2006 Bagchi Edn2 PGR Ovulation Mol Endo

download 2006 Bagchi Edn2 PGR Ovulation Mol Endo

of 12

Transcript of 2006 Bagchi Edn2 PGR Ovulation Mol Endo

  • 7/31/2019 2006 Bagchi Edn2 PGR Ovulation Mol Endo

    1/12

    A Novel Pathway Involving Progesterone Receptor,Endothelin-2, and Endothelin Receptor B ControlsOvulation in Mice

    Gopinath S. Palanisamy, Yong-Pil Cheon,* Jaeyeon Kim,* Athilakshmi Kannan, Quanxi Li,Marcey Sato, Srinivasa R. Mantena, Regine L. Sitruk-Ware, Milan K. Bagchi, and Indrani C. Bagchi

    Department of Veterinary Biosciences (G.S.P., Y.-P.C., A.K., Q.L., S.R.M., I.C.B.) and Department ofMolecular and Integrative Physiology (J.K., M.S., M.K.B.), University of Illinois at Urbana-Champaign,Urbana, Illinois 61802; and Population Council (R.L.S.-W.), New York, New York 10021

    The steroid hormone progesterone (P) plays a piv-

    otal role during ovulation. Mice lacking P receptor

    (Pgr) gene fail to ovulate due to a defect in follicular

    rupture. The P receptor (PGR)-regulated pathways

    that modulate ovulation, however, remain poorly

    understood. To identify these pathways, we per-formed gene expression profiling using ovaries

    from mice subjected to gonadotropin-induced su-

    perovulation in the presence and in the absence of

    CDB-2914, a synthetic PGR antagonist. Prominent

    among the genes that were down-regulated in re-

    sponse to CDB-2914 was endothelin (ET)-2, a po-

    tent vasoactive molecule. ET-2 mRNA was tran-

    siently induced in mural granulosa cells of the

    preovulatory follicles immediately preceding ovu-

    lation. This induction was absent in the ovaries of

    PGR null mice, indicating a critical role of this re-

    ceptor in ET-2 expression. To investigate the func-

    tional role of ET-2 during ovulation, we employed

    selective antagonists of endothelin receptors,

    ETR-A and ETR-B. Mice treated with an ETR-B

    antagonist exhibited a dramatic (>85%) decline in

    the number of released oocytes. Strong expression

    of ETR-B was observed in the mural and cumulusgranulosa cells of the preovulatory follicles as well

    as in the capillaries lining the inner border of the

    theca interna. We also identified cGMP-dependent

    protein kinase II, a previously reported PGR-regu-

    lated gene, as a downstream target of ET-2 during

    ovulation. Collectively, our studies uncovered a

    unique pathway in which ET-2, produced by PGR in

    mural granulosa cells, acts in a paracrine or auto-

    crine manner on multiple cell types within the pre-

    ovulatory follicle to control the final events leading

    to its rupture. (Molecular Endocrinology 20:

    27842795, 2006)

    OVULATION IN MAMMALS is a unique biologicalprocess in which a mature follicle within the ovaryruptures and releases a fertilizable oocyte. In most

    mammals, the follicle protrudes markedly from the

    ovarian surface at the time of ovulation, and in many

    instances a thin translucent stigma, the macula pellu-

    cida, forms at the apex of the follicle as a sign of

    impending rupture (1). It has been suggested that the

    rupture of the follicle is a culmination of a profound

    inflammatory response (2). Although morphological

    features of the ovulatory process are well described,

    the molecular cascade of events that ultimately leadsto follicular rupture remains largely unknown (24). It is

    widely thought, however, that a preovulatory surge ofthe LH initiates the expression of critical gene path-ways that underlie the morphological and biochemicalalterations that occur before ovulation.

    Several lines of evidence suggest that progesterone(P) receptor (PGR) is a key regulatory molecule in theovary, acting downstream of LH during ovulation. LHrapidly and selectively induces PGR expression in mu-ral granulosa cells of the preovulatory follicles (57).Earlier studies in rats indicated that ovulation is inhib-ited by treatment with either an anti-P antiserum, or

    epostane, a compound that blocks the synthesis of P(8, 9). Administration of PGR antagonists, such asRU486 and Org-31710, also inhibited ovulation, sug-gesting that P acting via its receptor controls ovulation(10, 11). Creation of a Pgr knockout (PRKO) mousemodel by Lydon et al. (12) and Conneely et al. (13)provided an unequivocal demonstration of the criticalrole played by this receptor in the ovulatory process.The PRKO mice failed to ovulate even when stimulatedwith exogenous gonadotropins. In these mutant mice,follicles developed normally to the preovulatory stage,and the characteristic expansion of the cumulus-oo-cyte complex (COC) appeared to occur. The follicles,however, failed to rupture, resulting in trapped oocyteswithin the ovarian tissue (12).

    First Published Online August 3, 2006* Y.-P.C. and J.K. contributed equally to the manuscript.

    Abbreviations: BW, Body weight; CG, chorionic gonado-tropin; cGK II, cGMP-dependent protein kinase II; COC, cu-mulus-oocyte complex; DIG, Digoxygenin; eNOS, endothelialNO synthase; ET, endothelin; ETR, ET receptor; NO, nitricoxide; P, progesterone; PGR, progesterone receptor; PMA,phorbol 12-myristate 13 acetate; PMSG, pregnant mare se-rum gonadotropin; PRKO, Pgrknockout; VIC, vasoactive in-testinal contractor; WT, wild type.

    Molecular Endocrinology is published monthly by TheEndocrine Society (http://www.endo-society.org), theforemost professional society serving the endocrinecommunity.

    0888-8809/06/$15.00/0 Molecular Endocrinology 20(11):27842795Printed in U.S.A. Copyright 2006 by The Endocrine Society

    doi: 10.1210/me.2006-0093

    2784

  • 7/31/2019 2006 Bagchi Edn2 PGR Ovulation Mol Endo

    2/12

    PGR is a well-known ligand-inducible transcriptionfactor (14, 15). It is likely that hormone-occupied PGRtriggers the expression of specific gene networks inthe granulosa cells of the ovarian follicles, and theproducts of these genes mediate the hormonal effectsduring ovulation. To understand the molecular basis ofthe ovulatory program, it is critical to identify the PGR-regulated molecules that are induced or suppressed atthe time of ovulation and analyze their functional rolesduring this complex physiological process.

    In this study, we employed CDB-2914, a novel an-tiprogestin, as a tool to uncover the PGR-regulatedpathways during ovulation in mice. This compoundbinds to PGR with high affinity and impairs its gene-regulatory function (1619). Our results indicated thatadministration of CDB-2914 strongly blocked LH-in-duced ovulation in mice. Using oligonucleotide mi-croarrays, we identified several genes the expressionof which is markedly repressed in response to CDB-

    2914 in mouse ovary at the time of ovulation. One ofthese genes encoded the vasoactive intestinal con-tractor (VIC), the mouse ortholog of human endothelin(ET)-2. ET-2 is a vasoactive molecule that mediatesvarious physiological functions such as vasodilation,vasoconstriction, and smooth muscle contraction inother tissues (2022). Here we present evidence that aunique signaling pathway in the preovulatory follicle,involving LH, PGR, ET-2, and its downstream targetmolecules, plays a critical role during ovulation.

