Am J Physiol Endocrinol Metab 296: E520 E531, First published January 13, 2009; doi: /ajpendo

Size: px
Start display at page:

Download "Am J Physiol Endocrinol Metab 296: E520 E531, First published January 13, 2009; doi: /ajpendo"

Transcription

1 Am J Physiol Endocrinol Metab 296: E520 E531, First published January 13, 2009; doi: /ajpendo KiSS-1 in the mammalian ovary: distribution of kisspeptin in human and marmoset and alterations in KiSS-1 mrna levels in a rat model of ovulatory dysfunction F. Gaytán, 1 M. Gaytán, 3 J. M. Castellano, 1 M. Romero, 1,3 J. Roa, 1 B. Aparicio, 3 N. Garrido, 3 J. E. Sánchez-Criado, 1 R. P. Millar, 4 A. Pellicer, 3 H. M. Fraser, 4 and M. Tena-Sempere 1,2 1 Department of Cell Biology, Physiology and Immunology, University of Córdoba, 2 CIBER, Fisiopatología de la Obesidad y Nutrición, Córdoba, and 3 Instituto Valenciano de Infertilidad, Valencia, Spain; and 4 Medical Research Council Human Reproductive Sciences Unit, Centre for Reproductive Biology, Queen s Medical Research Institute, Edinburgh, United Kingdom Submitted 6 November 2008; accepted in final form 7 January 2009 Gaytán F, Gaytán M, Castellano JM, Romero M, Roa J, Aparicio B, Garrido N, Sánchez-Criado JE, Millar RP, Pellicer A, Fraser HM, Tena-Sempere M. KiSS-1 in the mammalian ovary: distribution of kisspeptin in human and marmoset and alterations in KiSS-1 mrna levels in a rat model of ovulatory dysfunction. Am J Physiol Endocrinol Metab 296: E520 E531, First published January 13, 2009; doi: /ajpendo Kisspeptins, the products of the KiSS-1 gene acting via G protein-coupled receptor 54 (GPR54), have recently emerged as pivotal signals in the hypothalamic network triggering the preovulatory surge of gonadotropins and, hence, ovulation. Additional actions of kisspeptins at other levels of the hypothalamic-pituitary-ovarian axis have been suggested but remain to date scarcely studied. We report herein the pattern of expression of KiSS-1 and GPR54 in the human and nonhuman primate ovary and evaluate changes in ovarian KiSS-1 expression in a rat model of ovulatory dysfunction. KiSS-1 and GPR54 mrnas were detected in human ovarian tissue and cultured granulosa-lutein cells. In good agreement, kisspeptin immunoreactivity was observed in cyclic human and marmoset ovaries, with prominent signals in the theca layer of growing follicles, corpora lutea, interstitial gland, and ovarian surface epithelium. GPR54 immunoreactivity was also found in human theca and luteal cells. Administration of indomethacin to cyclic female rats disturbed ovulation and resulted in a dramatic drop in ovarian KiSS-1, but not GPR54, cyclooxygenase-2 (COX-2), or progesterone receptor, mrna levels at the time of ovulation; an effect mimicked by the selective COX-2 inhibitor NS398 and rescued by coadministration of PGE 2. Likewise, the stimulatory effect of human choriogonadotropin on ovarian KiSS-1 expression was partially blunted by indomethacin. In contrast, KiSS-1 mrna levels remained unaltered in another model of ovulatory failure, i.e., the RU486- treated rat. In summary, we document for the first time the expression of KiSS-1/kisspeptin and GPR54 in the human and nonhuman primate ovary. In addition, we provide evidence for the ability of inhibitors of COX-2, known to disturb follicular rupture and ovulation, to selectively alter the expression of KiSS-1 gene in rat ovary. Altogether, our results are suggestive of a conserved role of local KiSS-1 in the direct control of ovarian functions in mammals. G protein-coupled receptor 54; follicle; ovulation; rat THE HYPOTHALAMIC-PITUITARY-OVARIAN AXIS is a complex neurohormonal system responsible for the regulated secretion of ovarian hormones and the cyclic release of fertilizable oocytes Address for reprint requests and other correspondence: F. Gaytán and M. Tena-Sempere, Dept. of Cell Biology, Physiology and Immunology, Faculty of Medicine, Univ. of Córdoba, Avda. Menéndez Pidal s/n, Córdoba, Spain ( fi1tesem@uco.es). at ovulation (39, 47). The latter is triggered by the preovulatory surge of LH, evoked by the positive feedback effects of estrogen at midcycle (22). This hormonal surge induces the sequential expression of a series of genes in the ovary (8, 23, 50), which finally leads to the release of the cumulus-oocyte complex (COC) to the periovarian space. The central mechanisms responsible for the generation of the preovulatory surge of gonadotropins (22, 28), and the subsequent molecular events in the ovary, have been actively investigated recently. Proteolytic degradation of the extracellular matrix is mandatory for successful ovulation, and several proteolytic enzymes, including plasminogen activator-plasmin and matrix metalloproteinase (MMP) systems, have been involved in this process (4, 32, 52). Additionally, protease inhibitors, such as plasminogen activator inhibitors (PAIs) and tissue inhibitors of MMPs (TIMPs), are also concomitantly expressed during ovulation (4, 52). Indeed, the balance between proteolytic and antiproteolytic activities appears essential for the selective rupture of follicular apex, and conditions suspected to disturb such a balance are known to cause ovulatory dysfunction. Thus in cycling rats treated with the nonsteroidal anti-inflammatory drug indomethacin, a potent inhibitor of cyclooxygenase-1 (COX-1) and COX-2, follicle rupture occurs frequently at the basolateral follicular areas, with the COC being released to the ovarian stroma (16, 17, 19). In this model, degradation of the perifollicular tissues, invasion of blood and lymphatic vessels, degradation of the ovarian bursa, and invasion of the periovarian fat pad can be observed (17, 19). Such limited but detectable invasive capacity of granulosa and cumulus cells after COX blockade partially resembles tumor dissemination and trophoblast invasion of the maternal decidua. Whether the genes restraining cell invasion in tumor metastasis and placental formation also play also a role in preventing perifollicular invasion during normal ovulation is yet to be evaluated. KiSS-1 was originally identified as a metastasis-suppressor gene that encodes a family of structurally related peptides, termed kisspeptins, which acting via the G protein-coupled receptor 54 (GPR54) were shown to prevent tumor spread (33, 36, 37, 40). In addition, a role for kisspeptins in controlling trophoblast invasion was suggested (2). However, such antimetastatic and anti-invasive activities were recently eclipsed The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. E /09 $8.00 Copyright 2009 the American Physiological Society

2 by the demonstration of the fundamental roles of KiSS-1/ kisspeptins and GPR54 in reproductive physiology (36, 37, 40). Indeed, in the last 4 yr, KiSS-1 neurons in the forebrain have been demonstrated to play key functions in the dynamic control of gonadotropin secretion along the life span (20, 36, 37, 40), kisspeptins being very potent elicitors of LH and FSH secretion in different mammalian species, including humans (5, 6, 21, 27, 29, 30, 37, 42). Specifically in the female, the hypothalamic KiSS-1/GPR54 system seems to operate as central conduit not only for the negative, but also for the positive, feedback regulation of gonadotropins, thereby playing a substantial role in the generation of the preovulatory surge of LH (24, 38, 44). While the contention that kisspeptins act primarily at central (hypothalamic) levels to regulate ovarian function is well defined, the possibility of additional effects at other sites of the hypothalamic-pituitary-ovarian axis cannot be ruled out on the basis of the published experimental evidence (38). In fact, initial analyses in rodents suggested the expression of KiSS-1 and GPR54 genes in the rat ovary (48). In addition, discernible kisspeptin and GPR54 immunoreactivity was recently demonstrated in ovarian tissue sections from cyclic rats, where KiSS-1 gene expression was shown to fluctuate in a cycledependent manner under the regulation of pituitary LH (3). Likewise, cyclic KiSS-1 gene expression and kisspeptin-ir has been demonstrated in rat oviduct (18). Moreover, ovarian expression of GPR54 has been also recently documented in fish (10, 31). Taken together, these observations are suggestive of a potential (and, likely, evolutionary conserved) role of kisspeptins in the local control of ovarian function (38). Yet, the physiological relevance of such a role, if any, and whether the elements of the KiSS-1/GPR54 system are also expressed in the primate ovary remain unexplored to date. In this context, we report herein expression analyses of KiSS-1 and GPR54 in the human and marmoset monkey ovary and present evidence for altered ovarian expression of KiSS-1 gene in a rat model of ovulatory dysfunction due to systemic administration of indomethacin. MATERIALS AND METHODS Tissue (Human/Marmoset) Samples Human samples. Sections from normal human cyclic ovaries were obtained from the files of the Department of Pathology of University of Cordoba, upon approval of the local Committee of Bioethics. Tissues had been fixed in phosphate-buffered formaldehyde and routinely processed for paraffin embedding. Ovaries from oophorectomized women due to uterine pathology, who did not show ovarian disease, were not undergoing hormonal therapy, and displayed normal cyclicity, were used. The phase of the cycle was assigned by considering the menstrual history and dating of the endometrium and corpora lutea (9, 14). At least five ovaries from the follicular (from days 1 to 14) and luteal (from days 15 to 28) phase of the cycle were studied for kisspeptin-ir. Tissue fragments from human ovaries were also submitted to histological analyses for GPR54 immunoreactivity. These specimens were obtained at the Centre for Reproductive Biology (11), after approval by the Lothian Paediatrics and Reproductive Medicine Ethics Committee; all women gave informed consent. Gene expression analyses were conducted in human ovarian tissue and cultured granulosa-lutein cells obtained at the Hospital Universitario Dr. Peset (Valencia, Spain) and Instituto Valenciano de Infertilidad. Procedures were approved by the local Institutional Review Board on the use of human subjects in research and, when applicable, comply with the Spanish Law of Assisted Reproductive Technologies (14/2006). Fragments of ovarian tissue were obtained upon laparoscopic biopsy in regularly cyclic, reproductively healthy women. Cultures of human granulosa-lutein cells were carried out using granulosa cells obtained from women undergoing programmed ovarian gonadotropin stimulation for ART (assisted reproductive technique) procedures, as described in detail elsewhere (12, 26). These granulosa cells undergo incipient luteinization due to in vivo gonadotropin priming and in vitro culture; therefore, they are granulosalutein cells. Marmoset samples. Adult (2 3 yr old) female common marmoset monkeys (Callithrix jacchus), with a body weight of 350 g, regular (28 day) ovulatory cycles, and housed under standard conditions, were used. The ovaries used in this study had been collected previously, following standard procedures for immunohistochemical analysis (49). Experimental (Rat) Studies E521 Adult Wistar female rats bred in the vivarium of University of Córdoba were used. The animals were maintained under constant conditions of light (14 h of light, from 0700) and temperature (22 C). Experimental procedures were approved by Córdoba University Ethical Committee for animal experimentation and conducted in accordance with the European Union normative. In all experiments, the animals were monitored for estrous cyclicity by daily vaginal cytology; only rats with at least two consecutive regular 4-day estrous cycles were used for subsequent studies. The animals were killed by decapitation at the end of the experimental procedures, when ovaries were removed and processed for morphometric analysis or RNA isolation (see below). Unless otherwise stated, all reagents were obtained from Sigma (St. Louis, MO). As model of ovulatory dysfunction, female rats were treated with inhibitors of COX-1 and/or COX-2 (16, 17), and ovaries were collected for gene expression analysis and/or assessment of ovulation. In experiment 1, the effects of indomethacin, a nonselective COX inhibitor, on the expression levels of a panel of ovulation-related genes in the ovary were studied at the periovulatory period. Groups of cyclic female rats (n 6) were subcutaneously injected with indomethacin (1.0 mg/rat) or vehicle in the morning (1000) of proestrus, and ovarian samples were obtained upon decapitation of the animals in the evening of proestrus (2100) or early morning of estrus (0200), the latter being roughly coincident with ovulation. Ovarian tissue was obtained also from cyclic females (not treated with indomethacin) at the morning of proestrus and estrus. In all groups, ovaries were dissected out of the surrounding fat pad, snap frozen in liquid nitrogen, and stored at 80 C until used for RNA analyses. Considering that previous data from our group demonstrated the ability of human choriogonadotropin (hcg) to acutely increase KiSS-1 mrna levels in rat ovary (3), and given the inhibitory effects of indomethacin on KiSS-1 mrna levels detected in our initial experiments, in experiment 2, the effects of indomethacin on hcginduced KiSS-1 mrna expression in the ovary were explored. Cyclic female rats were subcutaneously injected in the morning of proestrus (900) with an effective dose of the potent gonadotropin-releasing hormone (GnRH) antagonist ORG (GnRH-ANT, 5 mg/kg; Organon, Oss, The Netherlands) and were subsequently treated at 1200 with an ovulatory bolus of hcg (25 IU/rat sc). Thereafter, groups of GnRH-ANT/hCG-treated animals (n 6) were subcutaneously injected with indomethacin (1.0 mg/rat) or vehicle and were sequentially killed by decapitation after 1, 3, 6, and 12 h for ovarian sampling. The effects of selective COX inhibitors on ovulatory efficiency and KiSS-1 gene expression were compared with those of indomethacin in experiment 3. Groups (n 6) of cyclic female rats, treated in the morning of proestrus (1100) with an effective dose of GnRH-ANT, were subsequently injected subcutaneously at 1300 with indomethacin

