Open Access
How to translate text using browser tools
1 April 2013 Recent Advances in the Understanding of Teleost Medaka Ovulation: The Roles of Proteases and Prostaglandins
Takayuki Takahashi, Chika Fujimori, Akane Hagiwara, Katsueki Ogiwara
Author Affiliations +
Abstract

Ovulation is the process of liberating oocytes from the preovulatory follicles, and is observed in the ovaries of virtually all female vertebrate animals. Compared with mammalian species, there have been far fewer studies that address the ovulatory mechanisms of non-mammalian species. We have examined the molecular mechanism of follicle rupture during ovulation using the teleost model, medaka, or Oryzias latipes. Follicle rupture in medaka ovulation involves the cooperation of the tissue inhibitor of metalloproteinase-2b protein with at least three matrix metalloproteinases (MMP): membrane type-1 MMP (MT1-MMP), MT2-MMP, and gelatinase A. Our studies also indicate that the serine protease, i.e., plasmin, participates in the rupture for only a few hours prior to the activation of MMP-mediated hydrolysis at ovulation. The involvement of prostaglandin E2 (PGE2) in medaka ovulation was also demonstrated. Cyclooxygenase-2 and PGE2 receptor subtype EP4b were respectively shown to be an enzyme responsible for PGE2 synthesis and a receptor for the generated ligand in the preovulatory follicles. Based on the results obtained from our studies of fish, we discuss the similarities and differences in vertebrate ovulation compared with mammalian species.

INTRODUCTION

Ovulation is a complex process that eventually results in the liberation of the oocytes from the preovulatory follicles that grow in the cortex of the ovary. This process is induced by a gonadotropin, i.e., luteinizing hormone (LH), which is delivered from the pituitary gland of vertebrates. In response to an LH surge, various changes occur in the follicular compartments of the preovulatory follicles; meiosis resumes in the oocyte to complete a series of events called oocyte maturation, while follicular cells surrounding the oocyte are activated to produce a variety of biologically active factors and proteins that are required for successful ovulation (Richards et al., 1998; Nagahama and Yamashita, 2008). The term “ovulation” is generally used for the entire process of follicular responses to LH, and the rupture of the follicle wall upon ovulation is one of the follicular responses (Tsafriri and Dekel, 1994). In this article, we follow these generally accepted concepts of “ovulation” and “follicle rupture”.

Historically, follicle rupture during ovulation in mammalian species was thought to be accomplished by the physical breakdown of the follicle wall in the apical region of the follicle due to increased intrafollicular pressure. However, this hypothesis was rejected in the early 1960s as it became clear that the follicles nearing ovulation do not experience significantly increased pressure (Espey and Lipner, 1994). The hypothesis that the rupture is a result of restricted proteolysis occurring at the apical region of the follicles has since gained support. Interestingly, the involvement of proteolytic enzymes in follicle rupture during ovulation was first suggested in 1916 (Schochet, 1916). Using mammalian species, a number of investigations addressing the roles of proteolytic enzymes in follicle rupture have since been conducted (Ohnishi et al., 2005; Espey and Richards, 2006; Curry TE and Smith, 2006). The results of these studies apparently indicate that proteases, especially matrix metalloproteinases (MMPs), play a role in follicle rupture during ovulation in mammals (Curry TE and Smith, 2006; Espey and Richards, 2006). However, studies of mice using gene knockouts of candidate proteases failed to demonstrate essential roles for these proteases in follicle rupture. Thus, it remains to be established whether proteases play indispensable roles in follicle rupture during ovulation in mammals. Nevertheless, our overall knowledge of mammalian ovulation has greatly advanced over the last several decades, and has aided in exploring the mechanisms that govern ovulation in non-mammalian vertebrates.

Over the past ten years, our research group has been studying ovulation using the teleost medaka as a non-mammalian vertebrate model. The aims of our study are 1) to understand to what extent the molecular mechanisms of ovulation may be conserved throughout vertebrates and 2) to approach important unsolved problems that are difficult to clarify using mammalian experimental systems. In the present review, we highlight the progress towards understanding follicle rupture during ovulation in medaka. We propose a “two-step extracellular matrix hydrolysis model,” in which both matrix metalloproteinases and serine proteinases play critical roles in follicle rupture. In addition, the involvement of prostaglandin E2 (PGE2) and its receptor in follicle rupture during ovulation in fish are discussed.

Oocyte maturation and ovulation in medaka

The medaka, Oryzias latipes, is a small freshwater teleost that offers advantages for use in genetics, developmental and reproductive biology, physiology, and toxicology (Iwamatsu et al., 1988; Nagahama, 1994; Ozato et al., 1992; Ishikawa, 2000; Wittbrodt et al., 2002; Kasahara et al., 2007). In particular, this non-mammalian vertebrate species has emerged as a powerful tool for the elucidation of reproductive processes, including the molecular mechanisms of ovulation. The fish usually spawn daily within 1 h of the onset of light for a number of consecutive days when maintained at ambient temperature (26°C) under a constant long photoperiod of 14 h light and 10 h dark. Using this method, the sequence of events leading to spawning, such as the completion of vitellogenesis, germinal vesicle breakdown and ovulation, can be timed accurately (Iwamatsu, 1978). Previous studies have elucidated the endocrinological background behind such reproductive events (Sakai et al., 1987; Sakai et al., 1988). In addition, the large follicles that are to ovulate on the next day are demonstrated to undergo a surge of gonadotropin at approximately 15–21 h before the expected time of ovulation (Iwamatsu, 1978). In this fish, germinal vesicle breakdown (GVBD), a critical process for oocyte maturation, occurs approximately 6 h before ovulation in the follicle that is destined to ovulate in vivo (Iwamatsu, 1978).

In the medaka, the ovary is a sac-like organ surrounded by an outermost thin layer that separates the ovary from the body cavity. The body of the ovary is surrounded by the surface germinal epithelium and contains growing follicles of various sizes (Fig. 1). A space between the outermost thin layer and the germinal epithelium of the ovary, called the ovarian cavity, is formed. Upon in vivo ovulation, oocytes are released from the body of the ovary into the ovarian cavity. In this review, we use the term “in vivo ovulation” to refer to the release of oocytes from the ovary body into the cavity. As we describe below, ovaries isolated from medaka or large preovulatory follicles dissected from the body of the fish ovary are employed for in vitro ovulation experiments. The term “in vitro ovulation” is used for oocytes' detaching from the ovarian follicle.