    RESULTS

    Administration of CDB-2914 Blocks Ovulation

    in Mice

    A previous study reported an antiovulatory activity ofCDB-2914 in rats (16). To investigate its effects onovulation in mice, gonadotropin-induced superovula-tion was performed in the absence and in the presenceof this drug. Mice were treated with pregnant mareserum gonadotropin (PMSG) for 48 h followed by

    treatment with human (h) chorionic gonadotropin (CG)to induce the ovulatory response. Vehicle or CDB-2914 was administered ip 1 h before the hCG injection.The number of released oocytes was counted 18 hafter hCG treatment. As shown in Fig. 1A, an averageof 3040 oocytes were released from the ovaries ofeach mouse subjected to the superovulation protocol.Histological analysis of the ovarian sections of vehicle-treated mice at 18 h after hCG injection exhibitednumerous corpora lutea, indicating efficient ovulation(Fig. 1B, panel a). Administration of increasing dosesof CDB-2914 led to a progressive decline in the num-ber of released oocytes when compared with micethat received vehicle alone (Fig. 1A). Ovarian sectionsof CDB-2914-treated mice showed the presence ofmany unruptured follicles at 18 h after hCG treatment,confirming impaired ovulation (Fig. 1B, panel b). Thefailure of follicular rupture upon CDB-2914 treatmentclosely resembled the ovarian phenotype of the PRKO

    mice and is consistent with the previously reportedantiprogestational activity of this drug. Collectively,our results indicated that CDB-2914 exerts its inhibi-tory effects on ovulation in mice by blocking the func-tion of PGR.

    Identification of ET-2 as a Downstream Target of

    Regulation by CDB-2914 in the Ovary

    We next employed CDB-2914 to identify genes thatare regulated by PGR during ovulation. Mice weresubjected to superovulation in the absence and in thepresence of CDB-2914 as described above, and ova-

    ries were collected 12 h after hCG treatment. mRNAwas isolated from vehicle or CDB2914-treated miceand hybridized to mouse oligonucleotide microarrays(Affymetrix, Santa Clara, CA) representing approxi-mately 11,000 genes as described previously (23). Ourstudies identified several mRNAs the expression ofwhich in the ovary altered at least 2-fold in response toCDB-2914 (Cheon, Y.C., and I.C. Bagchi, unpublishedresults). One of the mRNAs that was markedly down-regulated by CDB-2914 encoded the vasoactive intes-

    Fig. 1. Administration of CDB-2914 to Mice Inhibits OvulationA, Mice were treated with PMSG and 48 h later with hCG. One hour before hCG administration, animals (n 18) were treated

    with vehicle (n 6), CDB-2914 at 20 mg/kg BW (n 6), and CDB-2914 at 40 mg/kg BW (n 6), respectively. Oviducts werecollected 18 h after the hCG injection, and the number of released oocytes was counted. The data are represented as means SEM. B, Hematoxylin and eosin staining of ovarian sections of vehicle (panel a) or CDB-2914-treated (40 mg/kg BW; panel b) mice.The results are representative of three independent experiments. CL, Corpus luteum, UF, unruptured follicles.

    Palanisamy et al. P Regulation & Function of ET-2 in the Ovary Mol Endocrinol, November 2006, 20(11):27842795 2785

  • 7/31/2019 2006 Bagchi Edn2 PGR Ovulation Mol Endo

    3/12

    tinal contractor (VIC), the mouse ortholog of humanET-2 (2729). VIC differs from the human ET-2 in onlyone of 21 amino acid residues and mimics its biolog-ical function (2729). Therefore, in this paper, we willrefer to VIC as mouse ET-2.

    To verify the results of our microarray analysis, weexamined, by Northern blotting, ovarian RNAs ob-tained from mice subjected to superovulation in theabsence or in the presence of CDB-2914. Treatmentwith CDB-2914 did not alter the level of ovarian PgrmRNA induced by hCG (data not shown). As shown inFig. 2A, a strong signal corresponding to ET-2 mRNAwas observed at 12 h after hCG administration (lane 1).This signal was undetectable in the ovaries of CDB-2914-treated mice (lane 2), confirming the results ofour microarray analysis.

    ET-2 mRNA Expression Is Transiently Induced in

    the Ovary at the Time of Ovulation

    We next determined the temporal profile of ET-2 ex-pression during gonadotropin-induced superovula-tion. Total ovarian RNA was collected at 0, 4, 8, 12,and 16 h after hCG injection and subjected to Northernblot analysis. As shown in Fig. 2B, upper panel, ET-2transcript was undetectable at 08 h post hCG admin-istration. A dramatic rise in the level of this transcriptwas observed at 12 h, which declined to an undetect-able level by 16 h post hCG treatment.

    In mice subjected to gonadotropin-induced super-ovulation, follicular rupture typically occurs at 1112 hafter hCG administration. To further narrow down thewindow of expression of ET-2 gene during superovu-lation, we performed Northern blot analysis usingovarian RNA collected at 10, 11, 12, and 13 h afterhCG treatment (Fig. 2C). We found that the level of

    ET-2 mRNA rose sharply at 11 h, increased further at12 h, and then declined abruptly by 13 h. A transientsurge of ET-2 mRNA expression, therefore, occurs inthe ovary between 1112 h after hCG stimulation, andit precisely overlaps with the time of follicular rupture.

    PGR Regulates the Expression of ET-2 mRNA in

    Granulosa Cells of Preovulatory Follicles

    To investigate whether PGR controls the ovarian ex-pression of ET-2 mRNA, we analyzed the expressionof this gene in the ovaries of PRKO mice. Wild-type(WT) and PRKO females were subjected to gonado-

    tropin-induced superovulation. Ovarian RNA was iso-lated at 0, 4, 8, 12, and 16 h after hCG injection andanalyzed by Northern blotting. As shown in Fig. 3A,upper panel, a transient but robust expression of ET-2mRNA was observed in the ovarian tissues collectedfrom WT mice at 12 h post hCG. In contrast, theexpression of ET-2 mRNA was undetectable in PRKOmice, establishing that PGR is essential for inductionof ET-2 expression in the ovary during ovulation.

    We next examined the spatial expression of ET-2mRNA in the ovary at the time of ovulation by employ-ing in situ hybridization. As shown in Fig. 3B, using anantisense probe specific for ET-2, we observed a

    strong hybridization signal in the mural granulosa cellsof the preovulatory follicles at 12 h after hCG injection(Fig. 3B, panel a). No ET-2 mRNA was detected in theprimary or secondary follicles. Ovarian sections hy-bridized with the corresponding sense RNA probe ofequal length did not exhibit any significant signal (Fig.3B, panel b). We also failed to detect any signal cor-responding to ET-2 mRNA in the ovarian sections ofPRKO mice at 12 h after hCG injection (Fig. 3B, panelc). These results demonstrated that the expression ofET-2 is induced exclusively in the preovulatory folliclesduring ovulation, and this induction is dependent onPGR.

