DIRK VANDERSCHUEREN, LIESBETH VANDENPUT, STEVEN BOONEN, MARIE K. LINDBERG, ROGER BOUILLON, AND CLAES OHLSSON


Laboratory for Experimental Medicine and Endocrinology (D.V., L.V., S.B., R.B.), and Leuven University Centre for Metabolic Bone Diseases and Division of Geriatric Medicine (S.B.), Katholieke Universiteit Leuven, B-3000 Leuven, Belgium; and Division of Endocrinology, Department of Internal Medicine (M.K.L., C.O.), Sahlgrenska University Hospital, SE-41345 Goteborg, Sweden


C. Ohlsson, M.D., Ph.D., Division of Endocrinology, Department of Internal Medicine, Sahlgrenska University Hospital, SE-41345 Göteborg, Sweden; E-mail: Claes.Ohlsson@medic.gu.se.


Abstract

Loss of estrogens or androgens increases the rate of bone remodeling by removing restraining effects on osteoblasto- genesis and osteoclastogenesis, and also causes a focal imbalance between resorption and formation by prolonging the lifespan of osteoclasts and shortening the lifespan of osteoblasts. Conversely, androgens, as well as estrogens, maintain cancellous bone mass and integrity, regardless of age or sex. Although androgens, via the androgen receptor (AR), and estrogens, via the estrogen receptors (ERs), can exert these effects, their relative contribution remains uncertain. Recent studies suggest that androgen action on cancellous bone depends on (local) aromatization of androgens into estrogens. However, at least in rodents, androgen action on cancellous bone can be directly mediated via AR activation, even in the absence of ERs.


Androgens also increase cortical bone size via stimulation of both longitudinal and radial growth. First, androgens, like estrogens, have a biphasic effect on endochondral bone formation: at the start of puberty, sex steroids stimulate endochondral bone formation, whereas they induce epiphyseal closure at the end of puberty. Androgen action on the growth plate is, however, clearly mediated via aromatization in estrogens and interaction with ERα. Androgens increase radial growth, whereas estrogens decrease periosteal bone formation. This effect of androgens may be important because bone strength in males seems to be determined by relatively higher periosteal bone formation and, therefore, greater bone dimensions, relative to muscle mass at older age. Experiments in mice again suggest that both the AR and ERα pathways are involved in androgen action on radial bone growth. ERβ may mediate growth-limiting effects of estrogens in the female but does not seem to be involved in the regulation of bone size in males.


In conclusion, androgens may protect men against osteoporosis via maintenance of cancellous bone mass and expansion of cortical bone. Such androgen action on bone is mediated by the AR and ERα.


Abbreviations: AF, Activation function; ANDRKO, AR knockout; AR, androgen receptor; ArKO, aromatase knockout; BERKO, ERβ knockout; BMD, bone mineral density; cAIS, complete androgen insensitivity syn-drome; DERKO, double ER knockout; DHEA, dehydroepiandrosterone; DHEA-S, DHEA sulfate; DHT, 5α-dihydrotestosterone; DPA, dual photon absorptiometry; DXA, dual energy x-ray absorptiometry; E2, estradiol; ER, estrogen receptor; ERKO, ERα knockout; 17β-HSD, 17β-hy-droxysteroid dehydrogenase; IGFBP, IGF binding protein; IHH, isolated hypogonadotropic hypogonadism; KS, Klinefelter's syndrome; LBD, ligand-binding domain; OPG, osteoprotegerin; orch, orchidectomized or orchidectomy; ovx, ovariectomized or ovariectomy; PCOS, polycystic ovary syndrome; pQCT, peripheral QCT; QCT, quantitative computed tomography; RANK, receptor activator of nuclear factor KB; RANKL, RANK ligand; SERM, selective ER modulator; SPA, single photon absorp-tiometry; T, testosterone; Tfm, testicular feminized male.



Introduction


ANDROGENS INDUCE MALE sexual differentiation before birth and sexual maturation during puberty; in adult men, they maintain the function of the male genital system, including spermatogenesis. However, the role of androgens in other target organs-including muscle tissue; the cardiovascular, central nervous, and immune systems; and bone-is less well-established (1).


In the early 1940s, Albright and Reifenstein were among the first to refer to the antiosteoporotic and anabolic properties of androgens (2). From a public health perspective, osteoporosis is a greater problem in women than in men (3). This explains why most research efforts to explore skeletal effects of sex steroids have been devoted to estrogens. Moreover, androgens may be converted into estrogens via the P450 aromatase enzyme complex and may therefore act as prohormones for estrogens. In this respect, there is increasing evidence that at least part of the effects of androgens in men can be explained by their aromatization into estrogens (4, 5). Epiphyseal closure at the end of puberty, for example, is now generally accepted to be estrogen-dependent in both genders (6). In recent years, a specific role of androgens in skeletal homeostasis has even been questioned, although androgen receptors (ARs) in bone cells and AR-mediated actions on bone have been documented for more than a decade (7, 8).


The aim of this review is to address the question whether and how (through which receptors and/or pathways) an-drogens may affect bone strength and provide protection against osteoporosis. The clinical relevance of this question results from the recognition that, even in men, fractures due to skeletal fragility represent a huge public health problem.


Elderly men maintain cancellous bone integrity in com-parison with postmenopausal women, although their bone trabeculae become thinner. From a biomechanical perspective, compromised bone strength in men is the result of gender-related differences in the loss of bone mass during aging and, even more importantly, in the accumulation of bone mass during childhood and adolescence (9) (Table 1). During puberty, men develop a bigger bone size than women, due to increased periosteal apposition. In females, on the other hand, estrogens have an inhibitory effect on periosteal bone formation, whereas endocortical apposition is stimulated, narrowing the medullary cavity. Estrogens also stimulate epiphyseal closure earlier in women, resulting in longer bones in men. After puberty, the amount of bone formed on the periosteal surface is still greater in men (10, 11), whereas endocortical bone resorption is similar in both sexes, so that net bone loss is less in men (Table 1). The end result is a skeletal sexual dimorphism, characterized by a greater bone length, larger outer and inner bone perimeters, and a larger cortical volume in men compared with women. Therefore, adult men have greater bone mass than women, but this is due to a greater bone volume and not to a greater volumetric density. The greater areal bone mineral density (BMD) in males is thus only an artifact of the dual energy x-ray absorptiometry (DXA) software by expression of bone mass as projected areal (grams per square centimeter) instead of true or volumetric density (grams per cubic centimeter). In the current review, we will focus on the potential mechanisms through which androgens may prevent bone loss, increase bone mass (size), and improve bone strength.


In this manuscript, in vitro, experimental animal data and clinical human data with respect to skeletal androgen action will be reviewed in an attempt to define the possible impact of androgen action [through the estrogen receptor (ER) and AR pathways] on different aspects of skeletal homeostasis during growth and aging. Recent clinical and experimental data have indeed provided evidence that at least some of the skeletal androgen actions are not solely ER-dependent. Furthermore, we will discuss potential indirect nonbone cell- mediated effects of androgens on the skeleton.


Finally, the potential clinical benefits of androgen replace-ment in the context of male hypogonadism as well as in different patient groups will be discussed. Recent studies have explored different modes of action of androgens via the AR and ER pathways and may ultimately contribute to the potential use of selective AR or ER modulators in selected male target populations.


II.General Aspects of Androgen Action


A.Androgen metabolism

Androgens are C-19 steroids secreted primarily from the testes and the adrenals. The synthesis and metabolism of sex steroids have been extensively reviewed in several recent publications (4, 12). Therefore, we will just briefly mention the major pathways to facilitate the interpretation of experimental animal and clinical studies described in this review. The major gonadal androgen in males is testosterone (T), which is bound in the circulation to albumin and SHBG. It can be irreversibly converted in peripheral tissues by the enzyme 5α-reductase to the more potent 5α-dihydrotestos- terone (DHT). Both DHT and T can activate the AR (Fig. 1). T can also be converted to estradiol (E2) by an enzyme complex known as estrogen synthetase or aromatase followed by activation of the ERs. The adrenal cortex secretes large amounts of C-19 androgens including dehydroepiandros- terone (DHEA), DHEA-sulfate (DHEA-S), and androstenedi- one. These C-19 androgens can be metabolized either directly or indirectly in a rather complex pathway to estrone by the aromatase enzyme or to T by steroid sulfatase, 17β-hydroxy- steroid dehydrogenase (17β-HSD) and/or 3β-HSD (Fig. 1). Thus, depending on the relative activity of aromatase, 5α- reductase, 17β-HSD, 3β-HSD, and steroid sulfatase (13), T and C-19 androgens may predominantly activate either the AR or the ERs (Fig. 1). Several recent publications have demonstrated that aromatase (14 -25), 5α-reductase (14, 17, 18, 22, 26 -30), 17β-HSD (14 -17, 19, 23, 27, 31), 30-HSD (14, 32), and steroid sulfatase (13-15,17,19) are expressed in bone tissue, supporting the notion that local metabolism of androgens in bone tissue might be of physiological importance.


Figure 1. Simplified overview of the metabolism and action of sex steroids in men. 3β-HSD, 3β-Hydroxysteroid dehydrogenase. The sites of action of the different specific inhibitors of androgen and estrogen action, which are discussed in the present review, are indicated as follows: 1) aromatase inhibitor or aromatase-inactivated mice; 2) 5α-reductase inhibitor; 3) AR antagonist or AR-inactivated rodents; 4) ER antagonist; 5) ER α-inactivated mice; and 6) ERβ-inactivated mice.


Thus, sex steroids are partly synthesized locally in peripheral tissues, providing individual target tissues with the means to adjust formation and metabolism of sex steroids to their local requirements (Fig. 1). To elucidate the relative importance of androgen metabolism and action in bone, a variety of specific inhibitors/antagonists have been extensively used, including 5α-reductase inhibition (33-35), AR antagonists (36 -40), AR inactivation in rats (41) and mice (42-46), aromatase inhibitors (47-49), aromatase gene inactivation [in humans (50 -53) and mice (54 -56)], ER antagonists (36, 40, 57- 60), ERα gene inac-tivation [in humans (61) and mice (46,62- 67,69 -74)], and ERβ inactivation in mice (64- 67, 73, 75-81) (Fig. 1). A large part of this review will describe and discuss the results of these dif-ferent approaches to improving our understanding of the mechanism of action of androgens in bone tissue.


B.Mechanism of action of androgens

In this section, we will discuss the relative importance of the AR, ERα, and ERβ in mediating the effect of androgens on bone. The AR was cloned in 1988 (82,83). ERα was cloned in 1986 (84,85) and a second ER, ERβ, in 1995 (86). All of these receptors belong to the nuclear receptor family. They are all composed of three independent but interacting functional domains; the NH2-terminal or A/B domain, the C or DNA- binding domain, and the D/E/F or ligand-binding domain (LBD) (87) (Fig. 2). The sex steroid receptors are DNA-binding proteins that have the capacity to interact with specific DNA sequences, the androgen response element for the AR, and the estrogen response element for the ERs. The sequence homology in the DNA-binding domain is high among the sex steroid receptors (87). The N-terminal domain of these re-ceptors encodes a ligand-independent activation function (AF-1), a region of the receptor involved in protein-protein interactions, and transcriptional activation of target gene expression (87) (Fig. 2). The COOH-terminal region, or LBD, mediates ligand binding, receptor dimerization, nuclear translocation, and transcription of target gene expression (87) (Fig. 2). The classical mechanism of sex steroid action involves interaction with intracellular receptors, which are either cytoplasmic or nuclear. Binding of the sex steroids to their respective receptors leads to conformational changes of the protein that allow it to interact with the transcriptional machinery: directly or indirectly via protein-protein inter-actions with different transcription factors (88). The tran-scriptional activity of androgen-bound AR and estrogen- bound ERs is affected by tissue-specific coregulators, including factors enhancing transactivation (coactivators) and factors reducing transactivation (corepressors) (87, 89).


Figure 2. Diagramatic representation of the domain structure of nuclear receptors. The A/B domain at the NH2 terminus contains the AF-1 site where other transcription factors interact. The C/D domain contains the two-zinc finger structure that binds to DNA, and the E/F domain contains the ligand binding pocket as well as the AF-2 domain that directly contacts coactivator peptides. [Reproduced with permission from S. Nilsson et al.: Physiol Rev 81:1535-1565, 2001 (87).


C.Nongenomic effects of sex steroids

It was initially thought that the only mechanism for androgens/estrogens to affect transcription was by direct binding of activated AR to androgen response element or ERs to estrogen response element. But transcription can also be affected by protein-protein interactions with, for instance, the specificity protein-1, activation protein-1, and nuclear factor KB proteins (87). Furthermore, a variety of cell types respond to estrogens rapidly (within seconds/minutes), making a classical genomic mechanism of action unlikely (90). The importance of nongenomic mechanisms, in which the ligand interacts with plasma membrane/cytosolic receptors, is increasingly recognized to mediate the rapid responses to sex steroids (8, 91, 92). Nongenomic rapid effects of estrogens in vitro have been described for both osteoblasts and osteoclasts (8, 88, 91, 93). Furthermore, a plasma membrane ER is reported to partly mediate a nongenomic apo- ptotic effect of estrogens on osteoclasts (94).


The group of Manolagas (88) has demonstrated that the antiapoptotic effect of estrogens and androgens on osteoblasts in vitro is mediated by Src/Shc/ERK signaling via a nongenomic action of the classical receptors and is sex non-specific. This action is mediated by the LBD and is eliminated by nuclear targeting of the receptor protein (Fig. 3A). More recently, the same research group presented in vitro as well as in vivo data suggesting that the nongenomic effect of sex steroids involves kinase-mediated regulation of common transcription factors (95). Thus, nongenomic effects of sex steroids alter the activity of Elk-1, CCAAT enhancer binding protein-β (C/EBPβ), and cAMP-response element binding protein, or c-Jun/c-Fos by an extranuclear action of the ER or AR, resulting in activation of the Src/Shc/ERK pathway or down-regulation of c-Jun N-terminal kinase, respectively (95). Interestingly, a synthetic ligand, estren, which reproduces the nongenomic effects of sex steroids without affecting classical transcription, increases BMD in both ovariec- tomized (ovx) females and orchidectomized (orch) male mice, without affecting reproductive organs (uterus and seminal vesicles) (Fig. 3B). Such ligands merit investigation as potential therapeutic alternatives to hormone replacement for osteoporosis, in both women and men (88, 95, 96).


Figure 3. Nongenomic effects on BMD of the synthetic ligand estren. A, Model for ligand-induced dissociation of antiapoptotic from classical genomic activity of sex steroid receptors. The three diagrams depict conformational states of the receptor protein before and after interaction with the ligands, which are required to effect either the genomic (genotropic) or the antiapoptotic responses. The inactive unliganded receptor is depicted in the middle in gray. The change in conformation induced by interaction with a ligand that preferentially triggers transcriptional activity is depicted in the right in blue. The change in conformation induced by interaction with a ligand that preferentially triggers the antiapoptotic activity of the receptor (e.g., the estren) is depicted in the left in magenta. The green circle and green diamond represent the two ligands; please note the perfect and imperfect fit within the binding pocket, respectively. Ligands such as E2 will of course induce both conformations. Although in the antiapoptotic model we show direct contact between the receptor and Src, it is possible that adaptor protein(s) may bridge the interaction between the two molecules. Corresponding activation energy (Ea) of the receptor protein in the unliganded state (broken line), progressing to either the genotropic conformation (blue line) or the antiapoptotic conformation (magenta line), is shown at the bottom. B, Summary of the recent results presented by Kousteni et al. (96) and earlier studies (66, 70) regarding the effects of estrogens (E2), androgens (DHT), and the nongenomic acting synthetic ligand estren in gonadectomized mice. Estren reproduces the nongenomic effects of sex steroids without affecting classical transcription, and it increases BMD without affecting the reproductive organs. Thus, these results indicate that its effects on BMD are exerted via nongenomic effects, whereas the reproductive effects require genomic effects. I, Increase; NC, no change. [Panel A is reproduced with permission from S. Kousteni et al.: Cell 104:719-730, 2001 (88) with permission from Elsevier Science.].


Based on in vitro studies, Manolagas and co-workers (88, 97) have proposed that ERα, ERβ, and the AR can transmit the antiapoptotic effect of sex steroids with similar efficiency, irrespective of whether the ligand is an estrogen or an an-drogen. In contrast, however, several in vivo studies of trans-genic mouse models do not support the notion that estrogens have important AR-mediated physiological effects on can-cellous BMD or the concept that nonaromatizable androgens exert bone-sparing effects on cancellous BMD through direct activation of the ERs (42, 45, 66, 67, 70, 98) (see also Sections IV.E and IV.F). Although the hypothesis by Manolagas et al. that the bone-sparing effect of sex steroids is mediated via nongenomic mechanisms is extremely interesting and pro-vocative, additional investigation and confirmation by others are required before it can be fully accepted (99).


D.Expression of androgen and estrogen receptors in the skeleton

It is generally believed that an important part of the effect of androgens or their metabolites on the skeleton is exerted via a direct stimulation of the AR, ERα, and/or ERβ expressed locally in the skeleton. In this section, we will summarize in vitro and in vivo studies investigating the expression of these three receptors by growth plate chondrocytes, osteoblasts, osteocytes, osteoclasts, and/or by other bone- related cells.


1.Growth plate cartilage. Androgens exert important effects on pubertal growth, and a local effect on the growth plate is sup-ported by the fact that both cultured epiphyseal chondrocytes (100) and growth plate cartilage cells in vivo express AR (101-105) as detected by immunohistochemistry (101-105), binding studies (100), and in situ hybridization (105) (Table 2). The AR has been detected in all layers of the human growth plate at different ages (101-104), whereas in the rat it was expressed in proliferative and early hypertrophic chondrocytes at sexual maturation and only in prehypertrophic chondrocytes in older rats (105). Male rats displayed a higher AR expression in the growth plate and metaphyseal bone than female rats during sexual maturation (105). In contrast, no major sex difference regarding AR expression has been observed in human growth plate chondrocytes (100 -102).


Table 2. Expression of ARs and ERs in growth plate cartilage.


Several studies have detected ERα (102,104,106 -112) and ERβ (104,106,110,113) protein in the human, rabbit, and rat growth plate by using immunohistochemistry (Table 2), and the results regarding ERα have been confirmed by in situ hybridization (107). Most studies have detected ERα expression in all layers of the human and rabbit growth plate during both fetal stage and puberty (102, 104, 108, 110). In contrast, Kennedy et al. (111) detected ERα expression in the growth plate of immature but not mature rats. In addition, it is clear that the ERβ protein is expressed in the growth plate, but the expression pattern varies between studies. The first study by Nilsson et al. (113) localized ERβ mainly to the hypertrophic chondrocytes, whereas later studies have detected it in all layers of the growth plate (104, 106, 110) (Table 2). In con-clusion, AR, ERα, and ERβ are all expressed in the growth plate, indicating that androgens, either directly or after aro- matization, might influence the pubertal growth spurt and growth plate closure via a direct interaction with local sex steroid receptors. Future experiments, using growth plate- specific inactivation of the different sex steroid receptors, are needed to investigate whether these locally expressed recep-tors are of functional importance for the regulation of longi-tudinal bone growth.


2.Osteoblasts/osteocytes

Despite the obvious importance of androgens and estrogens in the regulation of adult bone metabolism, it has been difficult to detect AR and ERs in osteoblasts. Therefore, osteoblasts were for a long time not generally considered as primary target cells for sex steroids. The development of new and more sensitive techniques has resulted in the detection of the AR as well as ERα and ERβ expression by osteoblasts and osteocytes. The expression of the AR in cultured osteoblasts was first described in 1989 by Colvard et al. (114) using a nuclear binding assay. Several in vivo and in vitro studies have confirmed that the AR mRNA and protein are expressed by osteoblasts and osteocytes (14, 18, 101, 103, 105, 114- 122) (Table 3). AR binding has been demonstrated in vitro in osteosarcoma cell lines, osteoblast-like cell lines, and primary osteoblasts from several different species including human, rat, and mouse (18, 115, 118 -120, 123-126) (Table 3). The number of binding sites per cell appears to vary greatly from 70 to 14,000 binding sites per cell (127), depending on the assay technique, but it is in a range seen in other androgen target tissues. Human osteoblastic cells, isolated from cortical bone, expressed higher AR mRNA levels and AR binding than cells isolated from cancellous bone, whereas no major differences in AR expression in osteoblasts derived from males compared with osteoblasts derived from females were found (118). Most studies (116, 117, 119, 125), but not all (118, 128), indicate that androgen up-regulates the expression of its own receptor in osteoblasts.


Table 3. Expression of ARs and ERs in osteoblasts.


ER expression in bone cells was first reported in 1988 when specific binding sites for estrogens were identified in nuclear extracts from rat and human osteoblasts (129- 131) (Table 3). The number of estrogen binding sites in different studies varies between 60 and 4,500 binding sites per osteoblast, which is lower than in estrogen-responsive reproductive cells such as uterine and breast cells (132). Estrogens regulate osteoblast proliferation and expression of genes encoding enzymes, bone matrix proteins, hormone receptors, transcription factors, as well as growth factors and cytokines (133). However, conflicting results have been presented regarding the in vitro effects of estrogens on proliferation and differentiation of osteoblasts, which might be due to differences in the number of receptors per cell, animal species, skeletal localization of the osteoblasts, the stage of osteoblast differentiation, and the relative concentration of ERα vs. ERβ in the osteoblasts (132, 133). It is now well-established that both ERα and ERβ are expressed by osteoblasts and osteocytes, as studied both in vivo and in vitro; in several different species including human, rat, mouse, pig, and guinea pig; and using several different techniques including Western blot, Northern blot, RNase protection assay, PCR, in situ hybridization, and immunohistochemistry (Table 3). Some studies indicate that ERα expression increases with the increasing stage of differentiation of cultured osteoblasts (132, 134 -136). ERβ mRNA levels have been shown to either increase (135) or remain constant (134) with advancing cellular development. Thus, the ratio of ERα to ERβ and the estrogenic response may vary as the cells progress from preosteoblasts to mature osteoblasts (132). In a recent paper, the relative expression of ERα, ERβ, and the AR was followed simultaneously during differentiation of cultured osteoblasts (121), demonstrating that ERα levels were elevated during matrix maturation and then declined during mineralization. ERβ expression was rel-atively constant throughout differentiation, whereas AR levels were lowest during proliferation and then increased throughout differentiation, with highest levels in the most mature mineralizing cultures (121). One study in human developing bone demonstrated that ERα immunoreactiv- ity is strong in osteoblasts adjacent to the periosteal surface of the cortical bone, whereas ERβ immunoreactivity is dominant in osteoblasts in cancellous bone (106). Taken together, the AR and both ERα and ERβ are expressed by osteoblasts, but there is no consensus about their relative expression during differentiation and their localization within the skeleton.


3.Osteoclasts

AR expression has been detected in avian (137) and mouse (138) osteoclasts in vitro and in rat osteoclasts in vivo (17), whereas no expression has been detected in human osteoclasts in vivo (101, 103) (Table 4). Thus, the available data are contradictory regarding AR expression in osteoclasts, and it is generally believed that the major part of the effect of androgens on osteoclastogenesis and bone resorption is mediated by cells of the osteoblast lineage (139). However, some recent in vitro studies demonstrate that androgens can act directly on osteoclasts to promote their apoptosis (88, 96, 140). Furthermore, in bone marrow cell preparations, sex steroids have identical effects on osteoclas- togenesis in the presence or absence of cells of the osteoblastic lineage (140). An effect of estrogens, produced by aromatization of androgens, via ERs localized in osteoclasts is possible because most (106,141-151), but not all (107, 152, 153), studies have identified ERα and ERβ in osteoclasts (Table 4). In two separate studies, preosteoclasts, but not mature osteoclasts, were found to express ERα (154, 155). Taken together, the conflicting results regarding ER expression in osteoclasts suggest that osteoclasts express a low number of ERs, which may be close to the detection limit of the different assays used. This would be consistent with the fact that most but not all studies have detected ERs on os-teoclasts. Although the expression and the physiological role of sex steroid receptors in osteoclasts remain controversial, the available evidence suggests that the inhibitory effect of estrogens on osteoclastogenesis is largely mediated indirectly by cells of the osteoblast lineage, and not via a direct interaction with ERs on osteoclasts.


Table 4. Expression of ARs and ERs in osteoclasts.


