LH and hCG action on the same receptor results in quantitatively and qualitatively different intracellular signalling

Livio Casarini, Monica Lispi, Salvatore Longobardi, Fabiola Milosa, Antonio La Marca, Daniela Tagliasacchi, Elisa Pignatti, Manuela Simoni, Livio Casarini, Monica Lispi, Salvatore Longobardi, Fabiola Milosa, Antonio La Marca, Daniela Tagliasacchi, Elisa Pignatti, Manuela Simoni

Abstract

Human luteinizing hormone (hLH) and chorionic gonadotropin (hCG) act on the same receptor (LHCGR) but it is not known whether they elicit the same cellular and molecular response. This study compares for the first time the activation of cell-signalling pathways and gene expression in response to hLH and hCG. Using recombinant hLH and recombinant hCG we evaluated the kinetics of cAMP production in COS-7 and hGL5 cells permanently expressing LHCGR (COS-7/LHCGR, hGL5/LHCGR), as well as cAMP, ERK1/2, AKT activation and progesterone production in primary human granulosa cells (hGLC). The expression of selected target genes was measured in the presence or absence of ERK- or AKT-pathways inhibitors. In COS-7/LHCGR cells, hCG is 5-fold more potent than hLH (cAMP ED(50): 107.1±14.3 pM and 530.0±51.2 pM, respectively). hLH maximal effect was significantly faster (10 minutes by hLH; 1 hour by hCG). In hGLC continuous exposure to equipotent doses of gonadotropins up to 36 hours revealed that intracellular cAMP production is oscillating and significantly higher by hCG versus hLH. Conversely, phospho-ERK1/2 and -AKT activation was more potent and sustained by hLH versus hCG. ERK1/2 and AKT inhibition removed the inhibitory effect on NRG1 (neuregulin) expression by hLH but not by hCG; ERK1/2 inhibition significantly increased hLH- but not hCG-stimulated CYP19A1 (aromatase) expression. We conclude that: i) hCG is more potent on cAMP production, while hLH is more potent on ERK and AKT activation; ii) hGLC respond to equipotent, constant hLH or hCG stimulation with a fluctuating cAMP production and progressive progesterone secretion; and iii) the expression of hLH and hCG target genes partly involves the activation of different pathways depending on the ligand. Therefore, the LHCGR is able to differentiate the activity of hLH and hCG.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Dose-response and time-course experiments.
Figure 1. Dose-response and time-course experiments.
a. Dose-response experiment to r-hLH and r-hCG in COS7/LHCGR in the presence of 500 µM IBMX. Total cAMP was measured after 3 hours of incubation and the results of four independent experiments were plotted (Mean±SEM). b. Time-course experiment performed by continuous incubation of COS7/LHCGR for different time-points in the presence of 500 µM IBMX and gonadotropins at ED50 doses (500 pM r-hLH; 100 pM r-hCG). Intracellular cAMP was measured (n = 3; Mean±SEM). Asterisk indicates the significant differences of hLH vs hCG (t-test; p<0.05).
Figure 2. Long-term kinetics of intracellular cAMP…
Figure 2. Long-term kinetics of intracellular cAMP production and extracellular progesterone and cAMP release in a primary hGLC sample continuously exposed to r-hLH or r-hCG.
Absolute values of intracellular cAMP (bars) and extracellular progesterone (lines) production by hGLC cultured in the presence IBMX (500 µM) under continuous stimulation by hLH (500 pM) or hCG (100 pM) for up to 36 hours: (a) 0–12 hours, (b) 13–24 hours, (c) 25–36 hours. cAMP basal level of the unstimulated control cells is also shown for each time-point. d. Measurement of extracellular cAMP, released in culture medium. One representative of three independent measurement is shown for each graph. Please notice the different progesterone scale on the Y axis on right side.
Figure 3. Long-term kinetics of intracellular cAMP…
Figure 3. Long-term kinetics of intracellular cAMP production in hGLC continuously exposed to r-hLH or r-hCG.
hGLC were cultured in the presence IBMX (500 µM) under continuous stimulation by r-hLH (500 pM) or r-hCG (100 pM) for up to 36 hours: (a) 0–12 hours, (b) 13–24 hours, (c) 25–36 hours. Results are means±SEM (n = 3). Asterisks indicate significant differences between hCG and hLH at the given time point (n = 3; t-test; p<0.05).