    We also analyzed the PGR regulation of ET-2 ex-

    pression in primary cultures of granulosa cells. Previ-

    Fig. 2. ET-2 mRNA Expression in the OvaryA, Mice (n 4) were treated with PMSG and 48 h later with

    hCG. One hour before hCG administration, mice were in-jected with vehicle or CDB-2914 (40 mg/kg BW). The ovarieswere collected 12 h after hCG treatment, and mRNA (1 g)was analyzed by Northern blotting followed by hybridizationwith 32P-labeled cDNA probes corresponding to ET-2 and36B4, a housekeeping gene encoding a ribosomal protein.Lanes 1 and 2 represent mRNAs from mice treated with

    vehicle and CDB-2914, respectively. B, mRNA (1 g) ob-tained from ovaries of superovulated mice at 0, 4, 8, 12, and16 h after hCG administration (n 2 at each time point) wassubjected to Northern blot analysis. Top panel representssignals obtained after hybridization with a 32P-labeled ET-2cDNA probe. The bottom panel shows the same blot afterhybridization with a 32P-labeled 36B4 probe. C, mRNA (1 g)was isolated from ovaries of superovulated mice at 10, 11,12, and 13 h after hCG administration (n 3 at each timepoint) and subjected to Northern blot analysis. The intensitiesof the ET-2 mRNA signals were quantitated by densitometricscanning and normalized with respect to the 36B4 signals inthe same blot. The relative intensities representing ET-2mRNA levels at different times during superovulation werethen plotted. The normalized value of the 12 h samplewas setat 100%.

    2786 Mol Endocrinol, November 2006, 20(11):27842795 Palanisamy et al. P Regulation & Function of ET-2 in the Ovary

  • 7/31/2019 2006 Bagchi Edn2 PGR Ovulation Mol Endo

    4/12

    ous studies showed that treatment of cultures ofPMSG-primed granulosa cells with forskolin and phor-bol 12-myristate 13 acetate (PMA), which activate ad-

    enylyl cyclase and protein kinase C pathways, respec-tively, mimics LH signaling that, in turn, stimulates Pgrgene expression in these cells (31). Consistent withthese reports, we observed a marked induction of PgrmRNA within 4 h of addition of forskolin and PMA togranulosa cells (Fig. 4A). At 12 h, the level of ET-2mRNA rose steeply in these cells (Fig. 4, B and C). It isof interest to note that the temporal expression patternof Pgr and ET-2 mRNAs in granulosa cells in vitro isremarkably similar to that seen for these genes inresponse to LH in vivo. Addition of CDB-2914, whichblocks PGR function, inhibited the induction of ET-2mRNA in a dose-dependent manner (Fig. 4B). In

    agreement with this finding, when granulosa cells iso-lated from ovaries of PMSG-primed PRKO mice weretreated with forskolin and PMA, we failed to detect anyinduction of ET-2 mRNA (Fig. 4C). Collectively, theseresults strengthened our view that ET-2 is a down-stream target of PGR-dependent mechanisms in thepreovulatory follicles.

    Expression of ET Receptors (ETRs) in the Ovary

    The cellular functions of ET-2 are mediated via the ETRisoforms, ETR-A and ETR-B (2022). We, therefore,used Northern blotting to monitor the expression ofETR-A and ETR-B mRNAs in the mouse ovary at var-

    ious times after hCG administration. As shown in Fig.

    5A, signals corresponding to ETR-A and ETR-BmRNAs were observed in the ovaries at 0, 4, 8, or 12 h

    post hCG treatment, indicating constitutive expression

    of the receptors during the ovulatory period. Using32P-labeled ETR-A and ETR-B cDNA probes of iden-

    tical specific activity, we observed a relatively strongersignal of ETR-B compared with ETR-A in these tis-sues, suggesting that ETR-B is likely the predominant

    ET receptor in the ovary during ovulation. We alsoexamined the expression of these receptors in theovaries of PRKO mice and found that the expressionlevels of ETR-A and ETR-B in these mice are compa-

    rable to those seen in the WT animals (data notshown).

    We next localized the site of expression of ET-2receptors during ovulation. Mice were subjected to

    superovulation, ovaries were collected 12 h after hCGadministration, and immunohistochemistry was per-formed using ETR-A- and ETR-B-specific antibodies(Fig. 5B). Ovarian sections stained with the ETR-A

    antibody showed weak immunostaining in the muraland cumulus granulosa cells (Fig. 5B, panel b). Incontrast, the ETR-B antibody exhibited relativelystrong immunostaining in the mural cells and particu-

    larly intense staining was seen in the cumulus cells(Fig. 5B, panel c). Most interestingly, closer examina-tion of higher magnification micrographs revealed dis-tinct ETR-B immunostaining in the endothelial cells of

    large capillaries that lie along the inner border of the

    theca interna (Fig. 5B, panel e, indicated by arrows).

    Fig. 3. PGR Regulates Ovarian Expression of ET-2 mRNAA, Female WT and PRKO mice of same genetic background (129 Sv) were subjected to superovulation. mRNA (1 g) was

    obtained from ovaries of mice at 0, 4, 8, 12, and 16 h after hCG administration and analyzed by Northern blotting using 32P-labeledET-2 and 36B4 cDNA probes. The results are representative of two independent experiments. B, WT (129 Sv) and PRKO micewere subjected to superovulation, and ovaries were collected 12 h after hCG administration. Ovarian sections were analyzed by

    in situ hybridization employing a DIG-labeled antisense RNA probe specific for ET-2 gene. Panels a and b represent sections fromWT mice hybridized with antisense and sense ET-2 probes, respectively. Panel c is a section from PRKO mice hybridized withthe ET-2 antisense probe.

    Palanisamy et al. P Regulation & Function of ET-2 in the Ovary Mol Endocrinol, November 2006, 20(11):27842795 2787

  • 7/31/2019 2006 Bagchi Edn2 PGR Ovulation Mol Endo

    5/12

    Although ETR-B staining was observed in follicles of

    all stages, it was most prominent in the preovulatoryfollicles, particularly in the COC. These results indi-cated that ETR-B is the likely mediator of the func-tional effects of ET-2 during the final events leading tothe rupture of the preovulatory follicles. Because it isexpressed in multiple cell types within the tissue, itpresents multiple potential sites of action of ET-2 dur-ing the rupture process.

    Administration of ETR Antagonists Impairs

    Ovulation in the Mouse

    To address the role of ET-2 in ovulation, we employedselective antagonists of ETR-A and ETR-B. We used

    JKC-301, a specific inhibitor of ETR-A, and BQ-788, a

    specific inhibitor of ETR-B (32, 33). In superovulation

    experiments, each inhibitor was administered ip at 6 hafter hCG priming. The impact of these drugs on ovu-

    lation was assessed by counting the number of re-

    leased oocytes at 18 h after hCG injection (Fig. 6A).

