Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

758 Annika E. Langeneckert et al. DOI: 10.1002/eji.201847965 Eur. J. Immunol. 2019.

49: 758–769
Clinical

Allergy and inflammation

Research Article
CCL21-expression and accumulation of CCR7+ NK cells
in livers of patients with primary sclerosing cholangitis
Annika E. Langeneckert1 , Sebastian Lunemann1 , Glòria Martrus1 ,
Wilhelm Salzberger1 , Leonard U. Hess1 , Annerose E. Ziegler1 ,
Tobias Poch2 , Gevitha Ravichandran3 , Urte Matschl1 , Jens B. Bosse4 ,
Gisa Tiegs3 , Lutz Fischer5 , Martina Koch8 , Johannes Herkel2 ,
Karl J. Oldhafer6 , Christoph Schramm2,7 and Marcus Altfeld1

1
Research Department of Virus Immunology, Heinrich Pette Institute, Hamburg, Germany
2
I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg,
Germany
3
Institute for Experimental Immunology and Hepatology, University Medical Center
Hamburg-Eppendorf, Hamburg, Germany
4
Research Department of Structural Cell Biology of Viruses, Heinrich Pette Institute,
Hamburg, Germany
5
Department of Hepatobiliary Surgery and Transplant Surgery, University Medical Center
Hamburg-Eppendorf, Hamburg, Germany
6
Department of General and Abdominal Surgery, Asklepios Hospital Barmbek, Semmelweis
University of Medicine Hamburg, Germany
7
Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf,
Hamburg, Germany
8
Department for General, Visceral and Transplant Surgery, University Hospital Mainz,
Germany

The pathogenesis of primary sclerosing cholangitis (PSC), an autoimmune liver disease,


remains unknown. The aim of this study was to characterize peripheral blood and intra-
hepatic NK cells from patients with PSC. Peripheral blood samples from patients with
PSC, other autoimmune liver diseases, and from healthy control individuals were used,
as well as liver tissues from PSC patients undergoing liver transplantation. Multipa-
rameter flow cytometry showed that peripheral blood NK cells from PSC patients were
significantly enriched for CCR7+ and CXCR3+ cells, and CCR7+ but not CXCR3+ cells were
also significantly increased within intrahepatic NK cells. PSC patients undergoing liver
transplantation furthermore had significantly higher plasma levels of the CCR7-ligand
CCL21, and the CXCR3-ligands CXCL10 and CXCL11, and significantly higher levels of
CCL21, but not CXCL10, were detected in liver tissues. CCR7+ and CXCR3+ NK cells from
PSC patients exhibited significantly higher functional capacity in peripheral blood, but
not liver tissues, consistent with chronic activation of these NK cells in the inflamed
liver. These data show that PSC is characterized by intrahepatic CCL21 expression and
accumulation of CCR7+ NK cells in the inflamed liver tissue.

Correspondence: Prof. Marcus Altfeld


e-mail: marcus.altfeld@leibniz-hpi.de


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
Eur. J. Immunol. 2019. 49: 758–769 Allergy and inflammation 759

Keywords: Chemokines r Innate immunity r Liver inflammation r NK cells r Primary sclerosing


cholangitis

 Additional supporting information may be found online in the Supporting Information section
at the end of the article.

Introduction Results

Primary sclerosing cholangitis (PSC) is a chronic liver disease char- Increased frequencies of CXCR3+ and CCR7+ NK cells
acterized by inflammation and fibrosis of intrahepatic and extra- in peripheral blood of patients with PSC
hepatic bile ducts, ultimately leading to liver cirrhosis [1]. PSC
is associated with inflammatory bowel disease in approximately Earlier studies have indicated a role of chemokine receptors CCR7
80% of patients [1–4] and a high risk for cholangiocarcinoma [5]. and CXCR3 in the T-cell infiltration of inflamed liver tissues in
To date, liver transplantation represents the only effective ther- PSC and primary biliary cirrhosis (PBC) patients [35, 36]. How-
apy [1], although, recurrent PSC occurs within 4 years after trans- ever, only few studies have investigated the role of NK cells,
plantation in over 20% of cases [6]. The pathogenesis of PSC is which are highly enriched within human liver-resident lympho-
not well understood, and environmental, genetic, and immuno- cytes. We characterized NK cell populations derived from PBMCs
logical factors have been implicated. Genome-wide association from patients with PSC, PBC, or AIH (autoimmune hepatitis),
studies have identified PSC risk-genes associated with immune and from healthy control individuals using multi-parameter flow
cell function [7, 8] and infiltrates of immune cells are observed cytometry. Gating strategies to identify NK cell subpopulations
in the proximity of bile ducts of patients with PSC [9, 10]. A dys- and expression of surface molecules on NK cells are shown in Sup-
regulated cross-talk between activated cholangiocytes [11–14], porting Information Fig. 1. Overall frequencies of peripheral blood
and infiltrating immune cells has been implicated in the patho- natural killer cells (pNK) cells was similar between PSC patients
genesis of PSC [10, 15–20]. As the infiltration of liver portal and healthy control individuals (Fig. 1A), while frequencies of
areas by immune cells is one of the characteristic features of CD56bright CD16dim pNK cells were higher in PSC patients com-
PSC, chemokines might play an important role in this process pared to healthy control individuals (Fig. 1B, p = 0.02) and PBC
[21]. However, little is known about the role of NK cells in PSC patients (p = 0.05). CD56dim CD16bright pNK cells were decreased
pathogenesis. in PSC patients compared to healthy control individuals (Fig. 1B,
The main functions of NK cells are the defense against p = 0.01). Additionally tested markers expressed on the surface of
pathogens and cancers [22], as well as regulation of inflam- NK cells did not show any significant differences between patient
matory immune responses, thereby linking innate and adaptive groups, with the exception of NKp44, which was less expressed
immune responses [23]. NK cells are highly enriched among on pNK cells of PSC patients (Supporting Information Fig. 2,
lymphocytes in human liver tissues and have been suggested to p = 0.004) and NKp46, which was more highly expressed on ihNK
participate in balancing immune tolerance and defense against cells of PSC patients (Supporting Information Fig. 2, p = 0.01).
pathogens [24–27]. Expression of HLA-Bw4 and -C2, which A t-distributed stochastic neighbor embedding (t-SNE) analysis of
serve as ligands for the inhibitory killer-cell immunoglobulin-like high-dimensional data generated by multi-parameter flow cytom-
receptors (KIR) KIR3DL1 and KIR2DL1, respectively, was signif- etry showed higher expression of the chemokine receptors CCR7
icantly reduced in PSC patients [28]. Furthermore, MIC-A and and CXCR3 on pNK cells of PSC patients compared to healthy
MIC-B, which serve as ligands for activating NK-cell receptors, controls (Fig. 1C). Quantitative analysis of multi-parameter flow
were upregulated in PSC [29, 30], suggesting that NK cells cytometry data demonstrated a significantly higher proportion of
display a more activated status. In addition, higher frequencies pNK cells expressing CCR7 and CXCR3 (Fig. 1D, p = 0.001 and
of intrahepatic NK (ihNK) cells have been described in PSC p = 0.003), as well as higher expression levels (MFI) of CCR7
livers with lower fibrotic status [31], and suggested to have and CXCR3 (Fig. 1D, p = 0.04 and p = 0.02), in PSC patients
anti-fibrotic properties by inducing apoptosis in profibrogenic compared to healthy control individuals. Also, a tendency toward
myofibroblasts [32–34]. However, the precise role of NK cells and higher proportions of CXCR3+ pNK cells in PSC patients when
their interactions with other immune cells in livers affected by PSC compared to PBC patients (p = 0.08) and to AIH patients (p
still remain unknown. The aim of this study was to characterize = 0.07) was observed. A more detailed boolean-gating analysis
peripheral blood NK (pNK) cells and ihNK cells from PSC patients showed a significantly higher frequency of pNK cells coexpressing
and compare these to NK cells derived from patients with other CCR7 and CXCR3 (Fig. 1E, p = 0.0003) and a significantly lower
inflammatory liver diseases or nondiseased control individuals, to frequency of CCR7− CXCR3− NK cells (Fig. 1E, p = 0.007) in indi-
identify changes in the NK-cell compartment associated with PSC viduals with PSC. Altogether, a phenotype of CCR7+ and CXCR3+
pathogenesis. pNK cells was observed in peripheral blood from PSC patients.