Figure 4. Cellular localization of LHCGR and…
Figure 4. Cellular localization of LHCGR and morphological changes of hGLC under continuous exposure to hLH 500 (pM) by confocal microscopy.
a. Unstimulated cells (control) after 1 hour. b. hLH-treated cells after 1 hour. c. Unstimulated cells after 15 hours. d. LHCGR sequestration from cell surface in hLH-treated hGLC, after 15 hours. e. Unstimulated cells after 24 hours. f. Cell-rounding in hLH-treated cells after 24 hours. LHCGR is labeled in red (Tritc), the cytoplasmic marker ERK1/2 in green (Fitch) and cell nuclei marker (DAPI) in blue. The merging of the three images is in the lower right plate of each panel. Images are from one experiment and are representative of three independent experiments with similar results. White arrows indicated LHCGR; Scale bar: 10 µm. Western blot control for the anti-LHCGR antibody and non-permeabilized cells control for immunofluorescence staining were also included (Fig. S5).
Figure 5. Dose-response experiment evaluating the maximal…
Figure 5. Dose-response experiment evaluating the maximal phospho-ERK1/2 and phospho-AKT activation in hGLC by Western blotting.
The cells were stimulated for 15 minutes by different r-hLH or r-hCG doses and the phospho-ERK1/2 and phospho-AKT signals were normalized for total ERK. The image is representative of 3 independent experiments.
Figure 6. Time-course analysis of the phospho-ERK1/2…
Figure 6. Time-course analysis of the phospho-ERK1/2 activation in hGLC under 100 pM hLH or 100 pM hCG stimulation.
a. Comparison of phospho-ERK1/2 activation in hLH-stimulated vs unstimulated (control) at different time-points, by Western blotting (image is representative of 4 independent experiments; total ERK as normalizer). b. Comparison of phospho-ERK1/2 activation in hCG-stimulated vs unstimulated (control) at different time-points, by Western blotting (image is representative of 4 independent experiments; total ERK as normalizer). c, d. Relative semi-quantification of the optical density representing phospho-ERK1/2 activation (shown in figs. 6A,B) stimulated by (c) hLH or (d) hCG, compared to unstimulated (Mean±SEM; n = 4; * = significant vs unstimulated; t-test; p<0.05). e. Normalization of the phospho-ERK1/2 signals measured in hLH-stimulated samples (represented as relative units in Fig. 6C) over each unstimulated (Mean±SEM; n = 4; * = significant vs unstimulated; two-way analysis of variance; p<0.05). f. Normalization of the phospho-ERK1/2 signals measured in hCG-stimulated samples (represented as relative units in Fig. 6D) over each unstimulated (Mean±SEM; n = 4; * = significant vs unstimulated; two-way analysis of variance; p<0.05).
Figure 7. Time-course analysis of the phospho-AKT…
Figure 7. Time-course analysis of the phospho-AKT activation in hGLC under 100 pM hLH or 100 pM hCG stimulation.
a. Comparison of phospho-AKT activation in hLH-stimulated vs unstimulated (control) at different time-points, by Western blotting (image is representative of 4 independent experiments; total ERK as normalizer). b. Comparison of phospho-AKT activation in hCG-stimulated vs unstimulated (control) at different time-points, by Western blotting (image is representative of 4 independent experiments; total ERK as normalizer). c, d. Relative semi-quantification of the optical density representing phospho-AKT activation (shown in figs. 7A,B) stimulated by (C) hLH or (D) hCG, compared to unstimulated (Mean±SEM; n = 4; * = significant vs unstimulated; t-test; p<0.05). e. Normalization of the phospho-AKT signals measured in hLH-stimulated samples (represented as relative units in Fig. 7C) over each unstimulated (Mean±SEM; n = 4; * = significant vs unstimulated; two-way analysis of variance; p<0.05). f. Normalization of the phospho-AKT signals measured in hCG-stimulated samples (represented as relative units in Fig. 7D) over each unstimulated (Mean±SEM; n = 4; * = significant vs unstimulated; two-way analysis of variance; p<0.05).
