Exd 13915

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

| |

Received: 9 August 2018    Revised: 5 February 2019    Accepted: 7 March 2019

DOI: 10.1111/exd.13915

VIEWPOINT

How hormones may modulate human skin pigmentation in


melasma: An in vitro perspective

Muriel Cario1,2,3,4

1
Inserm 1035, Bordeaux, France
2 Abstract
Univ. Bordeaux, Bordeaux, France
3
Aquiderm, Bordeaux, France Melasma is a common acquired hyperpigmentary disorder occurring primarily in
4
National reference center for rare skin photo-­exposed areas and mainly affecting women of childbearing age. To decipher
Diseases, Bordeaux Hospital, Bordeaux,
France
the role of sex hormones in melasma, this viewpoint reviews the effects of sex hor-
Correspondence mones on cutaneous cells cultured in monolayers, in coculture, in 3D models and
Muriel Cario, Univ. Bordeaux, Inserm, explants in the presence or the absence of UV. The data show that sex steroid hor-
BMGIC, UMR1035, Bordeaux, France.
mones, especially oestrogen, can modulate in vitro pigmentation by stimulating mel-
Email: muriel.cario-andre@u-bordeaux.fr
anocytes and keratinocyte pro-­pigmentary factors, but not via fibroblast or mast cell
activation. In vitro data suggest that oestrogen acts on endothelial cell count, which
may in turn increase endothelin-­1 concentrations. However, data on explants re-
vealed that sex steroid even at doses observed during pregnancy cannot induce mel-
anogenesis alone nor melanosome transfer but that it acts in synergy with UVB. In
conclusion, we hypothesize that in predisposed persons, sex steroid hormones initi-
ate hyperpigmentation in melasma by amplifying the effects of UV on melanogenesis
via direct effects on melanocytes or indirect effects via keratinocytes and on the
transfer of melanosomes. They also help to sustain hyperpigmentation by increasing
the number of blood vessels and, in turn, the level of endothelin-­1.

KEYWORDS
endothelial cells, fibroblasts, keratinocytes, melanocytes, oestrogen, progesterone

1 |  H O R M O N E S A N D M E L A S M A I N V I VO to solar radiation seem to be the most important triggers.[1,5,6]


Melasma is mainly characterized by excessive epidermal pigmenta-
Melasma is a common acquired hyperpigmentary disorder occurring tion.[6–9] There are three subtypes: epidermal, dermal and mixed.[5]
primarily in photo-­exposed areas (face, neck, forearms) and mainly Dermal melasma is characterized by the presence of melanophages
affecting women of childbearing age. It can occur in all skin types in the superficial and deep dermis. However, melanophages are also
but is more common in intermediate phototypes (Hispanic, Asiatic, found in perilesional and non-­lesional skin, suggesting that pure
Mediterranean African, Middle-­Eastern populations) and rare in ex- dermal melasma does not exist.[7,9,10] An inapparent type with der-
treme phototypes (I, VI).[1,2] While a clear female predominance of 9 mal pigment deposition has been described in dark women.[5] The
or 10 to 1 is observed, the prevalence female/male differs according epidermal type occurs in 72% of pregnancy-­associated melasma,[4]
to the ethnic group, ranging from 6:1 to 39:1. [1]
While many factors and pregnancy-­induced melasma is associated with earlier onset.[1]
are associated with the onset of the disease, genetic predisposition, [3] Histologically, epidermal melasma is characterized by increased
hormonal influence, especially female sex hormone (oral contracep- melanin throughout the layers of the epidermis, and an increase in
tive use, hormone replacement therapy, pregnancy), [4]
and exposure melanosome number and transfer.[5] However, in Caucasoid women,

Experimental Dermatology. 2019;28:709–718. wileyonlinelibrary.com/journal/exd   © 2019 John Wiley & Sons A/S. |  709
Published by John Wiley & Sons Ltd
|
710       CARIO

melanosomes are transferred more individually than as polymelano- the onset of melasma.[28] Moreover, a testosterone metabolite,
somes.[5,7,11] While melanocytes are enlarged and have prominent 5α–androstane-­3 β–diol, competes with E2 to bind ERβ and thus
dendrites, the change in their number is still debated. elicits an oestrogenic response in cells.[29]
As a photoageing disorder,[12] melasma is also characterized by Oestrogen and progesterone act through receptors expressed in
solar elastosis.[6,7,13] Other features such as damage to the basal both the epidermis and dermis. In human (male and female) adult
membrane,[6,8] an increase in blood vessels and in dermal mast scalp skin, only oestrogen receptor (ER) β was observed in the
cells[6,13] and lymphohistiocytic infiltration have been described.[14] epidermis, fibroblast blood vessels and hair follicles, whereas in
The aetiology of melasma seems more complex than that of senile human neonatal foreskin both ER-­α and ER-­β were detected.[21,30]
lentigo, another photoageing disorder, which can be attributed Oestrogen and progesterone receptors are expressed differently in
[15]
mainly to dysregulation of UVA-­induced fibroblasts. As summa- melasma skin than in perilesional or non-­lesional skin (Table 1 [31–34]).
[12]
rized by Passeron and Picardo, solar radiation (UVA, UVB and The presence of these receptors and the effect of their ligands on
visible light) can induce melanogenesis in melasma by stimulating cutaneous cells were assessed in vitro (Table 2).
melanocytes directly or indirectly via secretion of melanogenic fac-
tors by keratinocytes,[16] fibroblasts,[15–18] endothelial cells[19] and
2 | E FFEC T O F S E X S TE RO I D H O R M O N E S
probably sebocytes. Chronic exposure to UVA1 and visible light
O N E PI D E R M A L PI G M E NTATI O N
seems to be implicated in dermal and basal membrane damage.[12]
On the other hand, sex hormones such as oestrogens, especially
2.1 | Direct effects of sex steroid hormones on
17β-­estradiol (E2) and progesterone, are factors involved in the reg-
melanocytes
ulation of pigmentation,[20] collagen content, skin ageing, skin mois-
ture and skin thickness.[21,22] Apart from FBS and phenol red which have a mild oestrogenic ef-
Estradiol, luteinizing hormone (LH) and follicle-­
s timulating fects (Table 3),[20,35,36] inter-­
individual variations which induce a
hormone (FSH) levels were found to be higher in the sera of specific response should be considered.[36,37] In women,
donor-­
women with melasma than in those of control women without estradiol and progesterone concentrations vary according to age,
melasma, whereas progesterone levels were similar in the two menstrual cycle, during pregnancy and at menopause (Table 4A, B).
groups.[23,24] In the sera of men with melasma, luteinizing hor- Men have lower levels of plasmatic estradiol and progesterone than
mone levels were higher than in those of controls without me- women (Table 4C).
lasma, whereas FSH levels were similar and testosterone levels
were lower.[25,26] In Indian males with melasma, no or only weak
2.1.1 | Effects of oestrogen on melanocytes
expression of oestrogen receptors, progesterone receptors and
stem cell factor was observed in lesional skin and no significant Exposure to 17β-­estradiol at concentrations found in males and non-­
changes in serum hormone levels were detected.[27] In a Japanese pregnant females (10−11 to 10−9 mol/L) induced an increase in mela-
male, oestrogen therapy for prostate cancer was incriminated in nin content in responsive melanocytes 3 days post-­treatment,[37,38]

