Journal Pre-proof Omeprazole inhibits IgE-mediated mast cell activation and allergic inflammation induced by ingested allergen in mice Cynthia Kanagaratham, PhD, Yasmeen S. El Ansari, MMSc, Benjamin F. Sallis, BSc, Brianna-Marie A. Hollister, BA, Owen L. Lewis, BS, Samantha C. Minnicozzi, MD, Michiko K. Oyoshi, PhD, Rachel Rosen, MD, MPH, Samuel Nurko, MD, Edda Fiebiger, PhD, Hans C. Oettgen, MD, PhD PII:
S0091-6749(20)30342-0
DOI:
https://doi.org/10.1016/j.jaci.2020.02.032
Reference:
YMAI 14452
To appear in:
Journal of Allergy and Clinical Immunology
Received Date: 10 October 2019 Revised Date:
25 February 2020
Accepted Date: 26 February 2020
Please cite this article as: Kanagaratham C, El Ansari YS, Sallis BF, Hollister B-MA, Lewis OL, Minnicozzi SC, Oyoshi MK, Rosen R, Nurko S, Fiebiger E, Oettgen HC, Omeprazole inhibits IgEmediated mast cell activation and allergic inflammation induced by ingested allergen in mice, Journal of Allergy and Clinical Immunology (2020), doi: https://doi.org/10.1016/j.jaci.2020.02.032. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2020 Published by Elsevier Inc. on behalf of the American Academy of Allergy, Asthma & Immunology.
1
Omeprazole inhibits IgE-mediated mast cell activation and allergic inflammation induced
2
by ingested allergen in mice
3 4
Cynthia Kanagaratham, PhD1,2, Yasmeen S. El Ansari, MMSc1,2, Benjamin F. Sallis, BSc1,
5
Brianna-Marie A. Hollister, BA1, Owen L. Lewis, BS1, Samantha C. Minnicozzi, MD1,2, Michiko K.
6
Oyoshi, PhD1,2, Rachel Rosen1,2, MD, MPH, Samuel Nurko1,2, MD, Edda Fiebiger, PhD1,2,* and
7
Hans C. Oettgen, MD, PhD1,2,*
8 9
1
10
2
11
*These authors contributed equally to this work.
Department of Pediatrics, Boston Children’s Hospital, Boston, MA, 02115, USA. Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
12 13
Conflicts: RR is a consultant for Takeda. All other authors declare that they have no relevant
14
conflicts of interest.
15 16
Corresponding author:
17
[email protected]
18
1 Blackfan Circle
19
Boston, MA 02115
20
USA
21
Phone: 1 (617) 919-6842
22
Fax: 1 (617) 730-0528
23 24
Research in this report was funded by NIH, grants 1R01AI119918-05 (HCO), DK097112-05 (RR)
25
and 5T32AI007512-33 (SCM). CK is supported by a postdoctoral fellowship from the Fonds de
26
recherche du Québec - Santé. EF was supported by a Bridge Grant from the Research Council
27
of Boston Children's Hospital, an Emerging Investigator Award from FARE, a Senior Research
28
Award from the Crohn's and Colitis Foundation, and an unrestricted gift from the Mead Johnson
29
Nutrition Company. This work was further supported by an NIH grant of the Harvard Digestive
30
Diseases Center (P30DK034854, Core C). Additional generous support was provided by a Sean
31
N. Parker Center Seed Grant, the Bunning Family Foundation, the Nanji Family Fund for Food
32
Allergy Research, the Rao Chakravorti Family Fund, and the Christine Olsen and Robert Small
33
Food Allergy Research Fund.
2
34
Abstract
35
Background: Patients with eosinophilic esophagitis have increased numbers of mucosal mast
36
cells. Administration of the proton pump inhibitor omeprazole can reduce both esophageal mast
37
cell and eosinophil numbers and attenuate type 2 inflammation in these subjects.
38
Objective: Given that maintenance of an acidic environment within granules is important for
39
mast cell homeostasis, we sought to evaluate the effects of omeprazole on mast cell functions
40
including development, IgE:FcεRI-mediated activation and responses to food allergen.
41
Methods: Mast cell degranulation, cytokine secretion and early signaling events in the FcεRI
42
pathway, including protein kinase phosphorylation and Ca2+ flux, were measured following IgE
43
crosslinking in murine bone marrow-derived mast cells and human cord blood-derived mast
44
cells. The effects of omeprazole on these responses were investigated as was its impact on
45
mast cell-dependent anaphylaxis and food allergy phenotypes in vivo.
46
Results: Murine and human mast cells treated with omeprazole exhibited diminished
47
degranulation and release of cytokines and histamine in response to allergen. In murine mast
48
cells, phosphorylation of protein kinases, ERK and SYK, was decreased. Differentiation of mast
49
cells from bone marrow progenitors was also inhibited. IgE-mediated passive anaphylaxis was
50
blunted in mice treated with omeprazole as was allergen-induced mast cell expansion and mast
51
cell activation in the intestine in a model of food allergy.
52
Conclusion: Our findings suggest that omeprazole targets pathways important for the
53
differentiation and activation of murine mast cells and for the manifestations of food allergy and
54
anaphylaxis.
55 56 57 58
3
59 60
Key Messages:
61
-Omeprazole blocks IgE-mediated mast cell degranulation, and prostaglandin D2 and cytokine
62
production in response to allergen as well as IgE-mediated hypersensitivity in vivo.
63
-Pretreatment with omeprazole results in decreased phosphorylation of protein kinases and
64
inhibits calcium flux into the cytosol.
65
-In a food allergy model, omeprazole inhibits mast cell expansion, type 2 allergic inflammation
66
and hypersensitivity responses to ingested allergen.
67 68
Capsule Summary:
69
Omeprazole inhibits IgE-mediated mast cell activation and allergic responses to food allergen.
70 71
Key Words: food allergy, anaphylaxis, mast cell, omeprazole, proton pump inhibitor
72 73
Abbreviations: OVA, ovalbumin; MCPT-1, mouse mast cell protease; EoE, eosinophilic
74
esophagitis; PPI, proton pump inhibitor
75
4
76 77
Introduction Food allergy is a major health problem in industrialized countries and its prevalence has
78
been increasing over the past decades1, 2. There is a lack of understanding of the mechanisms
79
leading to increased immunological sensitivity to foods in affected subjects and there are no
80
cures for the disease. Patients are advised to practice allergen avoidance and manage acute
81
reactions that arise after accidental exposures using injectable epinephrine2, 3. Insight into the
82
cellular and molecular pathways regulating hypersensitivity reactions to foods are badly needed
83
and may lead to innovative strategies for treatment. In its most common and severe form, food
84
allergy is mediated by the recognition of food antigens by food-specific IgE antibodies.
85
Reactions are triggered upon IgE:allergen-mediated activation of mast cells which harbor the
86
high affinity IgE receptor, FcεRI, on their cell surface4. Crosslinking of the receptor leads to
87
degranulation of mast cells and release of inflammatory mediators that lead to clinical
88
phenotypes including gastrointestinal responses (oral pruritus, abdominal pain, vomiting,
89
diarrhea) and systemic anaphylaxis including vasodilation and vascular leak with tissue edema.
90
Together, these physiologic changes can lead to decreased blood pressure with eventual shock
91
and to respiratory failure3.
92
The prevalence of chronic inflammatory forms of immunologically-mediated food
93
sensitivity is increasing. Many of these can belong to the category of eosinophilic
94
gastrointestinal disorders. The most common is eosinophilic esophagitis (EoE), which is
95
characterized by an elevated number of eosinophils in the esophagus and is one of the leading
96
causes of food impaction and esophageal strictures5-7. There is clearly overlap between the two
97
types of food sensitivity. For instance, in patients affected by EoE, tissue mast cell homeostasis
98
is dysregulated, and IgE-mediated food allergies are often also present8. Increased mast cell-
99
associated transcripts, including, for example, CPA3 and TPSB2, and mast cell numbers are
100
typically present in the esophageal tissue of EoE patients9-12. In a subset of EoE patients,
101
symptoms of esophageal dysfunction and eosinophilia are relieved following a course of PPI 5
102
therapy, giving rise to the term “PPI-responsive EoE”13, 14. Although PPIs are used primarily to
103
treat esophagitis and gastritis, they have also been shown to ameliorate symptoms, reverse
104
tissue inflammation, and normalize the allergic transcriptome this subset of patients. Moreover,
105
PPI treatment of subjects with PPI-responsive EoE results in a decrease in mast cell numbers
106
with RNA analysis showing a reduction in mast cell transcripts 11.
107
By covalently binding to the cysteine residues of H+/K+ ATPases of the parietal cells,
108
PPIs block the secretion of gastric acid, subsequently changing the pH of the cell and stomach
109
lumen16. Mast cells are known to be sensitive to changes in pH. The granules of the cell are
110
acidic compartments analogous to lysosomes17. Inhibition of proton pumps in mast cells by
111
bafilomycin A1, a selective inhibitor of vacuolar ATPase, affects the acidification of the granules
112
and consequently the processing and activity of granule contents18. Based on these
113
observations, we reasoned that PPIs might similarly affect the pH of mast cell compartments
114
and, consequently, the effector functions of the cells. We tested our hypothesis in in vitro and in
115
vivo, analyzing the effects of PPIs on the functions of cultured mast cells and in mast cell-
116
dependent mouse models of anaphylaxis and food allergy, respectively. Our results provide
117
evidence that PPIs interact with targets that mediate activating signals provided by FcεRI
118
ligation in mast cells, inhibit mediator release and attenuate the physiologic and inflammatory
119
responses to mast cell activation in vivo.
