ArticlePDF AvailableLiterature Review

Abstract

Psoriasis is a chronic immune-mediated inflammatory skin disease. Psoriasis lesions are characterized by hyper-proliferation of epidermal keratinocytes associated with inflammatory cellular infiltrate in both dermis and epidermis. The epidermis is the natural source of vitamin D synthesis by sunlight action. Recently, a role for vitamin D in the pathogenesis of different skin diseases, including psoriasis, has been reported. Indeed, significant associations between low vitamin D status and psoriasis have been systematically observed. Due to its role in proliferation and maturation of keratinocytes, vitamin D has become an important local therapeutic option in the treatment of psoriasis. To date, the successful treatment based on adequate dietary intake of vitamin D or oral vitamin D supplementation in psoriasis represent an unmet clinical need and the evidence of its beneficial effects remains still controversial. This information is important either for Dermatologists and Nutritionists to increases the knowledge on the possible bi-directional relationships between low vitamin D status and psoriasis and on the potential usefulness of vitamin D in psoriasis with the aim not only to reduce its clinical severity, but also for delineating the risk profile for co-morbidities cardiac risk factors that may result from psoriasis. In the current review, we analyzed the possible bi-directional links between psoriatic disease and vitamin D.
Vitamin D and its role in psoriasis: An overview
of the dermatologist and nutritionist
Luigi Barrea
1
&Maria Cristina Savanelli
1
&Carolina Di Somma
2
&
Maddalena Napolitano
3
&Matteo Megna
4
&Annamaria Colao
5
&Silvia Savastano
5
Published online: 7 February 2017
#The Author(s) 2017. This article is published with open access at Springerlink.com
Abstract Psoriasis is a chronic immune-mediated inflamma-
tory skin disease. Psoriasis lesions are characterized by hyper-
proliferation of epidermal keratinocytes associated with in-
flammatory cellular infiltrate in both dermis and epidermis.
The epidermis is the natural source of vitamin D synthesis
by sunlight action. Recently, a role for vitamin D in the path-
ogenesis of different skin diseases, including psoriasis, has
been reported. Indeed, significant associations between low
vitamin D status and psoriasis have been systematically ob-
served. Due to its role in proliferation and maturation of
keratinocytes, vitamin D has become an important local ther-
apeutic option in the treatment of psoriasis. To date, the suc-
cessful treatment based on adequate dietary intake of vitamin
D or oral vitamin D supplementation in psoriasis represent an
unmet clinical need and the evidence of its beneficial effects
remains still controversial. This information is important ei-
ther for Dermatologists and Nutritionists to increases the
knowledge on the possible bi-directional relationships be-
tween low vitamin D status and psoriasis and on the potential
usefulness of vitamin D in psoriasis with the aim not only to
reduce its clinical severity, but also for delineating the risk
profile for co-morbidities cardiac risk factors that may result
from psoriasis. In the current review, weanalyzedthe possible
bi-directional links between psoriatic disease and vitamin D.
Keywords Environmental factors .Vitamin D .Psoriasis .
Dermatologist .Nutritionist .Nutrition
1 Introduction
Psoriasis is a chronic immune-mediated inflammatory skin
disease, with a prevalence of about 2%3% in the general
population [1]. The primary manifestation of psoriasis most
commonly manifests on the skin, although inflammatory pro-
cesses can occur also in other organs [1]. Indeed, nowadays
psoriasis is considered a systemic pathology, including also
other conditions, from psoriatic arthritis to obesity and meta-
*Silvia Savastano
sisavast@unina.it
Luigi Barrea
luigi.barrea@unina.it
Maria Cristina Savanelli
cristysav@hotmail.com
Carolina Di Somma
cdisomma@unina.it
Maddalena Napolitano
maddy.napolitano@gmail.com
Matteo Megna
mat24@libero.it
Annamaria Colao
colao@unina.it
1
I.O.S. & COLEMAN Srl, Acerra, 80011 Naples, Italy
2
IRCCS SDN, Napoli Via Gianturco 113, 80143 Naples, Italy
3
Dipartimento di Medicina e Scienze della Salute BVincenzo Tiberio^,
Rheumatology Unit, University of Molise, Via Francesco De Sanctis
1, 86100 Campobasso, Italy
4
Dipartimento di Medicina Clinica e Chirurgia, Unit of Dermatology,
Federico II University Medical School of Naples, Via Sergio Pansini
5, 80131 Naples, Italy
5
Dipartimento di Medicina Clinica e Chirurgia, Unit of
Endocrinology, Federico II University Medical School of Naples, Via
Sergio Pansini 5, 80131 Naples, Italy
Rev Endocr Metab Disord (2017) 18:195205
DOI 10.1007/s11154-017-9411-6
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
bolic disease (MetS), which increased cardiovascular risk in
psoriatic patients [24]. Histologically, the dermatosis is char-
acterized by hyperproliferation of keratinocytes, impaired epi-
dermal barrier function at the sites of skin lesions, and skin
infiltration by activated inflammatory cells [5]. The aetiology
of psoriasis is not fully understood. Several factors contribute
to its development, such as auto-immunological, genetic, hor-
monal and psychosomatic issues [6].
Vitamin D, also known as the sunshine vitamin, has long
been known to be a hormone that regulates calcium-
phosphorous homeostasis and safeguards the integrity of the
skeletal system [7]. The epidermis is the natural source of
vitamin D synthesis by the action of ultraviolet light (UV) B
of the sun or other UVB source [8]. On the other hand, evi-
dence is accumulating that vitamin D might represent a key
modulator of immune and inflammation mechanisms [9].
Recently, a role for vitamin D in the pathogenesis of different
skin diseases, including psoriasis, has been reported [911].
However, the effectiveness of vitamin D supplementation as
adjunctive treatment option in psoriatic patients still remains
controversial [911]. In the current review, we analyzed the
possible bi-directional links between vitamin D and psoriatic
disease.
2 The dermatologists point of view
Patients suffering from psoriasis present a broad range of clin-
ical phenotypes. Psoriatic lesions are classified into plaque,
guttate, pustular, and erythrodermic types according to clinical
features, especially regarding lesions size and distribution [1].
Disease onset may occur at any age, including childhood, with
two peak age ranges, 16 to 22 and 57 to 60 years [2,3].
Psoriasis lesions are characterized by hyper-proliferation with
incomplete differentiation of epidermal keratinocytes and de-
creased keratinocyte apoptosis, associated with inflammatory
cellular infiltrate in both dermis and epidermis [12]. Psoriasis
Area and Severity Index (PASI) score is currently the pre-
ferred method for the assessment of the disease severity and
extent [13].
2.1 Effects of vitamin D on skin biology
The role of vitamin D as main regulator of skin physiology
is very complex (Table 1). The epidermis is composed of
four layers: basal layer (stratum basale), spinous layer (stra-
tum spinosum), stratum granulosum and stratum corneum.
The stem cells within the basal layer, which contact the
basement membrane, continually divide during the lifetime
of the organism, providing a source of cells which progres-
sively migrate upwards through the epidermis, differentiat-
ing and stratifying to form the barrier layer of the skin [11,
14]. The process of epidermal differentiation is complex,
sequential, and tightly controlled [11]. The precursor of
vitamin D, 7-dehydrocholesterol, is located in the mem-
branes of keratonocytes of the basal and spinous layer of
epidermis [10]. By the action of UVB (wavelength between
290 and 315 nm), via a photochemical reaction, the B ring
of 7-dehydrocholesterol is broken to form pre-vitamin D3
or cholecalciferol, which is subsequently converted first to
25-hydroxyvitamin D (25OHD) by the enzymes CYP27A1
and CYP2R1 and then to 1,25-hydroxyvitamin D
(1,25(OH)D or calcitriol) the active form of vitamin D, by
CYP27B1 [15]. Physiologically, the active form of vitamin
D and its receptor regulate the differentiation and prolifer-
ation of keratinocytes, the balance of the cutaneous im-
mune system and the process of apoptosis. The
1,25(OH)D has been shown to exert anti-proliferative ef-
fects on keratinocytes [16]. Numerous in vitro and in vivo
studies have demonstrated dose-dependent effects of vita-
min D on proliferation and differentiation of keratinocytes.
Of interest, low concentration of vitamin D promotes
keratinocyte proliferation in vitro, while at higher pharma-
cological doses a clear inhibitory effect became apparent
[14,17]. Moreover, 1,25(OH)D and analogs reduce
S100A7 levels, generally up-regulated in psoriatic skin, in
the reconstituted human epidermis stimulated by IL-22
[18], in interleukin (IL)-17-stimulated keratinocytes and
in skin of patients with psoriasis [19]. Indeed, 1,25(OH)D
regulates the cell proliferation in the stratum basale and
increases the synthesis of keratins (K1 and K10),
involucrin, transglutaminase, loricrin, and filaggrin, in the
stratum spinosum [11,14,15]. Furthermore, vitamin D
helps to regulate the synthesis of glycosylceramides need-
ful for the barrier integrity and permeability in the stratum
corneum [11,14,17]. These actions are due to the capacity
of vitamin D to regulate intracellular calcium level, through
induction of the calcium receptor, and the phospholipase C
enzymes [19,20]. A decrease or deficiency in 1,25(OH)D
or a loss-of-function of its receptor has been shown to dis-
rupt the differentiation of the epidermis, with reduced
levels of involucrin and loricrin and loss of keratohyalin
granules, resulting in hyperproliferation of the basal layer
[11,2123].
Table 1 Vitamin D actions on skin
Vitamin D actions on skin biology and psoriasis pathogenesis
Regulation of keratinocytes proliferation, differentiation and apoptosis
Regulation of cutaneous immune system (inhibition of T cell
proliferation, Tregs induction)
Down-regulation of pro-inflammatory cytokines
Stimulation of antimicrobial peptides expression
Regulation of barrier integrity and permeability
Vitamin D actions on skin biology and psoriasis pathogenesis
Treg s, Regulatory T cells
196 Rev Endocr Metab Disord (2017) 18:195205
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
2.2 Vitamin D regulation of apoptosis in keratinocytes
Calcitriol stimulates the synthesis of ceramide by inducing the
neutral Mg
2+
-dependent sphingomyelinase (thereby increas-
ing the conversion of sphingomyelin to ceramide) and in re-
turn, ceramide enhances the pro-differentiating effect of
calcitriol on keratinocytes in a feedback loop [10,24]. It has
been demonstrated that physiological concentrations of
calcitriol do not initiate apoptosis in cultured keratinocytes
but, in contrast, pharmacological concentrations of calcitriol
exert a pro-apoptotic effect on keratinocytes [25].
2.3 Effects of vitamin D on the cutaneous immune system
Psoriasis pathogenesis implicates the innate and adaptive seg-
ments of the immune system. In particular, it is centrally con-
trolled by T cells, in which an important role is played by T-
helper (Th)1, Th17 and Th22, interplaying with numerous cell
types via different cytokines, including tumour-necrosis
factor-α(TNF-α), IL-6 and IL-17 [26]. The activity of these
cells is modulated by specific T lymphocytes, named regula-
tory T cells (Treg) [9]. Regulatory T cells (Tregs) are able to
inhibit the immunological response and to preserve the cuta-
neous immunological homeostasis, preventing autoimmune
response against self-antigens [27].
