ArticlePDF Available

Vitamin D status in psoriasis patients treated with UVB therapy

Authors:
Vitamin D Status in Psoriasis Patients
Treated with UVB Therapy
Amra Osmančević
Department of Dermatology and Venereology
Sahlgrenska University Hospital
Institute of Clinical Sciences at
Sahlgrenska Academy
Göteborg, Sweden
2009
Title: Vitamin D status in psoriasis patients treated with UVB therapy
Author: Amra Osmančević, MD
E-mail:Amra.Osmancevic@vgregion.se
Department of Dermatology and Venereology, Sahlgrenska University Hospital,
Institute of Clinical Sciences at Sahlgrenska Academy, University of Gothenburg,
Göteborg, Sweden
Cover:
The cover picture illustrates the sun, the skin compartments (stratum basale,
stratum spinosum, stratum granulosum and stratum corneum) and the chemical
structure of vitamin D3. Ultraviolet B radiation stimulates the production of
vitamin D3 in stratum basale.
The Digital version can be downloaded from
http://hdl.handle.net/2077/19041
Printed by Geson Hylte Tryck AB, Kungsbacka, Sweden 2009
Mojim roditeljima
To my parents Magbula and Aziz Ahmic
"Nasi daleki preci su vjerovali da zivot treba ispuniti sa tri dobra djela, od djeteta covjeka podici,
kucu sagraditi i napisati knjigu zivota..." ”Zagubljene slike”, Sevko Kadric
”Our old ancestors believed that life could only be fulfilled through the completion of three good
deeds: bringing up one’s child to adulthood, building a house, and writing a book on life...”
”Borttappade bilder”, Sevko Kadric
1
Vitamin D status in psoriasis patients treated with UVB therapy
Amra Osmancevic
Department of Dermatology and Venereology, Institute of Clinical Sciences at
Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
Abstract
The thesis deals with the effect of ultraviolet B (UVB) 280-320 nm on vitamin D
production in psoriasis patients during treatment with phototherapy.
Background:
Psoriasis is a chronic, inflammatory disease affecting the skin and potentially
the joints. Both genetic and environmental factors are important in the aetiology of the
disease. Phototherapy (broadband UVB, narrowband UVB (NBUVB) and heliotherapy) is
commonly used as treatment of psoriasis.
Vitamin D3, or cholecalciferol, is produced in the basal epidermis by ultraviolet radiation
(290-315 nm) of 7-dehydrocholesterol and hydroxylated in the liver to the major circulating
metabolite 25-hydroxyvitamin D [25(OH)D]. Hydroxylation to 1,25-dihydroxyvitamin D
[1,25(OH)2D] in the kidneys is stimulated by parathyroid hormone (PTH) and suppressed by
phosphate. Sun exposure is the strongest factor influencing 25(OH)D.
Aims:
1) To study the effect of UVB on vitamin D synthesis in patients with psoriasis. 2)
To examine possible differences between NBUVB and broadband UVB on vitamin D
production in psoriatic patients. 3) To investigate the effect of UVB induced vitamin D on
bone, lipid and carbohydrate status in psoriasis patients.
Methods:
Serum 25(OH)D, 1,25(OH)2D, PTH, calcium and creatinine were measured
before and after the phototherapy in white, Caucasian patients with active plaque psoriasis.
Bone mineral density (BMD) was examined using Dual-Energy X-ray Absorptiometry
(DEXA) in postmenopausal women with psoriasis. Lipid and carbohydrate status were
assessed in patients treated with heliotherapy.
Results:
Psoriasis improved in all patients, with a 75% reduction in PASI (Psoriasis Area
and Severity Index) score on all regimes. Serum 25(OH)D increased and PTH decreased
after phototherapy. The increase in 25(OH)D was higher in the broadband treated patients
compared with NBUVB. There was no correlation between the dose of UVB and the
increase of 25(OH)D. Postmenopausal women with psoriasis had higher BMD both at the
hip and at the lumbar spine than age-matched controls. The ratio of low-density
lipoprotein (LDL) and high-density lipoprotein cholesterol (HDL), and the levels of
glycosylated haemoglobin A1c (HbA1c) decreased during heliotherapy.
Conclusion:
UVB and heliotherapy increased the serum 25(OH)D production, reduced
the serum PTH concentrations and improved psoriasis, lipid and carbohydrate status in the
patients. Vitamin D production in psoriasis patients increased less with NBUVB than with
broadband UVB phototherapy. Postmenopausal women with psoriasis had higher BMD
than age-matched controls, a finding that could be related to their higher body weight,
physical activity and the UVB exposure.
Key words: Vitamin D, PTH, psoriasis, bone mineral density, ultraviolet UVB
ISBN-978-91-628-7682-1
Göteborg 2009
”Vitamin D under ljusbehandling hos patienter med psoriasis”
Amra Osmancevic, Hudkliniken, Sahlgrenska Universitetssjukhuset
Svensk sammanfattning
Ultraviolett B (UVB) ljus respektive solljus under klimatvård på Gran Canaria
förbättrar psoriasis och höjer vitamin D nivåerna i blodet. Vitamin D produktionen
hos patienter med psoriasis ökar mer under behandling med bredband UVB (280-
320 nm) än med smalspektrum UVB, så kallat TL01 (311 nm).
Undersökning av en grupp postmenopausala kvinnor med psoriasis visade att de
hade bättre bentäthet än jämnåriga kvinnor från Göteborg. Detta kan delvis bero
på att kvinnorna med psoriasis hade högre kroppsvikt, fysisk aktivitet och positiv
effekt av UVB på vitamin D nivåerna.
Behandling med solljus under s.k. klimatvård på Gran Canaria hade gynnsam effekt
på psoriasis, ökade vitamin D och förbättrade blodfetterna samt
sockeromsättningen hos dessa patienter.
Psoriasispatienter med låga nivåer av vitamin D i blodet (<30 ng/ml) före
behandlingsstart, ökade mer i sina D-vitaminvärden efter ljusbehandlingarna än
dem som hade högre halter av D-vitamin vid start. Alla patienter gick upp i sina D-
vitaminvärden oavsett ålder, hudtyp eller svårighetsgrad av psoriasis. Effekt av
solljus på vitamin D under 2 veckors klimatvård på Gran Canaria kan jämföras med
effekten av behandling med UVB lampa 2-3 gånger/vecka under 2 till 3 månaders
tid.
UVB och solljus ökade vitamin D i blodet, förbättrade psoriasis, var associerat med
bättre benmassa, och hade positiv effekt på lipider och sockeromsättning.
Vitamin D status in psoriasis patients treated with UVB therapy
List of papers
This thesis is based on the following papers, which will be referred to in the text by
their Roman numerals I-IV:
I Osmancevic A, Landin-Wilhelmsen K, Larkö O, Mellström D, Wennberg AM,
Hulthén L, Krogstad AL. UVB therapy increases 25(OH) vitamin D synthesis in
postmenopausal women with psoriasis. Photodermatol Photoimmunol Photomed 2007;
23(5): 172-8.
II Osmancevic A, Landin-Wilhelmsen K, Larkö O, Mellström D, Wennberg AM,
Hulthén L, Krogstad AL. Risk factors for osteoporosis and bone status in
postmenopausal women with psoriasis treated with UVB therapy. Acta Derm
Venereol. 2008; 88(3):240-6.
III Osmancevic A, Landin-Wilhelmsen K, Larkö O, Wennberg AM, Krogstad
AL.Vitamin D production in psoriasis patients increases less with narrowband than
with broadband ultraviolet B phototherapy. Photodermatol Photoimmunol Photomed,
2009 (in press).
IV Osmancevic A, Nilsen LT, Landin-Wilhelmsen K, Søyland E, Abusdal Torjesen
P, Hagve TA, Nenseter M, Krogstad AL. Effect of climate therapy at Gran Canaria
on vitamin D production, blood glucose and lipids in patients with psoriasis. J Eur
Acad Dermatol Venereol, 2009 (in press).
1
Contents
CONTENTS ........................................................................................................ 1
ABBREVIATIONS ............................................................................................. 4
INTRODUCTION .............................................................................................. 5
VITAMINS ............................................................................................................ 5
VITAMIN D .......................................................................................................... 5
Nomenclature
................................................................................................ 5
Structure
......................................................................................................... 6
Historical perspective
................................................................................... 7
Sunshine as a means of health ...................................................................... 7
The fat-soluble vitamin ............................................................................... 8
Light equals vitamin D .............................................................................. 9
Photosynthesis
............................................................................................ 10
Factors that influence on vitamin D photosynthesis ......................................... 12
How much sunlight is necessary to satisfy the body’s requirement? ..................... 12
Metabolism
.................................................................................................. 13
Levels of 25(OH)D ................................................................................. 14
Mechanism of action
.................................................................................. 15
Effects on autoimmune diseases .................................................................. 17
Effects on hypertension .............................................................................. 18
Antimicrobial effects ................................................................................. 18
Substitution
................................................................................................. 19
Conclusions
................................................................................................. 19
PSORIASIS .......................................................................................................... 20
Effects of vitamin D3 analogues in psoriasis
............................................ 22
2
Psoriasis and comorbidities
....................................................................... 23
Osteoporosis ........................................................................................... 25
PHOTOTHERAPY ................................................................................................ 25
Mechanism of action
.................................................................................. 27
AIMS OF THE INVESTIGATION ................................................................ 29
MATERIAL AND METHODS ....................................................................... 30
SUBJECTS ........................................................................................................... 30
ANTHROPOMETRY AND BONE MEASUREMENT (PAPER I AND II) ........................ 30
VITAMIN D ANALYSES ....................................................................................... 31
OTHER BIOCHEMISTRY ...................................................................................... 31
PROCEDURES OF UV EXPOSURE AND UV MEASUREMENT ................................. 32
CONCOMITANT MEDICATION ............................................................................ 36
QUESTIONNAIRES .............................................................................................. 36
ETHICAL CONSIDERATIONS ............................................................................... 36
STATISTICS......................................................................................................... 37
RESULTS ........................................................................................................... 38
COMPARISON BETWEEN THE STUDIES ................................................................ 38
PAPER I ............................................................................................................. 43
PAPER II ............................................................................................................ 44
PAPER III .......................................................................................................... 46
PAPER IV........................................................................................................... 47
DISCUSSION .................................................................................................... 49
SERUM 25(OH)D IN PSORIASIS PATIENTS DURING TREATMENT WITH
PHOTOTHERAPY (PAPERS I, III AND IV) ............................................................ 49
3
SERUM 1,25(OH)2D IN PSORIASIS PATIENTS DURING TREATMENT WITH
PHOTOTHERAPY (PAPERS I, III AND IV) ............................................................ 54
SERUM PTH IN PSORIASIS PATIENTS DURING TREATMENT WITH PHOTOTHERAPY
(PAPERS I, III AND IV) ....................................................................................... 57
BONE STATUS IN POSTMENOPAUSAL WOMEN WITH PSORIASIS TREATED WITH
UVB PHOTOTHERAPY (PAPER I AND II) ............................................................ 58
LIPID STATUS AND BLOOD GLUCOSE IN PSORIASIS PATIENTS DURING TREATMENT
WITH HELIOTHERAPY (PAPER IV) ...................................................................... 61
Dyslipidemia .......................................................................................... 61
Metabolic syndrome .................................................................................. 61
Hypertension .......................................................................................... 62
Insulin resistance/diabetes ......................................................................... 63
Role of vitamin D in the pathogenesis of type 2 diabetes mellitus ....................... 64
CONCLUSIONS ............................................................................................... 65
FUTURE PROSPECTS .................................................................................... 66
ACKNOWLEDGEMENTS .............................................................................. 67
REFERENCES ................................................................................................. 69
4
Abbreviations
7-DHC 7- dehydrocholesterol
D3 cholecalciferol
25(OH)D 25-hydroxyvitamin D (or calcidiol)
1,25(OH)2D 1,25-dihydroxyvitamin D (or calcitriol)
PTH parathyroid hormone
VDR vitamin D receptor
VDP vitamin D binding protein
UVR ultraviolet radiation
UVB ultraviolet radiation B
UVA ultraviolet radiation A
NBUVB narrowband ultraviolet radiation B
PASI Psoriasis Area and Severity Index
HRT hormonal replacement therapy
DEXA Dual-Energy X-ray Absorptiometry
BMD bone mineral density
BMI body mass index
LDL low density lipoprotein cholesterol
HDL high density lipoprotein cholesterol
TNF-α tumour necrosis factor-α
HbA1c haemoglobin A1c
PUVA Psoarlen+UVA
MED Minimal Erythema Dose
SED Standard Erythema Dose
5
Introduction
Vitamins
William Fletcher was the first scientist who in 1905 determined that diseases
occurred if special factors (vitamins) were removed from food. He was researching
the causes of the disease beriberi when he discovered that eating unpolished rice
prevented beriberi and eating polished rice did not. William Fletcher believed that
there were special nutrients contained in the husk of the rice.
At the same time biochemist Frederick Gowland Hopkins discovered that certain
factors in food were important to health. In 1912, Polish scientist Cashmir Funk
named the special nutritional parts of food as a "vitamine" after "vita" meaning
life and "amine" from compounds found in the thiamine he isolated from rice
husks. Vitamine was later shortened to vitamin.
Vitamins B, C and D were all discovered as a result of research into diseases -
beriberi, scurvy and rickets, respectively. Supplementing an imbalanced diet with
certain foods was shown to prevent each of these diseases. Subsequently,
purification and analysis of disease-preventing compounds in these foods
established a new class of nutrients - in addition to proteins, fats and carbohydrates
- that are now known to be essential for human health.
Vitamin D
Nomenclature
Cholecalciferol or vitamin D3 is produced in the skin as a result of ultraviolet
irradiation of 7-dehydrocholesterol (7-DHC). Ergocalciferol or vitamin D2 is
produced by ultraviolet irradiation of the plant sterol ergosterol. “Calciferol” refers
to both of these compounds.
6
Structure
The two forms of vitamin D (D2 and D3) differ chemically in their side chains.
These structural differences also alter their metabolism, but in general, the
biological activity of their active metabolites is comparable.1
The only structural difference between vitamin D2 and D3 is in their side chains.
The side chain for vitamin D2 contains a double bond between C-22 and C-23 and
a C-24 methyl group (Figure 1).
Figure 1: Structure of vitamin D3 and D2 and their respective precursors,
7-dehydrocholesterol, and ergosterol (MacLaughlin and Holick, 1983).
7
Historical perspective
Sunshine as a means of health
The discovery of vitamin D in food and the realization that the body can produce
vitamin D independently are both intimately tied to research into the cause and
prevention of childhood rickets.
There has been some evidence since early Greek and Roman times of the bone-
deforming disease commonly known as rickets. The first scientific description of a
vitamin D deficiency, namely rickets, was provided in the 17th century by both Dr.
Daniel Whistler (1645) and Professor Francis Glisson (1650) (Figure 2).
Figure 2:
D.Whistler; F.Glisson. A Treatise of the Rickets: Being a Disease Common to Children.
London: P. Cole, 1651
Rickets is a disease of young children with a constellation of physical signs and
symptoms including deformities of the skeleton, such as bowed legs, enlargement
8
of the epiphyses of the long bones and rib cage (rachitic rosary), deformed pelvis,
enlarged head, curvature of the spine, poor dentition, and weak flabby legs.2 During
the Industrial Revolution in Europe children developed this disease as a result of a
sunless environment in the polluted inner cites. An autopsy study of children who
died of various causes in Leyden, The Netherlands, in the end of the 18th century,
revealed that 80-90% of these children had evidence of rickets.
In 1822 Sniadecki discovered the importance of exposure to sunlight for the
prevention and cure of rickets. He observed that children living in the inner city of
Warsaw, Poland, had a higher incidence of rickets than children living in rural areas.
He encouraged direct exposure of the skin to sunlight as one most efficient
methods for the prevention and cure of rickets.3
Little attention, however, was given to the environment as a cause of rickets until
1889, when a British Medical Association investigative committee reported that
rickets was infrequently seen in rural districts of the British Isles but was prevalent
in large industrial towns.4 In 1890 Palm collected clinical observations from a
number of his colleagues throughout the British Empire and the Orient. He found
that rickets was widespread in the industrial centres of Great Britain, whereas in
impoverished cities in China, Japan, and India, where people received poor
nutrition and lived in squalor, the disease was rare.5 Based on this epidemiological
survey he urged “the systematic use of sunbaths as a preventive and therapeutic
measure in rickets and other diseases, and the education of the public to the
appreciation of sunshine as a means of health.” However, it was difficult at the
time for people to believe that such a simple remedy as exposure to sunlight could
cure this bone-deforming disease, and little was done to use these astute
observations for curing rickets.
The fat-soluble vitamin
In the early 1800s it was a common practice, supported by folklore, to give children
cod liver oil to prevent and cure rickets. In the 1890s scientists began to search for
specific foods that could prevent rickets. Such reasoning was rooted in the
knowledge that two other diseases, scurvy and beriberi, could be prevented by the
addition of certain foods (such as citrus fruits that contain vitamin C, and whole
grain rice that contains vitamin B1) to the diet. However, it was not until 1918 when
Mellanby reported that he could make beagles rachitic and reverse the bone disease
9
with cod liver oil that the scientific community began to consider rickets as a
nutritional-deficiency disease. In 1921 he wrote, "The action of fats in rickets is due
to a vitamin or accessory food factor which they contain, probably identical with
the fat-soluble vitamin." Originally it was thought that the antirachitic factor in cod
liver oil was vitamin A. However, McColum et al. demonstrated that the
antirachitic activity that he called vitamin D was separate from vitamin A. They
exposed cod liver oil to heat and oxygen - vitamin A activity was destroyed while
when maintaining the antirachitic activity. The vitamin in cod liver oil was
designated “D” as vitamins A, B, and C had already been identified.
Light equals vitamin D
At the same time that Mellanby was demonstrating that rickets could be cured by
the ingestion of cod liver oil, Huldschinsky was exposing rachitic children to
radiation from a mercury arc lamp. He reported dramatic reversal of rickets within
four months after ultraviolet radiation therapy from the mercury arc lamp. In 1921
Hess and Unger exposed seven rachitic children in New York City to sunshine and
reported, by X-ray examination, that there was marked improvement in each child’s
rickets. This research had equivocally shown that exposure to sunlight alone could
prevent and cure this crippling bone disease. This led Hess and Weinstock to
investigate independently the use of ultraviolet irradiation of food (including wheat,
lettuce, vegetable oils and animal feed) as a way of imparting antirachitic activity.6
Steenbock appreciated the use of this technique and introduced the concept of
irradiating milk as a means of preventing rickets in children.7 This led to the
fortification of milk with vitamin D, which helped eradicate rickets in the countries
that used this practice.
The chemical structures of the D vitamins were determined in the 1930s in
Professor A. Windaus’s laboratory at the University of Göttingen in Germany.
Vitamin D was first isolated from the irradiation of the fungal sterol ergosterol and
was designated vitamin D1. However, the product was an impure mixture, and the
term was dropped. Vitamin D2, which could be produced by ultraviolet irradiation
of ergosterol, was chemically characterized in 1932. Vitamin D3 was not chemically
characterized until 1936 when it was shown to result from the ultraviolet irradiation
of 7-DHC. Almost simultaneously, the elusive antirachitic component of cod liver
oil was shown to be identical to the characterized vitamin D3. These results clearly
10
established that the antirachitic substance vitamin D was chemically a steroid, more
specifically a seco-steroid (Figure 1).
Once the structure of vitamin D was characterised and a simple process was
developed for its synthesis, vitamin D was directly added to milk, thereby making
the ultraviolet irradiation of milk obsolete. Ergocalciferol (D2) is commercially
made by irradiating and then purifying the ergosterol extracted from yeast.
Cholecalciferol (D3) is produced commercially by extracting 7-DHC from wool fat,
followed by UVB irradiation and purification.
Photosynthesis
Vitamin D is produced in the skin from 7-DHC, the last precursor in cholesterol
synthesis.8 In adult human skin, approximately 50% of provitamin D3 (7-DHC) is
found in the epidermis and the other half is found in the dermis.8 During exposure
to sunlight, the high-energy UVB photons with energies between 290 and 315 nm,
penetrate into the skin where they are absorbed by provitamin D3 -the immediate
precursor in cholesterol biosynthetic pathway. UVB photons break the B ring of
the cholesterol structure, cleavage between carbons 9 and 10 to form a 9,10 –
secosteroid known as previtamin D3. Previtamin D3 also has the capacity to
absorb ultraviolet radiation. This results in its isomerisation to two photoproducts
known as lumisterol and tachysterol.9 Both of these photoisomers are inert in
calcium metabolism. During prolonged exposure to the sun, the accumulation of
previtamin D3 is limited to about 10 to 15% of the original 7-DHC content
because the previtamin photoisomerizes to lumisterol 3 and tachysterol 3.8
Previtamin D3 is biologically inert and thermodynamically unstable. As a result, its
double bonds rearrange spontaneously to form vitamin D3 (Figure 3). At a
physiological temperature of 37° C, this process would take approximately 24 to 48
hours to reach completion. However, it is now recognised that previtamin D3 is
rapidly converted to vitamin D3 in human skin. Previtamin D3 exists in two
isomeric forms known as the cis,cis and cis,trans forms. Only the cis,cis conformer can
be converted to vitamin D3. The cis,trans form is thermodynamically more stable
but cannot be converted to vitamin D3, thus accounting for the prolonged
isomerisation time. It takes time for the cis,trans form to isomerise to the cis,cis
conformer before it can be converted to vitamin D3. However, a unique
mechanism operates that allows the efficient conversion of previtamin D3 to
vitamin D3 within hours after its formation in the skin. The previtamin D3 is
11
sandwiched between fatty acids in the bilipid membrane.10 Vitamin D3 is also able
to absorb ultraviolet radiation. Exposure to ultraviolet radiation results in
isomerisation of vitamin D3 to form at least three photoproducts, known as 5,6-
trans vitamin D3, supersterol I, and supersterol II (Figure 3).11 None of these
vitamin D3 photoproducts has any effect on calcium metabolism at physiological
concentrations. Once vitamin D3 is made, it undergoes a conformational change
that allows it to move from the membrane to the extracellular space and eventually
to the dermal capillary bed. Since most of the ultraviolet B radiation is absorbed in
the epidermis, more than 70% of previtamin D3 synthesis occurs here.8 Aging
decreases the thickness of both the epidermis and dermis, therefore there is an age-
dependent decline in provitamin D3 8,12 resulting in a decreased capacity of the
elderly to produce vitamin D3 in the skin.13
Figure 3: Photosynthesis of vitamin D in the skin. DBP= Vitamin D Binding Protein
12
Factors that influence on vitamin D photosynthesis
Any process that decreases or prevents ultraviolet B photons from reaching the
viable epidermis to be absorbed by provitamin D3 results in a diminution in the
photosynthesis of previtamin D3. Melanin and sunscreens are effective in
absorbing UVB radiation. Thus, an increase in skin pigmentation can markedly
diminish the cutaneous production of vitamin D3.14 Thus heavily pigmented people
require at least 5 to 10 times longer exposure than Caucasians to produce adequate
vitamin D3 in their skin.14 The application of a sunscreen with a sun protection
factor (SPF) of 8 will reduce, by more than 95%, the cutaneous production of
cholecalciferol.15 Most clothing completely absorbs UVB radiation thus preventing
the cutaneous production of vitamin D3 on the areas it covers.16 It is well-known
that sunlight exposure through glass will not result in any significant vitamin D3
production. The reason being that there are substances in glass, including lead, that
absorb UVB radiation.17
The zenith angle of the sun has a dramatic effect on the total number of UVB
photons reaching the earth’s surface. During the winter, at latitudes above 40°
north and below 40° south of the equator, the UVB photons are efficiently
absorbed by the ozone layer, essentially eliminating the ability of the skin to
produce vitamin D3.18 At latitudes above and below 34° south and north
respectively, there is cutaneous production of vitamin D3 all year round. The
latitude of Sweden is 62º north of the equator and in this geographic area UVB is
not transmitted in sunlight from October to March.
