Hum. Reprod.-2002-Barnes-88-91.pdf

Embed Size (px)

Citation preview

  • 8/10/2019 Hum. Reprod.-2002-Barnes-88-91.pdf

    1/4

    Human Reproduction Vol.17, No.1 pp. 8891, 2002

    CASE REPORT

    The role of LH and FSH in ovarian androgen secretion andovarian follicular development: Clinical studies in a patientwith isolated FSH deficiency and multicystic ovaries

    Randall B.Barnes1,5, Anne B.Namnoum2, Robert L.Rosenfield3 and Lawrence C.Layman4

    Departments of Obstetrics and Gynecology, 1University of Chicago, Chicago, IL 60637, 2Emory University, Atlanta, GA 30322,3Department of Pediatrics and Medicine, University of Chicago, Chicago, IL 60637, 4Department of Obstetrics and Gynecology,

    Medical College of Georgia, Augusta, GA 30912, USA

    5To whom correspondence should be addressed at: Dept of Obstetrics and Gynecology, University of Chicago, 5841 Maryland

    Avenue, Chicago, IL 60637, USA. E-mail: [email protected]

    Inactivating mutations have proven to be instructive in elucidating the role of FSH in human ovarian function. We

    performed a detailed reproductive endocrine evaluation of a patient with inactivating mutations in the FSH

    -subunit gene who was hypo-estrogenic and had LH excess. The patient underwent a pelvic ultrasound andovernight frequent blood sampling followed by a human chorionic gonadotrophin (HCG) stimulation test. One

    month later she received human recombinant FSH, followed 24 h later by a second HCG stimulation test. Despite

    a mean LH serum concentration and LH pulse characteristics typical for polycystic ovaries (PCOS), baseline and

    dexamethasone-suppressed free testosterone were lownormal. The administration of HCG led to minimal stimulation

    of 17-hydroxyprogesterone and androgens. The patient had multicystic ovaries containing follicles 35 mm in

    diameter and responded to FSH with prompt increases in estradiol and inhibin B. There were no clinical or

    laboratory consequences of LH excess in this FSH-deficient woman. These findings support the hypothesis that

    excessive LH stimulation alone does not cause ovarian hyperandrogenism. We also found that follicular development

    was present in the absence of FSH. These antral follicles had apparently developed normally, since estradiol and

    inhibin B increased promptly after FSH administration.

    Key words: androgens/FSH deficiency/LH excess/ovary

    Introduction

    Inactivating mutations have proven to be instructive in elucidat-

    ing the role of FSH in human ovarian function (Layman and

    McDonough, 2000; Themmen and Huhtaniemi, 2000). Our

    studies of a patient with compound heterozygous mutations of

    the FSH -subunit gene (Layman et al., 1997), undetectableserum FSH and elevated serum LH have elucidated two aspects

    of FSH action which challenge common wisdom. We report

    here evidence that FSH is not necessary for the development

    of small, healthy antral follicles readily responsive to FSH,

    which contrasts with studies in FSH-knockout mice (Kumar

    et al., 1997; Dierich et al., 1998). However, FSH is necessary

    for ovarian theca cell function, contrary to expectations from

    the 2-gonadotrophin, 2-cell model of ovarian steroidogenesis

    (Barnes et al., 2000). The latter finding has important

    implications for theories about the role of LH excess in the

    pathogenesis of polycystic ovary syndrome (PCOS).

    88 European Society of Human Reproduction and Embryology

    Case report

    Subject

    The patient presented at 16 years of age with primary

    amenorrhoea and absent breast development. Her baseline

    and gonadotrophin-releasing hormone (GnRH)-stimulatedFSH concentrations were undetectable by immunoassay, while

    LH concentrations at baseline (30 mIU/ml) and post-GnRH

    (150 mIU/ml) were elevated. The patient was subsequently

    treated with estrogen and underwent normal breast develop-

    ment. Analysis of the FSH -subunit gene demonstrated thatshe was a compound heterozygote for two different mutations,

    a two base-pair deletion in exon three at codon 61 (Val61X)

    inherited from her mother, and a missense mutation changing

    a cysteine to a glycine at codon 51 (Cys51Gly) inherited from

    her father. When these mutations were stably transfected into

    Chinese hamster ovary cells, they demonstrated no measurable

    FSH immuno- or bioactivity (Layman et al., 1997).

