Skip to main content
Molecular Endocrinology logoLink to Molecular Endocrinology
. 2013 May 28;27(7):1153–1171. doi: 10.1210/me.2013-1093

Research Resource: Preovulatory LH Surge Effects on Follicular Theca and Granulosa Transcriptomes

Lane K Christenson 1,*,, Sumedha Gunewardena 1, Xiaoman Hong 1, Marion Spitschak 1, Anja Baufeld 1, Jens Vanselow 1,*,
PMCID: PMC3706842  PMID: 23716604

Abstract

The molecular mechanisms that regulate the pivotal transformation processes observed in the follicular wall following the preovulatory LH surge, are still not established, particularly for cells of the thecal layer. To elucidate thecal cell (TC) and granulosa cell (GC) type-specific biologic functions and signaling pathways, large dominant bovine follicles were collected before and 21 hours after an exogenous GnRH-induced LH surge. Antral GCs (aGCs; aspirated by follicular puncture) and membrane-associated GCs (mGCs; scraped from the follicular wall) were compared with TC expression profiles determined by mRNA microarrays. Of the approximately 11 000 total genes expressed in the periovulatory follicle, only 2% of thecal vs 25% of the granulosa genes changed in response to the LH surge. The majority of the 203 LH-regulated thecal genes were also LH regulated in GCs, leaving a total of 57 genes as LH-regulated TC-specific genes. Of the 57 thecal-specific LH-regulated genes, 74% were down-regulated including CYP17A1 and NR5A1, whereas most other genes are being identified for the first time within theca. Many of the newly identified up-regulated thecal genes (eg, PTX3, RND3, PPP4R4) were also up-regulated in granulosa. Minimal expression differences were observed between aGCs and mGCs; however, transcripts encoding extracellular proteins (NID2) and matrix modulators (ADAMTS1, SASH1) dominated these differences. We also identified large numbers of unknown LH-regulated GC genes and discuss their putative roles in ovarian function. This Research Resource provides an easy-to-access global evaluation of LH regulation in TCs and GCs that implicates numerous molecular pathways heretofore unknown within the follicle.


In cattle the periovulatory transformation of the 17β-estradiol (E2)-producing dominant follicle during the follicular phase is accompanied by a profound morphologic reorganization of somatic cell layers (ie, granulosa and theca) to produce the ephemeral progesterone (P4)-producing corpus luteum (1, 2). This conversion or luteinization process of the dominant follicle has been well studied with respect to changes in steroid hormone production and the compartmentalization of gonadotropin receptors and steroidogenic enzymes (3, 4) and forms the basis of the “two-cell-two gonadotropin hypothesis of estrogen biosynthesis” (5). Although it is understood that paracrine interactions occur between the theca and granulosa tissue layers and that they are essential for folliculogenesis and likely luteal function, much yet remains to be discovered with respect to these interactions (6). This is particularly true for the role of the LH-responsive thecal cells (TCs) of which very little is known.

Immunocytochemical and in situ hybridization studies in rat have demonstrated that granulosa cells (GCs) can have markedly different levels of LH receptor (LHCGR) expression dependent upon their position within the follicular wall. Higher LHCGR levels are found in mural GCs located close to the basement membrane vs those close to the antrum (7, 8). Indeed, oocyte-derived factors present within the follicular fluid are known to affect the mural GCs, with those residing near the antrum being under a greater influence. An even greater difference in gene expression between GCs intimately associated with the oocyte (ie, corona radiata and cumulus) and the mural granulosa has already been established (810).

Expression profiling studies performed over the last couple of years in cattle and other species have shed light on changes that occur during the early and intermediate processes of follicular differentiation such as recruitment, selection, and dominance (1114). Furthermore, studies aimed at understanding the changes in gene expression following the ovulatory LH surge have been completed in rodent, domestic animals, and primate species. In mice and rats, early events (ie, 1 h after a preovulatory LH/human chorionic gonadotropin [hCG] surge) through 12 hours after the LH surge (ovulation occurs at ∼13–15 h) have been evaluated in GCs (1517). In primates, a recent analysis of whole periovulatory follicles collected at 0, 12, 24, and 36 hours post-LH in primates was completed (18). In cows and buffalo, suppression-subtractive hybridization (19) and gene-specific microarrays (20, 21) have been used to interrogate granulosa-specific gene expression changes occurring after the preovulatory LH surge. In the paper by Ndiaye et al (19), bovine GCs were evaluated before and 23 hours after hCG administration, and these authors reported only the down-regulated genes (32 in total). Another bovine study, conducted by Gilbert et al. (20), compared GC pre-LH surge gene expression to granulosa collected 6 and 22 hours post-GnRH (using custom arrays). Lastly a recent study, focusing on genes involved in steroid biosynthesis and using RNAseq-technology, was used to comparatively survey expression profiles in cells of the follicular wall of heifers and lactating cows (22). However, to date, no study has provided a comprehensive examination of the effect of the LH surge on all layers of the bovine follicular wall, granulosa, and theca interna, particularly evaluating the differential effects of location of GCs within the follicular wall (ie, antral vs membrane associated).

The present study examines large dominant bovine follicles collected before and 21 hours after a GnRH-induced preovulatory LH surge; follicles were dissected and antral (aspirated) GC (aGC)-, membrane-associated GC (mGC)-, and TC-specific expression profiles were analyzed by mRNA microarray analysis. The bovine model is useful because ultrasonography allows accurate tracking of the developing follicle and because a mono-ovulatory species enhances its relevance to human correlates as well as its general importance to animal husbandry of this economically important species. The results of this study provide the first comprehensive expression profiling analysis of both LH receptor-positive cell types within the periovulatory follicle and cell type-specific regulatory pathways, and functions affected by the preovulatory LH surge are identified.

Materials and Methods

Animals and tissue samples

Follicles were collected from normally cycling cows (n = 6) of the Holstein-Friesian breed after slaughter and immediately processed as described elsewhere (3, 23). Briefly, for collecting large dominant follicles before the preovulatory LH surge, numbers, sizes, and relative locations of follicles (≥5 mm) of a growing cohort were monitored by ultrasonography in normally cycling cows after ovulation and at 3 days and 21 hours before slaughtering using a transrectal 5 MHz linear transducer (Aloka SSD-500, Aloka GmbH, Meerbusch, Germany). In this way, the growth of individual follicles could be tracked. These cows were slaughtered at days 7 (n = 1) and 8 (n = 2) of the estrous cycle (day 0 = day of estrus) during the first follicular wave. In these animals the largest growing follicle (as determined by ultrasound monitoring) was collected, whereas stagnating or regressing follicles were easily identified and avoided. To collect the late preovulatory follicles after the LH surge, cows at day 8 (n = 3) of the cycle (first follicular wave) were ultrasonographically examined and treated with 500 μg PGF (PGF forte, Veyx Pharma GmbH, Schwarzenborn, Germany) to induce luteolysis of the functional corpus luteum. Forty-eight hours later, all animals were scanned and injected with 100 μg of a GnRH analog (Gonavet Veyx, Depherelin, Veyx Pharma GmbH) to induce the LH surge. The animals were slaughtered 23 hours after the GnRH injection, which elicits a preovulatory LH surge about 2 hours after administration, and the largest growing follicle of each animal was dissected away from the remaining ovarian tissue (3, 23).

All procedures involving living animals (injections, ultrasonographic examinations) were according to the German law for animal protection (TierSchG) and in compliance with the European legislation on the care and use of animals.

Isolation of different follicular cell types

Two separate fractions of GCs were successively isolated from each follicle, aGC and membrane-associated granulosa (mGC) previously described as mural granulosa in Refs. (3 and 23). Briefly, the aGC fraction, together with the antral fluid, were collected by puncturing the antral follicle with a 18G needle, and the antral fluid, together with the cumulus cells and free floating or only slightly adherent cells of the GC layer, were aspirated. It should be noted that the cumulus cells and oocyte were not separated from the other aGCs aspirated from the follicles, but they likely make up a very small percentage of the actual transcripts within the aGC preparation, because it is well established that large numbers of mural GCs are removed by aspiration. The fluid was centrifuged (2 min, 400 relative centrifugal force, room temperature), the cell-free supernatant was frozen and stored at −20°C for determination of steroid concentrations, and the sedimented cells were frozen in liquid nitrogen and stored at −80°C for RNA preparation. The follicle was then cut open with scissors, and the follicular wall was peeled away from the remaining ovarian stroma and submersed in Ca2+ and Mg2+-free PBS buffer (pH 7.4). Membrane-associated GCs (mGCs) were then gently scraped off with a small scalpel blade. Detached cells and surrounding buffer were collected with a pipette, centrifuged (2 min, 400 relative centrifugal force), after which the supernatant was discarded and the sedimented cells were frozen in liquid nitrogen and stored at −80°C until RNA preparation. To isolate the theca cells (TCs), the inner (antral) side of the follicular wall was again carefully scraped with a scalpel blade and washed several times in PBS in order to further eliminate residual GCs attached to the basement membrane. The remaining basement membrane and the TCs attached to its outer side were then transferred to RNAlater solution (Qiagen, Hilden, Germany), stored at 4°C overnight, and then transferred to −20°C for long-term storage until RNA preparation.

Determination of follicular P4 and E2 content

Concentrations of P4 were determined using a competitive single-antibody [3H]RIA (24). Briefly, follicular fluid of each sample (10 μL each) was diluted with assay buffer (90 μL). Ten microliters of the diluted samples were used to measure directly, ie, without extraction, P4 and E2 concentrations. The tracer, [1,2,6,7-3H] progesterone, was purchased from GE Healthcare (Freiburg, Germany). The antibody raised in rabbits was further purified by affinity chromatography on protein A superose (GE Healthcare). The intra- and interassay coefficients of variation (CVs) were 7.4% and 9.8%, respectively. Radioactivity was measured with a β-counter with integrated RIA calculation (TriCarb2900; PerkinElmer, Rodgau, Germany).

Concentrations of E2 were determined with an ultrasensitive [125I]RIA (DSL, Sinsheim, Germany). The standard curve was established between 5 and 750 pg/mL. Radioactivity was measured with an automatic γ-counter with integrated RIA calculation (Wizard; PerkinElmer). The detection limit of the method was 3 pg/mL. The intra- and interassay CVs were 8.4% and 10.2%, respectively.

The intrafollicular ratio of E2:P4 was calculated to characterize the endocrinologic status of isolated follicles. Before the preovulatory LH surge only E2-active (E2:P4 > 1) large dominant follicles were processed for cell type-specific transcriptome analysis because it has been demonstrated by others that E2-inactive follicles (E2:P4 < 1) isolated before the LH surge had reduced capacities to specifically bind gonadotropins and display clear histologic signs of atresia (25).