4.Other bone-related cells

It is generally believed that osteoblasts originate from pluripotent mesenchymal stem cells in the bone marrow. Several studies have demonstrated that bone marrow stromal cells express the AR (122, 156) as well as both ERα (122, 154, 157-160) and ERß (122, 157, 158) (Table 5). Furthermore, both AR and ERs have been detected on megakaryocytes (151, 156) and endothelial cells (101, 151, 156) within the bone compart-ment. Thus, besides growth plate chondrocytes and os-teoblasts, it is clear that several other types of cells within the skeleton express sex steroid receptors, which may be involved in mediating the effect of androgens on the skeleton.


Table 5. Expression of ARs and ERs in other bone-related cells.



III.Effects of Androgens in Vitro on Skeletal Cells


The effect of androgens on skeletal growth and on adult bone metabolism is exerted via direct effects on the different types of cells located within the bone compartment. Indirect effects via muscle or vascular cells may also be operative.


A.Growth plate chondrocytes

Androgens probably have direct effects on growth plate cartilage and thus on longitudinal bone growth. It has been documented, at least when using very strict culture conditions, that androgens regulate both proliferation and differentiation of cultured epiphyseal chondrocytes (100,161-164), supporting a direct effect of androgens on growth plate cartilage. A direct effect of androgens on epiphyseal growth and maturation is also supported by the fact that T, injected directly into the growth plate of rats, increases the growth plate width (165). Androgens, however, also have an important effect on GH secretion and its pulsatility during puberty, and this may indirectly mediate their effects on linear growth (166).


B.Osteoblasts/osteocytes

Most in vitro studies (18,126,167-172), but not all (115,173, 174), demonstrate that both DHT and T increase cell prolif-eration of cultured osteoblast progenitors derived from dif-ferent species. The effects on osteoblast differentiation are rather controversial, including stimulatory, no effect, and inhibitory effect on alkaline phosphatase, type I collagen, osteocalcin, and mineralization of extracellular bone matrix (18,115,125,167,169,170,173-177). These conflicting results might be due to differences in receptor concentration, animal species, skeletal localization of the osteoblasts, or the stage of osteoblast differentiation. However, in our opinion, most studies indicate that androgens induce a more differentiated osteoblast phenotype. Recent studies demonstrate that an-drogens decrease osteoblast and osteocyte apoptosis (88, 96). Thus, most in vitro studies support the notion that androgens stimulate proliferation of osteoblast progenitors and differ-entiation of mature osteoblasts while inhibiting apoptosis of osteoblasts.


Some of the local effects of androgens on bone might, as previously described for estrogens (4), be mediated via a regulation of cytokines and growth factors expressed locally in bone. The three most discussed androgen-regulated locally expressed factors include TGFβ, IGFs, and IL-6. TGFβ and the IGFs are involved in bone formation, whereas IL-6 increases osteoclastogenesis and androgens may therefore theoretically preserve bone via either an induction of TGFβ and IGFs or an inhibition of IL-6. We will here discuss results regarding androgen-induced regulation of these factors, but it should be emphasized that the functional role of these regulations is complex and full interpretation is not yet possible. TGFβ is highly expressed in bone tissue (the largest reservoir for TGFβ), and it is a mitogen for osteoblasts (178, 179). Several studies, both in vivo and in vitro, indicate that androgens increase TGFS expression and/or activity (115, 169, 180, 181). Furthermore, orch reduces bone content of TGFS, whereas T treatment increases TGFS content (181). It remains unclear to what extent this effect of T is mediated through the AR or ERs. In contrast, Hofbauer et al. (173) found that androgens decrease TGFS mRNA levels in a human osteoblastic cell line. IGFs and IGF-binding proteins (IGFBPs) exert important effects on osteoblast proliferation and differentiation (182, 183). Androgens have been shown to regulate the expression and/or the activity of IGFs either directly by regulating IGF expression or indirectly via a regulation of the expression of IGFBPs in several (168,175,184), but not all, studies (18,185). IL-6 is a cytokine believed to be involved in the bone loss associated with sex steroid deficiency. It increases osteoclastogenesis and bone resorption. Orch increases IL-6 secretion by bone marrow cells (186). DHT and T suppress the IL-6 production in both cultured bone marrow stromal cells and osteoblasts (187-189). Furthermore, androgens inhibit the expression of the gp80 and the gp130 subunits of the IL-6 receptor (190). Orch increases osteoclastogenesis, which is inhibited by androgens or IL-6 neutralizing antibody (188), and IL-6 knockout mice do not lose bone after orch (188), supporting the notion that inhibition of IL-6 production is at least partly involved in the antiresorptive effect of androgens on bone. Thus, TGFS, IGFs, and IL-6 are three major factors believed to be involved in the bone-sparing effect of androgens. However, other possible pathways for androgen regulation of bone metabolism have also been investigated. Androgens inhibit PTH- or IL- 1-induced prostaglandin E2 production (176) and PTH-in- duced cAMP production (170, 191), whereas they increase IL-1S production (192) and the mitogenic effect of fibroblast growth factor (168) in cultured osteoblasts. A recently published study demonstrates that DHT decreases osteoprote- gerin (OPG) levels (193), whereas it has previously been shown that estrogens increased OPG expression in cultured osteoblasts (194, 195). It remains to be investigated in vivo whether DHT and estrogens have opposite effects on OPG expression and, if so, whether it is of any physiological importance for the regulation of bone homeostasis.


C.Osteoclasts

Osteoclasts are derived from hematopoietic precursor cells of the colony-forming unit granulocyte-macrophage lineage within the bone marrow. The proliferation of these colony-forming unit granulocyte macrophages is up-regulated after orch. The terminal differentiation into mature osteoclasts requires close interaction and also cell-to-cell contact with stromal cells of the osteoblastic lineage in the bone marrow under tight control of the receptor activator of nuclear factor KB-ligand (RANKL)/OPG system (196). The osteoblastic stromal cells also appear to be essential for the bone-sparing action of androgens. This notion is supported by the fact that in the SAMP6 mouse model, in which osteoblast function is impaired due to an age-related decrease in osteoblast pro-genitors (197), the rise in remodeling after orch is blunted. Their failure to up-regulate osteoclastogenesis is secondary to defective osteoblast formation. Thus, orch-induced increased osteoclastogenesis is dependent on osteoblast/ preosteoblast-derived signals (139). Therefore, the removal of testes-derived sex steroids by orch in mice primarily leads to bone marrow changes. These changes, mediated by cells of the osteoblast lineage, are characterized by an increase of osteoblast precursors, which in turn indirectly stimulates osteoclastogenesis (198). A direct effect of androgens on preosteoclasts/osteoclasts is more controversial. However, Pederson et al. (137) have demonstrated that osteoclasts express AR and that DHT inhibits the resorptive capacity of isolated human, murine, and avian osteoclasts in vitro. Furthermore, recent in vitro data suggest that androgens may also directly modulate RANKL-induced osteoclast formation, independently of the bone marrow cells (199). Additionally, androgens, like estrogens, may regulate osteoclast survival, RANK expression in preosteoclasts, and activity of mature osteoclast independently of their effects on bone marrow stromal cells, at least in vitro. However, the contribution of these in vitro observations to in vivo activity of androgens remains to be clarified. A direct effect of estrogens on oste-oclasts in vivo is supported by the finding that E2 promotes apoptosis of murine osteoclasts (200). These results indicate that osteoclast precursors as well as osteoclasts are able to respond directly to androgens in vitro and thus are potential androgen target cells in vivo (137). In conclusion, it is apparent that some of the effect of androgens on osteoclastogenesis is indirectly mediated via cells of the osteoblasts lineage, although further investigation is needed to characterize a possible direct effect of androgens on osteoclasts in vivo.


IV.Effects of Androgens on the Rodent Skeleton


A.The rodent as a model for the study of skeletal androgen action

The rat is the best-characterized animal model for the study of skeletal androgen action. The skeleton of young and mature rats is mainly dependent on modeling. This modeling process involves both growing and shaping of the bones. It is a highly synchronized process of bone formation at one site and resorption at another, with the former exceeding the latter. Longitudinal bone growth occurs through endochondral bone formation, whereas radial bone growth is the result of periosteal apposition. The expansion of the medullary cavity is a combination of endocortical bone resorption and formation.


Young rats have been widely used as a model for the growing skeleton. The skeleton of rats, though, differs from the human skeleton because the growth plates never fully close (201). This should not be overinterpreted because by 12 months of age, the growth plate characteristics have stabilized, with no further significant change up to 24 months (202). This allows aged rats to be used as a model for human skeletal remodeling. Bone remodeling maintains the mechanical and structural integrity of the skeleton after puberty. Coupling of osteoblast and osteoclast actions ensures that the processes of bone resorption and formation occur at the same time and place, which allows old bone to be replaced by new bone. It is also important to mention that rodents do not experience spontaneous fractures. Rodent studies will therefore not answer the question whether androgens protect against osteoporotic fractures, but they may still contribute to our knowledge of how androgens influence skeletal structure and density.


Several experimental procedures have been used to eval-uate skeletal androgen action and metabolism in male and female rodents. These include (surgical and chemical) castration and administration of AR antagonists, ER antag-onists, aromatase inhibitors, selective ER modulators (SERMs), and type II 5α-reductase inhibitors, either alone or in combination with sex steroid replacement. Because the skeletal effects of these experimental conditions often differ between cortical and cancellous bone compartments, these compartments will be considered separately. Some of these interventions may also induce extraskeletal effects-including changes in body composition, growth, and food intake- that may indirectly interfere with skeletal homeostasis (see Section V). Tables 6- 8 summarize the most important findings concerning androgen action in male and female rats, respectively.


More recently, mice have been introduced as a model to study skeletal androgen action. Mice with targeted disruption of the different receptors or enzymes involved in androgen action have been described, and their skeletal phenotype will be reviewed. At the end of each section, we will indicate to what extent these animal data may contribute to our understanding of how androgens affect skeletal structure and density.


B.Skeletal consequences of gonadectomy in rodents

1. Skeletal effects of gonadectomy in rats. Surgical castration, as induced by ovx in the female rat and orch in the male rat, represents the most frequently used procedure to study skel-etal sex steroid action. Chemical castration, as induced by GnRH agonists, has similar skeletal effects as surgical cas-tration in female rats (37), but its impact on bone has not been studied in male rats. Both orch and ovx dramatically reduce serum levels of T and E2 in male and female rats. However, these procedures do not totally eliminate E2 production, be-cause adrenal androgens can be transformed into estrogens after aromatization. In both genders, castration has consid-erable impact on cancellous and cortical bone compartments. However, whereas this response appears to be similar in cancellous bone, it is different in cortical bone.


Skeletal cancellous changes are characterized by an increase in cancellous bone turnover, resulting in bone loss in gonadectomized rats, irrespective of gender, age, or strain (203-211) (Table 6). Cancellous bone loss after gonadectomy can be detected not only by histomorphometry but also by peripheral quantitative computed tomography (pQCT) and microcomputed tomography. Biochemical markers of bone resorption (e.g., urinary deoxypyridinoline) and formation (e.g., serum osteocalcin) reflect the early increase of cancellous bone turnover after gonadectomy in both genders (40, 204, 209, 211-214). These changes in the cancellous bone compartment are reminiscent of high-turnover osteoporosis after menopause (215) and explain why the ovx female and the orch male rat model have gained wide acceptance as animal servations in postmenopausal osteoporosis in humans, the number of osteoclasts is increased after ovx (39,203,207,212, 217, 218) or orch (208, 210, 211, 219). Osteoblast number, surface, and mineral apposition rates are up-regulated in an attempt to fill the increased number of resorption cavities created by these osteoclasts (203, 208, 210, 211, 220). Although relative changes in histomorphometric and biochemical indices of bone resorption and formation indices cannot predict final outcomes on cancellous bone mineral content, orch-induced bone resorption tends to be increased more and longer than formation, even at the level of the individual remodeling unit (211, 220). Therefore, and despite increased bone formation at the tissue level, net cancellous bone loss occurs as a result of an imbalance with bone resorption exceeding bone formation in each bone mineral unit. The microanatomical mechanism responsible for this cancellous bone loss after orch in rats is a reduction in trabecular number (33, 211, 220) and thickness (211).


Table 6. Skeletal effects of orch and ovx in rats.


In contrast to the human skeleton, the rodent cortical skeleton has no Haversian canals, which explains why intracortical bone loss is not readily observed in ovx and orch rodents. Some studies have reported increased cortical porosity in older orch rats (205, 221), probably due to orch-induced medullary expansion through increased resorption at the endocortical site. In contrast to the changes in cancellous bone, the responses to castration at the periosteal site and growth plate are essentially sexually dimorphic, especially in young growing rats (Table 6). Periosteal and longitudinal bone formation appear to be increased in some (40, 57, 58, 222-224), but not all (203, 204, 207), studies on ovx and decreased in the orch model (207, 210, 213, 219, 224) (Fig. 4). The end result will be cortical bone expansion and increased bone length in the ovx female but decreased cortical bone volume and bone length in the orch male. This increase/ decrease in cortical bone volume after gonadectomy (in female and male rats, respectively) is primarily due to a relative increase/decrease in periosteal bone formation and not (or to a lesser extent) to significant changes of the inner endocor- tical perimeter (225). The implication is that sex hormone deficiency may have a more severe impact on the integrity of the male skeleton compared with the female skeleton.


Figure 4. A, The effect of ovx on periosteal bone formation rate. The mean ± SE (ver-tical bar) and tetracycline labeling period (horizontal line) for intact controls (O) and ovx (•) rats are shown as a function of time after ovx. P < 0.01 for all ovx time points compared with intact controls. B, The effect of orch on periosteal bone formation rate. The mean ± SE and tetracycline labeling period for intact controls (A) and orch (0) are shown as a function of time after orch. P < 0.01 for all orch time points compared with the same labeling period in intact controls. [Reproduced from R. T. Turner et al.: J Orthop Res 8:612-617, 1990 (234) with permission from Orthopaedic Research Society.].


Gender-related differences in the response of periosteal bone to gonadectomy also explain why methods that measure areal rather than volumetric bone density, such as DXA, tend to overestimate bone loss in the orch male while underesti-mating bone loss in the ovx female. Moreover, cortical bone loss in the orch rat model, in contrast to changes in cancellous bone, is due to failure to gain new bone and not to net bone loss. These gender-specific responses in cortical bone volume are particularly important during active growth but less sig-nificant in elderly, slow-growing rats (209, 220) (Table 6). In older rats, longitudinal bone growth does not decrease after orch (205, 206, 209).


As summarized in Table 6, castration induces postmeno-pausal-like bone loss in the cancellous bone compartment in both genders, irrespective of age or strain of the rat. The intracortical bone compartment is relatively resistant to sur-gical castration, whereas the responses at the periosteal site and the growth plate appear to be sexually dimorphic, es-pecially in younger rats, with androgens stimulating and estrogens inhibiting periosteal bone expansion.


2.Skeletal effects of gonadectomy in mice. Reduced cortical bone growth, as described in rats, has also been observed in male orch mice (70, 226, 227). Female mice, in contrast to rats, do not experience significant cortical bone expansion after ovx (228, 229).


In cancellous bone, both ovx (228 -231) and orch (66, 70, 188, 227, 231) induce bone loss. This cancellous bone loss is associated with elevated markers of bone turnover (70, 229), and ovx and orch mice are therefore increasingly being ac-cepted as animal models for the study of steroid action in postmenopausal-like osteoporosis (139).


C.Skeletal effects of androgen replacement in rodents 1.

Skeletal effects of androgen replacement in rats a. Effects of aromatizable and nonaromatizable androgens.

The aromatizable androgen T is a very effective bone-sparing agent. T not only fully prevents cancellous bone loss in orch rats (208, 209, 221) but is also bone-sparing in ovx rats (232), even at subphysiological concentrations (233) (Fig. 5) and irrespective of age. In addition, T antagonizes periosteal expansion in ovx rats (234), but increases periosteal bone formation in orch rats (209, 221, 234) (Table 7). Weaker aro- matizable androgens, such as DHEA (223, 235) and andro- stenedione (218, 236), are evidently bone-sparing in the ovx rat model but have not been studied in male orch rats (Table 7). In the ovx rat model, bone protection by these androgens is exerted through the AR and not the ERs, as illustrated by the fact that the effect is blunted by concomitant administration of AR antagonists but not by aromatase inhibitors or ER antagonists (218, 235, 236).


Table 7. Skeletal effects of sex steroids or sex steroid-related agents in orch and ovx rats.


Figure 5. In the aged orch male rat model, subphysiological T replacement, which only partially prevents atrophy of ventral prostate and seminal vesicles, is already bone-sparing (233). In contrast, only su- praphysiological doses of DHT, resulting in hypertrophy of androgensensitive organs, prevent cancellous bone loss (237). The difference in bone-sparing capacity between T and DHT may be related to the fact that T can be aromatized in bone and activates one or both ERs.


The nonaromatizable androgen DHT also exerts bone-sparing action in orch (208, 209, 234, 237) and ovx rats (235, 238, 239) (Table 7). However, DHT appears to be less effective than T in the elderly orch rat model (208, 237), in particular on cortical bone (Fig. 5). In this model, higher doses of DHT than T are indeed needed to obtain at least some bone- protective action on cancellous bone, but this occurs at the expense of side effects like hypertrophy of the ventral prostate and seminal vesicles. Moreover, high-dose DHT fails to prevent orch-induced cortical thinning (237).


In conclusion, in both ovx female and orch male rats, aromatizable and nonaromatizable androgens show bone- protective action, especially at cancellous bone sites. Non- aromatizable androgens seem to be less effective than aro- matizable androgens. A possible explanation for the relative lack of efficacy of nonaromatizable androgens compared with aromatizable androgens may be the aromatization of the latter via estrogens and stimulation of the ERs.


b. Effects of estrogens. The bone-sparing effects of aroma- tizable androgens may depend on activation of the AR, the ERs, or both. Estrogens (including phytoestrogens) have well-documented bone-sparing effects, not only in ovx rats (204, 232, 235, 239 -242) but also in orch rats (209, 213, 237) (Table 7).


Overall, in gonadectomized rat models, aromatizable and nonaromatizable androgens and estrogens (Table 7) appear to be bone-sparing in the cancellous bone compartment, ir-respective of gender or age. Table 7 also indicates that sex steroid action on cortical bone is not only less well docu-mented but also less consistent. Both androgens and low- dose estrogens tend to stimulate cortical bone, however only in male rodents.


2.Skeletal effects of sex steroids in mice. T effectively prevents cancellous bone loss in orch mice (70, 188). DHT (96) and estrogens (66,70,96,228,231) also appear to be bone-sparing after castration, in both genders. Similar bone-sparing effects have been observed with phytoestrogens in ovx (243) and orch mice (244). So, in accordance with experiments in rats, androgens and estrogens both protect against cancellous bone loss in mice, irrespective of gender. Additionally, T and estrogens increase the cortical area in orch mice (226, 245).


D.Skeletal effects of selective manipulation of estrogen and androgen action in rodents

1. Effects of selective pharmacological modulation in rats. Ad-ministration of AR antagonists, ER antagonists, SERMs, and aromatase inhibitors will selectively interfere with the AR, the ERs, and the aromatization of androgens into estrogens, respectively (Fig. 1).


Aromatase inhibitors impair both skeletal development (cortical expansion) and maintenance (integrity of the can-cellous compartment) in male rats (47, 48) (Table 8). The bone-phenotypic changes induced by administration of an aromatase inhibitor are thus similar to those observed after orch, although bone turnover seems to be less elevated. Moreover, E2 prevents bone loss induced by an aromatase inhibitor in male rats, supporting the concept that pharma-cological administration of estrogen is protective for male bone (49).


Table 8. Skeletal effects of selective pharmacological modulation of androgen and estrogen action in intact male and female rats.


Administration of a selective ER antagonist, ICI 182,780, which induces cancellous bone loss in female intact and estrogen-repleted ovx rats (40, 57, 58), does not impair skeletal homeostasis in T-supplemented orch rats (59) or intact male rats (60) (Table 8). Yet, SERMs, some of which are being used in the treatment of postmenopausal osteoporosis, have been reported to restore ovx-induced bone loss in female rats (246, 247) (Table 7) as well as orch-induced (Table 7) and age-related bone loss (Table 8) in male rats (248, 249). Similarly, tibolone, a drug with mixed androgenic, estrogenic, and progestogenic properties, prevents bone loss in ovx rats (212) (Table 7). The bone-sparing effect of tibolone is reversed by concomitant administration of an antiestrogen but not by an antiprogestogen or antiandrogen, suggesting that bone protection by tibilone after ovx occurs through the ER path-way (232).


The skeletal effects of AR antagonists in rats are not con-sistent (Table 8). In older studies, AR antagonists like flut-amide or cyproterone acetate were found to induce bone loss in rats, irrespective of gender; however, in these studies, bone mass was only assessed by bone calcium content or kinetics (36, 37). Several histomorphometric studies have reported significantly reduced bone formation or an increase in bone resorption after AR antagonist administration, but without concomitant bone loss (38 -40). Confirmation of AR antag-onist-induced osteopenia by methods such as histomor- phometry is therefore required. Finally, finasteride, a type II 5α-reductase inhibitor that blocks the conversion of T into DHT, does not interfere with skeletal homeostasis in male rats (33) (Table 8).


In conclusion, selective modulation of the AR and ER pathways generally supports the concept of a dual mode of androgen action (through both the AR and ER pathways), at least in cancellous bone, both in male and female rat models (Table 8). Effects of AR antagonists in rats, on the other hand, are less well-established.


2. Effects of selective pharmacological modulation in mice. In intact male mice, the AR antagonist casodex does not affect bone density, whereas another AR antagonist, epitestosterone, which is also a 5α-reductase inhibitor, decreases bone density (250). The AR antagonist cyproterone acetate prevents orch- induced bone loss, suggesting that this agent may act as an AR agonist in bone (251). SERMs also prevent orch-induced (227, 231) and ovx-induced bone loss (231, 252) in mice. Overall, these studies (although limited in number) provide further evidence for a similar action of sex steroids in cancellous bone. Along these lines, the activator of nongenomic estrogen-like signaling, estren, has a similar (or even greater) anabolic effect on (especially cortical) bone than DHT in male orch mice or than E2 in female ovx mice, despite a much lower affinity for the ER (96) (see also Section II). These effects of estren, although obtained with a much higher dose than with E2 and DHT, occur without stimulation of reproductive organs in either sex.


E.Skeletal effects of selective manipulation of androgen and estrogen action in transgenic mice

1. Description of transgenic animal strains. The cyp19 aromatase gene has been inactivated [aromatase knockout (ArKO) mice] by two independent groups (253, 254), and a similar skeletal phenotype has been reported (54 -56). ERβ has been inactivated by three independent groups [ERβ knockout (BERKO) mice] (79, 255, 256), and the skeletal phenotypes of all three mouse strains are rather similar (64 - 67, 73, 75- 81, 98). Two independent groups have inactivated ERα [ERα knockout (ERKO) mice] (256, 257). The first and most studied ERKO mouse strain was generated in the laboratories of Korach and Smithies (257). A recent study indicates that these mice are not completely ERα-inactivated (258), as supported by the observation that they express one or two N- terminally modified ERα transcripts associated with minor ER activity regarding uterine weight and endothelial nitric oxide production (258). The remaining ERα activity is suggested to be mediated via a truncated ERα with remaining AF-2 activity, whereas there is no AF-1 activity left (258). This truncated ERα isoform has been detected in bone of these mice and has been shown to have effects on gene transcription in cultured human osteoblasts (259). These mice will be referred to as ERKOAF-W~ because they do not have any remaining AF-1. The second ERα-inactivated mouse model was developed in the laboratory of Chambon (256). There is no remaining ERα activity in these mice, and both AF-1 and AF-2 activity are absent in these animals, which will be referred to as ERKOAF-1/AF-2-/_. The skeletal phenotype has been reported for both the ERKOAF-W- mice (62-67,69,260, 261) and the ERKOAF-1/AF-2-/- mice (46, 71-74, 98). Most skeletal phenotypes (including the male skeletal phenotype and the skeletal responses to estrogen treatment in female ovx mice) are identical for the ERKOAF-W- (66, 67, 70) and the ERKOaf-1/af-2-/- mice (72, 73). However, a clear difference between the two ERKO models is seen in female gonadal intact mice (see Section IV.E., 2 and 3). Possible factors that might explain the differences between the two ovarian intact female ERKO models include: 1) differences in genetic background; 2) differences in dietary/environmental estrogen; 3) differences in the feedback regulation of sex steroids between the ERKOAF-W- and ERKOAF-1/AF-2-/_ mice, in which both E2 and T are dramatically increased but the magnitude of this disturbance might differ between the two ERKO models; 4) remaining AF-2 activity in ERKOaf-w- mice; and 5) other unknown reasons.


