Veterinary Pathology Online

the postmenopausal ovary, which can metabolize cholesterol into oxysterols but have limited capac- ity for the synthesis of steroid hormones, particu- larly sex ...
2MB taille 4 téléchargements 310 vues
Veterinary Pathology Online http://vet.sagepub.com/

Gonadectomy-induced Adrenocortical Neoplasia in the Domestic Ferret (Mustela putorius furo) and Laboratory Mouse M. Bielinska, S. Kiiveri, H. Parviainen, S. Mannisto, M. Heikinheimo and D. B. Wilson Vet Pathol 2006 43: 97 DOI: 10.1354/vp.43-2-97 The online version of this article can be found at: http://vet.sagepub.com/content/43/2/97

Published by: http://www.sagepublications.com

On behalf of:

American College of Veterinary Pathologists

Additional services and information for Veterinary Pathology Online can be found at: Email Alerts: http://vet.sagepub.com/cgi/alerts Subscriptions: http://vet.sagepub.com/subscriptions Reprints: http://www.sagepub.com/journalsReprints.nav Permissions: http://www.sagepub.com/journalsPermissions.nav

Downloaded from vet.sagepub.com by guest on July 29, 2010

Vet Pathol 43:97–117 (2006)

REVIEW ARTICLE Gonadectomy-induced Adrenocortical Neoplasia in the Domestic Ferret (Mustela putorius furo) and Laboratory Mouse M. BIELINSKA, S. KIIVERI, H. PARVIAINEN, S. MANNISTO, M. HEIKINHEIMO,

AND

D. B. WILSON

Departments of Pediatrics and Molecular Biology & Pharmacology, Washington University School of Medicine, St. Louis Children’s Hospital, St. Louis, MO (MB, MH, DBW); Children’s Hospital, Program for Developmental and Reproductive Biology, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland (SK, HP, SM, MH); University and University Central Hospital of Tampere, Tampere, Finland (MH) Abstract. Sex steroid–producing adrenocortical adenomas and carcinomas occur frequently in neutered ferrets, but the molecular events underlying tumor development are not well understood. Prepubertal gonadectomy elicits similar tumors in certain inbred or genetically engineered strains of mice, and these mouse models shed light on tumorigenesis in ferrets. In mice and ferrets, the neoplastic adrenocortical cells, which functionally resemble gonadal steroidogenic cells, arise from progenitors in the subcapsular or juxtamedullary region. Tumorigenesis in mice is influenced by the inherent susceptibility of adrenal tissue to gonadectomy-induced hormonal changes. The chronic elevation in circulating luteinizing hormone that follows ovariectomy or orchiectomy is a prerequisite for neoplastic transformation. Gonadectomy alters the plasma or local concentrations of steroid hormones and other factors that affect adrenocortical tumor development, including inhibins, activins, and Mu¨llerian inhibiting substance. GATA-4 immunoreactivity is a hallmark of neoplastic transformation, and this transcription factor might serve to integrate intracellular signals evoked by different hormones. Synergistic interactions among GATA-4, steroidogenic factor-1, and other transcription factors enhance expression of inhibin-a and genes critical for ectopic sex steroid production, such as cytochrome P450 17a-hydroxylase/17,20 lyase and aromatase. Cases of human adrenocortical neoplasia have been linked to precocious expression of hormone receptors and to mutations that alter the activity of G-proteins or downstream effectors. Whether such genetic changes contribute to tissue susceptibility to neoplasia in neutered ferrets and mice awaits further study. Key words: Adrenal tumor; ferrets; luteinizing hormone; mice; orchiectomy; ovariectomy; steroidogenesis.

more than 3 years,31,60,110 hinders prospective studies of tumor development in this species. The phenomenon of gonadectomy-induced adrenocortical neoplasia also has been observed in mice, rats, guinea pigs, and hamsters.104 Of these, the laboratory mouse is the most useful experimental model because of its well-characterized genetics. In certain inbred21 or genetically engineered35,72,89 strains of mice, adrenocortical neoplasms develop within weeks to months of gonadectomy with near complete penetrance. Although the similarities in adrenocortical neoplasia between inbred mice and ferrets are mentioned in veterinary medicine textbooks,23,98 no concerted effort has been made to apply knowledge gleaned from studies of tumorigenesis in mice to ferrets. This comparative review highlights the histologic changes that accompany gonadectomy-induced

Introduction Sex steroid–producing adrenocortical adenomas and carcinomas occur frequently in neutered ferrets and cause significant morbidity.60,97 It has been proposed that competent cells in the ferret adrenal cortex undergo neoplastic transformation and adopt features of gonadal steroidogenic cells in response to the hormonal changes that follow gonadectomy.23 Chronically elevated serum luteinizing hormone (LH) levels have been implicated in the pathobiology of these neoplasms,111,125 but the molecular events underlying tumorigenesis remain poorly characterized. Although the ferret is a valuable experimental animal for studies of reproductive endocrinology,4 it is not standardized with regard to genotype and therefore not ideal for studies of tumorigenesis. Moreover, the latency of adrenocortical neoplasia in neutered ferrets, which averages 97

Downloaded from vet.sagepub.com by guest on July 29, 2010

98

Bielinska, Kiiveri, Parviainen, Mannisto, Heikinheimo, and Wilson

Vet Pathol 43:2, 2006

Fig. 1. Steroid hormone biosynthesis in normal (solid lines) versus neoplastic (dashed lines) adrenocortical cells of the mouse and ferret. The repertoire of enzymes distal to the common pathway determines the steroidogenic capacity of a given cell.

adrenocortical neoplasia in both species and presents updated information on the molecular mechanisms of tumorigenesis deduced from the mouse models. The relevance to human adrenocortical tumors is also discussed. Overview of the Structure, Function, and Development of the Adrenal Cortex The adrenal cortex is a major source of steroid hormones that are synthesized from cholesterol through the sequential activities of a series of cytochrome P450 enzymes (Fig. 1). In most mammals, the adrenal cortex is divided into three main layers: the zona glomerulosa, zona fasciculata, and zona reticularis (Table 1). Mineralocorticoids are produced in the outermost layer, the zona glomerulosa, whereas glucocorticoids and androgen precursors are synthesized in the zona fasciculata and zona reticularis. Two less prominent layers, the zona intermedia and zona juxtamedullaris, have also been described in the ferret and other carnivores, although the functional significance of these layers is unclear.33 Cortisol is the principal glucocorticoid secreted by the adrenal cortex of the ferret.23 Under physiologic conditions, the ferret adrenal gland produces only limited amounts of the androgenic steroids dehydroepian-

drosterone (DHEA), DHEA sulfate (DHEA-S), or androstenedione.14,99 In the mouse adrenal, the zona glomerulosa and zona fasciculata are well defined, but there is no discernable zona reticularis. The adrenal cortex of the young mouse contains an additional layer, the X-zone, which is adjacent to the medulla and is analogous to the fetal zone of the human adrenal cortex.45 The function of the Xzone, which regresses at puberty in males and during the first pregnancy in females, is unknown. No correlate of the mouse X-zone or human fetal zone has been described in the ferret. Adrenocortical cells in the adult mouse lack cytochrome P450 17a-hydroxylase/C17–C20 lyase (P450c17), a dual-function enzyme required for synthesis of cortisol and sex steroids.46 Consequently, corticosterone is the principal glucocorticoid secreted by the mouse adrenal cortex, and under normal conditions, androgenic steroids are not produced in this tissue. During embryonic development, steroidogenic cells in the adrenal cortex and gonads arise from progenitors in the urogenital ridge (Fig. 2).45 Differentiation of the so-called adrenogonadal primordium is thought to be influenced by growth factors secreted by the underlying mesonephros.76 Progenitors of adrenocortical cells migrate medial-

Downloaded from vet.sagepub.com by guest on July 29, 2010

99

Gonadectomy-induced Adrenocortical Neoplasia

Vet Pathol 43:2, 2006

Table 1.

Comparative anatomy and physiology of the adrenal cortex. Mouse

Morphologically distinct zones

Zona glomerulosa Zona fasciculata X-zone

P450c17 17a-hydroxylase{ Major glucocorticoid produced P450c17 17,20 lyase{ Production of androgenic steroids

Ferret

Human

2 Corticosterone

Zona glomerulosa Zona intermedia Zona fasciculata Zona reticularis Zona juxtamedullaris* ++ Cortisol

Zona glomerulosa Zona fasciculata Zona reticularis Fetal zone ++ Cortisol

2 2

+/2 Minimal

++ ++

* It is unclear whether there is a transient zone analogous to the rodent X-zone or human fetal zone in the developing ferret. { The 17a-hydroxylase and 17,20 lyase activities of P450c17 might be differentially regulated in a given cell type.3 In the absence of 17a-hydroxylase activity, the major glucocorticoid produced is corticosterone rather than cortisol. 17,20 Lyase activity is required for synthesis of androgenic steroids. P450c17 activities in the ferret adrenal are inferred from measurements of steroid hormone levels in the circulation of healthy, gonadectomized animals.14

Fig. 2. Development of the adrenal gland and gonads from the urogenital ridge. Key regulatory genes at the various developmental steps are shown in boxes. Fig. 3. Gonadal ridge derivatives; DBA/2J mouse. Embryonic day 13.5. Arrowhead points to epithelium lining the medial coelomic bay. Dorsal aorta (ao), cardinal vein (cv), gonad (g), mesonephric tubules (mt), sympathoblasts (s). Circles indicate the nascent adrenals. GATA-4, avidin–biotin complex peroxidase method. Bar 5 100 mm.

Downloaded from vet.sagepub.com by guest on July 29, 2010

100

Bielinska, Kiiveri, Parviainen, Mannisto, Heikinheimo, and Wilson

Vet Pathol 43:2, 2006

Fig. 4. Hormone secretion and regulatory pathways in the hypothalamo–pituitary–adrenal/gonadal axes of nongonadectomized and gonadectomized animals. R and —| indicate positive and negative regulation, respectively. Following gonadectomy, feedback inhibition of the hypothalamus by sex steroids is reduced, as indicated by the dashed line.

ly and rostrally, eventually associating with neural crest derivatives that will give rise to the adrenal medulla; cells destined to become gonadal stroma move laterally and associate with primordial germ cells (Fig. 3). Mutations in certain genes expressed in the urogenital ridge or adrenogonadal primordium, such as Wilms’tumor1 (Wt-1), Wnt4, and steroidogenic factor-1 (Sf-1) disrupt development of both gonadal and adrenal steroidogenic cells, underscoring the close relationship between these two lineages.45,76,80 The further differentiation, growth, and survival of steroidogenic cells in the adrenal gland are controlled by a diverse array of hormones, including adrenocorticotropic hormone (ACTH; Fig. 4), angiotensin-II, endothelin-1, vasopressin, and insulin-related growth factors.27,102,127 In certain instances, endocrine and paracrine factors traditionally associated with the function of gonadal steroidogenic cells, such as LH (Fig. 4), inhibins, and activins, can also influence the differentiation, proliferation, and function of adrenocortical progenitor cells.6 These reproductive factors are of particular relevance to gonadectomy-induced tumorigenesis.

