Quick viewing(Text Mode)

A New Hypothesis for How Sex Steroid Hormones Regulate Bone Mass

A New Hypothesis for How Sex Steroid Hormones Regulate Bone Mass

A new hypothesis for how sex regulate bone mass

Joseph Lorenzo

J Clin Invest. 2003;111(11):1641-1643. https://doi.org/10.1172/JCI18812.

Commentary

It has been proposed — but remains controversial — that ’s effects on various tissues may be mediated by different pathways. Researchers have identified a synthetic that activates only a subset of these pathways, suggesting that bypass of the traditional estrogen pathway can prevent bone loss without associated side effects on reproductive organs.

Find the latest version: https://jci.me/18812/pdf COMMENTARIES A new hypothesis for how sex steroid 3′,5′-cyclic adenosine monophosphate (cAMP) levels within 15 seconds. hormones regulate bone mass Pietras and Szego (7) found that the outer surface of endometrial cells con- Joseph Lorenzo tained estrogen-binding sites. In recent years it has become apparent The University of Connecticut Health Center, Farmington, Connecticut, USA that rapid effects of steroid hormones are mediated by interactions with It has been proposed — but remains controversial — that estrogen’s components of various signal trans- effects on various tissues may be mediated by different cell signaling duction pathways, including adenylyl pathways. Researchers have identified (see related article beginning on cyclase, MAPKs, and PI3K. page 1651) a synthetic ligand that activates only a subset of these path- Interestingly, the receptors that bind ways, suggesting that bypass of the traditional estrogen pathway can pre- sex steroid ligands and initiate these vent bone loss without associated side effects on reproductive organs. responses can be identical to or differ- ent from known steroid receptors J. Clin. Invest. 111:1641–1643 (2003). doi:10.1172/JCI200318812. (Figure 1). Estrogen can bind the G –coupled homolog Sex steroid hormones exert a variety of receptor (AR). It is now GPR30 and activate extracellular sig- important actions. In addition to their appreciated that the relative balance of nal–regulated kinases (ERKs) 1 and 2 role as regulators of reproductive func- receptors, coactivator, and corepressor in SK-BR-3 breast cancer cells (8). tions, they have potent effects on the is a critical determinant of the ERK-1 and ERK-2 are members of the nervous and cardiovascular system and ability of this classical pathway to initi- MAPK family. Significantly, SK-BR-3 are major determinants of the develop- ate responses. Since the relative con- cells are devoid of both ERα and ERβ. ment and the structural integrity of the centrations of these molecules is cell Activation of ERKs by estrogen in skeleton. and reg- specific, sex steroid hormones can have SK-BR-3 cells was not prevented by an ulate such diverse cellular actions as vastly different functions in different ER antagonist, which blocks binding proliferation, morphogenesis, differ- tissues of the same organism. Varia- of estrogen to ER (8). entiation, and apoptosis, or pro- tions in the recruitment of coregulato- Estrogen is also capable of inducing grammed cell death. However, our ry molecules also appear to be the rapid effects via membrane-bound understanding of the pathways by mechanism by which selective estrogen ERs. Typically, the majority of either which sex steroid hormones influence receptor modulators (SERMs) produce ERα or ERβ is found in the nucleus. cellular functions is incomplete. their tissue-specific effects (3). However, small amounts (2%) can The classical mechanism of steroid A second mechanism of action for associate with the cell membrane. action involves interactions the classic pathway involves protein- Accumulation of sex steroid hormone with intracellular receptors (Figure 1), protein interactions (Figure 1). Typi- receptors in the cell membrane is which are either cytoplasmic or nuclear cally, in this pathway, receptor-ligand patchy, suggesting the presence of (1, 2). Subsequently, the receptor-ligand complexes interact with transcrip- structures that are critical for their complex binds to specific cis-active ele- tion factors to alter the ability of the localization. Both ERα and ERβ as ments of the promoter regions in the transcription factor to influence well as AR can accumulate in caveolae, DNA of responsive . A key role of transcription. An example of this which are 50- to 100-nm flask-shaped sex steroid hormones in the regulation mechanism is the capacity of ligated- subcompartments in the cell mem- of gene transcription is the recruitment ER complexes to influence the func- brane that contains the 22-kDa trans- of a complex of coactivators and core- tion of activator protein-1 and spe- membrane phosphoprotein caveolin. pressors to the receptor-ligand-DNA cific protein-1 (4, 5). However, a After binding ligand, ERs or ARs in the . This is true for estrogen common characteristic of both of cell membrane can interact with c-Src receptors (ERs) α and β as well as the these classic pathways is that changes and activate MAPK pathways (1, 9). in gene transcription typically occur In earlier investigations, Kousteni Address correspondence to: Joseph Lorenzo, after 30–60 minutes. and colleagues examined the role of The University of Connecticut Health Center, In contrast, it is now appreciated estrogen in preventing the bone loss Division of L3104, MC-1317, that more rapid nonclassical pathways that develops relatively rapidly in mice Farmington, Connecticut 06030-1317, USA. Phone: (860) 679-8199; Fax: (860) 679-1040; of sex steroid hormone action allow after sex steroid hormones are with- E-mail: [email protected]. both estrogens and androgens to drawn (10, 11). In this issue of the JCI, Conflict of interest: The author has declared influence cellular function. Typically, Kousteni et al. (12) now present data that no conflict of interest exists. these effects occur within seconds to that link the rapid activation of MAPK Nonstandard abbreviations used: estrogen minutes. In 1967, Szego and Davis (6) by nonclassical pathways to the ability receptor (ER); androgen receptor (AR); β selective modulator (SERM); demonstrated that 17 - of sex steroid hormones to regulate extracellular signal–regulated kinase (ERK). given intravenously increased uterine apoptosis in bone cells.