120
Methods
121
Murine mast cell culture. Bone marrow-derived mast cells (BMMCs) were cultured as
122
previously described19. Briefly, BMMCs were derived from bone marrow precursor cells of
123
BALB/c mice. Cells were cultured in RPMI-1640 medium supplemented with 10% FCS (Gibco),
124
100U/ml penicillin (Gibco), 100µg/ml streptomycin (Gibco), 1% Minimum Essential Medium non-
125
essential amino acids (Gibco), 10mM HEPES buffer (Gibco), 55μM 2-mercaptoethanol (Gibco),
126
10μg/ml gentamicin (Life Technologies), and 20ng/mL each of IL-3 and SCF (complete media).
6
127
Once cultures were mature (>90% of cells c-Kit+ FcεRIα+) cytokines were reduced to 10ng/ml
128
(gating strategy for mast cells is provided in Fig E1A). IgE-induced degranulation (LAMP-1
129
expression), cytokine release, calcium flux, protein tyrosine phosphorylation and changes in
130
granule and cytosolic pH were determined as detailed in the Online Repository.
131
Cord blood-derived human mast cell culture. Cord blood-derived mast cells (CBMCs) were
132
generously provided by the laboratory of Dr. J. Boyce (Brigham and Women’s Hospital, Boston,
133
MA). Human mast cells were derived from cord blood mononuclear cells as previously
134
described20. Mature human mast cells were always maintained and cultured in the presence of
135
SCF (100ng/mL). For activation experiments, human mast cells were primed with IL-4 (10ng/mL)
136
for three days in upregulate their surface expression of FcεRI. On the third day of IL-4
137
treatment cells were incubated with human myeloma IgE (100ng/ml; Chemicon International,
138
Tecaluma, CA). Unbound IgE was washed away and cells were stimulated with rabbit anti-
139
human IgE (100ng/ml). Details quantification of degranulation by measuring LAMP-1 expression,
140
and release of histamine, prostaglandin D2 and cytokines are detailed in the Online Repository.
141
Treatment of mast cell cultures with omeprazole. Omeprazole solid (Sigma, catalog number
142
O104) was first activated for 30 minutes in PBS at a pH of 4 to convert it to the active
143
sulfonamide form. An equal volume of complete media was added to the activated omeprazole
144
to neutralize the pH before adding it to cell cultures. Cells were treated with omeprazole at a
145
final concentration of 50µM as previously reported21-23. Drug vehicle (DMSO) was prepared in
146
the same manner, including acidification and neutralization, at equal volumes.
147
Mouse studies. All work was performed under protocols reviewed and approved by Boston
148
Children’s Hospital Institutional Animal Care and Use Committee. All mice used in this study
149
were bred and maintained under specific pathogen-free conditions in individually ventilated
150
cages. To induce food allergy, 6-8-week-old BALB/c mice were sensitized for three consecutive
151
weeks by intraperitoneal injection of 100µg of ovalbumin (OVA; Sigma, catalog number A5503)
7
152
adsorbed to 1.5mg of aluminum hydroxide (Imject Alum, Pierce, Rockford, IL) in 0.2mL of sterile
153
PBS. Following sensitization, animals were divided into two groups: omeprazole- or vehicle-
154
treated. Omeprazole treatment was administered intragastrically at a dose of 12.5mg/kg per
155
mouse (prepared in sterile PBS), as previously described23, and untreated animals were given
156
equal volumes of vehicle (DMSO). Two hours post treatment, the mice were intragastrically
157
challenged with 50mg of ovalbumin. Animals were re-challenged two days later and sacrificed
158
two hours following the second challenge. Tissues were collected for follow-up experiments as
159
described in the Online Repository. For passive systemic anaphylaxis experiments, female
160
BALB/c mice were treated with 1.6mg esomeprazole in 0.2mL (a formulation of the drug for
161
intravenous injection in humans) or with vehicle (PBS) for 4 consecutive days by intraperitoneal
162
injection. On the third day, mice were intraperitoneally sensitized with 10µg IgE anti-DNP (clone
163
SPE-7, Sigma-Aldrich, St. Louis, MO). Two hours after the final treatment mice were challenged
164
intraperitoneally with 75µg DNP-BSA. Body temperature was recorded every 5 minutes for 60
165
minutes via implanted transponders24.
166
Mast cell differentiation assay. To study the effects of omeprazole on the differentiation of
167
progenitor cells to BMMCs, cells were treated with 50µM of omeprazole. Media were changed
168
every three days and differentiation was assessed every six days by measuring the percentage
169
of c-Kit and FcεRIα double-positive cells by flow cytometry.
170
Interleukin 4 (IL-4)-induced mast cell expansion in the small intestine. Mice were injected
171
with complexes of 2µg of IL-4 (Peprotech) and 10µg of anti-IL-4 (Biolegend) on days 0, 3 and 6.
172
Mice were treated with esomeprazole daily from day -1 to day 6 by intraperitoneal injection.
173
Blood and small intestine were collected on day 7.
174
8
175
Results
176
Omeprazole blocks the degranulation of bone marrow-derived murine mast cells and cord
177
blood-derived human mast cells
178
To test the effects of omeprazole on mast cells, we first examined its effects on IgE-
179
mediated mast cell degranulation and cytokine production using cultured cells. Surface
180
expression of lysosomal-associated membrane protein 1 (LAMP-1) following stimulation with
181
antigen for 10 minutes was used as an indicator of the degree of mast cell degranulation. At
182
baseline, in the absence of antigen-specific IgE (IgE anti-TNP), exposure of BMMCs to antigen
183
(TNP-OVA) had no effect, and LAMP-1 expression was minimal (2.293% ± 0.5031%) (Fig 1A).
184
As expected, upon stimulation with TNP-OVA, mast cells sensitized with IgE anti-TNP exhibited
185
nearly a 30-fold increase in LAMP-1 expression on their surface (60.95% ± 5.118%). In contrast,
186
IgE-sensitized BMMCs preincubated with 50µM omeprazole for two hours prior to stimulation
187
with TNP-OVA exhibited markedly suppressed IgE-induced degranulation (8.775% ± 0.7180%
188
LAMP-1+). and histamine release (Fig 1B). A dose response analysis revealed that suppression
189
of degranulation by omeprazole was exerted in a dose-dependent manner over a range of 10 to
190
50µM in BMMCs (Fig E1B). The concentration of omeprazole used did not affect the viability of
191
murine mast cells (Fig E1C).
192
In order to test whether omeprazole similarly affects activation of human mast cells we
193
took advantage of human cord blood-derived mast cells. These were sensitized with myeloma
194
IgE and activated with anti-IgE in the presence or absence of omeprazole. As observed with
195
murine BMMC, LAMP-1 expression and histamine release were suppressed by omeprazole in a
196
dose dependent manner (Fig 2 A and B).
197
Though crosslinking of the IgE receptor is the best-known trigger of mast cell
198
degranulation and the one likely to mediate immunologically specific responses to foods,
199
stimulation through other receptors can also activate mast cells. We next tested if omeprazole
9
200
can block non-IgE receptor-mediated mast cell activation, such as through G-protein coupled
201
adenosine receptors. Stimulation of BMMCs with 100µM adenosine caused a 6-fold increase in
202
LAMP-1 expression compared to unstimulated cells (25.03% ± 1.313% vs 3.847% ± 0.777%,
203
Fig 1C). Treatment with omeprazole prior to adenosine stimulation restrained surface LAMP-1
204
at levels similar to those observed in unstimulated cells (3.310% ± 1.640%).
205
Upon IgE activation, transcription and translation of pro-inflammatory cytokines that
206
contribute to allergic inflammation also occurs in mast cells. To assess these transcriptional
207
changes, we used a predesigned NanoString panel composed of probes for the assessment of
208
genes related to Th2 inflammation25. Transcriptional profiling of allergen-stimulated, IgE-
209
sensitized BMMCs performed two hours following antigen exposure revealed the induction of a
210
number of genes, including some encoding cytokines associated with allergic inflammation (Fig
211
1D). Pretreatment with omeprazole at 50µM for two hours attenuated the IgE:FcεRI-induced
212
expression in BMMCs of pro-inflammatory genes such as, CCL5, IL-1β, IL-18, TGFβ, and IL-6
213
as well as IL-13 which is specifically related to type 2 inflammation. We further investigated the
214
effect of omeprazole on select mast-cell-specific cytokines that are known to be synthesized de
215
novo and released from the cells. Six hours after allergen challenge, IgE-sensitized BMMCs
216
produced significantly greater amounts of IL-4, IL-6, IL-13 and TNF-α than unstimulated cells
217
(Fig 1E-H). In cells pretreated with omeprazole, the production and release of these cytokines
218
was reduced. Likewise, in human mast cell cultures we observed a dose responsive decrease in
219
the secretion of PGD2 and cytokines, IL-5 and IL-13 (Fig 2C-E). Taken together, these results
220
show that omeprazole treatment blocks the immediate release of preformed mediators
221
contained in mast cell granules, the FcεRI-induced transcriptional program and the eventual
222
production and secretion of cytokines.
