Skip to main content

Spatiotemporal expression pattern of miR-205, miR-26a-5p, miR-17-5p, let-7b-5p, and their target genes during different stages of corpus luteum in Egyptian buffaloes

Abstract

Background

No doubt that the corpus luteum (CL) plays a vital role in the regulation of female cyclicity in mammals. The scenarios among microRNAs (miRNAs) and their target genes and steroid hormones {estradiol (E2) and progesterone (P4)} are required for better understanding the molecular regulation of CL during its formation, maturation, and regression. We aimed to (I) study the changes in the relative abundance of miR-205, miR-26a-5p, miR-17-5p, and let-7b-5p and their target genes: LHCGR, CASP3, PCNA, AMH, and PLA2G3, during different stages of corpus luteum in Egyptian buffaloes, and (II) and to address different scenarios between steroid concentrations in the serum and the expression pattern of selected miRNAs and their targets.

Methods

The paired ovaries and blood samples were collected from apparently healthy 50 buffalo cows at a private abattoir. The ovaries bearing CL were macroscopically divided according to their morphological structure and color into hemorrhagic (CLH), developing (CLD), mature (CLM), regressed (CLR), and albicans (CLA). Small pieces from different stages of CL (CLH, CLD, CLM, CLR, and CLA) were cut and immediately kept at − 80 °C for total RNA isolation and qRT-PCR. The serum was separated for steroid level estimation.

Results

The LHCGR was expressed during different stages of CL, and the peak of expression was at the mid-luteal stage. The CASP3 revealed a stage-specific response at different stages of CL. The PCNA has an essential role in cellular proliferation in buffaloes CL. Both expression patterns of PLA2G3 and AMH were found over the various developmental and regression stages. It was noticed that miR-205 is conserved to target LHCGR and CASP3 transcripts. Moreover, CASP3 and AMH were targeted via miR-26a-5p. Additionally, the CASP3 and PLA2G3 were targeted via let-7b-5p. The P4 level reached its peak during CLM. There were positive and negative strong correlations between miRNAs (miR-26a-5p and miR-205), target genes (LHCGR and CASP3) during different stages of CL, and steroid hormones in the serum.

Conclusions

Taken together, the orchestrated pattern among miRNAs, target genes, and steroid hormones is essential for maintaining the proper development and function of CL in buffalo cows.

Background

Buffaloes (Bubalus bubalis) play a significant role in the agricultural economy of many developing countries, via providing milk, meat, and draught power [1]. The world population of buffaloes was estimated to be ~ 204.3 million according to FAOSTAT [2]. In developing countries, buffaloes are mostly reared under the smallholder farming system [3, 4]. In Egypt, there are around 347.6 thousand buffaloes, which provide 79.15% of milk and 86% of meat production [5]. However, buffaloes are well adaptive to the harsh environment, and their reproductive efficiency is hampered by late puberty, poor oestrous expression, long postpartum anoestrus, long calving interval, poor response to multiple ovulation and embryo transfer (MOET), seasonality, and low conception rates [6,7,8]. These obstacles are major factors of economic losses to buffalo breeders either in Egypt [3], as well as all over the world [9].

Under normal physiological regulation of ovarian tissues, the development of the mammalian corpus luteum (CL) occurs rapidly in a time-dependent manner within 1 week after ovulation, with morphologic and biochemical changes in the theca interna and granulosa cells of the preovulatory follicle [10]. In the absence of pregnancy, CL should be regressed into non-functional vascular fibrous tissue, in order to animal enters into a new estrous cycle [11]. Improper regulations of luteinization, and angiogenesis during the early luteal phase, result in poor progesterone secretion, which could compromise embryo development, and subsequently reduce female fertility. In case of the absence of pregnancy, inadequate control of the regression process of CL associates with a long female acyclicity [10, 11]. The aberrations of CL function could be a reason for anestrum and delaying animals from entering a new estrous cycle, as well as very early embryonic mortality due to insufficient progesterone secretion [10, 12]. It was reported that CL is an ephemeral gland, which has unique characters, due to its nature of rapid growth, differentiation, cellular remodeling, and death [13, 14]. The complex interactions among growth factors, immune cells, endothelial cells, and cytokines should be highly regulated in order to support the structure and function of CL at each developmental stage (formation, maintenance, and regression) [14,15,16].

The precise regulation of CL is required during its formation, maturation, and regression to support the structure and function of CL [15]. The regulatory mechanisms of CL function have been well investigated at transcriptional and post-transcriptional levels, in humans [17], mice [18], cattle [19], and ovine species [20]. However, little is known about the transcriptional as well as post-transcription regulations of CL formation, maturation, and regression in Egyptian buffaloes. In a previous study in bovine, it was shown that luteinizing hormone/choriogonadotropin receptor (LHCGR) was expressed in CL throughout the estrous cycle, and the peak of expression was at the mid-luteal stage compared to the other luteal stages [21]. Apoptosis-related cysteine peptidase (CASP3) is one of the genes that are related to programmed cell death (apoptosis) pathways and revealed stage-specific responses during structural luteolysis of CL in bovine [22]. It was demonstrated that the proliferating cell nuclear antigen (PCNA) is a marker for cellular proliferation during the S phase of the cell cycle, in bovine CL [23] and porcine CL [24]. Furthermore, anti-Mullerian hormone (AMH) shows a strong relation to CL numbers after superovulation in high-producing dairy cows [25] and could be used as a biomarker for female fertility as well as superovulation in farm animals [26]. The phospholipase A2 (PLA2G) family plays a role in CL regulation via précis regulation of uterine prostaglandins biosynthesis in bovine [27]. In addition, it was observed that the PLA2G3 transcript was highly expressed in buffalo primary cultured granulosa cells (GC) from different follicular sizes compared to their uncultured one [28]. However, the regulation of CL function could be altered at the post-transcriptional level via microRNAs [29].

