Luteinizing Hormone Signaling in Preovulatory ... - Semantic Scholar

7 downloads 0 Views 400KB Size Report
Jan 2, 2008 - factor shedding. LH- but not amphiregulin-induced oocyte maturation and EGFR phosphorylation were sensitive to protein synthesis inhibition.
0888-8809/08/$15.00/0 Printed in U.S.A.

Molecular Endocrinology 22(4):924–936 Copyright © 2008 by The Endocrine Society doi: 10.1210/me.2007-0246

Luteinizing Hormone Signaling in Preovulatory Follicles Involves Early Activation of the Epidermal Growth Factor Receptor Pathway Sara Panigone,* Minnie Hsieh,* Maoyong Fu, Luca Persani, and Marco Conti Division of Reproductive Biology (S.P., M.H., M.F., M.C.), Department of Obstetrics and Gynecology, Stanford, University School of Medicine, Stanford, California 94305; and Department of Medical Sciences (S.P., L.P.), University of Milan, Lab of Experimental Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Auxologico Italiano, Milano 20095, Italy LH activates a cascade of signaling events that are propagated throughout the ovarian preovulatory follicle to promote ovulation of a mature egg. Critical to LH-induced ovulation is the induction of epidermal growth factor (EGF)-like growth factors and transactivation of EGF receptor (EGFR) signaling. Because the timing of this transactivation has not been well characterized, we investigated the dynamics of LH regulation of the EGF network in cultured follicles. Preovulatory follicles were cultured with or without recombinant LH and/or specific inhibitors. EGFR and MAPK phosphorylation were examined by immunoprecipitation and Western blot analyses. By semiquantitative RT-PCR, increases in amphiregulin and epiregulin mRNAs were detected 30 min after recombinant LH stimulation of follicles and were maximal after 2 h. LH-induced EGFR phosphorylation also increased after 30 min and reached a maximum at 2 h. EGFR activation precedes oocyte maturation and is cAMP dependent, because forskolin similarly acti-

vated EGFR. LH-induced EGFR phosphorylation was sensitive to AG1478, an EGFR kinase inhibitor, and to inhibitors of matrix metalloproteases GM6001 and TNF␣ protease inhibitor-1 (TAPI-1), suggesting the involvement of EGF-like growth factor shedding. LH- but not amphiregulin-induced oocyte maturation and EGFR phosphorylation were sensitive to protein synthesis inhibition. When granulosa cells were cultured with a combination of neutralizing antibodies against amphiregulin, epiregulin, and betacellulin, EGFR phosphorylation and MAPK activation were inhibited. In cultured follicles, LH-induced MAPK activation was partially inhibited by AG1478 and GM6001, indicating that this pathway is regulated in part by the EGF network but also involves additional pathways. Thus, complex mechanisms are involved in the rapid amplification and propagation of the LH signal within preovulatory follicles and include the early activation of the EGF network. (Molecular Endocrinology 22: 924–936, 2008)

T

Gs protein and activates adenylyl cyclase to elevate intracellular cAMP levels. The initial cAMP signal subsequently branches out to several signaling pathways including those downstream of protein kinase A (PKA), cAMP-activated guanine nucleotide exchange factors (cAMP-GEFs or EPAC) and phospholipase C (1–3). Although LH directly stimulates theca and granulosa cells, its effects on cumulus cells and oocytes are probably indirect. LH induces marked responses in cumulus cells and oocytes, yet these cells express few or no LHRs and fail to respond when directly stimulated by LH (4). In recent years, members of the epidermal growth factor (EGF)-like growth factor family have emerged as likely mediators of LH action in the follicle. Specifically, amphiregulin (AREG), epiregulin (EREG), and betacellulin (BTC) are rapidly induced by LH or its analog human chorionic gonadotropin (hCG) (5–7) and are thought to function in an autocrine and paracrine manner to propagate LH signals throughout the preovulatory follicle. In vitro, these growth factors promote many events that are stimulated by LH, including cumulus expansion and oocyte meiotic resumption (5, 6).

HE LH SURGE PROMOTES major changes in ovarian preovulatory follicles (POFs), including terminal differentiation of follicular cells and oocyte maturation, events required for ovulation of a fertilizable egg. The LH effects are mediated via activation of a member of the G-protein-coupled receptor (GPCR) superfamily, the LH receptor (LHR), which couples to

First Published Online January 10, 2008 * S.P. and M.H. contributed equally to this work. Abbreviations: ADAM, A disintegrin and metalloprotease; AREG, amphiregulin; BTC, betacellulin; COC, cumulus-oocyte complex; CREB, cAMP-response element-binding protein; DMSO, dimethylsulfoxide; DPBS, Dulbecco’s PBS; EGF, epidermal growth factor; EGFR, EGF receptor; EREG, epiregulin; FBS, fetal bovine serum; Fsk, forskolin; GPCR, Gprotein-coupled receptor; GVBD, germinal vesicle breakdown; hCG, human chorionic gonadotropin; HRP, horseradish peroxidase; LHR, LH receptor; PKA, protein kinase A; PMSG, pregnant mare serum gonadotropin; POF, preovulatory follicle; rLH, recombinant LH; TBST, Tris-buffered saline with 0.1% Tween 20. Molecular Endocrinology is published monthly by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the endocrine community. 924

Panigone et al. • Early Activation of EGFR by LH in Follicles

The growth factor effects require EGF receptor (EGFR, also called ErbB1) activity, because EGFR tyrosine kinase inhibitors block both processes (5, 6), as well as several other gonadotropin-dependent functions of granulosa cells (8–11). Recently, we reported that disruption of the EGF signaling network in mice impairs LH-induced ovulation, demonstrating the importance of this pathway to LH action also in vivo (12). LHinduced EGFR phosphorylation was significantly reduced in POFs from Areg⫺/⫺ Egfrwa2/wa2 double-mutant mice. This decrease in LH-induced EGFR phosphorylation was associated with impaired oocyte meiotic resumption in hCG-primed Areg⫺/⫺ Egfrwa2/wa2 mouse ovaries in vivo. The EGF-like growth factors are produced as transmembrane precursors that are cleaved at the cell surface by members of the matrix metalloprotease (MMP) or a disintegrin and metalloprotease (ADAM) families (13, 14). The mature growth factors bind to dimers of EGFR family members, resulting in activation of intrinsic receptor tyrosine kinase activity and autophosphorylation of specific tyrosine residues in the cytoplasmic domain. Target proteins are subsequently phosphorylated and/or recruited for the activation of downstream signaling cascades, including the MAPK pathway, to elicit distinct biological effects. GPCRs also can activate the MAPK cascade via G␤␥-c-Src (15) or effectors such as phosphatidylinositide-3-kinase or PKC (16, 17). In addition, GPCR-induced transactivation of the EGF signaling network has been linked to activation of the MAPK cascade (18, 19). Early studies described an intracellular mechanism for GPCR-mediated EGFR transactivation, involving the intracellular mediators Src, PKC, or Ca2⫹ (20–26). However, later findings have also documented a ligand-dependent mechanism of EGFR transactivation. In brief, agonist-stimulated GPCR signaling has been shown to induce metalloproteasedependent shedding of EGF-like growth factors and EGFR activation (27, 28). This mechanism of GPCRinduced EGFR transactivation has been described as the triple-membrane-passing signal (27). Because the exact timing of EGFR transactivation in relationship to oocyte reentry into the cell cycle has not been established, we sought to investigate the dynamics of LH regulation of the EGF network in cultured mouse POFs. Here, we show that LH induces EGFR phosphorylation within 30 min of stimulation. Based on this observation, we hypothesized that the early activation of EGFR induced by LH may involve shedding of a preexisting or rapidly formed pool of EGF-like growth factors. We provide evidence that the early LH-induced EGFR transactivation is likely mediated by cAMP and PKA signaling, involves metalloprotease activity, and impacts the MAPK cascade. This rapid activation of the EGF network serves as a means for amplifying the initial LH signal, and propagating LH effects throughout the POF.

