Novel role for anti-Müllerian hormone in the regulation of GnRH neuron excitability and hormone secretion

Irene Cimino, Filippo Casoni, Xinhuai Liu, Andrea Messina, Jyoti Parkash, Soazik P Jamin, Sophie Catteau-Jonard, Francis Collier, Marc Baroncini, Didier Dewailly, Pascal Pigny, Mel Prescott, Rebecca Campbell, Allan E Herbison, Vincent Prevot, Paolo Giacobini, Irene Cimino, Filippo Casoni, Xinhuai Liu, Andrea Messina, Jyoti Parkash, Soazik P Jamin, Sophie Catteau-Jonard, Francis Collier, Marc Baroncini, Didier Dewailly, Pascal Pigny, Mel Prescott, Rebecca Campbell, Allan E Herbison, Vincent Prevot, Paolo Giacobini

Abstract

Anti-Müllerian hormone (AMH) plays crucial roles in sexual differentiation and gonadal functions. However, the possible extragonadal effects of AMH on the hypothalamic-pituitary-gonadal axis remain unexplored. Here we demonstrate that a significant subset of GnRH neurons both in mice and humans express the AMH receptor, and that AMH potently activates the GnRH neuron firing in mice. Combining in vivo and in vitro experiments, we show that AMH increases GnRH-dependent LH pulsatility and secretion, supporting a central action of AMH on GnRH neurons. Increased LH pulsatility is an important pathophysiological feature in many cases of polycystic ovary syndrome (PCOS), the most common cause of female infertility, in which circulating AMH levels are also often elevated. However, the origin of this dysregulation remains unknown. Our findings raise the intriguing hypothesis that AMH-dependent regulation of GnRH release could be involved in the pathophysiology of fertility and could hold therapeutic potential for treating PCOS.

Figures

Figure 1. Specificity tests of anti-AMHR2 antibody…
Figure 1. Specificity tests of anti-AMHR2 antibody and identification of AMHR2-expressing cells in the adult mouse brain.
(ap) Coronal sections immunolabelled with the indicated antibodies, from AMHR2::Cre+/+ (AMHR2-wt, number of immunostained P120 female brains, n=5) and AMHR2::Cre −/− (AMHR2-null, number of immunostained P120 female brains, n=5). AMHR2 expression was also analysed in AMHR2::Cre+/;Z/EG reporter line (h; number of immunostained P60 female brains, n=3). The vector of Z/EG mouse line was designed to provide lacZ expression before Cre excision and EGFP expression after Cre excision and is referred to as Z/EG (lacZ/EGFP). AMHR2 expression was found in the cortex (a,i,m), dentate gyrus of the hippocampus (a,k,o), organum vasculosum of the lamina terminalis (OVLT; c) hypothalamic median eminence (me) and dorsomedial hypothamaus (DMH) arcuate nucleus (Arc; e,f,h). Moreover, AMHR2-expressing cells were found in hypothalamic tanycytes (t) lining the third ventricle (3V) and endothelial (EC) cells (f,h). Brains of AMHR2-null mice were totally depleted of AMHR2 immunoreactivity when using an antibody against an intracellular epitope of the AMHR2 (b,d,g,j,l) or an antibody against the extracellular region of the receptor (n,p). Experiments were replicated at least three times. Scale bars, (a,b) 200 μm; (ce,g) 100 μm; (f,h) 25 μm; (ip) 40 μm (inset in k and o). DAPI, 4′,6-diamidino-2-phenylindole.
Figure 2. AMHR2 is expressed in mouse…
Figure 2. AMHR2 is expressed in mouse and human GnRH neurons.
