You are on page 1of 15

0888-8809/03/$15.

00/0 Molecular Endocrinology 17(5):777–791


Printed in U.S.A. Copyright © 2003 by The Endocrine Society
doi: 10.1210/me.2002-0363

MINIREVIEW

Vitamin D: More Than a “Bone-a-Fide” Hormone


AMELIA 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 progress in unraveling the complexities of the vi-
proper development and maintenance of mineral tamin D endocrine system by focusing on four main
ion homeostasis and skeletal integrity. Beyond areas of research: the resolution of the vitamin D
these classical roles, recent evidence suggests receptor crystal structure, the molecular details of
that the bioactive metabolite of vitamin D, 1,25- 1,25-dihydroxyvitamin D3-mediated transcription,
dihydroxyvitamin D3, functions in diverse physio- murine knockout models of key genes in the en-
logical processes, such as hair follicle cycling, docrine system, and alternative vitamin D recep-
blood pressure regulation, and mammary gland tors and ligands. (Molecular Endocrinology 17:
development. This minireview explores the current 777–791, 2003)

V ITAMIN D WAS discovered nearly a century ago


as the nutrient that prevented rickets, a devastat-
ing skeletal disease characterized by undermineral-
calcium mobilization from the bone by promoting the
differentiation of precursor cells into mature, bone-
resorbing osteoclasts (4).
ized bones (1). Since that time, our concept of vitamin The hormonal or bioactive form of vitamin D is
D and, in particular, its most bioactive derivative, 1,25- 1,25-(OH)2D3. It is generated from sequential hy-
dihydroxyvitamin D3 [1,25-(OH)2D3], has evolved from droxylations of vitamin D3, a secosteroid precursor
that of an essential micronutrient to that of a hormone that is obtained from the diet or produced in the skin
involved in a complex endocrine system that directs upon exposure to UV light (5, 6). The first hydroxy-
mineral homeostasis, protects skeletal integrity, and lation of vitamin D3 occurs at the C-25 position and
modulates cell growth and differentiation in a diverse is catalyzed by vitamin D-25-hydroxylase in the liver
array of tissues. 1,25-(OH)2D3 acts in concert with PTH to produce 25-hydroxyvitamin D3 [25(OH)D3], the
to tightly regulate the concentration of serum calcium major circulating form of vitamin D in mammals.
and phosphate, thereby maintaining proper skeletal 25(OH)D3 is the substrate for a second hydroxylase,
mineralization (Fig. 1). A major function of 1,25- the renal 25(OH)D3-1␣-hydroxylase (1␣OHase),
(OH)2D3 is to promote intestinal absorption of calcium resulting in the production of the most bioactive
and phosphate. However, it also may have direct ef- metabolite, 1,25-(OH)2D3. A classic endocrine feed-
fects on the bone (2), in which continuous remodeling back system operates to tightly control serum levels
must occur to sustain structural integrity. For example, of 1,25-(OH)2D3 (5, 6). For example, renal 1␣OHase
in vitro studies indicate that 1,25-(OH)2D3 stimulates activity is stimulated by low serum calcium and
phosphorus levels and by PTH. The expression of
osteoblasts, the resident bone-forming cells, to termi-
1␣OHase is negatively regulated by high levels of
nally differentiate and to deposit calcified matrix (3).
1,25-(OH)2D3. Inactivation, or catabolism, of vitamin
Conversely, when dietary sources are inadequate to
D metabolites is initiated by the ubiquitous enzyme
maintain normocalcemia, 1,25-(OH)2D3 may stimulate
25-hydroxyvitamin D3-24-hydroxylase (24OHase) to
generate either 24,25(OH)2D3 or 1,24,25(OH)3D3.
Abbreviations: AF-2, Activation function-2; CBP, cAMP
response element binding protein-binding protein; DBD,
The 24-hydroxylated metabolites are further de-
DNA-binding domain; 1,25-(OH)2D3, 1,25-dihydroxyvitamin graded and eventually excreted as either calcitroic
D3; 1␣OHase, 25(OH)D3-1␣-hydroxylase; 25-(OH)D3, 25- acid or 23-carboxyl derivatives. This catabolic pro-
hydroxyvitamin D3; 24-OHase, 25-(OH)D3-24-hydroxylase; cess is also carefully regulated as 1,25-(OH)2D3
DRIP, VDR-interacting protein; HAT, histone acetyltrans-
stimulates 24OHase expression to prevent exces-
ferase; HVDRR, hereditary vitamin D-resistant rickets; LBD,
ligand-binding domain; LCA, lithocholic acid; NCoA, nuclear sive synthesis of the hormone.
receptor coactivator; RAR, retinoic acid receptor; RXR, reti- The biological effects of 1,25-(OH)2D3 are mediated
noid X receptor; SKIP, ski-interacting protein; SRC, steroid through the vitamin D receptor (VDR), a member of the
receptor coactivator; TIF2, transcription intermediary fac-
nuclear receptor superfamily of ligand-activated tran-
tor-2; TRAP, thyroid receptor-activating protein; VDR, vitamin
D receptor; VDRE, vitamin D response element; VDRKO, VDR scription factors (7, 8). Binding of 1,25-(OH)2D3 to VDR
knockout. initiates a cascade of macromolecular interactions ul-

777

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
778 Mol Endocrinol, May 2003, 17(5):777–791 Minireview • Sutton and MacDonald

Fig. 1. Metabolism and Mineral Homeostatic Functions of the Vitamin D Endocrine System
Bioactive 1,25-(OH)2D3 is generated by sequential hydroxylations of its precursor vitamin D3 in the liver and the kidney.
1,25-(OH)2D3 operates in a negative feedback loop by inducing expression of the catabolic enzyme 24-OHase and by inhibiting
expression of the anabolic enzyme 1␣OHase. In response to low serum calcium, PTH is produced and stimulates 1␣OHase
expression in the kidney and promotes calcium mobilization from the bone and reabsorption from the kidney. 1,25-(OH)2D3, in
turn, induces calcium absorption in the intestine and calcium release from the skeleton.

timately leading to transcription of select target genes (DR-3 elements). Thus, 1,25-(OH)2D3 target gene se-
(9). 1,25-(OH)2D3 associates with the VDR and pro- lectivity is conferred, in part, through ligand binding,
motes its heterodimerization with retinoid X receptor VDR-RXR heterodimerization, and high-affinity bind-
(RXR), a common heterodimeric partner for other class ing to DR-3 VDREs. Beyond these initial steps, the
II nuclear receptors (10). The liganded VDR-RXR het- precise molecular mechanisms involved in target gene
erodimer is the functionally active transcription factor activation by VDR are less evident. Recent attention
in 1,25-(OH)2D3-mediated transcription. The het- has turned to so-called coactivator proteins that inter-
erodimer binds with high affinity to vitamin D response act directly with VDR and other nuclear receptors in a
elements (VDREs) in the promoters of target genes. ligand-dependent manner (11). These coactivators par-
VDREs are characterized by two direct hexameric re- ticipate in an intricate multiprotein complex together with
peats with an intervening spacer of three nucleotides the basal transcriptional machinery and histone modi-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
Minireview • Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777–791 779

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 Fig. 2. Domain Structure of VDR and Two-Step Model of
several unique features (Fig. 2A and Refs. 5 and 9). The VDR-Mediated Transcription
hypervariable amino-terminal A/B domain of VDR is A, Functional domains of VDR. A/B, Amino-terminal region;
unusually short and, in contrast to that of most other DBD, DBD showing two zinc finger modules (Zn); LBD, LBD
nuclear receptors, is generally thought to lack potent including the long insertion domain and helix 12 encompass-
transactivation domains. However, as discussed later ing AF-2. B, Temporal association of coactivators during
(see VDR Isoforms), there is increasing evidence that VDR-mediated transcription. The liganded (D) VDR-RXR het-
the VDR A/B domain helps determine the overall trans- erodimer recruits SRCs and CBP/p300, resulting in acetyla-
tion (Ac) of histones. The open chromatin template allows for
activation capacity of the VDR (12). The DNA-binding
binding of the DRIP complex and entry of the core transcrip-
domain (DBD, or region C) of VDR is similar to that of tional machinery.
other nuclear receptors and is characterized by two
zinc-binding modules that direct sequence-specific
binding of receptors to DNA (13). The ligand-binding tallize VDR failed, likely due to the presence of a
domain (LBD, or region E) is a multifunctional globular unique insertion sequence in the LBD (see Fig. 2A) that
domain that mediates selective interactions of the re- is largely unordered, leading to decreased protein sol-
ceptor with its cognate hormone (13), with other nu- ubility. Removal of this insertion domain allowed for
clear receptor partners (14), and with comodulatory or efficient crystallization and structure determination of
adapter proteins (15). The LBD contains the ligand- the VDR LBD complexed to 1,25-(OH)2D3 (23). Al-
dependent activation function-2 (AF-2), which is cru- though the lack of this domain may compromise the
cial to ligand-activated transcription. Mutation of the interpretation of the VDR structure, the mutant VDR
AF-2 renders the nuclear receptor transcriptionally in- displays normal ligand binding and similar transacti-
active despite retaining the ability to bind ligand (14– vation properties in vitro (24). Thus, the absence of the
16). The DBD and LBD are bridged by the hinge region insertion sequence does not alter the conformation
(domain D), which is thought to confer rotational flex- significantly so as to compromise VDR function.
ibility between the DBD and LBD and allow for recep- The structure of the VDR LBD is similar to that of
tor dimerization and interaction with the DNA (17). other nuclear receptors, being most closely related to
Although detailed crystal structures for several nu- that of the retinoic acid receptor (RAR)␥ LBD (23). The
clear receptors have been available for nearly a de- VDR LBD is organized into 13 ␣-helices and 3
cade (18–22), the structure of the VDR LBD was not ␤-sheets, which together form a hydrophobic ligand-
solved until recently (23). Numerous attempts to crys- binding pocket. This pocket is larger than that of RAR␥