MicroRNAs (miRNAs) are small, non-coding RNA molecules that act as post-transcriptional regulators of gene expression by inhibiting translation or degrading mRNA through partial or complete base pairing with the 3′-UTRs of the target mRNAs [30]. The function of miRNAs occurs at multiple hierarchical levels of gene regulatory networks, through targeting hundreds of genes [31]. It has become clear that miRNAs are instrumental players in many events such as inflammatory immune responses, cellular differentiation, apoptosis, and tissue remodeling. The fine-tuning regulation of the expression of these genes is fundamental in directing these processes for normal female fertility [32]. It was stated that multiple arrays of miRNAs are involved in CL development and controlled its function [33]. For instance, the upregulation of miR-34a during the midcycle of luteal cells resulted in the inhibition of cell proliferation and increasing progesterone production [29]. The role of miRNAs could be extended to maintaining the early pregnancy via targeting the genes responsible for CL regression leading to sustaining of progesterone secretion by luteal tissue [34].

Despite the big role of CL in the regulation of female cyclicity, little is known about the regulatory dynamics either at the transcriptional or post-transcription level, which are involved in CL formation, maturation, and regression in buffaloes. Therefore, the current work aimed to (I) investigate the changes in the relative abundance of miR-205, miR-26a-5p, miR-17-5p, and let-7b-5p and their target genes: LHCGR, CASP3, PCNA, AMH, and PLA2G3, during different stages of corpus luteum in Egyptian buffaloes, and (II) and to figure out different scenarios between steroid concentrations {estradiol (E2) and progesterone (P4)} in the serum and the expression pattern of selected miRNAs and their target genes. Thus, the current study could give us a better understanding of the molecular basis (transcriptional and/or post-transcription levels) of CL formation, maturation, and regression and subsequently could contribute to improve fertility of Egyptian buffaloes, via maintaining normal physiological function of CL.

Methods

Chemicals

All chemicals and reagents were obtained from Qiagen (Hilden, Germany), Thermo Fisher Scientific (Wilmington, USA), unless otherwise stated.

Samples collection (ovarian tissues and blood) and morphological classification of CL

The ovaries (n = 100) were collected in pairs from apparently healthy 50 non-pregnant buffalo cows of unknown reproductive history, at Bahtim private abattoir, Al-Qaliobia, Egypt {from the months of September (2018) to February (2020)}. Directly after slaughtering, blood and ovarian tissues were collected. The ovaries were placed in chilled normal saline (0.9% NaCl) supplemented with 50 μg/l gentamycin (Sigma-Aldrich) and then transported on ice to the laboratory. At the lab, the serum was separated and stored at − 20 °C for the determination of steroid hormones (E2 and P4). Meanwhile, the ovaries were washed 3–4 times in warm saline, disinfected once in 70% ethanol, and then washed again with warm saline. Afterwards, the ovaries bearing CL were macroscopically categorized according to their morphological structure and color into hemorrhagic (CLH) (bloody apex over the rupture point), developing (CLD) (visible vascularization around the periphery of CL), mature (CLM) (fully developed CL with visible vascularization around its periphery; the apex is red or brown, and the rest is grayish), regressed (CLR) (the red or brown color disappeared and no vasculature visible on its surface), and albicans (CLA) (the red or brown color disappeared and the entire CL gray/white) [35,36,37]. Furthermore, different stages of CL were confirmed via histological examination (data not shown). Small pieces from different stages of CL (CLH, CLD, CLM, CLR, and CLA) were cut and immediately kept at − 80 °C for total RNA isolation (Supplementary Fig. 1).

In silico analysis for the selected candidate miRNAs

The prediction tools of miRNA such as DIANA-microT v3.0 (http://diana.cslab.ece.ntua.gr/microT/) and miRecords (http://mirecords.biolead.org/) were used for the filtrations of miRNA hits according to their potential relevance for cellular proliferation, apoptosis, regulation of seroidogenesis, and lipid mediators at least in four different search algorithms. We found that miR-205, miR-26a-5p, miR-17-5p, and let-7b-5p could be selected as potential targets to investigated genes in the current study.

