Sie sind auf Seite 1von 6

Journal of Steroid Biochemistry & Molecular Biology 103 (2007) 451–456

In vivo relevance for photoprotection by the


vitamin D rapid response pathway
K.M. Dixon a , S.S. Deo a , A.W. Norman b , J.E. Bishop b , G.M. Halliday c ,
V.E. Reeve d , R.S. Mason a,∗
a Department of Physiology and Bosch Institute, University of Sydney, NSW 2006, Australia
b Department of Biochemistry, University of California, Riverside, CA 92521, USA
c Department of Medicine (Dermatology), University of Sydney, NSW 2006, Australia
d Department of Veterinary Science, University of Sydney, NSW 2006, Australia

Abstract

Vitamin D is produced by exposure of 7-dehydrocholesterol in the skin to UV irradiation (UVR) and further converted in the skin to
the biologically active metabolite, 1,25-dihydroxyvitamin D3 (1,25(OH)2 D3 ) and other compounds. UVR also results in DNA damage
producing cyclobutane pyrimidine dimers (CPD). We previously reported that 1,25(OH)2 D3 at picomolar concentrations, protects human skin
cells from UVR-induced apoptosis, and decreases CPD in surviving cells. 1,25(OH)2 D3 has been shown to generate biological responses
via two pathways—the classical steroid receptor/genomic pathway or a rapid, non-genomic pathway mediated by a putative membrane
receptor. Whether the rapid response pathway is physiologically relevant is unclear. A cis-locked, rapid-acting agonist 1,25(OH)2 lumisterol3
(JN), entirely mimicked the actions of 1,25(OH)2 D3 to reduce fibroblast and keratinocyte loss and CPD damage after UVR. The effects
of 1,25(OH)2 D3 were abolished by a rapid-acting antagonist, but not by a genomic antagonist. Skh:hr1 mice exposed to three times the
minimal erythemal dose of solar-simulated UVR and treated topically with 1,25(OH)2 D3 or JN immediately after UVR showed reduction
in UVR-induced UVR-induced sunburn cells (p < 0.01 and <0.05, respectively), CPD (p < 0.01 for both) and immunosuppression (p < 0.001
for both) compared with vehicle-treated mice. These results show for the first time an in vivo biological response mediated by a rapid-acting
analog of the vitamin D system. The data support the hypothesis that 1,25(OH)2 D3 exerts its photoprotective effects via the rapid pathway
and raise the possibility that other D compounds produced in skin may contribute to the photoprotective effects.
© 2006 Elsevier Ltd. All rights reserved.

Keywords: Vitamin D; 1,25-Dihydroxyvitamin D3 ; Vitamin D analogs; Cyclobutane pyrimidine dimers (CPD); Sunburn cells; Ultraviolet radiation; Immuno-
suppression; Skh:hr1 hairless mice

1. Introduction in the liver, followed by the kidney, forming the biologically


active metabolite 1,25-dihydroxyvitamin D3 (1,25(OH)2 D3 ).
Exposure to ultraviolet radiation (UVR) from sunlight can There is evidence that the entire pathway to 1,25(OH)2 D3 is
result in a variety of responses in the skin including muta- present in skin [4].
genic DNA damage such as cyclobutane pyrimidine dimers Previous studies in this laboratory have shown that
(CPD) [1] and immunosuppression [2], both of which play 1,25(OH)2 D3 not only reduces the level of UVR-induced cell
a role in skin carcinogenesis. UVR is also the catalyst for death in human skin cells, but also reduces CPD in surviving
the formation of vitamin D3 in skin. Upon the interaction of cells in a dose-dependent manner when added to skin cell
UVR with 7-dehydrocholesterol in skin, pre-vitamin D3 is cultures prior to and/or after UVR [5–7].
formed, which thermally isomerizes to vitamin D3 [3]. Vita- The various biological responses produced by 1,25(OH)2
min D3 undergoes two sequential hydroxylation reactions D3 appear to be mediated by two major pathways. The
well-studied genomic pathway depends on the interaction
∗ Corresponding author. between 1,25(OH)2 D3 and the nuclear vitamin D recep-
E-mail address: rebeccam@physiol.usyd.edu.au (R.S. Mason). tor (VDR) followed by interaction of this steroid receptor

0960-0760/$ – see front matter © 2006 Elsevier Ltd. All rights reserved.
doi:10.1016/j.jsbmb.2006.11.016
452 K.M. Dixon et al. / Journal of Steroid Biochemistry & Molecular Biology 103 (2007) 451–456

Fig. 1. Structures of 1,25(OH)2 -lumisterol3 (JN) and 1,25(OH)2 D3 in cis- and trans-form.