    Whereas administration of JKC-301 led to a statisti-

    cally significant reduction (25%; P 0.05) in the

    number of released oocytes compared with the vehi-

    cle-treated control group, similar treatment with BQ-

    788 resulted in a more striking, approximately 75%,

    decline in the rate of ovulation. BQ-788, therefore,

    appeared to be a more effective blocker of ovulation

    than JKC-301.

    We next examined whether the timing of adminis-

    tration of the antagonist BQ-788 has any effect on the

    Fig. 4. PGR Regulates Expression of ET-2 mRNA in Primary Cultures of Granulosa CellsA, Mice were treated with PMSG and ovaries were collected at 48 h. Granulosa cells were obtained from preovulatory follicles

    by needle puncture and cultured overnight in DMEM-F12 medium containing 1% fetal bovine serum. The cells were treated withP (10 nM), PMA (20 nM), and forskolin (Fo, 10 M) for 0 and 4 h. Total RNA was isolated from these cells and analyzed byquantitative RT-PCR (Q-PCR) using gene-specific primers for PGR and 36B4. Expression of PgrmRNA was normalized against36B4 mRNA expression. Fold changes were computed relative to the expression level of PGR in cells collected from mice at 0 h.B, The cells were treated with or without indicated concentrations of P, PMA, forskolin, and CDB-2914 for 0 h (column 1) and 12 h(columns 25). Total RNAwas isolated from these cells and subjected to Q-PCR using ET-2 and 36B4 primers. Expression of ET-2mRNA was normalized against 36B4 mRNA expression. Fold changes were computed relative to the expression level of ET-2 incells collected from mice at 0 h. C, Granulosa cells from WT or PRKO mice were treated with P (10 nM), PMA (20 nM), and forskolin(10 M) for 0, 4, 8, 12, and 16 h. Total RNA was isolated from these cells and subjected to Q-PCR using primers for ET-2 and36B4. Expression of ET-2 mRNA was normalized against 36B4 mRNA expression. Fold changes were computed relative to theexpression level of ET-2 in cells collected from WT mice at 0 h.

    2788 Mol Endocrinol, November 2006, 20(11):27842795 Palanisamy et al. P Regulation & Function of ET-2 in the Ovary

  • 7/31/2019 2006 Bagchi Edn2 PGR Ovulation Mol Endo

    6/12

    magnitude of inhibition of ovulation. The drug wasadministered at 4, 6, or 8 h after hCG injection, and thenumber of released oocytes was counted at 18 h. Asshown in Fig. 6B, administration of BQ-788 at timesthat are increasingly closer to the time of ovulationresulted in a progressive decline in the number ofreleased oocytes. We observed greater than 85% in-hibition in oocyte release when the drug was givenonly 4 h before the time of ovulation.

    We also performed a histological analysis of the

    ovaries of mice treated with BQ-788 at 8 h after hCG

    injection. As shown in Fig. 6C, left panel, ovarian sec-tions of vehicle-treated control mice showed numer-ous corpora lutea, indicating successful ovulation. Incontrast, ovarian sections from BQ-788-treated miceshowed many unruptured follicles and only a few cor-pora lutea (Fig. 6C, right panel). Taken together, theseresults indicated that the blockade of ETR-B by BQ-788 effectively suppressed ovulation by inhibiting fol-licular rupture.

    Because ET-2 is a downstream target of PGR reg-

    ulation during the ovulatory period, we considered the

    Fig. 5. Expression Profiles of ETR-A and ETR-B in Mouse Ovary during OvulationA, mRNA (1 g) was isolated from ovaries of superovulated mice at 0, 4, 8, and 12 h after hCG administration and subjected

    to Northern blot analysis employing 32P-labeled ETR-A (top panel), ETR-B (middle panel), and 36B4 (bottom panel) cDNA probes.B, Ovaries from superovulated mice were collected at 12 h after hCG treatment and subjected to immunohistochemical analysisusing preimmune serum (a), ETR-A antibody (b), and ETR-B antibody (c). Panels d and e show higher magnification ( 20) of anovarian section obtained from a superovulated mouse at 12 h after hCG treatment. Panel d, Hematoxylin and eosin staining. The

    red spots indicate blood vessels. Panel e, Immunostaing using ETR-B antibody. Reddish brown stains in the mural and cumulus

    granulosa layers indicate specific ETR-B immunostaining. The arrows indicate large capillaries that also exhibit ETR-B immu-nostaining.

    Palanisamy et al. P Regulation & Function of ET-2 in the Ovary Mol Endocrinol, November 2006, 20(11):27842795 2789

  • 7/31/2019 2006 Bagchi Edn2 PGR Ovulation Mol Endo

    7/12

    possibility that it may mediate some of the effects ofthis receptor during ovulation. We, therefore, investi-gated whether certain of the known PGR-regulatedgenes in granulosa cells are also regulated by ET-2. Totest this possibility, we examined the expression of thegene encoding cGMP-dependent protein kinase II

    (cGK II, Prkg2), a molecule recently reported to beregulated by PGR-induced mechanisms (34). Amarked induction of cGK II mRNA was seen in culturedgranulosa cells 12 h after administration of forskolinand PMA (Fig. 7A). As expected, this induction wassuppressed when PGR function was inhibited byCDB-2914. We next examined the effects of the block-ade of the ET-2 pathway on cGK II mRNA expressionin granulosa cells. As shown in Fig. 7B, addition of theETR-B antagonist BQ-788 to cultured granulosa cellsdid not significantly alter the level of ET-2 mRNA. Wenoted, however, a marked reduction in the level of cGKII mRNA in response to BQ-788, indicating that theexpression of this gene is regulated by signaling

    downstream of ET-2 in granulosa cells (Fig. 7C).

    DISCUSSION

    Steroidal antagonists that bind PGR and block its

    gene-regulatory activity are powerful tools to identify

    the molecular pathways that mediate the function of

    this receptor in an adult target tissue (23). In this study,

    we used CDB-2914, a novel PGR-selective antago-nist, to identify the gene networks that are regulated

    by this receptor during ovulation. Compared with

    RU486, a well-known and widely used PGR antago-

    nist, CDB-2914 displays improved specificity and ef-

    ficacy due to its lower antiglucocorticoid activity and

    better binding affinity to PGR (17, 18). Previous studies

    in the rat showed that CDB-2914 displays dose-de-

    pendent antiovulatory activity when administered on

    the day of proestrus (16). In the present study, we

    found that administration of CDB-2914 to mice under-

    going gonadotropin-induced superovulation effec-

    tively blocked the release of oocytes from the ovary,

    presumably by inhibiting PGR-dependent pathways.