TA B L E   1   Expression of oestrogen and progesterone receptors in melasma skin (M) as compared to perilesional (PL) or non-­lesional skin
(NL); nd: not detected; na: not analysed

Progesterone
Ethnic origin Technique Localization Oestrogen receptor receptor Ref.

American M/NL IHC Increase na [31]


Korean M/PL IHC Epidermis Slight Increase ERβ Increase [32]
Dermis: Increase ERβ = [66]
Around small blood vessels
Fibroblast-­like cells
(probably telocytes)
Brazilian M/NL IHC Epidermis Increase ERβ Increase [33]
Dermis = =
Transcriptomic Whole skin = =
Korean M/NL Transcriptomic Whole skin, Variable expression of ERα and ERβ na [34]
14 women 3 women: great Increase ERβ and ERα;
2 women: great Increase ERβ;
2 women: Decrease ERα;
3 women: Decrease ERβ
CARIO |
      711

TA B L E   2   Expression of oestrogen and progesterone receptors but that their expression in vitro depends on stimulation by exogenous
in cells factors.
Progesterone Activation of GPER and PAQR7 respectively stimulates or inhib-
Oestrogen receptors receptors its cAMP signalling, the same pathway used by α-­MSH to regulate

Cells ERα ERβ GPER PR PAQR7 Ref.


pigmentation, so they counteract each other.[20] In order to, oestro-
gen via fixation on ERα and in association with Sp1 can modulate the
Melanocytes yes yes yes [35,36]
level of expression of the progesterone receptor by binding to ERE/
no no yes no yes [20]
Sp1 site (oestrogen response element and sp1 sites)[45] and proges-
Keratinocytes yes yes [47]
terone can inhibit ER expression.[41]
Mast cells yes yes [81]
Working on prostate stromal cells, Smith et al hypothesized that
yes no [78]
high titres of estradiol saturate the ER, inducing in turn a decrease
Fibroblasts yes yes [62]
in ER synthesis.45,46 Thus, we hypothesize that, in vitro, physiologi-
Sebocytes yes yes [61]
cal doses of oestrogen[10,11] stimulate the production of melanin by
Endothelial yes Do yes [71]
melanocytes, but that the latter regulate transcription of their ER to
cells express
maintain their homeostasis. Since we have sometimes observed that
oestrogen has no effect, we suppose that the threshold of sensitivity
varies according to the strain of melanocytes, or that transcriptional
Telocytes yes yes [66,67]
regulation of ER is induced more or less quickly. At doses found
during pregnancy and with short-­term exposure, oestrogen induced
whereas it was decreased after 10 days.[35] Under stimulation with melanin production, whereas progesterone decreased it in vitro. On
25 nmol/L (2.5.10−8 mol/L) 17β-­estradiol, a concentration observed the other hand, no data were found on the effect on melanocytes of
during pregnancy[39] or 2.5.10−8 mol/L ethinyl estradiol, which is exposure to both oestrogen and progesterone in vitro.
often used in oral contraceptive pills, melanin synthesis was in- We hypothesize that in vivo and in basal conditions, an equilib-
creased in melanocytes cultured in monolayers.[20] Modifications rium between oestrogen and progesterone pathways maintains pig-
in melanin production were inversely correlated with melanocyte mentation at its basal level. However, during hormone replacement
proliferation.[35,37] Experiments conducted with several strains of therapy and pregnancy, levels of oestrogen and progesterone might
melanocytes showed that some melanocytes did not respond to be modified and the ratio between the two hormones might vary.
stimulation by oestrogen, even at a high dose (10−7M) [36] (Table 5). This would explain the onset of melasma in women, who are more
sensitive to sex steroid hormones or are less able to regulate the
level of their receptors.
2.1.2 | Effect of progesterone on melanocytes
Few authors have analysed the effects of progesterone on pigmen-
2.2 | Indirect effect of sex steroid hormones on
tation, and no data have been reported with progesterone concen-
pigmentation via keratinocytes
trations found in males and non-­pregnant females. Progesterone
at 5.10 −7M, a physiological concentration observed in the third
2.2.1 | Effects of oestrogen on melanocytes via
trimester of pregnancy,[39] decreased pigmentation in melanocytes
keratinocytes
cultured in monolayers[20] (Table 5). Female melanocytes derived
from iPS and male melanocytes from various body locations (facial In keratinocytes, E2 (10 −11-­10 −9M) induced ERα synthesis in a dose-­
vs foreskin) and of different age (adult vs. newborn) had a similar dependent manner, whereas E2 did not modify ERβ levels.[47] On
[20]
response to oestrogen and progesterone (n = 3 for each group). the other hand, oestrogen at concentrations found in pregnancy
upregulated ERα and ERβ (34). E2 induced proliferation of keratino-
cytes in vitro[47] via non-­genomic and genomic pathways. However,
2.1.3 | Expression of sex steroid hormone receptors
an increase in proliferation was observed at basal concentrations
in melanocytes
(10 −11-­10 −9M), whereas, according to authors and thus probably
Oestrogen and progesterone may act through several receptors. For the donors, the high concentrations found in pregnant women
example, adult and foreskin melanocytes did[35,36] or did not[20] express (10 −8-­10 -7M) have no effect on proliferation[48] or reach a plateau at
ER and PR at mRNA and protein levels. However, while Natale et al did 10 -8 mol/L.[49] Estradiol is inactivated by oestrogen sulfotransferase,
not find ER and PR, they found that melanocytes responded to oestro- which is more highly expressed in calcium-­differentiated keratino-
gen via fixation to GPER (G-­protein coupled oestrogen receptor) and to cytes than in proliferating keratinocytes, suggesting that the ac-
progesterone via fixation to PAQR7 (progestin and adipoQ receptor 7), tion of estradiol is suppressed in differentiated epidermal layers.[50]
[20]
two G protein-­coupled receptors. Since various factors such as TPA, Estradiol (10 -8 mol/L, concentration observed during pregnancy)
[40–44]
PMA, CT, insulin, IGF-­1 and EGF modulate ER and PR, we hypoth- also enhanced the production of GM-­C SF, a well-­known melano-
esize that melanocytes are able to express ER, GPER, PR and PAQR7 genic factor,[16] in keratinocytes via the PKC and ERK pathways.[51]
|
712       CARIO

