You are on page 1of 11

ORIGINAL ARTICLE

The Major Orphan Forms of Ichthyosis Are


Characterized by Systemic T-Cell Activation
and Th-17/Tc-17/Th-22/Tc-22 Polarization
in Blood
Tali Czarnowicki1,2, Helen He1, Alexandra Leonard1, Kunal Malik1,3, Shai Magidi4, Stephanie Rangel5,
Krishna Patel5, Kara Ramsey5, Morgan Murphrey5, Teresa Song1, Yeriel Estrada1, Hue-Chi Wen1,
James G. Krueger2, Emma Guttman-Yassky1,2,6 and Amy S. Paller5,6

The ichthyoses are rare skin disorders with immune and barrier aberrations. Identifying blood phenotypes may
advance targeted therapeutics. We aimed to compare frequencies of skin homing/cutaneous lymphocyte antigen (þ)
versus systemic/cutaneous lymphocyte antigen (e) “polar” CD4þ/CD8þ and activated T-cell subsets in ichthyosis
versus atopic dermatitis, psoriasis, and control blood, with appropriate clinical correlations. Flow cytometry was used
to measure IFN-g, IL-13, IL-9, IL-17, and IL-22 cytokines in CD4þ/CD8þ T cells, with inducible co-stimulator molecule
and HLA-DR defining mid- and long-term T-cell activation, respectively. We compared peripheral blood from 47
patients with ichthyosis (congenital ichthyosiform erythroderma, lamellar ichthyosis, epidermolytic ichthyosis, and
Netherton syndrome) with 43 patients with atopic dermatitis and 24 patients with psoriasis and 59 age-matched
controls. Clinical measures included the ichthyosis severity score, with subsets for erythema and scaling, trans-
epidermal water loss, and pruritus. All ichthyoses had excessive inducible co-stimulator molecule activation
(P < 0.001), particularly epidermolytic ichthyosis. Significantly elevated IL-17- (P < 0.05) and IL-22-producing (P < 0.01)
T cells characterized ichthyoses, mainly Netherton syndrome and congenital ichthyosiform erythroderma (P < 0.05).
Increased T helper 2/cytotoxic T cell 2/T helper 9 (P < 0.05) and similar IFN-g frequencies (P > 0.1) versus controls were
also noted. IL-17/IL-22-producing cells clustered with clinical measures, whereas IFN-g clustered with age. Our data
show peripheral blood IL-17/IL-22 activation across the ichthyoses, correlating with clinical measures. Targeted
therapies should dissect the relative contribution of polar cytokines to disease pathogenesis.
Journal of Investigative Dermatology (2018) 138, 2157e2167; doi:10.1016/j.jid.2018.03.1523

INTRODUCTION defective epidermal barrier (Kawashima et al., 2005) with


The ichthyoses encompass a spectrum of syndromic and scaling and variable cutaneous erythema. Patients suffer from
nonsyndromic dermatoses with varying underlying genetic reduced quality of life and recurrent infections, posing an
bases (Oji et al., 2010). Their shared features include a economic burden on health care (Dreyfus et al., 2015).
The best-characterized and most common phenotype is
1
Department of Dermatology and the Immunology Institute, Icahn School ichthyosis vulgaris, owing to FLG (encoding filaggrin) gene
of Medicine at Mount Sinai, New York, New York, USA; 2Laboratory for
mutations (Smith et al., 2006). The more prevalent rare forms
Investigative Dermatology, The Rockefeller University, New York, New
York, USA; 3College of Medicine, SUNY Downstate, Brooklyn, New York, of ichthyosis are lamellar ichthyosis (LI; usually TGM1,
USA; 4Department of Pharmacological Sciences, Mount Sinai Center for encoding transglutaminase 1) and congenital ichthyosiform
Bioinformatics, BD2K-LINCS Data Coordination and Integration Center, erythroderma (CIE; multiple causative genes) (Takeichi and
Icahn School of Medicine at Mount Sinai School, New York, New York,
USA; and 5Departments of Dermatology and Pediatrics, Northwestern
Akiyama, 2016), collectively known as autosomal recessive
University Feinberg School of Medicine, Illinois, USA congenital ichthyosis (ARCI; Oji et al., 2010), keratinopathic
6
These authors contributed equally to this work. ichthyosis, epidermolytic ichthyosis (EI), involving mutations
Correspondence: Amy S. Paller, Department of Dermatology, Northwestern in KRT1 and KRT10 (Vahlquist et al., 2017), and Netherton
University Feinberg School of Medicine, 676 N. St. Clair, Suite 1600, syndrome (NS), with its atopic manifestations and underlying
Chicago, Illinois 60611-2997, USA. E-mail: apaller@northwestern.edu deficiency of a serine peptidase inhibitor, Kazal type 5
Abbreviations: AD, atopic dermatitis; ARCI, autosomal recessive congenital (SPINK5 mutations) (Sarri et al., 2017).
ichthyosis; CIE, congenital ichthyosiform erythroderma; CLA, cutaneous Recently, the cutaneous ichthyosis phenotype of these
lymphocyte antigen; EI, epidermolytic ichthyosis; IASI, ichthyosis area
severity index; IASI-E, ichthyosis area severity index-erythema; IASI-S, ich- orphan ichthyoses was linked to robust IL-17/tumor necrosis
thyosis area severity index-scaling; ICOS, inducible co-stimulator molecule; factor-a cytokine activation, which is highly associated with
LI, lamellar ichthyosis; NS, Netherton syndrome; Tc, cytotoxic T cell; Tcm, disease severity and transepidermal water loss (TEWL), the
central memory T cell; Tem, effector memory T cell; TEWL, transepidermal
water loss; Th, T helper; Treg, T-regulatory cell
functional barrier measure (Paller et al., 2017b). These data
are supported by studies on a limited number of patients
Received 5 February 2018; revised 15 March 2018; accepted 23 March 2018;
accepted manuscript published online 14 April 2018; corrected proof with restricted panels, mostly in NS (Briot et al., 2009; Fontao
published online 7 June 2018 et al., 2011), showing a pro-Th2/TARC/CCL17/TSLP signature

ª 2018 The Authors. Published by Elsevier, Inc. on behalf of the Society for Investigative Dermatology. www.jidonline.org 2157
T Czarnowicki et al.
Th17/Tc17/Th22/Tc22-Skewing in Ichthyosis Blood

