ArticlePDF Available

Analysis of serum markers of cellular immune activation in patients with bullous pemphigoid

Authors:

Abstract and Figures

Experimental models of bullous pemphigoid (BP), the most frequent subepidermal autoimmune bullous disease, revealed that the immune response leading to blister formation represents an incompletely understood complex process involving different inflammatory cells. In contrast to previous reports commonly focusing on limited molecular and cellular phenotypes of the disease, the aim of this study was to investigate a broad spectrum of markers of cellular immune activation in patients with BP. We found that serum levels of soluble CD4, myeloperoxidase, S100A12, eosinophil cationic protein, and soluble P-selectin were significantly elevated in patients with active BP compared with normal controls. Mast cell tryptase and neopterin serum levels significantly decreased at the time of clinical remission of the patients. Additionally, serum concentrations of soluble IL-2 receptor, mast cell tryptase, and soluble P-selectin were significantly associated with levels of circulating anti-BP180 autoantibodies. Our findings confirm and extend previous reports suggesting some concomitant involvement of a panel of molecules representative for a wide spectrum of cellular players (T cells, mast cells, neutrophils, eosinophils, macrophages, and platelets) orchestrating the inflammatory reaction in BP. These data may favor the employment of broad-spectrum or combined immunosuppressants, potentially together with an anticoagulant treatment, over cell- or molecule-specific targeted therapy in patients with this disorder. This article is protected by copyright. All rights reserved.
No caption available
… 
Content may be subject to copyright.
1248
|
wileyonlinelibrary.com/journal/exd Experimental Dermatology. 2017;26:1248–1252.
© 2017 John Wiley & Sons A/S.
Published by John Wiley & Sons Ltd
Accepted: 8 May 2017
DOI: 10.1111/exd.13382
ORIGINAL ARTICLE
Analysis of serum markers of cellular immune activation in
patients with bullous pemphigoid
Katja Bieber1| Anna Lara Ernst1| Stefan Tukaj2| Maike M. Holtsche3|
Enno Schmidt1,3| Detlef Zillikens1,3| Ralf J. Ludwig1,3 | Michael Kasperkiewicz3
1Lübeck Institute of Experimental
Dermatology, University of Lübeck, Lübeck,
Germany
2Department of Molecular Biology, University
of Gdańsk, Gdańsk, Poland
3Department of Dermatology, University of
Lübeck, Lübeck, Germany
Correspondence
Michael Kasperkiewicz, Department of
Dermatology, University of Lübeck, Lübeck,
Germany.
Email: Michael.Kasperkiewicz@uk-sh.de
Funding information
Deutsche Forschungsgemeinschaft
Abstract
Experimental models of bullous pemphigoid (BP), the most frequent subepidermal
autoimmune bullous disease, revealed that the immune response leading to blister
formation represents an incompletely understood complex process involving different
inflammatory cells. In contrast to previous reports commonly focusing on limited
molecular and cellular phenotypes of the disease, the aim of this study was to investi-
gate a broad spectrum of markers of cellular immune activation in patients with BP.
We found that serum levels of soluble CD4, myeloperoxidase, S100A12, eosinophil
cationic protein and soluble P- selectin were significantly elevated in patients with
active BP compared with normal controls. Mast cell tryptase and neopterin serum
levels significantly decreased at the time of clinical remission of the patients.
Additionally, serum concentrations of soluble IL- 2 receptor, mast cell tryptase and
soluble P- selectin were significantly associated with levels of circulating anti- BP180
autoantibodies. Our findings confirm and extend previous reports suggesting some
concomitant involvement of a panel of molecules representative for a wide spectrum
of cellular players (T cells, mast cells, neutrophils, eosinophils, macrophages and
platelets) orchestrating the inflammatory reaction in BP. These data may favour the
employment of broad- spectrum or combined immunosuppressants, potentially
together with an anticoagulant treatment, over cell- or molecule- specific targeted
therapy in patients with this disorder.
KEYWORDS
autoantibody, inflammation, pemphigoid
1 | INTRODUCTION
Bullous pemphigoid (BP) is the most frequent autoimmune subepider-
mal blistering dermatosis, characterized by autoantibodies directed
against the dermal–epidermal junction proteins BP180/BP230 typi-
cally causing pruritic bullous eruptions.[1] While it is well known that
autoantibodies against the 16th non- collagenous (NC16A) epitope
of the BP180 ectodomain are directly pathogenic and serologically
parallel the clinical course of disease,[2] knowledge on other biological
indicators of BP activity is incomplete.
The immune response leading to blister formation in BP rep-
resents a complex process involving different inflammatory cells and
molecules, including NC16A- reactive CD4 T cells, B cells, comple-
ment factors, mast cells, neutrophils, eosinophils, macrophages and
granulocyte- derived basal membrane zone- degrading proteases and
reactive oxygen species.[1,3,4] Previous studies demonstrated that
symptoms of patients with BP are associated with a plethora of raised
cytokines and chemokines in sera and blister fluids.[1,5,6] Moreover, the
Katja Bieber and Anna Lara Ernst are contributed equally to this work
    
|
 1249
BIEBER Et al.
activation of the coagulation cascade seems to be involved in the dis-
ease pathophysiology.[7-9]
It is conceivable that the effect of a single mediator cannot
produce all the aspects of the clinically expressed lesion, but it is
rather the interplay of different factors orchestrating the inflamma-
tory reaction which may better explain the clinical outcome in BP.
Therefore, and in contrast to previous reports mainly focusing on
single or restricted molecular and cellular phenotypes of this dis-
ease,[10-18] the aim of the study was to compare the degree of as-
sociations between a broad spectrum of markers of cellular immune
activation and disease activity in patients with BP. We investigated
serum parameters related to activation of T cells (soluble CD4 [sCD4]
and soluble IL- 2 receptor [sIL- 2R]), B cells (IgD), mast cells (mast cell
tryptase [MCT]), neutrophils (myeloperoxidase [MPO], S100A12 and
soluble L- selectin [sL- selectin]), eosinophils (eosinophil cationic pro-
tein [ECP]), macrophages (neopterin) and platelets (soluble P- selectin
[sP- selectin]).
2 | METHODS
2.1 | Patients
Sera were obtained from 25 BP patients with active skin lesions (mean
age 73±9 years, 15 females, 10 males) and elevated circulating anti-
BP180 NC16A autoantibodies (mean 504±1080 U/mL, range 53-
5543 U/mL) who were admitted to the Department of Dermatology
of the University of Lübeck. In addition, another set of sera was
analysed from 10 of the 25 patients with BP, from whom follow- up
data were available, after a mean follow- up period of 61±40 weeks.
