Accepted Manuscript The effect of penicillin administration in early life on murine gut microbiota and blood lymphocyte subsets Jaroslaw Daniluk, Urszula Daniluk, Malgorzata Rusak, Milena Dabrowska, Joanna Reszec, Magdalena Garbowicz, Kinga Humińska, Andrzej Dabrowski PII:
S1075-9964(17)30058-6
DOI:
10.1016/j.anaerobe.2017.03.015
Reference:
YANAE 1711
To appear in:
Anaerobe
Received Date: 24 December 2016 Revised Date:
4 March 2017
Accepted Date: 15 March 2017
Please cite this article as: Daniluk J, Daniluk U, Rusak M, Dabrowska M, Reszec J, Garbowicz M, Humińska K, Dabrowski A, The effect of penicillin administration in early life on murine gut microbiota and blood lymphocyte subsets, Anaerobe (2017), doi: 10.1016/j.anaerobe.2017.03.015. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. 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.
ACCEPTED MANUSCRIPT
1
The effect of penicillin administration in early life on murine gut microbiota
2
and blood lymphocyte subsets.
3 Jaroslaw Daniluk1, Urszula Daniluk2, Malgorzata Rusak3, Milena Dabrowska3,
5
Joanna Reszec4, Magdalena Garbowicz5, Kinga Humińska5, Andrzej Dabrowski1
RI PT
4
6 7
1
8
Bialystok ul. M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
9
2
SC
Department of Gastroenterology and Internal Medicine, Medical University of
M AN U
Department of Pediatrics, Gastroenterology and Allergology, Medical University of
10
Bialystok ul. J. Waszyngtona 17, 15-274 Bialystok, Poland
11
3
12
Waszyngtona 15A, 15-269 Bialystok, Poland
13
4
14
Waszyngtona 13, 15-269 Bialystok, Poland
15
5
16
Poland
TE D
Department of Medical Pathomorphology, Medical University of Bialystok ul. J.
Genomic Laboratory, DNA Research Center ul. Mickiewicza 31, 60-385 Poznan,
EP
17
Department of Haematological Diagnostics, Medical University of Bialystok ul. J.
Corresponding author: Jaroslaw Daniluk, Department of Gastroenterology and
19
Internal Medicine, Medical University of Bialystok ul. M. Sklodowskiej-Curie 24a, 15-
20
276 Bialystok, Poland
21
Phone: 48 85 746 82 34
22
fax: 48 85 746 85 06
23
Email:
[email protected],
[email protected]
AC C
18
1
ACCEPTED MANUSCRIPT
24
Abstract Background and aim: Antibiotics have many beneficial effects but their
26
uncontrolled use may lead to increased risk of serious diseases in the future. Our
27
hypothesis is that an early antibiotic exposition may affect immune system by
28
altering gut microbiota. Therefore, the aim of the study was to determine the effect of
29
penicillin treatment on gut microorganisms and immune system of mice. Methods:
30
21-days old C57BL6/J/cmdb male mice were treated with low-dose of penicillin
31
(study group) or water only (control group) for 4 weeks. Tissue and stool samples for
32
histology or microbiome assessment and peripheral blood for CBC and flow
33
cytometry evaluation were collected. Results: We found high variability in microbiota
34
composition at different taxonomic levels between littermate mice kept in the same
35
conditions, independently of treatment regimen. Interestingly, low-dose of penicillin
36
caused significant increase of Parabacteroides goldsteinii in stool and in colon tissue
37
in comparison to control group (9.5% vs. 4.9%, p=0.008 and 10.7% vs. 6.1%,
38
p=0.008, respectively). Moreover, mice treated with penicillin demonstrated
39
significantly elevated percentage of B cells (median 10.5% vs 8.0%, p=0.01) and
40
decrease in the percentage of total CD4+ cell (median 75.4% vs 82.5%, p=0.0039)
41
with subsequent changes among subsets - increased percentage of regulatory T
42
cells (Treg), T helper 1 (Th1) and T helper 2 (Th2) cells. Conclusion: Our study
43
showed significant effect of penicillin on B and T cells in peripheral blood of young
44
mice. This effect may be mediated through changes in gut microbiota represented
45
by the expansion of Parabacteroides goldsteinii.
46
Key words: lymphocytes, microbiota, Parabacteroides goldsteinii, penicillin
AC C
EP
TE D
M AN U
SC
RI PT
25
47 2
ACCEPTED MANUSCRIPT
48
1. Introduction Discovery of penicillin in 1928 by Sir Alexander Fleming, saved many lives
50
and revolutionized treatment of bacterial infections. Antibiotics have also found
51
application in agriculture, due to their effect on weight gain (up to 15%) in farm
52
animals [1]. Nowadays easy access to the antibiotics led to their overuse in humans
53
and caused development of bacterial resistance to the treatment.