    Fig. 6. Mice Treated with an ETR-B Antagonist Exhibit Impaired Follicular RuptureA, Mice (n 12) were subjected to superovulation and divided into three groups of four animals each. Each group was then

    injected with vehicle or JKC-301 (10 mg/kg BW) or BQ-788 (10 mg/kg BW). The injections (ip) were given at 6 h after hCG

    treatment. The number of released oocytes was counted 18 h after hCG administration. The data are represented as means

    SEM. B, Mice (n 4 at each time point) were subjected to superovulation and treated with BQ-788 (10 mg/kg BW) at indicatedtimes after hCG treatment. Control represents mice treated with vehicle alone at 6 h after hCG administration. The number ofreleased oocytes was counted 18 h after hCG treatment. C, Hematoxylin and eosin-stained ovarian sections of mice treated withvehicle (left panel) or BQ-788 (right panel) at 8 h after hCG injection. The ovarian sections were collected at 18 h after hCGadministration. CL, Corpus luteum; UF, unruptured follicles.

    2790 Mol Endocrinol, November 2006, 20(11):27842795 Palanisamy et al. P Regulation & Function of ET-2 in the Ovary

  • 7/31/2019 2006 Bagchi Edn2 PGR Ovulation Mol Endo

    8/12

    To obtain a comprehensive understanding of the genenetworks underlying PGR function during ovulation,we performed gene expression profiling in the ovariantissue under conditions in which the activity of thisreceptor is inhibited by CDB-2914. Our study demon-strated that ET-2 expression, which occurs immedi-ately preceding ovulation, is blocked when PGR isoccupied by CDB-2914. Therefore, we identified ET-2as a downstream target of PGR regulation during theovulatory process.

    The lack of ET-2 expression in the ovaries of PGRnull mice upon gonadotropin stimulation confirmed

    the role of this receptor in ET-2 induction during ovu-

    lation. This conclusion was further strengthened byour in vitro studies using primary cultures of granulosacells. Previous studies showed that addition of fors-kolin and PMA to granulosa cells isolated from PMSG-primed mice mimics LH action and induces Pgr ex-pression (31). In our experiments, treatment of primarycultures of granulosa cells with these reagents led to aremarkable induction of ET-2 mRNA in these cells. NoET-2 mRNA was detected when granulosa cells ob-tained from PRKO mice were treated similarly, indicat-ing that PGR is essential for ET-2 induction. In situhybridization analysis revealed that ET-2 mRNA ex-

    pression occurred only in the mural granulosa cells of

    Fig. 7. ET-2 Regulates Expression of cGK II mRNA in Primary Culture of Granulosa CellsA, Granulosa cells were treated with P, PMA, forskolin (Fo), and indicated concentrations of CDB-2914 for 0 h (column 1) and

    12 h (columns 25). Total RNA was isolated from these cells and analyzed by Q-PCR using gene-specific primers for cGK II and36B4. Expression of cGK II mRNA was normalized against 36B4 mRNA expression. Fold changes were computed relative to theexpression level of cGK II in cells collected from mice at 0 h. B and C, The cells were treated with or without indicatedconcentrations of P, PMA, forskolin, and ETR-B antagonist, BQ788, for 0 h (column 1) and 12 h (columns 2 and 3), and analyzedfor the expression of ET-2 (panel B) and cGK II (panel C). Expression of ET-2 and cGK II mRNA was normalized against 36B4expression. Fold changes were computed relative to the expression level of ET-2 or cGK II in cells collected from mice at 0 h.

    Palanisamy et al. P Regulation & Function of ET-2 in the Ovary Mol Endocrinol, November 2006, 20(11):27842795 2791

  • 7/31/2019 2006 Bagchi Edn2 PGR Ovulation Mol Endo

    9/12

    the preovulatory follicles. These cells are also knownto be the exclusive sites of Pgr expression in thepreovulatory follicles during ovulation (57). Therefore,PGR and ET-2 expression are spatially linked. Ourresults, however, did not reveal whether PGR inducesET-2 expression directly or indirectly. During super-ovulation of mice, PGR expression peaks at 24 h afterhCG stimulation. The peak expression of ET-2, on theother hand, occurs only at 1112 h after hCG treat-ment. Given this difference in their temporal expres-sion patterns, it is possible that PGR regulates ET-2through indirect mechanisms. Direct regulation of atarget gene by PGR involves interaction of the recep-tor with a P response element motif in its regulatoryregion or tethering to a promoter-bound transcriptionfactor (14, 15). An in silico analysis of the 5-regulatoryregion of ET-2 did not reveal any consensus P re-sponse element motif, although interaction via tether-ing to another transcription factor remains a possibil-

    ity. It is also conceivable that PGR induces a mediatormolecule, which in turn regulates the expression ofET-2. Further work would be needed to decipher themechanisms by which PGR regulates ET-2expression.

    The ET family of ligands is composed of ET-1, ET-2,and ET-3 (2022). Interestingly, of these three struc-turally related isoforms, only ET-2 is robustly induceddownstream of LH and PGR during the ovulatory pro-cess in mice. In Northern blotting experiments, wefailed to detect any mRNA signal corresponding toET-1 or ET-3 in the ovary under the superovulationconditions (data not shown). There is an extensive

    literature of the biological roles of ETs in diverse tis-sues (2022). The physiological effects of ETs are me-diated by two distinct seven transmembrane G pro-tein-coupled receptor subtypes, ETR-A and ETR-B(20). Germline mutation of either ETR leads to perinatalor juvenile lethality in the mouse (35, 36). Mice defi-cient in ETR-A exhibited defects in craniofacial andcardiac neural crest development (35). In contrast,mice deficient in ETR-B suffer from severe melanocyteand enteric neuron defects and die as juveniles (36).

    To determine the consequences of loss of ET-2signaling through each receptor during ovulation, weused two selective peptide inhibitors: JKC-301 for

    ETR-A and BQ-788 for ETR-B (32, 33, 36). Thesepeptides structurally resemble ETs and act as com-petitive inhibitors. Systemic administration of BQ-788,the ET-B antagonist, before ovulation led to as muchas 85% reduction in the number of released ova.These results are highly consistent with the recentfindings of Ko et al. (37), who showed that a directintraovarian injection of a small amount of tezosentan,a pan-antagonist of ETR, immediately before ovulationefficiently blocked follicular rupture in the rat. Theseresults strongly implied that the blockade of ovulationby the ETR antagonists is a consequence of disruptionof ET function within the ovary itself rather than asecondary effect due to the inhibitory action of this

    drug in another tissue. The use of ETR-A- and ETR-

    B-specific antagonists in our studies allowed us todissect the individual roles of these receptor subtypesduring ovulation. Our results, indicating that BQ-788 ismore effective than JKC-301 in blocking ovulation,suggest that ETR-B plays a more critical role thanETR-A in mediating the effects of ET-2 during thisprocess. A likely explanation of this finding is the ob-served difference in the expression levels of ETR-Aand ETR-B in the ovary. By the criteria of Northernblotting and immunohistochemical analyses, the ex-pression level of ETR-B appears to be substantiallyhigher than that of ETR-A.