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
760 Annika E. Langeneckert et al. Eur. J. Immunol. 2019. 49: 758–769

Figure 1. Comparison of pNK cells from healthy control individuals, PSC, PBC, and AIH patients using flow cytometry. (A) NK cell frequencies
in peripheral blood from healthy control individuals, PSC, PBC, and AIH patients. (B) CD56bright and CD56dim distribution in pNK cells of healthy
control individuals, PSC, PBC, and AIH patients. (C) PMBCs from each group were gated for NK cells and concatenated. t-SNE plots show density and
the expression of CD56, CD16, CCR7, and CXCR3 on NK cells from healthy control individuals, PSC, PBC, and AIH patients. Color coding indicates
the density and marker expression. (D) Quantitative analysis of flow cytometry data of pNK cells expressing CCR7 and CXCR3 and the expression
levels on CCR7+ and CXCR3+ NK cells. (E) Boolean-gating showing all possible combinations of marker coexpression of CCR7 and CXCR3 on pNK
cells from healthy control individuals and PSC patients. For graphs (A–E): Data were pooled from five independent experiments with a maximum
of two donor samples for each group measured in one experiment; Mann–Whitney test was used for all statistical analyses and a p-value <0.05
was considered statistically significant; white circles: control (n = 10); black circles: PSC (n = 10); dark gray triangles: PBC (n = 10); light gray hexagon
AIH (n = 9); red lines indicate the median.


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
Eur. J. Immunol. 2019. 49: 758–769 Allergy and inflammation 761

Liver-derived NK cells from PSC patients exhibited in single CXCR3+ ihNK cells in PSC patients (Fig. 3C, p = 0.03).
higher expression of tissue-residency markers Of note, CCR7- as well as CXCR3-expression on ihNK cells did not
differ between CD56bright CD16dim and CD56dim CD16bright NK cell
To determine whether changes in peripheral blood NK cell sub- subsets (Supporting Information Fig. 3E).
sets were reflective of changes in ihNK cells, liver tissue from To assess possible functional differences between CCR7+ and
PSC patients undergoing liver transplantation and nontumorous CXCR3+ NK cells in PSC patients, we quantified degranulation
liver tissue from patients with liver metastases were analyzed (CD107a expression) and intracellular cytokine production of NK
using flow cytometry. Total ihNK cell frequencies and distribu- cells following stimulation with K562 cells [40]. In peripheral
tions of CD56bright CD16dim and CD56dim CD16bright NK cells did blood of PSC patients, CCR7+ and CXCR3+ NK cells showed a
not significantly differ between PSC patients and nondiseased con- significantly higher proportion of CD107a+ cells following stim-
trols (Supporting Information Fig. 3A–C). Further comparisons to ulation with K562 cells compared to CCR7− and CXCR3− NK
liver samples derived from individuals with alcoholic liver disease cells (Fig. 3D and E, p = 0.02, p = 0.007). In contrast, intra-
(ALD) and assessment of the influence of ulcerative colitis (UC) hepatic CCR7+ and CXCR3+ NK cells from PSC patients exhibited
as well as of treatment with ursodeoxycholic acid (UDCA) within a slightly lower proportion of CD107a+ cells compared to intra-
PSC patients did not show significant differences within NK cell hepatic CCR7− and CXCR3− NK cells (Fig. 3D and E, p = 0.12,
populations (Supporting Information Fig. 3A–C). Comparing NK p = 0.06). Furthermore, CXCR3− ihNK cells exhibited higher fre-
cells derived from peripheral blood and intrahepatic tissues using quencies of IFNγ+ cells compared to CXCR3+ ihNK cells (Fig.
t-SNE analysis, we observed that tissue-residency markers were 3E, p = 0.06), while no differences were observed for CCR7+
expressed to a higher extend on liver-derived NK cells (Fig. 2A), NK cell subsets (Fig. 3D and E). No significant differences were
in line with previous data [37–39]. Quantitative analysis revealed observed in the production of TNFα between NK cell subsets (Fig.
a significantly higher expression of CD16 (Fig. 2B, p = 0.02), of 3D and E), or in the functional capacities of CCR7+ /CXCR3+
the tissue-residency markers CD49a and CD69 (Fig. 2B, CD49a: and CCR7− /CXCR3− pNK cell and ihNK cell subsets from other
p = 0.01; CD69: p = 0.03), and a trend toward higher CXCR6- autoimmune diseases (Supporting Information Fig. 4). Finally,
expression (Fig. 2B, p = 0.06) on ihNK cells derived from PSC assessment of ADCC capability did not show any significant dif-
patients. Furthermore, ihNK cells also contained a significantly ferences between CCR7+ /CXCR3+ and CCR7− /CXCR3− NK cells
higher frequency of CD49a+ cells in PSC patients compared to from PSC patients, or between NK cells derived from PSC patients
control individuals (Fig. 2B, p = 0.01). Boolean-gating of tissue- and the other study groups (p > 0.05 for all comparisons, data not
residency markers expressed on ihNK cells showed a significantly shown). Taken together, ihNK cells from PSC patients exhibited
decreased population of NK cells lacking tissue residency markers higher proportions of CCR7+ NK cells, but lower levels of CXCR3-
(CD49− CD69− CXCR6− ) in PSC (Fig. 2C, p = 0.04). In summary, expression compared to nondiseased controls, and CCR7+ and
ihNK cells showed a significantly higher expression of liver res- CXCR3+ NK cells derived from peripheral blood of PSC patients
idency markers CD49a, CD69, and CXCR6 when comparing NK showed increased degranulation.
cells from PSC and from nondiseased control livers.