complex with vitamin D response elements present in the The sham-irradiated plate was subjected to similar conditions
promoter regions of the target genes, resulting in up- or down- but covered in aluminium foil for the duration of irradiation.
regulation of transcription [8]. There is now considerable The UVR source for in vitro studies consisted of one
evidence to suggest that 1,25(OH)2 D3 can also utilize other UVA and one UVB fluorescent lamp, with irradiance of
signal transduction mechanisms in order to generate rapid, 203 mJ/cm2 UVB and 1168 mJ/cm2 UVA (Phillips, Amster-
non-genomic responses. A rapid-acting, non-genomic path- dam, Holland), filtered through 0.5 mm cellulose tri-acetate
way for 1,25(OH)2 D3 action has been proposed to generate a (Eastman Chemical Products, Kingsport, TN) to eliminate
variety of cell specific responses within seconds to minutes. wavelengths below 290 nm [6,7].
This involves the hormone binding to a membrane receptor, Cell loss after UVR was measured as previously described
the identity of which is still not clear [9,10]. [7]. Cyclobutane pyrimidine dimers (CPD) were detected
In the current study, vitamin D analogs were used by immunohistochemistry and image analysis as described
to differentiate between the vitamin D pathways. The previously [7].
steroid hormone, 1,25(OH)2 D3 , because it is conforma- Female Skh:hr1 (albino, inbred) hairless mice were used
tionally flexible, is able to generate biological responses for in vivo studies. Housing and irradiation conditions were as
either by the classical genomic pathway or by the previously described [7]. Mice were subjected to three times
membrane receptor-initiated rapid response path. It has the minimal erythemal dose (MED) of UVR (3.98 kJ/m2
been shown that analogs permanently locked in the cis- UVB and 63.8 kJ/m2 UVA).
configuration, as in 1␣,25(OH)2 lumisterol3 (JN), can activate Treatments were diluted in a base lotion containing
only the rapid response pathway [11] (Fig. 1). The com- ethanol, propylene glycol and water at a final solvent ratio
pound 1␤,25(OH)2 D3 (HL) acts only as a rapid response of 2:1:1. Mice were treated with vehicle, 1,25(OH)2 D3 , or
antagonist, whilst (23S)-25-dehydro-1␣-hydroxyvitamin D3 - the low calcemic, rapid-acting analog JN. All treatments
26,23-lactone (TEI-9647) acts as a genomic antagonist. No were applied dorsally, once, immediately after irradiation
specific agonist of the genomic pathway has been reported. only. Skin samples were taken from UV-irradiated back skin
In our preliminary studies we showed that the photoprotec- 24 h post-UVR, fixed in Histochoice (Amresco) and paraffin-
tive effects of 1,25(OH)2 D3 in human skin cells were likely embedded for sunburn cell analysis by routine haematoxylin
to be acting via the rapid pathway [6], and showed that the and eosin staining. Sunburn cells were identified under a light
photoprotective effects of 1,25(OH)2 D3 were present in vivo. microscope as cells with pyknotic nuclei surrounded by an
We now provide further data to support these proposals, and eosinophilic cytoplasm, and were counted along the length
show for the first time, an in vivo photoprotective effect of a of each skin section. Detection of CPD was by immunohis-
low-calcemic analog that can only activate the rapid pathway. tochemical analysis as previously described [7].
The contact hypersensitivity response was performed as
previously described [7]. Briefly, mice were sensitized 1 week
2. Methods after irradiation with 100 ␮L of 2% oxazolone (Sigma, St.
Louis, MO, USA) (w/v) in absolute alcohol applied to non-
Fibroblasts and keratinocytes from human neonatal fore- irradiated abdominal skin. Sensitization was repeated on the
skins were cultured in 96-well plates on 5 mm glass coverslips subsequent day. The sensitized mice were challenged 2 weeks
coated with poly-l-lysine as described in [12]. Cells were after irradiation. Ear thickness measurements were recorded
treated with 1,25(OH)2 D3 or analogs 24 h prior to irradiation before challenge and at 20 h after challenge.
and media containing treatments were added immediately Results are based on quadruplicates of each treatment from
after UVR. Treatments were replaced with Martinez buffer experiments performed at least twice with similar results.
solution containing d-glucose for the duration of irradiation. Comparisons between treatment groups were made by one-
K.M. Dixon et al. / Journal of Steroid Biochemistry & Molecular Biology 103 (2007) 451–456 453