3 E522 (1.0 mg/rat), the selective inhibitor of COX-1 SC560 (10 mg/kg), or the selective inhibitor of COX-2 NS398 (10 mg/kg). The animals received at 1700 an ovulatory dose of hcg (25 IU/rat sc) and were killed by decapitation after 3 h (2000). In addition, groups of treated females (n 6) were maintained until the morning of estrus, when ovaries were embedded in paraffin and serially sectioned for morphometric assessment of ovulation efficiency (17). Finally, we aimed to provide functional evidence for the association between altered prostaglandin synthesis (after COX-2 inhibition) and disturbed KiSS-1 mrna expression. To this end, in experiment 4, cyclic female rats (n 6), injected with indomethacin (1.0 mg/rat) in the morning (1000) of proestrus, were subsequently treated with two subcutaneously boluses of PGE 2 (500 g/injection; at 1900 and 2000) or vehicle, and ovarian samples were obtained in the evening of proestrus (2100). Ovarian tissue from proestrous females not treated with indomethacin served as controls. In addition, in experiment 5, ovarian KiSS-1 mrna levels were assayed in another model of ovulatory failure, the RU486-treated female rat. Cyclic female rats (n 6) were subcutaneously injected in the morning (1000) of proestrus with an effective dose of RU486 (2 mg/rat) or vehicle, and ovarian samples were obtained in the evening (2100) of proestrus. Analysis of RNA Expression by RT-PCR Total RNA was isolated from human ovarian fragments and cultured granulosa-lutein cells using TRIzol reagent, following the instructions of the manufacturer. Samples of rat ovarian tissue were processed for RNA isolation as described previously (3). In human samples, qualitative detection of KiSS-1 and GPR54 mrna was conducted by RT-PCR, using primer pairs and conditions included in Table 1; coamplification of the 240-bp fragment of S11 ribosomal protein mrna was used as internal control. In rat ovarian samples, expression of KiSS-1 and GPR54 mrnas was assessed by RT-PCR, optimized for semiquantitative detection, using primer pairs and conditions described in Table 1, in keeping with our previous reference (3). Expression of the genes encoding COX-2 and progesterone Fig. 1. Expression of the genes encoding kisspeptins (KiSS-1) and G proteincoupled receptor 54 (GPR54) in fragments of human ovarian tissue and cultured granulosa-lutein cells. Representative RT-PCR assays from three independent ovarian samples (1 3) and cultures of granulosa-lutein cells are presented. Amplicons of the expected 350-bp (KiSS-1) and 118-bp (GPR54) size were obtained, the identity of which was confirmed by direct sequencing. Amplification of ribosomal protein S11 transcripts served as an internal control. receptor (PR-AB) was also assayed, following previously published protocols (see Table 1), as these genes are induced in rat ovary by the preovulatory surge of LH (25, 35). As an internal control for RT and reaction efficiency, amplification of a 290-bp fragment of L19 ribosomal protein mrna was carried out. For semiquantitative analyses, the number of amplification cycles was chosen for each target based in our previous optimization curves, testing different numbers of PCR cycles, to attain exponential amplification conditions. Specificity of PCR products was confirmed by direct sequencing (Central Sequencing Service, University of Cordoba, Cordoba, Spain). When relevant, quantification of the intensity of RT-PCR signals was carried out by Table 1. Oligo-primer pairs and PCR conditions used for amplification of KiSS-1, GPR54, and RP-S11 mrnas in the human ovary, as well as KiSS-1, GPR54, COX-2, and PR-AB transcripts in the rat ovary Target Oligo Primers Expected Size PCR Cycles/Temp. Human KiSS-1 (hkiss1) 350 bp 38/56 C 5 -TGA ACT CAC TGG TTT CTT GGC A-3 5 -CGA AGG AGT TCC AGT TGT AGT T-3 Human GPR54 (hgpr54) 118 bp 37/63 C 5 -AGT CGG GAA CTC ACT GGT CAT C-3 5 -GGT ACG CAG CAC AGA AGG AAA-3 Human RP-S11 (S11) 240 bp 26/58 C 5 -CAT TCA GAC GGA GCG TGC TTA C-3 5 -TGC ATC TTC ATC TTC GTC AC-3 Rat KiSS-1 (rkiss1) 202 bp 34/62.5 C 5 -TGG CAC CTG TGG TGA ACC CTG AAC-3 5 -ATC AGG CGA CTG CGG GTG GCA CAC-3 Rat GPR54 (rgpr54) 197 bp 34/62.5 C 5 -TGT GCA AAT TCG TCA ACT ACA TCC-3 5 -AGC ACC GGG GCG GAA ACA GCT GC-3 Rat COX-2 (rcox-2) 244 bp 28/55 C 5 -GGG AAG CCT TCT CCA ACC-3 5 -GAA CCC AGG TCC TCG CTT-3 Rat PR-AB (rprab) 326 bp 33/57.5 C 5 -CCC ACA GGA GTT TGT CAA GCT C-3 5 -TAA CTT CAG ACA TCA TTT CCG G-3 Rat RP-L19 (rl19) 290 bp 24/55 C 5 -GAA ATC GCC AAT GCC AAC TC-3 5 -ACC TTC AGG TAC AGG CTG TG-3 For each target, the primer pair used for amplification is included. In addition, the expected size of the generated cdna products, number of cycles selected for exponential amplification, and annealing temperature for RT-PCR analysis are indicated for each signal. KiSS-1, kisspeptin gene; GPR54, G protein-coupled receptor 54; PR-AB, progesterone receptor; COX-2, cyclooxygenase-2.

4 E523 densitometric scanning using an image analysis system (1-D Manager; TDI, Madrid, Spain), and values of the specific targets were normalized to those of internal controls. In all assays, liquid controls and reactions without RT resulted in negative amplification. Kisspeptin Immunohistochemistry Archival samples of cyclic human ovaries, as well as fixed marmoset ovaries, were used. Processing of marmoset ovaries was as described in detail elsewhere (3, 49), followed by standard paraffin embedding. Ovarian sections (5- m thick) were incubated overnight with a primary anti-human kisspeptin-13 (4 13) antibody (Bachem, Bubendorf, Switzerland) that recognizes all forms of kisspeptins sharing the common 10 amino acid terminal region. Final dilution of the antibody was 1:800 in keeping with previous studies in the rat ovary (3). The sections were processed according to the avidin-biotinperoxidase complex technique, as described elsewhere (46). As positive controls, first trimester human placental samples (generously supplied from the archives of the Pathology Department of the Fig. 2. Immunolocalization of kisspeptins in the cyclic human (A E) and marmoset (F J) ovaries. In humans follicles (A), cytoplasmic immunostaining was restricted to steroidogenic theca cells, whereas in mature corpora lutea (B) strong immunoreactivity was observed in both theca-lutein and granulosa-lutein cells. Intense kisspeptin immunoreactivity was also detected in secondary interstitial cells (C), hilus cells (D), and ovarian surface epithelium (E). In the marmoset monkey ovary, kisspeptin immunostaining was detected in the theca layer of antral follicles (F and G), luteal tissue (F and H), secondary interstitial cells (I), and ovarian surface epithelium (J) in a pattern similar that found in the human ovary (hematoxylin counterstaining).

5 E524 University of Cordoba) were used, as previously described (3). In addition, immunohistochemical controls included omission of the primary anti-kisspeptin antibody and reactions after preabsorption of the antiserum overnight at 4 C with 1 g/ml of mouse kisspeptin [KiSS-1 ( )-NH 2; Phoenix Pharmaceuticals, Belmont, CA]; procedures that completely abolished kisspeptin immunolabeling. GPR54 Immunohistochemistry The peptide sequence CVLGEDNAPL in the carboxyl terminal tail domain of the human GPR54 was custom synthesized by EZ Biolab (Westfield, IN) as immunogen. This sequence is not present in any other proteins in the human genome. The peptide was conjugated via the cysteine to keyhole limpet hemocyanin, and 1 mg was injected into two rabbits in Freund s complete adjuvant, followed by further booster immunizations of 1 mg in Freund s incomplete adjuvant at 2, 4, and 7 wk. The rabbits were bled out (30 ml) at 10 wk and sera precipitated with 33% (NH 4)SO 4, reconstituted in PBS, dialyzed against PBS, and lyophilized. The titer in ELISA was 1:6,000 for both antisera. Antiserum 1210 was used for the findings reported, but antiserum 1212 gave the same results (data not shown). The specificity of the antisera was demonstrated by absence of staining in different ovarian compartments after preabsorption of antisera with the peptide immunogen and when the preimmune serum was used. Immunohistochemistry on ovarian tissue was conducted as follows. Tissue was fixed in 4% neutral buffered formaldehyde or 4% paraformaldehyde before paraffin waxing. Five-micrometer paraffin sections were cut and mounted onto superfrost plus slides (Thermo Shandon). Sections were dewaxed and rehydrated following standard procedures. After quenching of endogenous peroxidase activity, slides were incubated overnight with primary antibody at a 1:750 dilution. Sections were then serially incubated with biotinylated swine antirabbit IgG (Dako, Glostrup, Sweden; 1:500 dilution) and streptavidinperoxidase (Dako; 1:1,000 dilution) and were visualized with diaminobenzidine, as per manufacturers instructions. Presentation of Data and Statistics For gene expression analysis in rat studies, semiquantitative RT- PCR assays were carried out in duplicate from at least four independent RNA samples of each experimental group. Semiquantitative data are presented as means SE. Results were analyzed for statistically significant differences using one- or two-way ANOVA, followed by Student-Newman-Keuls multiple range test (SigmaStat 2.0). P 0.05 was considered significant. RESULTS KiSS-1/GPR54 Gene Expression and Pattern of Kisspeptinand GPR54 Immunoreactivity in Human and Marmoset Ovary Expression of KiSS-1 and GPR54 genes was demonstrated in the cyclic human ovary, as well as in cultured granulosalutein cells (Fig. 1). RT-PCR amplification of human ovarian RNA, using a primer pair designed to amplify a 350-bp fragment of KiSS-1 mrna, resulted in the generation of an amplicon of expected size, the identity of which was confirmed Fig. 3. Immunolocalization of GPR54 in human antral follicles and mature corpora lutea. Clear-cut GPR54 immunoreactivity was detected in the theca layer of antral follicles (A), and the granulosa-lutein cells in the corpus luteum (B and C) that was abolished in negative control sections, including preadsorption of the first antibody with the corresponding peptide (D). Strong immunostaining was also observed in some stromal cells that were particularly abundant in the central cavity and the inner aspect of the granulosa-lutein layer (arrows in C), corresponding to macrophages, as evidenced by parallel immunostaining with the specific marker CD68 (E; hematoxylin counterstaining).