Figure 8. Evaluation of the gene expression…
Figure 8. Evaluation of the gene expression induced in hGLC by hLH or hCG stimulation after 12 hours, performed by real-time PCR.
Specific ERK1/2- or AKT-pathways inhibitor (U0126 and LY294002 respectively) were also used where indicated. a. Increase in the gene expression of the EGF-like factors amphiregulin (AREG) and epiregulin (EREG) induced by LH or hCG. b. Effects of the hLH or hCG stimulation and of the ERK1/2- or AKT pathway early inhibition on neuregulin 1 (NRG1) gene expression. c. Effects of the hLH or hCG stimulation and of the ERK1/2- or AKT pathway early inhibition on NRG1 gene expression. In each treatments, RPS7 gene expression was used as normalizer (mean±SEM; n = 4; * = significant vs control; ° = significant vs hLH-stimulated; t-test; p<0.05).

References

    1. Ascoli M, Fanelli F, Segaloff DL (2002) The lutropin/choriogonadotropin receptor, a 2002 perspective. Endocr Rev 23 2:141–74.
    1. Guan R, Feng X, Wu X, Zhang M, Zhang X, et al. (2009) Bioluminescence resonance energy transfer studies reveal constitutive dimerization of the human lutropin receptor and a lack of correlation between receptor activation and the propensity for dimerization. J Biol Chem 284 12:7483–94.
    1. Khan-Sabir N, Beshay VE, Carr BR (2008) The Normal Menstrual Cycle and the Control of Ovulation. In: Endotext, Chapter 3. 2008. Avaiable:
    1. Birken S, Maydelman Y, Gawinowicz MA, Pound A, Liu Y, et al. (1996) Isolation and characterization of human pituitary chorionic gonadotropin. Endocrinology 137 4:1402–11.
    1. Stewart F, Allen WR (1981) Biological functions and receptor binding activities of equine chorionic gonadotrophins. J Reprod Fertil 62 2:527–36.
    1. Cole LA (2009) New discoveries on the biology and detection of human chorionic gonadotropin. Reprod Biol Endocrinol 7: 8.
    1. Santen RJ, Bardin CW (1973) Episodic luteinizing hormone secretion in man. Pulse analysis, clinical interpretation, physiologic mechanisms. J Clin Invest 52 10:2617–28.
    1. Penny R, Olambiwonnu NO, Frasier SD (1977) Episodic fluctuations of serum gonadotropins in pre- and post-pubertal girls and boys. J Clin Endocrinol Metab 45 2:307–11.
    1. Diebel ND, Bogdanove EM (1978) Analysis of luteinizing hormone and follicle-stimulating hormone release kinetics during a dynamic secretory event, the postpartum preovulatory surge in the rat, based on quantitative changes in stored and circulating luteinizing hormone and follicle-stimulating hormone and metabolic clearance data for these hormones. Endocrinology 103 3:665–73.
    1. Lähteenmäki P (1978) The disappearance of HCG and return of pituitary function after abortion. Clin Endocrinol (Oxf) 9 2:101–12.
    1. Mann K, Lamerz R, Hellmann T, Kümper HJ, Staehler G, et al. (1980) Use of human chorionic gonadotropin and alpha-fetoprotein radioimmunoassays: specificity and apparent half-life determination after delivery and in patients with germ cell tumors. Oncodev Biol Med 1 4–5:301–12.
    1. Norman RJ, Buchholz MM, Somogyi AA, Amato F (2000) hCGbeta core fragment is a metabolite of hCG: evidence from infusion of recombinant hCG. J Endocrinol 164 3:299–305.
    1. Ludwig M, Westergaard LG, Diedrich K, Andersen CY (2003) Developments in drugs for ovarian stimulation. Best Pract Res Clin Obstet Gynaecol 17 2:231–47.
    1. Rose MP, Gaines Das RE, Balen AH (2000) Definition and measurement of follicle stimulating hormone. Endocr Rev 21 1:5–22.
    1. Lunenfeld B (2002) Development of gonadotrophins for clinical use. Reprod Biomed Online 4 Suppl 1: 11–7.
    1. Galet C, Ascoli M (2005) The differential binding affinities of the luteinizing hormone (LH)/choriogonadotropin receptor for LH and choriogonadotropin are dictated by different extracellular domain residues. Mol Endocrinol 19 5:1263–76.