Increased plasma levels of CCL21, CXCL10, and


IhNK cells from PSC patients exhibited higher CXCL11, and increased intrahepatic levels of CCL21 in
expression of the chemokine receptor CCR7 PSC patients

IhNK cells from PSC patients were subsequently compared To determine whether changes in chemokines levels were associ-
to nondiseased control individuals to assess whether higher ated with observed changes in chemokine-receptor expression on
chemokine-receptor expression observed on pNK cells from PSC NK cells, we quantified levels of CCL21, CXCL10, and CXCL11 in
patients also occurred in liver tissues. Similar to peripheral blood, plasma samples of PSC patients, late stage PSC patients receiving
ihNK cells from PSC patients were significantly enriched for CCR7+ liver transplantation (lsPSC), PBC patients and AIH patients, as
NK cells (Fig. 3A and B, p = 0.009) although the MFI for CCR7 was well as healthy control individuals. Significant changes in plasma
similar in both groups (Fig. 3B). Interestingly, while the frequency chemokine levels were observed for CCL21 between lsPSC patients
of CXCR3+ ihNK cells did not differ between PSC patients and and healthy control individuals, and also between lsPSC patients
nondiseased control individuals (Fig. 3B, p = 0.4), CXCR3 expres- and AIH patients (Fig. 4A, p = 0.03 and p = 0.02). CXCL10 and
sion levels were significantly decreased in PSC (Fig. 3B, p = 0.01). CXCL11 plasma levels were furthermore significantly elevated in
In addition, the frequencies of CCR7+ and CXCR3+ ihNK cells were patients with early-stage PSC (Fig. 4A, p = 0.02 and p = 0.001)
also significantly higher in PSC patients compared to patients with and lsPSC (Fig. 4A, p = 0.003 and p = 0.04) compared to healthy
ALD (Supporting Information Fig. 3D, p = 0.02 and p = 0.02). control individuals. These data demonstrate that plasma levels of
No influence on CCR7 and CXCR3 expression was observed by the ligands for the chemokine receptors CCR7 and CXCR3 are elevated
presence of UC or treatment with UDCA within PSC patients (Sup- in patients at early and late stages of PSC disease. To investigate
porting Information Fig. 3D). Boolean-gating revealed an increase whether local chemokine expression in liver tissues might account
of single CCR7+ ihNK cells (Fig. 3C, p = 0.002) and a decrease for these observations, immunofluorescence staining of liver


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
762 Annika E. Langeneckert et al. Eur. J. Immunol. 2019. 49: 758–769

Figure 2. Tissue-residency marker expression in pNK cells and ihNK cells. (A) t-SNE Plots from one representative PSC patient and one represen-
tative nondiseased control show density and expression of CD56, CD16, CD49a, CD69, and CXCR6 on pNK cells and ihNK cells. (B) Quantitative
analysis of flow cytometry data from surface markers CD16, CD49a, CD69, and CXCR6 on ihNK cells from PSC patients and nondiseased controls.
(C) Results of boolean-gating represented in pie charts showing all possible combinations of marker coexpression of CD49a, CD69, and CXCR6 on
ihNK cells from PSC patients and nondiseased controls. For graphs B and C: data were pooled from five independent experiments in which two
donor samples for each group were measured in one experiment; medians were used for boolean-gating analyses; Mann–Whitney test was used
for statistical analyses and a p-value <0.05 was considered statistically significant; white circles: control (n = 10); black circles: PSC (n = 10); red
lines indicate the median.


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
Eur. J. Immunol. 2019. 49: 758–769 Allergy and inflammation 763

Figure 3. Chemokine receptor expression on ihNK cells from nondiseased controls and PSC patients. (A) t-SNE plots from one representative
individual showing density and expression of CCR7 and CXCR3 on ihNK cells. (B) Proportions of CCR7+ and CXCR3+ ihNK cells and MFI of CCR7+
and CXCR3+ NK cells from PSC patients and nondiseased controls. (C) Boolean-gating of CCR7 and CXCR3 on ihNK cells. For graphs (A–C): Data were
pooled from five independent experiments in which two donor samples per group were measured in one experiment; Mann–Whitney test was
used for statistical analyses and a p-value <0.05 was considered statistically significant; White circles: nondiseased control (n = 10); black circles:
PSC (n = 10); Red lines indicate the median. Plots representing CD107a, IFNγ, and TNFα expression in CCR7+ (red) and CCR7− (black) pNK cells and
ihNK cells (D) or in CXCR3+ (red) and CXCR3− (black) pNK cells and ihNK cells (E) from PSC patients upon coincubation with K562 cells. For graphs
(D and E): pNK cells (n = 8); ihNK cells (n = 5); data were pooled from two independent experiments with four donor samples per group measured
in one experiment for pNK cells in graph D; for ihNK cells in graph E, data were pooled from two independent experiments with maximum of
three samples per group measured in one experiment; cells incubated without target cells were used as controls; values were normalized to
unstimulated control; Wilcoxon signed rank test was used for statistical analyses and a p-value <0.05 was considered as statistically significant.


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
764 Annika E. Langeneckert et al. Eur. J. Immunol. 2019. 49: 758–769

Figure 4. Chemokine concentration levels in plasma, CCL21, and CXCL10 expression in liver sections and migration toward CCL21. (A) Plasma
concentration level of CCL21, CXCL10, and CXCL11 from nondiseased controls, PSC patients, lsPSC patients, PBC patients, and AIH patients was
determined by a Luminex assay. White circles: nondiseased control (n = 8); black circles: PSC (n = 8); gray squares: lsPSC (n = 8); dark gray triangles:
PBC (n = 7); light gray hexagon: AIH (n = 7). Red lines indicate the median. Mann–Whitney test was used for statistical analyses and a p-value
<0.05 was considered as statistically significant. Data were pooled from one experiment with two technical replicates per donor measured in one
experiment. (B) Dual immunostaining of cryosections from one representative PSC patient and one representative nondiseased control individual
with anti-CCL21, donkey anti-goat NL557-conjugated antibody and DAPI. Plot shows quantitative analysis of CCL21 expression in % of total cells.
(C) Dual immunostaining of cryosections from one PSC patient and one nondiseased control individual with anti-CXCL10, donkey anti-goat NL557-
conjugated antibody, and DAPI. Plot shows quantitative analysis of CXCL10 expression in % of total cells. For B and C: scale bar is set to 30 μm;
a Nikon Ti-E with Yokogawa W1 Spinning Disk, an Andor 888 camera, an objective 100 × 1.49 NA and Nikon NIS software was used for analysis;
further analyses were assessed using Fiji software; data were pooled from three different experiments using three or more independent images
per donor; for the analysis of assays using replicate images linear-mixed effect regression models with a random intercept were used to take into
account the intraparticipant correlation of the repeated measurements; a p-value <0.05 was considered as statistically significant. (D) Quantitative
analysis of flow cytometry data from transmigration assays using isolated pNK cells from control individuals (n = 5, white circles) and PSC patients
(n = 5, black circles) migrating toward medium containing CCL21. RP10 medium without CCL21 served as control. Wilcoxon signed rank test was
used for statistical analyses. A p-value <0.05 was considered as statistically significant. Data were pooled from three independent experiments
with a maximum of two measured samples in one group and two technical replicates per donor measured in one experiment.