During this time, they received immunosuppressive treatment lead-
ing to a decline in serum anti- BP180 NC16A autoantibodies and a
complete resolution of skin symptoms. Thus, the follow- up serum
marker evaluation was performed on patients free from clinical dis-
ease who still received maintenance immunosuppression (complete
remission on therapy). Control sera were collected from 56 age- and
gender- matched healthy donors (mean age 73±13 years, 34 females,
22 males). The diagnosis of BP in our study patients was based on
typical clinical findings as well as detection of linear deposits of IgG
and/or C3 at the dermal–epidermal junction and serum anti- BP180
NC16A autoantibodies by direct immunofluorescence microscopy
and enzyme- linked immunosorbent assay (ELISA), respectively.
The investigations were conducted under approval from the Ethics
Committee of the University of Lübeck and with written informed
consent.
2.2 | ELISA
ELISA was used to measure serum levels of anti- BP180 NC16A
IgG autoantibodies (Euroimmun, Lübeck, Germany), sCD4
(Novateinbio, Woburn, USA), sIL- 2R (ThermoScientific, Waltham,
USA), IgD (Bethyl Laboratories Inc., Montgomery, USA), MCT
(neoScientific, Boston, USA), MPO (BioLegend, San Diego, USA),
S100A12 (Biorbyt, Cambridge, UK), sL- selectin (eBioscience, San
Diego, USA), ECP (Novateinbio, Woburn, USA), neopterin (IBL
International, Hamburg, Germany) and sP- selectin (eBioscience,
San Diego, USA) according to the respective manufacturer’s
instructions.
2.3 | Statistical analysis
Data were analysed by t test, Mann- Whitney U test, Wilcoxon signed-
rank test or Spearman’s rank correlation test using GraphPad prism
5 (San Diego, CA, USA). A P- value <.05 was considered statistically
significant.
3 | RESULTS
3.1 | Serum soluble markers of immune
activation in patients with active BP vs controls
Serum levels of sCD4, sIL- 2R, IgD, MCT, MPO, S100A12, sL- selectin,
ECP, neopterin and sP- selectin were measured by ELISA in patients
with active BP and age- and gender- matched healthy individuals. In
comparison with controls, sera from patients with BP contained signif-
icantly higher levels of sCD4, MPO, S100A12, ECP and sP- selectin. In
addition, some trend towards higher serum concentrations was found
for MCT (P=.1) and neopterin (P=.06), whereas the remaining markers
were similar between both groups (Figure 1).
FIGURE1 Serum levels of sCD4, sIL- 2R, IgD, mast cell tryptase
(MCT), myeloperoxidase (MPO), S100A12, sL- selectin, eosinophil
cationic protein (ECP), neopterin and sP- selectin measured by
enzyme- linked immunosorbent assay in 25 patients with active
bullous pemphigoid (BP) and 56 normal controls. The dots/squares
and horizontal bars indicate individual and mean values in each
group, respectively. *P<.05, **P<.01, ***P<.001
1250 
|
   BIEBER Et al.
3.2 | Immune activation markers during
follow- up of BP patients
We next analysed the serum concentrations of immune activation
markers during the course of the disease in 10 patients with BP. We
observed a significant reduction of MCT and neopterin as well as
a slight tendency towards a decline in sIL- 2R (P=.1) in parallel with
complete healing of skin lesions after a mean follow- up period of
61 weeks, during which the patients received immunosuppressive
therapy (Figure 2). In addition, a concomitant decrease in levels of
circulating anti- BP180 autoantibodies was observed in these patients
with BP (data not shown), and the concentrations of some of the ac-
tivation markers were significantly associated (sIL2- R, MCT and sP-
selectin) or showed some trend towards a relationship (sCD4, MPO,
ECP and neopterin) with the levels of these autoantibodies (Table 1).
4 | DISCUSSION
Most data regarding the pathogenic importance of cellular autoim-
munity against BP180 were generated in animal models in which either
injection of rabbit antimurine BP180 antibodies or transfer of human
BP autoantibodies into BP180- knockout mice rescued by the human
ortholog duplicate major characteristics of the human disease.[19]
Accordingly, it is believed that following interaction of autoreactive T
and B cells with consecutive production and binding of autoantibodies
to the dermal–epidermal junction, a sequence of events involving dif-
ferent effector cells is initiated in BP. These pathophysiological steps
include release of interleukin 6 (IL- 6) and IL- 8 from basal keratinocytes,
(ii) complement activation, (iii) mast cell degranulation, (iv) infiltration
of inflammatory cells (neutrophils, eosinophils, macrophages and T
cells) into the upper dermis along with secretion of inflammatory me-
diators and (v) release of proteases and reactive oxygen species from
granulocytes that ultimately induce dermal–epidermal splitting.[1,3,4]
In addition, a Th1/Th2/Th17 mixed cytokine and chemokine profile
including interleukin 1β, interleukin 5, interleukin 6, interleukin 8, in-
terleukin 10, interleukin 15, interleukin 17, tumor necrosis factor α,
chemokine ligand 2 (CCL2), CCL5, CCL11, CCL13, CCL18 and CXCL10
can be found in serum samples and blister fluids of patients with BP,
partially paralleling patients’ disease activity.[1,5,6] The data reported
here confirm and extend previous studies suggesting a broad and si-
multaneous immune cell activation in patients with BP.
In this study, we used various serum soluble markers known to
reflect the activation state of different cell types: sCD4 and sIL- 2R, cell
surface molecules released by T cells upon their stimulation[20]; IgD, an
immunoglobulin associated with B- cell development/maturation and
class- switched autoreactive B cells[21]; MCT, MPO, and ECP, granule
proteins liberated from activated mast cells, neutrophils and eosino-
phils, respectively[22-24]; S100A12, a calcium- binding protein secreted
by activated neutrophils[25]; neopterin, a pyrazino- pyrimidine com-
pound synthesized from activated macrophages[26]; and sL- selectin
and sP- selectin, adhesion molecules indicative of neutrophil and plate-
let activation, respectively.[27]
We could show that the serum levels of sCD4, MPO, S100A12,
ECP and sP- selectin were significantly elevated in patients with ac-
tive BP compared with normal controls. Some trend towards higher
serum concentrations was also found for MCT and neopterin during
ongoing disease, both of which significantly decreased along with
a slight decline tendency for sIL- 2R at the time of clinical remission
of the patients. Additionally, some activation markers were signifi-
cantly associated (sIL2- R, MCT and sP- selectin) or showed some
trend towards a relationship (sCD4, MPO, ECP and neopterin) with
anti- BP180 autoantibody titres. These results are in agreement with
previous studies which reported increased concentrations of sCD4,
sIL- 2R, MCT, MPO, and ECP in BP blister fluids and/or serum, partly
FIGURE2 Individual enzyme- linked immunosorbent assay-
based serum levels of sCD4, sIL- 2R, IgD, mast cell tryptase
(MCT), myeloperoxidase (MPO), S100A12, sL- selectin, eosinophil
cationic protein (ECP), neopterin and sP- selectin at time of
cutaneous symptoms and during remission of skin lesions in 10
bullous pemphigoid patients after a mean period of 61 weeks of
immunosuppressive therapy. Horizontal black bars indicate the mean.