    1. Gromoll J, Eiholzer U, Nieschlag E, Simoni M (2000) Male hypogonadism caused by homozygous deletion of exon 10 of the luteinizing hormone (LH) receptor: differential action of human chorionic gonadotropin and LH. J Clin Endocrinol Metab 85 6:2281–6.
    1. Müller T, Gromoll J, Simoni M (2003) Absence of exon 10 of the human luteinizing hormone (LH) receptor impairs LH, but not human chorionic gonadotropin action. J Clin Endocrinol Metab 88 5:2242–9.
    1. Schiffer Z, Keren-Tal I, Deutsch M, Dantes A, Aharoni D, et al. (1996) Fourier analysis of differential light scattering for the quantitation of FSH response associated with structural changes in immortalized granulosa cells. Mol Cell Endocrinol 118 1–2:145–53.
    1. Amsterdam A, Sasson R, Keren-Tal I, Aharoni D, Dantes A, et al. (2003) Alternative pathways of ovarian apoptosis: death for life. Biochem Pharmacol 66 8:1355–62.
    1. Amsterdam A, Keren-Tal I, Aharoni D, Dantes A, Land-Bracha A, et al. (2003) Steroidogenesis and apoptosis in the mammalian ovary. Steroids 68 10–13:861–7.
    1. Ben-Ami I, Armon L, Freimann S, Strassburger D, Ron-El R, et al. (2009) EGF-like growth factors as LH mediators in the human corpus luteum. Hum Reprod 24 1:176–84.
    1. Park J-Y, Su Y-Q, Ariga M, Law E, Jin S-LC, et al. (2004) EGF-like growth factors as mediators of LH action in the ovulatory follicle. Science 303 5658:682–4.
    1. Donadeu FX, Ascoli M (2005) The differential effects of the gonadotropin receptors on aromatase expression in primary cultures of immature rat granulosa cells are highly dependent on the density of receptors expressed and the activation of the inositol phosphate cascade. Endocrinology 146 9:3907–16.
    1. Lindeberg M, Carlström K, Ritvos O, Hovatta O (2007) Gonadotrophin stimulation of non-luteinized granulosa cells increases steroid production and the expression of enzymes involved in estrogen and progesterone synthesis. Hum Reprod 22 2:401–6.
    1. Rainey WH, Sawetawan C, Shay JW, Michael MD, Mathis JM, et al. (1994) Transformation of human granulosa cells with the E6 and E7 regions of human papillomavirus. J Clin Endocrinol Metab 78 3:705–10.
    1. Nordhoff V, Sonntag B, von Tils D, Götte M, Schüring AN, et al. (2011) Effects of the FSH receptor gene polymorphism p.N680S on cAMP and steroid production in cultured primary human granulosa cells. Reprod Biomed Online 23 2:196–203.
    1. Amsterdam A, Hanoch T, Dantes A, Tajima K, Strauss JF, et al. (2002) Mechanisms of gonadotropin desensitization. Mol Cell Endocrinol 187 1–2:69–74.
    1. Mosmann T (1983) Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 65 1–2:55–63.
    1. Lin PC, Li X, Lei ZM, Rao ChV (2003) Human cervix contains functional luteinizing hormone/human chorionic gonadotropin receptors. J Clin Endocrinol Metab 88 7:3409–14.
    1. Menon B, Franzo-Romain M, Damanpour S, Menon KMJ (2011) Luteinizing hormone receptor mRNA down-regulation is mediated through ERK-dependent induction of RNA binding protein. Mol Endocrinol 25 2:282–90.
    1. Palaniappan M, Menon KM (2010) Human chorionic gonadotropin stimulates theca-interstitial cell proliferation and cell cycle regulatory proteins by a cAMP-dependent activation of AKT/mTORC1 signaling pathway. Mol Endocrinol 24 9:1782–93.
    1. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)). Method Methods 25 4:402–8.
    1. Hofer AM, Lefkimmiatis K (2007) Extracellular calcium and cAMP: Second messengers as “third messenger”? Physiology (Bethesda) 22: 320–7.