(Akagi et al., 2013; Briot et al., 2009) with innate (IL-1b, between ichthyosis overall and healthy controls, patients with
tumor necrosis factor-a) and T-cell activation (IL-2) cytokine psoriasis and AD, and subsequently evaluated differences
increases (Renner et al., 2009). A previous study in nine among the various ichthyosis subtypes (ARCI-LI, ARCI-CIE,
children with NS using flow cytometry found normal EI, and NS).
numbers of T, B, and T-regulatory cells (Tregs), with small To enhance representation of the distribution of values on
increases in natural killer cells in affected individuals (Renner the significance of results, comparison plots incorporate both
et al., 2009). Another study on 10 children and one adult means (black) and medians (red)  standard error, with
with NS showed altered B-cell expression and decreased respective P-values. Although results discuss mean values,
natural killer cytotoxic activity (Hannula-Jouppi et al., 2016). both are detailed in Supplementary Tables S1 and S2 online.
Comprehensive blood phenotyping of a larger number of
Ichthyosis is characterized by increased mid/ICOS and late/
these pediatric and adult patients with ichthyosis is not
HLA-DR T-cell activation
available.
Tcm and Tem cells, key components of adaptive immunity,
There remains a huge unmet need for safer, more effective
are characterized by diverse homing capacities (Sallusto
treatments for ichthyoses. Current treatments are empirical,
et al., 2004). Both express the skin homing marker CLA,
often ineffective, and largely directed at reducing scale.
but Tcm are also CCR7þ, allowing them to migrate into
These include topical moisturizers, keratolytics, retinoids,
lymph nodes, creating an immunologic reserve (Sallusto
vitamin D analogs, corticosteroids, and calcineurin in-
et al., 1999). Ichthyosis showed the highest skin-homing
hibitors, all off-label. Systemic retinoids have variable effi-
CD4þTem frequencies (normal: 20%, psoriasis: 21%, AD:
cacy and multiple potential side effects (DiGiovanna and
23%, ichthyosis: 28%; P < 0.01; Figure 1a and b) and higher
Robinson-Bostom, 2003). Experimental gene therapies are
CLAþCD8þ Tem (12% vs. 9%, P ¼ 0.05; Figure 1c) and Tcm
early in preclinical development (Gorell et al., 2014).
(13.5% vs. 9.8%, P ¼ 0.03; Figure 1d) cells than controls
Although cutaneous and systemic immune inflammation has
(Supplementary Table S1).
been successfully targeted in atopic dermatitis (AD) and
T-cell activation via the T-cell receptor leads to sequential
psoriasis (Czarnowicki et al., 2015c; Guttman-Yassky et al.,
expression of T-cell activation markers. ICOS, a mid-
2017), little attention has been paid to targeting inflammation
activation marker, is upregulated within 24 hours (Tafuri
in ichthyosis. Indeed, specific immune-targeting therapeutics
et al., 2001), whereas HLA-DR indicates chronic T-cell acti-
have shown efficacy for NS (anti-tumor necrosis factor-a)
vation (Ferenczi et al., 2000). Ichthyosis showed increased
(Fontao et al., 2011; Paller et al., 2017a) and ichthyotic dis-
pan activation of ICOS and HLA-DR in both the skin-homing/
order resulting from mutations in DSP, encoding desmopla-
CLAþ and systemic/CLAe compartments, particularly among
kin (ustekinumab) (Paller et al., 2017a).
CD4þ populations (Figure 1eel). Most striking was ICOS
Cutaneous lymphocyte antigen (CLAþ) memory T cells are
activation, in both CD4þ and CD8þ subsets (CD4þ Tem:
peripheral biomarkers in inflammatory skin disorders
CLAþ: ichthyosis 43.5% vs. normal 20.5%, P < 0.0001;
(Czarnowicki et al., 2017b). We evaluated activated CD4þ/
CLAe: ichthyosis 18.5% vs. normal 11.7%, P < 0.001
CD8þ T cells and their differentiation to IFN-g-, IL-13-, IL-22-,
[Figure 1eef]; and CD8þ Tcm: CLAþ: ichthyosis 29.8% vs.
IL-17A-, and IL-9-producing T cells within skin homing/CLAþ
normal 15.6%, P < 0.0001; CLAe: ichthyosis 6.3% vs.
and systemic/CLAe compartments in blood of patients with
normal 4.2%, P ¼ 0.001 [Supplementary Figure S1a, b, e,
ichthyosis, compared to patients with AD and psoriasis
and f online]). These frequencies were even higher than in
and healthy controls. Blood of patients with ichthyosis had
AD, a highly inflammatory skin disease with excessive T-cell
increases in T-cell activation, and in IL-22-, IL-17A-, IL-13-,
activation (Czarnowicki et al., 2015c). Conversely, HLA-DR
and IL-9-producing T cells. IL-17/IL-22 axes were linked with
demonstrated differential expression, with increases
disease severity, whereas the IFN-g pathway was associated
restricted to CD4þ subsets (Tem: CLAþ: ichthyosis 10% vs.
with disease chronicity.
normal 5.5%, P ¼ 0.004; CLAe: ichthyosis 4.2% vs. normal
2.2%, P ¼ 0.05; Tcm: CLAþ: ichthyosis 8.8% vs. normal
RESULTS
4.6%, P ¼ 0.004; CLAe: ichthyosis 4% vs. normal 4%,
Flow cytometry was used to measure frequencies of IFN-g-,
P ¼ 0.03 [Figure 1g, h, k, and l]), but significantly lower
IL-9-, IL-13-, IL-17-, and IL-22-producing T cells, defining
frequencies in CD8þ Tem/Tcm/CLAþ/CLAe cells (P < 0.02;
T helper (Th) 1/cytotoxic T cell (Tc)1, Th9/Tc9, Th2/Tc2,
Supplementary Figure S1c, d, g and h, Supplementary
Th17/Tc17, and Th22/Tc22 subsets in CD4þ/CD8þ T cells,
Table S1).
respectively. Cell surface staining was used to measure
expression of mid (inducible co-stimulator molecule [ICOS]) Prominent Th17/Th22 signatures characterize ichthyoses
and late (HLA-DR) activation markers in central (Tcm/ T-cell activation is followed by T-cell subset differentiation.
CCR7þCD45ROþ) and effector (Tem/CCR7eCD45ROþ) Thus, we next evaluated the frequencies of polar T-cell sub-
memory T cells in skin homing/CLAþ and systemic/CLAe sets, including IFN-g-, IL-9-, IL-13-, IL-17-, and IL-22-
subsets. To assess immune polarity, samples were analyzed in producing CD4þ/CD8þ T cells, in both the cutaneous/CLAþ
parallel with blood from healthy controls and previously and systemic/CLAe compartments.
published cohorts of moderate-to-severe AD and psoriasis Congruent with our recent skin findings (Paller et al.,
(Czarnowicki et al., 2015b, 2015c), representing different 2017b), IL-17 was increased in ichthyosis versus control
immune polarizations (Th2/Th22 and Th1/Th17, respec- blood, particularly among skin-homing CD4þ T cells (CLAþ:
tively). To understand the common immune skewing in ich- 4.5% vs. 2.4%, P ¼ 0.004; Figure 2a), demonstrating levels
thyosis, we first compared frequencies of T-cell populations either similar to or higher than in psoriasis (CD8þ/CLAþ/CLAe:

2158 Journal of Investigative Dermatology (2018), Volume 138


T Czarnowicki et al.
Th17/Tc17/Th22/Tc22-Skewing in Ichthyosis Blood

a + +
CLA CD4 Tem cells b CLA+CD4+ Tcm cells c + +
CLA CD8 Tem cells d CLA+CD8+ Tcm cells
* + 6 *
100 * *
+ +
* +
*** **
*** 30 40 **

75
4
Frequency (%)

30
20
50
20
2
25 10
10

0 0 0

Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis

e ICOS+CLA+CD4+ Tem cells


f ICOS+CLA–CD4+ Tem cells
g HLA-DR+CLA+CD4+ Tem cells
h HLA-DR+CLA–CD4+ Tem cells
*** *** * **
*** *** *
* ** *** 40 *
*** * *** * ** +
*** *** 60 *
100 *** 60 *** **
*** *** ***
*** ** ***
*** ***
30
Frequency (%)

75
40
40
20
50

20
20 10
25

0 0 0
0
Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis

i ICOS+CLA+CD4+ Tcm cells


j k HLA-DR+CLA+CD4+ Tcm cells
l
ICOS+CLA–CD4+ Tcm cells + – +
HLA-DR CLA CD4 Tcm cells
*** *** *** ***
*** + *** *** ***
*** * 40 * ***
*** *** ** *** 30 ***
*** *** 60 *** **
90 *** * **
* * *
* *** ** ***
** *** *** ***
** **
** 30
Frequency (%)

20
60 40
20

30 20 10
10

0 0 0 0

Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis

Figure 1. T-cell memory subset activation. (aed) CLA CD4 /CD8 Tcm (CD45RO CCR7 ), Tem (CD45RO CCR7 ) subset frequencies, and (eel) ICOSþ and
þ þ þ þ þ þ e

HLA-DRþ activation in CLAþ/e CD4þ Tcm/Tem cells in control (red), psoriasis (green), AD (purple), and ichthyosis (blue). Bar plots represent means (black)/
medians (red)  SEMs. P-values are designated as ***<0.001, **<0.01, *<0.05, þ<0.1. AD, atopic dermatitis; CLA, cutaneous lymphocyte antigen; ICOS,
inducible co-stimulator molecule; SEM, standard error of the mean/median; Tcm, central memory T cell; Tem, effector memory T cell.