RNA isolation and cDNA synthesis

Total RNA was extracted from different stages of CL (six independent biological replicates), using miRNeasy mini kit (Qiagen, Hilden, Germany). We extracted total RNA according to the manufacturer’s protocol. Furthermore, any possible contaminations of genomic DNA were removed via on-column DNA digestion, using the RNase-free DNase kit (Qiagen, Hilden, Germany). The extracted total RNA was subsequently stored at − 80 °C. The concentration of total RNA was determined by Nano-drop 2000/c (Thermo Fisher Scientific, Wilmington, USA), as well as confirmation of the presence of intact RNA, using agarose gel electrophoresis 2%. The samples that had clear 28 and 18S ribosomal RNA bands were selected for quantification. The cDNA was synthesized from the isolated total RNA, using the RevertAid First Strand cDNA Synthesis Kit (Thermo Fisher Scientific, USA). The RNA concentration was adjusted by RNase free water to a maximum volume of 10 μl RNA, then added 1 μl of oligo (dT)18 and 1 μl of random primers to have a total volume of 12 μl and kept on ice shortly until prepared a mixture of; 4 μl 5× reaction buffer, 1 μl ribolock RNase inhibitor, 2 μl dNTP mix, and 1 μl revertAid RT. Afterwards, 8 μl of the mixture was added to the RNA samples and then mixed well by pipetting. Thermocycler was adjusted according to the following program: 25 °C for 5 min, 42 °C for 60 min, 70 °C for 5 min, and hold at 4 °C. The synthesized cDNA was confirmed using GAPDH primer in a PCR reaction and then kept at − 20 °C.

The mRNA and miRNA quantifications

The transcript level of the candidate genes (PCNA, CASP3, LHCGR, PLA2G3, and AMH) was performed in CL obtained at different developmental stages using Maxima SYBR Green/ROX qPCR Master Mix (2×) (Thermo Fisher Scientific, USA), using StratageneMx 3000P instrument (Agilent Technologies, USA). Primer3 as an online tool was used to design the specific primers of the candidate genes (http://bioinfo.ut.ee/primer3-0.4.0/) (Table 1). The master mix which contained cDNA, optimized amount of forward, and reverse primers diluted in water and SYBR green master mix were subjected to thermal cycle initiating with a denaturation step at 95 °C for 10 min, 40 cycles at 95 °C for 15 s, 60 °C for 30 s, and 72 °C for 30 s followed by melting curve analysis in order to verify the amplification specificity. The expression level of housekeeping genes (ACTB and GAPDH) was used for normalization, and their stability over various groups was checked using the NormFinder software [38].

Table 1 List of primers used

For miRNA expression analysis, 10 ng of total RNA was reverse transcribed via MultiScribe reverse transcriptase. The specific primers of candidate miRNAs (let-7b-5p, miR-26a-5p, miR-17-5p, and miR205) were separately added for each sample according to the manufacturer’s instructions. The master mix of the real-time PCR was done in a volume of 10 μl, using 0.7 μl of RT product, 0.5 μl of particular primers with probes (Table 2), and TaqMan Universal PCR Master Mix II (Thermo Fisher Scientific, Wilmington, USA). Amplification was carried out using a Stratagene Mx 3000P instrument (Agilent Technologies, USA) with initial denaturation for 10 min at 95 °C, followed by 40 cycles of 15 s at 95 °C and 60 s at 60 °C. The normalization was done using the geometric means of 5S and U6. Finally, the expression level of the mRNAs and miRNAs was expressed as an arbitrary unit calculated by 2−ΔCt = 2−(Ct target gene-Ct housekeeping gene) [39].

Table 2 List of miRNAs names, miRbase accession numbers, and their mature sequences

Determination of steroid levels (E2 and P4) in the serum

The concentrations of E2 and P4 were determined in the serum using commercial ELISA kits (Chemux BioScience, South San Francisco, USA), according to the manufacturer’s instructions. The optical density (OD) value was measured, using ELISA microplate reader (BioTek ELx800, Vermont, USA), at 450 nm wavelength for both kits. For P4 ELISA kits, sensitivity, intra-assay, and inter-assay coefficients were 0.05 ng/ml, 5.8%, and 9.0%, respectively. For E2 ELISA kits, sensitivity, intra-assay, and inter-assay coefficients were 6.0 pg/ml, 12.1%, and 11.2%, respectively.

Statistical analysis

The data were represented in means ± SEM and statistically analyzed using the Kruskal-Wallis one-way ANOVA test followed by Dunn’s multiple comparisons test by the GraphPad Prism version 9.0 software (GraphPad Software, Inc., San Diego, CA, USA). The results were considered significant at P < 0.05.

Results

The expression pattern of candidate genes in a stage-dependent manner

The mRNA transcript of the candidate genes (PCNA, CASP3, LHCGR, PLA2G3, AMH) was summarized in Fig. 1. The expression of PCNA was significantly upregulated at the CLH stage compared to other developmental and regression stages. However, the mRNA expression pattern of CASP3 was significantly upregulated at the CLD stage in comparison with the other developmental stages. The same pattern was observed at CLR and CLA, which were significantly different, but less than the CLD stage. Unfortunately, there were no significant differences observed between CLH and CLM. Regarding the LHCGR expression profile, the highest expression level was found at the CLM stage, which was significantly different compared to other stages. On the other hand, the lowest expression pattern of LHCGR was found at CLA compared to the other stages. No significant difference was identified between CLD and CLR stages. Additionally, the same expression pattern of PLA2G3 and AMH was found over the various developmental and regression stages. Their expression was significantly increased at the CLD stage with respect to other developmental stages. Nevertheless, there were no significant differences among CLH, CLM, CLR, and CLA stages in terms of PLA2G3 and AMH mRNA transcriptional levels.

