d vitamin d- more than a bone-a-fide hormone

Upload: ytreffal

Post on 08-Apr-2018

217 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    1/15

    MINIREVIEW

    Vitamin D: More Than a Bone-a-Fide HormoneAMELIA L. M. SUTTON AND PAUL N. MACDONALD

    Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106

    The vitamin D endocrine system is critical for the

    proper development and maintenance of mineral

    ion homeostasis and skeletal integrity. Beyond

    these classical roles, recent evidence suggests

    that the bioactive metabolite of vitamin D, 1,25-

    dihydroxyvitamin D3, functions in diverse physio-

    logical processes, such as hair follicle cycling,

    blood pressure regulation, and mammary gland

    development. This minireview explores the current

    progress in unraveling the complexities of the vi-

    tamin D endocrine system by focusing on four main

    areas of research: the resolution of the vitamin D

    receptor crystal structure, the molecular details of

    1,25-dihydroxyvitamin D3-mediated transcription,

    murine knockout models of key genes in the en-

    docrine system, and alternative vitamin D recep-

    tors and ligands. (Molecular Endocrinology 17:

    777791, 2003)

    VITAMIN D WAS discovered nearly a century agoas the nutrient that prevented rickets, a devastat-ing skeletal disease characterized by undermineral-

    ized bones (1). Since that time, our concept of vitamin

    D and, in particular, its most bioactive derivative, 1,25-

    dihydroxyvitamin D3 [1,25-(OH)2D3], has evolved from

    that of an essential micronutrient to that of a hormone

    involved in a complex endocrine system that directs

    mineral homeostasis, protects skeletal integrity, and

    modulates cell growth and differentiation in a diverse

    array of tissues. 1,25-(OH)2D3 acts in concert with PTHto tightly regulate the concentration of serum calcium

    and phosphate, thereby maintaining proper skeletal

    mineralization (Fig. 1). A major function of 1,25-

    (OH)2D3 is to promote intestinal absorption of calcium

    and phosphate. However, it also may have direct ef-

    fects on the bone (2), in which continuous remodeling

    must occur to sustain structural integrity. For example,

    in vitro studies indicate that 1,25-(OH)2D3 stimulates

    osteoblasts, the resident bone-forming cells, to termi-

    nally differentiate and to deposit calcified matrix (3).

    Conversely, when dietary sources are inadequate to

    maintain normocalcemia, 1,25-(OH)2D3 may stimulate

    calcium mobilization from the bone by promoting the

    differentiation of precursor cells into mature, bone-resorbing osteoclasts (4).

    The hormonal or bioactive form of vitamin D is1,25-(OH)

    2D3.

    It is generated from sequential hy-

    droxylations of vitamin D3, a secosteroid precursorthat is obtained from the diet or produced in the skinupon exposure to UV light (5, 6). The first hydroxy-lation of vitamin D

    3occurs at the C-25 position and

    is catalyzed by vitamin D-25-hydroxylase in the liver

    to produce 25-hydroxyvitamin D3 [25(OH)D3], themajor circulating form of vitamin D in mammals.25(OH)D

    3is the substrate for a second hydroxylase,

    the renal 25(OH)D3

    -1-hydroxylase (1OHase),

    resulting in the production of the most bioactivemetabolite, 1,25-(OH)2D3. A classic endocrine feed-back system operates to tightly control serum levelsof 1,25-(OH)

    2D3

    (5, 6). For example, renal 1OHaseactivity is stimulated by low serum calcium and

    phosphorus levels and by PTH. The expression of1OHase is negatively regulated by high levels of1,25-(OH)

    2D3

    . Inactivation, or catabolism, of vitaminD metabolites is initiated by the ubiquitous enzyme

    25-hydroxyvitamin D3-24-hydroxylase (24OHase) togenerate either 24,25(OH)2D3 or 1,24,25(OH)3D3.The 24-hydroxylated metabolites are further de-graded and eventually excreted as either calcitroicacid or 23-carboxyl derivatives. This catabolic pro-cess is also carefully regulated as 1,25-(OH)

    2D3

    stimulates 24OHase expression to prevent exces-

    sive synthesis of the hormone.The biological effects of 1,25-(OH)2D3 are mediated

    through the vitamin D receptor (VDR), a member of thenuclear receptor superfamily of ligand-activated tran-scription factors (7, 8). Binding of 1,25-(OH)

    2D3

    to VDRinitiates a cascade of macromolecular interactions ul-

    Abbreviations: AF-2, Activation function-2; CBP, cAMPresponse element binding protein-binding protein; DBD,DNA-binding domain; 1,25-(OH)

    2D3

    , 1,25-dihydroxyvitaminD3

    ; 1OHase, 25(OH)D3

    -1-hydroxylase; 25-(OH)D3

    , 25-hydroxyvitamin D

    3; 24-OHase, 25-(OH)D

    3-24-hydroxylase;

    DRIP, VDR-interacting protein; HAT, histone acetyltrans-ferase; HVDRR, hereditary vitamin D-resistant rickets; LBD,ligand-binding domain; LCA, lithocholic acid; NCoA, nuclearreceptor coactivator; RAR, retinoic acid receptor; RXR, reti-noid X receptor; SKIP, ski-interacting protein; SRC, steroidreceptor coactivator; TIF2, transcription intermediary fac-tor-2; TRAP, thyroid receptor-activating protein; VDR, vitaminD receptor; VDRE, vitamin D response element; VDRKO, VDRknockout.

    0888-8809/03/$15.00/0 Molecular Endocrinology 17(5):777791Printed in U.S.A. Copyright 2003 by The Endocrine Society

    doi: 10.1210/me.2002-0363

    777by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    2/15

    timately leading to transcription of select target genes

    (9). 1,25-(OH)2

    D3

    associates with the VDR and pro-

    motes its heterodimerization with retinoid X receptor

    (RXR), a common heterodimeric partner for other class

    II nuclear receptors (10). The liganded VDR-RXR het-

    erodimer is the functionally active transcription factor

    in 1,25-(OH)2D3-mediated transcription. The het-

    erodimer binds with high affinity to vitamin D response

    elements (VDREs) in the promoters of target genes.

    VDREs are characterized by two direct hexameric re-

    peats with an intervening spacer of three nucleotides

    (DR-3 elements). Thus, 1,25-(OH)2

    D3

    target gene se-

    lectivity is conferred, in part, through ligand binding,

    VDR-RXR heterodimerization, and high-affinity bind-

    ing to DR-3 VDREs. Beyond these initial steps, the

    precise molecular mechanisms involved in target gene

    activation by VDR are less evident. Recent attention

    has turned to so-called coactivator proteins that inter-

    act directly with VDR and other nuclear receptors in a

    ligand-dependent manner (11). These coactivators par-

    ticipate in an intricate multiprotein complex together with

    the basal transcriptional machinery and histone modi-

    Fig. 1. Metabolism and Mineral Homeostatic Functions of the Vitamin D Endocrine System

    Bioactive 1,25-(OH)2

    D3

    is generated by sequential hydroxylations of its precursor vitamin D3

    in the liver and the kidney.

    1,25-(OH)2

    D3

    operates in a negative feedback loop by inducing expression of the catabolic enzyme 24-OHase and by inhibiting

    expression of the anabolic enzyme 1OHase. In response to low serum calcium, PTH is produced and stimulates 1OHase

    expression in the kidney and promotes calcium mobilization from the bone and reabsorption from the kidney. 1,25-(OH)2

    D3

    , in

    turn, induces calcium absorption in the intestine and calcium release from the skeleton.

    778 Mol Endocrinol, May 2003, 17(5):777791 Minireview Sutton and MacDonald

    by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    3/15

    fiers to stimulate expression of 1,25-(OH)2D3-regulated

    genes.

    Over the past five years, remarkable strides have

    been made in clarifying the physiological functions

    and the concomitant therapeutic potential of vitamin D

    and its derivatives. Structural studies provide new in-

    sight into the ligand-binding pocket of VDR com-plexed to 1,25-(OH)

    2D3 and several potent synthetic

    analogs and, thereby, present a scaffold on which to

    build future VDR-targeted therapies. A more complete

    molecular picture of VDR-mediated transcription is

    emerging as the details of coactivator action are un-

    raveled. Murine knockout models of VDR as well as

    key enzymes involved in vitamin D metabolism reveal

    the essential roles of vitamin D in vivo. Finally, inves-

    tigators have begun to identify novel ligands and al-

    ternative VDRs, from synthetic analogs to potential

    membrane receptors, that signal new directions for the

    field. Although the range of recent advances extends

    far, we chose to focus this minireview on these four

    areas of vitamin D research. Together, these areas of

    progress have not only affirmed classic paradigms in

    vitamin D physiology, but they also have opened up

    new avenues of exploration for future research.

    VDR CRYSTAL STRUCTURE

    The VDR shares discrete structural and functional do-

    mains with other nuclear receptors, but it also exhibits

    several unique features (Fig. 2A and Refs. 5 and 9). The

    hypervariable amino-terminal A/B domain of VDR is

    unusually short and, in contrast to that of most othernuclear receptors, is generally thought to lack potent

    transactivation domains. However, as discussed later

    (see VDR Isoforms), there is increasing evidence that

    the VDR A/B domain helps determine the overall trans-

    activation capacity of the VDR (12). The DNA-binding

    domain (DBD, or region C) of VDR is similar to that of

    other nuclear receptors and is characterized by two

    zinc-binding modules that direct sequence-specific

    binding of receptors to DNA (13). The ligand-binding

    domain (LBD, or region E) is a multifunctional globular

    domain that mediates selective interactions of the re-

    ceptor with its cognate hormone (13), with other nu-

    clear receptor partners (14), and with comodulatory or

    adapter proteins (15). The LBD contains the ligand-dependent activation function-2 (AF-2), which is cru-

    cial to ligand-activated transcription. Mutation of the

    AF-2 renders the nuclear receptor transcriptionally in-

    active despite retaining the ability to bind ligand (1416). The DBD and LBD are bridged by the hinge region

    (domain D), which is thought to confer rotational flex-

    ibility between the DBD and LBD and allow for recep-

    tor dimerization and interaction with the DNA (17).

    Although detailed crystal structures for several nu-

    clear receptors have been available for nearly a de-

    cade (1822), the structure of the VDR LBD was notsolved until recently (23). Numerous attempts to crys-

    tallize VDR failed, likely due to the presence of a

    unique insertion sequence in the LBD (see Fig. 2A) that

    is largely unordered, leading to decreased protein sol-

    ubility. Removal of this insertion domain allowed for

    efficient crystallization and structure determination of

    the VDR LBD complexed to 1,25-(OH)2D3 (23). Al-though the lack of this domain may compromise the

    interpretation of the VDR structure, the mutant VDR

    displays normal ligand binding and similar transacti-

    vation properties in vitro (24). Thus, the absence of the

    insertion sequence does not alter the conformation

    significantly so as to compromise VDR function.