TA B L E   3   Summary of melanocyte culture

Seeded melanocytes/cm² Type of Time of


Origin, age and phototype support Medium Supplements [Ho] Treatment incubation Ref.

Neonatal foreskin ECM coated EMEM 2% FCS 17β-­estradiol 10 -12 3/day 3 d [37]
2.10 4 or uncoated to 10 -9 mol/L + 1 time 4 h
Iscove 0.1% BSA,
age: 1-­4 months prior assay
medium 80 μg/mL soya bean
lecithin,
30 μg/mL transferrin
Neonatal foreskin uncoated EMEM 5% chelexed FCS 17β-­estradiol 10 -12 1 24 h [38]
1-­4.10 4 to 10 -9 mol/L
Passage 4 to 6
Korean Adult foreskin nd MCDB 4% chelexed FBS, Oestrogen 1/day 6 d [36]
5.3.10 4 153 8 nM TPA, or progesterone
phototype 0.6 ng/mL bFGF, 10 -9 mol/L and
III-­V 5 μg/mL insulin, 10 -7
1 g/mL tocopherol, 10 000
units/mL penicillin,
10 000 g/mL streptomycin
Foreskin nd M 254 0.2% BPE, 2.5.10 -­8 nd 4 d [20
Lightly pigmented 0.5% FBS, 17β–estradiol
2.6.106 1 μg/mL insulin-­like growth
factor-­I ,
Foreskin heavily pigmented 5.10 -7
5 μg/mL Bovine transferrin,
2.6.106 Progesterone
3 ng/mL bFGF,
0.18 μg/mL Hydrocortisone,
3 μg/m Heparin,
10 ng/m PMA,
10 000 units/mL penicillin,
10 000 g/mL streptomycin
Foreskin Vitronectin F12 Wo 20 ng/mL EGF, 17β–estradiol nd 4 and 10 d [35]
2.10 4 coated phenol 5 μg/mL insulin, 10 -12 and
red 40 ng/ml CT, 10 -9 mol/L
5 μg/mL transferrin,
1 μmol/L Hydrocortisone,
15 μg/mL Endothelial cell
growth supplement,
1.10 -7 mol/L retinol,
85 nmol/L TPA,
1% chelexed FCS

[Ho], hormone concentration; bFGF, basic fibroblast growth factor; BPE, bovine pituitary extract; BSA, bovine serum albumin; CT, cholera toxin; EGF,
epidermal growth factor; FBS, heat-inactivated fetal bovine serum; FCS, heat-inactivated fetal calf serum; Nd, not described; PMA, Phorbol 12-myristate
13-acetate; TPA, 12-O-tetradecanoyl phorbol 13-acetate; Wo, without.
Chelexed serum has no oestrogen.

In melasma lesional skin, PDZK1 (PDZ domain protein kidney 1), during pregnancy and luteal phase (10 -8-­10 -7M) did not have any ef-
[52]
an oestrogen-­induced factor, is overexpressed in keratinocytes fect on the proliferation of keratinocytes.[48] Until now, there is no
[34]
and melanocytes. In a melanocyte-­keratinocyte coculture, over- evidence indicating that progesterone stimulates the secretion of
expression of PDZK1 increased the oestrogen (10 -7-­10 -8M)-­induced melanogenic factors by keratinocytes.
tyrosinase expression, whereas knock-­down of PDZK1 reduced the Due to the expression of oestrogen sulfotransferase in differ-
oestrogen-­induced upregulation of tyrosinase. Oestrogen induces entiated keratinocytes, the effect of oestrogen is mainly observed
tyrosinase by way of MITF. This overexpression also stimulated mel- in the basal layer,[50] suggesting that oestrogen mainly affects the
anosome transfer via increased phosphorylation of ERM (ezrin/ra- epidermal melanin unit. In the epidermal melanin unit, the number of
dixin/moesin) and Rac1 (Ras-­related C3 botulinum toxin substrate 1) melanocytes and keratinocytes is tightly regulated,[53] so if oestro-
[34]
but not via PAR-­2. (Figure 1 A) gen induces proliferation of the basal keratinocytes, the number of
melanocytes increases. It is well known that keratinocytes secrete
exosomes and factors that regulate melanogenesis, differentiation
2.2.2 | Effects of progesterone on melanocytes via
and proliferation of melanocytes.[54,55] We hypothesize that oes-
keratinocytes
trogen (10 -7-­10 -8 mol/L) induces an increase in basal keratinocytes
An increased proliferation of keratinocytes was observed with basal which, in turn, secrete factors inducing proliferation of melanocytes,
concentrations of progesterone (10 -11-­10 -9M). Concentrations found tyrosinase activity and melanosome transfer leading to melasma.
CARIO |
      713

TA B L E   4   Concentration of estradiol and progesterone in females (A, B) [82–86] and males (C) [85–88]

(A)

Age (years) 0 to <6 6 to <11 11 to <15 15 to <19


-10 -10 -10
Estradiol (M) 0.73-­1.94 × 10 0.73-­2.16 × 10 0.73-­3.19 × 10 0.73-­
4.07 × 10 -10
Progesterone (M) 2.7 10 −10 -­4.7 × 10 −8

(B)

Menstrual cycle Weeks of pregnancy

Stage Follicular Ovulation Luteal <12 28 39 Menopause


−10 −9 −10 −9 −8 −8
Estradiol (M) 1.1-­3.6 × 10 0.47-­1.28 × 10 1.8-­6.6 × 10 ~8 ×10 ~4 × 10 ~8 × 10 <2.2 × 10 −10
−9 −9 −8 −8 −7 −7
Progesterone (M) 0.47-­2.2 × 10 ~8 × 10 0.6-­7.95 × 10 ~6 × 10 ~1.3 × 10 ~2.2 × 10 0.2-­5 × 10 −9