Mol Endocrinol, April 2008, 22(4):924–936

925

RESULTS Rapid Induction of EGFR Phosphorylation and Areg and Ereg Expression by Recombinant LH (rLH) in Mouse POFs LH/hCG has previously been shown to induce EGFR phosphorylation in mouse ovaries in vivo (5). To more closely investigate the time course of LH-induced EGFR phosphorylation specifically in POFs, a better controlled model of POF culture was used. POFs were cultured in the presence of rLH for 0, 0.5, 1, 2, and 4 h. Immunoprecipitation and Western blot analyses were performed to evaluate EGFR phosphorylation levels in the stimulated follicles. The anti-phospho-EGFR antibody used recognizes the Y1068 residue, a major autophosphorylation site that binds Grb2 and that is involved in MAPK activation (29–31). EGFR phosphorylation levels increased about 3-fold over control within 30 min of rLH stimulation, and phosphorylation levels were maximal after 2 h (Fig. 1A). The rLH-induced expression of Areg and Ereg mRNAs was similarly evaluated in mouse POFs cultured for the same times as above. By semiquantitative RT-PCR analysis, statistically significant increases in both Areg and Ereg mRNAs were detected within 30 min after rLH stimulation (Fig. 1B). Expression of both genes was maximal after 2 h stimulation and decreased at 4 h. This pattern of LH-induced Areg and Ereg expression in POFs is consistent with the results of earlier studies showing that LH/hCG induces the rapid and transient expression of Areg and Ereg mRNAs in wild-type mouse ovaries (5) and rat follicles (6) within 3 h of stimulation. Interestingly, the early induction of Areg and Ereg expression and EGFR activation by LH precedes the onset of oocyte meiotic resumption by at least 1 h (Fig. 1C). Protein Synthesis Is Required for LH-Induced Oocyte Maturation If de novo EGF-like protein synthesis were required for LH transactivation of EGFR and oocyte maturation, then inhibition of protein synthesis should block the LH- but not AREG-mediated effects. POFs were incubated in the presence of either protein synthesis inhibitor puromycin or cycloheximide before stimulation by either rLH or AREG. After 4 h, oocytes were examined for germinal vesicle breakdown (GVBD), a hallmark of oocyte meiotic resumption. In this follicle culture model, rLH or AREG stimulation resulted in approximately 80–90% GVBD (Fig. 2A). In agreement with previous reports, puromycin concentrations that decrease amino acid incorporation by more than 90% (see Materials and Methods) significantly inhibited rLH-induced GVBD in cultured follicles (⬃70% inhibition), indicating that protein synthesis is necessary for the LH effect. Conversely, inhibition of AREG-induced GVBD was minimal (Fig. 2A). Treatment with a different

926 Mol Endocrinol, April 2008, 22(4):924–936

A

Panigone et al. • Early Activation of EGFR by LH in Follicles

pEGFR EGFR rLH (h):

0

0.5

1

2

4

pEGFR/EGFR (fold Induction)

8 6

*

4 2 0 0

Relative Expression (% maximum)

B

1

2

3

4

5

LH-Induced MAPK1/3 Activation Is Dependent in Part on EGFR Activity

100 80 60 5

*

Cont

4

40

rLH 0.5h

3

*

2

20

1 0 Areg

0

C

Ereg

0

1

2

3

4

5

0

1

2

3

4

5

100

%GVBD

translation inhibitor, cycloheximide, alone promoted oocyte meiotic resumption (data not shown), confounding interpretation of the experimental results. The effect of this latter inhibitor was not further investigated. More importantly, puromycin also inhibited the LH stimulation of EGFR phosphorylation. After 2 h culture of POFs, rLH-induced EGFR phosphorylation was significantly inhibited by puromycin at the 10 ␮g/ml concentration and was completely prevented by 50 ␮g/ml (Fig. 2B). Puromycin had no significant effect on AREG-stimulated EGFR phosphorylation even at high doses. These data are consistent with the view that LH-induced synthesis of AREG (and/or additional EGF-like growth factors) and EGFR activation are required for oocyte meiotic resumption.

75 50 25 0

Hours after rLH Fig. 1. Time Course of Gene Expression, EGFR Activation, and Oocyte Meiotic Resumption in POFs A, Time course of EGFR phosphorylation in POFs. EGFR was immunoprecipitated from extracts of POFs cultured in the presence of rLH (5 IU) for 0, 0.5, 1, 2, and 4 h. Western blot analyses were performed to detect phosphorylated EGFR and EGFR proteins. The ratio of phosphorylated EGFR to total EGFR was determined for each time point, and the relative values are expressed as fold induction over control. Representative blots are shown. Data are the mean ⫾ SEM of at least three separate experiments. *, P ⬍ 0.05 compared with control (0 h) time point. B, Time course of Areg and Ereg mRNA expression in POFs. The induction of Areg (f) and Ereg (Œ) mRNAs by rLH (5 IU) was examined in cultured POFs by semiquantitative RT-PCR. Gene expression levels were normalized to Rpl19 levels at each time point and are expressed as percentage of maximal induction. Data shown are the mean ⫾ SEM of three separate experiments. Inset shows Areg and Ereg expression levels in control (Cont) vs. 0.5-h rLH-stimulated follicles. *, P ⬍ 0.05. C, Time course of GVBD

One of the pathways downstream of the EGFR activation implicated in oocyte meiotic resumption is the MAPK signaling cascade. Therefore, the time course of MAPK1/3 (also called ERK1/2) phosphorylation was examined in cultured follicles by Western blot analyses with phospho-ERK1/2-specific antibodies. In POFs, an increase in MAPK phosphorylation was induced by rLH within 30 min, and phosphorylation was maximal after 2 h of culture (Fig. 3A). Increased MAPK phosphorylation was also detected in cumulus-oocyte complexes (COCs) isolated from POFs 30 min after LH stimulation (data not shown), consistent with previous findings showing MAPK activation in cumulus cells isolated 30 min after hCG stimulation in mice, with maximal activation 2–4 h after hCG (32). Immunohistochemical staining was performed on pregnant mare serum gonadotropin (PMSG)-primed mouse ovaries stimulated with hCG for different times to localize phosphorylated MAPK in vivo. Signal for phosphorylated MAPK was detected predominantly in mural granulosa cells of large antral follicles 15–30 min after hCG and then was observed in both mural granulosa and cumulus cell compartments of POFs 1 and 2 h after hCG (Fig. 3B). After 4 h hCG stimulation, signal for phosphorylated MAPK remained elevated in cumulus cells but appeared reduced in mural granulosa cells. To understand the mechanisms underlying the rapid propagation of the LH signal within ovarian POFs, the involvement of LH-induced EGFR activation in the early activation of MAPK was investigated. When POFs were preincubated with the EGFR tyrosine ki-

in POFs. POFs (20–30 follicles per group) were cultured in the presence of rLH (5 IU) and punctured at the indicated times to release COCs. Oocytes were denuded of cumulus cells and evaluated for evidence of GVBD, a hallmark of oocyte meiotic resumption. Data are expressed as the mean percent GVBD ⫾ SEM of at least four separate experiments.

Panigone et al. • Early Activation of EGFR by LH in Follicles

A

** *

67

100

73

%GVBD

75

69

50 67 25 137

136

A 10 R EG PU R +A R EG

rL 10 H PU R +r LH

C on tr ol 10 PU R

0

B pEGFR

pEGFR/EGFR (fold induction)

EGFR

Mol Endocrinol, April 2008, 22(4):924–936

927

nase inhibitor AG1478, rLH-induced EGFR phosphorylation was significantly inhibited after 30 min (Fig. 4, A and E), demonstrating the effectiveness of this concentration of AG1478 (0.5 ␮M) in blocking the LHinduced EGFR tyrosine kinase activity. Next, activation of the downstream MAPK signaling cascade was examined. As shown in Fig. 3A, rLH stimulated an increase in MAPK1/3 phosphorylation in cultured POFs within 30 min (Fig. 4B). However, pretreatment of POFs with AG1478 resulted in approximately 60% inhibition of LH-induced MAPK1/3 phosphorylation (Fig. 4, B and E). In addition, when MAPK phosphorylation was examined in POFs from wild-type, Areg⫹/⫹ Egfrwa2/wa2, and Areg⫺/⫺ Egfrwa2/wa2 mice cultured without or with rLH for 2 h, rLH-induced MAPK activation was significantly reduced in the Areg⫺/⫺ Egfrwa2/wa2 follicles but not completely prevented (Fig. 5). Taken together, these results suggest that regulation of the MAPK signaling pathway is dependent in part on EGFR activity but likely involves additional LH-regulated intracellular pathways.