Mice with inactivated ERα and ERβ [double ER knockout (DERKO)], developed in two independent laboratories, have been reported to have rather similar skeletal phenotypes, both in intact mice and in estrogen-treated gonadectomized mice (64 - 67, 72,73,98). AR knockout mice (ANDRKO mice) have recently been developed by two independent groups using the cre/loxP system (42-44), and their skeletal phenotypes have recently been described (42-44, 262, 263).


It is of importance to note that the sex steroid levels (both E2 and T) are increased in ERKO and DERKO, due to disturbed feedback regulation (65, 73,264). In contrast, the skeletal phenotype of single ERβ inactivation is not influenced by altered sex steroids levels. By comparing the skeletal phe-notype of wild-type, ArKO, BERKO, ERKO, DERKO, and ANDRKO mouse strains, it may be possible to gain new insight in the relative importance of the AR, ERα, and ERβ in mediating the skeletal effects of androgens. The different transgenic mouse strains described in this section are inac-tivated from birth, which results in disturbed skeletal growth and maturation. Thus, the adult skeletal phenotype in these mice is a combination of effects on growth/maturation and adult bone remodeling. Future transgenic mouse models with inducible gene inactivation will be useful in separating the effects on growth/maturation from the effects on adult bone remodeling and will therefore be more relevant to further explore the protective effect of sex steroids on the adult skeleton. Nevertheless, the currently available transgenic mouse models have already been very informative. In the following section, the effects on longitudinal appendicular skeletal growth as well as the effects on cortical and cancellous bone in these mice will be discussed.


2. Longitudinal appendicular skeletal growth

a.Females. ERβ-inactivated female mice develop increased femoral length after sexual maturation (65, 73, 75, 78). Ac-tually, the normal length difference between males and fe-males is not observed in young adult female BERKO mice (65, 75, 76, 78) (Fig. 6A). An altered length of femur is often associated with a disturbed GH-IGF-axis (265-268). Interest-ingly, serum levels of IGF-I are increased in young adult female BERKO mice and correlate to the effect on femoral length, which might indicate that ERβ is an inhibitor of the GH-IGF-I axis (65). However, GH secretion has not yet been studied in these mice. Several independent studies using female ERKOaf-1-/- mice have demonstrated that these mice have a reduced length of the femur, which develops during sexual maturation and is still present at 18 months of age (62,63,65,260). In contrast, in one recent study in female ERKOaf-1/af-2-/ - mice, no effect on femoral length was seen at 16 wk of age (73). The length of the femur is largely unchanged in adult female ArKO (54) and DERKO (65, 73) mice. In summary, ERβ represses longitudinal appendicular skeletal growth during sexual maturation in female mice. Some (but not all studies) in ERKO models indicate that ERα may stimulate appendicular skeletal growth in female mice. Finally, a normal appendicular skeletal growth is observed in the absence of the opposing effects of ERα and ERβ activation as seen in female ArKO and DERKO mice.


Figure 6. Increased length (A) and increased cortical cross-sectional area (B) of the femur in young adult female but not male BERKO mice (open bars) compared with wild-type mice (filled bars). Thus, the skeletal sexual dimorphism is diminished in the ERβ -/- mice compared with wild-type mice. LOW, Low BMD; HIGH, high BMD. [Panel A is reproduced with permission from S. Windahl et al.: J Clin Invest 104:895-901, 1999 (75).]


b.Males. The longitudinal appendicular skeletal growth is unchanged in male BERKO mice (64, 73, 75) (Fig. 6A), although several independent studies using male ERKOaf-1-/- mice have clearly demonstrated that these mice have a reduced appendicular skeletal growth during sexual maturation (62-64). The decreased appendicular skeletal growth of these mice is associated with decreased serum levels of IGF-I (64). The appendicular growth of the male ERKOaf-1/af-2-/- mice has been analyzed by two independent groups with conflicting results, demonstrating unchanged growth in one study (73) and, similar to all the ERKOaf-1-/- studies, decreased growth in another study (46). Thus, most available data indicate that the appendicular growth is decreased in male ERKO mice. Femoral length is also decreased in male ArKO (54) and DERKO (65,73) mice, whereas bone length is reported to be unchanged in male ANDRKO mice (263). In summary, ER activation, but not AR activation, is involved in the regulation of male longitudinal appendicular skeletal growth in mice. Most studies indicate that the effect of estrogens is mediated via ERα and not via ERβ (46, 62- 64, 75) (Fig. 6).


3.Cortical bone

a. Females. Studies in two different BERKO mouse strains have demonstrated an increase in cortical cross-sectional bone area due to increased radial cortical bone growth (Fig. 6B). This effect is seen after sexual maturation (65, 75, 78, 81). In contrast, no significant effect on cortical bone parameters was found in female BERKO mice, developed in a third laboratory (73). In line with the effects on appendicular skeletal growth, the effect on cortical thickness differs dramatically between the gonadal intact female ERKOaf-1-/- mouse model (65), a mouse model with increased thickness, and the gonadal intact female ERKOaf-1/af-2_/_ (73) with decreased thickness. The cortical thickness is similar in the female ERKO and DERKO mice. Female ArKO mice have a decreased cortical crosssectional bone area (56). In summary, it is clear that ERβ activation decreases cortical thickness in female mice and that estrogen deficiency, due to aromatase inactivation, results in a decreased cortical area in female mice. The role of ERα in the regulation of cortical bone in gonadal intact female mice is unclear, because conflicting results have been presented.


b. Males. Cortical bone parameters reflecting the size of the cortical bone, including periosteal circumference, endocor- tical circumference, cortical thickness, and cortical cross-sectional area, are decreased in male ERKO (64, 70, 73), DERKO (64, 73) (Fig. 7), and ArKO mice (56). The decreased amount of cortical bone is associated with a reduced periosteal apposition rate (73). In contrast, the cortical bone parameters are unchanged in male BERKO mice (64, 73, 75). Recent data indicate that male ANDRKO mice have a decreased cortical bone area (44,263). In summary, ERα and AR but not ERβ enhance cortical radial bone growth in male mice.


Figure 7. Effects ofER inactivation on skeletal growth in gonadal intact male mice. Representative DXA scans (A) and middiaphyseal pQCT scans (B) of femora in young adult wild-type, ERKO, BERKO, and DERKO mice. LOW, Low BMD; HIGH, high BMD. Femoral length (A), a measure of appendicular growth, and cross-sectional area (B) are significantly reduced in ERKO and DERKO mice. [Reproduced with permission from O. Vidal et al.: Proc Natl Acad Sci USA 97: 5474-5479, 2000 (64). © National Academy of Sciences, USA.].


4.Cancellous bone in gonadal intact mice

a. Females. Surprisingly, female BERKO mice are protected against age-related cancellous bone loss (73, 76, 78, 80). This finding indicates that ERβ, in the presence of low age-related estrogen levels, might act as a competitor for the stimulatory effect of ERα on cancellous bone in old female mice. An inhibitory role of ERβ in the presence of ERα is supported by the observation that ERβ reduces ERα-regulated gene transcription (69). However, the estrogenic response to pharma-cological treatment with estrogen in gonadal intact female BERKO mice is not significantly altered compared with wild-type mice (81). Unexpectedly, female gonadal intact ERKO mice have an increased amount of cancellous bone (65, 73), probably caused by increased serum levels of T acting via the AR (98). Furthermore, female ERKO mice have masculinized livers, which also might affect bone mass accrual through circulating IGF-I (269). Female gonadal intact DERKO (65,73) and ArKO mice (54 -56) have a decreased amount of cancellous bone. These findings indicate that ER activation as well as AR activation has the capacity to preserve the amount of cancellous bone in gonadal intact female mice.


b. Males. Cancellous bone is unaffected in male BERKO mice (64, 73, 75, 76). Unexpectedly, the amount of cancellous bone is increased, associated with reduced bone turnover in adult gonadal intact male ERKO and DERKO mice (66, 70, 73). The increased cancellous bone mass is caused by elevated T levels acting via the AR, because treatment with an antiandrogen results in cancellous bone loss in the gonadal intact male ERKO mice (66, 73, 98). Male ArKO mice (55, 56) and ANDRKO mice (42-44, 263) have reduced cancellous bone mass. In the ANDRKO model, this osteopenia is associated with an increased bone turnover (42-44,263). Regarding male ArKO mice, one study has reported increased bone resorption (56), whereas another group has documented decreased bone turnover markers (both formation and resorption) (55). Taken together, these data suggest that the AR but not ERβ is required for the maintenance of cancellous bone mass in males. The specific role of ERα is difficult to determine using the gonadal intact male ERKO mice because their T levels are increased. Indeed, all studies on gonadal intact animals with mutated sex steroid receptors have to be considered carefully because endogenous sex steroid levels might be modified.


5.Effect of sex steroids on cancellous bone in gonadectomized mice. In the experiments described in this section, adult mice of the different genotypes were gonadectomized, and the effects of E2 (activating the ERs), T (potentially activating both the AR and the ERs) or DHT (activating the AR) on cancellous bone were investigated.


a. Females. Ovx-induced cancellous bone loss in BERKO mice is prevented by treatment with either E2 or DHT (Refs. 67, 72, 98; and M. K. Lindberg, and C. Ohlsson, unpublished personal data). In contrast, physiological E2 replacement therapy exerts no effects (67) or minor effects (72, 98) on cancellous bone in ovx ERKO mice. However, pharmacological treatment of ovx ERKO, reaching very high concentrations of E2, results in an increased amount of cancellous bone (98). The amount of cancellous bone is not increased by physiological E2 treatment in ovx DERKO mice (67, 98), whereas this treatment restores the cancellous bone in female ArKO mice (56). Both DHT and T increase the cancellous bone mass in ovx ERKO and DERKO mice (Ref. 98; and M. K. Lindberg, and C. Ohlsson, unpublished personal data). These findings seem to indicate that the cancellous bone-sparing effect of physiological levels of estrogens in ovx mice is mainly mediated via ERα. AR activation has the capacity to increase cancellous bone mass in ovx mice, but the physiological role of this effect is unclear (Refs. 96 and 98; and M. K. Lindberg, and C. Ohlsson, unpublished personal data).


b. Males. Orch-induced cancellous bone loss is prevented by either E2 or DHT in BERKO mice and by either T or DHT, but not by E2, in ERKO and DERKO mice, demonstrating that (in contrast to ERβ activation) ERα and AR have the capacity to increase the amount of cancellous bone in male mice (66, 70, 98, 245). T (in contrast to DHT) increases the amount of cancellous bone in male gona- dectomized ANDRKO mice, supporting the concept that ER activation preserves the cancellous bone in the absence of a functional AR (42, 262, 263). These findings suggest that the AR and ERα, but not ERβ, can independently mediate the cancellous bone-sparing effects of sex steroids in male mice. 6. Conclusion of studies in sex steroid-related transgenic mouse models. Both ERα and the AR are involved in mediating the effects of androgens on cortical cross-sectional area and adult cancellous bone remodeling, although only ERα regulates longitudinal appendicular growth in male mice (Tables 9 and 10). ERβ, however, is not involved in the regulation of the male skeleton. In contrast, both ERα and ERβ are of importance for the regulation of the female skeleton. The ERα is the main receptor responsible for the cancellous bone-sparing effect of physiological levels of estrogens in female mice. However, ERβ activation suppresses the cancellous bone mass in old female mice, indicating that ERβ suppresses the cancellous bone-sparing effect of ERα in female mice with low serum levels of estrogens. The AR has the capacity to preserve cancellous bone in female mice, but its physiological role in female bone metabolism is unclear.


Table 10. Summary of the role of ERα, ERβ, and AR activation on skeletal parameters in mice with different sex steroid-related gene inactivations.


F.Skeletal effects of androgen resistance in rodents

1. Androgen resistance in rats. Experiments in androgen- resistant testicular feminized male (Tfm) rats provide further support for the concept of a dual mode of action of sex steroids. Tfm rats are unresponsive to androgens due to a single base mutation in the LBD of the AR gene. These genotypic male but phenotypic female rats have a more female-like (cortical) bone structure (270). In contrast to orch rats, they do not experience spontaneous cancellous bone loss. Vanderschueren et al. (41) have hypothesized that bone loss in these Tfm rats is prevented by the increased serum levels of estrogens. Indeed, after orch, Tfm rats lose bone, suggesting the importance of T as a precursor for estrogens in the protection of bone (41). Again, androgen-induced extraskeletal effects may have important indirect skeletal consequences in these rats. In the Tfm rat, GH secretion follows a more female-like pattern because of lack of neonatal androgenization (270) (see also Section V). This female-like GH profile may explain, at least partly, the more female type of bone structure. Moreover, high estrogen concentrations in Tfm rats may further inhibit growth.


The hypothesis that androgens may prevent cancellous bone loss via both AR and ER pathways is thus supported by experiments in the Tfm rat model. According to the Tfm phenotype, cortical bone structure depends on a functional AR. In Tfm rats, however, female-like GH secretion and an increased degree of aromatization may be confounding factors creating a growth-inhibitory environment.


2. Androgen resistance in mice. Tfm mice, on the other hand, have a high-turnover cancellous bone phenotype (226), similar to the phenotype observed in the ANDRKO mouse model (42, 43, 262). In line with observations in ANDRKO mice, Tfm mice have low levels of T and E2 (as opposed to the high levels of T and E2 in the Tfm rat), which may confound both models. Sex steroid replacement in orch Tfm mice allows further investigation of the role of the AR in mediating T action in this model. In orch Tfm mice, cortical bone appears to be completely unresponsive to T action, whereas cancellous bone is preserved by T (226) (Table 9). Again, these data suggest that T action on cortical bone is at least partly AR-dependent, whereas T action on cancellous bone may depend on both AR and ER activation.


Table 9. Summary of the skeletal phenotypes in mice with different sex steroid-related gene inactivations


G.Animal data in support of a dual mode of androgen action on the skeleton

Androgens (both aromatizable and nonaromatizable) are able to maintain skeletal integrity of the cancellous compart-ment in rodents via the AR, irrespective of age and gender. Estrogens also prevent cancellous bone loss (through acti-vation of the ERα only) in male rodents, even at low con-centrations. This has led to our concept of a dual mode of action of androgens on bone via both ERα and the AR, although the relevance of ERβ activation remains uncertain. In addition, androgens are able to stimulate bone formation at the outer periosteal bone surface, at least in male rodents and during skeletal growth. Again, in this process of androgen-mediated periosteal bone formation, both the AR and ERα are involved. In female rodents, on the other hand, both ERα and ERβ are of importance for skeletal development and maintenance.


V.Indirect Mechanisms of Action of Androgens with Skeletal Implications


Sex steroids may differentially and sex-specifically regulate longitudinal and radial bone growth in rodents, leading to the typical sexually dimorphic skeletal changes (longer and broader bones in males) at the end of puberty (see Section IV). These opposite effects of androgens and estrogens may depend on stimulation of sex steroid receptors expressed locally in the skeleton (see Section II.D). Alternatively, androgens may also indirectly affect skeletal homeostasis through interactions with body growth, body composition, and the GH-IGF-I axis.


A Androgens, body growth, and body composition

Androgen-induced changes in growth and body com-position may have an important impact on skeletal changes: in growing rats, gain in body weight (associated with increased food intake) is impaired after orch in most (33,47, 205,207,219,224,271), but not all studies (208,210, 234), whereas the opposite is observed in the ovx rat (57, 58, 203, 204, 271, 272) (Table 6). These opposite changes in body weight are fully prevented by androgens in male and by estrogens in female gonadectomized growing rodents (Table 7). It is important to emphasize that body weight remains mostly unchanged in older gonadectomized rats (205, 206, 221, 272) (Table 6). Male rats also lose muscle mass and gain fat mass after orch (233, 237). It is therefore tempting to speculate that orch-induced muscle loss may lower mechanical strain. Decreases of longitudinal and radial bone growth after orch might therefore essentially represent an adaptation of skeletal modeling during growth to orch-induced changes in mechanical strain rather than a direct action of sex steroids on bone. The effects of gonadectomy-induced changes in body weight and composition on skeletal modeling and remodeling remain hypothetical but can and should be further explored. Conversely, increased skeletal growth in response to androgens (Table 7) might reflect increased mechanical loading. In line with this assumption, actions of T on bone and lean body mass (a surrogate for muscle mass) seem to be closely interrelated, at least in orch rodent models (233).


In addition, several links exist between adipocytes and bone. There is increasing evidence for some adipocytokines, such as leptin, to be the mediators via the central nervous system, in particular the nucleus paraventricularis (273). The communication between cells of the immune system occurs via very similar cytokines and receptors (e.g., RANK- RANKL-OPG) as the ones used by the different bone cells. Moreover, there are several arguments for a direct interaction between immune cells (e.g., B and T lymphocytes) and bone cells (274). Because sex steroids have a direct influence on immune cells (androgens and estrogens influence hemato-poiesis and notably B lymphopoiesis), indirect sex steroid effects on bone via immune cells might be plausible but need further exploration.


In humans, androgens stimulate longitudinal growth in the male, whereas estrogens appear to have a biphasic effect in both sexes (6). Low-dose estrogens stimulate the pubertal growth spurt, but higher concentrations inhibit linear growth and promote growth plate closure. Males may therefore be taller and have a higher peak bone mass because they are exposed to relatively lower estrogen concentrations during a longer prepubertal and pubertal period. Although it is assumed that sex steroids have similar actions on radial as on longitudinal growth, this remains to be established in humans.


Puberty in males is characterized not only by increased longitudinal growth but also by a greater gain in muscle mass compared with females (275). Increased muscle mass may result in enhanced mechanical loading, which is considered to be the most important stimulus for skeletal modeling (276). It is therefore tempting to speculate that changes in body composition (more muscle, less fat) contribute to greater bone size in men. However, the extent to which androgens increase muscle and thereby indirectly stimulate skeletal modeling remains unknown. Although T supplementation stimulates fat-free mass and muscle size in hy- pogonadal men (277), the skeletal implications of this type of anabolic response remain to be established. In vitro data support the concept that mechanical strain and sex steroids might impact on bone through similar modes of action. ERα has indeed been shown to mediate the strain-related proliferation of osteoblasts of both sexes (278, 279), but the relevance of this interaction between mechanical strain and sex steroids in vivo remains unclear.


B. Androgens and the GH-IGF-I axis

Postnatal growth is primarily regulated by the GH-IGF-I axis. Inhibition of IGF-I action- either directly (280, 281), or indirectly via disruption of the GH receptor (282, 283)- dramatically decreases growth rate in male and female mice, similar to the growth delay observed in GH-deficient or GH-resistant humans (265). This decrease in growth rate affects cortical bone (280, 283) but has no effect on cancellous bone mass (280,283,284). Interaction with the GH-IGF-I axis may therefore provide yet another indirect mechanism by which sex steroids regulate sexually dimorphic skeletal changes, including longitudinal bone growth and radial cortical bone growth (166, 285). The concept that a functional GH-IGF-I axis is essential for skeletal sexual dimorphism is supported by observations in the GH receptor or IGF-I knockout mice, which are both characterized by the absence of gender differences in growth rate and skeletal size. Therefore, the male cortical phenotype requires a functional and active GH-IGF-I axis. This axis is already differently imprinted in males and females during the neonatal period (286). Such imprinting depends on neonatal androgen secretion. The importance of androgen programming of the GH-IGF-I system for male growth is supported by observations in androgen-resistant Tfm rats; in these androgen- resistant male rats without neonatal androgen secretion, female-like GH profiles are associated with female-like growth rates and bone size (285) (see also Section IV).


During puberty, a rise in GH-IGF-I activity occurs in both sexes. After puberty, serum IGF-I levels tend to be higher in men and male rats compared with females (287, 288), whereas no gender-related difference has been observed in mice (65, 76, 280).


High-dose estrogens lower serum levels of IGF-I in the rat (49, 209, 289), whereas low doses, via a stimulation of GH secretion, are stimulatory in both sexes (237). In humans, androgens indirectly stimulate IGF-I secretion after aroma- tization into estrogens (290). Furthermore, aromatase inhib-itors lower serum IGF-I levels (49), whereas antiestrogens like ICI 182,780 that do not penetrate the blood-brain barrier, do not affect serum IGF-I levels in male rats (59). These findings indicate that androgens are centrally aromatized, followed by an ER-mediated regulation of GH secretion. Estrogens stimulate GH via ERα, as indicated by the lower serum IGF-I in both genders of ERKO mice (see Section IV). In addition, the growth-limiting effect of high-dose estrogens seems to depend more on an interaction with the GH-IGF-I axis than the growth-stimulatory action of androgens. Indeed, the expected stimulation of radial bone growth does not occur after ovx in GH-deficient and hypophysectomized female rats, whereas periosteal bone formation and growth rate decrease after orch in GH-deficient male rats (224, 225, 291). In addition, the nonaromatizable androgen DHT may stimulate longitudinal growth in boys suffering from delayed puberty without concomitant increases of serum IGF-I (292).


Taken together, both human and animal data (although limited in number) support the concept that androgens, apart from a direct action on bone cells, also stimulate (skeletal) growth indirectly after aromatization into estrogens and stimulation of pituitary GH secretion.


VI.Effects of Androgens on the Human Skeleton


A Skeletal consequences of castration, male hypogonadism, and androgen resistance in men

1. Skeletal effects of castration in men. Surgical (orch) or chemical (administration of GnRH agonists) castration induces a complete and sudden decline in the levels of sex steroids in men. Such androgen deprivation therapy in adult men with advanced prostate cancer is followed by rapid bone loss (293), similar to the loss of skeletal integrity in women after surgical ovx or during early menopause. Lumbar spine bone density decreases by about 5-10% within 1 yr after castration (294 -302), with a continuing further decrease of bone loss thereafter. More recently, significant bone loss, albeit to a lesser extent, has also been confirmed at appendicular skeletal sites, including the hip (296, 298 -301, 303-305) and radius (299, 306, 307). Recent data suggest that many men with prostate carcinoma may already have reduced bone densities before androgen deprivation therapy (308). In these patients, additional bone loss after castration is likely to further in-crease their risk of osteoporotic fractures (303, 309 -312) (Fig. 8). In addition to the decline in bone density, androgen deficiency-induced changes in body composition, including decreased lean body and muscle mass (299, 300, 313), may further enhance fracture risk.


Figure 8. Cumulative incidence of first osteoporotic fractures in men with prostate cancer with and without orchidectomy. Intervals are times from castration to last follow-up or to first osteoporotic fracture and from diagnosis, respectively. Numbers in parentheses indicate men remaining at various intervals. [Reproduced with permission from H. W. Daniell: J Urol 157:439-444, 1997 (303).].


Castration increases bone turnover, as assessed by bio-chemical parameters of bone resorption and formation (294, 295, 298, 299, 301, 302, 307). These observations indicate that the mechanism of bone loss in androgen-deficient men is similar to that induced by gonadal insufficiency of women or animals. An imbalance in favor of bone resorption induces net bone loss, especially at cancellous bone sites with large remodeling surfaces. Histomorphometric confirmation of this high turnover bone loss in these men is still lacking, but suppression of bone turnover preserves bone density. In particular, the use of bisphosphonates-including etidronate (301), iv pamidronate (298), or zoledronic acid (314)- has been shown to prevent bone loss in patients with prostate carcinoma and should therefore be considered after castration. Overall, these findings support the concept that sex steroid deficiency in men, as in women, induces highturnover osteoporosis.