Fig. 2. Protective effects of 1,25(OH)2 D3 on UVR-induced skin cell loss (a) and CPD (b) reversed by rapid-acting antagonist HL but not by genomic antagonist
TEI-9647. Cells were treated for 24 h with vehicle, 1,25(OH)2 D3 , TEI-9647, HL, or a combination of 1,25(OH)2 D3 and either TEI-9647 or HL. Treatments
were replaced with Martinez solution for the duration of irradiation, and post-treatments added immediately after irradiation. Cell loss was determined 24 h
after UVR (a). Significantly different from vehicle: *** p < 0.001 and * p < 0.05. CPD were detected 3 h after UVR (b). Significantly different from vehicle:
** p < 0.01.

way analysis of variance (ANOVA) using the GraphPad Instat As shown in Fig. 2a, the addition of the genomic path-
statistical program (GraphPad Software Inc., San Diego, CA). way antagonist, TEI-9647 to skin fibroblasts had no effect on
the photoprotective action of 1,25(OH)2 D3 . Mean cell loss
in vehicle-treated skin fibroblasts was 36.3 ± 3.0%. This was
3. Results reduced to 21.0 ± 2.2% in cells treated with 1,25(OH)2 D3
(p < 0.001). Mean loss in cells treated with TEI-9647 or HL
The level of UVR-induced cell loss was reduced in alone was not significantly different to that of vehicle-treated
cells treated with either 1,25(OH)2 D3 or JN, compared with cells. When cells were treated with a combination of TEI-
vehicle-treated cells. In pooled data from two separate exper- 9647 and 1,25(OH)2 D3 , mean cell loss was 28.8 ± 1.7%,
iments, mean cell loss in vehicle-treated skin fibroblasts was significantly lower than in vehicle-treated cells (p < 0.05)
37.1 ± 4.0%. This was significantly reduced to 15.4 ± 4.5% and not significantly different to mean loss in cells treated
in cells treated with 1,25(OH)2 D3 (p < 0.01). The rapid- with 1,25(OH)2 D3 alone. In contrast, the protective effect of
acting, low-calcemic analog JN completely mimicked the 1,25(OH)2 D3 on cell loss was abolished in the presence of
effects of 1,25(OH)2 D3 , reducing cell loss to 15.2 ± 5.8% HL (Fig. 2a).
(p < 0.01). Similar results were obtained in keratinocytes. The proportion of positive CPD staining in cells treated
Furthermore, both 1,25(OH)2 D3 and JN significantly reduced with a combination of TEI-9647 and 1,25(OH)2 D3 did not
CPD in keratinocytes from 18.6 ± 6.0 to 3.7 ± 1.0 (p < 0.01) differ from that in cells treated with 1,25(OH)2 D3 alone
and 5.3 ± 1.7 (p < 0.05), respectively. (Fig. 2b). In cells treated with the rapid response pathway
454 K.M. Dixon et al. / Journal of Steroid Biochemistry & Molecular Biology 103 (2007) 451–456

Fig. 3. Effects of rapid response pathway antagonist HL and genomic pathway antagonist TEI-9647 on UV-induced CPD in skin fibroblasts. Images are
representative of fields captured during image analysis for (A) UV-irradiated vehicle; (B) UV-irradiated 1␣,25(OH)2 D3 ; (C) UV-irradiated Tei-9647; (D)
UV-irradiated Tei-9647 + 1␣,25(OH)2 D3 ; (E) UV-irradiated HL; (F) UV-irradiated HL + 1␣,25(OH)2 D3 .

antagonist HL, there was no difference in the level of 1,25(OH)2 D3 as well as JN, significantly reduced lev-
CPD compared with vehicle-treated cells. When cells were els of UVR-induced immunosuppression in Skh:hr1 mice.
treated with a combination of HL and 1,25(OH)2 D3 , the As shown in Fig. 5, the level of immunosuppression in
inhibitory effects of 1,25(OH)2 D3 on CPD were abolished vehicle-treated mice at 20 h post-challenge was 23.4 ± 1.3%.
(Figs. 2b and 3). In mice treated with 1,25(OH)2 D3 at a dose of 22.8 pmol/cm2
Sunburn cells (apoptotic keratinocytes) in mouse skin this was reduced to 3.9 ± 1.5% (p < 0.001). JN at the same
were reduced by both 1,25(OH)2 D3 and JN. In mice treated dose (22.8 pmol/cm2 ) also significantly reduced immunosup-
with 1,25(OH)2 D3 at doses of 22.8 and 4.6 pmol/cm2 , the pression at 20 h post-challenge to −3.0 ± 1.5% (p < 0.001)
numbers of sunburn cells per 245 linear micrometres of (Fig. 5).
skin were reduced from 0.5 ± 0.1 to 0.2 ± 0.1 (p < 0.01) and
0.4 ± 0.1 (p < 0.05), respectively. In mice treated with JN
at a dose of 22.8 pmol/cm2 , sunburn cells were reduced to 4. Discussion
0.3 ± 0.0 (p < 0.05) (Fig. 4).
Topical 1,25(OH)2 D3 or JN at similar doses reduced CPD We have previously provided preliminary evidence to sup-
measured 24 h post-UVR from 9 ± 3% to 3 ± 1% (p < 0.01) port the proposal that the rapid pathway is involved in the
and 4 ± 2% (p < 0.01), respectively. photoprotective effect of 1,25(OH)2 D3 [6,7]. In the current
K.M. Dixon et al. / Journal of Steroid Biochemistry & Molecular Biology 103 (2007) 451–456 455