10
223 224
Omeprazole treatment dampens activating signaling pathways in mast cells To obtain a better understanding of how omeprazole attenuates mast cell activation, we
225
investigated early events in the well-characterized signaling cascade activated by FcεRI
226
crosslinking. This pathway drives both degranulation with release of preformed mediators of
227
anaphylaxis and the upregulation of inflammatory cytokine transcription and secretion4. To test
228
the effect of omeprazole on signaling, we stimulated IgE anti-TNP bound BMMCs via FcεRI
229
crosslinking with TNP-OVA for 2.5 to 30-minutes with or without pretreatment with omeprazole.
230
The degree of phosphorylation of upstream (SYK) and downstream (ERK) protein kinases was
231
measured to assess the strength of activation of the pathway. As previously extensively
232
described by others26, 27, crosslinking of the IgE receptor resulted in phosphorylation of both
233
SYK and ERK detectable as early as 2.5 minutes after stimulation (Fig 3A). Quantification of the
234
ratio in signal intensities of phosphorylated to total protein showed that the IgE-induced
235
phosphorylation was significantly weaker at all time points in omeprazole-treated cells following
236
antigen stimulation.
237
Another early signal critical for degranulation and cytokine production by mast cells is
238
increased cytosolic calcium. FcεRI crosslinking is associated with the release of calcium from
239
endoplasmic reticulum stores into the cytosol. This is necessary for granule fusion with the
240
plasma membrane and exocytosis of the contents28. Blocking calcium increase can disrupt the
241
full range of mast cell activation phenotypes following IgE receptor crosslinking. Using the Ca2+-
242
chelating fluorophore, Fluo 4, we observed a robust increase in cytosolic Ca2+ ([Ca2+]i)
243
immediately after antigen stimulation (Fig 3B). Compared to IgE-sensitized untreated BMMCs,
244
omeprazole-treated mast cells exhibited a dramatic suppression of increase in [Ca2+]i in a dose
245
responsive manner. These results show that omeprazole exerts suppressive effects on several
246
of the key signaling events elicited by cross-linking of FcεRI.
11
247 248
Omeprazole dampens passive IgE-mediated systemic anaphylaxis Systemic anaphylaxis is the most dramatic clinical outcome of an allergic reaction and is
249
caused by the multi-tissue effects of mediators released by activated mast cells and basophils
250
following crosslinking of the cell surface IgE receptor24, 29-31. Our in vitro data showed that
251
treatment of mast cells with omeprazole blocked their IgE:FcεRI-induced degranulation. Using a
252
model of passive systemic anaphylaxis, we assessed if the inhibition observed in vitro is
253
recapitulated in vivo and whether diminished mast cell activation in the presence of omeprazole
254
is accompanied by attenuation of the physiologic manifestations of anaphylaxis. Mice were
255
passively sensitized with anti-DNP IgE and challenged intraperitoneally with DNP-BSA.
256
Changes in core body temperature were measured as an indicator of vasodilation (with
257
increased cutaneous blood flow and cooling) and shock. As expected, untreated BALB/c mice
258
exhibited a significant drop in temperature following antigen challenge (Fig 4A). To investigate
259
the effects of proton pump inhibitors on anaphylaxis, we used esomeprazole which is a single (S)
260
enantiomer of omeprazole and available as an injectable formulation. Esomeprazole treatment
261
for 4 days prior to challenge significantly dampened the rate of temperature decline to less than
262
half of control. Release of the mast cell granule constituent protease, MCPT-1, and histamine
263
were also reduced to less than 50% that observed in esomeprazole-treated animals (Fig 4B
264
and C).
265
Treatment with omeprazole alters the pH in the cytosol and subcellular compartments of the
266
mast cell
267
Based on the known action of omeprazole as a proton pump inhibitor in gastric parietal
268
cells, we sought to determine whether it affects the pH of the acidic storage compartments and
269
cytosol of mast cells. Using Lysosensor Blue, a dye that accumulates in acidic intracellular
270
compartments, we assessed the effects of omeprazole on pH in mast cell granules. As a control
271
we treated BMMCs with bafilomycin, a blocker of V-ATPase that has previously been shown to
272
increase the pH of mast cell granules18. In comparison to vehicle-treated cells, the fluorescence 12
273
intensity of Lysosensor Blue in both omeprazole- and bafilomycin A1-treated cells was
274
significantly decreased, indicating that the intracellular granule compartment pH was increased
275
(Fig 5A).
276
In an alternate approach to detecting changes in the pH of intracellular vesicles, we
277
performed a FITC-dextran quenching experiment. FITC-dextran is taken up into endosomes
278
which fuse with acidic intracellular vesicles. The FITC signal is quenched by the acidic pH within
279
those compartments. Chloroquine, a blocker of endosome-lysosome fusion, was included as a
280
control for this experiment. Compared to untreated cells, both chloroquine- and omeprazole-
281
treated cells exhibited higher FITC fluorescence intensity than did untreated cells, consistent
282
with an increased pH in those normally acidic organelles (Fig 5B). As granules represent a
283
significant fraction of mast cell volume, we reasoned that inhibition of proton accumulation in
284
those compartments might be accompanied by an opposite effect, namely reduction of pH, in
285
the cytosol. We tested this hypothesis using pHrodoGreen, a dye that becomes trapped in the
286
cytosol and whose fluorescence is inversely proportional to the pH of the cytosol. The
287
pHrodoGreen assay showed that BMMCs treated with omeprazole had a more acidic cytosol
288
environment compared to untreated cells (Fig 5C). Notably, the cytosolic pH was unaffected by
289
pretreatment with bafilomycin. Furthermore, mast cells treated with bafilomycin have previously
290
been characterized as having altered morphology with swollen granules that have diminished
291
staining with May Grünwald/Giemsa (MGG) 18. In our hands, we observed similar changes (Fig
292
E2). This dramatic phenotype was not observed in omeprazole treated cells. However, unlike
293
the DMSO-treated control cells, they did exhibit a few punctate MGG negative regions (Fig E2).
294
These findings suggest that the effects of omeprazole on mast cell function might involve the
295
target of bafilomycin, the V-ATPase or a related proton pump.
296
Omeprazole blocks mast cell differentiation in vitro and in vivo
297
As our findings had shown that treatment with omeprazole affects the signaling
298
pathways and activation of mature mast cells, we next evaluated whether they might affect the
13
299
differentiation of precursor cells into mature mast cells. Culturing murine bone marrow stem
300
cells in the presence of IL-3 and SCF induces differentiation into mature mast cells. Mast cell
301
maturation in such cultures can be assessed by monitoring the appearance of c-Kit and FcεRIα
302
double-positive cells (Fig E1A). Under normal growth conditions, more than 80% (86.65% ±
303
1.350%) of cells in such cultures acquire the double-positive mast cell phenotype after five
304
weeks (Fig 6A). However, less than 15% (12.75% ± 0.75%) of cells cultured in the presence of
305
50µM of omeprazole were mature. In order to determine whether omeprazole might similarly
306
affect mast cell differentiation in vivo, we took advantage of an IL-4-driven model. We and
307
others have reported that exogenous administration of IL-4 as an immune complex with anti-IL4
308
(IL4C) to prolong the cytokine’s half-life (three injections over one week), elicits an expansion of
309
intestinal mast cells (Fig 6B)19, 32. Using this approach along with flow cytometric enumeration of
310
intestinal mast cells, we found that daily treatment with esomeprazole by intraperitoneal
311
injection decreased IL-4-induced mast cell expansion (Fig 6B). These in vivo data corroborate
312
our tissue culture findings in showing that omeprazole can block the maturation of precursor
313
cells into mast cells.
314
Treatment with omeprazole blocks allergic responses in a mast cell-dependent mouse model of
315
food allergy
316
In addition to driving immediate hypersensitivity reactions, mast cells are known to play
317
key roles in initiating and sustaining allergic inflammatory responses19, 31, 33, 34. They provide a
318
critical tissue source of cytokines that activate mucosal antigen presenting cells, as well as IL-4
319
which primes and consolidates T-helper 2 responses. Our analysis of the effects of omeprazole
320
on cultured mast cells showed a dramatic suppression of cytokine production following IgE-
321
mediated activation by allergen (Fig 1C-D). Our final aim was to investigate whether this effect
322
would be reflected in vivo by an altered response to food allergen. To test this possibility, we
323
adapted a well-characterized mast cell-dependent model of food allergy initially described by
14
324
Rothenberg and colleagues (Fig 7A)31. In this model, mice sensitized intraperitoneally with
325
allergen and then repeatedly enterally challenged to develop strong IgE responses and food
326
sensitivity. The response to repeated allergen challenge was accompanied by intestinal mast
327
cell expansion and a correlation has been demonstrated between the number of mast cells in
328
the small intestine and the severity of allergic reactions29.