    1. Lawrence TS, Ginzberg RD, Gilula NB, Beers WH (1979) Hormonally Induced Cell Shape Changes in Cultured Rat Ovarian Granulosa Cells. The Journal of Cell Biology 80 1:21–36.
    1. Molskness TA, Zelinski-Wooten MB, Hild-Petito SA, Stouffer RL (1991) Comparison of the steroidogenic response of luteinized granulosa cells from rhesus monkeys to luteinizing hormone and chorionic gonadotropin. Biol Reprod 45 2:273–81.
    1. Duijkers IJ, Beerens MC, Coelingh Bennink HJ, Huisman JA, Rombout F, et al. (1995) Pharmacokinetics of two human menopausal gonadotrophin preparations after single intravenous administration during pituitary suppression. Hum Reprod 10 6:1367–72.
    1. Zeleznik AJ (1998) In vivo responses of the primate corpus luteum to luteinizing hormone and chorionic gonadotropin. Proc Natl Acad Sci USA 95 18:11002–7.
    1. Cailleux-Bounacer A, Reznik Y, Cauliez B, Menard JF, Duparc C, et al. (2008) Evaluation of endocrine testing of Leydig cell function using extractive and recombinant human chorionic gonadotropin and different doses of recombinant human LH in normal men. Eur J Endocrinol 159 2:171–8.
    1. Van Hell, Matthijsen R, Overbeek GA (1964) Effects of human menopausal gonadotrophin preparations in different bioassay methods. Acta Endocrinol 47: 409–18.
    1. Cole LA (2009) hCG and hyperglycosylated hCG in the establishment and evolution of hemochorial placentation. J Reprod Immunol 82 2:112–8.
    1. Nagirnaja L, Rull K, Uusküla L, Hallast P, Grigorova M, et al. (2010) Genomics and genetics of gonadotropin beta-subunit genes: Unique FSHB and duplicated LHB/CGB loci. Mol Cell Endocrinol 329 1–2:4–16.
    1. Stanton PG, Pozvek G, Burgon PG, Robertson DM, Hearn MT (1993) Isolation and characterization of human LH isoforms. J Endocrinol 138 3:529–43.
    1. Stanton PG, Burgon PG, Hearn MT, Robertson DM (1996) Structural and functional characterisation of hFSH and hLH isoforms. Mol Cell Endocrinol 125 1–2:133–41.
    1. Almeida BE, Oliveira JE, Carvalho CM, Dalmora SL, Bartolini P, et al. (2010) Analysis of human luteinizing hormone and human chorionic gonadotropin preparations of different origins by reversed-phase high-performance liquid chromatography. J Pharm Biomed Anal 53 1:90–7.
    1. Lottersberger C, Hoermann R, Mann K, Schwarz S, Berger P (2003) Tumor- and pregnancy-derived isoforms of human chorionic gonadotropin: biological and diagnostic relevance. Horm Res 59 3:125–34.
    1. Cole LA (2012) hCG, the wonder of today's science. Reprod Biol Endocrinol 10: 24.
    1. Amoresano A, Siciliano R, Orrù S, Napoleoni R, Altarocca V, et al. (1996) Structural characterisation of human recombinant glycohormones follitropin, lutropin and choriogonadotropin expressed in Chinese hamster ovary cells. Eur J Biochem 242 3:608–18.
    1. Lambert A, Talbot JA, Anobile CJ, Robertson WR (1998) Gonadotrophin heterogeneity and biopotency: implications for assisted reproduction. Mol Hum Reprod 4 7:619–29.
    1. Gervais A, Hammel YA, Pelloux S, Lepage P, Baer G, et al. (2003) Glycosylation of human recombinant gonadotrophins: characterization and batch-to-batch consistency. Glycobiology 13 3:179–89.
    1. Talbot JA, Mitchell R, Hoy AM, Lambert A, Gauher A, et al. (1996) Recombinant human luteinizing hormone: a partial physicochemical, biological and immunological characterization. Mol Hum Reprod 2 10:799–806.