2. Skeletal effects of male hypogonadism. Hypogonadism is often defined as T levels well below the normal values. Still, this definition of hypogonadism may cause problems in some cases due to the wide range of T levels observed. In this section, we will discuss the skeletal effects of overt male hypogonadism, characterized by sustained low T concentra-tions (due to testicular and/or hypothalamic-pituitary dys-function) and clinical manifestations of hypogonadism. In Section VI.C.2, the skeletal impact in partially hypogonadal men will be addressed. Men with hypogonadism have sig-nificantly lower bone density (in particular at cancellous bone sites like the spine) than age-matched controls (315, 316). Little is known, however, about the impact of male hypogonadism on bone remodeling. Some data suggest that bone resorption and, to a lesser extent, bone formation may be increased in adult hypogonadal men, in line with observations in postmenopausal women (317), whereas other reports have provided histomorphometric evidence for low bone formation in the context of male hypogonadism (318, 319). Deterioration of the trabecular bone architecture of the distal tibia, as determined by high-resolution magnetic resonance micro-imaging, was recently reported in untreated hypogonadal men (320). In the study by Baran et al. (318), follow-up biopsies showed increased bone formation during supplementation with T, supporting the concept that T stim-ulates bone formation. In addition to histomorphometric evidence for a low rate of bone formation, Francis et al. (319) also observed calcium malabsorption and low serum 1,25- dihydroxyvitamin D3 concentrations in hypogonadal men. These findings contrast with other studies, documenting nor-mal calcium and 1,25-dihydroxyvitamin D3 metabolism in hypogonadal men (317) and suggesting that male hypo-go- nadism may affect skeletal homeostasis independently of cal-cium and vitamin D. Experiments using the orch rat model support this assumption (209).


The extent to which low bone density in the context of male hypogonadism is associated with an increased risk for fracture remains to be clarified. Case control studies report a higher than expected prevalence of hypogonadism in men with spine or hip fracture (321, 322). In men with hip fractures, increased bone resorption (but not the low formation rate) has even been documented to be a direct consequence of their T deficiency (323). However, prospective evidence to establish a cause-and-effect relationship between hypogonadism and future fracture risk is currently lacking.


Male hypogonadism can be due to a variety of diseases, which may all have a specific impact on skeletal integrity. Nevertheless, comparative data suggest similar impairment in bone density in men with different etiologies of hypo-gonadism, suggesting that hypogonadism per se and not the primary disease entity is responsible for the bone loss (324).


In young hypogonadal men, skeletal growth is impaired and, in line with animal data, bone mass and density will be even more severely affected. Decreased radial, spinal, and femoral bone densities and lower peak bone mass have been reported in adult men with delayed puberty (325-327), but areal bone density assessment based on projectional methods should be interpreted with caution. These methods may overestimate the bone mineral deficit during growth because of the associated failure to expand bone during delayed puberty. In this regard, measurements of volumetric density using pQCT are more appropriate. In adult men with a history of late puberty, pQCT fails to show decreased bone density, indicating that impairment of bone size maybe more important in these men than changes in bone tissue compo-sition (328).


Isolated hypogonadotropic hypogonadism (IHH) repre-sents the most complete and early form of male hypogonad-ism. In contrast to most other types of hypogonadotropic hypogonadism, men with IHH have isolated sex steroid de-ficiency without other metabolic abnormalities, making IHH a good model to examine the effects of sex steroids and sex steroid deficiencies in men. Compared with age-matched controls, patients with IHH have lower bone density at the spine and radius, not only before but also after growth plate closure (329). Interestingly, both areal and volumetric bone density are reduced. This suggests that true bone composition is impaired in the context of IHH. In these patients, assessing bone turnover has produced inconsistent results, demonstrating histomorphometric evidence for low-turnover osteoporosis in some patients (330) but increased levels of markers of bone formation and resorption in others (331).


Lower bone density (at radius and spine) is well docu-mented in hyperprolactinemic hypogonadal men as well (332). Moreover, reversal of the hypogonadism significantly increases cortical bone density, irrespective of the serum levels of prolactin, suggesting that it is T deficiency and not prolactin excess that impairs skeletal homeostasis in these patients (333).


Finally, Klinefelter's syndrome (KS) is the most frequent form of hypergonadotropic hypogonadism. According to most studies, with one exception (334), bone density is decreased in KS (335-339). Low bone density has even been reported in patients who have already been receiving longterm T replacement (338, 339), questioning the role of androgen deficiency as the cause of the bone deficit (as well as the role of androgen replacement to maintain bone density in this patient group) and suggesting other, disease-specific effects on bone that may be independent of the associated hypogonadism. Varying degrees of hypogonadism have been observed in patients with KS, and only those with severe hypogonadism may experience bone loss (334, 337). However, because of the small numbers of subjects and the lack of appropriate control groups in most studies dealing with KS, controversy is likely to persist until large prospective and controlled studies document the skeletal impact of different degrees of hypogonadism (and their response to T replacement).


Recently, the pivotal role of estrogens with respect to skel-etal development and maintenance in both sexes has received much attention (4, 5). It is now well-established that estrogens are responsible for skeletal maturation and epiphyseal closure, not only in women but also in men. However, the extent to which other components of bone formation, including trabecular thickening and periosteal formation, are regulated by aromatization of androgens into estrogens or directly by androgens remains unknown. Moreover, the degree of estrogen deficiency during male hypogonadism may vary according to the capacity to aromatize androgens. Patients with very low androgens also have limited capacity for aromatization. Hypogonadism should therefore be regarded as a combination of varying degrees of androgen and estrogen deficiency, which may impact bone differently.


3.Skeletal effects of androgen resistance in men. According to most (340 -343) but not all (344) studies, patients with the complete androgen insensitivity syndrome (cAIS) have lower areal bone density at the spine and hip when compared with age- and sex-matched controls. These findings suggest that androgens may also directly stimulate bone density via the AR and not only indirectly, after aromatization into estrogens. However, bone density values in androgen-resistant patients may be confounded by surgical castration and hormone replacement therapy. In line with this assumption, Marcus et al. (344) reported that compliance with estrogen is an important determinant of bone density in this patient group (Fig. 9). In addition, estrogen increases bone density at the spine and hip in most patients with cAIS (342, 343), although these measurements have not always been adjusted for their tall stature. Finally, androgen resistance in humans is not associated with impaired longitudinal growth, providing further evidence for the pivotal role of estrogens in longitudinal bone growth and epiphyseal closure in men. Whether androgen resistance in men affects periosteal bone formation, as would be expected from animal research, is not known.


Figure 9. Box plots of lumbar spine and femoral neck (FN) BMD z- scores for 18 women with cAIS by degree of compliance with estrogen replacement therapy. Values were compared with nominal population means by single sample t tests. Open boxes, Poor and fair compliance; hatched boxes, good and excellent compliance. The vertical lines of the box define the 25th and 75th percentiles, and the error bars denote the 10th and 90th percentiles. *, P = 0.06; **, P = 0.02; ***, P = 0.0001 (compared with normative mean z-score of 0). #, P = 0.005 (good vs. fair compliance). [Reproduced with permission from R. Marcus et al.: J Clin Endocrinol Metab 85:1032-1037, 2000 (344). © The Endocrine Society.].


Overall, bone studies in hypogonadal and androgen- resistant men are confounded by varying states of androgen and/or estrogen deficiency. Currently available data are par-ticularly hampered by the inability of areal bone density measurements to distinguish between bone size and com-position. The specific role of estrogens and androgens in the regulation of periosteal bone formation in humans also re-mains unknown (345). Only recently, Ahlborg et al. (346) have reported that the increased bone loss after menopause in women is associated with increased periosteal apposition, and accordingly, increased bone size (346,347). Because bone size is a major determinant of bone strength, further research in this area is urgently required.


B. Skeletal effects of androgens in women

The role of androgens in female skeletal homeostasis has not been well-established. In women, serum androgen con-centrations vary considerably; although T concentrations are lower than in men, serum concentrations of other, weaker androgens like androstenedione and DHEA-S are similar (348). Androgens may therefore contribute to clinically relevant differences in bone density between women.


Androgens might stimulate skeletal development during puberty. Most data in support of such a bone-stimulatory action are based on a number of studies, providing evidence for increased density in women with polycystic ovary syn-drome (PCOS) (Ref. 349 and references therein). One of the limitations of the PCOS model of androgen excess is the potential confounding by differences in body mass index, body composition, and menstrual irregularities (oligo- and amenorrhea) that may affect skeletal homeostasis indepen-dently of the androgen excess and are difficult to control in the case-control design of these studies. Moreover, androgen excess is often defined clinically by the presence of hirsutism, which represents only a rough determinant of androgen exposure in women. Despite these limitations, studies in PCOS have provided increasing and consistent evidence that hirsute women have higher peak bone density than age- matched controls, even after correction for body mass index (350, 351). It is important to note that this increase in bone density has been confirmed by cancellous pQCT and thus reflects real changes in bone tissue composition, rather than changes in bone size. Whether these positive effects relate to AR-mediated action or to aromatization of androgens in estrogens remains unknown. Obese patients with PCOS tend to have higher bone densities than their nonobese counter-parts, suggesting that aromatization within fat tissue may be important. It remains to be clarified whether and to what extent other typical metabolic abnormalities associated with PCOS, including low levels of SHBG (with correspondingly increased bioavailable hormone concentrations) and high concentrations of insulin, also affect skeletal homeostasis in PCOS. Finally, differences in body composition may be ad-ditional, clinically important mechanical determinants of skeletal integrity. In line with this assumption, site-specific differences in bone density have been observed in patients with PCOS (352). One particular area of concern relates to the bone safety of some of the treatment regimens in hirsute women, in par-ticular GnRH agonists and AR antagonists, alone or in com-bination. The AR antagonist flutamide, when administered alone, does not appear to induce changes in lumbar spine density (353), whereas another AR antagonist, spironolactone, when combined with linesterol, induces a decline in bone density at the same site (354). However, these findings are based on uncontrolled, short-term studies. Suppression of estrogen and androgen levels using GnRH agonist therapy induces postmenopausal-like bone loss in hirsute women. As expected, this bone loss is prevented by estrogen-progestin replacement therapy (355). More surprisingly, the AR an-tagonist spironolactone (but not flutamide) maintained bone density in hirsute women treated with GnRH agonists during a 6-month study (356). The mechanism of this bonesparing effect remains unclear and is in contrast to the earlier reported bone loss during spironolactone treatment in women who are not receiving a GnRH agonist (354).


In postmenopausal women, a potential bone-sparing action of androgens is less documented. Menopause is asso- dated with a 70% decline in the circulating levels of adrenal androgens (especially DHEA-S) (348, 357), but the extent to which this decline contributes to bone loss and fracture risk has not been assessed prospectively. In cross-sectional studies, no consistent relationship has emerged between serum levels of DHEA-S and bone density (358, 359). Moreover, it remains unclear to what extent these adrenal androgens have direct AR-mediated skeletal effects or mainly represent a source for aromatization into estrogens.


C.Skeletal effects of androgen replacement

Although the skeleton of prepubertal boys already re-sponds to T, as shown in an elegant calcium kinetics study by Mauras et al. (360), the benefits of T replacement have been most extensively documented in hypogonadal men. The in-terest of the scientific community in the potential benefits of androgen administration currently extends to young men with delayed puberty, elderly men with partial androgen deficiency, eugonadal men and glucocorticoid-treated men suffering from osteoporosis, and even postmenopausal women. Therefore, we will discuss the skeletal effects of T in the well-established indication of male hypogonadism and other new potential indications separately.


1. Skeletal effects of androgen replacement in male hypogonadism. Long-term prospective and retrospective data with respect to the efficacy of T replacement in male hypogonadism indicate that, after a 2-yr initial increase, bone density is maintained (Fig. 10). However, the reported gain in bone density is highly variable (324, 333, 361-366), and an increase has not even been documented in all studies (367). This large vari ation in effects on bone density may relate to differences in treatment duration as well as to different methodologies and sites to assess bone density. Table 11 summarizes the effects of androgens on bone in hypogonadal men. Overall, cancellous bone sites like the spine are more responsive than cortical sites like the radius or hip (363,365), and measurements based on pQCT or QCT (which do not fully correct for possible androgen-induced changes in fat) show much greater responses than approaches using DXA, dual photon absorp-tiometry (DPA), or single photon absorptiometry (SPA).


Table 11. Effects of androgen replacement on bone density in hypogonadal men.


Figure 10. Increase in BMD during long-term T substitution therapy up to 16 yr in 72 hypogo- nadal patients. Circles indicate hypogonadal patients with first pQCT measurement before initiation of T substitution therapy; squares show those patients already receiving T therapy at the first QCT. The dark-shaded area indicates the range of high fracture risk, the unshaded area indicates the range without significant fracture risk, and the light-shaded area indicates the intermediate range in which fractures may occur. [Reproduced with permission from H. M. Behre et al.: J Clin Endocrinol Metab 82:2386-2390, 1997 (364). © The Endocrine Society.].


Table 1. Sexually dimorphic age-related changes in humans.


The overall impression is that T, similar to estrogen in postmenopausal women, primarily acts as an antiresorptive rather than an anabolic agent. Not only are the reported increases and the pattern of bone density gain in line with this assumption, but also the observed responses of biochemical bone turnover markers. Most studies show a consistent decline in bone resorption after T replacement in hypogonadal men (331, 363, 365-367). A similar decrease in the levels of markers of bone formation, as would be expected during treatment with a typical antiresorptive agent, has been observed in some studies (363, 365). However, in various trials, markers of bone formation have been reported to initially increase after T replacement in hypogonadal men (362, 366 -368) or human choriogonadotropin (331). In addition to its effects on bone turnover, androgen replacement in hypogonadal men may have beneficial effects on body composition, which in turn may protect bone. Several studies have reported increases in lean body mass (331,363,367,369) or muscle strength (277,367,368,370) after im or transdermal T replacement, but again, other trials have been unable to confirm these findings (365, 371).


The mode of T administration may be an important de-terminant of its efficacy and safety. Although im adminis-tration of T may result in supraphysiological effects on bone, as suggested by a 5% (uncontrolled) gain of lumbar bone density within 2 yr in eugonadal T-treated men with osteo-porosis (372), transdermal administration tends to reach more physiological concentrations of T (369). Preliminary retrospective data have not revealed differences in efficacy (or safety) between these two modes of administration (364), but prospective comparative trials have not been performed as yet.


For ethical reasons, most T replacement studies in hypo- gonadal men have been uncontrolled. Consequently, there is little or no information about the natural evolution of bone density in these patients or about the skeletal benefits of T in addition to the benefits provided by calcium and vitamin D. Moreover, the effects of androgen replacement have been studied in small-sized studies of patients with different eti-ologies and variable degrees of estrogen and androgen de-ficiency. Lack of efficacy of T replacement in maintaining bone mass has been suggested in studies including KS patients (338, 339). However, this assumption was based on subnormal bone densities found in a small number of patients with KS who were receiving long-term T therapy. It is likely that the need for T replacement and the patient's response in terms of bone density gain will depend on his pretreatment levels of circulating T and bone density. Indeed, according to a large retrospective study, patients with the lowest initial BMD gain most during therapy (364). Of particular importance are findings that the loss of skeletal integrity may be partially irreversible. This is probably the case in patients with IHH who experienced a failure to gain normal bone mass during puberty. In these patients, T is likely to maintain bone density without completely restoring peak bone mass (330).


Finally, the question remains whether the impact of an-drogen supplementation relates primarily to activation of the AR. Most density-endpoint studies have used T replacement. Because T is an aromatizable androgen, all of these studies may reflect both androgen and estrogen replacement and, thus, do not allow us to draw specific conclusions regarding direct AR-mediated action only. An uncontrolled study in eugonadal men suggests that the beneficial effects of T on bone density may be related more to increases in E2 levels than to changes in T concentrations (372). No studies on the skeletal effects of the nonaromatizable androgen DHT have been reported in hypogonadal men.


Overall, the beneficial effects of the aromatizable androgen T on bone density are well-established in the context of male hypogonadism. In addition to the bone-sparing effect, a T- induced gain in lean body mass, as reflected in increased muscle mass and strength, may provide an additional benefit. However, it remains unknown to what extent these positive effects of T replacement will ultimately protect hy- pogonadal men against osteoporosis and osteoporotic fracture occurrence.


2. Skeletal effects of androgen replacement in other indications. Although the overall benefits of T replacement are well-documented in hypogonadal men and although T may not only affect bone and muscle mass but also mood, sexual function, and hematopoiesis (373,374), the potential benefits of T replacement in indications other than overt hypogonadism are not well-established and remain the subject of considerable debate.


The large group of elderly men with modest or partial degrees of T deficiency, as reflected by low levels of bio-available T, represents the most important potential target population for T replacement. Aging men are increasingly at risk of bone loss and osteoporotic fracture. However, the extent to which low levels of bioavailable T contribute to age-related bone loss in men remains unknown. In contrast to well-documented correlations between bioavailable E2 and bone density (4,5), studies in elderly men have failed to show strong or consistent associations between bioavailable T and bone density (359, 375, 376). Small studies have reported favorable changes on bone markers after T replacement in these men, but again, findings have not been consistent and their clinical significance remains unclear (377379). Because the relationship between bone density or bone turnover and fracture risk is complex and, in men, largely unsettled, additional research is required to examine whether and to what extent beneficial effects on bone turnover or density may ultimately translate in clinical benefits in terms of fracture risk reduction.


bone turnover or density may ultimately translate in clinical benefits in terms of fracture risk reduction. Two randomized double-blind placebo-controlled studies in elderly men with partial androgen deficiency have been large enough and of sufficient duration to evaluate the effects of T on bone density (Table 12). In the trial by Snyder et al. (378), the use of transdermal T during 36 months increased bone density, but no additional benefit could be documented compared with the control group receiving calcium and vitamin D supplementation (Fig. 11). In a study by Kenny et al. (379), transdermal T maintained bone density, whereas significant bone loss was observed in the placebo group, despite calcium and vitamin D supplementation. It is possible that both studies tend to underestimate the potential beneficial effect of T. As indicated, the placebo groups in both studies were supplemented with calcium and vitamin D, and this may have partially blunted any age-associated bone loss. In addition, a significant proportion of the men included in the Snyder study (378) were not really hypogonadal but had T levels in the low normal range. A post hoc analysis has highlighted the importance of pretreatment androgen status on treatment outcome: the lower the pretreatment serum T levels, the greater the effect of T on bone density. In addition, both studies were performed with transdermal T, which might be less effective than im T, the more commonly used androgen in hypogonadal men. Finally, these studies do not answer the critically important question about a potential threshold serum T level that may warrant T replacement in elderly men. Animal data suggest that it may be lower than the lower range of young normal men (233).


Table 12. Effects of androgen replacement on bone density in men with partial androgen deficiency.


Figure 11. Mean (± SE) BMD of the lumbar spine (L2-L4) as a percentage of the basal value in 108 men over 65 yr of age who were treated with either T or placebo (54 men each). BMD increased significantly (P < 0.001) from 0-36 months in both groups, but the increase was not significantly different between the two groups at 36 months. [Reproduced with permission from P. J. Snyder et al.: J Clin Endocrinol Metab 84:1966-1972, 1999 (378). © The Endocrine Society.


More recently, the effects of the nonaromatizable androgen DHT have also been evaluated in men with serum T levels in the low normal range. However, this study was of short duration (3 months) and only evaluated one marker of bone turnover (osteocalcin), which was not affected (371).


T replacement might be of more benefit to men who have both T deficiency and an additional increase in the risk for osteoporosis. A placebo-controlled cross-over study in glucocorticoid-treated men suffering from osteoporosis re-ported a 5% gain in lumbar spine bone density after 1 yr of im administration of T (380). However, no long-term data are available on the effects of T supplementation on bone loss and fracture risk in steroid-induced osteoporosis.


Delayed puberty represents another potential indication for T therapy. A 6-month study in a small number of adolescents with delayed puberty suggests that treatment with a low dose of T may increase bone density. However, the ultimate impact of this treatment on adult bone density was not documented (326).


Finally, in postmenopausal osteoporosis, the potential role of T administration remains to be defined as well (381, 382). The combination of low-dose T (or methyl-T) and estrogen administration may reverse the inhibitory effects of estrogen on biochemical markers of bone formation (383). Limited data also suggest that combined estrogen and T replacement may result in an additional increase in bone density compared with estrogen alone (382,384). However, the potential side effects of long-term androgen replacement are a concern and, as long as long-term safety and efficacy data are lacking, androgen and estrogen combination therapy cannot be recommended in postmenopausal women.


In recent years, several studies have addressed the skeletal effects of the weaker androgen DHEA. A placebo-controlled double-blind study in elderly women (>70 yr) suggested that 50 mg daily of DHEA increased serum levels of DHEA-S, with a concomitant decline in biochemical markers of bone resorption and a modest increase in bone density at some sites (385). Such beneficial effects of DHEA have not been confirmed in men (385), except in one uncontrolled small study (386). The women and men included in these studies had variable concentrations of DHEA-S at baseline. It is plausible that women and men with the lowest serum levels of DHEA would have the greatest benefit, but this remains to be established. In these trials, DHEA treatment increased T and E2 concentrations (385-387), indicating that DHEA may act as a prohormone for these more potent sex steroids. However, further prospective controlled long-term research has to determine the bone-sparing potential of DHEA, alone or in combination with established osteoporotic treatments.


In summary, current scientific evidence does not allow recommending T replacement or other androgens to promote bone health in indications other than male hypogonadism.


D.Skeletal effects of selective modulation of androgen and estrogen action in men

In line with published animal data, AR antagonists, ER antagonists, SERMs, aromatase inhibitors, and type II 5α- reductase inhibitors all potentially interfere with male skel-etal homeostasis. However, few studies have investigated the potential skeletal benefits and side effects of these drugs in men.


Finasteride, a type II 5α-reductase inhibitor, does not de-crease vertebral bone density (388) or increase bone turnover markers (35) in men suffering from benign prostate hyper-plasia, according to two small-sized short-term studies. In accordance, the recent finding that type I 5α-reductase is predominantly expressed in osteoblasts (30) supports the absence of effects on bone in these clinical studies with a type II 5α-reductase inhibitor. These findings suggest that selective interference with the AR pathway in men may be relatively safe with respect to bone, but data remain scarce. Therefore, surveillance of bone density still remains advisable, especially in men with clinical risk factors for osteoporosis.


The skeletal risks and benefits of selective interaction with the ER pathway in men remain unsettled as well. Interfering with estrogen production in men is likely to be associated with deleterious effects on bone, as reflected in the animal studies. In an uncontrolled study, administration of the aro-matase inhibitor anastrazole to elderly men for 9 wk was associated with an increase in bone resorption (389). In con-trast, estrogen suppression via administration of anastrazole to young male adolescents did not affect calcium kinetics (390). Long-term studies should further clarify the impact of aromatase inhibitors on skeletal integrity in men.


In agreement with animal data, high-dose estrogens are bone-sparing in selected populations such as patients with prostate carcinoma after surgical castration (306) and male- to-female transsexuals receiving chemical castration (391, 392). In elderly men, preliminary evidence suggests that short-term low-dose estrogen may reduce bone resorption as assessed by biochemical markers (393). However, because of its potential feminizing side effects, this type of therapy cannot be recommended. In contrast, SERMs could potentially have a more acceptable risk-benefit profile in men. However, Doran et al. (394) recently failed to show a significant bone-sparing effect of raloxifene in elderly men, but this conclusion was based on a short-term evaluation of bone turnover over a 6-month period. Further studies are needed to explore the potential skeletal benefit of selective stimulation of the ER pathway in men. It remains a tempting hypothesis that selected populations of elderly men, particularly those with low serum estrogen concentrations, would benefit from treatment with SERMs. This assumption is supported by the observation that raloxifene suppresses bone resorption in elderly men with low E2 levels (394). A potential alternative is the recently reported gender-neutral synthetic steroid, estren, which increases bone mass without affecting reproductive organs in both male and female rodents (395, 396). Whether estren will have similar beneficial effects for the treatment of osteoporosis in humans remains to be determined.


The relative importance of androgens and estrogens for bone turnover in men still remains unresolved. Falahati-Nini et al. (397) first studied elderly men under conditions of physiological T and E2 replacement and then assessed the impact on bone turnover of withdrawing both T and E2, withdrawing either T or E2, or continuing both during 3 wk. Their findings establish E2 as the dominant sex steroid regulating bone resorption in normal elderly men, whereas both T and E2 independently maintain bone formation. More recently, an analogous pharmacological intervention study was designed by Leder et al. (398), inducing combined T and E2 deficiency, T and E2 sufficiency, or selective E2 deficiency in young men during a longer experimental period (12 wk). Both androgens and estrogens appeared to be independent mediators of bone resorption in young adult men and may have similar effects on osteoblast function as well. The differences reported between the two studies most likely represent differences in study subjects or study design, or the (in)ability to separate direct effects of sex steroid deprivation on osteoblast function from indirect ones (remodelingcoupled increases in osteoblast activity). Longer-term studies are needed for a comprehensive picture of the relative role of sex steroids in regulating bone turnover in men.