329
Examination of chloroacetate esterase (CAE)-stained jejunal sections revealed a robust
330
expansion of mast cells following ovalbumin challenge in mice that had been sensitized to the
331
antigen (Fig 7D), but not in unsensitized controls (Fig 7B and C). This was reduced by
332
omeprazole treatment (Fig 7E). Enumeration of mast cells in sections from multiple mice
333
confirmed these observations (Fig 7F). Following allergen challenge by gavage, omeprazole-
334
treated mice also exhibited greatly reduced release of MCPT-1 into the serum (Fig 7G). The
335
increase in total and allergen specific IgE (OVA-IgE) in the serum following antigen challenge in
336
sensitized mice was attenuated in omeprazole treated mice (Fig 7 H and I). mRNA expression
337
of several cytokines associated with allergic inflammation, IL-4, IL-5, IL-13 and TNF-α, was also
338
reduced in omeprazole-treated, allergen-sensitized and challenged animals compared to
339
untreated animals (Fig 7 J-M). These findings indicate that omeprazole affects pathways
340
involved in mast cell differentiation and activation in response to ingested allergen and alters the
341
intestinal cytokine environment.
342
15
343 344
Discussion Our study provides strong evidence that the proton pump inhibitor omeprazole exerts
345
significant effects on pathways regulating mast cell homeostasis and function and can modulate
346
mast cell-dependent allergic disease phenotypes. We demonstrate that omeprazole attenuates
347
mast cell activation induced by FcεRI crosslinking and by adenosine, with decreased
348
degranulation and cytokine responses. Suppression occurs early in the signaling cascade with
349
decreased phosphorylation of the FcεRI-proximal tyrosine protein kinase SYK, as well as the
350
downstream serine protein kinase ERK, along with attenuation of increased [Ca2+]i. The
351
physiologic impact of the inhibitory effects of omeprazole on mast cell function were clearly
352
evident in in vivo models of anaphylaxis and mast cell-dependent food allergy. Furthermore, we
353
observe that omeprazole at 200µM can also block the activation of basophils, another major cell
354
involved in allergic phenotypes and anaphylaxis (Fig E3).
355
PPIs have been reported to have clinical benefit in eosinophilic esophagitis35. It was
356
initially postulated that the omeprazole responsiveness of some subjects with distal esophageal
357
eosinophilia indicated that their esophageal inflammation was triggered by acid reflux and not
358
food-induced type 2 inflammation. However, there is evidence that the mechanism of action of
359
omeprazole can be attributed to anti-type 2 inflammatory effects. For instance, oral
360
administration of omeprazole prior to allergen instillation into mouse airways reduced the
361
recruitment of inflammatory cells, including eosinophils21. In PPI-responsive EoE, there is now
362
general agreement, based largely on transcriptomic analyses, that omeprazole treatment
363
attenuates type 2 inflammation11. There is evidence that omeprazole impacts a range of
364
signaling intermediates. For instance, studies on epithelial cells have shown that omeprazole
365
decreases IL-4-, IL-13-, and IFN-ß-induced phosphorylation of STAT621.
366
The mechanism whereby omeprazole exerts its effects on mast cells remains to be
367
determined. On gastric parietal cells, omeprazole binds to and inhibits the gastric ATPase.
16
368
However, we have found that expression of this proton pump is quite low in mast cells. We
369
speculate that omeprazole could be binding to the V-ATPase, the target of bafilomycin A1.
370
Treatment with bafilomycin has previously been shown to alter mast cell morphology, inhibit the
371
maturation of mast cell granules and processing granule contents and impair mast cell
372
activation18. Furthermore, it has been reported that omeprazole binds to the catalytic sites of V-
373
ATPases purified from adrenal chromaffin granules and reconstituted into liposomes36. In our
374
hands, bafilomycin and omeprazole exerted similar effects on mast cell granule pH (Fig 5 A).
375
Yet, we have reason to speculate that additional targets for omeprazole might exist in mast cells
376
as omeprazole, but not bafilomycin, affects cytosolic pH (Fig 5C) and changes observed in
377
granule morphology are not identical by both drugs (Fig E2).
378
Several studies describe a link between the use of acid suppression and the
379
development of food allergies37. Elevating the pH of the stomach can decrease the activity of
380
stomach enzymes, such as pepsin, that are needed for protein digestion. It is hypothesized that
381
incomplete digestion might allow larger peptides harboring allergenic epitopes to enter the small
382
intestine and induce antigen sensitization38. In our studies, omeprazole had an anti-allergic
383
effect, but it was only administered once sensitization was established. Animals are sensitized
384
to either ovalbumin or with anti-DNP IgE via the intraperitoneal route, and so our experimental
385
design allows us to study the effects of omeprazole on allergen challenge-induced inflammation
386
and avoid its effects on sensitization.
387
Other research groups have also described anti-inflammatory effects of PPIs. For
388
example, omeprazole has been shown to decrease the migration of polymorphonuclear
389
neutrophils (PMN) towards the chemoattractant IL-839. Correlating with this decrease in
390
migration, a decrease in the expression of adhesion molecules CD11b and CD18 on PMNs has
391
also been observed40. Furthermore, production of IL-8 by epithelial cells following stimulation
392
with IL-1β is also inhibited39. These effects are beneficial in attenuating the inflammation
393
observed in infections, such as from Helicobacter pylori. In models of sepsis, injection with 17
394
omeprazole pre- or post-endotoxic shock can increase survival time by limiting cytokine
395
production23. Treatment of monocytes and animals with omeprazole inhibits TLR agonist-
396
induced secretion of IL-1β and TNF-α23.
397
Our studies establish that omeprazole can interfere with pathways involved in mast cell
398
differentiation and activation as well as allergic inflammation. These observations provide insight
399
into the beneficial effects of this agent in some patients with EoE. In future studies, it will be of
400
great interest to identify the specific pharmacologic target(s) of omeprazole in mast cells, and to
401
consider the development of higher affinity and more specific small molecule inhibitors.
402 403 404 405 406
Acknowledgements The authors are grateful to Dr. Joshua Boyce and Dr. Chunli (Lily) Feng for providing
407
human cord blood derived- mast cells. We are also grateful to Dr. Takashi Fujimura, Ms.
408
Catherine Lavallee for advice and assistance with animal studies, and to Ms. Yvonne Nguyen
409
for technical assistance.
18
410
Figure Legends
411
Figure 1. Omeprazole blocks IgE-induced mast cell degranulation, cytokine secretion and
412
transcriptional changes. A) Representative histogram and bar plots for LAMP-1 expression by
413
BMMCs following sensitization with IgE anti-TNP, treatment with drug vehicle (DMSO) or
414
omeprazole, and challenge with TNP-OVA for 10 minutes. B) Histamine release into culture
415
supernatant from IgE anti-TNP sensitized BMMCs following activation with TNP-OVA for 5
416
minutes. C) LAMP-1 expression of BMMCs following activation with adenosine. D) Heatmap of
417
transcriptional changes in IgE sensitized BMMCs pre-treated with and without omeprazole
418
followed by antigen stimulation for two hours. Direct mRNA counts as measured by the
419
nCounter® Digital Analyzer System (NanoString) were normalized to internal positive, negative
420
and housekeeping gene controls and presented as standard deviation from the row mean. E-H)
421
Release of cytokines (IL-4, IL-6, IL-13, TNF-α) by IgE-sensitized BMMCs following omeprazole
422
treatment and antigen stimulation for six hours. Statistical analysis by one-way analysis of
423
variance. Data are shown for one experiment representative of two independent experiments for
424
LAMP1, histamine and cytokine secretion assay; transcriptional profiling by NanoString was
425
performed once. *P<0.05, **P<0.01, ***P<0.001, and ****P<0.0001
426
Figure 2. Omeprazole blocks IgE-induced human mast cell degranulation and cytokine
427
secretion. Bar plots showing LAMP-1 expression (A) and histamine release (B) by human mast
428
cells following sensitization with IgE, treatment with drug vehicle (DMSO) or omeprazole, and
429
challenge with anti-IgE for 10 and 5 minutes, respectively. Secretion of PGD2 (C), IL-5 (D) and
430
IL-13 (E) by IgE-sensitized human following omeprazole treatment and stimulation with anti-IgE
431
for six hours. Statistical analysis by one-way analysis of variance. *P<0.05, **P<0.01,
432
***P<0.001, and ****P<0.0001.
433
Figure 3. Effect of omeprazole on calcium flux and signaling pathways downstream of FcεRI. A)
434
Representative SYK and ERK phosphorylation blots and compiled ratios of phospho
19
435
protein/total protein intensities in IgE-sensitized BMMCs treated or untreated (DMSO) with
436
omeprazole for two hours. Statistics calculated following two-way ANOVA. B) Mobilization of
437
calcium from intracellular stores following antigen stimulation of IgE-sensitized BMMCs treated
438
with omeprazole for two hours or untreated (DMSO). P-value calculated by two-way ANOVA
439
between DMSO and omeprazole (50 µM). Data are shown for one experiment each
440
representative of at least 2 independent experiments.