RI PT
49
Despite of their benefit in infection treatment, antibiotics have profound
55
influence on the structure and function of gut microbes. Our knowledge about human
56
microbiome has dramatically increased over the last decade due to development of
57
novel sequence-based molecular tests, like 16s rRNA-based sequencing, which
58
became the gold standard technique in microbiome identification [2]. Commensal
59
microorganisms prevent colonization of the gut by pathogens. They are involved in
60
the process of appropriate development and maturation of innate and acquired
61
immune system during the childhood, and maintaining homeostasis in the adults [3,
62
4]. The fetal gastrointestinal tract is sterile but immediately after the birth it is
63
colonized by many different microorganisms [5]. Recently, it has been reported that
64
any perturbations in microorganisms composition caused by antibiotics or changes
65
in the diet habits during the first two years of live, may predispose to the
66
development of chronic diseases like obesity, type 1 diabetes, rheumatoid arthritis,
67
coeliac disease, liver diseases and inflammatory bowel disease (IBD) [6-8]. It has
68
been also suggested that antibiotic exposure during the first year of life increases
69
the risk of asthma development [9]. Jernberg et al reported that even a short, 1-week
70
long course of antibiotic regimen causes significant and prolonged (up to 2 years)
AC C
EP
TE D
M AN U
SC
54
3
ACCEPTED MANUSCRIPT
71
changes in microbiota composition [10]. Therefore, the aim of our study was to
72
determine the effect of early antibiotic (penicillin) exposition on gut microorganisms
73
and immune system of mice.
75
RI PT
74 2. Material and methods 2.1. Animals
77
C57BL6/J/cmdb male mice (Medical University of Bialystok; Bialystok, Poland) were
78
maintained in specific pathogen-free (SPF) conditions. All in vivo experiments were
79
performed according to EU Directive 2010/63/EU and approved by the Local
80
Committee for Experiments with the Use of Laboratory Animals, Bialystok, Poland.
81
21-days old mice, just after the weaning, were randomly divided into two groups:
82
study group – animals receiving penicillin dissolved in drinking water (n=10) and
83
control group – mice receiving drinking water only (n=10), ad libitum for 28 days.
84
Dose of the antibiotic, 1 µg of penicillin / 1 gram of body weight, was determined on
85
the basis of previous literature data and assumption that daily consumption of water
86
is 15 ml per 100 g of mice [11, 12]. Fresh containers with water and water with
87
antibiotic were changed three times a week. Mice were housed 5 per cage and they
88
were allowed to eat standard chow diet ad libitum. Consumption of water or water
89
with penicillin was determined on daily basis for each cage. Body weight of each
90
mouse was measured every fourth day.
92
M AN U
TE D
EP
AC C
91
SC
76
2.2. Sample Collection
4
ACCEPTED MANUSCRIPT
After 28 days, mice were anesthetized and sacrificed. Blood samples were collected
94
by cardiac puncture and divided into two parts - complete blood count (CBC) and
95
flow cytometry analysis. The small and large intestine, pancreas, and liver were
96
harvested aseptically and stored for further histological evaluation. For microbiota
97
assessment samples were collected directly from tissue (distal part of small intestine
98
and ascending colon), and from the stool (lumen of the caecum), and immediately
99
snap-frozen and stored (-80°C).
SC
RI PT
93
100
2.3. CBC and flow cytometry analysis of isolated cells
102
To determine CBC, 200 µl of peripheral blood was used and analyzed in the
103
Department of Haematological Diagnostics, Medical University of Bialystok.
104
Peripheral subpopulations of lymphocytes were assessed using flow cytometry. To
105
prepare samples, approximately 200 µl of peripheral blood was treated with RBC
106
lysis buffer (Sigma-Aldrich) for 10 min at room temperature, and the remaining cells
107
were washed twice with cold PBS and centrifuged at 1,200 rpm for 10 min. Cells
108
were stained with the appropriate combinations of the following antibodies: FITC–
109
anti-CD3e (145-2C11; BD Pharmigen), APC-anti-CD4 (MR4-5; BD Pharmigen), PE-
110
anti-CD25 (3C7; BD Pharmigen), PE-Cy7-anti-CD127 (SB/199; BD Pharmigen), and
111
Mouse T Lymphocyte Subset Antibody Cocktail with Isotype Control (BD Pharmigen)
112
containing PE-Cy 7- anti-CD3e (145-2C11), PE-anti-CD4 (RM4-5), APC-anti- CD8a
113
(53-6.7), Mouse B Lymphocyte Subset Antibody Cocktail with Isotype Control (BD
114
Pharmigen) with PE-Cy 7- anti-CD45R/B220 (145-2C11); PE-anti-CD23 (RM4-5);
115
APC-anti- sIgM (53-6.7). The Mouse Th1/Th2/Th17 Phenotyping Kit (PerCP-Cy5.5-
AC C
EP
TE D
M AN U
101
5
ACCEPTED MANUSCRIPT
anti-CD4 (RM4-5); PE-anti-IL-17A (TC11-18H10.1); FITC-anti-INF-GMA (XMG1.2);
117
APC-anti-CD4 (11B11) BD Pharmigen) were used according to the manufacturer’s
118
instruction for the detection of CD4+IL-17+, CD4+IFNγ+, CD4+IL-4+ expression.