(C)

Age (years) 0 to <19 18-­3 0 20-­47 56-­71


-10 −10
Estradiol (M) 0.73-­1.46 × 10 ~2 × 10 ~1.4 × 10 −10
Progesterone (M) 2.1 × 10 −10 -­1.3 × 10 −8 ~6 × 10 −10

~, around

Nevertheless, the melanocyte/keratinocyte ratio may vary accord- collagen synthesis in culture fibroblasts from postmenopausal
ing to the patient's sensitivity or feedback control on the oestrogen women but had no effect on MMP-­2 and MMP-­9.[63] Oestrogen at
receptor. Progesterone does not seem to play a role in the modula- 10 -7 mol/L (pregnancy concentration) did not induce the prolifera-
tion of pigmentation by keratinocytes in melasma. tion of fibroblasts from women aged between 28 and 41 years or
the production of collagen, nor activate cyclin D1 and D3. However,
it induced morphological changes via reorganization of the actin
3 | E FFEC T S O F S E X S TE RO I D H O R M O N E S cytoskeleton and focal adhesion.[64] Contradictory results were
O N D E R M A L CO M PA RTM E NT A N D observed in another study conducted on dermal fibroblasts of un-
I N D I R EC T E FFEC T O N E PI D E R M A L determined age, since oestrogen (10 -8-­10 -7 mol/L), progesterone
PI G M E NTATI O N (10 -7 mol/L) and a combination of oestrogen (10 -8) and progesterone
(10 -7 mol/L) decreased the proliferation of fibroblasts and MMP 1
It is interesting to examine the influence of hormones in the der- and increased collagen I synthesis.[65]
mal compartment for two reasons. First, in melasma, the number of We did not find any evidence that the effects of oestrogen and
blood vessels and mast cells is greater in lesional skin than in perile- progesterone on fibroblasts modulate pigmentation, but they do
sional skin.[6] Second, dermal regulation of pigmentation has already modulate collagen synthesis. Thus, in melasma, oestrogen modula-
been demonstrated in vitro,[17,56,57] in vivo [18]
and in pigmentary tion of fibroblasts does not seem to be involved in pigmentation.
disorders such as senile lentigo[15] and vitiligo[58,59] and has been However, we hypothesize that high doses of oestrogen in some
[19]
hypothesized in melasma. Furthermore, in systemic scleroderma patients decrease the synthesis of collagen and thus facilitate the
(SSc) whose main skin feature is fibrosis, characteristic modifica- protrusion of melanocytes and the dermal deposition of melanin.[8]
tions of the fibroblast functional phenotype are thought to be due In melasma, fibroblast-­
like cells, located around small blood
to an interplay between damaged endothelial cells, immune cells vessels, expressed highly ERβ (Table 1).[32] These cells are probably
and their soluble mediators.[60] As in SSc, there might also be an in- telocytes (interstitial Cajal-­like cell (ICLC), CD34+stromal cells or
terplay in melasma. In addition, since most dermal cells possess sex CD34+ fibrocytes). Telocytes are localized in the papillary dermis
steroid hormone receptors (Table 2), dermal cells may influence epi- just beneath the basement membrane,[66] around blood vessels and
[16]
dermal pigmentation directly or indirectly via other dermal cells. surrounding sebaceous glands, expressed oestrogen and progester-
one receptors in vitro and seemed to play a role in angiogenesis[66,67]
and tissue homeostasis.[67] Indeed, they secrete extracellular vesi-
3.1 | Indirect effects of sex steroid hormones on
cles[36,68] which allow communication with neighbouring cells such
pigmentation via regulation of fibroblast secretions
as fibroblasts, immune cells nerve endings and endothelial cells.[69,70]
In dermis, fibroblasts express ERα and β at the mRNA and protein We may hypothesize that these cells participate in the increase in
level.[61,62] Oestrogen at 2-­5 10−9 (basal concentration) stimulated the number of vessels in melasma.
|
714       CARIO

TA B L E   5   Effects of estradiol and progesterone on melanocytes and melanogenesis

Tyrosine
Number of hydroxylase
Hormone cultures Concentration Proliferation activity Tyrosinase activity Melanin Ref.
−12 
17-­Estradiol 4 10 mol/L NE nd NE nd [38]
−11 
4 10 mol/L NE Slight increase
4 10 −10 and Decreasea Increase a
10 −9 mol/L
23 10 −12 to 13: Dose-­ nd 13: Dose-­ Increase (production [37]
10 −9 mol/L dependent dependent and extrusion)
decrease increase
8:NE 8:NE
nd 10 −12 and Dose-­dependent nd Dose-­dependent Dose-­dependent [35]
10 −9 mol/L increase at 10 d decrease at 4 d decrease at 10 d
3 0.5-­2.5. nd nd nd Dose-­dependent [20]
10−8 mol/L increase
Ethinyl 3 0.5-­2.5. nd nd nd Dose-­dependent [20]
estradiol 10 −8 mol/L increase
Estrone 4 10 −9 mol/L NE nd Increase NE [37]
−9 
Estriol 4 10 mol/L NE nd Increase NE [37]
Oestrogen 8 10 −9 mol/L 3: Increase, 4 (a) Increase nd nd [36]
1 (c): NE
1 (b): decrease, 4: NE
3:NE
8 10 −7 mol/L 3: Increase, 4 (a) Increase
1(c): decrease
4: NE 4: NE
Progesterone 8 10 −9 mol/L 3: Increase, 4 (a) increase nd nd [36]
1 (c): NE
1 (b): decrease, 3: 4: NE
NE
8 10 −7 mol/L 3: Increase, 4 (a) Increase
1 (c): NE
1: increase, 4:NE
1: decrease,
2: NE
3 10 −8-­ nd nd nd Dose-­dependent [20]
5.10 −7 mol/L decrease

nd, not determined; NE, no effect.