There is an increasing interest on broad regulatory effects
exerted by vitamin D on cells of the adaptive and innate im-
mune system [28]. Indeed, vitamin D acts as a pluripotent
immunomodulator that inhibits proliferation of T lympho-
cytes, induces generation of CD25+/CD4+ Tregs, a phenotype
of T cells promoting tolerance and inhibiting immunity after
stimulation with antigen. Moreover, vitamin D induces the
expression of the C-C chemokine receptor type 10 on surface
of T lymphocytes, a receptor involved in T cell-mediated skin
inflammation, leading their migration from dermal blood ves-
sels to epidermal kerotinocytes [29]. Finally, vitamin D helps
to defend from opportunistic infections, by inducing autoph-
agy in human macrophages, and to support the innate skin
barrier, by stimulating endogenous antimicrobial peptides ex-
pression in resident epithelial cells of the skin [9].
Antimicrobial peptides, like cathelicidins and defensins, have
not only properties against bacteria, fungi and viruses, but also
other immune regulatory properties, including cytokine and
chemokine release, antigen presentation, cell proliferation, in-
creasing vascular permeability, angiogenesis and wound
healing [30].
2.4 Vitamin D and psoriasis linkage
In psoriasis, vitamin D is involved in the maintenance of cu-
taneous barrier homeostasis. Several studies identified an as-
sociation between polymorphisms of vitamin D receptor
(VDR) and psoriasis susceptibility [9]. Richetta et al., have
found that the A-1012G promoter polymorphism of the
VDR gene is associated with psoriasis risk through a lower
expression of VDR mRNA, favoring conditions that may alter
cutaneous barrier and the development of psoriatic lesions
[31]. In addition, in psoriatic skin a decreased expression of
VDR and reduced tight-junction proteins is associated [32].
Tight junctions are fundamental to regulate adhesion and per-
meability of keratinocytes, and to polarize cutaneous cell dif-
ferentiation, to regulate extracellular calcium gradient,
interacting with nuclear and cytoplasmic proteins and
influencing the regulation of specific genes involved in
keratinocytes differentiation and proliferation [32,33].
Different studies have focused on the possible role of low
vitamin D status in the pathogenesis of psoriasis [3436].
Indeed, several studies reported that vitamin D is a key
modulator of inflammation function [37,38]. The active me-
tabolite of vitamin D exert an anti-inflammatory effect on the
inflammatory profile of human monocytes/macrophage
[3942], down-regulating the expression and production of
several pro-inflammatory cytokines including TNF-α,IL-
1β,IL-6,andIL-8[43]. Moreover, dendritic cells differentia-
tion, maturation, chemotaxis and antigen presentation seem to
be dampened, and hydrogen peroxide secretion in human
monocytes is also activated by 1,25(OH)D resulting in an
increased oxidative burst potential [9,44]. These anti-
inflammatory effects support a role a low vitamin D status in
the pathogenesis of psoriasis. Recent studies have shown that
1,25(OH)D values are significantly lower in psoriatic patients
than in control subjects, even after adjusting for confounding
factors in a multivariate analysis [11,35]. In another study,
vitamin D levels were lower in women with psoriasis in com-
parison with men, a difference not observed among controls
[45]. Therefore, low levels of vitamin D are negatively asso-
ciated with markers of inflammatory activation (C-reactive
protein, CRP) and obesity [35]. Moreover, other studies
showed that serum vitamin D levels were also reduced in
patients with psoriatic arthritis and being inversely linked to
disease activity [46,47].
2.5 Topical vitamin D in psoriasis treatment
The beneficial effects of vitamin D induced by exposure
to sunlight in the treatment of psoriasis have been
known for decades. The effectiveness on psoriasis of
vitamin D and its derivatives (calcitriol, calcipotriol,
tacalcitol, hexafluoro-1,25(OH)D and maxacalcitol) have
been known since 1985, being confirmed in numerous
trials [45,48]. The therapy with vitamin D, is one of
the most popularly prescribed topical medications for
this disease as the first-line, singly or in combination
with topical corticosteroids, and numerous studies doc-
umented the efficacy and safety of using topical
calcipotriol in the treatment of cases of localized plaque
Rev Endocr Metab Disord (2017) 18:195205 197
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
psoriasis [4954].Vitamin D analogs are particularly
helpful for hard-to-treat areas such as the face or ingui-
nal regions that are sensitive to steroid-induced atrophy
[55]. Vitamin D analogs do not exhibit tachyphylaxis,
as seen with corticosteroids, and topical treatment can
be continued indefinitely without serious adverse side
effects [9,55]. Additionally, they are effective in the
treatment of psoriatic skin lesions in children and elder-
ly population [5658]. A recent meta-analysis on the
effectiveness of topical vitamin D therapies evidenced
not only comparable efficacies to corticosteroids when
used as monotherapy, but also superior effects when
vitamin D used in combination with a potent topical
steroid.Accordingtotheseresults, as topical vitamin
D derivatives demonstrated a favorable safety profile,
with Bsteroid-sparing^effects, and should be considered
an indispensable component of the current physicians
arsenal in the treatment of psoriasis [11].
The therapeutic effects of topical vitamin D occur via
aVDR mediated genomic mechanism resulting in inhi-
bition of keratinocyte proliferation and mediated non-
genomic mechanisms inducing keratinocyte differentia-
tion by increasing intracellular calcium levels [59]. The
anti-inflammatory effects may also result from inhibition
of production of IL-2, IL-6, and interferon-gamma
(IFN-γ). Further, topical calcipotriol inhibits human beta
defensin and proinflammatory cytokines which are
found in increased levels in psoriatic lesions [60].
Allelic variations in individual VDR genes may deter-
mine a different response to treatment: the isoform A
of VDR is associated with a greater therapeutic response
in psoriatic patients [61].
3 The nutritionists point of view
Severe psoriasis has been associated with nutritional defi-
ciencies because of an accelerated loss of nutrients, in
particular of vitamin D, from the hyperproliferation and
desquamation of the epidermal layer of skin [6264].
Vitamin D supplementation is of particular interest to
Nutritionists for two important reasons. First, besides its
topical use, oral vitamin D supplementation represents an
important adjunctive treatment option for psoriatic patients
[9]; second, vitamin D supplementation might be very im-
portant for the prevention of psoriasis-related comorbidity
[65], hypertension [66] and metabolic syndrome [67].
3.1 Food and vitamin D
There are two ways to meet vitamin D requirements in mam-
mals: via nutrition and via synthesis inskin by fromthe sun or
other UVB source [8]. Several recommendations were
published regarding the dietary intake of vitamin D [6870].
In particular, these recommendations they refer to dietary ref-
erence intakes for calcium and vitamin D updated by the
Institute of Medicine (IOM) in 2010 [71]. To date, as the
evidence for extra-skelatal effects of vitamin D are inconsis-
tent and insufficient, the intake recommendations were based
on beneficial effects of vitamin D only on skeletal health. The
Recommended Dietary Allowances (RDAs) covering require-
ments of 97.5% of the population are shown in Table 2.
Other organizations recommended different RDAs: the
Endocrine Society suggested that adults aged 1950 years re-
quire at least 600 international unit (IU) of vitamin D daily to
maintain bone and muscular function [72]. However, the task
force further annotated that 15002000 IU per day are neces-
sary to consistently raise the serum level of 25(OH)D above
30 ng/mL. Finally, for older adults aged 60 and above, the
International Osteoporosis Foundation (IOF) recommended
a RDA of 8001000 IU in order to reach a serum 25(OH)D
level of 30 ng/mL [73].
The diet is an important determinant of vitamin D status
[71,74]. Some studies have calculated that the amount of
vitamin D intake that would ensure that the majority of the
population (97.5%) maintains plasma 25(OH)D concentra-
tions >25 nmol/l throughout the year is 8.7 mg/d [75]. Only
few foods contain naturally vitamin D, and these foodstuffs
are mainly of animal origin. The vitamin D status is the sum
from the combination of synthesis in the skin after sun expo-
sure and intake of the two main dietary forms of vitamin D:
ergocalciferol (vitamin D2) and cholecalciferol (vitamin D3)
[76]. The first is produced by plants, although fruits and veg-
etables in the human diet contain only minimal amounts of
this nutrient, despite significant amounts of vitamin D2 are
Table 2 Vitamin D dietary reference intakes by life stage
Life-stage group RDA (intake that
covers needs
of 97.5% of
population)
Serum 25OHD level
(corresponding to
the RDA)*
IU/d mcg/d ng/ml
Infants (012 months) 400** 10** 20
170 yr 600 15 20
+ 70 yr 800 20 20
Pregnant 600 15 20
Lactating 600 15 20
The Recommended Dietary Allowances (RDAs) covering requirements
by life stage
RDA,Recommended Dietary Allowance; IU,International Unit
*Measures of serum 25(OH)D levels corresponding to the RDA and
covering the requirements of at least 97.5% of the population
**Reflects adequate intake reference value rather than RDA. RDAs have
not been established for infants
198 Rev Endocr Metab Disord (2017) 18:195205
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
available through dietary supplements. The mushrooms pro-
vide variable amounts of vitamin D2, and if exposed to ultra-
violet light under controlled conditions, the mushrooms can
improve their content of vitamin D2 [77,78]. The vitamin D3
instead, is naturally found in some animal foods, particularly
in fatty fish, including salmon, herring, and mackerel, and in
fish oils [79]. The fish (especially fatty fish and fish liver) have
the highest natural content of vitamin D [80] Also egg yolk
has a high vitamin D3 content [80], which strongly correlates
with the content of vitamin D3 of the hens feed [77].
Depending on the hensdiet[81] and UVB exposure [82],
the vitamin D3 and 25(OH)D3 were transferred from the
hen to the egg yolk. Regarding of meat products, the content
of vitamin D depends on the contents of vitamin D in the
fodder, the fat content of the meat product, and latitude where
the animals have grazed [83]. Finally, the vitamin D3 is also
available through dietary supplements, and is the form present
in vitamin D-fortified foods such as milk, orange juice, and
cereals. The fortification of food with vitamin D has been
consideredto be the most promising strategywith the broadest
reach and impact [8486]. In fact, in countries where this
strategy was adopted proved to have a significant influence
on the daily vitamin D intake in the average adult [87]. The
food sources of vitamin D are listed in Table 3[88].
3.2 Bioavailability and influence of processing
and cooking
The vitamin D, like other fat-soluble vitamins (A, E, K), is
absorbed incorporated in mixed micelles from the intestine
into the enterocytes by non-saturable passive diffusion.
Subsequently, the vitamin D is transported in the chylomi-
crons via lymph to the circulation [89]. The more polar me-
tabolite 25(OH)D is absorbed better and faster than vitamin D
because it is also taken up directly from the proximal jejunum
into the portal vein [79]. There are few data on its availability
from natural sources. The absorption of vitamin D from sup-
plements may differ depending on the used vehicle substance,
such as oils, powders, ethanol [90]. For example, it has been
reported that the bioavailability of vitamin D from fortified
hard cheese is equivalent to supplements [91] and that vitamin
D bioavailability is not influenced by the fat content of the
fortified milk [92].
The cooking does not much influence the vitamin D
content of animal foods. Mattila et al. [93] found that, in
eggs boiled for 10 min, the vitamin D3 concentration was
16% lower and 25(OH)-D-3 content was 611% lower
compared with raw eggs. In addition, also in fish, the
cooking effect was moderate: baking various kinds of fish
(e.g., perch, rainbow trout, Baltic herring) in the oven at
172 °C or 200 °C for 20 min induced a vitamin D3 loss of
<10%, calculated on a dry matter basis [93]. The stability of
vitamin D3 and 25(OH)D and vitamin D2 in foodstuffs
during cooking has been shown to vary widely with heating
process and foodstuffs, with reported retentions in eggs,
margarine and bread after boiling, frying and baking of
between 40% and 88% [94].