119
Flow cytometric data were acquired using a FACS Canto II cytometer with BD
120
FACSDiva Software v6.1.3 (BD Biosciences) and analyzed with Worksheet software
121
(BD).
122
SC
RI PT
116
2.4. Histology and Immunohistochemistry
124
The organs were fixed with 10 % PBS-buffered formalin for 24h, embedded in
125
paraffin, cut sagittally into 5-µm sections, stained with hematoxylin and eosin (H&E),
126
and examined by light microscopy (Olympus BX45) for histological analysis. For
127
each animal, ten fields at a magnification of ×100 were captured randomly from the
128
four different parts of the intestine.
129
To determinate the lymphocytic infiltration we used antibodies against T cytotoxic
130
cells, T helpers and against lymphocytes B. Following the deparaffinization and
131
rehydration, epitope retrieval was carried out in the EnVision Flex Target Retrieval
132
Solution (DAKO) at low pH. Endogenous peroxidases were blocked by incubating
133
the sections in methanol and 3% hydrogen peroxidase for 40 minutes. Next, slides
134
were incubated with proper anti-mouse antibodies against CD3, CD4, CD8 and
135
CD20 in 1:100 dilutions for 1 hour at room temperature. Visualization reagent
136
EnVision (DAKO) was applied for 30 minutes and followed by DAB solution for 10
137
minutes. The slides were than counterstained with H&E and examined under the
AC C
EP
TE D
M AN U
123
6
ACCEPTED MANUSCRIPT
138
light microscope. The intensity of immunostaining was evaluated in random 10 fields
139
under ×20 magnification. Appropriate positive and negative controls were performed.
140 2.5. Faecal microbiota analysis
142
The bacterial composition of small and large intestine, and stool samples of each
143
group was carried out using 16S ribosomal RNA (rRNA) gene sequencing on the
144
MiSeq apparatus (Illumina) via next-generation sequencing (NGS) technology. In
145
order to analyze bacterial components, the V3-V4 hypervariable region of the 16S
146
rRNA gene was firstly amplified from the genomic DNA extracted from fecal samples
147
by
148
GACTACHVGGGTATCTAATCC-3’)
149
sequences, compatible with Illumina’s indices. Incorporation of primers with indexes
150
and adapters was followed by second PCR reactions. After each amplification step,
151
the PCR products were purified by using AMPure XP beads (Beckman Coulter
152
Genomic, CA, USA). DNA amplified fragments were normalized to 2nM by Qubit®
153
dsDNA HS kit (high
154
sequencing. 8pM libraries were loaded on the MiSeq plarform (Illumina Inc., San
155
Diego, CA, USA). Sequencing was performed with 300-nucleotide-long paired-end
156
reads on the Illumina sequencer (MiSeq) according to the instructions of the
157
manufacturer. The analysis output file generated for the 16S metagenomics
158
workflow provided classification of reads for each sample. Obtained data were
159
analyzed and interpreted using specialized software packages: Quantitative Insights
primers:
F
(5’-CCTACGGGNGGCWGCAG-3’) containing
TE D
two
sensitivity
DNA;
Life
additional
and
overhang
R
(5’-
adapter
Technologies) and pooled prior to
AC C
EP
using
M AN U
SC
RI PT
141
7
ACCEPTED MANUSCRIPT
160
into Microbial Ecology (QIIME), Greengenes, Metagenome Analyzer (MEGAN) I
161
Ribosomal Database Project (RDP).
162 2.6. Data analysis
164
Data were analyzed using Statistica 10 software. Statistical significance was
165
determined by U Mann-Whitney test; p<0.05 was considered statistically significant.
RI PT
163
SC
166 3. Results
168
3.1. Body weight, CBC and histological evaluation
169
During the whole study period, body weights of animals were within the range of
170
growth for C57BL6/J/cmdb male mice. There was no statistically significant
171
difference in body weight change between control and study groups (Table 1). There
172
was no difference in consumption of water in control (mean 21.8 ml/cage/day) or
173
water with penicillin in study group (mean 23.0 ml/cage/day). After 28 days of
174
penicillin administration no differences in white blood count, red blood count or
175
platelets count were observed between the groups. However, 4 weeks of antibiotic
176
exposure significantly increased percentage of blood neutrophils in comparison to
177
control group (6.0% vs. 4.0%, p=0.0002). This was accompanied by significant
178
decrease of lymphocytes percentage in the treatment group in comparison to control
179
(92.5% vs. 95.0% respectively, p=0.0002).
180
Histological examination of the small and large intestinal biopsies revealed no
181
abnormalities between study groups (Fig. 1). Tissue specimen evaluation did not
AC C
EP
TE D
M AN U
167
8
ACCEPTED MANUSCRIPT
182
show any signs of mucosal layer disruption or increased infiltration of inflammatory
183
cells in any of the experimental groups.