441
Figure 4. Effect of omeprazole on passive systemic anaphylaxis. A) Change in core body
442
temperature of mice treated with omeprazole or drug vehicle, sensitized with SPE-7, and
443
challenged with DNP-BSA. Statistical analysis by two-way analysis of variance. B) Serum
444
concentration of MCPT-1 in animals from PSA experiment at endpoint. C) Plasma histamine
445
concentration in animals 5 minutes after allergen challenge. Statistical analysis by unpaired t-
446
test. Data are from one experiment representative of 2 independent experiments with n = 4-5 for
447
temperature drop and MCPT1 levels; histamine levels are from one experiment with n = 3-4
448
animals. **P<0.01 and ***P<0.001
449
Figure 5. pH of cellular compartments following omeprazole treatment of BMMCs. A)
450
Measurement of changes in pH by of intracellular vesicles by Lysosensor blue following
451
treatment with omeprazole and bafilomycin. B) Loss of quenching of FITC-dextran in BMMCs
452
treated with omeprazole or chloroquine. C) Measurement of changes in cytosolic pH by
453
pHrodoGreen following treatment with omeprazole, chloroquine or bafilomycin. Statistical
454
analysis by one-way analysis of variance. Data are shown from one representative of two
455
experiments. MFI, mean fluorescence intensity. *P<0.05, **P<0.01, ***P<0.001 , and
456
****P<0.0001
457
Figure 6. Omeprazole inhibits differentiation of myeloid precursor cells to mast cells. A)
458
Maturation of precursor cells to BMMCs in the presence or absence of omeprazole. Statistical
459
analysis by two-way analysis of variance. B) Percentage of small intestinal mast cells among
460
CD45+ cells in mice intraperitoneally injected with immune complexes of IL-4 and anti-IL-4 (IL4C) 20
461
to induce mastocytosis and intragastrically treated with omeprazole. For in vitro mast cell
462
differentiation, data is from one experiment involving three independent cultures; the in vivo
463
experiment was repeated twice, and data are shown for one experiment. *P<0.05
464
Figure 7. Treatment with omeprazole blocks inflammation in a mouse model of ovalbumin-
465
induced food allergy. A) Experimental design (S: sensitization, C: challenge, PPI: proton pump
466
inhibitor). B-E) Representative CAE-stained histological sections of small intestine from mice
467
from each experimental group. F) Summary of average counts of mast cells per high power field.
468
G) Serum concentrations of MCPT-1 at the experimental endpoint. H and I) Serum total and
469
OVA-specific IgE concentrations at experimental endpoint. J-M) Expression of cytokine
470
transcripts in the small intestines of mice from each experimental group at study endpoint. UT:
471
Untreated, OM: Omeprazole, OVA: Ovalbumin. Data are shown are from one experiment
472
representative of three independent experiments with n = 3-6 per group. Statistical analysis by
473
one-way analysis of variance. *P<0.05, **P<0.01
474
21
475
References
476
1.
477 478
treatment. Annu Rev Med 2009; 60:261-77. 2.
479 480
Sicherer SH, Sampson HA. Food allergy: Epidemiology, pathogenesis, diagnosis, and treatment. J Allergy Clin Immunol 2014; 133:291-307; quiz 8.
3.
481 482
Sicherer SH, Sampson HA. Food allergy: recent advances in pathophysiology and
Renz H, Allen KJ, Sicherer SH, Sampson HA, Lack G, Beyer K, et al. Food allergy. Nat Rev Dis Primers 2018; 4:17098.
4.
483
Oettgen HC, Burton OT. IgE receptor signaling in food allergy pathogenesis. Curr Opin Immunol 2015; 36:109-14.
484
5.
Furuta GT, Katzka DA. Eosinophilic Esophagitis. N Engl J Med 2015; 373:1640-8.
485
6.
O'Shea KM, Aceves SS, Dellon ES, Gupta SK, Spergel JM, Furuta GT, et al.
486 487
Pathophysiology of Eosinophilic Esophagitis. Gastroenterology 2018; 154:333-45. 7.
488 489
Spergel J, Aceves SS. Allergic components of eosinophilic esophagitis. J Allergy Clin Immunol 2018; 142:1-8.
8.
Pelz BJ, Wechsler JB, Amsden K, Johnson K, Singh AM, Wershil BK, et al. IgE-
490
associated food allergy alters the presentation of paediatric eosinophilic esophagitis. Clin
491
Exp Allergy 2016; 46:1431-40.
492
9.
Hsu Blatman KS, Gonsalves N, Hirano I, Bryce PJ. Expression of mast cell-associated
493
genes is upregulated in adult eosinophilic esophagitis and responds to steroid or dietary
494
therapy. J Allergy Clin Immunol 2011; 127:1307-8 e3.
495
10.
Niranjan R, Mavi P, Rayapudi M, Dynda S, Mishra A. Pathogenic role of mast cells in
496
experimental eosinophilic esophagitis. Am J Physiol Gastrointest Liver Physiol 2013;
497
304:G1087-94.
498
11.
Wen T, Dellon ES, Moawad FJ, Furuta GT, Aceves SS, Rothenberg ME. Transcriptome
499
analysis of proton pump inhibitor-responsive esophageal eosinophilia reveals proton
500
pump inhibitor-reversible allergic inflammation. J Allergy Clin Immunol 2015; 135:187-97.
22
501
12.
502 503
Abe Y, Sasaki Y, Yagi M, Yaoita T, Nishise S, Ueno Y. Diagnosis and treatment of eosinophilic esophagitis in clinical practice. Clin J Gastroenterol 2017; 10:87-102.
13.
Liacouras CA, Furuta GT, Hirano I, Atkins D, Attwood SE, Bonis PA, et al. Eosinophilic
504
esophagitis: updated consensus recommendations for children and adults. J Allergy Clin
505
Immunol 2011; 128:3-20 e6; quiz 1-2.
506
14.
Lucendo AJ, Arias A, Molina-Infante J. Efficacy of Proton Pump Inhibitor Drugs for
507
Inducing Clinical and Histologic Remission in Patients With Symptomatic Esophageal
508
Eosinophilia: A Systematic Review and Meta-Analysis. Clin Gastroenterol Hepatol 2016;
509
14:13-22 e1.
510
15.
Dohil R, Newbury RO, Aceves S. Transient PPI responsive esophageal eosinophilia may
511
be a clinical sub-phenotype of pediatric eosinophilic esophagitis. Dig Dis Sci 2012;
512
57:1413-9.
513
16.
514 515
J Neurogastroenterol Motil 2013; 19:25-35. 17.
516 517
Johnson RG, Carty SE, Fingerhood BJ, Scarpa A. The internal pH of mast cell granules. FEBS Lett 1980; 120:75-9.
18.
518 519
Shin JM, Kim N. Pharmacokinetics and pharmacodynamics of the proton pump inhibitors.
Pejler G, Hu Frisk JM, Sjostrom D, Paivandy A, Ohrvik H. Acidic pH is essential for maintaining mast cell secretory granule homeostasis. Cell Death Dis 2017; 8:e2785.
19.
Burton OT, Darling AR, Zhou JS, Noval-Rivas M, Jones TG, Gurish MF, et al. Direct
520
effects of IL-4 on mast cells drive their intestinal expansion and increase susceptibility to
521
anaphylaxis in a murine model of food allergy. Mucosal Immunol 2013; 6:740-50.
522
20.
Ochi H, Hirani WM, Yuan Q, Friend DS, Austen KF, Boyce JA. T helper cell type 2
523
cytokine-mediated comitogenic responses and CCR3 expression during differentiation of
524
human mast cells in vitro. J Exp Med 1999; 190:267-80.
525 526
21.
Cortes JR, Rivas MD, Molina-Infante J, Gonzalez-Nunez MA, Perez GM, Masa JF, et al. Omeprazole inhibits IL-4 and IL-13 signaling signal transducer and activator of
23
527
transcription 6 activation and reduces lung inflammation in murine asthma. J Allergy Clin
528
Immunol 2009; 124:607-10, 10 e1.
529
22.
Zhang X, Cheng E, Huo X, Yu C, Zhang Q, Pham TH, et al. Omeprazole blocks STAT6
530
binding to the eotaxin-3 promoter in eosinophilic esophagitis cells. PLoS One 2012;
531
7:e50037.
532
23.
Balza E, Piccioli P, Carta S, Lavieri R, Gattorno M, Semino C, et al. Proton pump
533
inhibitors protect mice from acute systemic inflammation and induce long-term cross-
534
tolerance. Cell Death Dis 2016; 7:e2304.
535
24.
Mathias CB, Hobson SA, Garcia-Lloret M, Lawson G, Poddighe D, Freyschmidt EJ, et al.
536
IgE-mediated systemic anaphylaxis and impaired tolerance to food antigens in mice with
537
enhanced IL-4 receptor signaling. J Allergy Clin Immunol 2011; 127:795-805 e1-6.
538
25.
Platzer B, Baker K, Vera MP, Singer K, Panduro M, Lexmond WS, et al. Dendritic cell-
539
bound IgE functions to restrain allergic inflammation at mucosal sites. Mucosal Immunol
540
2015; 8:516-32.
541
26.
542 543
Immunol Rev 2009; 228:149-69. 27.