Description of results given only when results differed between strains.
(a): Represents a group of four melanocyte strains which behave similarly. (b), (c): Represent two special strains of melanocytes.
a
Similar value for 10 -10 and 10 -9 mol/L.

stimulates MITF phosphorylation, tyrosinase and TRP-­2 .[19] UVB-­


3.2 | Indirect effects of sex steroid hormones
or UVA-­irradiated HDMEC secretome stimulates melanogenesis
on pigmentation via regulation of endothelial
more strongly via an increase in SCF but not in ET-­1 , FGF-­2 and
cell secretions
POMC, which are well-­k nown melanogenic paracrine factors.[72]
Endothelial cells express ERα and GPER and should express Since UV did not induce ET-­1 in HDMEC and since ET-­1 is in-
ERβ.[71] Regazzetti et al observed an increase in pigmenta- creased in melasma lesional skin, [1] we believe that estradiol is
[19]
tion near to vessels in various pigmentary disorders and the involved in the increase in ET-­1 , although no study has yet been
number of vessels is increased in melasma, [6] so vessels may done on HDMEC and sex hormones. However, on other endothe-
be involved in hyperpigmentation in melasma. Human dermal lial cells, estradiol (2.10 −10 -­3 .7 10 −7M) did not induce any effect
microvascular endothelial cell (HDMEC) secretome promotes on endothelin-­1 (ET-­1) secretion, nor did it inhibit its secretion
melanogenesis via ET-­1 /EDNRB (endothelin receptor B), which but led to an increase in eNOS.[73,74] On the other hand, E2
CARIO |
      715

3.3 | Is regulation of mast cell metabolism by sex


steroid hormones involved in the modulation of
epidermal pigmentation?
Dermal mast cells secrete histamine, which can stimulate mela-
nin synthesis, proliferation and migration of melanocytes,[6] and
tryptase, which may be involved in dermal and basal membrane dam-
age.[6] Progesterone (10 -7M, concentration during pregnancy) inhib-
ited substance P-­induced release of histamine by rat mast cells.[76]
Thus, the absence of substance P in melasma may be related to pro-
gesterone.[77] E2-­induced release of LTC4 (leukotriene C4) is known
to be mitogenic for melanocytes in culture at concentrations ranging
between 10 -10 and 10 -9 mol/L; that is, basal concentrations below
the high concentrations observed during pregnancy.[78,79] Since the
main factors secreted by mast cells, histamine and tryptase, are not
released by hormone treatment but are efficiently released by UV
radiation, the involvement of mast cells in melasma may be due to
solar radiation rather than to sex steroid hormones.

3.4 | Effects of sex steroid hormones on


regulation of pigmentation in 3D model and explants
In 3D reconstructs which allow crosstalk between melanocytes
and keratinocytes, E2 at 2.5.10 -8M increased melanin levels but not
the number of melanocytes, whereas progesterone at 5.10−7 mol/L
decreased pigmentation but not the number of melanocytes[20]
(Figure 1B). 2.5.10−8 mol/L E2 and 5.10−7 mol/L progesterone are
F I G U R E   1   Schematic representation of effect of oestrogen values that occur during pregnancy,[39] so these two hormones have
and progesterone at concentration found during pregnancy an opposite effect regarding pigmentation in reconstructed epider-
on pigmentation in vitro and hypothesis of development of mis. Thus, we hypothesize that, in the absence of other stimulation
hyperpigmentation in melasma. Effect of sex steroid hormones such as UV irradiation and predisposing factors due to opposing
on melanocytes and keratinocytes cultured in monolayer and in
effect of these two molecules, pigmentation is maintained during
coculture (A) or in 3D (B). Oestrogen stimulated melanin production
directly via increase in tyrosinase and indirectly via secretion pregnancy.
of GM-­C SF by keratinocytes, whereas progesterone inhibited In skin explants, which is an even more complex model, 6-­day
melanin production. Oestrogen stimulated also melanin transfer. exposure to 10−9 mol/L and 10−7 mol/L oestrogen increased pigmen-
Effects of sex steroid hormones on explants before (C) and after tation in only 2/5 explants, whereas 10 -7M progesterone did not
irradiation (D). Oestrogen and progesterone induced no effect,
modify pigmentation.[36] Oestrogen and progesterone (10−9 mol/L
whereas oestrogen + progesterone+ UVB induced transfer of single
and 10−7 mol/L) did not induce any significant change, especially in
melanosome. Modelization of effect of oestrogen on initiation
of hyperpigmentation (E) and sustaining of hyperpigmentation the formation of rete ridges on skin explants.[36] Moreover, treatment
(F) in melasma. First exposure to high level of oestrogen and of Caucasoid skin explants with 10 -8 mol/L oestrogen or 10 -8 mol/L
UVB induced melanin production and transfer second oestrogen progesterone did not change the number of melanosomes or their
stimulated angiogenesis and thus ET-1 concentration is increased. transfer, whereas 50 mJ.cm−² UVB induced a slight increase in
Green arrow activation, red arrow inhibition
transfer. On the contrary, 10−8 mol/L oestrogen + 50 mJ.cm−² UVB
induced a dramatic increase in the number and transfer of single
induced via ERα an increase in VEGF, proliferation and migration melanosomes.[80] Treatment with 10 -8M progesterone + 50 mJ.cm-²
of endothelial cells, whereas via GPER triggered an increase in UVB was more efficient than UVB alone in increasing the transfer
angiogenesis.[71] Thus, E2 may be responsible for the increased of single melanosomes and seemed to be implicated in alteration
number, density and size of vessels in melasma and the effect on of the basal membrane,[80] which may facilitate the loss of melano-
pigmentation may just be due to the high number of vessels se- cytes and melanin in the dermis (free melanin or melanophages).[6]
creting endothelin 1 rather than to an increase in the level of ET-­1 No additive effect of 10 -8 mol/L progesterone on the number and
secreted by each type of endothelial cell. In addition, endothelial transfer of single melanosomes was observed in explants treated
cells may also secrete NO due to an increase in eNOS and thus with both hormones and UVB as compared to oestrogen + UVB[80]
75
stimulate melanogenesis. (Figure 1C,D). Thus, the change in the number and transfer of single
|
716       CARIO