How much sunlight is necessary to satisfy the body’s requirement?
This is an especially relevant question in view of concern about the damaging
effects of excessive exposure to sunlight. An adult Caucasian exposed to sunlight or
a (tanning bed) lamp (~32 mJ/cm2) that emits UVB radiation produces ~1 ng of
cholecalciferol/cm2 skin.2 A study was conducted where medical students received
a whole-body exposure to 1 minimal erythema dose (MED) of solar simulated
sunlight together with graded doses of vitamin D. The circulating levels of vitamin
D were subsequently measured at various intervals. It was found that a whole-body
exposure to 1 MED of UVB radiation resulted in a blood level of vitamin D that
was comparable to taking between 10,000 and 25,000 IU of vitamin D orally.17
13
Metabolism
It has been estimated that between 90 and 95% of all people obtain their vitamin D
requirement from exposure to sunlight.17 The vitamin D3 produced in the skin or
ingested from the diet can be stored in body fat and released into the circulation at
during times when there is an inadequate cutaneous production of vitamin D3. In
obese children and adults, the cholecalciferol is sequestered deep in the body fat,
making it less bioaviable.2 Thus, obese individuals are only able to increase their
blood levels of vitamin D by approximately 50% compared with normal-weighted
individuals.19 However, vitamin D is biologically inert and must be metabolized in
the liver on carbon 25 to form the major circulating form of vitamin D, 25-
hydroxycholecalciferol (25(OH)D) or calcidiol, (Figure 4).
UVB
Cholecalciferol
(Vitamin D
3)
7-Dehydrocholesterol
Liver
25-hydroxyvitamin D
3
Kidney
1,25-dihydroxyvitamin D
3
Vitamin D
3
Vitamin D
2
Dietary intake
Maintains calcium balance
In the body
Skin
Figure 4: Metabolism of vitamin D
14
25(OH)D is used clinically to measure vitamin D status for vitamin D deficiency,
sufficiency, and intoxication.17 Because 25(OH)D is biologically inert at
physiological concentrations, it must be converted in the kidney to its activated
form 1,25 dihydroxycholecalciferol [1,25(OH)2D] or calcitriol, (Figure 4).
Vitamin D-binding protein (DBP), a 458-amino acid polymorphic human serum
protein, is the major plasma carrier of vitamin D3 and of all its metabolites, which
include 25(OH)D and 1,25(OH)2D.20
The skin occupies a central position within the vitamin D system. Epidermal
keratinocytes also express the vitamin D hydroxylase enzymes 25-hydroxylase
(CYP27A1) and 1α-hydroxylase (CYP27B1), enabling them to convert vitamin D3
into 25(OH)D and 1,25(OH)2D, the biologically active form of vitamin D3.21 In
addition, keratinocytes are vitamin D target cells as they contain the vitamin D
receptor (VDR) and respond to 1,25(OH)2D with changes in proliferation,
differentiation and cytokine production.22 Taken together, these findings indicate
the existence of a unique photoendocrine vitamin D system in keratinocytes - this
is corroborated by the demonstration of 1,25(OH)2D synthesis and vitamin D
effects in UVB-irradiated skin or keratinocytes.21,23 In contrast to epidermal
keratinocytes, dermal fibroblasts only have the capacity for photoproduction of
25(OH)D but not 1,25(OH)2D.24 25(OH)D may then act as a paracrine factor to
keratinocytes. Monocytes, like keratinocytes, have the full machinery to
photoproduce 1,25(OH)2D.23
The physiologic significance of the cutaneous photosynthesis of 1,25(OH)2D is
poorly understood. As the amounts of UVB-produced 1,25(OH)2D in
keratinocytes are small23 and circulating 25(OH)D and 1,25(OH)2D are hardly
detectable in hepatectomized or nephrectomized animals25, cutaneous production
of active vitamin D does not appear to play a major role outside the skin. However,
locally produced 1,25(OH)2D may contribute to UVB effects within the skin, such
as its therapeutic action on psoriasis.26 Furthermore, photoproduced 1,25(OH)2D
might serve as an endogenous protection mechanism against UVB-dependent
DNA damage, apoptosis and release of proinflammatory cytokines such as IL-6.27,28
Levels of 25(OH)D
A 25(OH)D level less than <30 ng/ml (75 nmol/l) is considered to be suboptimal
vitamin D status - this is the minimal level of 25(OH)D necessary to suppress
15
parathyroid hormone secretion.29-31 A 25(OH)D level of between 21 ng/ml (52
nmol/l) and 29 ng/ml (74 nmol/l) is considered to be vitamin D insufficiency.31-33
The cut-off level for serum 25(OH)D, which is taken as a diagnostic value for
vitamin D deficiency, has varied over the years.33-35
Mechanism of action
1,25(OH)2D interacts with its nuclear VDR, which in turn binds with the retinoic
acid-X-receptor. This complex is recognised by specific gene sequences known as
the vitamin D responsive elements (VDRE) to unlock genetic information that is
responsible for its biologic actions. In the intestine 1,25(OH)2D induces the
expression of an epithelial calcium channel, calcium-binding protein (calbindin),
and a variety of other proteins to help the transport of calcium from food into the
circulation.36 1,25(OH)2D also interacts with the VDR in the osteoblast and
stimulates the expression of receptor activator of NFκβ ligand (RANKL) similar to
PTH.37 Thus, 1,25(OH)2D maintains calcium homeostasis by increasing the
efficiency of intestinal calcium absorption and mobilizing calcium stores from the
skeleton.
PTH, hypocalcaemia, and hypophosphatemia are the major stimulators for the
renal production of 1,25(OH)2D.37 During pregnancy, lactation, and growth sex
steroids, prolactin, growth hormone, and insulin-like growth factor 1 (IGF-1) play a
role in enhancing the renal production of 1,25(OH)2D to satisfy increased calcium
needs. 37
Vitamin D deficiency results in a decrease in the efficiency of intestinal absorption
of dietary calcium and phosphorus.37 This causes a transient lowering of the ionized
calcium, which is immediately corrected by the increased production and secretion
of PTH. PTH sustains the blood-ionized calcium by interacting with its membrane
receptor on mature osteoblasts, which induces the expression of RANKL.37 This
plasma membrane receptor protein is recognised by RANK that is present on the
plasma membrane of preosteoclasts. The intimate interaction between RANKL
and RANK results in increased production and maturation of osteoclasts.37,38
Osteoclasts release hydrochloric acid and collagenases to destroy bone, resulting in
the mobilization of calcium stores from the skeleton. Thus vitamin D deficiency
induced secondary hyperparathyroidism results in skeletal wasting that can
precipitate and exacerbate osteoporosis.2 Vitamin D deficiency and attendant
16
secondary hyperparathyroidism also causes loss of phosphorus into the urine and
lowering of serum phosphorus levels. This results in an inadequate calcium x
phosphorus product, causing poor or defective mineralization of the bone matrix
laid down by osteoblasts.2 In children, the effect of body weight and gravity on a
poorly mineralized skeleton results in the classic bony rachitic deformities in the
lower limbs (bowed legs and knocked knees). Adults have enough mineral in their
skeleton to prevent skeletal deformities. However, in vitamin D deficient state, the
newly laid-down osteid cannot be properly mineralised, leading to osteomalacia.
Unlike osteoporosis, which is a silent disease until fracture occurs, osteomalacia is
associated with either widespread or localised throbbing bone pain. The likely cause
is that the unmineralised osteoid becomes hydrated and provides little support for
the sensory fibres in periosteal covering.39 Osteomalacia cannot be distinguished
from osteoporosis/osteopenia by neither X-ray analyses nor by bone densitometry
- they appear identical.
Besides the small intestine and the osteoblast, VDR has been identified in almost
every tissue and cell in the body, including brain, heart, skin, pancreas, breast, colon
and immune cells.37,40 1,25(OH)2D helps regulate cell growth and maturation,
stimulates insulin secretion, inhibits renin production, and modulates the functions
of activated T and B lymphocytes and macrophages.37,41
It is documented that risk of morbidity or mortality from colon, prostate, breast,
ovarian, oesophageal, non-Hodgkin’s lymphoma, and a variety of other aggressive
cancers is related to living at higher latitudes and being at higher risk of vitamin D
deficiency.42-45 Initially the explanation for why increased sun exposure decreased
the risk of fatality from common cancers was due to the increased production of
vitamin D in the skin, leading to the increased production of 1,25(OH)2D in the
kidneys.42 Because it was known that the VDR existed in most tissues in the body
and that 1,25(OH)2D was a potent inhibitor of both normal and cancer cell
growth10,37 it was assumed that the increased renal production of 1,25(OH)2D could
downregulate cancer cell growth and therefore mitigate the cancer’s activity and
decrease mortality. However, it was also known that the production of 1,25(OH)2D
in the kidneys was tightly controlled and that increased intake of vitamin D or
exposure to sunlight did not result in an increase in circulating concentrations of
1,25(OH)2D.37 However, this still did not explain the anti-cancer effect of sunlight -
vitamin D concentration. The skin not only makes cholecalciferol, but it also has
the enzymatic machinery to convert 25(OH)D to 1,25(OH)2D, similar to activated
17
macrophages.46,47 In 1998 Schwartz et al.48 reported normal and malignant prostate
cancer cells also had the enzymatic machinery to make 1,25(OH)2D. Increased
exposure to sunlight or vitamin D intake leads to increased production of
25(OH)D. Higher concentrations of 25(OH)D are used by prostate cells to make
1,25(OH)2D, which help keep prostate cell proliferation in check and therefore
decreases the risk of malignancy.37 It has since been observed that breast, colon,
lung, brain and a wide variety of other cells in the body are able to produce
1,25(OH)2D.37,49-53 Thus it has been suggested that raising blood levels of
25(OH)D provides most of the body’s tissues with enough substrate to make
1,25(OH)2D locally to act as a sentinel to help control cellular growth and
maturation and decrease the risk of malignancy.37 Both prospective and
retrospective studies revealed that, if the 25(OH)D level is at least 20 ng/ml, than
there is an approximate 30-50% decreased risk of developing and dying of colon,
prostate, and breast cancers.37,42,45,54
There are several studies suggesting that increased exposure to limited amounts of
sunlight decreases the risk of developing and dying of the most deadly form of skin
cancer, melanoma.55,56
Effects on autoimmune diseases
Activated T and B lymphocytes, monocytes, and macrophages have VDR.41,53,57,58
1,25(OH)2D interacts with its VDR in immune cells and has a variety of effects on
regulating lymphocyte function, cytokine production, macrophage activity, and
monocyte maturation.37,41,53,58,59 Thus, 1,25(OH)2D is a potent immunomodulator.
Insights into the important role of vitamin D in the prevention of autoimmune
diseases have come from a variety of animal studies. Nonobese diabetic mice that
typically develop type I diabetes by 200 days reduced their risk of developing this
disease by 80% when they received a physiological dose of 1,25(OH)2D daily.60
Mice that were pretreated with 1,25(OH)2D before they were injected with myelin
to induce a multiple-sclerosis-like disease were immune from it.61 Similar
observations were made in a mouse model that develops Crohn’s disease.62 These
animal model studies have given important insights into the role of 1,25(OH)2D in
reducing the risk of developing common autoimmune diseases such as multiple
sclerosis63 and rheumatoid arthritis.64 Most compelling is the observation that
children in Finland who received 2000 IU of vitamin D daily from one year and
were followed up for the next 25 years had an 80% decreased risk of developing
18
type I diabetes, whereas children who were vitamin D deficient had a four-fold
increased risk of developing this disease later in life.65
Effects on hypertension
In 1997 Rostand 66 reported that people living at higher latitudes throughout the
world were at higher risk of developing hypertension. He suggested that this may
be related to being more prone to developing vitamin D deficiency. To determine
the possible link between sun exposure and the protective effect in preventing
hypertension, Krause et al. 67 exposed a group of hypertensive adults to a tanning
bed that emitted light and UVB and UVA radiation similar to summer sunlight. A
similar group of hypertensive adults was exposed to a similar tanning bed that
emitted light and UVA radiation similar to winter sunlight, i.e., no UVB radiation.
All subjects were exposed 3 times a week for 3 months. After 3 months, it was
observed that hypertensive patients who were exposed to the tanning bed that
emitted UVB radiation had a 180% increase in their circulating concentrations of
25(OH)D. There was no change in blood levels of 25(OH)D in the comparable
group that was exposed to the tanning bed that did not emit UVB radiation. The
patients who had increased 25(OH)D levels also had a decrease of systolic and
diastolic blood pressures by 6 mm Hg, bringing them into the normal range. The
placebo group (exposed to light that did not emit UVB radiation) did not change
their 25(OH)D levels and no effect was observed on their blood pressure.
It has also been observed that patients with cardiovascular heart disease are more
likely to develop heart failure if they are vitamin D deficient.68 The exact
mechanism responsible for vitamin D sufficiency protecting against cardiovascular
heart disease is not fully understood. It is known that 1,25(OH)2D is one of the
most potent hormones for downregulation of the blood pressure hormone renin in
the kidneys.69 Furthermore vascular smooth muscle cells have a VDR that in the
presence of 1,25(OH)2D induces relaxation and thereby vasodilatation.70,71
Antimicrobial effects
In addition to phagocytes and cytokines, a major component of the innate immune
system is a diverse combination of cationic peptides that include the α- and β-
defensins and cathelicidins, which have potent microbicidal activities at low
concentrations.72
19
The antimicrobial peptide cathelicidin is a vitamin D target gene,73 and induces
upregulation of CYP27B1 and VDR in monocytes. Cathelicidin expression was also
shown to be increased in human skin in vivo by topical application of active vitamin
D compounds such as calcipotriol and 1,25(OH)2D3.74
Cathelicidin plays also the role in wound healing and inflammatory skin disease.75
Cathelicidin is not only an effector molecule of innate immunity by its antimicrobial
activity but it also exhibits biological activities on adaptive immunity, angiogenesis
and cell proliferation and migration.72 Therefore, its function is pivotal for wound
repair.76 In addition to its upregulation in the skin following cutaneous injury, high
expression of cathelicidin and of antimicrobial peptides has also been noted in
psoriasis, accounting for the rare occurrence of skin infections in this condition.72
Substitution
Today, companies such as Hoffmann-La Roche and BASF produce large quantities
of the primary form of vitamin D, also known as vitamin D3 or cholecalciferol.
Cholesterol from animal products (such as lanolin from sheep wool) is purified and
used as starting material for purification of the precursor 7-DHC, which is then
converted into vitamin D3 by irradiation. This synthetic vitamin D3 is added to
many foods, particularly milk products; it is also a key ingredient in the
multivitamin supplements that many people take regularly.
Doses of calciferols are often expressed in international units (IU), there being 40
IU/µg of cholecalciferol and 38.8 IU/µg of ergocalciferol.77
Conclusions
It has been estimated that the body requires daily 3000-5000 IU of vitamin D.2,78
The most likely reason for this is that essentially every tissue and cell in the body
has a VDR and thus a requirement for vitamin D.2 Vitamin D is critically important
for the maintenance of calcium metabolism and good skeletal health throughout
life.79,80 The revelations that vitamin D regulates the immune system, controls
cancer cell growth and regulates the blood pressure hormone renin provides an
explanation for why vitamin D sufficiency has been observed to be so beneficial in
the prevention of many chronic illnesses that plague both children and adults.
People of darker skin colour are more prone to vitamin D deficiency at Northern
20
latitudes. Vitamin D deficiency has been recognised even in some of the sunniest
climates, including Saudi Arabia and India.81,82
Thus, vitamin D status has such important health implications that a measurement
of 25(OH)D should be part of a routine physical examination for children and
adults of all ages.
In the absence of sun exposure, 1000 IU of cholecalciferol a day is necessary to
maintain a healthy blood level of 25(OH)D of between 32 and 40 ng/ml (80 -100
nmol/l).2 Increasing the intake of vitamin D fortified foods and increasing fatty fish
consumption will help satisfy the requirement for vitamin D in the absence of
exposure to sunlight. Multivitamins typically contain 400 IU of vitamin D. Thus,
diet plus supplementary vitamin D can result in attaining the recommended 1000
IU of cholecalciferol.
It has been estimated that exposure to sunlight for usually no more than 5-15
minutes per day (between 10 am and 3 pm) on the limbs or hands, face and arms
during the spring, summer, and autumn (not during the winter unless located below
35° north) provides the required 1000 IU of cholecalciferol.2 Limited exposure
should be followed by the application of a broad spectrum sunscreen with an SPF
of at least 15 to prevent damaging effects due to excessive exposure to sunlight and
to prevent sun burning.2
Psoriasis
Psoriasis is a chronic, non-contagious skin disease characterized by red, inflamed
cutaneous lesions covered with silvery-white scale. The term psoro comes from the
Greek word for itch; psoriasis corresponds with the term itchy. In 1841 Ferdinand
Hebra, a Viennese dermatologist, was the first to ascribe the name 'psoriasis'. He
described the clinical picture of psoriasis that is used today. The hereditary factor of
psoriasis had already been established by that time.
Psoriasis affects both genders equally and can occur at any age, although it most
commonly appears for the first time between the ages of 15 and 25. The prevalence
of psoriasis in Western populations is estimated to be around 2-3%. The disease
affects the skin and potentially the joints. The prevalence of psoriatic arthritis in
psoriasis patients is estimated to be between 25-31%.83
21
Both genetic and environmental factors are important in the aetiology of the
disease. Psoriasis can be triggered by certain external stimuli, for example, bacterial
infections or injuries.84 Genetic susceptibility factors that contribute to
predisposition to psoriasis are being identified85, but so far, the cause of disease is
not fully understood.
The disease is an immune-mediated disorder in which excessive reproduction of
skin cells is secondary to factors produced by the immune system. T-cells, dendritic
antigen-presenting cells (APC) and cytokine networks are recognized as playing a
major role in the pathogenesis of psoriasis.85 Dysregulation of T-cell APC
interactions and over expression of proinflammatory cytokines lead to the
characterised hyperproliferation and decreased differentiation of keratinocytes
expressed by the increased cell turnover in epidermis.85 It is not known what
initiates the activation of these cells. Such triggers activate the otherwise dormant
innate immunity in the skin.
In patients with psoriasis, innate immunity then becomes hyperactivated.
Pathomorphologically, this is represented by the activity of natural killer (NK) cells,
increased antigen presentation by Langerhans cells, an influx of CD4+ T cells,
neutrophils, an activation of tumour necrosis factor α (TNF-α)–releasing
macrophages86-88 and hyperprolifreation of keratinocytes. This activation leads
finally to the recruitment of effector T-helper cell type 1 (TH-1) cells. The
epidermis of patients with psoriasis responds in a typical pattern to T-cell activation
and cytokine production. TH-1 cells produce large amounts of interferon gamma
(IFN- ), TNF-α, and interleukin (IL) 2 and activate macrophages to secrete even
more TNF-α.88,89 TNF-α and IFN- , have been shown to play a key role in
psoriasis.90 IFN- is responsible for the epidermal hyperplasia.91 IFN- can trigger
psoriasis at injection sites and T cells in psoriatic plaques produce it in high
amounts. High TNF-α levels have been found in psoriatic skin as well as serum and
are considered to derive mainly from activated macrophages.86,92 Furthermore,
TNF-α and IFN- levels in serum correlate with disease activity of psoriatic
patients.93,94 Why the T cells become active and remain in that state is as yet unclear.
Chronic inflammation of psoriatic lesions suggests that there is an underlying
inborn “error” in the regulatory T-cell population and the persistence of a yet
unknown trigger that induces an exaggerated innate immune response.84 Recently it
has been shown that regulatory T cells are functionally deficient in patients with
psoriasis and not present in sufficient numbers in lesions to achieve a
22
downregulation of the hyperresponse.95 Furthermore, the basement membrane
zone of patients with psoriasis seems to be altered structurally in a way that basal
layers of keratinocytes can be more easily promoted to proliferate than usual,
particularly in the presence of the inflammatory T-cell cytokines.95,96
Disease management is dependent on severity, psychosocial effects and the
patient's lifestyle. The treatment consists of various local and systemic treatments.
Local treatments include creams and ointments containing tar, dithranol, salicylic
acid or vitamin D-related compounds (calcipotriol (Daivonex)®, calcitriol (Silkis)®
or tacalcitol (Curatoderm)®). Occasionally, corticosteroid-containing ointments are
used for a short time. Vitamin D3 analogues inhibit proliferation, induce terminal
differentiation of human keratinocytes and exhibit immunomodulating properties.97
Several studies have shown that calcipotriol as well as calcitriol and tacalcitol are
efficacious, safe and can be used on a long-term basis for psoriasis.98-101 Vitamin D3
analogues can be used in combination with phototherapy.102
Effects of vitamin D3 analogues in psoriasis
Psoriasis is characterized by keratinocyte hyperproliferation, abnormal keratinocyte
differentiation, and immune-cell infiltration into the epidermis and dermis. The
most common form of psoriasis is plaque psoriasis or psoriasis vulgaris. At the
molecular level, psoriasis lesions show a prominent loss of loricrin and filaggrin in
the suprabasal layers of the epidermis and abnormal overexpression of other
differentiation markers such as involucrin, transglutaminase I (TGase I), psoriasin,
migration inhibitory factor related protein-8, and skin-derived antileukoproteinase.
The expression of normal suprabasal keratins K1 and K10 is inhibited and replaced
by the expression of the hyperproliferative keratins K6 and K16.
