Skip to main content
Erschienen in: Reproductive Biology and Endocrinology 1/2012

Open Access 01.12.2012 | Research

Effect of luteal-phase support on endometrial microRNA expression following controlled ovarian stimulation

verfasst von: Yulian Zhao, Howard Zacur, Chris Cheadle, Ning Ning, Jinshui Fan, Nikos F Vlahos

Erschienen in: Reproductive Biology and Endocrinology | Ausgabe 1/2012

Abstract

Background

Studies suggested that microRNAs influence cellular activities in the uterus including cell differentiation and embryo implantation. In assisted reproduction cycles, luteal phase support, given to improve endometrial characteristics and to facilitate the implantation process, has been a standard practice. The effect of different types of luteal phase support using steroid hormones in relation to endometrial miRNA profiles during the peri-implantation period has not seen described. This study was designed to evaluate the expression of miRNAs during the luteal phase following controlled ovarian stimulation for IVF and the influence of different luteal phase support protocols on miRNA profiles.

Methods

The study was approved by the Johns Hopkins Hospital Institutional Review Board. Endometrial biopsies were obtained on the day of oocyte retrieval from 9 oocyte donors (group I). An additional endometrial biopsy was obtained 3–5 days later (Group II) after the donors were randomized into three groups. Group IIa had no luteal-phase support, group IIb had luteal support with micronized progesterone (P), and Group IIc had luteal support with progesterone plus 17-beta-estradiol (P + E). Total RNA was isolated and microarray analysis was performed using an Illumina miRNA expression panel.

Results

A total of 526 miRNAs were identified. Out of those, 216 miRNAs were differentially regulated (p < 0.05) between the comparison groups. As compared to the day of retrieval, 19, 11 and 6 miRNAs were differentially regulated more than 2 fold in the groups of no support, in the P support only, and in the P + E support respectively, 3–5 days after retrieval. During the peri-implantation period (3–5 days after retrieval) the expression of 33 and 6 miRNAs increased, while the expression of 3 and 0 miRNAs decreased, in the P alone and in the P + E group respectively as compared to the no steroid supplementation group.

Conclusion

Luteal support following COS has a profound influence on miRNA profiles. Up or down regulation of miRNAs after P or P + E support suggest a role(s) of luteal support in the peri-implantation uterus in IVF cycles through the regulation of associated target genes.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1477-7827-10-72) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

YZ, NV designed the protocol, developed the study, managed specimens, and drafted the manuscript. HZ provided valuable suggestions for the study and actively involved in manuscript preparation. NN participated in data analysis and manuscript preparation. CC, JF carried out microarray analysis, performed the statistical analysis, and contributed in manuscript preparation. All authors read and approved the final manuscript.
Abkürzungen
miRNA
MicroRNAs
COS
Controlled ovarian stimulation
GnRH
Gonadotropic releasing hormones
IVF
In vitro fertilization
hCG
Human chorionic gonadotropin
LH
Luteinizing hormone
FSH
Follicle-stimulating hormone
P
Progesterone
P + E
Progesterone plus 17-beta-estradiol
IPA
Ingenuity pathway analysis.

Background

MicroRNAs (miRNAs) are a class of single-stranded, non-coding small RNAs that regulate gene expression at the translational level and play fundamental roles in several biological processes, including cell differentiation, proliferation, development and apoptosis [13]. It is believed that mammalian miRNAs are responsible for the regulation of over 60% of all human genes [4]. Either by controlling mRNA degradation or by translational repression, miRNAs have emerged as key regulators of gene expression [5, 6]. Each miRNA is predicated to have a broad range of target mRNAs and each mRNA may be regulated by multiple miRNAs [7, 8].
The role of miRNAs in the female reproductive system and particularly in the endometrium has been the focus of several studies in recent years [9, 10]. So far it has been established that miRNAs are indeed expressed in the human endometrium and they are also subjected to hormonal regulation [10, 11]. Hawkins et al. were able to identify a number of miRNAs that were differentially regulated in endometriotic tissues as compared to normal endometrium [12]. The overall regulatory role of miRNAs in the pathophysiology of endometriosis has been reviewed extensively by Ohlsson Teaque et al.[13].
Ovarian stimulation protocols with gonadotropins have been invariably associated with luteal phase deficiency and poor implantation rates [14, 15]. While the exact reasons for this phenomenon are still unclear, luteal phase support, given to improve endometrial characteristics and to facilitate the implantation process, has been a standard practice. Progesterone is a universally accepted agent for luteal phase support and can be administered orally, intramuscularly, or vaginally [16, 17]. Estrogens in the form of 17β- estradiol or estradiol valerate have also been used for luteal phase support [18], although studies aimed to evaluate the concept of estrogen addition during the luteal phase have lead to inconclusive results [14, 19] . It has been suggested that during ovarian stimulation for IVF, the endometrial receptivity starts to occur in mid luteal phase after oocyte retrieval [20]. Prior to, and during the implantation process, the expression of multiple endometrial genes and gene products is highly regulated [2123]. The role of miRNAs in regulating cellular processes during the endometrial transition has recently attracted a great deal of attention [10, 2428]. For example, Kuokkanen et al. reported distinct miRNA gene expression signatures in the late proliferative and mid-secretory phase endometrial epithelium [24]. However, the effect of different types of luteal support in relation to endometrial miRNA profiles during the period of peri-implantation has not been described. In this study, we have investigated the impact of two commonly used luteal phase support protocols, progesterone alone and progesterone plus estrogen, on the expression profiles of 526 miRNAs in the human endometrium following ovarian stimulation with a gonadotropin/ GnRH antagonist protocol.

Methods

Oocyte donors and ovarian stimulation

The study was approved by the Johns Hopkins Hospital Institutional Review Board. Nine oocyte donors who enrolled in the Johns Hopkins oocyte donation program participated in the study. All donors were 21 to 31 years of age and underwent a standard screening protocol for oocyte donation, in accordance with the recommendations of the American Society for Reproductive Medicine [29]. The risks of the procedure were discussed in detail, with particular emphasis on the risks associated with the endometrial biopsy and the use of steroids during luteal phase, and written informed consents were obtained.
Study subjects underwent ovarian stimulation according to a gonadotropin / GnRH antagonist protocol as described previously [30]. Briefly, ovarian stimulation was initiated with gonadotropins on the second day of vaginal bleeding following discontinuation of oral contraceptive pills. On the 6th day of stimulation, a daily subcutaneous evening dose of 0.25 mg ganirelix acetate (Schering-Plough Corp, West Orange, NJ, USA) was added. When at least three follicles reached a mean diameter of 18 mm, ovulation was triggered with a single dose of hCG (Profasi, 10,000 IU; Serono Inc. Rockland, MA, USA). Sonographically guided transvaginal oocyte retrieval was performed 34–36 hours after the hCG administration. The retrieved oocytes were used for IVF procedures and the resulting embryos were either transferred to matched recipients or cryopreserved for future use.

Luteal-phase support and tissue collection

Endometrial biopsies on oocyte donors were performed using a Pipelle catheter (Unimar, Wilton, CT) on the day of oocyte retrieval and served as baseline (group I). At that time, the donors were randomized into three groups, with three subjects in each group. Group IIa received no luteal phase support after retrieval. Group IIb had luteal phase support with micronized progesterone (P) in the form of vaginal suppositories (200 mg every 6 h starting from the day after retrieval). Group IIc received a daily oral dose of 2 mg 17β-estradiol in addition to the micronized progesterone (P + E). Endometrial biopsies were obtained again 3–5 days (each of treatment groups contains 2 samples from day 3 and 1 sample from day 5) after retrieval. All specimens were stored in liquid nitrogen at −196°C immediately after the biopsy.