    In contrast to the expression of its ligand ET-2,which is seen only transiently in the preovulatory fol-licles preceding ovulation, ETR-B expression was de-tected in follicles of all stages of development in thePMSG-treated ovary. Its expression increased onlymodestly upon hCG stimulation. Immunohistochemi-cal localization of the ETR-B provided interesting clues

    regarding the site(s) of action and function of ET-2 inthe preovulatory follicle. We found that ETR-B is ex-pressed in three different cell types within the ovary:mural granulosa cells, cumulus cells, and the endo-thelial cells of the wreath of capillaries bordering thetheca interna. Interestingly, the expression of ETR-Bwas more intense in the COC compared with the muralgranulosa cells. We postulate that ET-2 produced bythe mural granulosa cells of the preovulatory folliclesacts in an autocrine or paracrine manner at one ormore sites of ETR-B expression within the ovary tocontrol follicular rupture.

    The existence of ETR-B in the endothelial cells of the

    large capillaries in the theca interna indicates the pos-sibility that ET-2 could regulate the ovulatory processby ETR-mediated vasodilation and increased vascularpermeability. It is believed that certain aspects of theovulatory process resemble an acute inflammatory re-action involving marked changes in permeability of thefollicle-blood barriers, leading to accumulation of fol-licle fluid and increase of intrafollicular pressure (1, 38).Earlier studies showed that in rats subjected to super-ovulation, the ovarian blood volume increases signifi-cantly starting at 4 h after hCG treatment and reachesa peak that is markedly higher than the control level at10 h post hCG near the time of follicular rupture (39).

    This increased blood flow is known to be important forovulation, contributing to the hyperemia and edemathat occur before follicular rupture (40). ETs are knownto signal through a nitric oxide (NO)-mediated path-way. They stimulate endothelial nitric oxide (NO) syn-thase (eNOS) under various physiological conditions,which in turn catalyzes the production of NO fromL-arginine (41, 42). The NO diffuses across the endo-thelium into neighboring smooth muscle and inducesvasodilation. Interestingly, previous studies haveshown that the expression of eNOS required for NOproduction reaches a peak at 12 h after hCG injectionand plays a critical role in ovulation (4346). It is tempt-ing to speculate that ET-2 produced by the mural

    granulosa cells acts on the endothelial cells of the

    2792 Mol Endocrinol, November 2006, 20(11):27842795 Palanisamy et al. P Regulation & Function of ET-2 in the Ovary

  • 7/31/2019 2006 Bagchi Edn2 PGR Ovulation Mol Endo

    10/12

    blood vessels in the theca interna to induce NO sig-naling and in this manner plays a role in the inductionof local vasodilation (Fig. 8).

    Expression of ETR-B in the mural and cumulus gran-ulosa cells presents additional possible mechanismsof ET-2 action during ovulation (Fig. 8). It is conceiv-able that ET-2 secreted by the mural granulosa cells ofthe preovulatory follicles acts back on these samecells in an autocrine manner. The intense expressionof ETR-B in the COC is also intriguing, suggesting aparacrine role of ET-2 at this site. An important findingof our study is the identification of cGK II as a down-stream target of ET-2 in the preovulatory follicle. Re-cent work by Richards and co-workers (34) indicatedthat PGR-dependent pathways are responsible for theinduction of the cGK II gene in the granulosa cells andCOC at the time of ovulation. We propose that ET-2synthesized in response to PGR in the granulosa cellsacts on ETR-B in mural cells in an autocrine manner.

    ET-2 can also act on cumulus cells of the COC in aparacrine manner to induce synthesis of cGK II inthese cells. The functional significance of cGK II ex-pression in mural granulosa cells or COC is presentlyunclear. It is of interest, however, to note that manycomponents of the cGMP pathway are present in thegranulosa cells and activated during LH-induced ovu-lation. Both membrane-bound and soluble guanylatecyclases, which produce cGMP, are expressed inthese cells (30, 47). As mentioned above, NO signal-

    ing, which activates soluble guanylate cyclases, is alsoinduced in the ovarian tissue during ovulation (43). Thefuture challenge is to determine the precise nature ofthe molecular circuitry involving PGR, ET-2, NOS/NO,cGMP-producing enzymes, and cGK II signaling path-way that operates downstream of LH signaling to im-pact on the function of mural granulosa cells and theCOC during the ovulatory process.

    MATERIALS AND METHODS

    Reagents

    CDB-2914 was a generous gift from the Population Council(New York, NY). PMSG and hCG were purchased from SigmaChemical Co. (St. Louis, MO). The ETR inhibitors BQ-788 andJKC-301 were purchased from EMD Biosciences, Inc. (SanDiego, CA) and Alexis Biochemicals (San Diego, CA),

    respectively.

    Animals and Tissue Collection

    All experiments involving animals were conducted in accor-dance with the National Institutes of Health (NIH) standardsfor the use and care of animals and were approved by theInstitutional Animal Care and Use Committee at the Univer-sity of Illinois at Urbana-Champaign.

    To induce superovulation, the immature CD-1, 129Sv, orPRKO mice (2428 d old) were injected ip with 5 IU of PMSGand 48 h later by 5 IU of hCG. The animals were killed atvarious times after hCG injection, and ovaries were collectedfor Northern blot analysis. PRKO were bred and homozy-gotes were confirmed by genotyping as described previously(12, 23).

    To assess the effect of CDB-2914 on superovulation inmice, the compound was dissolved in sesame oil and admin-istered ip at 20 or 40 mg/kg body weight (BW) 1 h before hCGinjection. The oviducts were collected at 18 h after hCGinjection, and oocytes were counted. For histological analy-sis, ovaries were collected and fixed in 4% paraformaldehydeat 4 C. After paraffin embedding, ovarian sections were sub-

    jected to hematoxylin and eosin staining (Sigma ChemicalCo.).

    To confirm Pgr regulation of ovarian gene expression,CDB-2914 was administered ip to mice subjected to super-ovulation 1 h before hCG injection. Ovaries were collectedfrom these mice at 12 h after hCG, and mRNA was extractedand analyzed.

    GeneChip Analysis

    For microarray analysis, RNA samples were processed andanalyzed using mouse Affymetrix GeneChips following the

    Affymetrix protocol as described previously (23).

    RNA Analysis

    Northern blot analysis was performed as described previ-ously (23).

    The in situ hybridization was performed as described pre-viously (24). Briefly, tissues were fixed in 4% paraformalde-hyde at 4 C. Cryostat sections were cut at 8 m and attachedto 3-aminopropyl triethylsilane (Sigma)-coated slides.Digoxygenin (DIG)-labeled sense or antisense RNA probecomplementary to nucleotides 240 to 643 of mouse ET-2cDNA was used. DIG-labeled RNA probes were synthesized

    Fig. 8. A Hypothetical Signaling Network Involving LH/CG,PGR, ET-2, and ETR-B Controls Ovulation

    The pituitary surge of LH/CG before ovulation acts on themural granulosa cells of the preovulatory follicles. Signalingvia LH receptor in these cells induces PGR, which in turnleads to the expression of ET-2. ET-2 acts in an autocrine orparacrine manner via ETR-B on mural and/or cumulus gran-ulosa cells (GC) and endothelial cells of large capillaries toinduce downstream signaling molecules, which include cGKII. LH signaling in the ovary is also known to induce nitricoxide synthases, eNOS and iNOS, which are thought to beinvolved in cGMP production. cGMP activates cGK II, whichphosphorylates as yet unknown downstream molecules thatare likely to participate in pathways that control follicularrupture. iNOS, Inducible nitric oxide synthase.