544 545
Suzuki R, Scheffel J, Rivera J. New insights on the signaling and function of the highaffinity receptor for IgE. Curr Top Microbiol Immunol 2015; 388:63-90.
28.
546 547
Gilfillan AM, Rivera J. The tyrosine kinase network regulating mast cell activation.
Holowka D, Wilkes M, Stefan C, Baird B. Roles for Ca2+ mobilization and its regulation in mast cell functions: recent progress. Biochem Soc Trans 2016; 44:505-9.
29.
Ahrens R, Osterfeld H, Wu D, Chen CY, Arumugam M, Groschwitz K, et al. Intestinal
548
mast cell levels control severity of oral antigen-induced anaphylaxis in mice. Am J Pathol
549
2012; 180:1535-46.
550
30.
Lin RY, Schwartz LB, Curry A, Pesola GR, Knight RJ, Lee HS, et al. Histamine and
551
tryptase levels in patients with acute allergic reactions: An emergency department-based
552
study. J Allergy Clin Immunol 2000; 106:65-71.
24
553
31.
Brandt EB, Strait RT, Hershko D, Wang Q, Muntel EE, Scribner TA, et al. Mast cells are
554
required for experimental oral allergen-induced diarrhea. J Clin Invest 2003; 112:1666-
555
77.
556
32.
Madden KB, Whitman L, Sullivan C, Gause WC, Urban JF, Jr., Katona IM, et al. Role of
557
STAT6 and mast cells in IL-4- and IL-13-induced alterations in murine intestinal epithelial
558
cell function. J Immunol 2002; 169:4417-22.
559
33.
Galli SJ, Tsai M. IgE and mast cells in allergic disease. Nat Med 2012; 18:693-704.
560
34.
Mukai K, Tsai M, Saito H, Galli SJ. Mast cells as sources of cytokines, chemokines, and
561 562
growth factors. Immunol Rev 2018; 282:121-50. 35.
563 564
Bohm M, Richter JE. Treatment of eosinophilic esophagitis: overview, current limitations, and future direction. Am J Gastroenterol 2008; 103:2635-44; quiz 45.
36.
Moriyama Y, Patel V, Ueda I, Futai M. Evidence for a common binding site for
565
omeprazole and N-ethylmaleimide in subunit A of chromaffin granule vacuolar-type H(+)-
566
ATPase. Biochem Biophys Res Commun 1993; 196:699-706.
567
37.
Jordakieva G, Kundi M, Untersmayr E, Pali-Scholl I, Reichardt B, Jensen-Jarolim E.
568
Country-wide medical records infer increased allergy risk of gastric acid inhibition. Nat
569
Commun 2019; 10:3298.
570
38.
571 572
Pali-Scholl I, Jensen-Jarolim E. Anti-acid medication as a risk factor for food allergy. Allergy 2011; 66:469-77.
39.
Handa O, Yoshida N, Fujita N, Tanaka Y, Ueda M, Takagi T, et al. Molecular
573
mechanisms involved in anti-inflammatory effects of proton pump inhibitors. Inflamm Res
574
2006; 55:476-80.
575
40.
Yoshida N, Yoshikawa T, Tanaka Y, Fujita N, Kassai K, Naito Y, et al. A new mechanism
576
for anti-inflammatory actions of proton pump inhibitors--inhibitory effects on neutrophil-
577
endothelial cell interactions. Aliment Pharmacol Ther 2000; 14 Suppl 1:74-81.
578
25
Figure 1
1000
) µM
SO M
(5 0
ul
ep
U
ra
ns
zo
le
tim
D
at ed
) µM (5 0 le
zo ra
O m
O m ep
TNF-α (pg/ml)
200
100
) 0µ
M
M SO zo
le
(5
PTN o
ra
le zo
o
O VA
) 0µ
M
SO
TN
(5
N
ra ep
N
(5 e zo l
o N
****
300
0
PO VA
) 0µ
M
SO M
PTN
D
O VA
) (5 0 e
ra zo l ep O m
TNF-α
H
0
µM
SO M D
PO VA TN o N
M SO
P-
(5
TN
200
0
0
D
O VA
M ) 0µ
SO M
200
400
M
2
400
****
D
4
600
600
ra
IL-6 (pg/ml)
IL-4 (pg/ml)
6
***
800
IL-13
ep
***
8
G
IL-6
O m
F
IL-4
O m
E
o
+
N
+
+
IL-13 (pg/ml)
-
+
le
+
zo
+
ra
-
ep
-
0
O m
TNP-OVA
D
E Ig o N
Omeprazole (50!M)
10
0
0
IgE
20
D
20
2000
TNP-Ova TNP-OVA Omeprazole Omepazole
***
ep
40
30
O m
60
D
Adenosine
**
3000
****
Histamine (nM)
LAMP1+ (% of Live BMMCs)
80
C
Histamine
-
+
-
+
-
-
+
+
IgE lo
2 Nano
Relb Tnfa Ccl7 Il1b Ccl5 Tgfb1 Il13 Il4 Nrf1 Ccl2 Csf2 Mcpt8 Socs2 Il6 Raldh1a1 Cd103 Shfm1 Il25 Fcer1a Cpa3 Fcer1b Ahrr Cxcr5 Ptgs2 ltc4s Il15 Arnt Pdl1 Slc6a4 Ahr Mcpt2 Mcpt1 Tlr4 Nqo1 Ccl3 Clc4 Pla2g12a
3 IL1b
-Log10 (p-value)
B
LAMP1
LAMP1+ (% of Live BMMCs)
A
2
IL13
1
-4
-2
L
Most signifi
Gene Symbo
CPA3
TGFb1 Il1b
Most signif
Gene Symbo Tlr4 Nqo1 -3
-1.5
0
1.5
3
Il15
o an
IL-5
**** **** ****
60
40
20
E
0
200
0
**** **** ****
4000
2000
IL-13
**** **** ****
150
100
50
0
an tiO Ig m E ep D ra O M z m ep ole SO O razo (50 m µM ep le ( ) 1 ra zo 0 0 µ le M ) (2 00 µM )
Histamine
o
6000
PGD2 (pg/mL)
B
N
an tiO Ig m E ep D ra O M z m ep ole SO O razo (50 m µM ep le ( 10 ) ra zo 0µ le M ) (2 00 µM )
o
20
tiO Ig m E ep D ra O M z m ep ole SO O razo (50 m µM ep le ( ) 1 ra zo 00µ le M ) (2 00 µM )
40
N
60
an
80
* **** ****
o
80
Histamine (nM)
LAMP-1
N
o
an tiO Ig m E ep D ra O M z m ep ole SO O razo (50 m µM ep le ( ) 1 ra zo 00µ le M ) (2 00 µM )
N
A
IL-13 (pg/mL)
D
tiO Ig m E ep D ra O M z m ep ole SO O razo (50 m µM ep le ( ) 1 ra zo 00µ le M ) (2 00 µM )
N
IL-5 (pg/mL)
LAMP1+ (% of Live CBMCs)
Figure 2 C PGD2
8000
0
* * **
6000
4000
2000 0
Figure 3
DMSO
A
Minutes
0
2.5
5
10
Omeprazole 20
30
0
2.5
5
10
20
B
30
Percent change [(F-F0)/F0] *100
Total ERK β-actin
DMSO Omeprazole (50uM)
40 20
Fold increase from time 0
p<0.01
60
20
40
DMSO Omeprazole (50uM)
p< 0.0001 30
0 20 10
100 DMSO Ionomycin
Time (mins)
30
20
10
5
5 2.
0
30
20
10
5
5 2.