Medaka in vitro ovulation model

In vitro ovulation experiments can be performed using not only whole ovaries (Ogiwara et al., 2010) but also large preovulatory follicles dissected from the ovaries of the spawning fish (Schroeder and Pendergrass, 1976; Ogiwara et al., 2005). In vitro follicle ovulation experiments under various conditions have been used in our laboratory and are summarized in Fig. 2. For the preovulatory follicles that have undergone an LH surge in vivo between 15 and 21 h before ovulation (Iwamatsu, 1978), we often isolate the follicles 12 or 3 h before the expected time of ovulation. The follicles isolated in this manner spontaneously ovulate in vitro without requiring the addition of recombinant medaka LH to the culture medium. Compared with the in vivo situation, in vitro ovulation of the follicles takes a few more hours. Further, we have recently established an in vitro ovulation method for large preovulatory follicles isolated from the ovary prior to LH-priming in vivo (Ogiwara et al., 2013); the follicles successfully ovulate when cultured in the presence of recombinant medaka LH. In our in vitro follicle culture supplemented with recombinant medaka LH, we isolate the follicles from the ovary 22 h before ovulation, which is the time before the endogenous gonadotropin surge. These cultured follicles undergo GVBD and ovulation with a delay of approximately 3 and 8 h, compared with follicles that ovulate in vivo. Despite the delay in the timing of oocyte maturation and follicle ovulation in the LH-induced in vitro culture system, our in vitro follicle culture system has proven to be a useful experimental model for studying the ovulatory process in medaka.

Fig. 1.

Schematic representation of the medaka ovary. The medaka ovary is composed of apparently symmetric right and left parts that are connected in the center. The whole body of the ovary is wrapped by a thin layer that does not allow ovulated oocytes to escape out of the ovary. Ovulated oocytes move to the posterior cloaca and are eventually spawned. OC, ovarian cavity; Ov-Oc, ovulated oocyte, Fc, follicle; GE, germinal epithelium; and Ovd, oviduct.

f01_239.jpg

Fig. 2.

In vitro culture methods established for medaka preovulatory follicles. Mature female medaka acclimated to artificial reproductive conditions (photoperiod, 10-h dark/ 14-h light; temperature, 27°C) ovulate in vivo on a 24-h cycle at the start of the light period. The timings of the LH surge and GVBD in vivo are also shown. In our in vitro follicle culture system, preovulatory follicles are isolated from the fish ovary 22, 12, or 3 h before the expected time of ovulation. Incubation of the follicles isolated 22 h before ovulation is conducted in the presence of recombinant medaka LH, while the follicles isolated 12 or 13 h before ovulation are incubated without recombinant medaka LH. For each in vitro incubation, the timings of GVBD and ovulation are indicated.

f02_239.jpg

To date, relevant in vitro methods using ovary fragments and ovarian follicles have been established for many teleost species. These species include zebrafish (Li et al., 1993; Liu and Ge, 2002; Lister and Van Der Kraak, 2010), Atlantic croaker (Patino and Thomas, 1990; Tubbs et al., 2010), rainbow trout (Bobe et al., 2004; Crespo et al., 2010), brook trout (Goetz et al., 1982), goldfish (Kagawa and Nagahama, 1981; Goetz, 1993), sea lamprey (Gazourian et al., 1997), Coho salmon (Luckenbach et al., 2010), European sea bass (Sorbera et al., 2001), and killifish (Raldua et al., 2005). These experimental models generally serve as good systems for studying oocyte maturation. However, in these teleost species, mature, healthy and intact oocytes cannot come off the follicle or ovarian fragments even if they have been primed by gonadotropins in vivo. To the best of our knowledge, the in vitro culture method using medaka preovulatory follicles is currently the only experimental system useful for both oocyte maturation and ovulation studies.

Table 1.

Expression of proteolytic enzymes and the inhibitors in the medaka ovary.

t01_239.gif

Follicle rupture by two-step ECM hydrolysis mechanism

There are clear differences in the tissue structures of ovarian follicles in mammalian and non-mammalian species. The large follicle in mammals consists of a round oocyte and two types of somatic cells: the granulosa cells and the theca cells. Some of the granulosa cells surround the oocyte and form the cumulus oocyte complex (COC), which protrudes toward the interior of an antrum filled with follicular fluid. The remaining granulosa cells are positioned just below the basement membrane in multiple cell layers known as the membrane granulosa. Theca cells, which are present on the outside of the basement membrane, also exist in multiple cell layers enriched with extracellular matrix (ECM) components. In contrast, a common tissue structure observed in the large follicles of non-mammalian species consists of a single layer of granulosa cells surrounding an oocyte, a single layer of theca cells, and a basement membrane between the two somatic cell layers. No COC is formed during folliculogenesis of non-mammalian vertebrate animals. Despite what appear to be differences in the follicular tissue structure in mammalian and non-mammalian species, the degradation of ECM components present in the extracellular space of the follicle layer of the preovulatory follicle in a regulated manner is a common requirement for successful ovulation. Previous studies have established that collagen type I is abundantly present in the tunica albuginea and theca externa of fully grown follicles in mammalian ovaries (Espev, 1967), while collagen type IV is oresent in the basement membrane separating the granulosa and theca cell layers (Berkholtz et al., 2006; Lind et al., 2006). Our recent work and that of another laboratory using the teleost medaka (Horiguchi et al., 2008; Kato et al., 2010) and Prochilodus argenteus (Santos et al., 2008) indicate that, as in mammalian species, collagens type I and IV are localized in the theca cell layer and basement membrane of the large preovulatory follicle, respectively. Thus, in terms of ECM degradation associated with follicle rupture during ovulation, a similar, if not identical, mechanism involving proteolytic enzymes capable of hydrolyzing ECM components is highly expected to operate across the vertebrates.

A variety of proteolytic enzymes are expressed in the medaka ovary (Table 1). In our attempts to identify the proteolytic enzymes responsible for follicle rupture during ovulation, various protease inhibitors were tested in preovulatory follicles using the in vitro ovulation system. Serine protease inhibitors, such as aprotinin, leupeptin, antipain and soybean trypsin inhibitor, and MMP inhibitors, such as EDTA, o-phenanthroline and GM6001, strongly inhibited follicle ovulation in vitro (Ogiwara et al., 2005; Ogiwara et al., 2012), suggesting the involvement of two different proteolytic enzyme systems. Our detailed study demonstrated that the serine protease plasmin plays an indispensable role in follicle rupture during medaka ovulation (Ogiwara et al., 2012). Interestingly, plasmin participates in the rupture for only a few hours prior to the activation of MMP-mediated hydrolysis at ovulation. We have proposed a sequential two-step ECM hydrolysis mechanism for follicle rupture in medaka ovulation (Fig. 3). In the first step of ECM hydrolysis, which occurs approximately 5–7 h before ovulation, active plasmin is produced by the proteolytic processing of liver-derived precursor plasminogen in the preovulatory follicle that is destined to ovulate. More recently, we have found that active plasmin is capable of hydrolyzing laminin, a major ECM component constituting the basement membrane, in vivo as well as in vitro (Our unpublished results). Only a few hours of detectable active plasmin in the follicle suggests the presence of well-regulated mechanisms for plasminogen activation. Participation of a plasminogen activator inhibitor(s) in this process is highly likely. As a second step, approximately 0–3 h before ovulation, another proteolytic system involving MT1-MMP, MT2-MMP and gelatinase A is activated for further ECM degradation events. Gelatinase A, which is activated by MT1-MMP, hydrolyzes type IV collagen, a principle component of the basement membrane, and MT2-MMP degrades the type I collagen present in the theca cell layer (Ogiwara et al., 2005). The activation of gelatinase A by MT1-MMP occurs in the plasma membrane of the ovulating oocyte, and this activation process is regulated by the tissue inhibitor of metalloproteinase-2b (TIMP-2b) (Ogiwara et al., 2005).