  • 8/10/2019 Hum. Reprod.-2002-Barnes-88-91.pdf

    2/4

    Clinical studies in a patient with FSH deficiency

    Figure 1. Outline of clinical research center protocols.Dexamethasone, 0.5 mg four times daily, was started on day 1 andcontinued until the blood sample was drawn 24 h after HCG.

    Reproductive endocrine evaluation

    The patient underwent a detailed reproductive endocrine

    evaluation at 21 years of age in the University of Chicago

    General Clinical Research Center (GCRC), after dis-

    continuing the oral contraceptive pill for one month and giving

    informed consent. The study protocols were approved by the

    University of Chicago Institutional Review Board. Her heightwas 155 cm and her weight was 53.2 kg. There was no

    hirsutism (Ferriman and Gallway score of 4; normal 8),

    breasts and pubic hair were Tanner stage 5, and the pelvic

    examination was normal. An initial blood sample was drawn for

    immunoactive FSH, LH and free testosterone. The patient then

    received dexamethasone 0.5 mg four times daily for 4 days

    prior to and continuing throughout the endocrine evaluation

    to suppress adrenal function. Following admission to the

    GCRC, she underwent a transvaginal pelvic ultrasound fol-

    lowed by blood sampling every 10 min from 7.00 p.m. to 6.00

    a.m. Serum immunoactive FSH and LH were determined for

    each sample; inhibin A and B, bioactive FSH and free -

    subunit were determined in a pooled sample. The followingmorning, a human chorionic gonadotrophin (HCG) test was

    performed. A blood sample was drawn for baseline testoster-

    one, free testosterone and estradiol. The following steroid

    intermediates were also determined: 17-hydroxyprogesterone

    (17-Prog), androstenedione, 17-hydroxypregnenolone and

    dehydroepiandrosterone. HCG 5000 IU was then administered

    i.m. and blood was drawn 24 h later for repeat steroid

    measurements.

    The patient remained off any sex steroids for another month,

    then returned to the GCRC for a second HCG study which

    was like the first except she received 300 IU of recombinant

    human FSH s.c. in the morning, 24 h prior to the HCG test.

    A blood sample was drawn 24 h later for steroids and inhibin

    A and B, and HCG 5000 IU was administered. She then

    returned 24 h after HCG (48 h after FSH) for a final blood

    sample for steroid measurements (Figure 1).

    All samples for steroids were frozen at 20C and

    measured in the same assay using previously published methods

    (Barnes et al., 1989; Rosenfieldet al., 1994). Serum immuno-

    active LH and FSH were measured using a -subunit-specificassay (Delfia, Wallach, Finland; lower limit of sensitivity

    0.15 mIU/ml for LH and 0.2 mIU/ml for FSH). Bioactive

    FSH, inhibin A, inhibin B and free -subunit were measuredusing previously published methods (Christin-Maitre et al.,

    89

    Figure 2. Transvaginal ultrasound of the left ovary before FSHtreatment. The ovary is fusiform in shape, measuring ~61.5 cm.The stroma is scant but there are multiple follicles throughout theovary, the largest measuring 5 mm. The right ovary (not shown)was similar to the left.

    1996; Lavoieet al., 1998; Weltet al., 1999). LH pulse analysis

    was performed using the ULTRA program (Van Cauter andCopinschi, 1981).

    Results

    The ovarian ultrasound prior to FSH treatment is shown in

    Figure 2. The ovaries were multicystic containing follicles

    35 mm in diameter, but unlike typical polycystic ovaries

    (Adams et al., 1986), there was no dense stroma and the

    follicles were scattered throughout the ovary rather than being

    predominately subcapsular.