Gonadectomy-associated Adrenocortical Neoplasia in the Ferret In ferrets, adrenocortical neoplasia affects both genders but is more easily recognized in jills.60,97,129 The prevalence of this condition has been attributed to early gonadectomy.110 Adrenocortical tumors are common in ferrets living in the United States, where most commercially raised ferrets are castrated or ovariohysterectomized before 6 weeks of age. In countries in which preadolescent neutering of domestic ferrets is not routine, such as the United Kingdom, the incidence of adrenocortical neoplasia is markedly lower.110 Other factors hypothesized to predispose ferrets to neoplasia include inbreeding at commercial facilities, unnatural photoperiodic stimulation, and diet.23,98 The neoplastic cells that accumulate in the ferret adrenal after prepubertal gonadectomy functionally resemble gonadal steroidogenic cells. The ectopic production of sex steroids by these tumor cells causes a syndrome known as adrenal-associated endocrinopathy (AAE) or hyperadrenocorticism. Clinical signs of this syndrome attributable to sex steroids include bilateral symmetric alopecia, enlargement of the vulva (in jills), squamous meta-

Downloaded from vet.sagepub.com by guest on July 29, 2010

Vet Pathol 43:2, 2006

Gonadectomy-induced Adrenocortical Neoplasia

plasia of prostatic ductular epithelium (in hobs), and resumption of mating behavior (in hobs).23 The diagnosis of AAE is confirmed by documenting elevated plasma concentrations of 17a-hydroxyprogesterone, androstenedione, DHEA-S, or estradiol.23,99 Ferrets with AAE can also exhibit Cushingoid features, such as thin skin, muscular atrophy, and pot-bellied appearance,23 raising the possibility that excess glucocorticoid production contributes to the syndrome. Plasma concentrations of cortisol are infrequently elevated in ferrets with AAE,23,99 but in some cases, the urinary corticoid : creatinine ratio is elevated and resistant to suppression by dexamethasone,113 consistent with ACTH-independent cortisol production by the neoplastic adrenal gland. In approximately 85% of ferrets with AAE, only one adrenal gland is enlarged.100,128 Histologic examination might reveal nodular hyperplasia, adenoma, or carcinoma of the adrenal cortex. Adenomas are well-demarcated lesions composed mainly of polyhedral cells with small, hyperchromatic nuclei (Fig. 5).101 Adrenocortical carcinomas are generally large, proliferative lesions that invade beyond the adrenal capsule.101 Carcinomas often contain mixtures of small basophilic ovoid cells, large polyhedral cells, and large clear cells with small dense nuclei and vacuolated cytoplasm (Fig. 6). Anaplastic variants of adrenocortical carcinoma exhibit myxoid differentiation (Fig. 7).85 A separate spindle cell component might be seen in benign or malignant adrenocortical tumors in ferrets (Fig. 8), although its prognostic importance is unclear.26,81 Unlike adjacent normal adrenocortical tissue, gonadectomyassociated tumors in ferrets express markers characteristic of gonadal steroidogenic cells. Among these markers are inhibin-a and two nuclear factors that regulate gene expression in gonadal somatic cells, transcription factor GATA4 (Fig. 9) and estrogen receptor-a (ERa; Fig. 10).81,84,85 The pathophysiologic significance of these markers is discussed below. Mouse Models of Gonadectomy-induced Adrenocortical Neoplasia Inbred mouse strain

Prepubertal gonadectomy induces sex steroid– producing adrenocortical tumors in certain inbred mouse strains: CE mice develop carcinomas, whereas strains C3H and DBA/2J develop adenomas.9,21 These adrenal neoplasms arise in the subcapsular region and invade the deeper layers of the cortex, displacing normal adrenocortical tissue (Figs. 11a, b, 12a, b). The neoplasms are

101

composed of two populations of cells: small, basophilic cells, known as A cells, and large, lipid-laden cells, termed B cells.9,21 Type A cells express two markers of gonadal stroma, GATA-4 and the receptor for Mu¨llerian inhibiting substance (MIS), but lack expression of steroidogenic markers.8,9 The related transcription factor, GATA-6, which is found in steroidogenic cells of the normal adrenal cortex, is downregulated in the neoplastic A cells.8,9 Type B cells express markers of gonadal steroidogenic cells, including GATA-4, SF-1, LH receptor (LHR), inhibin-a, MIS, ERa, P450c17, and an ‘‘ovarian-specific’’ exon 1 variant of aromatase.8,9 Type B cells produce androgens, estrogens, or both. Although the terms ‘‘A cell’’ and ‘‘B cell’’ are vague and lackluster, they have been in use for 60 years and have not been supplanted by more precise or descriptive wording. Functionally, type A cells resemble stromal cells of the postmenopausal ovary, which can metabolize cholesterol into oxysterols but have limited capacity for the synthesis of steroid hormones, particularly sex steroids.39 Type B cells resemble follicular theca cells from women with polycystic ovary syndrome, a condition characterized by chronically elevated LH levels.43 Traditional adrenocortical cell markers, such as cell surface hormone receptors (e.g., the ACTH receptor [melanocortin-2 receptor, MC2R]) or enzymes dedicated to the synthesis of corticosterone or aldosterone (e.g., steroid 21hydroxylase [P450c21], P450 11b-hydroxylase [P450c11, Fig. 11b, 12b], aldosterone synthase [P450aldo]), become excluded from the neoplastic tissue.8,9,130 Inhibin-a promoter–SV40 T-antigen transgenic mice

Intact inhibin-a promoter–SV40 T-antigen transgenic mice develop gonadal sex cord stromal tumors.44,90 When these mice are subjected to prepubertal gonadectomy, they develop sex steroid–producing adrenocortical carcinomas by 5– 7 months of age. These adrenocortical carcinomas generally arise in the X-zone, but subcapsular patches of type A and B cells can accompany the X-zone lesions (Fig. 13a, b).44,90 The molecular markers of adrenocortical neoplasia in these mice, LHR, inhibin-a, GATA-4 (Fig. 13c), and P450c17, are similar to those described in gonadectomized DBA2/J mice.9 The adrenocortical tumors in inhibin-a promoter–SV40 T-antigen transgenic mice secrete large amounts of estrogen.79,91 GATA-6, a factor associated with growth inhibition in some cell types,77 is downregulated during adrenocortical tumorigenesis in these transgenic mice (Fig. 13d).52

Downloaded from vet.sagepub.com by guest on July 29, 2010

102

Bielinska, Kiiveri, Parviainen, Mannisto, Heikinheimo, and Wilson

Vet Pathol 43:2, 2006

Fig. 5. Adrenal gland, adenoma; neutered ferret. Note the polyhedral cells with hyperchromatic nuclei. HE. Bar 5 100 mm. Fig. 6. Adrenal gland, adrenocortical carcinoma; neutered ferret. Note the mixture of small basophilic ovoid cells, large polyhedral cells, and clear cells with small dense nuclei and vacuolated cytoplasm. HE. Bar 5 100 mm. Fig. 7. Liver, metastatic myxoid adrenocortical carcinoma; neutered ferret. The asterisk indicates a mucin pool. HE. Bar 5 100 mm.

Downloaded from vet.sagepub.com by guest on July 29, 2010

Vet Pathol 43:2, 2006

Gonadectomy-induced Adrenocortical Neoplasia

Inhibin-a null mice

103

Salient features of adrenocortical tumor development in ferrets and each of the mouse models are compared in Table 2. The similarities are striking; in each case, gonadectomy leads to an increase in circulating LH, ectopic expression of LHR, and differentiation toward a gonadal phenotype. In ferrets, ectopic sex steroid production contributes to alopecia and other characteristic signs of AAE. In mice, the side effects of estrogen or testosterone production by the adrenal are more subtle but are present in reproductive tissues (e.g., vaginal mucosa, uterine epithelium, seminal vesicles) and nonreproductive tissues that exhibit sexual dimorphism (e.g., Bowman’s capsule and submaxillary glands).21 Thus, gonadectomy-induced adrenocortical tumorigenesis appears to follow similar, if not identical, molecular pathways in ferrets and mice.

mammals,64 and in gonadectomized ferrets sex steroid–producing tumors histologically indistinguishable from adrenocortical tumors have been observed at another extragonadal site, the ovarian pedicle.83 On the other hand, the cell of origin could be a multipotential progenitor capable of differentiation into either adrenocortical or gonadal-like steroidogenic cells, depending on the hormonal milieu and other environmental factors.83 Another theoretical, albeit less likely, possibility is that neoplastic progenitors are of extra-adrenal origin and invade the adrenal capsule by direct contact or via the blood stream.16 An additional source of controversy is the relationship of subcapsular A and B cells in mouse adrenocortical neoplasms (or their histologic counterparts in ferret tumors—the small basophilic cell and large clear cell). Early investigators speculated that B cells might be descendants of A cells, but direct evidence of this is lacking.21 It is equally plausible that A and B cells arise from separate progenitors. Their temporal appearance raises the possibility that type A cells produce factors that promote B cell differentiation. Stromal cells of the postmenopausal ovary synthesize growth factor– binding proteins.39 Assuming that A cells are the functional equivalent of postmenopausal stromal cells,39 these neoplastic cells might serve to insulate sex steroid–producing B cells from the effects of growth factors that promote differentiation into corticosteroid-producing cells. Conversely, B cells might affect the proliferation or survival of A cells through secretion of paracrine factors (see below). Lineage-tracing studies and tests of clonality are needed to definitively establish the origin and interrelationship of A and B cells.

Controversies Regarding the Origin and Interrelatedness of Neoplastic Adrenocortical Cells

Signaling Molecules and Pathways Implicated in Gonadectomy-induced Neoplasia

The cell of origin for gonadectomy-induced adrenocortical neoplasms in ferrets and mice remains elusive. It might be a committed gonadal progenitor that is ectopically located in the adrenal. Rare cells with morphologic features of Leydig cells have been reported in the adrenal cortex of other

Given the similarities between neoplastic adrenocortical cells and normal gonadal somatic cells, it comes as no surprise that reproductive hormones along with their receptors and intracellular effectors play prominent roles in adrenocortical tumorigenesis in ferrets and mice (Table 3). The binding

Mice lacking inhibin-a develop gonadal stromal cell tumors that secrete large amounts of activin, which cause a cancer cachexia-like wasting syndrome and death by 18 weeks of age.72,74 Gonadectomized inhibin-a null mice develop sex steroid– producing tumors of the adrenal cortex with a latency of 5–7 months.72 As in the foregoing model, the adrenocortical tumors appear to arise from the X-zone, but subcapsular cells with histologic features of type A and B cells have been observed. Tumor development is accompanied by upregulated expression of LHR, P450c17, and aromatase, and the neoplastic cells secrete both androgen and estrogen.5,11 Adrenocortical Tumorigenesis is Similar in Ferrets and Mice

r Fig. 8. Adrenal gland, anaplastic adrenocortical carcinoma; neutered ferret. Note the spindle cell component (arrows). HE. Bar 5 100 mm. Fig. 9. Adrenal gland, adrenocortical carcinoma; neutered ferret. Nuclear GATA-4 expression is evident in both large and small tumor cells. GATA-4, avidin–biotin complex peroxidase method. Bar 5 100 mm. Fig. 10. Adrenal gland, adrenocortical carcinoma; neutered ferret. Nuclear ERa expression is evident in the neoplastic cells. ERa, avidin–biotin complex peroxidase method. Bar 5 250 mm.