VII.General Conclusions


Bone development and growth are similar in boys and girls up to the start of puberty. Thereafter, skeletal sexual dimorphism evolves with a greater bone mass in adult males than in adult females. The volumetric or true density of bone is, however, similar in both sexes. Men have more bone because of greater bone volume as a result of higher periosteal bone formation rates. A similar sexual dimorphism is also observed in many other species (e.g., rodents), and a wide variety of data suggest androgens and estrogens to be the hormones responsible for this sexual dimorphism.


After a period of peak bone mass, age-related bone loss occurs in both genders, but men experience less age-related net bone loss, again in contrast to the accelerated bone re-sorption in women. Androgen deficiency in men induces cancellous bone loss similar to estrogen deficiency in post-menopausal women. The histological and biochemical changes induced by castration in men are, again, similar to changes observed in postmenopausal women; the rate of bone remodeling is increased after loss of sex steroids, resulting in enhanced osteoclastogenesis and an increase in the number of osteoblast progenitors, with the former exceeding the latter. This imbalance between resorption and formation, together with a delay of osteoclast apoptosis, is responsible for a decrease in trabecular bone volume, thickness, and connectivity (198, 395).


Recent studies using various sex steroid-related transgenic mouse models as well as selective pharmacological modu-lations of the different pathways of androgen action have expanded our understanding of the relative roles of the AR, ERα, and ERβ in mediating the effects of androgens on the skeleton. Androgen action on bone differs according to the skeletal compartment, species, sex, and maturation or age (Table 13). In all species investigated thus far, androgens as well as estrogens maintain cancellous bone mass and integ-rity, regardless of age or sex. Sex steroids reduce cancellous bone turnover primarily via a down-regulation of osteoclas- togenesis after interaction with bone marrow osteoblast pre-cursor cells (and maybe also via direct action on osteoclasts). Moreover, recent data indicate that androgens as well as estrogens also induce apoptosis of osteoclasts and prevent osteoblast apoptosis. Although androgens via the AR and estrogens via the ERs can induce these effects, it is less clear what the relative contribution of each system is in the normal life cycle of male and female animals and humans. Recent data suggest that androgen action on cancellous bone depends (only) on (local) aromatization of androgens into estrogens. However, at least in rodents, androgen action on cancellous bone can be directly mediated via AR activation, even in the absence of ERs. These data establish the dual mode of action of T via both the AR and ERα in males (Table 13). In contrast, estrogen effects on cancellous bone are always mediated via ERα. In females, ERβ is involved in longitudinal and radial bone growth as well as in the regulation of cancellous bone.


Table 13. General overview of androgen action on bone in males.


Androgens increase cortical bone size via stimulation of both longitudinal and radial growth. First, androgens, like estrogens, have a biphasic effect on endochondral bone for-mation; at the start of puberty, sex steroids stimulate endo-chondral bone formation, whereas they induce epiphyseal closure at the end of puberty. Androgen action on growth plate closure is, however, clearly mediated via aromatization in estrogens and interaction with ERα (Table 13). Indeed, the bone phenotype of men suffering from estrogen deficiency (due to a mutation in the aromatase gene) or resistance (sec-ondary to a mutation in the ERα gene) is characterized by the absence of a pubertal growth spurt and delayed epiphyseal closure. In growing rodents, who do not experience epiphy-seal closure, androgens and estrogens seem to stimulate lon-gitudinal growth via ERα in the growth plate. Low concen-trations of estrogens (as present in males) appear to be stimulatory, whereas higher concentrations of estrogens (as in females) are inhibitory for longitudinal bone growth. The role, if any, of AR stimulation with respect to longitudinal growth and epiphyseal closure is not well-established in any species.


Androgens increase radial growth, whereas estrogens de-crease periosteal bone formation. However, such action has only been documented in rodents and poorly or only partially in humans (346). Androgens stimulate periosteal bone formation in male rodents during puberty (Table 13). Whether androgens also stimulate periosteal bone formation after puberty remains to be clarified. This latter effect of androgens may be important because bone strength in males seems to be determined by relatively higher periosteal bone formation and, therefore, greater bone dimensions, relative to muscle mass at older age. Indeed, because volumetric bone density is similar in both genders and bone fragility and subsequent fracture incidence is greater in females, bone size is a major determinant of bone strength. Understanding the hormonal and humoral mechanisms of periosteal bone growth or, more generally, the growth and maintenance of the cortical area is of major importance. However, little is known about the differences in cellular behavior of the two major bone compartments (cancellous and cortical), and the lack of understanding of the effects of sex steroid hormones on these two compartments is only a reflection of this statement. Indeed, it remains unclear to what extent androgens directly interact with (periosteal) osteoblasts or their precursors or, alternatively, stimulate osteoblasts secondary to mechanical loading via androgen-mediated increases of body growth and muscle. The relative importance of direct effects of androgens via local sex steroid receptors in osteoblasts might soon be further clarified because mice with osteoblast- specific inactivations of the AR, ERα, and ERβ are currently being developed. Experiments in mice suggest that both the AR and ERα pathways are involved in androgen action on radial bone growth in males, but the relative importance and interaction between both pathways is unclear (Table 13). There are conflicting data from rodent studies; most data indicate that the AR is the mediator of periosteal expansion (Tfm rats and ANDRKO mice). However, ERα stimulation (in the absence of ERβ) can also stimulate periosteal growth and aromatase inhibitors in the presence of the AR, and normal androgen levels inhibit periosteal growth in rats. A possible scenario, still to be confirmed, would be that periosteal expansion can be achieved by AR activation and, to a lesser extent, by ERα but is inhibited by ERβ activated by relatively high estrogen concentrations. This hypothesis could explain why bone volume is arrested earlier in girls than boys during puberty and why periosteal expansion reoccurs in postmenopausal women. The molecular mechanism of action, even whether this occurs via direct or indirect mechanisms, and the role of the GH-IGF-I axis all have to be further clarified.


Treatment of severe androgen deficiency with androgen replacement therapy is beneficial for bone. Whether replace-ment therapy is also warranted in partially androgen-deficient elderly men is not unequivocally shown, and the overall effects on all target tissues and quality of life and survival should be further explored. Selective AR modulators, com-bining agonistic effects on bone and other target tissues (en-dothelium, brain, etc.) and antagonistic or neutral effects on prostate, might be equally attractive as SERMs for women. In view of the essential role of ERα on cancellous bone, some SERMs with the appropriate tissue selectivity might even be useful for elderly men with osteoporosis. Vice versa, selective AR modulators might be equally interesting for metabolic bone diseases in women. To achieve such goals, the molecular targets of ligand-activated AR/ERs mediating their action on bone morphology and metabolism have to be much better identified. New technologies of genetics and proteomics combined with the availability of a whole set of cell-specific transgenic mice may soon provide new insights.


In summary, the AR is present in nearly all bone cells, and the major androgen, T, has a major impact on skeletal growth and maintenance, not only via signaling through the AR, but also via ERs. During skeletal remodeling, both receptor path-ways generate similar and additive effects on bone. For skeletal longitudinal growth, ERα is the crucial pathway, whereas for periosteal growth, activation of both the AR and ERα appears to be relevant.



Acknowledgements


This study was supported by the Swedish Medical Research Council, the Swedish Foundation for Strategic Research, the Lundberg Foundation, the Torsten and Ragnar Soderberg's Foundation, the Emil and Vera Cornell Foundation and Petrus and Augusta Hedlunds Foundation, as well as Grants G.0221.99, G.0417.03, and G.0171.03 from the Fund for Scientific Research-Flanders, Belgium (F.W.O.-Vlaanderen), and University Grant OT/01/39 from the Katholieke Universiteit Leuven. D.V. and S.B. are both senior clinical investigators of the Fund for Scientific Research-Flanders (F.W.O.-Vlaanderen). D. V. and L.V. contributed equally to this work.





References


Tenover JL 1999 Testosterone replacement therapy in older adult men. Int J Androl 22:300 -306 

Albright F, Reifenstein EC 1948 Metabolic bone disease: osteoporosis. In: Albright F, Reifenstein EC, eds. The parathyroid glands and metabolic bone disease. Baltimore: Williams and Wilkins; 145-204 

Vanderschueren D, Boonen S, Bouillon R 2000 Osteoporosis and osteoporotic fractures in men: a clinical perspective. Baillieres Best Pract Res Clin Endocrinol Metab 14:299 -315 

Riggs BL, Khosla S, Melton LJ 2002 Sex steroids and the construction and conservation of the adult skeleton. Endocr Rev 23: 279 -302 

Khosla S, Melton III LJ, Riggs BL 2002 Estrogen and the male skeleton. J Clin Endocrinol Metab 87:1443-1450 

Juul A 2001 The effects of oestrogens on linear bone growth. Hum Reprod Update 7:303-313 

Vanderschueren D, Bouillon R 1995 Androgens and bone. Calcif Tissue Int 56:341-346 

Compston JE 2001 Sex steroids and bone. Physiol Rev 81:419 -447 

Duan Y, Turner CH, Kim BT, Seeman E 2001 Sexual dimorphism in vertebral fragility is more the result of gender differences in age-related bone gain than bone loss. J Bone Miner Res 16:2267-2275 

Seeman E 2002 Pathogenesis of bone fragility in women and men. Lancet 359:1841-1850 

Russo CR, Lauretani F, Bandinelli S, Bartali B, Di Iorio A, Volpato S, Guralnik JM, Harris T, Ferrucci L 2003 Aging bone in men and women: beyond changes in bone mineral density. Osteoporos Int 14:531-538 

Longcope C 1998 Metabolism of estrogens and progestins. In: Fraser Jr IS, Lobo RA, Whitehead MI, eds. Estrogens and progestens in clinical practice. New York: Churchill Livingstone; 89 -94 

Fujikawa H, Okura F, Kuwano Y, Sekizawa A, Chiba H, Shimo- daira K, Saito H, Yanaihara T 1997 Steroid sulfatase activity in osteoblast cells. Biochem Biophys Res Commun 231:42-47 

Saito H, Yanaihara T1998 Steroid formation in osteoblast-like cells. J Int Med Res 26:1-12 

Janssen JM, Bland R, Hewison M, Coughtrie MW, Sharp S, Arts J, Pols HA, van Leeuwen JP 1999 Estradiol formation by human osteoblasts via multiple pathways: relation with osteoblast function. J Cell Biochem 75:528-537 

Eyre LJ, Bland R, Bujalska IJ, Sheppard MC, Stewart PM, Hewison M 1998 Characterization of aromatase and 17 ß-hydroxysteroid de-hydrogenase expression in rat osteoblastic cells. J Bone Miner Res 13:996-1004 

Van Der Eerden BC, Van De Ven J, Lowik CW, Wit JM, Karperien M 2002 Sex steroid metabolism in the tibial growth plate of the rat. Endocrinology 143:4048 -4055 

Nakano Y, Morimoto I, Ishida O, Fujihira T, Mizokami A, Tani- moto A, Yanagihara N, Izumi F, Eto S 1994 The receptor, metabolism and effects of androgen in osteoblastic MC3T3- E1 cells. Bone Miner 26:245-259 

Purohit A, Flanagan AM, Reed MJ 1992 Estrogen synthesis by osteoblast cell lines. Endocrinology 131:2027-2029 

Tanaka S, Haji M, Nishi Y, Yanase T, Takayanagi R, Nawata H 1993 Aromatase activity in human osteoblast-like osteosarcoma cell. Calcif Tissue Int 52:107-109 

Nawata H, Tanaka S, Tanaka S, Takayanagi R, Sakai Y, Yanase T,Ikuyama S, Haji M1995 Aromatase inbone cell: association with osteoporosis in postmenopausal women. J Steroid Biochem Molec Biol 53:165-174 

Shimodaira K, Fujikawa H, Okura F, Shimizu Y, Saito H, Yanaihara T 1996 Osteoblast cells (MG-63 and HOS) have aromatase and 5 a-reductase activities. Biochem Mol Biol Int 39:109 -116 

Sasano H, Uzuki M, Sawai T, Nagura H, Matsunaga G, Kashimoto O, Harada N 1997 Aromatase in human bone tissue. J Bone Miner Res 12:1416-1423 

Shozu M, Simpson ER 1998 Aromatase expression of human osteoblast-like cells. Mol Cell Endocrinol 139:117-129 

Oz OK, Millsaps R, Welch R, Birch J, Zerwekh JE 2001 Expression of aromatase in the human growth plate. J Mol Endocrinol 27: 249-253 

Audi L, Carrascosa A, Ballabriga A 1984 Androgen metabolism by human fetal epiphyseal cartilage and its chondrocytes in primary culture. J Clin Endocrinol Metab 58:819-825 

Bruch HR, Wolf L, Budde R, Romalo G, Schweikert HU 1992 Androstenedione metabolism in cultured human osteoblast-like cells. J Clin Endocrinol Metab 75:101-105 

Vittek J, Altman K, Gordon GG, Southren AL 1974 The metabolism of 7a-3H-testosterone by rat mandibular bone. Endocrinology 94:325-329 

Schweikert HU, Rulf W, Niederle N, Schafer HE, Keck E, Kruck F 1980 Testosterone metabolism in human bone. Acta Endocrinol (Copenh) 95:258-264 

Issa S, Schnabel D, Feix M, Wolf L, Schaefer HE, Russell DW, Schweikert HU 2002 Human osteoblast-like cells express predominantly steroid 5α-reductase type 1. J Clin Endocrinol Metab 87: 5401-5407 

Feix M, Wolf L, Schweikert HU 2001 Distribution of 17ß-hydroxy- steroid dehydrogenases in human osteoblast-like cells. Mol Cell Endocrinol 171:163-164 

Kuwano Y, Fujikawa H,Watanabe A, Shimodaira K, Sekizawa A, Saito H, Yanaihara T 1997 3ß-Hydroxysteroid dehydrogenase activity in human osteoblast-like cells. Endocr J 44:847-853 

Rosen HN, Tollin S, Balena R, Middlebrooks VL, Moses AC, Yamamoto M, Zeind AJ, Greenspan SL 1995 Bone density is normal in male rats treated with finasteride. Endocrinology 136: 1381-1387 

Matzkin H, Chen J, Lewyshon O, Ayalon D, Braf Z 1992 Effects of long term treatment with finasteride (MK-906), a 5-a reductase inhibitor, on circulating LH, FSH, prolactin and estradiol. Horm Metab Res 24:498-499 

Tollin SR, Rosen HN, Zurowski K, Saltzman B, Zeind AJ, Berg S, Greenspan SL 1996 Finasteride therapy does not alter bone turnover in men with benign prostatic hyperplasia-a clinical research center study. J Clin Endocrinol Metab 81:1031-1034 

Feldmann S, Minne HW, Parvizi S, Pfeifer M, Lempert UG, Bauss F, Ziegler R 1989 Antiestrogen and antiandrogen administration reduce bone mass in the rat. Bone Miner 7:245-254 

Goulding A, Gold E 1993 Flutamide-mediated androgen blockade evokes osteopenia in the female rat. J Bone Miner Res 8:763-769 

Lea C, Kendall N, Flanagan AM 1996 Casodex (a nonsteroidal antiandrogen) reduces cancellous, endosteal, and periosteal bone formation in estrogen-replete female rats. Calcif Tissue Int 58: 268-272 

Gallagher AC, Chambers TJ, Tobias JH 1996 Androgens contribute to the stimulation of cancellous bone formation by ovarian hormones in female rats. Am J Physiol 270:E407-E412 

Lea CK, Flanagan AM 1999 Ovarian androgens protect against bone loss in rats made oestrogen-deficient by treatment with ICI 182,780. J Endocrinol 160:111-117 

Vanderschueren D, Van Herck E, Geusens P, Suiker A, Visser W, Chung K, Bouillon R 1994 Androgen resistance and deficiency have different effects on the growing skeleton of the rat. Calcif Tissue Int 55:198-203 

Kawano H, Sato T, Yamada T, Matsumoto T, Kawaguchi H, Kato S 2002 Both androgen and estrogen signalings contribute to the maintenance of bone mass in males: analyses of male androgen receptor deficient mice. J Bone Miner Res 17(Suppl 1):S365 (Abstract) 

Yeh S, Tsai MY, Xu Q, Mu XM, Lardy H, Huang KE, Lin H, Yeh SD, Altuwaijri S, Zhou X, Xing L, Boyce BF, Hung MC, Zhang S, Gan L, Chang C 2002 Generation and characterization of androgen receptor knockout (ARKO) mice: an in vivo model for the study of androgen functions in selective tissues. Proc Natl Acad Sci USA 99:13498-13503 

Sato T, Kawano H, Kato S 2002 Study of androgen action in bone by analysis of androgen-receptor deficient mice. J Bone Miner Metab 20:326-330 

Vandenput L, Boonen S, Erben RG, Van Herck E, Swinnen JV, Bouillon R, Vanderschueren D 2002 Anabolic effects of testosterone on bone require the presence of a functional androgen receptor in male mice. J Bone Miner Res 17(Suppl 1):S290 (Abstract) 

Tozum TF, Oppenlander M, Chen C, Robins DM, McCauley LK 2002 Impact of estrogen and androgen receptors on skeletal metabolism. J Bone Miner Res 17(Suppl 1):S289 (Abstract) 

Vanderschueren D, Van Herck E, Nijs J, Ederveen AG, De Coster R, Bouillon R1997 Aromatase inhibition impairs skeletal modeling and decreases bone mineral density in growing male rats. Endocrinology 138:2301-2307 

Vanderschueren D, Van Herck E, De Coster R, Bouillon R 1996 Aromatization of androgens is important for skeletal maintenance of aged male rats. Calcif Tissue Int 59:179-183 

Vanderschueren D, Boonen S, Ederveen AG, De Coster R, Van Herck E, Moermans K, Vandenput L, Verstuyf A, Bouillon R 2000 Skeletal effects of estrogen deficiency as induced by an aromatase inhibitor in an aged male rat model. Bone 27:611-617 

Morishima A, Grumbach MM, Simpson ER, Fisher C, Qin K1995 Aromatase deficiency in male and female siblings caused by a novel mutation and the physiological role of estrogens. J Clin Endocrinol Metab 80:3689-3698 

Bilezikian JP, Morishima A, Bell J, Grumbach MM 1998 Increased bone mass as a result of estrogen therapy in a man with aromatase deficiency. N Engl J Med 339:599-603 

Carani C, Qin K, Simoni M, Faustini-Fustini M, Serpente S, Boyd J, Korach KS, Simpson ER 1997 Effect of testosterone and estradiol in a man with aromatase deficiency. N Engl J Med 337:91-95 

Herrmann BL, Saller B, Janssen OE, Gocke P, Bockisch A, Sperling H, Mann K, Broecker M 2002 Impact of estrogen replacement therapy in a male with congenital aromatase deficiency caused by a novel mutation in the CYP19 gene. J Clin Endocrinol Metab 87:5476-5484 

Oz OK, Zerwekh JE, Fisher C, Graves K, Nanu L, Millsaps R, Simpson ER 2000 Bone has a sexually dimorphic response to aromatase deficiency. J Bone Miner Res 15:507-514 

Oz OK, Hirasawa G, Lawson J, Nanu L, Constantinescu A, Antich PP, Mason RP, Tsyganov E, Parkey RW, Zerwekh JE, Simpson ER 2001 Bone phenotype of the aromatase deficient mouse. J Steroid Biochem Mol Biol 79:49-59 

Miyaura C, Toda K, Inada M, Ohshiba T, Matsumoto C, Okada T, Ito M, Shizuta Y, Ito A 2001 Sex- and age-related response to aromatase deficiency in bone. Biochem Biophys Res Commun 280: 1062-1068 

Sibonga JD, Dobnig H, Harden RM, Turner RT 1998 Effect of the high-affinity estrogen receptor ligand ICI 182,780 on the rat tibia. Endocrinology 139:3736-3742 

Gallagher A, Chambers TJ, Tobias JH 1993 The estrogen antagonist ICI 182,780 reduces cancellous bone volume in female rats. Endocrinology 133:2787-2791 

Vandenput L, Swinnen JV, Van Herck E, Verstuyf A, Boonen S, Bouillon R, Vanderschueren D 2002 The estrogen receptor ligand ICI 182,780 does not impair the bone-sparing effects of testosterone in the young orchidectomized rat model. Calcif Tissue Int 70: 170-175 

Turner RT, Evans GL, Dobnig H 2000 The high-affinity estrogen receptor antagonist ICI 182,780 has no effect on bone growth in young male rats. Calcif Tissue Int 66:461-464 

Smith EP, Boyd J, Frank GR, Takahashi H, Cohen RM, Specker B, Williams TC, Lubahn DB, Korach KS 1994 Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. N Engl J Med 331:1056-1061 

Schmidt A, Seedor JG, Gentile MA, Gentile BL, Pennypacker BL, Rodan GA, Kimmel DB 1999 Femoral bone density and length in male and female estrogen receptor-a (ERα) knockout mice. J Bone Miner Res 14(Suppl 1):S456 (Abstract) 

Couse JF, Korach KS 1999 Estrogen receptor null mice: what have we learned and where will they lead us? Endocr Rev 20:358-417 

Vidal O, Lindberg MK, Hollberg K, Baylink DJ, Andersson G, Lubahn DB, Mohan S, Gustafsson JA, Ohlsson C 2000 Estrogen receptor specificity in the regulation of skeletal growth and maturation in male mice. Proc Natl Acad Sci USA 97:5474-5479 

Lindberg MK, Alatalo SL, Halleen JM, Mohan S, Gustafsson JJ, Ohlsson C 2001 Estrogen receptor specificity in the regulation of the skeleton in female mice. J Endocrinol 171:229-236 

Lindberg MK, Moverare S, Skrtic S, Alatalo S, Halleen J, Mohan S, Gustafsson JA, Ohlsson C 2002 Two different pathways for the maintenance of trabecular bone in adult male mice. J Bone Miner Res 17:555-562 

Lindberg MK, Weihua Z, Andersson N, Moverare S, Gao H, Vidal O, Erlandsson M, Windahl S, Andersson G, Lubahn DB, Carlsten H, Dahlman-Wright K, Gustafsson JA, Ohlsson C 2002 Estrogen receptor specificity for the effects of estrogen in ovariectomized mice. J Endocrinol 174:167-178 

Deleted in proof Lindberg MK, Moverare S, Skrtic S, Gao H, Dahlman-Wright K, Gustafsson JA, Ohlsson C 2003 Estrogen receptor (ER)-|8 reduces ERα-regulated gene transcription, supporting a "Ying Yang" relationship between ERα and ERβ in mice. Mol Endocrinol 17:203-208 

Vandenput L, Ederveen AG, Erben RG, Stahr K, Swinnen JV, Van Herck E, Verstuyf A, Boonen S, Bouillon R, Vanderschueren D 2001 Testosterone prevents orchidectomy-induced bone loss in estrogen receptor-a knockout mice. Biochem Biophys Res Commun 285:70-76 

Sims NA, Dupont S, Resche-Rigon M, Clement-Lacroix P, Bouali Y, DaPonte F, Galien R, Gaillard-Kelly M, Baron R 2000 In vivo analysis of male and female estrogen receptor a, f and double knockouts reveals a dual role for ERf in bone remodeling. J Bone Miner Res 15(Suppl 1):S160 (Abstract) 

Sims NA, Clement-Lacroix P, Minet D, Dupont S, Krust A, Resche-Rigon M, Gaillard-Kelly M, Chambon PRB 2001 Estrogen receptor a is the major receptor regulating bone response to estradiol in gonadectomized female mice and testosterone plays a role in intact male and female ERα knockout mice. J Bone Miner Res 16(Suppl 1):S431 (Abstract) 

Sims NA, Dupont S, Krust A, Clement-Lacroix P, Minet D, Resche-Rigon M, Gaillard-Kelly M, Baron R 2002 Deletion of estrogen receptors reveals a regulatory role for estrogen receptors-f in bone remodeling in females but not in males. Bone 30:18-25 

McCauley LK, Brogley M, Tozüm TF, Patel K, Oppenlander M, Scheller A, Robins DM, Analysis of androgen and estrogen action in mice deficient for both AR and ERα. Program of the 84th Annual Meeting of The Endocrine Society, San Francisco, CA, 2002, p 83 (Abstract OR14-2) 

Windahl SH, Vidal O, Andersson G, Gustafsson JA, Ohlsson C 1999 Increased cortical bone mineral content but unchanged trabecular bone mineral density in female ERf (-/-) mice. J Clin Invest 104:895-901 

Windahl SH, Hollberg K, Vidal O, Gustafsson JA, Ohlsson C, Andersson G 2001 Female estrogen receptor f-/- mice are partially protected against age-related trabecular bone loss. J Bone Miner Res 16:1388-1398 