6 E525 by direct sequencing. Likewise, positive amplification, of correct size and sequence, was obtained using a primer pair spanning a 118-bp fragment of GPR54 cdna. Similar RT- PCR profiles were obtained for KiSS-1 and GPR54 transcripts in RNA samples from cultured human granulosa-lutein cells. The presence and pattern of cellular distribution of kisspeptins in the human and marmoset ovary were demonstrated by immunohistochemistry, using a specific polyclonal antibody raised against human kisspeptin-10, an antiserum that was used previously for immunodetection of kisspeptins in human placenta and rat ovary (3). This antibody recognizes all forms of kisspeptin sharing the C-terminal region. As a positive control, strong cytoplasm staining was detected in syncytiotrophoblast cells of placental villi f 3-mo-old human placenta (data not shown). Clear-cut kisspeptin-ir was detected in the human cyclic ovary, where specific signals were mainly located at parenchymal steroidogenic cells. In detail, in growing follicles, kisspeptin-ir appeared in the theca layer of growing follicles, whereas granulosa cells were negative (Fig. 2A). After ovulation, im- Fig. 4. Expression of KiSS-1, GPR54, progesterone receptor (PR)-AB, and cyclooxygenase-2 (COX-2) genes in the rat ovary along the proestrus-to-estrus transition, after systemic administration of indomethacin (INDO) to cyclic female rats. A: representative RT-PCR reactions are shown of ovarian samples taken at the evening of proestrus (P2100) and the early morning of estrus (E0200), from animals treated or not with indomethacin, are shown. For comparative purposes, reactions from samples taken from cyclic females at the morning of proestrus (P1000) and the morning of estrus (E1000) are also presented. For each group, 3 representative independent samples are shown. Parallel amplification of L-19 ribosomal protein mrna served as internal control. B: semiquantitative values of mrna levels of the different targets are means SE of at least 6 independent determinations. Groups with different superscript letters are statistically different (P 0.05, ANOVA followed by Student-Newman-Keuls multiple range test). OD, optical density.

7 E526 munostaining was also present in the nonfully luteinized granulosa cells of just-ruptured postovulatory follicles. Similarly, both theca-lutein and granulosa-lutein cells in mature corpora lutea showed intense immunostaining (Fig. 2B). Otherwise, the stromal component of the corpus luteum was negative. In addition, strong kisspeptin-ir was present in androgen-producing cells, i.e., in the hypertrophic theca cells in atretic follicles (data not shown) and its derivatives, the secondary interstitial cells (Fig. 2C), as well as in hilus Leydig cells (Fig. 2D). Finally, while the ovarian stroma was negative, strong immunostaining was detected in the ovarian surface epithelium (Fig. 2E). In the marmoset ovary, the cellular pattern of kisspeptin-ir was grossly similar to that of the human ovary. Strong immunostaining was present in the theca layer of antral follicles (Fig. 2, F G) and, after ovulation, in steroidogenic luteal cells (Fig. 2, F and H). Kisspeptin-IR was also detected in interstitial cells derived from atretic follicles (Fig. 2I), as well as in the ovarian surface epithelium (Fig. 2J). In contrast, the ovarian stroma was negative for kisspeptin-ir. In addition to the ligand, immunodetection of the canonical kisspeptin receptor GPR54 was conducted in ovarian samples using a polyclonal antiserum raised against the human receptor. Of note, this antibody did not appear to properly recognize the antigen in marmoset tissues. This could be due to the fact that the amino acid sequence CVLGEDNAPL in the C-terminal region of the human GPR54, used as immunogen, corresponds to CV- LGEAGAPL in the monkey, where the heavily charged DN sequence is substituted by the neutral AG residues (underlined). Thus our analyses were solely conducted in human ovaries. Moreover, immunostaining for GPR54 seemed highly sensitive to tissue fixation conditions, and archival material gave barely detectable signal and rather variable results. Accordingly, immunostaining was carried out in freshly isolated, adequately fixed fragments of human ovarian tissue containing corpora lutea and sparse follicles. In these preparations, theca cells of growing follicles showed clear-cut GPR54 immunoreactivity, whereas the granulosa layer appeared negative (Fig. 3A). Intense GPR54 immunoreactivity was also detected in mature corpora lutea (Fig. 3B). In detail, strong cytoplasmic immunostaining was observed in steroidogenic granulosa-lutein cells, while the signal was negligible in theca-lutein cells (Fig. 3C). Specificity of GPR54 immunoreactivity was confirmed by omission or preadsorption (Fig. 3D) of the primary antibody, which consistently abolished luteal immunostaining. Of note, intense immunolabeling was also noticed in irregularly shaped stromal cells that were particularly abundant at the inner border of the granulosa-lutein layer (Fig. 3C), cells that corresponded to macrophages, as indicated by parallel immunostaining with the specific marker CD68 (Fig. 3E). Yet, the nature of this signal is not totally clear as, in contrast to other ovarian cell types and compartments, staining of macrophages was not fully prevented by preadsorption of the primary antibody with the peptide. The fact that our antiserum did not properly recognize marmoset GPR54 made it impossible for us to check if this phenomenon is also detectable in other species. Expression of KiSS-1 and Ovulation-Related Genes in a Rat Model of Ovulatory Dysfunction In addition to expression analyses in the human and marmoset, we conducted functional studies aimed at providing (indirect) evidence for the putative roles of the ovarian KiSS- 1/GPR54 system in the local control of ovulation. To this end, the expression levels of KiSS-1, GPR54, and other ovulationrelated genes, such as COX-2 and progesterone receptor (45), were explored in a rat model of ovulatory dysfunction, namely the cyclic female treated with indomethacin (19). Changes in mrna levels of target genes were selectively assayed at the periovulatory period, namely at 2100 of proestrus and 0200 of estrus. For reference purposes, expression analyses were also conducted in ovaries from control females at the morning (1000) of proestrus and estrus. As shown in Fig. 4, relative levels of KiSS-1 and GPR54 mrnas in control ovaries remained rather invariant along the periovulatory period. Conversely, the expression of the transcript encoding both A and B forms of progesterone receptor, as well as COX-2 mrna levels, rose significantly at the periovulatory period and were dramatically suppressed thereafter. Of note, treatment of cyclic females with indomethacin in the morning of proestrus failed to induce any detectable change in the pattern of ovarian GPR54, PR-AB, and COX-2 mrna expression along the proestrus-to-estrus transition. In striking contrast, relative expression levels of KiSS-1 in the ovary were dramatically suppressed at 2100 proestrus and 0200 estrus by pretreatment with indomethacin (Fig. 4). Given the clear-cut inhibitory effects of indomethacin on ovarian KiSS-1 mrna levels, we explored its ability to blunt hcg-induced KiSS-1 overexpression. Injection of hcg at 1200 proestrus resulted in the expected increase in KiSS-1 mrna levels in the ovary (3), which peaked at 3 h and progressively declined thereafter. In clear contrast, cotreatment with an effective dose of indomethacin fully prevented the rise in ovarian expression of KiSS-1 mrna after hcg stimulation (Fig. 5). Effects of Selective COX Inhibitors on Ovarian KiSS-1 Gene Expression and Ovulation To dissect out the relative contribution of COX-1 and COX-2 pathways to the above phenomenon, the effects of Fig. 5. Relative expression levels of KiSS-1 mrna in ovarian samples, from rats injected with gonadotropin-releasing hormone antagonist (GnRH-ANT) and an ovulatory dose of human choriogonadotropin (hcg; 25 IU/rat) at different timepoints after treatment ( ) ornot( ) with indomethacin. Semiquantitative values of mrna levels of the different targets are means SE of at least 6 independent determinations. Groups with different superscript letters are statistically different (P 0.05, ANOVA followed by Student-Newman-Keuls multiple range test).

8 indomethacin were compared with those of the selective inhibitor of COX-1 SC560 and the selective blocker of COX-2 NS398. In this experiment, the effects of the inhibitors on KiSS-1 mrna levels in the ovary (at 3 h after administration) were correlated with their impact on ovulatory efficiency (21 h after treatment). As observed in control cyclic females, rats treated with the selective COX-1 inhibitor displayed no alterations of the ovulatory process, with a preserved number of effective ovulations (i.e., oocytes released at the oviduct) and no oocytes being trapped in corpora lutea or released to the ovarian stroma. In clear contrast, in all rats treated with indomethacin or the selective inhibitor of COX-2, a significant decrease in the percentage of effective ovulations was noticed, the magnitude of which was higher in indomethacin-treated rats ( 50% decrease vs. 25% reduction in females treated with the selective COX-2 inhibitor). Likewise, trapping of expanded COCs inside luteinized follicles and, to a lesser extent, oocytes released to the ovarian stroma were frequently seen after indomethacin or NS398 treatments (Fig. 6). Of note, these results in adult females are roughly analogous to those obtained previously in gonadotropin-primed immature rats (13) and provide the basis for our gene expression analysis. In keeping with ovulatory data, ovarian KiSS-1 mrna levels remained unaltered after systemic administration of SC560 but were equally and significantly decreased after treatment with either indomethacin or the COX-2 inhibitor NS398 (Fig. 7A). Effects of PGE 2 on Ovarian KiSS-1 Expression Replacement experiments, testing the effects of PGE 2 administration to indomethacin-treated rats, were also carried out. As shown in Fig. 7B, indomethacin injection in the morning of proestrus evoked the expected decrease in ovarian KiSS-1 mrna levels at proestrus evening. Such a lowering in KiSS-1 levels was fully prevented by coadministration of two boluses of an effective dose of PGE 2 (15). Further proof for the selectivity of the effects of COX-2 inhibitors was provided by the demonstration that ovarian KiSS-1 expression is not altered in proestrus rats after treatment with the anti-progestagen RU486 (Fig. 8), a treatment known to induce ovulatory defects distinct from those of indomethacin (13). DISCUSSION E527 To our knowledge, this study provides the first comprehensive overview of the pattern of expression of KiSS-1/kisspeptins and GPR54 in the human and nonhuman (marmoset) Fig. 6. Effects of selective COX inhibitors on ovulatory efficiency in the rat. Cyclic females were treated with a selective COX-1 inhibitor (SC560), selective COX-2 inhibitor (NS398), or indomethacin (inhibitor of both COX-1 and COX- 2). Top: some representative sections of ovaries from indomethacin-treated (A) and NS398-treated (B and C) rats are shown, demonstrating eggs released to the ovarian interstitium (A and C), trapped inside corpus luteum, (B), and invasion of blood vessels with formation of follicular fluid emboli (A). Bottom: quantification of the above changes is presented. In detail, percentages of oocytes found in the oviduct, trapped inside corpus luteum, or released to the ovarian stroma are shown. Groups with different superscript letters are statistically different (P 0.05, ANOVA followed by Student-Newman-Keuls multiple range test). NF, not found; veh, vehicle.