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
Eur. J. Immunol. 2019. 49: 758–769 Allergy and inflammation 765

tissue cryosections from PSC patients and nondiseased control diseases [46]. While the cross-sectional design of our study did
individuals was performed using CCL21 and CXCL10 antibodies. not allow us to determine whether CCR7- and CXCR3-expression
As shown in Fig. 4B, the CCR7-ligand CCL21 was significantly on NK cells occurred before or as a consequence of liver inflam-
higher expressed in liver sections from PSC patients compared mation, these data suggest a role of these chemokine receptors in
to nondiseased control livers (p = 0.006), while CXCL10 was the progressive pathogenesis of PSC.
expressed to a slightly lower extend on liver sections from PSC Studies of peripheral blood immune cells provide limited infor-
patients (Fig. 4C, p = 0.33). To begin to assess whether elevated mation into tissue-specific inflammatory processes. Only few stud-
CCL21 expression in liver tissues might attract CCR7+ NK cells ies have characterized ihNK cells in PSC. Berglin et al. observed
to the liver, we performed a NK cell migration assay using trans- scattered NK cells preferentially located within the parenchyma
well experiments. NK cells isolated from PBMCs from PSC patients distant from fibrotic areas of PSC livers using immunohistochem-
showed enhanced migration toward the chemokine CCL21 com- istry, and no increased ihNK cell numbers compared to normal
pared to control medium without CCL21 (Fig. 4D, p = 0.06), while controls [31]. In contrast, another study showed lower numbers
there was no difference in the migration of NK cells from control of ihNK cells in PSC [17]. In this study, we observed no significant
individuals (Fig. 4D, p = 0.19). Taken together, these data sug- differences in ihNK cell numbers and subsets (CD56bright CD16dim ,
gest that local production of the CCR7-ligand CCL21 may result in CD56dim CD16bright ) between PSC and nondiseased control individ-
recruitment of CCR7+ NK cells into liver tissues of patients with uals using multi-parameter flow cytometry, but observed higher
PSC. frequencies of liver-resident NK cells in PSC livers. While the use
of nontumorous liver tissue from liver resections due to hepatic
metastases as nondiseased controls has caveats, as liver-resident
Discussion lymphocyte populations can be affected by the underlying diseases
and treatments, the accumulation of NK cells expressing markers
PSC is a chronic progressive inflammatory disease characterized of liver residency appears to be a common feature of inflammatory
by the infiltration of liver portal areas with immune cells [15, 17, liver diseases [38, 47, 48]. We furthermore observed a significant
20]. Previous studies have shown an increased expression of the higher frequency of CCR7+ NK cells in livers, but not higher fre-
chemokine receptors CCR7 and CXCR3 on peripheral blood and quencies of CXCR3+ NK cells. In contrast to peripheral blood,
intrahepatic T cells during PSC and PBC [21, 35], as well as an CXCR3-expression on NK cells in PSC livers was reduced, possibly
accumulation of the CCR7 ligand CCL21 within portal areas of PSC suggesting a ligand-induced downregulation of this receptor. As
livers [21] and the CXCR3 ligands CXCL9 and CXCL10 in portal CXCR3 is essential for cellular trans-migration from hepatic sinu-
tracts of PBC livers [35]. NK cells express chemokine receptors soids into the liver parenchyma [49], expression of CXCR3 might
that can attract them to sites of tissue inflammation, but the role be expendable for ihNK cells. Although the expression of CCR7 has
of NK cells in the pathogenesis of PSC remains unknown. Here, we been implicated into the cellular egress from tissue into lymphatic
describe higher proportions of CCR7+ NK cells in peripheral blood vessels [50], CCR7+ NK cell egress from livers of PSC patients
and livers of PSC patients that were associated with elevated levels might be prevented due to expression of CCL21 within hepatic
of the CCR7-ligand CCL21, suggesting that CCL21-expression and portal fields. Taken together, these data suggest that expression
infiltration of CCR7+ NK cells might contribute to progressive liver of liver-homing receptors and chemokine receptors contributes to
inflammation observed in PSC. the accumulation of CCR7+ ihNK cells in PSC patients.
Several studies of chronic progressive liver inflammation Chemokines play a critical role in the recruitment of lympho-
have assessed changes in lymphocyte populations in periph- cytes to inflamed tissues. We detected increased plasma levels of
eral blood of PSC patients, and shown elevated T-cell num- CCL21 (CCR7-ligand) in PSC patients compared to healthy con-
bers [21] and increased NK cell frequencies [17, 41]. Com- trol individuals and patients with other autoimmune liver dis-
paring pNK cells between healthy control individuals and indi- eases, suggesting specific upregulation of this chemokine during
viduals with PSC, PBC, and AIH, we observed a significantly late stages of PSC disease. CCL21 is usually expressed on high
increased CD56bright CD16dim pNK cell population in PSC patients, endothelial venules that have been observed in inflammatory liver
but no changes in overall NK cell numbers. This might be due diseases like PSC, and a subset of CD11c+ cells in portal tracts of
to increased inflammation, as previously suggested [42–45]. PSC PSC livers have been identified as a major source of CCL21 [21].
patients exhibited higher frequencies of CCR7+ and CXCR3+ NK Previous studies showed that isolated blood lymphocytes and
cells in the peripheral blood, and higher surface expression of intrahepatic lymphocytes (IHLs) from PSC patients responded to
these two chemokine receptors. Our data are in line with previous CCL21 in micro chemotaxis chamber assays in vitro, but these
studies demonstrating an increased expression of these chemokine studies used total lymphocyte populations [21]. In this study, we
receptors on CD4+ T cells of PSC patients [21, 35]. CCR7 is mainly observed that isolated pNK cells from PSC patients but not from
expressed on CD56bright CD16dim NK cells, enabling these cells to healthy control individuals exhibit a trend toward higher migra-
respond to CCL21 [45], and has been shown to be required for the tion toward CCL21. We furthermore observed that CCL21 was
trafficking to lymph nodes [45]. CXCR3 serves as a receptor for significantly higher expressed in liver sections from PSC patients
several inflammatory chemokines (CXCL9, CXCL10, and CXCL11) compared to nondiseased control livers, in line with a previous
and has been described to be upregulated in inflammatory liver study in PSC [21], further supporting a role of this chemokine in