melanosomes may be attributed to the synergistic effect of oes- might also induce local hyperoxia that would stimulate melano-
trogen and UVB. Even though only short-­term effects such as gene genesis, an effect already observed in melanocytes both in mono-
regulation and melanosome transfer may be observed, these models layers and in 3D cultures (Cario et al, unpublished data), and would
are useful since they show that primary changes such as morpho- sustain hyperpigmentation. If irradiation is chronic, this local dis-
logical changes in melanosomes occur after acute exposure to oes- turbance might also stimulate keratinocytes, melanocytes, mast
trogen and UV. Thus, long-­term exposure may maintain and amplify cells and fibroblasts, which could potentiate and maintain hyper-
changes. Experiments on explants, which are more complex than 3D pigmentation and modify the basal membrane.
models, showed that sex steroid hormones alone are not able to in- Altogether, the data suggest that sex steroid hormones in me-
duce melasma. lasma, especially oestrogen, are not able alone to induce hyperpig-
mentation but act in synergy with UVB. However, oestrogen may
sustain hyperpigmentation by increasing the number of blood ves-
4 |  CO N C LU S I O N sels and thus stimulating the secretion of endothelin-­1. Since the
sensitivity of cells to sex steroid hormones varies greatly, we hy-
While the cells in the monolayer have receptors and can respond pothesize that sex steroid hormones are responsible for the differ-
to sex steroid hormones, the effects are more difficult to decipher ence in susceptibility to melasma (Figure 1E,F).
in three-­dimensional models. While oestrogen increases melano-
genesis in the melanocyte monolayer and induces the production
C O N FL I C T O F I N T E R E S T
of melanogenic factors by keratinocytes, the same dose failed to
increase it in explants. Therefore, it is likely that explants are able The author declares no conflict of interest.
to maintain skin homeostasis during short-­term exposure due to
the presence of the cutaneous cells (melanocytes, keratinocytes,
AU T H O R C O N T R I B U T I O N
fibroblasts, endothelial cells). On the other hand, this is not pos-
sible when a second trigger like UV irradiation is applied. Since UV Muriel Cario analysed the data and wrote the paper.
irradiation alone induced only a slight increase in transfer in this
model as compared to oestrogen+ UVB, the explant model con-
ORCID
firms that melasma can occur only on photo-­exposed skin in pa-
tients subjected to high levels of oestrogen. Moreover, the number Muriel Cario  https://orcid.org/0000-0003-0462-5684
of mast cells and blood vessels in explants did not increase, so we
hypothesize that mast cells and endothelial cells do not contribute
to triggering lesions in melasma. In addition, these effects were REFERENCES

observed with UVB, suggesting that UVA is implicated by amplify- [1] A. C. Handel, L. D. B. Miot, H. A. Miot, An. Bras. Dermatol. 2014, 89,
ing and/or sustaining the modifications observed in melasma. 771.
[2] A. C. Handel, P. B. Lima, V. M. Tonolli, L. D. Miot, H. A. Miot, Br. J.
In vitro studies on monolayers have shown that the effect of
Dermatol. 2014, 171, 588.
sex hormones on cells, especially melanocytes and fibroblasts, [3] R. Pichardo, Q. Vallejos, S. R. Feldman, M. R. Schulz, A. Verma, S. A.
varies according to the concentrations used and to the donors. In Quandt, T. A. Arcury, Int. J. Dermatol. 2009, 48, 22.
vitro, it seems that high-­p hototype melanocytes are more sensitive [4] A. G. Martin, S. Leal-Khouri, Int. J. Dermatol. 1992, 31, 375.
[5] N. P. Sanchez, M. A. Pathak, S. Sato, T. B. Fitzpatrick, J. L. Sanchez ,
to progesterone, whereas low-­p hototype ones are more sensitive
M. C. Mihm, J. Am. Acad. Dermatol. 1981, 4, 698.
to oestrogen.[20] This might explain why melasma occurs earlier in [6] S.-H. Kwon, Y.-J. Hwang, S.-K. Lee, K.-C. Park, Int. J. Mol. Sci. 2016,
low phototypes than in higher ones. In 3D models (reconstructed 17. [cited 2016 Aug 8] https://doi.org/www.ncbi.nlm.nih.gov/pmc/
epidermis and explants), oestrogen seemed to be more involved articles/pmc4926358/.
in regulating pigmentation than progesterone, but the effect was [7] W. H. Kang, K. H. Yoon, E.-S. Lee, J. Kim, K.-B. Lee, H. Yim, S. Sohn,
S. Im, Br. J. Dermatol. 2002, 146, 228.
again donor-­d ependent, thereby confirming that genetic predis-
[8] B. Torres-Álvarez, I. G. Mesa-Garza, J. P. Castanedo-Cázares, C.
position is an important factor in the effect of hormones. Both UV Fuentes-Ahumada, C. Oros-Ovalle, J. Navarrete-Solis, B. Moncada,
irradiation alone and sex hormones alone are able to induce pig- Am. J. Dermatopathol. 2011, 33, 291.
mentation. However, one may hypothesize that UV irradiation and [9] H. Y. Kang, J.-P. Ortonne, Ann. Dermatol. 2010, 22, 373.
[10] P. E. Grimes, N. Yamada, J. Bhawan, Am. J. Dermatopathol. 2005, 27, 96.
sex hormones are necessary to maintain hyperpigmentation, es-
[11] K. Konrad, K. Wolff, Arch. Dermatol. 1973, 107, 853.
pecially oestrogen in predisposed patients. In women, oestrogen [12] T. Passeron, M. Picardo, Pigment Cell Melanoma Res 2017, 31, 461.
and progesterone levels vary during the menstrual cycle and preg- [13] R. Hernández-Barrera, B. Torres-Alvarez, J. P. Castanedo-Cazares,
nancy and their balance is modified.[39] Indeed, one may postulate C. Oros-Ovalle, B. Moncada, Clin. Exp. Dermatol. 2008, 33, 305.
[14] O. A. Ogbechie-Godec, N. Elbuluk, Dermatol Ther (Heidelb) 2017, 7,
that a local increase in estradiol in the papillary dermis induces
305.
the proliferation of endothelial cells. Such a local increase in blood [15] M. Salducci, N. André, M. Martin, R. Fitoussi, K. Vie, M. Cario-
vessels might modify local levels of oestrogen, progesterone, ET-­1 Andre, Pigment Cell Melanoma Res 2014, 27, 502.
and SCF. In addition, an increase in the number of endothelial cells [16] G. Imokawa, Pigment Cell Res. 2004, 17, 96.
CARIO |
      717