RNA preparation and miRNA analysis

Total RNA was isolated and extracted from individual endometrial samples using the Trizol Reagent method (Invitrogen, Carlsbad, California 92008, cat. no. 15596–026). The quality of the RNA samples was assessed using an Agilent 2100 Bioanalyzer (Agilent Technologies, Palo Alto, CA). The integrity of miRNA was assessed by a miRNA specific RT-PCR using an ABI (Applied Biosystems; Foster City, CA) Taqman assay for U6 snRNA (AB Assay ID 001973). The results indicated an average Ct of 20.1 (SD 0.84) for all samples with a minimum Ct of 18.3 and maximum Ct of 22.
Illumina miRNA expression profiling (Catalog # MI-501-1001) was carried out according to manufacturer’s recommended protocols. Briefly 200ngs of total RNA for each sample was polyadenylated and converted to cDNA using a biotinylated oligo-dT primer with a universal PCR sequence at its 5’-end. Biotinylated cDNA was annealed to query oligos. Each query oligo consisted of a universal PCR priming site at the 5’end, an address sequence that complements a corresponding capture sequence on the array, and a microRNA-specific sequence at the 3’end. This mixture was bound to streptavidin-conjugated paramagnetic particles to select the cDNA/oligo complexes; second strand cDNA synthesis was completed by primer extension. All cDNA templates were amplified with a pair of common PCR primers. The primer on the strand complementary to the array was fluorescently labeled for subsequent hybridization to the arrays.
Validation of the selected miRNAs, shown to be regulated by Illumina miRNA microarray, was performed by RT-PCR. QRT-PCR was performed using the RT2 ProfilerTM Human miFinder miRNA PCR Array (MAH-001A) from SuperArray (SABiosciences, Gaithersburg, MD). RT2 Profiler™ PCR Arrays are designed for relative quantitative QRT-PCR based on SYBR Green detection and performed on a one sample/one plate 96-well format, using primers for a preset list of 88 most abundantly expressed and best characterized micro RNA sequences. In brief, miRNA was converted to cDNA via a universal tailing and reverse transcription reaction. CDNA volumes were adjusted to ~2.5 ml with SuperArray RT2 Real-Time SYBR Green/ROX PCR 2X Master Mix (PA-012) and 25 μl of cDNA mix was added to all wells. The PCR plate was sealed and spun at 1500 rpm X 4 min. Real time PCR was performed on an Applied Biosystem (Foster City, CA) 7300 Real Time PCR System. ABI instrument settings included setting reporter dye as “SYBR”, passive reference is “ROX”; Delete UNG Activation, and add Dissociation Stage.
To correlate differentially expressed miRNAs and their regulated genes, we used differentially regulated and selected miRNAs against an established miRNA database for predicted target genes (Sanger miRBase, v9.1, February 2007 release). MicroRNA data was also analyzed through the use of Ingenuity Pathway Analysis (IPA, Ingenuity® Systems, http://​www.​ingenuity.​com). Pathway enrichments were calculated using the NIAID DAVID functional enrichment tool [31, 32].

Statistical analysis

Preliminary analysis of the scanned data was performed using Illumina BeadStudio software which returns single intensity data values/miRNA following the computation of a trimmed mean average for each probe type represented by a variable number of bead probes/gene on the array. Data was globally normalized by scaling each array to a common median value, and significant changes in gene expression between class pairs were calculated using the Student t-test. Significant gene lists were calculated by selecting genes which satisfied a significance threshold criteria of t-test p-values less than or equal to 0.05 and a fold change ± 2 or greater.
Relative miRNA expression derived from QRT-PCR was calculated by using the 2-Ct method, in which Ct indicates cycle threshold, the fractional cycle number where the fluorescent signal reaches detection threshold [33]. The normalized ΔCt value of each sample is calculated using an endogenous control small molecular weight RNA (U6 snRNA). Fold change values are presented as average fold change = 2-(average Ct) for genes in treated relative to control samples. The criteria of significance used for the RT-PCR results were the same as used for the Illumina miRNA arrays.

Results

Demographic characteristics

Demographic characteristics for all study participants were similar in all treatment groups. The mean age of the study participants was 24 years and mean body mass index was 21.3 ± 1.2 kg/m2. Overall, the baseline serum FSH, LH and E2 levels, the length of the stimulation , total amount of gonadotropins used, peak estradiol levels, and number of oocytes retrieved were comparable (P > 0.05) between the groups (Table 1).
Table 1
Group characteristics
Characteristics
Group IIa (no support)
Group IIb (P support)
Group IIc (P + E support)
P
N
3
3
3
 
Age (yr)
25.7 ± 3.2
23.6 ± 0.8
22.8 ± 1.2
0.494
BMI (kg/m 2 )
23.3 ± 1.4
21.6 ± 1.8
20.2 ± 1.2
0.096
Day 2 FSH (mIU/ml)
4.5 ± 0.9
5.6 ± 1.1
4.0 ± 0.3
0.178
Day 2 LH (mIU/ml)
2.4 ± 0.8
4.0 ± 1.3
5.2 ± 0.2
0.507
Day 2 E2 (pg/ml)
36.7 ± 11.6
34.5 ± 12.5
20.5 ± 3.5
0.646
Gonadotropins used (IU)
2850 ± 525
2400 ± 645
2625 ± 675
0.387
Peak E2 level (pg/ml)
1928 ± 100.0
2514 ± 400
2625 ± 480
0.563
Days of stimulation
10.3 ± 1.1
9.3 ± 1.2
10.1 ± 0.7
0.588
No. of oocytes
14.5 ± 5
18.4 ± 3
16.0 ± 4
0.398

MiRNA profiles and comparisons between groups

To establish endometrial miRNA profiles, we used a microarray platform consisting of 526 miRNA probes. Triplicates of each group samples were used, which proved that genes from same condition of samples are reproducible. Levels of miRNA expression are similar in the same sample groups including the samples from either day 3 or day5. The fluorescent intensity of each expressed transcript in each sample group was compared to the median fluorescence intensity of each transcript in the paired comparison group. Individual transcripts with increased (red) and decreased (green) miRNA abundance in the given comparisons were identified, as shown in the hierarchical clustering map in Figure 1. It is demonstrated that there is a high degree of overall concordance between and within treatments for later versus early luteal phase and, in particular a striking concordance, for hormone treated versus non-treated groups at days 3–5 after oocyte retrieval. Following global normalization, the mean expression value for each group was subjected to statistical analysis. A 2 fold change in the expression was arbitrarily selected as a cut-off level. Individual miRNAs that have shown a significant change in their expression (greater than 2fold and/or p < 0.05 between the comparison groups) are shown in an Additional file 1: Table S1 with a total of 248 miRNAs listed.
Initially we compared miRNA expression in the endometrial samples obtained on the day of retrieval to those obtained 3–5 days later (Figure 2, the 3 comparison columns on the left). In the group with no luteal phase support, 14 miRNAs (HS_202.1, HS_209.1, HS_284.1, hsa-miR-202*:9.1, hsa-miR-346, hsa-miR-363*, hsa-miR-504, hsa-miR-569, hsa-miR-302d, hsa-miR-632, HS_17, HS_145.1, hsa-miR-133b, hsa-miR-144:9.1) were down-regulated and 5 miRNAs were up-regulated (HS_130, hsa-miR-876-5p, hsa-miR-876-3p, hsa-miR-122, hsa-miR-9) at greater than 2 fold changes. In the P alone group, 4 miRNAs (hsa-miR-144:9.1, hsa-miR-486-5p, HS_97, HS_203) were down regulated and 7 (HS_163, hsa-miR-614, hsa-miR-610, hsa-miR-559, hsa-miR-876-5p, HS_18, hsa-miR-876-3p) were up regulated, while in the P + E support group, 1 miRNA (hsa-miR-449a) was underexpressed and 5 (HS_276.1, hsa-miR-876-5p, HS_18, HS_111, hsa-miR-876-3p) were overexpressed .
Subsequently, we compared miRNA gene expression between the different treatment groups during mid-luteal phase at 3–5 days after retrieval, as shown in Figure 2, the 3 comparison columns on the right. In the progesterone support group an overexpression (more than 2 fold increase) was observed for 33 miRNAs (HS_149, HS_166.1, HS_175, HS_202.1, HS_209.1, HS_284.1, HS_41, hsa-miR-1468, hsa-miR-202*:9.1,hsa-miR-346, hsa-miR-504, hsa-miR-512-5p, hsa-miR-560:9.1, hsa-miR-563, hsa-miR-638, hsa-miR-663, hsa-miR-302d, hsa-miR-302b*, hsa-miR-632, hsa-miR-622, HS_17, HS_163, hsa-miR-518b, HS_108.1, hsa-miR-614, hsa-miR-610, HS_263.1, HS_30, hsa-miR-512-3p, HS_32, HS_282, HS_169, HS_145.1) and in the P + E support group for 6 miRNAs (HS_149, HS_276.1, HS_41, hsa-miR-563, HS_17, hsa-miR-144:9.1) as compared to the no steroid supplementation group. On the other hand, underexpression of 3 miRNAs (HS_176, HS_97, HS_203) were seen only in P support group. In the comparison between E + P and P supplementation groups, 5 miRNAs (hsa-miR-144:9.1, hsa-miR-486-5p, HS_176, HS_97, HS_203) were up-regulated and none were down regulated at greater than 2 fold levels.