Fig. 1
figure 1

Expression profile of PCNA, CASP3, LHCGR, PLA2G3, and AMH transcripts at different stages of corpus luteum in Egyptian buffaloes. Bars are presented as mean ± SEM. a,b,c,dStatistical differences between different stages of CL: hemorrhagic (CLH), developing (CLD), mature (CLM), regressed (CLR), and albicans (CLA)

Spatiotemporal of miRNA expression pattern during different stages of CL in buffalo cows

The in silico analysis of miRNA showed that the PCNA is targeted by miR-17-5p. Furthermore, miR-205 is conserved to target the LHCGR and CASP3 genes. Moreover, CASP3 and AMH are targeted via miR-26a-5p. Additionally, the CASP3 and PLA2G3 were targeted via let-7b-5p. Hence, the expression level of those miRNAs was checked, and the results were shown in Fig. 2. Unlike mRNA expression patterns, the expression pattern of miRNAs did not take the same and/or obvious pattern of mRNA over the various developmental stages of corpus luteum. The expression pattern of miR-17-5p was significantly downregulated at the CLH stage compared to CLD and CLM stages. The highest expression level was identified at the CLD stage followed by the CLM stage, which were significantly different from each other. However, the results did not show any significant differences among the CLH, CLR, and CLA stages. Furthermore, the expression of miR-26a-5p and let-7b-5p had the same pattern throughout the different developmental stages of corpus luteum in Egyptian buffalo. The expression level of those miRNAs at the CLM stage was increased in comparison with other stages followed by the CLD stage. However, their expression level was significantly decreased at CLH, CLR, and CLA compared to CLM and CLD. Meanwhile, the expression pattern among the three previously mentioned stages was not significant. Regarding the expression pattern of miR-205, the highest expression level was recorded at the CLH stage followed by CLD and subsequently CLR and CLA. The statistical analysis did not reveal any significant differences between CLR and CLA stages. However, the lowest expression level was found at the CLM stage, which was significantly different compared to other developmental stages.

Fig. 2
figure 2

Relative abundance of miR-17-5p, miR-205, let-7b-5p, and miR-26a-5p at different stages of corpus luteum in Egyptian buffaloes. Bars are presented as mean ± SEM. a,b,c,dStatistical differences between different stages of CL: hemorrhagic (CLH), developing (CLD), mature (CLM), regressed (CLR), and albicans (CLA)

Alteration of blood hormonal levels (E2 and P4) in a stage-dependent manner of corpus luteum formation, development, and regression

Following the mRNA and miRNA expression level detection, the concentrations of E2 and P4 in the blood serum were determined, and the results were shown in Fig. 3. The results indicated that the highest concentration of E2 was found at CLH and CLA stages, which were significantly different compared to other stages and insignificant in-between. However, no significant differences were observed among CLD, CLM, and CLR. On the other hand, the concentration of P4 was gradually increased from the CLH stage till the CLM stage. The peak of P4 concentration was recorded at the CLM stage and then gradually decreased till the CLA stage. The lowest level of P4 was found at the CLA stage. Generally, the differences among investigated stages were statistically different in terms of P4 concentration.

Fig. 3
figure 3

The level of steroid hormones (E2 and P4) in the serum at different stages of corpus luteum in Egyptian buffaloes. Bars are presented as mean ± SEM. a,b,c,dStatistical differences between different stages of CL: hemorrhagic (CLH), developing (CLD), mature (CLM), regressed (CLR), and albicans (CLA)

Correlation analysis

At mature CL stage (Table 3), the correlation between the arbitrary units of LHCGR mRNA, miR-205, and serum P4 concentration was positive and strong r = 0.9816, r = 0.8490, respectively (P < 0.01). On the other hand, the correlation between the arbitrary units of LHCGR mRNA, miR-205, and serum E2 concentration did not show any significant differences. At the developing CL stage (Table 3), the correlation between the arbitrary units of miR-26a-5p and serum P4 concentration was negative and strong r = − 0.8091 (P < 0.01). Furthermore, it was negative and strong correlation r = − 0.9650 (P < 0.01) between the arbitrary units of CASP3 and serum E2 level (Table 3).

Table 3 The correlation between miRNAs, target genes, and serum steroid levels (P4 and E2) during different stages of corpus luteum in buffaloes

Discussion

Currently, it is believed that the corpus luteum formation, function, and regression are controlled by a comprehensive profile of various mRNAs transcripts, which could be targeted and regulated by miRNAs. This regulation is required in order to provide a functional corpus luteum and subsequently maintain the pregnancy in mammals. Most of the previous studies focused on providing a snapshot of the miRNAs’ pattern without investigating their target genes. Hence, we aimed in our study to investigate the alteration of miRNAs (miR-205, miR-26a-5p, miR-17-5p, and let-7b-5p) and their target genes (LHCGR, CASP3, PCNA, AMH, and PLA2G3) during different stages of corpus luteum development in Egyptian buffaloes. To our knowledge, this is the first study to investigate the role of mRNA-miRNA interaction in regulating corpus luteum formation, function, and regression in Egyptian buffaloes.