[17] S. J. Hedley, C. Layton, M. Heaton, K. H. Chakrabarty, R. A. Dawson, [48] R. Urano, K. Sakabe, K. Seiki, M. Ohkido, J. Dermatol. Sci. 1995, 9,
D. J. Gawkrodger, S. McNeil, Pigment Cell Res. 2002, 15, 49. 176.
[18] M. Cario-André, C. Pain, Y. Gauthier, V. Casoli, A. Taieb, Pigment Cell [49] N. Kanda, S. Watanabe, J Invest Dermatol 2004, 123, 319.
Res. 2006, 19, 434. [50] A. Kushida, K. Hattori, N. Yamaguchi, T. Kobayashi, Biol. Pharm. Bull.
[19] C. Regazzetti, G. M. De Donatis, H. H. Ghorbel, N. Cardot-Leccia, 2011, 34, 1147.
D. Ambrosetti, P. Bahadora, B. Chignon, J.-P. Lacour, R. Ballotti, A. [51] N. Kanda, S. Watanabe, J Invest Dermatol 2004, 123, 329.
Mahns, T. Passeron, J Invest Dermatol 2015. [cited 2015 Aug 31] [52] M. G. Ghosh, D. A. Thompson, R. J. Weigel, Cancer Res. 2000, 60,
https://doi.org/www.nature.com.gate2.inist.fr/jid/journal/vaop/ 6367.
naam/abs/jid2015332a.html [53] G. A. Scott, A. R. Haake, J Invest Dermatol 1991, 97, 776.
[20] C. A. Natale, E. K. Duperret , J.Zhang, R Sadeghi, A. Dahal, K. [54] A. Lo Cicero, C. Delevoye, F. Gilles-Marsens, D. Loew, F. Dingli, C.
T. O‘Brien, R. Cookson, J. D. Winkler, T. W. Ridky, eLife 2016, 5, Guéré, N. André, K. Vié, G. van Niel, G. Raposo, Nat. Commun. 2015,
e15104. 6, 7506.
[21] S. Verdier-Sévrain, F. Bonté, B. Gilchrest, Exp. Dermatol. 2006, 15, 83. [55] T. Hirobe, Pigment Cell Res. 2005, 18, 2.
[22] S. Verdier-Sévrain, Climacteric 2007, 10, 289. [56] T. Hirobe, K. Hasegawa, R. Furuya, R. Fujiwara, K. Sato, J. Dermatol.
[23] I. Hassan, I. Kaur, R. Sialy, R. J. Dash, J. Dermatol. 1998, 25, 510. Sci. 2013, 71, 45.
[24] K Mahmood, M. Nadeem, S. Aman, A. Hameed, A. H. Kazmi, [57] V. Casoli, M. Cario-André, P. Costet, C. Pain, A. Taïeb. Pigment Cell
Asianet-­Pakistan, 2012. [cited 2018 Apr 24] https://doi.org/trove. Res. 2004, 17, 87.
nla.gov.au/work/161673123 [58] D. Kovacs, E. Bastonini, M. Ottaviani, C. Cota, E. Migliano, M. L.
[25] R. Sialy, I. Hassan, I. Kaur, R. J. Dash, J. Dermatol. 2000, 27, 64. Dell'Anna, M. Picardo, J Invest Dermatol 2018, 138, 394.
[26] R. Sarkar, P. Puri, R. K. Jain, A. Singh, A. Desai, J. Eur. Acad. Dermatol. [59] D. Kovacs, G. Cardinali, N. Aspite, C. Cota, F. Luzi, B. Bellei, S.
Venereol. 2010, 24, 768. Briganti, A. Amantea, M. R. Torrisi, M. Picardo, Br. J. Dermatol. 2010,
[27] S. Handa, D. De, G. Khullar, B. D. Radotra, N. Sachdeva, Clin. Exp. 163, 1020.
Dermatol. 2018, 43, 36. [60] C. Chizzolini, N. C. Brembilla, E. Montanari, M. E. Truchetet,

[28] A. Ogita, Y. Funasaka, S. Ansai, S. Kawana, H. Saeki, Ann. Dermatol. Autoimmun. Rev. 2011, 10, 276.
2015, 27, 763. [61] E. Makrantonaki, J. Adjaye, R. Herwig, T. C. Brink, D. Groth, C.
[29] Z. Weihua, S. Makela, L. C. Andersson, S. Salmi, S. Saji, J. I. Webster, Hultschig, H. Lehrach, C. C. Zouboulis, Aging Cell 2006, 5, 331.
E. V. Jensen, S. Nilsson, M. Warner, J. A. Gustafsson, Proc. Natl [62] J. Haczynski, R. Tarkowski, K. Jarzabek, M. Slomczynska, S.
Acad. Sci. USA 2001, 98, 6330. Wolczynski, D. A. Magoffin, J. Jakowicki, A. Jakimiuk, Int. J. Mol.
[30] M. J. Thornton, A. H. Taylor, K. Mulligan, F. Al-Azzawi, C. C. Lyon, J. Med. 2002, [cited 2018 Apr 10]. http://www.spandidos-publica-
O'Driscoll, A. G. Messenger, Exp. Dermatol. 2003, 12, 181. tions.com/10.3892/ijmm.10.2.149
[31] R. Lieberman, L. Moy, J. Drugs Dermatol. 2008, 7, 463. [63] A. Surazynski, K. Jarzabek, J. Haczynski, P. Laudanski, J. Palka, S.
[32] Y. H. Jang, J. Y. Lee, H. Y. Kang, E. S. Lee, Y. C. Kim, J. Eur. Acad. Wolczynski, Int. J. Mol. Med. 2003, 12, 803.
Dermatol. Venereol. 2010, 24, 1312. [64] J. Carnesecchi, M. Malbouyres, R. de Mets, M. Balland, G. Beauchef,
[33] A. Ade Tamega, H. A. Miot, N. P. Moço, M. G. Silva, M. E. Marques, K. Vié, C. Chamot, C. Lionnet, F. Ruggiero, J.-M. Vanacker, PLoS ONE
L. D. Miot, Int. J. Cosmet. Sci. 2015, 37, 222. 2015, 10, e0120672.
[34] N.-H. Kim, K. A. Cheong, T. R. Lee, A. Y. Lee, J Invest Dermatol 2012, [65] N. Philips, S. Auler, R. Hugo, S. Gonzalez, Enzyme Research 2011,
132, 2622. 2011, e427285.
[35] S. H. Jee, S. Y. Lee, H. C. Chiu, C. C. Chang, T. J. Chen, Biochem. [66] Y. Kang, Z. Zhu, Y. Zheng, W. Wan, C. G. Manole, Q. Zheng, J. Cell
Biophys. Res. Commun. 1994, 199, 1407. Mol. Med. 2015, 19, 2530.
[36] S. Im, E. S. Lee, W. Kim, W. On, J. Kim, M. Lee, W. H. Kang, J. Korean [67] L. Chaitow, Journal of Bodywork and Movement Therapies 2017, 21, 231.
Med. Sci. 2002, 17, 58. [68] D. Cretoiu, M. Gherghiceanu, E. Hummel, H. Zimmermann, O.