Downloaded from vet.sagepub.com by guest on July 29, 2010

104

Bielinska, Kiiveri, Parviainen, Mannisto, Heikinheimo, and Wilson

Downloaded from vet.sagepub.com by guest on July 29, 2010

Vet Pathol 43:2, 2006

Vet Pathol 43:2, 2006

Gonadectomy-induced Adrenocortical Neoplasia

of hormones to cell surface or nuclear receptors triggers signaling cascades that affect the proliferation, differentiation, and survival of steroidogenic cells. Often these signaling pathways culminate in the binding of transcription factors to cisregulatory elements in genes critical for cell growth or function. Hormonal signaling can also modulate the activity of cell cycle regulatory proteins. Pituitary hormones

Luteinizing hormone. This dimeric glycoprotein hormone is composed of a common gonadotropin a-subunit and hormone-specific b-subunit. LH is secreted from the pituitary in a pulsatile fashion in response to a surge of gonadotropin releasing hormone (GnRH; Fig. 4). LH binds to a Gprotein–coupled surface receptor, LHR, which is present on gonadal steroidogenic cells. Activation of this receptor has pleiotropic effects on cell growth, differentiation, and steroid production, which are mediated through several signaling pathways, including cAMP/protein kinase A (PKA; Fig. 14), mitogen-activated protein (MAP) kinase, and phosphatidylinositol-3-kinase.35,120 This in turn leads to phosphorylation/activation of transcription factors and enhanced expression of steroidogenic enzyme genes. The net effect is increased production of sex steroids, which feedback on the hypothalamic–pituitary axis and decrease LH secretion (Fig. 4). LHR has also been detected in lesser amounts on extragonadal cells, although the physiologic significance of this extragonadal LHR is uncertain.63 Several lines of evidence suggest that LH signaling plays a central role in adrenocortical tumorigenesis in neutered ferrets. A highly regulated system of hormonal and physical cues ensures that LH is produced at the appropriate time and concentration in the intact ferret.4 Gonadectomy disrupts feedback inhibition of the hypothalamic– pituitary axis, leading to increased numbers of gonadotrophs in the pars distalis adenohypophy-

105

sis112 and chronically elevated plasma LH levels.15,105,107 In the mature ferret, LH levels can be modulated further by photoperiodic stimulation.4 In neutered ferrets with adrenocortical tumors, plasma sex steroid levels are higher during the breeding season (March–August in the northern hemisphere), when prolonged daylight augments LH secretion by the pituitary.40,97,111 Conversely, alopecia and other signs of AAE can remit during autumn and winter in response to the decline in LH associated with a shortening photoperiod.23 Administration of GnRH increases plasma sex steroid levels in ferrets with adrenocortical tumors,111 whereas inhibition of gonadotropin secretion with leuprolide acetate or deslorelin acetate can decrease sex steroid production by tumors and temporarily ameliorate clinical signs of AAE.125,126 Pharmacologic and genetic studies in mice support the premise that LH has a central role in gonadectomy-induced adrenocortical neoplasia. Chronic gonadotropin elevation is a prerequisite for tumor development in all three of the mouse models. Hypophysectomy abrogates gonadectomyinduced adrenocortical tumorigenesis in inbred mice.37 Likewise, pharmacologic suppression of gonadotropin secretion with a GnRH antagonist precludes tumor development in inhibin-a promoter–SV40 T-antigen mice.93 Gonadectomy-induced adrenocortical neoplasia is prevented by introduction of an hpg (GnRH null) mutation into either inhibin-a promoter–SV40 T-antigen transgenic mice or inhibin-a null mice.56,89 Studies on NU/J nude mice, another inbred strain that is susceptible to gonadectomy-induced adrenocortical neoplasia, suggest that chronic LH elevation can induce adrenocortical tumorigenesis in the absence of gonadectomy.8 With the use of xenografts of human chorionic gonadotropin (hCG)–secreting cells, persistent gonadotropin stimulation was found to induce adrenocortical tumors in nongonadectomized NU/J nude mice.

r Fig. 11. Adrenal gland; intact DBA/2J mouse. A dashed line marks the capsule. P450c11, an enzyme required for corticoid synthesis, is expressed throughout the cortex. Fig. 11a. HE. Fig. 11b. P450c11, in situ hybridization with 33P-probe, dark-field view. Bar 5 600 mm. Fig. 12. Adrenal gland, adenoma; ovariectomized DBA/2J mouse. A dashed line marks the capsule. The cortex is infiltrated by tumor that lacks the capacity to produce corticoids. Fig. 12a. HE. Fig. 12b. P450c11, in situ hybridization with 33P-probe, dark-field view. Bar 5 600 mm, t5tumor. Fig. 13. Adrenal gland, adrenocortical carcinoma; gonadectomized inhibin-a promoter–SV40 T-antigen transgenic mouse. The boxed region in panel a is shown in panel b. Subcapsular A cells (arrows) and juxtamedullary carcinoma cells (circles) express GATA-4 but little GATA-6. Fig. 13a. HE. Fig 13b. HE. M5medulla. Fig 13c. GATA-4, in situ hybridization with 33P-probe, dark-field view. M5medulla. Fig 13d. GATA6, in situ hybridization with 33P-probe, dark-field view. Bars 5 100 mm, M5medulla.

Downloaded from vet.sagepub.com by guest on July 29, 2010

5–7 months Penetrance approaches 100% Estrogen

LHR, P450c17, aromatase

5–7 months Penetrance approaches 100% Estrogen

LHR, GATA-4, inhibin-a

Carcinomas, spindleCarcinomas, spindle-shaped shaped A cells, and sex A cells, and sex steroid– steroid–producing B cells producing B cells

Subcapsular and X-zone Subcapsular and X-zone

Molecular markers

Latency Incidence or penetrance Sex steroids produced

Variable, often near corticomedullary Subcapsular junction Nodular hyperplasia, adenomas, Nodular adenomas and and carcinomas; spindle cell carcinomas; composed component or myxoid differenof spindle-shaped A tiation might be present cells and sex steroid– producing B cells 3.5 years 1–6 months Reported incidence varies (0.5–20%) Penetrance of 40–100%, depending on the strain Estrogen, androstenedione, 17Estrogen (DBA/2J, CE, hydroxyprogesterone, DHEA-S NU/J) or androgens (CE) LHR, GATA-4, inhibin-a, ERa, LHR, GATA-4, inhibin-a, vimentin P450c17

Inhibin-a Null Mice Ferrets

Inbred Strains of Mice (e.g., DBA2/J, CE)

Inhibin-a Promoter–SV40 T-antigen Transgenic Mice

Bielinska, Kiiveri, Parviainen, Mannisto, Heikinheimo, and Wilson

Tumor location within adrenal Histology

Table 2.

Comparison of gonadectomy-induced adrenocortical neoplasia in domestic ferrets and three mouse models.

106

Vet Pathol 43:2, 2006

Table 3. Signaling molecules and cell cycle regulatory proteins implicated in gonadectomy-induced adrenocortical neoplasia. Endocrine or paracrine factors LH Prolactin Activins Inhibins MIS Wnts Sex steroids Transcription factors GATA-4 and -6 SF-1 FOG-2 DAX-1 CBP C/EBPb SMADs ERa Cell cycle regulatory proteins Cyclin D2 p27Kip1 p21Cip1

Transgenic mice with constitutively elevated LH/ hCG secretion have also been used to study the effects of chronic gonadotropin stimulation on adrenocortical function and tumorigenesis. Three such transgenic models have been generated: bLHb-CTP mice, which express an analog of LH in females,67 and two transgenic lines that overexpress hCG in both sexes.34,71,103 Female mice that bear both the bLHb-CTP and inhibin-a promoter– SV40 T-antigen transgenes develop adrenal tumors in the absence of gonadectomy.75 Interestingly, nongonadectomized female inhibin-a null mice bearing a bLHb-CTP transgene fail to develop adrenocortical carcinomas.5 In this instance, the secretion of activin by the gonadal tumors is postulated to inhibit adrenal tumor formation by accelerating the apoptosis of cells in the X-zone.5 For LH to have a direct effect on cells, its cognate receptor must be present. LHR is expressed in ovarian theca, granulosa, and luteal cells and in testicular Leydig cells. This receptor is also evident in the fetal but not adult adrenal of mice, a developmental expression pattern resembling those of GATA-4 and P450c17.2,9,51,90 Low-level expression of LHR mRNA and protein can be detected in the adrenal cortex of intact ferrets, but these receptors appear to be nonfunctional.86,111 Adrenocortical tumorigenesis in mice and ferrets is

Downloaded from vet.sagepub.com by guest on July 29, 2010

Vet Pathol 43:2, 2006

Gonadectomy-induced Adrenocortical Neoplasia

accompanied by dramatic and sustained increases in the levels of LHR mRNA and protein.84,111 This differs from the situation that exists in most LHresponsive cells, wherein chronic exposure to LH results in downregulation or desensitization of its receptor. The mechanism responsible this paradoxical upregulation of LHR in the neoplastic tissue is unknown. It is conceivable, albeit unproven, that the subcapsular or juxtamedullary regions contain rare cells that express functional LHR and transform or proliferate in response to chronic gonadotropin stimulation. Another possibility is that adrenocortical tumor formation is accompanied by increased expression of GATA-4, which then activates transcription of LHR in a feed-forward amplification loop.90 Concomitant with the upregulation of LHR in sex steroidogenic adrenocortical tumor cells, the ACTH receptor, MC2R, is downregulated.9 Thus, a switch in signaling accompanies adrenocortical tumorigenesis; the neoplastic tissue responds to LH but is no longer responsive to ACTH. Prolactin. Another pituitary hormone, prolactin, can affect steroidogenesis in gonadal and adrenocortical cells of some species by modulating LHR expression.18,47,68 In bLHb-CTP mice, prolactin appears to enhance LHR expression in normal adrenocortical cells, leading to expansion of the zona fasciculata and excess corticosterone production, which in turn is associated with retroperitoneal fat deposition and elevated serum leptin levels.47 Prolactin can potentiate LH-induced androgen production in ovarian interstitial cells.66,82 Given the established role of LHR in gonadectomy-induced adrenocortical tumorigenesis, it is tempting to speculate that changes in prolactin influence either tumor development or ectopic sex steroid production in ferrets. Like LH, prolactin secretion in ferrets is controlled by complex hormonal and physical cues (e.g., photoperiod) that are disrupted by gonadectomy.106 After neutering, 30–40% of the adenohypophyseal cells in healthy ferrets exhibit prolactin immunoreactivity.112 In female ferrets, prolactin has been shown to mediate the induction of puberty by long days.106 There is no direct evidence, however, that gonadectomy-induced changes in prolactin affect adrenocortical tumor formation in ferrets. Factors produced by gonadal and neoplastic adrenocortical tissue