In bovine, several miRNAs were differentially expressed among various developmental stages of CL, which are involved in the vital pathways [33]. Interestingly, the investigated miRNAs: miR-205, miR-26a-5p, miR-17-5p, and let-7b-5p, were a part of those miRNAs, which play essential roles in cell proliferation, as well as angiogenesis pathways. The miR-17-5p is found to be involved in cell proliferation, cell function (angiogenesis), and cell regression. In the current study, this miRNA was found to exhibit an opposite pattern with the expression of the PCNA gene, especially during the early and development stages, which is in agreement with the findings of Gecaj et al. [33]. These findings indicated that the low expression of the level of miR-17-5p during the early developmental stage is needed for promoting the proliferation of the corpus luteum. However, mirroring the findings was observed in cancer cells, which showed that miR-17-5p could promote cell proliferation through inhibiting the P21 gene, which is considered a cell cycle inhibitor [40]. On the other hand, the apoptosis process required a functionally CASP3 gene especially during CL regression [41], which is consistent with the current results. Surprisingly, the expression of CASP3 is dramatically increased at the CLD stage and then decreased with gradually increasing during CL regression, due to its functional role during the early stage of leutolysis [42]. Accordingly, the expression of its candidate miRNAs (miR-205, miR-26a-5p, and let-7b-5p) showed a reduction during the early stages of CL regression, as has been shown in the current work. Unexpectable, our results showed a downregulation of let-7b-5p and miR-26a-5p expression during the CLH, whereas the expression of miR-205 was downregulated during the CLM stage, which were unmatched with the regulation of miRNAs to the CASP3 gene [30]. These results revealed that the regulation of miRNAs to their targets could occur in a stage-dependent manner due to the dynamism of interacting the miRNAs with their targets as well as other factors such as the abundance of miRNAs [43], which needs further investigation.

Several mechanisms are well-coordinated to regulate CL growth and luteolysis. The phospholipase A2 (PLA2) family is a large family that plays a pivotal role in the regulation of an array of cell functions such as lipid metabolism [44], prostaglandins biosynthesis [27], and tumor angiogenesis [45]. As known, the endothelial cells, which represent more than 50% of CL cells count, and blood vessels are essential for CL functions [46], which is supporting our notion. The upregulation of PLA2G3 during CL development is required for its function, as has been shown in the current study. Accordingly, the expression of let-7b-5p was downregulated and then upregulated at the CLM stage. Additionally, the prostaglandin F2 alpha is involved in the luteolysis process [47]. Therefore, it is required to be synthesized during the late stage of CL development. However, our results were on the opposite side, which showed a downregulation pattern of PLA2G3 and its candidate miRNA during the late stages of CL formation. Furthermore, the same pattern was observed for the AMH gene and its candidate miRNA (miR-26a-5p). The AMH is a member of the transforming growth factor-beta (TGF-β) family. In buffalo, the AMH gene is located in chromosome 9 [26], where it is mapped to chromosome 7 in cattle [48]. It is responsible for controlling the follicle number as well as the selection of dominant follicles. Its expression reached to peak from primordial till secondary follicles and then decline once the selection of dominant follicle [26]. It reduces the sensitivity of granulosa cells to FSH in growing follicles [49]. So far, its physiologicalcal role in CL function remains elusive. Additionally, the inhibation of AMH during CLH and CLA stages could be attributed to high level of E2, which is in agreement with Grynberg et al. [50]. Therefore, our results open a new window for investigating the role of AMH during CL formation and regression, which needs confirmatory research.

To maintain CL function, LHCGR is required due to the binding of luteinizing hormone and human chorionic gonadotropin (hCG) to it. The binding of LH to LHCGR stimulates a signaling cascade leading to produce steroid hormones [51]. Accordingly, our results showed that the peak of LHCGR expression was found in fully functioned CL and then gradually decreased till regression. Meanwhile, the P4 concentration reached its peak during CLM, whereas the concentration was decreased at CLH and CLA. Supporting this notion, the freshly isolated luteinized granulosa cells showed a reduction in LHCGR and then recovered by hCG [52], which resulted in lower P4 production. Moreover, the high levels of LHCGR in the current study were offset by a reduction in miR-205-5p and vice versa, which leads us to lay the cornerstone for understanding the regulatory mechanism of LHCGR and P4 secretion.

Concerning the correlation findings, both positive and negative strong correlations between miRNAs (miR-26a-5p and miR-205), target genes (LHCGR and CASP3) during different stages of CL (developing and mature), and steroid hormones (E2 and P4) in the serum showed the precise regulation to offer a suitable milieu for CL (development and function) [13, 53]

Conclusions

The dynamics of proliferation (PCNA) and apoptosis (CASP3) genes during CL formation, development, and regression could be regulated via miR-17-5p, miR-205, miR-26a-5p, and let-7b-5p, respectively. Furthermore, the same pattern of expression either in mRNA or candidate miRNA was observed for PLA2G3 and AMH, which needs further investigation. Additionally, the LHCGR, which showed differential alteration in its expression over different stages of CL development, is regulated via miR-205 that contributed to the alteration in P4 concentration. Ultimately, harmony in mRNA-miRNA regulations is essential for functional corpus luteum, which could contribute to the understanding of the molecular mechanism behind CL development. This study opens a new window for understanding the molecular mechanism of CL development in order to map the gene network and their epigenetics regulatory mechanisms (Supplementary Fig. 2).