The Journal of Clinical Investigation | June 2003 | Volume 111 | Number 11 1641 Figure 1 Sex steroid hormones can affect cellular function by a variety of mechanisms. The illustration depicts the mechanisms by which estrogen influences cells. The classical path- ways (I and II) depend on direct interaction of estrogen with its receptor in the nucleus. Once activated, the estrogen-receptor com- plex can directly mediate gene transcription (I) or interact with transcription factors (II) to influence their activity. The nonclassical pathways (III and IV) work more rapidly and depend on the ability of estrogen to interact with either nonsteroid hormone receptors (III) or steroid hormone receptors in the membrane (IV). Both nonclassical pathways activate kinases that ultimately regulate tran- scription of specific genes. Adapted with per- mission from ref. 16.

Kousteni et al. (9) previously demon- signaling mechanisms in mediating rent study (12), the authors postulate strated in cell culture that the ability of the ability of sex steroid hormones to that estren has a different mechanism estrogens or androgens to regulate block apoptosis in cell culture models. of action than do SERMs, since the lat- apoptosis was mediated by the ligand- In addition, key to their studies is the ter exclusively use the classical path- binding domain of sex steroid hormone use of the synthetic ligand 4-estren- ways to regulate gene transcription in receptors that were localized exclusively 3α,17β-diol, which the authors refer to a cell-specific manner. The authors to the cell membrane. This response as estren. This compound showed no believe, based on their in vitro data, was seen in HeLa cells transfected with classical sex steroid hormone activity that estren influences cellular function ERs or ARs and in osteoblast and osteo- in the examined cell culture systems. through its ability to influence cell cyte cell culture models. The ability of However, estren was a potent activator membrane–mediated nonclassical sex steroid hormones to regulate apop- of the rapid cell membrane–mediated responses to sex steroid hormones. tosis in these models was not specific Src-MAPK pathways in cell culture This is a provocative and controver- for a particular sex steroid hormone models. Furthermore, it blocked apop- sial hypothesis that requires addition- receptor, since estrogens or androgens tosis in osteoblast models and stimu- al investigation before it can be fully were equally effective in mediating lated apoptosis in osteoclast models. accepted. The skeleton is a complex responses in HeLa cells transfected with In contrast, raloxifene — a SERM cur- organ system that is regulated by the either ERs or ARs. The authors hypoth- rently approved to treat osteoporosis interactions of both bone-forming esized that a major effect of sex steroid — did not mimic the apoptosis-regu- osteoblasts (derived from mesenchy- hormones in preventing bone loss lating effects of estren in cell culture mal precursor cells) and bone-resorb- results from inhibition of apoptosis in models. In addition, studies published ing osteoclasts (derived from hemato- osteoblasts and stimulation of apopto- last year by these authors (11) demon- poietic precursors). Compounding the sis in osteoclasts. strated that estren, at concentrations complexity of the signals that regulate In the current study (12), the authors 300-fold higher than those of 17β- bone are interactions use a series of dominant negative estradiol, blocked the bone loss that with marrow cells, which are in close analogs of components of the Src, occurred after ovariectomy in a mouse association to bone, and the responses PI3K, ERK, and JNK pathways to model. Based on the results of both of bone cells to the myriad cytokines demonstrate the critical role of these their previous work (9, 11) and the cur- that are produced in the bone micro-