    The structure of the VDR LBD is similar to that of

    other nuclear receptors, being most closely related to

    that of the retinoic acid receptor (RAR)LBD (23). The

    VDR LBD is organized into 13 -helices and 3

    -sheets, which together form a hydrophobic ligand-

    binding pocket. This pocket is larger than that of RAR

    Fig. 2. Domain Structure of VDR and Two-Step Model of

    VDR-Mediated Transcription

    A, Functional domains of VDR. A/B, Amino-terminal region;

    DBD, DBD showing two zinc finger modules (Zn); LBD, LBDincluding the long insertion domain and helix 12 encompass-

    ing AF-2. B, Temporal association of coactivators during

    VDR-mediated transcription. The liganded (D) VDR-RXR het-

    erodimer recruits SRCs and CBP/p300, resulting in acetyla-

    tion (Ac) of histones. The open chromatin template allows for

    binding of the DRIP complex and entry of the core transcrip-

    tional machinery.

    Minireview Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777791 779

    by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    4/15

    due to variations in the positions of helices H2 and H3n

    and the H6H7 loop. Helix 12, containing the ligand-dependent AF-2, of ligand-bound VDR is positioned

    similarly to other nuclear receptors (20), highlighting its

    central importance in creating a coactivator interaction

    surface (see VDR Coactivators). In fact, several resi-

    dues of H12 directly contact the ligand, indicating thatthe ligand conformation may modulate H12 conforma-

    tion and, therefore, coactivator binding and transcrip-

    tional activity. When additional structures of liganded

    VDR complexed with various coactivators are solved,

    they will likely provide a molecular framework on which

    to develop new compounds to modulate the vitamin D

    endocrine system.

    In this regard, numerous synthetic analogs have

    already been developed that mimic the advantageous

    effects of 1,25-(OH)2

    D3

    without the hypercalcemic

    side effects (see Novel Vitamin D Ligands). Specula-

    tion about the mechanisms behind the selective, pleio-

    tropic effects of 1,25-(OH)2

    D3

    analogs centers on the

    concept that these analogs induce distinct conforma-

    tions in VDR compared with that of the natural ligand,

    ultimately resulting in analog-selective gene regulation

    (25, 26). Protease digestions and coactivator binding

    studies provide experimental support for this model

    (25, 27). However, the VDR ligand-binding cavity is

    larger than that of many other nuclear receptors, and

    the ligand occupies less than half of this volume. Con-

    sequently, the VDR ligand-binding pocket can accom-

    modate rather significant structural changes in the

    ligand including 1,25-(OH)2

    D3

    analogs with bulky side

    chains (23, 28). Indeed, the crystal structures of VDR

    complexed with the MC1288 and KH1060 analogs

    show that these low calcemic analogs do not inducedifferent conformations in the VDR compared with the

    natural ligand (29). Thus, other mechanisms must be

    considered to explain the different potencies and cal-

    cemic profiles of the analogs. One potential answer

    resides in the observation that the VDR-analog com-

    plexes are more energetically stable than the VDR-

    1,25-(OH)2D3 complex (29). The increased half-life of

    the activated VDR may result in altered transcriptional

    activity, which may explain the differences both in

    potencies and in target gene selectivity between the

    natural and synthetic ligands. Alternatively, the solid-

    state crystal structure may not reveal subtle dynamic

    conformational changes in solubilized VDR evoked by

    various analogs.

    VDR COACTIVATORS

    The existence of limiting accessory factors or adapter

    proteins in steroid hormone receptor action was pro-

    posed in the late 1980s based on the squelchingphenomenon, in which the LBD of one receptor inter-

    feres with ligand-activated transcription mediated by a

    second receptor (30). A decade later, these comodu-

    latory proteins were identified as specific molecules

    that interact with nuclear receptors and influence their

    transactivation potential (3133). The emergence ofcoactivators, and their inhibitory counterparts core-

    pressors, provides new insight into the molecular

    mechanism of nuclear receptor-mediated transcrip-

    tion. Upon association with its cognate hormone, the

    receptor LBD undergoes a subtle conformationalchange (34). The critical change occurs in helix 12, the

    carboxy-terminal-helix containing the ligand-depen-

    dent AF-2. In response to ligand binding, helix 12 folds

    over top of the globular LBD and caps the ligand-

    binding cavity (20). This ligand-dependent conforma-

    tional shift creates a hydrophobic cleft composed of

    helices 3, 4, 5, and 12 (3537). The hydrophobic cleftserves as a docking surface for many nuclear receptor

    coactivators by interacting with a complementary hy-

    drophobic domain in the coactivator containing the

    consensus LXXLL motif, also referred to as the nuclear

    receptor box (38). Although these studies provide an

    elegant structural model for ligand-activated tran-scription by nuclear receptors and LXXLL-containing

    coactivators, the precise mechanisms governing nu-

    clear receptor-mediated transactivation are less clear.

    The ability of coactivators to interact with components

    of the preinitiation complex, with other transcription

    factors, and with histone-modifying proteins implies

    that a complex integration of transactivator cues oc-

    curs at the promoter of nuclear receptor target genes.

    The growing number of coactivators identified in the

    last decade adds yet another level of complexity to the

    paradigm of nuclear receptor-mediated transcription.

    Extensive reviews on comodulatory proteins can be

    found elsewhere (3941). Here, we will highlight a few

    significant developments in the VDR coactivator field.

    Steroid receptor coactivator (SRC)-1 [or nuclear re-

    ceptor coactivator (NCoA)1] is the founding member of

    the LXXLL motif-containing SRC family of coactivators

    (33). This family also includes transcriptional interme-

    diary factor-2 (TIF2; Refs. 42 and 43) and receptor-

    associated coactivator-3 (4448). The SRCs interactwith VDR and potentiate its transcriptional activity (15,

    49). Each of the SRCs possess an autonomous tran-

    scriptional activation domain, as evidenced by their

    ability to enhance transcription when fused to a het-

    erologous DNA-binding sequence such as GAL4.

    SRCs stimulate transcription possibly by recruiting

    other transcription factors to the promoter. For exam-ple, SRCs interact with cAMP response element bind-

    ing protein (CBP)/p300, a histone acetyltransferase

    (HAT) that remodels chromatin structure at the pro-

    moter (46, 47, 50). SRCs also possess intrinsic HAT

    activity (51). CBP/p300 directly associates with nu-

    clear receptors and, together with SRCs, synergisti-

    cally stimulates transcription (52, 53). Thus, SRCs di-

    rectly alter chromatin structure and recruit other

    factors that modify histones, potentially providing

    more accessible promoter templates on which the

    transcriptional machinery assembles and initiates

    transcription of target genes.

    780 Mol Endocrinol, May 2003, 17(5):777791 Minireview Sutton and MacDonald

    by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    5/15

    A large multiprotein complex called DRIP (VDR-

    interacting proteins) was identified as a coactivator for

    VDR and other nuclear receptors (54, 55). Many com-

    ponents of this complex were discovered separately

    as thyroid receptor activating protein (TRAP) and the

    mammalian Mediator complex (56, 57). The diversity

    of transactivator interactions with the DRIP/TRAP/Mediator complex clearly suggests a more fundamen-

    tal role for this complex in stimulus-activated tran-

    scriptional processes. In VDR-mediated transcription,

    DRIP205/TRAP220 acts as an anchoring subunit of

    the complex by interacting directly with VDR/RXR het-

    erodimers through one of two LXXLL motifs (58). Bio-

    chemical depletion of DRIP in cell-free transcription

    assays shows that DRIP is essential for VDR-activated

    transcription in vitro (54). Because the DRIP complex

    does not contain SRCs and is not associated with HAT

    activity (58), it is likely that DRIP and SRCs potentiate

    the transcriptional activation of VDR through distinct

    mechanisms. Chromatin immunoprecipitations stud-

    ies indicate that a coactivator exchange occurs in the

    transcriptional complex on native nuclear receptor-

    responsive promoters (5961). Specifically, SRCs ap-pear to enter the transcriptional complex first and dis-

    sociate followed by binding of the DRIP multimeric

    complex (60, 61). DRIP is also known to recruit the

    RNA polymerase II holoenzyme to VDR upon ligand

    binding (62). Although these data conflict, to some

    extent, with previous studies that show simultaneous

    association of SRCs and DRIP with activated nuclear

    receptor complexes (59), they do suggest a temporal

    model in which SRCs enter the complex first to re-

    model the chromatin, followed by DRIP complex entry

    and subsequent recruitment of RNA polymerase II(Fig. 2B).

    In addition to DRIP and SRCs, several other proteins

    that potentiate VDR-mediated transcription have been

    described. One example is NCoA-62/ski-interacting

    protein (SKIP), which is a coactivator unrelated to

    DRIPs, SRCs, and other LXXLL-containing coactiva-

    tors (63). It interacts with VDR and other nuclear re-

    ceptors and augments their transcriptional activity.

    Bx42, the Drosophila melanogasterortholog of NCoA-

    62/SKIP, is also implicated in transcriptional pro-

    cesses activated by the insect steroid ecdysone (64).

    NCoA-62/SKIP was identified independently through

    its interaction with ski, placing it as part of the TGF-

    -dependent Smad transcriptional complex (65). It isalso implicated in a number of other transcriptional

    pathways. NCoA-62/SKIP lacks LXXLL motifs and se-

    lectively associates with the VDR-RXR heterodimer

    through the LBD, but through a domain that is distinct

    from the H3-H5/H12 interactions surface (66). NCoA-

    62/SKIP binds VDR simultaneously with SRC-1 to

    form a ternary complex that synergistically enhances

    VDR-stimulated transcription (66), suggesting a poten-

    tial interplay between different coactivator classes for

    maximal activity. Recently, NCoA-62/SKIP was iden-

    tified in subcomplexes of the spliceosome (67, 68).

    This, combined with NCoA62/SKIPs ability to contact

    varied transcription factors including the VDR, sug-

    gests a potentially important role for this and other

    coactivators in coupling nuclear receptor-mediated

    transcription with mRNA splicing (69).