Activins and inhibins. Activin and inhibin are dimeric glycoproteins belonging to the TGFb superfamily. They function as endocrine hormones

107

or paracrine factors that act within the pituitary, gonads, and other tissues, including the adrenal glands.6,17,94 These signaling molecules can elicit various effects on the growth and function of steroidogenic cells, depending on the cell type, developmental stage, receptors present, and the overall hormonal milieu. Activin promotes the differentiation of granulosa cells, but in theca cells it inhibits LH-dependent androgen production.17,122 A variety of gonadal and extragonadal cell types produce activin. In normal mammals, the serum levels of activin do not fall significantly after orchiectomy or ovariectomy, suggesting that serum levels of this factor mainly reflect extragonadal production.61 Nevertheless, local concentrations of activin might change in response to gonadectomy. Activin uses the TGFb signaling pathway, which includes transmembrane receptor kinases and their downstream mediators, the Smad proteins.115 Activin receptors are expressed in the murine adrenal gland,6 and activins have been shown to inhibit adrenocortical cell growth and steroid production and to enhance apoptosis of X-zone cells.5,56 Activin secreted by tumorous adrenals in gonadectomized inhibin-a–deficient mice inhibits carcinoma formation in the contralateral adrenal, possibly through effects on the X-zone.6 Inhibins are produced primarily by gonadal somatic cells. Consequently, gonadectomy leads to a profound and rapid fall in circulating inhibin levels. Inhibins have no direct effect on steroidogenesis or cell survival but can antagonize activin signaling by competing for activin receptors.122 The synthesis of inhibins also limits the availability of bA- and bB-subunits for activin synthesis.122 Although studies of inhibin-a null mice have established that this gene might function as a tumor suppressor, the inactivation of this locus is not essential for adrenocortical tumorigenesis. Indeed, robust expression of inhibin-a is evident in adrenocortical tumors from inbred mice,9 ferrets,85 and humans.122 Mu¨llerian inhibiting substance. MIS, also known as anti-Mu¨llerian hormone, is a distinctive member of the TGFb superfamily. Whereas most factors in this family have a range of functions in different organs, MIS acts only on reproductive organs. MIS is produced by gonadal somatic cells but not by normal adrenocortical cells. In the testis, MIS is produced in Sertoli cells, whereas in the ovary, it is secreted by granulosa cells. Binding of MIS to its receptor, MISIIR (MIS receptor type II), initiates downstream signaling via Smad proteins, analogous to the activin signaling pathway.25,124 MIS

Downloaded from vet.sagepub.com by guest on July 29, 2010

108

Bielinska, Kiiveri, Parviainen, Mannisto, Heikinheimo, and Wilson

causes Mu¨llerian duct regression during sexual differentiation and regulates postnatal Leydig cell development through its effects on cell proliferation and gene expression.131 MIS decreases the expression of P450c17 in Leydig cells and aromatase in granulosa cells.13,88 Male MIS-deficient mice are predisposed to Leydig cell hyperplasia, suggesting that MIS can inhibit Leydig cell proliferation.73 In rats, MIS inhibits rat adult Leydig cell regeneration after ethylene dimethanesulfonate ablation.108 Circumstantial evidence suggests a possible role for MIS in gonadotropin-induced adrenocortical neoplasia. MIS immunoreactivity has been observed in sex steroid–producing adrenocortical neoplasms of the ferret.83 In the gonadectomyinduced adrenocortical neoplasms that arise in inbred mice, MIS immunoreactivity localizes to A and B cells, whereas transcripts encoding MISIIR are found exclusively in A cells.8 It is tempting to speculate that MIS, acting in an autocrine or paracrine fashion, can inhibit proliferation or steroidogenesis in A cells, facilitate their centripetal migration, or both. Genetic studies suggest that inhibin-a and MIS exert a synergistic effect on tumorigenesis in gonads. In inhibin-a null mice, the absence of MIS causes earlier onset and more aggressive development of testicular tumors.73 Whether the loss or overexpression of MIS can affect gonadectomy-induced adrenocortical tumorigenesis is unknown. Wnt proteins. Wnt proteins have been implicated in a variety of normal and pathologic developmental processes, including neoplasia.87 Members of the Wnt family bind to Frizzled receptors on the cell surface and initiate a cytoplasmic signaling cascade that results in the stabilization of b-catenin and its subsequent translocation to the nucleus.87 Once in the nucleus, b-catenin can enhance gene expression through interactions with various transcription factors or coactivators, including TCF/ LEF family members, Smad factors, and CREBbinding protein (CBP). Wnt-2b is expressed in subcapsular cells of the adrenal62 and Wnt-4 in the zona glomerulosa.30 Targeted mutagenesis of Wnt4 leads to aberrant development of both the adrenal cortex and gonads; Wnt4 null mice have decreased expression of P450aldo in the adrenal and ectopic expression of the adrenal-specific marker P450c21 in gonadal tissue.30,41 In the adrenal glands of gonadectomized inbred NU/J nude mice, b-catenin immunoreactivity has been observed in the nuclei of B cells (and to a lesser extent A cells), suggesting that the Wnt signaling pathway might participate

Vet Pathol 43:2, 2006

in adrenocortical tumorigenesis.8 b-catenin has been shown to synergize with SF-1 in the transcriptional activation of inhibin-a in steroidogenic cells, which could account for the robust expression of this gene in type B cells.28 Sex steroids. In mouse models of gonadectomyassociated adrenocortical neoplasia, the administration of sex steroids can inhibit tumor formation or ectopic sex steroid production.56,92 Besides decreasing the number of pituitary gonadotrophs and suppressing pituitary gonadotropin secretion (Fig. 4), sex steroids can directly inhibit steroid production by normal or neoplastic adrenocortical cells.42,65 In the setting of elevated LH/hCG and testosterone levels, as is the case in intact NU/J mice bearing hCG-secreting xenografts, neoplastic cells accumulate in the adrenal cortex, but these cells express little P450c17, suggesting that they have limited capacity for sex steroid production.8 Expression of ERa in adrenocortical tumors of both mice8 and ferrets81 (Fig. 10) supports the notion that the neoplastic steroidogenic cells can respond directly to estrogen. Transcription factors

GATA transcription factors and their coactivators. GATA factors are a group of DNA-binding proteins that recognize the consensus motif (A/ T)GATA(A/G). One member of this family, GATA-4, is expressed in the genital ridge and in gonadal somatic cells of fetal and adult mammals.29,48,123 This transcription factor and its putative target gene, P450c17, are also transiently expressed in the fetal adrenal of the mouse but are absent from the adrenal cortex of postnatal animals.46,51,84 In gonadal steroidogenic cells of rodents, Gata4 is expressed during periods of active proliferation29,49,120 and can be induced by gonadotropins.52 Ectopic expression of GATA-4 in the adrenal heralds the appearance of neoplastic cells.9,84 In the ferret adrenal, GATA-4 immunoreactivity can be seen in both small and large neoplastic cells (Fig. 9) and seems to correlate with the degree of nuclear and cytoplasmic atypia.84 GATA-4 is particularly abundant in anaplastic myxoid adrenocortical carcinomas but is absent from the spindle cell component of adrenocortical tumors.84 In the mouse models of gonadectomy-induced adrenocortical neoplasia, GATA-4 is expressed in A cells, B cells, and juxtamedullary carcinoma cells (Fig. 13c).9 GATA-4 is a transcriptional activator of genes involved in sex steroid biogenesis, such as the genes

Downloaded from vet.sagepub.com by guest on July 29, 2010

Vet Pathol 43:2, 2006

Gonadectomy-induced Adrenocortical Neoplasia

109

Fig. 14. Regulation of StAR expression. The binding of LH to its receptor leads to increases in the level of cAMP (black circle with white star). Binding of cAMP to the regulatory subunit (R) of PKA allows dissociation of the catalytic subunit (C) and its translocation to the nucleus, where it phosphorylates target transcription factors, including GATA-4. SF-1 is essential for basal StAR expression and binds another widely expressed transcription factor, Sp1. C/EBPb (CCAAT-enhancer binding protein) acts synergistically with GATA-4. DAX-1 and FOG-2 function as repressors. Modified from Manna et al.69 and Tremblay and Viger.120

for the steroidogenic acute regulatory protein32 (STAR; Fig. 14), P450c17,22 and aromatase.116,119 In addition, GATA-4 has been shown to drive expression of the inhibin-a and -b subunit genes19,119 as well as LHR.90 Studies of genetically engineered mice suggest that fully functional GATA-4 is required for the proper differentiation of steroidogenic cells in the fetal testis.117 Several posttranslational modifications can modulate the activity of GATA-4. For example, phosphorylation of GATA-4 by PKA or MAP kinase enhances its ability to activate steroidogenic enzyme genes.120 Sumoylation of GATA-4 limits cooperative interactions between this factor and others that compose transcriptional complexes.55 In addition, there is evidence that GATA-4 acts as an antiapoptotic factor in various cell types, including gonadal steroidogenic cells.53 In neoplastic cells, upregulation of this factor can lead to enhanced expression of ‘‘gonadal-specific’’ genes harboring GATA binding sites in their promoters. Among the

upregulated genes, LHR could play an important role in the phenotypic switch from ACTH responsiveness to LH responsiveness and sex steroid production. Thus, GATA-4 is presumed to play a role in the induction, maintenance, or both of the neoplastic phenotype. Another member of the GATA family, GATA-6, is expressed in steroidogenic cells of the testis, ovary, fetal adrenal, and adult adrenal. On the basis of its tissue distribution and in vitro studies, GATA-6 appears to regulate steroidogenesis in adrenal and gonads rather than merely gonads, as is the case with GATA-4.22,59,120 In addition to activating genes critical for the differentiation and function of steroidogenic cells,70,109,120 GATA-6 might limit cellular proliferation by modulating the expression of cell cycle regulatory proteins.77 Increased expression of GATA-6 correlates with growth arrest and upregulation of the cell cycle inhibitor p21Cip1 in various nonsteroidogenic cell types.77 Adrenocortical tumor development in