184 3.2. Bacterial composition of the gut
186
To determine possible changes in bacterial composition after low-dose of penicillin
187
treatment, we detected gut taxonomic groups by 16S rRNA sequencing. We looked
188
at the microbiota composition in the control and study group in gut tissue (distal
189
ileum and ascending colon) and faecal samples (obtained directly form caecum
190
lumen). Surprisingly, independently of treatment regimen (control or penicillin), we
191
found substantial diversity of microbiota both at phyla and species level between
192
littermates of C57BL6/J/cmdb male mice living in the same cage. Despite exactly the
193
same environmental conditions (drinking water, chow diet, bedding, air ventilation
194
system) microbiota of littermates at phyla level differed up to 40% for Firmicutes and
195
Barcteroidetes (Figure 2A). This phenomenon was found both in control and study
196
mice. Similar observations were made at other taxonomic levels, including species.
197
(Figure 2B). Next, we evaluated the effect of low-dose of penicillin on the gut
198
microbiota composition in comparison to control group without antibiotic. We did not
199
find any significant changes in the presence of two major phyla groups – Firmicutes
200
and Bacteroidetes (representing ~ 90% of gut microbiota) after penicillin treatment in
201
small intestine, colon or fecal specimens (Figure 3A, B). Similarly, we did not find
202
any significant alterations in other, less abundant, phyla of gut microbiota in
203
evaluated specimens (data not shown). Interestingly, at the species taxonomic level,
204
we found significant increase of bacteria Parabacteroides goldsteinii in stool and
AC C
EP
TE D
M AN U
SC
RI PT
185
9
ACCEPTED MANUSCRIPT
tissue samples from large intestine after penicillin treatment, in comparison to control
206
group (9.5% vs. 4.9%, p=0.008 and 10.7% vs. 6.1%, p=0.008, respectively) (Figure
207
3C). Similar increase of P. goldsteinii presence after antibiotic exposure was
208
observed in small intestine tissue samples, however, in this case the results did not
209
reach statistical significance (19.6% vs. 12.6%, p=0.098).
210
RI PT
205
3.3. Lymphocyte subsets in peripheral blood
212
To determine whether penicillin affects the lymphocyte subpopulations, we
213
performed flow cytometry on peripheral blood mononuclear cells (PBMC) from mice
214
in both groups. Comparing to controls, mice treated with penicillin demonstrated
215
significantly elevated percentage of B cells (CD19) (median 10.5% vs 8.0%, p=0.01)
216
and decreased percentage of total CD4+ (median 75.4% vs 82.5%, p=0.0039),
217
(Table 2). However among subsets of CD4+ lymphocytes, increased percentage of
218
CD4+CD25+CD127low (Treg) cells (median 14.5% vs 8.1%, p=0.002) and CD4+
219
cells producing IL-4 (Th2) (median 10.7% vs 7.8%, p=0.03) was detected in mice
220
treated with penicillin. No significant differences in CD4+IL-17+ (TH17) cells (median
221
4.9% vs 3.5%) or CD4+IFNγ+ (Th1) (median 7.9% vs 6.6%) were observed between
222
groups.
223
3.4. Lymphocyte subsets in gut tissue
224
We also evaluated if changes in blood lymphocyte subpopulations after penicillin
225
treatment were accompanied by any alterations in lymphocyte subsets infiltration in
226
small or large intestine. Interestingly, immunohistochemical staining of small
AC C
EP
TE D
M AN U
SC
211
10
ACCEPTED MANUSCRIPT
227
intestine and colon revealed no abnormalities or differences in lymphocyte subsets
228
(CD3+, CD4+, CD8+, CD20+) between control and study group (Figure 4).
229 4. DISCUSSION
RI PT
230
Despite relatively stable composition of the gut microbiome in adult healthy
232
individuals, there is an extremely high interpersonal diversity [4]. Data from Human
233
Microbiome Project showed clear differences in Bacteroides/Firmicutes (two most
234
abundant phyla in gut) ratio in healthy volunteers [13]. Similar observations were
235
found at lower taxonomic units, like species [13, 14]. The cause and reason for this
236
variability is currently unknown, however it may be driven by genetic factors,
237
environmental factors (i.e. lifestyle and geographical differences between European,
238
American and Asian population), diet or drugs (i.e. antibiotics) [4, 15]. The key
239
question is, if the differences in the gut microbiome composition may predispose to
240
the development of the diseases in selected persons. In our study, we have found
241
significant differences in gut microbiome composition between mice. Surprisingly,
242
this general phenomenon was independent of the treatment regimen. In our study,
243
the animals were kept in specific pathogen-free conditions, with individually
244
ventilated cages. They received the same autoclaved water and chow diet.