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
766 Annika E. Langeneckert et al. Eur. J. Immunol. 2019. 49: 758–769

NK cell recruitment during PSC. Ligands for CXCR3 have also been control donors enrolled into the Hamburger Gesundkohorte (n
shown to be overexpressed in inflammatory liver diseases, which = 10). Liver tissue samples were collected from patients under-
can lead to an infiltration of CXCR3-expressing cells [35, 36]. We going liver transplantation in the Department of Hepatobiliary
indeed detected significantly elevated plasma levels of the CXCR3- and Transplant Surgery of the UKE (PSC n = 10; ALD n = 11;
ligands CXCL10 and CXCL11 in PSC patients. However, we did PBC n = 2; AIH n = 2 and PBC/AIH overlap n = 1) or individ-
not detect elevated CXCL10-expression in PSC livers, but rather uals undergoing liver resection due to liver tumor metastases in
lower expression than in nondiseased livers. We cannot exclude the Department of General and Visceral Surgery at the Asklepios
that other CXCR3-ligands, such as CXCL11 or CXCL9, might have Clinic Hamburg-Barmbek (nondiseased n = 10). Characteristics
been upregulated in liver tissues and triggered the downregula- of study participants are summarized in Supporting Information
tion of CXCR3-expression observed on liver-resident NK cells in Table 1 and Supporting Information Table 2.
PSC livers.
Very little is known regarding the functional capacity of NK
cells in inflammatory liver diseases. We analyzed the ability of Ethics
pNK cells and ihNK cells to degranulate following stimulation with
MHC-devoid K562 cells. Peripheral blood CCR7+ and CXCR3+ All study subjects provided informed written consent according to
NK cells from PSC patients both exhibited higher proportions of study protocols approved by the Ärztekammer Hamburg (PV4898,
CD107a+ NK cells, suggesting that CCR7+ and CXCR3+ NK cells PV4081, PV4780).
might contribute to the inflammation observed during PSC. Fur-
thermore, comparing CCR7+ /CXCR3+ and CCR7− /CXCR3− pNK
cells and ihNK cells from patients with AIH and PBC for their Isolation of lymphocytes from peripheral blood and
ability to degranulate and to produce cytokines did not show any liver tissue
significant differences, indicating that the differences observed
in NK cell function were most pronounced in patients with PSC. PMBCs were isolated from peripheral blood using standard den-
In contrast to these observation in peripheral blood, CCR7+ and sity gradient centrifugation (Ficoll, Biochrom GmbH, Berlin,
CXCR3+ ihNK cells exhibited less activity against K562 cells than Germany). Cells were cryopreserved in 90%fetal bovine serum
CCR7− and CXCR3− NK cells. These data are in line with a previ- (FBS)/10%DMSO and stored in liquid nitrogen. IHLs were
ous study reporting no cytotoxicity of NK cells against K562 cells isolated as previously described [51] and cryopreserved in
in PSC [17]. The inability of intrahepatic CCR7+ NK cells as well 90%FBS/10%DMSO in liquid nitrogen.
as the overall ihNK cell population from PSC patients to strongly
react to additional target cell stimulation might be due to increased
baseline activation, as we observed a higher baseline of intracellu- Flow cytometry
lar cytokine levels in CCR7+ ihNK cells compared to CCR7− ihNK
cells and also CCR7+ pNK cells. Additional studies will be required Cryopreserved PBMC and liver samples were thawed, washed with
to determine whether interventions targeting these chemokine- PBS and stained with surface antibodies and live dead marker
receptor-expressing NK cells accumulating in inflamed livers might Zombie AquaTM (BioLegend, San Diego, CA, USA) for 20 min at
help to prevent the progressive inflammation that is a character- RT in the dark, fixed in 4%PFA/PBS and subsequently analyzed
istic of PSC. Taken together, our data suggest liver recruitment of using a LSR FortessaTM (BD Bioscience, San Jose, CA, USA). A
highly functional CCR7+ NK cells associated with elevated CCL21- list of antibodies used for staining of immune cells is provided in
expression within PSC livers, indicating that T and NK cells might Supporting Information Table 3. For flow cytometry analysis, we
play in concert during inflammatory processes that lead to the adhered to the “Guidelines for the use of flow cytometry and cell
progressive destruction of the bile ducts. These data provide a sorting in immunological studies” [52].
rational for the targeting of lymphocyte trafficking and homing
into inflamed liver tissues as a strategy to disrupt the detrimental
inflammatory cycle that is characteristic of PSC pathogenesis. Immunohistochemistry and fluorescence microscopy

Liver tissues from PSC patients and patients undergoing liver


resection were snap-frozen at –80°C. Tissues were mounted and
Materials and methods cut into 8 μm sections at –10°C using a cryostat (CM3050, Leica,
Wetzlar, Germany). Slides were air-dried and stored at –80°C.
Patient samples Liver tissue was fixed with 4%PFA/PBS for 20 min at 4°C and per-
meabilized with 0.1%TritonX100/D for 5 min at RT. Slides were
A total of 75 study subjects were included in this study. Peripheral blocked with 1%donkey serum/PBS for 1 h at RT. Anti-CCL21
blood samples were derived from individuals diagnosed with PSC and anti-CXCL10 were applied as first antibodies overnight at
(n = 10), PBC (n = 10), and AIH (n = 9) recruited at the Uni- 4°C. Control slides were incubated without the aforementioned
versity Medical Center Hamburg-Eppendorf (UKE), and healthy antibodies (Supporting Information Fig. 5). Secondary antibodies


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
Eur. J. Immunol. 2019. 49: 758–769 Allergy and inflammation 767

were applied for 60 min at RT. Slides were then stained with cytometry data was done using viSNE (Cytobank, Inc., Santa Clara,
DAPI (Thermo Fisher Scientific, Waltham, MA, USA) for 5 min at CA, USA). Plots were generated and statistical comparisons per-
RT and mounted with Fluorescence Mounting Medium (Agilent, formed in Prism7 (GraphPad Sofware, La Jolla, CA, USA). For
Santa Clara, CA, USA). Samples were visualized using fluores- comparisons between two groups Mann–Whitney test was used.
cence microscopy (Nikon Ti-E with Yokogawa W1 Spinning Disk, For comparisons between two paired groups Wilcoxon signed rank
Andor 888 camera, objective 100 × 1.49 NA, Nikon NIS software). test was used. For all tests, p < 0.05 was considered statistically
significant. Significant and close to significant p values are shown
in all figures. Medians are shown as red lines in all Figures, in
Chemokine quantification in plasma samples Supporting Information Figures median is shown in black. Due
to limited sample sizes from different patient cohorts no techni-
Plasma samples from PSC, PBC, and AIH patients as well as healthy cal or biological replicates were measured except for data in Fig.
control donors and PSC patients undergoing liver transplantation 4A and D. The analysis of fluorescence microscopy images was
(late stage PSC) were stored at –80°C. Plasma concentrations of carried out using Fiji software. For the analysis of assays using
CCL21, CXCL10, and CXCL11 were quantified using a Bio-Plex Pro replicate images, linear-mixed effect regression models with a ran-
Human Cytokine Kit, a Bio-Plex200 analyzer, and Bioplex Analysis dom intercept were used to take into account the intraparticipant
Software (Biorad Laboratories, Herkules, CA, USA). correlation of the repeated measurements.