Venn diagram analysis of differentially expressed miRNA genes

A total of 216 miRNAs were differentially regulated (p < 0.05) between the study groups. MiRNAs with significant changes in common (shared miRNAs) between groups are shown in Figure 3. Panel A shows changes in miRNA expressions between day 3–5 and day of retrieval. Among the 3 comparison groups, 3 miRNAs (hsa-miR-876-3p, hsa-miR-155, and hsa-miR-503) were shared by all 3 groups and 5, 10 and 13 miRNAs respectively were shared in each pair of groups. Panel B compares groups on day 3–5 at all possible combinations. Group IIb vs. IIa and Group IIc vs. IIa shared 4 miRNAs (HS_241.1, hsa-miR-346, hsa-miR-503, and hsa-miR-99a); Group IIc vs. IIa and Group IIc vs. IIb shared 1 miRNA (hsa-miR-766) and Group IIb vs. IIa and Group IIc vs. IIb shared 3 miRNAs (hsa-miR-501-5p, hsa-miR-512-5p and hsa-miR-146a).

Validation analysis

Array based RT-PCR with 88 miRNAs was used to validate our Illumina array expression findings. We were able to map 19 miRNAs between the two platforms. Of these, 14/19 demonstrated concordance at the level of the direction of regulation (increased or decreased) at a hypergeometric probability of p < 0.014. Nine representative miRNAs were selected for groups IIa vs. I and IIc vs. IIa as indicated in Figure 4. The trends for up-regulation and down- regulation of these miRNAs were consistent between the two array measurements.

MiRNA and target genes

To explore the biological relationship between differentially expressed miRNAs and their regulated genes, we used differentially regulated (p < 0.05) miRNAs on day 3–5 after oocyte retrieval against an established miRNA database for predicted target genes (Sanger miRBase, v9.1, February 2007 release). Interestingly, there are large numbers of predicted target genes for a given miRNA per miRBase. We were able to identify nineteen miRNAs and their selected target genes in this defined study categories which are shown in Table 2.
Table 2
Selected miRNAs and gene targets (comparisons are at day3-5 after oocyte retrieval)
miRNA
P vs. no
P + E vs. no
P + E vs. P
Predicted target genes
hsa-miR-335
↑↑
  
IMP2,CD79B,WWP1,AP3S1,HOXD8,MAX,SP1,MAP2,MAK3,STARD7,CAP350,PANK2,SRPR,PPP6C,LASS5,ATP1B1
hsa-miR-346
 
IMP1,EIF3S1,BCL6,ABCC12,LIF,FSTL4,KGFLP2,KRAS,RAF7,FGF7,TMEM28,IGSF4B,PPP1R9B,COL2A1,HCG3, CALN1,HBP1,SF1
hsa-miR-448
  
DOCK9,PPM2C,NTF3,CAP1,MAP2K6,ITM1,PRKAR2B,PAPPA,CDC2L6,CNTN4,IGF1R,SOCS5,CLK1,HOXA11,WWP1,FOXJ3,WDR22,MPPED2,ADD1,PRKA2
hsa-miR-504
  
DCX,ATP1B4,IL1RAPL1,MNT,KLF13,PRKAR2A,IL16,LIF,FXR2,NRF1,CAMK2G,MMD,LOC284296,DND1,CNTFR, SORT1,NFIX
hsa-miR-512-5p
↑↑
 
PIK3R1,CTNNB1,EMX2,SOX21,RIPK5,MBD6,SRPK, VNN3,ERP29,PHF15,FBXW11,LOC285074,MAP1A,CHD9
hsa-miR-520 g
↑↑
  
ETF1,CAMK2N1,NLK,TNFSF11,CNR1,EFTUD1,HMGB3,FBN2,ENC1,MARK4,TFEB,TNFSF12,PRKAR2A,TNKS1BP1,EIF4E,PPP3CA,IMP1,MAP3K14,TMTC2,TTN,GTF2IRD2,PTK2B,DNAJB5,TNRC6A,VEGF,EIF4G2,FOXO1A,MAP3K9
hsa-miR-204
  
RUNX2,SOX4,NRBF2,MAP1LC3B,CDC42,ATP2B1,AKAP1,MAP3K3,CENTD1,IGF2R,NTRK2,TGFBR2,AP3M1,
    
NEUROG1,P53CSV,TCF7L1,CDH2, CDC25B,TCF12,ELF2
hsa-miR-369-3p
  
TCF8,PKIA,TLN1,CHD7,NCK2,CD2AP,CDC2L6,ELMOD1,CCNE2,FOXG1B,HOXB3,ADAMTS19,GIT2,ADAMTS3,TCF12, SRPK2,ADAMTS6,MAP2,ADAM10,FOXO1A,VEGFC
hsa-miR-328
  
AK6,ITGA5,PRX,IGSF4C,MAX,SOX11,PTPN9,DPH2, HIST1H4D,USP37,VSIG4,RPP14,SF4,ULK2,FGD1,PLAG1
hsa-miR-186
  
APLP2,ITGA6,RPS6KB1,CDC42,PRDM10,IGSF11,EFCBP1,TCF20,CAST,LMBR1,TMED2,TGFBR2,ICMT,IL2,CCNT2,HOXB8,PAK7,FOXD1,PTGES3,MAP3K2,VEGF,COL3A1, SRPK2,MAPKAP1,C16orf52,MAP2
hsa-miR-517a
  
AMMECR1,ACACA,NPAS4,BSN,HNRPU,PTK2B,CDKN2A,CBLN2,RAPGEFL1,LOC201895,FOXJ3,PHF13,TMCC1
hsa-miR-365
  
↓↓
EIF4E3,MAP2K7,LAMP2,ENTPD7,PCNP,ADAMTS6, COL7A1,PPP5C,REV3L,PTGDR,KCNH2,RBM12,PKD2L2
hsa-miR-221
↓↓
  
CDC2L6,TIMP3,EIF4E3,NTF3,IMP2,HTLF,CDV3,NL,EIF5A2,NRK,PAK1,CDKN1C,FAT2,LIFR,TMCC1,MAP3K10,VGLL4,FAM13A1,TCF12,HOXC10,MAPK10,HMGCLL1,ADAM11,CD4,CTCF
hsa-miR-495
↓↓
  
MAPK6,CDK6,EML4,ILF3,PTK9,PRR7,HBEGF,HOMER1,MARK3,SP4,TGFB2,LHX2,HOXC6,PRKX,AP3M1,SOX9,GMFB,HMGCLL1,FOXO3A,EDG3,NKRF,HOXB9,TIMP2, IGSF4,CD164,TNFRSF21
hsa-miR-146a
 
FBXL10,IRAK1,TRAF6,CD79B,SP8,FLJ33814,SFRS6,NPAS4,CXorf22,EIF4G2,MMP16,USP3,KCTD15,SMAD4,LOC440944,SEC23IP,BCORL1,TM6SF2,DLGA1
hsa-miR-99a
 
EPDR1,FZD8,HS3ST2,EIF2C2,HS3ST3B1,FGFR3,BAZ2A,MBNL1,CYP26B1,KBTBD8,SMARCA5,FRAP1,ZZEF1,ICMT,C4orf16,ADCY1,MTMR3,CTDSPL,HOXA,RAVER2, INSM1,TRIB2,SLC44A1
hsa-miR-181c,d
↑↑
  
