PreprintPDF Available

Preexisting memory CD4 T cells in naïve individuals confer robust immunity upon vaccination

Authors:

Abstract and Figures

Antigen recognition through the T cell receptor (TCR) αβ heterodimer is one of the primary determinants of the adaptive immune response. Vaccines activate naïve T cells with high specificity to expand and differentiate into memory T cells that allow for a quick and robust T cell response upon exposure to the pathogen or re-exposure to the vaccine antigen. This is why the induction of memory T cells is a key feature in vaccine development. However, it has become increasingly evident that antigen-specific memory CD4 and CD8 T cells exist in unexposed antigen-naïve hosts and it is likely that exposure to one antigen might alter the TCR repertoire of memory T cells to a different unrelated antigen. In this study, we utilize high-throughput sequencing to profile the memory CD4 TCRβ repertoire and track vaccine-specific TCRβ clonotypes following the de novo administration of hepatitis B (HepB) vaccine in healthy HepB-naïve individuals and show that vaccinees with preexisting vaccine-specific memory CD4 T cell clonotypes elicited earlier and higher antibody concentrations and mounted a more robust CD4 T cell response to the vaccine. We further identify vaccine-specific TCRβ sequence patterns that can be used to predict which individuals will have an early and more vigorous vaccine-elicited immunity to HepB vaccine. Moreover, we find that an expansion of 4-1BB+ memory TREG is a prominent feature in individuals with delayed and modest vaccine-induced immunity. Our approach shows that modeling pre-vaccination TCRβ repertoire enables prediction of both antibody and CD4 responses to vaccines.
Content may be subject to copyright.
1
Preexisting memory CD4 T cells in naïve individuals confer robust
1
immunity upon vaccination
2
Running title: Preexisting memory T cells confer immunity to vaccination
3
George Elias 1,2%, Pieter Meysman 2,3,4%, Esther Bartholomeus 2,5%, Nicolas De Neuter
4
2,3,4, Nina Keersmaekers 2,6, Arvid Suls 2,5, Hilde Jansens 7, Aisha Souquette 8, Hans De Reu
5
1,9, Evelien Smits 1,9, Eva Lion1,9, Paul G. Thomas 8, Geert Mortier 2,5, Pierre Van Damme
6
2,10, Philippe Beutels 2,6, Kris Laukens 2,3,4%, Viggo Van Tendeloo 1,11%, Benson Ogunjimi
7
2,6,12,13%
8
9
% = equal contribution
10
1Laboratory of Experimental Hematology (LEH), Vaccine and Infectious Disease Institute, University of Antwerp,
11
Antwerp, Belgium.
12
2Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), Antwerp, Belgium.
13
3Adrem Data Lab, Department of Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium.
14
4Biomedical Informatics Research Network Antwerp (biomina), University of Antwerp, Antwerp, Belgium.
15
5Department of Medical Genetics, University of Antwerp/Antwerp University Hospital, Edegem, Belgium
16
6Centre for Health Economics Research & Modeling Infectious Diseases (CHERMID), Vaccine & Infectious Disease
17
Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
18
7Department of Clinical Microbiology, Antwerp University Hospital
19
8Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
20
9Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
21
10Centre for the Evaluation of Vaccination (CEV), Vaccine and Infectious Disease Institute, University of Antwerp,
22
Antwerp, Belgium.
23
11Janssen Research & Development, Immunosciences WWDA, Johnson & Johnson, Beerse, Belgium
24
12Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute,
25
University of Antwerp, Antwerp, Belgium
26
13Department of Paediatrics, Antwerp University Hospital, Antwerp, Belgium.
27
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
2
Corresponding authors: George Elias, Benson Ogunjimi
28
E-mail address:
29
george.elias@student.uantwerpen.be, benson.ogunjimi@uantwerp.be
30
Laboratory of Experimental Hematology (LEH), Vaccine and Infectious Disease Institute,
31
University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
32
33
34
35
36
Footnotes
37
This work was funded by
38
1. University of Antwerp [BOF Concerted Research Action (PS ID 30730) Methusalem
39
funding; Industrial Research Fund SBO].
40
2. Research Foundation Flanders (1861219N grant to B. Ogunjimi and FWO SB grant to N.
41
De Neuter.
42