9 E528 the local modulation of ovulation was hypothesized (3, 38). This possibility was further suggested by the previous demonstration of the ability of kisspeptins to regulate protease activity in certain cell systems (2, 51), matrix proteases being crucially involved in the ovulatory process (4, 32). To shed light on such putative role, functional expression analyses of KiSS-1 and GPR54 genes were implemented in a rat model of ovulatory dysfunction induced by systemic administration of COX inhibitors (17, 19). The pivotal role of COX-2 in ovulation has been well established by a number of experimental studies that demonstrated that ovarian expression of COX-2 gene is induced by the preovulatory surge of gonadotropins (43, 45) and pharmacological blockade of COX-2 disrupts ovulation (19). Of note, detailed morphometric analyses proved recently that, instead of merely preventing follicular rupture, indomethacin causes dysorganization of the process of selective proteolysis of follicular apex (16, 17, 19). Given the documented ability of kisspeptins to modulate MMP-2 and MMP-9 expression in other cellular systems (2, 51), we found this model specially suited for the assessment of potential roles of the KiSS-1/ GPR54 system in the local modulation of ovulation. Our studies demonstrated that both periovulatory (Fig. 4, A and B) and hcg-induced (Fig. 5A) expression of KiSS-1 gene in the rat ovary is strongly inhibited by indomethacin, i.e., after blockade of COX activity. This effect seems to be specific, as Fig. 7. Effects of the selective COX inhibitors or PGE 2 replacement on the expression levels of KiSS-1 mrna in rat ovary. A: relative expression levels of KiSS-1 mrna are shown from ovarian samples of rats treated with the selective COX-1 inhibitor (SC560), the selective COX-2 inhibitor (NS398), or indomethacin (as inhibitor of both COX-1 and COX-2). Semiquantitative values of mrna levels are means SE of at least 3 independent determinations done in duplicate. B: relative expression levels of KiSS-1 mrna are shown from ovarian samples taken at the evening of proestrus (P2100) from rats treated with indomethacin and submitted or not to replacement with two boluses of PGE 2. Semiquantitative values of mrna levels are means SE of at least 3 independent determinations done in duplicate. Groups with different superscript letters are statistically different (P 0.05, ANOVA followed by Student-Newman-Keuls multiple range test). primate ovary. As we (3) previously demonstrated in the rat, KiSS-1 and GPR54 genes are expressed in the cyclic human ovary, as well as in human granulosa-lutein cells. In good agreement, kisspeptin- and GPR54 immunoreactivities were also detected in human (kisspeptin/ GPR54) and marmoset (kisspeptin) ovaries. Of note, the pattern of cellular distribution of kisspeptin-ir within ovarian tissues was grossly similar between the human and monkey. Moreover, the location of kisspeptin and GPR54 immunoreactivity seems to also be similar to that in the rat ovary (3). The above commonalities are not only suggestive of a conserved function of the KiSS- 1/GPR54 pair in the local control of ovarian function but also support the usefulness of our experimental approach where joint expression analyses in primates and functional studies in rodent models might help to provide an integral view of the ovarian KiSS-1/GPR54 system in mammals. The physiological relevance of ovarian expression of KiSS-1 and GPR54 is yet to be fully defined but based on its pattern of cellular distribution and hormonally regulated expression in the rat ovary, a potential role of kisspeptins in Fig. 8. Effects of the anti-progestagen RU486 on expression levels of KiSS-1 mrna in rat ovary at the evening of proestrus (P2100). Top: a representative RT-PCR assay of KiSS-1 mrna levels in control (vehicle) and RU486 groups is presented. For each group, 3 representative independent samples are shown. Parallel amplification of L-19 ribosomal protein mrna served as internal control. Bottom: semiquantitative values of mrna levels are means SE of at least 3 independent determinations done in duplicate. No significant difference was detected between groups (Student t-test).

10 it was rescued by PGE 2 replacement but not observed in an unrelated model of ovulatory failure, as in the RU486-treated rat. Moreover, indomethacin failed to alter the expression of other ovulation-related genes, such as COX-2 itself and progesterone receptor, the mrna levels of which were clearly induced at the time preceding ovulation, in keeping with previous observations (35, 43), but were not disturbed by COX inhibition. Likewise, GPR54 mrna levels remained unaltered after indomethacin injection. Although not exhaustive, this set of data is suggestive of the potential association between dysregulated KiSS-1 expression at the time preceding ovulation and the severe impairment of ovulatory efficiency after treatment with indomethacin. This effect seems to be selectively mediated by inhibition of COX-2, as its blockade, but not that of COX-1, mimicked the actions of indomethacin. The molecular mechanisms underlying the potential COX-2-mediated regulation of KiSS-1 expression in the ovary, and its subsequent impact on the ovulatory process, remain to be settled. Yet, from a physiological standpoint, the demonstration of the concurrent decrease in ovarian KiSS-1 expression and disturbed ovulation, despite otherwise normal neuroendocrine events, as evidenced by preserved induction of COX-2 and progesterone receptor genes at the proestrus-to-estrus transition, indirectly points out that temporal and spatial dynamics in local kisspeptin expression might play a role in the tuning of the ovulatory process. The indispensable role of hypothalamic KiSS-1 and GPR54 in the neuroendocrine control of reproduction in general, and ovulation in particular, has been well settled in the last years (37); a major hierarchical role that is not disputed by our present data. Nonetheless, the fact that GnRH/gonadotropin replacement seemed sufficient to rescue the profound hypogonadal state of GPR54-null humans and mice might have obscured additional, less prominent, roles of kisspeptins and GPR54 at other levels of the gonadotropic axis. Concerning the ovary, the relevance of local kisspeptin/ GPR54 signaling is questioned by the fact that women and mice with GPR54 inactivation were successfully induced to ovulate after standard gonadotropin priming (mouse) or GnRH therapy (human) (34, 41), observations that preclude an indispensable role of direct kisspeptin actions for ovulation. However, the efficiency of the ovulatory process after GnRH/gonadotropin priming in the absence of GPR54 signaling has not been thoroughly studied. Thus it remains possible, although yet to be proven, that in spite of detectable oocyte release after protocols of pharmacological stimulation, GPR54-null females may show subtle (qualitative and/or quantitative) alterations in the ovulatory process, such as a decreased percentage of successful ovulation and/or dysregulated follicular rupture. Indeed, the conserved patterns of kisspeptin and GPR54 expression in the primate and rodent ovary, as well as the regulated expression of KiSS-1 gene by key players in the control of ovulation, such as gonadotropins and COX-2, strongly suggest that, although not strictly indispensable for oocyte release, ovarian kisspeptin may participate in the fine tuning of some facets of the ovulatory process. Besides their potential involvement in the control of ovulation, the patterns of tissue distribution of kisspeptins and GPR54 in primate and rodent ovaries are strongly suggestive of regulatory roles on other ovarian functions, the nature and E529 actual relevance of which remain, as yet, unexplored. These might include 1) active tissue remodeling of the follicular and luteal compartments of the ovary along the cycle (7, 45); 2) cell migration and remodeling at the ovarian surface epithelium during postovulatory wound repair (1); and 3) hormone production by steroidogenically active cells of the human ovary, such as theca and luteal cells, as well as the interstitial gland and hilus cells. Detailed structural and functional analyses of the ovaries of gonadotropin-primed GPR54 / mice might help to unravel the roles, if any, of local kisspeptins in the modulation of these functions in the female gonad. In summary, we present herein compelling evidence for the expression, at the gene and peptide levels, of KiSS-1 and GPR54 in the human and nonhuman primate ovary, with obvious similarities with the patterns reported previously in the rat ovary (3). In addition, we document the capacity of inhibitors of COX-2, which induce severe alterations of the ovulatory process, to selectively suppress KiSS-1 expression in the rat ovary at the time of ovulation or after gonadotropin stimulation. Although the indispensable role of the hypothalamic KiSS-1 system in the central control of ovarian function remains undisputed, our data are suggestive of a conserved, likely subordinated, role of local KiSS-1 in the direct control of ovarian physiology in mammals, the relative importance of which awaits to be fully elucidated. ACKNOWLEDGMENTS We thank M. R. Millar for conducting the GPR54 immunohistochemistry. GRANTS This work was supported by Grants BFU , BFU , and BFU (Ministerio de Educacion y Ciencia, Spain) and by funds from Instituto de Salud Carlos III (Red de Centros RCMN C03/08 and Project PI042082; Ministerio de Sanidad, Spain). The Centro de Investigacion Biomedica en Red is an initiative of Instituto de Salud Carlos III (Ministerio de Sanidad, Spain). REFERENCES 1. Auersperg N, Wong AS, Choi KC, Kang SK, Leung PC. Ovarian surface epithelium: biology, endocrinology, and pathology. Endocr Rev 22: , Bilban M, Ghaffari-Tabrizi N, Hintermann E, Bauer S, Molzer S, Zoratti C, Malli R, Sharabi A, Hiden U, Graier W, Knofler M, Andreae F, Wagner O, Quaranta V, Desoye G. Kisspeptin-10, a KiSS-1/metastin-derived decapeptide, is a physiological invasion inhibitor of primary human trophoblasts. J Cell Sci 117: , Castellano JM, Gaytan M, Roa J, Vigo E, Navarro VM, Bellido C, Dieguez C, Aguilar E, Sanchez-Criado JE, Pellicer A, Pinilla L, Gaytan F, Tena-Sempere M. Expression of KiSS-1 in rat ovary: putative local regulator of ovulation? Endocrinology 147: , Curry TE Jr., Osteen KG. Cyclic changes in the matrix metalloproteinase system in the ovary and uterus. Biol Reprod 64: , Dhillo WS, Chaudhri OB, Patterson M, Thompson EL, Murphy KG, Badman MK, McGowan BM, Amber V, Patel S, Ghatei MA, Bloom SR. Kisspeptin-54 stimulates the hypothalamic-pituitary gonadal axis in human males. J Clin Endocrinol Metab 90: , Dhillo WS, Chaudhri OB, Thompson EL, Murphy KG, Patterson M, Ramachandran R, Nijher GK, Amber V, Kokkinos A, Donaldson M, Ghatei MA, Bloom SR. Kisspeptin-54 stimulates gonadotropin release most potently during the preovulatory phase of the menstrual cycle in women. J Clin Endocrinol Metab 92: , Duncan WC, Illingworth PJ, Fraser HM. Expression of tissue inhibitor of metalloproteinases-1 in the primate ovary during induced luteal regression. J Endocrinol 151: , Espey LL, Richards JS. Temporal and spatial patterns of ovarian gene transcription following an ovulatory dose of gonadotropin in the rat. Biol Reprod 67: , 2002.