    Palanisamy et al. P Regulation & Function of ET-2 in the Ovary Mol Endocrinol, November 2006, 20(11):27842795 2793

  • 7/31/2019 2006 Bagchi Edn2 PGR Ovulation Mol Endo

    11/12

    from the cDNAs using T3 or T7 RNA polymerase and DIG-labeled nucleotides, according to the manufacturers speci-fications (Roche Applied Science, Indianapolis, IN).

    Immunohistochemistry

    Polyclonal antibodies against ETR-A and ETR-B receptorswere purchased from Alexis Chemical (Carlsbad, CA) andCalbiochem (La Jolla, CA), respectively, and diluted 1:500 forimmunohistochemistry. The antibodies were raised againstC-terminal peptide of rat ET-A and ET-B receptors and re-acted with rat, mouse, and human antigens. Paraffin-embed-ded ovarian tissues were sectioned at 4 m and mounted onslides. Sections were washed in PBS for 20 min and thenincubated in a blocking solution containing 10% normal rab-bit serum for 10 min before incubation in primary antibodyovernight at 4 C. Immunostaining was performed using Avi-din-Biotin kit for rabbit primary antibody (Vector Laboratories,Burlingame, CA) and the diaminobenzidine chromogen. Sec-tions were counterstained with hematoxylin, mounted, andexamined under bright field. Red deposits indicate the sitesof immunostaining.

    Inhibitor Studies

    The ETR inhibitors JKC-301 (10 mg/kg BW) and BQ-788 (10mg/kg BW) were administered ip to mice subjected to super-ovulation at 4, 6, and 8 h after hCG injection. The oviductswere collected at 18 h after hCG injection, and oocytes werecounted. The ovaries from mice treated with BQ-788 werealso collected for histological analysis.

    Statistical Analysis

    Thedata were rank transformed, and ANOVA (25) followed byDunnetts one-sided test against the control were performed(26). Data are reported as mean SD. P 0.05 was consid-

    ered as statistically significant.

    Acknowledgments

    The PRKO mice were kindly provided by F. J. DeMayo ofBaylor College of Medicine (Houston, TX).

    Received February 22, 2006. Accepted July 24, 2006.Address all correspondence and requests for reprints to:

    Indrani C. Bagchi, Department of Veterinary Biosciences,University of Illinois at Urbana-Champaign, Urbana, Illinois61802. E-mail: [email protected].

    This work was supported by National Institutes of Health(NIH) Grants U54 HD 299901-12 (to I.C.B. and R.L.S.-W.),

    and R01-HD-044611 (to M.K.B.). This investigation was con-ducted in a facility constructed with support from ResearchFacilities Improvement Program Grant C06 RR16515-01 fromthe National Center for Research Resources, NIH (to I.C.B.).

    Disclosure Statement: The authors have nothing todeclare.

    REFERENCES

    1. Espey LL 2001 An overview of 37 years of research onovulation. In: Adashi EY, ed. Ovulation: evolving scientificand clinical concepts. New York: Springer-Verlag; 116

    2. Richards JS, Russell DL, Ochsner S, Espey LL 2002Ovulation: new dimensions and new regulators of theinflammatory-like response. Annu Rev Physiol 64:6992

    3. Richards JS 2001 Perspective: the ovarian follicleaperspective in 2001. Endocrinology 142:21842193

    4. Richards JS 2005 Ovulation: new factors that prepare theoocyte for fertilization. Mol Cell Endocrinol 234:7579

    5. Park OK, Mayo KE 1991 Transient expression of proges-terone receptor messenger RNA in ovarian granulosacells after the preovulatory luteinizing hormone surge.

    Mol Endocrinol 5:9679786. Park-Sarge OK, Mayo KE 1994 Regulation of the proges-

    terone receptor gene by gonadotropins and cyclic aden-osine 3,5-monophosphate in rat granulosa cells. Endo-crinology 134:709718

    7. Robker RL, Russell DL, Espey LL, Lydon JP, OMalleyBW, Richards JS 2000 Progesterone-regulated genes inthe ovulation process: ADAMTS-1 and cathepsin L pro-teases. Proc Natl Acad Sci USA 97:46894694

    8. Mori T, Suzuki A, Nishimura T, Kambegawa A 1977 In-hibition of ovulation in immature rats by anti-progester-one antiserum. J Endocrinol 73:185186

    9. Snyder BW, Beecham GD, Schane HP 1984 Inhibition ofovulation in rats with epostane, an inhibitor of 3 -hy-droxysteroid dehydrogenase. Proc Soc Exp Biol Med176:238242

    10. Loutradis D, Bletsa R, Aravantinos L, Kallianidis K,Michalas S, Psychoyos A 1991 Preovulatory effects ofthe progesterone antagonist mifepristone (RU486) inmice. Hum Reprod 6:12381240

    11. Pall M, Mikuni M, Mitsube K, Brannstrom M 2000 Time-dependent ovulation inhibition of a selective progester-one-receptor antagonist (Org 31710) and effects on ovu-latory mediators in the in vitro perfused rat ovary. BiolReprod 63:16421647

    12. Lydon JP, DeMayo FJ, Funk CR, Mani SK, Hughes AR,Montgomery Jr CA, Shyamala G, Conneely OM,OMalley BW 1995 Mice lacking progesterone receptorexhibit pleiotropic reproductive abnormalities. GenesDev 9:22662278

    13. Conneely OM, Mulac-Jericevic B, DeMayo F, Lydon JP,OMalley BW 2002 Reproductive functions of progester-one receptors. Recent Prog Horm Res 57:339355

    14. Tsai MJ, OMalley BW 1994 Molecular mechanisms ofaction of steroid/thyroid receptor superfamily members.

    Annu Rev Biochem 63:45148615. Li X, OMalley BW 2003 Unfolding the action of proges-

    terone receptors. J Biol Chem 278:392613926416. Reel JR, Hild-Petito S, Blye RP 1998 Antiovulatory and

    postcoital antifertility activity of the antiprogestin CDB-2914 when administered as single, multiple, or continu-ous doses to rats. Contraception 58:129136

    17. Attardi, BJ, Burgenson J, Hild SA, Reel JR, Blye RP 2002CDB-4124 and its putative monodemethylated metabo-lite, CDB-4453, are potent antiprogestins with reducedantiglucocorticoid activity: in vitro comparison to mife-pristone and CDB-2914. Mol Cell Endocrinol 188:111123

    18. Wagner BL, Pollio G, Giangrande P, Webster JC, BreslinM, Mais DE, Cook CE, Vedeckis WV, Cidlowski JA, Mc-Donnell DP 1999 The novel progesterone receptor an-tagonists RTI 3021012 and RTI 3021022 exhibit com-plex glucocorticoid receptor antagonist activities:implications for the development of dissociated antipro-gestins. Endocrinology 140:14491458