75

LH-Induced EGFR Signaling Requires Matrix Metalloprotease Activity

50

EGFR transactivation occurs either by binding of shed EGF-like growth factors or by an intracellular mechanism. To determine whether EGF-like growth factor shedding is involved in LH-induced EGFR transactivation, POFs were pretreated with the broad spectrum metalloprotease inhibitor GM6001 before rLH stimulation. GM6001 completely inhibited rLH-induced EGFR phosphorylation after 30 min stimulation (Fig. 4, C and E). Similarly, preincubation of POFs in the presence of TAPI-1, a metalloprotease inhibitor with some specificity for TNF␣ converting enzyme/ADAM17, also blocked rLH-induced EGFR phosphorylation (data not shown). Consistent with their effects on EGFR activation, GM6001 and TAPI-1 inhibited LH-induced MAPK1/3 phosphorylation by about 50% (Fig. 4, D and E, and data not shown). In addition, GM6001 and TAPI-1 each inhibited LH-induced oocyte meiotic resumption in cultured follicles by at least 66%; in contrast, GM6001 failed to inhibit oocyte maturation induced by EREG (data not shown). Thus, metalloprotease activity involved in EGF-like growth factor shedding is required for EGFR activation and signaling.

*

25

C on

tr

o 10 l PU R 10 r PU LH R +r LH 10 PU AR R EG +A R E 50 G 50 PU P 50 U R R PU + R rLH +A R EG

0

Fig. 2. Effect of the Protein Synthesis Inhibitor Puromycin on rLH- and AREG-Induced GVBD and EGFR Phosphorylation in Cultured Follicles A, Effect of puromycin on rLH-and AREG-induced oocyte maturation. POFs were preincubated with vehicle or puromycin (10 ␮g/ml; 10PUR) for 30 min, followed by culture in the absence or presence of rLH (5 IU) or AREG (100 nM) for 4 h. After release from follicles, oocytes were denuded of cumulus cells for evaluation of GVBD. The total number of oocytes scored per treatment group is shown above each bar. Data are the mean percent GVBD ⫾ SEM of four to eight separate experiments. *, P ⬍ 0.05; **, P ⬍ 0.01. B, Effect of puromycin on rLH- and AREG-induced EGFR phosphorylation. POFs were preincubated with vehicle or puromycin (10 or 50 ␮g/ml) for 30 min and then cultured in the absence or presence of rLH (5 IU) or AREG (100 nM) for 2 h. EGFR was immunoprecipitated from protein extracts of POFs, and Western blot analyses were performed to detect phosphorylated EGFR and EGFR proteins. The ratio of phosphorylated EGFR to total EGFR was determined for each time point, and the relative values are expressed as fold induction over control. Results for the rLH and 10PUR ⫹ rLH groups are expressed as the mean ⫾ SEM of three separate experiments. *, P ⬍ 0.05. Except control, values for the other treatment groups are from experiments performed fewer than three times. Representative blots are shown.

LH-Mediated EGFR Transactivation Involves cAMP and PKA Signaling To determine whether EGFR transactivation induced by LH is mediated by cAMP signaling, POFs were stimulated with forskolin (Fsk), a direct activator of adenylyl cyclase. Fsk promoted an increase in EGFR phosphorylation to levels comparable to those observed with rLH after 30 min (Fig. 6A). In addition, Fsk-induced EGFR phosphorylation was completely inhibited by both AG1478 and GM6001, suggesting

928 Mol Endocrinol, April 2008, 22(4):924–936

A

*

pMAPK/MAPK (fold induction)

1 2 .5

Panigone et al. • Early Activation of EGFR by LH in Follicles

B

**

PMSG

1 0 .0

*** 7 .5 5 .0

15’ hCG 2 .5 0 .0

pMAPK MAPK 0

0.5

1

2

4

30’ hCG

Hours after rLH

1h hCG

2h hCG

4h hCG

Fig. 3. Time Course of MAPK Phosphorylation in Vitro and in Vivo A, Time course of MAPK activation in POFs. MAPK phosphorylation was examined by Western blot analyses using protein extracts from POFs cultured in the presence of rLH (5 IU) for 0, 0.5, 1, 2, and 4 h. The ratio of phosphorylated MAPK to total MAPK was determined for each time point, and the relative values are expressed as fold induction over control. Representative blots are provided. Data are shown as the mean ⫾ SEM of five separate experiments. *, P ⬍ 0.05; **, P ⬍ 0.01; ***, P ⬍ 0.001. B, Immunolocalization of activated MAPK in mouse ovaries. Phosphorylated MAPK was localized in hormone-primed ovaries by immunohistochemical analyses using an anti-phospho-p44/42 MAPK antibody. No signal above background was observed in PMSG-primed mouse ovaries. After 15–30 min hCG stimulation, staining for phosphorylated MAPK was detected predominantly in mural granulosa cells of POFs. At 1 h hCG, signal was evident in both mural granulosa and cumulus cell compartments and remained elevated after 2 h. Theca cells of preantral and antral follicles were also positive at these times. After 4 h, signal was still detected in cumulus cells and appeared decreased in granulosa cells of POFs.

Panigone et al. • Early Activation of EGFR by LH in Follicles

Mol Endocrinol, April 2008, 22(4):924–936

B 5

pMAPK/MAPK (fold induction)

*

4 3 2 1 0

+

+ -

5 4 3 2 1

5

rLH (0.5h) AG1478

D

4 3 2 1

-

+

+ -

+ +

5

* 4

3

2

1

-

+

+ -

+ +

-

+

+ -

1

rLH (0.5h) GM6001

H

0

0

1

pEGFR/EGFR (fold induction)

+ +

*

C

rLH (0.5h) GM6001

6

0

-

pMAPK/MAPK (fold induction)

rLH (0.5h) AG1478

*

7

r

pEGFR/EGFR (fold induction)

A

929

+ +

E pEGFR EGFR pMAPK MAPK Cont AG GM

rLH rLH rLH +AG +GM

Fig. 4. LH-Induced EGFR Phosphorylation and MAPK Activation Is Impaired by the Inhibitors AG1478 and GM6001 A, Inhibition of LH-induced EGFR phosphorylation by the EGFR tyrosine kinase inhibitor AG1478. POFs were preincubated with vehicle (DMSO) or AG1478 (0.5 ␮M) for 30 min, followed by culture in the presence of rLH (5 IU) for 30 min. EGFR protein was immunoprecipitated from POF extracts, and Western blot analyses were performed to detect phosphorylated and nonphosphorylated EGFR. The ratio of phosphorylated EGFR to total EGFR was normalized to control. Results are expressed as the mean ⫾ SEM of four separate experiments. *, P ⬍ 0.05. B, Inhibition of LH-induced MAPK activation by AG1478. LH-stimulated MAPK phosphorylation levels were examined by Western blot analyses in cell lysates of POFs that were preincubated with DMSO or AG1478 (0.5 ␮M) for 30 min, followed by stimulation with rLH (5 IU) for another 30 min. The ratio of phosphorylated MAPK to total MAPK was normalized to control. Results are expressed as the mean ⫾ SEM of four separate experiments. *, P ⬍ 0.05. C, Inhibition of LH-induced EGFR phosphorylation by the metalloprotease inhibitor GM6001. Mouse POFs were incubated with DMSO or GM6001 (20 ␮M) for 30 min, followed by culture for 30 min in the presence of rLH (5 IU). EGFR protein was immunoprecipitated from POF extracts, and Western blot analyses were performed to detect phosphorylated and nonphosphorylated EGFR. Data are shown as the mean ⫾ SEM of six separate experiments. *, P ⬍ 0.05. D, Inhibition of LH-induced MAPK activation by GM6001. LH-stimulated MAPK phosphorylation was analyzed by Western blot using protein extracts from POFs that were preincubated with DMSO or GM6001 (20 ␮M) for 30 min, followed by culture with rLH (5 IU) for another 30 min. Results are shown as the mean ⫾ SEM of four separate experiments. *, P ⬍ 0.005. E, Representative blots are shown for phosphorylated and nonphosphorylated EGFR and MAPK after treatment without or with the inhibitors AG1478 or GM6001 and rLH. AG, AG1478; Cont, control; GM, GM6001.