*P<.05, **P<.01
TABLE1 Correlation coefficients between serum levels of cell activation markers and anti- BP180 autoantibodies in BP patients
sCD4 sIL- 2R IgD MCT MPO S100A12 sL- selectin ECP Neopterin sP- selectin
Anti- BP180
r.416 .512 −.024 .722 .401 .066 −.109 .392 .404 .572
P.067 .021 .917 <.001 .079 .781 .647 .087 .077 .008
    
|
 1251
BIEBER Et al.
correlating with cytokines, autoantibodies and clinical disease sever-
ity in patients with BP.[10-18] The reported mean blister fluid concen-
trations for sCD4 (42.4 U/mL), sIL- 2R (2070 U/mL), MCT (4.35 U/L),
MPO (3498- 16 863 μg/L) and ECP (282- 1559 μg/L) in these patients
were found to be significantly higher compared with those in their sera
and/or suction blisters of healthy controls.[10-12,14-16,18]
The significantly increased serum concentrations of these acti-
vation markers in our study underscore the previously described key
contribution of the respective T cells, neutrophils and eosinophils to
the disease pathophysiology. Results from the BP180- humanised
mouse model revealed the importance of NC16A- reactive CD4 T cells
and accorded with evidence from previous in vitro studies with human
cells that demonstrated a restriction of NC16A- reactive CD4 T cells to
the BP susceptibility gene HLA- DQB1*0301.[1] In addition, a variety
of BP models suggest that autoantibody- dependent neutrophil and
eosinophil degranulation with the liberation of proteinases or cyto-
toxic agents is essential, damaging the basal membrane zone directly
and causing dermo- epidermal junction separation.[1,6] Interestingly, in
addition to classical inflammatory cells, platelets seem to play a role in
the disease as sP- selectin was observed to be significantly increased in
patients’ sera and to correlate with circulating autoantibodies. In fact,
although the role of the blood coagulation system including platelets
has not yet been investigated in experimental models of BP, some ev-
idence exists on the relationship between inflammation and coagula-
tion. It has been reported that patients with BP show elevated levels of
tissue factor, D- dimer, prothrombin fragment F1+2 and mean platelet
volume and that they have an increased risk for thrombotic events.[7-9]
It is interesting to note that, with the exception of MCT and ne-
opterin, no other immune cell activation marker was found to be sig-
nificantly decreased at follow- up. This suggests some latent persistent
molecular inflammatory process during both complete remission on
therapy and normalization of circulating anti- BP180 autoantibodies
of the patients. However, one has to take into account the different
limitations of this study as mentioned below.
A fraction of the investigated cell activation markers was not dif-
ferent (sIL- 2R, IgD and sL- selectin) or did not quite reach a statistically
significant level (MCT and neopterin) when compared with healthy
controls. This finding does not exclude the possibility that these fac-
tors or their associated cell types are involved in the disease process,
considering that some of these markers significantly declined during
healing of skin lesions (MCT and neopterin) or paralleled autoantibody
serum levels (sIL2- R and MCT) in our patient cohort. There may be
several mutually non- exclusive influencing variables that could have
accounted for these potentially false- negative and partly incongruent
results: (i) the relatively small population size; (ii) the reported pres-
ence of higher concentrations of activation markers in blister fluids
than in sera of patients with BP[10,11,15,16,18]; (iii) the fact that differ-
ent biomarkers corresponding to the same cell type exist which may
yield different results[20,28-32]; (iv) differences in disease duration and
time intervals from active disease stage (ie initial sample) to reaching
remission (ie follow- up sample); (v) the impact of immunosuppressive
(pre- )treatment on the different cell types; (vi) diverse stability of the
investigated markers in the circulation as well as during procession and
storage of blood samples[33]; and (vii) ELISA- related factors (eg anti-
body quality, kit manufacturer, measurement conditions and technical
issues related to dynamic ranges and dilutions).[33]
It also needs to be mentioned that the serological profile described
in our study may not be specific for BP as the tested markers represent
indicators of inflammation in a variety of other disease conditions.[20-27]
For example, all investigated markers that were significantly increased
in our patient cohort have been also found to be elevated in common
inflammatory dermatoses such as psoriasis and/or atopic dermati-
tis.[34-38] Especially platelets, which in addition to playing a central role
in normal haemostasis, have emerged as important players in various
types of inflammatory and non- inflammatory disorders including cere-
brovascular and neurodegenerative diseases.[27,28,39,40] Regarding the
latter, it is worth noting that the main risk factors for BP include neuro-
logic diseases comprising stroke and dementia,[41] both of which were
documented in the medical history of four of our study patients. Thus,
unspecific serum immune marker alterations reflecting rather general
aspects of inflammation or comorbidities cannot be fully excluded in our
study.
In summary, our findings confirm and extend previous reports sug-
gesting some concomitant involvement of a panel of molecules rep-
resentative for a wide spectrum of cellular players (T cells, mast cells,
neutrophils, eosinophils, macrophages and platelets) orchestrating the
inflammatory reaction in BP. These data may favour the employment
of broad- spectrum or combined immunosuppressants, potentially to-
gether with an anticoagulant treatment, over cell- or molecule- specific
targeted therapy in patients with this disorder.
ACKNOWLEDGEMENTS
This work was supported by Deutsche Forschungsgemeinschaft
(DFG) Excellence Cluster “Inflammation at Interfaces” (EXC 306/2)
and the Clinical Research Unit “Pemphigoid Diseases” (KFO 303/1).
CONFLICT OF INTEREST
None declared.
AUTHOR CONTRIBUTION
M.K. and R.J.L. conceived and designed the experiments; K.B. and
A.L.E. performed the experiments; K.B., A.L.E., S.T., E.S., M.M.H., D.Z.,
R.J.L. and M.K. analysed and interpreted the data. M.K. wrote the
manuscript. All authors reviewed the manuscript.
REFERENCES
[1] E. Schmidt, D. Zillikens, Lancet 2013, 381, 320.
[2] E. Schmidt, K. Obe, E. B. Bröcker, D. Zillikens, Arch. Dermatol. 2000,
136, 174.
[3] H. Ujiie, H. Shimizu, Exp. Dermatol. 2012, 21, 901.
[4] H. Iwata, Y. Kitajima, Exp. Dermatol. 2013, 22, 381.
[5] J. Plée, S. Le Jan, J. Giustiniani, C. Barbe, P. Joly, C. Bedane, P. Vabres,
F. Truchetet, F. Aubin, F. Antonicelli, P. Bernard, Sci. Rep. 2015, 5,
18001.
1252 
|
   BIEBER Et al.
[6] M. Riani, S. Le Jan, J. Plée, A. Durlach, R. Le Naour, G. Haegeman, P.
Bernard, F. Antonicelli, J. Allergy Clin. Immunol. 2017, 139, 863.e3.
[7] E. N. Rifaioglu, B. B. Sen, Ö. Ekiz, E. Buyukkaya, N. Sen, Platelets 2014,
25, 264.