3.3 The nutritionist and vitamin D supplementations
Supplements are the most important determinant of varia-
tion in vitamin D intake [85,86,95]A number of studies
showed that the daily intake of vitamin D was higher in
adults using vitamin D supplements than in those without
vitamin D supplementations (348 IU vs 84 IU of vitamin
D) [86]. Most nutritionists recommend the use of vitamin
D3 to treat and prevent vitamin D deficiency, because sev-
eral studies indicating a higher efficacy for vitamin D3 in
raising serum 25(OH)D concentrations when compared to
vitaminD2[96]. Although a significant inter-individual
variation exists, due to different variables including body
weight, sunlight exposure and calcium intake, it has been
calculated that supplementation of 1000 IU of vitamin D3
daily leads to an approximate increase in 25(OH)D levels
by 1020 ng/mL (2550 mmol/L), [9799]. Findings from
randomised placebo-controlled trials conducted during the
winter have shown that each 1 mg of supplemental vitamin
D is associated with an increase in serum 25(OH)D of be-
tween 0.7 nmol/L [100]and2nmol/L[75].
Several studies have observed the safety concerns re-
garding the dosage of vitamin D supplementation.
However, oral vitamin D intakes of up to 10,000 IU daily
were not associated with any harmful effects [101], since
this dose is comparable to the maximum cutaneous vitamin
Table 3 Food sources of vitamin D[88]
Food Amount IUs per serving**
Cod liver oil 1 tablespoon 1360
Swordfish, cooked 3 oz* 566
Salmon (sockeye), cooked 3 oz 477
Tuna fish, canned in water, drained 3 oz 154
Orange juice fortified with vitamin D
(check product labels, as amount
of added vitamin D varies)
1 cup 137
Milk, nonfat, reduced fat, and whole,
vitamin D-fortified
1 cup 115124
Sardines, canned in oil, drained 2 sardines 46
Liver, beef, cooked 3 oz 42
Egg (vitamin D is found in yolk) 1 large 41
Cheese, Swiss 1 oz 6
The food sources of vitamin D
*1 oz = 28,3495 g
**IU= International Units
Rev Endocr Metab Disord (2017) 18:195205 199
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
D production, and reports of vitamin D intoxication from
cutaneous synthesis alone do not exist [101]. In this con-
text, the IOM and the European Food and Safety Authority
(EFSA) recommend a safe tolerable upper intake level of
4000 IU vitamin D per day for all adults, including preg-
nant and lactating women [102], although, to date, very
few studies have investigated the long-term effects of a
vitamin D intake above the suggested threshold of
4000 IU per day [103,104]. To reach a steady state, after
two to three consecutive months of treatment with vitamin
D supplementation, its necessary a re-measurement of
25(OH)D serum levels [98].
Thus, the Nutritionists should consider a general vitamin D
supplementation in populations at high risk for vitamin D
deficiency, such as psoriatic patients [105]. The compounds
of vitamin D commonly used in clinical trials varied from
1,25(OH)D, the physiologically active form of vitamin D, to
1αOHD, alfa-calcidol, requiring only liver metabolism to be
converted to the active form to vitamin D or cholecalciferol,
requiring both liver and kidney metabolism to become active.
Perez et al. observed an overall 88% of clinical improvement
of psoriasis with oral vitamin D with a decrease in mean PASI
scores [61]. The results were confirmed in different studies on
limited number of patients, reporting moderate or greater im-
provement in psoriasis in 2550% of subjects [106109]. It
has also been proposed the therapeutic use of systemic alpha-
calcidol in patients with psoriatic arthritis [61]. Another study
demonstrated that a combination ofacitretin and oral calcitriol
resulted in a faster reduction of PASI in patients of chronic
plaque psoriasis [110]. A study on a large population sample
including 70,437 US females over a period of 14 years, ex-
amined the vitamin D intake levels and the incidence of pso-
riasis in the population. After adjusting for confounding var-
iables, the study found that there was no significant associa-
tion between vitamin D intake (dietary, supplementary, and
total vitamin D) and the risk of incident psoriasis. Thus, the
authors proposed that there was no role of dietary or supple-
mental vitamin D intake to prevent the development of psori-
asis [111]. However, current recommendations for dietary in-
take of vitamin D are based only on its effect on skeletal
health, while no information is present as regards psoriasis.
In addition, a discrepancy between recommended RDAs and
actual daily vitamin D intake exists. Therefore, solutions like
food fortification and personalized diet or vitamin D supple-
mentation in psoriatic patients need to be developed. In addi-
tion, taking into account the common association among pso-
riasis, obesity and Mets of which will be discussed in the next
chapter, the advantage of the potential use of oral vitamin D
supplementations to treat psoriasis and metabolic syndrome
concurrently through its anti-inflammatory effects was strong-
ly supported by a comprehensive meta-analysis including the
results of clinical trials using vitamin D supplementation in
psoriasis [106].
3.4 Vitamin D, obesity and psoriasis
Many studies showed the link between psoriasis, obesity
and MetS [112,113].Obesityisanimportant risk factor
for psoriasis [114,115]. The relationship between the two
conditions is probably bidirectional, with obesity, mainly
visceral obesity, predisposing to psoriasis and psoriasis
favouring obesity [116]. In particular, there was a 2-fold
increased risk for psoriasis development in the setting of
obesity as compared with normal weight subjects [117].
In addition, for each unit increment increase in body mass
index (BMI) was reported a 9% higher risk for psoriasis
onset and a 7% higher risk for increased of PASI score
[118]. A further evidence of the link between, obesity,
inflammation and cardiovascular diseases in patients with
psoriasis is provided by several studies reporting a corre-
lation between PASI score and increased of CRP levels
[119] and between psoriasis and waist circumference
[120]. CRP is an acute phase protein significantly associ-
ated with obesity, representing the most sensitive markers
of inflammation and an independent risk for cardiovascu-
lar disease. Waist circumference represents a surrogate
measure of fat distribution highly correlated with visceral
fat [121],themainsourceofinflammatorycytokinesin
obesity [122]. In particular, obesity and psoriasis are both
pro-inflammatory conditions, in which the adipokines bal-
ance is shifted in favor to action pro-inflammatory
adipokines [123,124]. On the one hand, the dominance
of pro-inflammatory adipokines favors either the develop-
ment and the maintenance of obesity and its conse-
quences, including cardiovascular diseases, type 2 diabe-
tes, and MetS [125]; on the other hand, the production of
inflammatory cytokines in visceral obesity represents the
link involved in the complex mechanisms leading to the
exacerbation of psoriasis and to psoriasis co-morbidities
[126]. Indeed, psoriasis is frequently associated with
cardio-metabolic co-morbidities and with an increased
cardiovascular mortality [127,128].
Similarly, there are significant associations between low
vitamin D status and increased risk of obesity and obesity-
related co-morbidities and cardiovascular mortality [127].
In particular, higher BMI leads to lower vitamin D status,
with an inverse association between mortality risk and vi-
tamin D levels, although this association could be indirectly
mediated by the obesity per se [129]. Ju SYet al. performed
a meta-analysis of the dose-response relationship between
blood vitamin D levels and the risk of MetS. Using data on
blood 25(OH)D levels, a monotonically decreasing rela-
tionship was observed for low levels of blood 25(OH)D in
the pooled analyses of 16 cross-sectional studies (OR =0.89
for 30 nmol/L, OR =0.80 for 60 nmol/L, OR =0.71 for
90 nmol/L, and OR =0.63 for 120 nmol/L). A 25 nmol/L
increase in 25(OH)D levels was associated with a 13%
200 Rev Endocr Metab Disord (2017) 18:195205
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
decrease in the risk of MetS in cross-sectional studies [130].
Several mechanisms might account for the low vitamin D
status in obese subjects, [131]. Besides a low dietary intake
of vitamin D linked to restrictive weight-loss diet regimen,
the most likely explanations for low vitamin D status in
obesity are sequestration of vitamin D in the adipose tissue
[132], volumetric dilution related to the greater volume of
distribution of 25(OH)D, less exposure of skin to sunlight
due to less outdoor activity and air pollution [133].The
associations of low vitamin D status with obesity on the
one hand, and with psoriasis on the other hand, lend support
to the hypothesis for considering vitamin D as a further link
between obesity and psoriasis. In this contest, a vicious
cycle could operate among low vitamin D status, obesity,
and psoriasis, with additive detrimental effects on cardio-
metabolic risk in obese psoriatic patients. According to this
hypothesis, vitamin D supplementation might be of partic-
ular usefulness for the prevention of psoriasis-related
comorbidities.
4Conclusions
Due to its role in proliferation and maturation of keratinocytes,
vitamin D has become an important local therapeutic option in
the treatment of psoriasis. Indeed, significant associations be-
tween low vitamin D status and psoriasis have been system-
atically observed. Although the exact role of vitamin D in the
pathogenesis of psoriasis is unclear, understanding the possi-
ble bi-directional relationships between low vitamin D status
and psoriasis is also important for delineating the risk profile
for co-morbidities that may result from psoriasis, such as obe-
sity, type 2 diabetes, and MetS. To date, the successful treat-
ment based on adequate dietary intake of vitamin D or oral
vitamin D supplementation in psoriasis represent an unmet
clinical need and the evidence of its beneficial effects remains
still controversial. Nevertheless, the Nutritionists should con-
sider a general vitamin D supplementation in populations at
high risk for vitamin D deficiency, such as psoriatic patients.
This information is important either for Dermatologists and
Nutritionists to increase the knowledge on the potential use-
fulness of vitamin D in psoriasis with the aim to reduce not
only its clinical severity, but also cardiac risk factors and pso-
riasis co-morbidities. Future well-designed dietary interven-
tion trials with vitamin D supplementations on large popula-
tion samples are needed to define the specific dose of vitamin
D supplementations for psoriasis.
1,25(OH)D, 1,25-hydroxyvitamin D or calcitriol; 25OHD,
25-hydroxyvitamin D; BMI, body mass index; CRP, C-
reactive protein; IL, interleukin; MetS, metabolic disease;
PASI, psoriasis area and severity index; RDAs, recommended
dietary allowances; TNF, tumor necrosis factor; VDR, vitamin
Dreceptor.
Acknowledgements We would like to acknowledge all the
Collaborators of this review: Antonio Improta, Dr. Lidia Albanese and
Dr. Vincenza Grazia Mele.
Compliance with ethical standards
Conflict of interest statement and funding sources The authors de-
clare that they have no conflict of interest. There was no funding source
for this work.
Open Access This article is distributed under the terms of the Creative
Commons Attribution 4.0 International License (http://
creativecommons.org/licenses/by/4.0/), which permits unrestricted use,
distribution, and reproduction in any medium, provided you give appro-
priate credit to the original author(s) and the source, provide a link to the
Creative Commons license, and indicate if changes were made.
References
1. Napolitano M, Caso F, Scarpa R, Megna M, Patrì A, Balato N,
Costa L. Psoriatic arthritis and psoriasis: differential diagnosis.
Clin Rheumatol. 2016;35(8):1893901.
2. Reich K. The concept of psoriasis as a systemic inflammation:
implications for disease management. J Eur Acad Dermatol
Venereol. 2012;26(Suppl 2):311.
3. Napolitano M, Megna M, Monfrecola G. Insulin resistance and
skin diseases. Sci World J. 2015;2015:479354.
4. Lønnberg AS, Skov L. Co-morbidity in psoriasis: mechanisms
and implications for treatment. Expert Rev Clin Immunol.