The observations that keratinocytes and T cells express VDR and that 1,25-(OH)2D
is a potent stimulator of keratinocyte differentiation provided a reasonable basis for
the clinical use of VDR ligands for the treatment of psoriasis.103,104 The first clinical
evidence to support the use of vitamin D analogues was obtained fortuitously when
a patient treated orally with 1 -hydroxyvitamin D3 for osteoporosis showed
remarkable remission of psoriatic lesions.105 Subsequently, promising clinical results
were obtained in studies using oral 1 -hydroxyvitamin D3, oral and topical 1,25-
(OH)2D (calcitriol), and topical 1,24-(OH)2D. In these clinical trials, approximately
23
70–80% of the patients showed improvement, and complete clearance of the target
lesions was observed in 20–25% of patients.106
The antipsoriatic activity of VDR ligands could be attributed to their
differentiation, antiproliferative, and immunomodulatory properties. VDR ligands
exhibit multipronged effects in psoriatic lesions and affect the function of
keratinocytes, T cells, and APC. VDR ligands promoted differentiation and
inhibited the proliferation of keratinocytes.106,107 Differentiation of keratinocytes
results in the formation of a cornified envelope (CE) that provides the barrier
function of the skin. The expression of involucrin, a component of the CE, and
TGase I, the enzyme that cross-links the components of CE, was increased by
1,25(OH)2D and other VDR ligands.108 Treatment of keratinocytes with the
medium containing high calcium also stimulated keratinocyte differentiation by
increasing the expression of involucrin and TGase I. 1,25(OH)2D promoted
keratinocyte differentiation, increased the expression of calcium receptor in
keratinocytes109 and indirectly induced the expression of keratin 1, involucrin,
TGase I, loricrin, and filaggrin, which are required for CE formation. VDR ligands
decreased the expression/level of proinflammatory cytokines IL-2, IFN- , IL-6, and
IL-8110-113 in T cells, all of which play a role in cutaneous inflammation, and
proliferation of T lymphocytes and keratinocytes. Furthermore, topical calcipotriol
increased antinflammatory cytokine IL-10 and decreased IL-8 in psoriatic lesions
114, and 1,25-(OH)2D also increased the expression of IL-10 receptor in
keratinocytes.115
APCs or dendritic cells (DCs) also play an important role in psoriasis and
autoimmune diseases because they are involved in autoantigen presentation. It
appears that APCs are one of the major targets of 1,25-(OH)2D-mediated
immunosuppressive action and VDR ligands prevent the differentiation,
maturation, activation, and survival of DCs, leading to T cell hyporesponsiveness.116
1,25-(OH)2D also increased the expression of IL-10 and decreased the expression
of IL-12, two major cytokines that are involved in Th1-Th2 balance.117
Psoriasis and comorbidities
Psoriasis is considered a chronic and debilitating inflammatory disease associated
with serious comorbiditites.118,119 The chronic inflammation in psoriasis can
predispose patients to other inflammatory conditions. For example, individuals
24
with psoriasis are at increased risk for insulin resistance, obesity, dyslipidemia, and
hypertension - components that characterize the metabolic syndrome. The
metabolic syndrome is an important driver of adverse cardiovascular outcomes. It
is likely that proinflammatory cytokines, such as TNF-α, and other factors that are
overproduced in patients with psoriasis likely contribute to the increased risk for
development of metabolic syndrome.120,121
The high prevalence of atherosclerosis is also reported in psoriasis patients. In the
pathogenesis of this phenomenon high serum lipid levels have been suggested.
High serum lipid levels are common in psoriasis and may be responsible for an
elevated prevalence of cardiovascular events in these patients.122 It may be useful to
perform early screening and treatment of hyperlipidaemia in psoriasis to prevent
atherosclerosis and its complications. Inflammation plays a key role in the
pathogenesis of psoriasis and a number of chronic inflammatory systemic diseases
listed above. Activated inflammatory cells and pro-inflammatory cytokines
contribute to the development of psoriatic lesions and play an important role in the
breakdown of atherosclerotic plaques. Psoriasis and atherosclerosis also have
similar histological characteristics involving T cells, macrophages and monocytes.
In particular, extravasation of T cells through the epithelium is characteristic of
both psoriatic and atherosclerotic plaques.
Inflammatory factors have also been associated with insulin resistance and β-cell
failure, both are key features of type 2 diabetes mellitus.123 There is evidence that
vitamin D may stimulate pancreatic insulin secretion directly. Vitamin D exerts its
effect through nuclear receptors that are found in a wide variety of tissues,
including T and B lymphocytes, skeletal muscle, and the pancreatic islet β-cells.124
The stimulatory effects of vitamin D on insulin secretion may be manifest only
when calcium levels are adequate.124 There is some evidence that increased PTH
activity is associated with, and possibly causes, reduced insulin sensitivity.124 The
prevalence of impaired glucose tolerance and diabetes mellitus is increased in
patients with primary hyperparathyroidism.125,126
Vitamin D has a wide range of effects on the immune system: it promotes the
differentiation of monocytes into macrophages thus increasing their cytotoxic
activity; reduces the antigen-presenting activity of macrophages to lymphocytes;
prevents dendritic cell maturation; inhibits T lymphocyte-mediated
immunoglobulin synthesis in B cells and inhibits delayed-type hypersensitivity
25
reactions.34,127,128 In contrast, vitamin D exerts an antiproliferative effect on
activated lymphocytes while suppressing the generation and activity of new NK
cells.129,130 Furthermore vitamin D has been reported to downregulate the
production of several cytokines: IL-2, IL-6 and IL-12, IFN- , TNF-α, and TNF-
β.128,131 Alternations in vitamin D status and/or action may affect insulin sensitivity,
β-cell function or both. Furthermore, several vitamin D-relatid genes are associated
with different pathogenetic traits of the disease. Therefore, vitamin D and its
related metabolic and immune pathways may be involved in the pathogenesis of
type 2 diabetes mellitus at both environmental and genetic levels.123
Osteoporosis
Information about prevalence of osteoporosis among patients with psoriasis and
epidemiology of risk factors for osteoporosis in this group is rare. Previous studies
did not detect any differences in bone mineral density (BMD) between patients
with psoriasis and healthy controls.132 It is not known if psoriasis disease itself has
any impact on bone metabolism.
However, a previous study on psoriasis patients showed no evidence that patients
with chronic plaque psoriasis, despite risk factors, had low BMD, although the
subgroup with joint involvement appeared to be at higher risk of osteoporosis and
therefore required prevention.132 Reduced BMD has been linked to palmoplantar
pustular psoriasis.133 The existence of less severe periarticular osteoporosis is
considered possible but there is no data concerning the existence of systemic
osteoporosis in patients with psoriatic arthritis.134
Vitamin D is important for bone metabolism.135 Vitamin D deficiency thus
contributes to the pathogenesis of osteoporosis and hip fractures.136 The
supplementation strategy with calcium and vitamin D supplements is cost saving
for osteoporotic fracture.137
Phototherapy
It has been known for more than 2000 years that several skin diseases improve
upon exposure to the sun (heliotherapy), but the systematic investigation of
phototherapeutic modalities did not start until the beginning of the 20th century.
UV radiation that reaches the skin is either reflected or absorbed by structures of
the skin. UVC (<280 nm) is mostly absorbed in the stratum corneum, and UVA
(320-400 nm) shows deeper penetration than UVB (280-320 nm).139-143 Thus, UVB
26
is mainly absorbed by epidermal components including keratinocytes, melanin and
Langerhans' cells.144 Biological effects of UV radiation are generated through
interaction with absorbing molecules called chromophores. In the case of UVB, the
most important chromophores are proteins such as keratin, melanin, collagen and
elastin, urocanic acid, DNA and previtamin D.8,145-147
In 1903, Niels Finsen received the Nobel Prize for developing phototherapy as a
treatment for tuberculosis of the skin. Around the turn of the 19th century,
Sardemann used for the first time ever a carbon arc lamp to treat psoriasis in a
patient.148 In 1923, Alderson then described “heliotherapy in psoriasis”. He used a
quartz-jacketed mercury discharge lamp to treat his first patients.149 Three years
later Goeckerman150 demonstrated the beneficial effect of natural sunlight in
combination with tar for psoriasis vulgaris “Goekermann regimen”.151 In 1953,
Ingram initiated the combination of UVB radiation, dithranol and tar-bathing for
psoriasis.152 In 1974, the combination of oral psoralen intake and subsequent UVA
irradiation was reported by Parrish153 and Wolff154, publications that marked the
initiation of modern psoralen+UVA (PUVA) photochemotherapy for psoriasis.
Phototherapy is thus an old and established treatment modality for this disease.
According to Feldman et al.155, with regard to efficacy, safety and cost-
effectiveness, UVB phototherapy appears to be the best first-line treatment for the
control of generalized psoriasis. Data from Fischer & Alsins156 and Parrish &
Jaenicke157 subsequently showed that wavelengths around 311 nm provoke least
erythema while being most effective for clearing psoriasis. According to these
results a fluorescent bulb was developed (TL-01), emitting a major peak at 311 (±
2nm) and a minor peak at 305 nm. This treatment was later called narrowband
UVB (NBUVB) and following its introduction several studies were published on its
superior efficacy in phototherapy of psoriasis.158-161 There is a large body of
evidence indicating that NBUVB is more effective than broadband UVB as a
monotherapeutic agent in the treatment of psoriasis even in children.160,162-164.
Whereas broadband UVB is considered to be most effective close to the minimal
erythema dose (MED), NBUVB has also been shown to be effective in
suberythemogenic doses.165 However, Diffey166 could show in a mathematical
model that clearance of psoriasis plaques is achieved faster with higher MED rates.
For treatment of psoriasis with NBUVB, three rather than two or five radiations a
week are effective167,168 and low incremental regimens are sufficient according to
27
Wainwright et al.169, who showed this regimen to be as effective as high incremental
regimens but less erythemogenic.
Ultraviolet radiation clears psoriasis with varying efficacy depending on the
wavelength. Wavelength dependency was determined with monochromatic UV
radiation and resulted in the action spectrum for psoriasis as established by
Parrish.149,156,157 Wavelengths of less than 300 nm also achieve significant clearing of
psoriasis, but cause more side effects in terms of erythema and burning and are
thus less efficient.170 Further conclusions from this spectral analysis were that
throughout the UVB and UVA ranges, the daily dose to clear psoriasis is equivalent
to the MED.148 In other words, if a UV source emits the psoriasis action spectrum,
one MED per day would be the best dosage regimen to clear the patients
psoriasis.161 On the other hand the production of vitamin D is mediated by UVB
dose equivalent to one MED.
Mechanism of action
Phototherapy (broadband UVB, NBUVB and heliotherapy) are commonly used
treatment modalities for widespread psoriasis.
Ultraviolet radiation influences the pathologic immune response in psoriasis in
multiple ways. Firstly, it alters the deviated antigen presentation, which is a major
trigger in psoriasis lesions. Langerhans cell numbers decrease by 90% after 7 UV
treatment sessions.171 Furthermore the remaining dendritic cells acquire
cytoskeleton damage by photo-oxidative stress, and this reduces subsequently their
ability to express high numbers of costimulatory surface markers to efficiently
stimulate T cells. UV radiation also alters the cytokine secretion pattern of
macrophages at the site of inflammation and can diminish their numbers by
induction of apoptosis.172 Under the influence of UV radiation, they produce IL-10,
an important immunosuppressive cytokine that shifts the TH-1 environment back
toward a TH-2 setting. Furthermore, macrophages produce IL-15 under the
influence of UV radiation, a cytokine that recruits new T cells into the plaque.
These new inactivated T-cells replace the originally present, hyperactivated set of T
cells that become apoptotic under UV irradiation.173 T cells are already susceptible
to very low doses of UVB and are thus more or less a selective target of
photons.174
28
The downregulation of T-cell activity goes along with a decrease in IL-2, a general
cytokine of T-cell activation in skin after UV irradiation.175 This can partly be
attributed to another effect of UV radiation: among the new T-cells recruited into
the lesion are regulatory T-cells, and UVB and PUVA have been shown to induce
regulatory T-cell production and thus promote intrinsic immunologic control of
pathologic overreaction.95,176,177 Ultraviolet radiation, however, also acts on the
innate defence because it has been shown to switch off the activity of neutrophil
and causes a dose-dependent inhibition of NK cell function.178,179 Doses that easily
reach the dermis in clinical practice have been shown to diminish NK cell activity
by at least 50%.171 This effect is exerted through reactive oxygen species induced by
UV, in particular the superoxide anion (O2) plays a key role here.171
UV radiation increases the IL-1 receptor antagonist in the epidermis more than
inducing IL-1, a fact that strongly promotes keratinocyte differentiation and thus
contributes to restoring the disturbed epidermal microarchitecture of psoriasis.
Interestingly the effects on TNF-α are controversial. Ultraviolet B increases TNF-α,
whereas UVA radiation reduces it. Ultraviolet radiation therefore does not inhibit
this major cytokine of psoriasis pathology.180
The effects of UV radiation on psoriasis can be divided into 2 mechanisms;
immediate effects and delayed effects. Immediate effects are, for example, cell
membrane damage by lipid peroxidation, DNA damage, induction of cytoplasmatic
transcription factors, and isomerization of chromophores such as urocanic
acid.170,181-184 These effects are largely cytopathic and induce growth arrest or even
apoptosis. Delayed effects result from cells surviving the immediate effects of
photon bombardment and lead to a modulation of the psoriasis microarchitecture,
particular the TH-1–dominated immune response.166,170
Heliotherapy, natural sunlight, has been used as a UV source throughout history
long before modern phototherapy was developed. Heliotherapy has been shown to
be generally effective in clearing psoriasis.185 Climatotherapy comprises alternative
treatment methods employing the healing capacities of natural resources, including
sunlight, temperature, humidity, barometric pressure and air. No other skin disease
responds to heliotherapy as dramatically as psoriasis vulgaris.
29
Aims of the investigation
The overall aims of the 4 studies were to 1) increase the knowledge about the
effects of UVB on vitamin D production during treatment with phototherapy and
heliotherapy in patients with psoriasis; 2) identify any differences between the
effect of NBUVB and broadband UVB on vitamin D synthesis in this group of
patients; and 3) investigate the effects of UVB-induced vitamin D synthesis on
bone, lipid and carbohydrate status in psoriasis patients.
Paper I. The aim was to examine whether broadband UVB therapy was capable of
inducing vitamin D synthesis, thereby supplying psoriasis patients with vitamin D.
Paper II. The aim was to investigate bone mineral density (BMD) and factors
contributing to osteoporosis in a group of postmenopausal women with psoriasis
who had been exposed to UVB therapy versus an age-matched control population.
Paper III. The aim was to study whether the effect of NBUVB on vitamin D
synthesis differed from the effect of broadband UVB in this respect.
Paper IV. The aim was to investigate the effect of climate therapy on vitamin D
synthesis, blood glucose and lipids, vitamin B12, C reactive protein and
haemoglobin in patients with psoriasis.
30
Material and methods
Subjects
White, Caucasian patients with active plaque psoriasis were included in the studies
(Paper I–IV). Clinical characteristics of the subjects included in the different studies
are presented in Table 1.
Study
Number
of
patients
Sex
M/F
Age (year)
(mean±SD)
PASI
score
before
PASI
score
after
Age at onset
of psoriasis
(mean±SD)
Paper
I
24 0/24 69 ± 5.9 6-12
(range)
1-4
(range)
34.0 ± 23
Paper
II
35 0/35 69.3 ±6.3 37.0 ±23.5
Paper
III
68 51/17 54.1±16.0 9.0±4.7 2.6±1.6 26.3 ± 14.2
Paper
IV
20 14/6 47.2±10.7 9.8±4.5 2.4±1.7
Table 1: Clinical characteristics of the psoriatic subjects included in study I–IV of this Thesis.
PASI (Psoriasis Area and Severity Index)
Anthropometry and bone measurement (paper I and II)
Body weight and height were measured in underwear and without shoes to the
nearest 0.5 kg and cm, respectively. Body mass index (BMI) was calculated as body
weight divided by height squared (kg/m2), measured at the time of the Dual-Energy
X-ray Absorptiometry (DEXA) examination.
Bone mineral density (BMD) was examined by DEXA with Hologic Delphi A at
the hip and the lumbar spine after UVB therapy. BMD results were recorded as the
T-score (SD from the mean peak value in young sex-matched adults) and the Z-
31
score (difference in SD from the mean of a healthy, age- and sex-matched sample)
for each subject. The Z-score was adjusted in the BMD measurements from
population data and inlaid as a control value in Hologic Delphi A.
Vitamin D analyses
Paper I and III
Serum samples for 25(OH)D and 1,25(OH)2D were obtained at baseline and after
the last dose of radiation. Serum 25(OH)D and 1,25(OH)2D were measured using
the 125I (radioimmunoassay) RIA method (DiaSorin, Stillwater, MN, USA). A
serum 25(OH)D concentration of 30 ng/ml was considered as vitamin D
insufficiency (Paper I).
Paper IV
Similar vitamin D methods were used but analysed at the Hormone Laboratory,
Aker University Hospital, Oslo, Norway. The serum concentrations of 25(OH)D
and 1,25(OH)2D were measured by RIA and competitive RIA, respectively
(DiaSorin, Stillwater, MN, USA).
Other biochemistry
Paper I and III
Blood samples for creatinine, calcium and intact parathyroid hormone (PTH) were
obtained at baseline and after the last dose of radiation. Serum PTH was measured
using the immunochemical luminescence method (mass concentration), serum
calcium by photometry, 600 nm and serum creatinine by using an enzymatic
method (µmol/l). The serum concentrations of thyroid hormones, and osteocalcin,
calcium and creatinine were measured before the first and after the last dose of
radiation (Paper I). Thyroxine hormone (free T4) and thyroid-stimulating hormone
(TSH) were examined by electrochemical luminescence ECLIA (nmol/l) and
(mU/l) respectively. Serum osteocalcin was examined using an immunochemical
radio immunoassay (µg/l).
32
Paper IV
The serum concentrations of calcium, ionised calcium (Ca++), PTH, plasma folate,
homocysteine, vitamin B12, erythrocyte haemoglobin (EHb), erythrocyte folate
(EFo), erythrocyte hematocrit (EHct), C reactive protein (CRP) and micro CRP
(mCRP) were obtained before the sun exposure, after one day and after 15 days of
exposure. The serum concentrations of creatinine, glucose, Apolipoprotein A1
(APO-A1), Apolipoprotein B (APO-B), Lipoprotein A (Lp(a)), total cholesterol,
high-density lipoprotein cholesterol (HDL), low-density lipoprotein cholesterol
(LDL), triglyceride and glycosylated haemoglobin A1c (HbA1c) were analysed using
routine laboratory methods, (Department of Medical Biochemistry, Rikshospitalet,
Oslo, Norway) before and after 15 days of sun exposure.
Procedures of UV exposure and UV measurement
Paper I
Patients were treated with broadband UVB (280-320 nm, PhilipsTL12 and Corona
4, ESSHÅ, Elagentur, AB, Värnamo, Sweden) 2 to 3 times/week for 8 to 12 weeks
(mean number of treatments 23.3±5.5) The radiation source was a fluorescent
cabinet that enclosed subjects, consisting of fluorescent tubes mounted in a round
assembly, placed at a distance of 30 cm apart from the body surface of the standing
subject (Figure 5). The irradiance was measured by PUVA Combi Light (ESSHÅ,
Elagentur, AB, Värnamo, Sweden).
The subjects were exposed (whole body) to individually adjusted doses of UVB
depending on skin phototype and erythemal response to therapy. The median
cumulative UVB dose during the series of exposures was 0.9 J/cm2 (range 0.4 to
5.8 J/cm2).
Paper III
The patients were previously treated with UV therapy. The selection of UV lamp
was based on patients’ previous experience of the respective broadband or
narrowband therapy. They were treated with the same lamp (NBUVB or TL12)
throughout the study. Twenty-six patients were treated with broadband UVB (280-
320 nm, Philips TL12 and Corona 4, ESSHÅ, Elagentur, AB, Värnamo, Sweden)
33
and 42 patients were treated with NBUVB (311-312 nm, Corona 4, ESSHÅ,
Elagentur, AB, Värnamo, Sweden).
Figure 5: Psoriasis. Treatments with UVB lamp.
All patients were treated according to standardised schedule adjusted to each
patient 2 to 3 times/week for 8 to 12 weeks. The patients reached approximately
the same outcome with similar total number of treatments (mean number of
treatments 27.2±5.3 in the broadband UVB treated group and 28.1±5.6 in the
NBUVB treated group).
The radiation source consisted of fluorescent tubes mounted in a round assembly
and placed at a distance of 30 cm apart from the body surface of the standing
subject. Irradiation was measured by PUVA Combi Light (ESSHÅ, Elagentur, AB,
Värnamo, Sweden).
Subjects were exposed (whole body) to individually adjusted doses of UVB
depending on skin phototype and erythemal response to therapy. The median
cumulative UVB dose during the series of exposures in the group treated with
broadband UVB was 12 J/cm2 and in the group treated with NBUVB 37 J/cm2.
Paper IV
The study was carried out during 15 days in March 2006 at Valle Marina Treatment
Centre, Gran Canaria (27oN, 15oW) (Figure 6). The patients followed a strict
exposure schedule the first day of the study, exposing first the front side of the
body for 30 minutes, the back side for 30 minutes, followed by 15 minutes
exposure of each side during the period between 11 am to 1 pm local time. They
were allowed to stay outside after lunch, when an adequate amount (2 mg/cm2) of
34
sunscreen with sun protection factor (SPF) of 25 (Pediatrics Photoprotector ISDN,
25B-10A-IR) was used for the whole body.186 For remaining days patients were
advised to gradually increase the hours of exposure per day, and to limit the use of
sunscreen to parts of the body that are susceptible to burning.
Figure 6: Valle Marina
The patients registered time spent in the sun every day per 20 minutes from 9 am
to 5 pm local time, as well as the use/amount of sunscreen and SPF factor. Spectral
UVB (280–315 nm), UVA (315–400 nm) and CIE-weighted UV irradiances were
measured every hour using 2 broadband instruments (Solar Light Co PMA 2100
with UVB sensor PMA 2101 and UVA sensor PMA 2110, and Gigahertz-Optik
GmbH X1 1 Optometer with UVB and UVA sensors XD-501).The CIE-action
spectrum is a reference action spectrum for UV induced erythema in Caucasian
human skin, valid for the UV region from 250–400 nm.187 Sensors were calibrated
and intercompared against a spectroradiometer (Brewer#185, measurement range
286.5–365 nm, extended for UVA 365–400 nm) at Izaña, Tenerife, prior to the
study (by Mr. Alberto Redondas, Instituto Nacional de Meteorologia (INM), Spain)
according to internationally accepted procedures (Figure 7).188,189 The overall
35
measurement uncertainty can be estimated within ±25%, and is due to the
uncertainty in different instruments, temperature variations, azimuth variations and
non-ideal cosine response in broadband sensors. Spectral UVB and UVA
irradiances, in addition to CIE-weighted UVB and UVA irradiances, were
calculated for the whole period using a radiation transfer model, libRadtran for
irradiance calculations.190
Figure 7: Calibrated broadband CIE-weighted UVB and spectral UVA-probes against
spectroradiometer at Izaña, Tenerife, at the INM (Instituto Nacional de Meteorologia España),
by Alberto Redondas
The model was run for the following conditions: cloudless sky, albedo of 0.05, sea
level and ozone values from the TOMS satellite.191 The UV irradiances were
adjusted according to measurements taken at the treatment centre to account for
the real weather situation and possible discrepancies from model parameters, such
as different albedo and aerosol amount. UV doses were estimated for each patient
after 1 day and after 15 days of sun exposure by combining calculated UV
irradiances with sun exposure time from patients’ diaries.
Results are presented as spectral UVB and UVA doses, as well as CIE-weighted
UV doses in Standard Erythema Dose (SED) (1 SED = 100 J/m2 = 0.01 J/cm2).
UV doses for each patient were set equal to the ambient UV doses divided by two,
since only half the body can be exposed at any time. Doses on the first day
36
excluded exposure time when sunscreen is used, since patients reported using
approximately the proper amount of sunscreen after lunch (30 ml, SPF 25).
All exposure time is included for remaining days since patients reported using small
amounts of sunscreen only on easily burned locations.
Concomitant medication
The patients were without any psoriasis medication for 4 weeks prior to the
interventions in study I, III and IV.
Questionnaires
Medical, family and gynaecological history, nutrient intake, physical activity, history
of psoriasis, previous psoriasis treatment and sun exposure in Paper II were
obtained with questionnaires. Previous (at the age of 25 years) body height and
weight were asked for.