1642 The Journal of Clinical Investigation | June 2003 | Volume 111 | Number 11 environment. The relatively simple cell will require in vivo experiments that ing sites for oestrogen at the outer surfaces of iso- lated endometrial cells. Nature. 265:69–72. culture systems that Kousteni et al. (9, use sophisticated technologies to dis- 8. Filardo, E.J., Quinn, J.A., Bland, K.I., and Frackel- 12) have examined to date are unlikely sect in detail both the cell-specific and ton, A.R., Jr. 2000. Estrogen-induced activation of to mimic the complex interactions the subcellular compartment–specific Erk-1 and Erk-2 requires the -coupled receptor homolog, GPR30, and occurs via trans- that regulate the skeleton in vivo. It effects that these agents have on bone activation of the epidermal receptor also is unclear whether the effects of mass. Once data from such in vivo through release of HB-EGF. Mol. Endocrinol. 14:1649–1660. sex steroid hormone withdrawal on studies become available, a more com- 9. Kousteni, S., et al. 2001. Nongenotropic, sex-non- the skeleton are mediated predomi- plete understanding of the many specific signaling through the estrogen or andro- nantly by the ability of these agents to effects that sex steroid hormones have gen receptors: dissociation from transcriptional activity. Cell. 104:719–730. regulate the differentiation of osteo- on the skeleton should be appreciated. 10. Jilka, R.L., et al. 1992. Increased osteoclast devel- blasts and osteoclasts from their pre- opment after estrogen loss: mediation by inter- cursor cells or, as Kousteni et al. pos- 1. Cato, A.C., Nestl, A., and Mink, S. 2002. Rapid leukin-6. Science. 257:88–91. actions of steroid receptors in cellular signaling 11. Kousteni, S., et al. 2002. Reversal of bone loss in tulate (12), the ability of sex steroid pathways. Sci. STKE. 138:RE9. mice by nongenotropic signaling of sex . hormones to regulate apoptosis. There 2. Shupnik, M.A. 2002. Oestrogen receptors, Science. 298:843–846. is now strong evidence in murine mod- receptor variants and oestrogen actions in the 12. Kousteni, S., et al. 2003. Kinase-mediated regula- hypothalamic-pituitary axis. J. Neuroendocrinol. tion of common transcription factors accounts for els that estrogen withdrawal is associ- 14:85–94. the bone-protective effects of sex steroids. J. Clin. ated with an increased number of 3. Riggs, B.L., and Hartmann, L.C. 2003. Selective Invest. 111:1651–1664. doi:10.1172/JCI200317261. estrogen-receptor modulators: mechanisms of 13. Manabe, N., et al. 2001. Connection between B osteoclast precursor cells in the mar- action and application to clinical practice. N. Engl. lymphocyte and osteoclast differentiation path- row (10), an effect that has been linked J. Med. 348:618–629. ways. J. Immunol. 167:2625–2631. to the regulation of B-lymphopoiesis 4. Paech, K., et al. 1997. Differential ligand activation 14. Sato, T., Shibata, T., Ikeda, K., and Watanabe, K. of estrogen receptors ERalpha and ERbeta at AP1 2001. Generation of bone-resorbing osteoclasts (13–15). Since the target cell for this sites. Science. 277:1508–1510. from B220+ cells: its role in accelerated osteoclas- response is unknown, it is unclear 5. Saville, B., et al. 2000. Ligand-, cell-, and estrogen togenesis due to estrogen deficiency. J. Bone Miner. what pathways estren or estrogen uses receptor subtype (alpha/beta)-dependent activa- Res. 16:2215–2221. tion at GC-rich (Sp1) promoter elements. J. Biol. 15. Katavic, V., et al. 2003. The surface antigen CD45R to produce this response. Hence, stud- Chem. 275:5379–5387. identifies a population of estrogen-regulated ies of the mechanism by which sex 6. Szego, C.M., and Davis, J.S. 1967. Adenosine 3′,5′- murine marrow cells that contain osteoclast pre- monophosphate in rat uterus: acute elevation by cursors. Bone. In press. steroid hormones or estren mediate estrogen. Proc. Natl. Acad. Sci. U. S. A. 58:1711–1718. 16. Miller, G. 2002. Divorcing estrogen’s bright and their bone-sparing effects ultimately 7. Pietras, R.J., and Szego, C.M. 1977. Specific bind- dark sides. Science. 298:723–724.