    1. le Cotonnec JY, Porchet HC, Beltrami V, Munafo A (1998) Clinical pharmacology of recombinant human luteinizing hormone: Part I. Pharmacokinetics after intravenous administration to healthy female volunteers and comparison with urinary human luteinizing hormone. Fertil Steril 1998 69 2:189–94.
    1. Trinchard-Lugan I, Khan A, Porchet HC, Munafo A (2002) Pharmacokinetics and pharmacodynamics of recombinant human chorionic gonadotrophin in healthy male and female volunteers. Reprod Biomed Online 4 2:106–15.
    1. Gibson-Berry KL, Chase DJ (1990) Continuous and pulsatile infusions of luteinizing hormone have identical effects on steroidogenic capacity and sensitivity of Leydig cells in rats passively immunized against gonadotropin-releasing hormone. Endocrinology 126 6:3107–15.
    1. Albrecht ED, Pepe GJ (1990) Placental steroid hormone biosynthesis in primate pregnancy. Endocr Rev 11 1:124–50.
    1. Evans AC, Martin F (2000) Kinase pathways in dominant and subordinate ovarian follicles during the first wave of follicular development in sheep. Anim Reprod Sci 64 3–4:221–31.
    1. Ryan KE, Glister C, Lonergan P, Martin F, Knight PG, et al. (2008) Functional significance of the signal transduction pathways Akt and Erk in ovarian follicles: in vitro and in vivo studies in cattle and sheep. J Ovarian Res 1 1:2.
    1. Godinho RO, Costa VL Jr (2003) Regulation of intracellular cyclic AMP in skeletal muscle cells involves the efflux of cyclic nucleotide to the extracellular compartment. Br J Pharmacol 138 5:995–1003.
    1. Segaloff DL (2010) Constitutive activity of the lutropin receptor and its allosteric modulation by receptor heterodimerization. Meth Enzymol 484: 231–52.
    1. Rozenfeld R, Devi LA (2011) Exploring a role for heteromerization in GPCR signalling specificity. Biochem J 433 1:11–8.
    1. Krishnamurthy H, Kishi H, Shi M, Galet C, Bhaskaran RS, et al. (2003) Postendocytotic trafficking of the follicle-stimulating hormone (FSH)-FSH receptor complex. Mol Endocrinol 17 11:2162–76.
    1. Quellari M, Desroches A, Beau I, Beaudeux E, Misrahi M (2003) Role of cleavage and shedding in human thyrotropin receptor function and trafficking. Eur J Biochem 270 17:3486–97.
    1. Magalhaes AC, Dunn H, Ferguson SS (2012) Regulation of GPCR activity, trafficking and localization by GPCR-interacting proteins. Br J Pharmacol 165 6:1717–36.
    1. Karlsson AB, Maizels ET, Flynn MP, Jones JC, Shelden EA, et al. (2010) Luteinizing hormone receptor-stimulated progesterone production by preovulatory granulosa cells requires protein kinase A-dependent activation/dephosphorylation of the actin dynamizing protein cofilin. Mol Endocrinol 24 9:1765–81.
    1. Noma N, Kawashima I, Fan H-Y, Fujita Y, Kawai T, et al. (2011) LH-induced neuregulin 1 (NRG1) type III transcripts control granulosa cell differentiation and oocyte maturation. Mol Endocrinol 25 1:104–16.
    1. Chin EC, Abayasekara DRE (2004) Progesterone secretion by luteinizing human granulosa cells: a possible cAMP-dependent but PKA-independent mechanism involved in its regulation. J Endocrinol 183 1:51–60.
    1. Gupta C, Chapekar T, Chhabra Y, Singh P, Sinha S, et al. (2012) Differential response to sustained stimulation by hCG and LH on goat ovarian granulosa cells. Indian J Med Res 331–40.
    1. Brown C, LaRocca J, Pietruska J, Ota M, Anderson L, et al. (2010) Subfertility caused by altered follicular development and oocyte growth in female mice lacking PKB alpha/Akt1. Biol Reprod 82 2:246–56.
    1. Yamashita S, Tai P, Charron J, Ko C, Ascoli M (2011) The Leydig cell MEK/ERK pathway is critical for maintaining a functional population of adult Leydig cells and for fertility. Mol Endocrinol 25 7:1211–22.