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
780 Mol Endocrinol, May 2003, 17(5):777–791 Minireview • Sutton and MacDonald

due to variations in the positions of helices H2 and H3n that interact with nuclear receptors and influence their
and the H6–H7 loop. Helix 12, containing the ligand- transactivation potential (31–33). The emergence of
dependent AF-2, of ligand-bound VDR is positioned coactivators, and their inhibitory counterparts core-
similarly to other nuclear receptors (20), highlighting its pressors, provides new insight into the molecular
central importance in creating a coactivator interaction mechanism of nuclear receptor-mediated transcrip-
surface (see VDR Coactivators). In fact, several resi- tion. Upon association with its cognate hormone, the
dues of H12 directly contact the ligand, indicating that receptor LBD undergoes a subtle conformational
the ligand conformation may modulate H12 conforma- change (34). The critical change occurs in helix 12, the
tion and, therefore, coactivator binding and transcrip- carboxy-terminal ␣-helix containing the ligand-depen-
tional activity. When additional structures of liganded dent AF-2. In response to ligand binding, helix 12 folds
VDR complexed with various coactivators are solved, over top of the globular LBD and caps the ligand-
they will likely provide a molecular framework on which binding cavity (20). This ligand-dependent conforma-
to develop new compounds to modulate the vitamin D tional shift creates a hydrophobic cleft composed of
endocrine system. helices 3, 4, 5, and 12 (35–37). The hydrophobic cleft
In this regard, numerous synthetic analogs have serves as a docking surface for many nuclear receptor
already been developed that mimic the advantageous coactivators by interacting with a complementary hy-
effects of 1,25-(OH)2D3 without the hypercalcemic drophobic domain in the coactivator containing the
side effects (see Novel Vitamin D Ligands). Specula- consensus LXXLL motif, also referred to as the nuclear
tion about the mechanisms behind the selective, pleio- receptor box (38). Although these studies provide an
tropic effects of 1,25-(OH)2D3 analogs centers on the elegant structural model for ligand-activated tran-
concept that these analogs induce distinct conforma- scription by nuclear receptors and LXXLL-containing
tions in VDR compared with that of the natural ligand, coactivators, the precise mechanisms governing nu-
ultimately resulting in analog-selective gene regulation clear receptor-mediated transactivation are less clear.
(25, 26). Protease digestions and coactivator binding The ability of coactivators to interact with components
studies provide experimental support for this model
of the preinitiation complex, with other transcription
(25, 27). However, the VDR ligand-binding cavity is
factors, and with histone-modifying proteins implies
larger than that of many other nuclear receptors, and
that a complex integration of transactivator cues oc-
the ligand occupies less than half of this volume. Con-
curs at the promoter of nuclear receptor target genes.
sequently, the VDR ligand-binding pocket can accom-
The growing number of coactivators identified in the
modate rather significant structural changes in the
last decade adds yet another level of complexity to the
ligand including 1,25-(OH)2D3 analogs with bulky side
paradigm of nuclear receptor-mediated transcription.
chains (23, 28). Indeed, the crystal structures of VDR
Extensive reviews on comodulatory proteins can be
complexed with the MC1288 and KH1060 analogs
found elsewhere (39–41). Here, we will highlight a few
show that these low calcemic analogs do not induce
significant developments in the VDR coactivator field.
different conformations in the VDR compared with the
natural ligand (29). Thus, other mechanisms must be Steroid receptor coactivator (SRC)-1 [or nuclear re-
considered to explain the different potencies and cal- ceptor coactivator (NCoA)1] is the founding member of
cemic profiles of the analogs. One potential answer the LXXLL motif-containing SRC family of coactivators
resides in the observation that the VDR-analog com- (33). This family also includes transcriptional interme-
plexes are more energetically stable than the VDR- diary factor-2 (TIF2; Refs. 42 and 43) and receptor-
1,25-(OH)2D3 complex (29). The increased half-life of associated coactivator-3 (44–48). The SRCs interact
the activated VDR may result in altered transcriptional with VDR and potentiate its transcriptional activity (15,
activity, which may explain the differences both in 49). Each of the SRCs possess an autonomous tran-
potencies and in target gene selectivity between the scriptional activation domain, as evidenced by their
natural and synthetic ligands. Alternatively, the solid- ability to enhance transcription when fused to a het-
state crystal structure may not reveal subtle dynamic erologous DNA-binding sequence such as GAL4.
conformational changes in solubilized VDR evoked by SRCs stimulate transcription possibly by recruiting
various analogs. 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-
VDR COACTIVATORS moter (46, 47, 50). SRCs also possess intrinsic HAT
activity (51). CBP/p300 directly associates with nu-
The existence of limiting accessory factors or adapter clear receptors and, together with SRCs, synergisti-
proteins in steroid hormone receptor action was pro- cally stimulates transcription (52, 53). Thus, SRCs di-
posed in the late 1980s based on the “squelching” rectly alter chromatin structure and recruit other
phenomenon, in which the LBD of one receptor inter- factors that modify histones, potentially providing
feres with ligand-activated transcription mediated by a more accessible promoter templates on which the
second receptor (30). A decade later, these comodu- transcriptional machinery assembles and initiates
latory proteins were identified as specific molecules transcription of target genes.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
Minireview • Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777–791 781

A large multiprotein complex called DRIP (VDR- varied transcription factors including the VDR, sug-
interacting proteins) was identified as a coactivator for gests a potentially important role for this and other
VDR and other nuclear receptors (54, 55). Many com- coactivators in coupling nuclear receptor-mediated
ponents of this complex were discovered separately transcription with mRNA splicing (69).
as thyroid receptor activating protein (TRAP) and the Although a variety of coactivator proteins have been
mammalian Mediator complex (56, 57). The diversity identified for VDR and other nuclear receptors, their
of transactivator interactions with the DRIP/TRAP/ physiological significance and their discrete and/or
Mediator complex clearly suggests a more fundamen- redundant functions in different signal-activated tran-
tal role for this complex in stimulus-activated tran- scriptional systems remain unclear. Mice with individ-
scriptional processes. In VDR-mediated transcription, ual targeted deletions of the three SRCs have been
DRIP205/TRAP220 acts as an anchoring subunit of developed (70–72). These models show that the SRCs
the complex by interacting directly with VDR/RXR het- share several similar functions, especially in the devel-
erodimers through one of two LXXLL motifs (58). Bio- opment and maintenance of the female reproductive
chemical depletion of DRIP in cell-free transcription system. However, the individual SRCs also serve dis-
assays shows that DRIP is essential for VDR-activated tinct physiological roles. For example, ablation of TIF2/
transcription in vitro (54). Because the DRIP complex glucocorticoid receptor-interacting protein 1 results in
does not contain SRCs and is not associated with HAT testicular defects, whereas deletion of either SRC-1 or
activity (58), it is likely that DRIP and SRCs potentiate receptor-associated coactivator 3 does not affect
the transcriptional activation of VDR through distinct male reproduction (71). As the effect of these deletions
mechanisms. Chromatin immunoprecipitations stud- on 1,25-(OH)2D3-mediated transcription has not been
ies indicate that a coactivator exchange occurs in the reported in any of these three knockout models, the in
transcriptional complex on native nuclear receptor- vivo relevance of SRCs in VDR-activated transcription
responsive promoters (59–61). Specifically, SRCs ap- remains to be determined. Deletion of the receptor-
pear to enter the transcriptional complex first and dis- interacting subunit of the DRIP complex, DRIP205/
sociate followed by binding of the DRIP multimeric TRAP220, results in attenuated thyroid hormone-stim-
complex (60, 61). DRIP is also known to recruit the ulated transcription but does not affect retinoic acid
RNA polymerase II holoenzyme to VDR upon ligand responses. Again, 1,25-(OH)2D3 responses have not
binding (62). Although these data conflict, to some been examined in this model, so it is unknown whether
extent, with previous studies that show simultaneous DRIP is required for VDR-activated transcription in
association of SRCs and DRIP with activated nuclear vivo. Silencing of the Caenorhabditis elegans ortholog
receptor complexes (59), they do suggest a temporal of NCoA-62/SKIP by RNA interference results in early
model in which SRCs enter the complex first to re- embryonic lethality due to a potential general tran-
model the chromatin, followed by DRIP complex entry scription defect (73). Although delineation of the phys-
and subsequent recruitment of RNA polymerase II iological function of NCoA-62/SKIP awaits develop-
(Fig. 2B). ment of a mammalian knockout model, these data
In addition to DRIP and SRCs, several other proteins suggest that NCoA-62/SKIP plays a fundamental role
that potentiate VDR-mediated transcription have been in RNA polymerase II-mediated transcription. More
described. One example is NCoA-62/ski-interacting studies are needed to build an integrative vision of
protein (SKIP), which is a coactivator unrelated to how the entire ensemble of coactivator proteins asso-
DRIPs, SRCs, and other LXXLL-containing coactiva- ciates and stimulates the transcriptional activity of
tors (63). It interacts with VDR and other nuclear re- VDR and other nuclear receptors. Initial forays into
ceptors and augments their transcriptional activity. deciphering this complex process have begun with the
Bx42, the Drosophila melanogaster ortholog of NCoA- application of chromatin immunoprecipitation assays
62/SKIP, is also implicated in transcriptional pro- and in vivo imaging of fluorescently tagged nuclear
cesses activated by the insect steroid ecdysone (64). receptors and coactivators to assess the temporal
NCoA-62/SKIP was identified independently through assembly of transcription factors and nuclear recep-
its interaction with ski, placing it as part of the TGF- tors on native promoters (59–61, 74, 75). These strat-
␤-dependent Smad transcriptional complex (65). It is egies, combined with in vitro systems composed of
also implicated in a number of other transcriptional purified components and chromatin-packaged tem-
pathways. NCoA-62/SKIP lacks LXXLL motifs and se- plates, will be required for a more complete under-
lectively associates with the VDR-RXR heterodimer standing of the molecular details of VDR-activated
through the LBD, but through a domain that is distinct transcription.
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- KNOCKOUT MODELS
tial interplay between different coactivator classes for
maximal activity. Recently, NCoA-62/SKIP was iden- Much of our understanding of the physiology of the
tified in subcomplexes of the spliceosome (67, 68). vitamin D endocrine system has stemmed from classic
This, combined with NCoA62/SKIP’s ability to contact dietary manipulations and from the analysis of inher-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
782 Mol Endocrinol, May 2003, 17(5):777–791 Minireview • Sutton and MacDonald