[8] M. Cugno, A. Tedeschi, A. Borghi, P. Bucciarelli, R. Asero, L. Venegoni,
S. Griffini, E. Grovetti, E. Berti, A. V. Marzano, PLoS ONE 2015, 10,
e0129456.
[9] M. Cugno, A. V. Marzano, P. Bucciarelli, Y. Balice, G. Cianchini, P.
Quaglino, P. Calzavara Pinton, M. Caproni, M. Alaibac, C. De Simone,
A. Patrizi, E. Cozzani, M. Papini, A. Tedeschi, E. Berti, F. R. Rosendaal;
INVENTEP Study Group, Thromb. Haemost. 2016, 115, 193.
[10] S. Tsuda, M. Miyasato, K. Iryo, T. Nakama, K. Kato, Y. Sasai, J. Dermatol.
1992, 19, 270.
[11] D. Zillikens, A. Ambach, M. Schuessler, R. Dummer, A. A. Hartmann, G.
Burg, Arch. Dermatol. Res. 1992, 284, 141.
[12] W. Czech, J. Schaller, E. Schöpf, A. Kapp, J. Am. Acad. Dermatol. 1993,
29, 210.
[13] M. Caproni, G. M. Palleschi, D. Falcos, A. D’Agata, G. Cappelli, P.
Fabbri, Int. J. Dermatol. 1995, 34, 177.
[14] L. D’Auria, M. Pietravalle, A. Mastroianni, C. Ferraro, A. Mussi, C.
Bonifati, B. Giacalone, F. Ameglio, Arch. Dermatol. Res. 1998, 290, 25.
[15] L. D’Auria, M. Pietravalle, P. Cordiali-Fei, F. Ameglio, Exp. Dermatol.
2000, 9, 131.
[16] C. C. Sun, J. Wu, T. T. Wong, L. F. Wang, M. T. Chuan, Br. J. Dermatol.
2000, 143, 1235.
[17] A. Frezzolini, G. Cianchini, M. Ruffelli, B. Didona, P. Puddu, Clin. Exp.
Immunol. 2004, 137, 595.
[18] A. Tedeschi, A. V. Marzano, M. Lorini, Y. Balice, M. Cugno, J. Eur. Acad.
Dermatol. Venereol. 2015, 29, 813.
[19] K. Bieber, S. Sun, N. Ishii, M. Kasperkiewicz, E. Schmidt, M. Hirose, J.
Westermann, X. Yu, D. Zillikens, R. J. Ludwig, Exp. Dermatol. 2010, 19, 2.
[20] M. Shipkova, E. Wieland, Clin. Chim. Acta 2012, 413, 1338.
[21] K. Chen, A. Cerutti, Curr. Opin. Immunol. 2011, 23, 345.
[22] J. Vitte, Mol. Immunol. 2015, 63, 18.
[23] Y. Kato, J. Clin. Biochem. Nutr. 2016, 58, 99.
[24] K. R. Acharya, S. J. Ackerman, J. Biol. Chem. 2014, 289, 17406.
[25] J. Pietzsch, S. Hoppmann, Amino Acids 2009, 36, 381.
[26] S. De Rosa, P. Cirillo, M. Pacileo, G. Petrillo, G. L. D’Ascoli, F. Maresca, F.
Ziviello, M. Chiariello, Curr. Vasc. Pharmacol. 2011, 9, 188.
[27] R. P. McEver, Cardiovasc. Res. 2015, 107, 331.
[28] P. Ferroni, S. Riondino, N. Vazzana, N. Santoro, F. Guadagni, G. Davì,
Thromb. Haemost. 2012, 108, 1109.
[29] S. Bugatti, B. Vitolo, R. Caporali, C. Montecucco, A. Manzo, Biomed.
Res. Int. 2014, 681678.
[30] J. Rojas, J. Salazar, M. S. Martínez, J. Palmar, J. Bautista, M. Chávez-
Castillo, A. Gómez, V. Bermúdez, Scientifica (Cairo) 2015, 2015,
851252.
[31] D. D. Metcalfe, R. Pawankar, S. J. Ackerman, C. Akin, F. Clayton, F. H.
Falcone, G. J. Gleich, A. M. Irani, M. W. Johansson, A. D. Klion, K. M.
Leiferman, F. Levi-Schaffer, G. Nilsson, Y. Okayama, C. Prussin, J. T.
Schroeder, L. B. Schwartz, H. U. Simon, A. F. Walls, M. Triggiani, World
Allergy Organ. J. 2016, 9, 7.
[32] D. Muthas, A. Reznichenko, C. A. Balendran, G. Böttcher, I. G. Clausen,
C. Kärrman Mårdh, T. Ottosson, M. Uddin, T. T. MacDonald, S. Danese,
M. Berner Hansen, Scand. J. Gastroenterol. 2016, 52, 125.
[33] X. Zhou, M. S. Fragala, J. E. McElhaney, G. A. Kuchel, Curr. Opin. Clin.
Nutr. Metab. Care 2010, 13, 541.
[34] W. Czech, J. Krutmann, E. Schöpf, A. Kapp, Br. J. Dermatol. 1992, 126,
351.
[35] G. Michaëlsson, W. Kraaz, B. Gerdén, E. Hagforsen, I. P. Lundin, L.
Lööf, O. Sj-oberg, A. Scheynius, Br. J. Dermatol. 1996, 135, 371.
[36] O. De Pità, M. Ruffelli, S. Cadoni, A. Frezzolini, G. F. Biava, R. Simom,
V. Bottari, G. De Sanctis, J. Dermatol. Sci. 1996, 13, 118.
[37] D. Wilsmann-Theis, J. Wagenpfeil, D. Holzinger, J. Roth, S. Koch, S.
Schnautz, T. Bieber, J. Wenzel, J. Eur. Acad. Dermatol. Venereol. 2016,
30, 1165.
[38] C. Yazici, K. Köse, S. Utaş, E. Tanrikulu, N. Taşlidere, Arch. Dermatol.
Res. 2016, 308, 207.
[39] B. Nieswandt, I. Pleines, M. Bender, J. Thromb. Haemost. 2011,
9(Suppl. 1), 92.
[40] N. S. Gowert, L. Donner, M. Chatterjee, Y. S. Eisele, S. T. Towhid, P.
Münzer, B. Walker, I. Ogorek, O. Borst, M. Grandoch, M. Schaller, J.
W. Fischer, M. Gawaz, S. Weggen, F. Lang, M. Jucker, M. Elvers, PLoS
ONE 2014, 9, e90523.
[41] Y. C. Lai, Y. W. Yew, W. C. Lambert, J. Eur. Acad. Dermatol. Venereol.
2016, 30, 2007.
How to cite this article: Bieber K, Ernst AL, Tukaj S, et al.
Analysis of serum markers of cellular immune activation in
patients with bullous pemphigoid. Exp Dermatol.