    19. Blithe DL, Nieman LK, Blye RP, Stratton P, Passaro M2003 Development of the selective progesterone recep-tor modulator CDB-2914 for clinical indications. Steroids68:10131017

    20. Rubanyi GM, Polokoff MA 1994 Endothelins: molecularbiology, biochemistry, pharmacology, physiology, andpathophysiology. Pharmacol Rev 46:325415

    21. Goto K 2001 Basic and therapeutic relevance of endo-thelin-mediated regulation. Biol Pharm Bull 24:12191230

    2794 Mol Endocrinol, November 2006, 20(11):27842795 Palanisamy et al. P Regulation & Function of ET-2 in the Ovary

  • 7/31/2019 2006 Bagchi Edn2 PGR Ovulation Mol Endo

    12/12

    22. Kusserow H, Unger T 2004 Vasoactive peptides, theirreceptors and drug development. Basic Clin PharmacolToxicol 94:512

    23. CheonYP, Li Q, Xu X, DeMayo FJ, Bagchi IC, Bagchi MK2002 A genomic approach to identify novel progesteronereceptor regulated pathways in the uterus during implan-tation. Mol Endocrinol 16:28532871

    24. Zhu LJ, Cullinan-Bove K, Polihronis M, Bagchi MK, Bag-chi IC 1998 Calcitonin is a progesterone-regulatedmarker that forecasts the receptive state of endometriumduring implantation. Endocrinology 139:39233934

    25. Conover WV, Iman RL 1981 Rank transformations as abridge between parametric and non-parametric statis-tics. Am Stat 35:124129

    26. Dunnett CW 1955 A multiple comparision procedure forcomparing several treatments with a control. J Am Stat

    Assoc 50:1096112127. Yanagisawa M, Kurihara H, Kimura S, Tomobe Y, Koba-

    yashi M, Mitsui Y, Yazaki Y, Goto K, Masaki T 1988 Anovel potent vasoconstrictor peptide produced by vas-cular endothelial cells. Nature 332:411415

    28. Saida K, Mitsui Y, Ishida N 1989 A novel peptide, vaso-active intestinal contractor, of a new (endothelin) peptidefamily. Molecular cloning, expression, and biological ac-tivity. J Biol Chem 264:1461314616

    29. Bloch KD, Hong CC, Eddy RL, Shows TB, QuertermousT 1991 cDNA cloning and chromosomal assignment ofthe endothelin 2 gene: vasoactive intestinal contractorpeptide is rat endothelin 2. Genomics 10:236242

    30. Shi F, Stewart RL, Perez E, Chen JY, LaPolt PS 2004Cell-specific expression and regulation of soluble gua-nylyl cyclase 1 and 1 subunits in the rat ovary. BiolReprod 70:15521561

    31. Robker RL, Russell DL, Yoshioka S, Sharma CS, LydonJP, OMalley BW, Espey LL, Richards JS 2000 Ovulation:a multi-gene, multi-step process. Steroids 65:559570

    32. Webb ML, Meek TD 1997 Inhibitors of endothelin. MedRes Rev 17:1767

    33. Ishikawa K, Ihara M, Noguchi K, Mase T, Mino N, SaekiT, Fukuroda T, FukamiT, Ozaki S, Nagase T, Nishikibe M,Yano M 1994 Biochemical and pharmacological profile ofa potent and selective endothelin B-receptor antagonist,BQ-788. Proc Natl Acad Sci USA 91:48924896

    34. Sriraman V., Rudd MD, Lohmann SM, Mulders SM, Rich-ards JS 2006 Cyclic guanosine 5-monophosphate-de-pendent protein kinase II is induced by luteinizing hor-mone and progesterone dependent mechanisms ingranulosa cells and cumulus oocyte complexes of ovu-lating follicles. Mol Endocrinol 20:348361

    35. Clouthier DE, Hosoda K, Richardson JA, Williams SC,Yanagisawa H, Kuwaki T, Kumada M, Hammer RE,Yanagisawa M 1998 Cranial and cardiac neural crestdefects in endothelin-A receptor-deficient mice. Devel-opment 125:813824

    36. Hosoda K, Hammer RE, Richardson JA, Baynash AG,Cheung JC, Giaid A, Yanagisawa M 1994 Targeted and

    natural (piebald lethal) mutations of endothelin-B recep-tor gene produce megacolon associated with spottedcoat color in mice. Cell 79:12671276

    37. Ko C, Gieske MC, Al-Alem L, Hahn YK, Su W, Gong MC,Iglarz M, Koo Y 2006 Endothelin-2 in ovarian folliclerupture. Endocrinology 147:17701779

    38. Espey LL 1994 Current status of the hypothesis thatmammalian ovulation is comparable to an inflammatoryreaction. Biol Reprod 50:233238

    39. Tanaka N, Espey LL, Okamura H 1989 Increase in ovar-ian blood volume during ovulation in the gonadotropin-primed immature rat. Biol Reprod 40:762768

    40. Espey LL, Lipner H 1994 Ovulation. In: Knobil E, Neill JD,eds. The physiology of reproduction. New York: RavenPress; 725780

    41. Zhang M, Luo B, Chen SJ, Abrams GA, Fallon MB 1999

    Endothelin-1 stimulation of endothelial nitric oxide syn-thase in the pathogenesis of hepatopulmonary syn-drome. Am J Physiol 277:G944G952

    42. Marrero MB, Venema VJ, Ju H, He H, Liang H, CaldwellRB, Venema RC 1999 Endothelial nitric oxide synthaseinteractions with G-protein-coupled receptors. BiochemJ 3453:235240

    43. Jablonka-Shariff A, Olson LM 1997 Hormonal regulationof nitric oxide synthases and their cell-specific expres-sion during follicular development in the rat ovary. En-docrinology 138:460468

    44. Shukovski L, Tsafriri A 1994 The involvement of nitricoxide in the ovulatory process in the rat. Endocrinology135:22872290

    45. Nakamura Y, Kashida S, Nakata M, Takiguchi S, Yama-gata Y, Takayama H, Sugino N, Kato H 1999 Changes in

    nitric oxide synthase activity in the ovary of gonadotropintreated rats: the role of nitric oxide during ovulation.Endocr J 46:529538

    46. Faletti A, Perez Martinez S, Perotti C, de Gimeno MA1999 Activity of ovarian nitric oxide synthase (NOs) dur-ing ovulatory process in the rat: relationship with pros-taglandins (PGs) production. Nitric Oxide 3:340347

    47. Huang H, Acuff CG, Steinhelper ME 1996 Isolation, map-ping, and regulated expression of the gene encodingmouse C-type natriuretic peptide. Am J Physiol.271:H1565H1575

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

    Palanisamy et al. P Regulation & Function of ET-2 in the Ovary Mol Endocrinol, November 2006, 20(11):27842795 2795