Fig. 3.

A two-step ECM hydrolysis model of follicle rupture during ovulation in the medaka. In the preovulatory follicle, ECM proteins of the follicle layer are intact before proteolytic systems are activated (A). At 5–7 h before ovulation, the urokinase-type plasminogen activator (uPA)/plasmin proteolytic system is activated to hydrolyze laminin, a major component of the basement membrane (B). Subsequently, the MMP proteolytic system involving MT1-MMP, MT2-MMP and gelatinase A is activated. Gelatinase A activated by MT1-MMP hydrolyzes collagen type IV, another major ECM protein of the basement membrane, while MT2-MMP degrades collagen type I, the ECM protein residing in the GE/TC layer of the follicle (C). GE, germinal epithelium; TC, theca cell; BM, basement membrane; GC, granulosa cell; EM, egg membrane; and Oc, oocyte.

f03_239.jpg

Fig. 4.

Calculation of degraded surface area in follicles relative to the total surface area upon ovulation. (A) The process by which the oocyte of the ovulating follicle ruptures is shown. A yellow arrow indicates the site of follicle layer degradation. A red arrowhead indicates a hole remained in the follicle that had lost the oocyte by ovulation. Bars indicate 1 mm. A schematic representation of the process is also shown at the right. (B) The surface area possibly degraded upon in vitro follicle ovulation is represented by hatched lines. To calculate the hatched area size relative to the total surface area of the follicle, the equation P = 50(1-cos θ) was used. The values of θ were experimentally determined. FL, follicle layer; Oc, oocyte.

f04_239.jpg

To what extent are the follicle layers of periovulatory follicles degraded for successful ovulation? This value was estimated on the basis of morphological observations of the follicles that ovulated in vitro. In vitro follicle ovulation starts with the appearance of a small hole on the surface of spherical follicles around the vegetal pole (Fig. 4A). The hole then enlarges with a concomitant appearance on the surface of the ovulating oocyte that is covered up with a thin layer of follicle cells before the start of ovulation. When the hole reaches a certain size, the oocyte frees itself from the follicle layer. The oocyte, which is just about to come off the follicle layer, becomes dumbbell-shaped, indicating that an extensive degradation of the follicle layer may not be necessary for in vitro medaka follicle ovulation. We determined the extent to which the follicle layer of ovulating follicles could be spatially deteriorated upon ovulation using the equation P = 50(1-cos θ), where P is the percent of the degrading surface area relative to the total surface area (Fig. 4B). Values of θ could be determined by morphological observations of in vitro ovulating follicles. The θ values were found to be 54.6 ± 3.4 (the mean ± SEM of six independent determinations, n = 6), given that P = 20.9 ± 1.8 (n = 6). These results indicate that follicle ovulation could occur by the dissolution of as little as 1/5 of the total ECM components in the layer of a fully-grown spherical follicle.

We have recently found that the treatment of preovulatory follicles prior to LH surge with recombinant medaka LH in the in vitro experimental system drastically induces the expression of MT2-MMP, but not gelatinase A, MT1-MMP, or TIMP-2b (our unpublished results). Our data also indicate that induction of MT2-MMP may be mediated by nuclear progesterone receptor (nPR) (Ogiwara et al., 2013).

INVOLVEMENT OF PROSTAGLANDINS IN OVULATION

Prostaglandins, prostaglandin synthesis, and the receptors in teleosts

Prostaglandins (PGs) play roles in a wide range of physiological processes (Simmons et al., 2004; Sugimoto and Narumiya, 2007). PGs are produced from arachidonic acid through the sequential actions of cyclooxygenase (COX) and specific PG synthases. Previous studies have established that COX plays a key regulatory role in PG synthesis. In mammals, two COX paralogs, a constitutive (COX-1) and inducible enzyme (COX-2), have been identified. In contrast, teleosts have additional copies of COX-1 and/or COX-2. For example, the medaka genome contains two COX-1 genes (ptgs1a and ptgs1b) and one COX-2 gene (ptgs2). This fact is thought to be the result of a teleost-specific genome duplication and subsequent genome loss event (Jarving et al., 2004; Ishikawa and Herschman, 2007; Ishikawa et al., 2007; Havird et al., 2008). It is generally accepted that PGs have a fundamental role in the mechanism of ovulation (Espey and Richards, 2006). Recent studies using mice lacking the gene encoding COX-2 or the PGE2 receptor EP2 have elucidated the role of PGs in the process of cumulus oocyte complex (COC) expansion during ovulation (Hizaki et al., 1999; Richards et al., 2002).

As in mammalian species, ovarian PG synthesis in non-mammalian vertebrates is known to occur during spontaneous or artificially-induced ovulation. In some teleosts, indomethacin, which is a non-selective inhibitor of COX, has been reported to effectively block ovulation (Cetta and Goetz, 1982; Patino et al., 2003; Lister and Van Der Kraak, 2008). Other investigations have reported that PGs induce in vivo and in vitro ovulation (Jalabert and Szollosi, 1975; Stacey and Pandey, 1975; Goetz and Theofan, 1979; Kagawa and Nagahama, 1981; Goetz and Nagahama, 1985; Pankhurst, 1985; Kagawa et al., 2003; Lister and Van Der Kraak, 2008). The possible involvement of PGs in ovulation has also been documented for amphibians (Schuetz, 1986; Chang et al., 1995; Chang et al., 1997; Ramos et al., 2008; Sena and Liu, 2008). These previous studies strongly suggest that PGs have a conserved role in ovulation in vertebrates, including teleost fish. Generally, the COC is formed only for the grown ovarian follicles of mammalian vertebrates; the role of PGs in the expansion of the COC in preovulatory follicles is not applicable to non-mammalian vertebrate species.