    Although a variety of coactivator proteins have been

    identified for VDR and other nuclear receptors, their

    physiological significance and their discrete and/orredundant functions in different signal-activated tran-

    scriptional systems remain unclear. Mice with individ-

    ual targeted deletions of the three SRCs have been

    developed (7072). These models show that the SRCsshare several similar functions, especially in the devel-

    opment and maintenance of the female reproductive

    system. However, the individual SRCs also serve dis-

    tinct physiological roles. For example, ablation of TIF2/

    glucocorticoid receptor-interacting protein 1 results in

    testicular defects, whereas deletion of either SRC-1 or

    receptor-associated coactivator 3 does not affect

    male reproduction (71). As the effect of these deletions

    on 1,25-(OH)2

    D3

    -mediated transcription has not been

    reported in any of these three knockout models, the in

    vivo relevance of SRCs in VDR-activated transcription

    remains to be determined. Deletion of the receptor-

    interacting subunit of the DRIP complex, DRIP205/

    TRAP220, results in attenuated thyroid hormone-stim-

    ulated transcription but does not affect retinoic acid

    responses. Again, 1,25-(OH)2

    D3

    responses have not

    been examined in this model, so it is unknown whether

    DRIP is required for VDR-activated transcription in

    vivo. Silencing of the Caenorhabditis elegans ortholog

    of NCoA-62/SKIP by RNA interference results in early

    embryonic lethality due to a potential general tran-

    scription defect (73). Although delineation of the phys-

    iological function of NCoA-62/SKIP awaits develop-ment of a mammalian knockout model, these data

    suggest that NCoA-62/SKIP plays a fundamental role

    in RNA polymerase II-mediated transcription. More

    studies are needed to build an integrative vision of

    how the entire ensemble of coactivator proteins asso-

    ciates and stimulates the transcriptional activity of

    VDR and other nuclear receptors. Initial forays into

    deciphering this complex process have begun with the

    application of chromatin immunoprecipitation assays

    and in vivo imaging of fluorescently tagged nuclear

    receptors and coactivators to assess the temporal

    assembly of transcription factors and nuclear recep-

    tors on native promoters (5961, 74, 75). These strat-

    egies, combined with in vitro systems composed ofpurified components and chromatin-packaged tem-

    plates, will be required for a more complete under-

    standing of the molecular details of VDR-activated

    transcription.

    KNOCKOUT MODELS

    Much of our understanding of the physiology of the

    vitamin D endocrine system has stemmed from classic

    dietary manipulations and from the analysis of inher-

    Minireview Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777791 781

    by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    6/15

    ited disorders in humans (Ref. 76 and reviewed in Ref.

    77). The recent development of murine genetic models

    in which key genes in this endocrine system have been

    systematically eliminated highlights the essential role

    of 1,25-(OH)2

    D3

    in maintaining mineral homeostasis as

    well as reveals more subtle actions of this hormone in

    other physiological processes.

    VDR Knockout (VDRKO) Mice

    Two groups independently created mouse strains with

    targeted deletions in the VDR gene by disrupting either

    exon 2 (78) or exon 3 (79). Not surprisingly, the VDRKO

    mice displayed all of the features of the human disease

    hereditary vitamin D-resistant rickets (HVDRR), a rare

    genetic disorder caused by mutations in the VDR gene

    (Ref. 76 and reviewed in Ref. 77). The VDRKO mice are

    viable and develop normally until the weaning period.

    However, shortly after weaning, VDR-null mice exhibit

    alopecia and growth retardation accompanied by pro-

    gressive hypocalcemia, hypophosphatemia, and com-

    pensatory hyperparathyroidism. These metabolic im-

    balances result in severe skeletal defects, including

    decreased bone mineral density, thinned bone cortex,

    and widened undermineralized growth plates. How-

    ever, when VDRKO mice are fed a rescue diet rich in

    calcium and phosphorus to normalize serum calcium

    and PTH levels, the mice develop normally without

    bone abnormalities (80). The skeletons of these mice

    appear grossly, histologically, and biometrically nor-

    mal (81). This indicates that the bone defect in VDR-

    null mice is secondary to the malabsorption of calcium

    in the intestine and is not due to the lack of a direct

    effect of 1,25-(OH)2D3 on the bone. The impaired in-testinal absorption of calcium in the VDRKO mice is

    linked to diminished intestinal expression of several

    1,25-(OH)2

    D3

    -regulated genes putatively involved in

    calcium transport, including calbindin D9K, calcium

    transport protein-1, and epithelial calcium channel (82,

    83). In addition to the intestinal defect in calcium ab-

    sorption, mice that express a mutant VDR that lacks

    the DBD have decreased renal reabsorption of calcium

    (84). These studies reinforce the concept that both the

    intestine and the kidney are essential VDR target or-

    gans in maintaining calcium homeostasis.

    The lack of a skeletal phenotype in the VDRKO mice

    weaned onto the rescue diet is somewhat surprising in

    light of the vast literature supporting direct, primaryroles of VDR in both osteoblast and osteoclast biology

    (85). On the surface, the VDRKO studies indicate that

    1,25-(OH)2

    D3

    is not essential for normal bone devel-

    opment and for maintaining skeletal integrity beyond

    its classic role in calcium and phosphate absorption in

    the intestine. However, a standard caveat with gene

    disruption is that the developing animal may acquire

    adaptive mechanisms or utilize redundant compensa-

    tory pathways to bypass the effects of the gene dele-

    tion. Conditional knockout approaches that ablate

    VDR in a temporally controlled or tissue-specific man-

    ner may be more informative. Such strategies could be

    designed to explore the role of VDR at stage-selective

    checkpoints in skeletal maturation, at which the pos-

    sibility of developing compensatory mechanisms has

    been minimized. Finally, the skeletal phenotypes of

    normocalcemic VDRKO mice may be more pro-

    nounced at different life stages or under different

    physiological stresses. Thus, it will be important todetermine whether normocalcemic VDRKO mice are

    more susceptible to age-related or ovariectomy-

    induced loss of bone mineral density and whether they

    are compromised in their ability to repair skeletal

    fractures.

    In contrast to the skeletal phenotype, the mineral-

    rich diet does not correct the alopecia (i.e. the absence

    of functional hair follicles) observed in VDRKO mice

    (80). Although the VDRKO keratinocytes proliferate

    and differentiate normally, they fail to properly initiate

    hair regrowth after depilation (86, 87). Due to the lack

    of feedback regulation of the anabolic 1OHase en-

    zyme and the catabolic 24OHase, the VDRKO animals

    have abnormally high levels of 1,25-(OH)2

    D3

    . Thus,

    one potential cause of alopecia in VDRKO animals is

    1,25-(OH)2D3 toxicity. To address this possibility,

    VDRKO mice were raised and bred for five generations

    in a UV light-free environment and on a diet lacking

    vitamin D derivatives (87). Despite having undetect-

    able levels of 1,25-(OH)2

    D3

    , fifth-generation vitamin

    D-deficient VDRKO mice still have alopecia. Thus,

    1,25-(OH)2

    D3

    toxicity does not cause alopecia in

    VDRKO mice. Because wild-type littermates of

    VDRKO mice raised under the same vitamin D-defi-

    cient conditions do not display alopecia, Demay and

    colleagues (87) proposed that VDR may regulate hair

    follicle cycling in a ligand-independent fashion. Furthersupport for this hypothesis comes from the observa-

    tion that mice with a targeted deletion in 25(OH)D3

    -

    1-OHase (see below), the biosynthetic enzyme that

    produces 1,25-(OH)2D3, do not display alopecia (88).

    In vitro studies indicate that VDR associates with var-

    ious transcription factors and induces select genes in

    the absence of ligand (63, 8991). Alternatively, unli-ganded VDR may repress a subset of target genes in

    a manner analogous to other nuclear receptors

    through corepressor interactions (9294). Such VDR-RXR-repressed genes could be involved in negatively

    regulating hair follicle cycling. Although these studies

    introduce a novel and potentially significant concept in

    VDR biology, identifying target genes and establishingmolecular mechanisms that govern the function of

    unliganded VDR in keratinocytes are important goals

    for future research.

    The global tissue distribution of VDR suggests that

    1,25-(OH)2

    D3

    plays important roles in physiological

    processes beyond mineral homeostasis and keratino-

    cyte function. For example, VDRKO mice have im-

    paired reproductive function (78). Both male and fe-

    male VDR-null mice exhibit diminished estradiol levels

    and elevated gonadotropins, indicating a gonadal dys-

    function in the secretion of sex hormones (95). Histo-

    logical analysis of reproductive glands shows abnor-

    782 Mol Endocrinol, May 2003, 17(5):777791 Minireview Sutton and MacDonald

    by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    7/15

    mal ovarian follicle development in the females and

    dilated seminiferous tubules with diminished spermat-

    ogenesis in the males. However, serum calcium nor-

    malization with the rescue diet mostly corrects the

    hormonal imbalances and histological abnormalities

    (95) and completely restores fertility (95, 96), suggest-

    ing that the gonadal dysfunction in VDRKO mice pri-marily results from hypocalcemia.

    Mammary gland development is also emerging as a

    biological process that is impacted by 1,25-(OH)2

    D3

    .

    The mammary glands of VDRKO mice demonstrate a

    hyperproliferative phenotype as evidenced by in-

    creased numbers of terminal end buds and enhanced

    ductal branching compared with wild-type littermates

    (97). VDRKO mammary glands also show accelerated

    ductal development and increased proliferation in re-

    sponse to exogenously administered estrogen and

    progesterone (97). Although dietary calcium supple-

    mentation normalizes estrogen levels in VDRKO mice,

    the abnormal mammary phenotype is retained. These

    data indicate a significant developmental role for VDRin the mammary gland potentially in restricting ductal

    growth. Combined with the observations that 1,25-

    (OH)2

    D3

    and its analogs inhibit the growth and induce

    the differentiation of breast cancer cell lines (98, 99),

    the mammary phenotype in the VDRKO model indi-

    cates that 1,25-(OH)2

    D3

    and its derivatives may be

    useful therapies for breast cancer.