ETF1,COL16A1,NLK,TNFSF11,MAP3K3,MAP2K1,ITGA3,TCERG1,MAPT,MAPK1,MAP1B,CDH13,ITGB8,PCGF2,ADAMTS18,LMBRD2,MMP14,CD163,LIF,ADAMTS6, TNFRSF11B,CDC42BPA
hsa-miR-200b
  
TCF8,NTF3,CYLN2,HMGB3,PRKAR2B,MPP5,GIT2,MAP4K3,FLJ21103,E2F3,CSNK1G3,MMD2,ZNF53, EIF5B,ERRFI1
hsa-miR-196b
 
 
IMP1,CDYL,COL14A1,SSR1,IMP3,CDV3,CALM3,COL24A1,CDKN1B,ELF4,HOXC8,HMGA2,HOXA5,MAP4K3,PARP6,COL3A1,HOXA1,TNFSF12,COL1A2, HOXA7,HOXB6
or = up or down regulated, p < 0.05; ↑↑ or ↓↓= up or down regulated, p < 0.01.
See website http://​www.​mirbase.​org (http://​microrna.​sanger.​ac.​uk) regarding additional predicted target genes.
In order to further investigate the possible biological implications for those miRNAs which were cross validated by both QRT-PCR and Illumina array data (Figure 4), the relationship of these microRNAs and their known gene targets was evaluated using the IPA miRNA Target Filter software. This group of miRNAs is regulated between day 3–5 and day 0 and also at day 3–5 between P + E (Grps IIc) and no support (Grps IIa) groups. IPA was able to identify 7 out of the 9 miRNAs from Figure 4 (excluding hsa-miR-144, and hsa-mir-181b). The gene targets were identified for these miRNAs based upon the selection of the most stringent criteria requiring experimental observation of a given miRNA and its target. Gene targets were further filtered for known involvement in endocrine system disorders. The results of this analysis (Table 3) that shows pathway enrichments were calculated for the entire gene set. The findings of the analysis demonstrated a significant involvement of genes of extracellular matrix, cell proliferation, and response to steroid hormone stimulus between days 3–5 versus day 0 at no steroid support groups (Table 3, Grps IIa-I). Interestingly, this effect was almost completely abrogated by progesterone and estrogen treatment (Table 3, GrpsIIc-IIa) for genes of cellular proliferation and response to steroid hormones but not for extracellular matrix.
Table 3
Cross validated miRNAs and their selected target genes
Symbol
GrpsIIa-I [ILL-FC]
GrpsIIc-IIa [ILL-FC]
Source
Symbol
Entrez gene name
Pathway (enrichment)
P Value
FDR
miR-223-3p (GUCAGUU)
1.389
1.111
1
VIM
vimentin
   
miR-223-3p (GUCAGUU)
1.389
1.111
1,2
RHOB
ras homolog family member B
   
miR-223-3p (GUCAGUU)
1.389
1.111
1
IRS1
insulin receptor substrate 1
   
miR-29b-3p (AGCACCA)
1.375
1.068
2,3
TUBB2A
tubulin, beta 2A class IIa
   
miR-29b-3p (AGCACCA)
1.375
1.068
1,2,3,4
SPARC
secreted protein, acidic, cysteine-rich (osteonectin)
extracellular matrix
1.29E-07
4.44E-06
miR-29b-3p (AGCACCA)
1.375
1.068
1,2,3
PIK3R1
phosphoinositide-3-kinase, regulatory subunit 1 (alpha)
   
miR-29b-3p (AGCACCA)
1.375
1.068
2,3
MYBL2
v-myb myeloblastosis viral oncogene homolog (avian)-like 2
   
miR-29b-3p (AGCACCA)
1.375
1.068
1,2
COL5A3
collagen, type V, alpha 3
extracellular matrix
1.29E-07
4.44E-06
miR-29b-3p (AGCACCA)
1.375
1.068
32,3
COL5A2
collagen, type V, alpha 2
extracellular matrix
1.29E-07
4.44E-06
miR-29b-3p (AGCACCA)
1.375
1.068
1,2,4
COL4A1
collagen, type IV, alpha 1
extracellular matrix
1.29E-07
4.44E-06
miR-29b-3p (AGCACCA)
1.375
1.068
1,2,3,4
COL1A2
collagen, type I, alpha 2
extracellular matrix
1.29E-07
4.44E-06
miR-29b-3p (AGCACCA)
1.375
1.068
1,2,4
COL15A1
collagen, type XV, alpha 1
extracellular matrix
1.29E-07
4.44E-06
miR-9-5p (CUUUGGU)
2.104
−1.802
1
NFKB1
nuclear factor of kappa light polypeptide gene enhancer in B-cells 1
   
miR-9-5p (CUUUGGU)
2.104
−1.802
1,2
FOXO1
forkhead box O1
   
miR-9-5p (CUUUGGU)
2.104
−1.802
1,2
FOXG1
forkhead box G1
positive regulation of cell proliferation
1.21E-08
1.67E-06
miR-9-5p (CUUUGGU)
2.104
−1.802
1,2,3
CDH1
cadherin 1, type 1, E-cadherin (epithelial)
   
miR-181a-5p (ACAUUCA)
1.376
−1.24
1,4
TRA@
T cell receptor alpha locus
   
miR-181a-5p (ACAUUCA)
1.376
−1.24
1,2
TIMP3
TIMP metallopeptidase inhibitor 3
   
miR-181a-5p (ACAUUCA)
1.376
−1.24
1,2
NOTCH4
notch 4
positive regulation of cell proliferation
1.21E-08
1.67E-06
miR-181a-5p (ACAUUCA)
1.376
−1.24
1,2
KRAS
v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog
response to steroid hormone stimulus
6.24E-07
4.65E-05
miR-181a-5p (ACAUUCA)
1.376
−1.24
1,2,4
HOXA11
homeobox A11
   
miR-181a-5p (ACAUUCA)
1.376
−1.24
1,2
GATA6
GATA binding protein 6
   
miR-181a-5p (ACAUUCA)
1.376
−1.24
1,2
ESR1
estrogen receptor 1
response to steroid hormone stimulus
6.24E-07
4.65E-05
miR-181a-5p (ACAUUCA)
1.376
−1.24
1,2
CDKN1B
cyclin-dependent kinase inhibitor 1B (p27, Kip1)
positive regulation of cell proliferation
1.21E-08
1.67E-06
miR-181a-5p (ACAUUCA)
1.376
−1.24
1,2,3,4
BCL2
B-cell CLL/lymphoma 2
response to steroid hormone stimulus
6.24E-07
4.65E-05
miR-196a-5p (AGGUAGU)
1.092
−1.342
1
IKBKB
inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta
   
miR-196a-5p (AGGUAGU)
1.092
−1.342
1,2,4
HOXC8
homeobox C8
   
miR-196a-5p (AGGUAGU)
1.092
−1.342
1,3
ANXA1
annexin A1
   
miR-99a-5p (ACCCGUA)
1.427
−1.48
1,2,3
MTOR
mechanistic target of rapamycin (serine/threonine kinase)
positive regulation of cell proliferation
1.21E-08
1.67E-06
miR-99a-5p (ACCCGUA)
1.427
−1.48
1,2,3
IGF1R
insulin-like growth factor 1 receptor
positive regulation of cell proliferation
1.21E-08
1.67E-06
miR-99a-5p (ACCCGUA)
1.427
−1.48
1,2,3
FGFR3
fibroblast growth factor receptor 3
positive regulation of cell proliferation
1.21E-08
1.67E-06
miR-128 (CACAGUG)
1.184
−1.345
1,2
TXNIP
thioredoxin interacting protein
response to steroid hormone stimulus
6.24E-07
4.65E-05
miR-128 (CACAGUG)
1.184
−1.345
2,3
TGFBR1
transforming growth factor, beta receptor 1
response to steroid hormone stimulus
6.24E-07
4.65E-05
miR-128 (CACAGUG)
1.184
−1.345
1,2
LDLR
low density lipoprotein receptor
response to steroid hormone stimulus
6.24E-07
4.65E-05
miR-128 (CACAGUG)
1.184
−1.345
1,2
E2F3
E2F transcription factor 3
positive regulation of cell proliferation
1.21E-08
1.67E-06
miR-128 (CACAGUG)
1.184
−1.345
1,2
ADORA2B
adenosine A2b receptor
   
Ingenuity Pathway Analysis (Ingenuity® Systems, http://​www.​ingenuity.​com). MiRNA Target Filter was applied using the strictest criteria (experimentally observed microRNA/gene targets only) filtered for genes previously identified for involvement in endocrine system disorders. Each of the seven miRNAs has multiple gene targets. Fold changes between groups as determined by Illumina miRNA array measurements are shown. Ingenuity target identifications are generated from multiple databases (Source). Pathway enrichments were calculated for the entire gene set using the NIAID DAVID functional enrichment tool [31, 32]. Genes that feature in both the cellular proliferation and the steroid hormone pathways are in bold (KRAS, BCL2, TGFBR1).
[ILL-FC] = Illumina array-fold change; Source = 1.miRecords, 2.TargetScan Human, 3.Ingenuity Expert Findings, 4.TarBase; FDR = False Discover Rate.