2017;26:1248–1252. https://doi.org/10.1111/exd.13382
... La cavidad bucal es uno de los principales órganos en los que se desarrollan los cuadros clínicos de distintas patologías. 1 Las alteraciones gingivales mediadas por factores sistémicos y locales pueden incluir procesos autoinmunes encargados de generar degradación en la zona de la membrana basal, dando así origen al desarrollo de lesiones en forma de ampolla. 2,3 Estos padecimientos en la cavidad oral son: pénfigo, penfigoide, algunos tipos de gingivitis no inducida por placa y liquen plano. Sus manifestaciones incluyen el desarrollo de úlceras, máculas eritematosas, pápulas urticariales, erosiones, vesículas y ampollas que al reventarse ocasionan mucho dolor; suelen curarse rápido y dejan una zona residual pigmentada, pero no una cicatriz. ...
... 56 Serum levels of soluble E-selectin and vascular endothelial growth factor in BP were associated with those of circulating autoantibodies, which supported the interaction between activated endothelial inflammatory and immune reactions. 57,58 Eosinophils in BP and PV are also believed to involve in coagulation activation at skin levels. 59 Third, antiphospholipid antibodies have been detected in patients with BP and PV. ...
Article
Full-text available
Background Bullous pemphigoid (BP) and pemphigus vulgaris (PV) share similar pathophysiology with venous thromboembolism (VTE) involving platelet activation, immune dysregulation, and systemic inflammation. Nevertheless, their associations have not been well established. Methods and Results To examine the risk of incident VTE among patients with BP or PV, we performed a nationwide cohort study using Taiwan's National Health Insurance Research Database and enrolled 12 162 adults with BP or PV and 12 162 controls. A Cox regression model considering stabilized inverse probability weighting was used to calculate the hazard ratios (HRs) for incident VTE associated with BP or PV. To consolidate the findings, a meta‐analysis that incorporated results from the present cohort study with previous literature was also conducted. Compared with controls, patients with BP or PV had an increased risk for incident VTE (HR, 1.87 [95% CI, 1.55–2.26]; P <0.001). The incidence of VTE was 6.47 and 2.20 per 1000 person‐years in the BP and PV cohorts, respectively. The risk for incident VTE significantly increased among patients with BP (HR, 1.85 [95% CI, 1.52–2.24]; P <0.001) and PV (HR, 1.99 [95% CI, 1.02–3.91]; P =0.04). In the meta‐analysis of 8 studies including ours, BP and PV were associated with an increased risk for incident VTE (pooled relative risk, 2.17 [95% CI, 1.82–2.62]; P <0.001). Conclusions BP and PV are associated with an increased risk for VTE. Preventive approaches and cardiovascular evaluation should be considered particularly for patients with BP or PV with concomitant risk factors such as hospitalization or immobilization.
... Platelets are a major source and site of storage of SDF-1 (67) and recently many proteins associated with platelet degranulation have been detected in BP blister fluid (68). Also, elevated serum levels of sP-selectin in BP patients reflect platelet activation (69). T2D patients have enhanced platelet activation and increased risk for atherosclerosis (70). ...
Article
Full-text available
The use of dipeptidyl peptidase 4 (DPP4) inhibitors, (also known as gliptins), is associated with an increased risk of bullous pemphigoid (BP), an autoimmune blistering skin disease. To explore the mechanism behind gliptin-associated BP we investigated circulating autoantibodies against the major BP autoantigen BP180 in serum samples from patients with type 2 diabetes (T2D) with preceding gliptin medication (n = 136) or without (n = 136). Sitagliptin was the most frequently prescribed gliptin (125/136 patients). Using an ELISA assay, we showed that IgG autoantibodies against the immunodominant NC16A domain of BP180 were found in 5.9% of gliptin treated and in 6.6% of non-gliptin treated T2D patients. We found that 28% of gliptin treated patients had IgG autoantibodies recognizing the native full-length BP180 in ELISA, but among non-gliptin treated the seropositivity was even higher, at 32%. Further ELISA analysis of additional serum samples (n = 57) found no major changes in the seropositivity against BP180 during a follow-up period of about nine years. In immunoblotting, full-length BP180 was recognized by 71% of gliptin treated and 89% of non-gliptin treated T2D patients, but only by 46% of the age-and sex-matched controls. The chemokine stromal derived factor-1(SDF-1/CXCL12) is one of the major substrates of DPP4. Immunostainings showed that the expression of SDF-1 was markedly increased in the skin of BP patients, but not affected by prior gliptin treatment. We found that the use of gliptins decreased the serum level of SDF-1α in both BP and T2D patients. Our results indicate that the autoantibodies against the linear full-length BP180 are common in patients with T2D, but seropositivity is unaffected by the use of sitagliptin.
... There is also evidence to support a role for mast cells in other chronic inflammatory diseases and cancer. For example, high numbers of mast cells and elevated levels of mast cell-derived mediators are seen in the tissues and blood of patients with multiple sclerosis 62 , rheumatoid arthritis 63 , bullous pemphigoid 64 and various types of cancer (reviewed in ref. 65 ), and correlate with the intensity of inflammation in some of these diseases 66 . Also, studies in Kit mutant mice suggested that mast cells drive pathology in autoimmune diseases (reviewed in ref. 20 ), although the results were inconsistent between different mouse strains 67,68 and were not reproduced using KITindependent mast cell-deficient mice in certain models, for example in models of type 1 diabetes 69 . ...
Article
Mast cells have crucial roles in allergic and other inflammatory diseases. Preclinical approaches provide circumstantial evidence for mast cell involvement in many diseases, but these studies have major limitations — for example, there is still a lack of suitable mouse models for some mast cell-driven diseases such as urticaria. Some approaches for studying mast cells are invasive or can induce severe reactions, and very few mediators or receptors are specific for mast cells. Recently, several drugs that target human mast cells have been developed. These include monoclonal antibodies and small molecules that can specifically inhibit mast cell degranulation via key receptors (such as FcεRI), that block specific signal transduction pathways involved in mast cell activation (for example, BTK), that silence mast cells via inhibitory receptors (such as Siglec-8) or that reduce mast cell numbers and prevent their differentiation by acting on the mast/stem cell growth factor receptor KIT. In this Review, we discuss the existing and emerging therapies that target mast cells, and we consider how these treatments can help us to understand mast cell functions in disease.
... It cannot only secrete inflammatory cytokines to promote subsequent infiltration of inflammatory cells such as neutrophils in skin lesions, but also release mast cell protease (MCP)-4 to activate major proteases such as MMP-9 that cause BP tissue damage (77). Trypsin in serum and BF, as reliable indicators of mast cell activation, seems to be consistent with the disease activity (78,79), but more research is required to obtain a high-level evidence. ...