In teleosts, the particular molecular species of PGs involved in ovulation appears to differ by species. PGF and PGE2 are the two major PGs that are thought to control fish ovulation (Stacey and Pandey, 1975; Goetz and Theofan, 1979). PGF and its metabolite 15-keto-PGF are well known to be postovulatory prostaglandin pheromones (Sorensen et al., 1988; Sorensen and Goetz, 1993; Stacey and Sorensen, 2002; Munakata and Kobayashi, 2010) that trigger female sexual behavior in a variety of externally fertilizing species. A close association between ovulation and PGF was reported for rainbow trout (Jalabert and Szollosi, 1975), carp (Epler et al., 1985), brook trout (Goetz et al., 1982), and goldfish (Stacey and Pandey, 1975; Sorensen et al., 1988), while PGE2 was found to play a dominant role in ovulation for yellow perch (Goetz and Theofan, 1979) and medaka (Fujimori et al., 2011 ; Fujimori et al., 2012).

The presence of PGF and/or PGE2 was demonstrated by direct measurement using the ovaries of zebrafish (Lister and Van Der Kraak, 2008; Lister and Van Der Kraak, 2009), yellow perch (Berndtson et al., 1989; Goetz, 1997), goldfish (Goetz, 1991), European sea bass (Sorbera et al., 2001), brook trout (Cetta and Goetz, 1982; Goetz, 1991), and medaka (Fujimori et al., 2011). Recently, the PGE2 receptor subtypes EPs, EP1, EP2, EP3, and EP4, as well as a PGF receptor (FP) from zebrafish (Villablanca et al., 2007; Kwok et al., 2012) were characterized. In addition, the expression of EP1, EP2, EP3, and EP4 transcripts in the medaka ovary was examined (Fujimori et al., 2011). Teleost fishes generally contain both EP and FP receptors, while medaka appears to contain only EP receptors. Indeed, our attempt to isolate the FP receptor using medaka tissues was not successful (our unpublished results). FP receptor sequences for zebrafish, fugu, tilapia, cod, coelacanth, and stickleback are available from the Ensembl Genome Database, whereas that of the medaka FP receptor is lacking in the database. Further, no sequence information for medaka PGF synthase, which is responsible for converting PGH2 to PGF, is available from the Ensembl Database. This result may indicate that the medaka fish is incapable of producing PGF, thus lacking the PGF/FP signaling system. Further thorough investigations are required for determining whether the medaka possesses a PGF receptor and/or PGF synthase, however.

Roles of prostaglandins in medaka ovulation

The indispensable role of PGs in medaka ovulation was demonstrated by the inhibition of in vitro follicle ovulation using culture medium containing indomethacin, a COX inhibitor, and GW627338X, an EP4 antagonist (Fujimori et al., 2011). The medaka fish contains three COX genes, ptgs1a, ptgs1b, and ptgs2. Of these, ptgs2 is expressed most abundantly in the ovary (Fujimori et al., 2011). During a 24-h spawning cycle, the ptgs2 mRNA levels in the ovary are fairly constant. Consistent with this finding, ovarian PGE2 levels do not fluctuate in the spawning cycle. This finding was rather surprising because, as established by previous studies using mammalian species (Espey and Richards, 2006) and teleost species (Grosser et al., 2002; Ishikawa et al., 2007; Lister and Van Der Kraak, 2009; Zou et al., 1999; Ishikawa and Herschman, 2007), the expression of COX-2 enzyme was reported to be inducible. As revealed by immunohistochemical analysis using a specific antibody against the medaka COX-2 protein, the follicle layer and oocyte cytoplasm of the large preovulatory follicle contain the protein (Fig. 5). The strongest signal was observed in the theca cells of the follicle, suggesting that the thecal cells predominantly produce PGE2 in the follicles that are destined to ovulate. The EP4b receptor, a subtype of six medaka PGE2 receptors, was expressed dominantly in the fish ovary, and transcripts of the PG receptor were expressed in the follicle cells of large preovulatory follicles. Further, EP4b receptor mRNA expression was drastically induced in the preovulatory follicles as ovulation approached (Fujimori et al., 2012). The expression of EP4b mRNA was inducible in vitro not only by pregnant mare serum gonadotropin (PMSG) (Fujimori et al., 2012) but also by recombinant medaka LH (our unpublished result). We have recently shown that the EP4b antagonist GW627368X completely abolishes the in vitro ovulation of large follicles even when added only 1 h before the time of ovulation (Fujimori et al., 2012). This result suggests that PGE2 functions to induce the ovulation of large preovulatory follicles by binding to the EP4 receptor just before the time of ovulation. Further, this result suggests that PGE2/EP4b signaling is required for fish ovulation at the time that follicle rupture occurs. More recently, we found that nuclear progesterone receptor (nPR) but not membrane progesterone receptor (mPR) is involved in the induction of EP4b expression (Hagiwara et al., unpublished results). Figure 6 shows a model for EP4b expression induced by LH in which the transcription factor nPR is implicated. In this model, we assume that the theca cells of the preovulatory follicle are mainly responsible for the production of PGE2 and that granulosa cells are the cells expressing EP4b receptor at the time of ovulation.

Similarities and differences in ovulation between mammals and medaka

Previous studies on mammalian ovulation have revealed that proteases, prostaglandins, and progesterone are critically involved in the process (Espey and Richards, 2006). Compared with the sheer number of references arguing for their roles in mammalian ovulation, information on the roles of the compounds in ovulation of the teleost medaka is very limited. Nevertheless, existing evidence indicates that they are indispensable for medaka ovulation as well, although clear differences exist in the precise roles and mechanisms in ovulation of the compounds between mammals and medaka.

Fig. 5.

Immunohistochemical localization of COX-2 in the preovulatory follicle of the medaka ovary. Paraffin sections (10 µm) of the mature female medaka ovary were incubated with normal mouse serum (A) or anti-medaka COX-2 serum (B). Signals were detected using an AEC kit (Vector Laboratories, Burlingame, CA), according to the manufacturer's instructions. GE/TC, germinal epithelium/theca cell layer; GC, granulosa cell layer; EM, egg membrane of the oocyte; Oc-cy, oocyte cytoplasm.

f05_239.jpg

Fig. 6.

Endocrine regulation of EP4b expression in the preovulatory follicle of the medaka. The LH surge results in the increased production of steroid hormone 17α-hydroxyprogesterone (17α-HP) via the activation of LH receptors on the granulosa cell (GC). 17αHP is then converted to 17α, 20β-dihydroxyprogesterone (DHP). The gonadotropin surge simultaneously induces the expression of nuclear progesterone receptor (nPR) in the GC. nPR binds to DHP and acts as a critical transcription factor for EP4b gene expression. Translated EP4b receptor protein is expressed on the surface of GC in the follicle that is about to ovulate. The interaction of EP4b with PGE2, which may be generated from arachidonic acid (AR) in the theca cell (TC), evokes an intracellular signal transduction reaction(s) in GC that eventually leads to ovulation. BM, basement membrane; GE, germinal epithelium; EM, egg membrane; Oc, oocyte.

f06_239.jpg

The potential roles in ovulation of many proteases have been studied using mammalian species. Three proteolytic enzyme systems, namely, plasminogen activator (PA)/ plasmin, MMPs, and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) enzymes, have been targets of intensive studies. The results of these studies indicate that MMPs have a significant function in the degradation of the follicle wall (Espey and Richards, 2006; Curry TE and Osteen, 2003). The idea that the PA/plasmin system may be important for ovulation does not appear to be firmly supported (Ny et al., 1999; Leonardsson et al., 1995; Ny et al., 1997; Curry TE and Smith, 2006). Further, the implication of ADAMTS enzymes in ovulation remains to be investigated (Espey and Richards, 2006; Curry TE and Smith, 2006). In medaka ovulation, a sequential action of PA/plasmin and MMPs is required for successful ovulation (Ogiwara et al., 2005; Ogiwara et al., 2012). A significant role of the PA/ plasmin system is unique in fish ovulation because, as described above, recent evidence has argued against an essential role of the proteolytic enzyme system for follicle rupture during ovulation in mammals.