930 Mol Endocrinol, April 2008, 22(4):924–936

C ontrol rLH 2h

1.0

Relative Induction

Panigone et al. • Early Activation of EGFR by LH in Follicles

*

0.8 0.6 0.4 0.2 0.0

Areg+/+ Egfr+/+

Areg+/+ Areg-/wa2/wa2 Egfr Egfrwa2/wa2

Fig. 5. LH-Induced MAPK Activation Is Reduced in Areg⫺/⫺ Egfrwa2/wa2 Preovulatory Follicles LH-induced MAPK phosphorylation was analyzed by Western blot using proteins extracted from wild-type (Areg⫹/⫹ Egfr⫹/⫹), Areg⫹/⫹ Egfrwa2/wa2, and Areg⫺/⫺ Egfrwa2/wa2 POFs that were cultured in the absence or presence of 5 IU rLH for 2 h. The ratios of phosphorylated MAPK to total MAPK levels were normalized to the wild-type level after 2 h rLH stimulation. Data represent the mean ⫾ SEM of five separate experiments. *, P ⬍ 0.001.

that the cAMP cascade lies upstream of both matrix metalloprotease activity and EGFR transactivation. The inhibitors tested could exert their effect by interfering with the LH activation of cAMP signaling. This possibility was investigated by following the phosphorylation of cAMP-response element-binding protein (CREB), a downstream target of PKA. In POFs cultured under the conditions used above, LH induced phosphorylation of the transcription factor CREB within 30 min, and the inhibitor AG1478, GM6001, or TAPI-1 had no effect on this phosphorylation (Fig. 6B). Thus, LH activation of cAMP signaling is not impaired by these inhibitors under the conditions used, and CREB is not a downstream target of the EGFR signaling pathway. The involvement of PKA downstream of cAMP in LH-induced EGFR transactivation was examined by incubation with the PKA inhibitor H89. When POFs were exposed to H89, rLH-stimulated EGFR phosphorylation was significantly inhibited (Fig. 6C), indicating a role for PKA in LH-stimulated EGFR transactivation. EGF-Like Growth Factors Implicated in Early LH Signaling In an independent strategy to define the involvement of EGF-like growth factors in the rapid LH-induced EGFR transactivation and activation of downstream signaling, POFs were cultured in the presence of neutralizing antibodies against AREG, EREG, and BTC for 30 min before rLH stimulation. A combination of the three antibodies was used based on the likely redun-

dant functions of these growth factors (12). As a control, POFs were cultured in the presence of mouse IgG1. In this experimental paradigm, no effect of the neutralizing antibodies on GVBD was observed after 4 h, possibly due to an inefficient ability of the antibodies to concentrate in the follicle (data not shown). To circumvent this problem, the effects of the three neutralizing antibodies on LH-induced EGFR phosphorylation was examined in cultured granulosa cells. After 30 min preincubation with the antibodies, cells were stimulated with vehicle or Fsk for 30 min. Fsk stimulated EGFR phosphorylation, and this effect was completely inhibited by the neutralizing antibodies but not by the nonspecific IgG1 antibody (Fig. 7A). Fskinduced MAPK1/3 phosphorylation was also significantly inhibited in granulosa cells preincubated with the combination of AREG, EREG, and BTC neutralizing antibodies (Fig. 7B). Taken together, the results suggest that one or more of the EGF-like growth factors are involved in the early EGFR phosphorylation and MAPK activation that occur downstream of LH and cAMP signaling in vivo.

DISCUSSION It is established that LH regulation of granulosa cells requires activation of the cAMP signaling pathway as well as modification and amplification of the primary signal by the recruitment of additional signaling modules. Among the signaling cascades activated in addition to PKA are the PKB/AKT, PKC, and MAPK pathways (1, 2). Activation of these additional pathways generally occurs through intracellular routes, often through cascades of phosphorylations, and is required for granulosa cell differentiation and ovulation. Here we describe a distinct mode by which the LH-dependent cAMP signal leads to activation of a tyrosine kinase-mediated pathway. LH induces rapid transactivation of the EGFR in cultured follicles through de novo synthesis of EGF-like growth factors and through metalloprotease-mediated shedding of these ligands. In concert with other pathways, EGFR signaling regulates MAPK activation in granulosa and cumulus cells to mediate oocyte maturation. Using the in vitro POF model, we establish that LH induces the rapid expression and activation of components of the EGF network in follicular cells. LH or Fsk induced EGFR phosphorylation as early as 30 min after stimulation. Although small compared with the large increase in EGFR phosphorylation that occurs after 2–4 h exposure to LH, this early increase is significant. This transactivation of EGFR occurs at the same time that small but significant increases in Areg and Ereg mRNAs are detected by RT-PCR under the same culture conditions. Previous studies have also shown Fsk stimulation of Areg mRNA in COCs (33) and Areg and Ereg mRNAs in human granulosa cells (34). We also show that LH- and Fsk-induced EGFR trans-

Panigone et al. • Early Activation of EGFR by LH in Follicles

A

pEGFR EGFR

-

rLH (0.5h) Fsk (0.5h) AG1478 GM6001

B

+ -

+ -

+ + -

+ +

pCREB CREB

rLH (0.5h) AG1478 GM6001

-

+ -

+

+ -

+ + -

+ +

-

+ -

+

+ -

+ + -

+ +

pCREB CREB rLH (0.5h) GM6001 TAPI-1

*

C Relative Induction

4 3 2 1 0

pEGFR EGFR rLH (0.5h) H89

-

+

+ -

+ +

Fig. 6. Activation of cAMP Signaling Is Involved in EGFR Transactivation by LH A, Fsk stimulation of EGFR phosphorylation in POFs. POFs were preincubated for 30 min in the absence or presence of DMSO, AG1478 (0.5 ␮M), or GM6001 (20 ␮M) followed by 30 min culture in the presence of rLH (5 IU) or Fsk (50 ␮M). EGFR was immunoprecipitated from POF extracts, and Western blot analyses were performed. Fsk was as effective as rLH in promoting EGFR phosphorylation. AG1478 and GM6001 each inhibited Fsk-induced EGFR phosphorylation. B, LHinduced CREB phosphorylation. POFs were preincubated for 30 min in the absence or presence of DMSO, AG1478 (0.5 ␮M), GM6001 (20 ␮M), or TAPI-1 (30 ␮M) followed by culture in the presence of rLH (5 IU) for another 30 min. Western blot analyses were performed to detect phosphorylated and nonphosphorylated CREB protein. The inhibitors AG1478, GM6001, and TAPI-1 had no effect on LH-stimulated CREB phosphorylation. C, LH-induced EGFR phosphorylation involves PKA. POFs were preincubated for 30 min with DMSO

