanReprod.2005. 20: 138–146.
  33. Opiela O, Romanek J, Lipiński, Smorąg Z. Effect of Hyaluronan on Developmental Competence and Quality of Oocytes and Obtained Blastocysts from In Vitro Maturation of Bovine Oocytes.BioMed Research International. 2014; 519189.
  34. Borgatti M, Rizzo R, Dal Canto M-B, Fumagalli D, Renzini M-M, Fadini R, Stignani M, Baricordi O-R, Gambari R. Release of sICAM-1 in Oocytes andIn VitroFertilizedHuman Embryos. PLoS ON December 2008. 3: 12, e3970.
  35. Thomson A, Greer M-R, Young A, Boswell F, Telfer J-F, Cameron I-T, Norman J-E, Campbell S. Expression of intercellular adhesion molecules ICAM-1 and ICAM-2in human endometrium: regulation by interferon-g. Mol Hum Reprod. 1999;5:1:64–70.
  36. Borgatti M, Rizzo R, Dal Canto M-B, Fumagalli D, Renzini M-M, Fadini R, Stignani M, Baricordi O-R, Gambari R. Release of sICAM-1 in Oocytes andIn VitroFertilizedHuman Embryos. PLoS ON December 2008. 3: 12, e3970.
  37. Jurisicova A, Casper R-F, MacLusky N-J, Librach CL. Embryonic human leukocyteantigen-G expression: Possible implications for human preimplantation development. FertilSteril. 1996;65:997–1002.
  38. Uzzi B, Rizzo R, Criscuoli L, Noci I, Melchiorri L, Scarselli B, Bencini EMenicucci A. Baricordi O-R.HLA-G expression in early embryos is a fundamental prerequisite for the obtainment of pregnancy. European J Immunol. 2002;32:311–5.
  39. Yao YQ, Barlow D-H, Sargent J-L. Differential expression of alternatively splicedtranscripts of HLA-G in human preimplantation embryos and inner cellmasses. J Immunol. 2005;175:8379–85.
  40. Jurisicova A, Casper R-F, MacLusky N-J, Librach CL. Embryonic human leukocyteantigen-G expression: Possible implications for human preimplantation development. FertilSteril. 1996;65:997–1002.
  41. Yao YQ, Barlow D-H, Sargent J-L. Differential expression of alternatively splicedtranscripts of HLA-G in human preimplantation embryos and inner cellmasses. J Immunol. 2005;175:8379–85.
  42. Rebmann V, Switala M, Eueb I, Schwah E, Merzenich M, Wilde H-G. Rapid evaluation of soluble HLA-G levels in supernatants of in vitrofertilized embryos. Human Immunol.2007. 68: 251–258.
  43. Tsuia K, Lin L, Horng H-C, Shian C-B, Chenga J-T, Wang P-H. Gene expression of cumulus cells in women with poor ovarian response after dehydroepiandrosterone supplementation. Taiwanese Journal of Obstetrics Gynecology. 2014;53(4):559–65.
  44. Van Rooij, Bancsi F-J-M-L, Broekmans F, Looman C-W-N, Habbema D, Velde E-R. Women older than 40 years of age and those with elevated follicle-stimulating hormone levels differ in poor response rate and embryo quality in in vitro fertilization. Fertility sterility. 2003;79:3.
  45. Perna L, Zhang Y, Mons U, Holleczek B, Saum K-U, Brenner H. Epigenetic age acceleration predicts cancer, cardiovascular, and all-cause mortality in a German case cohort. Clin Epigenetics. 2016;8:64.