ited disorders in humans (Ref. 76 and reviewed in Ref. designed to explore the role of VDR at stage-selective
77). The recent development of murine genetic models checkpoints in skeletal maturation, at which the pos-
in which key genes in this endocrine system have been sibility of developing compensatory mechanisms has
systematically eliminated highlights the essential role been minimized. Finally, the skeletal phenotypes of
of 1,25-(OH)2D3 in maintaining mineral homeostasis as normocalcemic VDRKO mice may be more pro-
well as reveals more subtle actions of this hormone in nounced at different life stages or under different
other physiological processes. physiological stresses. Thus, it will be important to
determine whether normocalcemic VDRKO mice are
VDR Knockout (VDRKO) Mice more susceptible to age-related or ovariectomy-
induced loss of bone mineral density and whether they
Two groups independently created mouse strains with are compromised in their ability to repair skeletal
targeted deletions in the VDR gene by disrupting either fractures.
exon 2 (78) or exon 3 (79). Not surprisingly, the VDRKO In contrast to the skeletal phenotype, the mineral-
mice displayed all of the features of the human disease rich diet does not correct the alopecia (i.e. the absence
hereditary vitamin D-resistant rickets (HVDRR), a rare of functional hair follicles) observed in VDRKO mice
genetic disorder caused by mutations in the VDR gene (80). Although the VDRKO keratinocytes proliferate
(Ref. 76 and reviewed in Ref. 77). The VDRKO mice are and differentiate normally, they fail to properly initiate
viable and develop normally until the weaning period. hair regrowth after depilation (86, 87). Due to the lack
However, shortly after weaning, VDR-null mice exhibit of feedback regulation of the anabolic 1␣OHase en-
alopecia and growth retardation accompanied by pro- zyme and the catabolic 24OHase, the VDRKO animals
gressive hypocalcemia, hypophosphatemia, and com- have abnormally high levels of 1,25-(OH)2D3. Thus,
pensatory hyperparathyroidism. These metabolic im- one potential cause of alopecia in VDRKO animals is
balances result in severe skeletal defects, including 1,25-(OH)2D3 toxicity. To address this possibility,
decreased bone mineral density, thinned bone cortex, VDRKO mice were raised and bred for five generations
and widened undermineralized growth plates. How- in a UV light-free environment and on a diet lacking
ever, when VDRKO mice are fed a rescue diet rich in vitamin D derivatives (87). Despite having undetect-
calcium and phosphorus to normalize serum calcium able levels of 1,25-(OH)2D3, fifth-generation vitamin
and PTH levels, the mice develop normally without D-deficient VDRKO mice still have alopecia. Thus,
bone abnormalities (80). The skeletons of these mice 1,25-(OH)2D3 toxicity does not cause alopecia in
appear grossly, histologically, and biometrically nor- VDRKO mice. Because wild-type littermates of
mal (81). This indicates that the bone defect in VDR- VDRKO mice raised under the same vitamin D-defi-
null mice is secondary to the malabsorption of calcium cient conditions do not display alopecia, Demay and
in the intestine and is not due to the lack of a direct colleagues (87) proposed that VDR may regulate hair
effect of 1,25-(OH)2D3 on the bone. The impaired in- follicle cycling in a ligand-independent fashion. Further
testinal absorption of calcium in the VDRKO mice is support for this hypothesis comes from the observa-
linked to diminished intestinal expression of several tion that mice with a targeted deletion in 25(OH)D3-
1,25-(OH)2D3-regulated genes putatively involved in 1␣-OHase (see below), the biosynthetic enzyme that
calcium transport, including calbindin D9K, calcium produces 1,25-(OH)2D3, do not display alopecia (88).
transport protein-1, and epithelial calcium channel (82, In vitro studies indicate that VDR associates with var-
83). In addition to the intestinal defect in calcium ab- ious transcription factors and induces select genes in
sorption, mice that express a mutant VDR that lacks the absence of ligand (63, 89–91). Alternatively, unli-
the DBD have decreased renal reabsorption of calcium ganded VDR may repress a subset of target genes in
(84). These studies reinforce the concept that both the a manner analogous to other nuclear receptors
intestine and the kidney are essential VDR target or- through corepressor interactions (92–94). Such VDR-
gans in maintaining calcium homeostasis. RXR-repressed genes could be involved in negatively
The lack of a skeletal phenotype in the VDRKO mice regulating hair follicle cycling. Although these studies
weaned onto the rescue diet is somewhat surprising in introduce a novel and potentially significant concept in
light of the vast literature supporting direct, primary VDR biology, identifying target genes and establishing
roles of VDR in both osteoblast and osteoclast biology molecular mechanisms that govern the function of
(85). On the surface, the VDRKO studies indicate that unliganded VDR in keratinocytes are important goals
1,25-(OH)2D3 is not essential for normal bone devel- for future research.
opment and for maintaining skeletal integrity beyond The global tissue distribution of VDR suggests that
its classic role in calcium and phosphate absorption in 1,25-(OH)2D3 plays important roles in physiological
the intestine. However, a standard caveat with gene processes beyond mineral homeostasis and keratino-
disruption is that the developing animal may acquire cyte function. For example, VDRKO mice have im-
adaptive mechanisms or utilize redundant compensa- paired reproductive function (78). Both male and fe-
tory pathways to bypass the effects of the gene dele- male VDR-null mice exhibit diminished estradiol levels
tion. Conditional knockout approaches that ablate and elevated gonadotropins, indicating a gonadal dys-
VDR in a temporally controlled or tissue-specific man- function in the secretion of sex hormones (95). Histo-
ner may be more informative. Such strategies could be logical analysis of reproductive glands shows abnor-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
Minireview • Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777–791 783

mal ovarian follicle development in the females and study suggests that VDR negatively regulates the ex-
dilated seminiferous tubules with diminished spermat- pression of renin, allowing for decreased angiotensin
ogenesis in the males. However, serum calcium nor- production and lower blood pressure. The relevance of
malization with the rescue diet mostly corrects the this study to human hypertension is not entirely clear
hormonal imbalances and histological abnormalities because there are no reports of hypertensive HVDRR
(95) and completely restores fertility (95, 96), suggest- patients. Regardless, these are provocative observa-
ing that the gonadal dysfunction in VDRKO mice pri- tions in the VDRKO model that may stimulate a more
marily results from hypocalcemia. extensive examination of 1,25-(OH)2D3 and its syn-
Mammary gland development is also emerging as a thetic analogs as potential therapies for some forms of
biological process that is impacted by 1,25-(OH)2D3. hypertension.
The mammary glands of VDRKO mice demonstrate a
hyperproliferative phenotype as evidenced by in- VDR/RXR Double Knockout
creased numbers of terminal end buds and enhanced
ductal branching compared with wild-type littermates VDR heterodimerizes with RXR to modulate transcrip-
(97). VDRKO mammary glands also show accelerated tion of target genes in response to 1,25-(OH)2D3. To
ductal development and increased proliferation in re- examine the effect of abolishing both of the active
sponse to exogenously administered estrogen and partners in 1,25-(OH)2D3 signaling, Yagishita and col-
progesterone (97). Although dietary calcium supple- leagues (106) crossed VDRKO mice with RXR␥-null
mentation normalizes estrogen levels in VDRKO mice, mice to generate VDR/RXR␥-double-knockout mice.
the abnormal mammary phenotype is retained. These The phenotype of the double-knockout mice is nearly
data indicate a significant developmental role for VDR identical with that of the single-VDR-knockout mouse,
in the mammary gland potentially in restricting ductal including growth retardation, hypocalcemia, hy-
growth. Combined with the observations that 1,25- pophosphatemia, hyperparathyroidism, rickets, and
(OH)2D3 and its analogs inhibit the growth and induce alopecia. Upon closer inspection, however, a unique
the differentiation of breast cancer cell lines (98, 99), abnormality was noted in the growth plates of long
the mammary phenotype in the VDRKO model indi- bones of VDR/RXR␥-knockout mice that is not present
cates that 1,25-(OH)2D3 and its derivatives may be in either of the single-knockout mice. Specifically,
useful therapies for breast cancer. VDR/RXR␥-null mice have a defect in the development
An important immunomodulatory function for 1,25- of hypertrophic chondrocytes, the most mature type of
(OH)2D3 and VDR has been indicated by decades of in chondrocyte in the growth plate. Normalization of se-
vitro studies. For example, 1,25-(OH)2D3 potently in- rum calcium and phosphorus rescues all of the skel-
hibits proliferation and drives the differentiation of leu- etal anomalies except for the disordered growth
kemic cells along the monocyte/macrophage lineage plates. Because this chondrocyte defect is not present
(100). However, VDRKO mice lack a striking immune in either of the single-knockout strains, the authors
system phenotype. Although O’Kelly et al. (101) ob- suggested that a functionally redundant VDR-related
served that VDRKO mice have abnormal T cell re- receptor exists that selectively heterodimerizes with
sponses due to diminished cytokine production by RXR␥. Such a receptor would likely share a consider-
macrophages, the calcium-rich rescue diet was not able amount of sequence similarity with the classical
tested in this study. Thus, it is not known whether VDR because it must 1) bind to and be transcriptionally
these abnormalities can be attributed to a lack VDR, to activated by 1,25-(OH)2D3 or its metabolites; 2) het-
1,25-(OH)2D3 toxicity, or to hypocalcemia. Mathieu erodimerize with RXR␥; and 3) recognize and stimulate
et al. (102) found that VDRKO mice had a defect in transcription of the same target genes as classical
calcium-dependent T cell proliferation resulting in pro- VDR in chondrocytes. Although the public genome
tection from experimentally induced autoimmune dia- databases do not indicate that highly related se-
betes and that these abnormalities could be corrected quences exist, potential candidates might include the
by restoring serum calcium to normal levels. The nor- most closely related nuclear receptors, such as farne-
mal development of the immune system in the VDRKO soid X receptor, steroid xenobiotic receptor/pregnane
mice suggests that VDR is not essential for immune X receptor, and liver X receptor (107). Alternatively, any
function or that other compensatory pathways exist. nuclear receptor unrelated to VDR that selectively het-
Several clinical studies have proposed that 1,25- erodimerizes with RXR␥ and binds 1,25-(OH)2D3 or its
(OH)2D3 may also be beneficial to the cardiovascular metabolites might fulfill this role.
system by decreasing blood pressure (103, 104). Con-
sistent with these observations in humans, VDRKO 25(OH)D3-1␣-Hydroxylase Knockout
mice have increased renin expression, resulting in
higher levels of angiotensin II, increased water intake, Arguably, one of the most significant advances in the
electrolyte disturbances, elevated blood pressure, and vitamin D field over the past five years has been the
cardiac hypertrophy (105). Furthermore, high levels of identification and cloning of the renal 1␣OHase,
renin and angiotensin II persist despite normalization the enzyme responsible for the regulated synthesis of
of mineral ion levels with the rescue diet. This re- the active hormone (108, 109). To study the effects
sponse appears to be at the transcriptional level, as of the absence of 1,25-(OH)2D3, two groups indepen-
1,25-(OH)2D3 suppresses renin promoter activity. This dently disrupted the 1␣OHase gene in mice (88, 110).

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
784 Mol Endocrinol, May 2003, 17(5):777–791 Minireview • Sutton and MacDonald