Availability of data and materials

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Abbreviations

CL:

Corpus luteum

miRNAs:

MicroRNAs

E2:

Estradiol

P4:

Progesterone

CLH:

Hemorrhagic CL

CLD:

Developing CL

CLM:

Mature CL

CLR:

Regressed CLR

CLA:

Albicans CL

MOET:

Multiple ovulation and embryo transfer

LHCGR:

Luteinizing hormone/choriogonadotropin receptor

CASP3:

Apoptosis-related cysteine peptidase

PCNA:

Proliferating cell nuclear antigen

AMH:

Anti-Mullerian hormone

PLA2G3:

Phospholipase A2 G3

References

  1. Minervino AHH, Zava M, Vecchio D, Borghese A (2020) Bubalus bubalis: a short story. Front Vet Sci 7:570413

    Article  Google Scholar 

  2. FAOSTAT (2019) http://faostat.fao.org.

  3. Ali A, Abdel-Razek A, Derar R, Abdel-Rheem HA, Shehata SH (2009) Forms of reproductive disorders in cattle and buffaloes in Middle Egypt. Reprod Domest Anim 44:580–586

    Article  Google Scholar 

  4. Warriach HM, McGill DM, Bush RD, Wynn PC, Chohan KR (2015) A review of recent developments in buffalo reproduction - a review. Asian-Australas J Anim Sci 28:451–455

    Article  Google Scholar 

  5. FAOSTAT (2018) http://faostat.fao.org.

  6. Perera BM (2011) Reproductive cycles of buffalo. Anim Reprod Sci 124:194–199

    Article  Google Scholar 

  7. Salzano A, De Canditiis C, Della Ragione F, Prandi A, Zullo G et al (2018) Evaluation of factors involved in the failure of ovum capture in superovulated buffaloes. Theriogenology 122:102–108

    Article  Google Scholar 

  8. Fatima LA, Evangelista MC, Silva RS, Cardoso AP, Baruselli PS et al (2013) FSH up-regulates angiogenic factors in luteal cells of buffaloes. Domest Anim Endocrinol 45:224–237

    Article  Google Scholar 

  9. Sah SK, Nakao T (2006) Characteristics of repeat breeding buffaloes in Nepal. J Reprod Dev 52:335–341

    Article  Google Scholar 

  10. Miyamoto A, Shirasuna K, Shimizu T, Bollwein H, Schams D (2010) Regulation of corpus luteum development and maintenance: specific roles of angiogenesis and action of prostaglandin F2alpha. Soc Reprod Fertil Suppl 67:289–304

    Google Scholar 

  11. Donadeu FX, Schauer SN, Sontakke SD (2012) Involvement of miRNAs in ovarian follicular and luteal development. J Endocrinol 215:323–334

    Article  Google Scholar 

  12. Esposito L, Salzano A, Russo M, de Nicola D, Prandi A, et al. (2020) Corpus luteum color doppler ultrasound and pregnancy outcome in buffalo during the transitional period. Animals (Basel) 10(7):1181. https://doi.org/10.3390/ani10071181.

  13. Stocco C, Telleria C, Gibori G (2007) The molecular control of corpus luteum formation, function, and regression. Endocr Rev 28:117–149

    Article  Google Scholar 

  14. Skarzynski DJ, Ferreira-Dias G, Okuda K (2008) Regulation of luteal function and corpus luteum regression in cows: hormonal control, immune mechanisms and intercellular communication. Reprod Domest Anim 43(Suppl 2):57–65

    Article  Google Scholar 

  15. Stouffer RL, Bishop CV, Bogan RL, Xu F, Hennebold JD (2013) Endocrine and local control of the primate corpus luteum. Reprod Biol 13:259–271

    Article  Google Scholar 

  16. Galvao AM, Ferreira-Dias G, Skarzynski DJ (2013) Cytokines and angiogenesis in the corpus luteum. Mediators Inflamm 2013:420186

    Article  Google Scholar 

  17. van den Driesche S, Myers M, Gay E, Thong KJ, Duncan WC (2008) HCG up-regulates hypoxia inducible factor-1 alpha in luteinized granulosa cells: implications for the hormonal regulation of vascular endothelial growth factor A in the human corpus luteum. Mol Hum Reprod 14:455–464

    Article  Google Scholar 

  18. Yang YL, Ren LR, Sun LF, Huang C, Xiao TX et al (2016) The role of GPR1 signaling in mice corpus luteum. J Endocrinol 230:55–65

    Article  Google Scholar 

  19. Hojo T, Al-Zi’abi MO, Skarzynski DJ, Acosta TJ, Okuda K (2009) Changes in the vasculature of bovine corpus luteum during the estrous cycle and prostaglandin F2alpha-induced luteolysis. J Reprod Dev 55:512–517

    Article  Google Scholar 

  20. Sboros V (2014) The ovine corpus luteum angiogenesis model: a tool for developing imaging technology. Annu Int Conf IEEE Eng Med Biol Soc: 4280-4282. https://doi.org/10.1109/EMBC.2014.6944570.