    An important immunomodulatory function for 1,25-

    (OH)2D3 and VDR has been indicated by decades of in

    vitro studies. For example, 1,25-(OH)2D3 potently in-

    hibits proliferation and drives the differentiation of leu-

    kemic cells along the monocyte/macrophage lineage

    (100). However, VDRKO mice lack a striking immune

    system phenotype. Although OKelly et al. (101) ob-served that VDRKO mice have abnormal T cell re-

    sponses due to diminished cytokine production by

    macrophages, the calcium-rich rescue diet was not

    tested in this study. Thus, it is not known whether

    these abnormalities can be attributed to a lack VDR, to

    1,25-(OH)2

    D3

    toxicity, or to hypocalcemia. Mathieu

    et al. (102) found that VDRKO mice had a defect in

    calcium-dependent T cell proliferation resulting in pro-

    tection from experimentally induced autoimmune dia-

    betes and that these abnormalities could be corrected

    by restoring serum calcium to normal levels. The nor-

    mal development of the immune system in the VDRKO

    mice suggests that VDR is not essential for immune

    function or that other compensatory pathways exist.Several clinical studies have proposed that 1,25-

    (OH)2

    D3

    may also be beneficial to the cardiovascular

    system by decreasing blood pressure (103, 104). Con-

    sistent with these observations in humans, VDRKO

    mice have increased renin expression, resulting in

    higher levels of angiotensin II, increased water intake,

    electrolyte disturbances, elevated blood pressure, and

    cardiac hypertrophy (105). Furthermore, high levels of

    renin and angiotensin II persist despite normalization

    of mineral ion levels with the rescue diet. This re-

    sponse appears to be at the transcriptional level, as

    1,25-(OH)2D3 suppresses renin promoter activity. This

    study suggests that VDR negatively regulates the ex-

    pression of renin, allowing for decreased angiotensin

    production and lower blood pressure. The relevance of

    this study to human hypertension is not entirely clear

    because there are no reports of hypertensive HVDRR

    patients. Regardless, these are provocative observa-

    tions in the VDRKO model that may stimulate a moreextensive examination of 1,25-(OH)

    2D3

    and its syn-

    thetic analogs as potential therapies for some forms of

    hypertension.

    VDR/RXR Double Knockout

    VDR heterodimerizes with RXR to modulate transcrip-

    tion of target genes in response to 1,25-(OH)2D3. To

    examine the effect of abolishing both of the active

    partners in 1,25-(OH)2D3 signaling, Yagishita and col-

    leagues (106) crossed VDRKO mice with RXR-null

    mice to generate VDR/RXR-double-knockout mice.

    The phenotype of the double-knockout mice is nearly

    identical with that of the single-VDR-knockout mouse,including growth retardation, hypocalcemia, hy-

    pophosphatemia, hyperparathyroidism, rickets, and

    alopecia. Upon closer inspection, however, a unique

    abnormality was noted in the growth plates of long

    bones of VDR/RXR-knockout mice that is not present

    in either of the single-knockout mice. Specifically,

    VDR/RXR-null mice have a defect in the development

    of hypertrophic chondrocytes, the most mature type of

    chondrocyte in the growth plate. Normalization of se-

    rum calcium and phosphorus rescues all of the skel-

    etal anomalies except for the disordered growth

    plates. Because this chondrocyte defect is not present

    in either of the single-knockout strains, the authors

    suggested that a functionally redundant VDR-relatedreceptor exists that selectively heterodimerizes with

    RXR. Such a receptor would likely share a consider-

    able amount of sequence similarity with the classical

    VDR because it must 1) bind to and be transcriptionally

    activated by 1,25-(OH)2

    D3

    or its metabolites; 2) het-

    erodimerize with RXR; and 3) recognize and stimulate

    transcription of the same target genes as classical

    VDR in chondrocytes. Although the public genome

    databases do not indicate that highly related se-

    quences exist, potential candidates might include the

    most closely related nuclear receptors, such as farne-

    soid X receptor, steroid xenobiotic receptor/pregnane

    X receptor, and liver X receptor (107). Alternatively, any

    nuclear receptor unrelated to VDR that selectively het-

    erodimerizes with RXRand binds 1,25-(OH)2

    D3

    or its

    metabolites might fulfill this role.

    25(OH)D3-1-Hydroxylase Knockout

    Arguably, one of the most significant advances in the

    vitamin D field over the past five years has been the

    identification and cloning of the renal 1OHase,

    the enzyme responsible for the regulated synthesis of

    the active hormone (108, 109). To study the effects

    of the absence of 1,25-(OH)2

    D3

    , two groups indepen-

    dently disrupted the 1OHase gene in mice (88, 110).

    Minireview Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777791 783

    by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    8/15

    The phenotype of the 1OHase-null mice is strikingly

    similar to that of the VDR-knockout mouse, including

    hypocalcemia, hyperparathyroidism, growth retarda-

    tion, and osteomalacia, consistent with rickets (88,

    110). The 1OHase-mutant female mice are anovula-

    tory and, therefore, infertile (88). Another key differ-

    ence between VDRKO mice and the 1OHase-ablatedmice is that the 1OHase-knockout animals do not

    display alopecia (88, 110). The role of hypocalcemia/

    hypophosphatemia in any aspect of the abnormal

    phenotype of the 1OHase-ablated mice has yet to be

    addressed using the rescue diet. Importantly, the de-

    velopment of knockout models of both the receptor

    and ligand of 1,25-(OH)2

    D3

    will provide powerful tools

    to delineate the overlapping and distinct roles of VDR

    and its ligand in diverse processes such as mineral

    homeostasis, hair follicle cycling, mammary gland de-

    velopment, and blood pressure regulation.

    24OHase Knockout

    24OHase metabolizes both the bioactive 1,25-(OH)2

    D3

    and its precursor, 25(OH)D3

    . 24OHase gene transcrip-

    tion is positively regulated by 1,25-(OH)2

    D3

    , thereby

    completing a negative feedback loop to prevent ex-

    cessive hormone synthesis. One of the major products

    of 24OHase, namely 24,25(OH)2

    D3

    , is considered to

    be an inactive metabolite, an initial product destined

    for further degradation and eventual excretion as cal-

    citroic acid (5). However, there is substantial evidence

    that 24,25(OH)2

    D3

    has biological activity of its own,

    most strikingly in the function of chondrocytes, or

    cartilage-forming cells (111113). To determine theconsequences of abolishing this enzyme and, there-

    fore, its 24-hydroxylated products, a knockout modelof 24OHase was created (114). This mutation results in

    reduced embryonic viability and aberrant intramem-

    branous ossification. The obvious possibility is that the

    diminished levels of 24,25(OH)2

    D3

    causes these ab-

    normalities. However, the phenotype is not rescued in

    24OHase-knockout progeny by feeding pregnant mice

    exogenous 24,25(OH)2

    D3

    . Alternatively, the phenotype

    could be caused by the abnormally high levels of

    1,25-(OH)2

    D3

    resulting from the deletion of this cata-

    bolic enzyme. Indeed, crossing the 24OHase-null mice

    with VDR-null mice completely rescues the decreased

    embryonic viability and ossification defects, thus sup-

    porting the concept that 1,25-(OH)2D3 toxicity leads to

    the abnormal phenotype in the 24OHase-knockoutmice. Moreover, this model also suggests that 24-

    hydroxylated metabolites are not required for normal

    intramembranous ossification (114).

    BEYOND 1,25-(OH)2D3 AND VDR: NOVEL

    LIGANDS AND ALTERNATIVE RECEPTORS

    Novel Vitamin D Ligands

    The therapeutic potential of 1,25-(OH)2

    D3

    continues to

    expand. In addition to treating disorders of mineral

    metabolism and diseases of the skeleton, such as

    rickets, osteoporosis, and renal osteodystrophy, 1,25-

    (OH)2

    D3

    has significant therapeutic potential for pa-

    thologies such as cancer, autoimmune syndromes,

    and psoriasis. However, 1,25-(OH)2

    D3

    itself has a nar-

    row therapeutic window limited by the development of

    toxic hypercalcemia. The increase in calcium isachieved both by enhanced intestinal absorption and

    by liberation of calcium from the skeleton, eventually

    leading to decreased bone mass at higher doses. This,

    of course, counteracts the beneficial effects of 1,25-

    (OH)2D3 for the treatment of bone diseases. Conse-

    quently, more than 800 synthetic 1,25-(OH)2

    D3

    ana-

    logs have been developed in attempt to preserve the

    favorable activities of 1,25-(OH)2

    D3

    while avoiding the

    side effects (115).

    Calcipotriol (MC 903) is an analog that has been

    used to treat psoriasis for nearly 15 yr, and it is cur-

    rently considered a first-line therapy for the disease

    (116). Calcipotriol improves psoriasis by inhibiting pro-

    liferation and promoting differentiation of keratino-

    cytes, but it does not cause hypercalcemia or de-

    creased bone mass. This selectivity can be attributed

    to calcipotriols low affinity for vitamin D binding pro-tein, the major vitamin D transport protein in the cir-

    culation (117), and the fact that it is applied topically,

    thus restricting its actions to the skin. 1,25-(OH)2

    D3

    analogs are also valuable in treating renal osteodys-

    trophy, a devastating consequence of chronic renal

    failure. Kidney disease, due to a variety of causes,

    often leads to reduced 1,25-(OH)2

    D3

    production and

    impaired phosphate excretion resulting in abnormally

    high PTH levels. The decreased calcitriol levels com-

    bined with secondary hyperparathyroidism, in turn,result in increased bone turnover (118). Treatment with

    1,25-(OH)2

    D3

    is effective in suppressing PTH levels

    and improving the initial skeletal abnormalities. How-

    ever, due to the narrow therapeutic window, 1,25-

    (OH)2

    D3

    often causes hypercalcemia and additional

    bone disease from inappropriately low bone turnover.

    Two 1,25-(OH)2D3 analogs are currently approved for

    treatment of secondary hyperparathyroidism in the

    United States. 19-Nor-D2

    (paricalcitol; Ref. 119) and

    1(OH)D2

    (doxercalciferol; Ref. 120) are as effective as

    1,25-(OH)2D3 in reducing PTH levels but do not result

    in significant hypercalcemia. Although the mecha-

    nisms of the selectivity of these two analogs remain

    unclear, they represent significant improvements intreatment regimens for renal osteodystrophy.

    Although neither vitamin D3

    nor 1,25-(OH)2

    D3

    ana-

    logs are currently Food and Drug Administration

    (FDA)-approved for treating osteoporosis in the United

    States, these compounds are widely used to prevent

    and treat osteoporosis throughout the world (121,

    122). Furthermore, vitamin D3

    is currently recom-

    mended as a dietary supplement in addition to any

    pharmacological treatment for all patients with de-

    creased bone mass or osteoporosis (123). Still, the

    higher doses of 1,25-(OH)2

    D3

    required for maximal

    improvement in bone density cause significant hyper-

    784 Mol Endocrinol, May 2003, 17(5):777791 Minireview Sutton and MacDonald

    by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    9/15

    calcemia. Peleg and colleagues (124) tested 1,25-

    (OH)2

    D3

    analogs for their ability to improve bone min-

    eral density in the ovariectomized rat model of

    osteoporosis. One novel analog, Ro-26-9228, protects

    against osteopenia, but it does not increase serum

    calcium except at very high doses. These observa-

    tions are potentially explained by the tissue-selectiveaction of Ro-26-9228, which stimulates osteocalcin

    and osteopontin expression in osteoblasts but does

    not affect calbindin D9K

    or plasma membrane calcium

    pump expression in the intestine (124). Shevde et al.