2016;28:18.
5. Nestle FO, Kaplan DH, Barker J. Psoriasis. N Engl J Med.
2009;361(5):496509.
6. Lebwohl M. Psoriasis. Lancet. 2003;361(9364):1197204.
7. Holick MF. Vitamin D deficiency. N Engl J Med. 2007;357:26681.
8. Kovács S, Wilkens MR, Liesegang A. Influence of UVB exposure
on the vitamin D status and calcium homoeostasis of growing
sheep and goats. J Anim Physiol Anim Nutr (Berl).
2015;99(Suppl S1):112.
9. Mattozzi C, Paolino G, Richetta AG, Calvieri S. Psoriasis, vitamin
D and the importance of the cutaneous barrier's integrity: an up-
date. J Dermatol. 2016;43(5):50714.
10. Wadhwa B, Relhan V, Goel K, Kochhar AM, Garg VK. Vitamin D
and skin diseases: a review. Indian J Dermatol Venereol Leprol.
2015;81(4):34455.
11. Soleymani T, Hung T, Soung J. The role of vitamin D inpsoriasis:
a review. Int J Dermatol. 2015;54(4):38392.
12. Lowes MA, Suárez-Fariñas M, Krueger JG. Immunology of pso-
riasis. Annu Rev Immunol. 2014;32:22755.
13. Harari M, Shani J, Hristakieva E, Stanimirovic A, Seidl W,
Burdo A. Clinical evaluation of a more rapid and sensitive
psoriasis assessment severity score (PASS), and its comparison
with the classic method of psoriasis area and severity index
(PASI), before and after climatotherapy at the Dead-Sea. Int J
Dermatol. 2000;39(12):9138.
14. Reichrath J. Vitamin D and the skin: An ancient friend, revisited.
Exp Dermatol. 2007;16(7):61825.
15. Bikle DD. Protective actions of vitamin D in UVB induced skin
cancer. Photochem Photobiol Sci. 2012;11(12):180816.
16. Matsumoto K, Hashimoto K, Nishida Y, Hashiro M, Yoshikawa
K. Growth-inhibitory effects of 1,25-dihydroxyvitamin D3 on
Rev Endocr Metab Disord (2017) 18:195205 201
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
normal human keratinocytes cultured in serum-free medium.
Biochem Biophys Res Commun. 1990;166(2):91623.
17. Gniadecki R. Stimulation versus inhibition of keratinocyte growth
by 1,25-dihydroxyvitamin D3: dependence on cell culture condi-
tions. J Invest Dermatol. 1996;106(3):5106.
18. Datta Mitra A, Raychaudhuri SP, Abria CJ, Mitra A, Wright R,
Ray R, Kundu-Raychaudhuri S. 1α,25-dihydroxyvitamin-D3-3-
bromoacetate regulates AKT/mTOR signaling cascades: a thera-
peutic agent for psoriasis. J Invest Dermatol. 2013;133(6):1556
64.
19. Hegyi Z, Zwicker S, Bureik D, Peric M, Koglin S, Batycka-Baran
A, Prinz JC, Ruzicka T, Schauber J, Wolf R. Vitamin D analog
calcipotriol suppresses the Th17 cytokine-induced proinflamma-
tory S100 "alarmins" psoriasin (S100A7) and koebnerisin
(S100A15) in psoriasis. J Invest Dermatol. 2012;132(5):141624.
20. Tu CL, Oda Y, Komuves L, Bikle DD. The role of the calcium-
sensing receptor in epidermal differentiation. Cell Calcium.
2004;35:26573.
21. Xie Z, Bikle DD. Cloning of the human phospholipase C-gamma1
promoter and identification of a DR6-type vitamin D-responsive
element. J Biol Chem. 1997;272:65737.
22. Bikle DD. Vitamin D regulated keratinocyte differentiation. J Cell
Biochem. 2004;92:43644.
23. Xie Z, Komuves L, Yu QC, Elalieh H, Ng DC, Leary C, Chang S,
Crumrine D, Yoshizawa T, Kato S, Bikle DD. Lack of the vitamin
D receptor is associated with reduced epidermal differentiation
and hair follicle growth. J Invest Dermatol. 2002;118:116.
24. Okasaki T, Bell RM, Hannun YA. Sphingomyelin turnover in-
duced by vitamin D3 in HL-60 cells. Role in cell differentiation.
J Biol Chem. 1989;264:1907680.
25. Lehmann B, Querings K, Reichrath J. Vitamin D and skin: new
aspects for dermatology. Exp Dermatol. 2004;13(Suppl 4):115.
26. Luan C, Chen X, Hu Y, Hao Z, Osland JM, Chen X,
Gerber SD, Chen M, Gu H, Yuan R. Overexpression and
potential roles of NRIP1 in psoriasis. Oncotarget. 2016;
doi:10.18632/oncotarget.12371.
27. Buckner JH. Mechanisms of impaired regulation by CD4(+)
CD25(+) FOXP3(+) regulatory T cells in human autoimmune
diseases. Nat Rev Immunol. 2010;10:84959.
28. Sloka S, Silva C, Wang J, Yong VW. Predominance of Th2polar-
ization by vitamin D through a STAT6-dependent mechanism. J
Neuroinflammation. 2011;8:56.
29. Van Etten E, Decallone B, Verlinden L, Verstuyf A, Bouillon R,
Mathieu C, et al. Analogs of 1α,25-dihydroxy vitamin D3 as
pluripotent immunomodulators. J Cell Biochem. 2003;88:2236.
30. Heilborn JD, Weber G, Gronberg A, Dieterich C, Stahle M.
Topical treatment with the vitamin D analogue calcipotriol en-
hances the upregulation of the antimicrobial protein hCAP18/
LL-37 during wounding in human skin in vivo. Exp Dermatol.
2010;19:3328.
31. Richetta AG, Silvestri V, Giancristoforo S, Rizzolo P, D'Epiro
S, Graziano V, Mattozzi C, Navazio AS, Campoli M, D'Amico
C, Scarnò M, Calvieri S, Ottini L. A-1012G promoter polymor-
phism of vitamin D receptor gene is associated with psoriasis
risk and lower allele-specific expression. DNA Cell Biol.
2014;33(2):1029.
32. Visconti B, Paolino G, Carotti S, Pendolino AL, Morini S,
Richetta AG, Calvieri S. Immunohistochemical expression of
VDR is associated with reduced integrity of tight junction com-
plex in psoriatic skin. J Eur Acad Dermatol Venereol. 2015;29:
203842.
33. Kirschner N, Rosenthal R, Günzel D, Moll I, Brandner JM. Tight
junctions and differentiation a chicken or the egg question? Exp
Dermatol. 2012;21:1715.
34. Gisondi P, Rossini M, Di Cesare A, Idolazzi L, Farina S, Beltrami
G. Vitamin D status in patients with chronic plaque psoriasis. Br J
Dermatol. 2012;166:50510.
35. Orgaz-Molina J, Buendía-Eisman A, Arrabal-Polo MA, Ruiz JC,
Arias-Santiago S. Deficiency of serum concentration of 25-
hydroxyvitamin D in psoriatic patients: a case-control study. J
Am Acad Dermatol. 2012;67:9318.
36. El-Moaty Zaher HA, El-Komy MH, Hegazy RA, Mohamed El
Khashab HA, Ahmed HH. Assessment of interleukin-17 and vi-
tamin D serum levels in psoriatic patients. J Am Acad Dermatol.
2013;69:8402.
37. Mehmood ZH, Papandreou D. An updated mini review of vitamin
D and obesity: Adipogenesis and inflammation state. Open Access
Maced J Med Sci. 2016;4(3):52632.
38. de Gonçalves Carvalho CM, Ribeiro SM. Aging, low-grade sys-
temic inflammation and vitamin D: a mini-review. Eur J Clin Nutr.
2016; doi:10.1038/ejcn.2016.177.
39. Zhang Y, Leung DY, Richers BN, Liu Y, Remigio LK, Riches
DW, Goleva E. Vitamin D inhibits monocyte/macrophage proin-
flammatory cytokine production by targeting MAPK phospha-
tase-1. J Immunol. 2012;188(5):212735.
40. Giulietti A, van Etten E, Overbergh L, Stoffels K, Bouillon R,
Mathieu C. Monocytes from type 2 diabetic patients have a pro-
inflammatory profile. 1,25-dihydroxyvitamin D(3) works as anti-
inflammatory. Diabetes Res Clin Pract. 2007;77(1):4757.
41. Neve A, Corrado A, Cantatore FP. Immunomodulatory effects of
vitamin D in peripheral blood monocyte-derived macrophages
from patients with rheumatoid arthritis. Clin Exp Med.
2014;14(3):27583.
42. Pilz S, Kienreich K, Rutters F, de Jongh R, van Ballegooijen AJ,
Grübler M, Tomaschitz A, Dekker JM. Role of vitamin D in the
development of insulin resistance and type 2 diabetes. Curr Diab
Rep. 2013;13(2):26170.
43. Calton EK, Keane KN, Newsholme P, Soares MJ. The impact of
vitamin D levels on inflammatory status: a systematic review of
immune cell studies. PLoS One. 2015;10(11):e0141770.
44. Yang M, Yang BO, Gan H, et al. Anti-inflammatory effect of 1,
25dihydroxyvitamin D (3) is associated with crosstalk between
signal transducer and activator of transcription 5 and the vitamin
D receptor in human monocytes. Exp Ther Med. 2015;9:173944.
45. Zuchi MF, Azevedo Pde O, Tanaka AA, Schmitt JV, Martins LE.
Serum levels of 25-hydroxy vitamin D in psoriatic patients. An
Bras Dermatol. 2015;90(3):4302.
46. Kincse G, Bhattoa PH, Herédi E, Varga J, Szegedi A, Kéri J,
Gaál J. Vitamin D3 levels and bone mineral density in pa-
tients with psoriasis and/or psoriatic arthritis. J Dermatol.
2015;42(7):67984.
47. Grazio S, NaglićĐB, AnićB, GrubišićF, Bobek D, Bakula M,
Kavanagh HS, Kuna AT, CvijetićS. Vitamin D serum level, dis-
ease activity and functional ability in different rheumatic patients.
Am J Med Sci. 2015;349(1):469.
48. Miyachi Y, Ohkawara A, Ohkido M, Harada S, Tamaki K,
Nakagawa H, Hori Y, Nishiyama S. Long-term safety and efficacy
of high-concentration (20 microg/g) tacalcitol ointment in psoria-
sis vulgaris. Eur J Dermatol. 2002;12(5):4638.
49. Kircik L. Efficacy and safety of topical calcitriol 3 microg/g oint-
ment, a new topical therapy for chronic plaque psoriasis. J Drugs
Dermatol. 2009;8(8 Suppl):s916.
50. Oquendo M, Abramovits W, Morrell P. Topical vitamin D analogs
available to treat psoriasis. Skinmed. 2012;10:35660.
51. Mason A, Mason J, Cork M, Hancock H, Dooley G. Topical
treatments for chronic plaque psoriasis: an abridged Cochrane
systematic review. J Am Acad Dermatol. 2013;69:799807.
52. Prieto-Pérez R, Cabaleiro T, Daudén E, Ochoa D, Román M,
Abad-Santos F. Pharmacogenetics of topical and systemic treat-
ment of psoriasis. Pharmacogenomics. 2013;14:162334.
202 Rev Endocr Metab Disord (2017) 18:195205
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
53. Ahn CS, Awadalla F, Huang KE, Yentzer B, Dabade TS, Feldman
SR. Patterns of vitamin d analog use for the treatment of psoriasis.