    1. Choi J-H, Chen C-L, Poon SL, Wang H-S, Leung PCK (2009) Gonadotropin-stimulated epidermal growth factor receptor expression in human ovarian surface epithelial cells: involvement of cyclic AMP-dependent exchange protein activated by cAMP pathway. Endocr Relat Cancer 16 1:179–88.
    1. Yamashita Y, Hishinuma M, Shimada M (2009) Activation of PKA, p38 MAPK and ERK1/2 by gonadotropins in cumulus cells is critical for induction of EGF-like factor and TACE/ADAM17 gene expression during in vitro maturation of porcine COCs. J Ovarian Res 2: 20.
    1. Ulloa-Aguirre A, Crépieux P, Poupon A, Maurel M-C, Reiter E (2011) Novel pathways in gonadotropin receptor signaling and biased agonism. Rev Endocr Metab Disord 12 4:259–74.
    1. Insel PA, Zhang L, Murray F, Yokouchi H, Zambon AC (2012) Cyclic AMP is both a pro-apoptotic and anti-apoptotic second messenger. Acta Physiol (Oxf) 204 2:277–87.
    1. Keren-Tal I, Suh BS, Dantes A, Lindner S, Oren M, et al. (1995) Involvement of p53 expression in cAMP-mediated apoptosis in immortalized granulosa cells. Exp Cell Res 218 1:283–95.
    1. Amsterdam A, Dantes A, Liscovitch M (1994) Role of phospholipase-D and phosphatidic acid in mediating gonadotropin-releasing hormone-induced inhibition of preantral granulosa cell differentiation. Endocrinology 135 3:1205–11.
    1. Aharoni D, Dantes A, Oren M, Amsterdam A (1995) cAMP-mediated signals as determinants for apoptosis in primary granulosa cells. Exp Cell Res 218 1:271–82.
    1. Sun Q-Y, Miao Y-L, Schatten H (2009) Towards a new understanding on the regulation of mammalian oocyte meiosis resumption. Cell Cycle 8 17:2741–7.
    1. Johnson AL, Bridgham JT, Swenson JA (2001) Activation of the Akt/protein kinase B signaling pathway is associated with granulosa cell survival. Biol Reprod 64 5:1566–74.
    1. Peter AT, Dhanasekaran N (2003) Apoptosis of granulosa cells: a review on the role of MAPK-signalling modules. Reprod Domest Anim 38 3:209–13.
    1. Craig J, Orisaka M, Wang H, Orisaka S, Thompson W, et al. (2007) Gonadotropin and intra-ovarian signals regulating follicle development and atresia: the delicate balance between life and death. Front Biosci 12: 3628–39.
    1. Motola S, Popliker M, Tsafriri A (2008) Response of follicle cells to ovulatory stimuli within the follicle and in primary culture. Mol Cell Endocrinol 282 1–2:26–31.
    1. Zamah AM, Hsieh M, Chen J, Vigne JL, Rosen MP, et al. (2010) Human oocyte maturation is dependent on LH-stimulated accumulation of the epidermal growth factor-like growth factor, amphiregulin. Hum Reprod 25 10:2569–78.
    1. Sekiguchi T, Mizutani T, Yamada K, Kajitani T, Yazawa T, et al. (2004) Expression of epiregulin and amphiregulin in the rat ovary. J Mol Endocrinol 33 1:281–91.
    1. McAllister JM, Mason JI, Byrd W, Trant JM, Waterman MR, et al. (1990) Proliferating human granulosa-lutein cells in long term monolayer culture: expression of aromatase, cholesterol side-chain cleavage, and 3 beta-hydroxysteroid dehydrogenase. J Clin Endocrinol Metab 71 1:26–33.
    1. Fan H-Y, Liu Z, Johnson PF, Richards JS (2011) CCAAT/enhancer-binding proteins (C/EBP)-α and -β are essential for ovulation, luteinization, and the expression of key target genes. Mol Endocrinol 25 2:253–68.
    1. Baer G, Loumaye E (2003) Comparison of recombinant human luteinising hormone (r-hLH) and human menopausal gonadotropin (hMG) in assisted reproductive technology. Curr Med Res Opin 19 2:83–8.

Source: PubMed

3
Subscribe