The phenotype of the 1␣OHase-null mice is strikingly metabolism and diseases of the skeleton, such as
similar to that of the VDR-knockout mouse, including rickets, osteoporosis, and renal osteodystrophy, 1,25-
hypocalcemia, hyperparathyroidism, growth retarda- (OH)2D3 has significant therapeutic potential for pa-
tion, and osteomalacia, consistent with rickets (88, thologies such as cancer, autoimmune syndromes,
110). The 1␣OHase-mutant female mice are anovula- and psoriasis. However, 1,25-(OH)2D3 itself has a nar-
tory and, therefore, infertile (88). Another key differ- row therapeutic window limited by the development of
ence between VDRKO mice and the 1␣OHase-ablated toxic hypercalcemia. The increase in calcium is
mice is that the 1␣OHase-knockout animals do not achieved both by enhanced intestinal absorption and
display alopecia (88, 110). The role of hypocalcemia/ by liberation of calcium from the skeleton, eventually
hypophosphatemia in any aspect of the abnormal leading to decreased bone mass at higher doses. This,
phenotype of the 1␣OHase-ablated mice has yet to be of course, counteracts the beneficial effects of 1,25-
addressed using the rescue diet. Importantly, the de- (OH)2D3 for the treatment of bone diseases. Conse-
velopment of knockout models of both the receptor quently, more than 800 synthetic 1,25-(OH)2D3 ana-
and ligand of 1,25-(OH)2D3 will provide powerful tools logs have been developed in attempt to preserve the
to delineate the overlapping and distinct roles of VDR favorable activities of 1,25-(OH)2D3 while avoiding the
and its ligand in diverse processes such as mineral side effects (115).
homeostasis, hair follicle cycling, mammary gland de- Calcipotriol (MC 903) is an analog that has been
velopment, and blood pressure regulation. used to treat psoriasis for nearly 15 yr, and it is cur-
rently considered a first-line therapy for the disease
24OHase Knockout
(116). Calcipotriol improves psoriasis by inhibiting pro-
liferation and promoting differentiation of keratino-
24OHase metabolizes both the bioactive 1,25-(OH)2D3
cytes, but it does not cause hypercalcemia or de-
and its precursor, 25(OH)D3. 24OHase gene transcrip-
creased bone mass. This selectivity can be attributed
tion is positively regulated by 1,25-(OH)2D3, thereby
to calcipotriol’s low affinity for vitamin D binding pro-
completing a negative feedback loop to prevent ex-
tein, the major vitamin D transport protein in the cir-
cessive hormone synthesis. One of the major products
culation (117), and the fact that it is applied topically,
of 24OHase, namely 24,25(OH)2D3, is considered to
thus restricting its actions to the skin. 1,25-(OH)2D3
be an inactive metabolite, an initial product destined
analogs are also valuable in treating renal osteodys-
for further degradation and eventual excretion as cal-
trophy, a devastating consequence of chronic renal
citroic acid (5). However, there is substantial evidence
failure. Kidney disease, due to a variety of causes,
that 24,25(OH)2D3 has biological activity of its own,
often leads to reduced 1,25-(OH)2D3 production and
most strikingly in the function of chondrocytes, or
impaired phosphate excretion resulting in abnormally
cartilage-forming cells (111–113). To determine the
high PTH levels. The decreased calcitriol levels com-
consequences of abolishing this enzyme and, there-
fore, its 24-hydroxylated products, a knockout model bined with secondary hyperparathyroidism, in turn,
of 24OHase was created (114). This mutation results in result in increased bone turnover (118). Treatment with
reduced embryonic viability and aberrant intramem- 1,25-(OH)2D3 is effective in suppressing PTH levels
branous ossification. The obvious possibility is that the and improving the initial skeletal abnormalities. How-
diminished levels of 24,25(OH)2D3 causes these ab- ever, due to the narrow therapeutic window, 1,25-
normalities. However, the phenotype is not rescued in (OH)2D3 often causes hypercalcemia and additional
24OHase-knockout progeny by feeding pregnant mice bone disease from inappropriately low bone turnover.
exogenous 24,25(OH)2D3. Alternatively, the phenotype Two 1,25-(OH)2D3 analogs are currently approved for
could be caused by the abnormally high levels of treatment of secondary hyperparathyroidism in the
1,25-(OH)2D3 resulting from the deletion of this cata- United States. 19-Nor-D2 (paricalcitol; Ref. 119) and
bolic enzyme. Indeed, crossing the 24OHase-null mice 1␣(OH)D2 (doxercalciferol; Ref. 120) are as effective as
with VDR-null mice completely rescues the decreased 1,25-(OH)2D3 in reducing PTH levels but do not result
embryonic viability and ossification defects, thus sup- in significant hypercalcemia. Although the mecha-
porting the concept that 1,25-(OH)2D3 toxicity leads to nisms of the selectivity of these two analogs remain
the abnormal phenotype in the 24OHase-knockout unclear, they represent significant improvements in
mice. Moreover, this model also suggests that 24- treatment regimens for renal osteodystrophy.
hydroxylated metabolites are not required for normal Although neither vitamin D3 nor 1,25-(OH)2D3 ana-
intramembranous ossification (114). logs are currently Food and Drug Administration
(FDA)-approved for treating osteoporosis in the United
States, these compounds are widely used to prevent
BEYOND 1,25-(OH)2D3 AND VDR: NOVEL and treat osteoporosis throughout the world (121,
LIGANDS AND ALTERNATIVE RECEPTORS 122). Furthermore, vitamin D3 is currently recom-
mended as a dietary supplement in addition to any
Novel Vitamin D Ligands pharmacological treatment for all patients with de-
creased bone mass or osteoporosis (123). Still, the
The therapeutic potential of 1,25-(OH)2D3 continues to higher doses of 1,25-(OH)2D3 required for maximal
expand. In addition to treating disorders of mineral improvement in bone density cause significant hyper-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
Minireview • Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777–791 785

calcemia. Peleg and colleagues (124) tested 1,25- variations in the VDR gene, transcript, and protein
(OH)2D3 analogs for their ability to improve bone min- sequences been discovered. At least 14 distinct tran-
eral density in the ovariectomized rat model of scripts of human VDR have been identified that differ
osteoporosis. One novel analog, Ro-26-9228, protects in their 5⬘ ends (135). These transcripts arise from
against osteopenia, but it does not increase serum alternative mRNA splicing and differential promoter
calcium except at very high doses. These observa- usage. Most of these variant transcripts utilize the
tions are potentially explained by the tissue-selective same initiator codon, producing a VDR that is 427
action of Ro-26-9228, which stimulates osteocalcin amino acids in length. However, two transcripts have
and osteopontin expression in osteoblasts but does upstream in-frame methionines that potentially gener-
not affect calbindin D9K or plasma membrane calcium ate N-terminal extensions in VDR of 50 or 23 amino
pump expression in the intestine (124). Shevde et al. acids (135). Low levels of endogenous VDRB1 protein,
(125) found that another analog, 2-methylene-19-nor- the 50-amino-acid-extended variant, have been de-
(20S)-1␣,25(OH)2D3 (2MD), potently stimulates bone tected in osteoblast, colon cancer, and kidney cell
formation in vitro and markedly improves bone mass in lines (136). Interestingly, VDRB1 has reduced tran-
ovariectomized rats without dramatically increasing scriptional activity compared with classical VDR.
serum calcium. Such studies support the concept that Whether the levels of expression of these isoforms are
more selective 1,25-(OH)2D3 analogs will be useful substantial and whether these isoforms result in al-
therapies for osteoporosis by enhancing bone mineral tered biological activity in vivo remains unresolved.
density without causing toxic hypercalcemia. Multiple polymorphic variations also exist in VDR in
In addition to these and numerous other designer the human population (137). The vast majority of these
analogs, a recent study suggests the possibility that polymorphisms do not result in a structural alteration
natural compounds other than 1,25-(OH)2D3 may in the VDR protein, with the exception of the Fok I
serve as tissue-selective activators of VDR-mediated variant (138). The Fok I polymorphism is located at the
responses. An unexpected ligand for VDR was discov- original initiator ATG, which is part of a Fok I endonu-
ered from studies with bile acid compounds (126). clease site. In some humans, there is an ATG 224 ACG
Metabolic lipophilic molecules such as bile acids ac- transition at the ⫹1 position, eliminating the transla-
tivate many of the orphan nuclear receptors, including tional initiation site and Fok I recognition sequence.
farnesoid X receptor and steroid xenobiotic receptor/ This transition results in the use of an in-frame methi-
pregnane X receptor (127–129). Recently, Makishima onine as the initiator codon at the ⫹4 position. Thus,
et al. (126) screened classical nuclear receptors to either a 427 (Met-1) or a 424 (Met-4) amino acid pro-
identify those that were activated by the bile acid tein is expressed. Numerous epidemiological studies
lithocholic acid (LCA). Surprisingly, they found that suggest an association between the shorter form of
LCA and its metabolites directly bind and activate VDR and increased bone mineral density in humans
VDR. However, this activation requires micromolar (139–143). The molecular mechanism of this associa-
concentrations of LCA, whereas VDR is activated by tion remains unclear, but there is suggestive evidence
nanomolar amounts of 1,25-(OH)2D3. Nonetheless, that the Met-4 VDR displays enhanced transcriptional
LCA- or 1,25-(OH)2D3-liganded VDR also stimulates activity due to increased interaction with basal tran-
the expression of endogenous CYP3A, the P450 en- scription factor II B (12). Although these findings re-
zyme responsible for degradation of LCA in the liver main controversial (144), they raise the possibility that
and the intestine. LCA is implicated as a toxin that the N terminus possesses some type of structure that
promotes colorectal carcinogenesis (130, 131), influences transcriptional activity. This, combined with
whereas 1,25-(OH)2D3 is protective against colon can- the observations that other N-terminal extensions of
cer (132). Thus, induction of CYP3A by 1,25-(OH)2D3 VDR may have reduced transcriptional activity, sug-
and by LCA itself may represent a detoxification path- gests that there may be inhibitory domains at the
way for LCA, as well as explain the potential preven- extreme N terminus of VDR that decrease its transac-
tative effects of 1,25-(OH)2D3 in colon cancer. Al- tivation potential.
though this study awaits further in vivo confirmation, it
clearly raises the possibility that VDR may be activated A Membrane Receptor? Rapid, Nongenomic
by other naturally occurring ligands. Effects of Vitamin D and Its Metabolites

VDR Isoforms According to the classical paradigm of nuclear recep-


tor action, ligand-activated nuclear receptors recruit
Many nuclear receptors, such as RAR, RXR, and thy- the basal transcriptional machinery and other activator
roid receptor, have multiple isoforms that are encoded complexes to the promoters of target genes to induce
by separate genes (133). Unlike these nuclear recep- transcription. Because these responses require tran-
tors, only one human VDR genetic locus has been scription and translation of target genes, they are typ-
identified (134), and the genomic database does not ically delayed by at least 30 min. However, more rapid
indicate additional highly related sequences. Although (within seconds to minutes) effects in response to
the cDNA encoding the human VDR was cloned nearly steroid hormones are also apparent. The rapid nature
15 yr ago (8), only recently have several significant of these effects and their relative insensitivity to tran-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
786 Mol Endocrinol, May 2003, 17(5):777–791 Minireview • Sutton and MacDonald