J Drugs Dermatol. 2013;12:90610.
54. Mostafa WZ, Hegazy RA. Vitamin D and the skin: focus on a
complex relationship: a review. J Adv Res. 2015;6(6):793804.
55. Tremezaygues L, Reichrath J. Vitamin D analogs in the treatment
of psoriasis: where are we standing and where will we be going?
Dermatoendocrinology. 2011;3:1806.
56. Balato N, Patruno C, Napolitano M, Patrì A, Ayala F, Scarpa R.
Managing moderate-to-severe psoriasis in the elderly. Drugs
Aging. 2014;31(4):2338.
57. Napolitano M, Megna M, Balato A, Ayala F, Lembo S, Villani A,
Balato N. Systemic treatment of pediatric psoriasis: a review.
Dermatol Ther (Heidelb). 2016;6(2):12542.
58. Megna M, Napolitano M, Balato N, Monfrecola G, Villani A,
Ayala F, Balato A. Efficacy and safety of ustekinumab in a group
of 22 elderly patients with psoriasis over a 2-year period. Clin Exp
Dermatol. 2016;41(5):5646.
59. Van Der Kerkhof PC. Biological activity of vitamin D analogues
in the skin, with special reference to antipsoriatic mechanisms. Br
J Dermatol. 1995;132:67582.
60. Peric M, Koglin S, Dombrowski Y, Gross K, Bradac E, Büchau A,
et al. Vitamin D analog differentially control antimicrobial pep-
tide/"alarmin" expression in psoriasis. PLoS One. 2009;4:e6340.
61. Perez A, Raab R, Chen TC, Turner A, Holick MF. Safety and
efficacy of oral calcitriol (1,25 dihydroxyvitamin D) for the treat-
ment of psoriasis. Br J Dermatol. 1996;134:10708.
62. Wolters M. Diet and psoriasis: experimental data and clinical ev-
idence. Br J Dermatol. 2005;153(4):70614.
63. Barrea L, Macchia PE, Tarantino G, Di Somma C, Pane E, Balato
N, Napolitano M, Colao A, Savastano S. Nutrition: a key environ-
mental dietary factor in clinical severity and cardio-metabolic risk
in psoriatic male patients evaluated by 7-day food-frequency ques-
tionnaire. J Transl Med. 2015;13:303.
64. Barrea L, Balato N, Di Somma C, Macchia PE, Napolitano M,
Savanelli MC, Esposito K, Colao A, Savastano S. Nutrition and
psoriasis: is there any association between the severity of the dis-
ease and adherence to the Mediterranean diet? J Transl Med.
2015;13:18. doi:10.1186/s12967-014-0372-1.
65. Muscogiuri G, Orio F, Colao A. Letter to the editor: vitamin D: a
wonder drug for the cure of type 2 diabetes? J Clin Endocrinol
Metab. 2016;101(4):L434.
66. Qi D, Nie X, Cai J. The effect of vitamin D supplementation on
hypertension in non-CKD populations: a systemic review and
meta-analysis. Int J Cardiol. 2016;227:17786.
67. Orgaz-Molina J, Magro-Checa C, Arrabal-Polo MA, Raya-
Álvarez E, Naranjo R, Buendía-Eisman A, Arias-Santiago S.
Association of 25-hydroxyvitamin D with metabolic syndrome
in patients with psoriasis: a case-control study. Acta Derm
Venereol. 2014;94(2):1425.
68. Wise J. NICE advises certain groups to take daily vitamin D sup-
plement. BMJ. 2014;348:g3349.
69. Cashman KD, Kiely M. EURRECA-estimating vitamin D re-
quirements for deriving dietary reference values. Crit Rev Food
Sci Nutr. 2013;53(10):1097109.
70. Cashman KD, Kiely M. Recommended dietary intakes for vitamin
D: where do they come from, what do they achieve and how can
we meet them? J Hum Nutr Diet. 2014;27(5):43442.
71. Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM,
Clinton SK, Durazo-Arvizu RA, Gallagher JC, Gallo RL, Jones
G, Kovacs CS, Mayne ST, Rosen CJ, Shapses SA. The 2011
report on dietary reference intakes for calcium and vitamin D from
the Institute of Medicine: what clinicians need to know. J Clin
Endocrinol Metab. 2011;96(1):538.
72. Holick MF, Binkley NC, Bischoff-Ferrari HA, Gordon CM,
Hanley DA, Heaney RP, Murad MH, Weaver CM. Evaluation,
treatment, and prevention of vitamin D deficiency: an Endocrine
Society clinical practice guideline. J Clin Endocrinol Metab.
2011;25:191130.
73. Dawson-Hughes B, Mithal A, Bonjour JP, Boonen S, Burckhardt
P, Fuleihan GE, Josse RG, Lips P, Morales-Torres J, Yoshimura N.
IOF position statement: vitamin D recommendations for older
adults. Osteoporos Int. 2010;21(7):11514.
74. Lamberg-Allardt C, Brustad M, Meyer HE, Steingrimsdottir L.
Vitamin D - a systematic literature review for the 5th edition of
the Nordic nutrition recommendations. Food Nutr Res. 2013;57.
doi:10.3402/fnr.v57i0.22671.
75. Cashman KD, Hill TR, Lucey AJ, Taylor N, Seamans KM,
Muldowney S, Fitzgerald AP, Flynn A, Barnes MS, Horigan G,
Bonham MP, Duffy EM, Strain JJ, Wallace JM, Kiely M.
Estimation of the dietary requirement for vitamin D in healthy
adults. Am J Clin Nutr. 2008;88(6):153542.
76. Lanham-New SA, Buttriss JL, Miles LM, Ashwell M, Berry JL,
Boucher BJ, Cashman KD, Cooper C, Darling AL, Francis RM,
Fraser WD, de Groot CP, Hyppönen E, Kiely M, Lamberg-Allardt
C, Macdonald HM, Martineau AR, Masud T, Mavroeidi A,
Nowson C, Prentice A, Stone EM, Reddy S, Vieth R, Williams
CM. Proceedings of the rank forum on vitamin D. Br J Nutr.
2011;105(1):14456.
77. Outila TA, Mattila PH, Piironen VI, Lamberg-Allardt CJ.
Bioavailability of vitamin D from wild edible mushrooms
(Cantharellus tubaeformis) as measured with a human bioassay.
Am J Clin Nutr. 1999;69(1):958.
78. Calvo MS, Whiting SJ, Barton CN. Vitamin D fortification in the
United States and Canada: current status and data needs. Am J
Clin Nutr. 2004;80(6 Suppl):1710S6S.
79. Ovesen L, Brot C, Jakobsen J. Food contents and biological ac-
tivity of 25-hydroxyvitamin D: a vitamin D metabolite to be reck-
oned with? Ann Nutr Metab. 2003;47(34):10713.
80. Schmid A, Walther B. Natural vitamin D content in animal prod-
ucts. Adv Nutr. 2013;4(4):45362.
81. Mattila PH, Valkonen E, Valaja J. Effect of different vitamin D
supplementations in poultry feed on vitamin D content of eggs and
chicken meat. J Agric Food Chem. 2011;59(15):8298303.
82. Kühn J, Schutkowski A, Hirche F, Baur AC, Mielenz N, Stangl
GI. Non-linear increase of vitamin D content in eggs from chicks
treated with increasing exposure times of ultraviolet light. J
Steroid Biochem Mol Biol. 2015;148:713.
83. Liu J, Greenfield H, Strobel N, Fraser DR. The influence of lati-
tude on the concentration of vitamin D3 and 25-hydroxy-vitamin
D3 in Australian red meat. Food Chem. 2013;140(3):4325.
84. Rejnmark L, Avenell A, Masud T, Anderson F, Meyer HE,
Sanders KM, Salovaara K, Cooper C, Smith HE, Jacobs ET,
Torgerson D, Jackson RD, Manson JE, Brixen K, Mosekilde L,
Robbins JA, Francis RM, Abrahamsen B. Vitamin D with calcium
reduces mortality: patient level pooled analysis of 70,528 patients
from eight major vitamin D trials. J Clin Endocrinol Metab.
2012;97(8):267081.
85. Kiely M, Black LJ. Dietary strategies to maintain adequacy of
circulating 25-hydroxyvitamin D concentrations. Scand J Clin
Lab Invest Suppl. 2012;243:1423.
86. Black LJ, Walton J, Flynn A, Cashman KD, Kiely M. Small in-
crements in vitamin D intake by Irish adults over a decade show
that strategic initiatives to fortify the food supply are needed. J
Nutr. 2015;145(5):96976.
87. Seamans KM,Cashman KD. Existing and potentially novel func-
tional markers of vitamin D status: a systematic review. Am J Clin
Nutr. 2009;89(6):1997S2008S.
88. U.S. Department of Agriculture, Agricultural Research
Service. 2011. USDA National Nutrient Database for standard
reference, release 24. Nutrient Data Laboratory Home Page,
http://www.ars.usda.gov/ba/bhnrc/ndl.
Rev Endocr Metab Disord (2017) 18:195205 203
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
89. van den Berg H. Bioavailability of vitamin D. Eur J Clin Nutr.
1997;51(Suppl 1):S769.
90. Grossmann RE, Tangpricha V. Evaluation of vehicle substances
on vitamin D bioavailability: a systematic review. Mol Nutr Food
Res. 2010;54(8):105561.
91. Wagner D, Sidhom G, Whiting SJ, Rousseau D, Vieth R. The
bioavailability of vitamin D from fortified cheeses and supple-
ments is equivalent in adults. J Nutr. 2008;138(7):136571.
92. Tangpricha V, Koutkia P, Rieke SM, Chen TC, Perez AA, Holick
MF. Fortification of orange juice with vitamin D: a novel approach
for enhancing vitamin D nutritional health. Am J Clin Nutr.
2003;77(6):147883.
93. Mattila P, Lehikoinen K, Kiiskinen T, Piironen V. Cholecalciferol
and 25-hydroxycholecalciferol content of chicken egg yolk as
affected by the cholecalciferol content of feed. J Agric Food
Chem. 1999;47(10):408992.
94. Jakobsen J, Knuthsen P. Stability of vitamin D in foodstuffs during
cooking. Food Chem. 2014;148:1705.
95. Cashman KD. Vitamin D: Dietary requirements and food fortifi-
cation as a means of helping achieve adequate vitamin D status. J
Steroid Biochem Mol Biol. 2015;148:1926.
96. Tripkovic L, Lambert H, Hart K, Smith CP, Bucca G, Penson S,
Chope G, Hyppönen E, Berry J, Vieth R, Lanham-New S.
Comparison of vitamin D2 and vitamin D3 supplementation in
raising serum 25-hydroxyvitamin D status: a systematic review
and meta-analysis. Am J Clin Nutr. 2012;95(6):135764.
97. Shab-Bidar S, Bours S, Geusens PP, Kessels AG, van den Bergh
JP. Serum 25(OH)D response to vitamin D3 supplementation: a
meta-regression analysis. Nutrition. 2014;30(9):97585.
98. Autier P, Gandini S, Mullie P. A systematic review: influence of
vitamin D supplementation on serum 25-hydroxyvitamin D con-
centration. J Clin Endocrinol Metab. 2012;97(8):260613.
99. Zittermann A, Ernst JB, Gummert JF, Börgermann J. Vitamin D
supplementation, body weight and human serum 25-
hydroxyvitamin D response: a systematic review. Eur J Nutr.
2014;53(2):36774.