As in mammalian species, COX-2 is responsible for the generation of PGE2 in the preovulatory follicles that are destined to ovulate in the medaka. However, a notable difference in the expression of COX-2 between mammalian species and the teleost medaka is their responsiveness to gonadotropins. In mammals, an LH surge or human chorionic gonadotropin treatment drastically induces the expression of COX-2 in ovarian granulosa cells and cumulus cells (Wong et al., 1989; Joyce et al., 2001; Sirois et al., 2004). In contrast, the medaka counterpart is constitutively expressed (Fujimori et al., 2011), and the expression levels are not affected by gonadotropins such as PMSG and recombinant medaka LH (our unpublished observation). Instead, PGE2 receptor subtype EP4b is readily induced by the treatment of recombinant medaka LH. Thus, it has been concluded that the effect of PGE2 on ovulation is regulated through the expression of EP4b receptor in the preovulatory follicles in the fish. Another clear difference in the role of PGE2 in ovulation between mammalian species and the medaka is that this bioactive compound is involved in the process of COC expansion during mammalian ovulation, while it has a direct role in follicle rupture during fish ovulation.

Mammalian ovaries begin producing a significant amount of progesterone in response to an LH surge (Bahr, 1978; Goff and Henderson, 1979; Hubbard and Greenwald, 1982). An LH surge also induces the expression of nuclear progesterone receptors (nPRs) (Li and O'Malley, 2003). The synthesis of progesterone and nPR both take place in the follicle cells of the preovulatory follicles, and their association results in the formation of an active transcription factor that directly regulates the expression of a variety of ovulation-related genes (Li and O'Malley, 2003; Ellman et al., 2009; Robker et al., 2009; Sriraman et al., 2010). In the medaka, 17α, 20β-dihydroxy-4-pregnen-3-one (DHP) is the naturally occurring steroid hormone (Sakai et al., 1987; Fukuda et al., 1994) that functions as a maturation-inducing hormone (MIH). The levels of DHP in the fish ovary rapidly increase after an LH surge (Sakai et al., 1987). Emerging evidence suggests that DHP has a dual role in the preovulatory follicle; the steroid hormone is essential not only for oocyte maturation, but also for ovulation in the medaka. Drastically induced expression of nPR was observed in the follicle cells of the fish ovarian follicle after treatment with PMSG (Nagahama and Yamashita, 2008) or recombinant medaka LH (our unpublished results). Because the expression of the two ovulation-related proteins, MT2-MMP and EP4b, in the medaka preovulatory follicle appears to be closely related to nPR, the activation of nPR is likely a prerequisite for the transcription of ovulation-related genes in the fish. Our recent morphological observation that the LH receptor, but not the FSH receptor, is localized to the follicle cells of the large preovulatory follicles (Ogiwara et al., 2013) is consistent with the idea that, as for mammalian species, nPR-mediated gene expression of ovulation-related proteins occurs in the granulosa cells of the follicles nearing ovulation.

CONCLUSIONS

Significant progress has been achieved in improving our understanding of the control of teleost oocyte maturation in recent years (Nagahama and Yamashita, 2008; Lessman, 2009; Thomas, 2012). In contrast, information on the mechanism of ovulation for non-mammalian vertebrate species has been very limited. However, results from our recent studies using the teleost medaka have highlighted the mechanism of follicle rupture during ovulation. Using the fish model, we have determined the proteases and the inhibitor that are involved in the rupture and have elucidated their respective roles in the process. We have also clarified the involvement of PGE2 in follicle rupture during fish ovulation. The accumulation of our knowledge of medaka ovulation has enabled us to consider the differences and similarities between the ovulatory process in mammalian and non-mammalian vertebrates at the molecular level. Although much has been learned about ovulation of the fish, much more remains to be solved. Areas of future study include the following: 1) defining the regulatory mechanisms of ovulation, particularly the LH-dependent induction mechanism of MT2-MMP and EP4b via the action of the transcription factor nPR and 2) determining the nature of the effect of PGE2 on the follicle cells of ovulating follicles at the time of ovulation. Another exciting challenge is to unravel the mystery of how these two important biological processes, i.e., oocyte maturation and ovulation, are properly timed in the follicle that is destined to ovulate.

ACKNOWLEDGMENTS

The authors thank the many wonderful colleagues who have contributed to the work described herein. This work was supported by Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan.

REFERENCES

1.

JM Bahr ( 1978) Simultaneous measurement of steroids in follicular fluid and ovarian venous blood in the rabbit. Biol Reprod 18: 193–197 Google Scholar

2.

CB Berkholtz , BE Lai , TK Woodruff , LD Shea ( 2006) Distribution of extracellular matrix proteins type I collagen, type IV collagen, fibronectin, and laminin in mouse folliculogenesis. Histochem Cell Biol 126: 583–592 Google Scholar

3.

AK Berndtson , FW Goetz , P Duman ( 1989) In vitro ovulation, prostaglandin synthesis, and proteolysis in isolated ovarian components of yellow perch (Perca flavescens): effects of 17α, 20βdihydroxy-4-pregnen-3-one and phorbol ester. Gen Comp Endocrinol 75: 454–465 Google Scholar

4.

J Bobe , T Nguyen , B Jalabert ( 2004) Targeted gene expression profiling in the rainbow trout (Oncorhynchus mykiss) ovary during maturational competence acquisition and oocyte maturation. Biol Reprod 71: 73–82 Google Scholar

5.

F Cetta , FW Goetz ( 1982) Ovarian and plasma prostaglandin E and F levels in brook trout (Salvelinus fontinalis) during pituitary-induced ovulation. Biol Reprod 27: 1216–1221 Google Scholar

6.

KJ Chang , JW Kim , J Lee , WB Im , HB Kwon , AW Schuetz ( 1995) Prostaglandin production and ovulation during exposure of amphibian ovarian follicles to gonadotropin or phorbol ester in vitro. Gen Comp Endocrinol 100: 257–266 Google Scholar

7.