scriptional and translational inhibitors, such as actino- important tools to decipher the molecular require-
mycin D and cycloheximide, precludes the possibility ments of classical VDR that mediate the genomic
that the traditional “genomic” model is operating. Re- and nongenomic effects of 1,25-(OH)2D3 in vivo.
cent attention to these rapid, “nongenomic” hormone
effects has spawned renewed interest in this long-
standing area of membrane-initiated signaling in the
steroid hormone field (145). Over two decades ago, SUMMARY: FRONTIERS IN VITAMIN D
1,25-(OH)2D3 was shown to evoke transcellular move-
ment of calcium across chick enterocytes within sev- The molecular, cellular, structural, and genetic studies
eral minutes (146, 147). This phenomenon is theorized of the past decade have impacted our understanding
to be adaptively beneficial for a hypocalcemic animal of the vitamin D endocrine system in a number of
in that rapid absorption of calcium occurs without a significant ways. First and foremost, the genetic mod-
delayed response involving transcription and transla- els solidify the fundamental roles that both 1,25-
tion of calcium-binding proteins or calcium transport- (OH)2D3 and VDR play in ensuring that an organism
ers (147). In addition to the enterocyte, the osteoblast obtains sufficient calcium and phosphate from the
is a target for 1,25-(OH)2D3-induced rapid calcium environment to sustain life and normal development.
mobilization from internal stores, a process that in- Lack of either functional VDR or active 1,25-(OH)2D3
volves a membrane-initiated signaling cascade includ- leads to profound, life-threatening hypocalcemia and
ing phospholipase C activation and inositol triphos- undermineralized skeletal tissue. Previous classical
phate formation (148). This process also occurs in nutritional manipulations and identification of mutated
skeletal muscle cells, in which 1,25-(OH)2D3 induces VDR as the causative defect in HVDRR have estab-
calcium release from the sarcoplasmic reticulum (149), lished this central function of 1,25-(OH)2D3 long before
potentially through MAPK activation (150). These are the creation of genetic mouse models. However, the
just a few of the many examples of in vitro systems in development and analysis of such models are central
which these nongenomic actions of 1,25-(OH)2D3 in formulating a detailed physiological picture of 1,25-
have been studied. (OH)2D3 signaling and in reinforcing a direct link be-
Although the rapid effects of 1,25-(OH)2D3 and tween the various genes within the endocrine system
numerous other steroids are well documented, the and mineral ion dysregulation.
field is hindered by the inability to identify the puta- On the other hand, the knockout studies provide
tive membrane receptors that trigger these non- somewhat of a surprise and, to a certain extent, a bit
genomic effects. Although suggestive biochemical of a disappointment for those interested in 1,25-
and immunological data (151) indicate that the (OH)2D3 and bone biology. In particular, specific cells
membrane VDR is a distinct gene product, a true of the osteoblast and osteoclast lineage have gar-
protein representing this receptor remains elusive. A nered considerable research attention over the past
promising new approach in the nongenomic field is several decades as important direct targets of 1,25-
the use of 1,25-(OH)2D3 analogs to discriminate be- (OH)2D3 in preserving skeletal integrity. However, the
tween receptors that mediate membrane-initiated striking skeletal defects observed in the VDRKO are
events and those that mediate the classical nuclear corrected by simply providing the animals with sup-
effects of 1,25-(OH)2D3. Song et al. (152) showed plemental dietary calcium. Does this mean that 1,25-
that 6-s-cis-locked analogs of 1,25-(OH)2D3 stimu- (OH)2D3 does not act as a direct “bone-a-fide” hor-
late rapid phosphorylation of MAPK in leukemic mone in the skeleton? Clearly, more cellular and
cells, yet these analogs bind poorly to VDR and are genetic approaches are needed to fully answer this
weak activators of VDR-mediated transcription. question and to test whether VDR and 1,25-(OH)2D3
These studies also support the possibility of two play more limited or specialized roles in the developing
separate gene products involved in either rapid, or aging skeleton. Although the jury is still out on the
nongenomic signaling or in classical transcriptional bone, striking new biologies are emerging from the
activation by 1,25-(OH)2D3. In fact, two preliminary VDRKO studies, indicating potential functions for
reports indicate that annexin II, a membrane-asso- 1,25-(OH)2D3 in diverse processes such as hair follicle
ciated calcium-binding protein, may bind 1,25- cycling, blood pressure regulation, and mammary
(OH)2D3 and function as its membrane receptor gland development that are independent of mineral ion
(153, 154). In contrast to these studies, analysis of homeostasis. Moreover, studies on alopecia in
mice carrying a mutated VDR lacking a DBD implies VDRKO mice raise the exciting possibility that VDR
that the classical nuclear VDR mediates both acts in a ligand-independent fashion and may stimu-
genomic and nongenomic responses (84). Osteo- late further exploration into the molecular and cellular
blast cultures derived from VDR-mutant mice are functions of unliganded VDR.
unable to initiate a rapid calcium flux in response to Beyond the physiological information gleaned from
1,25-(OH)2D3, suggesting that some nongenomic the genetic mouse models, recent progress in other
responses require a functional nuclear VDR. Al- areas of the vitamin D field is revealing additional
though a full array of other nongenomic responses molecular details of the endocrine system. Description
needs to be tested, the VDRKO strains will provide of the crystal structure of VDR has supplied an atomic

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
Minireview • Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777–791 787

view of the VDR protein, highlighting the expansive entiation and bone resorption by 1,25-dihydroxyvitamin
binding pocket associated with both natural and syn- D3. Proc Natl Acad Sci USA 80:5907–5911
5. Jones G, Strugnell SA, DeLuca HF 1998 Current under-
thetic ligands. Further refinement of this structure
standing of the molecular actions of vitamin D. Physiol
complexed with coactivators undoubtedly will allow Rev 78:1193–1231
for the future rational design of selective VDR-targeted 6. Horst RL, Reinhardt TA 1997 Vitamin D metabolism. In:
drugs. Some of the additional molecular requirements Feldman D, Glorieux, FH, Pike JW, eds. Vitamin D. San
for the transcriptional activity of 1,25-(OH)2D3-ligan- Diego: Academic Press; 13–32
ded VDR have been elucidated and are beginning to 7. McDonnell DP, Mangelsdorf DJ, Pike JW, Haussler MR,
O’Malley BW 1987 Molecular cloning of complementary
be assembled into an integrated model of coactivator DNA encoding the avian receptor for vitamin D. Science
cooperativity. Furthermore, it is becoming increasingly 235:1214–1217
clear that some actions of 1,25-(OH)2D3 cannot be 8. Baker AR, McDonnell DP, Hughes M, Crisp TM, Man-
explained by the traditional model of 1,25-(OH)2D3- gelsdorf DJ, Haussler MR, Pike JW, Shine J, O’Malley
bound VDR acting solely as a transcription factor in the BW 1988 Cloning and expression of full-length cDNA
encoding human vitamin D receptor. Proc Natl Acad Sci
nucleus. Novel VDR ligands, including synthetic ana-
USA 85:3294–3298
logs and natural compounds such as bile acids, are 9. Haussler MR, Whitfield GK, Haussler CA, Hsieh JC,
broadening our understanding of the therapeutic po- Thompson PD, Selznick SH, Dominguez CE, Jurutka
tential and physiological intricacies of vitamin D. Like- PW 1998 The nuclear vitamin D receptor: biological and
wise, alternative receptors, both related to and distinct molecular regulatory properties revealed. J Bone Miner
from the classical nuclear VDR, are emerging as sig- Res 13:325–349
10. Mangelsdorf DJ, Evans RM 1995 The RXR heterodimers
nificant participants mediating the biological effects of and orphan receptors. Cell 83:841–850
vitamin D compounds. The four areas of progress 11. McKenna NJ, Lanz RB, O’Malley BW 1999 Nuclear
covered in this minireview have filled in many gaps in receptor coregulators: cellular and molecular biology.
our knowledge of vitamin D, but they also have raised Endocr Rev 20:321–344
a multitude of new questions that await answering with 12. Jurutka PW, Remus LS, Whitfield GK, Thompson PD,
Hsieh J-C, Zitzer H, Tavakkoli P, Galligan MA, Dang
the new molecular, pharmacological, and genetic tools HTL, Haussler CA, Haussler MR 2000 The polymorphic
developed in recent years. N terminus in human vitamin D receptor isoforms influ-
ences transcriptional activity by modulating interaction
Acknowledgments with transcription factor IIB. Mol Endocrinol 14:401–420
13. McDonnell DP, Scott RA, Kerner SA, O’Malley BW, Pike
JW 1989 Functional domains of the human vitamin D3
We apologize to many colleagues whose excellent primary
receptor regulate osteocalcin gene expression. Mol En-
publications may not have been cited in this minireview due
to space limitations. docrinol 3:635–644
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 to
Received November 1, 2002. Accepted March 5, 2003. form a complex with a receptor auxiliary factor required
Address all correspondence and requests for reprints to: for high affinity binding to the vitamin D-responsive
Paul N. MacDonald, Ph.D., Department of Pharmacology, element. Mol Endocrinol 8:159–172
Case Western Reserve University, 10900 Euclid Avenue, 15. Masuyama H, Brownfield CM, St-Arnaud R, MacDonald
Cleveland, Ohio 44106. E-mail: pnm2@po.cwru.edu. PN 1997 Evidence for ligand-dependent intramolecular
This work was supported by NIH Grants R01-DK-50348 folding of the AF-2 domain in vitamin D receptor-acti-
and R01-DK-53980 (to P.N.M.), by an award from the Medical vated transcription and coactivator interaction. Mol En-
Scientist Training Program NIH Grant T32-GM-007250 (to docrinol 11:1507–1517
A.L.M.S.), and by a Pharmaceutical Manufacturers’ Associa- 16. Danielian PS, White R, Lees JA, Parker MG 1992 Iden-
tion Foundation Pre-Doctoral Fellowship (to A.L.M.S.). tification of a conserved region required for hormone
dependent transcriptional activation by steroid hor-
mone receptors. EMBO J 11:1025–1033
17. Mangelsdorf DJ, Thummel C, Beato M, Herrlich P,
REFERENCES Schutz G, Umesono K, Blumberg B, Kastner P, Mark M,
Chambon P, Evans RM 1995 The nuclear receptor
superfamily: the second decade. Cell 83:835–839
1. Brown AJ, Dusso A, Slatopolsky E 1999 Vitamin D. Am J
Physiol 277:F157–F175 18. Schwabe JW, Chapman L, Finch JT, Rhodes D 1993
2. van Leeuwen JP, van Driel M, van den Bemd GJ, Pols The crystal structure of the estrogen receptor DNA-
HA 2001 Vitamin D control of osteoblast function and binding domain bound to DNA: how receptors discrim-
bone extracellular matrix mineralization. Crit Rev Eu- inate between their response elements. Cell 75:567–578
karyot Gene Expr 11:199–226 19. Wagner RL, Apriletti JW, McGrath ME, West BL, Baxter
3. Owen TA, Aronow MS, Barone LM, Bettencourt B, Stein JD, Fletterick RJ 1995 A structural role for hormone in
GS, Lian JB 1991 Pleiotropic effects of vitamin D on the thyroid hormone receptor. Nature 378:690–697
osteoblast gene expression are related to the prolifera- 20. Renaud JP, Rochel N, Ruff M, Vivat V, Chambon P,
tive and differentiated state of the bone cell phenotype: Gronemeyer H, Moras D 1995 Crystal structure of the
dependency upon basal levels of gene expression, du- RAR-␥ ligand-binding domain bound to all-trans reti-
ration of exposure, and bone matrix competency in noic acid. Nature 378:681–689
normal rat osteoblast cultures. Endocrinology 128: 21. Bourguet W, Ruff M, Chambon P, Gronemeyer H, Mo-
1496–1504 ras D 1995 Crystal structure of the ligand-binding do-
4. Bar-Shavit Z, Teitelbaum SL, Reitsma P, Hall A, Pegg main of the human nuclear receptor RXR-␣. Nature
LE, Trial J, Kahn AJ 1983 Induction of monocytic differ- 375:377–382

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
788 Mol Endocrinol, May 2003, 17(5):777–791 Minireview • Sutton and MacDonald