P < 0.01; Figure 2bed), a prototypical Th17/IL-17 disease Figure 3aed). IL-9-producing cells, recently associated with
(Raychaudhuri, 2013). Even more prominent were the psoriasis (Ruiz-Romeu et al., 2017; Singh et al., 2013) and
increased frequencies of IL-22-producing CD4þ (CLAþ: 7% vs. AD (Ma et al., 2014), were also upregulated in ichthyosis,
4%, P ¼ 0.006; CLAe: 3.4% vs. 1.65%, P ¼ 0.001; Figure 2e specifically in CD4þ (CLAþ: 6.9% vs. 3.7%, P ¼ 0.01; CLAe:
and f) and CD8þ (CLAþ: 6.6% vs. 2%, P ¼ 0.004; CLAe: 1.5% 0.4% vs. 0.2%, P ¼ 0.02; Figure 3e and f), but not CD8þ T cells
vs. 0.5%, P ¼ 0.001; Figure 2g and h) T cells in ichthyosis (P > 0.1; Figure 3g and h). Finally, IFN-g was largely similar
versus controls, seen consistently across the cutaneous and between ichthyoses and control (P > 0.1; Figure 3iel).
systemic compartments. IL-22 levels in ichthyosis were similar
or higher (CD8þ/CLAe: 1.5% vs. 0.6%, P ¼ 0.004; Figure 2h) Epidermolytic ichthyosis/EI shows the highest ICOS and
than in AD, an inflammatory disease with characteristic IL-22 HLA-DR expression
skewing in blood and skin (Czarnowicki et al., 2015b; Gittler To detect whether inflammatory profiles of ichthyosis sub-
et al., 2012). Intriguingly, similar to AD (Czarnowicki et al., types vary, we next compared disease subcategories and
2015b), IL-13-producing CD4þ/CD8þ cells (besides CD4þ/ controls. Although all subtypes had increased Tem ICOS
CLAþ cells, P > 0.1) were increased in ichthyosis to levels activation, EI showed highest expressions in both CLAþ
much higher than in psoriasis across all subsets (P < 0.01; (55.3% vs. 20.5%, P ¼ 0.0001; Figure 4a) and CLAe (29%

www.jidonline.org 2159
T Czarnowicki et al.
Th17/Tc17/Th22/Tc22-Skewing in Ichthyosis Blood

CD4+ T cells CD8+ T cells


a IL-17+CLA+ b IL-17+CLA– c IL-17+CLA+ d +
IL-17 CLA

30 + + 40 **
** 10.0 *** +
* ** **
** * +
**
* 6
* 30
7.5
20
Frequency (%)

Frequency (%)
4
20
5.0

10
10 2
2.5

0 0.0 0 0

Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis

e IL-22+CLA+
f IL-22+CLA–
g +
IL-22 CLA
+ h +
IL-22 CLA

**
**
*
** + ***
60 + +
***
*** * ** **
*** ***
*** *** *
*** 15 **
75 ** *
*** *** *** 10
*** ***
Frequency (%)

Frequency (%)
40
10 50

5
20
5 25

0 0 0 0

Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis
D D þ þ þ/e þ þ
Figure 2. IL-17 /IL-22 cytokine frequencies. (aed) IL-17 and (eeh) IL-22 frequencies in CLA , CD4 /CD8 T cells in control (red), psoriasis (green), AD
(purple), and ichthyosis (blue). Bar plots represent means (black)/medians (red)  SEMs. P-values are designated as ***<0.001, **<0.01, *<0.05, þ<0.1. AD,
atopic dermatitis; CLA, cutaneous lymphocyte antigen; SEM, standard error of the mean/median.

vs. 11.7%, P ¼ 0.01; Figure 4b) CD4þ, but not CD8þ Tem Th17/Tc17 and Th22/Tc22 cells cluster with ichthyosis
populations (Supplementary Table S2; Supplementary clinical measures
Figure S2a and b online). HLA-DR Tem activation was We also assessed how clinical characteristics, including
largely similar among ichthyoses (Figure 4c and d; ichthyosis severity (total ichthyosis area severity index [IASI],
Supplementary Figure S2c and d). EI also showed the with subsets for erythema [IASI-E] and scaling [IASI-S]),
highest Tcm ICOS (CLAþ: 37% vs. 14.7%, P ¼ 0.008; CLAe: pruritus, and TEWL, a functional barrier measure, relate to
15.6% vs. 7.2%, P ¼ 0.02) and HLA-DR activation, polar T-cell subsets. Unsupervised hierarchical clustering of
restricted to CD4þ subsets (Figure 4eeh; Supplementary all parameters was performed using Pearson correlations, as
Figure S2eeh). shown in the correlation heatmap and dendrogram in
Figure 6 (red: positive; blue: negative correlations, stars and
plus signs: significant correlations). Selected individual cor-
NS and CIE demonstrate the highest IL-17/L-22 frequencies relation scatter plots are presented in Supplementary
NS showed elevated IL-17 and IL-22 frequencies in CLAþ or Figures S4 and S5 online. As shown in Figure 6, a tight
CLAe, CD4þ or CD8þ cells (P < 0.05 for all), not just versus cluster gathering of TEWL, IASI-S, IASI, IASI-E, and IL-17-
controls but also compared with other ichthyosis subtypes producing cells (purple square) was adjacent to a Th22/
except CIE (P > 0.1) (Figure 5, Supplementary Table S2). Tc22 cluster (green square). Clinical severity and TEWL
Contrary to the Th2 fingerprint reported in past NS serum measures (red ellipses) branched around IL-17/IL-22 axes,
studies (Akagi et al., 2013; Briot et al., 2009), our results whereas pruritus clustered with IL-13-producing cells (yellow
showed comparable IL-13 frequencies in NS and controls square). IFN-g clustered with age (blue square). Correlations
(Supplementary Figure S3aed online). In contrast, CIE and LI are listed in Supplementary Table S3 online.
had increased IL-13 frequencies, particularly among the IASI-E/inflammation and IASI-S/scaling components highly
CLAe CD4þ (CIE 1.8% vs. control 0.7%, P ¼ 0.007; correlated with total IASI (P < 0.0001; Supplementary
Supplementary Figure S3b) and CD8þ (CIE 1.3% vs. control Figure S4a and b). Th17 (r ¼ 0.4, P ¼ 0.004;
0.3%, P ¼ 0.01; LI 1.4% vs. control 0.3%, P ¼ 0.005; Supplementary Figure S4c)/Tc17 (r ¼ 0.34, P ¼ 0.016;
Supplementary Figure S3d) subsets. Th9/Tc9 occurrence was Supplementary Figure S4d) and Th22 (r ¼ 0.43, P ¼ 0.001;
upregulated in NS and LI (Supplementary Figure S3eeh). Supplementary Figure S4e)/Tc22 (r ¼ 0.32, P ¼ 0.02;
Only EI showed significantly lower IFN-g levels than controls Supplementary Figure S4f) correlated significantly with total
(P < 0.05; Supplementary Figure S3iel). IASI. Analogous correlations were observed between Th17/

2160 Journal of Investigative Dermatology (2018), Volume 138


T Czarnowicki et al.
Th17/Tc17/Th22/Tc22-Skewing in Ichthyosis Blood

CD4+ T cells CD8+ T cells

a IL-13+CLA+ b IL-13+CLA–
c IL-13+CLA+ d IL-13+CLA–
*** *** *** 10.0 *
*** *** 50 *** **
*** 8 *** *** +
*** * *** *** *
*** * *** *
*** * *** ***
*** ** 40 ** ***
*** *** *** 7.5 ***
40 *** 6 ** *** **
** ***
Frequency (%)

Frequency (%)
30
5.0
4
20
20

2 2.5
10

0 0 0 0.0

Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis

e IL-9+CLA+ f IL-9+CLA– g IL-9+CLA+ h IL-9+CLA–


60 *** + 30 +
** 1.5 +
***
*** +
**
* * **
* +
4
40
Frequency (%)

20

Frequency (%)
1.0

20 2
10 0.5

0 0 0 0.0

Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis

i IFN-γ+CLA+ j +
IFN-γ CLA
– k IFN-γ+CLA+ l IFN-γ+CLA–
+
* +
* 120
* *
* *
60
60 60
Frequency (%)

Frequency (%)

80
40
40 40

40
20 20 20

0 0 0 0

Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis Normal Psoriasis AD Ichthyosis

Figure 3. IL-13D/IL-9D/IFN-gD cytokine frequencies. (aed) IL-13þ, (eeh) IL-9þ, and (iel) IFN-gþ frequencies in CLAþ/e, CD4þ/CD8þ T cells in control (red),
psoriasis (green), AD (purple), and ichthyosis (blue). Bar plots represent means (black)/medians (red)  SEMs. P-values are designated as ***<0.001, **<0.01,
*<0.05, þ<0.1. AD, atopic dermatitis; CLA, cutaneous lymphocyte antigen; SEM, standard error of the mean/median.