Mol Endocrinol, April 2008, 22(4):924–936

931

activation is sensitive to metalloprotease inhibitors, suggesting the involvement of cAMP signaling and a requirement for shed EGF-like growth factors. Indeed, Fsk-induced EGFR phosphorylation was prevented in cultured granulosa cells by neutralizing antibodies against the AREG, EREG, and BTC growth factors. Given the rapid phosphorylation of one of the major EGFR autophosphorylation sites (Y1068), these results argue for a ligand-dependent rather than an intracellular-dependent mechanism of EGFR transactivation. Although the intracellular mediator Src has been reported to phosphorylate the EGFR autophosphorylation sites in addition to distinct residues (35), LH-induced phosphorylation of the Y1068 residue is blocked when EGF-like growth factor processing or availability is inhibited. When POFs were cultured in the presence of a protein synthesis inhibitor, LH-induced but not AREGinduced EGFR phosphorylation was inhibited. In a similar fashion, LH-induced but not AREG-induced oocyte maturation was prevented. A likely explanation of this finding is that LH action requires de novo synthesis of EGF-like growth factors. That EGFR phosphorylation is indispensable for LH-stimulated oocyte maturation is indicated by pharmacological inhibition with the different inhibitors used in this study and by the analyses of oocyte maturation in follicles with genetic disruptions in the EGF network (12). In both cases, a decrease or absence of EGFR phosphorylation was associated with a failure of LH to induce oocyte maturation. That LH still produces a robust signal under these conditions is indicated by the effect of LH on CREB phosphorylation and MAPK activation. Based on the above findings, we propose that LH signaling branches to activation of the EGF network through complex mechanisms during the ovulatory process. Initially, transactivation occurs either through very rapidly synthesized growth factors or mobilization of a small preformed pool of EGF-like growth factors that may not depend on de novo synthesis. This pool could not be detected by fluorescence-activated cell sorting analysis of granulosa cells before exposure to LH (5). This initial small activation is then greatly amplified and stabilized by the de novo synthesis of EGFlike growth factors as we have previously described. With this study, we also show that LH-induced EGFR transactivation impacts the MAPK cascade, consistent with the previous observation that AREG induces MAPK phosphorylation in COCs (33). LH-induced MAPK activation is diminished but not abolished when EGFR phosphorylation is blocked either by an EGFR tyrosine kinase inhibitor or inhibitors of

or H89 (20 ␮M) and then stimulated by rLH (5 IU) for 30 min. EGFR was immunoprecipitated from POF extracts and Western blot analyses performed. The ratio of phosphorylated EGFR to total EGFR was normalized to control. Results are shown as the mean ⫾ SEM of three separate experiments and representative blots. *, P ⬍ 0.05.

A

pEGFR/EGFR (fold induction)

932 Mol Endocrinol, April 2008, 22(4):924–936

*

3 .0 2 .5 2 .0 1 .5 1 .0 0 .5

G

k

l

0 .0

pEGFR EGFR

B

pMAPK/MAPK (fold induction)

Fsk (0.5h) nAb IgG1

-

+ -

5

+ + -

+ +

+ + -

+ +

*

4 3 2 1 0

pMAPK MAPK Fsk (0.5h) nAb IgG1

-

+ -

Fig. 7. Fsk-Induced EGFR Phosphorylation and MAPK Activation Is Inhibited by AREG, EREG, and BTC Neutralizing Antibodies in Cultured Granulosa Cells A, Inhibition of Fsk-induced EGFR phosphorylation by EGFlike growth factor neutralizing antibodies. Granulosa cells were serum starved before culture for 30 min in the presence of a combination of mouse neutralizing antibodies against AREG (3 ␮g/ml), EREG (2.5 ␮g/ml), and BTC (2 ␮g/ml) or identical concentration of mouse anti-IgG antibody as a control. Cells were next stimulated with Fsk (10 ␮M) or vehicle (DMSO) for an additional 30 min. EGFR was immunoprecipitated from granulosa cell extracts, and Western blot analyses were performed to detect phosphorylated and nonphosphorylated EGFR. Data shown are the mean ⫾ SEM of four separate experiments. *, P ⬍ 0.05. B, Inhibition of Fsk-induced MAPK activation by EGF-like growth factor neutralizing antibodies. Fsk-stimulated MAPK phosphorylation levels were examined by Western blot analyses in extracts of granulosa cells cultured for 30 min in the absence or presence of the three neutralizing antibodies against AREG, EREG, and BTC or mouse anti-IgG antibody as a control followed by 30 min stimulation with Fsk (10 ␮M) or vehicle (DMSO). Data are shown as the mean ⫾ SEM of five separate experiments. *, P ⬍ 0.05. Representative blots are shown for each experiment.

EGF-like growth factor shedding or action. The finding that substantial MAPK activation is observed in Areg⫺/⫺ Egfrwa2/wa2 follicles is consistent with the

Panigone et al. • Early Activation of EGFR by LH in Follicles

pharmacological data. The incomplete inhibition of MAPK activity in the absence of EGFR phosphorylation suggests that additional pathways contribute to the LH-mediated activation of MAPK in the POF. These may be intracellular pathways that may be directly downstream of cAMP. It is possible that additional mechanisms are also operating to regulate MAPK activity at times earlier than 30 min LH stimulation. Regardless of these mechanisms, both the pharmacological and genetic manipulations define an experimental paradigm whereby oocyte maturation does not occur when cAMP accumulation is induced by LH (12) and when MAPK activation is still substantial in the follicle. This finding suggests that neither cAMP nor MAPK activation by itself is sufficient to signal oocyte maturation unless the EGF network is activated. This observation is consistent with reports showing that MAPK is necessary but not sufficient to induce oocyte maturation. MAPK kinase inhibitors prevent LH-induced oocyte maturation in follicles or FSH-induced maturation in COCs (32, 36, 37); however, activation of MAPK in cumulus cells by recombinant growth and differentiation factor 9 alone or in combination with FSH in the presence of hypoxanthine fails to promote oocyte maturation in COCs (38). Thus, synergism between several pathways yet to be identified is likely required. All the pharmacological or genetic manipulations that interfere with activation of the EGF network prevent LH-mediated induction of oocyte meiotic resumption. These observations reinforce our hypothesis that transactivation of the EGFR mediates the signal that induces oocyte meiotic resumption. The detailed time course of EGFR phosphorylation documents that the timing of this activation is compatible with the proposed mediatory role in downstream events. The EGFR phosphorylation observed after 30–60 min of LH stimulation precedes oocyte GVBD by at least 2 h. This time interval is identical to the time required for EGF-like growth factors to induce maturation in the POF in vitro (5). Assuming that the time required for the oocyte to process the maturation stimulus into GVBD requires 60 min, as inferred from spontaneous maturation, there is an interval of 1 h between the EGFR phosphorylation and the trigger of the signal for oocyte maturation. Some of this time may be required to propagate the signal from mural to cumulus cells and for amplification of the signal. However, the exact steps that are activated during this window of time in somatic cells and that are required to signal oocyte reentry into the cell cycle remain elusive. Connexin phosphorylation by PKA or MAPK and closure of gap junction communication between the somatic cells and the oocyte is a possible candidate step (39). This closure should lead to a decrease in the diffusion of any inhibitory signals. However, MAPK is still substantially activated when EGFR phosphorylation is prevented, and LH-mediated activation of PKA still occurs in view of the phosphorylation of CREB. Under these conditions, oocyte maturation is not induced.

Panigone et al. • Early Activation of EGFR by LH in Follicles

Pending additional experiments directly probing the junctional permeability in follicles where EGFR activation is prevented, connexin phosphorylation should still occur in view of the increase in cAMP and MAPK activation. It is possible that additional signals downstream of EGFR phosphorylation are indispensable for meiotic maturation. Indeed, EGFR activation produces signaling through a plethora of cascades that include activation of the Ras-MAPK, c-Jun N-terminal kinase (JNK), Src, phosphatidylinositide-3-kinase, phospholipase C, signal transducer and activator of transcription (STAT), and Cbl pathways, as well as regulation of cytoskeleton modification, cell adhesion, and lipid mediator production (14, 40, 41). Thus, each one or a combination of these pathways may be involved in the final signals for oocyte meiotic resumption. In conclusion, we demonstrate the rapid transactivation of the EGF network by LH in POFs that likely serves to amplify the initial LH signal, thus propagating the LH effects within the follicle. This early transactivation of the EGF network may be a critical signal for oocyte maturation. These findings provide new insight into the mechanisms regulating oocyte meiotic resumption and have important implications for improving the in vitro maturation of oocytes.