[37] S. D. McLeod, M. Ranson, R. S. Mason, J. Steroid Biochem. Mol. Biol. Simionescu, L. E. Popescu, J. Cell Mol. Med. 2015, 19, 714.
1994, 49, 9. [69] J. Yang, Y. Li, F. Xue, W. Liu, S. Zhang, Am. J. Transl. Res. 2017, 9,
[38] M. Ranson, S. Posen, R. S. Mason, J Invest Dermatol 1988, 91, 593. 5375.
[39] M. Abbassi-Ghanavati, L. G. Greer, F. G. Cunningham, Obstet. [70] Y. Bei, F. Wang, C. Yang, J. Xiao, J. Cell Mol. Med. 2015, 19, 1441.
Gynecol. 2009, 114, 1326. [71] A. Trenti, S. Tedesco, C. Boscaro, L. Trevisi, C. Bolego, A. Cignarella,
[40] A. González-Arenas, M. Á. Peña-Ortiz, V. Hansberg-Pastor, B.
Int. J. Mol. Sci. 2018, 19, 000.
Marquina-Sanchez, N. Baranda-Avila, K. Nava-Castro, A. Cabrera- [72] M. Kim, T. Shibata, S. Kwon, T. J. Park, H. Y. Kang, Sci. Rep. 2018, 8,
Wrooman, J. Gonzalez-Jorge, I. Camacho-Arrroyo, .Endocrinology 4235.
2015, 156, 1010. [73] L. J. Pearson, T. G. Yandle, M. G. Nicholls, J. J. Evans, Peptides 2008,
[41] J. J. Pinzone, H. Stevenson, J. S. Strobl, P. E. Berg, Mol. Cell. Biol. 29, 1057.
2004, 24, 4605. [74] C. S. Wingrove, J. C. Stevenson, Eur. J. Endocrinol. 1997, 137, 205.
[42] M. B. Martin, P. Garcia-Morales, A. Stoica, H. B. Solomon, M. [75] H.-Y. Jo, C.-K. Kim, I.-B. Suh, S.-W. Ruy, K.-S. Ha, Y.-G. Kwon, Y.-M
PierceD Katz, S. Zhang, M. Danielsen, M. Saceda, J. Biol. Chem. Kim, J. Dermatol. 2009, 36, 10.
1995, 270, 25244. [76] M. Vasiadi, D. Kempuraj, W. Boucher, D. Kalogeromitros, T. C.

[43] R. Heimann, R. H. Rice, M. K. Gross, E. L. Coe, J. Cell. Physiol. 1985, 123, Theoharides, Int J Immunopathol Pharmacol 2006, 19, 787.
197. [77] H. Bak, H. J. Lee, S.-E. Chang, J.-H. Choi, M.-N. Kim, B.-J. Kim,
[44] M. Cutolo, B. Villaggio, A. Bisso, A. Sulli, D. Coviello, J. M. Dayer, Dermatol. Surg. 2009, 35, 1244.
Eur. Cytokine Netw. 2001, 12, 368. [78] M. Zaitsu, S.-I. Narita, K. C. Lambert, J. J. Grady, D. M. Estes, E.
[45] L. N. Petz, Y. S. Ziegler, J. R. Schultz, H. Kim, J. K. Kemper, A. M. M. Curran, E. G. Brooks, C. S. Watson, R. M. Goldblum, T. Midoro-
Nardulli, J. Steroid Biochem. Mol. Biol. 2004, 88, 113. Horiuti, Mol. Immunol. 2007, 44, 1977.
[46] P. Smith, N. P. Rhodes, Y. Ke, C. S. Foster, Prostate Cancer Prostatic [79] J. G. Morelli, S. S. Hake, R. C. Murphy, D. A. Noris. J Invest Dermatol
Dis 2002, 5, 105. 1992, 98, 55.
[47] S. Verdier-Sevrain, M. Yaar, J. Cantatore, A. Traish, B. A. Gilchrest, [80] Y. Gauthier, M. Cario, C. Pain, S. Lepreux, L. Benzekri, A. Taieb, Br. J.
FASEB J 2004, 18, 1252. Dermatol. 2018. In press
|
718       CARIO

[81] O. Zierau, A. C. Zenclussen, F. Jensen, Front Immunol. 2012, 3. [87] K. Y. Ho, W. S. Evans, R. M. Blizzard, J. D. Veldhuis, G. R. Merriam,
[cited 2018 May 5]. https://www.ncbi.nlm.nih.gov/pmc/articles/ E. Samojlik, R. Furlanetto, A. D. Rogol, D. L. Kaiser, M. O. Thorner, J.
PMC3377947/ Clin. Endocrinol. Metab. 1987, 64, 51.
[82] S. F. Pang, P. L. Tang, G. W. Tang, A. W. Yam, K. W. Ng, J. Pineal Res. [88] C. A. Strott, T. Yoshimi, M. B. Lipsett, J Clin Invest 1969, 48, 930.
1987, 4, 21.
[83] S. N. Raheem, M. Atoum, H. Al-Hourani, M. Rasheed, N. Nimer, T.
Almuhrib, Neoplasma 2010, 57, 74.
How to cite this article: Cario M. How hormones may
[84] A. Zlotnik, B. F. Gruenbaum, B. Mohar, R. Kuts, S. E. Gruenbaum,
modulate human skin pigmentation in melasma: an in vitro
S. Ohayon, M. Boyko, Y. Klin, E. Sheiner, G. Shaked, Y. Shapira, V. I.
Teichberg, Biol. Reprod. 2011, 84, 581. perspective. Exp Dermatol. 2019;28:709‐718. https://doi.
[85] O. P. Soldin, E. G. Hoffman, M. A. Waring, S. J. Soldin. Clin. Chim. org/10.1111/exd.13915
Acta 2005, 355, 205.
[86] M. W. Elmlinger, W. Kühnel, M. B. Ranke, Clin. Chem. Lab. Med.
2002, 40, 1151.

You might also like