Tc17/Th22/Tc22 and IASI-E (Supplementary Figure S5aed). significantly correlated with age (P < 0.04; Supplementary
IASI-S correlated with both CD8þ/CLAþ cells (r ¼ 0.31, P ¼ Figure S4m and n). Ichthyosis severity showed a trend of
0.02; Supplementary Figure S5e) and Th9/Tc9 cells (P < 0.05; amelioration with age (Supplementary Figure S4o). Lastly,
Supplementary Figure S5f and g). Although Th22 cells IL-22-producing cells highly correlated with both IL-17 and
correlated positively with TEWL (r ¼ 0.28, P ¼ 0.06; IL-13 CD4þ/CD8þ T-cell subsets (P < 0.01 for all;
Supplementary Figure S4g), negative correlations were Supplementary Figure S5hek).
observed between Th9/Tc9 (r ¼ e0.35, P ¼ 0.02 for both; To assess how skin-homing T cells in blood correlate with
Supplementary Figure S4h and i) and TEWL. Tc2 (r ¼ 0.44, cutaneous T cells and disease severity in ichthyosis, we per-
P ¼ 0.003; Supplementary Figure S4j) significantly correlated formed immunostaining for CD3þ T cells in skin from avail-
with the 5D-pruritus scale, with lesser correlations found able ichthyosis samples (n ¼ 27). CD3þ cell counts
with Th17 (r ¼ 0.32, P ¼ 0.04) and Th22 (r ¼ 0.28, P ¼ 0.07; correlated significantly with ICOS-activated systemic Tcm
Supplementary Figure S4k and l). To evaluate for the effect of cells (r ¼ 0.45, P ¼ 0.02; Supplementary Figure S5l), with a
disease chronicity, we also correlated parameters with age. trend toward significance for IASI-E (r ¼ 0.34, P ¼ 0.08;
IFN-g, previously associated with disease chronicity Supplementary Figure S5m), but not IASI-S (P ¼ 0.9;
(Czarnowicki et al., 2015a, 2017a; Gittler et al., 2012), was Supplementary Figure S5n, Supplementary Table S3).

www.jidonline.org 2161
T Czarnowicki et al.
Th17/Tc17/Th22/Tc22-Skewing in Ichthyosis Blood

a ICOS+CLA+CD4+ Tem cells b + – +


ICOS CLA CD4 Tem cells c HLA-DR+CLA+CD4+ Tem cells d HLA-DR+CLA-CD4+ Tem cells
* ** +
* * 30
60 *
* ** +
* *
75 *
*** *
***
100 *** *** 20
***
Frequency (%)

*** +
* 40
*** +
***
** 50

50 10
20
25

0
0 0 0

Normal NS LI EI CIE Normal NS LI EI CIE Normal NS LI EI CIE Normal NS LI EI CIE

e ICOS+CLA+CD4+ Tcm cells f ICOS+CLA–CD4+ Tcm cells g HLA-DR+CLA+CD4+ Tcm cells h HLA-DR+CLA-CD4+ Tcm cells
+
** *** +
*
*** 60 *
** + + 30
**
90 ** *
*
+ *
*
* 40
* **
Frequency (%)

*
40 20
60

20 10
20
30

0
0 0 0

Normal NS LI EI CIE Normal NS LI EI CIE Normal NS LI EI CIE Normal NS LI EI CIE


D þ þ þ/e þ
Figure 4. Ichthyosis subtypes CD4 T-cell activation. (aeh) ICOS and HLA-DR activation in CLA CD4 Tcm/Tem cells in control (red), NS, LI, EI, and CIE
(shades of blue). Bar plots represent means (black)/medians (red)  SEMs. P-values are designated as ***<0.001, **<0.01, *<0.05, þ<0.1. CIE, congenital
ichthyosiform erythroderma; CLA, cutaneous lymphocyte antigen; EI, epidermolytic ichthyosis; ICOS, inducible co-stimulator molecule; LI, lamellar ichthyosis;
NS, Netherton syndrome; SEM, standard error of the mean/median; Tcm, central memory T cell; Tem, effector memory T cell.

Total Tregs are increased in ichthyosis and correlate with biomarkers to monitor treatment responses and advance
T-cell activation. targeted therapeutic development.
Ichthyosis showed significantly increased total (defined Similar to our recent findings of Th17-dominant inflam-
as CD25þCD127eCCR4þ) (Gorski et al., 2010) (P < 0.01), mation in skin (Paller et al., 2017b), the current data
but not skin-homing (CCR4þCLAþ) Treg frequencies versus establish shared elevation of IL-17 frequencies in ichthyoses
control, psoriasis, and AD, with largely similar blood across skin homing/CLAþ and systemic/CLAe CD4þ
distribution among ichthyosis subtypes (Supplementary and CD8þ populations, supporting their systemic immune
Figure S6aed online). Similar to AD report (Czarnowicki dysregulation and hinting at possible associated comorbid-
et al., 2015c), Tregs significantly correlated with skin- ities. IL-22 was profoundly upregulated in ichthyosis to
homing Tcm/Tem, HLA-DR activated T-cell subsets levels even higher than in AD, a disease with a prominent
(P < 0.013 for all; Supplementary Figure S6eeh, IL-22 signature (Czarnowicki et al., 2015b; Gittler et al.,
Supplementary Table S3). 2012). Furthermore, Th17/Tc17/Th22/Tc22 subsets corre-
late with total IASI and IASI-E, and cluster close to TEWL,
DISCUSSION 5D pruritus score and IASI-S, suggesting their possible role
This study details all recognized effector T-cell subsets within in ichthyosis skin manifestations. The close link of barrier
CLAþ and CLAe compartments in the circulation of the four measures to dysregulated cytokines indicates ongoing
more common subtypes among orphan ichthyoses (ARCI-LI, crosstalk between these components in ichthyosis, similar to
ARCI-CIE, EI, and NS) compared with healthy controls. AD and psoriasis (Czarnowicki et al., 2014; Guttman-Yassky
Because of the suggested phenotypical similarities to psori- et al., 2011). NS and CIE show the highest IL-17/IL-22
asis and AD in skin, ichthyosis blood samples were analyzed expression, reflecting their more erythrodermic phenotype
in parallel with previously published AD (Th2/Th22-skewed) (Richard, 1993) and higher IASI-E and TEWL values
and psoriasis (Th1/Th17-polarized) samples (Czarnowicki (Supplementary Table S4 online), further highlighting the
et al., 2015c). We not only found a common blood pheno- association between inflammation and barrier impairment.
type in the largest group of patients with ichthyosis, but also IL-22 and IL-17 are extensively involved in protective anti-
characterized the unique blood profiles of different subtypes. bacterial immune responses (Cho et al., 2010; Jin and
Expanding the ichthyosis phenotyping to blood, including Dong, 2013), antimicrobial peptide secretion, regulation
identification of activation markers and polarized cytokines, of chronic inflammation, and maintenance of epithelial
carries the potential for the development of noninvasive integrity (Sonnenberg et al., 2011; Wolk et al., 2004). The

2162 Journal of Investigative Dermatology (2018), Volume 138


T Czarnowicki et al.
Th17/Tc17/Th22/Tc22-Skewing in Ichthyosis Blood

CD4+ T cells CD8+ T cells

a IL-17+CLA+ b IL-17+CLA– c IL-17+CLA+ d IL-17+CLA–


+ + +
** * 50
+ 15
*
+
+
* 40 7.5 *
20 +
Frequency (%)

Frequency (%)
*
10 + *
30 *
5.0

10 20
5
2.5
10

0 0 0 0.0

Normal NS LI EI CIE Normal NS LI EI CIE Normal NS LI EI CIE Normal NS LI EI CIE

e IL-22+CLA+
f IL-22+CLA–
g +
IL-22 CLA
+ h IL-22+CLA–

***
+ +
25 +
*
+
+ +
+
** * 15
** * 100
** 20 + *
60 *** **
** * *
** **
+
Frequency (%)

Frequency (%)
***
** 75 *
15 *
+ **
+ **
10
40
10 50

5
20
5 25

0 0 0 0

Normal NS LI EI CIE Normal NS LI EI CIE Normal NS LI EI CIE Normal NS LI EI CIE


D D þ þ þ/ þ þ
Figure 5. Ichthyosis subtypes IL-17 /IL-22 cytokine frequencies. (aed) IL-17 and (e-h) IL-22 frequencies in CLA , CD4 /CD8 T-cells control (red), NS,
LI, EI, and CIE (shades of blue). Bar plots represent means (black)/medians (red)  SEMs. P-values are designated as ***<0.001, **<0.01, *<0.05, þ<0.1. CIE,
congenital ichthyosiform erythroderma; CLA, cutaneous lymphocyte antigen; EI, epidermolytic ichthyosis; LI, lamellar ichthyosis; NS, Netherton syndrome;
SEM, standard error of the mean/median.