What is the real role of antimicrobial proteins and in paralleled the evolving methodology of polypeptides that can mediate several protein biochemistry and purification, other inflammatory responses? and molecular biology. In this histori- cal context, the initial focus remained on the antibacterial action of newly iso- Peter Elsbach lated proteins and peptides, reflecting an ongoing search for new antibiotics Department of Medicine, New York University School of Medicine, New York, New York, USA and the relative ease of conducting bioassays of bacterial viability (2, 3). Antimicrobial peptides are effector molecules of innate immunity with microbicidal and pro- or anti-inflammatory activities. Their role is now Discovery of proteins and widening following evidence that one such multifunctional , polypeptides with antibacterial LL-37, induces angiogenesis, a process essential for host defense, wound properties healing, and tissue repair (see related article beginning on page 1665). Numerous proteins and polypeptides with antimicrobial activity in vitro J. Clin. Invest. 111:1643–1645 (2003). doi:10.1172/JCI200318761. have now been isolated and/or cloned from a broad range of both simple The last two decades have heralded implicated in many aspects of the and complex organisms, including impressive progress in the identifica- host response to infection and other humans. Thus, endogenous-polypep- tion of a broad array of structurally inflammatory stimuli. tide antibiotics became prominent and functionally diverse polypeptides Almost half a century ago, Hirsch targets in the study of antimicrobial described the antimicrobial properties host defenses per se and also as poten- of phagocytin, a crude protein fraction tial pharmacologic agents (3). Whole Address correspondence to: Peter Elsbach, Department of Medicine, New York of polymorphonuclear leukocytes (1), families of such gene products have University School of Medicine, 550 First thereby setting the stage for a growing been identified in plants, insects, and Avenue, New York, New York 10016, USA. interest in the biologic activities of pro- other animals (4) in settings that are Phone: (212) 263-5633; Fax: (212) 263-3952; E-mail: [email protected]. tein components of inflammatory consistent with prominent roles in Conflict of interest: The author has declared cells. The subsequent exploration of innate immunity. How well have these that no conflict of interest exists. the functional capabilities of defined roles been defined so far?

The Journal of Clinical Investigation | June 2003 | Volume 111 | Number 11 1643