Downloaded from vet.sagepub.com by guest on July 29, 2010

110

Bielinska, Kiiveri, Parviainen, Mannisto, Heikinheimo, and Wilson

inbred mice and inhibin-a promoter–SV40 Tantigen transgenic mice is accompanied by a decrease in GATA-6 in subcapsular A cells (Fig. 13d) and a contemporaneous increase in GATA-4 expression in these same cells (Fig. 13c).52 Gata6 expression persists in B cells, underscoring its importance in sex steroidogenesis. The mechanisms underlying the reciprocal changes in GATA-4 and GATA-6 in A cells are unknown; studies on knockout embryonic stem cells suggest that GATA-4 might directly or indirectly inhibit GATA-6 expression.78 GATA factors often execute their activity through interactions with the friend-of-GATA (FOG) family of coactivators or corepressors.1,95 One member of the FOG family, FOG-2, is essential for steroidogenic cell differentiation in the testis,117 and Fog2 is expressed in neoplastic B cells that accumulate in the adrenal of gonadectomized inbred mice.8 Another coactivator, CBP, can also augment the activity of GATA factors.36. CBP serves a critical role in the activation of cAMPinducible promoters or enhancers, which typify most steroidogenic enzyme genes. SF-1 and DAX-1. SF-1 is an orphan nuclear receptor expressed in steroidogenic cells of both the adrenal cortex and gonads.135 SF-1–deficient mouse embryos exhibit aberrant adrenal and gonadal development,135 and haplo-insufficiency for this factor impairs the compensatory adrenocortical cell growth that follows unilateral adrenalectomy.7 SF1 activates a wide array of genes involved in both adrenal and gonadal steroidogenesis. This transcription factor cooperates with GATA-4, GATA6, or both in the activation of genes expressed in gonads, including Mis and genes encoding steroid biosynthetic enzymes (Fig. 14).118,135 Similarly, SF1 cooperates with GATA-6 in the transcriptional activation of genes in normal adrenocortical tissue.59 Like GATA-4, SF-1 undergoes posttranslational modifications that modulate its activity, including phosphorylation and sumoylation.55 Because SF-1 is expressed in steroidogenic cells found in both normal and neoplastic tissue, it is not useful clinically as a specific marker of gonadectomyinduced adrenocortical neoplasia. In light of its established role in gonadal cell steroidogenesis, SF1 probably is an essential factor in the production of ectopic sex steroids by the adrenocortical tumors. The absence of SF-1 from type A cells suggests that this transcription factor might not be essential for the induction of tumorigenesis but rather is essential for maintenance of the steroidogenic cell phenotype.

Vet Pathol 43:2, 2006

The orphan nuclear hormone receptor DAX-1 has an expression pattern resembling that of SF1.38 DAX-1 binds to SF-1 in vitro and functions as a repressor of steroidogenesis, generally suppressing the same genes that SF-1 activates (Fig. 14).58,134 In Dax1 knockout mice, the adrenal cortex develops normally and produces steroid hormones, but X-zone degeneration is impaired.132,133 DAX-1 is downregulated in the neoplastic cells that accumulate in the adrenal cortex of gonadectomized inbred mice.8 Cell cycle regulatory proteins

There is evidence that adrenocortical tumorigenesis involves changes in cell proliferation, which are controlled by cyclins and their associated cyclindependent kinases (CDKs).114 Cyclins bind and activate CDKs and these cyclin–CDK complexes phosphorylate critical intracellular substrates, such as the product of the retinoblastoma tumor suppressor gene, thereby facilitating cell cycle progression.114 Mice bearing null mutations in both inhibin-a and Ccnd2, which encodes cyclin D2, develop gonadal and adrenocortical tumors more slowly than inhibina null mice.10 Thus, cyclin D2 can antagonize the suppressive effects of inhibin on adrenocortical tumorigenesis. In inhibin-a null mice, the absence of the CDK inhibitor p27Kip1 causes earlier onset and more aggressive gonadal tumors.10,12 However, the loss of p27Kip1 has little effect on adrenocortical tumor progression in gonadectomized inhibin-a null mice. As noted above, increased expression of GATA-6 correlates with growth arrest, downregulation of cyclin A, and the upregulation of another CDK inhibitor, p21Cip1, in various nonsteroidogenic cell types.77 Of note, this same correlation has not been observed in transplanted adrenal tissue or human adrenocortical tumors.50 Genetic Basis of Strain Susceptibility to Gonadectomy-induced Adrenocortical Neoplasia Adrenocortical neoplasms develop in only a minority of neutered ferrets, suggesting that genetic changes, perhaps inadvertently selected through inbreeding, influence susceptibility of adrenal tissue to neoplastic transformation. The same is true in mice, as borne out by the inbred and genetically engineered mouse models of adrenocortical neoplasia. Studies of inhibin-a promoter–SV40 Tantigen mice demonstrate that forced expression of a potent oncogene in the adrenal can alter tissue susceptibility to gonadectomy-induced tumor formation. Similarly, the phenotype of inhibin-a null mice shows that decreased expression of a tumor suppressor gene (inhibin-a), presumably in adrenal

Downloaded from vet.sagepub.com by guest on July 29, 2010

Vet Pathol 43:2, 2006

Gonadectomy-induced Adrenocortical Neoplasia

tissue per se, can affect susceptibility. Organ transplantation experiments with inbred mice suggest that strain susceptibility to gonadectomyinduced neoplasia resides in adrenal tissue; prepubertal adrenal glands transplanted from a susceptible strain into a gonadectomized nonsusceptible strain still develop tumors.37,121 However, the genetic changes (in the transplanted tissue) that account for strain-specific differences in susceptibility have not been elucidated. Inbred mouse strain susceptibility to gonadectomy-induced adrenocortical tumorigenesis appears to correlate with the in vivo and in vitro capacity of normal gonadal tissue to produce steroid hormones in response to gonadotropin stimulation. In general, ‘‘low-steroid’’ strains (e.g., CE, DBA/2J), which exhibit poor fertility, are susceptible to gonadectomy-induced tumorigenesis, whereas ‘‘high-steroid’’ strains (e.g., C57BL/6), which display superior reproductive performance, are resistant to gonadectomy-induced tumorigenesis.9 Intriguingly, a polymorphism in the Sf1 coding sequence appears to discriminate between low- and highsteroid strains.24 Although the physiologic consequence of this polymorphism is not yet known, such a mutation could alter the capacity of SF-1 to regulate adrenal versus gonadal-type lineage specification, either directly or through synergistic interactions with other transcription factors. Alterations in the level of SF-1 have been shown to disrupt compensatory adrenal growth response to unilateral adrenalectomy,7 so an inhibitory SF-1 mutation might preferentially affect adrenal cell proliferation and shift the balance in favor of ectopic gonadal cell differentiation or proliferation. It seems plausible that inherited genetic changes can alter the sensitivity of adrenal tissue to reproductive hormones or paracrine factors like LH and activin. Such changes might enhance cell proliferation or survival and thereby predispose cells to acquire additional (i.e., secondary) mutations that activate oncogenes or inhibit tumor suppressor genes. The genetically engineered mouse models of gonadectomy-induced tumorigenesis lend credence to such secondary events. Cells derived from adrenocortical tumors that arise in inhibina null mice are unresponsive to activin in vitro.6 This acquired property might explain how these cells are able to proliferate and secrete sex steroids despite high local concentrations of activin. Ambiguities in Adrenocortical Tumor Classification Confound the Discussion of Pathogenesis When categorizing adrenocortical lesions, it can be difficult to separate cell proliferation, often

111

termed ‘‘hyperplasia,’’ from so-called ‘‘adenomas.’’ Traditionally, pathologists have used the term ‘‘hyperplasia’’ to describe localized replacement of cortical tissue by abnormal cells, with an increase in the width of the cortex but minimal extension beyond the capsule. When the proliferation of the abnormal tissue is more extensive and localized, the term ‘‘adenoma’’ has been used. Actually no sharp distinctions can be made between the two. In the mouse, both types of lesions will progress and eventually become transplantable neoplasms provided the animal lives long enough,21 and the same could hold true for the ferret. Alternatively, the word ‘‘metaplasia’’ has been used to describe the transformation of adrenocortical tissue into cells resembling gonadal stroma.9 Although each of these terms has merit, we have elected to discuss gonadectomy-induced adrenocortical lesions under the rubric of ‘‘neoplasm,’’ acknowledging that tumorigenesis might be a multistep process that begins as hyperplasia or metaplasia. Human Adrenocortical Tumors As in ferrets and mice, adrenocortical tumor formation in humans appears to be influenced by both the hormonal milieu and the inherent susceptibility of the adrenal gland. Subcapsular adrenocortical neoplasms with histologic features resembling luteinized ovarian stroma (‘‘thecal metaplasia’’) have been reported in postmenopausal women20 and men with acquired testicular atrophy,96 suggesting that the hormone changes associated with gonadal failure (e.g., elevated LH, decreased sex steroids, etc.) might contribute to tumor development in humans. Analyses of heritable and spontaneous types of human adrenocortical tumors have documented alterations in either cell surface receptors or their downstream effectors that make adrenocortical tissue more susceptible to hormone-associated neoplastic transformation (Table 4).54,57 In a majority of cases, these genetic alterations lead to adrenocortical neoplasms that produce excess glucocorticoids, but in a few cases, notably those involving youngchildren, the adrenocortical tumors produce large amounts of sex steroids and inhibin-a, like the tumors in gonadectomized ferrets and mice. Whether such genetic changes contribute to tissue susceptibility to neoplasia in neutered ferrets or mice awaits further study. The inbred mouse models, coupled with the rapidly expanding database of genomic DNA sequences and single nucleotide polymorphisms in various strains, afford a means to characterize the alleles and genetic modifiers influencing gonadectomy-induced adre-

Downloaded from vet.sagepub.com by guest on July 29, 2010

112

Bielinska, Kiiveri, Parviainen, Mannisto, Heikinheimo, and Wilson

Table 4. Genetic alternations observed in heritable and spontaneous types of human adrenocortical tumors. Ectopic or precocious expression of cell surface receptors for: Gastric inhibitory polypeptide Vasopressin b-Adrenergic agonists IL-1 LH G-protein mutations Activating mutations in Gsa (McCune–Albright syndrome) Inactivating mutations in Gi2a Increased PKA activity caused by mutations in its regulatory subunit (Carney complex) Decreased expression of the CDK inhibitor P57kip2 (Beckwith–Wiedemann syndrome) Inactivating mutations in tumor suppressor TP53 (Li–Fraumeni syndrome)

5

6

7

8

9

nocortical neoplasia. In addition, these models will facilitate studies of the signaling pathways that control cell fate decisions in normal and neoplastic steroidogenic lineages. 10

Acknowledgements We thank Drs. Richard Peterson and Charles Capen (The Ohio State University) and Dr. Matti Kiupel (Michigan State University) for providing tumor specimens from neutered ferrets. We thank Drs. Nafis Rahman (University of Turku) and Ilpo Huhtaniemi (Imperial College London) for providing tissue sections of gonadectomized inhibin-a promoter–SV40 T-antigen transgenic mice. Rosie Wilson provided technical assistance. The Finnish Pediatric Research Foundation; Juselius Foundation; NIH HL61006; and DK52574, MOD 1-FY02-203, and AHA 0455623Z supported this work.