Exposure to sun during the summer was graded from 1 to 4, grade 1 was sun
exposure <½ h/day, grade 2 was sun exposure ½–1 h/day, grade 3 was sun
exposure 1–2 h, and grade 4 was defined as sun exposure >2 h/sunny summer day.
Time spent outdoors per day (independent of sun exposure) was registered and
graded from 1 to 4 in a similar way.
Travel to sunny countries in the last two and ten years respectively was analysed
and graded from 0 to 4. Grade 0 was no travel to sunny countries, grade 1 was 1-4
travels, grade 2 was 5-10 travels, grade 3 was 10-20 travels and grade 4 was >20
travels in the last 10 years.
Ethical considerations
Each patient was given written information about the aim of the study. The study
was approved by the Ethics Committee at the University of Gothenburg and the
Swedish National Data Inspection Board (Paper I, II and III) and Regional
Committee of Medical Ethics and the Norwegian National Data Inspection Board
(Paper IV). Declaration of Helsinki protocols was followed and the written
informed consent of patients was obtained.
37
Statistics
Data are given as mean ± SD or median (min-max) unless otherwise stated. Simple
descriptive statistics and univariate correlations were performed using the statistics
routines of software (Excel, Microsoft Inc, SPSS, Version 15).
Student’s paired t-test was used for comparisons of blood test results before and
after sun exposure. Associations between variables were tested by Pearson and
Spearman correlation analysis. Probability values (2-sided) were considered
significant at values of <0.05.
38
Results
Comparison between the studies
Phototherapy induced vitamin D production in patients with psoriasis.
Serum levels of 25(OH)D increased during the treatment with artificial UV
(broadband UVB (p<0.00001), NBUVB (p<0.0001)) and during the heliotherapy
(p<0.0001) (Table 2, Figure 8 and 9).
Postmenopausal
women treated
with broadband
UVB (Paper I)
Broadband
(Paper III)
Narrowband
(Paper III)
Heliotherapy
(Paper IV)
25(OH)D(ng/ml)
before
after
36.8 ±17.0
59.6±18.7
37.9±16.9
69.4±19.7
34.8±11.9
55.3±17.6
22.9±6.0
41.8±6.3
1,25(OH)2D (pg/ml)
before
after
53.2±17.0
61.7±11.0
59.4±16.8
66.5±19.3
62.1±25.6
62.0±19.1
58.6±16.8
73.1±23.7
PTH (ng/l)
before
after
63.3±26.2
48.4±17.3
31.6±17.2
23.3±12.8
33.7±17.7
29.2±14.1
41.8±17.3
38.6±12.9
Table 2: Changes in serum concentrations of 25(OH)D, 1,25(OH)2D and PTH in psoriasis
patients treated with broadband UVB, NBUVB and heliotherapy. (Mean ±SD)
The increase in 25(OH)D was higher in the broadband treated patients compared
to NBUVB treated group (p=0.008) and compared to patients treated with
heliotherapy (p<0.01). The increase in 25(OH)D during two weeks of
climatotherapy was similar to the increase in 25 (OH)D during the treatment with
NBUVB.
39
Study
HeliotherapyNBUVBBroadband UVBPostmenopausal
women
(broadband UVB)
Mean 25(OH)D ng/ml
80
60
40
20
0
Error bars: 95% CI
25(OH)D after ng/ml
25(OH)D before ng/m
l
Figure 8: The mean 25(OH)D serum concentrations before and after the treatment with
broadband UVB lamp (Paper I and III), NBUVB lamp (Paper III) and heliotherapy (Paper IV),
respectively. Error bars: 95% CI.
The serum concentrations of 25(OH)D increased in postmenopausal women with
psoriasis treated with broadband UVB. There was no difference in the increase of
25(OH)D between the postmenopausal women treated with broadband UVB,
younger patients treated with broadband, NBUVB, or heliotherapy, respectively.
Patients treated with broadband UVB during winter months increased their serum
levels of 25(OH)D from 28.5±8.8 to 64.6±24.1 ng/ml (p<0.001) and patients
treated with NBUVB increased their 25(OH)D from 28.3±6.8 to 47.2±15.5 ng/ml
40
(p<0.001); (p=0.012 between lamps). Patients treated with 2 weeks heliotherapy
increased 25(OH)D from 22.9±6.0 to 41.8±6.3 ng/ml (p<0.001), respectively,
(p<0.015 between broadband UVB treated patients during winter and patients
treated with heliotherapy).
Two weeks of heliotherapy were as effective as treatment with NBUVB 2-3 times
per week for 2 to 3 months. Serum concentration of 25(OH)D at the start in Paper
I and III was however, higher than in patients treated with heliotherapy
(p=0.0001). The mean serum concentration of 25(OH)D after UV therapy was also
higher in patients treated with artificial lamps (p<0.0001) compared with
heliotherapy.
Figure 9: Changes in serum concentrations of 25(OH)D induced by broadband UVB, NBUVB
and heliotherapy in each psoriatic patient.
41
PTH decreased after the treatment with broadband UVB and after heliotherapy
(Table 2). The decrease in PTH was most prominent in the group of
postmenopausal women treated with broadband UVB, Paper I.
The suppression of PTH in postmenopausal women treated with broadband UVB
was higher than in patients treated with NBUVB (p=0.005), Paper III, and in
patients treated with heliotherapy (p=0.026), Paper IV, respectively (Table 2, Figure
10 and 11).
Study
HeliotherapyNBUVBBroadband UVBPostmenopausal
women
(broadband UVB)
Mean PTH (ng/l) after - before
5,0
0,0
-5,0
-10,0
-15,0
-20,0
-25,0
Error Bars: 95% CI
Figure 10: Changes in serum PTH after different phototherapies in psoriatic patients.
42
Figure 11: Changes in serum concentrations of PTH induced by broadband UVB, NBUVB and
heliotherapy in each psoriatic patient.
1,25(OH)2D increased more during the heliotherapy than with NBUVB (p=0.02).
There was no correlation between the dose of UVB and the increase of 25(OH)D,
or 1,25(OH)2D respectively.
Psoriasis improved in all patients, with a reduction in PASI score of about 75% on
all regimens. PASI at start was similar in all groups and improved similarly on all
regimens.
43
Paper I
Twenty-four women aged 60–81 years with a mean BMI of 27.7±4 kg/m2 were
treated with broadband UVB, 2–3 times/week during 8–12 weeks. Psoriasis
improved in all patients, with a reduction in PASI from 6–12 before to 1–4 after
UV therapy.
Vitamin 25(OH)D
Serum levels of 25(OH)D increased from 36.8 ±17.0 ng/ml before the UVB
treatment to 59.6 ± 18.7 ng/ml after (p<0.001) (Table 2, Figure 8).
Two patients showed a reduction in their circulating vitamin D levels following
phototherapy. Both subjects had high level of 25(OH)D at the start of the
treatment – one subject due to travel to a sunny country one month before the
UVB therapy, and one subject commenced her course of UVB at the start of the
autumn following summer, so their vitamin D levels were falling after the summer
peak.
Before UVB therapy, a serum concentration of 25(OH)D below 30 ng/ml was
found in 10 patients (42%) and serum concentrations of 25(OH)D of >30 ng/ml
were present in 14 patients. All increased 25(OH)D after UVB (p=0.001), while
PTH decreased (p=0.001). The increase in 25(OH)D was enhanced in patients
with low vitamin D values.
The serum concentration of osteocalcin was similar in patients with low and normal
vitamin D before and after UVB therapy.
Six of the patients had taken vitamin and mineral supplements (vitamin E, Omega
3 and multivitamins including vitamin D3 (200 IU/day)). Despite this intake of
supplements, two of these patients had vitamin D insufficiency (25(OH)D <30
ng/ml). All these women experienced an increase in serum 25(OH)D similar to
that in patients without supplements.
Serum calcium, osteocalcin, thyroid hormones and creatinine were unaltered after
the UVB therapy.
Serum PTH
Serum PTH decreased after UVB exposure from 63.3±26 ng/l to 48.4±17.3 ng/l
(p<0.001) in all women (Table 2, Figure 10).
44
Serum PTH decreased from 69.6±30 ng/l to 49.8±17 (=29%) (p<0.001) in
patients 70 years of age and from 55.9±19 ng/l to 46.6±19 (=17%) (p=0.039) in
patients <70 years of age.
Secondary hyperparathyroidism (PTH >65 ng/l) was found in 7 patients (29%)
before treatment. In these patients, the median PTH value decreased from 98.2
ng/l before the UVB therapy to 63.7 ng/l (54%) after (p< 0.001). None of the
patients had hypercalcaemia. The median 25(OH)D in these patients was 22.6
ng/ml before UVB treatment and 64.7 ng/ml (186%) after.
Bone status
The mean Z-score value (SD from the mean peak value in age-matched adults) was
+0.54 SD (±1.1) at the hip and +0.50 SD (±1.4) at the lumbar spine.
The mean T-score value (SD from mean peak value in young adults) was -0.90 SD
(±1.15) at the hip and -1.49 SD (±1.4) at the lumbar spine.
Normal BMD (T-score >-1 SD) was found in 10 of the youngest patients,
osteopenia (T-score between –1 to -2.5 SD) in 5 patients and osteoporosis (T-score
<-2.5 SD) in 9 patients.
Mean serum levels of 25(OH)D before the UVB treatment were 38.4±21 ng/ml in
the group with normal BMD, 36.0±13 ng/ml in the group with osteopenia and
35.5±16 ng/ml in the women with osteoporosis (ns. between groups).
After the UVB treatment, serum 25(OH)D increased in all groups (p<0.01),
respectively. The mean serum level of 1,25(OH)2D did not differ between the
different BMD groups before or after the UVB treatment. There was no
relationship between psoriasis onset and bone status. Any changes in BMD were
not expected to occur in 8-12 weeks of UV therapy.
Paper II
Anthropometry and bone status
Thirty-five women with psoriasis were compared with 2448 age-matched control
women from the Geriatric out-patient clinic, Göteborg, regarding risk factors for
osteoporosis. Both the Z score and T score at the hip and lumbar spine were
higher in women with psoriasis than in controls.
45
A positive family history for kyphosis was less common in patients than in controls
(p=0.0086). The duration of HRT was shorter in patients than in controls
(p=0.045).Prevalence of myocardial infarction and angina pectoris was more
common in the patient group. Patients had higher consumption of cheese and
coffee and were more physically active than controls. The degree of physical
activity in patients was 3.7±2.2 of 1–4 (low to high) per week vs. 1.8±1 in controls
(p=0.0001).
Subjects with normal BMD (n=14) were younger (67±6 years) than the patients
with osteopenia and osteoporosis (72±6 and 70±6 years, respectively). BMI and
the number of UVB treatments did not differ between groups. The age at onset of
psoriasis correlated positively with BMD in all patients.
All fractures occurred after falling during patients’ leisure time and 87% involved a
wrist fracture.
The patients with normal BMD had highest childbirth, latest menopause and
longest duration of HRT.
Milk consumption varied from 0 to 5 glasses per day but did not differ between
groups. Coffee consumption varied between 1 to 8 cups/day and was highest in the
patients with osteoporosis. Vitamin and mineral supplementations (vitamin E,
omega 3, calcium or multivitamin) did not differ between groups. Current smoking
was more common in patients with osteopenia and osteoporosis.
Physical leisure time activities were highest in patients with normal BMD. The
activities were mainly described as Grade 3 (regular exercise within different
activities depending on the season such as swimming, skiing, walking and
gardening).
Sun exposure of two hours or more during summer was highest in patients with
normal BMD. The patients with normal BMD had the highest number of trips to
sunny countries in the last 2 years. Spending time outdoors (independent on sun
exposure) was similar in all groups and varied between 1–2 h/day.
None of the patients had systemic treatment with corticosteroids but 27 (77%) had
topical treatment with cortisone ointments alone or in combination with vitamin D
analogue (calcipotriol). Patients with normal BMD used more calcipotriol while
osteoporosis patients used more topical corticosteroid.
46
Paper III
Twenty-six psoriatic patients (7 women and 19 men) with broadband UVB
treatment were compared with 42 (10 women and 32 men) psoriatic patients with
NBUVB treatment for 8 to 12 weeks.
Serum 25(OH)D increased after both broadband and NBUVB (Table 2, Figures 8
and 9). The increase in 25(OH)D was higher in the broadband treated patients
(p=0.008) compared with NBUVB. Serum PTH decreased on broadband UVB
(p<0.05).
Secondary hyperparathyroidism (PTH >65ng/ml) was present in 5 men and their
PTH values normalised after UVB therapy. 25(OH)D increased similarly in patients
with 25(OH)D <30ng/ml as in patients with 25(OH)D 30 ng/ml at baseline.
There were no changes in 1,25 (OH)2D, creatinine or serum calcium in the groups
after treatment.
There was no correlation between the dose of UVB and the increase of 25(OH)D.
The treatment time was four times longer for the NBUVB treated patients than for
patients treated with the broadband UVB.
Patients with skin type II (n=23), obtained as expected, a lower median dose of
UVB (broadband 6.9 J/cm2 and narrowband 31.4 J/cm2) than patients with skin
type III (n=44, dose of broadband UVB 16.4 J/cm2 and 51.8 J/cm2 dose of
NBUVB) and IV (n=1, treated with broadband UVB, dose 15.7 J/cm2). Patients
with skin type II (n=23) increased in 25(OH)D from 33.8±16.8 to 56.5±20.7
ng/ml (p<0.001) and patients with skin type III (n=44) increased from 36.9±12.3
to 64.0±18.0 ng/ml (p<0.001), respectively (ns between skin type II and III).
Psoriasis improved in all patients, with a similar reduction in PASI score in both
groups. Improvement in psoriasis correlated positively with increase in 25(OH)D
(p=0.047).
The baseline 25(OH)D levels were lower in those who started in winter compared
with those who started during spring (p=0.0001).
The age difference was explained by the later introduction of the NBUVB lamps
(younger patients).
47
Paper IV
Sun exposure for 15 days led to reduction in the PASI score in patients with
psoriasis. Furthermore, the sun exposure increased serum concentrations of both
25(OH)D and 1,25(OH)2D.
Patients with 25(OH)D 20 ng/ml (50 nmol/l) at baseline (1 woman and 5 men)
increased more in 25(OH)D than patients with 25(OH)D >50 nmol/l (p=0.03).
The serum concentrations of 1,25(OH)2D increased after 15 days of sun exposure
(p=0.01 day 1–15 and p=0.004 day 2–15).
The serum concentrations of PTH decreased (p=0.04 day 2–15).
BMI was unaltered during the study period. The sun exposure improved the lipid
and carbohydrate status of the patients. The LDL/HDL cholesterol ratio decreased
from 2.4 to 1.9 (p<0.001). The serum concentrations of APO-A1, EHb, folic acid,
HDL, homocysteine and uric acid increased during 15 days of climate therapy. The
serum concentrations of APO-B, vitamin B12 and HbA1c decreased during the sun
exposure period. The serum concentrations of calcium, creatinine, EHct, glucose,
CRP, mCRP, total cholesterol, LDL, Lp(a) and triglyceride were not influenced by
climate therapy.
Diastolic blood pressure increased from 84.5±9.7 to 90.9±10.6 mmHg (p=0.007)
while systolic blood pressure and pulse were unaltered after 2 weeks of sun
exposures.
Daily sun exposure time per patient increased throughout the treatment period.
The patients received on average 5.1±2.3 SED (median=4.0 SED), range 2.6–10.3
SED on the first day of exposure. The mean dose was similar for the patients with
skin type II and III. The mean dose after 15 days sun exposure was 166±25 SED
(range 104–210), 135 and 169 SED for the patients with skin type II and III,
respectively. The variation between minimum and maximum patient doses each day
was large. The patients´ sunbathing with sunscreen during the first day reported a
use of approximately 30 ml of cream each on body sites easily burned. Although 14
out of the 20 patients reported erythema after the first day of sun exposure, but
none reported blistering erythema.
The reduction in PASI score was 73%, but there was no correlation between the
improvement in PASI score and vitamin D or the UV dose, respectively.
48
Furthermore, we did not find any correlation between changes in the single
component of PASI score, including area, erythema, infiltration and desquamation,
and the dose of received UVB.
Serum concentration of 1,25(OH)2D at baseline correlated positively to serum
concentration of 25(OH)D at baseline (p<0.0005) and negatively to increase of
25(OH)D (p=0.023). The increase in 25(OH)D correlated to the increase in
1,25(OH)2D after 15 days of climate therapy (p<0.0005). Serum concentrations of
25(OH)D at baseline correlated positively to serum HDL at baseline (r=0.46,
p=0.040).
49
Discussion
Serum 25(OH)D in psoriasis patients during treatment
with phototherapy (Papers I, III and IV)
Serum 25(OH)D levels increased in psoriasis patients following treatment with
heliotherapy, broadband UVB and NBUVB phototherapy. Psoriasis improved in all
patients, with a reduction in PASI score of about 75% on all regimens. UVB and
sun exposure are the strongest factors influencing 25(OH)D.8,18,33,138,192 The same
wavelength of the UVB spectrum (290-315 nm) that is responsible for D3 vitamin
synthesis in the skin also improves psoriasis lesions, and has therefore been used in
psoriasis therapy.
Vitamin D3 production in patients with psoriasis increased less with NBUVB than
with broadband UVB phototherapy. One explanation might be that the optimal
wavelength for initiation of the vitamin D3 pathway was 300±5 nm in vitro and in
vivo193 which is in the broadband UVB range (280-320 nm). The synthesis of
vitamin D3 was stimulated by wavelengths between 290-315 nm, but not longer
than 315 nm. The present results from our study (Paper III) showed that a
wavelength of 311 nm was effective for inducing vitamin D3 synthesis, but not to
the same extent as wavelengths in the broadband UVB range. UVB treatment of
psoriasis was a sufficiently time-consuming procedure to increase vitamin D3 also
with NBUVB. The time required for NBUVB to have an effect can reduce the
difference in the potential for vitamin D3 production between the two lamps. The
treatment time correlated strongly with the type of lamp (patients treated with
NBUVB required 4 times longer exposure times than patients treated with
broadband UVB). This is consistent with other studies demonstrating that the dose
response of the erythemal spectra of NBUVB should be about 4.2 times that of
broadband UVB 194. The dose of UVB also correlated with the type of lamp, but
we could not find any correlation between the dose of UVB and the increase of
25(OH)D levels. This might be explained by autoregulation of skin synthesis,
storage, and slow, steady release of vitamin D3 from the skin into the circulation.8
The serum concentrations of 25(OH)D almost doubled during 15 days of climate
therapy (Paper IV). Patients with lower 25(OH)D levels at baseline responded
better to sunlight and phototherapy (Paper I and IV) which is consistent with other
50
studies.195 All patients reached serum levels of 30 ng/ml (75 nmol/l) after two
weeks of sun exposure. A circulating level of 25(OH)D of >30 ng/ml, or >75
nmol/l, appears to be necessary to maximize the health benefits of vitamin D.195
Sun exposure is the major source of vitamin D3 for most humans.195 Skin pigment,
sunscreen use, aging, time of day, season, and latitude all affect previtamin D3
synthesis.196
The ability of the skin to produce vitamin D3 declines with age 12 due to
insufficient sunlight exposure29,197 and a reduction in the functional production
capacity of the skin.12,13,198 All the patients in our study (Paper I) were post-
menopausal women and we were unable to see any clear negative correlation
between age and vitamin D3 synthesis in line with a previous population study
carried out in the same city.138 Age did not correlate with the increase in 25(OH)D
in our intervention studies (Paper I, III, and IV).
The increase in 25(OH)D during 2 weeks of heliotherapy was very similar to the
increase in 25(OH)D during treatment with broadband UVB and NBUVB for 2 to
3 months (Paper I and III). During prolonged exposure to the sun, the
accumulation of previtamin D3 is limited to about 10 to 15% of the original 7-
DHC content, because the previtamin also undergoes photoisomerization into two
biologically inert photoproducts, lumisterol-3 and tachysterol-3.8
The vitamin D3 production is a unique, autoregulated mechanism which occurs at
two levels. Excessive sun exposure does not lead to overdosing of vitamin D3 due
to conversion of previtamin D3 to inactive photoproducts (lumisterol 3 and
tachisterol 3) as well as conversion of vitamin D3 to its isomers in the skin (5,6-
trans vitamin D3, supersterol I, supersterol II) which are thought to have low
calcemic effect at physiological concentrations. Vitamin D3 is synthesized in the
skin and released steadily and slowly from the skin into the circulation.8
There was no difference in the increase of 25(OH)D between the different skin
types in the present studies. The reason could be that the subjects were exposed to
individually adjusted doses of UVB depending on skin phototype and erythemal
response to therapy. All patients had previous experience of UVB therapy for their
psoriasis disease. As expected, fair-skinned patients required lower doses of UVB
(broadband and narrowband) than patients with skin type III and IV. This finding
is consistent with other studies examining the effect of skin pigmentation on
51
vitamin D3 synthesis.199 Melanin pigment in human skin competes for, and absorbs
the UVB photons responsible for the vitamin D3 synthesis.199
During the winter months vitamin D3 production is insufficient to meet the
optimal requirements in both younger and older adults at Northern latitudes.200 As
seen in a previous study201, lower baseline 25(OH)D levels were found in those
who initiated treatment in winter. Psoriasis lesions usually deteriorate during winter,
and many patients are therefore given repeated UVB treatment during this season.
In addition to healing psoriasis lesions, UVB therapy also provides these patients
with vitamin D3 during the winter months, when levels of 25(OH)D in Northern
countries are generally low. Broadband UVB induced vitamin D3 production and
suppressed PTH (Paper I, III and IV). The increase in 25(OH)D was similar for
the broadband and NBUVB lamps during the spring period. The influence of
ambient UVB on vitamin D status during the spring months (March-June) might
explain this result.
We found no correlation between the increase of the dose of UVB and the increase
of serum 25(OH)D levels within the groups. This might be due to the fact that
serum concentrations of 25(OH)D were measured at different time points and a
plateau level was reached after three weeks, which had also been seen in a previous
study.202 A recent in vitro study has demonstrated that the dose-response
relationship of UV exposure and cholecalciferol synthesis was nonlinear. It was
hypothesized that exposure to additional UV may not result in a proportional
increase in vitamin D levels.203
The correlation between sunlight measures and serum 25(OH)D has been shown
to be weak.204 Patients (Paper IV) reached their plateau of daily sun exposure after
the first week. It might be that the vitamin D3 production was most prominent
during the first week, when the patients had experienced redness and some of them
even got sunburned.
The increase of 25OHD during 15 days of climate therapy was significant even
though the patients used sunscreens on body sites susceptible to sunburn, and even
though the skin was affected by psoriasis lesions. An SPF-8 sunscreen reduces the
skin's production of vitamin D3 by 95%. Clothing completely blocked all solar
UVB radiation and thereby prevented vitamin D3 production.15
We found no correlation between the reduction in PASI score and serum
concentrations of 25(OH)D during climate therapy, consistent with another study
52
(Paper I). Furthermore, there was no dose-dependent correlation between vitamin
D metabolite levels, PASI score and the received UV dose.