    (125) found that another analog, 2-methylene-19-nor-

    (20S)-1,25(OH)2

    D3

    (2MD), potently stimulates bone

    formation in vitro and markedly improves bone mass in

    ovariectomized rats without dramatically increasing

    serum calcium. Such studies support the concept that

    more selective 1,25-(OH)2

    D3

    analogs will be useful

    therapies for osteoporosis by enhancing bone mineral

    density without causing toxic hypercalcemia.

    In addition to these and numerous other designer

    analogs, a recent study suggests the possibility that

    natural compounds other than 1,25-(OH)2

    D3

    may

    serve as tissue-selective activators of VDR-mediated

    responses. An unexpected ligand for VDR was discov-

    ered from studies with bile acid compounds (126).

    Metabolic lipophilic molecules such as bile acids ac-

    tivate many of the orphan nuclear receptors, including

    farnesoid X receptor and steroid xenobiotic receptor/

    pregnane X receptor (127129). Recently, Makishimaet al. (126) screened classical nuclear receptors to

    identify those that were activated by the bile acid

    lithocholic acid (LCA). Surprisingly, they found that

    LCA and its metabolites directly bind and activate

    VDR. However, this activation requires micromolarconcentrations of LCA, whereas VDR is activated by

    nanomolar amounts of 1,25-(OH)2

    D3

    . Nonetheless,

    LCA- or 1,25-(OH)2

    D3

    -liganded VDR also stimulates

    the expression of endogenous CYP3A, the P450 en-

    zyme responsible for degradation of LCA in the liver

    and the intestine. LCA is implicated as a toxin that

    promotes colorectal carcinogenesis (130, 131),

    whereas 1,25-(OH)2

    D3

    is protective against colon can-

    cer (132). Thus, induction of CYP3A by 1,25-(OH)2

    D3

    and by LCA itself may represent a detoxification path-

    way for LCA, as well as explain the potential preven-

    tative effects of 1,25-(OH)2

    D3

    in colon cancer. Al-

    though this study awaits further in vivo confirmation, it

    clearly raises the possibility that VDR may be activatedby other naturally occurring ligands.

    VDR Isoforms

    Many nuclear receptors, such as RAR, RXR, and thy-

    roid receptor, have multiple isoforms that are encoded

    by separate genes (133). Unlike these nuclear recep-

    tors, only one human VDR genetic locus has been

    identified (134), and the genomic database does not

    indicate additional highly related sequences. Although

    the cDNA encoding the human VDR was cloned nearly

    15 yr ago (8), only recently have several significant

    variations in the VDR gene, transcript, and protein

    sequences been discovered. At least 14 distinct tran-

    scripts of human VDR have been identified that differ

    in their 5 ends (135). These transcripts arise from

    alternative mRNA splicing and differential promoter

    usage. Most of these variant transcripts utilize the

    same initiator codon, producing a VDR that is 427amino acids in length. However, two transcripts have

    upstream in-frame methionines that potentially gener-

    ate N-terminal extensions in VDR of 50 or 23 amino

    acids (135). Low levels of endogenous VDRB1 protein,

    the 50-amino-acid-extended variant, have been de-

    tected in osteoblast, colon cancer, and kidney cell

    lines (136). Interestingly, VDRB1 has reduced tran-

    scriptional activity compared with classical VDR.

    Whether the levels of expression of these isoforms are

    substantial and whether these isoforms result in al-

    tered biological activity in vivo remains unresolved.

    Multiple polymorphic variations also exist in VDR in

    the human population (137). The vast majority of these

    polymorphisms do not result in a structural alteration

    in the VDR protein, with the exception of the Fok I

    variant (138). The Fok I polymorphism is located at the

    original initiator ATG, which is part of a Fok I endonu-

    clease site. In some humans, there is an ATG 224 ACG

    transition at the 1 position, eliminating the transla-

    tional initiation site and Fok I recognition sequence.

    This transition results in the use of an in-frame methi-

    onine as the initiator codon at the 4 position. Thus,

    either a 427 (Met-1) or a 424 (Met-4) amino acid pro-

    tein is expressed. Numerous epidemiological studies

    suggest an association between the shorter form of

    VDR and increased bone mineral density in humans

    (139143). The molecular mechanism of this associa-tion remains unclear, but there is suggestive evidencethat the Met-4 VDR displays enhanced transcriptional

    activity due to increased interaction with basal tran-

    scription factor II B (12). Although these findings re-

    main controversial (144), they raise the possibility that

    the N terminus possesses some type of structure that

    influences transcriptional activity. This, combined with

    the observations that other N-terminal extensions of

    VDR may have reduced transcriptional activity, sug-

    gests that there may be inhibitory domains at the

    extreme N terminus of VDR that decrease its transac-

    tivation potential.

    A Membrane Receptor? Rapid, NongenomicEffects of Vitamin D and Its Metabolites

    According to the classical paradigm of nuclear recep-

    tor action, ligand-activated nuclear receptors recruit

    the basal transcriptional machinery and other activator

    complexes to the promoters of target genes to induce

    transcription. Because these responses require tran-

    scription and translation of target genes, they are typ-

    ically delayed by at least 30 min. However, more rapid

    (within seconds to minutes) effects in response to

    steroid hormones are also apparent. The rapid nature

    of these effects and their relative insensitivity to tran-

    Minireview Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777791 785

    by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    10/15

    scriptional and translational inhibitors, such as actino-

    mycin D and cycloheximide, precludes the possibility

    that the traditional genomic model is operating. Re-cent attention to these rapid, nongenomic hormoneeffects has spawned renewed interest in this long-

    standing area of membrane-initiated signaling in the

    steroid hormone field (145). Over two decades ago,1,25-(OH)

    2D3 was shown to evoke transcellular move-

    ment of calcium across chick enterocytes within sev-

    eral minutes (146, 147). This phenomenon is theorized

    to be adaptively beneficial for a hypocalcemic animal

    in that rapid absorption of calcium occurs without a

    delayed response involving transcription and transla-

    tion of calcium-binding proteins or calcium transport-

    ers (147). In addition to the enterocyte, the osteoblast

    is a target for 1,25-(OH)2

    D3

    -induced rapid calcium

    mobilization from internal stores, a process that in-

    volves a membrane-initiated signaling cascade includ-

    ing phospholipase C activation and inositol triphos-

    phate formation (148). This process also occurs in

    skeletal muscle cells, in which 1,25-(OH)2

    D3

    induces

    calcium release from the sarcoplasmic reticulum (149),

    potentially through MAPK activation (150). These are

    just a few of the many examples of in vitro systems in

    which these nongenomic actions of 1,25-(OH)2

    D3

    have been studied.

    Although the rapid effects of 1,25-(OH)2

    D3

    and

    numerous other steroids are well documented, the

    field is hindered by the inability to identify the puta-

    tive membrane receptors that trigger these non-

    genomic effects. Although suggestive biochemical

    and immunological data (151) indicate that the

    membrane VDR is a distinct gene product, a true

    protein representing this receptor remains elusive. Apromising new approach in the nongenomic field is

    the use of 1,25-(OH)2

    D3

    analogs to discriminate be-

    tween receptors that mediate membrane-initiated

    events and those that mediate the classical nuclear

    effects of 1,25-(OH)2

    D3

    . Song et al. (152) showed

    that 6-s-cis-locked analogs of 1,25-(OH)2

    D3

    stimu-

    late rapid phosphorylation of MAPK in leukemic

    cells, yet these analogs bind poorly to VDR and are

    weak activators of VDR-mediated transcription.

    These studies also support the possibility of two

    separate gene products involved in either rapid,

    nongenomic signaling or in classical transcriptional

    activation by 1,25-(OH)2

    D3

    . In fact, two preliminary

    reports indicate that annexin II, a membrane-asso-ciated calcium-binding protein, may bind 1,25-

    (OH)2

    D3

    and function as its membrane receptor

    (153, 154). In contrast to these studies, analysis of

    mice carrying a mutated VDR lacking a DBD implies

    that the classical nuclear VDR mediates both

    genomic and nongenomic responses (84). Osteo-

    blast cultures derived from VDR-mutant mice are

    unable to initiate a rapid calcium flux in response to

    1,25-(OH)2

    D3

    , suggesting that some nongenomic

    responses require a functional nuclear VDR. Al-

    though a full array of other nongenomic responses

    needs to be tested, the VDRKO strains will provide

    important tools to decipher the molecular require-

    ments of classical VDR that mediate the genomic

    and nongenomic effects of 1,25-(OH)2

    D3

    in vivo.

    SUMMARY: FRONTIERS IN VITAMIN D

    The molecular, cellular, structural, and genetic studies

    of the past decade have impacted our understanding

    of the vitamin D endocrine system in a number of

    significant ways. First and foremost, the genetic mod-

    els solidify the fundamental roles that both 1,25-

    (OH)2

    D3

    and VDR play in ensuring that an organism

    obtains sufficient calcium and phosphate from the

    environment to sustain life and normal development.

    Lack of either functional VDR or active 1,25-(OH)2

    D3

    leads to profound, life-threatening hypocalcemia and

    undermineralized skeletal tissue. Previous classical

    nutritional manipulations and identification of mutated

    VDR as the causative defect in HVDRR have estab-

    lished this central function of 1,25-(OH)2

    D3

    long before

    the creation of genetic mouse models. However, the

    development and analysis of such models are central

    in formulating a detailed physiological picture of 1,25-

    (OH)2

    D3

    signaling and in reinforcing a direct link be-

    tween the various genes within the endocrine system

    and mineral ion dysregulation.