McDougall KE, Perry MJ, Tobias JH 2001 Mice with a targeted gene deletion in estrogen receptor-f show enhanced osteogenic responsiveness to 17f-estradiol. Bone 28(Suppl 1):S99 (Abstract) 

Ke HZ, Chidsey-Frink KL, Qi H, Crawford DT, Simmons HA, Brown TA, Petersen DN, Allen MR, McNeish JD, Thompson DD 2001 The role of estrogen receptor-f (ER-f ) in the early age-related bone gain and later age-related bone loss. J Bone Miner Res 16(Suppl 1):S160 (Abstract) 

Ke HZ, Brown TA, Chidsey-Frink KL, Crawford DD, Simmons HA, Petersen DN, Allen MR, McNeish JD, Thompson DD 2000 Estrogen is efficacious in preventing ovariectomy-induced cancellous and cortical bone loss in estrogen receptor-f knockout (BERKO) mice. J Bone Miner Res 15(Suppl 1):S161 (Abstract) 

Ke HZ, Qi H, Crawford DD, Simmons HA, Zhang Q, Brown TA, Petersen DN, Jee WSS, Thompson DD 2002 Improved survival rate and bone status in senile female mice lacking estrogen recep- tor-f. J Bone Miner Res 17(Suppl 1):S133 (Abstract) 

McDougall KE, Perry MJ, Gibson RL, Bright JM, Colley SM, Hodgin JB, Smithies O, Tobias JH 2002 Estrogen-induced osteogenesis in intact female mice lacking ERß. Am J Physiol Endocrinol Metab 283:E817-E823 

Chang CS, Kokontis J, Liao ST 1988 Molecular cloning of human and rat complementary DNA encoding androgen receptors. Science 240:324-326 

Lubahn DB, Joseph DR, Sar M, Tan J, Higgs HN, Larson RE, French FS, Wilson EM 1988 The human androgen receptor: complementary deoxyribonucleic acid cloning, sequence analysis and gene expression in prostate. Mol Endocrinol 2:1265-1275 

Green S, Walter P, Kumar V, Krust A, Bornert JM, Argos P, Chambon P 1986 Human oestrogen receptor cDNA: sequence, expression and homology to v-erb-A. Nature 320:134-139 

Greene GL, Gilna P, Waterfield M, Baker A, Hort Y, Shine J 1986 Sequence and expression of human estrogen receptor complementary DNA. Science 231:1150-1154 

Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA 1996 Cloning of a novel estrogen receptor expressed in rat prostate and ovary. Proc Natl Acad Sci USA 93:5925-5930 

Nilsson S, Makela S, Treuter E, Tujague M, Thomsen J, Andersson G, Enmark E, Pettersson K, Warner M, Gustafsson JA 2001 Mechanisms of estrogen action. Physiol Rev 81:1535-1565 

Kousteni S, Bellido T, Plotkin LI, O'Brien CA, Bodenner DL, Han L, Han K, DiGregorio GB, Katzenellenbogen JA, Katzenellenbogen BS, Roberson PK, Weinstein RS, Jilka RL, Manolagas SC 

2001 Nongenotropic, sex-nonspecific signaling through the estrogen or androgen receptors: dissociation from transcriptional activity. Cell 104:719-730 

Heinlein CA, Chang C 2002 Androgen receptor (AR) coregulators: an overview. Endocr Rev 23:175-200 

McEwen BS, Alves SE 1999 Estrogen actions in the central nervous system. Endocr Rev 20:279-307 

Brubaker KD, Gay CV 1999 Evidence for plasma membranemediated effects of estrogen. Calcif Tissue Int 64:459-462 

Wehling M 1997 Specific, nongenomic actions of steroid hormones. Annu Rev Physiol 59:365-393 

Endoh H, Sasaki H, Maruyama K, Takeyama K, Waga I, Shimizu T, Kato S, Kawashima H 1997 Rapid activation of MAP kinase by estrogen in the bone cell line. Biochem Biophys Res Commun 235:99-102 

Kameda T, Mano H, Yuasa T, Mori Y, Miyazawa K, Shiokawa M, Nakamaru Y, Hiroi E, Hiura K, Kameda A, Yang NN, Hakeda Y, Kumegawa M 1997 Estrogen inhibits bone resorption by directly inducing apoptosis of the bone-resorbing osteoclasts. J Exp Med 186:489-495 

Kousteni S, Han L, Chen JR, Almeida M, Plotkin LI, Bellido T, Manolagas SC 2003 Kinase-mediated regulation of common transcription factors accounts for the bone-protective effects of sex steroids. J Clin Invest 111:1651-1664 

Kousteni S, Chen JR, Bellido T, Han L, Ali AA, O'Brien CA, Plotkin L, Fu Q, Mancino AT, Wen Y, Vertino AM, Powers CC, Stewart SA, Ebert R, Parfitt AM, Weinstein RS, Jilka RL, Manolagas SC 2002 Reversal of bone loss in mice by nongenotropic signaling of sex steroids. Science 298:843-846 

Manolagas SC, Kousteni S, Jilka RL 2002 Sex steroids and bone. Recent Prog Horm Res 57:385-409 

Sims NA, Clement-Lacroix P, Minet D, Fraslon-Vanhulle C, Gaillard-Kelly M, Resche-Rigon M, Baron R 2003 A functional androgen receptor is not sufficient to allow estradiol to protect bone after gonadectomy in estradiol receptor-deficient mice. J Clin Invest 111:1319-1327 

Lorenzo J 2003 A new hypothesis for how sex steroid hormones regulate bone mass. J Clin Invest 111:1641-1643 

Carrascosa A, Audi L, Ferrandez MA, Ballabriga A 1990 Biological effects of androgens and identification of specific dihydrotestosterone-binding sites in cultured human fetal epiphyseal chondrocytes. J Clin Endocrinol Metab 70:134-140 

Abu EO, Horner A, Kusec V, Triffitt JT, Compston JE 1997 The localization of androgen receptors in human bone. J Clin Endocrinol Metab 82:3493-3497 

Ben-Hur H, Thole HH, Mashiah A, Insler V, Berman V, Shezen E, Elias D, Zuckerman A, Ornoy A 1997 Estrogen, progesterone and testosterone receptors in human fetal cartilaginous tissue: immunohistochemical studies. Calcif Tissue Int 60:520-526 

Noble B, Routledge J, Stevens H, Hughes I, Jacobson W 1999 Androgen receptors in bone-forming tissue. Horm Res 51:31-36 

Nilsson O, Chrysis D, Pajulo O, Boman A, Holst M, Rubinstein J, Ritzen ME, Savendahl L, Expression of ERα, ERβ and AR in the human pubertal growth plate. Program of the 84th Annual Meeting of The Endocrine Society, San Francisco, CA, 2002, p 439 (Abstract P2-507) 

van der Eerden BC, van Til NP, Brinkmann AO, Lowik CW, Wit JM, Karperien M 2002 Gender differences in expression of androgen receptor in tibial growth plate and metaphyseal bone of the rat. Bone 30:891-896 

Bord S, Horner A, Beavan S, Compston J 2001 Estrogen receptors a and f are differentially expressed in developing human bone. J Clin Endocrinol Metab 86:2309-2314 

Kusec V, Virdi AS, Prince R, Triffitt JT 1998 Localization of estrogen receptor-a in human and rabbit skeletal tissues. J Clin Endocrinol Metab 83:2421-2428 

Ben-Hur H, Mor G, Blickstein I, Likhman I, Kohen F, Dgani R, Insler V, Yaffe P, Ornoy A 1993 Localization of estrogen receptors in long bones and vertebrae of human fetuses. Calcif Tissue Int 53:91-96 

Braidman IP, Davenport LK, Carter DH, Selby PL, Mawer EB, Freemont AJ 1995 Preliminary in situ identification of estrogen target cells in bone. J Bone Miner Res 10:74-80 

Nilsson O, Abad V, Chrysis D, Ritzen EM, Savendahl L, Baron J 2002 Estrogen receptor-a and-f are expressed throughout postnatal development in the rat and rabbit growth plate. J Endocrinol 173:407-414 

Kennedy J, Baris C, Hoyland JA, Selby PL, Freemont AJ, Braidman IP 1999 Immunofluorescent localization of estrogen receptor-a in growth plates of rabbits, but not in rats, at sexual maturity. Bone 24:9-16 

Nasatzky E, Schwartz Z, Soskolne WA, Brooks BP, Dean DD, Boyan BD, Ornoy A 1994 Evidence for receptors specific for 17 f-estradiol and testosterone in chondrocyte cultures. Connect Tissue Res 30:277-294 

Nilsson LO, Boman A, Savendahl L, Grigelioniene G, Ohlsson C, Ritzen EM, Wroblewski J 1999 Demonstration of estrogen receptor-f immunoreactivity in human growth plate cartilage. J Clin Endocrinol Metab 84:370-373 

Colvard DS, Eriksen EF, Keeting PE, Wilson EM, Lubahn DB, French FS, Riggs BL, Spelsberg TC1989 Identification of androgen receptors in normal human osteoblast-like cells. Proc Natl Acad Sci USA 86:854- 857 

Benz DJ, Haussler MR, Thomas MA, Speelman B, Komm BS 1991 High-affinity androgen binding and androgenic regulation of a 1(I)- procollagen and transforming growth factor-f steady state messenger ribonucleic acid levels in human osteoblast-like osteosarcoma cells. Endocrinology 128:2723-2730 

Zhuang YH, Blauer M, Pekki A, Tuohimaa P 1992 Subcellular location of androgen receptor in rat prostate, seminal vesicle and human osteosarcoma MG-63 cells. J Steroid Biochem Mol Biol 41: 693-696 

Wiren KM, Zhang X, Chang C, Keenan E, Orwoll ES 1997 Transcriptional up-regulation of the human androgen receptor by androgen in bone cells. Endocrinology 138:2291-2300 

Kasperk C, Helmboldt A, Borcsok I, Heuthe S, Cloos O, Niethard F, Ziegler R 1997 Skeletal site-dependent expression of the androgen receptor in human osteoblastic cell populations. Calcif Tissue Int 61:464-473 

Wiren K, Keenan E, Zhang X, Ramsey B, Orwoll E 1999 Homologous androgen receptor up-regulation in osteoblastic cells may be associated with enhanced functional androgen responsiveness. Endocrinology 140:3114-3124 

Czerwiec FS, Liaw JJ, Liu SB, Perez-Stable C, Grumbles R, Howard GA, Roos BA, Burnstein KL 1997 Absence of androgenmediated transcriptional effects in osteoblastic cells despite presence of androgen receptors. Bone 21:49-56 

Wiren KM, Chapman EA, Zhang XW 2002 Osteoblast differentiation influences androgen and estrogen receptor-a and-f expression. J Endocrinol 175:683-694 

Gruber R, Czerwenka K, Wolf F, Ho GM, Willheim M, Peterlik M 1999 Expression of the vitamin D receptor, of estrogen and thyroid hormone receptor a- and ß-isoforms, and of the androgen receptor in cultures of native mouse bone marrow and of stromal/ osteoblastic cells. Bone 24:465-473 

Orwoll ES, Stribrska L, Ramsey EE, Keenan EJ 1991 Androgen receptors in osteoblast-like cell lines. Calcif Tissue Int 49:183-187 

Liesegang P, Romalo G, Sudmann M, Wolf L, Schweikert HU 1994 Human osteoblast-like cells contain specific, saturable, high- affinity glucocorticoid, androgen, estrogen, and 1 a,25-dihydroxy- cholecalciferol receptors. J Androl 15:194-199 

Takeuchi M, Kakushi H, Tohkin M1994 Androgens directly stimulate mineralization and increase androgen receptors in human osteoblast-like osteosarcoma cells. Biochem Biophys Res Commun 204:905-911 

Masuyama A, Ouchi Y, Sato F, Hosoi T, Nakamura T, Orimo H 1992 Characteristics of steroid hormone receptors in cultured MC3T3-E1 osteoblastic cells and effect of steroid hormones on cell proliferation. Calcif Tissue Int 51:376-381 

Bland R 2000 Steroid hormone receptor expression and action in bone. Clin Sci (Lond) 98:217-240 

Hofbauer LC, Hicok KC, Schroeder MJ, Harris SA, Robinson JA, Khosla S 1997 Development and characterization of a conditionally immortalized human osteoblastic cell line stably transfected with the human androgen receptor gene. J Cell Bio- chem 66:542-551 

Eriksen EF, Colvard DS, Berg NJ, Graham ML, Mann KG, Spelsberg TC, Riggs BL 1988 Evidence of estrogen receptors in normal human osteoblast-like cells. Science 241:84-86 

Komm BS, Terpening CM, Benz DJ, Graeme KA, Gallegos A, Korc M, Greene GL, O'Malley BW, Haussler MR 1988 Estrogen binding, receptor mRNA, and biologic response in osteoblast-like osteosarcoma cells. Science 241:81-84 

Kaplan FS, Fallon MD, Boden SD, Schmidt R, Senior M, Haddad JG 1988 Estrogen receptors in bone in a patient with polyostotic fibrous dysplasia (McCune-Albright syndrome). N Engl J Med 319:421-425 

Komm BS, Bodine PVN 2001 Regulation of bone cell function by estrogens. In: Marcus R, Feldman D, Kelsey JL, eds. Osteoporosis, 2nd ed. San Diego: Academic Press; 305-337 

Spelsberg TC, Subramaniam M, Riggs BL, Khosla S 1999 The actions and interactions of sex steroids and growth factors/ cytokines on the skeleton. Mol Endocrinol 13:819-828 

Onoe Y, Miyaura C, Ohta H, Nozawa S, Suda T 1997 Expression of estrogen receptor ß in rat bone. Endocrinology 138:4509-4512 

Arts J, Kuiper GG, Janssen JM, Gustafsson JA, Lowik CW, Pols HA, van Leeuwen JP 1997 Differential expression of estrogen receptors a and ß mRNA during differentiation of human osteoblast SV-HFO cells. Endocrinology 138:5067-5070 

Bodine PV, Henderson RA, Green J, Aronow M, Owen T, Stein GS, Lian JB, Komm BS 1998 Estrogen receptor-a is developmentally regulated during osteoblast differentiation and contributes to selective responsiveness of gene expression. Endocrinology 139: 2048-2057 

Pederson L, Kremer M, Judd J, Pascoe D, Spelsberg TC, Riggs BL, Oursler MJ 1999 Androgens regulate bone resorption activity of isolated osteoclasts in vitro. Proc Natl Acad Sci USA 96:505-510 

Mizuno Y, Hosoi T, Inoue S, Ikegami A, Kaneki M, Akedo Y, Nakamura T, Ouchi Y, Chang C, Orimo H 1994 Immunocyto- chemical identification of androgen receptor in mouse osteoclastlike multinucleated cells. Calcif Tissue Int 54:325-326 

Weinstein RS, Jilka RL, Parfitt AM, Manolagas SC 1997 The effects of androgen deficiency on murine bone remodeling and bone mineral density are mediated via cells of the osteoblastic lineage. Endocrinology 138:4013-4021 

Chen JRK, S, Bellido T, Han L, Di Gregorio GB, Jilka RL, Manolagas SC 2001 Gender-independent induction of murine osteoclast apoptosis in vitro by either estrogens or non-aromatizable androgens. J Bone Miner Res 16 Suppl:S159 (Abstract) 

Fiorelli G, Gori F, Petilli M, Tanini A, Benvenuti S, Serio M, Bernabei P, Brandi ML 1995 Functional estrogen receptors in a human preosteoclastic cell line. Proc Natl Acad Sci USA 92: 2672-2676 

Oursler MJ, Osdoby P, Pyfferoen J, Riggs BL, Spelsberg TC 1991 

Avian osteoclasts as estrogen target cells. Proc Natl Acad Sci USA 88:6613- 6617 

Pederson L, Kremer M, Foged NT, Winding B, Ritchie C, Fitzpatrick LA, Oursler MJ 1997 Evidence of a correlation of estrogen receptor level and avian osteoclast estrogen responsiveness. J Bone Miner Res 12:742-752 

Hoyland JA, Mee AP, Baird P, Braidman IP, Mawer EB, Freemont AJ 1997 Demonstration of estrogen receptor mRNA in bone using in situ reverse-transcriptase polymerase chain reaction. Bone 20: 87-92 

Gunhan M, Gunhan O, Celasun B, Mutlu M, Bostanci H 1998 Estrogen and progesterone receptors in the peripheral giant cell granulomas of the oral cavity. J Oral Sci 40:57-60 

Pensler JM, Radosevich JA, Higbee R, Langman CB 1990 Osteoclasts isolated from membranous bone in children exhibit nuclear estrogen and progesterone receptors. J Bone Miner Res 5:797-802 

Oursler MJ, Pederson L, Fitzpatrick L, Riggs BL, Spelsberg T 1994 Human giant cell tumors of the bone (osteoclastomas) are estrogen target cells. Proc Natl Acad Sci USA 91:5227-5231 

Sunyer T, Lewis J, Collin-Osdoby P, Osdoby P 1999 Estrogen's bone-protective effects may involve differential IL-1 receptor regulation in human osteoclast-like cells. J Clin Invest 103: 1409-1418 

Mano H, Yuasa T, Kameda T, Miyazawa K, Nakamaru Y, Shiokawa M, Mori Y, Yamada T, Miyata K, Shindo H, Azuma H, Hakeda Y, Kumegawa M 1996 Mammalian mature osteoclasts as estrogen target cells. Biochem Biophys Res Commun 223:637-642 

Vidal O, Kindblom LG, Ohlsson C 1999 Expression and localization of estrogen receptor-f in murine and human bone. J Bone Miner Res 14:923-929 

Braidman IP, Hainey L, Batra G, Selby PL, Saunders PT, Hoyland JA 2001 Localization of estrogen receptor f protein expression in adult human bone. J Bone Miner Res 16:214-220 

CollierFM, Huang WH, Holloway WR, Hodge JM, Gillespie MT, Daniels LL, Zheng MH, Nicholson GC 1998 Osteoclasts from human giant cell tumors of bone lack estrogen receptors. Endocrinology 139:1258-1267 

Windahl SH, Norgard M, Kuiper GG, Gustafsson JA, Andersson G 2000 Cellular distribution of estrogen receptor f in neonatal rat bone. Bone 26:117-121 

Oreffo RO, Kusec V, Virdi AS, Flanagan AM, Grano M, Zambonin-Zallone A, Triffitt JT 1999 Expression of estrogen receptor-a in cells of the osteoclastic lineage. Histochem Cell Biol 111:125-133 

Huang WH, Lau AT, Daniels LL, Fujii H, Seydel U, Wood DJ, Papadimitriou JM, Zheng MH 1998 Detection of estrogen receptor a, carbonic anhydrase II and tartrate-resistant acid phosphatase mRNAs in putative mononuclear osteoclast precursor cells of neonatal rats by fluorescence in situ hybridization. J Mol Endocrinol 20:211-219 

Mantalaris A, Panoskaltsis N, Sakai Y, Bourne P, Chang C, Messing EM, Wu JH 2001 Localization of androgen receptor expression in human bone marrow. J Pathol 193:361-366 

Lim SK, Won YJ, Lee HC, Huh KB, Park YS 1999 A PCR analysis of ERα and ERf mRNA abundance in rats and the effect of ovariectomy. J Bone Miner Res 14:1189-1196 

Bonnelye E, Aubin JE 2002 Differential expression of estrogen receptor-related receptor a and estrogen receptors a and f in osteoblasts in vivo and in vitro. J Bone Miner Res 17:1392-1400 

Bellido T, Girasole G, Passeri G, Yu XP, Mocharla H, Jilka RL, Notides A, Manolagas SC 1993 Demonstration of estrogen and vitamin D receptors in bone marrow-derived stromal cells: up- regulation of the estrogen receptor by 1,25-dihydroxyvitamin-D3. Endocrinology 133:553-562 

Qu Q, Perala-Heape M, Kapanen A, Dahllund J, Salo J, Vaananen HK, Harkonen P 1998 Estrogen enhances differentiation of osteoblasts in mouse bone marrow culture. Bone 22:201-209 

Corvol MT, Carrascosa A, Tsagris L, Blanchard O, Rappaport R 1987 Evidence for a direct in vitro action of sex steroids on rabbit cartilage cells during skeletal growth: influence of age and sex. Endocrinology 120:1422-1429  

binding proteins in serum and other biological fluids: regulation and functions. Endocr Rev 18:801-831 

Canalis E 1997 Insulin-like growth factors and osteoporosis. Bone 21:215-216 

Gori F, Hofbauer LC, Conover CA, Khosla S 1999 Effects of androgens on the insulin-like growth factor system in an androgen- responsive human osteoblastic cell line. Endocrinology 140:5579- 5586 

Canalis E, Centrella M, McCarthy TL 1991 Regulation of insulinlike growth factor-II production in bone cultures. Endocrinology 129:2457-2462 

Zhang J, Pugh TD, Stebler B, Ershler WB, Keller ET 1998 Orchiectomy increases bone marrow interleukin-6 levels in mice. Calcif Tissue Int 62:219-226 

Hofbauer LC, Khosla S 1999 Androgen effects on bone metabolism: recent progress and controversies. Eur J Endocrinol 140: 271-286 

Bellido T, Jilka RL, Boyce BF, Girasole G, Broxmeyer H, Dalrymple SA, Murray R, Manolagas SC 1995 Regulation of interleukin-6, osteoclastogenesis, and bone mass by androgens. The role of the androgen receptor. J Clin Invest 95:2886-2895 Hofbauer LC, Ten RM, Khosla S 1999 The anti-androgen hy- droxyflutamide and androgens inhibit interleukin-6 production by an androgen-responsive human osteoblastic cell line. J Bone Miner Res 14:1330-1337 

Lin SC, Yamate T, Taguchi Y, Borba VZ, Girasole G, O'Brien CA, Bellido T, Abe E, Manolagas SC 1997 Regulation of the gp80 and gp130 subunits of the IL-6 receptor by sex steroids in the murine bone marrow. J Clin Invest 100:1980-1990 

Fukayama S, Tashjian Jr AH 1989 Direct modulation by estradiol of the response of human bone cells (SaOS-2) to human parathyroid hormone (PTH) and PTH-related protein. Endocrinology 124: 397-401 

Pivirotto LA, Cissel DS, Keeting PE 1995 Sex hormones mediate interleukin-1 ß production by human osteoblastic HOBIT cells. Mol Cell Endocrinol 111:67-74 

Hofbauer LC, Hicok KC, Chen D, Khosla S 2002 Regulation of osteoprotegerin production by androgens and anti-androgens in human osteoblastic lineage cells. Eur J Endocrinol 147:269-273 Hofbauer LC, Khosla S, Dunstan CR, Lacey DL, Spelsberg TC, Riggs BL 1999 Estrogen stimulates gene expression and protein production of osteoprotegerin in human osteoblastic cells. Endocrinology 140:4367-4370 

Saika M, Inoue D, Kido S, Matsumoto T 2001 17ß-estradiol stimulates expression of osteoprotegerin by a mouse stromal cell line, ST-2, via estrogen receptor-a. Endocrinology 142:2205-2212 Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT, Martin TJ 1999 Modulation of osteoclast differentiation and functionby the new members of the tumor necrosis factor receptor and ligand families. Endocr Rev 20:345-357 

Jilka RL, Weinstein RS, Takahashi K, Parfitt AM, Manolagas SC 1996 Linkage of decreased bone mass with impaired osteoblasto- genesis in a murine model of accelerated senescence. J Clin Invest 97:1732-1740 

Manolagas SC 2000 Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev 21:115-137 

Huber DM, Bendixen AC, Pathrose P, Srivastava S, Dienger KM, Shevde NK, Pike JW 2001 Androgens suppress osteoclast formation induced by RANKL and macrophage-colony stimulating factor. Endocrinology 142:3800-3808 

Hughes DE, Dai A, Tiffee JC, Li HH, Mundy GR, Boyce BF 1996 Estrogen promotes apoptosis of murine osteoclasts mediated by TGF-ß. Nat Med 2:1132-1136 

Kimmel DB 1991 Quantitative histologic changes in the proximal tibial growth cartilage of aged female rats. Cells Materials Suppl. 1:11-18 

Kalu DN 1991 The ovariectomized rat model of postmenopausal bone loss. Bone Miner 15:175-192 