Discussion

In the past few years, the field of miRNA research has evolved rapidly. Various studies have provided strong evidence for the widespread expression and the regulatory functions of miRNAs on gene expression under either physiologic or pathologic conditions. MicroRNAs have now been recognized as key players in the process of cell proliferation and differentiation. Global analysis of miRNAs in human tissues have showed that, in addition to the brain, the uterus, the cervix, and the ovaries have the highest restricted enrichment in individual miRNAs [34]. The identification of miRNA as well as the functional analysis of individual expressed miRNA in the uterus has shed light onto the cycling changes that occur in response to steroids and during pregnancy. The impact of the ovarian steroids on miRNA expression and regulation in the uterus has been evidenced by the fact that treatment with 17β estradiol or RU-486 resulted in differential regulation of miRNAs in the myometrium and leiomyomas [35].
In the present study, we have examined 526 different miRNAs in the human endometrium following COS and identified a rich number of miRNAs with at least 2 fold changes in the level of expression during the luteal phase (Figure 2, Additional file 1: Table S1). Statistical analysis identified that the changes were significant (p < 0.05) for 216 of miRNAs ( Additional file 1: Table S1). These changes were observed not only in the within the group analysis at different times during luteal phase (comparison between day 0 and day 3–5) but also in the analysis between groups at the same time frame (comparison between the treatment groups at day 3–5). As demonstrated in Figure 1and Figure 2, there was a substantial increase in miRNA expression in the groups treated with progesterone alone as compared to the no supplementation group. In genome-wide identification of endometrial miRNA in natural and stimulated cycles reported by Sha et al.[36], 22 miRNAs were significantly dysregulated on the day of hCG + 7 in stimulated cycles as compared with day of LH + 7 in natural cycles. Among those, 11 miRNAs exhibited putative estrogen response elements or progesterone response elements in the promoters. In a study of examining gene expression profile in natural cycle and stimulated cycles during luteal phase (LH + 2 or 7; hCG + 2 or 7), Haouzi et al.[37] demonstrated that COS regimens altered endometrial receptivity in comparison with natural cycle. These and our studies indicate that ovarian stimulation or altered steroid hormone levels may affect miRNA profiles, consequently, affect endometrial receptivity. Furthermore, we found that the addition of estradiol in the regimen resulted in a significant attenuation of effect of progestone (Figure 1, Figure 2) on the level of miRNA expression. These findings support the notion that the well known anti-proliferative effect of progesterone on the endometrium could be possibly exerted by a localized increase in miRNA expression. The addition of estradiol at the same time could reverse this effect partially by attenuating this increase. Whether this effect is directly or indirectly associated with ovarian stimulation or the type of drug delivery for luteal support (estradiol was administered orally whereas progesterone was administered vaginally in this study) requires further investigation.
By microarray, Northen blot and in situ hybridization, Hu et al. [38] was able to identify eight specific miRNAs that were significantly up-regulated at implantation sites. Chakrabarty et al. have showed in the mouse uterus, that two specific miRNAs, the mmu-miR-101a and the mmu-miR-199a*, were differentially expressed during implantation in coordination with the expression of cyclooxygenase-2(Cox-2), a gene critical for implantation [39]. Studies on temporal and spatial regulation of miRNAs in the rat uterus, during embryo implantation, have identified the let-7a and mir-320 specifically in the uterine endometrium with higher expression level on gestation day 6–7 [26, 27]. These evidences and our findings of differential expression of miRNAs in the peri-implantation period with and without luteal phase support suggest role(s) of miRNAs during the remodeling process of endometrium in association with implantation.
Neo-angiogenesis is a pivotal process in reproductive function where it regulates endometrial regeneration, corpus luteum formation and finally placentation. The regulatory function of miRNAs in the process of neo-angiogenesis has been illustrated in several in vitro and in vivo models [9]. For example, the role of miRNAs in the neo-angiogenesis has been reported in experiments with Dicerex ½ mouse embryos (altered function of Dicer required for miRNA processing) which suffer from defective angiogenesis, due to disruption in the expression of vascular endothelial growth factor (VEGF) as well as to its receptor flt-1 [40]. We have noticed in our study that several miRNAs including miR-520 g, miR-369-3p, and miR-186 (Table 2), with VEGF as predicted target gene, were differentially regulated during the peri-implantation period. More specifically there was a significant increase in the expression of miR-520 g in the group that received only progesterone as compared to the other groups. In contrast, in the same group, there was a pronounced suppression of miR-221, which is known to regulate endothelial nitric oxide synthase, one of the key regulators of endothelial biology and angiogenesis [41]. Whereas our findings support the regulatory effect of miRNAs in the process of neo-angiogenesis, the precise impact of this action remains obscure. Individual targets of specific miRNAs responsible for the phenotypes have been proposed in experimental settings, although it is likely that many miRNAs function through cooperative regulation of multiple mRNAs [7]. Indeed, Revel et al. evaluated the expression of miRNAs in the secretory endometrium of repeated implantation failure patients and identified 13 miRNAs were differentially expressed (10 were overexpressed and 3 were underexpressed), which putatively regulated the expression of 3800 genes.
In addition, in this study, based on the most stringent criteria requiring experimental observations, IPA miRNA Target analysis for cross validated microRNAs identified 7 out of 9 miRNAs and their gene targets which were further subjected for pathway analysis. The results revealed significant involvement of genes of extracellular matrix, cell proliferation, and response to steroid hormone stimulus from day 0 to day 3–5 after oocyte retrieval in a group with no steroid support (Table 3). Conversely, this effect was almost completely abolished by supplementation of progesterone and estrogen (Table 3, GrpsIIc-IIa) for genes of cellular proliferation and response to steroid hormones bur not for genes of extracellular matrix.
Under the influence of the ovarian steroids the human endometrium undergoes cyclic changes. Estradiol promotes epithelial cell proliferation, while progesterone inhibits this estrogen-induced effect, promotes differentiation, and has decidualizing effects on endometrial stroma later in the secretary phase. We hypothesize that ovarian steroids may regulate multiple genes related to the uterine tissue remodeling and endometrial receptivity, at least in part, through modulating miRNA expression profiles.
We realize that there are several limitations in this study. The relatively small sample size due to limited number of donors that have agreed to participate could represent one of those. Unfortunately due to the design of our experiment it was extremely difficult to obtain more specimens. Furthermore, due to the fact that the same women were biopsied twice during the same COS cycle the first biopsy may induce gene expression differences that are likely to be reflected in the miRNA expression profile of the second biopsy. Additional group(s) with only one biopsy for each subject for a given group and given day of biopsy would provide another layer of control to strengthen the findings in this study. On the other hand, the limited sample size also reflects the difficulty in obtaining these samples. In addition, although group II contains 3 samples in each sub-group, there are 2 samples from day 3 and 1 sample from day 5 which may potentially affect miRNA profiles. However, after normalization and careful comparison, samples from day 3 and day 5 showed similar expression level on miRNAs profile in the same treatment group. Since day 3–5 are all in mid-secretory period of the cycle, we combined day 3 and day5 samples as one stage of the luteal phase for analysis.
Despite these limitations nevertheless, our array-based global miRNA profiling describes, for the first time, the miRNA expression profile in the human endometrium during the luteal phase following COS for IVF and luteal support with steroid supplementation. We have shown that this profile is under considerable influence by ovarian steroids, even though the molecular mechanism of this interaction still remains unclear. Importantly, several miRNAs found to have enriched or depleted transcript load during the luteal phase may have specific roles in the control of endometrial receptivity. Further studies are necessary to create a detailed expression profile for these miRNAs in relation to their target genes in the endometrium throughout the natural cycle as well as the stimulated cycle for IVF. We plan to further investigate several significantly regulated miRNAs and associated target gene pathways in relation to endometrial receptivity and implantation. Functional study will also be designed to link the imperative miRNAs in potential clinical applications.