Improvement in psoriasis correlated positively with an increase in 25(OH)D3 levels
in Paper III (p=0.047; the group of patients treated with broadband UVB and
NBUVB) but not in the other studies (Paper I and IV). We do not have a good
explanation for this. Instead we propose that a larger population of psoriasis
patients should be examined to clarify this finding.
It has been alleged that NBUVB is more effective than broadband UVB for
reducing PASI scores, as well as safer and better tolerated by patients than PUVA
when taken at suberythemogenic doses.205 The NBUVB lamp seemed to be easier
to handle and better tolerated, giving it some advantages over the standard
broadband UVB206 which has led to a reduction in the usage of broadband UVB.
One of the drawbacks with the new lamp is that the radiation times are almost
doubled.207 The reduction of PASI scores in our study (Paper III) was similar in all
patients, irrespective of the choice of lamp. The patients were not allowed to apply
calcipotriol to psoriasis lesions during the study and several patients noticed a
somewhat delayed effect of the UVB therapy on the healing process. Several
studies have shown more rapid healing of psoriasis with the combination of
calcipotriol and UVB radiation than with monotherapy with either treatment.102,208
Our method for measuring and estimating UV doses (Paper IV) is simpler
compared with using personal UV dosimeters, but it is associated with more
uncertainties. These are discussed by Snellman et al209, but the predominant
uncertainty is probably due to the fact that the skin dose equals half the ambient
dose. This is appropriate as a first approximation, in particular for the abdomen
and back during sun bathing if the patients turn to expose these sites to an equal
extent. Measurements using personal dosimeters in other studies support this
assumption.209-211 For the extremities, doses are reported to be higher or lower
depending on activities, and all vary more than during sun bathing.209,211,212 The
upper extremities probably often receive more than 50% of the ambient UV209-212
and are therefore more susceptible to sunburn. These sites are often covered with
sunscreen through the treatment period.
The correlation between personal report of sun exposure and ambient UV light
measured by dosimeter (assessment of radiation dose) is weak.204
53
The positive correlation between serum 25(OH)D and serum 1,25(OH)2D (Paper
IV) suggests that the production of the latter is substrate-dependent in vitamin D-
deficient patients.136,213 Serum concentration of 25(OH)D at baseline was lower in
patients treated with heliotherapy (p=0.0001) than in patients treated with
broadband UVB and NBUVB, and in postmenopausal women with psoriasis
treated with broadband UVB.
Serum 25 OH vitamin D levels in the postmenopausal psoriatic women before
UVB therapy (Paper I) were similar to those in age- and location-matched control
women.138 The serum concentration of 25(OH)D at baseline was similar in the
patients treated with broadband UVB and NBUVB, and in postmenopausal
women with psoriasis treated with broadband UVB. After broadband UVB
treatment, serum 25(OH)D increased whilst serum PTH decreased. In addition to
the positive calciotropic effect of vitamin D on bone metabolism and the
suppression of PTH214,10, vitamin D also plays an important role in healing
psoriasis.215-217 This indicates that the positive effect of UVB on psoriasis might be
due to vitamin D3 production in the skin.
The increase in serum 25(OH)D found after UVB exposure was consistent with
the results from sun exposure studies, where sunlight was the strongest factor
influencing serum vitamin D in women in the general population.79,138
The regular use of sunbeds that emits vitamin D-producing ultraviolet radiation is
associated with higher 25(OH)D concentrations in healthy adults, and may thus be
of benefit to the skeleton in vitamin D insufficiency.79 Others have found that
vitamin D3 is more efficacious than vitamin D2 (the major vitamin in supplements)
for increasing serum 25(OH)D levels.218 UVB therapy increased serum 25(OH)D
levels even in patients taking vitamin D supplements. This is in line with previous
studies which reported that UV-induced vitamin D3 synthesis had a greater
influence on the serum levels of circulating calcidiol than the peroral intake of
supplements.10,218
The controlled use of UVB is a safe and effective method for the treatment of
psoriasis151 and could be used as treatment for vitamin D deficiency.136
The cut-off level for serum 25(OH)D, which is used as a diagnostic marker for
vitamin D deficiency, has varied over the years.33-35 The early biochemical changes
in vitamin D insufficiency include a rise in serum PTH, which begins to increase as
serum 25(OH)D levels fall below 30 ng/ml or 75 nmol/l.35 This level of 25(OH)D
54
has become the suggested cut-off point for vitamin D deficiency or
inadequacy.29,30,35,219
In those studies, it is not possible to draw conclusions about the ability of psoriatic
skin to produce vitamin D3. A larger study is needed to examine the correlation
between PASI and the capacity of psoriatic skin to produce vitamin D3 during
UVB exposure.
Serum 1,25(OH)2D in psoriasis patients during treatment
with phototherapy (Papers I, III and IV)
Vitamin D undergoes metabolism in the liver to 25(OH)D, and then in the kidney
to a number of metabolites, the most important of which is 1,25(OH)2D. This is
the classical pathway and quantitatively the most important for producing
1,25(OH)2D. However, the keratinocyte is fully capable of producing its own
1,25(OH)2D.46 The skin is the only tissue yet known in which the complete UVB-
induced pathway from 7-DHC via intermediates (previtamin D3, vitamin D3,
25(OH)D) to the final product 1,25(OH)2D, takes place under physiological
conditions.26 Levels of 1,25(OH)2D tended to increase during phototherapy, but
statistically significant increases were noticed only during heliotherapy, and only in
postmenopausal women with 25(OH)D3 below 30 ng/ml, and in women aged
70 years. One explanation might be that these patients had lower serum
concentrations of 25(OH)D at the start of the treatment.
It has been postulated that the synthesis of 1,25(OH)2D is tightly regulated, and
that increases in 25(OH)D concentrations due to exposure to sunlight have no
effect on serum 1,25(OH)2D levels.195,220 Some other studies221,222 have shown a
positive correlation between serum 25(OH)D and serum 1,25(OH)2D in vitamin
D-deficient patients, indicating substrate-dependent synthesis of 1,25(OH)2D. The
similar observation that both 25(OH)D and 1,25(OH)2D increased in vitamin D
deficient persons following UVB exposure223 or after vitamin D supplementation221
has been reported previously. Parathyroid hormone is a well-known stimulator of
1,25(OH)2D synthesis. The positive correlation between 1,25(OH)2D and intact
PTH reflects the physiological effects of PTH on the 1,25(OH)2D response to low
calcium. The positive relationship between PTH and 1,25(OH)2D, and the inverse
relationship between PTH and 25(OH)D has been shown in a previous study from
55
the same city.224 The persistent elevation of 1,25(OH)2D concentrations in patients
with low levels of 25(OH)D might be explained by PTH-induced chronic
stimulation of the renal 25(OH)D-1α-hydroxylase.223
The increase of 1,25(OH)2D levels between patients treated with heliotherapy and
patients treated with NBUVB differed (p=0.02). This might be also explained by
lower values of 25(OH)D at baseline in patients treated with heliotherapy.
Keratinocytes are capable of producing a variety of vitamin D metabolites,
including 1,25(OH)2D, 24,25(OH)2D, 1,24,25(OH)3D46 from exogenous and
endogenous sources of 25(OH)D. The process by which 1,25(OH)2D is produced
and catabolised is tightly regulated and coupled to the differentiation of these cells.
The epidermis is likely to contribute to circulating levels of 1,25(OH)2D, as human
keratinocytes rapidly and extensively convert 25(OH)D to 1,25(OH)2D.46,225 Peak
intracellular levels of 1,25(OH)2D are reached within 1 hour of adding 25(OH)D.46
However, when renal production of 1,25(OH)2D is normal, circulating levels of
1,25(OH)2D are sufficient to limit the contribution from epidermal production.225
It is now clear that epidermal 1α-hydroxylase and renal 1α-hydroxylase are identical
proteins. The 1α-hydroxylase molecule is a 56-kDa protein with 506 amino acids.
Production of 1,25(OH)2D regulates itself in the cell through a negative feedback
loop which is similar to that observed in the kidney, but differs from that seen in
the macrophage where this feedback is missing.46 An important difference between
keratinocytes and renal cells in the regulation of 25(OH)D metabolism by
1,25(OH)2D is that the concentration of 1,25(OH)2D required to inhibit the 1α-
hydroxylase and induce 24-hydroxylase in renal cells appears to be several times
greater than that required to achieve comparable effects in keratinocytes.46,225 Thus,
1,25(OH)2D production by keratinocytes is extremely sensitive to exogenous
calcitriol. Acute nephrectomy leads to a very low extrarenal production of
1,25(OH)2D226, and pig skin perfused with 25(OH)D showed low amounts of
calcitriol which increased after 4-8 hours of perfusion.227 In contrast to the renal
1α-hydroxylase which is tightly regulated by feedback mechanisms, the extrarenal
1α-hydroxylase is only constitutively expressed.21 As a consequence, there should
be no relevant feedback mechanisms between the systemic and epidermal vitamin
D pathways that ultimately depend on UVB radiation.21 Thus, the local UVB-
triggered production of calcitriol may primarily regulate epidermal cellular functions
in an auto- and paracrine manner, but this should not be crucial for systemic
56
vitamin D effects21 and systemic vitamin D deficiency does not stimulate epidermal
synthesis of 1,25(OH)2D.228
Cutaneous production of 1,25(OH)2D3 may regulate growth, differentiation,
apoptosis and other biological processes in the skin.229 The NBUVB has been
shown to have less capacity to induce a local skin production of 1,25(OH)2D3 at
44% of the monochromatic irradiation at 300 ±2.5 nm.193
There is no information in the literature whether psoriatic skin is capable of
synthesizing vitamin D.
It is not clear if the serum 25(OH)D level is linked to the level of the active form of
vitamin D3 present in the skin. It has been suggested that cutaneous conversion of
25(OH)D to 1,25(OH)2D does not play a role because the amount of free
25(OH)D3 that penetrates the cell membrane of epidermal keratinocytes is too
small to produce sufficient amounts of 1,25(OH)2D.229 The main form of
circulating 25(OH)D is presented in a complex with vitamin D-binding protein
(DBP) with only a very small amount (0.03%) available as the free form.
Furthermore, the deeper layers of the epidermis are not vascularised, which further
impairs the passage of the 25(OH)D3-DBP complex from blood to epidermal
keratinocytes.229
We found no correlation between reduction in PASI score and serum
concentrations 1,25(OH)2D. Nevertheless, the known therapeutic effect of UVB
light therapy for the treatment of psoriasis may be mediated via UVB-induced
production of 1,25(OH)2D.26 The calcitriol concentration in UVB-treated skin
measured by microdialysis was dependent on the UVB dose.230 In vitro studies have
shown that the substrate concentration of cholecalciferol in keratinocytes mainly
determines the synthesis rate of 1,25(OH)2D in these cells.231 Thus, higher synthesis
rates of cholecalciferol should result in a faster and more pronounced release of
1,25(OH)2D into the extracellular fluid. UVB-induced membrane damage to
epidermal keratinocytes may also increase the outflow of newly synthesized
calcitriol.230
The 1,25(OH)2D molecule and its analogues, as well as UVB phototherapy, exert
antiproliferative, prodifferentiative, and immune-modulatory effects on
keratinocytes that are of particular importance for the therapy of hyperproliferative
skin diseases such as psoriasis vulgaris.21,232 However, the full range of UVB and
vitamin D3 effects is not completely understood.
57
The observation that 1,25(OH)2D induces keratinocyte differentiation was first
made by Hosomi et al.233 and provided an explanation for the earlier and
unexpected finding of 1,25(OH)2D receptors in the skin. 1,25(OH)2D is likely to be
an autocrine or paracrine factor for epidermal differentiation, since it is produced
by the keratinocyte. The receptor for, and the production of 1,25(OH)2D vary with
the differentiation in a manner suggesting feedback regulation;234 both are reduced
in the later stages of differentiation. 1,25(OH)2D increases involucrin,
transglutaminase activity, and cornified envelope formation in keratinocytes.233
Serum PTH in psoriasis patients during treatment with
phototherapy (Papers I, III and IV)
Plasma concentrations of 1,25(OH)2D3 (calcitriol) depend mainly on renal
function, plasma concentrations of intact PTH, and dietary intake of calcium and
phosphate. High PTH and low dietary intake of calcium and phosphate are the
main stimulators of the production of calcitriol. PTH increases with increasing age,
possibly due to less sunlight exposure and/or reduced calcium/vitamin D intake.224
The clear concomitant decrease in serum PTH after UVB exposure in our studies
indicates that the skin has a good ability to synthesise vitamin D3 even at high age
and with part of the skin covered by psoriasis lesions. Our results indicate that in
women aged >70 years, UVB therapy radically increased vitamin D levels and
suppressed PTH levels in serum. However, after adjustment for age, sun exposure
was the only external factor that influenced serum vitamin D.138 This is in line with
the positive effect of UVB on the vitamin D3 synthesis in the women in our study,
irrespective of age.
Serum PTH levels in postmenopausal women with psoriasis was higher both
before and after UVB exposure compared with the population sample from the
same region.224 We found 7 patients (Paper I) and 5 patients (Paper III),
respectively, with secondary hyperparathyroidism due to vitamin D deficiency. In
these patients, PTH was suppressed by the UVB-induced increase in serum vitamin
D3. Irradiation with UVB was as effective as oral vitamin D3 for increasing serum
25(OH)D and suppressing secondary hyperparathyroidism.136 25(OH)D
concentrations below 30 ng/ml (75 nmol/l) resulted in secondary
hyperparathyroidism and a decrease in BMD.235
58
Decreased renal function, oestrogen deficiency and low calcium intake may also
induce secondary hyperparathyroidism.30 Broadband UVB induced vitamin D3
synthesis and suppressed PTH (Papers I, III and IV).
Bone status in postmenopausal women with psoriasis
treated with UVB phototherapy (Paper I and II)
Postmenopausal women with psoriasis (Paper I and II) had higher BMD both of
the hip and the lumbar spine compared with age-matched controls.
Multiple risk factors that contribute to low serum 25(OH)D and osteoporosis have
been identified. They include inadequate sun exposure18, insufficient intake of
fortified foods or vitamin D supplements236, low body mass index, white ethnicity,
lack of exercise, use of medications that accelerate vitamin D metabolism, diseases
that alter vitamin D metabolism such as malabsorption syndromes, and chronic
liver disease.30,35,237
In our study (Paper I), patients with 25(OH)D levels below 30 ng/ml and
secondary hyperparathyroidism had lower BMD in terms of both T and Z scores
of the hip and the lumbar spine compared with those with higher vitamin D levels,
consistent with another study.79
The second study (Paper II) showed that women with psoriasis had higher BMD
both of the hip and the lumbar spine compared with age-matched controls. Higher
body weight and BMI are factors which may have contributed to the higher BMD
in patients compared with controls. Information about the prevalence of
osteoporosis among psoriasis patients and the epidemiology of risk factors for
osteoporosis in this group are sparse. However, a previous study showed no
evidence that patients with chronic plaque psoriasis had low BMD despite risk
factors, although the subgroup with joint involvement appeared to be at higher risk
of osteoporosis and therefore required prevention therapy.132 Reduced BMD has
been linked to palmoplantar pustular psoriasis.133 The existence of less severe
periarticular osteoporosis is considered possible, but there are no data concerning
the prevalence of systemic osteoporosis in patients with psoriatic arthritis.134 In
general, bone loss increases with increasing age. BMD has been shown to be a
predictive indicator for bone fractures in healthy subjects and in patients with
osteoporosis.238
59
Vitamin D is important for bone metabolism.135 Vitamin D deficiency thus
contributes to the pathogenesis of osteoporosis and hip fractures.136
Supplementation strategies involving calcium and vitamin D supplements are cost-
saving for preventing osteoporotic fractures.137
Treatment with UVB in patients with psoriasis is most common during the darker
period of the year when UVB is lacking and levels of vitamin D are low in
Northern countries. Furthermore, UVB therapy heals psoriasis and supplies these
patients with vitamin D at levels similar to those in the general population,138 which
might have positive effects also on bone status.
A family history of fractures, physical activity, smoking and oestrogen substitution
are important factors influencing bone mass.239-241 Low body weight is related to
low skeletal muscle mass and increased risk of fractures.241,242 Muscle tissue and
strength are important for body balance and to prevent of falls.243 Previous studies
have confirmed a protective effect of weight gain against fractures.237 Moreover, the
relative risk of fractures (with the exception of spinal fractures) correlated positively
with current height, height at the age of 25, and height loss since the age of 25.244
The decrease in height since the age of 25 in patients with psoriasis with
osteoporosis compared with patients with normal BMD was consistent with
findings in other studies.237
A positive family history of hip fractures and especially kyphosis was more
common in controls than in patients with psoriasis. That could be a possible
explanation for the lower BMD of the lumbar spine in controls compared with
patients with psoriasis. Heredity for hip fractures was mainly found in psoriatic
patients with osteoporosis. A maternal history of hip fractures doubled the risk of
having a hip fracture.240,242 A history of fractures (particularly a family history of hip
fractures) confers an increased risk of fractures that is independent of BMD.245
Furthermore, the effect of family history is not general, but implies a site-specific
predisposition to fractures.246
Menopause in patients with psoriasis occurred on average at a higher age in the
group with normal BMD compared with the osteoporosis group. Early menopause
is widely regarded as a risk factor for osteoporosis.247 However, bone mass declines
and the risk of fractures increases with increasing age, especially after the
menopause along with diminishing endogenous secretion of oestradiol.237,239,241
60
Osteoporosis in postmenopausal women is more related to hormonal aberrations
than to lifestyle factors.237 Patients who used HRT had higher BMD.237,248 The
duration of HRT was longer in controls than in psoriasis patients. Within the
psoriasis group, the duration of HRT was longer in patients with normal BMD.
The duration of contraceptive usage was considerably longer in psoriasis patients
with normal BMD than in the other two groups. Prior use of, and the duration use
of oral contraceptive agents is associated with higher BMD.249
A history of postmenopausal symptoms in psoriasis patients did not influence
BMD, consistent with a previous study.250
Physical activity correlated positively with BMD in psoriasis patients. Physical
activity has been claimed to be beneficial to bone mass and protective against
fractures.251 Regular walking in middle-aged and elderly women is associated with a
reduced risk of vertebral deformity.252 Subjects who took a daily walk of at least 30
min had a significantly better climbing capacity, higher BMD and lower
concentration of serum triglycerides than subjects who walked less.253 Lifetime
exercise was also positively associated with BMD of the hip.254
Current smoking was positively correlated with osteoporosis in our study,
consistent with previous studies.255,256 Smoking is widely considered to be a risk
factor for future fracture.257,258
None of the patients had undergone systemic treatment with corticosteroids, but
77% had used topical treatment with corticosteroid ointments alone or in
combination with vitamin D analogues (calcipotriol). The usage of topical
corticosteroids was sparse, and the dosages were too low to have any systemic
effects or impair on BMD. Prior and current exposure to corticosteroids confers an
increased risk of fractures.259
Many of the patients, 63%, had joint pain (half of these patients had normal BMD)
and reported limited mobility or function in at least one joint, but none of them
had psoriasis arthritis. Higher body weight or BMI were positively associated with
joint pain, consistent with a previous study.260
The limitation of this study (Paper II) was the small number of patients with
psoriasis, and larger studies are needed before any firm conclusions regarding bone
status and possible risk factors in psoriatic patients can be drawn.
61
Lipid status and blood glucose in psoriasis patients
during treatment with heliotherapy (Paper IV)
Dyslipidemia
The ratio of low-density lipoprotein cholesterol (LDL) and high-density lipoprotein
cholesterol (HDL) decreased, and the levels of haemoglobin A1c (HbA1c) also
decreased in psoriasis patients during heliotherapy.
A strong link has been suggested between psoriasis and abnormalities in fatty acid
metabolism.118 Patients with psoriasis exhibit a dyslipidemic profile, including
increased levels of plasma cholesterol, triglycerides (TG), LDL, very low-density
lipoprotein (VLDL) cholesterol, and decreased levels of HDL cholesterol and
antioxidant capacity.
Consistent with these findings, a potentially atherogenic lipid profile has also been
observed in psoriasis patients compared with matched controls.261 A recent study
investigating the role of altered lipid metabolism in psoriasis suggests that a
dyslipidemic profile may precede the onset of psoriasis.262 Compared with healthy
controls, patients with recent onset of psoriasis showed significantly elevated levels
of HDL cholesterol and apolipoprotein (Apo) A-1 and had altered cholesterol/TG
ratios in VLDL particles.262 Adjustment for age, sex, smoking, physical exercise,
alcohol use, BMI, and systolic blood pressure did not influence the results, leading
to the conclusion that lipid abnormalities in psoriasis patients may be genetically
determined.262 Furthermore, HDL-associated Apo A-1 can inhibit the activation of
monocytes/macrophages by interfering with their interaction with stimulated T-
cells, leading to decreased TNF-α and IL-1 production.263 Therefore, it is possible
that lower levels of HDL may contribute to a state of chronic inflammation.263
Moreover, VLDL and LDL have been shown to enhance the proliferation and
LDL receptor expression of human epidermal keratinocytes in vitro, and increased
VLDL or LDL could thus contribute to the pathogenesis of psoriasis.264
Metabolic syndrome
Large epidemiological studies have confirmed that psoriasis and psoriatic arthritis
are associated with metabolic diseases including obesity, dyslipidemia and
diabetes.265 The metabolic syndrome comprises a cluster of risk factors including
62
central obesity, atherogenic dyslipidemia, hypertension and glucose intolerance, and
is a strong predictor of diabetes, cardiovascular diseases, and stroke.266,267 Patients
with psoriasis also show an increased prevalence of the metabolic syndrome
compared with nonpsoriatic dermatological patients.268 The importance of the
metabolic syndrome is that it may confer a higher cardiovascular risk than its
individual components.265
Psoriasis patients with metabolic syndrome had disease onset at an earlier age, and
longer disease duration compared with psoriasis patients without metabolic
syndrome268, suggesting that psoriasis duration is a risk factor for the metabolic
syndrome. On the other hand, abdominal obesity is a proinflammatory state with
the visceral adipose tissue providing a rich source of inflammatory mediators
known as adipocytokines. These include adiponectin, leptin, resistin and visfatin,
and may provide an important link between obesity, insulin resistance and related
inflammatory disorders. Other products of adipose tissue that have been
characterized include TNF-α and IL-6.269 These products all play well-defined roles
in the pathogenesis of psoriasis and at the interface between the immune and
metabolic systems. In line with this hypothesis is the evidence that obesity is a risk
factor for other inflammatory diseases such as atherosclerosis and asthma.
Hypertension
Diastolic blood pressure increased in patients with psoriasis following 15 days of
climate therapy. Other studies have described that sun-like UV irradiation
decreased resting pulse rate, decreased recovery pulse rate, and decreased systolic
blood pressure in healthy persons.270 We have no explanation for the present
contradictory finding, but it could be due to chance or possibly changes in physical
activity or alcohol or nutrient intake.
A higher prevalence of hypertension in psoriasis patients compared with controls
has been reported.271 The possibility exists that hypertension and psoriasis share
common risk factors.118 For example, angiotensin II, a product of angiotensin
converting enzyme (ACE), is known to regulate vascular tone and to stimulate the
release of proinflammatory cytokines.272 Increased levels of serum ACE in psoriasis
patients have been reported273, as well as an increase in plasma renin activity. The
prevalence of hypertension in psoriasis and the basis of this association require
further study.