0
200
300
Time (s)
0
0
0
Fold increase from time 0
40
ERK
SYK
Time (mins)
p<0.0001
P-SYK Total SYK β-actin P-ERK
80
60
Omeprazole (1 µM) Omeprazole (10 µM) Omperazole (50 µM)
Figure 4 B
1
400
MCPT-1
-2
p < 0.0001
-3 PBS Esomeprazole
-4 -5
0
5
10
15
20
25 30 35 Time (min)
40
45
50
55
60
300 200 100 0
8000
**
-1
MCPT-1 (ng/ml)
Temperature (°C)
0
C
Histamine (nM)
A
PBS
Esomeprazole
Histamine
***
6000 4000 2000 0
PBS
Esomeprazole
Ba
fil o
in
ne yc
ui
(2
15000
)
M
)
) M
M 0u 0n
(5
0u
SO
****
m
oq
(5
M
0
or
le
200
MFI PhrodoGreen (Live cKit+FcεRIα+)
400
D
)
uM
)
600
zo
(5 0
M
**
ra
ne
0u
****
hl
ui
(5
800
C
C
oq
le
****
ep
or
SO
FITC Dextran
O m
hl
M
B
C
zo
0
D
20000
MFI (FITC dextran)
40000
ra
)
)
M
nM
0u (2 0
(5
* **
ep
A1
le
M SO
Lysosensor Blue
O m
in
zo
D
60000
yc
m
ra
A
Ba fil o
ep
O m
MFI (Lysosensor Blue)
Figure 5 pHrodoGreen ****
****
10000
5000
0
Figure 6
le zo
Untreated Omeprazole (50uM)
ra
Days
0.0
ep
6 12 18 24 30 36
O m
0
IL 4C
0
0.2
+
20
0.4
C
40
0.6
IL 4
60
*
0.8
ve
80
B
ai
p<0.0001
N
100
% of Lin-cKit+FcεRIα+ of CD45+ cells
% of cKit+FcεRIα+ of live cells
A
Figure 7 A
S1
S2
S3
C1 C2 PPI
DAY 0
7
14
21 23 H
G
*
4000
Total IgE (ng/ml)
100
Total IgE
80 60 40 20 0
3000
2000 1000
1000
0
**
30 20 10 0 PBS OVA PBS OVA UT UT OM OM OVA OVA OVA OVA
PBS OVA PBS OVA UT UT OM OM OVA OVA OVA OVA
PBS OVA PBS OVA UT UT OM OM OVA OVA OVA OVA
K
IL-5 600
L
**
400
200
0 PBS OVA PBS OVA UT UT OM OM OVA OVA OVA OVA
IL-13 3000
**
2000
0
IL-4 40
OVA-IgE
**
3000
PBS OVA PBS OVA UT UT OM OM OVA OVA OVA OVA
J
I OVA-IgE (ng/ml)
Mast cell count
M
**
2000
1000
0
TNF-α 20
Relative expression
E OVA-OM-OVA
F
Relative expression
OVA-UT-OVA
C PBS-OM-OVA
Relative expression
D
PBS-UT-OVA
Relative expression
B
Average number of MC/HPF
HARVEST
**
15 10 5 0
PBS OVA PBS OVA UT UT OM OM OVA OVA OVA OVA
PBS OVA PBS OVA UT UT OM OM OVA OVA OVA OVA
Figure E1 A
Cells
Single cells
250K
Live cells
250K
cKIT+ FceRIa+ cells
250K
200K
150K
150K
150K
100K
100K
100K
50K
50K
50K
0
0
0
Comp-PE-A :: FceRIa
200K
FSC-H
200K
FSC-H
SSC-A
10
10
10
5
4
3
0
-10 0
50K
100K
150K
200K
250K
0
50K
100K
FSC-A
B
** ****
% Live cells of single cells
LAMP-1+ (% of Live BMMCs)
10
5
1
5
10
50
PO V D A M SO TN o
250K
-10
Omeprazole (µM)
0
10
3
10
4
10
5
3
-10
3
0
10
3
10
4
10
5
Comp-APC-A :: c-Kit
Comp-APC-Cy7-A :: Viability
C
0
N
200K
FSC-A
LAMP-1 15
150K
3
Viability 100 98
DMSO Omeprazole (50µM)
96 94 92 90 2
6
24
48
Hours of incubation
72
Figure E2 DMSO
Bafilomycin
Omeprazole
Figure E3
1
ONLINE REPOSITORY
2 3
Omeprazole inhibits IgE-mediated mast cell activation and allergic inflammation induced by
4
ingested allergen in mice
5 6
Cynthia Kanagaratham, PhD1,2, Yasmeen S. El Ansari, MMSc1,2, Benjamin F. Sallis, BSc1,2,
7
Brianna-Marie A. Hollister, BA1, Owen Lewis, BS1, Samantha C. Minnicozzi, MD1,2, Michiko K.
8
Oyoshi, PhD1,2, Rachel Rosen1,2, MD, MPH, Samuel Nurko1,2, MD, Edda Fiebiger, PhD1,2,* and
9
Hans C. Oettgen, MD, PhD1,2,*
10 11
1
12
2
Department of Pediatrics, Boston Children’s Hospital, Boston, MA, 02115, USA. Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
13 14
*These authors contributed equally to this work.
15 16
Corresponding author:
17
[email protected]
18
1 Blackfan Circle
19
Boston, MA 02115
20
USA
21
Phone: 1 (617) 919-6842
22
Fax: 1 (617) 730-0528
23 24
25 26 27
Methods
28
incubated with anti-TNP IgE (500ng/ml). Unbound IgE was washed away and cells were treated
29
with omeprazole at 50 µM for two hours. Following treatment, cells were stimulated with TNP-
30
OVA (100ng/ml) for 10 minutes at 37ºC while also being stained for cell surface marker c-Kit
31
(APC), degranulation marker LAMP-1 (PE), and viability (Viability-APCCy7). The reaction was
32
stopped with cold FACS buffer. Cells were pelleted and resuspended in FACS buffer prior to
33
acquisition on an LSR Fortessa (BD Biosciences, Franklin Lakes, NJ). Mast cells were identified
34
as live cells that are cKit+. The expression of LAMP-1 on the cell surface was used as a
35
measure of mast cell degranulation. For omeprazole treatment, cells were incubated with the
36
drug (50µM) for two hours, then stimulated with TNP-OVA.
37
For adenosine-mediated degranulation, cells were not incubated with IgE prior to treatment.
38
Cells were stimulated with 100µM adenosine (Sigma) in the presence of staining antibodies.
39
Human mast cell LAMP-1 assay. 105 cord blood mast cells (CBMCs) in 100µl were incubated
40
with human myeloma IgE (100ng/ml; Chemicon International, Tecaluma, CA). Unbound IgE was
41
washed away and cells were treated with omeprazole at various doses (50, 100, and 200µM) for
42
two hours. Following treatment, cells were stimulated with rabbit anti-human IgE (100ng/ml) for
43
10 minutes in the presence of viability dye (Viability-APCCy7), anti-LAMP-1 (PE), and anti-cKit
44
(APC). Cells were pelleted and resuspended in FACS buffer prior to acquisition on an LSR
45
Fortessa (BD). Mast cells were identified as live cells that are cKit+. The expression of LAMP-1
46
on the cell surface was used as a measure of mast cell degranulation. For omeprazole
47
treatment, cells were incubated with the drug (50µM) for two hours, then stimulated with anti-
48
IgE.
49
Histamine ELISA. Histamine concentration in plasma or cell culture supernatants was
50
measured using the histamine ELISA kit from SPI bio (Cayman Chemical, Ann Arbor, Michigan)
Murine mast cell LAMP-1 assay. 105 bone marrow-derived mast cells (BMMCs) in 100µl were
51
following the manufacturer’s instructions. For measurement of histamine concentration in
52
plasma, blood was collected 5 minutes after antigen challenge from mice. For measurement of
53
histamine concentration in cell culture, supernatants were collected 5 minutes after stimulation.
54
Cytokine release from BMMCs. BMMCs were sensitized with 500ng/mL anti-TNP IgE and
55
treated with omeprazole (50µM) for two hours. Following treatment, cells were stimulated with
56
100ng/mL of TNP-OVA for six hours. Supernatants were collected and immediately assayed for
57
to measure cytokine concentration.
58
Human mast cell cytokine secretion. CBMCs were sensitized with human myeloma IgE
59
(100ng/mL). Unbound IgE was washed away and cells were treated with omeprazole at various
60
doses (50, 100, and 200µM) for two hours. Following treatment, cells were stimulated with rabbit
61
anti-human IgE (100ng/ml) for 6 hours then supernatants were collected. Human mast cell IL-5
62
and IL-13 secretion was measured using a Cytometric Bead Array from BD Biosciences (San
63
Jose, California), according to the manufacturer’s instructions.
64
Transcriptional profiling of BMMCs by NanoString. BMMCs (106 cells in 1mL) were
65
sensitized overnight with 500ng/ml anti-TNP IgE. Unbound IgE was removed and cells were
66
treated with 50µM Omeprazole for 2 hours. Cells were then stimulated with 100ng/ml TNP-OVA
67
for 2h. Stimulation was stopped by washing with ice cold PBS, prior to lysis and homogenization
68
in RTL buffer (Qiagen) with β-mercaptoethanol (Sigma). RNA was isolated using the Qiagen
69
RNeasy mini kit according to the manufacturer’s instructions. Purified RNA (100 ng) was
70
hybridized with customized nCounter gene expression code sets as previously established for
71
unbiased analysis of Th2 inflammation1. Direct mRNA counts were determined by the
72
nCounter® Digital Analyzer System (NanoString Technologies, Seattle, WA;
73
www.nanostring.com) according to the manufacturer's protocol. Direct mRNA counts were
74
normalized to internal positive and negative controls and 5 house-keeping genes according to
75
manufacturer’s recommendations. Data is presented as standard deviation from the row mean.
76
PGD2 assay. PGD2 concentration in cell culture supernatants was measured using the
77
commercially available ELISA kit from Cayman Chemical following the manufacturer’s
78
instructions (Cayman). Samples to be assayed were prepared in the same manner as for
79
human mast cell cytokine secretion. Phosphoprotein detection. For each time point, 106
80
BMMCs were sensitized overnight with anti-TNP IgE (500ng/ml). Following sensitization, cells
81
treated with control or omeprazole for two hours then stimulated with 100ng/ml of TNP-OVA.