Article
Full-text available
Bullous pemphigoid (BP) is the most common autoimmune subepidermal blistering disorder in the elderly. Systemic and topical use of glucocorticoids and immunosuppressants has been shown to be effective in most patients. However, refractory BP patients are challenged to clinicians with severe clinical symptoms, resistance to treatment, and high relapse rate. How to predict and assess the refractory and severity of bullous pemphigoid is the key issue in clinical practice, and the urgent need for precision medicine in refractory patients is driving the search for biomarkers and biologics. Recently, some biomarkers, such as the level of specific autoantibodies and released cytokines, have been proposed as the potential parameters to reflect the disease severity and predict the treatment response and relapse of refractory BP. Moreover, new biologics targeting pathogenic antibodies, complement, Th2 axis, eosinophils, and Th17 axis have shown potent efficacy on refractory BP. Here, we review the literature and give an overview of emerging biomarkers and therapeutic strategies for refractory bullous pemphigoid to improve the prognosis of the patient.
Article
Pemphigoid diseases (PDs) are a group of autoimmune blistering diseases, including bullous pemphigoid, epidermolysis bullosa acquisita, mucous membrane pemphigoid, linear immunoglobulin A disease, and other rare variants. These diseases are characterized by the presence of autoantibodies that target proteins at the dermal-epidermal junction, resulting in the formation of tense blisters and erosions on the skin and/or mucosa. The current therapeutic approaches, such as systemic corticosteroid, are associated with significant adverse effects, highlighting that safer and more effective treatment options are an urgent clinical need. To address this unmet need, a comprehensive understanding of the detailed mechanisms underlying PDs is essential. Based on their histopathological infiltration in pemphigoid lesions, neutrophils have long been implicated as major contributors to the initiation and progression of the diseases. Numerous in vivo and in vitro studies have investigated the role of neutrophils in the pemphigoid pathology, revealing various pathological mechanisms induced by these cells, including the release of neutrophil elastase and matrix metalloproteinase-9, as well as the formation of neutrophil extracellular traps. The present review provides a comprehensive summary and critical evaluation of the current understanding regarding the role of neutrophils in PDs. In addition, it discusses the potential of targeting neutrophil-associated pathways as a novel therapeutic approach for the diseases.
Article
Full-text available
Background Bullous pemphigoid (BP) is an autoimmune skin-blistering disease. Systemic corticosteroids remain the first line treatment for moderate-to-severe BP with the potential for severe adverse events. Dupilumab has emerged as an alternative option for BP patients. Objective We evaluated the efficiency and safety of dupilumab on BP treatment and explored a mode of drug action in depth. Methods and results A multicenter retrospective cohort included 20 BP patients who received dupilumab with or without systemic corticosteroid in dupilumab group, and 20 matched BP patients who received corticosteroid alone in conventional group. Serum samples were collected from 20 patients (10 from dupilumab group and 10 from conventional group) at baseline and week 4. Compared to systemic corticosteroid alone, dupilumab with or without systemic corticosteroid was similarly efficacious in clinical remission at week4 (complete remission plus partial remission: 100%) and week24 (complete remission plus partial remission:100%), but allowing significant decreases in the cumulative doses of corticosteroids with reducing the incidence of adverse events. However, dupilumab did not decrease BP180 antibody despite an obvious clinical improvement. Comparative plasma proteomic analysis performed before and after treatment in 3 BP patients from dupilumab group revealed that drug use was associated with 30 differentially expressed proteins, including 26 down-regulated and 4 up-regulated proteins. The former consisted of immune related proteins involved in T/B cell interactions (inducible T-cell co-stimulator ligand, ICOSL) and in the activation of eosinophils (PRG2), mast cells (S100A12), and complement (CR2). TARC and ICOSL levels correlated with BP severity in patients who received either dupilumab or conventional treatment. Conclusion Dupilumab has similar efficacy in treating BP as conventional drugs, by inhibiting the activities of many types of immune cells and complement, and regulating the interactions between T and B cells.
Article
Pruritus is a hallmark feature in pemphigoid diseases, where it can be severe and greatly impact the quality of life of affected patients. Despite being a key symptom, the exact pathophysiological mechanisms involved in pruritus in pemphigoid are yet to be fully elucidated and effective therapies addressing them are limited. This review summarizes the present understanding of pruritus specific to pemphigoid diseases, especially the pruritogens that induce it, and the therapeutic options that have been explored so far. The majority of the available evidence is on bullous pemphigoid and epidermolysis bullosa acquisita. Histamine derived from basophils correlates with pruritus severity, with omalizumab demonstrating promising efficacy in pruritus for bullous pemphigoid. IL‐4/−13 contribute to itch in bullous pemphigoid with dupilumab being evaluated in clinical trials. Other pruritogens of interest include substance P, tryptase, and thymic stromal lymphopoetin, with therapies targeting them requiring further investigation. Scratching behaviors contribute directly to blister formation through various mechanisms, such as pathological autoantibody recruitment, T helper cell type 1 polarization, and exposure of intracellular autoantigens. Treatments addressing these pathways may contribute to decreasing disease severity. Additional studies are needed to fully characterize how pruritus is regulated in pemphigoid diseases, to help pave the way to develop novel and effective therapeutics that will not only address pruritic symptoms but also decrease disease severity.
Article
Bullous pemphigoid (BP) is an autoimmune blistering disorder with substantial morbidity and mortality. BP is regarded as a disorder driven by IgG due to BP180 and BP230 IgG autoantibodies, yet, new advances highlight the function of eosinophils and IgE autoantibodies in BP. Evidence supports that eosinophils are involved in BP pathogenesis, notably, these include the presence of IL-5, eotaxin, and eosinophil-colony stimulating factor in blister fluid, peripheral blood eosinophilia is present in nearly 50% of affected patients, eosinophils are found against the dermo-epidermal junction (DEJ) when BP serum is present, metalloprotease-9 is secreted by eosinophils at blister sites, blister fluid of BP patients contains eosinophil granule proteins which are located along the lamina lucida of the BMZ in patients with BP and correspond with disease clinically, eosinophil extracellular traps (EET) have been linked to DEJ splitting, IL-5 activated eosinophils cause DEJ separation when BP serum is present, and eosinophils are requisite to drive anti-BP180 IgE mediated blistering of the skin. Yet, the mechanism whereby eosinophils contribute to the pathogenesis of BP remains to be explored. In this review, we examine the role of eosinophils in BP while offering a basis to explain the pathomechanisms of eosinophils in BP.