NK cell degranulation and ADCC assay

Cryopreserved PBMCs (n = 8) and IHLs (n = 5) from PSC patients


were thawed and rested overnight in R10 supplemented with Acknowledgments: The authors thank all the donors from the
5 ng/mL IL-15. Cells were cocultured with K562 cells (DSMZ) LTX cohort at the University Clinical Center Hamburg-Eppendorf
and Raji cells (in the presence of Rituximab, Invivogen, Toulouse, (UKE), the AKB cohort at the Asklepios Hospital Barmbek (AKB)
France) at an effector to target (E:T) ratio of 5:1 in the presence and the healthy donor cohort at the Heinrich Pette Institute (HPI)
of anti-CD107a, as previously described [40]. After 1 h incuba- in Hamburg for their participation in our study. Furthermore, the
tion at 37°C, Brefeldin A (Sigma-Aldrich, Taufkirchen, Germany) authors would like to thank all nurses and surgeons of the Depart-
and Monensin (BD Bioscience, San Jose, CA, USA) were added ment of Hepatobiliary and Transplant Surgery (UKE) and of the
followed by four additional hours of incubation. Cells were sub- Department of General and Abdominal Surgery (AKB) for their
sequently stained with surface antibodies and live-dead marker involvement. M.A., J.H., C.S., G.T., and S.L. received funding from
Zombie AquaTM (Biolegend, San Diego, CA, USA) for 20 min at the SFB841 of the Deutsche Forschungsgesellschaft. C.S. is funded
RT. For intracellular cytokine staining a Cytofix/Cytoperm kit (BD by the Helmut and Hannelore Greve Foundation. A.L., M.A., C.S.,
Bioscience, San Jose, CA, USA), anti-IFNγ and anti-TNFα (Biole- J.H., G.T., T.P., and G.R. are supported by the CRU306 of the
gend, San Diego, CA, USA) was used. Samples were then analyzed Deutsche Forschungsgesellschaft. L.H. and A.Z. received financial
by flow cytometry. support from the graduate school of the SFB841. The funders had
no influence on study design, data collection, and analysis, the
decision to publish or contents of the manuscript. A.L., S.L., M.A.
NK cell migration assay designed the study. A.L., G.M., L.H., G.R., U.M., W.S., T.P., S.L.
performed experiments. A.L. analyzed the data. A.L. and M.A.
PBMCs from control individuals and PSC patients were thawed wrote the manuscript. M.K., K.O., L.F. provided patient samples.
and cultured with IL15 (5 ng/mL) in RPMI medium with 10% All authors critically reviewed the manuscript.
FCS for 2 h at 37°C. NK cells were isolated and migration toward
the chemokine CCL21 (10 ng/mL) was assessed using a transwell
system with a 5.0 μm pore polycarbonate membrane (Fisher Sci-
Conflict of interest: J.B.B. is a consultant of Crescenta Bio-
entific GmbH, Schwerte, Germany). NK cells with a concentration
sciences, NJ, USA and holds options in this company. All
of 6,7 cells/mL in RP10 were incubated for 4 h at 37°C and the
other authors declare no commercial or financial conflict of
amount of migrated cells was determined using CountBrightTM
interest.
Absolute Counting Beads (Fisher Scientific GmbH, Schwerte,
Germany).

References
Statistical analyses
1 Chapman, R., Fevery, J., Kalloo, A., Nagorney, D. M., Boberg, K. M., Shnei-
der, B. and Gores, G. J., AASLD PRACTICE GUIDELINES. Diagnosis and
Flow cytometry data were analyzed using FlowJoTM Version management of primary sclerosing cholangitis. Hepatology 2010; 51: 660–
10.4.2. (Ashland, OR, USA). Additional visualization of flow 678.


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
768 Annika E. Langeneckert et al. Eur. J. Immunol. 2019. 49: 758–769

2 Boonstra, K., Van Erpecum, K. J., Van Nieuwkerk, K. M. J., Drenth, J. P. mediated positioning of Th-17 cells in the inflamed liver. J. Hepatol. 2012.
H., Poen, A. C., Witteman, B. J. M., Tuynman, H. A. R. E. et al., Primary 57: 1044–1051.
sclerosing cholangitis is associated with a distinct phenotype of inflam-
19 Kamihira, T., Shimoda, S., Nakamura, M., Yokoyama, T., Takii, Y.,
matory bowel disease. Inflam. Bowel Dis. 2012: 1–7.
Kawano, A., Handa, M. et al., Biliary epithelial cells regulate autoreac-
3 Hirschfield, G. M., Karlsen, T. H., Lindor, K. D. and Adams, D. H., Primary tive T cells: implications for biliary-specific diseases. Hepatology 2005. 41:
sclerosing cholangitis. Lancet. 2013. 382: 1587–1599. 151–159.

4 O’Toole, A., Alakkari, A., Keegan, D., Doherty, G., Mulcahy, H. and 20 Eksteen, B., Grant, A. J., Miles, A., Curbishley, S. M., Lalor, P. F., Hübscher,
O’Donoghue, D., Primary sclerosing cholangitis and disease distribution S. G., Briskin, M. et al., Hepatic endothelial CCL25 mediates the recruit-
in inflammatory bowel disease. Clin. Gastroenterol. Hepatol. 2012. 10: 439– ment of CCR9 + gut-homing lymphocytes to the liver in primary scleros-
441. ing cholangitis. J. Exp. Med. 2004. 200: 1511–1517.