Fig. 4. Reduction in sunburn cells (apoptotic keratinocytes) by 1,25(OH)2 D3 in Skh:hr1 mouse skin. Mice were exposed to 3 MED solar-simulated UVR and
treated topically with vehicle or 1,25(OH)2 D3 immediately after exposure. Skin biopsies were taken 24 h after UVR. Results presented as mean number of
sunburn cells per 245 linear micrometres of skin section, n = 3. Significantly different from vehicle: ** p < 0.01 and * p < 0.05.

study, we show that the rapid-acting analog, JN, entirely mim- These functions are likely to be important, as there is
ics the photoprotective effects of 1,25(OH)2 D3 in human skin ample evidence that both production of CPD and immunosup-
cells. Furthermore, both the reduction in UVR-induced cell pression are key elements in skin carcinogenesis [2]. Since
loss and in CPD damage by 1,25(OH)2 D3 were fully reversed CPD are produced by the direct interaction of UVR with
by the rapid response antagonist HL and unaffected by the DNA, and the protective effects of vitamin D compounds are
genomic antagonist TEI-9647. seen following application after irradiation, it is reasonable
We have shown for the first time an in vivo complex to speculate that the CPD reduction is the result, at least in
biological response using an analog that is only capable part, of increased repair, probably in turn a result of increased
of acting via the rapid pathway. The cis-locked compound p53 expression and reduced nitric oxide products, previously
JN, which has no transcriptional activity, entirely mimicked demonstrated in the presence of 1,25(OH)2 D3 [7]. Repair
the effects of 1,25(OH)2 D3 at equivalent doses by reducing of CPDs reduces skin cell apoptosis, and prevents UVR-
UVR-induced sunburn cells, DNA damage and immuno- induced mutation but also immunosuppression [13], thus
suppression in Skh:hr1 hairless mice. These results provide providing a link between the three effects described in vivo.
strong and consistent evidence for a physiological role of the The process of vitamin D3 production in skin following
rapid response pathway in photoprotection. exposure to UVR takes several hours. The conversion of vita-
min D3 to 1,25(OH)2 D3 takes place over several further hours
[14]. Because of this long time-course, it is unlikely that the
1,25(OH)2 D3 produced locally in skin protects from the UVR
exposure responsible for its production. It is more likely that
the locally produced vitamin D compounds protect from fur-
ther UVR exposure, as is the case for UVR-induced increases
in cornification and pigmentation [15,16]. Overirradiation
products have some structural similarities to the rapid-acting
agonists [3] and it is possible to speculate that these com-
pounds may also contribute to the photoprotective effect.
The data presented are consistent with the proposal that
locally produced 1,25(OH)2 D3 contributes to endogenous
Fig. 5. Reduction in UVR-induced systemic immunosuppression by
photoprotection in skin. The data also provide evidence that
1,25(OH)2 D3 and JN in Skh:hr1 mice. Mice were exposed to 3 MED solar- the rapid response pathway for vitamin D plays an important
simulated UVR and treated immediately after exposure once with vehicle, physiological role in a whole animal.
1,25(OH)2 D3 or JN. Mice were sensitized on non-irradiated abdominal skin
7 and 8 d after UVR with 2% oxazolone. Mice were challenged on ears
7 d after sensitization and ear swelling recorded 20 h later. Results were
Acknowledgements
calculated as the difference between pre- and post-challenge ear thick-
ness measurements of non-irradiated mice as a proportion of the difference
between pre- and post-challenge ear thickness measurements of irradiated This work was supported by the National Health and
mice for each treatment. Immunosuppression expressed as 100% minus this Medical Research Council of Australia and by the Cancer
value, ± S.E.M. *** p < 0.001 significantly different from vehicle. n = 5. Council of New South Wales. K.M. Dixon was the recipient
456 K.M. Dixon et al. / Journal of Steroid Biochemistry & Molecular Biology 103 (2007) 451–456