(ac) Confocal representative photomicrographs showing GnRH and AMHR2 immunoreactivity in sagittal sections of the E12.5 embryonic nasal compartment (number of immunostained E12.5, n=5). Dashed line indicates the boundary between the vomeronasal organ (vno) and the nasal mesenchyme (nm). GnRH neurons migrating out of the vno express AMHR2. (di) Confocal photomicrographs showing GnRH (d,g) and AMHR2 (e,h) immunoreactivity in coronal sections through the hypothalamus of adult female mice (P90–120; number of immunostained brains, n=5). Images show widespread expression of AMHR2 at the level of the organum vasculosum of the lamina terminalis (OVLT). (gi) High-magnifications images of boxed area in d. Arrows point to GnRH neurons expressing AMHR2. (jm) Representative sagittal section of a human fetus at 9 weeks LMP, immunolabelled for GnRH and AMHR2 (number of immunostained fetuses, n=3). At this developmental stage, the majority of GnRH neurons are still located in the nasal region, at the beginning of their migratory path. Double immunofluorescence shows co-expression of these antigens in the same migratory neurons. Experiments were replicated at least three times. cp, cribriform plate; fb, forebrain; oe, olfactory epithelium; vfb, ventral forebrain. (np) Representative coronal section of an adult hypothalamus double labelled for GnRH and AMHR2. Human hypothalami were obtained between 24 and 36 h post mortem from two autopsied individuals: a 20-year-old female and a 72-year-old male subject; 17 out of 29 and 20 out of 35 GnRH neurons exhibited AMHR2 immunoreactivity, respectively (analyses have been performed in eight consecutive 16-μm thick coronal sections for each individual). Scale bars, (ac) 10 μm; (df) 100 μm; (gi) 20 μm, (j) 100 μm, (km) 40 μm, (np) 5 μm.
Figure 3. AMH receptor transcript expression in…
Figure 3. AMH receptor transcript expression in GnRH neurons.
(a) Schematic summarizing the isolation of GnRH-GFP cells. GFP-positive GnRH neurons were isolated by fluorescence-activated cell sorting from the nasal region of E12.5 embryos (n=3) and from the hypothalamic preoptic area of postnatal (females P12, n=3) and adult (females P90, n=3) female mice. (b) Real-time PCR analysis of expression levels of AMHR2 messenger RNA (mRNA) in GnRH cells sorted at E12.5, P12 and P90. AMHR2 transcript expression was detectable in GnRH neurons at all stages, although it reached its highest level at P90. Values are expressed relative to values at E12.5, set at 1, and shown as means±s.e.m. One-way analysis of variance, F(2,8)=7.6, P=0.02. Fisher's least significant difference post hoc test, *: P<0.05 between P90–P120 and E12.5 and P90–P120 versus P12. (c) Relative mRNA expression of AMH type-I receptors (Alk2, Alk3 and Alk6) in GnRH neurons isolated from the preoptic region of adult female mice (females P90, n=3). All receptor transcripts were detectable in adult GnRH neurons. (df) Nasal explants were generated from E11.5 GnRH-GFP embryos (n=7) and maintained in serum-free media for 7 days before immunohistochemistry procedures. Immunostaining for AMHR2 show co-localization with fully differentiated GnRH-GFP neurons. Experiments were replicated three times. LS, lateral septum; MS, medial septum; DBB, diagonal band of Broca; LSI, lateral septal nucleus, intermediate part; LSV, lateral septal nucleus, ventral part; OVLT, organum vasculosum of the lamina terminalis. Scale bars, (df) 10 μm.
Figure 4. AMH increases GnRH neuron firing…
Figure 4. AMH increases GnRH neuron firing and hormone secretion.
(a,b) Cell-attached voltage recordings of GnRH neurons from GnRH::GFP male mice stimulated with AMH. (c,d) Cell-attached voltage recordings of GnRH neurons from GnRH::GFP diestrous mice. AAB (AP5, CNQX and GABAzin) application shows that AMH excitation of GnRH neurons is not dependent upon amino-acid transmission. (e) Whole-cell current recording of a GnRH neuron in the presence of AAB and tetrodotoxin (TTX). **Spontaneous synaptic currents blocked by AAB+TTX. Dots indicate truncated currents induced by the ramp voltage protocol. (f) Current–voltage plots before (black) and during AMH response (AMH, red). The ‘AMH current' (IAMH, green) is obtained by the subtraction of control from AMH current at ramp potentials (−120 to −20 mV for 1.5 s). AMH current with a reversal potential around −30 mV is replotted as an inset with an expanded current axis. (g) Schematics illustrating ME dissection and explant preparation. Hypothalamic MEs were microdissected from adult diestrous or ovariectomized (OVX) female rats. (h) Quantification of GnRH secretion from ME explants of P120 diestrous rats stimulated or not with 125 nM AMH (Die, n=4; Die+AMH, n=4). GnRH mean concentration±s.e.m. Unpaired Student's t-test, t(6)=−6.01, ***P<0.001. (i) MEs dissected from OVX rats (n=4 each group). One-way analysis of variance, F(3,15)=30.9, P<0.0001. ***P<0.0001, **P<0.001 Fisher's least significant difference post hoc test. All the experiments were replicated at least three times.