    On the other hand, the knockout studies provide

    somewhat of a surprise and, to a certain extent, a bit

    of a disappointment for those interested in 1,25-

    (OH)2

    D3

    and bone biology. In particular, specific cells

    of the osteoblast and osteoclast lineage have gar-

    nered considerable research attention over the pastseveral decades as important direct targets of 1,25-

    (OH)2

    D3

    in preserving skeletal integrity. However, the

    striking skeletal defects observed in the VDRKO are

    corrected by simply providing the animals with sup-

    plemental dietary calcium. Does this mean that 1,25-

    (OH)2

    D3

    does not act as a direct bone-a-fide hor-mone in the skeleton? Clearly, more cellular and

    genetic approaches are needed to fully answer this

    question and to test whether VDR and 1,25-(OH)2

    D3

    play more limited or specialized roles in the developing

    or aging skeleton. Although the jury is still out on the

    bone, striking new biologies are emerging from the

    VDRKO studies, indicating potential functions for

    1,25-(OH)2D3 in diverse processes such as hair folliclecycling, blood pressure regulation, and mammary

    gland development that are independent of mineral ion

    homeostasis. Moreover, studies on alopecia in

    VDRKO mice raise the exciting possibility that VDR

    acts in a ligand-independent fashion and may stimu-

    late further exploration into the molecular and cellular

    functions of unliganded VDR.

    Beyond the physiological information gleaned from

    the genetic mouse models, recent progress in other

    areas of the vitamin D field is revealing additional

    molecular details of the endocrine system. Description

    of the crystal structure of VDR has supplied an atomic

    786 Mol Endocrinol, May 2003, 17(5):777791 Minireview Sutton and MacDonald

    by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    11/15

    view of the VDR protein, highlighting the expansive

    binding pocket associated with both natural and syn-

    thetic ligands. Further refinement of this structure

    complexed with coactivators undoubtedly will allow

    for the future rational design of selective VDR-targeted

    drugs. Some of the additional molecular requirements

    for the transcriptional activity of 1,25-(OH)2D3-ligan-ded VDR have been elucidated and are beginning to

    be assembled into an integrated model of coactivator

    cooperativity. Furthermore, it is becoming increasingly

    clear that some actions of 1,25-(OH)2

    D3

    cannot be

    explained by the traditional model of 1,25-(OH)2D3-

    bound VDR acting solely as a transcription factor in the

    nucleus. Novel VDR ligands, including synthetic ana-

    logs and natural compounds such as bile acids, are

    broadening our understanding of the therapeutic po-

    tential and physiological intricacies of vitamin D. Like-

    wise, alternative receptors, both related to and distinct

    from the classical nuclear VDR, are emerging as sig-

    nificant participants mediating the biological effects of

    vitamin D compounds. The four areas of progress

    covered in this minireview have filled in many gaps in

    our knowledge of vitamin D, but they also have raised

    a multitude of new questions that await answering with

    the new molecular, pharmacological, and genetic tools

    developed in recent years.

    Acknowledgments

    We apologize to many colleagues whose excellent primarypublications may not have been cited in this minireview dueto space limitations.

    Received November 1, 2002. Accepted March 5, 2003.Address all correspondence and requests for reprints to:

    Paul N. MacDonald, Ph.D., Department of Pharmacology,Case Western Reserve University, 10900 Euclid Avenue,Cleveland, Ohio 44106. E-mail: [email protected].

    This work was supported by NIH Grants R01-DK-50348and R01-DK-53980 (to P.N.M.), by an awardfrom theMedicalScientist Training Program NIH Grant T32-GM-007250 (toA.L.M.S.), and by a Pharmaceutical Manufacturers Associa-tion Foundation Pre-Doctoral Fellowship (to A.L.M.S.).

    REFERENCES

    1. Brown AJ, Dusso A, Slatopolsky E 1999 Vitamin D. Am JPhysiol 277:F157F1752. van Leeuwen JP, van Driel M, van den Bemd GJ, Pols

    HA 2001 Vitamin D control of osteoblast function andbone extracellular matrix mineralization. Crit Rev Eu-karyot Gene Expr 11:199226

    3. Owen TA, Aronow MS, Barone LM, Bettencourt B, SteinGS, Lian JB 1991 Pleiotropic effects of vitamin D onosteoblast gene expression are related to the prolifera-tive and differentiated state of the bone cell phenotype:dependency upon basal levels of gene expression, du-ration of exposure, and bone matrix competency innormal rat osteoblast cultures. Endocrinology 128:14961504

    4. Bar-Shavit Z, Teitelbaum SL, Reitsma P, Hall A, PeggLE, Trial J, Kahn AJ 1983 Induction of monocytic differ-

    entiation and bone resorption by 1,25-dihydroxyvitaminD3. Proc Natl Acad Sci USA 80:59075911

    5. Jones G, Strugnell SA, DeLuca HF 1998 Current under-standing of the molecular actions of vitamin D. PhysiolRev 78:11931231

    6. Horst RL, Reinhardt TA 1997 Vitamin D metabolism. In:Feldman D, Glorieux, FH, Pike JW, eds. Vitamin D. San

    Diego: Academic Press; 13327. McDonnell DP, Mangelsdorf DJ, Pike JW, Haussler MR,

    OMalley BW 1987 Molecular cloning of complementaryDNA encoding the avian receptor for vitamin D. Science235:12141217

    8. Baker AR, McDonnell DP, Hughes M, Crisp TM, Man-gelsdorf DJ, Haussler MR, Pike JW, Shine J, OMalleyBW 1988 Cloning and expression of full-length cDNAencoding human vitamin D receptor. Proc Natl Acad SciUSA 85:32943298

    9. Haussler MR, Whitfield GK, Haussler CA, Hsieh JC,Thompson PD, Selznick SH, Dominguez CE, JurutkaPW 1998 The nuclear vitamin D receptor: biological andmolecular regulatory properties revealed. J Bone MinerRes 13:325349

    10. Mangelsdorf DJ, Evans RM 1995 TheRXR heterodimers

    and orphan receptors. Cell 83:84185011. McKenna NJ, Lanz RB, OMalley BW 1999 Nuclearreceptor coregulators: cellular and molecular biology.Endocr Rev 20:321344

    12. Jurutka PW, Remus LS, Whitfield GK, Thompson PD,Hsieh J-C, Zitzer H, Tavakkoli P, Galligan MA, DangHTL, Haussler CA, Haussler MR 2000 The polymorphicN terminus in human vitamin D receptor isoforms influ-ences transcriptional activity by modulating interactionwith transcription factor IIB. Mol Endocrinol 14:401420

    13. McDonnell DP, Scott RA, Kerner SA, OMalley BW, PikeJW 1989 Functional domains of the human vitamin D3receptor regulate osteocalcin gene expression. Mol En-docrinol 3:635644

    14. Nakajima S, Hsieh JC, MacDonald PN, Galligan MA,Haussler CA, Whitfield GK, Haussler MR 1994 The C-terminal region of the vitamin D receptor is essential toform a complex with a receptor auxiliary factor requiredfor high affinity binding to the vitamin D-responsiveelement. Mol Endocrinol 8:159172

    15. Masuyama H, Brownfield CM, St-Arnaud R, MacDonaldPN 1997 Evidence for ligand-dependent intramolecularfolding of the AF-2 domain in vitamin D receptor-acti-vated transcription and coactivator interaction. Mol En-docrinol 11:15071517

    16. Danielian PS, White R, Lees JA, Parker MG 1992 Iden-tification of a conserved region required for hormonedependent transcriptional activation by steroid hor-mone receptors. EMBO J 11:10251033

    17. Mangelsdorf DJ, Thummel C, Beato M, Herrlich P,Schutz G, Umesono K, Blumberg B, Kastner P, Mark M,Chambon P, Evans RM 1995 The nuclear receptorsuperfamily: the second decade. Cell 83:835839

    18. Schwabe JW, Chapman L, Finch JT, Rhodes D 1993The crystal structure of the estrogen receptor DNA-binding domain bound to DNA: how receptors discrim-inatebetween their response elements. Cell 75:567578

    19. Wagner RL, Apriletti JW, McGrath ME, West BL, BaxterJD, Fletterick RJ 1995 A structural role for hormone inthe thyroid hormone receptor. Nature 378:690697

    20. Renaud JP, Rochel N, Ruff M, Vivat V, Chambon P,Gronemeyer H, Moras D 1995 Crystal structure of theRAR- ligand-binding domain bound to all-trans reti-noic acid. Nature 378:681689

    21. Bourguet W, Ruff M, Chambon P, Gronemeyer H, Mo-ras D 1995 Crystal structure of the ligand-binding do-main of the human nuclear receptor RXR-. Nature375:377382

    Minireview Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777791 787

    by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    12/15

    22. WeathermanRV, Fletterick RJ,ScanlanTS1999 Nuclear-receptor ligands and ligand-binding domains. Annu RevBiochem 68:559581

    23. Rochel N, Wurtz JM, Mitschler A, Klaholz B, Moras D2000 The crystal structure of the nuclear receptor forvitamin D bound to its natural ligand. Mol Cell5:173179

    24. Rochel N, Tocchini-Valentini G, Egea PF, Juntunen K,Garnier JM, Vihko P, Moras D 2001 Functional andstructural characterization of the insertion region in theligand binding domain of the vitamin D nuclear receptor.Eur J Biochem 268:971979

    25. Peleg S, Sastry M, Collins ED, Bishop JE, Norman AW1995 Distinct conformational changes induced by 20-epi analogues of 1,25-dihydroxyvitamin D3 are asso-ciated with enhanced activation of the vitamin D recep-tor. J Biol Chem 270:1055110558

    26. Carlberg C, Quack M, Herdick M, Bury Y, Polly P, ToellA 2001 Central role of VDR conformations for under-standing selective actions of vitamin D(3) analogues.Steroids 66:213221

    27. Yang W, Freedman LP 1999 20-Epi analogues of 1,25-dihydroxyvitamin D3 are highly potent inducers of DRIPcoactivator complex binding to the vitamin D3 receptor.J Biol Chem 274:1683816845

    28. Norman AW, Manchand PS, Uskokovic MR, OkamuraWH, Takeuchi JA, Bishop JE, Hisatake JI, Koeffler HP,Peleg S 2000 Characterization of a novel analogue of1,25(OH)

    2-vitamin D

    3with two side chains: interaction

    with its nuclear receptor and cellular actions. J MedChem 43:27192730

    29. Tocchini-Valentini G, Rochel N, Wurtz JM, Mitschler A,Moras D 2001 Crystal structures of the vitamin D re-ceptor complexed to superagonist 20-epi ligands. ProcNatl Acad Sci USA 98:54915496

    30. Tasset D, Tora L, Fromental C, Scheer E, Chambon P1990 Distinct classes of transcriptional activating do-mains function by different mechanisms. Cell 62:11771187

    31. Cavailles V, Dauvois S, Danielian PS, Parker MG 1994

    Interaction of proteins with transcriptionally active es-trogen receptors. Proc Natl Acad Sci USA 91:1000910013