22. Weatherman RV, Fletterick RJ, Scanlan TS 1999 Nuclear- 39. McKenna NJ, O’Malley BW 2002 Combinatorial control
receptor ligands and ligand-binding domains. Annu Rev of gene expression by nuclear receptors and coregula-
Biochem 68:559–581 tors. Cell 108:465–474
23. Rochel N, Wurtz JM, Mitschler A, Klaholz B, Moras D 40. McKenna NJ, O’Malley BW 2002 Minireview: nuclear
2000 The crystal structure of the nuclear receptor for receptor coactivators—an update. Endocrinology 143:
vitamin D bound to its natural ligand. Mol Cell 2461–2465
5:173–179 41. Rosenfeld MG, Glass CK 2001 Coregulator codes of
24. Rochel N, Tocchini-Valentini G, Egea PF, Juntunen K, transcriptional regulation by nuclear receptors. J Biol
Garnier JM, Vihko P, Moras D 2001 Functional and Chem 276:36865–36868
structural characterization of the insertion region in the 42. Hong H, Kohli K, Trivedi A, Johnson DL, Stallcup MR
ligand binding domain of the vitamin D nuclear receptor. 1996 GRIP1, a novel mouse protein that serves as a
Eur J Biochem 268:971–979 transcriptional coactivator in yeast for the hormone
25. Peleg S, Sastry M, Collins ED, Bishop JE, Norman AW binding domains of steroid receptors. Proc Natl Acad
1995 Distinct conformational changes induced by 20- Sci USA 93:4948–4952
epi analogues of 1␣,25-dihydroxyvitamin D3 are asso- 43. Voegel JJ, Heine MJ, Zechel C, Chambon P, Grone-
ciated with enhanced activation of the vitamin D recep- meyer H 1996 TIF2, a 160 kDa transcriptional mediator
tor. J Biol Chem 270:10551–10558 for the ligand-dependent activation function AF-2 of
26. Carlberg C, Quack M, Herdick M, Bury Y, Polly P, Toell nuclear receptors. EMBO J 15:3667–3675
A 2001 Central role of VDR conformations for under- 44. Li H, Gomes PJ, Chen JD 1997 RAC3, a steroid/nuclear
standing selective actions of vitamin D(3) analogues. receptor-associated coactivator that is related to
Steroids 66:213–221 SRC-1 and TIF2. Proc Natl Acad Sci USA 94:8479–8484
27. Yang W, Freedman LP 1999 20-Epi analogues of 1,25- 45. Anzick SL, Kononen J, Walker RL, Azorsa DO, Tanner
dihydroxyvitamin D3 are highly potent inducers of DRIP MM, Guan XY, Sauter G, Kallioniemi OP, Trent JM,
coactivator complex binding to the vitamin D3 receptor. Meltzer PS 1997 AIB1, a steroid receptor coactivator
J Biol Chem 274:16838–16845 amplified in breast and ovarian cancer. Science 277:
28. Norman AW, Manchand PS, Uskokovic MR, Okamura 965–968
WH, Takeuchi JA, Bishop JE, Hisatake JI, Koeffler HP, 46. Chen H, Lin RJ, Schiltz RL, Chakravarti D, Nash A, Nagy
Peleg S 2000 Characterization of a novel analogue of L, Privalsky ML, Nakatani Y, Evans RM 1997 Nuclear
1␣,25(OH)2-vitamin D3 with two side chains: interaction receptor coactivator ACTR is a novel histone acetyl-
with its nuclear receptor and cellular actions. J Med transferase and forms a multimeric activation complex
Chem 43:2719–2730 with P/CAF and CBP/p300. Cell 90:569–580
29. Tocchini-Valentini G, Rochel N, Wurtz JM, Mitschler A, 47. Torchia J, Rose DW, Inostroza J, Kamei Y, Westin S,
Moras D 2001 Crystal structures of the vitamin D re- Glass CK, Rosenfeld MG 1997 The transcriptional co-
ceptor complexed to superagonist 20-epi ligands. Proc activator p/CIP binds CBP and mediates nuclear-recep-
Natl Acad Sci USA 98:5491–5496 tor function. Nature 387:677–684
30. Tasset D, Tora L, Fromental C, Scheer E, Chambon P 48. Takeshita A, Cardona GR, Koibuchi N, Suen CS, Chin
1990 Distinct classes of transcriptional activating do- WW 1997 TRAM-1, a novel 160-kDa thyroid hormone
mains function by different mechanisms. Cell 62: receptor activator molecule, exhibits distinct properties
1177–1187 from steroid receptor coactivator-1. J Biol Chem 272:
31. Cavailles V, Dauvois S, Danielian PS, Parker MG 1994 27629–27634
Interaction of proteins with transcriptionally active es- 49. Kraichely DM, Collins 3rd JJ, DeLisle RK, MacDonald
trogen receptors. Proc Natl Acad Sci USA 91: PN 1999 The autonomous transactivation domain in
10009–10013 helix H3 of the vitamin D receptor is required for trans-
32. Halachmi S, Marden E, Martin G, MacKay H, Abbon- activation and coactivator interaction. J Biol Chem 274:
danza C, Brown M 1994 Estrogen receptor-associated 14352–14358
proteins: possible mediators of hormone-induced tran- 50. Ikeda M, Kawaguchi A, Takeshita A, Chin WW, Endo T,
scription. Science 264:1455–1458 Onaya T 1999 CBP-dependent and independent en-
33. Onate S, Tsai, SY, Tsai, M-J, O’Malley, BW 1995 Se- hancing activity of steroid receptor coactivator-1 in thy-
quence and characterization of a coactivator for the roid hormone receptor-mediated transactivation. Mol
steroid hormone receptor superfamily. Science 270: Cell Endocrinol 147:103–112
1354–1357 51. Spencer TE, Jenster G, Burcin MM, Allis CD, Zhou J,
34. Leid M 1994 Ligand-induced alteration of the protease Mizzen CA, McKenna NJ, Onate SA, Tsai SY, Tsai MJ,
sensitivity of retinoid X receptor ␣. J Biol Chem 269: O’Malley BW 1997 Steroid receptor coactivator-1 is a
14175–14181 histone acetyltransferase. Nature 389:194–198
35. Feng W, Ribeiro RCnJ, Wagner RL, Nguyen H, Apriletti 52. Chakravarti D, LaMorte VJ, Nelson MC, Nakajima T,
JW, Fletterick RJ, Baxter JD, Kushner PJ, West BL 1998 Schulman IG, Juguilon H, Montminy M, Evans RM 1996
Hormone-dependent coactivator binding to a hydro- Role of CBP/P300 in nuclear receptor signalling. Nature
phobic cleft on nuclear receptors. Science 280: 383:99–103
1747–1749 53. Kamei Y, Xu L, Heinzel T, Torchia J, Kurokawa R, Gloss
36. Nolte RT, Wisely GB, Westin S, Cobb JE, Lambert MH, B, Lin SC, Heyman RA, Rose DW, Glass CK, Rosenfeld
Kurokawa R, Rosenfeld MG, Willson TM, Glass CK, MG 1996 A CBP integrator complex mediates transcrip-
Milburn MV 1998 Ligand binding and co-activator tional activation and AP-1 inhibition by nuclear recep-
assembly of the peroxisome proliferator-activated tors. Cell 85:403–414
receptor-␥. Nature 395:137–143 54. Rachez C, Suldan Z, Ward J, Chang CP, Burakov D,
37. Shiau AK, Barstad D, Loria PM, Cheng L, Kushner PJ, Erdjument-Bromage H, Tempst P, Freedman LP 1998 A
Agard DA, Greene GL 1998 The structural basis of novel protein complex that interacts with the vitamin D3
estrogen receptor/coactivator recognition and the an- receptor in a ligand-dependent manner and enhances
tagonism of this interaction by tamoxifen. Cell 95: VDR transactivation in a cell-free system. Genes Dev
927–937 12:1787–1800
38. Heery DM, Kalkhoven E, Hoare S, Parker MG 1997 A 55. Rachez C, Lemon BD, Suldan Z, Bromleigh V, Gamble
signature motif in transcriptional co-activators mediates M, Naar AM, Erdjument-Bromage H, Tempst P, Freed-
binding to nuclear receptors. Nature 387:733–736 man LP 1999 Ligand-dependent transcription activation

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
Minireview • Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777–791 789