Figure 5. Intracerebroventricular (i.c.v.) administration of AMH…
Figure 5. Intracerebroventricular (i.c.v.) administration of AMH in vivo increases the LH secretion.
(a) Schematic depicting AMH injection into the lateral cerebral ventricle of diestrous mice. Trunk blood was collected from 3–4-month-old mice 15 or 30 min after the injection. (b) Following AMH administration (AMH 0.5 μM, n=3; AMH 1 μM, n=4; AMH 3 μM, n=6; saline, n=5), LH was measured. Values are expressed as means±s.e.m. One-way analysis of variance, F (3,19)=6.6, P=0.004. *P:<0.01, **P:<0.001, Fisher's least significant difference post hoc test. (c) Adult diestrous mice were injected with AMH 3 μM (n=20), or saline (n=7). Plasma LH secretion peaked at 15 min after injection and returned to basal levels 30 min later (n=7). AMH was injected i.c.v 2 h after an intraperitoneal injection of the GnRH antagonist (n=7). Treatment with an ALK receptor inhibitor prevented the AMH-dependent increase in LH secretion (n=6). One-way analysis of variance, F(4,47)=11.1, P<0.0001. *P<0.01, ***P<0.0001, Fisher's least significant difference post hoc test. (d) Schematic representation of AMH injection into the lateral cerebral ventricle of diestrous (3–4-month old) mice with 50 nM AMH or saline. Tail blood was collected every 10 min for 2 h and LH measured (n=7 control, n=8 AMH; eg). Asterisks in e and f indicate LH pulses in two -representative saline- and AMH-treated mice. Values are represented as means±s.e.m. Unpaired Student's t-test, t(13)=−2.56, *P<0.05. Experiments were replicated three times. MS, medial septum; LSI, lateral septal nucleus, intermediate part; LSV, lateral septal nucleus, ventral part; OVLT, organum vasculosum of the lamina terminalis.
Figure 6. Schematic representation of the proposed…
Figure 6. Schematic representation of the proposed mechanism of action of AMH on GnRH neurons in normal and PCOS women.
In normal women of reproductive age, the levels of circulating AMH are low and do not significantly fluctuate over the menstrual cycle. GnRH neurons express AMHR2 as well as type-I AMH receptors (ALK 2/3/6). In women with PCOS, circulating AMH levels are two to three times higher than in normal women. We hypothesize that AMH could bypass the blood–brain barrier by passing through the fenestrated capillaries at the level of the organum vasculosum of the lamina terminalis (OVLT) and the median eminence and act directly on GnRH dendrites and terminals, respectively. In the median eminence, AMH could also act indirectly on GnRH neurons via tanycytes and vascular endothelial cells, which also express AMHR2, possibly contributing with other dysregulated factors to the increase in GnRH and LH pulsatilities. The altered ratio of LH to FSH is known to be responsible for the ovarian androgen production, partly explaining the two central diagnostic features of PCOS: hyperandrogenemia and hyperandrogenism (hirsutism). FSH, follicle-stimulating hormone; LH, luteinizing hormone.

References

    1. Christian C. A. & Moenter S. M. The neurobiology of preovulatory and estradiol-induced gonadotropin-releasing hormone surges. Endocr. Rev. 31, 544–577 (2010) .
    1. Gonzalez-Martinez D., Hu Y. & Bouloux P. M. Ontogeny of GnRH and olfactory neuronal systems in man: novel insights from the investigation of inherited forms of Kallmann's syndrome. Front. Neuroendocrinol. 25, 108–130 (2004) .
    1. Tsutsumi R. & Webster N. J. GnRH pulsatility, the pituitary response and reproductive dysfunction. Endocr. J. 56, 729–737 (2009) .
    1. Broekmans F. J. & Fauser B. C. Diagnostic criteria for polycystic ovarian syndrome. Endocrine 30, 3–11 (2006) .
    1. Ehrmann D. A. Polycystic ovary syndrome. N. Engl. J. Med. 352, 1223–1236 (2005) .
    1. Taylor A. E. et al. Determinants of abnormal gonadotropin secretion in clinically defined women with polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 82, 2248–2256 (1997) .
    1. Pellatt L. et al. Anti-Mullerian hormone reduces follicle sensitivity to follicle-stimulating hormone in human granulosa cells. Fertil. Steril. 96, 1246–1251 (2011) .
    1. Jonard S. & Dewailly D. The follicular excess in polycystic ovaries, due to intra-ovarian hyperandrogenism, may be the main culprit for the follicular arrest. Hum. Reprod. Update 10, 107–117 (2004) .