    32. Halachmi S, Marden E, Martin G, MacKay H, Abbon-danza C, Brown M 1994 Estrogen receptor-associatedproteins: possible mediators of hormone-induced tran-scription. Science 264:14551458

    33. Onate S, Tsai, SY, Tsai, M-J, OMalley, BW 1995 Se-quence and characterization of a coactivator for thesteroid hormone receptor superfamily. Science 270:13541357

    34. Leid M 1994 Ligand-induced alteration of the proteasesensitivity of retinoid X receptor . J Biol Chem 269:1417514181

    35. Feng W, Ribeiro RCnJ, Wagner RL, Nguyen H, AprilettiJW, Fletterick RJ, Baxter JD, Kushner PJ, West BL 1998Hormone-dependent coactivator binding to a hydro-phobic cleft on nuclear receptors. Science 280:17471749

    36. Nolte RT, Wisely GB, Westin S, Cobb JE, Lambert MH,Kurokawa R, Rosenfeld MG, Willson TM, Glass CK,Milburn MV 1998 Ligand binding and co-activatorassembly of the peroxisome proliferator-activatedreceptor-. Nature 395:137143

    37. Shiau AK, Barstad D, Loria PM, Cheng L, Kushner PJ,Agard DA, Greene GL 1998 The structural basis ofestrogen receptor/coactivator recognition and the an-tagonism of this interaction by tamoxifen. Cell 95:927937

    38. Heery DM, Kalkhoven E, Hoare S, Parker MG 1997 Asignature motif in transcriptional co-activators mediatesbinding to nuclear receptors. Nature 387:733736

    39. McKenna NJ, OMalley BW 2002 Combinatorial controlof gene expression by nuclear receptors and coregula-tors. Cell 108:465474

    40. McKenna NJ, OMalley BW 2002 Minireview: nuclearreceptor coactivatorsan update. Endocrinology 143:24612465

    41. Rosenfeld MG, Glass CK 2001 Coregulator codes of

    transcriptional regulation by nuclear receptors. J BiolChem 276:3686536868

    42. Hong H, Kohli K, Trivedi A, Johnson DL, Stallcup MR1996 GRIP1, a novel mouse protein that serves as atranscriptional coactivator in yeast for the hormonebinding domains of steroid receptors. Proc Natl AcadSci USA 93:49484952

    43. Voegel JJ, Heine MJ, Zechel C, Chambon P, Grone-meyer H 1996 TIF2, a 160 kDa transcriptional mediatorfor the ligand-dependent activation function AF-2 ofnuclear receptors. EMBO J 15:36673675

    44. Li H, Gomes PJ, Chen JD 1997 RAC3, a steroid/nuclearreceptor-associated coactivator that is related toSRC-1 andTIF2. Proc Natl Acad Sci USA94:84798484

    45. Anzick SL, Kononen J, Walker RL, Azorsa DO, TannerMM, Guan XY, Sauter G, Kallioniemi OP, Trent JM,Meltzer PS 1997 AIB1, a steroid receptor coactivatoramplified in breast and ovarian cancer. Science 277:965968

    46. Chen H, Lin RJ, Schiltz RL, Chakravarti D, Nash A, NagyL, Privalsky ML, Nakatani Y, Evans RM 1997 Nuclearreceptor coactivator ACTR is a novel histone acetyl-transferase and forms a multimeric activation complexwith P/CAF and CBP/p300. Cell 90:569580

    47. Torchia J, Rose DW, Inostroza J, Kamei Y, Westin S,Glass CK, Rosenfeld MG 1997 The transcriptional co-activator p/CIP binds CBP and mediates nuclear-recep-tor function. Nature 387:677684

    48. Takeshita A, Cardona GR, Koibuchi N, Suen CS, ChinWW 1997 TRAM-1, a novel 160-kDa thyroid hormonereceptor activator molecule, exhibits distinct propertiesfrom steroid receptor coactivator-1. J Biol Chem 272:2762927634

    49. Kraichely DM, Collins 3rd JJ, DeLisle RK, MacDonaldPN 1999 The autonomous transactivation domain inhelix H3 of the vitamin D receptor is required for trans-activation and coactivator interaction. J Biol Chem 274:1435214358

    50. Ikeda M, Kawaguchi A, Takeshita A, Chin WW, Endo T,Onaya T 1999 CBP-dependent and independent en-hancing activity of steroid receptor coactivator-1 in thy-roid hormone receptor-mediated transactivation. MolCell Endocrinol 147:103112

    51. Spencer TE, Jenster G, Burcin MM, Allis CD, Zhou J,Mizzen CA, McKenna NJ, Onate SA, Tsai SY, Tsai MJ,OMalley BW 1997 Steroid receptor coactivator-1 is ahistone acetyltransferase. Nature 389:194198

    52. Chakravarti D, LaMorte VJ, Nelson MC, Nakajima T,Schulman IG, Juguilon H, Montminy M, Evans RM 1996Role of CBP/P300 in nuclear receptor signalling. Nature383:99103

    53. Kamei Y, Xu L, Heinzel T, Torchia J, Kurokawa R, GlossB, Lin SC, Heyman RA, Rose DW, Glass CK, RosenfeldMG 1996 A CBP integrator complex mediates transcrip-tional activation and AP-1 inhibition by nuclear recep-tors. Cell 85:403414

    54. Rachez C, Suldan Z, Ward J, Chang CP, Burakov D,Erdjument-Bromage H, Tempst P, Freedman LP 1998 Anovel protein complex that interacts with the vitamin D3receptor in a ligand-dependent manner and enhancesVDR transactivation in a cell-free system. Genes Dev12:17871800

    55. Rachez C, Lemon BD, Suldan Z, Bromleigh V, GambleM, Naar AM, Erdjument-Bromage H, Tempst P, Freed-man LP 1999 Ligand-dependent transcription activation

    788 Mol Endocrinol, May 2003, 17(5):777791 Minireview Sutton and MacDonald

    by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    13/15

    by nuclear receptors requires the DRIP complex. Nature398:824828

    56. Fondell JD, Ge H, Roeder RG 1996 Ligand induction ofa transcriptionally active thyroid hormone receptor co-activator complex. Proc Natl Acad Sci USA 93:83298333

    57. Jiang YW, Veschambre P, Erdjument-Bromage H,

    Tempst P, Conaway JW, Conaway RC, Kornberg RD1998 Mammalian mediator of transcriptional regulationand its possible role as an end-point of signal transduc-tion pathways. Proc Natl Acad Sci USA 95:85388543

    58. Rachez C, Gamble M, Chang CP, Atkins GB, Lazar MA,Freedman LP 2000 The DRIP complex and SRC-1/p160coactivators share similar nuclear receptor binding de-terminants but constitute functionally distinct com-plexes. Mol Cell Biol 20:27182726

    59. Shang Y, Hu X, DiRenzo J, Lazar MA, Brown M 2000Cofactor dynamics and sufficiency in estrogen recep-tor-regulated transcription. Cell 103:843852

    60. Burakov D, Crofts LA, Chang CP, Freedman LP 2002Reciprocal recruitment of DRIP/mediator and p160 co-activator complexes in vivo by estrogen receptor. J BiolChem 277:1435914362

    61. Sharma D, Fondell JD 2002 Ordered recruitment of

    histone acetyltransferases and the TRAP/Mediatorcomplex to thyroid hormone-responsive promoters invivo. Proc Natl Acad Sci USA 99:79347939

    62. Chiba N, Suldan Z, Freedman LP, Parvin JD 2000 Bind-ing of liganded vitamin D receptor to the vitamin Dreceptor interacting protein coactivator complex in-duces interaction with RNA polymerase II holoenzyme.J Biol Chem 275:1071910722

    63. Baudino TA, Kraichely DM, Jefcoat Jr SC, WinchesterSK, Partridge NC, MacDonald PN 1998 Isolation andcharacterization of a novel coactivator protein, NCoA-62, involved in vitamin D-mediated transcription. J BiolChem 273:1643416441

    64. Wieland C, Mann S, von Besser H, Saumweber H 1992The Drosophila nuclear protein Bx42, which is found inmany puffs on polytene chromosomes, is highlycharged. Chromosoma 101:517525

    65. Dahl R, Wani B, Hayman MJ 1998 The Ski oncoproteininteracts with Skip, the human homolog of DrosophilaBx42. Oncogene 16:15791586

    66. Zhang C, Baudino TA, Dowd DR,Tokumaru H, Wang W,MacDonald PN 2001 Ternary complexes and coopera-tive interplay between NCoA-62/Ski-interacting proteinand steroid receptor coactivators in vitamin D receptor-mediated transcription. J Biol Chem 276:4061440620

    67. Zhou Z, Licklider LJ, Gygi SP, Reed R 2002 Compre-hensive proteomic analysis of the human spliceosome.Nature 419:182185

    68. Makarov EM, Makarova OV, Urlaub H, Gentzel M, WillCL, Wilm M, Luhrmann R 2002 Small nuclear ribonu-cleoprotein remodeling during catalytic activation of thespliceosome. Science 298:22052208

    69. Auboeuf D, Honig A, Berget SM, OMalley BW 2002

    Coordinate regulation of transcription and splicing bysteroid receptor coregulators. Science 298:41641970. Xu J, Qiu Y, DeMayo FJ, Tsai SY, Tsai M-J, OMalley

    BW 1998 Partial hormone resistance in mice with dis-ruption of the steroid receptor coactivator-1 (SRC-1)gene. Science 279:19221925

    71. Gehin M, Mark M, Dennefeld C, Dierich A, GronemeyerH, Chambon P 2002 The function of TIF2/GRIP1 inmouse reproduction is distinct from those of SRC-1 andp/CIP. Mol Cell Biol 22:59235937

    72. Xu J, Liao L, Ning G, Yoshida-Komiya H, Deng C,OMalley BW 2000 The steroid receptor coactivatorSRC-3 (p/CIP/RAC3/AIB1/ACTR/TRAM-1) is requiredfor normal growth, puberty, female reproductive func-tion, and mammary gland development. Proc Natl AcadSci USA 97:63796384

    73. Kostrouchova M, Housa D, Kostrouch Z, Saudek V, RallJE 2002 SKIP is an indispensable factor for Caenorhab-ditis elegans development. Proc Natl Acad Sci USA99:92549259

    74. Stenoien DL, Nye AC, Mancini MG, Patel K, Dutertre M,OMalley BW, Smith CL, Belmont AS, Mancini MA 2001Ligand-mediated assembly and real-time cellular dy-

    namics of estrogen receptor -coactivator complexesin living cells. Mol Cell Biol 21:4404441275. Becker M, Baumann C, John S, Walker DA, Vigneron M,