by nuclear receptors requires the DRIP complex. Nature 73. Kostrouchova M, Housa D, Kostrouch Z, Saudek V, Rall
398:824–828 JE 2002 SKIP is an indispensable factor for Caenorhab-
56. Fondell JD, Ge H, Roeder RG 1996 Ligand induction of ditis elegans development. Proc Natl Acad Sci USA
a transcriptionally active thyroid hormone receptor co- 99:9254–9259
activator complex. Proc Natl Acad Sci USA 93: 74. Stenoien DL, Nye AC, Mancini MG, Patel K, Dutertre M,
8329–8333 O’Malley BW, Smith CL, Belmont AS, Mancini MA 2001
57. Jiang YW, Veschambre P, Erdjument-Bromage H, Ligand-mediated assembly and real-time cellular dy-
Tempst P, Conaway JW, Conaway RC, Kornberg RD namics of estrogen receptor ␣-coactivator complexes
1998 Mammalian mediator of transcriptional regulation in living cells. Mol Cell Biol 21:4404–4412
and its possible role as an end-point of signal transduc- 75. Becker M, Baumann C, John S, Walker DA, Vigneron M,
tion pathways. Proc Natl Acad Sci USA 95:8538–8543 McNally JG, Hager GL 194 2002 Dynamic behavior of
58. Rachez C, Gamble M, Chang CP, Atkins GB, Lazar MA, transcription factors on a natural promoter in living cells.
Freedman LP 2000 The DRIP complex and SRC-1/p160 EMBO Rep 3:1188–1181
coactivators share similar nuclear receptor binding de- 76. Hughes MR, Malloy PJ, Kieback DG, Kesterson RA,
terminants but constitute functionally distinct com- Pike JW, Feldman D, O’Malley BW 1988 Point muta-
plexes. Mol Cell Biol 20:2718–2726 tions in the human vitamin D receptor gene associated
59. Shang Y, Hu X, DiRenzo J, Lazar MA, Brown M 2000 with hypocalcemic rickets. Science 242:1702–1705
Cofactor dynamics and sufficiency in estrogen recep- 77. Malloy PJ, Pike JW, Feldman D 1999 The vitamin D
tor-regulated transcription. Cell 103:843–852 receptor and the syndrome of hereditary 1,25-dihy-
60. Burakov D, Crofts LA, Chang CP, Freedman LP 2002 droxyvitamin D-resistant rickets. Endocr Rev 20:
Reciprocal recruitment of DRIP/mediator and p160 co- 156–188
activator complexes in vivo by estrogen receptor. J Biol 78. Yoshizawa T, Handa Y, Uematsu Y, Takeda S, Sekine K,
Chem 277:14359–14362 Yoshihara Y, Kawakami T, Arioka K, Sato H, Uchiyama
61. Sharma D, Fondell JD 2002 Ordered recruitment of Y, Masushige S, Fukamizu A, Matsumoto T, Kato S
histone acetyltransferases and the TRAP/Mediator 1997 Mice lacking the vitamin D receptor exhibit im-
complex to thyroid hormone-responsive promoters in paired bone formation, uterine hypoplasia and growth
vivo. Proc Natl Acad Sci USA 99:7934–7939 retardation after weaning. Nat Genet 16:391–396
62. Chiba N, Suldan Z, Freedman LP, Parvin JD 2000 Bind- 79. Li YC, Pirro AE, Amling M, Delling G, Baron R, Bronson
ing of liganded vitamin D receptor to the vitamin D R, Demay MB 1997 Targeted ablation of the vitamin D
receptor interacting protein coactivator complex in- receptor: an animal model of vitamin D-dependent rick-
duces interaction with RNA polymerase II holoenzyme. ets type II with alopecia. Proc Natl Acad Sci USA 94:
J Biol Chem 275:10719–10722 9831–9835
63. Baudino TA, Kraichely DM, Jefcoat Jr SC, Winchester 80. Li YC, Amling M, Pirro AE, Priemel M, Meuse J, Baron R,
SK, Partridge NC, MacDonald PN 1998 Isolation and Delling G, Demay MB 1998 Normalization of mineral ion
characterization of a novel coactivator protein, NCoA- homeostasis by dietary means prevents hyperparathy-
62, involved in vitamin D-mediated transcription. J Biol roidism, rickets, and osteomalacia, but not alopecia in
Chem 273:16434–16441 vitamin D receptor-ablated mice. Endocrinology 139:
64. Wieland C, Mann S, von Besser H, Saumweber H 1992 4391–4396
The Drosophila nuclear protein Bx42, which is found in 81. Amling M, Priemel M, Holzmann T, Chapin K, Rueger
many puffs on polytene chromosomes, is highly JM, Baron R, Demay MB 1999 Rescue of the skeletal
charged. Chromosoma 101:517–525 phenotype of vitamin D receptor-ablated mice in the
65. Dahl R, Wani B, Hayman MJ 1998 The Ski oncoprotein setting of normal mineral ion homeostasis: formal his-
interacts with Skip, the human homolog of Drosophila tomorphometric and biomechanical analyses. Endocri-
Bx42. Oncogene 16:1579–1586 nology 140:4982–4987
66. Zhang C, Baudino TA, Dowd DR, Tokumaru H, Wang W, 82. Van Cromphaut SJ, Dewerchin M, Hoenderop JG,
MacDonald PN 2001 Ternary complexes and coopera- Stockmans I, Van Herck E, Kato S, Bindels RJ, Collen D,
tive interplay between NCoA-62/Ski-interacting protein Carmeliet P, Bouillon R, Carmeliet G 2001 Duodenal
and steroid receptor coactivators in vitamin D receptor- calcium absorption in vitamin D receptor-knockout
mediated transcription. J Biol Chem 276:40614–40620 mice: functional and molecular aspects. Proc Natl Acad
67. Zhou Z, Licklider LJ, Gygi SP, Reed R 2002 Compre- Sci USA 98:13324–13329
hensive proteomic analysis of the human spliceosome. 83. Li YC, Bolt MJG, Cao L-P, Sitrin MD 2001 Effects of
Nature 419:182–185 vitamin D receptor inactivation on the expression of
68. Makarov EM, Makarova OV, Urlaub H, Gentzel M, Will calbindins and calcium metabolism. Am J Physiol En-
CL, Wilm M, Luhrmann R 2002 Small nuclear ribonu- docrinol Metab 281:E558–E564
cleoprotein remodeling during catalytic activation of the 84. Erben RG, Soegiarto DW, Weber K, Zeitz U, Lieberherr
spliceosome. Science 298:2205–2208 M, Gniadecki R, Moller G, Adamski J, Balling R 2002
69. Auboeuf D, Honig A, Berget SM, O’Malley BW 2002 Deletion of deoxyribonucleic acid binding domain of the
Coordinate regulation of transcription and splicing by vitamin D receptor abrogates genomic and nongenomic
steroid receptor coregulators. Science 298:416–419 functions of vitamin D. Mol Endocrinol 16:1524–1537
70. Xu J, Qiu Y, DeMayo FJ, Tsai SY, Tsai M-J, O’Malley 85. Haussler MR, Haussler CA, Jurutka PW, Thompson PD,
BW 1998 Partial hormone resistance in mice with dis- Hsieh JC, Remus LS, Selznick SH, Whitfield GK 1997
ruption of the steroid receptor coactivator-1 (SRC-1) The vitamin D hormone and its nuclear receptor: mo-
gene. Science 279:1922–1925 lecular actions and disease states. J Endocrinol 154:
71. Gehin M, Mark M, Dennefeld C, Dierich A, Gronemeyer S57–S73
H, Chambon P 2002 The function of TIF2/GRIP1 in 86. Sakai Y, Demay MB 2000 Evaluation of keratinocyte
mouse reproduction is distinct from those of SRC-1 and proliferation and differentiation in vitamin D receptor
p/CIP. Mol Cell Biol 22:5923–5937 knockout mice. Endocrinology 141:2043–2049
72. Xu J, Liao L, Ning G, Yoshida-Komiya H, Deng C, 87. Sakai Y, Kishimoto J, Demay MB 2001 Metabolic and
O’Malley BW 2000 The steroid receptor coactivator cellular analysis of alopecia in vitamin D receptor knock-
SRC-3 (p/CIP/RAC3/AIB1/ACTR/TRAM-1) is required out mice. J Clin Invest 107:961–966
for normal growth, puberty, female reproductive func- 88. Panda DK, Miao D, Tremblay ML, Sirois J, Farookhi R,
tion, and mammary gland development. Proc Natl Acad Hendy GN, Goltzman D 2001 Targeted ablation of the
Sci USA 97:6379–6384 25-hydroxyvitamin D 1␣-hydroxylase enzyme: evidence

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
790 Mol Endocrinol, May 2003, 17(5):777–791 Minireview • Sutton and MacDonald

for skeletal, reproductive, and immune dysfunction. ulator of the renin-angiotensin system. J Clin Invest
Proc Natl Acad Sci USA 98:7498–7503 110:229–238
89. Masuyama H, Jefcoat Jr SC, MacDonald PN 1997 The 106. Yagishita N, Yamamoto Y, Yoshizawa T, Sekine K, Ue-
N-terminal domain of transcription factor IIB is required matsu Y, Murayama H, Nagai Y, Krezel W, Chambon P,
for direct interaction with the vitamin D receptor and Matsumoto T, Kato S 2001 Aberrant growth plate de-
participates in vitamin D-mediated transcription. Mol velopment in VDR/RXR ␥ double null mutant mice. En-
Endocrinol 11:218–228 docrinology 142:5332–5341
90. Lavigne AC, Mengus G, Gangloff YG, Wurtz JM, David- 107. Whitfield GK, Jurutka PW, Haussler CA, Haussler MR
son I 1999 Human TAF(II)55 interacts with the vitamin 1999 Steroid hormone receptors: evolution, ligands,
D(3) and thyroid hormone receptors and with derivatives and molecular basis of biologic function. J Cell Biochem
of the retinoid X receptor that have altered transactiva- (Suppl)32–33:110–122
tion properties. Mol Cell Biol 19:5486–5494 108. Takeyama K, Kitanaka S, Sato T, Kobori M, Yanagisawa
91. Tolon RM, Castillo AI, Jimenez-Lara AM, Aranda A 2000 J, Kato S 1997 25-Hydroxyvitamin D3 1␣-hydroxylase
Association with Ets-1 causes ligand- and AF2-inde- and vitamin D synthesis. Science 277:1827–1830
pendent activation of nuclear receptors. Mol Cell Biol 109. St-Arnaud R, Messerlian S, Moir JM, Omdahl JL, Glo-
20:8793–8802 rieux FH 1997 The 25-hydroxyvitamin D 1-␣-hydroxy-
92. Chen JD, Evans RM 1995 A transcriptional co-repressor lase gene maps to the pseudovitamin D-deficiency rick-
that interacts with nuclear hormone receptors. Nature ets (PDDR) disease locus. J Bone Miner Res 12:
377:454–457 1552–1559
93. Kurokawa R, Soderstrom M, Horlein A, Halachmi 110. Dardenne O, Prud’homme J, Arabian A, Glorieux FH,
S, Brown M, Rosenfeld MG, Glass CK 1995 Polarity- St-Arnaud R 2001 Targeted inactivation of the 25-
specific activities of retinoic acid receptors determined hydroxyvitamin D3-1␣-hydroxylase gene (CYP27B1)
by a co-repressor. Nature 377:451–454 creates an animal model of pseudovitamin D-deficiency
94. Horlein AJ, Naar AM, Heinzel T, Torchia J, Gloss B, rickets. Endocrinology 142:3135–3141
Kurokawa R, Ryan A, Kamei Y, Soderstrom M, Glass 111. Seo E-G, Einhorn TA, Norman AW 1997 24R,25-Dihy-
CK, Rosenfeld MG 1995 Ligand-independent repres- droxyvitamin D3: an essential vitamin D3 metabolite for
sion by the thyroid hormone receptor mediated by a both normal bone integrity and healing of tibial fracture
nuclear receptor co-repressor. Nature 377:397–404 in chicks. Endocrinology 138:3864–3872
95. Kinuta K, Tanaka H, Moriwake T, Aya K, Kato S, Seino 112. Seo EG, Norman AW 1997 Three-fold induction of renal
Y 2000 Vitamin D is an important factor in estrogen 25-hydroxyvitamin D3–24-hydroxylase activity and in-
biosynthesis of both female and male gonads. Endocri- creased serum 24,25-dihydroxyvitamin D3 levels are
nology 141:1317–1324 correlated with the healing process after chick tibial
96. Johnson LE, DeLuca HF 2001 Vitamin D receptor null fracture. J Bone Miner Res 12:598–606
mutant mice fed high levels of calcium are fertile. J Nutr 113. Boyan BD, Sylvia VL, Dean DD, Schwartz Z 2001 24,25-
131:1787–1791 (OH)2D3 regulates cartilage and bone via autocrine and
97. Zinser G, Packman K, Welsh J 2002 Vitamin D(3) recep- endocrine mechanisms. Steroids 66:363–374
tor ablation alters mammary gland morphogenesis. De- 114. St-Arnaud R, Arabian A, Travers R, Barletta F, Raval-
velopment 129:3067–3076 Pandya M, Chapin K, Depovere J, Mathieu C, Christa-
98. Elstner E, Linker-Israeli M, Said J, Umiel T, de Vos S, kos S, Demay MB, Glorieux FH 2000 Deficient min-
Shintaku IP, Heber D, Binderup L, Uskokovic M, Koef- eralization of intramembranous bone in vitamin
fler HP 1995 20-Epi-vitamin D3 analogues: a novel class D-24-hydroxylase-ablated mice is due to elevated 1,25-
of potent inhibitors of proliferation and inducers of dif- dihydroxyvitamin D and not to the absence of 24,25-
ferentiation of human breast cancer cell lines. Cancer dihydroxyvitamin D. Endocrinology 141:2658–2666
Res 55:2822–2830 115. Brown AJ 2001 Therapeutic uses of vitamin D ana-
99. Campbell MJ, Gombart AF, Kwok SH, Park S, Koeffler logues. Am J Kidney Dis 38:S3–S19
HP 2000 The anti-proliferative effects of 1␣,25(OH)2D3 116. Scott LJ, Dunn CJ, Goa KL 2001 Calcipotriol ointment.
on breast and prostate cancer cells are associated with A review of its use in the management of psoriasis. Am J
induction of BRCA1 gene expression. Oncogene 19: Clin Dermatol 2:95–120
5091–5097 117. Bouillon R, Allewaert K, Xiang DZ, Tan BK, van Baelen
100. Abe E, Miyaura C, Sakagami H, Takeda M, Konno K, H 1991 Vitamin D analogs with low affinity for the vita-
Yamazaki T, Yoshiki S, Suda T 1981 Differentiation of min D binding protein: enhanced in vitro and decreased
mouse myeloid leukemia cells induced by 1␣,25-dihy- in vivo activity. J Bone Miner Res 6:1051–1057
droxyvitamin D3. Proc Natl Acad Sci USA 78: 118. Slatopolsky E, Brown A, Dusso A 1999 Pathogenesis of
4990–4994 secondary hyperparathyroidism. Kidney Int Suppl 73:
101. O’Kelly J, Hisatake J, Hisatake Y, Bishop J, Norman A, S14–S19
Koeffler HP 2002 Normal myelopoiesis but abnormal T 119. Martin KJ, Gonzalez EA, Gellens M, Hamm LL, Abboud
lymphocyte responses in vitamin D receptor knockout H, Lindberg J 1998 19-Nor-1-␣-25-dihydroxyvitamin
mice. J Clin Invest 109:1091–1099 D2 (Paricalcitol) safely and effectively reduces the levels
102. Mathieu C, Van Etten E, Gysemans C, Decallonne B, of intact parathyroid hormone in patients on hemodial-
Kato S, Laureys J, Depovere J, Valckx D, Verstuyf A, ysis. J Am Soc Nephrol 9:1427–1432
Bouillon R 2001 In vitro and in vivo analysis of the 120. Tan Jr AU, Levine BS, Mazess RB, Kyllo DM, Bishop
immune system of vitamin D receptor knockout mice. CW, Knutson JC, Kleinman KS, Coburn JW 1997 Effec-
J Bone Miner Res 16:2057–2065 tive suppression of parathyroid hormone by 1␣-
103. Kristal-Boneh E, Froom P, Harari G, Ribak J 1997 As- hydroxy-vitamin D2 in hemodialysis patients with mod-
sociation of calcitriol and blood pressure in normoten- erate to severe secondary hyperparathyroidism. Kidney
sive men. Hypertension 30:1289–1294 Int 51:317–323
104. Lind L, Hanni A, Lithell H, Hvarfner A, Sorensen OH, 121. Tilyard MW, Spears GF, Thomson J, Dovey S 1992
Ljunghall S 1995 Vitamin D is related to blood pressure Treatment of postmenopausal osteoporosis with calcit-
and other cardiovascular risk factors in middle-aged riol or calcium. N Engl J Med 326:357–362
men. Am J Hypertens 8:894–901 122. Ikeda K, Ogata E 2000 Modulation of bone remodeling
105. Li YC, Kong J, Wei M, Chen Z-F, Liu SQ, Cao L-P 2002 by active vitamin D: its role in the treatment of osteo-
1,25-Dihydroxyvitamin D3 is a negative endocrine reg- porosis. Mech Ageing Dev 116:103–111