    1. Franks S., Stark J. & Hardy K. Follicle dynamics and anovulation in polycystic ovary syndrome. Hum. Reprod. Update 14, 367–378 (2008) .
    1. Pigny P., Jonard S., Robert Y. & Dewailly D. Serum anti-Mullerian hormone as a surrogate for antral follicle count for definition of the polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 91, 941–945 (2006) .
    1. Cook C. L., Siow Y., Brenner A. G. & Fallat M. E. Relationship between serum mullerian-inhibiting substance and other reproductive hormones in untreated women with polycystic ovary syndrome and normal women. Fertil. Steril. 77, 141–146 (2002) .
    1. Pellatt L. et al. Granulosa cell production of anti-Mullerian hormone is increased in polycystic ovaries. J. Clin. Endocrinol. Metab. 92, 240–245 (2007) .
    1. Pigny P. et al. Elevated serum level of anti-mullerian hormone in patients with polycystic ovary syndrome: relationship to the ovarian follicle excess and to the follicular arrest. J. Clin. Endocrinol. Metab. 88, 5957–5962 (2003) .
    1. Cate R. L. et al. Isolation of the bovine and human genes for Mullerian inhibiting substance and expression of the human gene in animal cells. Cell 45, 685–698 (1986) .
    1. Vigier B., Picard J. Y., Tran D., Legeai L. & Josso N. Production of anti-Mullerian hormone: another homology between Sertoli and granulosa cells. Endocrinology 114, 1315–1320 (1984) .
    1. Durlinger A. L. et al. Control of primordial follicle recruitment by anti-Mullerian hormone in the mouse ovary. Endocrinology 140, 5789–5796 (1999) .
    1. di Clemente N. et al. Cloning, expression, and alternative splicing of the receptor for anti-Mullerian hormone. Mol. Endocrinol. 8, 1006–1020 (1994) .
    1. Baarends W. M. et al. A novel member of the transmembrane serine/threonine kinase receptor family is specifically expressed in the gonads and in mesenchymal cells adjacent to the mullerian duct. Development 120, 189–197 (1994) .
    1. Josso N. & Clemente N. Transduction pathway of anti-Mullerian hormone, a sex-specific member of the TGF-beta family. Trends Endocrinol. Metab. 14, 91–97 (2003) .
    1. Lebeurrier N. et al. Anti-Mullerian-hormone-dependent regulation of the brain serine-protease inhibitor neuroserpin. J. Cell Sci. 121, 3357–3365 (2008) .
    1. Wang P. Y. et al. Mullerian inhibiting substance contributes to sex-linked biases in the brain and behavior. Proc. Natl Acad. Sci. USA 106, 7203–7208 (2009) .
    1. Jamin S. P., Arango N. A., Mishina Y., Hanks M. C. & Behringer R. R. Requirement of Bmpr1a for Mullerian duct regression during male sexual development. Nat. Genet. 32, 408–410 (2002) .
    1. Novak A., Guo C., Yang W., Nagy A. & Lobe C. G. Z/EG, a double reporter mouse line that expresses enhanced green fluorescent protein upon Cre-mediated excision. Genesis 28, 147–155 (2000) .
    1. Prevot V. et al. Gonadotrophin-releasing hormone nerve terminals, tanycytes and neurohaemal junction remodelling in the adult median eminence: functional consequences for reproduction and dynamic role of vascular endothelial cells. J. Neuroendocrinol. 22, 639–649 (2010) .
    1. Spergel D. J., Kruth U., Hanley D. F., Sprengel R. & Seeburg P. H. GABA- and glutamate-activated channels in green fluorescent protein-tagged gonadotropin-releasing hormone neurons in transgenic mice. J. Neurosci. 19, 2037–2050 (1999) .
    1. Parkash J. et al. Suppression of beta1-integrin in gonadotropin-releasing hormone cells disrupts migration and axonal extension resulting in severe reproductive alterations. J. Neurosci. 32, 16992–17002 (2012) .
    1. Schwanzel-Fukuda M. & Pfaff D. W. Origin of luteinizing hormone-releasing hormone neurons. Nature 338, 161–164 (1989) .
    1. Wray S., Grant P. & Gainer H. Evidence that cells expressing luteinizing hormone-releasing hormone mRNA in the mouse are derived from progenitor cells in the olfactory placode. Proc. Natl Acad. Sci. USA 86, 8132–8136 (1989) .