Wronski TJ, Walsh CC, Ignaszewski LA 1986 Histologic evidence for osteopenia and increased bone turnover in ovariectomized rats. Bone 7:119-123 

Kalu DN, Liu CC, Salerno E, Hollis B, Echon R, Ray M 1991  

Skeletal response of ovariectomized rats to low and high doses of 17|8-estradiol. Bone Miner 14:175-187 

Wink CS, Felts WJL 1980 Effects of castration on the bone structure of male rats: a model of osteoporosis. Calcif Tissue Int 32:77-82 

Verhas M, Schoutens A, L'Hermite-Baleriaux M, Dourov N, Verschaeren A, Mone M, Heilporn A 1986 The effect of orchi- dectomy on bone metabolism in aging rats. Calcif Tissue Int 39:74-77 

Turner RT, Hannon KS, Demers LM, Buchanan J, Bell NH 1989 Differential effects of gonadal function on bone histomorphometry in male and female rats. J Bone Miner Res 4:557-563 

Wakley GK, Schutte Jr HD, Hannon KS, Turner RT 1991 Androgen treatment prevents loss of cancellous bone in the orchidecto- mized rat. J Bone Miner Res 6:325-330 

Vanderschueren D, Van Herck E, Suiker AM, Visser WJ, Schot LP, Bouillon R 1992 Bone and mineral metabolism in aged male rats: short and long term effects of androgen deficiency. Endocrinology 130:2906-2916 

Gunness M, Orwoll E 1995 Early induction of alterations in cancellous and cortical bone histology after orchiectomy in mature rats. J Bone Miner Res 10:1735-1744 

Erben RG, Eberle J, Stahr K, Goldberg M 2000 Androgen deficiency induces high turnover osteopenia in aged male rats: a sequential histomorphometric study. J Bone Miner Res 15:1085-1098 

Ederveen AGH, Kloosterboer HJ 1999 Tibolone, a steroid with a tissue-specific hormonal profile, completely prevents ovariectomy- induced bone loss in sexually mature rats. J Bone Miner Res 14: 1963-1970 

Gaumet-Meunier N, Coxam V, Robins S, Pastoureau P, Pointillart A, Davicco MJ, Lebecque P, Barlet JP 2000 Gonadal steroids and bone metabolism in young castrated male rats. Calcif Tissue Int 66:470-475 

Vanderschueren D, Jans I, Van Herck E, Moermans K, Verhaeghe J, Bouillon R 1994 Time-related increase of biochemical markers of bone turnover in androgen-deficient male rats. Bone Miner 26: 123-131 

Turner RT, Riggs BL, Spelsberg TC 1994 Skeletal effects of estrogen. Endocr Rev 15:275-300 

Wakley GK, Turner RT 1991 Sex steroids and the regulation of bone volume in the rat. Cells Materials Suppl 1:85-91 

Kalu DN, Liu CC, Hardin RR, Hollis BW 1989 The aged rat model of ovarian hormone deficiency bone loss. Endocrinology 124:7-16 

Lea CK, Moxham V, Reed MJ, Flanagan AM 1998 Androstenedi- one treatment reduces loss of cancellous bone volume in ovariec- tomised rats in a dose-responsive manner and the effect is not mediated by oestrogen. J Endocrinol 156:331-339 

Schoutens A, Verhas M, L'Hermite-Baleriaux M, L'Hermite M, Verschaeren A, Dourov N, Mone M, Heilporn A, Tricot A 1984 Growth and bone haemodynamic responses to castration in male rats. Reversibility by testosterone. Acta Endocrinologica 107: 428- 432 

Yao W, Jee WSS, Chen J, Liu H, Tam CS, Cui L, Zhou H, Setterberg RB, Frost HM 2000 Making rats rise to erect bipedal stance for feeding partially prevented orchidectomy-induced bone loss and added bone to intact rats. J Bone Miner Res 15:1158-1168 

Prakasam G, Yeh JK, Chen M-M, Castro-Magana M, Liang CT, Aloia JF 1999 Effects of growth hormone and testosterone on cortical bone formation and bone density in aged orchiectomized rats. Bone 24:491-497 

Wronski TJ, Lowry PL, Walsh CC, Ignaszewski LA 1985 Skeletal alterations in ovariectomized rats. Calcif Tissue Int 37:324-328 

Turner RT, Lifrak ET, Beckner M, Wakley GK, Hannon KS, Parker LN 1990 Dehydroepiandrosterone reduces cancellous bone osteopenia in ovariectomized rats. Am J Physiol 258:E673-E677 

Zhang XZ, Kalu DN, Erbas B, Hopper JL, Seeman E 1999 The effects of gonadectomy on bone size, mass, and volumetric density in growing rats are gender-, site-, and growth hormone-specific. J Bone Miner Res 14:802- 809 

Kim BT, Mosekilde L, Duan Y, Zhang XZ, Tornvig L, Thomsen JS, Seeman E 2003 The structural and hormonal basis of sex differences in peak appendicular bone strength in rats. J Bone Miner Res 18:150-155 

Vandenput L 2002 The role of aromatization of androgens in oestrogens and of the oestrogen receptors in the development and maintenance of the male skeleton. A study in rats and mice. Acta Biomedica Lovaniensia. Leuven: Leuven University Press 

Broulik PD 2000 Tamoxifen prevents bone loss in castrated male mice. Horm Metab Res 32:181-184 

Bain SD, Bailey MC, Celino DL, Lantry MM, Edwards MW 1993 High-dose estrogen inhibits bone resorption and stimulates bone formation in the ovariectomized mouse. J Bone Miner Res 8: 435-442 

Daci E, Verstuyf A, Moermans K, Bouillon R, Carmeliet G 2000 Mice lacking the plasminogen activator inhibitor 1 are protected from trabecular bone loss induced by estrogen deficiency. J Bone Miner Res 15:1510-1516 

Poli V, Balena R, Fattori E, Markatos A, Yamamoto M, Tanaka H, Ciliberto G, Rodan GA, Costantini F 1994 Interleukin-6 deficient mice are protected from bone loss caused by estrogen depletion. EMBO J 13:1189-1196 

Onoe Y, Miyaura C, Ito M, Ohta H, Nozawa S, Suda T 2000 Comparative effects of estrogen and raloxifene on B lymphopoiesis and bone loss induced by sex steroid deficiency in mice. J Bone Miner Res 15:541-549 

Ederveen AG, Kloosterboer HJ 2001 Tibolone exerts its protective effect on trabecular bone loss through the estrogen receptor. J Bone Miner Res 16:1651-1657 

Vanderschueren D, Vandenput L, Boonen S, Van Herck E, Swin- nen JV, Bouillon R 2000 An aged rat model of partial androgen deficiency: prevention of both loss of bone and lean body mass by low-dose androgen replacement. Endocrinology 141:1642-1647 

Turner RT, Wakley GK, Hannon KS 1990 Differential effects of androgens on cortical bone histomorphometry in gonadectomized male and female rats. J Orthop Res 8:612-617 

Martel C, Sourla A, Pelletier G, Labrie C, Fournier M, Picard S, Li S, Stojanovic M, Labrie F 1998 Predominant androgenic component in the stimulatory effect of dehydroepiandrosterone on bone mineral density in the rat. J Endocrinol 157:433-442 

Lea CK, Flanagan AM 1998 Physiological plasma levels of androgens reduce bone loss in the ovariectomized rat. Am J Physiol 274:E328-E335 

Vandenput L, Boonen S, Van Herck E, Swinnen JV, Bouillon R, Vanderschueren D 2002 Evidence from the aged orchidectomized male rat model that 17|8-estradiol is a more effective bone-sparing and anabolic agent than 5α-dihydrotestosterone. J Bone Miner Res 17:2080-2086 

Tobias JH, Gallagher A, Chambers TJ 1994 5 a-Dihydrotestoster- one partially restores cancellous bone volume in osteopenic ovari- ectomized rats. Am J Physiol 267:E853-E859 

Coxam V, Bowman BM, Mecham M, Roth CM, Miller MA, Miller SC 1996 Effects of dihydrotestosterone alone and combined with estrogen on bone mineral density, bone growth, and formation rates in ovariectomized rats. Bone 19:107-114 

Wronski TJ, Cintron M, Doherty AL, Dann LM 1988 Estrogen treatment prevents osteopenia and depresses bone turnover in ovariectomized rats. Endocrinology 123:681-686 

Fanti P, Monier-Faugere MC, Geng Z, Schmidt J, Morris PE, Cohen D, Malluche HH 1998 The phytoestrogen genistein reduces bone loss in short-term ovariectomized rats. Osteoporos Int 8: 274-281 

Picherit C, Coxam V, Bennetau-Pelissero C, Kati-Coulibaly S, Davicco MJ, Lebecque P, Barlet JP 2000 Daidzein is more efficient than genistein in preventing ovariectomy-induced bone loss in rats. J Nutr 130:1675-1681 

Ishimi Y, Miyaura C, Ohmura M, Onoe Y, Sato T, Uchiyama Y, Ito M, Wang X, Suda T, Ikegami S 1999 Selective effects of genistein, a soybean isoflavone, on B-lymphopoiesis and bone loss caused by estrogen deficiency. Endocrinology 140:1893-1900 

Ishimi Y, Yoshida M, Wakimoto S, Wu J, Chiba H, Wang X, Takeda K, Miyaura C 2002 Genistein, a soybean isoflavone, affects bone marrow lymphopoiesis and prevents bone loss in castrated male mice. Bone 31:180-185 

Moverare S, Venken K, Eriksson AL, Andersson N, Skrtic S, Wergedal J, Mohan S, Salmon P, Bouillon R, Gustafsson JA, Vanderschueren D, Ohlsson C 2003 Differential effects on bone of estrogen receptor a and androgen receptor activation in or- chidectomized adult male mice. Proc Natl Acad Sci USA 100: 13573-13578 

Nuttall ME, Bradbeer JN, Stroup GB, Nadeau DP, Hoffman SJ, Zhao H, Rehm S, Gowen M 1998 ldoxifene: a novel selective estrogen receptor modulator prevents bone loss and lowers cholesterol levels in ovariectomized rats and decreases uterine weight in intact rats. Endocrinology 139:5224-5232 

Martel CL, Picard S, Richard V, Blanger A, Labrie C, Labrie F 2000 Prevention of bone loss by EM-800 and raloxifene in the ovariec- tomized rat. J Steroid Biochem Molec Biol 74:45-56 

Ke HZ, Qi H, Chidsey-Frink KL, Crawford DT, Thompson DD 2001 Lasofoxifene (CP-336,156) protects against the age-related changes in bone mass, bone strength, and total serum cholesterol in intact aged male rats. J Bone Miner Res 16:765-773 

Ke HZ, Qi H, Crawford DT, Chidsey-Frink KL, Simmons HA, Thompson DD 2000 Lasofoxifene (CP-336,156), a selective estrogen receptor modulator, prevents bone loss induced by aging and orchidectomy in the adult rat. Endocrinology 141:1338-1344 

Broulik PD, Starka L 1997 Effect of antiandrogens casodex and epitestosterone on bone composition in mice. Bone 20:473-475 

Robin-Jagerschmidt C, Clement-Lacroix P, Minet D, Hippomène V, Gaillard-Kelly M, Baron R, Resche-Rigon M 2002 A selective androgen receptor modulator (SARM) displays dissociated activity on bone and seminal vesicles in mice. J Bone Miner Res 17(Suppl 1):S132 (Abstract) 

Erlandsson MC, Jonsson CA, Lindberg MK, Ohlsson C, Carlsten H 2002 Raloxifene- and estradiol-mediated effects on uterus, bone and B lymphocytes in mice. J Endocrinol 175:319-327 

Fisher CR, Graves KH, Parlow AF, Simpson ER 1998 Characterization of mice deficient in aromatase (ArKO) because of targeted disruption of the CYP19 gene. Proc Natl Acad Sci USA 95:69656970 

Nemoto Y, Toda K, Ono M, Fujikawa-Adachi K, Saibara T, Onishi S, Enzan H, Okada T, Shizuta Y 2000 Altered expression of fatty acid-metabolizing enzymes in aromatase-deficient mice. J Clin Invest 105:1819-1825 

Krege JH, Hodgin JB, Couse JF, Enmark E, Warner M, Mahler JF, Sar M, Korach KS, Gustafsson JA, Smithies O 1998 Generation and reproductive phenotypes of mice lacking estrogen receptor f. Proc Natl Acad Sci USA 95:15677-15682 

Dupont S, Krust A, Gansmuller A, Dierich A, Chambon P, Mark M 2000 Effect of single and compound knockouts of estrogen receptors a (ERα) and f (ERf) on mouse reproductive phenotypes. Development 127:4277-4291 

Lubahn DB, Moyer JS, Golding TS, Couse JF, Korach KS, Smithies O 1993 Alteration of reproductive function but not prenatal sexual development after insertional disruption of the mouse estrogen receptor gene. Proc Natl Acad Sci USA 90:11162-11166 

Pendaries C, Darblade B, Rochaix P, Krust A, Chambon P, Korach KS, Bayard F, Arnal JF 2002 The AF-1 activation-function of ERα may be dispensable to mediate the effect of estradiol on endothelial NO production in mice. Proc Natl Acad Sci USA 99:2205-2210 

Denger S, Reid G, Kos M, Flouriot G, Parsch D, Brand H, Korach KS, Sonntag-Buck V, Gannon F 2001 ERα gene expression in human primary osteoblasts: evidence for the expression of two receptor proteins. Mol Endocrinol 15:2064-2077 

Vidal O, Lindberg M, Savendahl L, Lubahn DB, Ritzen EM, Gustafsson JA, Ohlsson C 1999 Disproportional body growth in female estrogen receptor-a-inactivated mice. Biochem Biophys Res Commun 265:569-571 

Lindberg M, Moverare S, Eriksson AL, Skrtic S, Gao H, Dahl- man-Wright K, Gustafsson J-A, Ohlsson C 2002 Identification of estrogen-regulated genes of potential importance for the regulation of trabecular bone mineral density. J Bone Miner Res 17:2183-2195 

Kawano HYT, Sato T, Matsumoto T, Kawaguchi H, Kato S 2003 Independent contributions of androgen and estrogen signalings in the maintenance of bone mass in males: analysis of androgen receptor deficient mice. Bone 32(Suppl 1):S94 (Abstract) 

Kawano H, Sato T, Yamada T, Matsumoto T, Sekine K, Wa- tanabe T, Nakamura T, Fukuda T, Yoshimura K, Yoshizawa T, Aihara KI, Yamamoto Y, Nakamichi Y, Metzger D, Chambon 

P, Nakamura K, Kawaguchi H, Kato S 2003 Suppressive function of androgen receptor in bone resorption. Proc Natl Acad Sci USA 100:9416-9421 

Couse JF, Yates MM, Walker VR, Korach KS 2003 Characterization of the hypothalamic-pituitary-gonadal axis in estrogen receptor (ER) null mice reveals hypergonadism and endocrine sex reversal in females lacking ERα but not ERf. Mol Endocrinol 17: 1039-1053 

Ohlsson C, Bengtsson BA, Isaksson OG, Andreassen TT, Sloot- weg MC 1998 Growth hormone and bone. Endocr Rev 19:55-79 

Yakar S, Rosen CJ, Beamer WG, Ackert-Bicknell CL, Wu Y, Liu JL, Ooi GT, Setser J, Frystyk J, Boisclair YR, LeRoith D 2002 Circulating levels of IGF-1 directly regulate bone growth and density. J Clin Invest 110:771-781 

Sjogren K, Liu JL, Blad K, Skrtic S, Vidal O, Wallenius V, LeRoith D, Tornell J, Isaksson OG, Jansson JO, Ohlsson C 1999 Liver- derived insulin-like growth factor I (IGF-I) is the principal source of IGF-I in blood but is not required for postnatal body growth in mice. Proc Natl Acad Sci USA 96:7088-7092 

Sjogren K, Sheng M, Moverare S, Liu JL, Wallenius K, Tornell J, Isaksson O, Jansson JO, Mohan S, Ohlsson C 2002 Effects of liver-derived insulin-like growth factor I on bone metabolism in mice. J Bone Miner Res 17:1977-1987 

Sueyoshi T, Yokomori N, Korach KS, Negishi M 1999 Developmental action of estrogen receptor-a feminizes the growth hor- mone-Stat5b pathway and expression of Cyp2a4 and Cyp2d9 genes in mouse liver. Mol Pharmacol 56:473-477 

Vanderschueren D, Van Herck E, Suiker AMH, Visser WJ, Schot LPC, Chung K, Lucas RS, Einhorn TA, Bouillon R 1993 Bone and mineral metabolism in the androgen-resistant (testicular feminized) male rat. J Bone Miner Res 8:801-809 

Saville PD 1969 Changes in skeletal mass and fragility with castration in the rat: a model of osteoporosis. J Am Geriatr Soc 17: 155-166 

Gurkan L, Ekeland A, Gautvik KM, Langeland N, Ronningen H, Solheim LF 1986 Bone changes after castration in rats. A model for osteoporosis. Acta Orthop Scand 57:67-70 

Ducy P, Schinke T, Karsenty G 2000 The osteoblast: a sophisticated fibroblast under central surveillance. Science 289:1501-1504 

Roggia C, Gao Y, Cenci S, Weitzmann MN, Toraldo G, Isaia G, Pacifici R 2001 Up-regulation of TNF-producing T cells in the bone marrow: a key mechanism by which estrogen deficiency induces bone loss in vivo. Proc Natl Acad Sci USA 98:13960-13965 

Tanner JM 1986 Normal growth and techniques of growth assessment. Clin Endocrinol Metab 15:411-451 

Frost HM 1987 The mechanostat: a proposed pathogenic mechanism of osteoporoses and the bone mass effects of mechanical and nonmechanical agents. Bone Miner 2:73-85 

Bhasin S, Storer TW, Berman N, Yarasheski KE, Clevenger B, Phillips J, Lee WP, Bunnell TJ, Casaburi R 1997 Testosterone replacement increases fat-free mass and muscle size in hypogo- nadal men. J Clin Endocrinol Metab 82:407-413 

Damien E, Price JS, Lanyon LE 1998 The estrogen receptor's in-volvement in osteoblasts' adaptive response to mechanical strain. J Bone Miner Res 13:1275-1282 

Damien E, Price JS, Lanyon LE 2000 Mechanical strain stimulates osteoblast proliferation through the estrogen receptor in males as well as females. J Bone Miner Res 15:2169-2177 

Bikle D, Majumdar S, Laib A, Powell-Braxton L, Rosen C, Beamer W, Nauman E, Leary C, Halloran B 2001 The skeletal structure of insulin-like growth factor I-deficient mice. J Bone Miner Res 16: 2320-2329 

Liu JP, Baker J, Perkins AS, Robertson EJ, Efstratiadis A1993 Mice carrying null mutations of the genes encoding insulin-like growth factor I (Igf-1) and type 1 IGF receptor (Igf1r). Cell 75:59-72 

Zhou Y, Xu BC, Maheshwari HG, He L, Reed M, Lozykowski M, Okada S, Cataldo L, Coschigamo K, Wagner TE, Baumann G, Kopchick JJ 1997 A mammalian model for Laron syndrome produced by targeted disruption of the mouse growth hormone re- ceptor/binding protein gene (the Laron mouse). Proc Natl Acad Sci USA 94:13215-13220 

Sjogren K, Bohlooly YM, Olsson B, Coschigano K, Tornell J, Mohan S, Isaksson OG, Baumann G, Kopchick J, Ohlsson C 2000 

Disproportional skeletal growth and markedly decreased bone mineral content in growth hormone receptor - / - mice. Biochem Biophys Res Commun 267:603-608 

Sims NA, Clement-Lacroix P, Da Ponte F, Bouali Y, Binart N, Moriggl R, Goffin V, Coshigano K, Gaillard-Kelly M, Kopchick J, Baron R, Kelly PA 2000 Bone homeostasis in growth hormone receptor-null mice is restored by IGF-I but independent of Stat5. J Clin Invest 106:1095-1103 

Jansson JO, Eden S, Isaksson O 1985 Sexual dimorphism in the control of growth hormone secretion. Endocr Rev 6:128-150 

Jansson JO, Ekberg S, Isaksson OG, Eden S 1984 Influence of gonadal steroids on age- and sex-related secretory patterns of growth hormone in the rat. Endocrinology 114:1287-1294 

Bouillon R, Bex M, Van Herck E, Laureys J, Dooms L, Lesaffre E, Ravussin E 1995 Influence of age, sex, and insulin on osteoblast function: osteoblast dysfunction in diabetes mellitus. J Clin Endo-crinol Metab 80:1194-1202 

Handelsman DJ, Spaliviero JA, Scott CD, Baxter RC 1987 Hormonal regulation of the peripubertal surge of insulin-like growth factor-I in the rat. Endocrinology 120:491-496 

Wakley GK, Evans GL, Turner RT 1997 Short-term effects of high dose estrogen on tibiae of growing male rats. Calcif Tissue Int 60:37-42 

Weissberger AJ, Ho KK 1993 Activation of the somatotropic axis by testosterone in adult males: evidence for the role of aromatiza- tion. J Clin Endocrinol Metab 76:1407-1412 

Yeh JK, Chen MM, Aloia JF 1997 Effects of 17 β-estradiol admin-istration on cortical and cancellous bone of ovariectomized rats with and without hypophysectomy. Bone 20:413-420 

Keenan BS, Richards GE, Ponder SW, Dallas JS, Nagamani M, Smith ER 1993 Androgen-stimulated pubertal growth: the effects of testosterone and dihydrotestosterone on growth hormone and insulin-like growth factor-I in the treatment of short stature and delayed puberty. J Clin Endocrinol Metab 76:996-1001 

Smith MR 2002 Osteoporosis during androgen deprivation therapy for prostate cancer. Urology 60:79- 85 

Stepan JJ, Lachman M, Zverina J, Pacovsky V, Baylink DJ 1989 Castrated men exhibit bone loss: effect of calcitonin treatment on biochemical indices of bone remodeling. J Clin Endocrinol Metab 69:523-527 

Goldray D, Weisman Y, Jaccard N, Merdler C, Chen J, Matzkin H 1993 Decreased bone density in elderly men treated with the gonadotropin-releasing hormone agonist decapeptyl (D-Trp6- GnRH). J Clin Endocrinol Metab 76:288-290 

Maillefert JF, Sibilia J, Michel F, Saussine C, Javier RM, Tavernier C 1999 Bone mineral density in men treated with synthetic gonadotropin-releasing hormone agonists for prostatic carcinoma. J Urol 161:1219-1222 

Wei JT, Gross M, Jaffe CA, Gravlin K, Lahaie M, Faerber GJ, Cooney KA1999 Androgen deprivation therapy for prostate cancer results in significant loss of bone density. Urology 54:607-611 

Smith MR, McGovern FJ, Zietman AL, Fallon MA, Hayden DL, Schoenfeld DA, Kantoff PW, Finkelstein JS 2001 Pamidronate to prevent bone loss during androgen-deprivation therapy for prostate cancer. N Engl J Med 345:948-955 

Stoch SA, Parker RA, Chen L, Bubley G, Ko YJ, Vincelette A, Greenspan SL 2001 Bone loss in men with prostate cancer treated with gonadotropin-releasing hormone agonists. J Clin Endocrinol Metab 86:2787-2791 

Berruti A, Dogliotti L, Terrone C, Cerutti S, Isaia G, Tarabuzzi R, Reimondo G, Mari M, Ardissone P, De Luca S, Fasolis G, Fontana D, Rocca RS, Angeli A 2002 Changes inbone mineral density, lean body mass and fat content as measured by dual energy x-ray absorptiometry in patients with prostate cancer without apparent bone metastases given androgen deprivation therapy. J Urol 167: 2361-2367 

Diamond T, Campbell J, Bryant C, Lynch W 1998 The effect of combined androgen blockade on bone turnover and bone mineral densities in men treated for prostate carcinoma: longitudinal eval-uation and response to intermittent cyclic etidronate therapy. Cancer 83:1561-1566 

Basaria S, Lieb J, Tang AM, DeWeese T, Carducci M, Eisenberger M, Dobs AS 2002 Long-term effects of androgen deprivation therapy in prostate cancer patients. Clin Endocrinol (Oxf) 56:779-786 

Daniell HW 1997 Osteoporosis after orchiectomy for prostate cancer. J Urol 157:439-444 

Daniell HW, Dunn SR, Ferguson DW, Lomas G, Niazi Z, Stratte PT 2000 Progressive osteoporosis during androgen deprivation therapy for prostate cancer. J Urol 163:181-186 