increased IL-17/IL-22 blood subsets are consistent with the et al., 2009). IL-9 is increased in ichthyosis blood, perhaps
elevated IL-17/IL-22-related markers in skin, including perpetuating the Th17/Th22 deviation. Although positively
antimicrobial peptides (Paller et al., 2017b). Although pos- correlated with IASI-S, IL-9 correlates negatively with TEWL,
itive correlations between IL-13 and IL-22 observed here are suggesting a complex interaction between Th9/Tc9 subsets
similar to AD findings (Czarnowicki et al., 2015b), IL-17 and barrier homeostasis. Similar to alopecia areata
and IL-22 associations might partially derive from the abil- (Czarnowicki et al., 2017a) and AD (Czarnowicki et al.,
ity of Th17 cells to produce both cytokines (Liang et al., 2015a; Gittler et al., 2012), in which IFN-g marks disease
2006). chronicity, Th1/Tc1 subsets correlate with age. Ichthyosis has
Despite the cutaneous resemblance to psoriasis (Paller similar or even higher Tem/Tcm cell frequencies than AD,
et al., 2017b), the ichthyosis blood profile blends character- suggesting excessive circulatory activation with concurrent
istics of both AD and psoriasis, with increased ICOS/HLA-DR immune reserve accumulation in lymph nodes, as indicated
and IL-22 activation, features of both AD and psoriasis, but by increased Tcm cells, potentially explaining disease chro-
also elevated IL-17 level characteristic of psoriasis and nicity, recurrent exacerbations, and treatment failures.
increased IL-13 frequencies typifying AD. Clustering (and ICOS, the mid-activation marker, is necessary for intact
positive correlations) of Th2/Tc2 cells and the 5D pruritus immune circuits (Tafuri et al., 2001) and shapes adaptive
scale not only support the IL-13 role as an itch mediator immunity (Coyle et al., 2000). Although ICOS augments Th2
(Mollanazar et al., 2016) but may also suggest a tool for responses (Tesciuba et al., 2008), it is also critical for Th17
patient targeted therapeutic stratification. development (Nurieva, 2005; Paulos et al., 2010). ICOS
Surprisingly, despite the accepted association between NS activation is profoundly increased across all Tcm/Tem/CD4þ/
and atopy (Samuelov and Sprecher, 2014), IL-13 is not CD8þ/CLAþ/systemic subsets in ichthyosis to levels even
increased in NS blood or skin (Paller et al., 2017b). Lack of higher than the exceedingly activated AD (Czarnowicki et al.,
Th2 skewing in NS may result from the reciprocal regulation 2015c), probably augmenting IL-17 responses in ichthyosis
by Th17 (Lynch et al., 2016), which is highest in NS. IL-9 has blood and skin (Paller et al., 2017b). The fact that the chronic
been implicated in allergic/atopic disorders (Wisniewski and activation marker HLA-DR (Reddy et al., 2004) is increased
Borish, 2011), including AD (Ma et al., 2014) and more in CD4þ Tem/Tcm cells, but significantly low among CD8þ
recently psoriasis (Ruiz-Romeu et al., 2017; Singh et al., subsets, suggests a specific T-cell activation sequence in
2013). IL-9 induces Th17 differentiation and promotes ichthyosis with persistent ICOS/HLA-DR CD4þ activation,
Th17-driven inflammation (Elyaman et al., 2009; Nowak and a minor contribution of CD8þ cells in chronic disease.

www.jidonline.org 2163
T Czarnowicki et al.
Th17/Tc17/Th22/Tc22-Skewing in Ichthyosis Blood

TEWL

5-D-Pruritus Scale
Age

IASI-S
+

CD4+IL13+CLA+
CD8+IL13+CLA+
CD4 IFNγ CLA

CD4+IL9+CLA+
CD4+CLA+
CD8+CLA+
+

CD8+IFNγ+CLA+

CD8 IL9+CLA+

+
CD4 IL17 CLA
+

CD8+IL17+CLA+
+

CD4+IL22+CLA+
IASI-E
IASI
CD4 IL9+
CD4+IL9+CLA–
+

CD8+IL9+
CD8+IL9+CLA–

CD8+IL22+CLA+
Correlation

+
+

CD4+IL13+
CD4+IL13+CLA–
1.0

CD8+IL13+
CD8+IL13+CLA–
CD4+IL22+
CD4 IL22+CLA–
CD4 IL17+
CD4+IL17+CLA–

CD8+IL17+
CD8 IL17+CLA–

CD8+IL22+
CD8+IL22+CLA–
CD4+IFNγ+CLA–
CD8+IFNγ+
CD8+IFNγ+CLA–

CD4+IFNγ+

0.5

+
0.0

+
+
-0.5

-1.0

Age ** * * *** ** * + * * **
+ + + + + +
CD4 IFNγ CLA *** ** ** *** *** * *
CD8+IFNγ+CLA+ *** *** *** *** **
CD8+IFNγ+ *** *** *** * * + + *
CD8+IFNγ+CLA– *** *** * * + + *
CD4+IFNγ+ *** + +

CD4+IFNγ+CLA– +

CD4+CLA+ *** ** * * ** * + +

CD8+CLA+ ** ** *** *** ** * * * * * * *


CD8+IL9+CLA+ *** *** *** *** * ** +

CD8+IL9+ *** *** *** ** * ** *


CD8+IL9+CLA– + * ** + * +

CD4+IL9+CLA+ *** ** + * + + + + +

CD4+IL9+ *** *
CD4+IL9+CLA–
TEWL + + + + +
IASI-S *** +
IASI *** + * ** ** * * ** ** * * *
IASI-E + * ** ** * * *** *** * ** ** ** *
+ + + + +
CD4 IL17 CLA *** *** *** *** *** *** *** * *
CD8+IL17+CLA+ *** *** *** *** *** *** ** ** **
CD4+IL17+ *** *** *** *** *** ** ** *** *** *
CD4+IL17+CLA– *** *** *** *** ** ** *** *** *
CD8+IL17+ *** *** *** * ** *** *** +

CD8+IL17+CLA– *** *** * ** *** *** +

CD8+IL13+ *** ** ** *** *** *** *** * ** *** ***


CD8+IL13+CLA– ** ** *** *** *** *** + ** *** **
CD4+IL22+ *** *** *** *** *** + * **
CD4+IL22+CLA– *** *** *** *** + * **
CD4+IL22+CLA+ *** *** *** * *
CD8+IL22+CLA+ *** *** + *
CD8+IL22+ *** + *
CD8+IL22+CLA– + *
5-D-Pruritus Scale **
CD4+IL13+ *** *** ***
CD4+IL13+CLA– * **
CD4+IL13+CLA+ ***
CD8+IL13+CLA+
Age
CD4+IFNγ+CLA+
CD8+IFNγ+CLA+
CD8+IFNγ+
CD8+IFNγ+CLA–
+

CD4+IFNγ+CLA–
CD4+CLA+
+

CD8+IL9+
CD8+IL9+CLA–
+

CD4+IL9+CLA–
TEWL
IASI-S
IASI
IASI-E
+

CD4+IL17+
CD4+IL17+CLA–
CD8+IL17+

CD8+IL13+
CD8+IL13+CLA–
CD4+IL22+

CD4+IL22+CLA+
+

5-D-Pruritus Scale

CD4+IL13+
CD4+IL13+CLA–
+

CD8+IL13+CLA+
CD8 IL17 CLA

CD4 IL22 CLA

CD8 IL22 CLA


CD4 IFNγ

CD8 CLA
CD8 IL9 CLA

CD4 IL9 CLA


CD4 IL9

CD4 IL17 CLA


CD8 IL17 CLA

CD8 IL22 CLA


CD8 IL22

CD4 IL13 CLA


+

+
+

+
+

+
+

***p<0.001 **p<0.01 *p<0.05 +p<0.1

Figure 6. Unsupervised hierarchical clustering of polarized cytokine subsets with ichthyosis clinical measures, using Pearson correlation as a similarity
metric. CLAþ/e Th17/Tc17 clustered with or adjacent to total IASI, IASI-E, IASI-S and TEWL (blue box), and close to CLAþ/e Th22/Tc22 (orange box). Th2/Th2
clustered with the 5-D Pruritus Scale (teal box). All clinical measures clustered close to each other (red ellipses). CLAþ/e Th1/Tc1 cells clustered with age
(red box). The heatmap shows the positive (red) or negative (blue) correlations of all parameters with color intensity reflecting the strength of the correlation
(e1 to þ1). Dendrograms are shown as a tree, representing the distance between variables. P-values are designated as ***<0.001, **<0.01, *<0.05, þ<0.1.
CLA, cutaneous lymphocyte antigen; IASI, ichthyosis area severity index; IASI-E, ichthyosis area severity index- erythema; IASI-S, ichthyosis area severity
index-scaling; Tc, cytotoxic T cell; TEWL, transepidermal water loss; Th, T helper.