11

12

13

References 1 Anttonen M, Ketola I, Parviainen H, Pusa AK, Heikinheimo M: FOG-2 and GATA-4 are coexpressed in the mouse ovary and can modulate Mu¨llerian-inhibiting substance expression. Biol Reprod 68:1333–1340, 2003 2 Apaja PM, Aatsinki JT, Rajaniemi HJ, PetajaRepo UE: Expression of the mature luteinzing hormone receptor in rodent urogenital and adrenal tissues is developmentally regulated at a posttranslational level. Endocrinology 146:3224–3232 3 Auchus RJ: Overview of dehydroepiandrosterone biosynthesis. Semin Reprod Med 22:281–288, 2004 4 Baum MJ: Use of the Ferret in Reproductive Endocrinology. In: Biology and Diseases of the

14

15

16

Vet Pathol 43:2, 2006

Ferret, ed. Fox JG, 2nd ed., pp. 521–535. Lea & Febiger, Philadelphia, PA, 1998 Beuschlein F, Looyenga BD, Bleasdale SE, Mutch C, Bavers DL, Parlow AF, Nilson JH, Hammer GD: Activin induces X-zone apoptosis that inhibits luteinizing hormone-dependent adrenocortical tumor formation in inhibin-deficient mice. Mol Cell Biol 23:3951–3964, 2003 Beuschlein F, Looyenga BD, Reincke M, Hammer GD: Role of the inhibin/activin system and luteinizing hormone in adrenocortical tumorigenesis. Horm Metab Res 36:392–396, 2004 Beuschlein F, Mutch C, Bavers DL, Ulrich-Lai YM, Engeland WC, Keegan C, Hammer GD: Steroidogenic factor-1 is essential for compensatory adrenal growth following unilateral adrenalectomy. Endocrinology 143:3122–3135, 2002 Bielinska M, Genova E, Boime I, Parviainen H, Kiiveri S, Rahman N, Leppa¨luoto J, Heikinheimo M, Wilson DB: Nude mice as a model for gonadotropin-induced adrenocortical neoplasia. Endocrinology 146:3975–3984 Bielinska M, Parviainen H, Porter-Tinge SB, Kiiveri S, Genova E, Rahman N, Huhtaniemi IT, Muglia LJ, Heikinheimo M, Wilson DB: Mouse strain susceptibility to gonadectomy-induced adrenocortical tumor formation correlates with the expression of GATA-4 and luteinizing hormone receptor. Endocrinology 144:4123–4133, 2003 Burns KH, Agno JE, Sicinski P, Matzuk MM: Cyclin D2 and p27 are tissue-specific regulators of tumorigenesis in inhibin-a knockout mice. Mol Endocrinol 17:2053–2069, 2003 Burns KH, Owens GE, Ogbonna SC, Nilson JH, Matzuk MM: Expression profiling analyses of gonadotropin responses and tumor development in the absence of inhibins. Endocrinology 144: 4492–4507, 2003 Cipriano SC, Chen L, Burns KH, Koff A, Matzuk MM: Inhibin and p27 interact to regulate gonadal tumorigenesis. Mol Endocrinol 15:985– 996, 2001 di Clemente N, Ghaffari S, Pepinsky RB, Pieau C, Josso N, Cate RL, Vigier B: A quantitative and interspecific test for biological activity of antimu¨llerian hormone: the fetal ovary aromatase assay. Development 114:721–727, 1992 Donovan BT, Matson C, Kilpatrick MJ: Effect of exposure to long days on the secretion of oestradiol, oestrone, progesterone, testosterone, androstenedione, cortisol and follicle-stimulating hormone in intact and spayed ferrets. J Endocrinol 99: 361–368, 1983 Donovan BT, ter Haar MB: Effects of luteinizing hormone releasing hormone on plasma folliclestimulating hormone and luteinizing hormone levels in the ferret. J Endocrinol 73:37–52, 1977 Douma S, Van Laar T, Zevenhoven J, Meuwissen R, Van Garderen E, Peeper DS: Suppression of anoikis and induction of metastasis by the

Downloaded from vet.sagepub.com by guest on July 29, 2010

Vet Pathol 43:2, 2006

17 18 19

20 21 22

23

24

25

26

27

28

29

30

31

Gonadectomy-induced Adrenocortical Neoplasia

neurotrophic receptor TrkB. Nature 430:1034– 1039, 2004 Drummond AE, Findlay JK, Ireland JJ: Animal models of inhibin action. Semin Reprod Med 22: 243–252, 2004 Eldridge JC, Lymangrover JR: Prolactin stimulates and potentiates adrenal steroid secretion in vitro. Horm Res 20:252–260, 1984 Feng ZM, Wu AZ, Zhang Z, Chen CL: GATA-1 and GATA-4 transactivate inhibin/activin beta-Bsubunit gene transcription in testicular cells. Mol Endocrinol 14:1820–1835, 2000 Fidler WJ: Ovarian thecal metaplasia in adrenal glands. Am J Clin Pathol 67:318–323, 1977 Firth CH, Dunn TB: Tumours of the adrenal gland. IARC Sci Publ 111:595–609, 1994 Fluck CE, Miller WL: GATA-4 and GATA-6 modulate tissue-specific transcription of the human gene for P450c17 by direct interaction with Sp1. Mol Endocrinol 18:1144–1157, 2004 Fox JG, Marini RP: Diseases of the endocrine system. In: Biology and Diseases of the Ferret, ed. Fox JG, 2nd ed., pp. 291–305. Lea & Febiger, Philadelphia, PA, 1998 Frigeri C, Tsao J, Cordova M, Schimmer BP: A polymorphic form of steroidogenic factor-1 is associated with adrenocorticotropin resistance in Y1 mouse adrenocortical tumor cell mutants. Endocrinology 143:4031–4037, 2002 Fynn-Thompson E, Cheng H, Teixeira J: Inhibition of steroidogenesis in Leydig cells by Mullerianinhibiting substance. Mol Cell Endocrinol 211: 99–104, 2003 Gliatto JM, Alroy J, Schelling SH, Engler SJ, Dayal Y: A light microscopical, ultrastructural and immunohistochemical study of spindle-cell adrenocortical tumours of ferrets. J Comp Pathol 113: 175–183, 1995 Grazzini E, Boccara G, Joubert D, Trueba M, Durroux T, Guillon G, Gallo-Payet N, Chouinard L, Payet MD, Serradeil LG: Vasopressin regulates adrenal functions by acting through different vasopressin receptor subtypes. Adv Exp Med Biol 449:325–334, 1998 Gummow BM, Winnay JN, Hammer GD: Convergence of Wnt signaling and steroidogenic factor1 (SF-1) on transcription of the rat inhibin alpha gene. J Biol Chem 278:26572–26579, 2003 Heikinheimo M, Ermolaeva M, Bielinska M, Rahman NA, Narita N, Huhtaniemi IT, Tapanainen JS, Wilson DB: Expression and hormonal regulation of transcription factors GATA-4 and GATA-6 in the mouse ovary. Endocrinol 138:3505–3514, 1997 Heikkila M, Peltoketo H, Leppaluoto J, Ilves M, Vuolteenaho O, Vainio S: Wnt-4 deficiency alters mouse adrenal cortex function, reducing aldosterone production. Endocrinology 143:4358–4365, 2002 Hillyer EV: Adrenal gland tumors in ferrets. J Am Vet Med Assoc 205:1660–1661, 1994

113

32 Hiroi H, Christenson LK, Strauss JF III: Regulation of transcription of the steroidogenic acute regulatory protein (StAR) gene: temporal and spatial changes in transcription factor binding and histone modification. Mol Cell Endocrinol 215: 119–126, 2004 33 Holmes RL: The adrenal glands of the ferret. J Anat 95:325–336, 1961 34 Huhtaniemi I, Rulli S, Ahtiainen P, Poutanen M: Multiple sites of tumorigenesis in transgenic mice overproducing hCG. Mol Cell Endocrinol 234: 117–126, 2005 35 Huhtaniemi I, Zhang FP, Kero J, Hamalainen T, Poutanen M: Transgenic and knockout mouse models for the study of luteinizing hormone and luteinizing hormone receptor function. Mol Cell Endocrinol 187:49–56, 2002 36 Hung HL, Lau J, Kim AY, Weiss MJ, Blobel GA: CREB-binding protein acetylates hematopoietic transcription factor GATA-1 at functionally important sites. Mol Cell Biol 19:3496–3505, 1999 37 Huseby RA, Bittner JJ: Differences in adrenal responsiveness to post-castrational alteration as evidenced by transplanted adrenal tissue. Cancer Res 11:954–961, 1951 38 Ito M, Yu R, Jameson JL: DAX-1 inhibits SF-1-mediated transactivation via a carboxyterminal domain that is deleted in adrenal hypoplasia congenita. Mol Cell Biol 17:1476– 1483, 1997 39 Jabara S, Christenson LK, Wang CY, McAllister JM, Javitt NB, Dunaif A, Strauss JF III: Stromal cells of the human postmenopausal ovary display a distinctive biochemical and molecular phenotype. J Clin Endocrinol Metab 88:484–492, 2003 40 Jallageas M, Boissin J, Mas N: Differential photoperiodic control of seasonal variations in pulsatile luteinizing hormone release in long-day (ferret) and short-day (mink) mammals. J Biol Rhythms 9:217–231, 1994 41 Jeays-Ward K, Hoyle C, Brennan J, Dandonneau M, Alldus G, Capel B, Swain A: Endothelial and steroidogenic cell migration are regulated by WNT4 in the developing mammalian gonad. Development 130:3663–3670, 2003 42 Johnson DC, Crane LH: Inhibitory and stimulatory effect of oestrogens upon ovarian 17a-hydroxylase/C17, 20-lyase in immature hypophysectomized rats treated with gonadotrophin. J Endocrinol 145: 59–67, 1995 43 Kaaijk EM, Sasano H, Suzuki T, Beek JF, van Der Veen F: Distribution of steroidogenic enzymes involved in androgen synthesis in polycystic ovaries: an immunohistochemical study. Mol Hum Reprod 6:443–447, 2000 44 Kananen K, Markkula M, Mikola M, Rainio EM, McNeilly A, Huhtaniemi I: Gonadectomy permits adrenocortical tumorigenesis in mice transgenic for the mouse inhibin a-subunit promoter/simian virus 40 T-antigen fusion gene: evidence for negative