11 E Fadare O, Zheng W. Histologic dating of the endometrium: accuracy, reproducibility, and practical value. Adv Anat Pathol 12: 39 46, Filby AL, van Aerle R, Duitman J, Tyler CR. The kisspeptin/gonadotropin-releasing hormone pathway and molecular signaling of puberty in fish. Biol Reprod 78: , Fraser HM, Bell J, Wilson H, Taylor PD, Morgan K, Anderson RA, Duncan WC. Localization and quantification of cyclic changes in the expression of endocrine gland vascular endothelial growth factor in the human corpus luteum. J Clin Endocrinol Metab 90: , Garrido N, Albert C, Krussel JS, O Connor JE, Remohi J, Simon C, Pellicer A. Expression, production, and secretion of vascular endothelial growth factor and interleukin-6 by granulosa cells is comparable in women with and without endometriosis. Fertil Steril 76: , Gaytan F, Bellido C, Gaytan M, Morales C, Sanchez-Criado JE. Differential effects of RU486 and indomethacin on follicle rupture during the ovulatory process in the rat. Biol Reprod 69: , Gaytan F, Morales C, Garcia-Pardo L, Reymundo C, Bellido C, Sanchez-Criado JE. Macrophages, cell proliferation, and cell death in the human menstrual corpus luteum. Biol Reprod 59: , Gaytan F, Tarradas E, Bellido C, Morales C, Sanchez-Criado JE. Prostaglandin E(1) inhibits abnormal follicle rupture and restores ovulation in indomethacin-treated rats. Biol Reprod 67: , Gaytan F, Tarradas E, Morales C, Bellido C, Sanchez-Criado JE. Morphological evidence for uncontrolled proteolytic activity during the ovulatory process in indomethacin-treated rats. Reproduction 123: , Gaytan M, Bellido C, Morales C, Sanchez-Criado JE, Gaytan F. Effects of selective inhibition of cyclooxygenase and lipooxygenase pathways in follicle rupture and ovulation in the rat. Reproduction 132: , Gaytan M, Castellano JM, Roa J, Sanchez-Criado JE, Tena-Sempere M, Gaytan F. Expression of KiSS-1 in rat oviduct: possible involvement in prevention of ectopic implantation? Cell Tissue Res 329: , Gaytan M, Morales C, Bellido C, Sanchez-Criado JE, Gaytan F. Non-steroidal anti-inflammatory drugs (NSAIDs) and ovulation: lessons from morphology. Histol Histopathol 21: , Gottsch ML, Clifton DK, Steiner RA. Kisspepeptin-GPR54 signaling in the neuroendocrine reproductive axis. Mol Cell Endocrinol : 91 96, Gottsch ML, Cunningham MJ, Smith JT, Popa SM, Acohido BV, Crowley WF, Seminara S, Clifton DK, Steiner RA. A role for kisspeptins in the regulation of gonadotropin secretion in the mouse. Endocrinology 145: , Herbison AE. Estrogen positive feedback to gonadotropin-releasing hormone (GnRH) neurons in the rodent: The case for the rostral periventricular area of the third ventricle (RP3V). Brain Res Rev 57: , Hernandez-Gonzalez I, Gonzalez-Robayna I, Shimada M, Wayne CM, Ochsner SA, White L, Richards JS. Gene expression profiles of cumulus cell oocyte complexes during ovulation reveal cumulus cells express neuronal and immune-related genes: does this expand their role in the ovulation process? Mol Endocrinol 20: , Kinoshita M, Tsukamura H, Adachi S, Matsui H, Uenoyama Y, Iwata K, Yamada S, Inoue K, Ohtaki T, Matsumoto H, Maeda K. Involvement of central metastin in the regulation of preovulatory luteinizing hormone surge and estrous cyclicity in female rats. Endocrinology 146: , Lim H, Paria BC, Das SK, Dinchuk JE, Langenbach R, Trzaskos JM, Dey SK. Multiple female reproductive failures in cyclooxygenase 2-deficient mice. Cell 91: , Melo MA, Meseguer M, Garrido N, Bosch E, Pellicer A, Remohi J. The significance of premature luteinization in an oocyte-donation programme. Hum Reprod 21: , Messager S, Chatzidaki EE, Ma D, Hendrick AG, Zahn D, Dixon J, Thresher RR, Malinge I, Lomet D, Carlton MB, Colledge WH, Caraty A, Aparicio SA. Kisspeptin directly stimulates gonadotropin-releasing hormone release via G protein-coupled receptor 54. Proc Natl Acad Sci USA 102: , Naftolin F, Garcia-Segura LM, Horvath TL, Zsarnovszky A, Demir N, Fadiel A, Leranth C, Vondracek-Klepper S, Lewis C, Chang A, Parducz A. Estrogen-induced hypothalamic synaptic plasticity and pituitary sensitization in the control of the estrogen-induced gonadotrophin surge. Reprod Sci 14: , Navarro VM, Castellano JM, Fernandez-Fernandez R, Tovar S, Roa J, Mayen A, Barreiro ML, Casanueva FF, Aguilar E, Dieguez C, Pinilla L, Tena-Sempere M. Effects of KiSS-1 peptide, the natural ligand of GPR54, on follicle-stimulating hormone secretion in the rat. Endocrinology 146: , Navarro VM, Castellano JM, Fernandez-Fernandez R, Tovar S, Roa J, Mayen A, Nogueiras R, Vazquez MJ, Barreiro ML, Magni P, Aguilar E, Dieguez C, Pinilla L, Tena-Sempere M. Characterization of the potent luteinizing hormone-releasing activity of KiSS-1 peptide, the natural ligand of GPR54. Endocrinology 146: , Nocillado JN, Levavi-Sivan B, Carrick F, Elizur A. Temporal expression of G-protein-coupled receptor 54 (GPR54), gonadotropinreleasing hormones (GnRH), and dopamine receptor D2 (drd2) in pubertal female grey mullet, Mugil cephalus. Gen Comp Endocrinol 150: , Ny T, Wahlberg P, Brandstrom IJ. Matrix remodeling in the ovary: regulation and functional role of the plasminogen activator and matrix metalloproteinase systems. Mol Cell Endocrinol 187: 29 38, Ohtaki T, Shintani Y, Honda S, Matsumoto H, Hori A, Kanehashi K, Terao Y, Kumano S, Takatsu Y, Masuda Y, Ishibashi Y, Watanabe T, Asada M, Yamada T, Suenaga M, Kitada C, Usuki S, Kurokawa T, Onda H, Nishimura O, Fujino M. Metastasis suppressor gene KiSS-1 encodes peptide ligand of a G-protein-coupled receptor. Nature 411: , Pallais JC, Bo-Abbas Y, Pitteloud N, Crowley WF Jr, Seminara SB. Neuroendocrine, gonadal, placental, and obstetric phenotypes in patients with IHH and mutations in the G-protein coupled receptor, GPR54. Mol Cell Endocrinol : 70 77, Park OK, Mayo KE. Transient expression of progesterone receptor messenger RNA in ovarian granulosa cells after the preovulatory luteinizing hormone surge. Mol Endocrinol 5: , Popa SM, Clifton DK, Steiner RA. The role of kisspeptins and GPR54 in the neuroendocrine regulation of reproduction. Annu Rev Physiol 70: , Roa J, Aguilar E, Dieguez C, Pinilla L, Tena-Sempere M. New frontiers in kisspeptin/gpr54 physiology as fundamental gatekeepers of reproductive function. Front Neuroendocrinol 29: 48 69, Roa J, Tena-Sempere M. KiSS-1 system and reproduction: comparative aspects and roles in the control of female gonadotropic axis in mammals. Gen Comp Endocrinol 153: , Schwartz NB. Neuroendocrine regulation of reproductive cyclicity. In: Neuroendocrinology in Physiology and Medicine, edited by Conn PM and Freeman ME. Totowa, NJ: Humana, 2000, p Seminara SB. Metastin and its G protein-coupled receptor, GPR54: critical pathway modulating GnRH secretion. Front Neuroendocrinol 26: , Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS Jr, Shagoury JK, Bo-Abbas Y, Kuohung W, Schwinof KM, Hendrick AG, Zahn D, Dixon J, Kaiser UB, Slaugenhaupt SA, Gusella JF, O Rahilly S, Carlton MB, Crowley WF Jr, Aparicio SA, Colledge WH. The GPR54 gene as a regulator of puberty. N Engl J Med 349: , Shahab M, Mastronardi C, Seminara SB, Crowley WF, Ojeda SR, Plant TM. Increased hypothalamic GPR54 signaling: a potential mechanism for initiation of puberty in primates. Proc Natl Acad Sci USA 102: , Sirois J, Sayasith K, Brown KA, Stock AE, Bouchard N, Dore M. Cyclooxygenase-2 and its role in ovulation: a 2004 account. Hum Reprod Update 10: , Smith JT, Popa SM, Clifton DK, Hoffman GE, Steiner RA. Kiss1 neurons in the forebrain as central processors for generating the preovulatory luteinizing hormone surge. J Neurosci 26: , Stocco C, Telleria C, Gibori G. The molecular control of corpus luteum formation, function, and regression. Endocr Rev 28: , Tena-Sempere M, Barreiro ML, Gonzalez LC, Gaytan F, Zhang FP, Caminos JE, Pinilla L, Casanueva FF, Dieguez C, Aguilar E. Novel expression and functional role of ghrelin in rat testis. Endocrinology 143: , Tena-Sempere M, Huhtaniemi I. Gonadotropins and gonadotropin receptors. In: Reproductive Medicine Molecular, Cellular and Genetic Fundamentals, edited by Fauser BCJM. New York: Parthenon, 2003, p Terao Y, Kumano S, Takatsu Y, Hattori M, Nishimura A, Ohtaki T, Shintani Y. Expression of KiSS-1, a metastasis suppressor gene, in trophoblast giant cells of the rat placenta. Biochim Biophys Acta 1678: , 2004.

KISSPEPTINS WERE originally identified as the products

KISSPEPTINS WERE originally identified as the products 0013-7227/06/$15.00/0 Endocrinology 147(10):4852 4862 Printed in U.S.A. Copyright 2006 by The Endocrine Society doi: 10.1210/en.2006-0117 Expression of KiSS-1 in Rat Ovary: Putative Local Regulator of

More information

Abbreviations: P- paraffin-embedded section; C, cryosection; Bio-SA, biotin-streptavidin-conjugated fluorescein amplification.

Abbreviations: P- paraffin-embedded section; C, cryosection; Bio-SA, biotin-streptavidin-conjugated fluorescein amplification. Supplementary Table 1. Sequence of primers for real time PCR. Gene Forward primer Reverse primer S25 5 -GTG GTC CAC ACT ACT CTC TGA GTT TC-3 5 - GAC TTT CCG GCA TCC TTC TTC-3 Mafa cds 5 -CTT CAG CAA GGA

More information

Supplementary Appendix

Supplementary Appendix Supplementary Appendix This appendix has been provided by the authors to give readers additional information about their work. Supplement to: Sherman SI, Wirth LJ, Droz J-P, et al. Motesanib diphosphate

More information

c Tuj1(-) apoptotic live 1 DIV 2 DIV 1 DIV 2 DIV Tuj1(+) Tuj1/GFP/DAPI Tuj1 DAPI GFP

c Tuj1(-) apoptotic live 1 DIV 2 DIV 1 DIV 2 DIV Tuj1(+) Tuj1/GFP/DAPI Tuj1 DAPI GFP Supplementary Figure 1 Establishment of the gain- and loss-of-function experiments and cell survival assays. a Relative expression of mature mir-484 30 20 10 0 **** **** NCP mir- 484P NCP mir- 484P b Relative

More information

Supplementary Document

Supplementary Document Supplementary Document 1. Supplementary Table legends 2. Supplementary Figure legends 3. Supplementary Tables 4. Supplementary Figures 5. Supplementary References 1. Supplementary Table legends Suppl.

More information

CD31 5'-AGA GAC GGT CTT GTC GCA GT-3' 5 ' -TAC TGG GCT TCG AGA GCA GT-3'

CD31 5'-AGA GAC GGT CTT GTC GCA GT-3' 5 ' -TAC TGG GCT TCG AGA GCA GT-3' Table S1. The primer sets used for real-time RT-PCR analysis. Gene Forward Reverse VEGF PDGFB TGF-β MCP-1 5'-GTT GCA GCA TGA ATC TGA GG-3' 5'-GGA GAC TCT TCG AGG AGC ACT T-3' 5'-GAA TCA GGC ATC GAG AGA

More information

Supplementary Table 3. 3 UTR primer sequences. Primer sequences used to amplify and clone the 3 UTR of each indicated gene are listed.