82
Stimulation was stopped with cold FACS buffer and cells were pelleted and lysed with
83
radioimmunoprecipitation assay (RIPA) buffer (Sigma) containing a cocktail of protease and
84
phosphatase inhibitors (Roche). Lysed cells were spun to collect protein supernatant. 10µl of
85
lysate was resolved on a 12% MiniProtean TGX precast gel (Biorad) and electrophoretically
86
transferred to a PVDF membrane (Millipore). Membranes were blocked with 5% BSA in TBST
87
for at least one hour. Membranes were probed with primary antibody overnight, washed with
88
TBST then probed with appropriate peroxidase conjugated secondary antibodies for one hour.
89
Membranes were developed with Supersignal chemiluminescent substrate reagent and imaged
90
on an iBright Western Blot scanner.
91
Calcium assay. BMMCs sensitized with anti-TNP IgE were treated with omeprazole (50µM) or
92
control. Intracellular calcium was stained using the Fluo 4NW calcium assay kit following the
93
manufacturer’s instructions. Change in the fluorescence of the calcium indicator was measured
94
using a Tecan Spark plate reader. Baseline fluorescence was measured for 30 seconds and
95
cells were then stimulated with 100ng/mL of TNP-OVA and changes in fluorescence were
96
measured for five minutes.
97
pH Assays. PPI-treated BMMCs (106 cells/mL) were incubated with FITC-Dextran overnight
98
(100µg/mL). Cells were washed and stained for cell surface markers c-Kit and FcεRIα, and for
99
viability. Cells were acquired by flow cytometry, and the intensity of the FITC signal of live c-Kit
100
and FcεRIα double-positive cells was assessed. Lysosensor Blue DND 167 (ThermoFisher) was
101
used to measure the changes in pH of acidic organelles following PPI treatment. Briefly, PPI-
102
treated BMMCs were washed and incubated with 1µM Lysosensor Blue for one hour. Cells were
103
washed and resuspended in media and the fluorescence intensity of the Lysosensor Blue was
104
measured using a Tecan Spark plate reader. To measure changes in intracellular pH following
105
treatment of BMMCs with PPI, pHRodoGreen AM intracellular pH indicator was used following
106
the manufacturer’s instructions. Briefly, PPI-treated cells were washed with live cell imaging
107
solution (ThermoFisher) then incubated with a mixture of pHRodoGreen and PowerLoad for 30
108
minutes. Cells were washed and resuspended in FACS buffer then stained for viability and cell
109
surface markers (c-Kit and FcεRIα). BMMCs were acquired on an LSR Fortessa and the
110
intensity of the intracellular pH indicator was measured in the population of live c-Kit and FcεRIα
111
double-positive cells.
112
Enzyme linked immunosorbent assay (ELISA). MCPT-1, OVA-IgE, and total IgE were
113
measured in serum isolated from mouse blood collected by intracardiac puncture. MCPT-1 was
114
measured using the MCPT-1 ELISA (ThermoFisher) following the manufacturer’s instructions.
115
Total IgE was measured using a direct sandwich ELISA using mouse IgE capture antibody
116
(Southern Biotech, #11101-01) and HRP-conjugated anti-mouse IgE (Southern Biotech, #1110-
117
05). OVA-IgE was measured using a direct sandwich ELISA using mouse IgE capture antibody
118
(Southern Biotech, #11101-01), and detected using biotinylated-OVA (US Biologicals, #O8075-
119
01) and Streptavidin-HRP. All ELISAs were developed with TMB and reactions were stopped
120
with 2N H2SO4. Cytokines in cell culture supernatants were measured using IL-6, IL-13, and
121
TNF-a Ready SET-Go! kits (ThermoFisher) following the manufacturer’s instructions. IL-4 in cell
122
culture supernatants was measured using the ELISA MAX Standard Set Mouse IL-4 kit
123
following the manufacturer’s instructions (Biolegend).
124
RT-qPCR. Jejunum sections were collected from mice and stored in RNAlater (ThermoFisher)
125
solution at 4oC overnight before freezing tissue at -80oC. RNA was extracted from 20 to 30mg of
126
tissue using the Qiagen RNeasy mini kit according to the manufacturer’s instructions. RNA
127
concentration was measured using a Nanodrop 2000. One µg of RNA was reverse transcribed
128
into cDNA using iScript cDNA Synthesis Kit (Biorad) following the manufacturer’s instructions.
129
Gene expression was measured using a QuantStudio 3 real time qPCR instrument
130
(ThermoFisher), Taqman Fast Advanced Mastermix (Applied Biosystems) and predesigned
131
probes (Life Technologies, Applied Biosystems). Expression of all genes was normalized to
132
Gapdh and calculated using the Cq values and the 2-ddCq method.
133
Intestinal cell isolation. Jejunum segments were isolated and flushed with PBS containing
134
10% FBS. The intestine was cut longitudinally and chopped into 0.5cm segments then
135
incubated in buffer containing 10mM EDTA and 1mM DTE for 20 minutes to remove the
136
epithelial layer. Tissue was finely chopped using a razor blade then digested with 971U/ml
137
collagenase VIII and 5µg/mL DNAse I (Applichem) in complete RPMI for 30 minutes. Cell
138
fraction was resuspended in 40% Percoll and overlaid above a 70% Percoll fraction. Cells were
139
centrifuged at 600xg for 20 minutes and leukocytes at the interface were collected.
140
Histology. Jejunums were collected from mice and segments of approximately 1cm were fixed
141
in 4% paraformaldehyde for a minimum of 48 hours. Dehydration, paraffin embedding,
142
sectioning and staining were done by the Harvard Medical Area Rodent Histopathology Core
143
(Harvard, Boston, MA). Images were captured on a Nikon E800 microscope (Nikon, Melville,
144
NY).
145
May Grünwald Giemsa Staining. BMMCs were treated with bafilomycin or omeprazole or drug
146
vehicle. 300,000 cells were adhered onto poly-lysine coated slides by cytospin (Shandon
147
Cytospin 3). Slides were stained with May Grünwald solution (Sigma) for 5 minutes, washed 3
148
times in deionized water, stained with Giemsa solution (Sigma) for 15 minutes then washed 6
149
times in deionized water. Slides were air dried then fixed with Permount (Sigma). Images were
150
captured on a Nikon E800 microscope (Nikon, Melville, NY).
151
Human basophil activation test. Human basophil activation tests were performed as
152
previously described 2 using the FLOW CAST Basophil activation test kit (Bühlmann
153
Laboratories, Schönenbuch, Switzerland). Briefly, 50µL aliquots of whole blood from healthy
154
donors were incubated with omeprazole (200µM) for two hours in a 37oC water bath. Following
155
treatment, samples were stimulated with 200ng/mL anti-IgE (Southern biotech) and
156
simultaneously stained for cell viability (Viability-APCCy7), anti-CCR3 (PE), anti-CD63 (FITC)
157
for 15 minutes at 37 oC. Red blood cells were lysed, and cells were washed and acquired on an
158
LSR Fortessa (BD). Basophils were identified as SSClow and CCR3+. The expression of CD63
159
on the cell surface was used as a measure of basophil activation.
160
161
Figure legends
162
Figure E1. Gating strategy for identification of bone marrow derived mast cells by flow
163
cytometry, dose responsive effect of omeprazole on mast cell degranulation, and viability of
164
cells following incubation with omeprazole. A) Cell population, and subsequently single cells,
165
are first identified based on forward and side scatter. Live cells are then selected from the single
166
cell gate based on negative staining for the viability dye. Live cells expressing cKIT and FcεRIa
167
are defined as bone marrow mast cells (BMMCs). B) LAMP-1 expression of IgE anti-TNP bound
168
BMMCs following incubation with various doses of omeprazole and degranulation with TNP-
169
OVA. C) Viability of BMMCs following incubation with omeprazole. Viability staining is applied to
170
all BMMC culture experiments analyzed by flow cytometry. For panel B, data are shown from
171
one experiment representative of two experiments. Statistical analysis by one-way analysis of
172
variance. **P<0.01, ****P<0.0001
173
Figure E2. Alterations in granule morphology following incubations with bafilomycin and
174
omeprazole. May Grünwald/Giemsa staining of cytospin preparations of BMMCs following
175
incubation with DMSO, bafilomycin (20 nM) or omeprazole (50 µM) for 24 hours.
176
Figure E3. Omeprazole blocks activation of basophils. Activation of basophils in whole blood
177
using anti-IgE following treatment with 200 µM omeprazole or vehicle for 2 hours. Basophil
178
activation was quantified by flow cytometry. Basophils were identified as SSClow, negative
179
staining for viability dye, and CCR3+. The expression of CD63 on the cell surface was used as a
180
measure of basophil activation. Data points represent four different individuals assayed in
181
duplicates for each condition. Statistical analysis between treatment groups was done using a
182
paired t-test. *P<0.05
183
184 185
References 1. Platzer B, Baker K, Vera MP, Singer K, Panduro M, Lexmond WS, et al. Dendritic cell-
186
bound IgE functions to restrain allergic inflammation at mucosal sites. Mucosal Immunol
187
2015; 8:516-32.
188
2.
Burton OT, Logsdon SL, Zhou JS, Medina-Tamayo J, Abdel-Gadir A, Noval Rivas M, et
189
al. Oral immunotherapy induces IgG antibodies that act through FcgammaRIIb to
190
suppress IgE-mediated hypersensitivity. J Allergy Clin Immunol 2014; 134:1310-7 e6.
191