RNA preparation for mRNA microarray analysis

Total RNA was isolated from granulosa and theca with Trizol (Ambion, Forest City, California), per the manufacturer's instruction. RNA quality was assessed with Agilent Bioanalyzer Nano Chips (Agilent Technologies, Santa Clara, California), and all samples had sharp 18S and 28S peaks as well as RNA integrity values over 8.0, indicating high-quality RNA. The RNA was subjected to Gene Chip Eukaryotic Target Labeling Assay (Affymetrix). Biotin-labeled cRNA was fragmented according to Affymetrix protocols. The fragmented cRNA from each sample was hybridized to individual Affymetrix bovine gene array chips using the GeneChip Fluidics Station 400 protocol (Affymetrix), and the hybridized chips were scanned using the Agilent GeneArray Scanner (Affymetrix). Before analysis, all probes that did not show significant probe level hybridization (measured by Affymetrix MAS5.0 algorithm) in any of the 18 samples were filtered out. The raw signal was background corrected, normalized, and gene-level summarized using the Robust Multichip Average (RMA) procedure (26) using default RMA settings. To allow for statistical analysis, 3 preovulatory dominant follicles isolated from 3 different cows (ie, experimental unit) and 3 periovulatory (eg, ∼21-post LH-surge) follicles isolated from 3 different cows were dissected and the aGC, mGC, and TC preparations were collected, such that a total of 18 individual bovine Affymetrix GeneChips were used. The array results have been uploaded in the GEO database (GSE46996).

cDNA synthesis and quantitative RT-PCR

Gene-specific primers for cDNA synthesis and amplification of different transcripts were designed according to published mRNA sequences (Supplemental Table 1 published on The Endocrine Society's Journals Online web site at http://mend.endojournals.org). For cDNA synthesis, 0.25 μg total RNA was reverse transcribed in a 25-μL reaction volume using M-MLV reverse transcriptase, RNase H Minus, Point Mutant (Promega, Mannheim, Germany) with gene-specific, or random and oligo-dT primers. The freshly synthesized cDNA samples were cleaned with the High Pure PCR Product Purification Kit (Roche, Mannheim, Germany) and eluted in 50 μL elution buffer. The identity of products generated with different primer pairs was determined by sequencing.

For qPCR, 0.5 and 0.25 μL of each purified cDNA sample were amplified with the LC 480 SYBR Green I Master Kit (Roche) in 12 μL total reaction volume. Measures from both reactions were averaged, considering the different amounts of cDNA. Amplification and quantification of products were performed in a LightCycler 480 instrument (Roche) under the following cycling conditions: preincubation at 95°C for 5 minutes, followed by 40 cycles denaturation at 95°C for 20 seconds, annealing at 60°C for 15 seconds, extension at 72°C for 15 seconds, and single-point fluorescence acquisition at 83°C for 10 seconds. The melting peaks of all samples were routinely determined by melting curve analysis in order to ascertain that only the expected products had been generated. Additionally, the length of all PCR products was monitored by agarose gel electrophoresis analysis (3% agarose, ethidium bromide stained). Cloned PCR products of the respective transcripts were used to generate external standard curves. Routinely, dilutions of standards covering 5 orders of magnitude (5 × 10−16 to 5 × 10−12 g DNA per reaction) were freshly diluted from stocks of 10−9 g DNA/μL and coamplified during each run. Abundance of target transcript was calculated relative to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) transcripts. In a previous study it has been shown that GAPDH transcript abundance is similar in the aGC, mGC, and TC fractions of large dominant bovine follicles before and after the LH surge (3).

Statistics and bioinformatics

Correlations between mRNA microarray data and levels of transcript abundance (relative to GAPDH transcripts), as determined by quantitative RT-PCR (qRT-PCR), were calculated by Pearson Product Moment analysis; pair-wise comparisons by t test analysis using the SigmaPlot 12.0 software (Jandel Scientific, San Rafael, California).

Gene set enrichment analysis was performed with the Ingenuity Pathway Analysis tool (Ingenuity IPA 8.0, Ingenuity Systems Inc., Redwood City, California) to map differentially expressed transcripts to specific biologic functions and canonical pathways. If the calculated P value was below .05, mapping of the respective transcript to a specific biologic function or canonical pathway was considered significant. For clarity, significant canonical pathways were combined to super ordinate categories (see Results).

Results

Morphologic and physiologic characterization of follicles before and after the LH surge

Ultrasonography of each developing follicle was completed to ensure that only the single largest growing follicles (ie, destined preovulatory follicles) were collected for subsequent analyses. Physiologic (antral E2 and P4 concentrations), morphologic (size, vascularization, color etc.), and molecular markers were used to confirm that the individual follicles collected from the 6 different cows were characteristic of either preovulatory (n = 3) and post-LH (n = 3) follicles before gene expression analysis. Before the LH surge, the mean growing follicle diameter was 13.8 ± 1.2 mm, and the ratio of antral fluid E2:P4 was 4.9 ± 1.5, which was significantly different from follicles of the post-LH group, which were 20.5 ± 0.9 mm and 0.6 ± 0.2, respectively (Supplemental Table 2). Analysis of PTGS2 transcript levels within the GCs of the LH-treated periovulatory follicles were markedly (∼280-fold) elevated in comparison to the preovulatory follicles (Table 1). Thus, the preovulatory follicles were E2 active, which is an important criterion for healthy (nonatretic) large dominant follicles (25), whereas those after LH treatment had a significantly reduced E2:P4 ratio and increased PTGS2 expression, which are both reliable biomarkers for impending ovulation (3, 23, 2729).

Table 1.

Hybridization Values, Fold Changes, and Correlation Coefficients of Microarray and qRT-PCR Comparison for LH-Responsive TC- and GC-Specific Genes

Gene Symbol Gene Name Fold Change qRT-PCRa
Microarray HSI − Mean RMA Hybridization
Correlation of qRT-PCR and HSI r Value
aGC
mGC
TC
aGC mGC TC bLH aLH bLH aLH bLH aLH
CYP17A1 Cytochrome P450; family XVII, 17α-hydroxylase −77 47 36 266 26 3452 45 1.00
FSHR FSH receptor −4.1 285 70 294 75 29 19 0.91
CYP19A1 Cytochrome P450; family 19; aromatase −398 −539 −80 6762 17 13603 25 1255 16 0.97
PTGS2 Prostaglandin-endoperoxide synthase 2 274 290 32 16 4287 13 3876 20 643 0.94
ADAMTS1 ADAM metallopeptidase with thrombospondin type 1 motif; 1 29 8.5 45 1272 184 1569 1138 2552 0.89
CCND2 Cyclin D2 −82 −27 5297 64 3993 146 796 296 0.92
CYP11A1 Cytochrome P450; family 11; subfamily A; polypeptide 1 8180 549 9733 974 7146 2852 0,83
HSD3B1 hydr.-δ-5-ster. dehydr.; 3β- and steroid δ-isom. 1 7002 5677 9315 4268 3571 2716 0.89
LHCGR LH/CG receptor −7.9 −17 −7.4 280 35 790 46 380 52 0.95
NR5A1 Nuclear receptor subfamily 5; group A; member 1 −2.8 529 352 405 228 642 229 0.73
NR5A2 Nuclear receptor subfamily 5; group A; member 2 8205 5549 9327 5559 1093 1164 0.89
PCNA Proliferating cell nuclear antigen −4.2 −2.7 2300 553 2263 841 2443 1942 0.81
PTX3 Pentraxin 3; long 644 711 108 14 9232 15 10921 85 9158 0.93
STAR Steroidogenic acute regulatory protein 57 287 823 963 9578 4471 0.96
TIMP1 TIMP metallopeptidase inhibitor 1 85 38 10 72 6151 241 9231 1439 14888 0.85

Boldface symbols are the cell-specific and LH-induced (PTGS2) validation genes we evaluated.

a

Only significant LH responses by array are depicted; bLH, before LH surge; aLH, after LH surge; correlation coefficients (r) calculated by Pearson Product Moment analysis including all 18 samples; all P values were <.001.

Microarray analysis, cell-specific markers, and qRT-PCR validation

Principal component analysis of the data revealed that the global gene expression patterns observed in aGCs, mGCs, and TCs from follicles before (n = 3) and after LH surge (n = 3) were tightly clustered within cell types and across treatment groups (before and after LH; Figure 1A). Analysis of the microarray data revealed that of the 24 016 probe sets which represent 11 548 independent annotated bovine genes and 7091 unannotated expressed sequence tags, approximately half were determined as significantly present, based on the significance of the probe level hybridization of their transcripts to the microarray in all 3 biologic replicates (calculated using Affymetrix MAS 5.0 algorithm) in all aGC, mGC, or TC samples either before and after LH, respectively (Figure 1, B and C). The majority of the probe sets, 10 770 and 11 124 were present in all 3 cell types before and after the LH surge, respectively. No striking differences in actual numbers of present genes within a cell type before and after the LH surge were found for the aGC, mGC, or TC samples (comparison of gene numbers across cell types in Figure 1, B and C). In order to estimate the degree of potential contamination of the GC preparations with thecal RNA and vice versa, the microarray hybridization signals of several well-established genes that are commonly associated with one cell type or the other (CYP19A1 and FSHR in GC; and CYP17A1 in TC) were examined (Table 1). Hybridization signal intensity (HSI: RMA normalized fluorescence intensity values surrogate to the transcript hybridization signal) for CYP19A1 in aGCs and mGCs before the LH surge were robust (6762- and 13606-HSI, respectively); however, TCs collected before LH surge also had detectable levels (1255-HSI) of CYP19A1, possibly indicating a small degree of contamination of the TC preparation with this highly expressed GC mRNA. Statistical analysis indicates that the aGC and mGC CYP19A1 levels were significantly (false discovery rate [fdr] < 0.01) greater (5.4- and 10.8-fold, respectively) than those in TCs before the LH surge. After the LH surge, the CYP19A1 HSI dropped to low levels (<25) in all cell types. Another established GC-specific transcript, FSHR, that has a lower initial HSI in aGCs and mGCs (285- and 294-HSI) before the LH surge, was 9.8- and 10.1-fold greater (fdr < 0.02) than the HSI level (<29) observed in TCs, respectively. After the LH surge FSHR expression dropped (70- and 75-HSI) as expected in aGCs and mGCs, respectively, whereas TC values remained low and nearly unchanged (29-HSI vs 19-HSI; Table 1). Examination of CYP17A1 in the 3 cellular preparations indicated that TC levels (3452-HSI) were 73.5- and 13.0-fold greater than those in aGCs (47-HSI) and mGCs (266-HSI). After the LH surge, CYP17A1 levels were low (HSI < 45) and not different across cell types.

Figure 1.

Figure 1.

mRNA Microarray Analysis of Present Transcripts within Cells of the aGCs, mGCs, and TC before and after the Preovulatory LH Surge. A, Principal component analysis shown for the 3 cell types, before (bLH) and after (aLH) the LH surge. Each symbol represents an individual replicate and the line connects those from the same cow. B and C, Venn diagrams showing members of probe sets significantly called present, based on the significance of the probe level hybridization to the microarray in all 3 biologic replicates (panel B represents before the LH surge and panel C represents after the LH surge).

Using a panel of 15 genes expressed at varying levels in GCs and TCs we compared the hybridization signals and relative qRT-PCR results for these genes (Table 1). The transcript abundance levels of these 15 selected transcripts, 7 of which were significantly up- or down-regulated by the LH surge, were correlated to qRT-PCR results for all 18 samples. The Pearson product moment correlation test indicated that all 15 transcripts showed a highly significant (P < .001) correlation between qRT-PCR and microarray hybridization values with correlation coefficients between 0.73 and 1.00 (Table 1). Because of this high correlation for genes expressed at divergent levels (low and high expression genes) combined with the tight clustering of biologic replicates observed in the principal component analysis analysis, we postulate that the microarray results are accurate and that they reflect actual relative changes before and after the LH surge for the remaining genes detected in the microarrays.

Cell-specific gene expression markers in GCs and TCs

Comparison of aGCs to mGCs before or after LH yielded few statistically significant differences. Only 1 gene, PLXNB2 (encoding plexin B2) was 2.67-fold greater (fdr < 0.05) in mGCs vs aGCs before LH, whereas 2 genes, HS6ST1 (heparan sulfate 6-O-sulfotransferase 1; 1.52-fold increase) and SSR1 (signal sequence receptor; α −1.88-fold decrease) were different (fdr < 0.05) between mGCs and aGCs after the LH surge. Even increasing the fdr to < 0.1 resulted in no further annotated genes being detected with a fold difference of ≥1.5-fold.