Article
iNKT (invariant natural killer T) cells are unconventional immunoregulatory T cells which contribute to B cell maturation, antibody and cytokine production. iNKT cells are implicated in the control of autoimmune inflammation in different disorders. For bullous pemphigoid (BP), the most frequent bullous autoimmune dermatosis, the role of iNKT cells has not yet been studied. We, therefore, aimed at investigating the frequency of iNKT cells in peripheral blood and biopsies from lesional and non-lesional skin from patients with BP and controls. Circulating CD3⁺iTCR⁺ iNKT cells were assessed by flow cytometry in peripheral blood from 30 patients with BP and from 29 controls (19 patients with skin tumors and 10 healthy controls). In 34 lesional and 13 non-lesional skin biopsies from BP patients and 17 biopsies from control individuals the number of Vα24⁺Vβ11⁺ iNKT cells was investigated by immunofluorescence staining. BP patients showed a significantly lower frequency of circulating iNKT cells compared to the control group. Patients with severe disseminated blistering tended to display lower iNKT cell numbers than patients with moderate disease severity. In lesional skin of BP patients, an enrichment of iNKT cells was detected compared to skin biopsies from controls. Similarly to control biopsies, non-lesional biopsies of BP patients contained only few iNKT cells. In conclusion, the deficiency of circulating iNKT cells associated with enrichment at the site of cutaneous inflammation suggests that iNKT cells may play a pathophysiologically relevant role in BP.
Article
Full-text available
Myeloperoxidase is an inflammatory enzyme that generates reactive hypochlorous acid in the presence of hydrogen peroxide and chloride ion. However, this enzyme also uses bromide ion or thiocyanate as a substrate to form hypobromous or hypothiocyanous acid, respectively. These species play important roles in host defense against the invasion of microorganisms. In contrast, these enzyme products modify biomolecules in hosts during excess inflammation, indicating that the action of myeloperoxidase is both beneficial and harmful. Myeloperoxidase uses other endogenous compounds, such as serotonin, urate, and l-tyrosine, as substrates. This broad-range specificity may have some biological implications. Target molecules of this enzyme and its products vary, including low-molecular weight thiols, proteins, nucleic acids, and lipids. The modified products represent biomarkers of myeloperoxidase action. Moderate inhibition of this enzyme might be critical for the prevention/modulation of excess, uncontrolled inflammatory events. Some phytochemicals inhibit myeloperoxidase, which might explain the reductive effect caused by the intake of vegetables and fruits on cardiovascular diseases.
Article
Full-text available
Biomarkers of disease activity have come into wide use in the study of mechanisms of human disease and in clinical medicine to both diagnose and predict disease course; as well as to monitor response to therapeutic intervention. Here we review biomarkers of the involvement of mast cells, basophils, and eosinophils in human allergic inflammation. Included are surface markers of cell activation as well as specific products of these inflammatory cells that implicate specific cell types in the inflammatory process and are of possible value in clinical research as well as within decisions made in the practice of allergy-immunology.
Article
Full-text available
Bullous pemphigoid (BP) is an inflammatory autoimmune bullous disease involving cytokines and proteases in the process of blister formation. Recently, IL-17 and IL-23 were evidenced in lesional skin and serum of BP patients at time of diagnosis, but their involvement in disease outcome has still not been investigated yet. We then analysed IL-17 and IL-23 serum levels during the first months of follow-up upon treatment. Compared with age- and sex- matched controls, high levels of IL-23 were observed at baseline in BP patients serum (P < 0.01), while IL-17 levels was not. However, some BP patients expressed high IL-17 serum level, independently of disease severity. In these patients, those with ongoing remission reduced IL-17 concentration upon treatment (P < 0.001), whereas IL-17 level remained elevated in patients who relapsed. Meanwhile, IL-23 serum levels increased during the first month of treatment in BP patients who later relapsed (P < 0.01) and MMP-9 serum level was not controlled. Accordingly, we found that both IL-17 and IL-23 increased MMP-9 secretion from leukocytes in-vitro. Then, we showed that elevated IL-17/IL-23 serum concentrations helped to discriminate BP patients who later relapsed. Such uncontrolled inflammatory response raises the question whether these molecules could become biological target for BP patients resistant to steroid treatment.
Article
Full-text available
Activation of blood coagulation has been demonstrated in bullous pemphigoid (BP), a rare autoimmune blistering disease, potentially leading to a prothrombotic state. In order to evaluate the incidence of venous thromboembolism (VTE) in BP, a cohort study was carried out on 432 BP patients (59 % females; median age 76 years, interquartile range [IQR]: 68-82). At diagnosis, autoimmune bullous skin disorder intensity score (ABSIS) was calculated. VTE incidence was standardised with rates of the general population. Multivariable Cox proportional hazard model was used to estimate the hazard ratio of VTE according to ABSIS and concomitant risk factors. During a median follow-up of 4.2 years, 31 objectively-diagnosed VTE events were recorded. The incidence rate of VTE (per 1000 patient-years) was 17.2 overall (95 % confidence interval [CI]: 11.1-23.2), 56.7 (95 %CI: 33.0-80.4) during acute phase (22 VTE) and 6.3 (95 %CI: 2.8-11.3) during remission (9 VTE). The standardised incidence ratio was 4.06 (95 %CI: 2.73-5.65), higher during the acute phase (14.86, 95 %CI: 9.20-21.88) than during remission (1.48, 0.66-2.63). The adjusted hazard ratio of VTE was 2.74 (95 %CI: 1.07-7.04) for ABSIS > 48 vs ABSIS < 28, and 2.56 (95 %CI: 1.00-6.70) in patients with ≥ 2 concomitant risk factors. In conclusion, BP patients have a 15-fold increased VTE risk during acute phase, proportional to disease severity and heightened by concomitant risk factors.
Article
Full-text available
Coagulation activation has been demonstrated in two prototypic autoimmune skin diseases, chronic autoimmune urticaria and bullous pemphigoid, but only the latter is associated with increased thrombotic risk. Two markers of coagulation activation (prothrombin fragment F1+2 and fibrin fragment D-dimer) were measured by immunoenzymatic methods in plasma samples from 30 patients with active chronic autoimmune urticaria, positive for autologous serum skin test, 30 patients with active bullous pemphigoid and 30 healthy subjects. In skin biopsies, tissue factor expression was evaluated by both immunohistochemistry and in situ hybridization. F1+2 and D-dimer levels were higher in active chronic autoimmune urticaria (276.5±89.8 pmol/L and 5.56±4.40 nmol/L, respectively) than in controls (145.2±38.0 pmol/L and 1.06±0.25 nmol/L; P=0.029 and P=0.011) and were much higher in active bullous pemphigoid (691.7±318.7 pmol/L and 15.24±9.09 nmol/L, respectively) (P<0.0001). Tissue factor positivity was evident in skin biopsies of both disorders with higher intensity in bullous pemphigoid. F1+2 and D-dimer, during remission, were markedly reduced in both disorders. These findings support the involvement of coagulation activation in the pathophysiology of both diseases. The strong systemic activation of coagulation in bullous pemphigoid may contribute to increase the thrombotic risk and provides the rationale for clinical trials on anticoagulant treatments in this disease.