    1. Fueshko S. & Wray S. LHRH cells migrate on peripherin fibers in embryonic olfactory explant cultures: an in vitro model for neurophilic neuronal migration. Dev. Biol. 166, 331–348 (1994) .
    1. Giacobini P. et al. Cholecystokinin modulates migration of gonadotropin-releasing hormone-1 neurons. J. Neurosci. 24, 4737–4748 (2004) .
    1. Kevenaar M. E. et al. Serum anti-mullerian hormone levels reflect the size of the primordial follicle pool in mice. Endocrinology 147, 3228–3234 (2006) .
    1. Prevot V. et al. Estradiol coupling to endothelial nitric oxide stimulates gonadotropin-releasing hormone release from rat median eminence via a membrane receptor. Endocrinology 140, 652–659 (1999) .
    1. Ojeda S. R., Urbanski H. F., Costa M. E., Hill D. F. & Moholt-Siebert M. Involvement of transforming growth factor alpha in the release of luteinizing hormone-releasing hormone from the developing female hypothalamus. Proc. Natl Acad. Sci. USA 87, 9698–9702 (1990) .
    1. Halmos G., Schally A. V., Pinski J., Vadillo-Buenfil M. & Groot K. Down-regulation of pituitary receptors for luteinizing hormone-releasing hormone (LH-RH) in rats by LH-RH antagonist Cetrorelix. Proc. Natl Acad. Sci. USA 93, 2398–2402 (1996) .
    1. Pinski J. et al. Chronic administration of the luteinizing hormone-releasing hormone (LHRH) antagonist cetrorelix decreases gonadotrope responsiveness and pituitary LHRH receptor messenger ribonucleic acid levels in rats. Endocrinology 137, 3430–3436 (1996) .
    1. Bedecarrats G. Y., O'Neill F. H., Norwitz E. R., Kaiser U. B. & Teixeira J. Regulation of gonadotropin gene expression by Mullerian inhibiting substance. Proc. Natl Acad. Sci. USA 100, 9348–9353 (2003) .
    1. Steyn F. J. et al. Development of a methodology for and assessment of pulsatile luteinizing hormone secretion in juvenile and adult male mice. Endocrinology 154, 4939–4945 (2013) .
    1. Sullivan S. D. & Moenter S. M. Prenatal androgens alter GABAergic drive to gonadotropin-releasing hormone neurons: implications for a common fertility disorder. Proc. Natl Acad. Sci. USA 101, 7129–7134 (2004) .
    1. Moore A. M., Prescott M. & Campbell R. E. Estradiol negative and positive feedback in a prenatal androgen-induced mouse model of polycystic ovarian syndrome. Endocrinology 154, 796–806 (2013) .
    1. Moore A. M., Prescott M., Marshall C. J., Yip S. H. & Campbell R. E. Enhancement of a robust arcuate GABAergic input to gonadotropin-releasing hormone neurons in a model of polycystic ovarian syndrome. Proc. Natl Acad. Sci. USA 112, 596–601 (2015) .
    1. Renaud E. J., MacLaughlin D. T., Oliva E., Rueda B. R. & Donahoe P. K. Endometrial cancer is a receptor-mediated target for Mullerian Inhibiting Substance. Proc. Natl Acad. Sci. USA 102, 111–116 (2005) .
    1. Segev D. L. et al. Mullerian inhibiting substance inhibits breast cancer cell growth through an NFkappa B-mediated pathway. J. Biol. Chem. 275, 28371–28379 (2000) .
    1. Hoshiya Y. et al. Mullerian inhibiting substance induces NFkB signaling in breast and prostate cancer cells. Mol. Cell. Endocrinol. 211, 43–49 (2003) .
    1. Barbie T. U., Barbie D. A., MacLaughlin D. T., Maheswaran S. & Donahoe P. K. Mullerian Inhibiting Substance inhibits cervical cancer cell growth via a pathway involving p130 and p107. Proc. Natl Acad. Sci. USA 100, 15601–15606 (2003) .
    1. Catlin E. A. et al. Mullerian inhibiting substance inhibits branching morphogenesis and induces apoptosis in fetal rat lung. Endocrinology 138, 790–796 (1997) .
    1. Josso N., Racine C., di Clemente N., Rey R. & Xavier F. The role of anti-Mullerian hormone in gonadal development. Mol. Cell. Endocrinol. 145, 3–7 (1998) .
    1. Josso N. & di Clemente N. TGF-beta Family Members and Gonadal Development. Trends Endocrinol. Metab. 10, 216–222 (1999) .