Comparison of TC expression to mGCs or aGCs before LH indicated that 3564 or 3650 genes, respectively, had a ≥1.5-fold (fdr < 0.05) difference in expression across these cell types; of these the majority (2696 genes) were consistently differentially regulated in both GC populations compared with TCs (Supplemental Table 3). After the LH surge 596 or 3833 genes were different (fdr < 0.05) at the ≥1.5-fold level between TCs and mGCs or aGCs, respectively; of these 541 were consistently differentially regulated in both cell populations compared with TCs (Supplemental Table 4). In summary, these data are therefore consistent with the observation that aGCs and mGCs are very similar and quite divergent from TCs.

Using a different approach to identify cell-specific genes (comparison of the significance of the transcript HSI calls, ie, absent vs present calls), we identified 34 genes as specific to the TC preparation: (ie, present in all 6 TC samples and absent [below background hybridization] in all 12 GC samples; with a significance of P < .05; Supplemental Table 5). A large number of these TC-specific transcripts are implicated in endothelial cell function (ie, PECAM1, TMEM100, LMO2, CD300LG, PEAR1). This is likely due to the unavoidable contamination of the TC with the capillary plexus surrounding the highly vascular dominant follicle. The complete lack of these endothelial markers in the avascular GC preparations of all 12 samples is reassuring. Analysis of the GCs identified 15 genes as present in either mGCs or aGCs and absent from all TC samples; 6 of the 15 genes were shown to be LH regulated in the GCs (Supplemental Table 6).

Cell-specific expression of gene transcripts influenced by the LH surge

To determine those genes that are regulated by the LH surge, we included only those probe sets that showed significant probe level hybridization (P ≤ .05, Affymetrix MAS 5.0 algorithm) and that exhibited an absolute fold change that was equal to or exceeded 1.5-fold and had a stringent Benjamini-Hochberg adjusted significance P value (ie, fdr) of 0.01 or less. This resulted in the selection of 2741 and 2417 differently expressed probe sets in aGCs and mGCs, and 226 in TC samples before and after LH, respectively (Figure 2). The relative number of gene transcripts which were down-regulated (aGCs 54%, mGCs 52%, and TCs 56%) was slightly more than numbers of gene transcripts which were up-regulated (Figure 2). Consistent with their similar tissue of origin, aGCs and mGCs shared 1783 transcripts that were similarly and significantly up- or down-regulated (Figure 2). A large number of LH-regulated probe sets (804 and 473) were differentially expressed in only aGCs or mGCs, respectively (Figure 2). However, in most cases these probe sets that were significant for either aGCs or mGCs had a calculated similar fold change (ie, within 2-fold) for the other cell type but failed to reach significance in the other cell type under the highly stringent conditions we established. Comparison of the fold changes for all 3060 probe sets that were LH regulated in GCs (see Supplemental Table 7), showed that only 29 genes exhibited more than a 3-fold difference (P < .05) in response to LH between aGCs and mGCs (Table 2).

Figure 2.

Figure 2.

Venn Diagram Showing Numbers of Significantly LH Affected Probe Set Hybridization Signals in GC and TC Preparations. Total numbers of up- (↑) or down-regulated (↓) probe sets are shown in brackets.

Table 2.

LH Regulated Genes That Are Differentially Expressed Between aGCs and mGCs

Gene Name or Chromosomal Location Gene Symbola Fold Change Between LH Response of aGCs vs mGCs P Value LH Regulated in aGCs fdr < 0.01 LH Regulated in mGCs fdr < 0.01 HSI − Mean RMA Hybridization Value Before LH
HSI − Mean RMA Hybridization Value After LH
aGC mGC aGC mGC
Nidogen 2 NID2 16.52 .002 Yes Yes 23 558 3616 5224
Similar to Collagen α-1(XIV) chain precursor LOC781493 8.91 .001 Yes No 14 138 371 417
Caveolin 1 CAV1 7.54 .001 Yes Yes 16 75 6548 6235
Fibronectin 1 FN1 5.10 .002 Yes No 89 744 1200 1978
Claudin 11 CLDN11 4.91 .011 Yes Yes 12 57 3518 3409
Dickkopf homolog 3 DKK3 4.77 .001 Yes Yes 15 92 2273 2952
Collagen, type V, α 2 COL5A2 3.82 .029 Yes No 15 256 215 956
CART prepropeptide CARTPT 3.68 .001 No Yes 45 155 46 43
Chr2:110382-336-110382643(−) (IGFBP5) 3.67 .033 Yes No 71 540 573 1181
Dihydropyrimidinase-like 3 DPYSL3 3.56 .001 Yes YES 21 81 252 277
ρ family GTPase 3 RND3 3.55 .001 Yes Yes 29 93 4046 3710
ADAM metallopeptidase with thrombospondin type 1 motif; 1 ADAMTS1 3.34 .001 Yes Yes 45 184 1272 1569
Chromosome 8 open reading frame 4 C27H8orf4 3.32 .025 Yes Yes 11 37 654 662
Cyclin D2 CCND2 −3.01 .002 Yes Yes 5297 3993 64 146
SPARC-related modular calcium binding 2 SMOC2 −3.02 .015 Yes No 942 717 192 442
Hydroxysteroid (17-β) dehydrogenase 1 HSD17B1 −3.05 .010 Yes Yes 2452 1039 49 62
Heme oxygenase 1 HMOX1 −3.07 .033 Yes No 367 218 90 164
NADH dehydrogenase (ubiquinone) 1 α subcomplex; 4-like 2 NDUFA4L2 −3.10 .005 Yes No 62 21 20 22
6-Phosphofructo-2-kinase/fructose-2;6-biphosphatase 3 PFKFB3 −3.28 .011 No Yes 283 95 504 556
Chromosome 12 open reading frame 32 C5H12orf32 −3.35 .002 Yes No 226 236 32 113
CD14 molecule CD14 −3.39 .048 No Yes 77 20 146 130
Neural precursor cell expressed; developmentally down-regulated 9 NEDD9 −3.41 .032 Yes No 130 118 21 65
KIAA0101 protein (PCNA-assoc factor) KIAA0101 −3.45 .018 Yes Yes 1782 1439 45 126
SAM and SH3 domain containing 1 SASH1 −3.51 .028 Yes No 105 65 16 35
chr2:88862297–88862784 (−) (Ankrd44) −4.04 .009 Yes No 405 163 70 114
chr11:16969669-16970066 (+) −4.26 .006 Yes Yes 701 267 27 43
Complement component 1; s subcomponent C1S −5.63 .005 No Yes 20 41 18 210
Guanylate cyclase activator 1A (retina) GUCA1A −6.07 .008 Yes No 485 97 21 25
chr1:73033634-73034079 (+) −8.03 .003 No Yes 19 200 102 1737
a

Gene symbols enclosed in parentheses are probe sets that recognize transcripts with high homology to a named gene in another species.

Chromosomal locations are based on the Cow October 2011 assembly. Dashes reflect negative or down regulated gene.

Of the 226 probe sets shown to be LH regulated in TCs, only 62 probe sets (representing 57 independent genes) were significantly regulated by LH in TC only. Most (74%) of these TC-specific LH-regulated transcripts (Table 3) were significantly down-regulated after LH-surge, with CYP17A1 encoding steroid 17α-hydroxylase (P450c17), the key enzyme of androgen biosynthesis showing the strongest response. Of the remaining 164 probe sets that were LH regulated in both TCs and GCs all but 2 of these showed the same (ie, either up or down) regulation across the different cell types. The 2 genes CACNA1H (calcium channel; voltage-dependent; T type; α 1H subunit) and SIPA1 (signal-induced proliferation-associated 1) were down-regulated (−2.13-fold, fdr < 0.01; −1.92-fold, fdr < 0.01) in TCs after LH. In contrast, CACNA1H and SIPA1 were up-regulated in aGCs (2.74-fold, fdr < 0.01; 1.46-fold, fdr > 0.5) or mGCs (1.05-fold, fdr > 0.5; 1.69-fold, fdr < 0.01), respectively. The remaining 162 differentially expressed probe sets (representing 136 independent genes), were divided into high, intermediate, and low expression (based on HSI) for depiction in (Table 4). Fold-changes varied widely with the greatest change being the up-regulation of PTX3 (108-, 711-, 643-fold) and the down-regulation of CYP19A1 (−80-, −539-, −397-fold), in TCs, mGCs, and aGCs, respectively. Comparison of these LH-regulated genes to previous studies indicated that a large number of the identified genes have not been previously shown to be LH-regulated in bovine GCs (see Table 4).

Table 3.

LH-Regulated Genes Specific to TCs

Gene Name or Chromosomal Location Gene Symbola Fold Change HSI − Mean RMA Hybridization Value
Before LH After LH
Cytochrome P450c17, 17α-hydroxylase/17;20 lyase CYP17A1 −77.1 3452 45
Ankyrin repeat and sterile α motif domain containing 4B ANKS4B −19.1 234 12
Solute carrier family 1; member 3 SLC1A3 −10.5 195 19
chr16:69743779-69744290 (−) (TRAF5) −9.5 107 11
Chemokine (C-X-C motif) ligand 14 CXCL14 −9.2 142 15
Tetraspanin 33 TSPAN33 −8.8 270 31
Crystallin; μ CRYM −7.8 328 42
Hydroxyprostaglandin dehydrogenase 15-(NAD) HPGD −7.1 875 124
Asporin ASPN −6.5 709 109
dCTP pyrophosphatase 1 DCTPP1 −5.5 2181 400
Oncoprotein induced transcript 3 OIT3 −5.0 222 45
ChrUn_JH123563:7386-7628 - AffyID Bt.16424.1.S1_at −4.9 609 124
Transmembrane 7 superfamily member 2 TM7SF2 −4.7 624 132
Ryanodine receptor 3 RYR3 −4.5 85 19
Pyridine nucleotide-disulfide oxidoreductase domain 2 PYROXD2 −4.4 216 50
Angiopoietin 4 ANGPT4 −4.0 192 48
BCL2-related protein A1 BCL2A1 −3.9 193 49
Protein phosphatase 2; regulatory subunit B; β PPP2R2B −3.9 45 12
Perilipin 5 PLIN5 −3.7 44 12
chr5:17947935-17948135 (−) −3.7 28 8
Guanylate binding protein 4 GBP4 −3.7 106 29
Transient receptor potential cation channel, subfamily M, member 4 TRPM4 −3.3 102 30
Chemokine (C-C motif) ligand 26 CCL26 −3.3 135 41
Small integral membrane protein 10 SMIM10 −3.1 147 48
Cytochrome P450; family 4; subfamily F; polypeptide 3 CYP4F3 −3.0 64 21
Potassium large conductance calcium-activated channel; subfamily M; β member 1 KCNMB1 −3.0 52 18
Nuclear receptor subfamily 5; group A; member 1 NR5A1 −2.8 642 229
Similar to DKFZP586J0619 protein INTS1 −2.6 347 131
ADP-ribosyltransferase 3 ART3 −2.6 94 36
NLR family; CARD domain containing 5 NLRC5 −2.6 59 23
Gap junction protein; β 5; 31.1 kDa GJB5 −2.5 243 98
Diacylglycerol O-acyltransferase homolog 2 (mouse) DGAT2 −2.2 41 18
Carbohydrate (keratan sulfate Gal-6) sulfotransferase 1 CHST1 −2.2 228 102
Mevalonate kinase MVK −2.0 571 288
Fibroblast growth factor 1 (acidic) FGF1 −1.9 30 16
RAN guanine nucleotide release factor RANGRF −1.9 115 60
Mitochondrial ribosome recycling factor MRRF −1.9 445 233
chr29:48560683-48560989 (+) (CCND1) −1.9 23 12
ATP synthase subunit g, mitochondrial-like ATP5 liter −1.7 2599 1561
Solute carrier family 25; member 34 SLC25A34 −1.7 33 20
Solute carrier family 25: member 23 SCL25A23 −1.6 67 41
Fragile X mental retardation; autosomal homolog 1 FXR1 −1.6 1542 970
Kruppel-like factor 10 KLF10 2.3 581 1311
ras Homolog gene family; member B RHOB 2.4 1375 3341
chr19:32921507-32921922 (+) (Hs3st3b1) 2.6 10 25
chr11:86149219-86149711 (−) (FAM84A) 2.7 23 60
Chromosome 21 open reading frame 7 ortholog C1H21ORF7 2.9 18 52
Adenomatosis polyposis coli down-regulated 1 APCDD1 3.0 71 213
Carbonic anhydrase IV CA4 3.8 45 169
chr8:52926349-52926740 (+) (Rorb) 3.9 11 42
Coagulation factor V (proaccelerin; labile factor) F5 4.0 13 53
chr8:26339534-26340004 (−) (Acer2) 4.3 78 340
Nuclear protein; transcriptional regulator; 1 NUPR1 4.9 63 306
ATP-binding cassette; subfamily G (WHITE); member 1 ABCG1 6.9 14 93
Solute carrier family 7; member 5 SLC7A5 7.7 54 419
Ceruloplasmin (ferroxidase) CP 8.2 20 166
Lysyl oxidase-like 4 LOXL4 18.8 98 1847
a

Gene symbols enclosed in parentheses are probes sets that recognize transcripts with high homology to a named gene in another species.