100. Heaney RP, Davies KM, Chen TC, Holick MF, Barger-Lux MJ.
Human serum 25-hydroxycholecalciferol response to extended
oral dosing with cholecalciferol. Am J Clin Nutr. 2003;77(1):
20410.
101. Zittermann A, Prokop S, Gummert JF, Börgermann J. Safety is-
sues of vitamin D supplementation. Anti Cancer Agents Med
Chem. 2013;13(1):410.
102. EFSA Panel on Dietetic Products, Nutrition and Allergies (NDA).
Scientific opinion on the tolerable upper intake level of vitamin D.
EFSA J. 2012;10:145.
103. Davidson MB, Duran P, Lee ML, Friedman TC. High-dose vita-
min D supplementation in people with prediabetes and
hypovitaminosis D. Diabetes Care. 2013;36(2):2606.
104. Jorde R, Strand Hutchinson M, Kjærgaard M, Sneve M, Grimnes
G. Supplementation with high doses of vitamin D to subjects
without vitamin D deficiency may have negative effects: pooled
data from four intervention trials in Tromsø. ISRN Endocrinol.
2013;2013:348705.
105. Brouwer-Brolsma EM, Bischoff-Ferrari HA, Bouillon R, Feskens
EJ, Gallagher CJ, Hypponen E, Llewellyn DJ, Stoecklin E,
Dierkes J, Kies AK, Kok FJ, Lamberg-Allardt C, Moser U, Pilz
S, Saris WH, van Schoor NM, Weber P, Witkamp R, Zittermann
A, de Groot LC. Vitamin D: Do we get enough? A discussion
between vitamin D experts in order to make a step towards the
harmonisation of dietary reference intakes for vitamin D across
Europe. Osteoporos Int 2013;24(5):15671577.
106. Fu LW, Vender R. Systemic role for vitamin d in the treatment of
psoriasis and metabolic syndrome. Dermatol Res Pract.
2011;2011:276079.
107. Lakka HM, Laaksonen DE, Lakka TA, Niskanen LK, Kumpusalo
E, Tuomilehto J, Salonen JT. The metabolic syndrome and total
and cardiovascular disease mortality in middle-aged men. JAMA.
2002;288(21):270916.
108. Abramovits W. Calcitriol 3 microg/g ointment: an effective and
safe addition to the armamentarium in topical psoriasis therapy. J
Drugs Dermatol. 2009;8(8 Suppl):s1722.
109. van de Kerkhof PC. The topical treatment of psoriasis. Clin Exp
Dermatol. 2005;30(2):2058.
110. Merola JF, Han J, Li T, Qureshi AA. No association between
vitamin D intake and incident psoriasis among US women. Arch
Dermatol Res. 2014;306(3):3057.
111. Ezquerra GM, Regana MS, Millet PU. Combination of acitretin
and oral calcitriol for treatment of plaque-type psoriasis. Acta
Derma Venereol. 2007;87:44950.
112. Azfar RS, Gelfand JM. Psoriasis and metabolic disease: epidemi-
ology and pathophysiology. Curr Opin Rheumatol. 2008;20(4):
41622.
113. Barrea L, Macchia PE, Di Somma C, Napolitano M, Balato A,
Falco A, Savanelli MC, Balato N, Colao A, Savastano S.
Bioelectrical phase angle and psoriasis: a novel association with
psoriasis severity, quality of life and metabolic syndrome. J Transl
Med. 2016;14(1):130.
114. Santos M, Fonseca HM, Jalkh AP, Gomes GP, Cavalcante
AS. Obesity and dyslipidemia in patients with psoriasis treat-
edatadermatologicclinicin Manaus. An Bras Dermatol.
2013;88:9136.
115. Barrea L, Nappi F, Di Somma C, Savanelli MC, Falco A, Balato
A, Balato N, Savastano S. Environmental Risk Factors in
Psoriasis: The Point of View of the Nutritionist. Int J Environ
Res Public Health. 2016;13(5)
116. Carrascosa JM, Rocamora V, Fernandez-Torres RM, Jimenez-
Puya R, Moreno JC, Coll-Puigserver N, et al. Obesity and psori-
asis: inflammatory nature of obesity, relationship between psoria-
sis and obesity, and therapeutic implications. Actas
Dermosifiliogr. 2014;105:3144.
117. Debbaneh M, Millsop JW, Bhatia BK, Koo J, Liao W. Diet and
psoriasis, part I: impact of weight loss interventions. J Am Acad
Dermatol. 2014;71:13340.
118. Wolk K, Mallbris L, Larsson P, Rosenblad A, Vingard E, Stahle
M. Excessive body weight and smoking associates with a high risk
of onset of plaque psoriasis. Acta Derm Venereol. 2009;89:4927.
119. Beygi S, Lajevardi V, Abedini R. C-reactive protein in psoriasis: a
review of the literature. J Eur Acad Dermatol Venereol: JEADV.
2014;28:70011.
120. Tobin AM, Hackett CB, Rogers S, Collins P, Richards HL, OShea
D, et al. Body mass index, waist circumference and HOMA-IR
correlate with the psoriasis area and severity index in patients with
psoriasisreceiving phototherapy. Br J Dermatol. 2014;171:4368.
121. Bosy-Westphal A, Booke CA, Blocker T, Kossel E, Goele K,
Later W, et al. Measurement site for waist circumference affects
its accuracy as an index of visceral and abdominal subcutaneous
fat in a Caucasian population. J Nutr. 2010;140:95461.
122. Jensen MD. Role of body fat distribution and the metabolic com-
plications of obesity. J Clin Endocrinol Metab. 2008;93:S5763.
123. Toussirot E, Aubin F, Dumoulin G. Relationships between adipose
tissue and psoriasis, with or without arthritis. Front Immunol.
2014;5:368.
124. Klöting N, Blüher M. Adipocyte dysfunction, inflammation
and metabolic syndrome. Rev Endocr Metab Disord.
2014;15(4):27787.
125. Prasad P, Kochhar A. Interplay of vitamin D and metabolic syn-
drome: a review. Diabetol Metab Syndr. 2016;10:10512.
126. Wolk K, Sabat R. Adipokines in psoriasis: an important link be-
tween skin inflammation and metabolic alterations. Rev Endocr
Metab Disord. 2016; doi:10.1007/s11154-016-9381-0.
204 Rev Endocr Metab Disord (2017) 18:195205
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
127. Devaraj S, Jialal G, Cook T, Siegel D, Jialal I. Low vitamin D
levels in northern American adults with the metabolic syndrome.
Horm Metab Res. 2011;43(1):724.
128. Baz-Hecht M, Goldfine AB. The impact of vitamin D deficiency
on diabetes and cardiovascular risk. Curr Opin Endocrinol
Diabetes Obes. 2010;17(2):1139.
129. Earthman CP, Beckman LM, Masodkar K, Sibley SD. The link
between obesity and low circulating 25-hydroxyvitamin D con-
centrations: considerations and implications. Int J Obes. 2012;36:
38796.
130. JuSY,JeongHS,KimDH.BloodvitaminDstatusandmeta-
bolic syndrome in the general adult population: a dose-
response meta-analysis. J Clin Endocrinol Metab. 2014;99(3):
105363.
131. Vimaleswaran KS, Berry DJ, Lu C, Tikkanen E, Pilz S, Hiraki LT,
Cooper JD, Dastani Z, Li R, et al. Causal relationship between
obesity and vitamin D status: Bi-directional Mendelian randomi-
zation analysis of multiple cohorts. PLoS Med. 2013;10:
e1001383.
132. Vanlint S. Vitamin D and Obesity. Nutrients. 2013;5:94956.
133. Barrea L, Savastano S, Di Somma C, Savanelli MC, Nappi F,
Albanese L, Orio F, Colao A. Low serum vitamin D-status, air
pollution and obesity: a dangerous liaison. Rev Endocr Metab
Disord. 2016; doi:10.1007/s11154-016-9388-6.
Rev Endocr Metab Disord (2017) 18:195205 205
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... On the other hand, evidence is accumulating that vitamin D might represent a key modulator of immune and inflammation mechanisms. Recently, a role for vitamin D in the pathogenesis of different skin diseases, including psoriasis, has been reported (12). ...
... 12: The distribution of plague psoriatic patients according to ABO Blood group system.The ABO blood group antigens play a role in the pathophysiology of some diseases and several researches have investigated this relationship in the field of dermatology, like psoriasis. In the present study, an attempt was made to find the distribution of blood group 43 types in patients with chronic plaque type psoriasis. ...
Thesis
Full-text available
Psoriasis is a complex, chronic, multifactorial, inflammatory disease that involves hyperproliferation of the keratinocytes in the epidermis, with an increase in the epidermal cell turnover rate. Environmental, genetic, and immunologic factors appear to play a role. Toll-like receptors (TLRs) are key transmembrane proteins in both the innate and adaptive immune responses which are known to be integral processes in psoriasis. Previous studies in different peoples have shown that genetic variations in TLR2, particularly those arising from polymorphisms, are often linked to different Skin diseases and disorders like psoriasis. Therefore, the aim of this case- control study was evaluated the association of the TLR-2 gene polymorphism at (rs4696480) position and some physiological parameter with psoriatic diseases in Iraqi patients at Anbar province
... While specific details about the doses and durations of vitamin D administration were not provided, the study showed clinical improvement in the Psoriasis Area and Severity Index (PASI) score for 88% of patients, and the other reported moderate or better improvements in 25-50% of patients with psoriasis. 30 McCullough et al. demonstrated significant clinical benefits in psoriasis, with doses ranging from 25,000 IU/day to 60,000 IU/day, without the emergence of toxicity or hypercalcemia. However, another study showed that the oral administration of 10,000 IU/day to 25,000 IU/day of vitamin D is considered safe for the general population. ...
... Numerous studies have provided evidence supporting the effectiveness and safety of employing topical calcipotriol in the treatment of localized plaque psoriasis. 30 The US Food and Drug Administration (FDA) has approved topical calcitriol for the treatment of psoriasis due to its minimal impact on serum calcium levels. Numerous studies have documented the effectiveness and safety of various vitamin D analogs, including calcitriol, calcipotriol, tacalcitol, hexafluoro-1,25-dihydroxyvitamin D3, and maxacalcitol, in the topical treatment of psoriasis. ...
Article
Full-text available
Vitamin D was recognized for its pivotal role in bone formation, but it is now recognized for its impact on the optimal functioning of various tissues throughout the body, including the brain, heart, muscles, immune system, and skin. Vitamin D is a fat-soluble prohormone steroid with endocrine, paracrine, and autocrine functions. Vitamin D receptors and the enzymes that can convert 25-hydroxyvitamin D [25(OH)D] into the active form, 1,25-hydroxyvitamin D [1,25(OH)D], have been identified in numerous cells throughout the body, including the skin. Vitamin D analogs are widely acknowledged for effective treatment of psoriasis vulgaris due to their anti-proliferative and pro-differentiating impact on keratinocytes. Beyond this, emerging roles for vitamin D in skin health include immunomodulatory and anti-apoptotic effects, suggesting potential applications in conditions like atopic dermatitis and infections including verucca vulgaris. Epidemiological studies have linked low vitamin D levels to autoimmune diseases like vitiligo, leading to the use of topical vitamin D in its treatment. Recent evidence also suggests that cutaneous vitamin D synthesis may contribute to preventing skin malignancies, and oral supplementation may reduce cancer mortality. Its role in the pathogenesis of various dermatological diseases has not been an exception and has been extensively studied in recent years. In this review, we will shed light on roles of vitamin D in various skin disorders.