Kiratli BJ, Srinivas S, Perkash I, Terris MK 2001 Progressive decrease in bone density over 10 years of androgen deprivation therapy in patients with prostate cancer. Urology 57:127-132 

Eriksson S, Eriksson A, Stege R, Carlstrom K 1995 Bone mineral density in patients with prostatic cancer treated with orchidectomy and with estrogens. Calcif Tissue Int 57:97-99 

Mittan D, Lee S, Miller E, Perez RC, Basler JW, Bruder JM 2002 Bone loss following hypogonadism in men with prostate cancer treated with GnRH analogs. J Clin Endocrinol Metab 87:3656-3661 

Smith MR, McGovern FJ, Fallon MA, Schoenfeld D, Kantoff PW, Finkelstein JS 2001 Low bone mineral density in hormone-naive men with prostate carcinoma. Cancer 91:2238-2245 

Townsend MF, Sanders WH, Northway RO, Graham Jr SD 1997 Bone fractures associated with luteinizing hormone-releasing hormone agonists used in the treatment of prostate carcinoma. Cancer 79:545-550 

Hatano T, Oishi Y, Furuta A, Iwamuro S, Tashiro K 2000 Incidence of bone fracture in patients receiving luteinizing hormonereleasing hormone agonists for prostate cancer. BJU Int 86:449-452 

Oefelein MG, Ricchuiti V, Conrad W, Seftel A, Bodner D, Goldman H, Resnick M 2001 Skeletal fracture associated with androgen suppression induced osteoporosis: the clinical incidence and risk factors for patients with prostate cancer. J Urol 166:1724- 1728 

Melton 3rd LJ, Alothman KI, Khosla S, Achenbach SJ, Oberg AL, Zincke H 2003 Fracture risk following bilateral orchiectomy. J Urol 169:1747-1750 

Smith MR, Finkelstein JS, McGovern FJ, Zietman AL, Fallon MA, Schoenfeld DA, Kantoff PW 2002 Changes in body composition during androgen deprivation therapy for prostate cancer. J Clin Endocrinol Metab 87:599-603 

Smith MR, Eastham J, Gleason DM, Shasha D, Tchekmedyian S, Zinner N 2003 Randomized controlled trial of zoledronic acid to prevent bone loss in men receiving androgen deprivation therapy for nonmetastatic prostate cancer. J Urol 169:2008-2012 

Leder BZ, Finkelstein JS 2002 Gonadal steroids and the skeleton in men. Clinical aspects. In: Orwoll ES, Bliziotes M, eds. Osteoporosis. Pathophysiology and clinical management. Totowa, NJ: Humana Press; 393-411 

Francis RM 1999 The effects of testosterone on osteoporosis in men. Clin Endocrinol (Oxf) 50:411-414 

Jackson JA, Kleerekoper M, Parfitt AM, Rao DS, Villanueva AR, Frame B 1987 Bone histomorphometry in hypogonadal and eugo- nadal men with spinal osteoporosis. J Clin Endocrinol Metab 65: 53-58 

Baran DT, Bergfeld MA, Teitelbaum SL, Avioli LV 1978 Effect of testosterone therapy on bone formation in an osteoporotic hypogo-nadal male. Calcif Tissue Res 26:103-106 

Francis RM, Peacock M, Aaron JE, Selby PL, Taylor GA, Thompson J, Marshall DH, Horsman A 1986 Osteoporosis in hypogonadal men: role of decreased plasma 1,25-dihydroxyvi- tamin D, calcium malabsorption, and low bone formation. Bone 7:261-268 

Benito M, Gomberg B, Wehrli FW, Weening RH, Zemel B, Wright AC, Song HK, Cucchiara A, Snyder PJ 2003 Deterioration of tra-becular architecture in hypogonadal men. J Clin Endocrinol Metab 88:1497-1502 

Seeman E, Melton IIILJ, O'Fallon WM, Riggs BL1983 Risk factors for spinal osteoporosis in men. Am J Med 75:977-983 

Stanley HL, Schmitt BP, Poses RM, Deiss WP 1991 Does hypo-gonadism contribute to the occurence of a minimal trauma hip fracture in elderly men? J Am Geriatr Soc 39:766-771 

Boonen S, Vanderschueren D, Cheng XG, Verbeke G, Dequeker J, Geusens P, Broos P, Bouillon R 1997 Age-related (type II) femoral neck osteoporosis in men: biochemical evidence for both hy- povitaminosis D- and androgen deficiency-induced bone resorption. J Bone Miner Res 12:2119-2126 

Leifke E, Körner H-C, Link TM, Behre HM, Peters PE, Nieschlag E 1998 Effects of testosterone replacement therapy on cortical and trabecular bone mineral density, vertebral body area and paraspinal muscle area in hypogonadal men. Eur J Endocrinol 138:51-58 

Finkelstein JS, Neer RM, Biller BM, Crawford JD, Klibanski A 1992 Osteopenia in men with a history of delayed puberty. N Engl J Med 326:600-604 

Bertelloni S, Baroncelli GI, Battini R, Perri G, Saggese G 1995 Short-term effects of testosterone treatment on reduced bone density in boys with constitutional delay of puberty. J Bone Miner Res 10:1488-1495 

Finkelstein JS, Klibanski A, Neer RM 1996 A longitudinal evaluation of bone mineral density in adult men with histories of delayed puberty. J Clin Endocrinol Metab 81:1152-1155 

Bertelloni S, Baroncelli GI, Ferdeghini M, Perri G, Saggese G 1998 Normal volumetric bone mineral density and bone turnover in young men with histories of constitutional delay of puberty. J Clin Endocrinol Metab 83:4280-4283 

Finkelstein JS, Klibanski A, Neer RM, Greenspan SL, Rosenthal DI, Crowley Jr WF 1987 Osteoporosis in men with idiopathic hypogonadotropic hypogonadism. Ann Intern Med 106:354-361 

Finkelstein JS, Klibanski A, Neer RM, Doppelt SH, Rosenthal DI, Segre GV, Crowley Jr WF 1989 Increases in bone density during treatment of men with idiopathic hypogonadotropic hypogonadism. J Clin Endocrinol Metab 69:776-783 

Guo C-Y, Jones TH, Eastell R 1997 Treatment of isolated hypogo-nadotropic hypogonadism effect on bone mineral density and bone turnover. J Clin Endocrinol Metab 82:658-665 

Greenspan SL, Neer RM, Ridgway EC, Klibanski A 1986 Osteoporosis in men with hyperprolactinemic hypogonadism. Ann Intern Med 104:777-782 

Greenspan SL, Oppenheim DS, Klibanski A 1989 Importance of gonadal steroids to bone mass in men with hyperprolactinemic hypogonadism. Ann Intern Med 110:526-531 

Luisetto G, Mastrogiacomo I, Bonanni G, Pozzan G, Botteon S, Tizian L, Galuppo P 1995 Bone mass and mineral metabolism in Klinefelter's syndrome. Osteoporos Int 5:455-461 

Smith DAS, Walker MS 1977 Changes in plasma steroids andbone density in Klinefelter's syndrome. Calcif Tissue Res 22:225-228 

Foresta C, Ruzza G, Mioni R, Meneghello A, Baccichetti C 1983 Testosterone and bone loss in Klinefelter syndrome. Horm Metab Res 15:56-57 

Horowitz M, Wishart JM, O'Loughlin PD, Morris HA, Need AG, Nordin BEC 1992 Osteoporosis and Klinefelter's syndrome. Clin Endocrinol (Oxf) 36:113-118 

Wong FH, Pun KK, Wang C 1993 Loss of bone mass in patients with Klinefelter's syndrome despite sufficient testosterone replacement. Osteoporos Int 3:3-7 

van den Bergh JP, Hermus AR, Spruyt AI, Sweep CG, Corstens FH, Smals AG 2001 Bone mineral density and quantitative ultrasound parameters in patients with Klinefelter's syndrome after long-term testosterone substitution. Osteoporos Int 12:55-62 

Munoz-Torres M, Jodar E, Quesada M, Escobar-Jimenez F 1995 Bone mass in androgen-insensitivity syndrome: response to hormonal replacement therapy. Calcif Tissue Int 57:94-96 

Soule SG, Conway G, Prelevic GM, Prentice M, Ginsburg J, Jacobs HS 1995 Osteopenia as a feature of the androgen insensitivity syndrome. Clin Endocrinol (Oxf) 43:671-675 

Mizunuma H, Soda M, Okano H, Kagami I, Miyamoto S, Ohsawa M, Ibuki Y 1998 Changes in bone mineral density after orchidec- tomy and hormone replacement therapy in individuals with androgen insensitivity syndrome. Hum Reprod 13:2816-2818 

Bertelloni S, Baroncelli GI, Federico G, Cappa M, Lala R, Saggese G 1998 Altered bone mineral density in patients with complete androgen insensitivity syndrome. Horm Res 50:309-314 

Marcus R, Leary D, Schneider DL, Shane E, Favus M, Quigley CA 2000 The contribution of testosterone to skeletal development and maintenance: lessons from the androgen insensitivity syndrome. J Clin Endocrinol Metab 85:1032-1037 

Orwoll ES 2003 Toward an expanded understanding of the role of the periosteum in skeletal health. J Bone Miner Res 18:949-954 

Ahlborg HG, Johnell O, Turner CH, Rannevik G, Karlsson MK 2003 Bone loss and bone size after menopause. N Engl J Med 349:327-334 

Seeman E 2003 Periosteal bone formation-a neglected determinant of bone strength. N Engl J Med 349:320-323 

Labrie F, Belanger A, Cusan L, Gomez J-L, CandasB 1997 Marked decline in serum concentrations of adrenal C19 sex steroid precursors and conjugated androgen metabolites during aging. Endocrinology 82:2396-2402 

Zborowski JV, Cauley JA, Talbott EO, Guzick DS, Winters SJ 2000 Bone mineral density, androgens, and the polycystic ovary: the complex and controversial issue of androgenic influence in female bone. J Clin Endocrinol Metab 85:3496-3506 

Buchanan JR, Hospodar P, Myers C, Leuenberger P, Demers LM 1988 Effect of excess endogenous androgens on bone density in young women. J Clin Endocrinol Metab 67:937-943 

Dagogo-Jack S, al-Ali N, Qurttom M 1997 Augmentation of bone mineral density in hirsute women. J Clin Endocrinol Metab 82: 2821-2825 

Good C, Tulchinsky M, Mauger D, Demers LM, Legro RS 1999 Bone mineral density and body composition in lean women with polycystic ovary syndrome. Fertil Steril 72:21-25 

Bertelloni S, Baroncelli GI, Sorrentino MC, Costa S, Battini R, Saggese G 1997 Androgen-receptor blockade does not impair bone mineral density in adolescent females. Calcif Tissue Int 61:1-5 

Prezelj J, Kocijancic A 1994 Antiandrogen treatment with spi-ronolactone and linestrenol decreases bone mineral density in eu- menorrhoeic women with androgen excess. Horm Metab Res 26: 46-48 

Simberg N, Tiitinen A, Silfvast A, Viinikka L, Ylikorkala O 1996 High bone density in hyperandrogenic women: effect of gonado-tropin-releasing hormone agonist alone or in conjunction with estrogen-progestin replacement. J Clin Endocrinol Metab 81: 646- 651 

Moghetti P, Castello R, Zamberlan N, Rossini M, Gatti D, Negri C, Tosi F, Muggeo M, Adami S 1999 Spironolactone, but not flutamide, administration prevents bone loss in hyperandrogenic women treated with gonadotropin-releasing hormone agonist. J Clin Endocrinol Metab 84:1250-1254 

Gurnell EM, Chatterjee VK 2001 Dehydroepiandrosterone re-placement therapy. Eur J Endocrinol 145:103-106 

Barrett-Connor E, Kritz-Silverstein D, Edelstein SL 1993 A prospective study of dehydroepiandrosterone sulfate (DHEAS) and bone mineral density in older men and women. Am J Epidemiol 137:201-206 

Greendale GA, Edelstein S, Barrett-Connor E 1997 Endogenous sex steroids and bone mineral density in older women and men: the Rancho Bernardo study. J Bone Miner Res 12:1833-1843 

Mauras N, Haymond MW, Darmaun D, Vieira NE, Abrams SA, Yergey AL 1994 Calcium and protein kinetics in prepubertal boys. Positive effects of testosterone. J Clin Invest 93:1014-1019 

Devogelaer JP, De Cooman S, Nagant DD 1992 Low bone mass in hypogonadal males. Effect of testosterone substitution therapy, a densitometric study. Maturitas 15:17-23 

Isaia G, Mussetta M, Pecchio F, Sciolla A, di Stefano M, Molinatti GM 1992 Effect of testosterone on bone in hypogonadal males. Maturitas 15:47-51 

Katznelson L, Finkelstein JS, Schoenfeld DA, Rosenthal DI, Anderson EJ, Klibanski A 1996 Increase in bone density and lean body mass during testosterone administration in men with acquired hypogonadism. J Clin Endocrinol Metab 81:4358-4365 

Behre HM, Kliesch S, Leifke E, Link TM, Nieschlag E 1997 Longterm effect of testosterone therapy on bone mineral density in hypogonadal men. J Clin Endocrinol Metab 82:2386-2390 

Snyder PJ, Peachey H, Berlin JA, Hannoush P, Haddad G, Dlewati A, Santanna J, Loh L, Lenrow DA, Holmes JH, Kapoor SC, Atkinson LE, Strom BL 2000 Effects of testosterone replacement in hypogonadal men. J Clin Endocrinol Metab 85:2670-2677 

Wang C, Swerdloff RS, Iranmanesh A, Dobs A, Snyder PJ, Cunningham G, Matsumoto AM, Weber T, Berman N 2001 Effects of transdermal testosterone gel onbone turnover markers andbone mineral density in hypogonadal men. Clin Endocrinol (Oxf) 54: 739-750 

Wang C, Eyre DR, Clark R, Kleinberg D, Newman C, IranmanesVeldhuis J, Dudley RE, Berman N, Davidson T, Barstow TJ, Sinow R, Alexander G, Swerdloff RS 1996 Sublingual testosterone replacement improves muscle mass and strength, decreases bone resorption, and increases bone formation markers in hypogonadal men-a clinical research center study. J Clin Endocrinol Metab 81: 3654-3662 

Morley JE, Perry HM, Kaiser FE, Kraenzle D, Jensen J, Houston K, Mattamel M 1993 Effects of testosterone replacement therapy in old hypogonadal males: a preliminary study. J Am Geriatr Soc 41:149-152 

Wang C, Swerdloff RS, Iranmanesh A, Dobs A, Snyder PJ, Cunningham G, Matsumoto AM, Weber T, Berman N 2000 Trans-dermal testosterone gel improves sexual function, mood, muscle strength, and body composition parameters in hypogonadal men. Testosterone Gel Study Group. J Clin Endocrinol Metab 85:2839- 2853 

Sih R, Morley JE, Kaiser FE, Perry III HM, Patrick P, Ross C 1997 Testosterone replacement in older hypogonadal men: a 12-month randomized controlled trial. J Clin Endocrinol Metab 82:1661-1667 

Ly LP, Jimenez M, Zhuang TN, Celermajer DS, Conway AJ, Handelsman DJ 2001A double-blind, placebo-controlled, randomized clinical trial of transdermal dihydrotestosterone gel on muscular strength, mobility, and quality of life in older men with partial androgen deficiency. J Clin Endocrinol Metab 86:4078-4088 

Anderson FH, Francis RM, Faulkner K 1996 Androgen supple-mentation in eugonadal men with osteoporosis- effects of 6 months of treatment on bone mineral density and cardiovascular risk factors. Bone 18:171-177 

Vermeulen A 2001 Androgen replacement therapy in the aging male-a critical evaluation. J Clin Endocrinol Metab 86:2380-2390 

Gruenewald DA, Matsumoto AM 2003 Testosterone supplementation therapy for older men: potential benefits and risks. J Am Geriatr Soc 51:101-115 

Khosla S, Melton 3rd LJ, Atkinson EJ, O'Fallon WM, Klee GG, Riggs BL 1998 Relationship of serum sex steroid levels and bone turnover markers with bone mineral density in men and women: a key role for bioavailable estrogen. J Clin Endocrinol Metab 83: 2266-2274 

Kenny AM, Prestwood KM, Marcello KM, Raisz LG 2000 Deter-minants of bone density in healthy older men with low testosterone levels. J Gerontol A Biol Sci Med Sci 55α:M492-M497 

Tenover JL 1992 Effect of testosterone supplementation in the aging male. J Clin Endocrinol Metab 75:1092-1098 

Snyder PJ, Peachey H, Hannoush P, Berlin JA, Loh L, Holmes JH, Dlewati A, Staley J, Santanna J, Kapoor SC, Attie MF, Haddad Jr JG, Strom BL 1999 Effect of testosterone treatment on bone mineral density in men over 65 years of age. J Clin Endocrinol Metab 84:1966-1972 

Kenny AM, Prestwood KM, Gruman CA, Marcello KM, Raisz LG 2001 Effects of transdermal testosterone on bone and muscle in older men with low bioavailable testosterone levels. J Gerontol A Biol Sci Med Sci 56:M266-M272 

Reid IR, Wattie DJ, Evans MC, Stapleton JP 1996 Testosterone therapy in glucocorticoid-treated men. Arch Intern Med 156:11731177 

Rosenberg MJ, King TDN, Timmons MC 1997 Estrogen-androgen for hormone replacement. J Reprod Med 42:394-404 

Davis S 1999 Androgen replacement in women: a commentary. J Clin Endocrinol Metab 84:1886-1891 

Raisz LG, Wiita B, Artis A, Bowen A, Schwartz S, Trahiotis M, Shoukri K, Smith J 1996 Comparison of the effects of estrogen alone and estrogen plus androgen on biochemical markers of bone formation and resorption in postmenopausal women. J Clin En-docrinol Metab 81:37-43 

Tobias JH, Compston JE 1999 Does estrogen stimulate osteoblast function in postmenopausal women? Bone 24:121-124 

Baulieu E-E, Thomas G, Legrain S, Lahlou N, Roger M, DebuirFaucounau V, Girard L, Hervy M-P, Latour F, Leaud M-C, Mokrane A, Pitti-Ferrandi H, Trivalle C, de Lacharriere O, 

Nouveau S, Rakoto-Arison B, Souberbielle J-C, Raison J, Le Bouc Y, Raynaud A, Girerd X, Forette F 2000 Dehydroepiandrosterone (DHEA), DHEA sulfate, and aging: contribution of the DHEAge study to a sociobiomedical issue. Proc Natl Acad Sci USA 97:4279- 4284 

Villareal DT, Holloszy JO, Kohrt WM 2000 Effects of DHEA replacement on bone mineral density and body composition in elderly women and men. Clin Endocrinol (Oxf) 53:561-568 

Morales AJ, Nolan JJ, Nelson JC, Yen SSC 1994 Effects of replacement dose of dehydroepiandrosterone in men and women of advancing age. J Clin Endocrinol Metab 78:1360-1367 

Matzkin H, Chen J, Weisman Y, Goldray D, Pappas F, Jaccard N, Braf Z 1992 Prolonged treatment with finasteride (a 5 a-reductase inhibitor) does not affect bone density and metabolism. Clin Endocrinol (Oxf) 37:432-436 

Taxel P, Kennedy DG, Fall PM, Willard AK, Clive JM, Raisz LG 2001 The effect of aromatase inhibition on sex steroids, gonado-tropins, and markers of bone turnover in older men. J Clin Endocrinol Metab 86:2869-2874 

Mauras N, O'Brien KO, Klein KO, Hayes V 2000 Estrogen suppression in males: metabolic effects. J Clin Endocrinol Metab 85: 2370-2377 

Lips P, Asscheman H, Uitewaal P, Netelenbos JC, Gooren L 1989 The effect of cross-gender hormonal treatment on bone metabolism in male-to-female transsexuals. J Bone Miner Res 4:657-662 

vanKesterenP,LipsP,GoorenLJG,AsschemanH,MegensJ 1998 Long-term follow-up of bone mineral density and bone metabolism in transsexuals treated with cross-sex hormones. Clin Endocrinol (Oxf) 48:347-354 

Taxel P, Fall PM, Albertsen PC, Dowsett RD, Trahiotis M, Zimmerman J, Ohannessian C, Raisz LG 2002 The effect of mi- cronized estradiol on bone turnover and calciotropic hormones in older men receiving hormonal suppression therapy for prostate cancer. J Clin Endocrinol Metab 87:4907-4913 

Doran PM, Riggs BL, Atkinson EJ, Khosla S 2001 Effects of ralox-ifene, a selective estrogen receptor modulator, on bone turnover markers and serum sex steroid and lipid levels in elderly men. J Bone Miner Res 16:2118-2125 

Rosen CJ 2003 Restoring aging bones. Sci Am 288:70-77 

Moggs JG, Deavall DG, Orphanides G 2003 Sex steroids, ANGELS and osteoporosis. Bioessays 25:195-199 

Falahati-Nini A, Riggs BL, Atkinson EJ, O'Fallon WM, Eastell R, Khosla S 2000 Relative contributions of testosterone and estrogen in regulating bone resorption and formation in normal elderly men. J Clin Invest 106:1553-1560 

Leder BZ, LeBlanc KM, Schoenfeld DA, Eastell R, Finkelstein JS 2003 Differential effects of androgens and estrogens on bone turnover in normal men. J Clin Endocrinol Metab 88:204-210 

Dayani N, Corvol MT, Robel P, Eychenne B, Moncharmont B, Tsagris L, Rappaport R 1988 Estrogen receptors in cultured rabbit articular chondrocytes: influence of age. J Steroid Biochem 31: 351-356 

Benz DJ, Haussler MR, Komm BS 1991 Estrogen binding and estrogenic responses in normal human osteoblast-like cells. J Bone Miner Res 6:531-541 

Etienne MC, Fischel JL, Milano G, Formento P, Formento JL, Francoual M, Frenay M, Namer M1990 Steroid receptors inhuman osteoblast-like cells. Eur J Cancer 26:807-810 

Slootweg MC, Ederveen AG, Schot LP, Schoonen WG, Kloosterboer HJ 1992 Oestrogen and progestogen synergistically stimulate human and rat osteoblast proliferation. J Endocrinol 133: R5-R8 

Davis VL, Couse JF, Gray TK, Korach KS 1994 Correlation between low levels of estrogen receptors and estrogen responsiveness in two rat osteoblast-like cell lines. J Bone Miner Res 9:983-991 

Suzuki S, Koga M, Takaoka K, Ono K, Sato B 1993 Effects of retinoic acid on steroid and vitamin D3 receptors in cultured mouse osteosarcoma cells. Bone 14:7-12 

Braidman IP, Baris C, Selby PL, Adams JE, Freemont AJ, Hoyland JA 2000 Preliminary report of impaired oestrogen receptor-a ex-pression in bone, but no involvement of androgen receptor, in male idiopathic osteoporosis. J Pathol 192:90-96 

Ankrom MA, Patterson JA, d'Avis PY, Vetter UK, Blackman MR, Sponseller PD, Tayback M, Robey PG, Shapiro JR, Fedarko NS 1998 Age-related changes in human oestrogen receptor a function and levels in osteoblasts. Biochem J 333:787-794 

Mahonen A, Maenpaa PH 1994 Steroid hormone modulation of vitamin D receptor levels in human MG-63 osteosarcoma cells. Biochem Biophys Res Commun 205:1179-1186 

Ikegami A, Inoue S, Hosoi T, Kaneki M, Mizuno Y, Akedo Y, Ouchi Y, Orimo H 1994 Cell cycle-dependent expression of estrogen receptor and effect of estrogen on proliferation of synchronized human osteoblast-like osteosarcoma cells. Endocrinology 135:782-789 

Sutherland MK, Hui DU, Rao LG, Wylie JN, Murray TM 1996 Immunohistochemical localization of the estrogen receptor in human osteoblastic SaOS-2 cells: association of receptor levels with alkaline phosphatase activity. Bone 18:361-369 

Hoshino S, Inoue S, Hosoi T, Saito T, Ikegami A, Kaneki M, Ouchi Y, Orimo H 1995 Demonstration of isoforms of the estrogen receptor in the bone tissues and in osteoblastic cells. Calcif Tissue Int 57:466-468 

Westerlind KC, Sarkar G, Bolander ME, Turner RT 1995 Estrogen receptor mRNA is expressed in vivo in rat calvarial periosteum. Steroids 60:484-487