Chromosomal locations are based on the Cow October 2011 assembly. Dashes reflect a negative or down regulated gene.

Table 4.

LH-Regulated Genes in GCs and mGCs and TCs

Gene Name or Chromosomal Location Gene Symbola aGC Fold Change mGC Fold Change TC Fold Change HSI − Mean RMA Hybridization Value Before LH
HSI − Mean RMA Hybridization Value After LH
aGC mGC TC aGC mGC TC
High Expression Genes
    Cytochrome P450; family 19; subfamily A; polypeptide 1 CYP19A1 −397.6 −539.3 −80.1 6762 13603 1255 17 25 16
    Carbohydrate sulfotransferase 8 CHST8 −103.8 −89.7 −15.6 2313 2044 270 22 23 17
    chr4:81639977-81640459 (+) −63.5 −44.1 −56.9 22293 23339 5402 351 529 95
    Hydroxysteroid (17-β) dehydrogenase 1 HSD17B1 −50.5 −16.5 −5.7 2452 1039 197 49 63 34
    Brain-expressed X-linked 2 BEX2 −47.6 −52.9 −37.3 15996 16449 7717 336 311 207
    Troponin I type 3 (cardiac) TNNI3 −41.0 −27.9 −10.5 10015 7713 5333 244 276 507
    Inhibin; β A INHBA −29.5 −29.4 −44.6 24933 27321 9370 846 930 210
    chr23:7266183-7266577 (+) (GCLC) −24.9 −18.8 −7.9 8260 8319 2630 331 442 335
    Solute carrier family 35, member G1 SLC35G1 −20.4 −22.5 −10.8 3422 3945 740 168 175 69
    Glutamic-pyruvate transaminase GPT −19.2 −26.5 −4.6 1748 1990 276 91 75 60
    Janus kinase 3 JAK3 −18.0 −17.7 −12.5 2269 2816 2496 126 159 200
    ST3 β-galactoside α-2;3-sialyltransferase 4 ST3GAL4 −15.8 −13.4 −3.0 8672 11352 2212 549 849 726
    Arsenic (+3 oxidation state) methyltransferase AS3MT −9.0 −9.5 −18.6 347 358 2497 38 38 134
    Serpin peptidase inhibitor; clade A; member 3 SERPINA3 −8.7 −7.6 −3.9 3539 2669 3235 408 351 832
    Stearoyl-CoA desaturase 5 SCD5 −6.3 −6.6 −2.7 7996 11245 5720 1265 1701 2133
    ADP-ribosylation factor GTPase activating protein 3 ARFGAP3 −6.1 −5.1 −2.7 6010 7954 5284 988 1571 1978
    chr27:17010375-17010926 (−) (SORBS2) −6.1 −4.9 −3.2 3427 2903 4173 565 592 1319
    Guanine nucleotide binding protein (G protein); γ 10 GNG10 −5.4 −4.7 −1.8 10398 9768 4266 1911 2068 2332
    chr25:38495947-38496497(−) −5.3 −5.9 −2.3 9275 7888 2375 1758 1339 1031
    Biliverdin reductase A BLVRA −5.0 −4.1 −2.2 4220 3271 1814 848 803 838
    chr27:36195193-36195591 (+) (PLEKHA2) −4.8 −4.2 −1.9 4214 3869 1608 872 926 835
    G protein-coupled receptor 125 GPR125 −4.6 −5.1 −2.4 2078 2619 1215 455 515 499
    Enoyl-CoA δ isomerase 1 ECI1 −3.6 −2.1 −2.2 1164 744 684 319 357 304
    Transcription elongation factor A (SII)-like 1 TCEAL1 −3.3 −2.5 −2.5 923 858 1418 277 344 562
    MAP/microtubule affinity-regulating kinase 1 MARK1 −3.2 −3.6 −3.0 2561 2565 1775 811 703 593
    Membrane-spanning 4-domains; subfamily A; member 8B MS4A8B −3.1 −6.0 −7.0 1775 3444 4092 564 575 585
    Ligand of numb-protein × 2 LNX2 −3.1 −3.0 −3.3 1899 1623 1337 611 544 411
    Ubiquitin specific peptidase 48 USP48 −2.5 −2.3 −1.8 1048 1140 640 418 493 347
    Transcriptional adaptor 3 TADA3 −2.0 −1.8 −1.9 1489 1180 827 749 648 425
    chr21:13524038-13524353 (−) LOC100848010 −2.0 −1.7 −2.0 2416 2418 2002 1237 1456 1019
    chr8:101776156-101776718 (+) (ZNF462) −1.8 −2.1 −2.0 845 896 535 461 429 270
    ORM1-like 2 (S. cerevisiae) ORMDL2 −1.6 −1.7 −1.6 965 1010 1103 593 608 696
    Progesterone receptor membrane component 1 PGRMC1 −1.3 −1.6 −1.6 1299 2355 2342 1023 1459 1480
    Myeloid cell leukemia sequence 1 MCL1 2.3 2.4 1.8 645 574 914 1495 1378 1622
    SWI/SNF related; matrix associated; actin dependent regulator of chromatin; subfamily a; member 1 SMARCA1 3.4 3.0 3.1 3258 3635 2445 11008 10934 7674
    WD repeat domain 26 WDR26 3.5 2.5 1.8 1323 1515 926 4579 3796 1701
    SLAIN motif family; member 1 SLAIN1 3.5 3.9 4.4 631 526 122 2236 2065 534
    chr3:8282216-8282683 (−) (ATF6) 4.2 3.6 2.1 904 1001 681 3828 3587 1428
    UDP-GlcNAc:βGal β-1;3-N-acetylglucosaminyltransferase 2 B3GNT2 4.4 4.4 3.5 1779 1735 847 7905 7630 2982
    chr1:50742017-50742481 (−) (CBLB) 4.7 4.6 3.4 237 267 151 1118 1218 513
    CDC42 small effector 2 CDC42SE2 5.5 5.5 2.6 696 705 510 3814 3859 1331
    TCDD-inducible poly(ADP-ribose) polymerase TIPARP 5.7 5.5 3.5 265 272 224 1513 1485 774
    Solute carrier family 25; member 17 SLC25A17 6.4 4.6 2.2 551 831 646 3551 3829 1428
    N-Acylsphingosine amidohydrolase (acid ceramidase) 1 ASAH1 6.5 6.0 3.9 324 342 567 2090 2051 2190
    Monoamine oxidase A MAOA 7.6 6.2 4.2 209 206 174 1583 1270 739
    Plasminogen activator; tissue PLAT 9.3 3.6 4.2 601 1897 801 5588 6754 3336
    Translocator protein (18 kDa) TSPO 9.3 8.3 3.5 156 170 279 1457 1413 977
    Retinol binding protein 1; cellular RBP1 10.2 16.5 7.6 1039 758 1013 10634 12524 7657
    chr28:30192805-30192992 (+) (SAMD8) 10.6 8.6 4.7 251 354 188 2660 3054 890
    Metallothionein 2A MT2A 12.9 14.8 22.6 890 629 306 11508 9286 6917
    chr12:67614705-67615018(+) (GPC6) 13.4 11.9 6.8 244 280 198 3267 3341 1341
    Spermidine/spermine N1-acetyltransferase 1 SAT1 15.0 12.7 2.7 1185 1213 4102 17742 15349 11043
    Regenerating islet-derived family; member 4 REG4 17.6 10.5 5.2 88 134 360 1540 1413 1873
    chr23:35873098-35873230 (−) (SOX4) 18.9 11.0 3.8 391 558 670 7400 6143 2564
    chr26:11594846-11595263 (−) (SLC16A12) 19.0 12.6 26.4 57 106 14 1079 1337 360
    Thrombospondin 2 THBS2 19.6 10.0 5.4 34 92 535 660 911 2863
    Nudix (nucleoside diphosphate linked moiety X)-type motif 10 NUDT10 22.0 22.4 12.1 118 130 54 2587 2910 650
    Regulator of calcineurin 1 RCAN1 23.1 20.2 3.9 167 181 288 3863 3647 1120
    SEMA5A sema domain, 7 thrombospondin repeats, transmembrane domain (TM) and short cytoplasmic domain, (semaphorin) 5A) SEMA5A 32.9 25.7 4.8 33 49 240 1101 1265 1144
    Nudix (nucleoside diphosphate linked moiety X)-type motif 11 NUDT11 34.9 29.7 13.9 176 258 187 6152 7668 2596
    FBJ murine osteosarcoma viral oncogene homolog FOS 40.3 40.2 7.2 122 134 431 4898 5396 3088
    Procollagen C-endopeptidase enhancer 2 PCOLCE2 43.1 21.6 7.6 25 55 189 1079 1181 1447
    TNF receptor superfamily; member 6b; decoy TNFRSF6B 51.6 81.1 10.0 28 25 36 1429 2005 361
    Neurotensin NTS 92.8 84.8 9.4 20 21 50 1887 1759 467
    TIMP metallopeptidase inhibitor 1 TIMP1 106.8 41.3 9.6 52 251 1563 5600 10386 15001
    Chromosome 5 open reading frame, human C12orf75 C5H12ORF75 108.6 76.2 18.9 26 37 156 2791 2803 2941
    Vanin 2 VNN2 116.0 140.9 39.6 29 22 30 3364 3056 1170
    chr5:5940972-5941399 (−) (PHLDA1) 116.0 104.7 16.3 23 25 39 2665 2589 641
    chr6:22210314-22210785 (+) (CXXC4) 141.4 83.9 18.6 17 24 19 2432 2032 351
    ρ Family GTPase 3 RND3 141.7 39.9 3.4 29 93 644 4046 3710 2217
    Prostaglandin-endoperoxide synthase 2 PTGS2 274.0 289.6 32.4 16 13 20 4287 3876 643
    Periostin; osteoblast specific factor POSTN 325.0 194.8 13.3 10 21 114 3121 4067 1514
    chr23:19331670-19332170 (+) (RUNX2) 602.3 276.5 25.3 9 20 61 5282 5645 1536
    Pentraxin 3; long PTX3 643.9 711.4 108.0 14 15 85 9232 10921 9158
Intermediate Expression Gene
    chr3:9463181-94632151 (+) (Dab1) −19.5 −15.6 −11.0 372 254 229 19 16 21
    Solute carrier family 5 (sodium/glucose cotransporter); member 11 SLC5A11 −19.1 −28.5 −13.1 227 369 329 12 13 25
    Ethanolamine kinase 2 ETNK2 −11.2 −8.6 −4.1 822 745 451 74 87 110
    Transmembrane protein 156 TMEM156 −9.1 −7.3 −3.2 728 468 169 80 65 53
    Troponin T type 1 (skeletal; slow) TNNT1 −8.3 −4.6 −15.4 148 105 898 18 23 58
    LH/CG receptor LHCGR −7.9 −17.2 −7.4 280 790 380 35 46 52
    Breast cancer antiestrogen resistance 3 BCAR3 −7.7 −6.6 −2.6 907 1074 328 118 162 125
    chr3:24537369-24537825 (−) (PDE4DIP) −4.5 −3.2 −2.7 397 446 865 88 140 320
    Aldolase C; fructose-bisphosphate ALDOC −4.4 −2.4 −2.5 939 563 582 212 236 232
    Protein phosphatase 1; regulatory (inhibitor) subunit 1A PPP1R1A −2.9 −3.9 −8.8 124 155 527 43 40 60
    unc-119 homolog (C. elegans) UNC119 −2.8 −2.8 −4.2 238 213 495 84 77 118
    Cox2 chaperone homolog COX20 −2.6 −2.0 −1.7 259 211 191 101 108 115
    Phosphofructokinase; muscle PFKM −2.5 −2.4 −2.6 327 442 616 132 186 236
    Estrogen receptor 1 ESR1 −2.4 −2.7 −1.9 391 426 174 162 160 91
    Propionyl CoA carboxylase; β polypeptide PCCB −2.2 −2.2 −2.2 362 674 977 165 303 446
    Chromosome 19 open reading frame, human C17orf101 C19H17orf101 −1.8 −1.7 −1.8 203 240 243 114 145 136
    chr10:70504545-70504958 (+) (TMEM260) −1.6 −2.0 −1.8 202 300 258 124 153 140
    chr18:53779152-53779618 (+) (ARHGAP35) −1.5 −1.7 −1.6 330 357 284 223 205 182
    Polyhomeotic homolog 2 PHC2 2.7 2.6 2.0 216 196 201 589 507 401
    Filamin A interacting protein 1 FILIP1 4.1 3.7 2.3 103 106 66 419 390 154
    Zinc finger; DHHC-type containing 23 ZDHHC23 4.2 2.6 2.4 118 165 56 499 427 133
    Retinol binding protein 4; plasma RBP4 4.7 4.2 3.9 50 83 50 237 349 195
    Similar to TPR repeat-containing protein 39 TTC39C 5.7 5.0 5.3 121 127 44 682 633 231
    chr8:99285533-99286070 (−) (ABCA1) 5.8 8.8 5.1 36 32 84 212 280 432
    Cylindromatosis CYLD 5.9 4.4 1.7 156 193 177 914 845 303
    A kinase (PRKA) anchor protein 11 AKAP11 6.5 4.9 3.2 135 149 75 875 729 238
    Inositol 1;4;5-trisphosphate 3-kinase A ITPKA 6.7 6.3 2.1 95 96 175 634 606 373
    Fibulin 1 FBLN1 6.7 4.4 4.0 97 226 130 652 994 523
    Solute carrier family 25; member 12 SLC25A12 9.5 9.7 3.3 95 94 141 905 913 468
    chr9:42277341-42277566 (+) 15.6 14.0 4.0 31 37 31 490 523 123
    TNF receptor superfamily; member 12A TNFRSF12A 16.5 20.3 6.0 17 30 81 286 617 486
    Poly (ADP-ribose) polymerase family; member 8 PARP8 21.6 19.3 6.6 47 44 48 1027 858 316
    Regulator of G-protein signaling 2 RGS2 22.1 14.7 4.4 22 26 26 490 376 113
    chr20:4115985-4116308 (−) (Sh3pxd2b) 23.8 18.3 4.6 27 36 119 649 662 550
    Protein phosphatase 4; regulatory subunit 4 PPP4R4 23.9 19.6 8.0 39 57 69 930 1113 547
    KIAA0226-like KIAA0226 liter 45.9 35.0 7.7 12 11 13 557 390 97
    Glutamate receptor; ionotrophic; AMPA 3 GRIA3 80.5 71.3 8.7 9 10 9 749 680 74
Low Expression Genes
    Rh family; B glycoprotein (gene/pseudogene) RHBG −11.1 −10.8 −7.0 99 98 89 9 9 13
    chr23:30536840-30537310 (−) (Hist1h4 h) −8.1 −9.0 −2.9 149 156 91 18 17 31
    Collectin subfamily member 11 COLEC11 −5.1 −7.1 −5.1 143 284 173 28 40 34
    Cytochrome P450; family 2; subfamily C; polypeptide 87 CYP2C87 −4.5 −4.5 −3.2 108 92 82 24 21 26
    Cytochrome P450; family 4; subfamily F; polypeptide 3 CYP4F3 −4.2 −6.4 −16.0 58 64 241 14 10 15
    Usher syndrome 1C binding protein 1 USHBP1 −4.1 −4.3 −3.2 140 127 159 34 30 50
    chr8:67614674-67615116 (−) −3.1 −3.4 −2.5 184 203 96 60 60 39
    Tetraspanin 33 TSPAN33 −2.6 −3.8 −8.8 31 48 186 12 12 21
    Chemokine (C-C motif) ligand 25 CCL25 −2.4 −2.5 −2.7 199 167 220 83 66 82
    Shisa homolog 3 (Xenopus laevis) SHISA3 −2.1 −2.9 −3.3 19 29 34 9 10 10
    SH3-domain GRB2-like 2 SH3GL2 −1.9 −3.1 −3.6 190 225 114 99 74 32
    Sushi domain containing 3 SUSD3 −1.4 −2.0 −3.7 45 58 116 31 29 31
    tRNA splicing endonuclease 54 homolog (S. cerevisiae) TSEN54 −1.3 −1.5 −1.8 95 126 167 71 83 92
    Teashirt zinc finger homeobox 2 TSHZ2 −1.3 −1.6 −2.2 55 74 195 41 46 89
    LY6/PLAUR domain containing 1 LYPD1 2.8 3.6 3.2 21 17 14 59 63 45
    chr10:27864596-27865146 (+) (KATNBL1) 3.0 2.4 1.9 101 100 49 301 246 90
    chr16:68337216-68337707 (−) (VASH2) 3.2 3.7 2.7 25 33 39 80 122 107
    chr2:53798017-53798235 (+) (ZEB2) 3.8 3.6 6.9 49 44 23 185 157 160
    Potassium large conductance calcium-activated channel; subfamily M; β member 4 KCNMB4 5.6 7.6 2.2 25 23 58 139 176 127
    Retinoic acid induced 14 RAI14 8.7 8.2 2.7 35 22 67 306 183 181
    Carbonic anhydrase XIII CA13 8.7 11.8 7.1 17 16 14 148 185 100
    RAS protein activator like 1 (GAP1 like) RASAL1 9.4 6.9 3.2 34 37 22 318 257 71
    Transmembrane protein 22 TMEM22 11.3 9.6 2.2 11 11 11 126 108 23
    chr11:22800828-22801173 (−) 13.7 14.0 3.9 14 13 14 197 188 53
    Meis homeobox 1 MEIS1 32.3 22.8 2.9 8 11 60 258 246 178
a