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.
Minireview • Sutton and MacDonald Mol Endocrinol, May 2003, 17(5):777–791 791

123. 2000 Osteoporosis prevention, diagnosis, and therapy. 140. Harris SS, Eccleshall TR, Gross C, Dawson-Hughes B,
NIH Consensus Statement. Bethesda, MD: National In- Feldman D 1997 The vitamin D receptor start codon
stitutes of Health; 17:1–45 polymorphism (FokI) and bone mineral density in pre-
124. Peleg S, Uskokovic M, Ahene A, Vickery B, Avnur Z menopausal American black and white women. J Bone
2002 Cellular and molecular events associated with the Miner Res 12:1043–1048
bone-protecting activity of the noncalcemic vitamin D 141. Ferrari S, Rizzoli R, Manen D, Slosman D, Bonjour JP
analog Ro-26-9228 in osteopenic rats. Endocrinology 1998 Vitamin D receptor gene start codon polymor-
143:1625–1636 phisms (FokI) and bone mineral density: interaction with
125. Shevde NK, Plum LA, Clagett-Dame M, Yamamoto H, age, dietary calcium, and 3⬘-end region polymorphisms.
Pike JW, DeLuca HF 2002 A potent analog of 1␣,25- J Bone Miner Res 13:925–930
dihydroxyvitamin D3 selectively induces bone forma- 142. Gennari L, Becherini L, Mansani R, Masi L, Falchetti A,
tion. Proc Natl Acad Sci USA 99:13487–13491 Morelli A, Colli E, Gonnelli S, Cepollaro C, Brandi ML
126. Makishima M, Lu TT, Xie W, Whitfield GK, Domoto H, 1999 FokI polymorphism at translation initiation site of
Evans RM, Haussler MR, Mangelsdorf DJ 2002 Vitamin the vitamin D receptor gene predicts bone mineral den-
D receptor as an intestinal bile acid sensor. Science sity and vertebral fractures in postmenopausal Italian
296:1313–1316 women. J Bone Miner Res 14:1379–1386
127. Chawla A, Repa JJ, Evans RM, Mangelsdorf DJ 2001 143. Ames SK, Ellis KJ, Gunn SK, Copeland KC, Abrams SA
Nuclear receptors and lipid physiology: opening the 1999 Vitamin D receptor gene Fok1 polymorphism pre-
X-files. Science 294:1866–1870 dicts calcium absorption and bone mineral density in
128. Lu TT, Repa JJ, Mangelsdorf DJ 2001 Orphan nuclear children. J Bone Miner Res 14:740–746
receptors as eLiXiRs and FiXeRs of sterol metabolism. 144. Gross C, Krishnan AV, Malloy PJ, Eccleshall TR, Zhao
J Biol Chem 276:37735–37738 XY, Feldman D 1998 The vitamin D receptor gene start
129. Xie W, Evans RM 2001 Orphan nuclear receptors: the codon polymorphism: a functional analysis of FokI vari-
exotics of xenobiotics. J Biol Chem 276:37739–37742 ants. J Bone Miner Res 13:1691–1699
130. Debruyne PR, Bruyneel EA, Karaguni IM, Li X, Flatau G, 145. Falkenstein E, Tillmann HC, Christ M, Feuring M, Weh-
Muller O, Zimber A, Gespach C, Mareel MM 2002 Bile ling M 2000 Multiple actions of steroid hormones—a
acids stimulate invasion and haptotaxis in human colo- focus on rapid, nongenomic effects. Pharmacol Rev
rectal cancer cells through activation of multiple onco- 52:513–556
genic signaling pathways. Oncogene 21:6740–6750 146. Nemere I, Yoshimoto Y, Norman AW 1984 Calcium
131. Kozoni V, Tsioulias G, Shiff S, Rigas B 2000 The effect
transport in perfused duodena from normal chicks: en-
of lithocholic acid on proliferation and apoptosis during
hancement within fourteen minutes of exposure to 1,25-
the early stages of colon carcinogenesis: differential
dihydroxyvitamin D3. Endocrinology 115:1476–1483
effect on apoptosis in the presence of a colon carcin-
147. Norman AW 1997 Rapid biological responses mediated
ogen. Carcinogenesis 21:999–1005
by 1␣,25-dihydroxyvitamin D3: a case study of tran-
132. Lamprecht SA, Lipkin M 2001 Cellular mechanisms of
scaltachia (rapid hormonal stimulation of intestinal cal-
calcium and vitamin D in the inhibition of colorectal
cium transport). In: Feldman FHG, Pike JW, eds. Vita-
carcinogenesis. Ann NY Acad Sci 952:73–87
133. Aranda A, Pascual A 2001 Nuclear hormone receptors min D. San Diego: Academic Press; 233–256
and gene expression. Physiol Rev 81:1269–1304 148. Le Mellay V, Grosse B, Lieberherr M 1997 Phospho-
134. Miyamoto K-i, Kesterson RA, Yamamoto H, Taketani Y, lipase C␤ and membrane action of calcitriol and estra-
Nishiwaki E, Tatsumi S, Inoue Y, Morita K, Takeda E, diol. J Biol Chem 272:11902–11907
Pike JW 1997 Structural organization of the human 149. Vazquez G, de Boland AR, Boland RL 1998 1␣,25-
vitamin D receptor chromosomal gene and its pro- Dihydroxy-vitamin-D3-induced store-operated Ca2⫹ in-
moter. Mol Endocrinol 11:1165–1179 flux in skeletal muscle cells. Modulation by phospho-
135. Crofts LA, Hancock MS, Morrison NA, Eisman JA 1998 lipase C, protein kinase C, and tyrosine kinases. J Biol
Multiple promoters direct the tissue-specific expression Chem 273:33954–33960
of novel N-terminal variant human vitamin D receptor 150. Morelli S, Buitrago C, Vazquez G, De Boland AR, Bo-
gene transcripts. Proc Natl Acad Sci USA 95: land R 2000 Involvement of tyrosine kinase activity in
10529–10534 1␣,25(OH)2-vitamin D3 signal transduction in skeletal
136. Sunn KL, Cock TA, Crofts LA, Eisman JA, Gardiner EM muscle cells. J Biol Chem 275:36021–36028
2001 Novel N-terminal variant of human VDR. Mol En- 151. Nemere I, Ray R, McManus W 2000 Immunochemical
docrinol 15:1599–1609 studies on the putative plasmalemmal receptor for
137. Wood RJ, Fleet JC 1998 The genetics of osteoporosis: 1,25(OH)2D3. I. Chick intestine. Am J Physiol Endocrinol
vitamin D receptor polymorphisms. Annu Rev Nutr 18: Metab 278:E1104–E1114
233–258 152. Song X, Bishop JE, Okamura WH, Norman AW 1998
138. Gross C, Eccleshall TR, Malloy PJ, Villa ML, Marcus R, Stimulation of phosphorylation of mitogen-activated
Feldman D 1996 The presence of a polymorphism at the protein kinase by 1␣,25-dihydroxyvitamin D3 in promy-
translation initiation site of the vitamin D receptor gene elocytic NB4 leukemia cells: a structure-function study.
is associated with low bone mineral density in post- Endocrinology 139:457–465
menopausal Mexican-American women. J Bone Miner 153. Baran DT, Quail JM, Ray R, Honeyman T 2000 Binding
Res 11:1850–1855 of 1␣,25-dihydroxyvitamin D3 to annexin II: effect of
139. Arai H, Miyamoto K, Taketani Y, Yamamoto H, Iemori Y, vitamin D metabolites and calcium. J Cell Biochem
Morita K, Tonai T, Nishisho T, Mori S, Takeda E 1997 A 80:259–265
vitamin D receptor gene polymorphism in the translation 154. Baran DT, Quail JM, Ray R, Leszyk J, Honeyman T 2000
initiation codon: effect on protein activity and relation to Annexin II is the membrane receptor that mediates the
bone mineral density in Japanese women. J Bone Miner rapid actions of 1␣,25-dihydroxyvitamin D3. J Cell Bio-
Res 12:915–921 chem 78:34–46

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 15 July 2014. at 05:20 For personal use only. No other uses without permission. . All rights reserved.

You might also like