Conclusions

The array-based study presented here has revealed several findings: 1) there is an expression of a unique set of miRNAs in the endometium following controlled ovarian stimulation; 2) the level of expression for these miRNAs undergoes significant changes during the peri-implantation period; 3) the expression is influenced by ovarian steroids; 4) expression of miRNAs may be associated with target genes and gene pathways. The miRNAs found to have enriched or depleted transcript load during the luteal phase may have specific roles in the control of endometrial receptivity during the peri- implantation period through regulation of their target genes. Further studies are necessary to create a detailed expression profile for these miRNAs as well as their associated target genes throughout the natural cycle and the stimulated cycle for IVF in the endometrium. Studies for specifically regulated miRNAs and their target genes as well specific gene pathways in relation to endometrial receptivity and implantation are also proposed.

Acknowledgement

This study was supported by Schering - Plough Research Institute grant #90048620.
We thank Johns Hopkins REI fellows for endometrial biopsies and Ms Tonya Watkins of Department of Medicine for technical assistance in microarray analysis.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

YZ, NV designed the protocol, developed the study, managed specimens, and drafted the manuscript. HZ provided valuable suggestions for the study and actively involved in manuscript preparation. NN participated in data analysis and manuscript preparation. CC, JF carried out microarray analysis, performed the statistical analysis, and contributed in manuscript preparation. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Bueno MJ, de Castro Pérez I, Malumbres M: Control of cell proliferation pathways by microRNAs. Cell Cycle. 2008, 20: 3143-3148.CrossRef Bueno MJ, de Castro Pérez I, Malumbres M: Control of cell proliferation pathways by microRNAs. Cell Cycle. 2008, 20: 3143-3148.CrossRef
2.
Zurück zum Zitat Engels BM, Hutvagner G: Principles and effects of microRNA-mediated post-transcriptional gene regulation. Oncogene. 2006, 25: 6163-6169. 10.1038/sj.onc.1209909.CrossRefPubMed Engels BM, Hutvagner G: Principles and effects of microRNA-mediated post-transcriptional gene regulation. Oncogene. 2006, 25: 6163-6169. 10.1038/sj.onc.1209909.CrossRefPubMed
3.
Zurück zum Zitat Jovanovic M, Hengartner MO: miRNAs and apoptosis: RNAs to die for. Oncogene. 2006, 25: 6176-6187. 10.1038/sj.onc.1209912.CrossRefPubMed Jovanovic M, Hengartner MO: miRNAs and apoptosis: RNAs to die for. Oncogene. 2006, 25: 6176-6187. 10.1038/sj.onc.1209912.CrossRefPubMed
5.
Zurück zum Zitat Ambros V, Chen X: The regulation of genes and genomes by small RNAs. Development. 2007, 134: 1635-1641. 10.1242/dev.002006.CrossRefPubMed Ambros V, Chen X: The regulation of genes and genomes by small RNAs. Development. 2007, 134: 1635-1641. 10.1242/dev.002006.CrossRefPubMed
6.
Zurück zum Zitat Bartel DP: MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004, 116: 281-297. 10.1016/S0092-8674(04)00045-5.CrossRefPubMed Bartel DP: MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004, 116: 281-297. 10.1016/S0092-8674(04)00045-5.CrossRefPubMed
7.
Zurück zum Zitat Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, Castle J, Bartel DP, Linsley PS, Johnson JM: Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature. 2005, 433: 769-773. 10.1038/nature03315.CrossRefPubMed Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, Castle J, Bartel DP, Linsley PS, Johnson JM: Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature. 2005, 433: 769-773. 10.1038/nature03315.CrossRefPubMed
8.
Zurück zum Zitat Revel A, Achache H, Stevens J, Smith Y, Reich R: MicroRNAs are associated with human embryo implantation defects. Hum Reprod. 2011, 26: 2830-2840. 10.1093/humrep/der255.CrossRefPubMed Revel A, Achache H, Stevens J, Smith Y, Reich R: MicroRNAs are associated with human embryo implantation defects. Hum Reprod. 2011, 26: 2830-2840. 10.1093/humrep/der255.CrossRefPubMed
9.
Zurück zum Zitat Pan Q, Luo X, Toloubeydokhti T, Chegini N: The expression profile of micro-RNA in endometrium and endometriosis and the influence of ovarian steroids on their expression. Mol Hum Reprod. 2007, 13 (11): 797-806. 10.1093/molehr/gam063.CrossRefPubMed Pan Q, Luo X, Toloubeydokhti T, Chegini N: The expression profile of micro-RNA in endometrium and endometriosis and the influence of ovarian steroids on their expression. Mol Hum Reprod. 2007, 13 (11): 797-806. 10.1093/molehr/gam063.CrossRefPubMed
10.
Zurück zum Zitat Pan Q, Chegini N: MicroRNA signature and regulatory functions in the endometrium during normal and disease states. Semin Reprod Med. 2008, 26: 479-493. 10.1055/s-0028-1096128.PubMedCentralCrossRefPubMed Pan Q, Chegini N: MicroRNA signature and regulatory functions in the endometrium during normal and disease states. Semin Reprod Med. 2008, 26: 479-493. 10.1055/s-0028-1096128.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Ohlsson Teague EM, Van der Hoek KH, Van der Hoek MB, Perry N, Wagaarachchi P, Robertson SA, Print CG, Hull LM: MicroRNA-regulated pathways associated with endometriosis. Mol Endocrinol. 2009, 23: 265-275.CrossRefPubMed Ohlsson Teague EM, Van der Hoek KH, Van der Hoek MB, Perry N, Wagaarachchi P, Robertson SA, Print CG, Hull LM: MicroRNA-regulated pathways associated with endometriosis. Mol Endocrinol. 2009, 23: 265-275.CrossRefPubMed
12.
Zurück zum Zitat Hawkins SM, Creighton CJ, Han DY, Zariff A, Anderson ML, Gunaratne PH, Matzuk MM: Functional MicroRNA involved in endometriosis. Mol Endocrinol. 2011, 25: 821-832. 10.1210/me.2010-0371.PubMedCentralCrossRefPubMed Hawkins SM, Creighton CJ, Han DY, Zariff A, Anderson ML, Gunaratne PH, Matzuk MM: Functional MicroRNA involved in endometriosis. Mol Endocrinol. 2011, 25: 821-832. 10.1210/me.2010-0371.PubMedCentralCrossRefPubMed
13.
Zurück zum Zitat Ohlsson Teague EM, Print CG, Hull ML: The role of microRNAs in endometriosis and associated reproductive conditions. Hum Reprod Update. 2010, 16: 142-165. 10.1093/humupd/dmp034.CrossRef Ohlsson Teague EM, Print CG, Hull ML: The role of microRNAs in endometriosis and associated reproductive conditions. Hum Reprod Update. 2010, 16: 142-165. 10.1093/humupd/dmp034.CrossRef
14.
Zurück zum Zitat Kovalevsky G, Patrizio P: High rates of embryo wastage with use of assisted reproductive technology: a look at the trends between 1995 and 2001 in the United States. Fertil Steril. 2005, 84: 325-330. 10.1016/j.fertnstert.2005.04.020.CrossRefPubMed Kovalevsky G, Patrizio P: High rates of embryo wastage with use of assisted reproductive technology: a look at the trends between 1995 and 2001 in the United States. Fertil Steril. 2005, 84: 325-330. 10.1016/j.fertnstert.2005.04.020.CrossRefPubMed