245
Littermate mice were at the same age, freshly weaned from mother. Nevertheless,
246
inter-animal differences in gut microbiome composition were still observed. This may
247
suggest that genetic heterogeneity between mice is a driving force of microbiota
248
composition in the case of identical housing (location, diet) and treatment (penicillin
249
badge and dose) conditions. Of course, we cannot completely rule out that mouse
AC C
EP
TE D
M AN U
SC
231
11
ACCEPTED MANUSCRIPT
husbandry condition influenced intestinal microbial composition, as it has been
251
shown recently [16]. However, reported risk factors of inter-animals microbial
252
differences and diversity, namely irradiated chow diet, open cages housing system,
253
presence of other strains at the same room, or low restrictive access policy were not
254
a case in our study. In our case, we compared microbiota of mice kept in the same
255
animal facility, room and even a cage, so the effect of husbandry policy on microbial
256
composition is highly unlikely.
SC
RI PT
250
Another important finding of our study was significant increase of
258
Parabacteroides goldsteinii in microbiota composition after penicillin exposure. P.
259
goldsteinii is a Gram-negative, strictly anaerobic, rod-shaped bacteria described for
260
the first time in 2005 [17]. Interestingly, P. goldsteinii can demonstrate resistance to
261
many commonly used antibiotics like penicillin, clindamycin, erythromycin,
262
piperacillin-tazobactam, meropenem, and may possess metallo-β-lactamase [18,
263
19]. The bacteria is a part of human gut microbiota, but it was also isolated from
264
abdominal abscesses and peritoneal fluid as a consequence of diverticulitis, caecal
265
inflammation, and necrotic ileum or from appendix tissue in the case of acute
266
appendicitis [17]. In our study, 4 weeks of mice exposure to low-doses of penicillin
267
was sufficient to significantly increase population of P. goldsteinii in the colon and
268
stool. Penicillin is a narrow spectrum antibiotic, more effective against Gram-positive
269
bacteria, which may explain selective increase of specific Gram-negative pathogens.
270
Elevated population of P. goldsteinii was accompanied by the increased percentage
271
of neutrophils in peripheral blood of antibiotic treated mice, comparing to control.
AC C
EP
TE D
M AN U
257
12
ACCEPTED MANUSCRIPT
272
However, it is difficult to ascertain if this phenomenon was the direct effect of
273
penicillin or pathogenic bacteria. The relationship between gut microbiome composition and function of
275
immune system is already known, but it still remains under close evaluation [11, 20,
276
21]. It has been recently shown that short-term treatment of mice with low-dose
277
penicillin started from the birth, has altered metabolism and enhanced the risk of
278
obesity [11]. This mechanism was, at least partially, mediated by changes in
279
expression of ileal genes involved in immunity. Low-dose penicillin decreased
280
expression of genes linked to antigen-presenting cells, T cells, B cells, and
281
phagocytic cells. To investigate the immune cells shifts in response to an altered
282
intestinal microbiota, we used murine model treated with low-dose of penicillin. Our
283
data did not show any histological abnormalities in intestinal tissues or other organs
284
of gastrointestinal tract like liver and pancreas (data not shown) in study group
285
comparing to control. Additionally, we found no significant alterations among
286
lymphocyte subsets in intestinal specimens after antibiotic exposure comparing to
287
control. One of the possible explanation to these contradictory results is the use of
288
different method for detection of immune cells in tissue in our study.
289
It was previously reported, that influence of gut microbiota on the T cell subsets’
290
balance extended beyond the intestinal lamina propria [22]. However, only limited
291
studies were focused on the relationship between intestinal flora alteration and
292
modification of peripheral blood immune cells. Banck et al. tested in vitro the effect
293
of 20 different antibiotics on function of lymphocytes isolated from the peripheral
294
blood, showing no influence of penicillin [20]. In our study, penicillin exposure
AC C
EP
TE D
M AN U
SC
RI PT
274
13
ACCEPTED MANUSCRIPT
resulted in the significant decrease of total CD4+ cells in blood. Similar pattern of
296
lymphocyte counts, with decreased counts of CD4+ cells in lymphoid organs, but the
297
same numbers of CD8+ and CD19+ as conventional colonized mice, was reported in
298
germ-free mice [23]. However, in this study lymphocytes were isolated from the
299
spleen, what has not been the case in our study. Low CD4+ count could be also the
300
effect of dysbiosis caused by penicillin, since some bacteria, like Bacteroides fragilis,
301
may affect the number of circulating CD4+ T cells [23]. Likewise, expansion of lamina
302
propria and systemic Treg cells depends on intestinal microbiota, mainly on the
303
presence of Clostridial strains [22]. In our study we were unable to identify the
304
specific lymphocyte subpopulation responsible for the decrease of total CD4+
305
lymphocytes. Unfortunately, selected CD4+ subpopulations representing only a small
306
fraction of total CD4+ lymphocytes showed the opposite trend in the low-dose of
307
penicillin group. Penicillin treated mice demonstrated increase of peripheral Treg
308
(CD4+CD25+CD127low), as well as Th1 (IFNγ), Th2 (IL-4) cells, and B cells,
309
although no functional studies were performed and we could not determine if this
310
effect was caused by penicillin itself or indirectly by microbial imbalance,
311
represented by increase of some specific bacterial strains, like Parabacteroides
312
goldsteinii. Enhanced Th2 response in our study could be related to allergic reaction
313
triggered by penicillin, since relationship between treatment with broad-spectrum
314
antibiotics and exaggerated Th2 cell responses with increased circulating basophil
315
populations, elevated IgE and allergic inflammation in mice was already reported
316
[24]. On the other hand, penicillin has been also shown as an exacerbating factor of
317
experimental autoimmune encephalomyelitis, the Th1 derived disease [25]. In this
AC C
EP
TE D
M AN U
SC
RI PT
295
14
ACCEPTED MANUSCRIPT
study, among evaluated beta-lactams, cefuroxime exposure resulted in down-
319
regulation of genes expression associated with Th2 and Treg differentiation.