KJ Chang , JW Kim , WB Im , HM Kang , HB Kwon ( 1997) Differential effects of gonadotropin and orthovanadate on oocyte maturation, ovulation, and prostaglandin synthesis by Rana ovarian follicles in vitro. J Exp Zool 277: 155–165 Google Scholar

8.

D Crespo , E Bonnet , N Roher , SA MacKenzie , A Krasnov , FW Goetz , et al. (2010) Cellular and molecular evidence for a role of tumor necrosis factor alpha in the ovulatory mechanism of trout. Reprod Biol Endocrinol 8: 34 Google Scholar

9.

TE Curry Jr , KG Osteen ( 2003) The matrix metalloproteinase system: changes, regulation, and impact throughout the ovarian and uterine reproductive cycle. Endocr Rev 24: 428–465 Google Scholar

10.

TE Curry Jr , MF Smith ( 2006) Impact of extracellular matrix remodeling on ovulation and the folliculo-luteal transition. Semin Reprod Med 24: 228–241 Google Scholar

11.

P Epler , K Bieniarz , E Marosz ( 1985) Effect of temperature, 17αhydroxy-20 β-dihydroprogesterone and prostaglandin F on carp oocyte maturation and ovulation in vitro. Gen Comp Endocrinol 58: 192–201 Google Scholar

12.

LL Espey ( 1967) Ultrastructure of the apex of the rabbit Graafian follicle during the ovulatory process. Endocrinology 81: 267–276 Google Scholar

13.

LL Espey , H Lipner ( 1994) Ovulation. In “The Physiology of Reproduction” Ed by E Knobil , JD Neill , Raven Press, New York, pp 725–780 Google Scholar

14.

LL Espey , JS Richards ( 2006) Ovulation. In “Physiology of Reproduction, Vol 1” 3rd ed, Ed by JD Neil et al., Academic Press, Amsterdam, pp 425–474 Google Scholar

15.

C Fujimori , K Ogiwara , A Hagiwara , S Rajapakse , A Kimura , T Takahashi ( 2011) Expression of cyclooxygenase-2 and prostaglandin receptor EP4b mRNA in the ovary of the medaka fish, Oryzias latipes: possible involvement in ovulation. Mol Cell Endocrinol 332: 67–77 Google Scholar

16.

C Fujimori , K Ogiwara , A Hagiwara , T Takahashi ( 2012) New evidence for the involvement of prostaglandin receptor EP4b in ovulation of the medaka, Oryzias latipes. Mol Cell Endocrinol. 362: 76–84 Google Scholar

17.

L Gazourian , KL Deragon , CF Chase , D Pati , HR Habibi , SA Sower ( 1997) Characteristics of GnRH binding in the gonads and effects of lamprey GnRH-I and -III on reproduction in the adult sea lamprey. Gen Comp Endocrinol 108: 327–339 Google Scholar

18.

FW Goetz ( 1991) Compartmentalization of prostaglandin synthesis within the fish ovary. Am J Physiol 260: R862–R865 Google Scholar

19.

FW Goetz ( 1993) Involvement of protein kinase C in agoniststimulated goldfish ovulation. Biol Reprod 48: 846–850 Google Scholar

20.

FW Goetz ( 1997) Follicle and extrafollicular tissue interaction in 17α, 20β-dihydroxy-4-pregnen-3-one-stimulated ovulation and prostaglandin synthesis in the yellow perch (Perca flavescens) ovary. Gen Comp Endocrinol 105: 121–126 Google Scholar

21.

FW Goetz , Y Nagahama ( 1985) The in vitro effects of cyclic nucleotides on prostaglandin-induced ovulation of goldfish (Carassius auratus). Zool Sci 2: 225–228 Google Scholar

22.

FW Goetz , G Theofan ( 1979) In vitro stimulation of germinal vesicle breakdown and ovulation of yellow perch (Perca flavescens) oocytes. Effects of 17α-hydroxy-20β-dihydroprogesterone and prostaglandins. Gen Comp Endocrinol 37: 273–285 Google Scholar

23.

FW Goetz , DC Smith , SP Krickl ( 1982) The effects of prostaglandins, phosphodiesterase inhibitors, and cyclic AMP on ovulation of brook trout (Salvelinus fontinalis) oocytes. Gen Comp Endocrinol 48: 154–160 Google Scholar

24.

AK Goff , KM Henderson ( 1979) Changes in follicular fluid and serum concentrations of steroids in PMS treated immature rats following LH administration. Biol Reprod 20: 1153–1157 Google Scholar

25.

T Grosser , S Yusuff , E Cheskis , MA Pack , GA FitzGerald ( 2002) Developmental expression of functional cyclooxygenases in zebrafish. Proc Natl Acad Sci U S A 99: 8418–8423 Google Scholar

26.

JC Havird , MM Miyamoto , KP Choe , DH Evans ( 2008) Gene duplications and losses within the cyclooxygenase family of teleosts and other chordates. Mol Biol Evol 25: 2349–2359 Google Scholar

27.

H Hizaki , E Segi , Y Sugimoto , M Hirose , T Saji , F Ushikubi , et al. (1999) Abortive expansion of the cumulus and impaired fertility in mice lacking the prostaglandin E receptor subtype EP(2). Proc Natl Acad Sci U S A 96: 10501–10506 Google Scholar

28.

M Horiguchi , C Fujimori , K Ogiwara , A Moriyama , T Takahashi ( 2008) Collagen type-I α1 chain mRNA is expressed in the follicle cells of the medaka ovary. Zool Sci 25: 937–945 Google Scholar

29.

CJ Hubbard , GS Greenwald ( 1982) Cyclic nucleotides, DNA, and steroid levels in ovarian follicles and corpora lutea of the cyclic hamster. Biol Reprod 26: 230–240 Google Scholar

30.

Y Ishikawa ( 2000) Medakafish as a model system for vertebrate developmental genetics. Bioessays 22: 487–495 Google Scholar

31.

TO Ishikawa , HR Herschman ( 2007) Two inducible, functional cyclooxygenase-2 genes are present in the rainbow trout genome. J Cell Biochem 102: 1486–1492 Google Scholar

32.

TO Ishikawa , KJ Griffin , U Banerjee , HR Herschman ( 2007) The zebrafish genome contains two inducible, functional cyclooxygenase-2 genes. Biochem Biophys Res Commun 352: 181–187 Google Scholar

33.

T Iwamatsu ( 1978) Studies on oocyte maturation of the medaka, Oryzias latipes. VI. Relationship between the circadian cycle of oocyte maturation and activity of the pituitary gland. J Exp Zool 206: 355–363 Google Scholar

34.

T Iwamatsu , T Ohta , E Oshima , N Sakai ( 1988) Oogenesis in the medaka Oryzias latipes. Stage of oocyte development. Zool Sci 5: 353–373 Google Scholar

35.