2164 Journal of Investigative Dermatology (2018), Volume 138


T Czarnowicki et al.
Th17/Tc17/Th22/Tc22-Skewing in Ichthyosis Blood

Tregs are crucial in maintaining immune tolerance (Long targeted treatment investigation, positive correlations be-
and Buckner, 2011), and their levels may influence activa- tween CLAþ/CLAe IL-17/IL-22-producing cells and clinical
tion of effector cells (McHugh and Shevach, 2002). Our data measures support their possible roles in ichthyosis. Further-
show significantly increased systemic Treg frequencies in more, our findings suggest that “nonsyndromic/skin-limited”
ichthyosis, even higher than in AD. Conversely, CLAþ Tregs ichthyoses may have systemic manifestations and support
were lower than AD and psoriasis, possibly resulting from the systemic therapeutic approaches for severely affected
complex counterequilibrium between Th17 cells and Tregs patients.
(Diller et al., 2016; Eisenstein and Williams, 2009). The
positive correlations between activated skin-homing memory MATERIALS AND METHODS
T cells and Treg may be secondary to the possible perpetu- Patient characteristics and samples
ation of inflammation via Tregs, in addition to their sup- Blood was obtained from 47 patients with ichthyosis (30 females, 17
pressive effects. males; ages 1e57 years, mean 22 years; 13 ARCI-LI, 18 ARCI-CIE, 8
Whether expanded Th17/Th22 cytokines are primary NS, 8 EI) (Paller et al., 2017b), 43 patients with moderate-to-severe
pathogenic factors in ichthyosis or secondary to chronic AD (19 females, 24 males; ages 18e81 years; mean 43 years), 24
antigenic stimulation and barrier impairment still needs patients with moderate-to-severe psoriasis (19 females, 24 males;
investigation. Our preliminary skin and blood data showing ages 18e81 years; mean 43 years), and 59 controls (30 females, 29
Th1/Th17/IL-23 elevation in a patient with an ichthyotic males; ages 6e66 years; mean 25 years). No age (P ¼ 0.32, one-way
syndrome resulting from DSP mutation led to initiation of analysis of variance), ethnicity (P ¼ 0.57, chi-square test), or sex
targeted Th1/Th17/IL-23 blockade with ustekinumab in two (P ¼ 0.18, chi-square test) disparities were observed between
patients. Ustekinumab treatment reduced skin erythema, ichthyosis patients and controls. Eighteen patients with ichthyosis
scaling, and TEWL, supporting a possible pathogenic role for were younger than 18 years. All patients were classified clinically,
IL-17 (Paller et al., 2017a). Furthermore, the significant Th17 but genotyping was performed on 96% of patients and was consis-
skewing in our recent ichthyosis skin phenotyping in 21 pa- tent with the clinical diagnosis. All NS resulted from SPINK5
tients with the four orphan forms studied herein (Paller et al., mutations; seven of the eight EI had a KRT10 and one a KRT1
2017b) engendered an ongoing clinical trial of secukinumab mutation; clinically patients with LI had TGM1 mutations, but one
(anti-IL17-antibody) in patients with ichthyosis had a NIPAL4 mutation; and the patients with CIE included three
(NCT03041038). with harlequin ichthyosis (all ABCA12 mutations), but others had
Ichthyoses are characterized by overt systemic inflamma- one of various genes mutated as have been described with CIE
tion, as reflected by increased T-cell activation and immune (ABCA12, ALOXE3, ALOX12B, CerS3, NIPAL4, and PNPLA1). Of
dysregulation of multiple polar cytokines among CLAe sub- the 47 patients, 2 (4%) took a retinoid orally (one acetretin, one
sets. These data further highlight ichthyosis as a systemic, isotretinoin), 3 (6%) used topical tazarotene to localized areas, 1 had
rather than skin-limited disease, similar to psoriasis and AD, used budesonide ointment, and 1 tacrolimus ointment for pruritus
both currently conceptualized as systemic disorders in the previous few days. No patient was treated with an oral
(Guttman-Yassky and Krueger, 2017; Guttman-Yassky et al., immunosuppressant medication.
2017). Both were linked to systemic comorbidities, including Although cytokine polarization was similar between children and
the increased risk of cardiovascular disease (Brunner et al., adults by sensitivity analysis, adults showed higher T-cell activation
2017; Puig, 2017; Silverberg and Greenland, 2015). In pso- than children, probably secondary to disease chronicity and ongoing
riasis, IL-17 was suggested to drive cutaneous and systemic immune stimulation. Written institutional review board-approved
inflammation, as well as the comorbidities (Krueger and informed consents were obtained from subjects (12 years) and
Brunner, 2017). Although there is no consensus on comor- parents (<18 years). Ichthyosis severity was calculated using the IASI
bid associations in ichthyoses, our results suggest the need for score, as previously described (Paller et al., 2017b), a nonvalidated
early monitoring for systemic extracutaneous involvement, scale that combines the IASI-E/inflammation and IASI-S into a total
particularly for metabolic and cardiovascular disease. It is IASI score. Additional clinical measures, including TEWL on the
possible that excessive T-cell activation and IL-17 polariza- upper arm and 5D pruritus score, were captured as well.
tion in ichthyosis promote systemic manifestations that, Demographic data are summarized in Supplementary Tables S4 and
similar to psoriasis (Conrad and Gilliet, 2018), might be S5 online.
addressed or even prevented through targeted therapeutics.
Isolation of peripheral blood mononuclear cells
Our study had limitations, including the fact that a small
Peripheral blood mononuclear cells were isolated from whole blood
subset of ichthyosis patients was younger than 18 years,
by Ficoll-Paque Plus (GE Healthcare, Uppsala, Sweden), as previ-
limiting our ability to compare absolute numbers between
ously described (Czarnowicki et al., 2017a) (see Supplementary
age groups. However, their immune polarity was comparable
Materials and Methods online).
to that of adults (data not shown). Also, although our study
presents the largest dataset of ichthyoses blood profiles, there Stimulation of blood cell populations for cytokine responses
were patient number variations across different subtypes due Ex vivo cell activation is required to detect cytokine production, as
to their rarity. less than 1% of nonstimulated cells produce cytokines. Whole blood
In sum, ichthyosis blood is accompanied by increased was incubated with phorbol 12-myristate 13-acetate (25 ng/ml) plus
systemic and skin-homing T-cell activation and multicytokine ionomycin (2 mg/ml) in the presence of brefeldin A (10 mg/ml) for 4
polarization, with IL-17/IL-22 predominance, similar to the hours at 37 C to induce cytokine responses. After stimulation, red
skin compartment. Although the pathogenic contribution of blood cells were lysed with FACS lysing solution to obtain leuko-
each cytokine pathway will only be determined through cytes (see Supplementary Materials and Methods).

www.jidonline.org 2165
T Czarnowicki et al.
Th17/Tc17/Th22/Tc22-Skewing in Ichthyosis Blood