Downloaded from vet.sagepub.com by guest on July 29, 2010

114

45 46

47

48

49

50

51

52

53

54

55

56

Bielinska, Kiiveri, Parviainen, Mannisto, Heikinheimo, and Wilson

autoregulation of the inhibin a-subunit gene. Mol Endocrinol 10:1667–1677, 1996 Keegan CE, Hammer GD: Recent insights into organogenesis of the adrenal cortex. Trends Endocrinol Metab 13:200–208, 2002 Keeney DS, Jenkins CM, Waterman MR: Developmentally regulated expression of adrenal 17ahydroxylase cytochrome P450 in the mouse embryo. Endocrinology 136:4872–4879, 1995 Kero J, Poutanen M, Zhang FP, Rahman N, McNicol AM, Nilson JH, Keri RA, Huhtaniemi IT: Elevated luteinizing hormone induces expression of its receptor and promotes steroidogenesis in the adrenal cortex. J Clin Invest 105:633–641, 2000 Ketola I, Pentikainen V, Vaskivuo T, Ilvesmaki V, Herva R, Dunkel L, Tapanainen JS, Toppari J, Heikinheimo M: Expression of transcription factor GATA-4 during human testicular development and disease. J Clin Endocrinol Metab 85:3925–3931, 2000 Ketola I, Rahman N, Toppari J, Bielinska M, Porter-Tinge SB, Tapanainen JS, Huhtaniemi IT, Wilson DB, Heikinheimo M: Expression and regulation of transcription factors GATA-4 and GATA-6 in developing mouse testis. Endocrinology 140:1470–1480, 1999 Kiiveri S, Liu J, Arola J, Heikkila¨ P, Lehtonene E, Voutilainen R, Heikinheimo M: Transcription factors GATA-6, SF-1, and cell proliferation in human adrenocortical tumors. Mol Cell Endocrinol 233:47–56 Kiiveri S, Liu J, Westerholm-Ormio M, Narita N, Wilson DB, Voutilainen R, Heikinheimo M: Differential expression of GATA-4 and GATA-6 in fetal and adult mouse and human adrenal tissue. Endocrinology 143:3136–3143, 2002 Kiiveri S, Siltanen S, Rahman N, Bielinska M, Lehto VP, Huhtaniemi IT, Muglia LJ, Wilson DB, Heikinheimo M: Reciprocal changes in the expression of transcription factors GATA-4 and GATA-6 accompany adrenocortical tumorigenesis in mice and humans. Mol Med 5:490–501, 1999 Kim Y, Ma AG, Kitta K, Fitch SN, Ikeda T, Ihara Y, Simon AR, Evans T, Suzuki YJ: Anthracyclineinduced suppression of GATA-4 transcription factor: implication in the regulation of cardiac myocyte apoptosis. Mol Pharmacol 63:368–377, 2003 Koch CA, Pacak K, Chrousos GP: The molecular pathogenesis of hereditary and sporadic adrenocortical and adrenomedullary tumors. J Clin Endocrinol Metab 87:5367–5384, 2002 Komatsu T, Mizusaki H, Mukai T, Ogawa H, Baba D, Shirakawa M, Hatakeyama S, Nakayama KI, Yamamoto H, Kikuchi A, Morohashi KI: SUMO1 modification of the synergy control motif of Ad4BP/SF-1 regulates synergistic transcription between Ad4BP/SF-1 and Sox9. Mol Endocrinol 18:2451–2462, 2004 Kumar TR, Donehower LA, Bradley A, Matzuk MM: Transgenic mouse models for tumour-suppressor genes. J Intern Med 238:233–238, 1995

Vet Pathol 43:2, 2006

57 Lacroix A, Baldacchino V, Bourdeau I, Hamet P, Tremblay J: Cushing’s syndrome variants secondary to aberrant hormone receptors. Trends Endocrinol Metab 15:375–382, 2004 58 Lalli E, Bardoni B, Zazopoulos E, Wurtz JM, Strom TM, Moras D, Sassone-Corsi P: A transcriptional silencing domain in DAX-1 whose mutation causes adrenal hypoplasia congenita. Mol Endocrinol 11:1950–1960, 1997 59 LaVoie HA: The role of GATA in mammalian reproduction. Exp Biol Med 228:1282–1290, 2003 60 Li X, Fox JG, Padrid PA: Neoplastic diseases in ferrets: 574 cases (1968–1997). J Am Vet Med Assoc 212:1402–1406, 1998 61 Lin SY, Morrison JR, Phillips DJ, de Kretser DM: Regulation of ovarian function by the TGF-beta superfamily and follistatin. Reproduction 126: 133–148, 2003 62 Lin Y, Liu A, Zhang S, Ruusunen T, Kreidberg JA, Peltoketo H, Drummond I, Vainio S: Induction of ureter branching as a response to Wnt-2b signaling during early kidney organogenesis. Dev Dyn 222:26–39, 2001 63 Lo KC, Lei Z, Rao C, Beck J, Lamb DJ: De novo testosterone production in luteinizing hormone receptor knockout mice after transplantation of leydig stem cells. Endocrinology 145:4011–4015, 2004 64 Magalhaes MC: A new crystal-containing cell in human adrenal cortex. J Cell Biol 55:126–133, 1972 65 Magoffin DA, Erickson GF: Direct inhibitory effect of estrogen on LH-stimulated androgen synthesis by ovarian cells cultured in defined medium. Mol Cell Endocrinology 28:81–89, 1982 66 Magoffin DA, Erickson GF: Prolactin inhibition of luteinizing hormone-stimulated androgen synthesis in ovarian interstitial cells cultured in defined medium: mechanism of action. Endocrinology 111:2001–2007, 1982 67 Mann RJ, Keri RA, Nilson JH: Consequences of elevated luteinizing hormone on diverse physiological systems: use of the LHbCTP transgenic mouse as a model of ovarian hyperstimulation-induced pathophysiology. Recent Prog Horm Res 58: 343–375, 2003 68 Manna PR, El Hefnawy T, Kero J, Huhtaniemi IT: Biphasic action of prolactin in the regulation of murine Leydig tumor cell functions. Endocrinology 142:308–318, 2001 69 Manna PR, Wang XJ, Stocco DM: Involvement of multiple transcription factors in the regulation of steroidogenic acute regulatory protein gene expression. Steroids 68:1125–1134, 2003 70 Martin LJ, Taniguchi H, Robert NM, Simard J, Tremblay JJ, Viger RS: GATA factors and the nuclear receptors SF-1/LRH-1 are key mutual partners in the regulation of the human HSD3B2 promoter. Mol Endocrinol 19:2358–2370 71 Matzuk MM, DeMayo FJ, Hadsell LA, Kumar TR: Overexpression of human chorionic gonado-

Downloaded from vet.sagepub.com by guest on July 29, 2010

Vet Pathol 43:2, 2006

72

73

74

75

76 77

78

79

80

81

82 83

84

Gonadectomy-induced Adrenocortical Neoplasia

tropin causes multiple reproductive defects in transgenic mice. Biol Reprod 69:338–346, 2003 Matzuk MM, Finegold MJ, Mather JP, Krummen L, Lu H, Bradley A: Development of cancer cachexia-like syndrome and adrenal tumors in inhibin-deficient mice. Proc Natl Acad Sci USA 91:8817–8821, 1994 Matzuk MM, Finegold MJ, Mishina Y, Bradley A, Behringer RR: Synergistic effects of inhibins and mu¨llerian-inhibiting substance on testicular tumorigenesis. Mol Endocrinol 9:1337–1345, 1995 Matzuk MM, Finegold MJ, Su JG, Hsueh AJ, Bradley A: Alpha-inhibin is a tumour-suppressor gene with gonadal specificity in mice. Nature 360: 313–319, 1992 Mikola M, Kero J, Nilson JH, Keri RA, Poutanen M, Huhtaniemi I: High levels of luteinizing hormone analog stimulate gonadal and adrenal tumorigenesis in mice transgenic for the mouse inhibin-a subunit promoter/Simian virus 40 Tantigen fusion gene. Oncogene 22:3269–3278, 2003 Morohashi K: The ontogenesis of the steroidogenic tissues. Genes Cells 2:95–106, 1997 Morrisey EE: GATA-6: the proliferation stops here—cell proliferation in glomerular mesangial and vascular smooth muscle cells. Circ Res 87: 638–640, 2000 Morrisey EE, Tang Z, Sigrist K, Lu MM, Jiang F, Ip HS, Parmacek MS: GATA6 regulates HNF4 and is required for differentiation of visceral endoderm in the mouse embryo. Genes Dev 12: 3579–3590, 1998 Nagata D, Suzuki E, Nishimatsu H, Yoshizumi M, Mano T, Walsh K, Sata M, Kakoki M, Goto A, Omata M, Hirata Y: Cyclin A downregulation and p21cip1 upregulation correlate with GATA-6-induced growth arrest in glomerular mesangial cells. Circ Res 87:699–704, 2000 Natoli TA, Alberta JA, Bortvin A, Taglienti ME, Menke DB, Loring J, Jaenisch R, Page DC, Housman DE, Kreidberg JA: Wt1 functions in the development of germ cells in addition to somatic cell lineages of the testis. Dev Biol 268: 429–440, 2004 Newman SJ, Bergman PJ, Williams B, Scase T, Craft D: Characterization of spindle cell compenent of ferret (Mustela putorius furo) adrenal cortical neoplasms—correlation to clinical parameters and prognosis. Vet Comp Oncol 2:113–124, 2004 Odell WD, Parker LN: Control of adrenal androgen production. Endocr Res 10:617–630, 1984 Patterson MM, Rogers AB, Schrenzel MD, Marini RP, Fox JG: Alopecia attributed to neoplastic ovarian tissue in two ferrets. Comp Med 53: 213–217, 2003 Peterson RA, Kiupel M, Bielinska M, Kiiveri S, Heikinheimo M, Capen CC, Wilson DB: Transcription factor GATA-4 is a marker of anaplasia in adrenocortical neoplasms of the domestic ferret