B Jalabert , D Szollosi ( 1975) In vitro ovulation of trout oocytes: effect of prostaglandins on smooth muscle-like cells of the theca. Prostaglandins 9: 765–778 Google Scholar

36.

R Jarving , I Jarving , R Kurg , AR Brash , N Samel ( 2004) On the evolutionary origin of cyclooxygenase (COX) isozymes: characterization of marine invertebrate COX genes points to independent duplication events in vertebrate and invertebrate lineages. J Biol Chem 279: 13624–13633 Google Scholar

37.

IM Joyce , FL Pendola , M O'Brien , JJ Eppig ( 2001) Regulation of prostaglandin-endoperoxide synthase 2 messenger ribonucleic acid expression in mouse granulosa cells during ovulation. Endocrinology 142: 3187–3197 Google Scholar

38.

H Kagawa , Y Nagahama ( 1981) In vitro effects of prostaglandins on ovulation in goldfish Carassius auratus. Bull Jap Soc Sci Fish 47: 1119–1121 Google Scholar

39.

H Kagawa , H Tanaka , T Unuma , H Ohta , K Gen , K Kuzawa ( 2003) Role of prostaglandin in the control of ovulation in the Japanese eel Anguilla japonica. Fisheries Sci 69: 234–241 Google Scholar

40.

M Kasahara , K Naruse , S Sasaki , Y Nakatani , W Qu , B Ahsan , et al. (2007) The medaka draft genome and insights into vertebrate genome evolution. Nature 447: 714–719 Google Scholar

41.

Y Kato , K Ogiwara , C Fujimori , A Kimura , T Takahashi ( 2010) Expression and localization of collagen type IV α1 chain in medaka ovary. Cell Tissue Res 340: 595–605 Google Scholar

42.

AH Kwok , Y Wang , FC Leung ( 2012) Molecular characterization of prostaglandin F receptor (FP) and E receptor subtype 1 (EP(1)) in zebrafish. Gen Comp Endocrinol 178: 216–226 Google Scholar

43.

G Leonardsson , XR Peng , K Liu , L Nordstrom , P Carmeliet , R Mulligan , et al. (1995) Ovulation efficiency is reduced in mice that lack plasminogen activator gene function: functional redundancy among physiological plasminogen activators. Proc Natl Acad Sci USA 92: 12446–12450 Google Scholar

44.

CA Lessman ( 2009) Oocyte maturation: converting the zebrafish oocyte to the fertilizable egg. Gen Comp Endocrinol 161: 53–57 Google Scholar

45.

X Li , BW O'Malley ( 2003) Unfolding the action of progesterone receptors. J Biol Chem 278: 39261–39264 Google Scholar

46.

S Li , Z Mao , W Han , Z Sun , W Yan , H Chen , S Yan ( 1993) In vitro oocyte maturation in the zebra fish, Brachydanio rerio, and the fertilization and development of the mature egg. Chin J Biotechnol 9: 247–255 Google Scholar

47.

AK Lind , B Weijdegard , P Dahm-Kahler , J Molne , K Sundfeldt , M Brannstrom ( 2006) Collagens in the human ovary and their changes in the perifollicular stroma during ovulation. Acta Obstet Gynecol Scand 85: 1476–1484 Google Scholar

48.

AL Lister , G Van Der Kraak ( 2008) An investigation into the role of prostaglandins in zebrafish oocyte maturation and ovulation. Gen Comp Endocrinol 159: 46–57 Google Scholar

49.

AL Lister , GJ Van Der Kraak ( 2009) Regulation of prostaglandin synthesis in ovaries of sexually-mature zebrafish (Danio rerio). Mol Reprod Dev 76: 1064–1075 Google Scholar

50.

A Munakata , M Kobayashi ( 2010) Endocrine control of sexual behavior in teleost fish. Gen Comp Endocrinol 165: 456–468 Google Scholar

51.

Y Nagahama ( 1994) Endocrine regulation of gametogenesis in fish. Int J Dev Biol 38: 217–229 Google Scholar

52.

Y Nagahama , M Yamashita ( 2008) Regulation of oocyte maturation in fish. Dev Growth Differ 50 Suppl 1 : S195–S219 Google Scholar

53.

A Ny , L Nordstrom , P Carmeliet , T Ny ( 1997) Studies of mice lacking plasminogen activator gene function suggest that plasmin production prior to ovulation exceeds the amount needed for optimal ovulation efficiency. Eur J Biochem 244: 487–493 Google Scholar

54.

A Ny , G Leonardsson , AC Hagglund , P Hagglof , VA Ploplis , P Carmeliet , T Ny ( 1999) Ovulation in plasminogen-deficient mice. Endocrinology 140: 5030–5035 Google Scholar

55.

K Ogiwara , N Takano , M Shinohara , M Murakami , T Takahashi ( 2005) Gelatinase A and membrane-type matrix metalloproteinases 1 and 2 are responsible for follicle rupture during ovulation in the medaka. Proc Natl Acad Sci USA 102: 8442–8447 Google Scholar

56.

K Ogiwara , T Ikeda , T Takahashi ( 2010) A new in vitro ovulation model for medaka based on whole ovary culture. Zool Sci 27: 762–767 Google Scholar

57.

K Ogiwara , K Minagawa , N Takano , T Kageyama , T Takahashi ( 2012) Apparent involvement of plasmin in early-stage follicle rupture during ovulation in medaka. Biol Reprod 86: 113 Google Scholar

58.

K Ogiwara, C Fujimori, S Rajapakse, T Takahashi (2013) Characterization of luteinizing hormone and luteinizing hormone receptor and their indispensable role in the ovulatory process of the medaka. PLoS ONE 8(1): e54482. doi:10.1371/journal.pone. 0054482 Google Scholar

59.

J Ohnishi , E Ohnishi , H Shibuya , T Takahashi ( 2005) Functions for proteinases in the ovulatory process. Biochim Biophys Acta 1751: 95–109 Google Scholar

60.

K Ozato , Y Wakamatsu , K Inoue ( 1992) Medaka as a model of transgenic fish. Mol Mar Biol Biotechnol 1: 346–354 Google Scholar

61.

NW Pankhurst ( 1985) Final maturation and ovulation of oocytes of the goldeye, Hiodon alasoides (Rahubesque), in vitro. Can J Zool 63: 1003–1009 Google Scholar

62.

R Patino , P Thomas ( 1990) Effects of gonadotropin on ovarian intrafollicular processes during the development of oocyte maturational competence in a teleost, the Atlantic croaker: evidence for two distinct stages of gonadotropin control of final oocyte maturation. Biol Reprod 43: 818–827 Google Scholar

63.