    McNally JG, Hager GL 194 2002 Dynamic behavior oftranscription factors on a natural promoter in living cells.EMBO Rep 3:11881181

    76. Hughes MR, Malloy PJ, Kieback DG, Kesterson RA,Pike JW, Feldman D, OMalley BW 1988 Point muta-tions in the human vitamin D receptor gene associatedwith hypocalcemic rickets. Science 242:17021705

    77. Malloy PJ, Pike JW, Feldman D 1999 The vitamin Dreceptor and the syndrome of hereditary 1,25-dihy-droxyvitamin D-resistant rickets. Endocr Rev 20:156188

    78. Yoshizawa T, HandaY, Uematsu Y, Takeda S, Sekine K,Yoshihara Y, Kawakami T, Arioka K, Sato H, UchiyamaY, Masushige S, Fukamizu A, Matsumoto T, Kato S

    1997 Mice lacking the vitamin D receptor exhibit im-paired bone formation, uterine hypoplasia and growthretardation after weaning. Nat Genet 16:391396

    79. Li YC, Pirro AE, Amling M, Delling G, Baron R, BronsonR, Demay MB 1997 Targeted ablation of the vitamin Dreceptor: an animal model of vitamin D-dependent rick-ets type II with alopecia. Proc Natl Acad Sci USA 94:98319835

    80. Li YC, Amling M,Pirro AE, Priemel M,Meuse J, Baron R,Delling G, Demay MB 1998 Normalization of mineral ionhomeostasis by dietary means prevents hyperparathy-roidism, rickets, and osteomalacia, but not alopecia invitamin D receptor-ablated mice. Endocrinology 139:43914396

    81. Amling M, Priemel M, Holzmann T, Chapin K, RuegerJM, Baron R, Demay MB 1999 Rescue of the skeletalphenotype of vitamin D receptor-ablated mice in thesetting of normal mineral ion homeostasis: formal his-tomorphometric and biomechanical analyses. Endocri-nology 140:49824987

    82. Van Cromphaut SJ, Dewerchin M, Hoenderop JG,Stockmans I, Van Herck E, Kato S, Bindels RJ, Collen D,Carmeliet P, Bouillon R, Carmeliet G 2001 Duodenalcalcium absorption in vitamin D receptor-knockoutmice: functional and molecular aspects. Proc Natl AcadSci USA 98:1332413329

    83. Li YC, Bolt MJG, Cao L-P, Sitrin MD 2001 Effects ofvitamin D receptor inactivation on the expression ofcalbindins and calcium metabolism. Am J Physiol En-docrinol Metab 281:E558E564

    84. Erben RG, Soegiarto DW, Weber K, Zeitz U, LieberherrM, Gniadecki R, Moller G, Adamski J, Balling R 2002Deletion of deoxyribonucleic acid binding domain of the

    vitamin D receptor abrogates genomic and nongenomicfunctions of vitamin D. Mol Endocrinol 16:1524153785. Haussler MR, Haussler CA, Jurutka PW, Thompson PD,

    Hsieh JC, Remus LS, Selznick SH, Whitfield GK 1997The vitamin D hormone and its nuclear receptor: mo-lecular actions and disease states. J Endocrinol 154:S57S73

    86. Sakai Y, Demay MB 2000 Evaluation of keratinocyteproliferation and differentiation in vitamin D receptorknockout mice. Endocrinology 141:20432049

    87. Sakai Y, Kishimoto J, Demay MB 2001 Metabolic andcellular analysis of alopecia in vitamin D receptor knock-out mice. J Clin Invest 107:961966

    88. Panda DK, Miao D, Tremblay ML, Sirois J, Farookhi R,Hendy GN, Goltzman D 2001 Targeted ablation of the25-hydroxyvitamin D 1-hydroxylase enzyme: evidence

    Minireview Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777791 789

    by on November 16, 2006mend.endojournals.orgDownloaded from

    http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/http://mend.endojournals.org/
  • 8/7/2019 D Vitamin D- More Than a Bone-a-Fide Hormone

    14/15

    for skeletal, reproductive, and immune dysfunction.Proc Natl Acad Sci USA 98:74987503

    89. Masuyama H, Jefcoat Jr SC, MacDonald PN 1997 TheN-terminal domain of transcription factor IIB is requiredfor direct interaction with the vitamin D receptor andparticipates in vitamin D-mediated transcription. MolEndocrinol 11:218228

    90. Lavigne AC, Mengus G, Gangloff YG, Wurtz JM, David-son I 1999 Human TAF(II)55 interacts with the vitaminD(3) and thyroid hormone receptors and with derivativesof the retinoid X receptor that have altered transactiva-tion properties. Mol Cell Biol 19:54865494

    91. Tolon RM, Castillo AI, Jimenez-Lara AM, Aranda A 2000Association with Ets-1 causes ligand- and AF2-inde-pendent activation of nuclear receptors. Mol Cell Biol20:87938802

    92. Chen JD,Evans RM 1995 A transcriptional co-repressorthat interacts with nuclear hormone receptors. Nature377:454457

    93. Kurokawa R, Soderstrom M, Horlein A, HalachmiS, Brown M, Rosenfeld MG, Glass CK 1995 Polarity-specific activities of retinoic acid receptors determinedby a co-repressor. Nature 377:451454

    94. Horlein AJ, Naar AM, Heinzel T, Torchia J, Gloss B,Kurokawa R, Ryan A, Kamei Y, Soderstrom M, GlassCK, Rosenfeld MG 1995 Ligand-independent repres-sion by the thyroid hormone receptor mediated by anuclear receptor co-repressor. Nature 377:397404

    95. Kinuta K, Tanaka H, Moriwake T, Aya K, Kato S, SeinoY 2000 Vitamin D is an important factor in estrogenbiosynthesis of both female and male gonads. Endocri-nology 141:13171324

    96. Johnson LE, DeLuca HF 2001 Vitamin D receptor nullmutant mice fed high levels of calcium are fertile. J Nutr131:17871791

    97. Zinser G, Packman K, Welsh J 2002 Vitamin D(3) recep-tor ablation alters mammary gland morphogenesis. De-velopment 129:30673076

    98. Elstner E, Linker-Israeli M, Said J, Umiel T, de Vos S,Shintaku IP, Heber D, Binderup L, Uskokovic M, Koef-

    fler HP 1995 20-Epi-vitamin D3 analogues: a novel classof potent inhibitors of proliferation and inducers of dif-ferentiation of human breast cancer cell lines. CancerRes 55:28222830

    99. Campbell MJ, Gombart AF, Kwok SH, Park S, KoefflerHP 2000 The anti-proliferative effects of 1,25(OH)2D3on breast and prostate cancer cells are associated withinduction of BRCA1 gene expression. Oncogene 19:50915097

    100. Abe E, Miyaura C, Sakagami H, Takeda M, Konno K,Yamazaki T, Yoshiki S, Suda T 1981 Differentiation ofmouse myeloid leukemia cells induced by 1,25-dihy-droxyvitamin D3. Proc Natl Acad Sci USA 78:49904994

    101. OKelly J, Hisatake J, Hisatake Y, Bishop J, Norman A,Koeffler HP 2002 Normal myelopoiesis but abnormal Tlymphocyte responses in vitamin D receptor knockoutmice. J Clin Invest 109:10911099

    102. Mathieu C, Van Etten E, Gysemans C, Decallonne B,Kato S, Laureys J, Depovere J, Valckx D, Verstuyf A,Bouillon R 2001 In vitro and in vivo analysis of theimmune system of vitamin D receptor knockout mice.J Bone Miner Res 16:20572065

    103. Kristal-Boneh E, Froom P, Harari G, Ribak J 1997 As-sociation of calcitriol and blood pressure in normoten-sive men. Hypertension 30:12891294

    104. Lind L, Hanni A, Lithell H, Hvarfner A, Sorensen OH,Ljunghall S 1995 Vitamin D is related to blood pressureand other cardiovascular risk factors in middle-agedmen. Am J Hypertens 8:894901

    105. Li YC, Kong J, Wei M, Chen Z-F, Liu SQ, Cao L-P 20021,25-Dihydroxyvitamin D3 is a negative endocrine reg-

    ulator of the renin-angiotensin system. J Clin Invest110:229238

    106. Yagishita N, Yamamoto Y, Yoshizawa T, Sekine K, Ue-matsu Y, Murayama H, Nagai Y, Krezel W, Chambon P,Matsumoto T, Kato S 2001 Aberrant growth plate de-velopment in VDR/RXR double null mutant mice. En-docrinology 142:53325341

    107. Whitfield GK, Jurutka PW, Haussler CA, Haussler MR1999 Steroid hormone receptors: evolution, ligands,and molecular basis of biologic function.J Cell Biochem(Suppl)3233:110122

    108. Takeyama K, Kitanaka S, Sato T, Kobori M, YanagisawaJ, Kato S 1997 25-Hydroxyvitamin D3 1-hydroxylaseand vitamin D synthesis. Science 277:18271830

    109. St-Arnaud R, Messerlian S, Moir JM, Omdahl JL, Glo-rieux FH 1997 The 25-hydroxyvitamin D 1--hydroxy-lase gene maps to the pseudovitamin D-deficiency rick-ets (PDDR) disease locus. J Bone Miner Res 12:15521559

    110. Dardenne O, Prudhomme J, Arabian A, Glorieux FH,St-Arnaud R 2001 Targeted inactivation of the 25-hydroxyvitamin D

    3-1-hydroxylase gene (CYP27B1)

    creates an animal model of pseudovitamin D-deficiencyrickets. Endocrinology 142:31353141

    111. Seo E-G, Einhorn TA, Norman AW 1997 24R,25-Dihy-droxyvitamin D3: an essential vitamin D3 metabolite forboth normal bone integrity and healing of tibial fracturein chicks. Endocrinology 138:38643872

    112. Seo EG, Norman AW 1997 Three-fold induction of renal25-hydroxyvitamin D324-hydroxylase activity and in-creased serum 24,25-dihydroxyvitamin D3 levels arecorrelated with the healing process after chick tibialfracture. J Bone Miner Res 12:598606

    113. Boyan BD, Sylvia VL, Dean DD, Schwartz Z 2001 24,25-(OH)

    2D3

    regulates cartilage and bone via autocrine andendocrine mechanisms. Steroids 66:363374

    114. St-Arnaud R, Arabian A, Travers R, Barletta F, Raval-Pandya M, Chapin K, Depovere J, Mathieu C, Christa-kos S, Demay MB, Glorieux FH 2000 Deficient