... When Vitamin D3 is used orally, patient compliance rates are less than 60% (Bubshait et al., 2018), which has resulted in several researchers looking for alternative delivery systems, one of which is topical. The effectiveness of vitamin D3 topically is of significant effect when used with potent topical steroids (Barrea et al., 2017). A particular study found that Vitamin D3 could reduce oedema after seven days of administration (Almeida Moreira Leal et al., 2020). ...
Article
Background: Vitamin D3 plays a role in immunity, especially in the body's regulation of the inflammatory response. The limitations of vitamin D3 are poor bioavailability and water-insoluble. Nanostructured Lipid Carriers (NLC) can increase the solubility of water-insoluble active ingredients. Objective: To develop NLC as a delivery agent of vitamin D3 for topical application. Method: NLC was created using a high-shear homogenisation method with various lipid ratios. NLC uses solid lipids (Monostearin) and liquid lipids (Miglyol 812) with variations in the ratios of F1 (7:3), F2 (8:2) and F3 (9:1). NLC is expected to be able to protect the active ingredients in the lipid matrix so that the stability of the active ingredients increases. Characterisation was performed in organoleptic, pH, particle size, viscosity, zeta potential, and entrapment efficiency. Result: Vitamin D3 NLC system had good characteristics: pH suitable for topical use, particle size < 600nm, PI < 0.5, Zeta potential -26.80 to -33.76. F1 resulted in the highest entrapment efficiency of 82.18%. Conclusion: Vitamin D3 NLC systems with different lipid concentration ratios affect the physical and chemical characteristics. Increasing the liquid lipid content in the Vit D3 NLC system can reduce particle size and viscosity and increase entrapment efficiency.
... Vitamin D deficiency is implicated in the pathophysiology of psoriasis, with the active form of vitamin D and its receptors playing a role in regulating keratinocytes. 3,4 Various studies highlight vitamin D's impact on keratinocyte proliferation and differentiation, crucial for maintaining the balance of the cutaneous immune system and apoptosis. Genetic variations in vitamin D receptors contribute to vitamin D resistance in autoimmune diseases, necessitating higher doses for clinical efficacy. ...
Article
Full-text available
Background: Insights into psoriasis as a systemic ailment have spurred the investigation of novel therapies due to dissatisfaction with current treatments and significant quality of life impairment. Vitamin D3 deficiency is implicated in psoriasis pathophysiology, with studies highlighting its impact on keratinocyte regulation and autoimmune disease resistance. This review critically evaluates evidence on high-dose oral vitamin D3 supplementation for psoriasis management, aiming to assess its effectiveness, treatment outcomes, and safety profile to inform clinician. Methods: This systematic review focused on full-text English literature published between 2017 and 2024 using the PRISMA 2020 guidelines. Editorials and review pieces published in the same journal as the submission without a DOI were not accepted. The literature was compiled using PubMed, ScienceDirect, and SagePub, among other online venues. Result: Our research team initially collected numerous publications from credible sources such as Science Direct, PubMed, and JAMA Dermatology. Employing a meticulous three-tier screening approach, we identified only five papers that were directly pertinent to our ongoing systematic assessment. Subsequently, we conducted a comprehensive examination of the entire text and further selected articles; case control study, double-blind, randomized, placebo-controlled study, randomized controlled trial, a bidirectional two-sample mendelian randomization analysis, and observational cross-sectional study. Conclusion: In conclusion, maintaining serum vitamin D levels above 30 ng/ml may benefit severe psoriasis, while oral vitamin D2 supplementation showed promise in mild cases. Larger trials are needed for severe psoriasis, and future research should explore higher doses. Additionally, a study found no support for monthly vitamin D3 supplementation in older mild psoriasis patients. We confirmed the causal link between vitamin D levels and psoriasis, identified associations with increased risk, and explored effects on atopic dermatitis. While no significant difference was found in serum vitamin D levels between psoriasis patients and controls, lower levels in psoriasis suggest various influences.
Chapter
Vitamin D, traditionally renowned for its function in regulating calcium levels and promoting skeletal health, has become a focus of scientific investigation because of its complex involvement in immune system modulation. This chapter focuses on its immune function and comprehensively explores the multifaceted functions of vitamin D, its relevance to autoimmune disorders, susceptibility to infections, and the potential usefulness of dietary vitamin D supplements. Recent discoveries have brought to light the existence of vitamin D receptors on immune cells, which facilitate the formation of active vitamin D metabolites. This underscores the dual action of vitamin D in protecting bone homeostasis while modulating immune responses. In particular, low blood levels of vitamin D have been linked with a higher susceptibility to autoimmune disorders, underscoring its critical importance. While vitamin D supplementation shows promise for immune enhancement, clinical findings have yielded inconclusive results, requiring further investigation. To optimize immune function and overall well-being, maintaining optimal vitamin D levels (40–60 ng/mL) through judicious sun exposure and judicious supplementation is recommended. This chapter is valuable for understanding the complex interplay between the immune system and vitamin D, which goes beyond its classical function in calcium regulation and skeletal health. This underscores the imperative to comprehensively understand the diverse functions of vitamin D, especially among individuals at increased threat of autoimmune conditions and increased susceptibility to infectious agents.
Article
Scalp psoriasis is a common manifestation of psoriasis that significantly impacts a patient's quality of life. About 80% of cases of psoriasis involve the scalp, making it the most frequently affected area of the body. The treatment of scalp psoriasis is particularly crucial because of its hard-to-treat nature and substantial adverse impacts on overall well-being. Along with the physical symptoms of discomfort and itching, psoriasis, especially when it affects the scalp, can cause severe psychological damage. Treating scalp psoriasis can be challenging due to its location and associated symptoms, such as scaling and pruritus, which is why various drugs have become widely used for refractory cases. Topical treatments like corticosteroids and vitamin D analogs manage scalp psoriasis by reducing inflammation and regulating skin cell growth. Tar-based shampoos, salicylic acid solutions, and moisturizers control scaling. Phototherapy with UVB light reduces inflammation. Severe cases may require systemic medications such as oral retinoids and immunosuppressants. While various therapies are accessible for scalp psoriasis, concerns arise due to their limited advantages and the absence of controlled studies assessing their effectiveness. Considering these challenges, there is a clear demand for innovative approaches to address this condition effectively. Recent advancements in topical therapies, phototherapy, systemic agents, and complementary therapies have shown promising results in managing scalp psoriasis. Also, the advent of biologics, specifically anti-IL-17 and anti-IL-23 drugs for scalp psoriasis, has seen significant improvements. The review highlights the lack of well-tolerated and effective treatments for scalp psoriasis and underscores the importance of further research in this area. The objective of this review is to clarify the different treatment options currently available or being investigated in clinical trials for managing scalp psoriasis.
Book
Full-text available
D Vitamini Kimyasal Yapısı ve Metabolizması Hülya Cenk D Vitamini Ve Genetik Aydın Rüstemoğlu D Vitamininin Normal Serum Düzeyleri, D Vitamin Düzeylerini Etkileyen Faktörler Ve D Vitamini Yetmezliği Sabiye Akbulut Serum D Vitamininin Ölçümü Andaç Uzdoğan, Çiğdem Yücel D Vitamini Biyoyararlanımı ve Doğal Beslenme Kaynakları Atilla Çifci, Halil İbrahim Yakut Sistemik D Vitamini Tedavi Ajanları, Biyoyararlanımı ve Tedavi Yönetimi Işıl Deniz Oğuz Topikal D Vitamini Tedavisi, Tedavi Yönetimi ve Kullanıldığı Hastalıklar Dursun Türkmen Deride D Vitamini Sentezi Mekanizmaları Abdullah Demirbaş, Ömer Faruk Elmas Güneşten Koruyucu Kullanımı ve D Vitamini Nursel Dilek, Yunus Saral D Vitamininin Deri Yapısı ve Fizyolojisine Etkisi Pelin Hızlı Deri Yaşlanması ve D Vitamini Ülker Gül Psoriasis ve D Vitamini Ülker Gül Psöriatik Artrit ve D Vitamini Mehmet Uçar Atopik Dermatit ve D Vitamini Ayşegül Ertuğrul, İlknur Bostancı Mast Hücresi ve Kutanöz Mastositozda D Vitamini Selçuk Doğan, Tülin Çataklı, İlknur Bostancı Ürtiker ve D Vitamini Kemal Özyurt Kaşıntı ve D Vitamini Kübra Yüce Atamulu Likenoid Dermatozlar ve D Vitamini Nihal Altunışık Vitiligo ve D Vitamini Ayşe Akbaş Melasma ve D Vitamini İbrahim Etem Arıca Rozase ve D Vitamini Nalan Saraç Akne ve D Vitamini Selma Korkmaz Hidradenitis Süpürativa ve D Vitamini Yılmaz Ulaş Seboreik Dermatit ve D Vitamini Dilek Başaran Otoimmün Büllöz Hastalıklar ve D Vitamini Sezgi Sarıkaya Solak Bağ Doku Hastalıkları ve D Vitamini Kevser Gök Behçet Hastalığı ve D Vitamini Şule Ketenci Ertaş, Ragıp Ertaş İdiyopatik Fotodermatozlar ve D Vitamini Bülent Nuri Kalaycı İktiyozis ve D Vitamini Tubanur Çetinarslan Epidermolizis Bülloza ve Vitamin D Eda Haşal Kseroderma Pigmentozum, Epidermodisplasia Verrusiformis ve D Vitamini Derya Yayla Nevüsler ve D Vitamini Serpil Şener, Suat Sezer Aktinik Keratoz ve Seboreik Keratozda D Vitamini Mahmut Sami Metin Deri Maliniteleri ve D Vitamini Sevda Önder Vaskülitler ve Vitamin D Havva Hilal Ayvaz Venöz Trombozis ve D Vitamini Cahit Yavuz Yara İyileşmesi ve D Vitamini Bülent Nuri Kalaycı Diyabetik Ayak Ülseri ve D Vitamini Gözde Ulutaş Demirbaş, Abdullah Demirbaş Granülomatöz Hastalıklar ve D Vitamini Selma Bakar Dertlioğlu Deri Enfeksiyonları ve Vitamin D Atıl Avcı Oral Mukoza Hastalıkları ve D Vitamini Ali İhsan Güleç Tırnak Sağlığı ve Hastalıklarında D Vitamini Hülya Cenk Alopesiler ve D Vitamini Munise Daye Hirsutizm ve D Vitamini Efşan Gürbüz Yontar Sistemik Kortikosteroid Kullanımında D Vitamini Desteği Selma Korkmaz Fototerapi ve D Vitamini Tuğba Özkök Akbulut Covıd-19 Ve Vitamin D Sibel Altunışık Toplu D Vitamini Tedavisinin Yan Etkileri ve D Vitamini Tedavisi Sürecinde Dikkat Edilecek Hususlar Dursun Türkmen, Nihal Altunışık D Vitamini Ve İlaç İlaç Etkileşimleri Şule Gökşin D Vitamini İntoksikasyonu Bedriye Müge SÖNMEZ
Chapter
As well as quantifying the burden of skin disease, epidemiology provides a direct method for assessing the causes of skin diseases. Identification of causes is a necessary step in pursuing the ultimate goal of disease prevention. This chapter covers the importance of thinking in terms of population rather than individuals, and provides a summary of the substantial global morbidity caused by skin disease. Risk factors from genetics to environment are outlined, along with describing the natural history and associations of common skin diseases. Finally, there is a description of how the principles and data from epidemiological studies provide the cornerstone to planning appropriate dermatology services. The chapter closes by providing the reader with a glossary of terms and a checklist to help with reading epidemiological studies.