    Immunoactive LH was 46 mIU/ml and FSH was undetectable

    (0.2 mIU/ml) in the single sample drawn before dexametha-

    sone. There was no immunoactive FSH detected in the samplescollected overnight (Table I), and no bioactive FSH was

    detected in the pooled sample. The mean immunoactive

    LH in the overnight samples was 46 mIU/ml; mean pulse

    amplitude was 15.8 mIU/ml and nine pulses were detected in

    11 h (Figure 3). This LH profile is similar to that reported for

    PCOS (Waldstreicher et al., 1988). Free -subunit measuredin the pooled 11 h sample was in the postmenopausal range

    (3501740 pg/ml; Table I).

    Although LH concentrations were elevated, the patient had

    no laboratory evidence of ovarian hyperandrogenism. The

    patients baseline free testosterone was 6 pg/ml (normal

    range 310 pg/ml; ovarian hyperandrogenism 10 pg/ml)

    (Rosenfield et al., 1994). After 4 days of dexamethasone

    suppression the free testosterone fell to 2 pg/ml (normal range

    8 pg/ml; ovarian hyperandrogenism 8 pg/ml; Rosenfield

    et al., 1994). The 17-Prog concentration 24 h after HCG was

    40 ng/dl which was 2.7 standard deviations (SD) below the

    mean in women with ovarian hyperandrogenism (288 ng/dl)

    (Levrantet al., 1997) and 1.5 SD below the mean of normal

    follicular phase women (139 ng/dl: Levrant et al., 1997).

    One month later, during the second GCRC study,

    estradiol increased substantially 24 h after FSH administration

    (2880 pg/mL). Inhibin B was near the lower limit of the

    normal range at baseline and rose to above the range for

  • 8/10/2019 Hum. Reprod.-2002-Barnes-88-91.pdf

    3/4

    R.B.Barnes et al.

    Table I. Gonadotrophin and inhibin concentrations in an FSH-deficient patient

    FAS LH FSH Bioactive FSH Inhibin A Inhibin B(pg/ml) (mIU/ml) (mIU/ml) (mIU/ml) (pg/ml) (pg/ml)

    Baselinea 1247 46 0.2 4 0.6 25.324 h after FSH 8.4 0.8 283Normal women 103257 015 320 440 0.680 15220

    aBaseline is mean of every 10 min samples over 11 h for LH and FSH. For all others, baseline is a value ofthe pooled 11 h sample. Normal values are from Christein-Maitre et al., 1996; Lavoie et al., 1998; Weltet al., 1999.FAS free alpha subunit.

    Figure 3. LH pulse analysis. LH was measured at 10 min intervalsover 11 h. Significant pulses are indicated by an asterisk.

    menstruating women 24 h after FSH injection (Table I). In

    contrast, inhibin A was present at low concentrations.

    The response of testosterone and the steroid intermediates

    to the two HCG tests have been previously reported (Barnes

    et al., 2000). In brief, all steroid concentrations 24 h after the

    second HCG injection (48 h after FSH) were greater than at

    24 h after the first HCG injection (HCG alone). The difference

    between the two HCG tests was most dramatic for testosterone,

    which was unaffected by the first HCG test (12 ng/dl before

    and after HCG), but increased from 16 to 41 ng/dl after the

    second, FSH-primed HCG test.

    Discussion

    This patient, with well characterized inactivating mutations in

    the FSH -subunit gene, was hypo-estrogenic with secondaryLH excess. Two features were noteworthy: antral follicular

    development was present in spite of the lack of FSH, and

    there were no clinical or laboratory consequences of the

    LH excess.