Article
Background: Outcome of bullous pemphigoid (BP), the most frequent autoimmune skin blistering disease, involves MMP-9, IL-17 and IL-23 release from infiltrated inflammatory cells. The chemokine CXCL10 has been associated with several autoimmune diseases but its participation to BP pathophysiology still needs to be clarified. Objective: We sought to assess whether BP outcome was associated with different CXCL10 levels, and to evaluate CXCL10 contribution to the described cytokine/protease inflammatory loop associated to disease outcome. Methods: BP skin biopsy specimens (n=16), sera (n=114), blister fluid (n=23) as well as primary inflammatory cells from BP patients were used to investigate CXCL10 expression and function. Results: At baseline, both resident such as keratinocytes and fibroblasts, and infiltrating immune cells expressed CXCL10 at lesional site in BP skin. CXCL10 levels were higher in BP patient blister fluids (p<0.0001) and sera (p<0.005) than in age- and sex-matched control sera (n=34). Furthermore, CXCL10 serum levels increased at day 60 only in patients who relapsed within the first year of treatment (n=33; p<0.005). Interestingly, cxcl10 expression could be upregulated by itself and IL-17 in inflammatory cells. Notably, neutrophils and monocytes from BP patients only, but not lymphocytes, responded to CXCL10 by increasing MMP-9 secretion through the activation of the ERK1/2, P38, PI3K signalling pathways. Finally, CXCL10-increased MMP-9 secretion was inhibited by methylprednisolone and also by compound A, a novel non-steroidal glucocorticoid receptor ligand. Conclusion: We showed that increased level of inflammatory biomarkers in BP such as CXCL10 favours neutrophil- and monocyte-associated MMP-9 release and disease relapse, and opened new therapeutic horizons in this autoimmune disease.
Article
Objectives: This review article describes the role of neutrophils in mucosal injury and the resulting crypt abscesses characteristic of ulcerative colitis. We also review selected biomarkers for monitoring neutrophil presence and activity in the mucosa as well as their potential as therapeutic targets. Material: We have collated and selectively reviewed data on the most prominent well-established and emerging neutrophil-related biomarkers and potential therapeutic targets (calprotectin, lactoferrin, CXCR1, CXCR2, MMP-9, NGAL, elafin, HNE, pANCAs, MPO, CD16, CD177, CD64, HNPs, SLPI and PTX3) in ulcerative colitis. Results: Systemic and intestinal neutrophil activity increases substantially in active ulcerative colitis, driving tissue damage and extra-intestinal manifestations. Calprotectin is a robust neutrophil and disease biomarker and a few neutrophil related targets are being clinically explored as therapeutic targets. Conclusion: We propose that targeting neutrophils and their inflammatory mediators per se is an opportunity that should be explored to identify new effective medical therapies. The overall clinical goal for neutrophil-targeted therapy will be to modulate, but not completely silence, neutrophil activity, thereby abolishing the destructive inflammation with associated acute and chronic tissue damage without compromising host-defense.
Article
Bullous pemphigoid (BP) is a chronic, autoimmune vesiculobullous disease that frequently occurs in the elderly population. Previous epidemiological studies have suggested an association between BP and neurological diseases; some studies, however, showed conflicting results. This study aimed to investigate if patients with BP have significantly higher risks for neurological disorders, compared to controls. A comprehensive search was performed using MEDLINE, EMBASE and Cochrane library databases. Case-control and cohort studies that assessed the relationship between BP and neurological diseases were included. DerSimonian and Laird random-effects models were utilized to calculate the pooled relative risks (RRs). Publication bias was evaluated qualitatively by constructing a funnel plot and quantitatively by conducting Egger's test. Fourteen studies, with 23 369 BP cases and 128 697 controls were included in this meta-analysis. Patients with BP were significantly more likely to have stroke (RR 2.68, 95% CI: 2.07-3.46), Parkinson's disease (PD; RR 3.42, 95% CI: 3.01-3.87), dementia (RR 4.46, 95% CI: 3.23-6.16), epilepsy (RR 2.98, 95% CI: 1.42-6.28), multiple sclerosis (RR 12.40, 95% CI: 6.64-23.17) and any aforementioned neurological disease (RR 4.93, 95% CI: 3.62-6.70), compared to controls. Moderate to high heterogeneity were observed for analyses of most neurological diseases, except for PD and multiple sclerosis. This study provided support for a significant association between BP and neurological diseases. Clinicians should be aware of this association and manage modifiable risk factors for neurological diseases accordingly.
Article
Oxidative stress may play a pivotal role in the pathogenesis of psoriasis, an inflammatory/hyperproliferative skin disease characterized by the cutaneous accumulation of neutrophils releasing reactive oxygen species, as revealed in a number of studies. This study was performed to demonstrate the presence of oxidative stress in psoriasis, as measured by protein oxidation markers. Twenty-nine psoriasis patients were selected based on disease severity assessment using body surface area as well as the psoriasis area severity index (PASI), and were grouped as mild (PASI ≤ 10) and moderate-to-severe (PASI > 10). The measured parameters in psoriatic patients and fourteen healthy volunteers were as follows: erythrocyte sedimentation rate (ESR), high sensitive C-reactive protein (CRP), myeloperoxidase (MPO) activity, neopterin, total lipid hydroperoxides (LHP), pyrrolized protein (PP), protein carbonyl compounds (PCC), advanced oxidation protein products (AOPP), thiol levels, along with complete blood count. Except lower thiols, all parameters were found to be higher in total patients as well as in subgroups, compared to controls. There was no significant difference among the subgroups. In conclusion, protein oxidation in psoriatics, not only in moderate-to-severe, but also in mild patients, may be explained by the findings of inflammation, phagocytic cell oxidation, and MPO-hypochlorous acid-oxidation reactions; as reflected by increased total/differential leucocytes counts, CRP, ESR as well as MPO, neopterin, AOPP, PCC, PP, LHP, and decreased thiol levels. Demonstrating the AOPP and PP formation for the first time, oxidants from active neutrophils/monocytes may play an important role in the pathogenesis of psoriasis, leading to oxidative stress, especially by protein oxidation.
Article
Psoriasis is a chronic skin disease with deregulation of proteins in the immune system. These proteins include members of the heterogeneous S100 family, which have been discussed as potential biomarkers for disease severity. The aim of this study was to evaluate the impact of S100A7, S100A8, S100A9 and S100A12 as possible markers for disease activity in patients with psoriasis skin disease. S100A7, S100A8, S100A9 and S100A12 mRNA expression was determined in the skin of patients with psoriasis and controls (N = 341) by gene expression analyses. In addition, S100 serum levels were investigated by ELISA in an independent cohort of psoriasis patients (i) untreated, with different manifestations (skin/joints), (ii) under treatment (etanercept) and (iii) healthy controls, (N = 55). All S100-subtypes included are significantly upregulated in psoriasis skin lesions when compared with atopic dermatitis, lichen ruber and healthy donors. In untreated psoriasis patients, S100A12-serum levels showed the closest association with disease activity (PASI) (r = 0.542; P < 0.01). Serum levels decreased under treatment with etanercept (P < 0.05). Among the investigated S100-proteins, S100A12 showed the closest association with disease activity and therapeutic response and might therefore provide a valuable biomarker for psoriasis. © 2015 European Academy of Dermatology and Venereology.