Mol Endocrinol, April 2008, 22(4):924–936

933

mice were injected ip with an ovulatory dose of 5 IU hCG, and ovaries were excised at selected times after injection. Areg⫹/⫹ Egfr⫹/⫹, Areg⫹/⫹ Egfrwa2/wa2, and Areg⫺/⫺ Egfrwa2/wa2 mice were generated and genotyped as previously described (12). All animal procedures were approved and followed the guidelines of the Stanford University Administrative Panel on Laboratory Animal Care. RNA Extraction and Semiquantitative RT-PCR

MATERIALS AND METHODS

Total RNA was extracted from POFs cultured in the presence of 5 IU rLH for 0, 0.5, 1, 2, or 4 h using Trizol Reagent (Invitrogen) as instructed by the manufacturer. Total RNA (100 ng) was reverse-transcribed using 1⫻ Thermocycle buffer, 3.75 mM MgCl2, 1 mM dNTPs (Promega Corp., Madison, WI), 500 ng poly-dT (Amersham Pharmacia, Newark, NJ), and 2.5 U avian myeloblastosis virus-reverse transcriptase (Promega) at 42 C for 75 min and 95 C for 5 min. Afterward, one fourth of each RT reaction was used for PCR amplification of the genes of interest, and 0.1 ␮M of each primer, 0.25 ␮Ci [32P]dCTP (PerkinElmer, Boston, MA), 0.625 U Taq polymerase in 1⫻ Thermocycle buffer, and 2 mM MgCl2 (Promega Corp.) were used in each reaction. PCR conditions were 94 C for 2 min followed by cycles of 94 C for 30 sec, 60 C for 45 sec, and 72 C for 1 min and a final extension at 72 C for 7 min. Specific primers used were as follows: Areg forward, 5⬘-CACAGCGAGGATGACAAGGACC3⬘; Areg reverse, 5⬘-AATGTCATTTCCGGTGTGGCTTGG-3⬘; Ereg forward, 5⬘-CATGGAGTCTGACTCTAGTC-3⬘; Ereg reverse, 5⬘-TCAGTGACTCACGAAGACCT-3⬘; and L19 (42, 43). The amplified PCR products were resolved on 5% polyacrylamide gels. The gels were dried, and radioactive bands were quantified using the Storm 840 PhosphorImager and ImageQuant software (Molecular Dynamics, Inc., Sunnyvale, CA).

Materials

POF Culture

The rLH (Luveris) was purchased from Serono International (Rockland, MA). PMSG, AG1478, H89, Fsk, and diphtheria toxin CRM mutant were purchased from Calbiochem (San Diego, CA), hCG and puromycin dihydrochloride from Sigma Chemical Co. (St. Louis, MO), and TAPI-1 and GM6001 (Galardin) from Biomol (Plymouth Meeting, PA). Rec-Protein GSepharose 4B was obtained from Zymed Laboratories (South San Francisco, CA). Complete, mini EDTA-free protease inhibitor cocktail tablets were purchased from Roche Applied Science (Indianapolis, IN). ECL and ECL Plus Western Blotting detection reagents were from Amersham Biosciences (Piscataway, NJ). Sheep anti-EGFR and anti-phospho-CREB (Ser133) antibodies were purchased from Upstate (Charlottesville, VA); anti-pan-ERK and anti-ERK1 from BD Biosciences (San Jose, CA); anti-pY1068-EGFR, anti-phosphoMAPK (p44/42), and anti-CREB from Cell Signaling Technology (Danvers, MA); and rabbit polyclonal anti-EGFR from Santa Cruz Biotechnology (Santa Cruz, CA). Antimouse IgG1 and antimouse AREG, antimouse EREG, and antimouse BTC neutralizing antibodies were purchased from R&D Systems (Minneapolis, MN). Leibowitz’s L15 medium, MEM, DMEM/F12, methionine-free DMEM, Dulbecco’s PBS (DPBS) without Ca2⫹ and Mg2⫹, and fetal bovine serum (FBS), were obtained from GIBCO (Carlsbad, CA). Vectastain ABC kit and diaminobenzidine chromogen were from Vector Laboratories (Burlingame, CA).

POFs were microdissected from PMSG-primed mouse ovaries under a stereomicroscope, in Leibowitz’s L15 medium supplemented with 5% FBS, 100 U penicillin, and 100 ␮g streptomycin. At least 30 follicles per treatment group were cultured in 1 ml MEM supplemented with 10% FBS and antibiotics at 37 C in glass vials under 95% O2/5% CO2. After equilibration, POFs were cultured in the absence or presence of 5 IU rLH or 50 ␮M Fsk. At the end of culture, POFs were washed in DPBS and collected for protein extraction. In some experiments, POFs were preincubated in the presence of the inhibitors AG1478 (0.5 ␮M), galardin (20 ␮M), TAPI-1 (30 ␮M), puromycin (10 ␮g/ml), H89 (20 ␮M), or vehicle [dimethylsulfoxide (DMSO)] for 30 min before hormone stimulation. For oocyte maturation studies, POFs were cultured in the presence of 5 IU rLH for the indicated times. POFs preincubated in the presence of puromycin were stimulated for 4 h with 5 IU rLH or 100 nM AREG. Follicles were punctured using 26.5-gauge needles (Becton Dickinson & Co., Franklin Lakes, NJ) to release COCs. Cumulus cells were gently removed using a Drummond microdispenser, and oocytes were examined for evidence of GVBD, a hallmark of oocyte meiotic resumption. COCs that were collected 4 h after culture were treated briefly with hyaluronidase (1␮g/ml) to facilitate removal of cumulus cells from oocytes.

Animals

Granulosa cells were isolated from ovaries of immature mice 46 h after PMSG stimulation by needle puncture in Leibowitz’s L15 medium supplemented with 5% FBS, penicillin, and streptomycin. Cells were passed through a 40-␮m cell strainer to remove oocytes, spun down, and resuspended in DMEM/F12 medium supplemented with 1% FBS, penicillin, and streptomycin. Cells were seeded at a density of about

Immature (22–24 d old) female C57BL/6 mice (Charles River Laboratories, Wilmington, MA) were injected ip with 5 IU PMSG to stimulate follicle development to the preovulatory stage. After 44–48 h, animals were euthanized and ovaries excised. For immunohistochemical analyses, PMSG-primed

Granulosa Cell Culture

934 Mol Endocrinol, April 2008, 22(4):924–936

Panigone et al. • Early Activation of EGFR by LH in Follicles

5–6 ⫻ 106 cells per well in six-well culture plates. On the next day, cells were cultured in 1 ml serum-free DMEM/F12 containing vehicle (DMSO) or 10 ␮M Fsk, an activator of adenylyl cyclase. At the end of culture, cells were washed once with DPBS and then collected in RIPA buffer [50 mM Tris (pH 7.5), 150 mM NaCl, 1% Nonidet P-40, 0.1% SDS, 0.25% deoxycholic acid, Complete, Mini, EDTA-free protease inhibitor cocktail, 1 mM EDTA, 1 mM Na-orthovanadate, 1 mM NaF, and 1 mM Na-pyrophosphate] for protein extraction. In some experiments, antimouse IgG1 or neutralizing antibodies against AREG (3 ␮g/ml), EREG (2.5 ␮g/ml), and BTC (2 ␮g/ml) were added to culture 30 min before treatment with Fsk.

IgG-HRP (1:3000), respectively. Specific signals were detected using ECL Western blotting reagent and visualized by autoradiography. For Western blots, specific bands were quantified by scanning and densitometric analyses using Scion Image software (Scion Corp., Frederick, MD).