    The point at which FSH is necessary for follicular develop-

    ment in the human ovary is debated (Gougeon, 1996; McGee

    and Hsueh, 2000). Despite having no detectable FSH, our

    patient had multicystic ovaries with follicles up to 5 mm in

    diameter. Similar sized follicles have been reported in some

    women with FSH receptor mutations (Aittomaki et al., 1996;

    Beau et al., 1998); however, those FSH receptor mutations

    90

    may not be completely inactivating. These findings are in

    contrast to FSH- and FSH receptor-knockout mice, in whichantral follicles are not maintained (Kumar et al., 1997; Dierich

    et al., 1998). In studies of human ovarian xenografts trans-

    planted into the kidney capsule of immunodeficient and hypo-

    gonadotrophic mice, FSH was required for the growth of

    follicles beyond the two-layer granulosa cell stage (Oktay

    et al., 1998), which is about the point at which the FSHreceptor gene isfirst expressed in human follicles (Oktay et al.,

    1997). Although the xenograft data are in contrast to our

    findings, it is likely that the endocrine and growth factor milieu

    of the in-situ human ovary is very different than that in the

    immunodeficient, hypogonadotrophic mouse.

    The prompt estradiol and inhibin B responses 24 h after

    exogenous FSH suggest that our patients antral follicles

    contained granulosa cells that had developed normally in the

    absence of FSH. Our findings are similar to those in two other

    patients with isolated FSH deficiency who ovulated and had a

    successful pregnancy after ~14 days of menotrophin therapy

    (Rabinowitz et al., 1979; Matthews et al., 1993). Ovulation

    after a short exposure to menotrophins implies that somehealthy follicles had reached the point of recruitability without

    FSH exposure, since ~14 days are required for an ovulatory

    follicle to develop from follicles a few millimeters in diameter,

    in contrast to the 3 months required to develop from the pre-

    antral stage (Gougeon, 1996). These earlier reports and our

    findings suggest that in the complete absence of FSH stimula-

    tion, human ovarian antral follicles can develop up to 5 mm

    in diameter.

    Our patient had no clinical or laboratory evidence of ovarian

    hyperandrogenism despite a mean LH concentration, LH pulse

    characteristics and ovarian follicular sizes typical for PCOS.

    On ultrasound her multicystic ovaries lacked the excess stroma

    of classic polycystic ovaries. Indeed, her ovaries produced

    little, if any, androgen. Her baseline and dexamethasone-

    suppressed free testosterone were lownormal. The administra-

    tion of HCG led to minimal stimulation of 17-Prog or other

    thecal cell steroids. However, we have previously shown that

    exogenously administered FSH augmented her LH- and HCG-

    stimulated production of testosterone and all steroids of thecal

    cell origin (Barneset al., 2000). This suggests that FSH action,

    probably via granulosa cell-produced paracrine intermediates,

    is necessary for thecal cells to respond to LH. One of many

    such paracrine factors is inhibin B, which increased markedly

    with FSH administration in this patient (Barnes 1998).

  • 8/10/2019 Hum. Reprod.-2002-Barnes-88-91.pdf

    4/4

    Clinical studies in a patient with FSH deficiency

    These findings are relevant to the role of LH excess in the

    pathogenesis of the ovarian hyperandrogenism of PCOS. It

    has been reported that 75% of women with clinical evidence

    of PCOS have an elevated LH concentration and 94% have

    an increased LH/FSH ratio (Taylor et al., 1997). These

    gonadotrophin secretory abnormalities have been thought to

    play an important role in the development of the ovarian

    hyperandrogenism characteristic of PCOS (Hall, 1993). In arelated study, we found that a woman with a constitutively

    activating mutation of the LH receptor, identified because she

    was the mother of two sons with gonadotrophin-independent

    precocious puberty, had no clinical or laboratory evidence of

    ovarian hyperandrogenism (Rosenthal et al., 1996). Thus,

    increased LH stimulation, even in the presence of FSH, appears

    to be insufficient to induce the hyperandrogenism and stromal

    hyperplasia of PCOS. The findings in the previous, as well as

    the current, case report support the hypothesis that thecal cell

    androgen secretion in response to excessive LH stimulation is

    strictly limited by intra-ovarian factors. Ovarian hyperandro-

    genism is more likely a result of escape from down-regulation

    by these intra-ovarian factors than a result of elevated LHconcentrations (Ehrmann et al., 1995). Taken together, our

    studies support the hypotheses that normal ovarian androgen

    production depends on both FSH and LH and that excessive

    ovarian androgen production is a result of abnormal intra-

    ovarian regulation, and not of excessive LH stimulation.