15.
Zurück zum Zitat Tavaniotou A, Albano C, Smitz J, Devroey P: Comparison of LH concentrations in the early and mid-luteal phase in IVF cycles after treatment with HMG alone or in association with the GnRH antagonist Cetrorelix. Hum Reprod. 2001, 16: 663-667. 10.1093/humrep/16.4.663.CrossRefPubMed Tavaniotou A, Albano C, Smitz J, Devroey P: Comparison of LH concentrations in the early and mid-luteal phase in IVF cycles after treatment with HMG alone or in association with the GnRH antagonist Cetrorelix. Hum Reprod. 2001, 16: 663-667. 10.1093/humrep/16.4.663.CrossRefPubMed
16.
Zurück zum Zitat Pabuccu R, Akar ME: Luteal phase support in assisted reproductive technology. Curr Opin Obstet Gynecol. 2005, 17: 277-281. 10.1097/01.gco.0000169105.62257.e3.CrossRefPubMed Pabuccu R, Akar ME: Luteal phase support in assisted reproductive technology. Curr Opin Obstet Gynecol. 2005, 17: 277-281. 10.1097/01.gco.0000169105.62257.e3.CrossRefPubMed
17.
Zurück zum Zitat Posaci C, Smitz J, Camus M, Osmanagaoglu K, Devroey P: Progesterone for the luteal support of assisted reproductive technologies: clinical options. Hum Reprod Suppl. 2000, 1: 129-148.CrossRef Posaci C, Smitz J, Camus M, Osmanagaoglu K, Devroey P: Progesterone for the luteal support of assisted reproductive technologies: clinical options. Hum Reprod Suppl. 2000, 1: 129-148.CrossRef
18.
Zurück zum Zitat Pritts EA, Atwood AK: Luteal phase support in infertility treatment: a meta-analysis of the randomized trials. Hum Reprod. 2002, 17: 2287-2299. 10.1093/humrep/17.9.2287.CrossRefPubMed Pritts EA, Atwood AK: Luteal phase support in infertility treatment: a meta-analysis of the randomized trials. Hum Reprod. 2002, 17: 2287-2299. 10.1093/humrep/17.9.2287.CrossRefPubMed
19.
Zurück zum Zitat Kolibianakis EM, Venetis CA, Papanikolaou EG, Diedrich K, Tarlatzis BC, Griesinger G: Estrogen addition to progesterone for luteal phase support in cycles stimulated with GnRH analogues and gonadotrophins for IVF: a systematic review and meta-analysis. Hum Reprod. 2008, 23: 1346-1354. 10.1093/humrep/den115.CrossRefPubMed Kolibianakis EM, Venetis CA, Papanikolaou EG, Diedrich K, Tarlatzis BC, Griesinger G: Estrogen addition to progesterone for luteal phase support in cycles stimulated with GnRH analogues and gonadotrophins for IVF: a systematic review and meta-analysis. Hum Reprod. 2008, 23: 1346-1354. 10.1093/humrep/den115.CrossRefPubMed
20.
Zurück zum Zitat Navot D, Veeck LL, Scott RT, Lui HC, Droesch K, Rosenwaks Z: The window of embryo transfer and efficiency of human conception in vitro. Ferti Steril. 1991, 55: 114-118. Navot D, Veeck LL, Scott RT, Lui HC, Droesch K, Rosenwaks Z: The window of embryo transfer and efficiency of human conception in vitro. Ferti Steril. 1991, 55: 114-118.
21.
Zurück zum Zitat Zhao Y, Garcia J, Kolp L, Cheadle C, Rodriguez A, Vlahos NF: The impact of luteal phase support on gene expression of extracellular matrix protein and adhesion molecules in the human endometrium during the window of implantation following controlled ovarian stimulation with a GnRH antagonist protocol. Fertil Steril. 2010, 94: 2264-2271. 10.1016/j.fertnstert.2010.01.068.CrossRefPubMed Zhao Y, Garcia J, Kolp L, Cheadle C, Rodriguez A, Vlahos NF: The impact of luteal phase support on gene expression of extracellular matrix protein and adhesion molecules in the human endometrium during the window of implantation following controlled ovarian stimulation with a GnRH antagonist protocol. Fertil Steril. 2010, 94: 2264-2271. 10.1016/j.fertnstert.2010.01.068.CrossRefPubMed
22.
Zurück zum Zitat Liu Y, Lee KF, Ng EH, Yeung WS, Ho PC: Gene expression profiling of human peri-implantation endometria between natural and stimulated cycles. Fertil Steril. 2008, 90: 2152-2164. 10.1016/j.fertnstert.2007.10.020.CrossRefPubMed Liu Y, Lee KF, Ng EH, Yeung WS, Ho PC: Gene expression profiling of human peri-implantation endometria between natural and stimulated cycles. Fertil Steril. 2008, 90: 2152-2164. 10.1016/j.fertnstert.2007.10.020.CrossRefPubMed
23.
Zurück zum Zitat Reddy KV, Mangale SS: Integrin receptors: the dynamic modulators of endometrial function. Tissue Cell. 2003, 35: 260-273. 10.1016/S0040-8166(03)00039-9.CrossRefPubMed Reddy KV, Mangale SS: Integrin receptors: the dynamic modulators of endometrial function. Tissue Cell. 2003, 35: 260-273. 10.1016/S0040-8166(03)00039-9.CrossRefPubMed
24.
Zurück zum Zitat Kuokkanen S, Chen B, Ojalvo L, Benard L, Santoro N, Pollard JW: Genomic profiling of microRNAs and messenger RNAs reveals hormonal regulation in microRNA expression in human endometrium. Biol Reprod. 2010, 82: 791-801. 10.1095/biolreprod.109.081059.PubMedCentralCrossRefPubMed Kuokkanen S, Chen B, Ojalvo L, Benard L, Santoro N, Pollard JW: Genomic profiling of microRNAs and messenger RNAs reveals hormonal regulation in microRNA expression in human endometrium. Biol Reprod. 2010, 82: 791-801. 10.1095/biolreprod.109.081059.PubMedCentralCrossRefPubMed
25.
Zurück zum Zitat Qian K, Hu L, Chen H, Li H, Liu N, Li Y, Ai J, Zhu G, Tang Z, Zhang H: Hsa-miR-222 is involved in differentiation of endometrial stromal cells in vitro. Endocrinology. 2009, 150: 4734-4743. 10.1210/en.2008-1629.CrossRefPubMed Qian K, Hu L, Chen H, Li H, Liu N, Li Y, Ai J, Zhu G, Tang Z, Zhang H: Hsa-miR-222 is involved in differentiation of endometrial stromal cells in vitro. Endocrinology. 2009, 150: 4734-4743. 10.1210/en.2008-1629.CrossRefPubMed
26.
Zurück zum Zitat Xia HF, Jin XH, Song PP, Cui Y, Liu CM, Ma X: Temporal and spatial regulation of miR-320 in the uterus during embryo implantation in the Rat. Int J Mol Sci. 2010, 11: 719-730. 10.3390/ijms11020719.PubMedCentralCrossRefPubMed Xia HF, Jin XH, Song PP, Cui Y, Liu CM, Ma X: Temporal and spatial regulation of miR-320 in the uterus during embryo implantation in the Rat. Int J Mol Sci. 2010, 11: 719-730. 10.3390/ijms11020719.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Xia HF, Jin XH, Song PP, Cui Y, Liu CM, Ma X: Temporal and spatial regulation of Let-7a in the uterus during embryo implantation in the Rat. J Reprod Dev. 2010, 56: 73-78. 10.1262/jrd.09-088K.CrossRefPubMed Xia HF, Jin XH, Song PP, Cui Y, Liu CM, Ma X: Temporal and spatial regulation of Let-7a in the uterus during embryo implantation in the Rat. J Reprod Dev. 2010, 56: 73-78. 10.1262/jrd.09-088K.CrossRefPubMed
28.