Immunoprecipitation and Immunoblotting Cultured POFs or granulosa cells were homogenized and lysed in ice-cold RIPA buffer containing a cocktail of EDTAfree protease inhibitors, 1 mM EDTA, 1 mM Na-orthovanadate, 1 mM NaF, and 1 mM Na-pyrophosphate. After centrifugation at 14,000 ⫻ g for 5 min at 4 C, supernatants were collected and protein concentrations assayed using the BCA protein assay kit (Pierce). A fixed amount of protein (⬃140–190 ␮g) was immunoprecipitated by incubation with 4 ␮g anti-EGFR antibody overnight at 4 C, followed by 2.5 h incubation with rec-Protein G-Sepharose 4B beads. After centrifugation, lysates were retained for additional assays. Pellets were washed in DPBS containing protease inhibitors, and then bound EGFR proteins were released from the pellets by boiling for 5 min in 3⫻ SDS buffer. The eluted samples were separated on 7.5% polyacrylamide gels and transferred to polyvinylidene difluoride membranes. Membranes were blocked in Tris-buffered saline with 0.1% Tween 20 (TBST) plus 5% nonfat dry milk, washed with TBST, probed with an anti-phospho-EGFR antibody (specific for autophosphorylation site Tyr1068) diluted 1:1000 in TBST plus 5% BSA (Sigma) overnight at 4 C, and then incubated for 1 h at room temperature with an antirabbit IgG horseradish peroxidase (HRP)-conjugated antibody (Amersham) diluted in TBST plus 0.5% nonfat dry milk (Bio-Rad). After washing in TBST, membranes were incubated with ECL Plus Western blotting reagent for detection of specific signal. Afterward, the same membranes were incubated in a solution containing 62.5 mM Tris-HCl (pH 6.8), 2% SDS, and 100 mM ␤-mercaptoethanol for 30 min at 50 C, washed in TBST, and reprobed for total EGFR. Membranes were blocked for 1 h at room temperature in TBST plus 1% nonfat dry milk, incubated with a polyclonal anti-EGFR antibody diluted 1:200 in TBST plus 0.2% nonfat dry milk for 2 h at room temperature, washed in TBST, and then incubated for 1 h at room temperature with an antirabbit IgG HRP-linked antibody diluted 1:5000 in TBST plus 0.2% nonfat dry milk. After washing in TBST, membranes were incubated with ECL Western blotting reagent for detection of specific signal. MAPK (p44/42) and CREB phosphorylation levels in cultured POFs were analyzed in protein lysates retained after EGFR immunoprecipitation. Briefly, 2 or 5 ␮g lysate, respectively, were separated on 10% polyacrylamide gels and transferred to polyvinylidene difluoride membranes. Membranes were blocked in TBST plus 0.2% nonfat dry milk, incubated for 2 h at room temperature with anti-phosphop44/42 MAPK (1:1000) or anti-phospho-CREB (1 ␮g/ml) antibody diluted in TBST plus 0.2% nonfat dry milk, washed in TBST, and then incubated for 1 h at room temperature with antirabbit IgG-HRP (1:5000) or antimouse IgG-HRP antibodies (1:3000), respectively. Specific signals were detected using ECL reagent. Afterward, membranes were stripped of bound antibodies as described above, blocked in TBST plus 1% nonfat dry milk, and then incubated with anti-pan-ERK or anti-ERK1 (diluted 1:2500 in TBST plus 0.2% nonfat dry milk) for 2 h at room temperature or with anti-CREB (diluted 1:1000 in TBST plus 0.2% nonfat dry milk) overnight at 4 C. After washing in TBST, membranes were incubated for 1 h at room temperature with antimouse IgG-HRP (1:5000) or antirabbit

Metabolic Labeling Intact POFs isolated from PMSG-primed mice were washed three times in methionine-free DMEM (GIBCO), supplemented with 10% dialyzed FBS, 100 U penicillin, 100 ␮g streptomycin, and 2 mM L-glutamine (GIBCO). Three follicles were then preincubated for 15 min in this medium with 50 ␮Ci/ml [35S]methionine (Amersham) to deplete intracellular pools of methionine. The follicles were incubated for an additional 30 min in the absence or presence of puromycin and then stimulated with 5 IU rLH for 4 h. After the incubations, follicles were washed twice in DPBS and homogenized on ice in 500 ␮l 5% (wt/vol) trichloroacetic acid solution. After centrifugation at 3000 rpm for 30 min at 4 C, the pellets were resuspended in 1 ml 5% trichloroacetic acid solution and centrifuged again at 3000 rpm for 30 min at 4 C. Afterward, the pellet was resuspended with 200 ␮l DPBS. The supernatant was reserved for use as a control. After vortexing and sonication of the samples to break down the cells, the amount of [35S]methionine incorporation was counted. Immunohistochemistry Excised ovaries were fixed in Bouin’s solution overnight at 4 C. After dehydration, the ovaries were embedded in Paraplast and sectioned at 6 ␮m onto Superfrost Plus slides. For immunohistochemical staining, sections were deparaffinized and rehydrated, treated with 3% H2O2 for 15 min to inactivate intrinsic peroxidase activity, and incubated with sodium citrate buffer for 20 min at 90 C for antigen retrieval. Washes were performed with automation buffer [50 mM Tris-HCl (pH 7.5), 150 mM NaCl, and 0.02% Tween 20]. After blocking for 20 min at room temperature with normal goat serum, sections were incubated for 1 h at room temperature with anti-phospho-p44/42 MAPK antibody diluted 1:100 in buffer containing 1% BSA. Sections were next incubated with biotinylated goat antirabbit antibody for 30 min at room temperature, followed by a 30-min incubation with the avidin/biotinylated enzyme complex (ABC). Blocking serum and antibodies were from the antirabbit Vectastain ABC kit (Vector Laboratories). Staining was performed using the diaminobenzidine chromogen. The staining reactions were stopped in distilled water, and sections were dehydrated and mounted with Permount. Statistics Data are presented as the mean ⫾ SEM. Data were analyzed by Student’s t test or one-way ANOVA and Bonferroni’s multiple comparison test where appropriate. P ⬍ 0.05 was considered statistically significant.

Acknowledgments Received May 10, 2007. Accepted January 2, 2008. Address all correspondence and requests for reprints to: Marco Conti, M.D., Professor, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California at San Francisco, 513 Parnassus, San Francisco, California 94143-0556. E-mail: [email protected]. Current address for M.H. and M.C.: Department of Obstetrics, Gynecology and Reproductive Sciences, University of California at San Francisco, San Francisco, California 94143. The work done in the authors’ laboratory was supported by the National Research Service Award F32-HD049966 (M.H.)

Panigone et al. • Early Activation of EGFR by LH in Follicles

from the National Institute of Child Health and Human Development (NICHD) and NICHD/National Institutes of Health Grant 1R01HD052909 (M.C.) and through a cooperative agreement (U54-HD31389 to M.C.) as part of the Specialized Cooperative Centers Program in Reproduction Research. A grant from Organon (M.C.) is also acknowledged. Disclosure Summary: S.P., M.H., M.F., and L.P. have nothing to declare. M.C. received grant support (10/06-9/07) from N.V. Organon.

REFERENCES 1. Richards JS 2001 New signaling pathways for hormones and cyclic adenosine 3⬘,5⬘-monophosphate action in endocrine cells. Mol Endocrinol 15:209–218 2. Conti M 2002 Specificity of the cyclic adenosine 3⬘,5⬘monophosphate signal in granulosa cell function. Biol Reprod 67:1653–1661 3. Wayne CM, Fan HY, Cheng X, Richards JS 2007 Folliclestimulating hormone induces multiple signaling cascades: evidence that activation of Rous sarcoma oncogene, RAS, and the epidermal growth factor receptor are critical for granulosa cell differentiation. Mol Endocrinol 21:1940–1957 4. Peng XR, Hsueh AJ, LaPolt PS, Bjersing L, Ny T 1991 Localization of luteinizing hormone receptor messenger ribonucleic acid expression in ovarian cell types during follicle development and ovulation. Endocrinology 129: 3200–3207 5. Park JY, Su YQ, Ariga M, Law E, Jin SL, Conti M 2004 EGF-like growth factors as mediators of LH action in the ovulatory follicle. Science 303:682–684 6. Ashkenazi H, Cao X, Motola S, Popliker M, Conti M, Tsafriri A 2005 Epidermal growth factor family members: endogenous mediators of the ovulatory response. Endocrinology 146:77–84 7. Sekiguchi T, Mizutani T, Yamada K, Kajitani T, Yazawa T, Yoshino M, Miyamoto K 2004 Expression of epiregulin and amphiregulin in the rat ovary. J Mol Endocrinol 33: 281–291 8. Andric N, Ascoli M 2006 A delayed gonadotropin-dependent and growth factor-mediated activation of the extracellular signal-regulated kinase 1/2 cascade negatively regulates aromatase expression in granulosa cells. Mol Endocrinol 20:3308–3320 9. Jo M, Curry Jr TE 2006 Luteinizing hormone-induced RUNX1 regulates the expression of genes in granulosa cells of rat periovulatory follicles. Mol Endocrinol 20: 2156–2172 10. Tirone E, D’Alessandris C, Hascall VC, Siracusa G, Salustri A 1997 Hyaluronan synthesis by mouse cumulus cells is regulated by interactions between follicle-stimulating hormone (or epidermal growth factor) and a soluble oocyte factor (or transforming growth factor ␤1). J Biol Chem 272:4787–4794 11. Orly J, Rei Z, Greenberg NM, Richards JS 1994 Tyrosine kinase inhibitor AG18 arrests follicle-stimulating hormone-induced granulosa cell differentiation: use of reverse transcriptase-polymerase chain reaction assay for multiple messenger ribonucleic acids. Endocrinology 134:2336–2346 12. Hsieh M, Lee D, Panigone S, Horner K, Chen R, Theologis A, Lee DC, Threadgill DW, Conti M 2007 Luteinizing hormone-dependent activation of the epidermal growth factor network is essential for ovulation. Mol Cell Biol 27:1914–1924 13. Holbro T, Hynes NE 2004 ErbB receptors: directing key signaling networks throughout life. Annu Rev Pharmacol Toxicol 44:195–217 14. Yarden Y, Sliwkowski MX 2001 Untangling the ErbB signalling network. Nat Rev Mol Cell Biol 2:127–137