85

86

87 88

89

90

91

92

93

94 95

96

115

(Mustela putorius furo). Vet Pathol 41:446–449, 2004 Peterson RA, Kiupel M, Capen CC: Adrenal cortical carcinomas with myxoid differentiation in the domestic ferret (Mustela putorius furo). Vet Pathol 40:136–142, 2003 Pietila EM, Tuusa JT, Apaja PM, Aatsinki JT, Hakalahti AE, Rajaniemi HJ, Petaja-Repo UE: Inefficient maturation of the rat luteinizing hormone receptor: A putative way to regulate receptor numbers at the cell surface. J Biol Chem 280:26622–26629 Prunier C, Hocevar BA, Howe PH: Wnt signaling: physiology and pathology. Growth Fact 22: 141–150, 2004 Racine C, Rey R, Forest MG, Louis F, Ferre A, Huhtaniemi I, Josso N, di Clemente N: Receptors for anti-mu¨llerian hormone on Leydig cells are responsible for its effects on steroidogenesis and cell differentiation. Proc Natl Acad Sci USA 95: 594–599, 1998 Rahman NA, Kananen RK, Paukku T, Mikola M, Markkula M, Hamalainen T, Huhtaniemi IT: Transgenic mouse models for gonadal tumorigenesis. Mol Cell Endocrinol 145:167–174, 1998 Rahman NA, Kiiveri S, Rivero-Muller A, Levallet J, Vierre S, Kero J, Wilson DB, Heikinheimo M, Huhtaniemi I: Adrenocortical tumorigenesis in transgenic mice expressing the inhibin a-subunit promoter/SV40 virus T-antigen transgene: relationship between ectopic expression of luteinizing hormone receptor and transcription factor GATA-4. Mol Endocrinol 18:2553–2569, 2004 Rahman NA, Kiiveri S, Siltanen S, Levallet J, Kero J, Lensu T, Wilson DB, Heikinheimo MT, Huhtaniemi IT: Adrenocortical tumorigenesis in transgenic mice: the role of luteinizing hormone receptor and transcription factors GATA-4 and GATA-6. Reprod Biol 1:5–9, 2001 Rilianawati., Kero J, Paukku T, Huhtaniemi I: Long-term testosterone treatment prevents gonadal and adrenal tumorigenesis of mice transgenic for the mouse inhibin-a subunit promoter/simian virus 40 T-antigen fusion gene. J Endocrinol 166:77–85, 2000 Rilianawati., Paukku T, Kero J, Zhang FP, Rahman N, Kananen K, Huhtaniemi I: Direct luteinizing hormone action triggers adrenocortical tumorigenesis in castrated mice transgenic for the murine inhibin a-subunit promoter/simian virus 40 T-antigen fusion gene. Mol Endocrinol 12:801–809, 1998 Risbridger GP, Schmitt JF, Robertson DM: Activins and inhibins in endocrine and other tumors. Endocr Rev 22:836–858, 2001 Robert NM, Tremblay JJ, Viger RS: Friend of GATA (FOG)-1 and FOG-2 differentially repress the GATA-dependent activity of multiple gonadal promoters. Endocrinology 143:3963–3973, 2002 Romberger CF, Wong TW: Thecal metaplasia in the adrenal gland of a man with acquired bilateral

Downloaded from vet.sagepub.com by guest on July 29, 2010

116

97 98

99

100

101 102

103

104 105 106

107

108

109

110

Bielinska, Kiiveri, Parviainen, Mannisto, Heikinheimo, and Wilson

testicular atrophy. Arch Pathol Lab Med 113: 1071–1075, 1989 Rosenthal KL: Adrenal gland disease in ferrets. Vet Clin North Am Small Anim Pract 27:401–418, 1997 Rosenthal KL: Endocrine diseases part II. In: Ferrets, Rabbits, and Rodents: Clinical Medicine and Surgery, ed. Hillyer EV and Quesenberry KE, pp. 91–98. W.B. Saunders Company, Philadephia, PA, 1997 Rosenthal KL, Peterson ME: Evaluation of plasma androgen and estrogen concentrations in ferrets with hyperadrenocorticism. J Am Vet Med Assoc 209:1097–1102, 1996 Rosenthal KL, Peterson ME, Quesenberry KE, Hillyer EV, Beeber NL, Moroff SD, Lothrop CD Jr: Hyperadrenocorticism associated with adrenocortical tumor or nodular hyperplasia of the adrenal gland in ferrets: 50 cases (1987–1991). J Am Vet Med Assoc 203:271–275, 1993 Rosol TJ, Yarrington JT, Latendresse J, Capen CC: Adrenal gland: structure, function, and mechanisms of toxicity. Toxicol Pathol 29:41–48, 2001 Rossi GP, Sacchetto A, Cesari M, Pessina AC: Interactions between endothelin-1 and the reninangiotensin-aldosterone system. Cardiovasc Res 43: 300–307, 1999 Rulli SB, Kuorelahti A, Karaer O, Pelliniemi LJ, Poutanen M, Huhtaniemi I: Reproductive disturbances, pituitary lactotrope adenomas, and mammary gland tumors in transgenic female mice producing high levels of human chorionic gonadotropin. Endocrinology 143:4084–4095, 2002 Russfield AB: Experimental endocrinopathies. Methods Achiev Exp Pathol 7:132–148, 1975 Ryan KD: Hormonal correlates of photoperiodinduced puberty in a reflex ovulator, the female ferret (Mustela furo). Biol Reprod 31:925–935, 1984 Ryan KD, Robinson SL: Prolactin or dopamine mediates the induction of puberty by long days in female ferrets. Endocrinology 125:2605–2611, 1989 Ryan KD, Siegel SF, Robinson SL: Influence of day length and endocrine status on luteinizing hormone secretion in intact and ovariectomized adult ferrets. Biol Reprod 33:690–697, 1985 Salva A, Hardy MP, Wu XF, Sottas CM, MacLaughlin DT, Donahoe PK, Lee MM: Mu¨llerian-inhibiting substance inhibits rat Leydig cell regeneration after ethylene dimethanesulphonate ablation. Biol Reprod 70:600–607, 2004 Saner KJ, Suzuki T, Sasano H, Pizzey J, Ho C, Strauss JF III, Carr BR, Rainey WE: Steroid sulfotransferase (SULT2A1) gene transcription is regulated by steroidogenic factor 1 (SF1) and GATA-6 in the human adrenal. Mol Endocrinol 19:184–197, 2004 Schoemaker NJ, Schuurmans M, Moorman H, Lumeij JT: Correlation between age at neutering and age at onset of hyperadrenocorticism in ferrets. J Am Vet Med Assoc 216:195–197, 2000

Vet Pathol 43:2, 2006

111 Schoemaker NJ, Teerds KJ, Mol JA, Lumeij JT, Thijssen JH, Rijnberk A: The role of luteinizing hormone in the pathogenesis of hyperadrenocorticism in neutered ferrets. Mol Cell Endocrinol 197:117–125, 2002 112 Schoemaker NJ, van der Hage MH, Flik G, Lumeij JT, Rijnberk A: Morphology of the pituitary gland in ferrets (Mustela putorius furo) with hyperadrenocorticism. J Comp Pathol 130: 255–265, 2004 113 Schoemaker NJ, Wolfswinkel J, Mol JA, Voorhout G, Kik MJ, Lumeij JT, Rijnberk A: Urinary glucocorticoid excretion in the diagnosis of hyperadrenocorticism in ferrets. Domest Anim Endocrinol 27:13–24, 2004 114 Sherr CJ, Roberts JM: CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev 13:1501–1512, 1999 115 Shi Y, Massague J: Mechanisms of TGF-b signaling from cell membrane to the nucleus. Cell 113:685– 700, 2003 116 Stocco C: In vivo and in vitro inhibition of cyp19 gene expression by prostaglandin F2a in murine luteal cells: implication of GATA-4. Endocrinology 145:4957–4966, 2004 117 Tevosian SG, Albrecht KH, Crispino JD, Fujiwara Y, Eicher EM, Orkin SH: Gonadal differentiation, sex determination and normal Sry expression in mice require direct interaction between transcription partners GATA4 and FOG2. Development 129:4627–4634, 2002 118 Tremblay JJ, Viger RS: Transcription factor GATA-4 enhances Mu¨llerian inhibiting substance gene transcription through a direct interaction with the nuclear receptor SF-1. Mol Endocrinol 13: 1388–1401, 1999 119 Tremblay JJ, Viger RS: GATA factors differentially activate multiple gonadal promoters through conserved GATA regulatory elements. Endocrinology 142:977–986, 2001 120 Tremblay JJ, Viger RS: Novel roles for GATA transcription factors in the regulation of steroidogenesis. J Steroid Biochem Mol Biol 85:291–298, 2003 121 Tullos HS, Kirschbaum A, Trentin JJ: Role of gonadotropic hormone in the initiation and progression of adrenal tumors in ovariectomized mice. Cancer Res 21:730–734, 1960 122 Vanttinen T, Liu J, Kuulasmaa T, Kivinen P, Voutilainen R: Expression of activin/inhibin signaling components in the human adrenal gland and the effects of activins and inhibins on adrenocortical steroidogenesis and apoptosis. J Endocrinol 178:479–489, 2003 123 Viger RS, Mertineit C, Trasler JM, Nemer M: Transcription factor GATA-4 is expressed in a sexually dimorphic pattern during mouse gonadal development and is a potent activator of the Mu¨llerian inhibiting substance promoter. Development 125:2665–2755, 1998

Downloaded from vet.sagepub.com by guest on July 29, 2010

Vet Pathol 43:2, 2006

Gonadectomy-induced Adrenocortical Neoplasia

124 Visser JA: AMH signaling: from receptor to target gene. Mol Cell Endocrinol 211:65–73, 2003 125 Wagner RA, Bailey EM, Schneider JF, Oliver JW: Leuprolide acetate treatment of adrenocortical disease in ferrets. J Am Vet Med Assoc 218: 1272–1274, 2001 126 Wagner RA, Piche CA, Jochle W, Oliver JW: Clinical and endocrine responses to treatment with deslorelin acetate implants in ferrets with adrenocortical disease. Am J Vet Res 66:910–914, 2005 127 Weber MM, Fottner C, Wolf E: The role of the insulin-like growth factor system in adrenocortical tumourigenesis. Eur J Clin Invest 30 (Suppl 3): 69–75, 2000 128 Weiss CA, Scott MV: Clinical aspects and surgical treatment of hyperadrenocorticism in the domestic ferret: 94 cases (1994–1996). J Am Anim Hosp Assoc 33:487–493, 1997 129 Weiss CA, Williams BH, Scott JB, Scott MV: Surgical treatment and long-term outcome of ferrets with bilateral adrenal tumors or adrenal hyperplasia: 56 cases (1994–1997). J Am Vet Med Assoc 215:820–823, 1999 130 Wood JR, Nelson VL, Ho C, Jansen E, Wang CY, Urbanek M, McAllister JM, Mosselman S, Strauss

131

132

133 134

135

117

JF III: The molecular phenotype of polycystic ovary syndrome (PCOS) theca cells and new candidate PCOS genes defined by microarray analysis. J Biol Chem 278:26380–26390, 2003 Wu X, Arumugam R, Baker SP, Lee MM: Pubertal and adult Leydig cell function in Mu¨llerian inhibiting substance-deficient mice. Endocrinology 146:589–595, 2005 Yu RN, Ito M, Jameson JL: The murine Dax-1 promoter is stimulated by SF-1 (steroidogenic factor-1) and inhibited by COUP-TF (chicken ovalbumin upstream promoter-transcription factor) via a composite nuclear receptor-regulatory element. Mol Endocrinol 12:1010–1022, 1998 Yu RN, Ito M, Saunders TL, Camper SA, Jameson JL: Role of Ahch in gonadal development and gametogenesis. Nat Genet 20:353–357, 1998 Zazopoulos E, Lalli E, Stocco DM, Sassone-Corsi P: DNA binding and transcriptional repression by DAX-1 blocks steroidogenesis. Nature 390:311– 315, 1997 Zhao L, Bakke M, Hanley NA, Majdic G, Stallings NR, Jeyasuria P, Parker KL: Tissue-specific knockouts of steroidogenic factor 1. Mol Cell Endocrinol 215:89–94, 2004

Request reprints from Dr. D. B. Wilson, Department of Pediatrics, Box 8208, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110 (USA). E-mail: [email protected].

Downloaded from vet.sagepub.com by guest on July 29, 2010