Gene symbols enclosed in parentheses are probes sets that recognize transcripts with high homology to a named gene in another species.

Chromosomal locations are based on the Cow October 2011 Assembly. Dashes reflect a negative or down regulated gene.

Gene set enrichment analysis

To uncover biologic functions that are significantly affected by the LH surge, differentially expressed transcripts were analyzed with the Ingenuity Pathway Analysis (IPA) tool. Altogether 1614, 1373, and 146 of the differentially expressed transcripts, which had been identified in aGCs, mGCs, and TCs, respectively, could be significantly assigned to specific IPA biofunctions. Both GC fractions were affected by LH and yielded very similar biofunctional categories, which included “cell cycle, cellular assembly and organization,” and “DNA replication, recombination, and repair,” and “cellular movement.” The “functions annotations” within these broad categories had increased “predicted activation states” that were broadly associated with cellular migration (eg, “organization of cytoplasm/cytoskeleton,” “formation of plasma membrane projections,” “microtubule dynamics;” Supplemental Table 8). In addition, these cells had significantly decreased “predicted activation states” that were associated the “functions annotations” (“segregation of chromosomes,” “proliferation of cells”: Supplemental Table 8).

IPA analysis of the 146 LH-regulated theca genes indicated that the following categories were highlighted: “lipid metabolism,” “small molecule biochemistry,” “endocrine/reproductive system development and function” and “cell death and survival” were highlighted.

The “functions annotations” within these categories all had significant decreased “predicted activation states” associated with “modification of androstenedione” or “apoptosis of epidermal cells” (Supplemental Table 9).

Discussion

This study describes, for the first time, the comparative gene expression profile of TCs cells from preovulatory follicles (before LH surge) to periovulatory follicles (collected ∼21 h after a GnRH-induced LH surge) of any species. Comparison of LH-regulated TC genes with the other LH-responsive cell (ie, granulosa) within the same follicle, also provides additional novel insights into ovarian physiology as well as helps with interpretation of previous “granulosa cell” only studies. LH responsiveness of the mural GCs has long been known to vary with respect to location of GCs with respect to distance from the basement membrane that separates it from the theca. Our results indicate that these differences are mostly quantitative because the actual genes that were expressed in the antral (aspirated) and membrane-associated (scraped) granulosa are not that different, with only a few genes exhibiting any major quantitative differences with respect to position within the follicular wall. Lastly, this study provides a robust and comprehensive accounting of gene expression changes that occur following the LH surge in bovine follicular cells and the identification of a number of novel LH-regulated genes in the bovine follicle, both in GCs and TCs.

Validation of experimental model and cellular preparations

The morphologic, physiologic, and molecular characterization (principal component analysis) of these dominant follicles collected before and after the LH surge, clearly indicate that very little variation was observed across cows, again indicating a very consistent experimental procedure. The increased mean size and the dramatic decrease in the E2:P4 ratio indicates that the conversion from an E2-active to E2-inactive follicles in the wake of the preovulatory LH surge is consistent with previous studies using this same paradigm to collect preovulatory follicles (before LH surge) and periovulatory follicles (collected ∼21 h after a GnRH-induced LH surge) as described earlier (3) as was the induction of PTGS2. The consistency of the array data was further validated by the very high correlations (R = 0.73 to 1.0) for 15 genes when compared with qRT-PCR levels. Because of these highly uniform correlation coefficients, we can be very confident in the relative fold-differences predicted by the arrays.