320
Unfortunately, the effect of penicillin on these genes was not explored. However, it
321
was shown, that penicillin-modified albumin was able to enter T cells, and enhanced
322
their
323
encephalomyelitis. [25]. In our study, penicillin exposure also resulted in significant
324
increase of Th1 (CD4+IFNγ+) lymphocytes as it was mentioned above. It is difficult to
325
speculate which subset of T cells dominated at the beginning of treatment and which
326
of these cells would dominate after longer (i.e. several months) exposure to
327
penicillin. Certainly, more research is needed to determine specific effects of
328
antibiotics on function of immune cells, both in peripheral blood and locally in
329
tissues. Our study has some limitations, including small sample size. However, our
330
data were consistent and low number of animals does not seem to affect the final
331
results. Moreover, in our experiment both study and control Group consisted
332
exclusively of male mice, so we cannot exclude that the results are gender specific.
333
Cox et al. have already shown that temporary exposure to low-dose penicillin may
334
have a sex-specific long term effect (metabolic alterations, changes in ileal
335
expression of genes involved in immunity) only during a specific time window –
336
before the birth of the pups and throughout the weaning process [11]. In this study,
337
there was no difference in body composition of older male and female mice after
338
antibiotic exposure. In our experiment, we used mice after the weaning period, so
339
the probability that the results were affected by specific gender is much lower. In
340
agriculture, administration of low doses of antibacterial agents promotes the growth
in
the
development
of
experimental
autoimmune
AC C
EP
TE D
M AN U
SC
pathogenicity
RI PT
318
15
ACCEPTED MANUSCRIPT
of farm animals independently of sex. We also did not look at the expression of
342
intestinal or colonic genes involved in immune reaction. Moreover, our observation
343
period was limited to 4 weeks only. It would be interesting to prolong the experiment
344
until the development of changes in mice body weight or autoimmune-related
345
diseases.
RI PT
341
In summary, the present work documents unique and significant effect of
347
penicillin on B and T cells in peripheral blood of young mice. This effect may be
348
mediated through changes in gut microbiota represented by the expansion of
349
Parabacteroides goldsteinii. Altering immune cells at a young age may have a
350
tremendous effect on future development of autoimmune or allergic diseases.
M AN U
SC
346
351 Funding
353
This research received grant from Medical University of Bialystok, Poland
354
(N/ST/ZB/16/001/1151)
355
Competing interests
356
The authors declare no competing financial interests.
357
Ethical approval
358
All in vivo experiments were performed according to EU Directive 2010/63/EU and
359
approved by the Local Committee for Experiments with the Use of Laboratory
360
Animals, Bialystok, Poland.
362
EP
AC C
361
TE D
352
References
16
ACCEPTED MANUSCRIPT
363
[1]
P. Butaye, L.A. Devriese, F. Haesebrouck. Antimicrobial growth promoters
364
used in animal feed: effects of less well known antibiotics on gram-positive bacteria.
365
Clin Microbiol Rev 16 (2003) 175-88.
366
[2]
367
"how to" guide for physicians. Am J Gastroenterol 109 (2014) 983-93.
368
[3]
369
maintain gut homeostasis. Curr Opin Immunol 22 (2010) 455-60.
370
[4]
371
al. Inflammation and colorectal cancer, when microbiota-host mutualism breaks.
372
World J Gastroenterol 20 (2014) 908-22.
373
[5]
374
Fierer, et al. Delivery mode shapes the acquisition and structure of the initial
375
microbiota across multiple body habitats in newborns. Proc Natl Acad Sci U S A 107
376
(2010) 11971-5.
377
[6]
378
a role in the pathogenesis of autoimmune diseases? Gut 64 (2015) 332-41.
379
[7]
380
personalized medicine. Curr Gastroenterol Rep 17 (2015) 15.
381
[8]
382
ageing, and beyond: gut microbiota and inflammatory status in seniors and
383
centenarians. PLoS One 5 (2010) e10667.
384
[9]
385
from antibiotic use in early life. Chest 131 (2007) 1753-9.