    Acknowledgements

    We are greatly indebted to Patrick Sluss,Reproductive Endocrine Unit,The Massachusetts General Hospital, Boston, MA, who performedthe FSH bioassay, FAS, inhibin A, and inhibin B assays; to ZubieSheikh, Department of Obstetrics and Gynecology, University ofChicago, for performing the transvaginal ultrasound; and to Jennifer

    M.Cunningham, Department of Medicine, University of Chicago, forperforming the LH pulse analysis. We also appreciate the helpfuldiscussions of J.Larry Jameson from the Center for Endocrinology,Metabolism, and Molecular Medicine, Northwestern University,Chicago, IL and William F.Crowly Jr from the ReproductiveEndocrine Unit and the National Center for Infertility Research, TheMassachusetts General Hospital, Boston, MA.

    L.C.L. was supported by NIH grant support PHS NICHD HD33004;R.L.R. was supported by NIH grant support RR-00055 (CRC).

    References

    Adams, J., Polson, D.W. and Franks, S. (1986) Prevalence of polycysticovaries in women with anovulation and idiopathic hirsutism. Br. Med. J.,293

    , 355359.Aittomaki, K., Herva, R., Stenman, U-H. et al. (1996) Clinical features ofprimary ovarian failure caused by a point mutation in the follicle-stimulating hormone receptor gene. J. Clin. Endocrinol. Metab., 81,37223726.

    Barnes, R.B. (1998) The pathogenesis of polycystic ovary syndrome: lessonsfrom ovarian stimulation studies. J. Endocrinol. Invest., 21 , 567579.

    Barnes, R.B., Rosenfield, R.L., Burstein, S. et al. (1989) Pituitaryovarianresponses to nafarelin testing in the polycystic ovary syndrome. N. Engl.

    J. Med., 320 , 559565.

    Barnes, R.B., Namnoum, A., Rosenfield, R.L. et al. (2000) Effect of follicle-stimulating hormone on ovarian androgen production in a woman withisolated follicle-stimulating hormone deficiency. N. Engl. J. Med., 343,11971198.

    91

    Beau, I., Touraine, P., Meduri, G. et al. (1998) Novel phenotype related topartial loss of function mutations of the follicle stimulating hormonereceptor. J. Clin. Invest., 102 , 13521359.

    Christin-Maitre, S., Taylor, A.E., Khoury, R.H. et al. (1996) Homologousin vitro bioassay for follicle-stimulating hormone (FSH) reveals increasedFSH biological signal during the mid- to late luteal phase of the humanmenstrual cycle. J. Clin. Endocrinol. Metab., 81, 20802088.

    Dierich, A., Sairam, M.R., Monaco, L. et al. (1998) Impairing follicle-stimulating hormone (FSH) signaling in vivo: targeted disruption of theFSH receptor leads to aberrant gametogenesis and hormonal imbalance.

    Proc. Natl Acad. Sci. USA, 95, 1361213617.Ehrmann, D.A., Barnes, R.B. and Rosenfield, R.L. (1995) Polycystic ovary

    syndrome as a form of functional ovarian hyperandrogenism due todysregulation of androgen secretion. Endocr. Rev., 16 , 322353.

    Gougeon, A. (1996) Regulation of ovarian follicular development in primates:facts and hypotheses. Endocr. Rev., 17, 121155.

    Hall, J.E. (1993) Polycystic ovarian disease as a neuroendocrine disorder ofthe female reproductive axis.Endocrinol. Metab. Clin. North Am., 22, 7592.

    Kumar, T.R., Wang, Y., Lu., N. et al. (1997) Follicle-stimulating hormone isrequired for ovarian follicle maturation but not male fertility.Nature Genet.,15, 201204.