Supplementary Table 3. 3 UTR primer sequences. Primer sequences used to amplify and clone the 3 UTR of each indicated gene are listed. Supplemental Figure 1. DLKI-DIO3 mirna/mrna complementarity. Complementarity between the indicated DLK1-DIO3 cluster mirnas and the UTR of SOX2, SOX9, HIF1A, ZEB1, ZEB2, STAT3 and CDH1with mirsvr and PhastCons

More information

Supplementary Materials

Supplementary Materials Supplementary Materials 1 Supplementary Table 1. List of primers used for quantitative PCR analysis. Gene name Gene symbol Accession IDs Sequence range Product Primer sequences size (bp) β-actin Actb gi

More information

Toluidin-Staining of mast cells Ear tissue was fixed with Carnoy (60% ethanol, 30% chloroform, 10% acetic acid) overnight at 4 C, afterwards

Toluidin-Staining of mast cells Ear tissue was fixed with Carnoy (60% ethanol, 30% chloroform, 10% acetic acid) overnight at 4 C, afterwards Toluidin-Staining of mast cells Ear tissue was fixed with Carnoy (60% ethanol, 30% chloroform, 10% acetic acid) overnight at 4 C, afterwards incubated in 100 % ethanol overnight at 4 C and embedded in

More information

Supplementary Figures

Supplementary Figures Supplementary Figures Supplementary Figure 1. H3F3B expression in lung cancer. a. Comparison of H3F3B expression in relapsed and non-relapsed lung cancer patients. b. Prognosis of two groups of lung cancer

More information

a) Primary cultures derived from the pancreas of an 11-week-old Pdx1-Cre; K-MADM-p53

a) Primary cultures derived from the pancreas of an 11-week-old Pdx1-Cre; K-MADM-p53 1 2 3 4 5 6 7 8 9 10 Supplementary Figure 1. Induction of p53 LOH by MADM. a) Primary cultures derived from the pancreas of an 11-week-old Pdx1-Cre; K-MADM-p53 mouse revealed increased p53 KO/KO (green,

More information

Supplemental Data. Shin et al. Plant Cell. (2012) /tpc YFP N

Supplemental Data. Shin et al. Plant Cell. (2012) /tpc YFP N MYC YFP N PIF5 YFP C N-TIC TIC Supplemental Data. Shin et al. Plant Cell. ()..5/tpc..95 Supplemental Figure. TIC interacts with MYC in the nucleus. Bimolecular fluorescence complementation assay using

More information

Supplementary Figure 1 MicroRNA expression in human synovial fibroblasts from different locations. MicroRNA, which were identified by RNAseq as most

Supplementary Figure 1 MicroRNA expression in human synovial fibroblasts from different locations. MicroRNA, which were identified by RNAseq as most Supplementary Figure 1 MicroRNA expression in human synovial fibroblasts from different locations. MicroRNA, which were identified by RNAseq as most differentially expressed between human synovial fibroblasts

More information

Nature Structural & Molecular Biology: doi: /nsmb Supplementary Figure 1

Nature Structural & Molecular Biology: doi: /nsmb Supplementary Figure 1 Supplementary Figure 1 U1 inhibition causes a shift of RNA-seq reads from exons to introns. (a) Evidence for the high purity of 4-shU-labeled RNAs used for RNA-seq. HeLa cells transfected with control

More information

Supplementary Figure 1 a

Supplementary Figure 1 a Supplementary Figure a Normalized expression/tbp (A.U.).6... Trip-br transcripts Trans Trans Trans b..5. Trip-br Ctrl LPS Normalized expression/tbp (A.U.) c Trip-br transcripts. adipocytes.... Trans Trans

More information

Figure S1. Analysis of genomic and cdna sequences of the targeted regions in WT-KI and

Figure S1. Analysis of genomic and cdna sequences of the targeted regions in WT-KI and Figure S1. Analysis of genomic and sequences of the targeted regions in and indicated mutant KI cells, with WT and corresponding mutant sequences underlined. (A) cells; (B) K21E-KI cells; (C) D33A-KI cells;

More information

Kisspeptin stimulates progesterone secretion via the Erk1/2 mitogen-activated protein kinase signaling pathway in rat luteal cells

Kisspeptin stimulates progesterone secretion via the Erk1/2 mitogen-activated protein kinase signaling pathway in rat luteal cells Kisspeptin stimulates progesterone secretion via the Erk1/2 mitogen-activated protein kinase signaling pathway in rat luteal cells Jing Peng, M.D., a Min Tang, M.D., b Bao-Ping Zhang, M.D., a Peng Zhang,

More information

CASE 41. What is the pathophysiologic cause of her amenorrhea? Which cells in the ovary secrete estrogen?

CASE 41. What is the pathophysiologic cause of her amenorrhea? Which cells in the ovary secrete estrogen? CASE 41 A 19-year-old woman presents to her gynecologist with complaints of not having had a period for 6 months. She reports having normal periods since menarche at age 12. She denies sexual activity,

More information

Supplementary Figure 1. ROS induces rapid Sod1 nuclear localization in a dosagedependent manner. WT yeast cells (SZy1051) were treated with 4NQO at

Supplementary Figure 1. ROS induces rapid Sod1 nuclear localization in a dosagedependent manner. WT yeast cells (SZy1051) were treated with 4NQO at Supplementary Figure 1. ROS induces rapid Sod1 nuclear localization in a dosagedependent manner. WT yeast cells (SZy1051) were treated with 4NQO at different concentrations for 30 min and analyzed for

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION doi: 10.1038/nature05883 SUPPLEMENTARY INFORMATION Supplemental Figure 1 Prostaglandin agonists and antagonists alter runx1/cmyb expression. a-e, Embryos were exposed to (b) PGE2 and (c) PGI2 (20μM) and

More information

Thiruvarur Dist.,Tamilnadu, India.

Thiruvarur Dist.,Tamilnadu, India. IOSR Journal of Mathematics (IOSR-JM) e-issn: 2278-5728, p-issn: 2319-765X. Volume 12, Issue 5 Ver. VI (Sep. - Oct.2016), PP 14-19 www.iosrjournals.org A Mathematical Weibull model for desensitization

More information

Supplementary Table 2. Conserved regulatory elements in the promoters of CD36.

Supplementary Table 2. Conserved regulatory elements in the promoters of CD36. Supplementary Table 1. RT-qPCR primers for CD3, PPARg and CEBP. Assay Forward Primer Reverse Primer 1A CAT TTG TGG CCT TGT GCT CTT TGA TGA GTC ACA GAA AGA ATC AAT TC 1B AGG AAA TGA ACT GAT GAG TCA CAG

More information

Table S1. Oligonucleotides used for the in-house RT-PCR assays targeting the M, H7 or N9. Assay (s) Target Name Sequence (5 3 ) Comments

Table S1. Oligonucleotides used for the in-house RT-PCR assays targeting the M, H7 or N9. Assay (s) Target Name Sequence (5 3 ) Comments SUPPLEMENTAL INFORMATION 2 3 Table S. Oligonucleotides used for the in-house RT-PCR assays targeting the M, H7 or N9 genes. Assay (s) Target Name Sequence (5 3 ) Comments CDC M InfA Forward (NS), CDC M

More information

The reproductive lifespan

The reproductive lifespan The reproductive lifespan Reproductive potential Ovarian cycles Pregnancy Lactation Male Female Puberty Menopause Age Menstruation is an external indicator of ovarian events controlled by the hypothalamicpituitary

More information

Culture Density (OD600) 0.1. Culture Density (OD600) Culture Density (OD600) Culture Density (OD600) Culture Density (OD600)

Culture Density (OD600) 0.1. Culture Density (OD600) Culture Density (OD600) Culture Density (OD600) Culture Density (OD600) A. B. C. D. E. PA JSRI JSRI 2 PA DSAM DSAM 2 DSAM 3 PA LNAP LNAP 2 LNAP 3 PAO Fcor Fcor 2 Fcor 3 PAO Wtho Wtho 2 Wtho 3 Wtho 4 DTSB Low Iron 2 4 6 8 2 4 6 8 2 22 DTSB Low Iron 2 4 6 8 2 4 6 8 2 22 DTSB

More information

Current Highlights The role of kisspeptin signalling in control of reproduction in genetically similar species

Current Highlights The role of kisspeptin signalling in control of reproduction in genetically similar species Current Highlights The role of kisspeptin signalling in control of reproduction in genetically similar species Amir Babiker (1), Adnan Al Shaikh (2) (1) King Saud University Medical City and King Saud

More information

Female Reproductive System. Lesson 10

Female Reproductive System. Lesson 10 Female Reproductive System Lesson 10 Learning Goals 1. What are the five hormones involved in the female reproductive system? 2. Understand the four phases of the menstrual cycle. Human Reproductive System

More information

Reproductive Hormones

Reproductive Hormones Reproductive Hormones Male gonads: testes produce male sex cells! sperm Female gonads: ovaries produce female sex cells! ovum The union of male and female sex cells during fertilization produces a zygote

More information

ABSTRACT. Key words: ovulation, ovary, human, follicle, collagen, MMP and TIMP. ISBN-10: ISBN-13:

ABSTRACT. Key words: ovulation, ovary, human, follicle, collagen, MMP and TIMP. ISBN-10: ISBN-13: HUMAN OVULATION Studies on collagens, gelatinases and tissue inhibitors of metalloproteinases Anna Karin Lind Department of Obstetrics and Gynecology Institute of Clinical Sciences Sahlgrenska University

More information

Citation for published version (APA): Oosterveer, M. H. (2009). Control of metabolic flux by nutrient sensors Groningen: s.n.

Citation for published version (APA): Oosterveer, M. H. (2009). Control of metabolic flux by nutrient sensors Groningen: s.n. University of Groningen Control of metabolic flux by nutrient sensors Oosterveer, Maaike IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it.

More information

Reproductive cyclicity 19. Introduction. Page 1. repro and its story lines. Male repro: a simpler way of control

Reproductive cyclicity 19. Introduction. Page 1. repro and its story lines. Male repro: a simpler way of control Reproductive cyclicity 19 Male repro: a simpler way of control Menstrual cycles: ovary / uterine anatomy and cell types, follicular phase, ovulation, luteal phase, cyclicity Race events: removal of P4

More information

Chapter 27 The Reproductive System. MDufilho

Chapter 27 The Reproductive System. MDufilho Chapter 27 The Reproductive System 1 Figure 27.19 Events of oogenesis. Before birth Meiotic events 2n Oogonium (stem cell) Mitosis Follicle development in ovary Follicle cells Oocyte 2n Primary oocyte

More information

Nature Immunology: doi: /ni.3836

Nature Immunology: doi: /ni.3836 Supplementary Figure 1 Recombinant LIGHT-VTP induces pericyte contractility and endothelial cell activation. (a) Western blot showing purification steps for full length murine LIGHT-VTP (CGKRK) protein:

More information

Female Reproductive Physiology. Dr Raelia Lew CREI, FRANZCOG, PhD, MMed, MBBS Fertility Specialist, Melbourne IVF

Female Reproductive Physiology. Dr Raelia Lew CREI, FRANZCOG, PhD, MMed, MBBS Fertility Specialist, Melbourne IVF Female Reproductive Physiology Dr Raelia Lew CREI, FRANZCOG, PhD, MMed, MBBS Fertility Specialist, Melbourne IVF REFERENCE Lew, R, Natural History of ovarian function including assessment of ovarian reserve

More information

Investigation: The Human Menstrual Cycle Research Question: How do hormones control the menstrual cycle?

Investigation: The Human Menstrual Cycle Research Question: How do hormones control the menstrual cycle? Investigation: The Human Menstrual Cycle Research Question: How do hormones control the menstrual cycle? Introduction: The menstrual cycle (changes within the uterus) is an approximately 28-day cycle that

More information

Enzyme linked immunosorbent assay (ELSA) investigation for Human Kisspeptin and progesterone Levels in female having regular menstrual cycle

Enzyme linked immunosorbent assay (ELSA) investigation for Human Kisspeptin and progesterone Levels in female having regular menstrual cycle Original Article Enzyme linked immunosorbent assay (ELSA) investigation for Human Kisspeptin and progesterone Levels in female having regular menstrual cycle * MSc, PhD Fac Med Baghdad 2011; Vol. 53, No.

More information

9.4 Regulating the Reproductive System

9.4 Regulating the Reproductive System 9.4 Regulating the Reproductive System The Reproductive System to unite a single reproductive cell from a female with a single reproductive cell from a male Both male and female reproductive systems include

More information

Kisspeptin and other neuropeptides. New opportunities for reproductive endocrinology Nobel Laureates. Richard A Anderson

Kisspeptin and other neuropeptides. New opportunities for reproductive endocrinology Nobel Laureates. Richard A Anderson Kisspeptin and other neuropeptides Higher centres Timing of puberty Stress New opportunities for reproductive endocrinology E2 +ve E2 -ve Richard A Anderson Obstetrics and Gynaecology University of Edinburgh

More information

Chapter 14 Reproduction Review Assignment

Chapter 14 Reproduction Review Assignment Date: Mark: _/45 Chapter 14 Reproduction Review Assignment Multiple Choice Identify the choice that best completes the statement or answers the question. 1. Use the diagram above to answer the next question.