RI PT
A.D. Tyler, M.I. Smith, M.S. Silverberg. Analyzing the human microbiome: a
SC
H. Chung, D.L. Kasper. Microbiota-stimulated immune mechanisms to
M AN U
M. Candela, S. Turroni, E. Biagi, F. Carbonero, S. Rampelli, C. Fiorentini, et
TE D
M.G. Dominguez-Bello, E.K. Costello, M. Contreras, M. Magris, G. Hidalgo, N.
EP
M.H. McLean, D. Dieguez, L.M. Miller, H.A. Young. Does the microbiota play
AC C
E. Bellaguarda, E.B. Chang. IBD and the gut microbiota--from bench to
E. Biagi, L. Nylund, M. Candela, R. Ostan, L. Bucci, E. Pini, et al. Through
A.L. Kozyrskyj, P. Ernst, A.B. Becker. Increased risk of childhood asthma
17
ACCEPTED MANUSCRIPT
386
[10]
C. Jernberg, S. Löfmark, C. Edlund, J.K. Jansson. Long-term ecological
387
impacts of antibiotic administration on the human intestinal microbiota. ISME J 1
388
(2007) 56-66.
389
[11]
390
Altering the intestinal microbiota during a critical developmental window has lasting
391
metabolic consequences. Cell 158 (2014) 705-21.
392
[12]
393
early life alter the murine colonic microbiome and adiposity. Nature 488 (2012) 621-
394
6.
395
[13]
396
microbiome. Nature 486 (2012) 207-14.
397
[14]
398
Genome Med 8 (2016) 51.
399
[15]
400
human gut microbial gene catalogue established by metagenomic sequencing.
401
Nature 464 (2010) 59-65.
402
[16]
403
Analysis of factors contributing to variation in the C57BL/6J fecal microbiota across
404
German animal facilities. Int J Med Microbiol 306 (2016) 343-55.
405
[17]
406
"Bacteroides goldsteinii sp. nov." isolated from clinical specimens of human
407
intestinal origin. J Clin Microbiol 43 (2005) 4522-7.
RI PT
L.M. Cox, S. Yamanishi, J. Sohn, A.V. Alekseyenko, J.M. Leung, I. Cho, et al.
M AN U
SC
I. Cho, S. Yamanishi, L. Cox, B.A. Methé, J. Zavadil, K. Li, et al. Antibiotics in
H.M.P. Consortium. Structure, function and diversity of the healthy human
TE D
J. Lloyd-Price, G. Abu-Ali, C. Huttenhower. The healthy human microbiome.
EP
J. Qin, R. Li, J. Raes, M. Arumugam, K.S. Burgdorf, C. Manichanh, et al. A
AC C
P. Rausch, M. Basic, A. Batra, S.C. Bischoff, M. Blaut, T. Clavel, et al.
Y. Song, C. Liu, J. Lee, M. Bolanos, M.L. Vaisanen, S.M. Finegold.
18
ACCEPTED MANUSCRIPT
408
[18]
T.J. Krogh, C.N. Agergaard, J. Møller-Jensen, U.S. Justesen. Draft Genome
409
Sequence of Parabacteroides goldsteinii with Putative Novel Metallo-β-Lactamases
410
Isolated from a Blood Culture from a Human Patient. Genome Announc 3 (2015).
411
[19]
412
report of Parabacteroides goldsteinii bacteraemia in a patient with complicated intra-
413
abdominal infection. Anaerobe 16 (2010) 223-5.
414
[20]
415
vitro. Antimicrob Agents Chemother 16 (1979) 554-60.
416
[21]
417
Induction of colonic regulatory T cells by indigenous Clostridium species. Science
418
331 (2011) 337-41.
419
[22]
420
microbiota and the immune system. Science 336 (2012) 1268-73.
421
[23]
422
immunomodulatory molecule of symbiotic bacteria directs maturation of the host
423
immune system. Cell 122 (2005) 107-18.
424
[24]
425
Commensal bacteria-derived signals regulate basophil hematopoiesis and allergic
426
inflammation. Nat Med 18 (2012) 538-46.
427
[25]
428
gene expression via covalent binding to cellular albumin. Proc Natl Acad Sci U S A
429
110 (2013) 2981-6.
RI PT
F.M. Awadel-Kariem, P. Patel, J. Kapoor, J.S. Brazier, E.J. Goldstein. First
SC
G. Banck, A. Forsgren. Antibiotics and suppression of lymphocyte function in
M AN U
K. Atarashi, T. Tanoue, T. Shima, A. Imaoka, T. Kuwahara, Y. Momose, et al.
TE D
L.V. Hooper, D.R. Littman, A.J. Macpherson. Interactions between the
EP
S.K. Mazmanian, C.H. Liu, A.O. Tzianabos, D.L. Kasper. An
AC C
D.A. Hill, M.C. Siracusa, M.C. Abt, B.S. Kim, D. Kobuley, M. Kubo, et al.