The purity of GC and TC sample preparations was verified by analysis of CYP17A1 (17α-hydroxylase) or CYP19A1 (aromatase) and FSHR, which have been reported to be restricted to the theca and granulosa layers, respectively (4) by in situ hybridization. Our results are consistent with earlier studies that show both steroidogenic genes and the FSHR gene are largely down-regulated by the preovulatory LH surge (3, 30, 31). The presence of CYP17A1 and CYP19A1 in mGC and TC, respectively, at very low levels, might be explained by slight contamination with the respective other cell type. Following the scraping of the basement membrane, this is probably not avoidable, nor is the incidental sloughing of some TC into the mGC preparation following scraping. Interestingly, 2 of the top LH down-regulated genes as reported by Gilbert et al. (20) in mural bovine GCs, CXCL14 (chemokine [C-X-C motif] ligand 14) and ASPN (asporin), were found in our experiments to be primarily expressed in TCs (142- and 709-HSI) compared with aGCs and mGCs (HSI < 20), respectively. Furthermore, we show that these 2 reportedly GC LH-regulated genes (20) were only LH-regulated in TCs (Table 3), decreasing significantly after the LH surge. This and other previous studies examining GC gene expression in periovulatory follicles did not examine purity of their cellular preparations and based their GC derivation solely on the method of isolation (scraping), which is identical to our procedure for mGCs. Consistent with their distance from the TCs, the aGCs had significantly less CYP17A1 than mGC, yet was still detectable (by hybridization) even though the experimental procedure (needle aspiration) was highly unlikely to allow a significant amount of cross-contamination. This leaves open several alternatives, 1) GCs/TCs may actually express very low levels of “contaminating” mRNA that are sufficient to be detected via the sensitive methods we have used or 2) alternatively, extracellular RNA in the form of exosome encapsulated/associated RNA released from GCs, TCs, or follicular fluid could be associating with the basement membrane or other cell preparations effectively contaminating them. The lack of detection of oocyte-derived transcripts (eg, BMP15 and GDF9 and ZP2, ZP3, and ZP4) in the aGC preparation illustrates that the levels of detectable contaminating RNAs (eg, CYP17A1, CYP19A1, etc.) is of a greater magnitude than that can be detected from a single oocyte; however, it is no reliable indication for the actual absence of oocyte, which might be present in these preparations. The high induction of PTGS2 expression in the GCs following the GnRH-induced LH surge is a conserved mechanism that occurs about 18 hours post-hCG treatment in bovine follicles in vivo and is considered a reliable marker for approaching ovulation (29, 32). Overall these results indicate a very small degree of contamination that may only be evident for those genes with extremely high expression such as CYP17A1 and CYP19A1, because low/intermediate expressing genes (FSHR), although still being detected, were far more cell selective. On the other hand, the uneven distribution of LHCGR transcripts particularly between the aGC and mGC samples indicates that both granulosa preparations, in fact, represent different fractions. As expected from data of others (7), LHCGR transcripts predominated in the GCs close to the basement membrane (mGC [HSI-790] compared with the aspirated cells [aGCs, HSI-280] or TCs [HSI-380] before LH, respectively).

Theca-specific transcriptome is considerably less affected by the preovulatory LH surge than the granulosa-specific transcriptome

Principal component analysis revealed that the LH surge exerted a significant influence on the expression profiles of theca and granulosa samples, leading to the arrangement of the samples into clearly defined clusters, before and after LH. Also theca and granulosa samples could be clearly separated due to their cell type-specific expression profiles. Whereas 25% and 21% of all expressed genes showed a significant reaction to the GnRH-induced LH surge in aGCs and mGCs respectively, only 2% of the genes present in TCs were significantly up- or down-regulated. The vast majority of genes that are LH regulated in granulosa and theca showed the same direction of change (up or down) in both cell types. The strongly varying extent of LH response in theca and granulosa, however, cannot be simply explained by a lower number of LH binding sites (LH receptors) on cells of the theca compared with those of the granulosa. Previous studies, as well as the present study, indicate that both cell types, granulosa and theca, expressed the corresponding LHCGR transcripts at relatively high levels before the onset of the LH surge (see Table 1 and Refs. 3, 4, and 33). One might speculate that the signal transduction pathway downstream of the LH receptor might be different in GCs and TCs during the periovulatory period. This is the first demonstration that the TC transcriptome shows considerably less alteration in the wake of the preovulatory LH surge compared with that of the GCs. This result might be expected if one considers the reduced morphologic and biochemical changes that occur in TCs (loss of androgen biosynthesis, minimal cell hypertrophy) compared with GCs (loss of cell proliferation, loss of aromatase activity, induction of numerous steroidogenic genes, extensive cell hypertrophy).

Novel genes identified in bovine TCs that are regulated by the LH surge

Although critical roles in periovulatory follicular function and subsequent luteal function have been defined for TCs, previous studies examining TC cell gene expression have been limited to individual genes (3, 34, 35) or cultured TC experiments (36). This study is therefore the first to globally identify thecal genes that exhibit changes in mRNA expression in vivo following the LH surge. In total 203 genes (combined results of Tables 3 and 4) exhibited differential expression in the TCs following the LH surge. However, only 57 of these 199 were specifically LH regulated in TCs (Table 3). Of the remaining 141 LH-regulated genes found common to both granulosa and theca, only about 10% or 19 of these LH-regulated genes could be absolutely confirmed to be LH regulated in theca because they had higher (HSI) expression levels than in corresponding GC preparations. A great number of the other 141 genes (Table 4) showed similar levels across the cell types, but without further analysis (ie, in situ hybridization or laser capture dissection followed by qRT-PCR confirmation) cannot be assured as truly LH-regulated genes in TC, and not cross-contamination by granulosa. The 19 additional thecal LH-regulated genes included AS3MT, SORBS2, TCEAL1, MS4A8B, ORMDL2, MCL1, ASAH1, SAT1, THBS2, FOS, PCOLCE2, TIMP1, RND3, TNNT1, PDE4DIP, PPP1R1A, PFKM, PCCB, SIPA1 (encoding arsenic methyltransferase, transcription elongation factor A-like 1, sorbin and SH3 domain containing 2, membrane spanning 4-domains; subfamily A; member 8B, ORM1-like 2, myeloid cell leukemia sequence 1, N-acylsphingosine amidohydrolase 1 [acid ceramidase], spermidine/speriine N1-acetyltransferase 1, thromospondin 2, FOS, procollagen C-endopeptidase enhancer 2, TIMP1, Rho family GTPase 3, troponin T type 1, phosphodiesterase 4D interacting protein [PDE4DIP], transcript variant 8, phosphofructokinase, propionyl CoA carboxylase; β polypeptide, protein phosphatase 1; regulatory subunit 1A, signal-induced proliferation-associated 1, respectively).

Not surprisingly, CYP17A1 mRNA, which encodes the critical TC enzyme involved in androgen biosynthesis had the greatest decrease (1/77 of pre-LH values) following the LH surge. Transcriptional regulation of the CYP17A1 gene has been well studied (31, 37) and steroidogenic factor-1 (SF-1 encoded by NR5A1) one of its primary transcriptional regulators was also decreased to approximately one-third of its pre-GnRH level following the LH surge in TC (Table 3). As an established mediator of steroidogenic activity and known transcriptional activator of CYP17A1, this loss of SF-1 in TCs and lack of LH effect on GCs is consistent with previous observations, which indicate that NR5A2 (also known as LRH-1) is the primary steroidogenic transcription factor in GCs (38). We additionally identified acid ceramidase (ASAH1) as one of 19 genes induced (∼4- to 6-fold) by LH that was common to both TCs and GCs. Recently, ASAH1, which catalyzes the conversion of ceramide to sphingoside (generating sphingoside 1 phosphate) was demonstrated to be induced in adrenocortical cells by ACTH/cAMP, and this was associated with changes in adrenal steroidogenesis, by decreasing the expression of a number of steroidogenic enzymes including CYP17A1 (39, 40). This is the first demonstration that LH may have a similar effect in TCs. Another unique aspect of sphinoside-1 phosphate is its antagonist actions on NR5A1 transcriptional activity (41). Thus, in addition to a decrease in SF-1 expression, acid ceremide-induced sphingosine may further block SF-1 action, ultimately leading to the sharp reduction in CYP17A1 expression we observed. With respect to the remaining LH-regulated thecal genes, a large number are associated with lipid metabolism and regulation of apoptosis (see Supplemental Table 9), both of which would be considered as important in luteinization and formation of the corpus luteum.

To our surprise we found PTX3 (pentraxin 3) as a highly abundant and LH-regulated gene also in TCs. It is well established that this protein plays a role as a modulator of the inflammatory response (42). In the context of the ovarian follicle, however, its expression has only been reported in the context of GCs of periovulatory follicles, where it mediates cumulus cell-oocyte interactions (43). In a recent paper it was also found that it interferes with fibroblast growth factor 2 actions in luteal endothelial cells, thus inhibiting angiogenesis (44). Suggestively, pentraxin 3 might be involved in the regulation of angiogenesis also in the intensely vascularized theca layer. However, the function of its heavy LH-induced up-regulation during the periovulatory period still seems obscure, because this protein is an inhibitor of angiogenesis, whereas the luteinization process is accompanied by intense vascular growth.

The thecal-specific gene with the greatest LH induction we identified was LOXL4 (encoding lysyl oxidase-like 4). This gene's expression increased 18.8-fold (HSI-98 to HSI-1847) following the LH surge, with no concomitant LH induction in aGCs or mGCs (HSI was <148 at all time points in granulosa). Lysyl oxidases are an extracellular copper enzyme that cross-link lysine side chains in mature fibrillar collagen or elastin, regulating the tensile strength and structural integrity of connective tissues (45). LOXL4 shares it amine oxidase motif and the scavenger receptor cysteine-rich protein domain with 4 other family members, lysyl oxidase (LOX) and lysyl oxidase like 1–4 (45). Our study detected expression of LOX, LOXL1, and LOXL2 in TCs and GCs, with TCs having higher levels of LOX (HSI-140) and LOXL1 (HSI-450) than GCs (HSI < 30 for both), whereas equal levels of LOXL2 (all HSI < 62) were found in TCs and GCs. However, no LH regulation was observed for these other lysyl oxidase family members within TC or GC compartments. Conversely, although whole-tissue Northern blot analyses have demonstrated presence of LOXL4 in human ovary as well as numerous other tissues (46), this is the first indication of LH regulation of this gene in any ovarian tissue. Previous studies examining LOX expression and activity have shown it to be down-regulated during early folliculogenesis by FSH (47, 48). In other cells, LOXL4 has been shown to block cell migration by suppressing the synthesis of laminins and α3 integrin and the activity of matrix metallopeptidase 2 (49), and recent studies have shown the full-length protein to be a tumor suppressor gene (50). Thus, the high induction of LOXL4 seen in TCs might be a critical regulatory protein within the extracellular matrix in the highly dynamic periovulatory follicle, and this gene awaits further study. Lastly, most of the newly identified TC genes have not been studied within the context of the periovulatory follicle; therefore this study provides a critical first step in our understanding this largely understudied follicular cell.

Variation in mural GC transcriptome based on location within the periovulatory follicle

Immunohistochemical analyses and in situ hybridization studies have shown that LHCGR levels can vary with distance from the basement membrane (7). The relative expression of LHCGR in our aGC and mGC preparations before the LH surge (Table 1) supports this observation although it did not reach significance. Recent studies examining the gene profile in GC types, mural vs cumulus granulosa, have observed large differences (5153). Our study did not aim to investigate this difference but instead wished to determine whether differences in the mural granulosa (ie, those near the antrum vs those near the membrane) could be elucidated. Indeed although our aGC samples would likely be contaminated with cumulus cells and the oocyte, we failed to detect oocyte-specific genes or the differences observed in these other papers (5153). Our study further indicates only minor differences in the 2 different mural GC preparations: mGCs (ie, membrane-associated) and aGCs (aspirated). This inability to detect changes could easily be attributed to a pronounced contamination of the aGC population with mGCs, a result that cannot be prevented using the current procedures. To further evaluate whether a quantitative difference might exist we compared the fold changes in response to LH across the cells (See Supplemental Table 7 for complete list) and identified a small number of genes (29) that were different (Table 2). We observed a preferential loss (one-fourth of its pre-LH levels) of CARTPT transcript in mGCs that was unaffected in aGCs. Decreased follicular fluid levels and expression of CARTPT have been previously tied to bovine follicle selection and increased health status of the dominant follicle in bovine (54). Many of the other genes that were quantitatively differentially expressed (>3-fold difference) between aGCs and mGCs were either extracellular matrix proteins or proteins that interact with or metabolize these proteins. Interestingly, extracellular matrix genes were also enriched in cumulus cells in comparison with mural granulosa in human preovulatory follicles (52). Extracellular matrix proteins separate the granulosa and theca (ie, basement membrane) or are of a specialized type (also known as focimatrix proteins) between GCs (55). Most of these proteins (ie, collagen α-1, claudin 11, a disintegrin-like and metallopeptidase with thrombospondin type 1 motif [ADAMTS1]), had slightly higher expression in the mGCs vs aGCs before the LH surge and fairly equivalent levels after the LH surge. Nidogen 2 and fibronectin 1, both extracellular matrix proteins, had clearly greater expression in the mGC (558- and 744-HSI) compared with almost background levels (23- and 89-HSI) in aGCs before LH, and almost equivalent levels after the LH surge (see Table 2). Two additional genes which modulate extracellular signaling, neural precursor cell expressed; developmentally down-regulated 9 (NEDD9) and SAM and SH3 domain containing 1 (SASH1) were LH-regulated in aGCs, exclusively. These two proteins have been linked to cancer cell migration and invasion (56, 57). Whether these genes may play a role in the ovulation by oocyte migration model as proposed by Akison et al. (58) is unknown. Lastly, several genes involved in cell cycle control, CCND2 (cyclin D2) and KIAA0101 (also known as proliferating cell nuclear antigen-associated factor) were both found to undergo a pronounced decrease in expression after the LH surge, with the decline being of slightly greater magnitude in the aGCs than in the mGCs (see Table 2).