R Patino , G Yoshizaki , D Bolamba , P Thomas ( 2003) Role of arachidonic acid and protein kinase C during maturation-inducing hormone-dependent meiotic resumption and ovulation in ovarian follicles of Atlantic croaker. Biol Reprod 68: 516–523 Google Scholar

64.

I Ramos , SB Cisint , CA Crespo , MF Medina , SN Fernandez ( 2008) Modulators of Bufo arenarum ovulation. Zygote 16: 65–72 Google Scholar

65.

JS Richards , DL Russell , RL Robker , M Dajee , TN Alliston ( 1998) Molecular mechanisms of ovulation and luteinization. Mol Cell Endocrinol 145: 47–54 Google Scholar

66.

JS Richards , DL Russell , S Ochsner , LL Espey ( 2002) Ovulation: new dimensions and new regulators of the inflammatory-like response. Annu Rev Physiol 64: 69–92 Google Scholar

67.

RL Robker , LK Akison , DL Russell ( 2009) Control of oocyte release by progesterone receptor-regulated gene expression. Nucl Recept Signal 7: e012 Google Scholar

68.

N Sakai , T Iwamatsu , K Yamauchi , Y Nagahama ( 1987) Development of the steroidogenic capacity of medaka (Oryzias latipes) ovarian follicles during vitellogenesis and oocyte maturation. Gen Comp Endocrinol 66: 333–342 Google Scholar

69.

N Sakai , T Iwamatsu , K Yamauchi , N Suzuki , Y Nagahama ( 1988) Influence of follicular development on steroid production in the medaka (Oryzias latipes) ovarian follicle in response to exogenous substrates. Gen Comp Endocrinol 71: 516–523 Google Scholar

70.

HB Santos , Y Sato , L Moro , N Bazzoli , E Rizzo ( 2008) Relationship among follicular apoptosis, integrin beta1 and collagen type IV during early ovarian regression in the teleost Prochilodus argenteus after induced spawning. Cell Tissue Res 332: 159–170 Google Scholar

71.

SS Schochet ( 1916) A suggestion as to the process of ovulation and ovarian cyst formation. Anat Rec 10: 447–457 Google Scholar

72.

PC Schroeder , P Pendergrass ( 1976) The inhibition of in-vitro ovulation from follicles of the teleost, Oryzias latipes, by cytochalasin B. J Reprod Fertil 48: 327–330 Google Scholar

73.

AW Schuetz ( 1986) Hormonal dissociation of ovulation and maturation of oocytes: ovulation of immatureamphibian oocytes by prostaglandin. Gamete Res 15: 99–113 Google Scholar

74.

J Sena , Z Liu ( 2008) Expression of cyclooxygenase genes and production of prostaglandins during ovulation in the ovarian follicles of Xenopus laevis. Gen Comp Endocrinol 157: 165–173 Google Scholar

75.

DL Simmons , RM Botting , T Hla ( 2004) Cyclooxygenase isozymes: the biology of prostaglandin synthesis and inhibition. Pharmacol Rev 56: 387–437 Google Scholar

76.

J Sirois , K Sayasith , KA Brown , AE Stock , N Bouchard , M Dore ( 2004) Cyclooxygenase-2 and its role in ovulation: a 2004 account. Hum Reprod Update 10: 373–385 Google Scholar

77.

LA Sorbera , JF Asturiano , M Carrillo , S Zanuy ( 2001) Effects of polyunsaturated fatty acids and prostaglandins on oocyte maturation in a marine teleost, the European sea bass (Dicentrarchus labrax). Biol Reprod 64: 382–389 Google Scholar

78.

PW Sorensen , FW Goetz ( 1993) Pheromonal and reproductive function of F prostaglandins and their metabolites in teleost fish. J Lipid Mediat 6: 385–393 Google Scholar

79.

PW Sorensen , TJ Hara , NE Stacey , FW Goetz ( 1988) F prostaglandins function as potent olfactory stimulants that comprise the postovulatory female sex pheromone in goldfish. Biol Reprod 39: 1039–1050 Google Scholar

80.

V Sriraman , M Sinha , JS Richards ( 2010) Progesterone receptorinduced gene expression in primary mouse granulosa cell cultures. Biol Reprod 82: 402–412 Google Scholar

81.

NE Stacey , S Pandey ( 1975) Effects of indomethacin and prostaglandins on ovulation of goldfish. Prostaglandins 9: 597–607 Google Scholar

82.

Y Sugimoto , S Narumiya ( 2007) Prostaglandin E receptors. J Biol Chem 282: 11613–11617 Google Scholar

83.

P Thomas ( 2012) Rapid steroid hormone actions initiated at the cell surface and the receptors that mediate them with an emphasis on recent progress in fish models. Gen Comp Endocrinol 175: 367–383 Google Scholar

84.

A Tsafriri , N Dekel ( 1994) Molecular mechanisms in ovulation. In “Molecular Biology of the Female Reproductive System” Ed by JK Findley , New York, Academic press, pp 207–258 Google Scholar

85.

C Tubbs , M Pace , P Thomas ( 2010) Expression and gonadotropin regulation of membrane progestin receptor alpha in Atlantic croaker (Micropogonias undulatus) gonads: role in gamete maturation. Gen Comp Endocrinol 165: 144–154 Google Scholar

86.

EJ Villablanca , A Pistocchi , FA Court , F Cotelli , C Bordignon , ML Allende , C Traversari , V Russo ( 2007) Abrogation of prostaglandin E2/EP4 signaling impairs the development of rag1+ lymphoid precursors in the thymus of zebrafish embryos. J Immunol 179: 357–364 Google Scholar

87.

J Wittbrodt , A Shima , M Schartl ( 2002) Medaka--a model organism from the far East. Nat Rev Genet 3: 53–64 Google Scholar

88.

WY Wong , DL DeWitt , WL Smith , JS Richards ( 1989) Rapid induction of prostaglandin endoperoxide synthase in rat preovulatory follicles by luteinizing hormone and cAMP is blocked by inhibitors of transcription and translation. Mol Endocrinol 3: 1714–1723 Google Scholar

89.

J Zou , NF Neumann , JW Holland , M Belosevic , C Cunningham , CJ Secombes , AF Rowley ( 1999) Fish macrophages express a cyclo-oxygenase-2 homologue after activation. Biochem J 340 (Pt 1): 153–159 Google Scholar
© 2013 Zoological Society of Japan
Takayuki Takahashi, Chika Fujimori, Akane Hagiwara, and Katsueki Ogiwara "Recent Advances in the Understanding of Teleost Medaka Ovulation: The Roles of Proteases and Prostaglandins," Zoological Science 30(4), 239-247, (1 April 2013). https://doi.org/10.2108/zsj.30.239
Received: 11 October 2012; Accepted: 1 November 2012; Published: 1 April 2013
KEYWORDS
LH
medaka
ovary
ovulation
progesterone receptor
prostaglandin E2
Back to Top