F. Mor, I.R. Cohen. Beta-lactam antibiotics modulate T-cell functions and
430
19
ACCEPTED MANUSCRIPT Table 1 Changes of the body weight, complete blood count and histological changes in the small and large colon after penicillin treatment (% change of body weight calculated to the day before penicillin administration). Sample size was 10 mice per
RI PT
group.
Penicillin administration
Histological changes
156.5 (150-159.3) NS 232.2 (183-240) NS 247 (191-267.4) NS
M AN U
141.3 (134.3-157) 222 (208-231.2) 251 (237-279.2)
2.56 (1.96-3.41) 95.5 (92-97) 4 (3-5) 776.5 (531-897) 9.1 (8.1-9.6) 13.8 (12.6-14.3) 44.4 (41.6-46.3) 48.5 (46.1-51.6)
2.58 (1.07-3.74) 92.5 (90-94) 6 (4-7) 773 (256-905) 9.2 (8.7-9.8) 13.8 (13.1-14.5) 44.6 (42-48.6) 48.4 (46-50.7)
no inflammation
no inflammation
TE D
% Change of body weight (range) Day 7 Day 14 Day 28 CBC value median (min-max): WBC x 103/uL Lymphocytes (% of WBC) Neutrophils (% of WBC) Platelets x 103/uL RBC x 106/uL Hb (g/dl) Ht (%) MCV (fl)
p
SC
Control
NS 0.0002 0.0002 NS NS NS NS NS NS
AC C
EP
CBC – complete blood count, WBC – white blood count, Hb – haemoglobin, Ht – haematocrit, MCV – mean corpuscular volume
ACCEPTED MANUSCRIPT Table 2 Changes in the percentage of lymphocyte subsets in peripheral blood between study groups. Sample size was 10 mice per group.
p
88.1 (74.2-93.2) 82.5 (77.6-92.7) 24.7 (13.2-26.9) 8.1 (3.8-12.9) 21.7 (17.8-31.4) 7.8 (7.4-9.2) 6.6 (5.5-16.3) 3.5 (2.4-4.7) 8.0 (4.9-11.2) 60.7 (56.8-61.4) 38.3 (35.5-40.4)
81.7(65.2-89.6) 75.4 (62.1-80.9) 28.7 (21-39.3) 14.5 (6.5-17.8) 28.2 (25.2-31.3) 10.7 (7.8-12.3) 7.9 (5.8-11) 4.9 (4.7-5.2) 10.5 (8.1-13.2) 55.2 (53.8-57.4) 42.1 (37.6 – 43.8)
0.029 0.0039 NS 0.002 0.007 0.03 NS 0.008 0.01 0.02 NS
EP AC C
SC
RI PT
Penicillin administration median (min-max)
TE D
CD3+ (%) CD4+ (%) CD8+ (%) CD4+CD25+CD127low (%) CD4+CD25+CD127high (%) CD4+ IL-4 (%) CD4+IL-17 (%) CD4+IFNγ (%) CD19+ (%) sIgM+CD45R+CD23- (%) sIgM+CD45R+CD23+ (%)
Control median (min-max)
M AN U
Lymphocyte subset
ACCEPTED MANUSCRIPT
RI PT
ig. 1. Representative H&E staining of small intestine (A) and colon (B) sections of mice after 28 days o
eatment with water vs. water plus penicillin. Histology evaluation revealed no abnormalities in mucosa of termina
AC C
EP
TE D
M AN U
SC
art of ileum (A) and ascending colon (B) in control and penicillin treated mice (control = 10, penicillin = 10).
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
Fig. 2. Differences in gut microbiota between C57BL6/J/cmdb male mice kept in the same conditions. Essent
RI PT
variations in small intestine, colon and faecal microbiota composition at phyla (A) or species (B) taxonomic level in mic
SC
ittermates were observed. Five circles in each square represent five separate mice housed in the same cage. Difference
AC C
EP
TE D
M AN U
n microbiota between littermates were observed both in control and penicillin treated mice.
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
RI PT
Fig. 3. Changes in small intestine, colon and faecal microbiota composition after 28 days of penicillin
treatment. At the phyla taxonomic level there was no significant difference in Firmicutes (A) and Bacteroidetes (B)
SC
presence between control and penicillin treated mice. At the species taxonomic level significant increase of
M AN U
Parabacteroides goldsteinii (C) after penicillin treatment was observed. Each bar represents average from five
AC C
EP
TE D
samples with SD.
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
RI PT
Fig. 4. Representative immunohistochemistry staining of small intestine sections of mice after 28 days of penicillin treatment. Histology evaluation revealed no differences in intestinal infiltration of lymphocytes T CD3+ (A),
AC C
EP
TE D
M AN U
SC
CD4+ (B) and CD8+ (C) and lymphocytes B CD20+ (D) (control = 10, penicillin = 10).
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT Highlights
EP
TE D
M AN U
SC
RI PT
Murine gut microbiota is highly variable between littermate mice Penicillin treatment increases abundance of pathogenic P. goldsteinii bacteria Antibiotic treatment has a profound effect on the immune system
AC C
• • •