More information

THE MENSTRUAL CYCLE INA S. IRABON, MD, FPOGS, FPSRM, FPSGE OBSTETRICS AND GYNECOLOGY REPRODUCTIVE ENDOCRINOLOGY AND INFERTILITY

THE MENSTRUAL CYCLE INA S. IRABON, MD, FPOGS, FPSRM, FPSGE OBSTETRICS AND GYNECOLOGY REPRODUCTIVE ENDOCRINOLOGY AND INFERTILITY THE MENSTRUAL CYCLE INA S. IRABON, MD, FPOGS, FPSRM, FPSGE OBSTETRICS AND GYNECOLOGY REPRODUCTIVE ENDOCRINOLOGY AND INFERTILITY REFERENCE Comprehensive Gynecology 7 th edition, 2017 (Lobo RA, Gershenson

More information

Female Reproductive System. Justin D. Vidal

Female Reproductive System. Justin D. Vidal Female Reproductive System Justin D. Vidal If you cannot identify the tissue, then it is probably part of the female reproductive system! Introduction The female reproductive system is constantly changing,

More information

Supplementary Figure 1

Supplementary Figure 1 Metastatic melanoma Primary melanoma Healthy human skin Supplementary Figure 1 CD22 IgG4 Supplementary Figure 1: Immunohisochemical analysis of CD22+ (left) and IgG4 (right), cells (shown in red and indicated

More information

KISSPEPTIN AND GNIH CONTROL OF GNRH IN FEMALE MAMMALS

KISSPEPTIN AND GNIH CONTROL OF GNRH IN FEMALE MAMMALS KISSPEPTIN AND GNIH CONTROL OF GNRH IN FEMALE MAMMALS M.J. Zamiri Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran mjzamiri@gmail.com Introduction Since the discovery

More information

Plasmids Western blot analysis and immunostaining Flow Cytometry Cell surface biotinylation RNA isolation and cdna synthesis

Plasmids Western blot analysis and immunostaining Flow Cytometry Cell surface biotinylation RNA isolation and cdna synthesis Plasmids psuper-retro-s100a10 shrna1 was constructed by cloning the dsdna oligo 5 -GAT CCC CGT GGG CTT CCA GAG CTT CTT TCA AGA GAA GAA GCT CTG GAA GCC CAC TTT TTA-3 and 5 -AGC TTA AAA AGT GGG CTT CCA GAG

More information

www.lessonplansinc.com Topic: Protein Synthesis - Sentence Activity Summary: Students will simulate transcription and translation by building a sentence/polypeptide from words/amino acids. Goals & Objectives:

More information

SUPPLEMENTARY DATA. Supplementary Table 1. Primer sequences for qrt-pcr

SUPPLEMENTARY DATA. Supplementary Table 1. Primer sequences for qrt-pcr Supplementary Table 1. Primer sequences for qrt-pcr Gene PRDM16 UCP1 PGC1α Dio2 Elovl3 Cidea Cox8b PPARγ AP2 mttfam CyCs Nampt NRF1 16s-rRNA Hexokinase 2, intron 9 β-actin Primer Sequences 5'-CCA CCA GCG

More information

Kisspeptin-54 Stimulates Gonadotropin Release Most Potently during the Preovulatory Phase of the Menstrual Cycle in Women

Kisspeptin-54 Stimulates Gonadotropin Release Most Potently during the Preovulatory Phase of the Menstrual Cycle in Women 0021-972X/07/$15.00/0 The Journal of Clinical Endocrinology & Metabolism 92(10):3958 3966 Printed in U.S.A. Copyright 2007 by The Endocrine Society doi: 10.1210/jc.2007-1116 Kisspeptin-54 Stimulates Gonadotropin

More information

Phylogenetic analysis of human and chicken importins. Only five of six importins were studied because

Phylogenetic analysis of human and chicken importins. Only five of six importins were studied because Supplementary Figure S1 Phylogenetic analysis of human and chicken importins. Only five of six importins were studied because importin-α6 was shown to be testis-specific. Human and chicken importin protein

More information

BIOLOGY 621 Identification of the Snorks

BIOLOGY 621 Identification of the Snorks Name: Date: Block: BIOLOGY 621 Identification of the Snorks INTRODUCTION: In this simulation activity, you will examine the DNA sequence of a fictitious organism - the Snork. Snorks were discovered on

More information

REPRODUCCIÓN. La idea fija. Copyright 2004 Pearson Education, Inc., publishing as Benjamin Cummings

REPRODUCCIÓN. La idea fija. Copyright 2004 Pearson Education, Inc., publishing as Benjamin Cummings REPRODUCCIÓN La idea fija How male and female reproductive systems differentiate The reproductive organs and how they work How gametes are produced and fertilized Pregnancy, stages of development, birth

More information

Hot Topics in Translational Endocrinology Endocrine Research

Hot Topics in Translational Endocrinology Endocrine Research JCEM ONLINE Hot Topics in Translational Endocrinology Endocrine Research The Effects of Kisspeptin-10 on Reproductive Hormone Release Show Sexual Dimorphism in Humans Channa N. Jayasena, Gurjinder M. K.

More information

OVARY The surface of the ovary is covered with surface epithelium

OVARY The surface of the ovary is covered with surface epithelium OVARY Cow The ovary, or female gonad, is: 1. an exocrine gland, producing oocytes 2. an endocrine gland, secreting hormones, i.e., estrogen and progesterone OVARY OVARY The surface of the ovary is covered

More information

Supplemental Information. Th17 Lymphocytes Induce Neuronal. Cell Death in a Human ipsc-based. Model of Parkinson's Disease

Supplemental Information. Th17 Lymphocytes Induce Neuronal. Cell Death in a Human ipsc-based. Model of Parkinson's Disease Cell Stem Cell, Volume 23 Supplemental Information Th17 Lymphocytes Induce Neuronal Cell Death in a Human ipsc-based Model of Parkinson's Disease Annika Sommer, Franz Maxreiter, Florian Krach, Tanja Fadler,

More information

Reproduction and Development. Female Reproductive System

Reproduction and Development. Female Reproductive System Reproduction and Development Female Reproductive System Outcomes 5. Identify the structures in the human female reproductive system and describe their functions. Ovaries, Fallopian tubes, Uterus, Endometrium,

More information

REPRODUCTION & GENETICS. Hormones

REPRODUCTION & GENETICS. Hormones REPRODUCTION & GENETICS Hormones http://www.youtube.com/watch?v=np0wfu_mgzo Objectives 2 Define what hormones are; Compare and contrast the male and female hormones; Explain what each hormone in the mail

More information

Fukushima-ku, Osaka. Synopsis. and LH release by investigating the effects of exogenous estrogen on the progesteroneinduced

Fukushima-ku, Osaka. Synopsis. and LH release by investigating the effects of exogenous estrogen on the progesteroneinduced Further Studies on the Causal Relationship between the Secretion of Estrogen and the Release of Luteinizing Hormone in the Rat FUMIHIKO KOBAYASHI, KATSUMI HARA AND TAMOTSU MIYAKE Shionogi Research Laboratory,

More information

Evidence that estrogen mediates the positive feedback effect on GnRH

Evidence that estrogen mediates the positive feedback effect on GnRH Advances in Environmental Biology, 3(3): 244-248, 2009 ISSN 1995-0756 2009, American-Eurasian Network for Scientific Information 244 This is a refereed journal and all articles are professionally screened

More information

Reproductive Endocrinology. Isabel Hwang Department of Physiology Faculty of Medicine University of Hong Kong Hong Kong May2007

Reproductive Endocrinology. Isabel Hwang Department of Physiology Faculty of Medicine University of Hong Kong Hong Kong May2007 Reproductive Endocrinology Isabel Hwang Department of Physiology Faculty of Medicine University of Hong Kong Hong Kong May2007 isabelss@hkucc.hku.hk A 3-hormone chain of command controls reproduction with

More information

Supplementary Figure 1

Supplementary Figure 1 Supplementary Figure 1 Supplementary Figure 1. Lats1/2 deleted ihbs and ihps showed decreased transcripts of hepatocyte related genes (a and b) Western blots (a) and recombination PCR (b) of control and

More information

Supplementary Figure 1a

Supplementary Figure 1a Supplementary Figure 1a Hours: E-cadherin TGF-β On TGF-β Off 0 12 24 36 48 24 48 72 Vimentin βactin Fig. S1a. Treatment of AML12 cells with TGF-β induces EMT. Treatment of AML12 cells with TGF-β results

More information

Two important cells in female are the theca cells and the granulose cells. Granulosa cells are affected by the two gonadotropin hormones; FSH and LH.

Two important cells in female are the theca cells and the granulose cells. Granulosa cells are affected by the two gonadotropin hormones; FSH and LH. 1 UGS physiology sheet #13 lecture 3 Dr.Saleem Khresha. Now we will start discussing the female reproductive system Ovarian Steroids Two important cells in female are the theca cells and the granulose

More information

10.7 The Reproductive Hormones

10.7 The Reproductive Hormones 10.7 The Reproductive Hormones December 10, 2013. Website survey?? QUESTION: Who is more complicated: men or women? The Female Reproductive System ovaries: produce gametes (eggs) produce estrogen (steroid

More information

SISTEMA REPRODUCTOR (LA IDEA FIJA) Copyright 2004 Pearson Education, Inc., publishing as Benjamin Cummings

SISTEMA REPRODUCTOR (LA IDEA FIJA) Copyright 2004 Pearson Education, Inc., publishing as Benjamin Cummings SISTEMA REPRODUCTOR (LA IDEA FIJA) How male and female reproductive systems differentiate The reproductive organs and how they work How gametes are produced and fertilized Pregnancy, stages of development,

More information

Cross-talk between mineralocorticoid and angiotensin II signaling for cardiac

Cross-talk between mineralocorticoid and angiotensin II signaling for cardiac ONLINE SUPPLEMENT TO Crosstalk between mineralocorticoid and angiotensin II signaling for cardiac remodeling An Di ZHANG,,3, Aurelie NGUYEN DINH CAT*,,3, Christelle SOUKASEUM *,,3, Brigitte ESCOUBET, 4,

More information

REPRODUCTIVE CYCLE OF FEMALE MAMMAL

REPRODUCTIVE CYCLE OF FEMALE MAMMAL REPRODUCTIVE CYCLE OF FEMALE MAMMAL Fig. 8-12 Secondary follicles growing follicles increase in number of layers of granulosa cells Tertiary follicles maturing follicles antrum formation fluid filled space

More information

Reproductive System. Testes. Accessory reproductive organs. gametogenesis hormones. Reproductive tract & Glands

Reproductive System. Testes. Accessory reproductive organs. gametogenesis hormones. Reproductive tract & Glands Reproductive System Testes gametogenesis hormones Accessory reproductive organs Reproductive tract & Glands transport gametes provide nourishment for gametes Hormonal regulation in men Hypothalamus - puberty

More information

MICROWELL ELISA LUTEINIZING HORMONE (LH) ENZYMEIMMUNOASSAY TEST KIT LH ELISA. Cat # 4225Z

MICROWELL ELISA LUTEINIZING HORMONE (LH) ENZYMEIMMUNOASSAY TEST KIT LH ELISA. Cat # 4225Z DIAGNOSTIC AUTOMATION, INC. 23961 Craftsman Road, Suite D/E/F, Calabasas, CA 91302 Tel: (818) 591-3030 Fax: (818) 591-8383 onestep@rapidtest.com technicalsupport@rapidtest.com www.rapidtest.com See external

More information

Supplementary Figure 1

Supplementary Figure 1 Supplementary Figure 1 3 3 3 1 1 Bregma -1.6mm 3 : Bregma Ref) Http://www.mbl.org/atlas165/atlas165_start.html Bregma -.18mm Supplementary Figure 1 Schematic representation of the utilized brain slice

More information

Astaxanthin prevents and reverses diet-induced insulin resistance and. steatohepatitis in mice: A comparison with vitamin E

Astaxanthin prevents and reverses diet-induced insulin resistance and. steatohepatitis in mice: A comparison with vitamin E Supplementary Information Astaxanthin prevents and reverses diet-induced insulin resistance and steatohepatitis in mice: A comparison with vitamin E Yinhua Ni, 1,2 Mayumi Nagashimada, 1 Fen Zhuge, 1 Lili

More information

Beta Thalassemia Case Study Introduction to Bioinformatics

Beta Thalassemia Case Study Introduction to Bioinformatics Beta Thalassemia Case Study Sami Khuri Department of Computer Science San José State University San José, California, USA sami.khuri@sjsu.edu www.cs.sjsu.edu/faculty/khuri Outline v Hemoglobin v Alpha

More information

LH (Bovine) ELISA Kit

LH (Bovine) ELISA Kit LH (Bovine) ELISA Kit Catalog Number KA2280 96 assays Version: 05 Intended for research use only www.abnova.com Table of Contents Introduction... 3 Intended Use... 3 Background... 3 Principle of the Assay...