63
Insulin resistance/diabetes
An association between psoriasis and increased serum fasting glucose levels,
hyperinsulinemia, insulin resistance, and type 2 diabetes has been demonstrated.274
A cross-sectional study found that psoriasis patients were more likely to be insulin
resistant and to have impaired glucose tolerance, higher fasting insulin levels, and
impaired β-cell function than non-psoriatic subjects.274 However, there were no
correlations between these measures and psoriasis disease severity and duration.274
Although the literature supports an association between psoriasis and insulin
resistance/type 2 diabetes, the confounding factor of obesity should be taken into
consideration. Obesity, especially abdominal obesity, is strongly associated with the
development of the metabolic syndrome and type 2 diabetes.275
Neimann et al. demonstrated that obesity and diabetes were more strongly
associated with severe psoriasis than were hypertension, hyperlipidemia, or
smoking.271 Therefore, lifestyle factors (e.g. lack of exercise, poor diet) could
contribute to the development of obesity and accompanying insulin resistance in
psoriasis patients.
Proinflammatory cytokines involved in psoriasis, such as TNF-α and IL-1β, play a
central role in the development of atherosclerosis.276 Atherosclerosis is considered
to be an inflammatory disease and involves the production of proinflammatory
cytokines by immune cells (macrophages, monocytes, T cells) at the sites of
atherosclerotic lesions.276
Another link between psoriasis and atherosclerosis involves
hyperhomocysteinemia.277 Hyperhomocysteinemia is considered to be a risk factor
for atherosclerotic vascular disease278, and increased levels of homocysteine are
implicated in atherosclerosis and vascular disease, in part through promotion of
endothelial injury and thrombotic pathways.279 In a case-control study
homocysteine levels were found to be higher in patients with psoriasis than in
healthy controls, and correlated with increasing psoriasis severity.280, 281 Although
folate deficiency may have a role in this condition, the exact mechanisms involved
in hyperhomocysteinemia remain unclear.280 Vitamin B12 (cyanocobalamin) can be
photolysed by UV light.282 UVB readily destroys tryptophan (Trp) residues of LDL
and HDL. Moreover, LDL and HDL cholesterol are natural carriers of vitamin E
and carotenoids. These two antioxidants are also rapidly bleached by UVB.283 The
64
decrease in serum vitamin B12 during climate therapy can be explained by its
photodegradation.
Role of vitamin D in the pathogenesis of type 2 diabetes mellitus
Increase in HDL-cholesterol and decrease in HbA1c during climate therapy could
be explained by several factors. One possible mechanism could be a direct effect of
vitamin D on insulin sensitivity.124 Another is that sun exposure usually implies
greater outdoor physical activity, leading to beneficial effects on lipids and insulin
sensitivity, unrelated to serum 25(OH)D concentrations.124 The diet might also
influence glucose and lipid metabolism. The observed associations between vitamin
D, insulin, and glucose metabolism in humans have not yet been confirmed by
intervention studies and, hence, a causal association has not been established.124
Although climate therapy did not change the basal glucose levels of the patients,
the HbA1c levels decreased about 10 %, indicating improved insulin sensitivity.
There are seasonal variations in lipid levels, with levels of total cholesterol, LDL,
triglyceride and lipoprotein A being highest in the winter when UVB induced
production of vitamin D3 is at its minimum.284 Serum concentrations of 25(OH)D
at baseline correlated positively with serum HDL at baseline, consistent with a
previously published study.285 We found no other correlation between vitamin D
and blood lipids.
Maintained body weight speaks against any caloric reduction as an explanation for
the improved glucose and lipid metabolism.
Heliotherapy enhanced vitamin D3 production in patients with psoriasis. This
might be one reason why positive effects were also seen on glucose and lipid
metabolism.
65
Conclusions
UVB therapy improved psoriasis scores, increased serum 25(OH)D
synthesis and reduced serum PTH concentrations in postmenopausal
women (paper I).
Postmenopausal women with psoriasis had higher BMD than age-matched
controls. Higher body weight, physical activity and UVB exposure could
explain this finding (paper II).
Serum concentrations of 25(OH)D in psoriasis patients increased less with
narrowband UVB than with broadband UVB phototherapy. Psoriasis
improved on both regimens (paper III).
Climate therapy administered in Gran Canaria enhanced vitamin D3
production and improved PASI scores in patients with psoriasis. Positive
effects were also seen on glucose and lipid metabolism (paper IV).
66
Future prospects
Extensive research has been carried out on the synthesis of vitamin D and the
importance of this vitamin for human health in general. This thesis contributes to
the research including vitamin D status in psoriasis patients during treatment with
phototherapy.
The studies showed that the production of 25(OH)D did not correlate to the dose
of UVB indicating a complexity in the regulation of the synthesis and metabolism
of vitamin D.
It is not known whether skin affected by psoriasis or eczema differ in vitamin D
production compared to normal skin. Our hypothesis is that different skin regions
vary in their production of vitamin D.
More research is needed to develop safe intentional recommendations for sun
exposure to obtain appropriate vitamin D production especially in the Scandinavian
population. It is also necessary to establish new recommendations for daily vitamin
D supplements in different patient groups.
Possible future research includes studies on:
The capacity of psoriatic skin to produce and metabolise vitamin D during
UVB exposure using the microdialysis technique.
The ability of different skin areas to synthesize vitamin D for optimizing the
recommendations for safe sun exposure.
Vitamin D status in different population groups at increased risk of vitamin
D deficiency including dark-skinned people and immigrants.
To clarify the effect of climate therapy and life style factors such as physical
activity and diet, on psoriasis, vitamin D, insulin sensitivity and lipid status.
To clarify the relationship between psoriasis, type 2 diabetes and
cardiovascular disease.
67
Acknowledgements
I wish to express my deep appreciation and sincere gratitude to:
Anne Lene Krogstad, my supervisor, for scientific guidance, generous support,
stimulating supervision, fruitful discussions, and for your kindness, elegance,
generosity and belief in me.
Kerstin Landin-Wilhelmsen, my assisting supervisor, my role model, for teaching
me so much about scientific writing and proof-reading, for your diligence and your
constructive help and support.
Olle Larkö, my co-supervisor, for your great interest in science, support and
encouragement.
Ann-Marie Wennberg, my co-supervisor, for your friendly support, guidance and
encouraging conversations.
I would like to thank my co-authors Dan Mellström, Lena Hulthen, Lill Tove
Nilsen, Elisabeth Søyland, Peter Abusdal Torjesen, Marit Nenseter and Tor
Arne Hagve for good and stimulating collaboration.
Martin Gillstedt, for statistical advice, and for taking the time and kindly helping
me with all the graphs.
Valter Sundh, for statistical advice and assistance.
Morgan Carlsson, for skilful preparation of posters and help with the illustrations.
Lena Mattsson, for assistance with patients and friendly support over the years.
Inger Forsell, for diligent help with administrative procedures.
Agneta Fälted, for assistance with patients, literature explorations, and friendly
support.
Staff at the outpatient clinic for psoriasis in Majorna for their help and
support, taking care of both patients and samples.
Staff and colleagues at the Department of Dermatology Sahlgrenska University
Hospital, Göteborg for their help and support.
68
Louise Hellner, Lisbet Selvén and staff at the Department for Clinical
Chemistry for their help with all the analyses.
Angelica Jarlert and staff at the Osteoporosis laboratory for the performance
of DEXA measurements.
The Section for Climate therapy, Dept of Rheumatology (Behandlingsreiser) for
their support in carrying out the study on psoriasis patients during heliotherapy at
Gran Canaria.
The staff at the Valle Marina Treatment Centre, at the laboratory of
Immunology and at the Department of Dermatology, Dr Negrin University
Hospital, Las Palmas for careful follow-up of the patients during the study, and
assistance with collecting and storing the samples.
My family:
My parents Magbula and Aziz Ahmic for your endless love and unfailing support
that always gave me confidence, peace and faith.
My husband Edin and our children Amar, Adin and Selma for all your patience,
understanding, support and for your love.
My brother Amer Ahmic and his family for gracious support.
All my relatives and friends in Bosnia and Herzegovina and in Sweden.
This work was supported by grants from the federal government under the ALF
agreement, the Göteborg Medical Society, the Regional Health Authority of West
Sweden, the Edward Welander and Finsen’s Foundation, the LUA Foundation at
Sahlgrenska University Hospital and grants from the Section for Climate therapy,
Dept of Rheumatology, Rikshospitalet University Hospital Oslo.
69
References
1 Bikle DD. Vitamin D: Production, metabolism, and mechanisms of action.
In: Diseases of Bone and Calcium Metabolism, Hyperparathyroidism, Vol. Chapter 3:
Endotex.com. 2004; 1-27.
2 Holick MF. The vitamin D epidemic and its health consequences. J Nutr
2005; 135: 2739S-48S.
3 Sniadecki J. Cited by W. Mozolowski: "Jedrzej Sniadecki (1768-1883) on the
cure of rickets". Nature 1939; 143:121.
4 Owen I. Geografical distribution of rickets, acute and subacute rheumatism,
chorea, cancer and urinary calculus in the British Islands. Br Med J 1889; 1:
113-6.
5 Palm T. The geographic distribution and etiology of rickets. Practitioner 1890;
45: 321-42.
6 Hess AF. On the Induction of Antirachitic Properties in Rations by
Exposure to Light. Science 1924; 60: 269.
7 Steenbock H. The Induction of Growth Promoting and Calcifying
Properties in a Ration by Exposure to Light. Science 1924; 60: 224-5.
8 Holick MF. The cutaneous photosynthesis of previtamin D3: a unique
photoendocrine system. J Invest Dermatol 1981; 77: 51-8.
9 Holick MF, MacLaughlin JA, Doppelt SH. Regulation of cutaneous
previtamin D3 photosynthesis in man: skin pigment is not an essential
regulator. Science 1981; 211: 590-3.
10 Holick MF. Vitamin D: A millenium perspective. J Cell Biochem 2003; 88:
296-307.
11 Webb AR, DeCosta BR, Holick MF. Sunlight regulates the cutaneous
production of vitamin D3 by causing its photodegradation. J Clin Endocrinol
Metab 1989; 68: 882-7.
12 MacLaughlin J, Holick MF. Aging decreases the capacity of human skin to
produce vitamin D3. J Clin Invest 1985; 76: 1536-8.
13 Need AG, Morris HA, Horowitz M et al. Effects of skin thickness, age, body
fat, and sunlight on serum 25-hydroxyvitamin D. Am J Clin Nutr 1993; 58:
882-5.
14 Clemens TL, Adams JS, Henderson SL et al. Increased skin pigment reduces
the capacity of skin to synthesise vitamin D3. Lancet 1982; 1: 74-6.
70
15 Matsuoka LY, Wortsman J, Hollis BW. Use of topical sunscreen for the
evaluation of regional synthesis of vitamin D3. J Am Acad Dermatol 1990; 22:
772-5.
16 Matsuoka LY, Wortsman J, Dannenberg MJ et al. Clothing prevents
ultraviolet-B radiation-dependent photosynthesis of vitamin D3. J Clin
Endocrinol Metab 1992; 75: 1099-103.
17 Holick MF. McCollum Award Lecture, 1994: vitamin D--new horizons for
the 21st century. Am J Clin Nutr 1994; 60: 619-30.
18 Webb AR, Kline L, Holick MF. Influence of season and latitude on the
cutaneous synthesis of vitamin D3: exposure to winter sunlight in Boston
and Edmonton will not promote vitamin D3 synthesis in human skin. J Clin
Endocrinol Metab 1988; 67: 373-8.
19 Wortsman J, Matsuoka LY, Chen TC et al. Decreased bioavailability of
vitamin D in obesity. Am J Clin Nutr 2000; 72: 690-3.
20 Verboven C, Rabijns A, De Maeyer M et al. A structural basis for the unique
binding features of the human vitamin D-binding protein. Nat Struct Biol
2002; 9: 131-6.
21 Bar M, Domaschke D, Meye A et al. Wavelength-dependent induction of
CYP24A1-mRNA after UVB-triggered calcitriol synthesis in cultured human
keratinocytes. J Invest Dermatol 2007; 127: 206-13.
22 Segaert S, Garmyn M, Degreef H et al. Retinoic acid modulates the anti-
proliferative effect of 1,25-dihydroxyvitamin D3 in cultured human
epidermal keratinocytes. J Invest Dermatol 1997; 109: 46-54.
23 Vantieghem K, Kissmeyer AM, De Haes P et al. UVB-induced production of
1,25-dihydroxyvitamin D3 and vitamin D activity in human keratinocytes
pretreated with a sterol Delta7-reductase inhibitor. J Cell Biochem 2006; 98:
81-92.
24 Vantieghem K, De Haes P, Bouillon R et al. Dermal fibroblasts pretreated
with a sterol Delta7-reductase inhibitor produce 25-hydroxyvitamin D3
upon UVB irradiation. J Photochem Photobiol B 2006; 85: 72-8.
25 Jones G, Strugnell SA, DeLuca HF. Current understanding of the molecular
actions of vitamin D. Physiol Rev 1998; 78: 1193-231.
26 Lehmann B, Querings K, Reichrath J. Vitamin D and skin: new aspects for
dermatology. Exp Dermatol 2004; 13 Suppl 4: 11-5.
27 De Haes P, Garmyn M, Degreef H et al. 1,25-Dihydroxyvitamin D3 inhibits
ultraviolet B-induced apoptosis, Jun kinase activation, and interleukin-6
production in primary human keratinocytes. J Cell Biochem 2003; 89: 663-73.
71
28 De Haes P, Garmyn M, Verstuyf A et al. 1,25-Dihydroxyvitamin D3 and
analogues protect primary human keratinocytes against UVB-induced DNA
damage. J Photochem Photobiol B 2005; 78: 141-8.
29 Chapuy MC, Preziosi P, Maamer M et al. Prevalence of vitamin D
insufficiency in an adult normal population. Osteoporos Int 1997; 7: 439-43.
30 Lips P. Vitamin D deficiency and secondary hyperparathyroidism in the
elderly: consequences for bone loss and fractures and therapeutic
implications. Endocr Rev 2001; 22: 477-501.
31 Malabanan A, Veronikis IE, Holick MF. Redefining vitamin D insufficiency.
Lancet 1998; 351: 805-6.
32 Dawson-Hughes B, Heaney RP, Holick MF et al. Estimates of optimal
vitamin D status. Osteoporos Int 2005; 16: 713-6.
33 Grant WB, Holick MF. Benefits and requirements of vitamin D for optimal
health: a review. Altern Med Rev 2005; 10: 94-111.
34 Zittermann A. Vitamin D in preventive medicine: are we ignoring the
evidence? Br J Nutr 2003; 89: 552-72.
35 Favus MJ. Postmenopausal osteoporosis and the detection of so-called
secondary causes of low bone density. J Clin Endocrinol Metab 2005; 90: 3800-
1.
36 Van Cromphaut SJ, Dewerchin M, Hoenderop JG et al. Duodenal calcium
absorption in vitamin D receptor-knockout mice: functional and molecular
aspects. Proc Natl Acad Sci U S A 2001; 98: 13324-9.
37 Holick MF. Vitamin D: importance in the prevention of cancers, type 1
diabetes, heart disease, and osteoporosis. Am J Clin Nutr 2004; 79: 362-71.
38 Huang JC, Sakata T, Pfleger LL et al. PTH differentially regulates expression
of RANKL and OPG. J Bone Miner Res 2004; 19: 235-44.
39 Holick MF. Sunlight and vitamin D: both good for cardiovascular health. J
Gen Intern Med 2002; 17: 733-5.
40 Stumpf WE, Sar M, Reid FA et al. Target cells for 1,25-dihydroxyvitamin D3
in intestinal tract, stomach, kidney, skin, pituitary, and parathyroid. Science
1979; 206: 1188-90.
41 Adorini L. 1,25-Dihydroxyvitamin D3 analogs as potential therapies in
transplantation. Curr Opin Investig Drugs 2002; 3: 1458-63.
42 Garland C, Shekelle RB, Barrett-Connor E et al. Dietary vitamin D and
calcium and risk of colorectal cancer: a 19-year prospective study in men.
Lancet 1985; 1: 307-9.
72
43 Garland CF, Garland FC. Do sunlight and vitamin D reduce the likelihood
of colon cancer? Int J Epidemiol 1980; 9: 227-31.
44 Grant WB. An estimate of premature cancer mortality in the U.S. due to
inadequate doses of solar ultraviolet-B radiation. Cancer 2002; 94: 1867-75.
45 Ahonen MH, Tenkanen L, Teppo L et al. Prostate cancer risk and
prediagnostic serum 25-hydroxyvitamin D levels (Finland). Cancer Causes
Control 2000; 11: 847-52.
46 Bikle DD, Nemanic MK, Gee E et al. 1,25-Dihydroxyvitamin D3 production
by human keratinocytes. Kinetics and regulation. J Clin Invest 1986; 78: 557-
66.
47 Adams JS, Gacad MA, Anders A et al. Biochemical indicators of disordered
vitamin D and calcium homeostasis in sarcoidosis. Sarcoidosis 1986; 3: 1-6.
48 Schwartz GG, Whitlatch LW, Chen TC et al. Human prostate cells
synthesize 1,25-dihydroxyvitamin D3 from 25-hydroxyvitamin D3. Cancer
Epidemiol Biomarkers Prev 1998; 7: 391-5.
49 Tangpricha V, Flanagan JN, Whitlatch LW et al. 25-hydroxyvitamin D-
1alpha-hydroxylase in normal and malignant colon tissue. Lancet 2001; 357:
1673-4.
50 Mawer EB, Hayes ME, Heys SE et al. Constitutive synthesis of 1,25-
dihydroxyvitamin D3 by a human small cell lung cancer cell line. J Clin
Endocrinol Metab 1994; 79: 554-60.
51 Cross HS, Bareis P, Hofer H et al. 25-Hydroxyvitamin D(3)-1alpha-
hydroxylase and vitamin D receptor gene expression in human colonic
mucosa is elevated during early cancerogenesis. Steroids 2001; 66: 287-92.
52 Maas RM, Reus K, Diesel B et al. Amplification and expression of splice
variants of the gene encoding the P450 cytochrome 25-hydroxyvitamin D(3)
1,alpha-hydroxylase (CYP 27B1) in human malignant glioma. Clin Cancer Res
2001; 7: 868-75.
53 Tsoukas CD, Provvedini DM, Manolagas SC. 1,25-dihydroxyvitamin D3: a
novel immunoregulatory hormone. Science 1984; 224: 1438-40.
54 Garland FC, Garland CF, Gorham ED et al. Geographic variation in breast
cancer mortality in the United States: a hypothesis involving exposure to
solar radiation. Prev Med 1990; 19: 614-22.
55 Berwick M, Armstrong BK, Ben-Porat L et al. Sun exposure and mortality
from melanoma. J Natl Cancer Inst 2005; 97: 195-9.
56 Kennedy C, Bajdik CD, Willemze R et al. The influence of painful sunburns
and lifetime sun exposure on the risk of actinic keratoses, seborrheic warts,
73
melanocytic nevi, atypical nevi, and skin cancer. J Invest Dermatol 2003; 120:
1087-93.
57 Bhalla AK, Amento EP, Clemens TL et al. Specific high-affinity receptors
for 1,25-dihydroxyvitamin D3 in human peripheral blood mononuclear cells:
presence in monocytes and induction in T lymphocytes following activation.
J Clin Endocrinol Metab 1983; 57: 1308-10.
58 Mathieu C, Adorini L. The coming of age of 1,25-dihydroxyvitamin D(3)
analogs as immunomodulatory agents. Trends Mol Med 2002; 8: 174-9.
59 Deluca HF, Cantorna MT. Vitamin D: its role and uses in immunology.
FASEB J 2001; 15: 2579-85.
60 Mathieu C, Waer M, Laureys J et al. Prevention of autoimmune diabetes in
NOD mice by 1,25 dihydroxyvitamin D3. Diabetologia 1994; 37: 552-8.
61 Cantorna MT, Hayes CE, DeLuca HF. 1,25-Dihydroxyvitamin D3 reversibly
blocks the progression of relapsing encephalomyelitis, a model of multiple
sclerosis. Proc Natl Acad Sci U S A 1996; 93: 7861-4.
62 Cantorna MT, Munsick C, Bemiss C et al. 1,25-Dihydroxycholecalciferol
prevents and ameliorates symptoms of experimental murine inflammatory
bowel disease. J Nutr 2000; 130: 2648-52.
63 Embry AF, Snowdon LR, Vieth R. Vitamin D and seasonal fluctuations of
gadolinium-enhancing magnetic resonance imaging lesions in multiple
sclerosis. Ann Neurol 2000; 48: 271-2.
64 Merlino LA, Curtis J, Mikuls TR et al. Vitamin D intake is inversely
associated with rheumatoid arthritis: results from the Iowa Women's Health
Study. Arthritis Rheum 2004; 50: 72-7.
65 Hypponen E, Laara E, Reunanen A et al. Intake of vitamin D and risk of
type 1 diabetes: a birth-cohort study. Lancet 2001; 358: 1500-3.
66 Rostand SG. Ultraviolet light may contribute to geographic and racial blood
pressure differences. Hypertension 1997; 30: 150-6.
67 Krause R, Buhring M, Hopfenmuller W et al. Ultraviolet B and blood
pressure. Lancet 1998; 352: 709-10.
68 Zittermann A, Schleithoff SS, Tenderich G et al. Low vitamin D status: a
contributing factor in the pathogenesis of congestive heart failure? J Am Coll
Cardiol 2003; 41: 105-12.
69 Li YC, Kong J, Wei M et al. 1,25-Dihydroxyvitamin D(3) is a negative
endocrine regulator of the renin-angiotensin system. J Clin Invest 2002; 110:
229-38.
74
70 Weishaar RE, Simpson RU. Involvement of vitamin D3 with cardiovascular
function. II. Direct and indirect effects. Am J Physiol 1987; 253: E675-83.
71 O'Connell TD, Weishaar RE, Simpson RU. Regulation of myosin isozyme
expression by vitamin D3 deficiency and 1,25-dihydroxyvitamin D3 in the
rat heart. Endocrinology 1994; 134: 899-905.
72 Zasloff M. Sunlight, vitamin D, and the innate immune defenses of the
human skin. J Invest Dermatol 2005; 125: xvi-xvii.
73 Wang TT, Nestel FP, Bourdeau V et al. Cutting edge: 1,25-dihydroxyvitamin
D3 is a direct inducer of antimicrobial peptide gene expression. J Immunol
2004; 173: 2909-12.
74 Weber G, Heilborn JD, Chamorro Jimenez CI et al. Vitamin D induces the
antimicrobial protein hCAP18 in human skin. J Invest Dermatol 2005; 124:
1080-2.
75 Segaert S, Simonart T. The epidermal vitamin D system and innate
immunity: some more light shed on this unique photoendocrine system?
Dermatology 2008; 217: 7-11.
76 Schauber J, Dorschner RA, Coda AB et al. Injury enhances TLR2 function
and antimicrobial peptide expression through a vitamin D-dependent
mechanism. J Clin Invest 2007; 117: 803-11.
77 Norman AW. Problems relating to the definition of an international unit for
vitamin D and its metabolites. J Nutr 1972; 102: 1243-6.
78 Barger-Lux MJ, Heaney RP, Dowell S et al. Vitamin D and its major
metabolites: serum levels after graded oral dosing in healthy men. Osteoporos
Int 1998; 8: 222-30.