  21. Yoshioka S, Abe H, Sakumoto R, Okuda K (2013) Proliferation of luteal steroidogenic cells in cattle. PLoS One 8:e84186

    Article  Google Scholar 

  22. Jonczyk AW, Piotrowska-Tomala KK, Skarzynski DJ (2019) Effects of prostaglandin F2alpha (PGF2alpha) on cell-death pathways in the bovine corpus luteum (CL). BMC Vet Res 15:416

    Article  Google Scholar 

  23. Kfir S, Basavaraja R, Wigoda N, Ben-Dor S, Orr I et al (2018) Genomic profiling of bovine corpus luteum maturation. PLoS One 13:e0194456

    Article  Google Scholar 

  24. Kurowska P, Mlyczynska E, Dupont J, Rak A (2020) Novel insights on the corpus luteum function: role of vaspin on porcine luteal cell angiogenesis, proliferation and apoptosis by activation of GRP78 receptor and MAP3/1 kinase pathways. Int J Mol Sci 21(18):6823

    Article  Google Scholar 

  25. Souza AH, Carvalho PD, Rozner AE, Vieira LM, Hackbart KS et al (2015) Relationship between circulating anti-Mullerian hormone (AMH) and superovulatory response of high-producing dairy cows. J Dairy Sci 98:169–178

    Article  Google Scholar 

  26. Umer S, Zhao SJ, Sammad A, Weldegebriall Sahlu B, Yunwei P et al (2019) AMH: could it be used as a biomarker for fertility and superovulation in domestic animals? Genes (Basel) 10(12):1009

    Article  Google Scholar 

  27. Tithof PK, Roberts MP, Guan W, Elgayyar M, Godkin JD (2007) Distinct phospholipase A2 enzymes regulate prostaglandin E2 and F2 alpha production by bovine endometrial epithelial cells. Reprod Biol Endocrinol 5:16

    Article  Google Scholar 

  28. Sosa ASA, Ibrahim S, Mahmoud KGM, Ayoub MM, Abdo MSS et al (2020) Expression profiling of primary cultured buffalo granulosa cells from different follicular size in comparison with their in vivo counterpart. Zygote 28:233–240

    Article  Google Scholar 

  29. Maalouf SW, Smith CL, Pate JL (2016) Changes in microRNA expression during maturation of the bovine corpus luteum: regulation of luteal cell proliferation and function by microRNA-34a. Biol Reprod 94:71

    Article  Google Scholar 

  30. Bartel DP (2004) MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116:281–297

    Article  Google Scholar 

  31. Makeyev EV, Maniatis T (2008) Multilevel regulation of gene expression by microRNAs. Science 319:1789–1790

    Article  Google Scholar 

  32. Pan Q, Chegini N (2008) MicroRNA signature and regulatory functions in the endometrium during normal and disease states. Semin Reprod Med 26:479–493

    Article  Google Scholar 

  33. Gecaj RM, Schanzenbach CI, Kirchner B, Pfaffl MW, Riedmaier I et al (2017) The dynamics of microRNA transcriptome in bovine corpus luteum during its formation, function, and regression. Front Genet 8:213

    Article  Google Scholar 

  34. Przygrodzka E, Sokolowska G, Myszczynski K, Krawczynski K, Kaczmarek MM (2020) Clustered microRNAs: the molecular mechanism supporting the maintenance of luteal function during early pregnancy. FASEB J 34:6582–6597

    Article  Google Scholar 

  35. Ali A, Abdel-Razek AK, Abdel-Ghaffar S, Glatzel PS (2003) Ovarian follicular dynamics in buffalo cows (Bubalus bubalis). Reprod Domest Anim 38:214–218

    Article  Google Scholar 

  36. Parillo F, Catone G, Maranesi M, Gobbetti A, Gasparrini B et al (2012) Immunolocalization, gene expression, and enzymatic activity of cyclooxygenases, prostaglandin e2-9-ketoreductase, and nitric oxide synthases in Mediterranean buffalo (Bubalus bubalis) corpora lutea during diestrus. Microsc Res Tech 75:1682–1690

    Article  Google Scholar 

  37. Ireland JJ, Murphee RL, Coulson PB (1980) Accuracy of predicting stages of bovine estrous cycle by gross appearance of the corpus luteum. J Dairy Sci 63:155–160

    Article  Google Scholar 

  38. Andersen CL, Jensen JL, Orntoft TF (2004) Normalization of real-time quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res 64:5245–5250

    Article  Google Scholar 

  39. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25:402–408

    Article  Google Scholar 

  40. Chen C, Lu Z, Yang J, Hao W, Qin Y et al (2016) MiR-17-5p promotes cancer cell proliferation and tumorigenesis in nasopharyngeal carcinoma by targeting p21. Cancer Med 5:3489–3499

    Article  Google Scholar 

  41. Carambula SF, Matikainen T, Lynch MP, Flavell RA, Goncalves PB et al (2002) Caspase-3 is a pivotal mediator of apoptosis during regression of the ovarian corpus luteum. Endocrinology 143:1495–1501