Novel genes identified in bovine GCs that are regulated by the LH surge

Our study identified a large number of heretofore unknown LH-regulated genes in bovine granulosa, even though this is not the first study to characterize the periovulatory GCs following the LH surge. Indeed, a recent study examining late post-LH changes of GC-specific expression profiles in bovine follicles (20) having remarkably different data was reported. Surprisingly, only 121 of LH-regulated genes are common between both studies. In addition, when comparing the transcript abundance data of individual genes, remarkable differences are evident. From the 15 cell marker transcripts shown in Table 1, 14 are represented on both microarray systems (ADAMTS1, CCND2, CYP11A1, CYP19A1, FSHR, LHCGR, NR5A1, NR5A2, PCNA, PTGS2, PTX3, STAR, TIMP1, HSD3B1). From these 14, only CCND2, STAR, and TIMP1 were differentially expressed in the Gilbert et al. (20) data set (see Supplemental Data 1). The fact that PTGS2 was not up-regulated in the study of Gilbert et al. (20), at 22 hours post-LH, is surprising. PTGS2 expression is a conserved mechanism that occurs about 18 hours post-hCG treatment in bovine follicles in vivo (29, 32) and can be considered as a reliable marker for approaching ovulation. A thorough comparison of the 2 studies is presented in Supplemental Data 1. Consistent with our study, of 23 granulosa-derived transcripts that were found down-regulated 23 hours after hCG application using substractive hybridization (19), 13 (ANAPC5, ARFGAP3, CYP19A1, FNDC3B, FSHR, GJA1, IDH3A, INHBA, LRP8, PRC1, RPA2, SCD, TRIB2) were confirmed. In this regard, however, the deviating animal models might be responsible for some of the differences observed between studies. In the study of Gilbert et al. (20) follicles were harvested following an ovarian superovulation regime. In contrast, the monitoring (ultrasonography during estrous cycle, signs of heat, morphologic parameters, antral E2 and P4 concentrations, appearance of PTGS2 transcripts) and hormone treatment regime used during the present study was developed to obtain only a single, well-characterized large dominant preovulatory follicle per animal, thus mimicking the natural cycling situation as close as possible.

We identified 2 members of the Ig-like domain-containing superfamily, IGSF1 and IGSF11, encoding Ig superfamily; members 1 and 11, as LH-regulated genes in GC. These genes have not been identified in GC before and, interestingly, both are regulated by LH in the opposite way: whereas the x-chromosomal IGSF1 is clearly up-regulated in aGCs and mGCs (10.1- and 10.9-fold in aGCs and mGCs, respectively), IGSF11 is significantly down-regulated in both GC populations (−4.9 and −6.7 fold in aGCs and mGCs, respectively). Generally, the Ig domain has evolved to serve diverse biologic functions including growth and development, signaling, adhesion, and protein-carbohydrate interactions (59). Specifically, IGSF11 has been clearly connected to carcinogenesis (60, 61). The role of these specific Igs in luteinizing GC and, in particular, the complementary regulatory characteristic is not yet clear. However considering their function in other tissues, both might play a role during the intense tissue reorganization of the granulosa layer during the formation of the corpus luteum. This might also be applicable to another interesting gene, AMIGO2 (adhesion molecule with Ig-like domain 2), which is up-regulated 6.0- and 3.0-fold in aGCs and mGCs, respectively, by LH. AMIGO2 is a member of the AMIGO/Alvin family, which are transmembrane proteins characterized by 6 leucine-rich repeats and a single Ig-like extracellular domain. AMIGO proteins are mainly found in the nervous system, where they function as novel cell adhesion molecules that facilitate neuronal growth (62). Correspondingly, during the formation of the corpus luteum AMIGO2 that is up-regulated to relatively high levels in aGCs and mGCs by the preovulatory LH surge (1020-HSI and 1117-HSI, respectively; Supplemental Table 7) might also be involved in mediating cell adhesion and regulating migration of GC.

In conclusion, the data of the present study demonstrate that 1) theca cell-specific transcriptome is considerably less affected by the preovulatory LH surge than that of GCs, consistent with the greater changes induced in GCs as they undergo luteinization, 2) aGCs and mGCs show very similar gene expression profiles, thus suggesting a widely equivalent molecular equipment of both GC preparations and 3) numerous novel LH-regulated genes in GCs and TCs were identified. This is particularly true for the TCs where, with the exception of a few known TC marker genes, almost all of the genes were identified as LH regulated for the first time. This research resource comparing the transcriptomic profiles of both thecal and granulosa follicular cells from preovulatory (pre-LH) and post-LH (ie, periovulatory) follicles provides an easily accessible and searchable resource that should greatly benefit the scientific community.

Supplementary Material

Supplemental Data
Supplemental Data

Acknowledgments

We thank Maren Anders of the Reproductive Biology Unit at the Leibniz Institute for Farm Animal Biology for excellent technical assistance.

This work was supported by Grant VA135/5-2 from the Deutsche Forschungsgemeinschaft (DFG) and by Grant HD061580 from the National Institute of Health (to L.K.C.).

Disclosure Summary: The authors have nothing to disclose.

Footnotes

Abbreviations:
aGC
antral (aspirated) granulosa cell
E2
17β-estradiol
fdr
false discovery rate
GAPDH
glyceraldehyde-3-phosphate dehydrogenase
GC
granulosa cell
hCG
human chorionic gonadotropin
HSI
hybridization signal intensity
IPA
Ingenuity Pathway Analysis
mGC
membrane-associated granulosa cell
P4
progesterone
qRT-PCR
quantitative RT-PCR
RMA
Robust Multichip Average
SF-1
steroidogenic factor-1
TC
thecal cell.