    1. Wang P. Y. et al. Mullerian inhibiting substance acts as a motor neuron survival factor in vitro. Proc. Natl Acad. Sci. USA 102, 16421–16425 (2005) .
    1. Wittmann W. & McLennan I. S. Anti-Mullerian hormone may regulate the number of calbindin-positive neurons in the sexually dimorphic nucleus of the preoptic area of male mice. Biol. Sex Differ. 4, 18 (2013) .
    1. Wittmann W. & McLennan I. S. The bed nucleus of the stria terminalis has developmental and adult forms in mice, with the male bias in the developmental form being dependent on testicular AMH. Horm. Behav. 64, 605–610 (2013) .
    1. Silverman A. J., Jhamandas J. & Renaud L. P. Localization of luteinizing hormone-releasing hormone (LHRH) neurons that project to the median eminence. J. Neurosci. 7, 2312–2319 (1987) .
    1. Merchenthaler I., Setalo G., Petrusz P., Negro-Vilar A. & Flerko B. Identification of hypophysiotropic luteinizing hormone-releasing hormone (LHRH) neurons by combined retrograde labeling and immunocytochemistry. Exp. Clin. Endocrinol. 94, 133–140 (1989) .
    1. Herbison A. E., Porteous R., Pape J. R., Mora J. M. & Hurst P. R. Gonadotropin-releasing hormone neuron requirements for puberty, ovulation, and fertility. Endocrinology 149, 597–604 (2008) .
    1. di Clemente N. et al. Processing of anti-mullerian hormone regulates receptor activation by a mechanism distinct from TGF-beta. Mol. Endocrinol. 24, 2193–2206 (2010) .
    1. Pepinsky R. B. et al. Proteolytic processing of mullerian inhibiting substance produces a transforming growth factor-beta-like fragment. J. Biol. Chem. 263, 18961–18964 (1988) .
    1. Ciofi P. et al. Brain-endocrine interactions: a microvascular route in the mediobasal hypothalamus. Endocrinology 150, 5509–5519 (2009) .
    1. Schaeffer M. et al. Rapid sensing of circulating ghrelin by hypothalamic appetite-modifying neurons. Proc. Natl Acad. Sci. USA 110, 1512–1517 (2013) .
    1. Langlet F., Mullier A., Bouret S. G., Prevot V. & Dehouck B. Tanycyte-like cells form a blood-cerebrospinal fluid barrier in the circumventricular organs of the mouse brain. J. Comp. Neurol. 521, 3389–3405 (2013) .
    1. Herde M. K., Geist K., Campbell R. E. & Herbison A. E. Gonadotropin-releasing hormone neurons extend complex highly branched dendritic trees outside the blood-brain barrier. Endocrinology 152, 3832–3841 (2011) .
    1. Laven J. S. et al. Anti-Mullerian hormone serum concentrations in normoovulatory and anovulatory women of reproductive age. J. Clin. Endocrinol. Metab. 89, 318–323 (2004) .
    1. Catteau-Jonard S. et al. Changes in serum anti-mullerian hormone level during low-dose recombinant follicular-stimulating hormone therapy for anovulation in polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 92, 4138–4143 (2007) .
    1. Kissell K. A. et al. Biological variability in serum anti-Mullerian hormone throughout the menstrual cycle in ovulatory and sporadic anovulatory cycles in eumenorrheic women. Hum. Reprod. 29, 1764–1772 (2014) .
    1. Goodarzi M. O., Dumesic D. A., Chazenbalk G. & Azziz R. Polycystic ovary syndrome: etiology, pathogenesis and diagnosis. Nat. Rev. Endocrinol. 7, 219–231 (2011) .
    1. Sir-Petermann T. et al. Increased anti-Mullerian hormone serum concentrations in prepubertal daughters of women with polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 91, 3105–3109 (2006) .
    1. Crisosto N. et al. Anti-Mullerian hormone levels in peripubertal daughters of women with polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 92, 2739–2743 (2007) .
    1. Beauvillain J. C. & Tramu G. Immunocytochemical demonstration of LH-RH, somatostatin, and ACTH-like peptide in osmium-postfixed, resin-embedded median eminence. J. Histochem. Cytochem. 28, 1014–1017 (1980) .
    1. Vidal A., Zhang Q., Medigue C., Fabre S. & Clement F. DynPeak: an algorithm for pulse detection and frequency analysis in hormonal time series. PloS ONE 7, e39001 (2012) .

Source: PubMed

3
購読する