Zurück zum Zitat Altmäe S, Martinez-Conejero JA, Esteban FJ, Ruiz-Alonso M, Stavreus-Evers A, Horcajadas JA, Salumets A: MicroRNAs miR-30b, miR-30d, and miR-494 Regulate Human Endometrial Receptivity. Reprod Sci. 2012, [Epub ahead of print] Altmäe S, Martinez-Conejero JA, Esteban FJ, Ruiz-Alonso M, Stavreus-Evers A, Horcajadas JA, Salumets A: MicroRNAs miR-30b, miR-30d, and miR-494 Regulate Human Endometrial Receptivity. Reprod Sci. 2012, [Epub ahead of print]
29.
Zurück zum Zitat American Society for Reproductive Medicine: Guidelines for oocyte donation. Fertil Steril. 2002, 77 (Suppl 5): S6-S8. American Society for Reproductive Medicine: Guidelines for oocyte donation. Fertil Steril. 2002, 77 (Suppl 5): S6-S8.
30.
Zurück zum Zitat Vlahos NF, Lipari CW, Bankowski B, Lai TH, King JA, Shih IM, Fragakis K, Zhao Y: Effect of luteal-phase support on endometrial L-selectin ligand expression after recombinant follicle-stimulating hormone and ganirelix acetate for in vitro fertilization. J Clin Endocrinol Metab. 2006, 91: 4043-4049. 10.1210/jc.2006-0520.CrossRefPubMed Vlahos NF, Lipari CW, Bankowski B, Lai TH, King JA, Shih IM, Fragakis K, Zhao Y: Effect of luteal-phase support on endometrial L-selectin ligand expression after recombinant follicle-stimulating hormone and ganirelix acetate for in vitro fertilization. J Clin Endocrinol Metab. 2006, 91: 4043-4049. 10.1210/jc.2006-0520.CrossRefPubMed
31.
Zurück zum Zitat Huang DW, Sherman BT, Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID Bioinformatics Resources. Nature Protoc. 2009, 4: 44-57.CrossRef Huang DW, Sherman BT, Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID Bioinformatics Resources. Nature Protoc. 2009, 4: 44-57.CrossRef
32.
Zurück zum Zitat Huang DW, Sherman BT, Lempicki RA: Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Re. 2009, 37: 1-13. 10.1093/nar/gkn923.CrossRef Huang DW, Sherman BT, Lempicki RA: Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Re. 2009, 37: 1-13. 10.1093/nar/gkn923.CrossRef
33.
Zurück zum Zitat Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)). Methods. 2001, 25: 402-408. 10.1006/meth.2001.1262.CrossRefPubMed Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)). Methods. 2001, 25: 402-408. 10.1006/meth.2001.1262.CrossRefPubMed
34.
Zurück zum Zitat Shingara J, Keiger K, Shelton J, Laosinchai-Wolf W, Powers P, Conrad R, et al: An optimized isolation and labeling platform for accurate microRNA expression profiling. RNA. 2005, 11: 1461-1470. 10.1261/rna.2610405.PubMedCentralCrossRefPubMed Shingara J, Keiger K, Shelton J, Laosinchai-Wolf W, Powers P, Conrad R, et al: An optimized isolation and labeling platform for accurate microRNA expression profiling. RNA. 2005, 11: 1461-1470. 10.1261/rna.2610405.PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Pan Q, Luo X, Chegini N: Differential expression of microRNAs in myometrium and leiomyomas and regulation by ovarian steroids. J Cell Mol Med. 2008, 12: 227-240.PubMedCentralCrossRefPubMed Pan Q, Luo X, Chegini N: Differential expression of microRNAs in myometrium and leiomyomas and regulation by ovarian steroids. J Cell Mol Med. 2008, 12: 227-240.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Sha AG, Liu JL, Jiang XM, Ren JZ, Ma CH, Lei W, Su RW, Yang ZM: Genome-wide identification of micro-ribonucleic acids associated with human endometrial receptivity in natural and stimulated cycles by deep sequencing. Fertil Steril. 2011, 96: 150-155. 10.1016/j.fertnstert.2011.04.072.CrossRefPubMed Sha AG, Liu JL, Jiang XM, Ren JZ, Ma CH, Lei W, Su RW, Yang ZM: Genome-wide identification of micro-ribonucleic acids associated with human endometrial receptivity in natural and stimulated cycles by deep sequencing. Fertil Steril. 2011, 96: 150-155. 10.1016/j.fertnstert.2011.04.072.CrossRefPubMed
37.
Zurück zum Zitat Haouzi D, Assou S, Mahmoud K, Tondeur S, Rème T, Hedon B, De Vos J, Hamamah S: Gene expression profile of human endometrial receptivity: comparison between natural and stimulated cycles for the same patients. Hum Reprod. 2009, 24: 1436-1445. 10.1093/humrep/dep039.PubMedCentralCrossRefPubMed Haouzi D, Assou S, Mahmoud K, Tondeur S, Rème T, Hedon B, De Vos J, Hamamah S: Gene expression profile of human endometrial receptivity: comparison between natural and stimulated cycles for the same patients. Hum Reprod. 2009, 24: 1436-1445. 10.1093/humrep/dep039.PubMedCentralCrossRefPubMed
38.
Zurück zum Zitat Hu S, Ren G, Liu JL, Zhao ZA, Yu YS, Su RW, Ma XH, Ni H, Lei W, Yang ZM: MicroRNA Expression and Regulation in Mouse Uterus during Embryo Implantation. J Bio Chem. 2008, 283: 23473-23484. 10.1074/jbc.M800406200.CrossRef Hu S, Ren G, Liu JL, Zhao ZA, Yu YS, Su RW, Ma XH, Ni H, Lei W, Yang ZM: MicroRNA Expression and Regulation in Mouse Uterus during Embryo Implantation. J Bio Chem. 2008, 283: 23473-23484. 10.1074/jbc.M800406200.CrossRef
39.
Zurück zum Zitat Chakrabarty A, Tranguch S, Daikoku T, Jensen K, Furneaux H, Dey S: MicroRNA Regulation of cyclooxygenase-2 during embryo implantation. PNAS. 2007, 104: 15144-15149. 10.1073/pnas.0705917104.PubMedCentralCrossRefPubMed Chakrabarty A, Tranguch S, Daikoku T, Jensen K, Furneaux H, Dey S: MicroRNA Regulation of cyclooxygenase-2 during embryo implantation. PNAS. 2007, 104: 15144-15149. 10.1073/pnas.0705917104.PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Yang WJ, Yang DD, Na S, Sandusky GE, Zhang Q, Zhao G: Dicer is required for embryonic angiogenesis during mouse development. J Biol Chem. 2005, 280: 9330-9335.CrossRefPubMed Yang WJ, Yang DD, Na S, Sandusky GE, Zhang Q, Zhao G: Dicer is required for embryonic angiogenesis during mouse development. J Biol Chem. 2005, 280: 9330-9335.CrossRefPubMed
41.
Zurück zum Zitat Suárez Y, Fernández-Hernando C, Pober JS, Sessa WC: Dicer dependent microRNAs regulate gene expression and functions in human endothelial cells. Circ Res. 2007, 100: 1164-1173. 10.1161/01.RES.0000265065.26744.17.CrossRefPubMed Suárez Y, Fernández-Hernando C, Pober JS, Sessa WC: Dicer dependent microRNAs regulate gene expression and functions in human endothelial cells. Circ Res. 2007, 100: 1164-1173. 10.1161/01.RES.0000265065.26744.17.CrossRefPubMed
Metadaten
Titel
Effect of luteal-phase support on endometrial microRNA expression following controlled ovarian stimulation
verfasst von
Yulian Zhao
Howard Zacur
Chris Cheadle
Ning Ning
Jinshui Fan
Nikos F Vlahos
Publikationsdatum
01.12.2012
Verlag
BioMed Central
Erschienen in
Reproductive Biology and Endocrinology / Ausgabe 1/2012
Elektronische ISSN: 1477-7827
DOI
https://doi.org/10.1186/1477-7827-10-72

Weitere Artikel der Ausgabe 1/2012

Reproductive Biology and Endocrinology 1/2012 Zur Ausgabe

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.