References

  • 1. Smith MF, McIntush EW, Smith GW. Mechanisms associated with corpus luteum development. J Anim Sci. 1994;72:1857–1872 [DOI] [PubMed] [Google Scholar]
  • 2. Murphy BD. 2004 Luteinization. In: Leung PC, Adashi EY, eds. The Ovary. 2nd ed Burlington, MA: Academic Press; 2004;185–199 [Google Scholar]
  • 3. Nimz M, Spitschak M, Schneider F, Fürbass R, Vanselow J. Down-regulation of genes encoding steroidogenic enzymes and hormone receptors in late preovulatory follicles of the cow coincides with an accumulation of intrafollicular steroids. Domest Anim Endocrinol. 2009;37:45–54 [DOI] [PubMed] [Google Scholar]
  • 4. Bao B, Garverick HA. Expression of steroidogenic enzyme and gonadotropin receptor genes in bovine follicles during ovarian follicular waves: a review. J Anim Sci. 1998;76:1903–1921 [DOI] [PubMed] [Google Scholar]
  • 5. Hillier SG, Whitelaw PF, Smyth CD. Follicular oestrogen synthesis: the 'two-cell, two-gonadotrophin model' revisited. Mol Cell Endocrinol. 1994;100:51–54 [DOI] [PubMed] [Google Scholar]
  • 6. Orisaka M, Tajima K, Tsang BK, Kotsuji F. Oocyte-granulosa-theca cell interactions during preantral follicular development. J Ovarian Res. 2009;2:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Peng XR, Hsueh AJ, LaPolt PS, Bjersing L, Ny T. Localization of luteinizing hormone receptor messenger ribonucleic acid expression in ovarian cell types during follicle development and ovulation. Endocrinology. 1991;129:3200–3207 [DOI] [PubMed] [Google Scholar]
  • 8. Gilchrist RB, Ritter LJ, Armstrong DT. Oocyte-somatic cell interactions during follicle development in mammals. Anim Reprod Sci. 2004;82–83:431–446:431–446 [DOI] [PubMed] [Google Scholar]
  • 9. Markholt S, Grøndahl ML, Ernst EH, Andersen CY, Ernst E, Lykke-Hartmann K. Global gene analysis of oocytes from early stages in human folliculogenesis shows high expression of novel genes in reproduction. Mol Hum Reprod. 2012;18:96–110 [DOI] [PubMed] [Google Scholar]
  • 10. Eppig JJ, Wigglesworth K, Pendola F, Hirao Y. Murine oocytes suppress expression of luteinizing hormone receptor messenger ribonucleic acid by granulosa cells. Biol Reprod. 1997;56:976–984 [DOI] [PubMed] [Google Scholar]
  • 11. Skinner MK, Schmidt M, Savenkova MI, Sadler-Riggleman I, Nilsson EE. Regulation of granulosa and theca cell transcriptomes during ovarian antral follicle development. Mol Reprod Dev. 2008;75:1457–1472 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Liu Z, Youngquist RS, Garverick HA, Antoniou E. Molecular mechanisms regulating bovine ovarian follicular selection. Mol Reprod Dev. 2009;76:351–366 [DOI] [PubMed] [Google Scholar]
  • 13. Zielak AE, Canty MJ, Forde N, et al. Differential expression of genes for transcription factors in theca and granulosa cells following selection of a dominant follicle in cattle. Mol Reprod Dev. 2008;75:904–914 [DOI] [PubMed] [Google Scholar]
  • 14. Hayashi KG, Ushizawa K, Hosoe M, Takahashi T. Differential genome-wide gene expression profiling of bovine largest and second-largest follicles: identification of genes associated with growth of dominant follicles. Reprod Biol Endocrinol. 2010;8:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Jo M, Gieske MC, Payne CE, et al. Development and application of a rat ovarian gene expression database. Endocrinology. 2004;145:5384–5396 [DOI] [PubMed] [Google Scholar]
  • 16. Espey LL, Richards JS. Temporal and spatial patterns of ovarian gene transcription following an ovulatory dose of gonadotropin in the rat. Biol Reprod. 2002;67:1662–1670 [DOI] [PubMed] [Google Scholar]
  • 17. Carletti MZ, Christenson LK. Rapid effects of LH on gene expression in the mural granulosa cells of mouse periovulatory follicles. Reproduction. 2009;137:843–855 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Xu F, Stouffer RL, Müller J, et al. Dynamics of the transcriptome in the primate ovulatory follicle. Mol Hum Reprod. 2011;17:152–165 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Ndiaye K, Fayad T, Silversides DW, Sirois J, Lussier JG. Identification of downregulated messenger RNAs in bovine granulosa cells of dominant follicles following stimulation with human chorionic gonadotropin. Biol Reprod. 2005;73:324–333 [DOI] [PubMed] [Google Scholar]
  • 20. Gilbert I, Robert C, Dieleman S, Blondin P, Sirard MA. Transcriptional effect of the LH surge in bovine granulosa cells during the peri-ovulation period. Reproduction. 2011;141:193–205 [DOI] [PubMed] [Google Scholar]
  • 21. Rao JU, Shah KB, Puttaiah J, Rudraiah M. Gene expression profiling of preovulatory follicle in the buffalo cow: effects of increased IGF-I concentration on periovulatory events. PLoS One. 2011;6:e20754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Walsh SW, Mehta JP, McGettigan PA, et al. Effect of the metabolic environment at key stages of follicle development in cattle: focus on steroid biosynthesis. Physiol Genomics. 2012;44:504–517 [DOI] [PubMed] [Google Scholar]
  • 23. Vanselow J, Spitschak M, Nimz M, Fürbass R. DNA methylation is not involved in preovulatory down-regulation of CYP11A1, HSD3B1, and CYP19A1 in bovine follicles but may have a role in permanent silencing of CYP19A1 in large granulosa lutein cells. Biol Reprod. 2010;82:289–298 [DOI] [PubMed] [Google Scholar]
  • 24. Schneider F, Brüssow KP. Effects of a preovulatory administered depot gonadotrophin-releasing hormone agonist on reproductive hormone levels and pregnancy outcome in gilts. Reprod Fertil Dev. 2006;18:857–866 [DOI] [PubMed] [Google Scholar]
  • 25. Ireland JJ, Roche JF. Development of antral follicles in cattle after prostaglandin-induced luteolysis: changes in serum hormones, steroids in follicular fluid, and gonadotropin receptors. Endocrinology. 1982;111:2077–2086 [DOI] [PubMed] [Google Scholar]
  • 26. Irizarry RA, Hobbs B, Collin F, et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003;4:249–264 [DOI] [PubMed] [Google Scholar]
  • 27. Nimz M, Spitschak M, Fürbass R, Vanselow J. The pre-ovulatory luteinizing hormone surge is followed by down-regulation of CYP19A1, HSD3B1, and CYP17A1 and chromatin condensation of the corresponding promoters in bovine follicles. Mol Reprod Dev. 2010;77:1040–1048 [DOI] [PubMed] [Google Scholar]
  • 28. Sirois J, Sayasith K, Brown KA, Stock AE, Bouchard N, Doré M. Cyclooxygenase-2 and its role in ovulation: a 2004 account. Human Reproduction Update. 2004;10:373–385 [DOI] [PubMed] [Google Scholar]
  • 29. Sirois J. Induction of prostaglandin endoperoxide synthase-2 by human chorionic gonadotropin in bovine preovulatory follicles in vivo. Endocrinology. 1994;135:841–848 [DOI] [PubMed] [Google Scholar]
  • 30. Voss AK, Fortune JE. Levels of messenger ribonucleic acid for cytochrome P450 17α-hydroxylase and P450 aromatase in preovulatory bovine follicles decrease after the luteinizing hormone surge. Endocrinology. 1993;132:2239–2245 [DOI] [PubMed] [Google Scholar]
  • 31. Conley AJ, Kaminski MA, Dubowsky SA, Jablonka-Shariff A, Redmer DA, Reynolds LP. Immunohistochemical localization of 3β-hydroxysteroid dehydrogenase and P450 17α-hydroxylase during follicular and luteal development in pigs, sheep, and cows. Biol Reprod. 1995;52:1081–1094 [DOI] [PubMed] [Google Scholar]
  • 32. Liu J, Carrière PD, Doré M, Sirois J. Prostaglandin G/H synthase-2 is expressed in bovine preovulatory follicles after the endogenous surge of luteinizing hormone. Biol Reprod. 1997;57:1524–1531 [DOI] [PubMed] [Google Scholar]
  • 33. Bao B, Garverick HA, Smith GW, Smith MF, Salfen BE, Youngquist RS. Changes in messenger ribonucleic acid encoding luteinizing hormone receptor, cytochrome P450-side chain cleavage, and aromatase are associated with recruitment and selection of bovine ovarian follicles. Biol Reprod. 1997;56:1158–1168 [DOI] [PubMed] [Google Scholar]
  • 34. Li Q, Jimenez-Krassel F, Ireland JJ, Smith GW. Gene expression profiling of bovine preovulatory follicles: gonadotropin surge and prostanoid-dependent up-regulation of genes potentially linked to the ovulatory process. Reproduction. 2009;137:297–307 [DOI] [PubMed] [Google Scholar]
  • 35. Mamluk R, Wolfenson D, Meidan R. LH receptor mRNA and cytochrome P450 side-chain cleavage expression in bovine theca and granulosa cells luteinized by LH or forskolin. Domest Anim Endocrinol. 1998;15:103–114 [DOI] [PubMed] [Google Scholar]
  • 36. Wood JR, Nelson VL, Ho C, et al. The molecular phenotype of polycystic ovary syndrome (PCOS) theca cells and new candidate PCOS genes defined by microarray analysis. J Biol Chem. 2003;278:26380–26390 [DOI] [PubMed] [Google Scholar]
  • 37. Komar CM, Berndtson AK, Evans AC, Fortune JE. Decline in circulating estradiol during the periovulatory period is correlated with decreases in estradiol and androgen, and in messenger RNA for p450 aromatase and p450 17α-hydroxylase, in bovine preovulatory follicles. Biol Reprod. 2001;64:1797–1805 [DOI] [PubMed] [Google Scholar]
  • 38. Zhao H, Li Z, Cooney AJ, Lan ZJ. Orphan nuclear receptor function in the ovary. Front Biosci. 2007;12:3398–3405 [DOI] [PubMed] [Google Scholar]
  • 39. Lucki N, Sewer MB. The cAMP-responsive element binding protein (CREB) regulates the expression of acid ceramidase (ASAH1) in H295R human adrenocortical cells. Biochim Biophys Acta. 2009;1791:706–713 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Lucki NC, Bandyopadhyay S, Wang E, Merrill AH, Sewer MB. Acid ceramidase (ASAH1) is a global regulator of steroidogenic capacity and adrenocortical gene expression. Mol Endocrinol. 2012;26:228–243 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Urs AN, Dammer E, Sewer MB. Sphingosine regulates the transcription of CYP17 by binding to steroidogenic factor-1. Endocrinology. 2006;147:5249–5258 [DOI] [PubMed] [Google Scholar]
  • 42. Kunes P, Holubcova Z, Kolackova M, Krejsek J. Pentraxin 3(PTX 3): an endogenous modulator of the inflammatory response. Mediators Inflamm. 2012;2012:920517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Varani S, Elvin JA, Yan C, et al. Knockout of pentraxin 3, a downstream target of growth differentiation factor-9, causes female subfertility. Mol Endocrinol. 2002;16:1154–1167 [DOI] [PubMed] [Google Scholar]
  • 44. Zalman Y, Klipper E, Farberov S, et al. Regulation of angiogenesis-related prostaglandin f2α-induced genes in the bovine corpus luteum. Biol Reprod. 2012;86:92. [DOI] [PubMed] [Google Scholar]
  • 45. Smith-Mungo LI, Kagan HM. Lysyl oxidase: properties, regulation and multiple functions in biology. Matrix Biol. 1998;16:387–398 [DOI] [PubMed] [Google Scholar]
  • 46. Mäki JM, Tikkanen H, Kivirikko KI. Cloning and characterization of a fifth human lysyl oxidase isoenzyme: the third member of the lysyl oxidase-related subfamily with four scavenger receptor cysteine-rich domains. Matrix Biol. 2001;20:493–496 [DOI] [PubMed] [Google Scholar]
  • 47. Harlow CR, Rae M, Davidson L, Trackman PC, Hillier SG. Lysyl oxidase gene expression and enzyme activity in the rat ovary: regulation by follicle-stimulating hormone, androgen, and transforming growth factor-β superfamily members in vitro. Endocrinology. 2003;144:154–162 [DOI] [PubMed] [Google Scholar]
  • 48. Slee RB, Hillier SG, Largue P, Harlow CR, Miele G, Clinton M. Differentiation-dependent expression of connective tissue growth factor and lysyl oxidase messenger ribonucleic acids in rat granulosa cells. Endocrinology. 2001;142:1082–1089 [DOI] [PubMed] [Google Scholar]
  • 49. Kim DJ, Lee DC, Yang SJ, et al. Lysyl oxidase like 4, a novel target gene of TGF-β1 signaling, can negatively regulate TGF-β1-induced cell motility in PLC/PRF/5 hepatoma cells. Biochem Biophys Res Commun. 2008;373:521–527 [DOI] [PubMed] [Google Scholar]
  • 50. Sebban S, Golan-Gerstl R, Karni R, Vaksman O, Davidson B, Reich R. Alternatively spliced lysyl oxidase-like 4 isoforms have a pro-metastatic role in cancer. Clin Exp Metastasis. 2013;30:103–117 [DOI] [PubMed] [Google Scholar]
  • 51. Gilchrist RB, Ritter LJ, Myllymaa S, et al. Molecular basis of oocyte-paracrine signalling that promotes granulosa cell proliferation. J Cell Sci. 2006;119:3811–3821 [DOI] [PubMed] [Google Scholar]
  • 52. Grøndahl ML, Andersen CY, Bogstad J, Borgbo T, Boujida VH, Borup R. Specific genes are selectively expressed between cumulus and granulosa cells from individual human pre-ovulatory follicles. Mol Hum Reprod. 2012;18:572–584 [DOI] [PubMed] [Google Scholar]
  • 53. Kõks S, Velthut A, Sarapik A, et al. The differential transcriptome and ontology profiles of floating and cumulus granulosa cells in stimulated human antral follicles. Mol Hum Reprod. 2010;16:229–240 [DOI] [PubMed] [Google Scholar]
  • 54. Smith GW, Sen A, Folger JK, Ireland JJ. Putative role of cocaine- and amphetamine-regulated transcript (CARTPT) in dominant follicle selection in cattle. Soc Reprod Fertil Suppl. 2010;67:105–117 [PubMed] [Google Scholar]
  • 55. Irving-Rodgers HF, Catanzariti KD, Aspden WJ, D'Occhio MJ, Rodgers RJ. Remodeling of extracellular matrix at ovulation of the bovine ovarian follicle. Mol Reprod Dev. 2006;73:1292–1302 [DOI] [PubMed] [Google Scholar]
  • 56. Chang JX, Gao F, Zhao GQ, Zhang GJ. Role of NEDD9 in invasion and metastasis of lung adenocarcinoma. Exp Ther Med. 2012;4:795–800 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Meng Q, Zheng M, Liu H, et al. SASH1 regulates proliferation, apoptosis, and invasion of osteosarcoma cell. Mol Cell Biochem. 2013;373:201–210 [DOI] [PubMed] [Google Scholar]
  • 58. Akison LK, Alvino ER, Dunning KR, Robker RL, Russell DL. Transient invasive migration in mouse cumulus oocyte complexes induced at ovulation by luteinizing hormone. Biol Reprod. 2012;86:125. [DOI] [PubMed] [Google Scholar]
  • 59. Srinivasan M, Roeske RW. Immunomodulatory peptides from IgSF proteins: a review. Curr Protein Pept Sci. 2005;6:185–196 [DOI] [PubMed] [Google Scholar]
  • 60. Watanabe T, Suda T, Tsunoda T, et al. Identification of immunoglobulin superfamily 11 (IGSF11) as a novel target for cancer immunotherapy of gastrointestinal and hepatocellular carcinomas. Cancer Sci. 2005;96:498–506 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Ailan H, Xiangwen X, Daolong R, et al. Identification of target genes of transcription factor activator protein 2 gamma in breast cancer cells. BMC Cancer. 2009;9:279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Chen Y, Aulia S, Li L, Tang BL. AMIGO and friends: an emerging family of brain-enriched, neuronal growth modulating, type I transmembrane proteins with leucine-rich repeats (LRR) and cell adhesion molecule motifs. Brain Res Rev. 2006;51:265–274 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Data
Supplemental Data

Articles from Molecular Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES