circadian clock function in the mammalian ovary

14
http://jbr.sagepub.com/ Journal of Biological Rhythms http://jbr.sagepub.com/content/early/2014/10/20/0748730414554222 The online version of this article can be found at: DOI: 10.1177/0748730414554222 published online 3 November 2014 J Biol Rhythms Michael T. Sellix Circadian Clock Function in the Mammalian Ovary Published by: http://www.sagepublications.com On behalf of: Society for Research on Biological Rhythms can be found at: Journal of Biological Rhythms Additional services and information for http://jbr.sagepub.com/cgi/alerts Email Alerts: http://jbr.sagepub.com/subscriptions Subscriptions: http://www.sagepub.com/journalsReprints.nav Reprints: http://www.sagepub.com/journalsPermissions.nav Permissions: What is This? - Nov 3, 2014 OnlineFirst Version of Record >> at Dicle Ãoeniversitesi on November 5, 2014 jbr.sagepub.com Downloaded from at Dicle Ãoeniversitesi on November 5, 2014 jbr.sagepub.com Downloaded from

Upload: m-t

Post on 07-Mar-2017

220 views

Category:

Documents


6 download

TRANSCRIPT

http://jbr.sagepub.com/Journal of Biological Rhythms

http://jbr.sagepub.com/content/early/2014/10/20/0748730414554222The online version of this article can be found at:

 DOI: 10.1177/0748730414554222

published online 3 November 2014J Biol RhythmsMichael T. Sellix

Circadian Clock Function in the Mammalian Ovary  

Published by:

http://www.sagepublications.com

On behalf of: 

  Society for Research on Biological Rhythms

can be found at:Journal of Biological RhythmsAdditional services and information for    

  http://jbr.sagepub.com/cgi/alertsEmail Alerts:

 

http://jbr.sagepub.com/subscriptionsSubscriptions:  

http://www.sagepub.com/journalsReprints.navReprints:  

http://www.sagepub.com/journalsPermissions.navPermissions:  

What is This? 

- Nov 3, 2014OnlineFirst Version of Record >>

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

JOURNAL OF BIOLOGICAL RHYTHMS, Vol XX No. X, Month 201X 1 –13DOI: 10.1177/0748730414554222© 2014 The Author(s)

1

CirCadian Timing SySTem in mammalS

The circadian timing system regulates daily rhythms of gene expression, hormone secretion, metabolism, and behavior (Albrecht, 2012; Marcheva et al., 2013). Circadian rhythms are generated at the molecular level by the rhythmic transcription of circa-dian clock genes. Most differentiated mammalian cells host an autoregulatory transcriptional-translational

feedback loop of clock gene transcription factors (Albrecht, 2012; Mohawk et al., 2012). The transcrip-tional enhancers BMAL1 and CLOCK are central to the oscillator, with CLOCK displaying histone acetyl transferase activity at target promoters (Etchegaray et al., 2003; Hirayama et al., 2007; Nader et al., 2009). Together, the BMAL1:CLOCK complex binds to E-box (CACGTG) sequences in target promoters and drives transcription of the period (per 1-3) and cryptochrome

554222JBRXXX10.1177/0748730414554222Journal Of Biological RhythmsSellix / molecular and Physiological Function of The Ovarian Oscillatorresearch-article2014

1. To whom all correspondence should be addressed: Michael T. Sellix, Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Rochester, NY 14623; e-mail: [email protected].

Circadian Clock Function in the Mammalian Ovary

Michael T. Sellix1 Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester

School of Medicine and Dentistry, Rochester, New York, USA

Abstract Rhythmic events in the female reproductive system depend on the coordinated and synchronized activity of multiple neuroendocrine and endo-crine tissues. This coordination is facilitated by the timing of gene expression and cellular physiology at each level of the hypothalamo-pituitary-ovarian (HPO) axis, including the basal hypothalamus and forebrain, the pituitary gland, and the ovary. Central to this pathway is the primary circadian pace-maker in the suprachiasmatic nucleus (SCN) that, through its myriad outputs, provides a temporal framework for gonadotropin release and ovulation. The heart of the timing system, a transcription-based oscillator, imparts SCN pace-maker cells and a company of peripheral tissues with the capacity for daily oscillations of gene expression and cellular physiology. Although the SCN sits comfortably at the helm, peripheral oscillators (such as the ovary) have unde-fined but potentially critical roles. Each cell type of the ovary, including theca cells, granulosa cells, and oocytes, harbor a molecular clock implicated in the processes of follicular growth, steroid hormone synthesis, and ovulation. The ovarian clock is influenced by the reproductive cycle and diseases that perturb the cycle and/or follicular growth can disrupt the timing of clock gene expres-sion in the ovary. Chronodisruption is known to negatively affect reproductive function and fertility in both rodent models and women exposed to shiftwork schedules. Thus, influencing clock function in the HPO axis with chronobiotics may represent a novel avenue for the treatment of common fertility disorders, particularly those resulting from chronic circadian disruption.

Keywords clock gene, reproduction, fertility, circadian rhythm, ovary, mouse

The BOdy ClOCkS review SerieS

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

2 JOURNAL OF BIOLOGICAL RHYTHMS / Month 201X

genes (cry1 and cry2). After some critical posttransla-tional modifications, including phosphorylation by Casein kinases, the PER and CRY proteins translocate to the nucleus, wherein they repress the activity of the BMAL1:CLOCK enhancer complex and suppress their own transcription. The inherent delay in nuclear translocation of the repressor complex, dictated by parallel phosphorylation and protein degradation pathways, underlies the near 24-h timing of the oscil-lator. In addition to the core loop, a secondary loop of interlocking transcriptional regulators including the repressor REV-ERBα and the enhancer retinoic acid-like orphan receptor α(RORα) provides stabil-ity of function by regulating BMAL1 expression. The core clock genes also control output genes or clock-controlled genes (CCGs). CCGs are the effec-tor molecules of the clock, mediating its temporal control over cell-type–specific gene expression and physiology (Hamada et al., 2004; Kennaway et al., 2003).

In mammals, the timing system has been described as a multioscillator hierarchy of coordi-nated and synchronized cell and tissue clocks (Menaker et al., 2013)). At the top of this hierarchy is the central pacemaker located in the suprachias-matic nucleus (SCN) of the basal hypothalamus. In addition to maintaining pacemaker function in the SCN, the molecular clock also regulates the timing of gene expression and physiology in peripheral tis-sues and extra-SCN brain regions (Abe et al., 2002; Guilding et al., 2009; Menaker et al., 2013; Yamazaki et al., 2000; Yoo et al., 2004). It is now widely believed, with minimal exceptions, that the majority of mam-malian cells are autonomous or semi-autonomous oscillators. This arrangement offers a unique set of challenges for the organism. Clearly, synchroniza-tion among central and peripheral oscillators (e.g., SCN, liver, pituitary gland, etc.) is a fundamental property of physiological homeostasis and a defin-ing feature of the timing system (Buijs et al., 2001; Guo et al., 2005; Menaker et al., 2013; Mohawk et al., 2012). The responsibility for this coordination falls largely on the timing of neural efflux from the SCN carried directly through peripheral nerves and indi-rectly through control of hypothalamic neuroendo-crine-releasing factors (e.g., GnRH; Albrecht, 2012; Menaker et al., 2013). Internal circadian organiza-tion is broadly defined as the coordinated and syn-chronized timing of central and peripheral clocks as well as the timing of each oscillator relative to the external (and internal) environment. A decline in circadian organization (circadian disruption or chro-nodisruption, as seen during chronic jet lag, rotating shift work, etc.) can lead to health issues including prediabetes, cancer, and cardiovascular disease (Evans and Davidson, 2013).

CirCadian Timing SySTem in endOCrine PhySiOlOgy: FrOm gOnadOTrOPh TO

β-Cell

Some of the earliest evidence for circadian clock function in the mammalian endocrine system was recorded well before the SCN was identified as the locus of the neural pacemaker (Moore and Eichler, 1972; Stephan and Zucker, 1972). In the mid-20th cen-tury, John Everett and Charles Sawyer established the role of a neural oscillator or “timer” in the temporal control of the ovulatory surge of luteinizing hormone (LH) secretion. They found that blocking the LH surge with a timed injection of anesthetic in the early afternoon on the day of ovulation (proestrus), a treat-ment that globally silenced neuronal activity, pre-vented the rise in LH (and thus ovulation) and delayed it 24 h (Everett and Sawyer, 1950; Sawyer et al., 1949). That is, rather than occurring several hours later when the effects of sedation waned, ovulation was delayed until the afternoon of the following day. These key data supported the intimate relationship between the circadian timing system and the hypo-thalamic-pituitary-gonadal (HPG) axis. Subsequent mechanistic studies solidified the SCN as the critical mediator of timing in neuroendocrine physiology (Guo et al., 2005; Hastings et al., 2007; Kalsbeek and Fliers, 2013). The SCN regulates rhythms of hormone secretion via a complex interplay of neuroendocrine, endocrine, and autonomic timing cues (Buijs et al., 2003a; Buijs et al., 2003b; Guo et al., 2005; Hastings et al., 2007; Kriegsfeld and Silver, 2006). Outside the brain, the molecular oscillator has been localized to each endocrine tissue, although functional signifi-cance in each hormone regulatory system is poorly understood. Well before it was determined that peripheral tissues were composed of cell-based oscil-lators, it was postulated that tissue autonomous clocks could contribute to the timing of hormone syn-thesis and secretion (Andrews, 1971; Andrews and Folk, 1964; Ungar and Halberg, 1962). Since the clon-ing of the first mammalian circadian clock gene (King et al., 1997) and the determination that clock genes are global regulators of cellular function (Albrecht, 2012), considerable evidence has accumulated link-ing the molecular clock to endocrine physiology (Bass and Takahashi, 2010; Huang et al., 2011; Kalsbeek et al., 2012; Prasai et al., 2011; Sellix and Menaker, 2011; Tonsfeldt and Chappell, 2012; Williams and Kriegsfeld, 2012). These discoveries have stimulated interest in the impact of chronodisruption in all its forms on endocrine physiology (Bass and Takahashi, 2010; Mahoney, 2010). The hypothalamo-pituitary-ovarian (HPO) axis represents an excellent model system for investigating the impact of chronodisrup-tion on physiology. The coordinated activity of

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

Sellix / MOLECULAR AND PHYSIOLOGICAL FUNCTION OF THE OVARIAN OSCILLATOR 3

neural, neuroendocrine, and endocrine oscillators is critical for HPO axis function (Fig. 1). Synchronization depends on communication among the various oscil-lators through direct (autonomic nervous cues) and indirect (neuroendocrine releasing factors, hormones) timing cues. Further, feedback from circulating hor-mones may act to modulate the phase and amplitude of tissue sensitivity to neuroendocrine and endocrine cues. By more closely exploring the activity of the clock and its target genes in endocrine tissues, novel functional relationships may emerge, providing insight into the connection between clock function and physiology. A clearer understanding of these relationships may eventually allow us to exploit tar-geted manipulation of the clock with chronobiotics,

paving the way for new and exciting means of treat-ment for complex and devastating fertility disorders.

diverSiTy and COmPlexiTy OF Ovarian ClOCk FunCTiOn

Ovarian clocks show significant phylogenetic diversity, having been detected in mammals, non-mammalian vertebrates, and some invertebrates (Sellix and Menaker, 2010). The molecular clock mechanism has been described in the ovary of the silkworm, moth, frog, and fruit fly, although the func-tional significance for fertility remains unclear (Kubo et al., 2010; Sellix and Menaker, 2010; Takeuchi et al., 2014). Limited evidence suggests that the clock may regulate fertility in drosophila, though core clock genes are not rhythmically expressed in the fly ovary (Beaver et al., 2003; Kotwica et al., 2009; Rush et al., 2006). Cells of the drosophila ovary lack functional CRY, which serves as a photoreceptor in drosophila responsible for the light-dependent degradation of timeless (Rush et al., 2006). Regardless, daily rhythms of oviposition (egg laying) are well described in dro-sophila and persist in mutant flies with a period closely matching that of locomotor activity (McCabe and Birley, 1998). These rhythms can be entrained by temperature cycles and are influenced almost equally by both temperature and light (Kannan et al., 2012). Flies lacking functional period or timeless gene expression do have reproductive deficits marked by irregular oocyte maturation and reduced fecundity (Beaver et al., 2003; TM et al., 2008). Surprisingly, flies lacking pigment-dispersing factor or functional ven-tral lateral neurons, known to be critical components of the drosophila timing system, continue to display daily rhythms of egg-laying (Howlader et al., 2006; Howlader and Sharma, 2006). Thus, while the loca-tion of the endogenous pacemaker for egg-laying rhythms in flies remains to be determined, evidence strongly suggests it is located outside of the ovary.

More is known regarding ovarian clock function in nonmammalian vertebrates, including domestic hens and quail (Nakao et al., 2004; Nakao et al., 2007). It is clear that the circadian clock plays a role in the timing of oviposition and egg laying in birds (Underwood et al., 1997). Nakao and colleagues explored the func-tional significance of the clock in avian follicles, link-ing it to transcriptional regulation of steroidogenic enzymes within granulosa cells (GCs; Nakao et al., 2004; Nakao et al., 2007). Given the dearth of evidence (nonmammalian vertebrates) and the general ambi-guity (invertebrates) regarding ovarian clock function in these species, this discussion will be limited to the evidence for clock function in the mammalian ovary.

Figure 1. The circadian timing system in the hypothalamo-pitu-itary-ovarian (hPO) axis. a schematic diagram of the hPO axis indicating the integration of the circadian timing system in hPO axis function. The ovary shows phasic sensitivity to gonadotro-pins (lh and FSh that may depend on the timing of the ovar-ian clock and/or numerous timing cues of central and peripheral origin. Time of day or “photic” cues are passed to the central pacemaker in the SCn via the rhT. The SCn then conveys this information to the pineal via the SCg and the pituitary indirectly via the activity of releasing hormone neurons in the basal hypo-thalamus and forebrain. The SCn may also modulate the timing of the ovarian clock more directly via autonomic nervous cues (SnS, PSnS). Cells of the ovarian follicle also express melatonin receptors, and melatonin could indirectly modulate the timing of the ovarian clock. adrenal glucocorticoids (e.g., corticosterone) may also act to synchronize the timing of the molecular clock in the pituitary and ovary. Ovarian steroid feedback affects the timing of clock gene expression in several tissues, including the uterus, pituitary, and ovary, and could further gate the response to gonadotropins. nervous signals are indicated by lines ending in triangles, while lines ending in solid dots indicate hormonal pathways. hv indicates light, rhT, retinohypothalamic tract; SnS/PSnS, sympathetic nervous system, parasympathetic ner-vous system; FSh, follicle-stimulating hormone; lh, luteinizing hormone; SCn, suprachiasmatic nucleus; SCg, superior cervical ganglion.

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

4 JOURNAL OF BIOLOGICAL RHYTHMS / Month 201X

CirCadian ClOCk FunCTiOn in The mammalian Ovary

The female reproductive system in mammals exhib-its elegant and precise rhythmicity, normally tightly entrained to the light:dark cycle by photoperiodic cues originating in the SCN (see Boden and Kennaway, 2006; Kennaway, 2005). In mammals, circulating gonadotropin LH and follicle-stimulating hormone (FSH) levels oscillate with a diurnal rhythm marked by significant afternoon “surges” on the day of ovula-tion (Bronson and Vom Saal, 1979; Goldman, 1999; Legan and Karsch, 1975; Moenter et al., 2003). In rodents, the preovulatory LH surge occurs in the late afternoon/early evening on proestrus (the third day of the 4- to 5-d reproductive cycle; Bronson and Vom Saal, 1979; Everett and Sawyer, 1950). By all accounts, this afternoon LH surge is necessary and sufficient for ovulation, luteinization, and subsequent formation of the corpus luteum (Karsch et al., 1997; Moenter et al., 2003). The ovarian follicle is subdivided both anatomi-cally and functionally into the mesenchyme-derived theca cells (TCs), cuboidal androgen- and estrogen-producing cells that line the outside of the follicle, and epithelial granulosa cells (GCs) that line the inside of the follicle, surround the oocyte, and primarily synthe-size estrogen (Kacsoh, 2000; Leung and Adashi, 2004). Following follicular rupture, both the TCs and GCs undergo luteinization, resulting in increased vascular-ization of the GC layer and a transition to increased production of progesterone (Leung and Adashi, 2004). The process of luteinization results in considerable changes in the pattern of steroidogenic enzyme expres-sion in both TCs and GCs (Espey and Richards, 2006). Normal patterns of steroid hormone production, mat-uration of the follicle, and ovulation depend on the synergistic interaction between these two cell types.

The timing of ovulation, limited to a temporal win-dow on the afternoon of proestrus, depends on the timing of the LH surge (Karsch et al., 1997; Kriegsfeld and Silver, 2006; Moenter et al., 2003). This dogma holds that the ovary simply acts as a vessel for devel-opment of the oocyte, responding to threshold levels of LH with initiation of an ovulatory program (Espey and Richards, 2006). However, it has been suggested that the LH surge must arrive at the appropriate time of day to be effective (Sellix et al., 2010). Our experi-ments reveal that delivery of exogenous LH (equine) to rats previously treated with a selective GnRH receptor antagonist (cetrotide) was effective at induc-ing ovulation only when the LH was provided dur-ing the dark phase (Sellix et al., 2010). We have also seen nearly identical responses to ovine FSH, which is also known to stimulate ovulation through cross-talk between FSH and LH receptors (Galway et al., 1990). This rhythm of ovulatory responsiveness to LH

persisted in constant darkness, suggesting that it is gated by the timing system (Sellix et al., 2010). It is possible that SCN-dependent cues of either a humoral (e.g., adrenal steroids) or neural (autonomic nerves) nature play some role in setting this window of sensi-tivity to gonadotropins. It is equally possible that the ovary contains a circadian oscillator that dictates rhythmic expression of key players in the ovulatory response pathway. This hypothesis challenges the notion that peripheral oscillators in the HPG axis are passive with regard to the timing of key reproductive events. This notion has been extended to other peripheral endocrine organs, including the pituitary, endocrine pancreas, and adrenal gland (Marcheva et al., 2010; Resuehr et al., 2007; Yoder et al., 2014), indi-cating that the circadian clocks in peripheral endo-crine organs may play an active role in the timing of sensitivity to various activating and inhibiting fac-tors. As with these other endocrine organs, recent dis-coveries have shed additional light on the functional significance of the clock in the mammalian ovary.

Rhythms of clock gene expression have been reported in the intact ovary (Fahrenkrug et al., 2006; He et al., 2007a, 2007b; Karman and Tischkau, 2006; Tischkau et al., 2011) and isolated GCs (Chen et al., 2013a; Chen et al., 2013b; Chu et al., 2012; Chu et al., 2011; He et al., 2007b; Yoshikawa et al., 2009) from both rats and mice. Clock gene expression has also been reported in oocytes and preimplantation embryos from mice (Amano et al., 2009; Hamatani et al., 2004; Johnson et al., 2002), rabbits, and cattle (Amano et al., 2010). Rhythms of clock gene expres-sion were first independently reported in the rat ovary (Fahrenkrug et al., 2006; Karman and Tischkau, 2006). Gonadotropin exposure in vivo induced cyclic expression of bmal1 and per2 mRNA in the ovaries of hypophysectomized prepubertal rats (Karman and Tischkau, 2006). Further, diurnal rhythms of per1 and per2 expression were observed that persisted across the 4- to 5-d estrus cycle (Fahrenkrug et al., 2006). Rhythms of clock gene expression were also detected in large preantral follicles, small antral follicles, Graafian follicles, and corpora lutea. The physiologi-cal significance of rhythmic clock gene expression in these cells is poorly defined. It is possible that the ovarian clock modulates the timing of ovulation by regulating the expression of gonadotropin receptors and/or regulates the timing and amplitude of steroid and peptide hormone secretion. It has been suggested that the timing of ovulation may depend on rhythmic sensitivity of the ovary to gonadotropins (Sellix, 2013; Sellix et al., 2010). While these data indicate that the clock in the ovary could drive rhythms of sensitivity to LH, there is little evidence supporting the cell autonomous nature of this sensitivity in follicular cells. We will focus on the mechanism whereby the

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

Sellix / MOLECULAR AND PHYSIOLOGICAL FUNCTION OF THE OVARIAN OSCILLATOR 5

clock in each different cell type of the ovary contrib-utes to ovarian physiology (summarized in Fig. 2).

Circadian Clock Function in Ovarian gCs

Most of what we currently know regarding clock function in the mammalian ovary relates to the timing of gene expression in mature or luteinized GCs from rats and mice. It has been reported that rhythms of clock gene expression are limited to mature GCs and luteal cells (Chu et al., 2012; Chu et al., 2011; He et al., 2007a, 2007b). It is well known that the LH surge induces a significant change in gene expression within the GCs lining the preovulatory follicle (Espey and Richards, 2002; Espey et al., 2003; Richards, 2005). However, it remains to be seen if these LH-responsive genes oscillate with a circadian rhythm in the absence of the surge. Several candidate genes associated with the response of GCs to LH can be considered CCGs, including prostaglandin synthase, LH receptor, ste-roidogenic enzymes, gap junction proteins, and non–clock gene transcription factors (Fig. 2; Chen et al., 2013a; Chen et al., 2013b; Sellix and Menaker, 2010).

A significant step in the response of the GC to LH is the increase in the level of prostanoids, including prostaglandin E2 (PGE2) and PGF2α (Sirois et al., 2004). Reports indicate that the expression of cyclo-oxygenase-2 (COX2), the rate-limiting enzyme for prostaglandin synthsis, is regulated by the BMAL1::CLOCK complex through E-boxes in the COX2 promoter (Sirois et al., 2004). These data indi-cate that successful follicular rupture, a process dependent on prostanoid signaling, may require proper transcriptional control by the circadian clock. Another likely candidate is liver receptor homolog 1 (LRH-1), which was first identified as an orphan nuclear receptor in the liver (Nitta et al., 1999). In the ovary, lrh-1 expression is limited to the GC layer (Liu et al., 2003). In the liver, LRH-1 binds to the core circa-dian clock protein CLOCK (Oiwa et al., 2007) and acts synergistically to drive CLOCK-BMAL1–mediated transcription (Oiwa et al., 2007). Whether LRH-1 has the same activity in ovarian cells is currently unknown. In the ovary, LRH-1 controls steroid hor-mone biosynthesis in GCs through direct activation of cytochrome P450 side chain cleavage (cyp11a1) transcription following luteinization (Kim et al., 2005). It has been suggested that regular lrh-1 expres-sion in mouse GCs is critical for ovulation (Duggavathi et al., 2008). Whether disruption of molecular clock function in GCs results from, or is linked to, abnor-mal lrh-1 or cox2 gene expression is unknown. Nevertheless, the reduced fecundity associated with lrh-1 deficiency and the link between lrh-1 or cox2 transcription and clock gene expression make it likely that disruption of the oscillator in the ovary will have detrimental effects on fertility.

Treatment with PGE2 in vivo has been shown to phase shift the rhythm of per1, d-element binding pro-tein (dbp), and rev-erbα mRNA expression in the heart, liver, and kidney (Tsuchiya et al., 2005). Most recently, it was revealed that luteinized GCs do in fact have robust circadian rhythms of ptgs2 and LH receptor (lhcgr) gene expression that are disrupted and in some cases abolished following attenuation of bmal1 expression (Chen et al., 2013b). These data indicate that rhythms of lhcgr expression are directly regu-lated by the BMAL1:CLOCK enhancer complex bind-ing to E-box motifs in the lhcgr promoter. Further, these data suggest that an increase in COX2 and LHCGR expression and/or PG secretory activity pre-ceding the arrival of the LH surge may facilitate the responsiveness of GCs to LH and the subsequent transition to a functional corpus luteum. Additional evidence for a link between the circadian clock and PG signaling was recently discovered in the bovine uterus (Isayama et al., in press). The expression of both reverbα and ptgs2 in bovine uterine stromal cells (USCs) was significantly down-regulated following

Figure 2. Circadian clock function in ovarian follicles. in granulosa cells (gCs), the clock (Bmal1:ClOCk heterodi-mer indicated as circles binding to gene promoters) regulates the timing and amplitude of several gene products associated with the ovulatory response to gonadotropins, including the lh receptor (lhCgr) and prostaglandin synthase (COx2). The clock in gCs also drives rhythms of enzymes critical for steroid hormone biosynthesis including steroidogenic acute regulatory protein (Star), 3β-hydroxysteroid dehydrogenase (3β-hSd), 11β-hydroxylase (CyP11a), and aromatase (CyP19). although clock genes are rhythmically expressed in theca cells (TCs), there is currently no evidence supporting cell-type–specific regula-tion of CCg expression in these cells. Star, steroidogenic acute regulatory protein; 3β-hSd, 3β-hydroxysteroid dehydrogenase; CyP11a, 11β-hydroxylase; CyP19, aromatase; lhCgr, luteiniz-ing hormone/chorionic gonadotropin receptor; COx2, cyclooxy-genase-2.

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

6 JOURNAL OF BIOLOGICAL RHYTHMS / Month 201X

treatment with bmal1 small interfering RNA (siRNA). Further, production of PGF2α was reduced in USCs following exposure to bmal1 siRNA. Finally, levels of ptgs2 mRNA in both USC and uterine epithelial cells were increased following treatment with the syn-thetic REVERBα antagonist SR8278, suggesting that REVERBα directly suppresses ptgs2 gene expression in uterine tissue, most likely through binding at rev-erb response elements in the ptgs2 promoter. This evi-dence supports the assertion that a similar activity of REVERBα controls the expression of ptgs2 in GCs. It was recently suggested that the appearance of rhyth-mic clock gene expression in maturing follicles depends on FSH-induced expression of gap junctions in GCs (Chen et al., 2013a). According to these reports, disruption of intercellular communication via gap junction blockers reduces the amplitude and length-ens the period of PER2 expression in rat GCs (Chen et al., 2013a), the suggestion being that gonadotropin-dependent communication among GCs could play a critical role in the appearance and/or maintenance of CCG expression in the follicle. Together, these data indicate that multiple core clock genes both directly or indirectly modulate the timing and amplitude of CCG expression in GCs.

Circadian Clock Function in Ovarian TCs

Because of the difficulty in isolating purified pop-ulations, evidence for cell autonomous clock function in ovarian TCs and interstitial (stromal) cells has been limited to histological analyses (Fahrenkrug et al., 2006; Gras et al., 2012; Karman and Tischkau, 2006). While the clock genes are rhythmically expressed in these cells, evidence for a clock-controlled program of cell-type–specific gene expression in theca and stromal cells remains elusive. Given TCs’ critical involvement in ovarian steroid production and their established role in both follicular growth and ovula-tion, it is imperative that we better define the role of the circadian clock in TCs going forward. The effects of circadian disruption on TCs may have consider-able implications for fertility, particularly as it relates to the impact of chronodisruption on androgen syn-thesis and secretion from these cells (Sen and Hammes, 2010).

Circadian Clock Function in the Oocyte

Clock genes are expressed in mouse, rabbit, and bovine oocytes (Amano et al., 2009; Amano et al., 2010; Hamatani et al., 2004; Johnson et al., 2002). In mice, clock gene mRNA fluctuates as a function of developmental stage (Amano et al., 2009). However, it has been suggested that, rather than driving daily

rhythms of gene expression, they are part of the maternal program responsible for normal oocyte maturation. The increased levels of cry1 mRNA expression in germinal vesicle oocytes suggests that it plays a role, along with clock and bmal1, in sup-pressing CCG expression during oocyte maturation and early embryonic development. In support of this hypothesis, it was determined that knockdown of cry1 mRNA expression with siRNA leads to slight inhibition of meiosis progression in murine oocytes (Amano et al., 2009). Together, these data support an oscillator-independent role for the clock genes in the development of the mature oocyte.

It was recently reported that mice with a condi-tional knockout of the BMAL1 locus in steroidogenic cells (steroidogenic-factor 1-Cre; Bmal1flx/flx) show severe deficits in implantation success and compro-mised progesterone secretion (Liu et al.,2014). Notably, these mice showed normal estrous cycles, rhythms of behavior and number of corpora lutea when compared with littermate controls. Normal implantation was rescued when WT ovaries were transplanted to SF1-Cre;BMAL1-/- mice and implan-tation rates were reduced when conversely, KO ova-ries were transplanted into WT hosts. These data strongly suggest that BMAL1 deletion in steroido-genic cells of the ovary disrupts implantation due to abnormal progesterone secretion. These effects are not due to disruption of the endometrial clock, as the SF1-Cre transgene used was reportedly not expressed in uterine tissue. This construct did result in deletion of BMAL1 in the brain, adrenal gland and pituitary. Nonetheless, these data support the hypothesis that the ovarian clock plays a role in fertility, linked in large part to the control of ovarian steroid hormone synthesis.

Circadian Clock in the Ovary: role in Steroidogenesis

Evidence from rodents (Alvarez et al., 2008; Chen et al., 2013a; Chen et al., 2013b; Ratajczak et al., 2009) and birds (Nakao et al., 2007) suggest that the circa-dian clock plays a significant role in the amplitude and timing of steroid hormone synthesis in the ovary. Circadian rhythms of steroidogenic acute regulatory protein (StAR), 3β-hydroxysteroid dehydrogenase, 11β-hydroxylase, and aromatase (cyp19) have been observed in mature GCs (Chen et al., 2013a; Chen et al., 2013b). These rhythms are altered or abolished following clock disruption (Chen et al., 2013b). Further, bmal1–/– mice have abnormally low levels of progesterone secretion due to reduced StAR expression (Ratajczak et al., 2009). These data suggest that chronodisruption alone may lead to abnormal patterns and/or levels of ovarian steroid hormone

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

Sellix / MOLECULAR AND PHYSIOLOGICAL FUNCTION OF THE OVARIAN OSCILLATOR 7

synthesis and secretion or exacerbate the already neg-ative effects of endocrine pathology. It was recently reported that mice with a conditional knockout of the BMAL1 locus in steroidogenic cells (steroidogenic-factor 1-Cre; Bmal1flx/flx) show severe deficits in implantation success and compromised progesterone secretion (Liu et al., 2014). Notably, these mice showed normal estrous cycles, rhythms of behavior and num-ber of corpora lutea when compared with littermate controls. Normal implantation was rescued when WT ovaries were transplanted to SF1-Cre;BMAL1-/- mice and implantation rates were reduced when con-versely, KO ovaries were transplanted into WT hosts. These data strongly suggest that BMAL1 deletion in steroidogenic cells of the ovary disrupts implantation due to abnormal progesterone secretion. These effects are not due to disruption of the endometrial clock, as the SF1-Cre transgene used was reportedly not expressed in uterine tissue. This construct did result in deletion of BMAL1 in the brain, adrenal gland and pituitary. Nonetheless, these data support the hypoth-esis that the ovarian clock plays a role in fertility, linked in large part to the control of ovarian steroid hormone synthesis.

iS The Ovarian ClOCk required FOr nOrmal rePrOduCTive FunCTiOn?

The cells of the ovarian follicle display clear phasic sensitivity to gonadotropins, suggesting the activity of a circadian oscillator in the GC, TC, or both (Yoshikawa et al., 2009). Data showing rhythmic expression of the LH receptor gene in GCs also sup-port this notion (Chen et al., 2013b). Further, our data suggest that the timing of ovulation depends not only on the rhythm of LH secretion, itself driven by the SCN, but also on the timing of ovarian sensitivity to gonadotropins potentially set by the ovarian clock (Sellix et al., 2010). While the location and contribu-tion to physiological function of the oscillator remain a subject of conjecture, considerable evidence sug-gests that it is located in the follicle. Evidence to either refute or confirm this hypothesis has yet to material-ize. Extrinsic inputs to the ovary, including adrenal glucocorticoids and sympathetic nervous cues, might also contribute to the timing of ovarian sensitivity. It is thus equally plausible that the SCN coordinates the timing of ovulation independently of, or in partner-ship with, the ovarian clock through non–gonadotro-pin-dependent signaling.

Impaired fertility and fecundity have been observed in behaviorally arrhythmic clock mutant mice (Alvarez et al., 2008; Boden et al., 2010; Dolatshad et al., 2006; Miller et al., 2004; Ratajczak et

al., 2009). However, these animals are still able to ovulate and produce fertilizable ova (Ratajczak et al., 2009). While these data suggest that mutations that globally disrupt the clock have negative effects on fertility, they do not address the specific role of the clock in ovarian follicles. Lack of an effect of a global mutation on ovulation may result from devel-opmental compensation or redundancy within the core clock (as in Clock-null mice, see Debruyne et al., 2006). Studies using targeted knockout of clock gene expression in specific follicular cells, as was recently reported in the pituitary (Chu et al.. 2013), should shed some additional light on clock function in specific compartments of the developing follicle. Overall, the evidence for autonomous clock function in follicular cells supports a role for the clock in ovarian steroid synthesis, follicular growth, and ovulation. Identifying a link between the timing of the clock, the timing of ovarian sensitivity to LH, and the expression of CCGs such as lrh-1, ptgs2, and gap junctions may lead to improved understanding of the molecular bases of infertility.

Ovarian ClOCk FunCTiOn and FerTiliTy diSOrderS: new diSCOverieS and

FuTure PrOmiSe

The timing of clock gene expression in the rat and mouse ovary is clearly influenced by the develop-mental stage of the follicle (Gras et al., 2012). Further, it has been suggested that the ovarian clock is influ-enced by the varying titer of serum steroid hormones across the cycle (Murphy et al., 2013; Nakamura et al., 2010). Limited evidence supports a modest feedback effect of estrogen and progesterone on circadian rhythms of behavior (Royston et al., 2014; Takahashi and Menaker, 1980). Ovariectomy in rats and ham-sters lengthens the free-running period of locomotor activity, an effect that can be reversed following estro-gen replacement (Albers, 1981; Morin et al., 1977). Although steroid receptors are present in SCN pace-maker neurons, steroid hormones at physiologically relevant concentrations do not appear to affect the timing of clock gene expression in the SCN (Murphy et al., 2013; Nakamura et al., 2008). However, estra-diol can modulate the timing of clock gene expression in other regions, including areas of the basal forebrain that send dense projections to the SCN, raising the possibility of indirect control of behavior by steroid hormone feedback (Perrin et al., 2006). While the means of steroid feedback remain unresolved, recent evidence points to estrogen receptor subtype–specific effects on rhythms of locomotor activity (Royston et al., 2014). Activated steroid receptors may act down-stream of the molecular clock, a possibility supported

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

8 JOURNAL OF BIOLOGICAL RHYTHMS / Month 201X

by the fact that estrogen can alter the expression of gap junctions in SCN neurons (Shinohara et al., 2001). SCN pacemaker neurons certainly express steroid hormone receptors, including estrogen, progestin, and androgen receptors (Iwahana et al., 2008; Karatsoreos et al., 2011; Vida et al., 2008). It is worth noting that androgens can affect the period of loco-motor activity, and gonadectomy alters the timing and amplitude of clock gene expression in SCN neu-rons (Daan et al., 1975; Karatsoreos et al., 2011; Karatsoreos et al., 2007). The effects of ovarian steroid hormones on peripheral clock function are less ambiguous, with clear effects of steroids on rhythms of clock gene expression in tissues including the uterus, liver, and ovary (He et al., 2007c; Hirao et al., 2011; Nakamura et al., 2005; Nakamura et al., 2010). Regardless of the mechanism, the timing system is clearly affected by steroid hormones, with tissue-spe-cific effects apparent in both central and peripheral oscillators.

While the feedback effects of ovarian steroids on central clock function remain to be clarified, recent evidence suggests that irregular patterns of hormone secretion can globally influence the timing system. A common feature of fertility disorders such as polycys-tic ovarian syndrome (PCOS) is disruption of neuro-endocrine and endocrine function leading to irregular patterns and timing of ovarian steroid hormone secre-tion (Franks and Hardy, 2010; Nisenblat and Norman, 2009; Padmanabhan and Veiga-Lopez, 2011). Altered timing of clock gene expression in peripheral tissues such as the ovary may occur as a result of, or be par-tially responsible for, the hormonal imbalances associ-ated with these disorders (Amaral et al., 2014). As an example, an irregularly high serum androgen level during sexual development is a common clinical fea-ture of PCOS (Shi and Vine, 2012). Excess androgen, or hyperandrogenemia, leads to a dramatic change in the timing system, particularly the timing of clock gene expression in the ovarian follicle and liver (Sellix et al., 2013). These data suggest that reproductive endocrinopathies, particularly those arising due to developmental programming by abnormal steroid environments (e.g., PCOS), can alter the timing of the clock in tissues associated with both reproductive and metabolic function. This effect on the timing of ovar-ian clock gene expression may compound the nega-tive effects of excess androgen on key components of the steroid biosynthetic and ovulatory response path-ways. It is possible that treatment with clock-modu-lating compounds or chronobiotics, such as the REVERBα ligands or cryptochrome modulators (Chen et al., 2012), could be used as novel strategies for improving reproductive success in individuals suffer-ing from this and other fertility disorders. Moreover, treatment with melatonin or melatonin receptor ago-nists commonly used for the treatment of insomnia or

non–24-h sleep disorder might also improve internal circadian organization and resolve reproductive and metabolic dysfunction (Cardinali et al., 2011; Pai and Majumdar, in press). Other environmental manipula-tions such as precisely timed exercise or caloric restric-tion that are known to alter circadian organization may also improve HPO axis function (Mendoza et al., 2012; Schroeder et al., 2012). It remains to be seen whether manipulation of the timing system by any means, either alone or in conjunction with other thera-pies, represents a viable approach for addressing reproductive dysfunction in women.

COnCluSiOn

The timing of endocrine rhythms is a pervasive aspect of mammalian reproductive physiology, criti-cal for life on a rotating planet. Although we have made great strides in our understanding of the tim-ing system’s role in endocrinology, we have seen only a glimpse of its potential function. The depth of inte-gration, at the cellular and molecular level, remains to be determined for the majority of hormones and hormone regulatory pathways. It is apparent that the timing system plays a significant role in ovarian physiology, participating in the processes of ovula-tion, steroid hormone synthesis, and oocyte matura-tion. While we certainly know a great deal, many critical questions remain. How does the clock regu-late the response of the ovary to hormones and neural cues, particularly at the level of each ovarian cell type? When is the molecular clock actually “turned on” in developing follicular cells? What is the mecha-nism regulating the ontogeny of the clock in ovarian cells? And, lastly, how might adjusting the timing of the clock in ovarian follicles, or altering the syn-chrony between the ovarian clock and other tissues of the HPO axis, improve physiological function and/or diminish the risk of endocrine pathophysiology? As we face a growing epidemic of infertility due to genetic disease and environmental factors such as obesity, diabetes, and endocrine-disrupting com-pounds, the information we gain from the study of clock function in these tissues may provide the key to identifying new and effective treatments. Further, answering these questions may yield additional insights into the study of stem cells, developmental biology, and cancer biology.

aCknOwledgmenTS

The author wishes to thank Ms. Amanda Mereness for proofreading and assistance with the preparation of the manuscript.

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

Sellix / MOLECULAR AND PHYSIOLOGICAL FUNCTION OF THE OVARIAN OSCILLATOR 9

reFerenCeS

Abe M, Herzog ED, Yamazaki S, Straume M, Tei H, Sakaki Y, Menaker M, and Block GD (2002) Circadian rhythms in isolated brain regions. J Neurosci 22:350-356.

Albers HE (1981) Gonadal hormones organize and mod-ulate the circadian system of the rat. Am J Physiol 241:R62.

Albrecht U (2012) Timing to perfection: the biology of cen-tral and peripheral circadian clocks. Neuron 74:246-260.

Alvarez JD, Hansen A, Ord T, Bebas P, Chappell PE, Giebultowicz JM, Williams C, Moss S, and Sehgal A (2008) The circadian clock protein BMAL1 is necessary for fertility and proper testosterone production in mice. J Biol Rhythms 23:26-36.

Amano T, Matsushita A, Hatanaka Y, Watanabe T, Oishi K, Ishida N, Anzai M, Mitani T, Kato H, Kishigami S, et al. (2009) Expression and functional analyses of circadian genes in mouse oocytes and preimplantation embryos: Cry1 is involved in the meiotic process independently of circadian clock regulation. Biol Reprod 80:473-483.

Amano T, Tokunaga K, Kakegawa R, Yanagisawa A, Takemoto A, Tatemizo A, Watanabe T, Hatanaka Y, Matsushita A, Kishi M, et al. (2010) Expression analysis of circadian genes in oocytes and preimplantation embryos of cattle and rabbits. Anim Reprod Sci 121:225-235.

Amaral FG, Castrucci AM, Cipolla-Neto J, Poletini MO, Mendez N, Richter HG, and Sellix MT (2014) Environmental control of biological rhythms: effects on development, fertility and metabolism. J Neuroendocrinol 26:603-612.

Andrews RV (1971) Circadian rhythms in adrenal organ cultures. Gegenbaurs Morphol Jahrb 117:89-98.

Andrews RV, and Folk GE Jr. (1964) Circadian metabolic patterns in cultured hamster adrenal glands. Comp Biochem Physiol 11:393-409.

Bass J, and Takahashi JS (2010) Circadian integration of metabolism and energetics. Science 330:1349-1354.

Beaver LM, Rush BL, Gvakharia BO, and Giebultowicz JM (2003) Noncircadian regulation and function of clock genes period and timeless in oogenesis of Drosophila melanogaster. J Biol Rhythms 18:463-472.

Boden MJ, and Kennaway DJ (2006) Circadian rhythms and reproduction. Reproduction 132:379-392.

Boden M, Varcoe T, Voultsios A, and Kennaway D (2010) Reproductive biology of female Bmal1 null mice. Reproduction 139:1077-1090.

Bronson FH, and Vom Saal FS (1979) Control of the preovu-latory release of luteinizing hormone by steroids in the mouse. Endocrinology 104:1247-1255.

Buijs RM, Chun SJ, Niijima A, Romijn HJ, and Nagai K (2001) Parasympathetic and sympathetic control of the pancreas: a role for the suprachiasmatic nucleus and other hypothalamic centers that are involved in the regulation of food intake. J Comp Neurol 431: 405-423.

Buijs RM, la Fleur SE, Wortel J, Van Heyningen C, Zuiddam L, Mettenleiter TC, Kalsbeek A, Nagai K, and Niijima A (2003a) The suprachiasmatic nucleus balances sym-pathetic and parasympathetic output to peripheral organs through separate preautonomic neurons. J Comp Neurol 464:36-48.

Buijs RM, van Eden CG, Goncharuk VD, and Kalsbeek A (2003b) The biological clock tunes the organs of the body: timing by hormones and the autonomic nervous system. J Endocrinol 177:17.

Cardinali DP, Pagano ES, Scacchi Bernasconi PA, Reynoso R, and Scacchi P (2011) Disrupted chronobiology of sleep and cytoprotection in obesity: possible thera-peutic value of melatonin. Neuro Endocrinol Lett 32: 588-606.

Chen H, Zhao L, Chu G, Kito G, Yamauchi N, Shigeyoshi Y, Hashimoto S, and Hattori MA (2013a) FSH induces the development of circadian clockwork in rat granulosa cells via a gap junction protein Cx43-dependent path-way. Am J Physiol Endocrinol Metab 304:E566-E575.

Chen H, Zhao L, Kumazawa M, Yamauchi N, Shigeyoshi Y, Hashimoto S, and Hattori MA (2013b) Down-regulation of core clock gene Bmal1 attenuates expression of pro-gesterone and prostaglandin biosynthesis-related genes in rat luteinizing granulosa cells. Am J Physiol Cell Physiol 304:C1131-C1140.

Chen Z, Yoo SH, Park YS, Kim KH, Wei S, Buhr E, Ye ZY, Pan HL, and Takahashi JS (2012) Identification of diverse modulators of central and peripheral circadian clocks by high-throughput chemical screening. Proc Natl Acad Sci USA 109:101-106.

Chu A, Zhu L, Blum ID, Mai O, Leliavski A, Fahrenkrug J, Oster H, Boehm U, and Storch KF (2013) Global but not gonadotrope-specific disruption of Bmal1 abolishes the luteinizing hormone surge without affecting ovulation. Endocrinology 154:2924-2935.

Chu G, Misawa I, Chen H, Yamauchi N, Shigeyoshi Y, Hashimoto S, and Hattori MA (2012) Contribution of FSH and triiodothyronine to the development of cir-cadian clocks during granulosa cell maturation. Am J Physiol Endocrinol Metab 302:E645-E653.

Chu G, Yoshida K, Narahara S, Uchikawa M, Kawamura M, Yamauchi N, Xi Y, Shigeyoshi Y, Hashimoto S, and Hattori MA (2011) Alterations of circadian clockworks during differentiation and apoptosis of rat ovarian cells. Chronobiol Int 28:477-487.

Daan S, Damassa D, Pittendrigh CS, and Smith ER (1975) An effect of castration and testosterone replacement on a circadian pacemaker in mice (Mus musculus). Proc Natl Acad Sci USA 72:3744.

Debruyne JP, Noton E, Lambert CM, Maywood ES, Weaver DR, and Reppert SM (2006) A clock shock: mouse CLOCK is not required for circadian oscillator func-tion. Neuron 50:465-477.

Dolatshad H, Campbell EA, O’Hara L, Maywood ES, Hastings MH, and Johnson MH (2006) Developmental

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

10 JOURNAL OF BIOLOGICAL RHYTHMS / Month 201X

and reproductive performance in circadian mutant mice. Hum Reprod 21:68-79.

Duggavathi R, Volle DH, Mataki C, Antal MC, Messaddeq N, Auwerx J, Murphy BD, and Schoonjans K (2008) Liver receptor homolog 1 is essential for ovulation. Genes Dev 22:1871-1876.

Espey LL, and Richards JS (2002) Temporal and spatial patterns of ovarian gene transcription following an ovulatory dose of gonadotropin in the rat. Biol Reprod 67:1662-1670.

Espey LL, and Richards JS (2006) Ovulation. In: Neill J, editor. Physiology of reproduction. St. Louis (MO): Elsevier Academic Press. p. 425-474.

Espey LL, Ujioka T, Okamura H, and Richards JS (2003) Metallothionein-1 messenger RNA transcription in steroid-secreting cells of the rat ovary during the peri-ovulatory period. Biol Reprod 68:1895-1902.

Etchegaray JP, Lee C, Wade PA, and Reppert SM (2003) Rhythmic histone acetylation underlies transcription in the mammalian circadian clock. Nature 421:177.

Evans JA, and Davidson AJ (2013) Health consequences of circadian disruption in humans and animal models. Prog Mol Biol Transl Sci 119:283-323.

Everett JW, and Sawyer CH (1950) A 24-hour periodicity in the “LH-release apparatus” of female rats, disclosed by barbiturate sedation. Endocrinology 47:198.

Fahrenkrug J, Georg B, Hannibal J, Hindersson P, and Gras S (2006) Diurnal rhythmicity of the clock genes Per1 and Per2 in the rat ovary. Endocrinology 147:3769-3776.

Franks S, and Hardy K (2010) Aberrant follicle develop-ment and anovulation in polycystic ovary syndrome. Annales d’endocrinologie 71:228-230.

Galway AB, Lapolt PS, Tsafriri A, Dargan CM, Boime I, and Hsueh AJ (1990) Recombinant follicle-stimulat-ing hormone induces ovulation and tissue plasmino-gen activator expression in hypophysectomized rats. Endocrinology 127:3023-3028.

Goldman BD (1999) The circadian timing system and repro-duction in mammals. Steroids 64:679.

Gras S, Georg B, Jorgensen HL, and Fahrenkrug J (2012) Expression of the clock genes Per1 and Bmal1 during follicle development in the rat ovary: effects of gonado-tropin stimulation and hypophysectomy. Cell Tissue Res 350:539-548.

Guilding C, Hughes AT, Brown TM, Namvar S, and Piggins HD (2009) A riot of rhythms: neuronal and glial circa-dian oscillators in the mediobasal hypothalamus. Mol Brain 2:28.

Guo H, Brewer JM, Champhekar A, Harris RB, and Bittman EL (2005) Differential control of peripheral circadian rhythms by suprachiasmatic-dependent neural signals. Proc Natl Acad Sci USA 102:3111-3116.

Hamada T, Antle MC, and Silver R (2004) Temporal and spatial expression patterns of canonical clock genes and clock-controlled genes in the suprachiasmatic nucleus. Eur J Neurosci 19:1741.

Hamatani T, Carter MG, Sharov AA, and Ko MS (2004) Dynamics of global gene expression changes during mouse preimplantation development. Dev Cell 6:117-131.

Hastings M, O’Neill JS, and Maywood ES (2007) Circadian clocks: regulators of endocrine and metabolic rhythms. J Endocrinol 195:187-198.

He PJ, Hirata M, Yamauchi N, Hashimoto S, and Hattori MA (2007a) The disruption of circadian clockwork in differentiating cells from rat reproductive tissues as identified by in vitro real-time monitoring system. J Endocrinol 193:413-420.

He PJ, Hirata M, Yamauchi N, Hashimoto S, and Hattori MA (2007b) Gonadotropic regulation of circadian clockwork in rat granulosa cells. Mol Cell Biochem 302:111-118.

He PJ, Hirata M, Yamauchi N, and Hattori MA (2007c) Up-regulation of Per1 expression by estradiol and pro-gesterone in the rat uterus. J Endocrinol 194:511-519.

Hirao J, Nishimura M, Arakawa S, Niino N, Mori K, Furukawa T, Sanbuissho A, Manabe S, Nishihara M, and Mori Y (2011) Sex and circadian modulatory effects on rat liver as assessed by transcriptome analyses. J Toxicol Sci 36:9-22.