5 Chapman, M. H., Webster, G. J. M., Bannoo, S., Johnson, G., Wittmann, 21 Grant, A. J., Goddard, S., Ahmed-Choudhury, J., Reynolds, G., Jackson, D.
J. and Pereira, S. P., Cholangiocarcinoma and dominant strictures in G., Briskin, M., Wu, L. et al., Hepatic expression of secondary lymphoid
patients with primary sclerosing cholangitis: a 25-year single-centre chemokine (CCL21) promotes the development of portal-associated lym-
experience. Eur. J. Gastroenterol. Hepatol. 2012. 24: 1051–1058. phoid tissue in chronic inflammatory liver disease. Am. J. Pathol. 2002.
160: 1445–1455.
6 Hildebrand, T., Pannicke, N., Dechene, A., Gotthardt, D. N., Kirchner, G.,
Reiter, F. P., Sterneck, M. et al., Biliary strictures and recurrence after 22 Vivier, E., Tomasello, E., Baratin, M., Walzer, T. and Ugolini, S., Functions
liver transplantation for primary sclerosing cholangitis: a retrospective of natural killer cells. Nat. Immunol. 2008. 9: 503–510.
multicenter analysis. Liver Transplant. 2016. 22: 42–52.
23 Vivier, E., Raulet, D., Moretta, A. and Caligiuri, M., Innate or adaptive
7 Melum, E., Franke, A., Schramm, C. and Weismüller, T. J., Genome-wide immunity? The example of natural killer cells. Science 2011. 331: 44–49.
association analysis in primary sclerosing cholangitis identifies two non-
24 Schleinitz, N., Vély, F., Harlé, J. R. and Vivier, E., Natural killer cells in
HLA susceptibility loci. Nat. Genet. 2015. 43: 17–19.
human autoimmune diseases. Immunology 2010. 131: 451–458.
8 Næss, S., Lie, B. A., Melum, E., Olsson, M., Hov, J. R., Croucher, P. J.
25 Jiao, G. and Wang, B., NK cell subtypes as regulators of autoimmune liver
P., Hampe, J. et al., Refinement of the MHC risk map in a Scandinavian
disease. Gastroenterol. Res. Pract. 2016. 2016: 1–6.
primary sclerosing cholangitis population. PLoS One. 2014. 9: 1–19.
26 Villard, J., The role of natural killer cells in human solid organ and tissue
9 Whiteside, T. L., Lasky, S., Si, L. and Van Thiel, D. H., Immunologic
transplantation. J. Innate Immun. 2011. 3: 395–402.
analysis of mononuclear cells in liver tissues and blood of patients with
primary sclerosing cholangitis. Hepatology 1985. 5: 468–474. 27 Racanelli V. and Rehermann B., The liver as an immunological organ.
Hepatology 2006. 43: S54–S62.
10 Ponsioen, C. Y., Etiologic and clinical studies in primary sclerosing
cholangitis (chapter IV Immunohistochemical analysis of inflammation 28 Karlsen, T. H., Boberg, K. M., Olsson, M., Sun, J. Y., Senitzer, D., Bergquist,
in primary sclerosing cholangitis. Eur. J. Gastroenterol. Hepato. 1999. 11: A., Schrumpf, E. et al., Particular genetic variants of ligands for natural
769–774. killer cell receptors may contribute to the HLA associated risk of primary
sclerosing cholangitis. J. Hepatol. 2007. 46: 899–906.
11 Björnsson, E., Cederborg, A., Åkvist, A., Simren, M., Stotzer, P. O. and
Bjarnason, I., Intestinal permeability and bacterial growth of the small 29 Wiencke, K., Spurkland, A., Schrumpf, E. and Boberg, K. M., Pri-
bowel in patients with primary sclerosing cholangitis. Scand. J. Gastroen- mary sclerosing cholangitis is associated to an extended B8-DR3 hap-
terol. 2005. 40: 1090–1094. lotype including particular MICA and MICB alleles. Hepatology 2001. 34:
625–630.
12 Katt, J., Schwinge, D., Schoknecht, T., Quaas, A., Sobottka, I., Burandt,
E., Becker, C. et al., Increased T helper type 17 response to pathogen 30 Pollheimer, M. J., Halilbasic, E., Fickert, P. and Trauner, M., Pathogenesis
stimulation in patients with primary sclerosing cholangitis. Hepatology of primary sclerosing cholangitis. Best Pract. Res. Clin. Gastroenterol. 2011.
2013. 58: 1084–1093. 25: 727–739.

13 Sartor, R. B., Microbial influences in inflammatory bowel diseases. Gas- 31 Berglin, L., Bergquist, A., Johansson, H., Glaumann, H., Jorns, C., Lune-
troenterology 2008. 134: 577–594. mann, S., Wedemeyer, H. et al., In situ characterization of intrahepatic
non-parenchymal cells in PSC reveals phenotypic patterns associated
14 Kulaksiz, H., Rudolph, G., Kloeters-Plachky, P., Sauer, P., Geiss, H. and
with disease severity. PLoS One 2014. 9: 1–11.
Stiehl, A., Biliary candida infections in primary sclerosing cholangitis. J.
Hepatol. 2006. 45: 711–716. 32 Melhem, A., Muhanna, N., Bishara, A., Alvarez, C. E., Ilan, Y., Bishara,
T., Horani, A. et al., Anti-fibrotic activity of NK cells in experimental liver
15 Hashimoto, E., Lindor, K. D., Homburger, H. A., Dickson, E. R., Czaja,
injury through killing of activated HSC. J. Hepatol. 2006. 45: 60–71.
A. J. and Wiesner, R. H. L. J., Immunohistochemical characterization
of hepatic lymphocytes in primary biliary cirrhosis in comparison with 33 Radaeva, S., Sun, R., Jaruga, B., Nguyen, V. T. and Tian, Z., Gao, B.,
primary sclerosing cholangitis and autoimmune chronic active hepatitis. Natural killer cells ameliorate liver fibrosis by killing activated stellate
Mayo Clin. Proc. 1993. 68: 1049–1055. cells in NKG2D-dependent and tumor necrosis factor-related apoptosis-
inducing ligand-dependent manners. Gastroenterology 2006. 130:
16 Senaldi, G., Portmann, B., Mowat, A. P., Mieli-Vergani G. and Vergani
435–452.
D., Immunohistochemical features of the portal tract mononuclear cell
infiltrate in chronic aggressive hepatitis. Arch. Dis. Child. 1992. 67: 1447– 34 Glässner, A., Eisenhardt, M., Krämer, B., Körner, C., Coenen, M., Sauer-
1453. bruch, T., Spengler, U. et al., NK cells from HCV-infected patients effec-
tively induce apoptosis of activated primary human hepatic stellate cells
17 Bo, X., Broome, U., Remberger, M. and Sumitran-Holgersson, S., Tumour
in a TRAIL-, FasL-and NKG2D-dependent manner. Lab. Investig. 2012. 92:
necrosis factor alpha impairs function of liver derived T lymphocytes
967–977.
and natural killer cells in patients with primary sclerosing cholangitis.
Gut. 2001. 49: 131–41. 35 Chuang, Y. H., Lian, Z. X., Cheng, C. M., Lan, R. Y., Yang, G. X., Moritoki,
Y., Chiang, B. L. et al., Increased levels of chemokine receptor CXCR3 and
18 Oo, Y. H., Banz, V., Kavanagh, D., Liaskou, E., Withers, D. R., Humphreys,
chemokines IP-10 and MIG in patients with primary biliary cirrhosis and
E., Reynolds, G. M. et al., CXCR3-dependent recruitment and CCR6-
their first degree relatives. J. Autoimmun. 2005. 25: 126–132.