Cell surface staining and intracellular staining on peripheral Cho JS, Pietras EM, Garcia NC, Ramos RI, Farzam DM, Monroe HR, et al.
blood mononuclear cells and stimulated and nonstimulated IL-17 is essential for host defense against cutaneous Staphylococcus aureus
infection in mice. J Clin Invest 2010;120:1762e73.
CD4/CD8 T cells
Peripheral blood mononuclear cells were stained with Conrad C, Gilliet M. Psoriasis: from pathogenesis to targeted therapies. Clin
Rev Allergy Immunol 2018;54:102e13.
fluorochrome-labeled antibodies to cell surface markers (CD3, CD8,
Coyle AJ, Lehar S, Lloyd C, Tian J, Delaney T, Manning S, et al. The CD28-
CD4, CD45RO, CCR7, ICOS, HLA-DR, and CLA). Stimulated and related molecule ICOS is required for effective T cell-dependent immune
nonstimulated blood cells were stained for cell surface markers responses. Immunity 2000;13:95e105.
(CD3, CD4, CD69, and CLA) and permeabilized with FACS/perm to Czarnowicki T, Esaki H, Gonzalez J, Malajian D, Shemer A, Noda S, et al.
stain for cytokines, including IL-13, IL-22, IL-9, IFN-g, and IL-17A Early pediatric atopic dermatitis shows only a cutaneous lymphocyte an-
(see Supplementary Materials and Methods). tigen (CLA)(þ) TH2/TH1 cell imbalance, whereas adults acquire CLA(þ)
TH22/TC22 cell subsets. J Allergy Clin Immunol 2015a;136:941e51.
Immunohistochemistry Czarnowicki T, Gonzalez J, Shemer A, Malajian D, Xu H, Zheng X, et al.
Immunohistochemistry staining for CD3 was performed on frozen Severe atopic dermatitis is characterized by selective expansion of
OCT-embedded cryostat sections using purified mouse anti-human circulating TH2/TC2 and TH22/TC22, but not TH17/TC17, cells within
the skin-homing T-cell population. J Allergy Clin Immunol 2015b;136:
mAb (Clone SK7, BD Biosciences), as described (Esaki et al., 104e15.
2015; Suarez-Farinas et al., 2015). Immunohistochemistry staining Czarnowicki T, He HY, Wen HC, Hashim PW, Nia JK, Malik K, et al.
was performed on a subset of 27 available patient samples. Cell Alopecia areata is characterized by expansion of circulating Th2/Tc2/
counts were quantified with ImageJ V1.42 (National Institute of Th22, within the skin-homing and systemic T-cell populations. Allergy
2018;73:713e23.
Health, Bethesda, MD). See Supplementary Materials and Methods
for details. Czarnowicki T, Krueger JG, Guttman-Yassky E. Skin barrier and immune
dysregulation in atopic dermatitis: an evolving story with important clinical
Statistical analysis implications. J Allergy Clin Immunol Pract 2014;2:371e9.
Welch’s t-test and the Wilcoxon Mann-Whitney test were used to Czarnowicki T, Malajian D, Shemer A, Fuentes-Duculan J, Gonzalez J,
Suarez-Farinas M, et al. Skin-homing and systemic T-cell subsets show
compare means and medians of variables, respectively. Unsuper-
higher activation in atopic dermatitis versus psoriasis. J Allergy Clin
vised hierarchical clustering of variables was performed using the R Immunol 2015c;136:208e11.
package hclust with Pearson correlation as a similarity metric with Czarnowicki T, Santamaria-Babi LF, Guttman-Yassky E. Circulating CLA(þ) T
average agglomeration algorithm and presented as a heatmap with a cells in atopic dermatitis and their possible role as peripheral biomarkers.
dendrogram (using the R package ape). Individual scatter plots Allergy 2017b;72:366e72.
display coefficients, 95% confidence intervals, and P-values. DiGiovanna JJ, Robinson-Bostom L. Ichthyosis: etiology, diagnosis, and
management. Am J Clin Dermatol 2003;4:81e95.
P-values were designated as ***<0.001, **<0.01, *<0.05, þ<0.1.
Diller ML, Kudchadkar RR, Delman KA, Lawson DH, Ford ML. Balancing
CONFLICT OF INTEREST inflammation: the link between Th17 and regulatory T cells [e-pub ahead
JGK: Institutional research grants and funding from Novartis, Pfizer, Boeh- of print] Mediators Inflamm 2016;6309219. https://doi.org/10.1155/2016/
ringer, Janssen, Abbvie, Amgen, Innovaderm, BMS, EMD Serono, Paraxial, 6309219.
Leo Pharma, Vitae, Kinta, Regeneron, Novan, AkrosNovartis, Sienna, UCB; Dreyfus I, Pauwels C, Bourrat E, Bursztejn AC, Maruani A, Chiaverini C, et al.
consulting fees from Novartis, Pfizer, Boehringer, Amgen, Lilly, BiogenIdec, Burden of inherited ichthyosis: a French national survey. Acta Derm
Janssen, Abbvie,Leo Pharma, Escalier, Acros, Roche, Valiant, Allergen, UCB, Venereol 2015;95:326e8.
Aurigene. EGY: Institutional grants from Celgene, Dermira, Janssen, Leo,
Merck Novartis, Regeneron BMS; consulting fees from Abbie, Allergen, Eisenstein EM, Williams CB. The T(reg)/Th17 cell balance: a new paradigm for
Amgen Anacor, Asana,BMS, Celgene, Celsus,Curadim, Dermira, Drais, autoimmunity. Pediatr Res 2009;65:26Re31R.
Escalier, Galderma, Genentech, GlaxoSmithKline, Glenmark, Kymab Limited, Elyaman W, Bradshaw EM, Uyttenhove C, Dardalhon V, Awasthi A, Imitola J,
Kyowa Kirin, LEAD, Leo, Novartis, Pfizer, Regeneron, Sanofi, Sienna, Ther- et al. IL-9 induces differentiation of TH17 cells and enhances function of
avance. The remaining authors state no conflict of interest. FoxP3þ natural regulatory T cells. Proc Natl Acad Sci USA 2009;106:
12885e90.
ACKNOWLEDGMENTS
Esaki H, Ewald DA, Ungar B, Rozenblit M, Zheng X, Xu H, et al. Identification
This research was supported by the Foglia Family Foundation Endowment. We
of novel immune and barrier genes in atopic dermatitis by means of laser
acknowledge Core resources provided by the Northwestern University Skin
capture microdissection. J Allergy Clin Immunol 2015;135:153e63.
Disease Research Center (NIAMS P30AR057216); genetic testing of most
patients was performed in the laboratory of Dr Keith Choate, Yale University. Ferenczi K, Burack L, Pope M, Krueger JG, Austin LM. CD69, HLA-DR and
We acknowledge the assistance of the Foundation for Ichthyosis and Related the IL-2R identify persistently activated T cells in psoriasis vulgaris lesional
Skin Types, which facilitated enrollment of many of these subjects at the 2016 skin: blood and skin comparisons by flow cytometry. J Autoimmun
FIRST meeting. 2000;14:63e78.
Fontao L, Laffitte E, Briot A, Kaya G, Roux-Lombard P, Fraitag S, et al.
SUPPLEMENTARY MATERIAL Infliximab infusions for Netherton syndrome: sustained clinical improve-
ment correlates with a reduction of thymic stromal lymphopoietin levels in
Supplementary material is linked to the online version of the paper at www.
the skin. J Invest Dermatol 2011;131:1947e50.
jidonline.org, and at https://doi.org/10.1016/j.jid.2018.03.1523.
Gittler JK, Shemer A, Suarez-Farinas M, Fuentes-Duculan J, Gulewicz KJ,
REFERENCES Wang CQF, et al. Progressive activation of T(H)2/T(H)22 cytokines and
selective epidermal proteins characterizes acute and chronic atopic
Akagi A, Kitoh A, Moniaga CS, Fujimoto A, Fujikawa H, Shimomura Y, et al.
dermatitis. J Allergy Clin Immunol 2012;130:1344e54.
Case of Netherton syndrome with an elevated serum thymus and
activation-regulated chemokine level. J Dermatol 2013;40:752e3. Gorell E, Nguyen N, Lane A, Siprashvili Z. Gene therapy for skin diseases.
Cold Spring Harb Perspect Med 2014;4:a015149.
Briot A, Deraison C, Lacroix M, Bonnart C, Robin A, Besson C, et al. Kalli-
krein 5 induces atopic dermatitis-like lesions through PAR2-mediated Gorski K, Sitko J, Thomas J, Gavin M, Cottrell S, Cox C, et al. CCR4 improves
thymic stromal lymphopoietin expression in Netherton syndrome. J Exp phenotypic identification of T-regulatory cells; validation and imple-
Med 2009;206:1135e47. mentation of clinical test. J Immunol 2010;184(Supplement). 49.26.
Brunner PM, Suarez-Farinas M, He H, Malik K, Wen HC, Gonzalez J, et al. Guttman-Yassky E, Krueger JG. Atopic dermatitis and psoriasis: two
The atopic dermatitis blood signature is characterized by increases in in- different immune diseases or one spectrum? Curr Opin Immunol
flammatory and cardiovascular risk proteins. Sci Rep 2017;7:8707. 2017;48:68e73.