of a N.S.W. Cancer Institute Research Scholar Award. With [8] A.W. Norman, et al., Different shapes of the steroid hormone
thanks to Dr. S. Ishizuka, The Teijin Institute for Biomed- 1alpha,25(OH)(2)-vitamin D(3) act as agonists for two different recep-
ical Research, Tokyo, Japan, for providing the TEI-9647 tors in the vitamin D endocrine system to mediate genomic and rapid
responses, Steroids 66 (3–5) (2001) 147–158.
compound. [9] A.W. Norman, M.T. Mizwicki, D.P. Norman, Steroid-hormone rapid
actions, membrane receptors and a conformational ensemble model,
Nat. Rev. 3 (1) (2004) 27–41.
References [10] I. Nemere, et al., Ribozyme knockdown functionally links a
1,25(OH)2 D3 membrane binding protein (1,25D3 -MARRS) and phos-
phate uptake in intestinal cells, Proc. Natl. Acad. Sci. U.S.A. 101 (19)
[1] R.B. Setlow, Cyclobutane-type pyrimidine dimers in polynucleotides,
(2004) 7392–7397.
Science 153 (734) (1966) 379–386.
[11] A.W. Norman, et al., Comparison of 6-s-cis- and 6-s-trans-locked
[2] V.E. Reeve, R.D. Ley, Animal models of ultraviolet radiation-induced
analogs of 1alpha,25-dihydroxyvitamin D3 indicates that the 6-s-cis
skin cancer, in: D. Hill, J.M. Elwood, D.R. English (Eds.), Prevention of
conformation is preferred for rapid nongenomic biological responses
Skin Cancer. Cancer Prevention–Cancer Causes, vol. 3, Kluwer Acad
and that neither 6-s-cis- nor 6-s-trans-locked analogs are preferred
Publ., Dordrecht, 2004, pp. 177–194.
for genomic biological responses, Mol. Endocrinol. 11 (10) (1997)
[3] M.F. Holick, McCollum Award Lecture, 1994: vitamin D—new hori-
1518–1531.
zons for the 21st century, Am. J. Clin. Nutr. 60 (4) (1994) 619–
[12] C. Gordon-Thomson, R.S. Mason, G.P. Moore, Regulation of epider-
630.
mal growth factor receptor expression in human melanocytes, Exp.
[4] B. Lehmann, et al., UVB-induced conversion of 7-dehydrocholesterol
Dermatol. 10 (5) (2001) 321–328.
to 1alpha,25-dihydroxyvitamin D3 in an in vitro human skin equivalent
[13] M.L. Kripke, et al., Pyrimidine dimers in DNA initiate systemic
model, J. Invest. Dermatol. 117 (5) (2001) 1179–1185.
immunosuppression in UV-irradiated mice, Proc. Natl. Acad. Sci.
[5] R.S. Mason, C.J. Holliday, R. Gupta, 1,25-Dihydroxyvitamin D and
U.S.A. 89 (16) (1992) 7516–7520.
photoprotection in skin cells, in: D. Tsambos, H. Merk (Eds.), Modern
[14] B. Lehmann, S. Abraham, M. Meurer, Role for tumor necrosis
Trends in Skin Pharmacology, Parissianos Medical Publications S.A.
factor-alpha in UVB-induced conversion of 7-dehydrocholesterol to
Athens, Athens, Greece, 2002, pp. 59–66.
1alpha,25-dihydroxyvitamin D3 in cultured keratinocytes, J. Steroid
[6] G. Wong, et al., 1,25-Dihydroxyvitamin D and three low-calcemic
Biochem. Mol. Biol. 89–90 (1–5) (2004) 561–565.
analogs decrease UV-induced DNA damage via the rapid response
[15] T.B. Fitzpatrick, Ultraviolet-induced pigmentary changes: benefits and
pathway, J. Steroid Biochem. Mol. Biol. 89–90 (1–5) (2004) 567–
hazards, Curr. Problems Dermatol. 15 (1986) 25–38.
570.
[16] N.S. Dissanayake, R.S. Mason, Modulation of skin cell functions by
[7] K.M. Dixon, et al., Skin cancer prevention: a possible role of 1,25dihy-
transforming growth factor-beta1 and ACTH after ultraviolet irradia-
droxyvitamin D3 and its analogs, J. Steroid Biochem. Mol. Biol. 97
tion, J. Endocrinol. 159 (1) (1998) 153–163.
(1–2) (2005) 137–143.

Das könnte Ihnen auch gefallen