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
Eur. J. Immunol. 2019. 49: 758–769 Allergy and inflammation 769

36 Nishioji, K., Okanoue, T., Itoh, Y., Narumi, S., Sakamoto, M., Nakamura, carcinoma are associated with tumor progression. J. Gastroenterol. Hepatol.
H., Morita, A. et al., Increase of chemokine interferon-inducible protein- 2011. 26: 1519–1526.
10 (IP-10) in the serum of patients with autoimmune liver diseases and
48 Wehr, A., Baeck, C., Heymann, F., Niemietz, P. M., Hammerich, L., Martin,
increase of its mRNA expression in hepatocytes. Clin. Exp. Immunol. 2001.
C., Zimmermann, H. W. et al., Chemokine receptor CXCR6-dependent
123: 271–279.
hepatic NK T cell accumulation promotes inflammation and liver fibrosis.
37 Stegmann, K. A., Robertson, F., Hansi, N., Gill, U., Pallant, C., J. Immunol. 2013. 190: 5226–5236.
Christophides, T., Pallett, L. J. et al., CXCR6 marks a novel subset of T-bet
49 Curbishley, S. M., Eksteen, B., Gladue, R. P., Lalor, P. and Adams, D.
lo Eomes hi natural killer cells residing in human liver. Sci. Rep. 2016. 6:
H., CXCR3 activation promotes lymphocyte transendothelial migration
1–10.
across human hepatic endothelium under fluid flow. Am. J. Pathol. 2005.
38 Martrus, G., Kautz, T., Lunemann, S., Richert, L., Glau, L., 167: 887–899.
Salzberger, W., Goebels, H. et al., Proliferative capacity exhibited
50 Bromley, S. K., Thomas, S. Y. and Luster, A. D., Chemokine receptor
by human liver-resident CD49a+CD25+ NK cells. PLoS One. 2017. 12:
CCR7 guides T cell exit from peripheral tissues and entry into afferent
e0182532.
lymphatics. Nat. Immunol. 2005. 6: 895–901.
39 Marquardt, N., Béziat, V., Nyström, S., Hengst, J., Ivarsson, M. A.,
51 Salzberger, W., Martrus, G., Bachmann, K., Goebels, H., Heß, L., Koch, M.,
Kekäläinen, E., Johansson, H. et al., Cutting edge: identification and char-
Langeneckert, A. et al., Tissue-resident NK cells differ in their expression
acterization of human intrahepatic CD49a + NK Cells. J. Immunol. 2015.
profile of the nutrient transporters Glut1, CD98 and CD71. PLoS One. 2018.
194: 2467–2471.
13: 1–19.
40 Alter, G., Malenfant, J. M. and Altfeld, M., CD107a as a functional marker
52 Cossarizza, A., Chang, H., Radbruch, A., Andr, I., Martin, B., Garc, M. D.
for the identification of natural killer cell activity. J. Immunol. Methods
and Gr, A., Guidelines for the use of flow cytometry and cell sorting in
2004. 294: 15–22.
immunological studies. Eur. J. Immunol. 2017. 47: 1584–1797.
41 Chuang, Y. H., Lian, Z. X., Tsuneyama, K., Chiang, B. L., Ansari, A.
A., Coppel, R. L. and Eric Gershwin, M., Increased killing activity and
decreased cytokine production in NK cells in patients with primary bil- Abbreviations: ALD: alcoholic liver disease · AIH: autoimmune hepatitis
iary cirrhosis. J. Autoimmun. 2006. 26: 232–240.
· CCR7: C-C chemokine receptor type 7 · CXCR3: C-X-C motif chemokine
42 Robertson, M. J., Role of chemokines in the biology of natural killer cells. receptor 3 · CXCL10: C-X-C motif chemokine 10 · CXCL11: C-X-C motif
J. Leukoc. Biol. 2002. 71: 173–183.
chemokine 11 · CXCL9: C-X-C motif chemokine 9 · CCL21: C-C motif
43 Fehniger, T. A., Cooper, M. A., Nuovo, G. J., Cella, M., Facchetti, F., chemokine ligand 21 · FBS: fetal bovine serum · IHL: intrahepatic lym-
Colonna, M. and Caligiuri, M. A., CD56 bright natural killer cells are
phocytes · KIR: killer-cell immunoglobulin-like receptor · pNK cells:
present in human lymph nodes and are activated by T cell-derived IL-2:
peripheral blood natural killer cells · ihNK cells: intrahepatic natural
a potential new link between adaptive and innate immunity. Blood 2003.
101: 3052–3057. killer cells · PSC: primary sclerosing cholangitis · PBC: primary bil-
iary cirrhosis · t-SNE: t-distributed stochastic neighbor embedding · UC:
44 Dalbeth, N., Gundle, R., Davies, R. J. O., Lee, Y. C. G., McMichael, A. J. and
Callan, M. F. C., CD56 bright
NK cells are enriched at inflammatory sites ulcerative colitis · UDCA: ursodeoxycholic acid
and can engage with monocytes in a reciprocal program of activation. J.
Immunol. 2004. 173:6418–6426. Full correspondence: Prof. Marcus Altfeld, Research Department of Virus
Immunology, Heinrich Pette Institute, Martinistraße 52, 20251
45 Mailliard, R. B., Alber, S. M., Shen, H., Watkins, S. C., Kirkwood, J. M.,
Hamburg, Germany
Herberman, R. B. and Kalinski, P., IL-18–induced CD83 + CCR7 + NK
e-mail: marcus.altfeld@leibniz-hpi.de
helper cells. J. Exp. Med. 2005. 202: 941–953.

46 Shields P. L., Morland C. M., Salmon M., Qin S., Hubscher S. G. and The peer review history for this article is available at
Adams D. H., Chemokine and chemokine receptor interactions pro- https://publons.com/publon/10.1002/eji.201847965
vide a mechanism for selective T cell recruitment to specific liver
compartments within hepatitis C-infected liver. J. Immunol. 1999. 163:
Received: 11/10/2018
6236–6243.
Revised: 22/1/2019
47 Zhu, J., Feng, A., Sun, J., Jiang, Z., Zhang, G., Wang, K., Hu, S. et al., Accepted: 19/2/2019
Increased CD4(+) CD69(+) CD25(-) T cells in patients with hepatocellular Accepted article online: 20/2/2019


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu

You might also like