2166 Journal of Investigative Dermatology (2018), Volume 138


T Czarnowicki et al.
Th17/Tc17/Th22/Tc22-Skewing in Ichthyosis Blood

Guttman-Yassky E, Krueger JG, Lebwohl MG. Systemic immune mechanisms Reddy M, Eirikis E, Davis C, Davis HM, Prabhakar U. Comparative analysis of
in atopic dermatitis and psoriasis with implications for treatment [e-pub lymphocyte activation marker expression and cytokine secretion profile in
ahead of print]. Exp Dermatol. https://doi.org/10.1111/exd.13336. stimulated human peripheral blood mononuclear cell cultures: an in vitro
Guttman-Yassky E, Nograles KE, Krueger JG. Contrasting pathogenesis of model to monitor cellular immune function. J Immunol Methods 2004;293:
atopic dermatitis and psoriasis—part I: clinical and pathologic concepts. 127e42.
J Allergy Clin Immunol 2011;127:1110e8. Renner ED, Hartl D, Rylaarsdam S, Young ML, Monaco-Shawver L, Kleiner G,
Hannula-Jouppi K, Laasanen SL, Ilander M, Furio L, Tuomiranta M, Marttila R, et al. Comel-Netherton syndrome defined as primary immunodeficiency.
et al. Intrafamily and interfamilial phenotype variation and immature im- J Allergy Clin Immunol 2009;124:536e43.
munity in patients with Netherton syndrome and Finnish SPINK5 founder Richard G. Autosomal recessive congenital ichthyosis. In: Adam MP,
mutation. JAMA Dermatol 2016;152:435e42. Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Mefford HC, et al., editors.
Jin W, Dong C. IL-17 cytokines in immunity and inflammation. Emerg Mi- GeneReviews. Seattle, WA: University of Washington, Seattle; 1993-
crobes Infect 2013;2:e60. 2018.
Kawashima J, Akiyama M, Takizawa Y, Takahashi S, Matsuo I, Shimizu H. Ruiz-Romeu E, Ferran M, de Jesus-Gil C, Garcia P, Sagrista M, Casanova JM,
Structural, enzymatic and molecular studies in a series of nonbullous et al. Microbe-dependent induction of IL-9 by CLA(þ) T cells in psoriasis
congenital ichthyosiform erythroderma patients. Clin Exp Dermatol and relationship with IL-17A. J Invest Dermatol 2018;138:580e7.
2005;30:429e31. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory
Krueger JG, Brunner PM. Interleukin-17 alters the biology of many cell types T cell subsets: function, generation, and maintenance. Annu Rev Immunol
involved in the genesis of psoriasis, systemic inflammation and associated 2004;22:745e63.
comorbidities. Exp Dermatol 2017;27:115e23. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of
Liang SC, Tan XY, Luxenberg DP, Karim R, Dunussi-Joannopoulos K, memory T lymphocytes with distinct homing potentials and effector
Collins M, et al. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells functions. Nature 1999;401:708e12.
and cooperatively enhance expression of antimicrobial peptides. J Exp Med Samuelov L, Sprecher E. Peeling off the genetics of atopic dermatitis-like
2006;203:2271e9. congenital disorders. J Allergy Clin Immunol 2014;134:808e15.
Long SA, Buckner JH. CD4þFOXP3þ T regulatory cells in human autoim- Sarri CA, Roussaki-Schulze A, Vasilopoulos Y, Zafiriou E, Patsatsi A,
munity: more than a numbers game. J Immunol 2011;187:2061e6. Stamatis C, et al. Netherton syndrome: a genotype-phenotype review. Mol
Lynch JP, Ferreira MA, Phipps S. Th2/Th17 reciprocal regulation: twists and Diagn Ther 2017;21:137e52.
turns in the complexity of asthma phenotypes. Ann Transl Med 2016;4:S59. Silverberg JI, Greenland P. Eczema and cardiovascular risk factors in 2 US
Ma L, Xue HB, Guan XH, Shu CM, Zhang JH, Yu J. Possible pathogenic role of adult population studies. J Allergy Clin Immunol 2015;135:721e8.
T helper type 9 cells and interleukin (IL)-9 in atopic dermatitis. Clin Exp Singh TP, Schon MP, Wallbrecht K, Gruber-Wackernagel A, Wang XJ, Wolf P.
Immunol 2014;175:25e31. Involvement of IL-9 in Th17-associated inflammation and angiogenesis of
McHugh RS, Shevach EM. The role of suppressor T cells in regulation of psoriasis. PLoS One 2013;8:e51752.
immune responses. J Allergy Clin Immunol 2002;110:693e702. Smith FJ, Irvine AD, Terron-Kwiatkowski A, Sandilands A, Campbell LE,
Mollanazar NK, Smith PK, Yosipovitch G. Mediators of chronic pruritus in atopic Zhao Y, et al. Loss-of-function mutations in the gene encoding filaggrin
dermatitis: getting the itch out? Clin Rev Allergy Immunol 2016;51:263e92. cause ichthyosis vulgaris. Nat Genet 2006;38:337e42.
Nowak EC, Weaver CT, Turner H, Begum-Haque S, Becher B, Schreiner B, Sonnenberg GF, Fouser LA, Artis D. Border patrol: regulation of immunity,
et al. IL-9 as a mediator of Th17-driven inflammatory disease. J Exp Med inflammation and tissue homeostasis at barrier surfaces by IL-22. Nat
2009;206:1653e60. Immunol 2011;12:383e90.
Nurieva RI. Regulation of immune and autoimmune responses by ICOS-B7h Suarez-Farinas M, Ungar B, Correa da Rosa J, Ewald DA, Rozenblit M,
interaction. Clin Immunol 2005;115:19e25. Gonzalez J, et al. RNA sequencing atopic dermatitis transcriptome profiling
provides insights into novel disease mechanisms with potential therapeutic
Oji V, Tadini G, Akiyama M, Blanchet Bardon C, Bodemer C, Bourrat E, et al.
implications. J Allergy Clin Immunol 2015;135:1218e27.
Revised nomenclature and classification of inherited ichthyoses: results of
the First Ichthyosis Consensus Conference in Soreze 2009. J Am Acad Tafuri A, Shahinian A, Bladt F, Yoshinaga SK, Jordana M, Wakeham A, et al.
Dermatol 2010;63:607e41. ICOS is essential for effective T-helper-cell responses. Nature 2001;409:
105e9.
Paller AS, Czarnowicki T, Renert-Yuval Y, Holland K, Huynh T, Sadlier M,
et al. The spectrum of manifestations in DSP (desmoplakin) SR6 domain Takeichi T, Akiyama M. Inherited ichthyosis: non-syndromic forms.
mutations: immunophenotyping and response to ustekinumab. J Am Acad J Dermatol 2016;43:242e51.
Dermatol 2018;78:495e505. Tesciuba AG, Shilling RA, Agarwal MD, Bandukwala HS, Clay BS,
Paller AS, Renert-Yuval Y, Suprun M, Esaki H, Oliva M, Huynh TN, et al. An Moore TV, et al. ICOS costimulation expands Th2 immunity by aug-
IL-17-dominant immune profile is shared across the major orphan forms of menting migration of lymphocytes to draining lymph nodes. J Immunol
ichthyosis. J Allergy Clin Immunol 2017b;139:152e65. 2008;181:1019e24.
Paulos CM, Carpenito C, Plesa G, Suhoski MM, Varela-Rohena A, Vahlquist A, Fischer J, Torma H. Inherited nonsyndromic ichthyoses: an up-
Golovina TN, et al. The inducible costimulator (ICOS) is critical for the date on pathophysiology, diagnosis and treatment. Am J Clin Dermatol
development of human T(H)17 cells. Sci Transl Med 2010;2:55ra78. 2018;19:51e66.
Puig L. Cardiometabolic comorbidities in psoriasis and psoriatic arthritis. Int J Wisniewski JA, Borish L. Novel cytokines and cytokine-producing T cells in
Mol Sci 2017;19:58. allergic disorders. Allergy Asthma Proc 2011;32:83e94.
Raychaudhuri SP. Role of IL-17 in psoriasis and psoriatic arthritis. Clin Rev Wolk K, Kunz S, Witte E, Friedrich M, Asadullah K, Sabat R. IL-22 increases
Allergy Immunol 2013;44:183e93. the innate immunity of tissues. Immunity 2004;21:241e54.

www.jidonline.org 2167

You might also like