More information

1. During the follicular phase of the ovarian cycle, the hypothalamus releases GnRH.

1. During the follicular phase of the ovarian cycle, the hypothalamus releases GnRH. 1. During the follicular phase of the ovarian cycle, the hypothalamus releases GnRH. 2. This causes the anterior pituitary to secrete small quantities of FSH and LH. 3. At this time, the follicles in the

More information

Biology 12 January 2004 Provincial Examination

Biology 12 January 2004 Provincial Examination Biology 12 January 2004 Provincial Examination ANSWER KEY / SCORING GUIDE CURRICULUM: Organizers 1. Cell Biology 2. Cell Processes and Applications 3. Human Biology Sub-Organizers A, B, C, D E, F, G, H

More information

Hormonal Control of Human Reproduction

Hormonal Control of Human Reproduction Hormonal Control of Human Reproduction Bởi: OpenStaxCollege The human male and female reproductive cycles are controlled by the interaction of hormones from the hypothalamus and anterior pituitary with

More information

Ovarian Follicular Development in the Untreated and

Ovarian Follicular Development in the Untreated and Ovarian Follicular Development in the Untreated and PMSG-treated Cyclic Rat Hajime MIYAMOTO, Goro KATSUURA and Takehiko ISHIBASHI Department of Animal Science, College of Agriculture, Kyoto University,

More information

LH and FSH. Women. Men. Increased LH. Decreased LH. By Ronald Steriti, ND, PhD 2011

LH and FSH. Women. Men. Increased LH. Decreased LH. By Ronald Steriti, ND, PhD 2011 LH and FSH By Ronald Steriti, ND, PhD 2011 Luteinizing hormone (LH) and follicle-stimulating hormone (FSH) are gonadotropins that stimulate the gonads - the testes in males, and the ovaries in females.

More information

Reproductive Health and Pituitary Disease

Reproductive Health and Pituitary Disease Reproductive Health and Pituitary Disease Janet F. McLaren, MD Assistant Professor Division of Reproductive Endocrinology and Infertility Department of Obstetrics and Gynecology jmclaren@uabmc.edu Objectives

More information

A smart acid nanosystem for ultrasensitive. live cell mrna imaging by the target-triggered intracellular self-assembly

A smart acid nanosystem for ultrasensitive. live cell mrna imaging by the target-triggered intracellular self-assembly Electronic Supplementary Material (ESI) for Chemical Science. This journal is The Royal Society of Chemistry 2017 A smart ZnO@polydopamine-nucleic acid nanosystem for ultrasensitive live cell mrna imaging

More information

Supplemental Information. Cancer-Associated Fibroblasts Neutralize. the Anti-tumor Effect of CSF1 Receptor Blockade

Supplemental Information. Cancer-Associated Fibroblasts Neutralize. the Anti-tumor Effect of CSF1 Receptor Blockade Cancer Cell, Volume 32 Supplemental Information Cancer-Associated Fibroblasts Neutralize the Anti-tumor Effect of CSF1 Receptor Blockade by Inducing PMN-MDSC Infiltration of Tumors Vinit Kumar, Laxminarasimha

More information

Endocrine control of female reproductive function

Endocrine control of female reproductive function Medicine School of Women s & Children s Health Discipline of Obstetrics & Gynaecology Endocrine control of female reproductive function Kirsty Walters, PhD Fertility Research Centre, School of Women s

More information

Serum Kisspeptin Levels in Korean Girls with Central Precocious Puberty

Serum Kisspeptin Levels in Korean Girls with Central Precocious Puberty ORIGINAL ARTICLE Pediatrics DOI: 10.3346/jkms.2011.26.7.927 J Korean Med Sci 2011; 26: 927-931 Serum Kisspeptin Levels in Korean Girls with Central Precocious Puberty Young Jun Rhie 1,2, Kee Hyoung Lee

More information

PATIENTS AND METHODS. Subjects

PATIENTS AND METHODS. Subjects PATIENTS AND METHODS Subjects Twenty-nine morbidly obese subjects involved in a gastric surgery program were enrolled in the study between October 25 and March 21. Bariatric surgery was performed in patients

More information

Differential effects of RU486 and indomethacin on follicle. rupture during the ovulatory process in the rat

Differential effects of RU486 and indomethacin on follicle. rupture during the ovulatory process in the rat BOR Papers in Press. Published on February 19, 2003 as DOI:10.1095/biolreprod.102.013755 Differential effects of RU486 and indomethacin on follicle rupture during the ovulatory process in the rat Francisco

More information

Is it the seed or the soil? Arthur Leader, MD, FRCSC

Is it the seed or the soil? Arthur Leader, MD, FRCSC The Physiological Limits of Ovarian Stimulation Is it the seed or the soil? Arthur Leader, MD, FRCSC Objectives 1. To consider how ovarian stimulation protocols work in IVF 2. To review the key events

More information

Sample Provincial exam Q s: Reproduction

Sample Provincial exam Q s: Reproduction Sample Provincial exam Q s: Reproduction 11. Functions Testosterone Makes the male sex organs function normally, and also inhibits hypothalamus s release of GnRH and thus LH & FSH and thus testosterone

More information

CIRCRESAHA/2004/098145/R1 - ONLINE 1. Validation by Semi-quantitative Real-Time Reverse Transcription PCR

CIRCRESAHA/2004/098145/R1 - ONLINE 1. Validation by Semi-quantitative Real-Time Reverse Transcription PCR CIRCRESAHA/2004/098145/R1 - ONLINE 1 Expanded Materials and Methods Validation by Semi-quantitative Real-Time Reverse Transcription PCR Expression patterns of 13 genes (Online Table 2), selected with respect

More information

PITUITARY GONADOTROPINS LH and FSH are structurally

PITUITARY GONADOTROPINS LH and FSH are structurally 0013-7227/05/$15.00/0 Endocrinology 146(9):3917 3925 Printed in U.S.A. Copyright 2005 by The Endocrine Society doi: 10.1210/en.2005-0337 Changes in Hypothalamic KiSS-1 System and Restoration of Pubertal

More information

Web Activity: Simulation Structures of the Female Reproductive System

Web Activity: Simulation Structures of the Female Reproductive System differentiate. The epididymis is a coiled tube found along the outer edge of the testis where the sperm mature. 3. Testosterone is a male sex hormone produced in the interstitial cells of the testes. It

More information

Female Reproduction 1 and 2: The Menstrual Cycle R.J. Witorsch, Ph.D.

Female Reproduction 1 and 2: The Menstrual Cycle R.J. Witorsch, Ph.D. Female Reproduction 1 and 2: The Menstrual Cycle R.J. Witorsch, Ph.D. OBJECTIVES: At the end of this block of lectures, the student should be able to: 1. Describe the functional anatomy of ovary with particular

More information

Effects of Catecholamines and Dibenamine on Ovulation in the Perfused Fowl Ovary

Effects of Catecholamines and Dibenamine on Ovulation in the Perfused Fowl Ovary Effects of Catecholamines and Dibenamine on Ovulation in the Perfused Fowl Ovary Tomoki HIGUCHI, Tomoki SOH, Frank HERTELENDY* and Kousaku TANAKA Faculty of Agriculture, Kyushu University, Higashi-ku,

More information

The intra-follicular molecular biology mandating advancement of egg retrieval in some women

The intra-follicular molecular biology mandating advancement of egg retrieval in some women The intra-follicular molecular biology mandating advancement of egg retrieval in some women David H. Barad, USA Director of Assisted Reproductive Technology, The Center for Human Reproduction New York

More information

AnS 214 SI Multiple Choice Set 4 Weeks 10/14-10/23

AnS 214 SI Multiple Choice Set 4 Weeks 10/14-10/23 AnS 214 SI Multiple Choice Set 4 Weeks 10/14-10/23 The following multiple choice questions pertain to material covered in the last two weeks' lecture sets. Answering the following questions will aid your

More information

Reproductive FSH. Analyte Information

Reproductive FSH. Analyte Information Reproductive FSH Analyte Information 1 Follicle-stimulating hormone Introduction Follicle-stimulating hormone (FSH, also known as follitropin) is a glycoprotein hormone secreted by the anterior pituitary

More information

I. Endocrine System & Hormones Figure 1: Human Endocrine System

I. Endocrine System & Hormones Figure 1: Human Endocrine System I. Endocrine System & Hormones Figure 1: Human Endocrine System Endocrine System: a) Endocrine glands are ductless since they lack specific vessels for the transport of hormones throughout the body. Instead,

More information

Reproductive physiology

Reproductive physiology Reproductive physiology Sex hormones: Androgens Estrogens Gestagens Learning objectives 86 (also 90) Sex Genetic sex Gonadal sex Phenotypic sex XY - XX chromosomes testes - ovaries external features Tha

More information

Endocrinology of the Female Reproductive Axis

Endocrinology of the Female Reproductive Axis Endocrinology of the Female Reproductive Axis girlontheriver.com Geralyn Lambert-Messerlian, PhD, FACB Professor Women and Infants Hospital Alpert Medical School at Brown University Women & Infants BROWN

More information

Kisspeptin: A Potential Factor for Unexplained Infertility and Impaired Embryo Implantation

Kisspeptin: A Potential Factor for Unexplained Infertility and Impaired Embryo Implantation Original Article Kisspeptin: A Potential Factor for Unexplained Infertility and Impaired Embryo Implantation Aaida Mumtaz, MBBS. 1#, Aqsa Khalid, MBBS. 1#, Zehra Jamil, M.Phil. 2, Syeda Sadia Fatima, Ph.D.

More information

Testes (male gonads) -Produce sperm -Produce sex hormones -Found in a sac called the scrotum -Suspended outside of the body cavity for temperature

Testes (male gonads) -Produce sperm -Produce sex hormones -Found in a sac called the scrotum -Suspended outside of the body cavity for temperature REPRODUCTION Testes (male gonads) -Produce sperm -Produce sex hormones -Found in a sac called the scrotum -Suspended outside of the body cavity for temperature reduction -Testes wall made of fibrous connective

More information

SUPPORTING INFORMATION

SUPPORTING INFORMATION SUPPORTING INFORMATION Biology is different in small volumes: endogenous signals shape phenotype of primary hepatocytes cultured in microfluidic channels Amranul Haque, Pantea Gheibi, Yandong Gao, Elena

More information

Physiology of Male Reproductive System

Physiology of Male Reproductive System Physiology of Male Reproductive System the anterior pituitary gland serves as the primary control of reproductive function at puberty Ant Pituitary secretes FSH & large amounts of LH (ICSH) FSH & LH cause

More information