Mol Endocrinol, April 2008, 22(4):924–936

935

15. Daaka Y, Luttrell LM, Lefkowitz RJ 1997 Switching of the coupling of the ␤2-adrenergic receptor to different G proteins by protein kinase A. Nature 390:88–91 16. Liebmann C 2001 Regulation of MAP kinase activity by peptide receptor signalling pathway: paradigms of multiplicity. Cell Signal 13:777–785 17. Lopez-Ilasaca M, Crespo P, Pellici PG, Gutkind JS, Wetzker R 1997 Linkage of G protein-coupled receptors to the MAPK signaling pathway through PI 3-kinase ␥. Science 275:394–397 18. Daub H, Weiss FU, Wallasch C, Ullrich A 1996 Role of transactivation of the EGF receptor in signalling by Gprotein-coupled receptors. Nature 379:557–560 19. Wetzker R, Bohmer FD 2003 Transactivation joins multiple tracks to the ERK/MAPK cascade. Nat Rev Mol Cell Biol 4:651–657 20. Daub H, Wallasch C, Lankenau A, Herrlich A, Ullrich A 1997 Signal characteristics of G protein-transactivated EGF receptor. EMBO J 16:7032–7044 21. Bokemeyer D, Schmitz U, Kramer HJ 2000 Angiotensin II-induced growth of vascular smooth muscle cells requires an Src-dependent activation of the epidermal growth factor receptor. Kidney Int 58:549–558 22. Luttrell LM, Della Rocca GJ, van Biesen T, Luttrell DK, Lefkowitz RJ 1997 G␤␥ subunits mediate Src-dependent phosphorylation of the epidermal growth factor receptor. A scaffold for G protein-coupled receptor-mediated Ras activation. J Biol Chem 272:4637–4644 23. Zwick E, Daub H, Aoki N, Yamaguchi-Aoki Y, Tinhofer I, Maly K, Ullrich A 1997 Critical role of calcium-dependent epidermal growth factor receptor transactivation in PC12 cell membrane depolarization and bradykinin signaling. J Biol Chem 272:24767–24770 24. Eguchi S, Numaguchi K, Iwasaki H, Matsumoto T, Yamakawa T, Utsunomiya H, Motley ED, Kawakatsu H, Owada KM, Hirata Y, Marumo F, Inagami T 1998 Calcium-dependent epidermal growth factor receptor transactivation mediates the angiotensin II-induced mitogenactivated protein kinase activation in vascular smooth muscle cells. J Biol Chem 273:8890–8896 25. Soltoff SP 1998 Related adhesion focal tyrosine kinase and the epidermal growth factor receptor mediate the stimulation of mitogen-activated protein kinase by the G-protein-coupled P2Y2 receptor. Phorbol ester or [Ca2⫹]i elevation can substitute for receptor activation. J Biol Chem 273:23110–23117 26. Tsai W, Morielli AD, Peralta EG 1997 The m1 muscarinic acetylcholine receptor transactivates the EGF receptor to modulate ion channel activity. EMBO J 16:4597–4605 27. Prenzel N, Zwick E, Daub H, Leserer M, Abraham R, Wallasch C, Ullrich A 1999 EGF receptor transactivation by G-protein-coupled receptors requires metalloproteinase cleavage of proHB-EGF. Nature 402:884–888 28. Fischer OM, Hart S, Gschwind A, Ullrich A 2003 EGFR signal transactivation in cancer cells. Biochem Soc Trans 31:1203–1208 29. Rojas M, Yao S, Lin YZ 1996 Controlling epidermal growth factor (EGF)-stimulated Ras activation in intact cells by a cell-permeable peptide mimicking phosphorylated EGF receptor. J Biol Chem 271:27456–27461 30. Downward J, Parker P, Waterfield MD 1984 Autophosphorylation sites on the epidermal growth factor receptor. Nature 311:483–485 31. Saito T, Okada S, Ohshima K, Yamada E, Sato M, Uehara Y, Shimizu H, Pessin JE, Mori M 2004 Differential activation of epidermal growth factor (EGF) receptor downstream signaling pathways by betacellulin and EGF. Endocrinology 145:4232–4243 32. Su YQ, Wigglesworth K, Pendola FL, O’Brien MJ, Eppig JJ 2002 Mitogen-activated protein kinase activity in cumulus cells is essential for gonadotropin-induced oocyte meiotic resumption and cumulus expansion in the mouse. Endocrinology 143:2221–2232

936 Mol Endocrinol, April 2008, 22(4):924–936

Panigone et al. • Early Activation of EGFR by LH in Follicles

33. Shimada M, Hernandez-Gonzalez I, Gonzalez-Robayna I, Richards JS 2006 Paracrine and autocrine regulation of epidermal growth factor-like factors in cumulus oocyte complexes and granulosa cells: key roles for prostaglandin synthase 2 and progesterone receptor. Mol Endocrinol 20:1352–1365 34. Freimann S, Ben-Ami I, Dantes A, Ron-El R, Amsterdam A 2004 EGF-like factor epiregulin and amphiregulin expression is regulated by gonadotropins/cAMP in human ovarian follicular cells. Biochem Biophys Res Commun 324:829–834 35. Stover DR, Becker M, Liebetanz J, Lydon NB 1995 Src phosphorylation of the epidermal growth factor receptor at novel sites mediates receptor interaction with Src and P85␣. J Biol Chem 270:15591–15597 36. Sela-Abramovich S, Chorev E, Galiani D, Dekel N 2005 Mitogen-activated protein kinase mediates luteinizing hormone-induced breakdown of communication and oocyte maturation in rat ovarian follicles. Endocrinology 146:1236–1244 37. Fan HY, Huo LJ, Chen DY, Schatten H, Sun QY 2004 Protein kinase C and mitogen-activated protein kinase cascade in mouse cumulus cells: cross talk and effect on meiotic resumption of oocyte. Biol Reprod 70:1178–1187

38. Su YQ, Denegre JM, Wigglesworth K, Pendola FL, O’Brien MJ, Eppig JJ 2003 Oocyte-dependent activation of mitogen-activated protein kinase (ERK1/2) in cumulus cells is required for the maturation of the mouse oocytecumulus cell complex. Dev Biol 263:126–138 39. Kalma Y, Granot I, Galiani D, Barash A, Dekel N 2004 Luteinizing hormone-induced connexin 43 downregulation: inhibition of translation. Endocrinology 145: 1617–1624 40. Singh AB, Harris RC 2005 Autocrine, paracrine and juxtacrine signaling by EGFR ligands. Cell Signal 17: 1183–1193 41. Meyer zu Heringdorf D, Lass H, Kuchar I, Alemany R, Guo Y, Schmidt M, Jakobs KH 1999 Role of sphingosine kinase in Ca2⫹ signalling by epidermal growth factor receptor. FEBS Lett 461:217–222 42. Park OK, Mayo KE 1991 Transient expression of progesterone receptor messenger RNA in ovarian granulosa cells after the preovulatory luteinizing hormone surge. Mol Endocrinol 5:967–978 43. Robker RL, Russell DL, Espey LL, Lydon JP, O’Malley BW, Richards JS 2000 Progesterone-regulated genes in the ovulation process: ADAMTS-1 and cathepsin L proteases. Proc Natl Acad Sci USA 97:4689–4694

Molecular Endocrinology is published monthly by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the endocrine community.