Hirayama J, Sahar S, Grimaldi B, Tamaru T, Takamatsu K, Nakahata Y, and Sassone-Corsi P (2007) CLOCK-mediated acetylation of BMAL1 controls circadian function. Nature 450:1086-1090.

Howlader G, Paranjpe DA, and Sharma VK (2006) Non-ventral lateral neuron-based, non-PDF-mediated clocks control circadian egg-laying rhythm in Drosophila mela-nogaster. J Biol Rhythms 21:13-20.

Howlader G, and Sharma VK (2006) Circadian regulation of egg-laying behavior in fruit flies Drosophila melano-gaster. J Insect Physiol 52:779-785.

Huang W, Ramsey KM, Marcheva B, and Bass J (2011) Circadian rhythms, sleep, and metabolism. J Clin Invest 121:2133-2141.

Isayama K, Chen H, Yamauchi N, and Hattori MA (in press) REV-ERBalpha inhibits the PTGS2 expression in bovine uterus endometrium stromal and epithelial cells exposed to ovarian steroids. J Reprod Dev.

Iwahana E, Karatsoreos I, Shibata S, and Silver R (2008) Gonadectomy reveals sex differences in circadian rhythms and suprachiasmatic nucleus androgen recep-tors in mice. Horm Behav 53:422-430.

Johnson MH, Lim A, Fernando D, and Day ML (2002) Circadian clockwork genes are expressed in the repro-ductive tract and conceptus of the early pregnant mouse. Reprod Biomed Online 4:140.

Kacsoh B (2000) Endocrine physiology. New York (NY): McGraw-Hill, Health Professions Division.

Kalsbeek A, and Fliers E (2013) Daily regulation of hor-mone profiles. Handb Exp Pharmacol:185-226.

Kalsbeek A, van der Spek R, Lei J, Endert E, Buijs RM, and Fliers E (2012) Circadian rhythms in the hypothalamo-pituitary-adrenal (HPA) axis. Mol Cell Endocrinol 349:20-29.

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

Sellix / MOLECULAR AND PHYSIOLOGICAL FUNCTION OF THE OVARIAN OSCILLATOR 11

Kannan NN, Reveendran R, Hari Dass S, Manjunatha T, and Sharma VK (2012) Temperature can entrain egg laying rhythm of Drosophila but may not be a stronger zeitgeber than light. J Insect Physiol 58:245-255.

Karatsoreos IN, Butler MP, Lesauter J, and Silver R (2011) Androgens modulate structure and function of the suprachiasmatic nucleus brain clock. Endocrinology 152:1970-1978.

Karatsoreos IN, Wang A, Sasanian J, and Silver R (2007) A role for androgens in regulating circadian behavior and the suprachiasmatic nucleus. Endocrinology 148:5487-5495.

Karman BN, and Tischkau SA (2006) Circadian clock gene expression in the ovary: effects of luteinizing hormone. Biol Reprod 75:624-632.

Karsch FJ, Bowen JM, Caraty A, Evans NP, and Moenter SM (1997) Gonadotropin-releasing hormone require-ments for ovulation. Biol Reprod 56:303-309.

Kennaway DJ (2005) The role of circadian rhythmicity in reproduction. Hum Reprod Update 11:91-101.

Kennaway DJ, Varcoe TJ, and Mau VJ (2003) Rhythmic expression of clock and clock-controlled genes in the rat oviduct. Mol Hum Reprod 9:503-507.

Kim JW, Havelock JC, Carr BR, and Attia GR (2005) The orphan nuclear receptor, liver receptor homolog-1, reg-ulates cholesterol side-chain cleavage cytochrome p450 enzyme in human granulosa cells. J Clin Endocrinol Metab 90:1678-1685.

King DP, Zhao Y, Sangoram AM, Wilsbacher LD, Tanaka M, Antoch MP, Steeves TD, Vitaterna MH, Kornhauser JM, Lowrey PL, et al. (1997) Positional cloning of the mouse circadian clock gene. Cell 89:641-653.

Kotwica J, Larson MK, Bebas P, and Giebultowicz JM (2009) Developmental profiles of PERIOD and DOUBLETIME in Drosophila melanogaster ovary. J Insect Physiol 55:419-425.

Kriegsfeld LJ, and Silver R (2006) The regulation of neuro-endocrine function: timing is everything. Horm Behav 49:557-574.

Kubo Y, Takeuchi T, Okano K, and Okano T (2010) Cryptochrome genes are highly expressed in the ovary of the African clawed frog, Xenopus tropicalis. PLoS One 5:e9273.

Legan SJ, and Karsch FJ (1975) A daily signal for the LH surge in the rat. Endocrinology 96:57-62.

Leung PCK, and Adashi EY (2004) The ovary. Amsterdam: Elsevier.

Liu DL, Liu WZ, Li QL, Wang HM, Qian D, Treuter E, and Zhu C (2003) Expression and functional analysis of liver receptor homologue 1 as a potential steroidogenic factor in rat ovary. Biol Reprod 69:508-517.

Liu Y, Johnson BP, Shen AL, Wallisser JA, Krentz KJ, Moran SM, Sullivan R, Glover E, Parlow AF, Drinkwater NR, Schuler LA, Bradfield CA (2014) Loss of BMAL1 in ovarian steroidogenic cells results in implantation fail-ure in female mice. Proc. of the Natl. Acad. of Sci., USA Sep 30;111(39):14295-300.

Mahoney MM (2010) Shift work, jet lag, and female repro-duction. Int J Endocrinol 2010:813764.

Marcheva B, Ramsey KM, Buhr ED, Kobayashi Y, Su H, Ko CH, Ivanova G, Omura C, Mo S, Vitaterna MH, et al. (2010) Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature 466:627-631.

Marcheva B, Ramsey KM, Peek CB, Affinati A, Maury E, and Bass J (2013) Circadian clocks and metabolism. Handb Exp Pharmacol 217:127-155.

McCabe C, and Birley A (1998) Oviposition in the period genotypes of Drosophila melanogaster. Chronobiol Int 15:119-133.

Menaker M, Murphy ZC, and Sellix MT (2013) Central control of peripheral circadian oscillators. Curr Opin Neurobiol 23:741-746.

Mendoza J, Gourmelen S, Dumont S, Sage-Ciocca D, Pevet P, and Challet E (2012) Setting the main circadian clock of a diurnal mammal by hypocaloric feeding. J Physiol 590:3155-3168.

Miller BH, Olson SL, Turek FW, Levine JE, Horton TH, and Takahashi JS (2004) Circadian clock mutation disrupts estrous cyclicity and maintenance of pregnancy. Curr Biol 14:1367-1373.

Moenter SM, DeFazio AR, Pitts GR, and Nunemaker CS (2003) Mechanisms underlying episodic gonadotropin-releasing hormone secretion. Front Neuroendocrinol 24:79-93.

Mohawk JA, Green CB, and Takahashi JS (2012) Central and peripheral circadian clocks in mammals. Ann Rev Neurosci 35:445-462.

Moore RY, and Eichler VB (1972) Loss of a circadian adre-nal corticosterone rhythm following suprachiasmatic lesions in the rat. Brain Res 42:201-206.

Morin LP, Fitzgerald KM, and Zucker I (1977) Estradiol shortens the period of hamster circadian rhythms. Science 196:305-307.

Murphy ZC, Pezuk P, Menaker M, and Sellix MT (2013) Effects of ovarian hormones on internal circadian orga-nization in rats. Biol Reprod 89:35.

Nader N, Chrousos GP, and Kino T (2009) Circadian rhythm transcription factor CLOCK regulates the transcriptional activity of the glucocorticoid recep-tor by acetylating its hinge region lysine clus-ter: potential physiological implications. FASEB J 23:1572-1583.

Nakamura TJ, Moriya T, Inoue S, Shimazoe T, Watanabe S, Ebihara S, and Shinohara K (2005) Estrogen differ-entially regulates expression of Per1 and Per2 genes between central and peripheral clocks and between reproductive and nonreproductive tissues in female rats. J Neurosci Res 82:622-630.

Nakamura TJ, Sellix MT, Kudo T, Nakao N, Yoshimura T, Ebihara S, Colwell CS, and Block GD (2010) Influence of the-estrous cycle on clock gene expression in repro-ductive tissues: effects of fluctuating ovarian steroid hormone levels. Steroids 75:203-212.

Nakamura TJ, Sellix MT, Menaker M, and Block GD (2008) Estrogen directly modulates circadian rhythms of PER2

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

12 JOURNAL OF BIOLOGICAL RHYTHMS / Month 201X

expression in the uterus. Am J Physiol Endocrinol Metab 295:E1025-E1031.