    Lavoie, H.B., Martin, K.A., Taylor, E., et al. (1998) Exaggerated free alpha-subunit levels during pulsatile gonadotropin-releasing hormone replacementin women with idiopathic hypogonadotropic hypogonadism. J. Clin.

    Endocrinol. Metab., 83 , 241247.

    Layman, L.C. and McDonough, P.G. (2000) Mutations of follicle stimulating

    hormone-beta and its receptor in human and mouse: genotype/phenotype.Mol. Cell. Endocrinol., 161 , 917.

    Layman, L.C., Lee, E.J., Peak, D.B. et al. (1997) Delayed puberty andhypogonadism caused by mutations in the follicle-stimulating hormone -subunit gene. N. Engl. J. Med., 337 , 607611.

    Levrant, S.G., Barnes, R.B. and Rosenfield, R.L. (1997) A pilot study of thehuman chorionic gonadotrophin test for ovarian hyperandrogenism. Hum.

    Reprod., 12, 14161420.

    Matthews, C.H., Borgato, S., Beck-Peccoz, P. et al. (1993) Primaryamenorrhoea and infertility due to a mutation in the -subunit of follicle-stimulating hormone. Nature Genet., 5 , 8386.

    McGee, E.A. and Hsueh, A.J.W. (2000) Initial and cyclic recruitment ofovarian follicles. Endocr. Rev., 21 , 200214.

    Oktay, O., Briggs D. and Gosden, R.G. (1997) Ontogeny of follicle-stimulatinghormone receptor gene expression in isolated human ovarian follicles. J.Clin. Endocrinol. Metab., 82 , 37483751.

    Oktay, O., Newton H., Mullan J. et al. (1998) Development of humanprimordial follicles to antral stages in SCID/hpg mice stimulated withfollicle-stimulating hormone.Hum. Reprod., 13 , 11331138.

    Rabinowitz, D., Benveniste, R., Lindner, J. et al. (1979) Isolated follicle-stimulating hormone deficiency revisted. N. Engl. J. Med., 300 , 126128.

    Rosenfield, R.L., Barnes, R.B. and Ehrmann, D.A. (1994) Studies of thenature of 17-hydroxyprogesterone hyperresponsiveness to gonadotropin-releasing hormone agonist challenge in functional ovarianhyperandrogenism.J. Clin. Endocrinol. Metab., 79, 16861692.

    Rosenthal, I.M., Refetoff, S., Rich, B. et al.(1996) Response to acute challengewith gonadotropin releasing hormone agonist in two half-brothers and theirmother with a constitutively activating mutation of the luteinizing hormonereceptor. J. Clin. Endocrinol. Metab., 81 , 38023806.

    Taylor, A.E., McCourt, B., Martin, K.A. et al. (1997) Determinants ofabnormal gonadotropinsecretion in clinically defined women with polycysticovary syndrome. J. Clin. Endocrinol. Metab., 82, 22482256.

    Themmen, A.P.N. and Huhtaniemi, I.T. (2000) Mutations of gonadotropins

    and gonadotropin receptors: elucidating the physiology and pathophysiologyof pituitarygonadal function. Endocr. Rev., 21, 551583.

    Van Cauter, E. and Copinschi, G. (eds) (1981) Human pituitaryhormones: circadian and episodic variations. Martinus Nijhoff, The Hague,pp. 221235.

    Waldstreicher, J., Santoro, N.F., Hall, J.E., et al. (1988) Hyperfunction of thehypothalamicpituitary axis in women with polycystic ovarian disease:indirect evidence for partial gonadotroph desensitization.J. Clin. Endocrinol.

    Metab., 66 , 165172.

    Welt, C.K., Adams, J.M., Sluss, P.M. et al. (1999) Inhibin A and inhibinB responses to gonadotropin withdrawal depends on stage of follicledevelopment.J. Clin. Endocrinol. Metab., 84 , 21632169.

    Received on February 28, 2001; accepted on August 21, 2001