79 Tangpricha V, Turner A, Spina C et al. Tanning is associated with optimal
vitamin D status (serum 25-hydroxyvitamin D concentration) and higher
bone mineral density. Am J Clin Nutr 2004; 80: 1645-9.
80 Bischoff-Ferrari HA, Dietrich T, Orav EJ et al. Positive association between
25-hydroxy vitamin D levels and bone mineral density: a population-based
study of younger and older adults. Am J Med 2004; 116: 634-9.
81 Sedrani SH. Vitamin D status of Saudi men. Trop Geogr Med 1984; 36: 181-7.
82 Sedrani SH, Elidrissy AW, El Arabi KM. Sunlight and vitamin D status in
normal Saudi subjects. Am J Clin Nutr 1983; 38: 129-32.
83 Gladman DD. Psoriatic arthritis. Dermatol Ther 2004; 17: 350-63.
84 Bos JD, de Rie MA, Teunissen MB et al. Psoriasis: dysregulation of innate
immunity. Br J Dermatol 2005; 152: 1098-107.
75
85 Bowcock AM, Krueger JG. Getting under the skin: the immunogenetics of
psoriasis. Nat Rev Immunol 2005; 5: 699-711.
86 Wang H, Peters T, Kess D et al. Activated macrophages are essential in a
murine model for T cell-mediated chronic psoriasiform skin inflammation. J
Clin Invest 2006; 116: 2105-14.
87 Stratis A, Pasparakis M, Rupec RA et al. Pathogenic role for skin
macrophages in a mouse model of keratinocyte-induced psoriasis-like skin
inflammation. J Clin Invest 2006; 116: 2094-104.
88 Clark RA, Kupper TS. Misbehaving macrophages in the pathogenesis of
psoriasis. J Clin Invest 2006; 116: 2084-7.
89 Austin LM, Ozawa M, Kikuchi T et al. The majority of epidermal T cells in
Psoriasis vulgaris lesions can produce type 1 cytokines, interferon-gamma,
interleukin-2, and tumor necrosis factor-alpha, defining TC1 (cytotoxic T
lymphocyte) and TH1 effector populations: a type 1 differentiation bias is
also measured in circulating blood T cells in psoriatic patients. J Invest
Dermatol 1999; 113: 752-9.
90 Ozawa M, Aiba S. Immunopathogenesis of psoriasis. Curr Drug Targets
Inflamm Allergy 2004; 3: 137-44.
91 Fierlbeck G, Rassner G, Muller C. Psoriasis induced at the injection site of
recombinant interferon gamma. Results of immunohistologic investigations.
Arch Dermatol 1990; 126: 351-5.
92 Ettehadi P, Greaves MW, Wallach D et al. Elevated tumour necrosis factor-
alpha (TNF-alpha) biological activity in psoriatic skin lesions. Clin Exp
Immunol 1994; 96: 146-51.
93 Bonifati C, Carducci M, Cordiali Fei P et al. Correlated increases of tumour
necrosis factor-alpha, interleukin-6 and granulocyte monocyte-colony
stimulating factor levels in suction blister fluids and sera of psoriatic
patients--relationships with disease severity. Clin Exp Dermatol 1994; 19: 383-
7.
94 Jacob SE, Nassiri M, Kerdel FA et al. Simultaneous measurement of multiple
Th1 and Th2 serum cytokines in psoriasis and correlation with disease
severity. Mediators Inflamm 2003; 12: 309-13.
95 Kagen MH, McCormick TS, Cooper KD. Regulatory T cells in psoriasis.
Ernst Schering Res Found Workshop 2006: 193-209.
96 Bata-Csorgo Z, Hammerberg C, Voorhees JJ et al. Kinetics and regulation of
human keratinocyte stem cell growth in short-term primary ex vivo culture.
Cooperative growth factors from psoriatic lesional T lymphocytes stimulate
76
proliferation among psoriatic uninvolved, but not normal, stem
keratinocytes. J Clin Invest 1995; 95: 317-27.
97 van der Vleuten CJ, Gerritsen MJ, Steijlen PM et al. A therapeutic approach
to erythrodermic psoriasis: report of a case and a discussion of therapeutic
options. Acta Derm Venereol 1996; 76: 65-7.
98 Bourke JF, Iqbal SJ, Hutchinson PE. The effects of UVB plus calcipotriol
on systemic calcium homeostasis in patients with chronic plaque psoriasis.
Clin Exp Dermatol 1997; 22: 259-61.
99 Fogh K, Kragballe K. Recent developments in vitamin D analogs. Curr
Pharm Des 2000; 6: 961-72.
100 Langner A, Ashton P, Van De Kerkhof PC et al. A long-term multicentre
assessment of the safety and tolerability of calcitriol ointment in the
treatment of chronic plaque psoriasis. Br J Dermatol 1996; 135: 385-9.
101 van de Kerkhof PC, Berth-Jones J, Griffiths CE et al. Long-term efficacy and
safety of tacalcitol ointment in patients with chronic plaque psoriasis. Br J
Dermatol 2002; 146: 414-22.
102 Kragballe K. Vitamin D and UVB radiation therapy. Cutis 2002; 70: 9-12.
103 Feldman D, Chen T, Hirst M et al. Demonstration of 1,25-dihydroxyvitamin
D3 receptors in human skin biopsies. J Clin Endocrinol Metab 1980; 51: 1463-
5.
104 Provvedini DM, Tsoukas CD, Deftos LJ et al. 1,25-dihydroxyvitamin D3
receptors in human leukocytes. Science 1983; 221: 1181-3.
105 Morimoto S, Kumahara Y. A patient with psoriasis cured by 1 alpha-
hydroxyvitamin D3. Med J Osaka Univ 1985; 35: 51-4.
106 Nagpal S, Lu J, Boehm MF. Vitamin D analogs: mechanism of action and
therapeutic applications. Curr Med Chem 2001; 8: 1661-79.
107 Takahashi H, Ibe M, Kinouchi M et al. Similarly potent action of 1,25-
dihydroxyvitamin D3 and its analogues, tacalcitol, calcipotriol, and
maxacalcitol on normal human keratinocyte proliferation and differentiation.
J Dermatol Sci 2003; 31: 21-8.
108 Pillai S, Bikle DD. Role of intracellular-free calcium in the cornified
envelope formation of keratinocytes: differences in the mode of action of
extracellular calcium and 1,25 dihydroxyvitamin D3. J Cell Physiol 1991; 146:
94-100.
109 Ratnam AV, Bikle DD, Cho JK. 1,25 dihydroxyvitamin D3 enhances the
calcium response of keratinocytes. J Cell Physiol 1999; 178: 188-96.
77
110 Manolagas SC, Provvedini DM, Tsoukas CD. Interactions of 1,25-
dihydroxyvitamin D3 and the immune system. Mol Cell Endocrinol 1985; 43:
113-22.
111 Muller K, Bendtzen K. 1,25-Dihydroxyvitamin D3 as a natural regulator of
human immune functions. J Investig Dermatol Symp Proc 1996; 1: 68-71.
112 Pinette KV, Yee YK, Amegadzie BY et al. Vitamin D receptor as a drug
discovery target. Mini Rev Med Chem 2003; 3: 193-204.
113 Tobler A, Gasson J, Reichel H et al. Granulocyte-macrophage colony-
stimulating factor. Sensitive and receptor-mediated regulation by 1,25-
dihydroxyvitamin D3 in normal human peripheral blood lymphocytes. J Clin
Invest 1987; 79: 1700-5.
114 Kang S, Yi S, Griffiths CE et al. Calcipotriene-induced improvement in
psoriasis is associated with reduced interleukin-8 and increased interleukin-
10 levels within lesions. Br J Dermatol 1998; 138: 77-83.
115 Michel G, Gailis A, Jarzebska-Deussen B et al. 1,25-(OH)2-vitamin D3 and
calcipotriol induce IL-10 receptor gene expression in human epidermal cells.
Inflamm Res 1997; 46: 32-4.
116 Penna G, Adorini L. 1 Alpha,25-dihydroxyvitamin D3 inhibits
differentiation, maturation, activation, and survival of dendritic cells leading
to impaired alloreactive T cell activation. J Immunol 2000; 164: 2405-11.
117 Adorini L, Penna G, Giarratana N et al. Tolerogenic dendritic cells induced
by vitamin D receptor ligands enhance regulatory T cells inhibiting allograft
rejection and autoimmune diseases. J Cell Biochem 2003; 88: 227-33.
118 Gottlieb AB, Chao C, Dann F. Psoriasis comorbidities. J Dermatolog Treat
2008; 19: 5-21.
119 Christophers E. Comorbidities in psoriasis. Clin Dermatol 2007; 25: 529-34.
120 Gottlieb AB, Dann F, Menter A. Psoriasis and the metabolic syndrome. J
Drugs Dermatol 2008; 7: 563-72.
121 Sommer DM, Jenisch S, Suchan M et al. Increased prevalence of the
metabolic syndrome in patients with moderate to severe psoriasis. Arch
Dermatol Res 2006; 298: 321-8.
122 Akhyani M, Ehsani AH, Robati RM et al. The lipid profile in psoriasis: a
controlled study. J Eur Acad Dermatol Venereol 2007; 21: 1330-2.
123 Palomer X, Gonzalez-Clemente JM, Blanco-Vaca F et al. Role of vitamin D
in the pathogenesis of type 2 diabetes mellitus. Diabetes Obes Metab 2008; 10:
185-97.
78
124 Tai K, Need AG, Horowitz M et al. Vitamin D, glucose, insulin, and insulin
sensitivity. Nutrition 2008; 24: 279-85.
125 Procopio M, Magro G, Cesario F et al. The oral glucose tolerance test reveals
a high frequency of both impaired glucose tolerance and undiagnosed Type
2 diabetes mellitus in primary hyperparathyroidism. Diabet Med 2002; 19:
958-61.
126 Taylor WH, Khaleeli AA. Prevalence of primary hyperparathyroidism in
patients with diabetes mellitus. Diabet Med 1997; 14: 386-9.
127 Luong K, Nguyen LT, Nguyen DN. The role of vitamin D in protecting
type 1 diabetes mellitus. Diabetes Metab Res Rev 2005; 21: 338-46.
128 Mauricio D, Mandrup-Poulsen T, Nerup J. Vitamin D analogues in insulin-
dependent diabetes mellitus and other autoimmune diseases: a therapeutic
perspective. Diabetes Metab Rev 1996; 12: 57-68.
129 Hewison M, Gacad MA, Lemire J et al. Vitamin D as a cytokine and
hematopoetic factor. Rev Endocr Metab Disord 2001; 2: 217-27.
130 Norman PE, Powell JT. Vitamin D, shedding light on the development of
disease in peripheral arteries. Arterioscler Thromb Vasc Biol 2005; 25: 39-46.
131 Lemire JM. Immunomodulatory actions of 1,25-dihydroxyvitamin D3. J
Steroid Biochem Mol Biol 1995; 53: 599-602.
132 Millard TP, Antoniades L, Evans AV et al. Bone mineral density of patients
with chronic plaque psoriasis. Clin Exp Dermatol 2001; 26: 446-8.
133 Nymann P, Kollerup G, Jemec GB et al. Decreased bone mineral density in
patients with pustulosis palmaris et plantaris. Dermatology 1996; 192: 307-11.
134 Frediani B, Allegri A, Falsetti P et al. Bone mineral density in patients with
psoriatic arthritis. J Rheumatol 2001; 28: 138-43.
135 Chapuy MC, Arlot ME, Duboeuf F et al. Vitamin D3 and calcium to prevent
hip fractures in the elderly women. N Engl J Med 1992; 327: 1637-42.
136 Chel VG, Ooms ME, Popp-Snijders C et al. Ultraviolet irradiation corrects
vitamin D deficiency and suppresses secondary hyperparathyroidism in the
elderly. J Bone Miner Res 1998; 13: 1238-42.
137 Lilliu H, Pamphile R, Chapuy MC et al. Calcium-vitamin D3
supplementation is cost-effective in hip fractures prevention. Maturitas 2003;
44: 299-305.
138 Landin-Wilhelmsen K, Wilhelmsen L, Wilske J et al. Sunlight increases serum
25(OH) vitamin D concentration whereas 1,25(OH)2D3 is unaffected.
Results from a general population study in Goteborg, Sweden (The WHO
MONICA Project). Eur J Clin Nutr 1995; 49: 400-7.
79
139 Wassberg C, Backvall H, Diffey B et al. Enhanced epidermal ultraviolet
responses in chronically sun-exposed skin are dependent on previous sun
exposure. Acta Derm Venereol 2003; 83: 254-61.
140 Chadwick CA, Potten CS, Nikaido O et al. The detection of cyclobutane
thymine dimers, (6-4) photolesions and the Dewar photoisomers in sections
of UV-irradiated human skin using specific antibodies, and the
demonstration of depth penetration effects. J Photochem Photobiol B 1995; 28:
163-70.
141 Campbell C, Quinn AG, Angus B et al. Wavelength specific patterns of p53
induction in human skin following exposure to UV radiation. Cancer Res
1993; 53: 2697-9.
142 Hoffmann K, Kaspar K, Altmeyer P et al. UV transmission measurements of
small skin specimens with special quartz cuvettes. Dermatology 2000; 201: 307-
11.
143 Farr PM, Diffey BL, Steele MC. A preliminary study on the in vivo
transmission of light through psoriatic plaques. Photodermatol 1984; 1: 87-90.
144 Krutmann J, Morita, A. and Elmets, A. Mechanisms of
photo(chemo)therapy. Springer pp. 56-59 2001.
145 Trautinger F. Mechanisms of photodamage of the skin and its functional
consequences for skin ageing. Clin Exp Dermatol 2001; 26: 573-7.
146 Clingen PH, Arlett CF, Cole J et al. Correlation of UVC and UVB
cytotoxicity with the induction of specific photoproducts in T-lymphocytes
and fibroblasts from normal human donors. Photochem Photobiol 1995; 61:
163-70.
147 Dalziel KL. Aspects of cutaneous ageing. Clin Exp Dermatol 1991; 16: 315-
23.
148 Diffey BL, Farr PM. An appraisal of ultraviolet lamps used for the
phototherapy of psoriasis. Br J Dermatol 1987; 117: 49-56.
149 Parrish JA. Ultraviolet phototherapy of psoriasis. Pharmacol Ther 1981; 15:
313-20.
150 Goeckerman WH. The treatment of psoriasis. Northwest Med (1925); 24 pp.
229-31.
151 Honigsmann H. Phototherapy for psoriasis. Clin Exp Dermatol 2001; 26: 343-
50.
152 Ingram JT. The approach to psoriasis. Br Med J 1953; 2: 591-4.
80
153 Parrish JA, Fitzpatrick TB, Tanenbaum L et al. Photochemotherapy of
psoriasis with oral methoxsalen and longwave ultraviolet light. N Engl J Med
1974; 291: 1207-11.
154 Wolff K, Honigsmann H, Gschnait F et al. [Photochemotherapy of psoriasis:
clinical experiences with 152 patients (author's transl)]. Dtsch Med Wochenschr
1975; 100: 2471-7, 1497.
155 Feldman SR, Garton R, Averett W et al. Strategy to manage the treatment of
severe psoriasis: considerations of efficacy, safety and cost. Expert Opin
Pharmacother 2003; 4: 1525-33.
156 Fischer T, Alsins J. Treatment of psoriasis with trioxsalen baths and
dysprosium lamps. Acta Derm Venereol 1976; 56: 383-90.
157 Parrish JA, Jaenicke KF. Action spectrum for phototherapy of psoriasis. J
Invest Dermatol 1981; 76: 359-62.
158 Green C, Ferguson J, Lakshmipathi T et al. 311 nm UVB phototherapy--an
effective treatment for psoriasis. Br J Dermatol 1988; 119: 691-6.
159 Karvonen J, Kokkonen EL, Ruotsalainen E. 311 nm UVB lamps in the
treatment of psoriasis with the Ingram regimen. Acta Derm Venereol 1989; 69:
82-5.
160 van Weelden H, De La Faille HB, Young E et al. A new development in
UVB phototherapy of psoriasis. Br J Dermatol 1988; 119: 11-9.
161 Berneburg M, Rocken M, Benedix F. Phototherapy with narrowband vs
broadband UVB. Acta Derm Venereol 2005; 85: 98-108.
162 Coven TR, Burack LH, Gilleaudeau R et al. Narrowband UV-B produces
superior clinical and histopathological resolution of moderate-to-severe
psoriasis in patients compared with broadband UV-B. Arch Dermatol 1997;
133: 1514-22.
163 Dawe RS, Cameron H, Yule S et al. A randomized controlled trial of
narrowband ultraviolet B vs bath-psoralen plus ultraviolet A
photochemotherapy for psoriasis. Br J Dermatol 2003; 148: 1194-204.
164 Pasic A, Ceovic R, Lipozencic J et al. Phototherapy in pediatric patients.
Pediatr Dermatol 2003; 20: 71-7.
165 Walters IB, Burack LH, Coven TR et al. Suberythemogenic narrow-band
UVB is markedly more effective than conventional UVB in treatment of
psoriasis vulgaris. J Am Acad Dermatol 1999; 40: 893-900.
166 Diffey BL. Towards optimal regimens for the UVB phototherapy of
psoriasis: a mathematical model. Acta Derm Venereol 2004; 84: 259-64.
81
167 Dawe RS, Wainwright NJ, Cameron H et al. Narrow-band (TL-01) ultraviolet
B phototherapy for chronic plaque psoriasis: three times or five times weekly
treatment? Br J Dermatol 1998; 138: 833-9.
168 Cameron H, Dawe RS, Yule S et al. A randomized, observer-blinded trial of
twice vs. three times weekly narrowband ultraviolet B phototherapy for
chronic plaque psoriasis. Br J Dermatol 2002; 147: 973-8.
169 Wainwright NJ, Dawe RS, Ferguson J. Narrowband ultraviolet B (TL-01)
phototherapy for psoriasis: which incremental regimen? Br J Dermatol 1998;
139: 410-4.
170 Zanolli M. Phototherapy treatment of psoriasis today. J Am Acad Dermatol
2003; 49: S78-86.
171 Duthie MS, Kimber I, Norval M. The effects of ultraviolet radiation on the
human immune system. Br J Dermatol 1999; 140: 995-1009.
172 Sethi G, Sodhi A. Role of p38 mitogen-activated protein kinase and caspases
in UV-B-induced apoptosis of murine peritoneal macrophages. Photochem
Photobiol 2004; 79: 48-54.
173 Krueger JG, Wolfe JT, Nabeya RT et al. Successful ultraviolet B treatment of
psoriasis is accompanied by a reversal of keratinocyte pathology and by
selective depletion of intraepidermal T cells. J Exp Med 1995; 182: 2057-68.
174 Ozawa M, Ferenczi K, Kikuchi T et al. 312-nanometer ultraviolet B light
(narrow-band UVB) induces apoptosis of T cells within psoriatic lesions. J
Exp Med 1999; 189: 711-8.
175 Okamoto H, Horio T, Maeda M. Alteration of lymphocyte functions by 8-
methoxypsoralen and long-wave ultraviolet radiation. II. The effect of in
vivo PUVA on IL-2 production. J Invest Dermatol 1987; 89: 24-6.
176 Kripke ML, Morison WL, Parrish JA. Systemic suppression of contact
hypersensitivity in mice by psoralen plus UVA radiation (PUVA). J Invest
Dermatol 1983; 81: 87-92.
177 Aubin F, Mousson C. Ultraviolet light-induced regulatory (suppressor) T
cells: an approach for promoting induction of operational allograft
tolerance? Transplantation 2004; 77: S29-31.
178 Weitzen ML, Bonavida B. Mechanism of inhibition of human natural killer
activity by ultraviolet radiation. J Immunol 1984; 133: 3128-32.
179 Kasahara S, Aizawa K, Okamiya M et al. UVB irradiation suppresses
cytokine production and innate cellular immune functions in mice. Cytokine
2001; 14: 104-11.
82
180 Schneider LA, Hinrichs R, Scharffetter-Kochanek K. Phototherapy and
photochemotherapy. Clin Dermatol 2008; 26: 464-76.
181 Schneider LA, Bloch W, Kopp K et al. 8-Isoprostane is a dose-related
biomarker for photo-oxidative ultraviolet (UV) B damage in vivo: a pilot
study with personal UV dosimetry. Br J Dermatol 2006; 154: 1147-54.
182 Potten CS, Chadwick CA, Cohen AJ et al. DNA damage in UV-irradiated
human skin in vivo: automated direct measurement by image analysis
(thymine dimers) compared with indirect measurement (unscheduled DNA
synthesis) and protection by 5-methoxypsoralen. Int J Radiat Biol 1993; 63:
313-24.
183 Greinert R, Boguhn O, Harder D et al. The dose dependence of cyclobutane
dimer induction and repair in UVB-irradiated human keratinocytes. Photochem
Photobiol 2000; 72: 701-8.
184 Brenneisen P, Blaudschun R, Gille J et al. Essential role of an activator
protein-2 (AP-2)/specificity protein 1 (Sp1) cluster in the UVB-mediated
induction of the human vascular endothelial growth factor in HaCaT
keratinocytes. Biochem J 2003; 369: 341-9.
185 Snellman E, Lauharanta J, Reunanen A et al. Effect of heliotherapy on skin
and joint symptoms in psoriasis: a 6-month follow-up study. Br J Dermatol
1993; 128: 172-7.
186 Colipa. International Sun Protection Factor Test Metho. In: The European
Cosmetic Toiletry and Perfumery Association, Vol. 12. 2006.
187 McKinlay AFaD, B. L. A Reference Action Spectrum for Ultraviolet
Induced Erythema in Human Skin. CIE-J 1987; 6,: 17-22.
188 Leszczynski K JK, Ylianttila L, Visuri R, Blumthaler M. Report of the
WMO/STUK intercomparison of erythemally-weighted solar UV
radiometers. In: Vol. Spring/Summer. Geneva:: World Meteorological
Organization (WMO). 1995.
189 Seckmeyer G BA, Bernard G, Blumthaler M, Booth C R., Lantz K,
McKenzie R L. Instruments to Measure Solar Ultraviolet Radiation. Part 2:
Broadband Instruments Measuring Erythemally Weighted Solar Irradiance. .
In, Vol. WMO/GAW Report No. 164. Geneva: Geneva: World
Meteorological Organization (WMO). 2008.
190 Mayer B, Kylling A. Technical note: The libRadtran software package for
radiative transfer calculations - description and examples of use. Atmos.
Chem. Phys. 2005; 5: 1855-77.
191 NASA. Total ozone mapping spectrometer. 2006.
83
192 Holick MF. Environmental factors that influence the cutaneous production
of vitamin D. Am J Clin Nutr 1995; 61: 638S-45S.
193 Lehmann B, Knuschke P, Meurer M. The UVB-induced synthesis of vitamin
D3 and 1alpha,25-dihydroxyvitamin D3 (calcitriol) in organotypic cultures of
keratinocytes: effectiveness of the narrowband Philips TL-01 lamp (311 nm).
J Steroid Biochem Mol Biol 2007; 103: 682-5.
194 Leenutaphong V, Sudtim S. A comparison of erythema efficacy of ultraviolet
B irradiation from Philips TL12 and TL01 lamps. Photodermatol Photoimmunol
Photomed 1998; 14: 112-5.