Nakao N, Yasuo S, Ebihara S, and Yoshimura T (2004) Circadian expression of clock genes in cultured chicken granulosa cells. Ninth meeting of the Society for Research on Biological Rhythms, Whistler, British Columbia.

Nakao N, Yasuo S, Nishimura A, Yamamura T, Watanabe T, Anraku T, Okano T, Fukada Y, Sharp PJ, Ebihara S, et al. (2007) Circadian clock gene regulation of ste-roidogenic acute regulatory protein gene expression in preovulatory ovarian follicles. Endocrinology 148: 3031-3038.

Nisenblat V, and Norman RJ (2009) Androgens and poly-cystic ovary syndrome. Curr Opin Endocrinol Diabetes Obes 16:224-231.

Nitta M, Ku S, Brown C, Okamoto AY, and Shan B (1999) CPF: an orphan nuclear receptor that regulates liver-specific expression of the human cholesterol 7alpha-hydroxylase gene. Proc Natl Acad Sci USA 96:6660-6665.

Oiwa A, Kakizawa T, Miyamoto T, Yamashita K, Jiang W, Takeda T, Suzuki S, and Hashizume K (2007) Synergistic regulation of the mouse orphan nuclear receptor SHP gene promoter by CLOCK-BMAL1 and LRH-1. Biochem Biophys Res Commun 353:895-901.

Padmanabhan V, and Veiga-Lopez A (2011) Developmental origin of reproductive and metabolic dysfunctions: androgenic versus estrogenic reprogramming. Semin Reprod Med 29:173-186.

Pai SA, and Majumdar AS (in press) Protective effects of melatonin against metabolic and reproductive distur-bances in polycystic ovary syndrome in rats. J Pharm Pharmacol.

Perrin JS, Segall LA, Harbour VL, Woodside B, and Amir S (2006) The expression of the clock protein PER2 in the limbic forebrain is modulated by the estrous cycle. Proc Natl Acad Sci USA 103:5591-5596.

Prasai MJ, Pernicova I, Grant PJ, and Scott EM (2011) An endocrinologist’s guide to the clock. J Clin Endocrinol Metab 96:913-22.

Ratajczak CK, Boehle KL, and Muglia LJ (2009) Impaired steroidogenesis and implantation failure in Bmal1-/- mice. Endocrinology 150:1879-1885.

Resuehr D, Wildemann U, Sikes H, and Olcese J (2007) E-box regulation of gonadotropin-releasing hormone (GnRH) receptor expression in immortalized gonado-trope cells. Mol Cell Endocrinol 278:36-43.

Richards JS (2005) Ovulation: new factors that prepare the oocyte for fertilization. Mol Cell Endocrinol 234:75-79.

Royston SE, Yasui N, Kondilis AG, Lord SV, Katzenellenbogen JA, and Mahoney MM (2014) ESR1 and ESR2 differentially regulate daily and circa-dian activity rhythms in female mice. Endocrinology 155:2613-2623.

Rush BL, Murad A, Emery P, and Giebultowicz JM (2006) Ectopic CRYPTOCHROME renders TIM light sensitive in the Drosophila ovary. J Biol Rhythms 21:272-278.

Sawyer CH, Everett JW, and Markee JE (1949) A neural factor in the mechanism by which estrogen induces the release of luteinizing hormone in the rat. Endocrinology 44:218-233.

Schroeder AM, Truong D, Loh DH, Jordan MC, Roos KP, and Colwell CS (2012) Voluntary scheduled exercise alters diurnal rhythms of behaviour, physiology and gene expression in wild-type and vasoactive intestinal peptide-deficient mice. J Physiol 590:6213-6226.

Sellix MT (2013) Clocks underneath: the role of peripheral clocks in the timing of female reproductive physiology. Front Endocrinol (Lausanne) 4:91.

Sellix MT, and Menaker M (2010) Circadian clocks in the ovary. Trends Endocrinol Metab 21:628-636.

Sellix MT, and Menaker M (2011) Circadian clocks in mam-malian reproductive physiology: effects of the “other” biological clock on fertility. Discov Med 11:273-281.

Sellix MT, Murphy ZC, and Menaker M (2013) Excess androgen during puberty disrupts circadian organiza-tion in female rats. Endocrinology 154:1636-1647.

Sellix MT, Yoshikawa T, and Menaker M (2010) A circadian egg timer gates ovulation. Curr Biol 20:R266-R267.

Sen A, and Hammes SR (2010) Granulosa cell-specific andro-gen receptors are critical regulators of ovarian develop-ment and function. Mol Endocrinol 24:1393-1403.

Shi D, and Vine DF (2012) Animal models of polycystic ovary syndrome: a focused review of rodent models in relationship to clinical phenotypes and cardiometa-bolic risk. Fertil Steril 98:185-193.

Shinohara K, Funabashi T, Nakamura TJ, and Kimura F (2001) Effects of estrogen and progesterone on the expression of connexin-36 mRNA in the suprachias-matic nucleus of female rats. Neurosci Lett 309:37-40.

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

Stephan FK, and Zucker I (1972) Circadian rhythms in drinking behavior and locomotor activity of rats are eliminated by hypothalamic lesions. Proc Natl Acad Sci USA 69:1583-1586.

Takahashi JS, and Menaker M (1980) Interaction of estra-diol and progesterone: effects on circadian locomo-tor rhythm of female golden hamsters. Am J Physiol 239:R497.

Takeuchi T, Kubo Y, Okano K, and Okano T (2014) Identification and characterization of cryptochrome4 in the ovary of western clawed frog Xenopus tropicalis. Zool Sci 31:152-159.

Tischkau SA, Jaeger CD, and Krager SL (2011) Circadian clock disruption in the mouse ovary in response to 2,3,7,8-tet-rachlorodibenzo-p-dioxin. Toxicol Lett 201:116-122.

TM, Hari Dass S, and Sharma VK (2008) Egg-laying rhythm in Drosophila melanogaster. J Genet 87:495-504.

Tonsfeldt KJ, and Chappell PE (2012) Clocks on top: the role of the circadian clock in the hypothalamic and pituitary regulation of endocrine physiology. Mol Cell Endocrinol 349:3-12.

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from

Sellix / MOLECULAR AND PHYSIOLOGICAL FUNCTION OF THE OVARIAN OSCILLATOR 13

Tsuchiya Y, Minami I, Kadotani H, and Nishida E (2005) Resetting of peripheral circadian clock by prostaglan-din E2. EMBO Rep 6:256-261.

Underwood H, Siopes T, and Edmonds K (1997) Eye and gonad: role in the dual-oscillator circadian system of female Japanese quail. Am J Physiol 272:R172-r182.

Ungar F, and Halberg F (1962) Circadian rhythm in the in vitro response of mouse adrenal to adrenocorticotropic hormone. Science 137:1058-1060.

Vida B, Hrabovszky E, Kalamatianos T, Coen CW, Liposits Z, and Kallo I (2008) Oestrogen receptor alpha and beta immunoreactive cells in the suprachiasmatic nucleus of mice: distribution, sex differences and regulation by gonadal hormones. J Neuroendocrinol 20:1270-1277.

Williams WP III, and Kriegsfeld LJ (2012) Circadian control of neuroendocrine circuits regulating female reproduc-tive function. Front Endocrinol (Lausanne) 3:60.

Yamazaki S, Numano R, Abe M, Hida A, Takahashi R, Ueda M, Block GD, Sakaki Y, Menaker M, and Tei H (2000) Resetting central and peripheral circadian oscil-lators in transgenic rats. Science 288:682-685.

Yoder JM, Brandeland M, and Engeland WC (2014) Phase-dependent resetting of the adrenal clock by ACTH in vitro. Am J Physiol Regul Integr Comp Physiol 306:R387-R393.

Yoo SH, Yamazaki S, Lowrey PL, Shimomura K, Ko CH, Buhr ED, Siepka SM, Hong HK, Oh WJ, Yoo OJ, et al. (2004) PERIOD2::LUCIFERASE real-time reporting of circadian dynamics reveals persistent circadian oscilla-tions in mouse peripheral tissues. Proc Natl Acad Sci USA 101:5339-5346.

Yoshikawa T, Sellix M, Pezuk P, and Menaker M (2009) Timing of the ovarian circadian clock is regulated by gonadotrophins. Endocrinology 150:4338-4347.

at Dicle Ãœniversitesi on November 5, 2014jbr.sagepub.comDownloaded from