    Article  Google Scholar 

  42. Peluffo MC, Bussmann L, Stouffer RL, Tesone M (2006) Expression of caspase-2, -3, -8 and -9 proteins and enzyme activity in the corpus luteum of the rat at different stages during the natural estrous cycle. Reproduction 132:465–475

    Article  Google Scholar 

  43. O’Brien J, Hayder H, Zayed Y, Peng C (2018) Overview of microRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol (Lausanne) 9:402

    Article  Google Scholar 

  44. Dennis EA (1997) The growing phospholipase A2 superfamily of signal transduction enzymes. Trends Biochem Sci 22:1–2

    Article  Google Scholar 

  45. Linkous AG, Yazlovitskaya EM, Hallahan DE (2010) Cytosolic phospholipase A2 and lysophospholipids in tumor angiogenesis. J Natl Cancer Inst 102:1398–1412

    Article  Google Scholar 

  46. Davis JS, Rueda BR, Spanel-Borowski K (2003) Microvascular endothelial cells of the corpus luteum. Reprod Biol Endocrinol 1:89

    Article  Google Scholar 

  47. Punetha M, Kumar S, Paul A, Jose B, Bharati J et al (2021) Deciphering the functional role of EGR1 in Prostaglandin F2 alpha induced luteal regression applying CRISPR in corpus luteum of buffalo. Biol Res 54:9

    Article  Google Scholar 

  48. Gao Q, Womack JE (1997) A genetic map of bovine chromosome 7 with an interspecific hybrid backcross panel. Mamm Genome 8:258–261

    Article  Google Scholar 

  49. Durlinger AL, Gruijters MJ, Kramer P, Karels B, Kumar TR et al (2001) Anti-Mullerian hormone attenuates the effects of FSH on follicle development in the mouse ovary. Endocrinology 142:4891–4899

    Article  Google Scholar 

  50. Grynberg M, Pierre A, Rey R, Leclerc A, Arouche N et al (2012) Differential regulation of ovarian anti-mullerian hormone (AMH) by estradiol through alpha- and beta-estrogen receptors. J Clin Endocrinol Metab 97:E1649–E1657

    Article  Google Scholar 

  51. Przygrodzka E, Plewes MR, Davis JS (2021) Luteinizing hormone regulation of inter-organelle communication and fate of the corpus luteum. Int J Mol Sci 22(18):9972. https://doi.org/10.3390/ijms22189972.

  52. Convissar S, Winston NJ, Fierro MA, Scoccia H, Zamah AM et al (2019) Sp1 regulates steroidogenic genes and LHCGR expression in primary human luteinized granulosa cells. J Steroid Biochem Mol Biol 190:183–192

    Article  Google Scholar 

  53. Abedel-Majed MA, Romereim SM, Davis JS, Cupp AS (2019) perturbations in lineage specification of granulosa and theca cells may alter corpus luteum formation and function. Front Endocrinol (Lausanne) 10:832

    Article  Google Scholar 

Download references

Acknowledgements

The authors are grateful to the Reproductive Genetic Lab, Department of Animal Reproduction and AI, Veterinary Research Institute, National Research Centre, for the great help and support.

Funding

This work was financially supported by a grant (ID:3/I-3-B-6116-1) from the International Foundation for Science (Sweden) and Organisation of Islamic Cooperation’s Standing Committee on Scientific and Technological Cooperation (COMSTECH).

Author information

Authors and Affiliations

Authors

Contributions

SI: conceptualization, methodology, statistics analysis, and writing and editing of the manuscript. MT: statistics analysis and writing and editing of the manuscript. AS and AE: methodology and sample collection. KM: conceptualization and review of the manuscript. HD and AEl: methodology and figure preparation. MN: final editing and review of the manuscript. The authors read and approved the final manuscript.

Corresponding author

Correspondence to Sally Ibrahim.

Ethics declarations

Ethics approval and consent to participate

The current study did not involve the handling of live animals, whereas the samples were obtained from the slaughterhouse, so the permission of the ethical committee was not required.

Consent for publication

The authors are declaring that the work described was an original manuscript that followed all the ethical regulations and that has not been published previously. Also, no conflict of interest exists in the submission of this manuscript, and the manuscript is approved by all authors to be published.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Figure S1

. Schematic diagram shown the whole experimental design. CLH: hemorrhagic corpus luteum, CLD: developing corpus luteum, CL M: mature corpus luteum, and CLA: corpus luteum albicans.

Additional file 2: Figure S2

. Schematic model summarized the scenario among miRNAs, mRNAs and serum P4 level at developing corpus luteum (CLD) as well as mature corpus luteum (CLM) stages in Egyptian buffalo cows.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ibrahim, S., Taqi, M.O., Sosa, A.S.A. et al. Spatiotemporal expression pattern of miR-205, miR-26a-5p, miR-17-5p, let-7b-5p, and their target genes during different stages of corpus luteum in Egyptian buffaloes. J Genet Eng Biotechnol 20, 37 (2022). https://doi.org/10.1186/s43141-022-00320-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43141-022-00320-9

Keywords