Article
Full-text available
Sphingolipid metabolism was examined in human promyelocytic leukemia HL-60 cells. Differentiation of HL-60 cells with 1 α, 25-dihydroxyvitamin D3 (vitamin D3; 100 nM) was accompanied by sphingomyelin turnover. Maximum turnover of [³H]choline-labeled sphingomyelin occurred 2 h following vitamin D3 treatment, with sphingomyelin levels decreasing to 77 ± 6% of control and returning to base-line levels by 4 h. Ceramide and phosphorylcholine were concomitantly generated. Ceramide mass levels increased by 55% at 2 h following vitamin D3 treatment and returned to base-line levels by 4 h. The amount of phosphorylcholine produced equaled the amount of sphingomyelin hydrolyzed, suggesting the involvement of a sphingomyelinase. Vitamin D3 treatment resulted in a 90% increase in the activity of a neutral sphingomyelinase from HL-60 cells. The inferred role of sphingomyelin hydrolysis in the induction of cell differentiation was investigated using an exogenous sphingomyelinase. When a bacterial sphingomyelinase was added at concentrations that caused a similar degree of sphingomyelin hydrolysis as 100 nM vitamin D3, it enhanced the ability of subthreshold levels of vitamin D3 to induce HL-60 cell differentiation. This study demonstrates the existence of a “sphingomyelin cycle” in human cells. Such sphingolipid cycles (Hannun, Y., and Bell, R. (1989) Science 243, 500–507) may function in a signal transduction pathway and in cellular differentiation.
Article
Full-text available
Background: The bioavailability of vitamin D from mushrooms in humans is unknown. Objective: We investigated the bioavailability of vitamin D from wild edible mushrooms (Cantharellus tubaeformis) using the increase in serum 25-hydroxyvitamin D concentrations as a measure of vitamin D bioavailability. Design: Twenty-seven volunteers with serum 25-hydroxyvitamin D concentrations <60 nmol/L (x̄: 38.5 nmol/L; range: 15–60 nmol/L) were randomly divided into 3 groups of 9 persons each. For 3 wk, excluding Saturdays and Sundays, group 1 received mushrooms (C. tubaeformis) providing 14 μg ergocalciferol/d with their lunch, group 2 (control) received an ergocalciferol supplement providing 14 μg/d, and group 3 (also a control) received no supplementation. Results: At the beginning of the study, mean serum 25-hydroxyvitamin D concentrations did not differ significantly among the groups (P = 0.280). When all 3 groups were considered, serum 25-hydroxyvitamin D concentrations showed different time-related changes among the groups during the study: group (P = 0.388), time (P = 0.000), and group × time (P = 0.001). When groups 1 and 2 were compared with group 3, serum 25-hydroxyvitamin D concentrations at 3 wk differed significantly between groups 1 and 3 (P = 0.032) as well as between groups 2 and 3 (P = 0.004). Serum 25-hydroxyvitamin D concentrations at 3 wk did not differ significantly between groups 1 and 2 (P = 0.317). Conclusions: We showed for the first time that ergocalciferol was well absorbed from lyophilized and homogenized mushrooms in humans and that vitamin D bioavailability can be studied in humans with such an experimental protocol.
Article
Full-text available
Nuclear receptor interacting protein 1 (NRIP1, also known as RIP140) is a co-regulator for various transcriptional factors and nuclear receptors, and has been shown to take part in many biological and pathological processes, such as regulating mammary gland development and inflammatory response. The aim of this study is to investigate the expression of NRIP1 and to explore its roles in the pathogenesis of psoriasis. Thirty active psoriasis patients and 16 healthy volunteers were enrolled for this study. qRT-PCR analyses found that both NRIP1 and RelA/p65 were elevated in psoriatic lesions compared to psoriatic non-lesions and normal controls, and also overexpressed in peripheral blood mononuclear cell (PBMCs) of psoriasis patients. Suppression of NRIP1 in HaCaT cells could significantly inhibit cell growth and induce apoptosis, and the suppression of NRIP1 in CD4? T cells isolated from psoriasis patients could downregulate the expression of RelA/p65 and decrease the secretion of IL-17. Furthermore, in Nrip1 knockout mice, IMQ-induced inflammation of skin was delayed and the RelA/p65 expression in lesions was reduced. In conclusion, our data suggests that NRIP1 is overexpressed both in skin and PBMCs of psoriasis patients and may be involved in the abnormal proliferation and apoptosis of keratinocytes, as well as the immune reaction through the regulation of RelA/p65. Therefore, NRIP1 may be a potential therapeutic target for psoriasis.
Article
Full-text available
The aim of this review is to provide a general overview of the possible associations among the vitamin D status, air pollution and obesity. Sunlight exposure accounts in humans for more than 90 % of the production of vitamin D. Among emerging factors influencing sunlight-induced synthesis of vitamin D, prospective and observational studies proved that air pollution constitutes an independent risk factor in the pathogenesis of vitamin D hypovitaminosis. In addition, environmental pollutants can affect risk of obesity when inhaled, in combination with unhealthy diet and lifestyle. In turn, obesity is closely associated with a low vitamin D status and many possible mechanisms have been proposed to explain this association. The associations of air pollution with low vitamin D status on the hand and with obesity on the other hand, could provide a rationale for considering obesity as a further link between air pollution and low vitamin D status. In this respect, a vicious cycle could operate among low vitamin D status, air pollution, and obesity, with additive detrimental effects on cardio-metabolic risk in obese individuals. Besides vitamin D supplementation, nutrient combination, used to maximize the protective effects against air pollution, might also contribute to improve the vitamin D status by attenuating the “obesogen” effects of air pollution.
Article
Full-text available
Vitamin D related research continues to expand and theorise regarding its involvement in obesity, as both hypovitaminosis D and obesity strike in pandemic proportions. Vitamin D plays an important role in immune system through Vitamin D Receptors (VDR), which are transcription factors located abundantly in the body. Due to this characteristic, it is potentially linked to obesity, which is a state of inflammation involving the release of cytokines from adipose tissue, and exerting stress on other organs in a state of positive energy balance. Research trials in the past couple of years and systematic reviews from SCOPUS and MEDLINE will be discussed. The role of Vitamin D throughout the lifespan (from fetal imprinting until older age), and in various other obesity mediated chronic conditions shall be highlighted. Various mechanisms attributed to the inverse relationship of Vitamin D and obesity are discussed with research gaps identified, particularly the role of adipokines, epigenetics, calcium and type of adipose tissue.
Article
Full-text available
Psoriasis is a chronic inflammatory skin disease most common in Europe, North America, and Australia. The etiology and pathomechanisms underlying the evolution and persistence of the skin alterations are increasingly being understood and have led to the development of effective anti-psoriatic therapies. Apart from the skin manifestations, psoriasis is associated with the metabolic syndrome (MetS), known to increase the risk of type 2 diabetes mellitus and cardiovascular disorders. Research of the last years demonstrated a dysregulated adipokine balance as an important link between inflammation, MetS, and consequential disorders. This article describes selected adipokines and their potential role in both metabolic comorbidity and skin inflammation in psoriasis.
Article
Full-text available
Psoriasis is a common, chronic, immune-mediated skin disease with systemic pro-inflammatory activation, where both environmental and genetic factors contribute to its pathogenesis. Among the risk factors for psoriasis, evidence is accumulating that nutrition plays a major role, per se, in psoriasis pathogenesis. In particular, body weight, nutrition, and diet may exacerbate the clinical manifestations, or even trigger the disease. Understanding the epidemiological relationship between obesity and psoriasis is also important for delineating the risk profile for the obesity-related comorbidities commonly found among psoriatic patients. Moreover, obesity can affect both drug’s pharmacokinetics and pharmacodynamics. Additionally, the overall beneficial effects on the obesity-associated comorbidities, clinical recommendations to reduce weight and to adopt a healthy lifestyle could improve the psoriasis severity, particularly in those patients with moderate to severe disease, thus exerting additional therapeutic effects in the conventional treatment in obese patients with psoriasis. Education regarding modifiable environmental factors is essential in the treatment of this disease and represents one of the primary interventions that can affect the prognosis of patients with psoriasis. The goal is to make psoriatic patients and health care providers aware of beneficial dietary interventions. The aim of this review is to assess the relevance of the environmental factors as modifiable risk factors in psoriasis pathogenesis, with particular regard to the involvement of obesity and nutrition in the management of psoriasis, providing also specific nutrition recommendations.
Article
Objectives: To investigate the vitamin D supplementation on blood pressure control by a systemic review and meta-analysis. Methods: Randomized controlled clinical trials were analyzed, which date from eight studies in databases including MEDLINE, EMBASE, Clinical trials, China Integrated Knowledge Resources Database and the Cochrane library. Results: Total 917 patients from eight randomized controlled trials (RCTs), treatment with vitamin D for more than 3months were analyzed. Meta-analysis showed that vitamin D supplementation slightly reduced the systolic blood pressure (SBP) by 1.964mmHg (95% CI, 0.362-3.566; P=0.016), but not lowered diastolic blood pressure (SMD: -0.087, 95% CI, -0.208-0.033; P=0.155). Subgroup analysis also showed that sBP lowering by vitamin D supplementation was not dose-dependent. Comparison to placebo, there is also no statistical difference in SBP lowering by vitamin D supplementation. Conclusions: This meta-analysis indicated that vitamin D is not an antihypertensive agent although it has a moderate SBP lowering effect. More RCTs are required to observe the role of vitamin D plus other antihypertensive drugs in blood pressure control, and define the optimum dose, dosing interval, and type of vitamin D to administer.
Article
We conducted a mini-review of the literature, focusing on aging-related systemic inflammation and its association with vitamin D (VitD). As main findings, data indicate that inflammation in aging seems to have multiple origins, including immunosenescence, and from intestinal and adipose tissue, where VitD may modulate these three factors, through mechanisms not totally known. From the selected studies, three randomized clinical trials of VitD supplementation showed improvements in inflammatory status, whereas two studies did not. Ten epidemiological studies showed associations between VitD and inflammation, whereas two studies did not. One case study reinforced this association. As such, we can assume a reasonable association between VitD and inflammation in the elderly, and a promising role of supplementation in some situations. However, most studies did not take into account environmental and individual factors such as the season of the year, latitude, skin color or even the use of some medicines; in addition, the doses, time of intervention and the sample sizes differed between the studies. In conclusion, although more controlled VitD studies, both clinical and epidemiological, are necessary, it is important to remember the network of factors involved in systemic inflammation in the elderly; an understanding of the dietetic and non-dietetic factors is needed to offer a realistic approach.European Journal of Clinical Nutrition advance online publication, 28 September 2016; doi:10.1038/ejcn.2016.177.
Article
Introduction: Psoriasis is a common, chronic, immune-mediated inflammatory disorder. The disease is associated with several co-morbidities including cardiovascular disease, metabolic syndrome, and psychiatric disorders. It is important to identify and treat these co-morbidities because they have a strongly negative effect on the overall health of patients with psoriasis. Unfortunately, these co-morbidities are often overlooked and/or left untreated. Therefore, the aim of this review is to discuss the mechanisms of how co-morbidities are associated with psoriasis as well as implications for the clinic to be able to recognize such co-morbidities. Areas covered: This is a review of studies investigating and discussing co-morbidities of psoriasis and screening. Literature was retrieved by searching on the PubMed database using individual and combined search terms related to relevant co-morbidities. Expert Commentary: Effective management of psoriasis involves targeting of both psoriasis and co-morbidities.