195 Holick MF, Chen TC. Vitamin D deficiency: a worldwide problem with
health consequences. Am J Clin Nutr 2008; 87: 1080S-6S.
196 Holick MF. Sunlight, UV-radiation, vitamin D and skin cancer: how much
sunlight do we need? Adv Exp Med Biol 2008; 624: 1-15.
197 Barth J, Gerlach B, Knuschke P et al. Serum 25(OH)D3 and ultraviolet
exposure of residents in an old people's home in Germany. Photodermatol
Photoimmunol Photomed 1992; 9: 229-31.
198 Holick MF, Matsuoka LY, Wortsman J. Age, vitamin D, and solar
ultraviolet. Lancet 1989; 2: 1104-5.
199 Chen TC, Chimeh F, Lu Z et al. Factors that influence the cutaneous
synthesis and dietary sources of vitamin D. Arch Biochem Biophys 2007; 460:
213-7.
200 Devgun MS, Paterson CR, Johnson BE et al. Vitamin D nutrition in relation
to season and occupation. Am J Clin Nutr 1981; 34: 1501-4.
201 Armas LA, Dowell S, Akhter M et al. Ultraviolet-B radiation increases serum
25-hydroxyvitamin D levels: the effect of UVB dose and skin color. J Am
Acad Dermatol 2007; 57: 588-93.
202 Porojnicu AC, Bruland OS, Aksnes L et al. Sun beds and cod liver oil as
vitamin D sources. J Photochem Photobiol B 2008; 91: 125-31.
203 Olds WJ, McKinley AR, Moore MR et al. In vitro model of vitamin D(3)
(Cholecalciferol) synthesis by UV radiation: Dose-response relationships. J
Photochem Photobiol B 2008; 93: 88-93.
204 McCarty CA. Sunlight exposure assessment: can we accurately assess vitamin
D exposure from sunlight questionnaires? Am J Clin Nutr 2008; 87: 1097S-
101S.
205 Barbagallo J, Spann CT, Tutrone WD et al. Narrowband UVB phototherapy
for the treatment of psoriasis: a review and update. Cutis 2001; 68: 345-7.
84
206 Storbeck K, Holzle E, Schurer N et al. Narrow-band UVB (311 nm) versus
conventional broad-band UVB with and without dithranol in phototherapy
for psoriasis. J Am Acad Dermatol 1993; 28: 227-31.
207 Larko O. Treatment of psoriasis with a new UVB-lamp. Acta Derm Venereol
1989; 69: 357-9.
208 Schiener R, Behrens-Williams SC, Pillekamp H et al. Calcipotriol vs.
tazarotene as combination therapy with narrowband ultraviolet B (311 nm):
efficacy in patients with severe psoriasis. Br J Dermatol 2000; 143: 1275-8.
209 Snellman E, Jansen CT, Lauharanta J et al. Solar ultraviolet (UV) radiation
and UV doses received by patients during four-week climate therapy periods
in the Canary Islands. Photodermatol Photoimmunol Photomed 1992; 9: 40-3.
210 Thieden E, Agren MS, Wulf HC. The wrist is a reliable body site for
personal dosimetry of ultraviolet radiation. Photodermatol Photoimmunol
Photomed 2000; 16: 57-61.
211 Diffey BL, Larko O, Swanbeck G. UV-B doses received during different
outdoor activities and UV-B treatment of psoriasis. Br J Dermatol 1982; 106:
33-41.
212 Herlihy E, Gies PH, Roy CR et al. Personal dosimetry of solar UV radiation
for different outdoor activities. Photochem Photobiol 1994; 60: 288-94.
213 Lips P. Vitamin D deficiency and osteoporosis: the role of vitamin D
deficiency and treatment with vitamin D and analogues in the prevention of
osteoporosis-related fractures. Eur J Clin Invest 1996; 26: 436-42.
214 DeLuca HF. The vitamin D story: a collaborative effort of basic science and
clinical medicine. Faseb J 1988; 2: 224-36.
215 Smith EL, Walworth NC, Holick MF. Effect of 1 alpha,25-dihydroxyvitamin
D3 on the morphologic and biochemical differentiation of cultured human
epidermal keratinocytes grown in serum-free conditions. J Invest Dermatol
1986; 86: 709-14.
216 Kragballe K, Wildfang IL. Calcipotriol (MC 903), a novel vitamin D3
analogue stimulates terminal differentiation and inhibits proliferation of
cultured human keratinocytes. Arch Dermatol Res 1990; 282: 164-7.
217 Binderup L, Bramm E. Effects of a novel vitamin D analogue MC903 on
cell proliferation and differentiation in vitro and on calcium metabolism in
vivo. Biochem Pharmacol 1988; 37: 889-95.
218 Armas LA, Hollis BW, Heaney RP. Vitamin D2 is much less effective than
vitamin D3 in humans. J Clin Endocrinol Metab 2004; 89: 5387-91.
85
219 Holick MF, Siris ES, Binkley N et al. Prevalence of Vitamin D Inadequacy
Among Postmenopausal North American Women Receiving Osteoporosis
Therapy. Obstet Gynecol Surv 2005; 60: 658-9.
220 Chesney RW, Rosen JF, Hamstra AJ et al. Absence of seasonal variation in
serum concentrations of 1,25-dihydroxyvitamin D despite a rise in 25-
hydroxyvitamin D in summer. J Clin Endocrinol Metab 1981; 53: 139-42.
221 Lips P, Wiersinga A, van Ginkel FC et al. The effect of vitamin D
supplementation on vitamin D status and parathyroid function in elderly
subjects. J Clin Endocrinol Metab 1988; 67: 644-50.
222 Bouillon RA, Auwerx JH, Lissens WD et al. Vitamin D status in the elderly:
seasonal substrate deficiency causes 1,25-dihydroxycholecalciferol deficiency.
Am J Clin Nutr 1987; 45: 755-63.
223 Adams JS, Clemens TL, Parrish JA et al. Vitamin-D synthesis and
metabolism after ultraviolet irradiation of normal and vitamin-D-deficient
subjects. N Engl J Med 1982; 306: 722-5.
224 Landin-Wilhelmsen K, Wilhelmsen L, Lappas G et al. Serum intact
parathyroid hormone in a random population sample of men and women:
relationship to anthropometry, life-style factors, blood pressure, and vitamin
D. Calcif Tissue Int 1995; 56: 104-8.
225 Kragballe K. Vitamin D in Dermatology. 2000.
226 Shultz TD, Fox J, Heath H, 3rd et al. Do tissues other than the kidney
produce 1,25-dihydroxyvitamin D3 in vivo? A reexamination. Proc Natl Acad
Sci U S A 1983; 80: 1746-50.
227 Bikle DD, Halloran BP, Riviere JE. Production of 1,25 dihydroxyvitamin D3
by perfused pig skin. J Invest Dermatol 1994; 102: 796-8.
228 Vanhooke JL, Prahl JM, Kimmel-Jehan C et al. CYP27B1 null mice with
LacZreporter gene display no 25-hydroxyvitamin D3-1alpha-hydroxylase
promoter activity in the skin. Proc Natl Acad Sci U S A 2006; 103: 75-80.
229 Lehmann B. The vitamin D3 pathway in human skin and its role for
regulation of biological processes. Photochem Photobiol 2005; 81: 1246-51.
230 Lehmann B, Sauter W, Knuschke P et al. Demonstration of UVB-induced
synthesis of 1 alpha,25-dihydroxyvitamin D3 (calcitriol) in human skin by
microdialysis. Arch Dermatol Res 2003; 295: 24-8.
231 Lehmann B, Knuschke P, Meurer M. UVB-induced conversion of 7-
dehydrocholesterol to 1 alpha,25-dihydroxyvitamin D3 (calcitriol) in the
human keratinocyte line HaCaT. Photochem Photobiol 2000; 72: 803-9.
86
232 van de Kerkhof PC. Biological activity of vitamin D analogues in the skin,
with special reference to antipsoriatic mechanisms. Br J Dermatol 1995; 132:
675-82.
233 Hosomi J, Hosoi J, Abe E et al. Regulation of terminal differentiation of
cultured mouse epidermal cells by 1 alpha,25-dihydroxyvitamin D3.
Endocrinology 1983; 113: 1950-7.
234 Merke J, Schwittay D, Furstenberger G et al. Demonstration and
characterization of 1,25-dihydroxyvitamin D3 receptors in basal cells of
epidermis of neonatal and adult mice. Calcif Tissue Int 1985; 37: 257-67.
235 Sahota O, Mundey MK, San P et al. The relationship between vitamin D and
parathyroid hormone: calcium homeostasis, bone turnover, and bone
mineral density in postmenopausal women with established osteoporosis.
Bone 2004; 35: 312-9.
236 Vieth R. Vitamin D supplementation, 25-hydroxyvitamin D concentrations,
and safety. Am J Clin Nutr 1999; 69: 842-56.
237 Landin-Wilhelmsen K, Wilhelmsen L, Bengtsson BA. Postmenopausal
osteoporosis is more related to hormonal aberrations than to lifestyle
factors. Clin Endocrinol (Oxf) 1999; 51: 387-94.
238 Fogelman I, Blake GM. Different approaches to bone densitometry. J Nucl
Med 2000; 41: 2015-25.
239 Riggs BL. Pathogenesis of osteoporosis. Am J Obstet Gynecol 1987; 156: 1342-
6.
240 Cummings SR, Nevitt MC, Browner WS et al. Risk factors for hip fracture in
white women. Study of Osteoporotic Fractures Research Group. N Engl J
Med 1995; 332: 767-73.
241 Johnell O, Gullberg B, Kanis JA et al. Risk factors for hip fracture in
European women: the MEDOS Study. Mediterranean Osteoporosis Study. J
Bone Miner Res 1995; 10: 1802-15.
242 Cummings SR, Melton LJ. Epidemiology and outcomes of osteoporotic
fractures. Lancet 2002; 359: 1761-7.
243 Aniansson A, Zetterberg C, Hedberg M et al. Impaired muscle function with
aging. A background factor in the incidence of fractures of the proximal end
of the femur. Clin Orthop Relat Res 1984: 193-201.
244 Gunnes M, Lehmann EH, Mellstrom D et al. The relationship between
anthropometric measurements and fractures in women. Bone 1996; 19: 407-
13.
87
245 Kanis JA, Johansson H, Oden A et al. A family history of fracture and
fracture risk: a meta-analysis. Bone 2004; 35: 1029-37.
246 Fox KM, Cummings SR, Powell-Threets K et al. Family history and risk of
osteoporotic fracture. Study of Osteoporotic Fractures Research Group.
Osteoporos Int 1998; 8: 557-62.
247 Seeman E, Cooper ME, Hopper JL et al. Effect of early menopause on bone
mass in normal women and patients with osteoporosis. Am J Med 1988; 85:
213-6.
248 Kanis JA. Estrogens, the menopause, and osteoporosis. Bone 1996; 19: 185S-
90S.
249 Kleerekoper M, Brienza RS, Schultz LR et al. Oral contraceptive use may
protect against low bone mass. Henry Ford Hospital Osteoporosis
Cooperative Research Group. Arch Intern Med 1991; 151: 1971-6.
250 Scoutellas V, O'Neill TW, Lunt M et al. Does the presence of
postmenopausal symptoms influence susceptibility to vertebral deformity?
European Vertebral Osteoporosis Study (EVOS) Group. Maturitas 1999; 32:
179-87.
251 Cooper C, Barker DJ, Wickham C. Physical activity, muscle strength, and
calcium intake in fracture of the proximal femur in Britain. Bmj 1988; 297:
1443-6.
252 Silman AJ, O'Neill TW, Cooper C et al. Influence of physical activity on
vertebral deformity in men and women: results from the European Vertebral
Osteoporosis Study. J Bone Miner Res 1997; 12: 813-9.
253 Frandin K, Grimby G, Mellstrom D et al. Walking habits and health-related
factors in a 70-year-old population. Gerontology 1991; 37: 281-8.
254 Greendale GA, Barrett-Connor E, Edelstein S et al. Lifetime leisure exercise
and osteoporosis. The Rancho Bernardo study. Am J Epidemiol 1995; 141:
951-9.
255 van der Klift M, de Laet CE, McCloskey EV et al. Risk factors for incident
vertebral fractures in men and women: the Rotterdam Study. J Bone Miner Res
2004; 19: 1172-80.
256 Rundgren A, Mellstrom D. The effect of tobacco smoking on the bone
mineral content of the ageing skeleton. Mech Ageing Dev 1984; 28: 273-7.
257 Kanis JA, Johnell O, Oden A et al. Smoking and fracture risk: a meta-
analysis. Osteoporos Int 2005; 16: 155-62.
88
258 Johansson C, Mellstrom D. An earlier fracture as a risk factor for new
fracture and its association with smoking and menopausal age in women.
Maturitas 1996; 24: 97-106.
259 Kanis JA, Johansson H, Oden A et al. A meta-analysis of prior corticosteroid
use and fracture risk. J Bone Miner Res 2004; 19: 893-9.
260 Huang C, Ross PD, Lydick E et al. Factors associated with joint pain among
postmenopausal women. Int J Obes Relat Metab Disord 1997; 21: 349-54.
261 Jones SM, Harris CP, Lloyd J et al. Lipoproteins and their subfractions in
psoriatic arthritis: identification of an atherogenic profile with active joint
disease. Ann Rheum Dis 2000; 59: 904-9.
262 Mallbris L, Granath F, Hamsten A et al. Psoriasis is associated with lipid
abnormalities at the onset of skin disease. J Am Acad Dermatol 2006; 54: 614-
21.
263 Burger D, Dayer JM. High-density lipoprotein-associated apolipoprotein A-
I: the missing link between infection and chronic inflammation? Autoimmun
Rev 2002; 1: 111-7.
264 Furumoto H, Arai H, Kuramitsu Y et al. Lipoproteins modulate growth and
differentiation of cultured human epidermal keratinocytes. Electrophoresis
2002; 23: 161-6.
265 Girolomoni G, Gisondi P. Psoriasis and metabolic comorbidities: the
importance of well-designed prospective studies. Commentary. Dermatology
2008; 217: 222-4.
266 Eckel RH, Grundy SM, Zimmet PZ. The metabolic syndrome. Lancet 2005;
365: 1415-28.
267 Wannamethee SG, Shaper AG, Lennon L et al. Metabolic syndrome vs
Framingham Risk Score for prediction of coronary heart disease, stroke, and
type 2 diabetes mellitus. Arch Intern Med 2005; 165: 2644-50.
268 Gisondi P, Tessari G, Conti A et al. Prevalence of metabolic syndrome in
patients with psoriasis: a hospital-based case-control study. Br J Dermatol
2007; 157: 68-73.
269 Tilg H, Moschen AR. Adipocytokines: mediators linking adipose tissue,
inflammation and immunity. Nat Rev Immunol 2006; 6: 772-83.
270 Barth J, Kohl V, Hanefeld M. [Behavior of lipids, other serum parameters
and cardiovascular functions in ultraviolet therapy]. Hautarzt 1994; 45: 702-7.
271 Neimann AL, Shin DB, Wang X et al. Prevalence of cardiovascular risk
factors in patients with psoriasis. J Am Acad Dermatol 2006; 55: 829-35.
89
272 Das UN. Is angiotensin-II an endogenous pro-inflammatory molecule? Med
Sci Monit 2005; 11: RA155-62.
273 Huskic J, Alendar F, Matavulj A et al. Serum angiotensin converting enzyme
in patients with psoriasis. Med Arh 2004; 58: 202-5.
274 Ucak S, Ekmekci TR, Basat O et al. Comparison of various insulin sensitivity
indices in psoriatic patients and their relationship with type of psoriasis. J
Eur Acad Dermatol Venereol 2006; 20: 517-22.
275 Carr DB, Utzschneider KM, Hull RL et al. Intra-abdominal fat is a major
determinant of the National Cholesterol Education Program Adult
Treatment Panel III criteria for the metabolic syndrome. Diabetes 2004; 53:
2087-94.
276 Libby P. Inflammation and cardiovascular disease mechanisms. Am J Clin
Nutr 2006; 83: 456S-60S.
277 Austin RC, Lentz SR, Werstuck GH. Role of hyperhomocysteinemia in
endothelial dysfunction and atherothrombotic disease. Cell Death Differ 2004;
11 Suppl 1: S56-64.
278 Clarke R, Daly L, Robinson K et al. Hyperhomocysteinemia: an independent
risk factor for vascular disease. N Engl J Med 1991; 324: 1149-55.
279 Thambyrajah J, Townend JN. Homocysteine and atherothrombosis--
mechanisms for injury. Eur Heart J 2000; 21: 967-74.
280 Malerba M, Gisondi P, Radaeli A et al. Plasma homocysteine and folate levels
in patients with chronic plaque psoriasis. Br J Dermatol 2006; 155: 1165-9.
281 Vanizor Kural B, Orem A, Cimsit G et al. Plasma homocysteine and its
relationships with atherothrombotic markers in psoriatic patients. Clin Chim
Acta 2003; 332: 23-30.
282 Grissom CB, Chagovetz AM, Wang Z. Use of viscosigens to stabilize
vitamin B12 solutions against photolysis. J Pharm Sci 1993; 82: 641-3.
283 Salmon S, Maziere JC, Santus R et al. UVB-induced photoperoxidation of
lipids of human low and high density lipoproteins. A possible role of
tryptophan residues. Photochem Photobiol 1990; 52: 541-5.
284 Kelly GS. Seasonal variations of selected cardiovascular risk factors. Altern
Med Rev 2005; 10: 307-20.
285 Carbone LD, Rosenberg EW, Tolley EA et al. 25-Hydroxyvitamin D,
cholesterol, and ultraviolet irradiation. Metabolism 2008; 57: 741-8.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
We tested the hypothesis that the age-related decline in skin thickness may contribute to the age-related decline in serum 25-hydroxyvitamin D [25(OH)D]. We measured skinfold thickness on the back of the hand, serum 25(OH)D, height, and weight in 433 normal postmenopausal women. We also noted the average daily hours of sunlight in the month in which the observations were made and in the preceding 2 mo. Serum 25(OH)D was positively related to hours of sunlight (with a time lag of 2 mo) and to skin thickness, and negatively to body mass index (wt/ht²). Serum 25(OH)D fell significantly after age 69 y. Seasonal variation of serum 25(OH)D was greater in lean than in fat subjects, which we attributed to the larger fat mass and consequent larger pool size in the latter group. The results suggest that the tendency for serum 25(OH)D to fall with age is due in part to the age-related decline in skin thickness.
Chapter
Full-text available
This chapter talks about vitamin D production, metabolism, mechanism of action, and clinical requirements. It discusses transport of vitamin D in the blood. The chapter focuses on internalization of vitamin D metabolites. It describes the regulation of calcium and phosphate metabolism. The chapter discusses classical target tissues of bone, intestine and kidney, and nonclassical target tissues of parathyroid glands and pancreas. It explains the immunobiology of vitamin D, and keratinocyte function in epidermis and hair follicles. Finally, the chapter deals with nutritional considerations and vitamin D treatment strategies. © 2013 American Society for Bone and Mineral Research. All rights reserved.
Article
Vitamin D is the sunshine vitamin for good reason. During exposure to sunlight, the ultraviolet B photons enter the skin and photolyze 7-dehydrocholesterol to previtamin D3 which in turn is isomerized by the body's temperature to vitamin D3. Most humans have depended on sun for their vitamin D requirement. Skin pigment, sunscreen use, aging, time of day, season, and latitude dramatically affect previtamin D3 synthesis. Vitamin D deficiency was thought to have been conquered, but it is now recognized that more than 50% of the world's population is at risk for vitamin D deficiency. This deficiency is in part due to the inadequate fortification of foods with vitamin D and the misconception that a healthy diet contains an adequate amount of vitamin D. Vitamin D deficiency causes growth retardation and rickets in children and will precipitate and exacerbate osteopenia, osteoporosis and increase risk of fracture in adults. The vitamin D deficiency pandemic has other serious consequences including increased risk of common cancers, autoimmune diseases, infectious diseases, and cardiovascular disease. There needs to be a renewed appreciation of the beneficial effect of moderate sensible sunlight for providing all humans with their vitamin D requirement for health.
Article
Background: Obesity is associated with vitamin D insufficiency and secondary hyperparathyroidism. Objective: This study assessed whether obesity alters the cutaneous production of vitamin D3 (cholecalciferol) or the intestinal absorption of vitamin D2 (ergocalciferol). Design: Healthy, white, obese [body mass index (BMI; in kg/m²) ≥ 30] and matched lean control subjects (BMI ≤ 25) received either whole-body ultraviolet radiation or a pharmacologic dose of vitamin D2 orally. Results: Obese subjects had significantly lower basal 25-hydroxyvitamin D concentrations and higher parathyroid hormone concentrations than did age-matched control subjects. Evaluation of blood vitamin D3 concentrations 24 h after whole-body irradiation showed that the incremental increase in vitamin D3 was 57% lower in obese than in nonobese subjects. The content of the vitamin D3 precursor 7-dehydrocholesterol in the skin of obese and nonobese subjects did not differ significantly between groups nor did its conversion to previtamin D3 after irradiation in vitro. The obese and nonobese subjects received an oral dose of 50000 IU (1.25 mg) vitamin D2. BMI was inversely correlated with serum vitamin D3 concentrations after irradiation (r = −0.55, P = 0.003) and with peak serum vitamin D2 concentrations after vitamin D2 intake (r = −0.56, P = 0.007). Conclusions: Obesity-associated vitamin D insufficiency is likely due to the decreased bioavailability of vitamin D3 from cutaneous and dietary sources because of its deposition in body fat compartments.
Article
Vitamin D deficiency is now recognized as a pandemic. The major cause of vitamin D deficiency is the lack of appreciation that sun exposure in moderation is the major source of vitamin D for most humans. Very few foods naturally contain vitamin D, and foods that are fortified with vitamin D are often inadequate to satisfy either a child's or an adult's vitamin D requirement. Vitamin D deficiency causes rickets in children and will precipitate and exacerbate osteopenia, osteoporosis, and fractures in adults. Vitamin D deficiency has been associated with increased risk of common cancers, autoimmune diseases, hypertension, and infectious diseases. A circulating level of 25-hydroxyvitamin D of >75 nmol/L, or 30 ng/mL, is required to maximize vitamin D's beneficial effects for health. In the absence of adequate sun exposure, at least 800–1000 IU vitamin D3/d may be needed to achieve this in children and adults. Vitamin D2 may be equally effective for maintaining circulating concentrations of 25-hydroxyvitamin D when given in physiologic concentrations.
Chapter
Ultraviolet (UV) radiation has been used for decades with great success and at a constantly increasing rate in the management of skin diseases and has thereby become an essential part of modern dermatological therapy [28]. Its success as a therapeutic agent has stimulated studies about the mechanisms by which UVB and UVA phototherapy work. The knowledge obtained from this work is an indispensable prerequisite for making treatment decisions on a rational rather than empirical basis. Modern dermatological phototherapy has just begun to profit from this knowledge, and it is very likely that this development will continue and provide dermatologists with improved phototherapeutic modalities and regimens for established and new indications. This chapter aims to provide an overview about current concepts of the mode of action of dermatological phototherapy. Special emphasis will be given to studies that have identified previously unrecognized immunosuppressive/anti-inflammatory principles of photo(chemo)therapy.