3. ALSAC at St. Jude Children’s Research Hospital and by HSN272201400006C and
43
R01AI107625 from the National Institute of Allergy and Infectious Disease.
44
45
46
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
3
Abstract
47
Antigen recognition through the T cell receptor (TCR) αβ heterodimer is one of the primary
48
determinants of the adaptive immune response. Vaccines activate naïve T cells with high
49
specificity to expand and differentiate into memory T cells that allow for a quick and robust T
50
cell response upon exposure to the pathogen or re-exposure to the vaccine antigen. This is why
51
the induction of memory T cells is a key feature in vaccine development. However, it has
52
become increasingly evident that antigen-specific memory CD4 and CD8 T cells exist in
53
unexposed antigen-naïve hosts and it is likely that exposure to one antigen might alter the TCR
54
repertoire of memory T cells to a different unrelated antigen. In this study, we utilize high-
55
throughput sequencing to profile the memory CD4 TCRβ repertoire and track vaccine-specific
56
TCRβ clonotypes following the de novo administration of hepatitis B (HepB) vaccine in healthy
57
HepB-naïve individuals and show that vaccinees with preexisting vaccine-specific memory CD4
58
T cell clonotypes elicited earlier and higher antibody concentrations and mounted a more robust
59
CD4 T cell response to the vaccine. We further identify vaccine-specific TCRβ sequence patterns
60
that can be used to predict which individuals will have an early and more vigorous vaccine-
61
elicited immunity to HepB vaccine. Moreover, we find that an expansion of 4-1BB+ memory
62
TREG is a prominent feature in individuals with delayed and modest vaccine-induced immunity.
63
Our approach shows that modeling pre-vaccination TCRβ repertoire enables prediction of both
64
antibody and CD4 responses to vaccines.
65
66
Keywords
67
CD4 T cell, T cell receptor, immune repertoire sequencing, vaccination
68
69
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
4
70
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
5
Introduction
71
Antigen recognition through the T cell receptor (TCR) is one of the key determinants of the
72
adaptive immune response 1. Antigen presentation via major histocompatibility complex (MHC)
73
(encoded by HLA genes), together with the right costimulatory and cytokine signals, are
74
responsible for T cell activation 2,3. In this system, every T cell receptor (TCR) αβ heterodimer
75
imparts specificity for a peptide-MHC (pMHC) complex. A highly diverse TCR repertoire
76
ensures that an effective T cell response can be mounted against pathogen-derived peptides 4.
77
High TCRαβ diversity is generated through V(D)J recombination at the complementary-
78
determining region 3 (CDR3) of TCRα and TCRβ chains, accompanied with junctional deletions
79
and insertions of nucleotides, further adding to the diversity 5.
80
Vaccines activate naïve T cells with high specificity to vaccine-derived peptides and induce their
81
expansion and differentiation into effective and multifunctional T cells. This is followed by a
82
contraction phase from which surviving cells constitute a long-lived memory T cell pool that
83
allows for a quick and robust T cell response upon a second exposure to the pathogen 6.
84
However, recent work has shown that a prior pathogen encounter is not a prerequisite for the
85
formation of memory T cells and that CD4 T cells with a memory phenotype can be found in
86
antigen-naïve individuals 7. The existence of memory-like CD4 T cells in naïve individuals 8 can
87
be explained by molecular mimicry, as the encounter with environmentally-derived peptides
88
activates cross-reactive T cells due to the highly degenerate nature of the CD4 T cell recognition
89
of peptide-MHC complex 9. Indeed, work that attempted to replicate the history of human
90
pathogen exposure in mice has shown that sequential infections altered the immunological
91
profile and remodeled the immune response to vaccination 10. The existence of memory CD4 T
92
cells specific to vaccine-derived peptides in unexposed individuals might confer an advantage in
93
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
6
vaccine-induced immunity. In the present study we used high-throughput sequencing to profile
94
the memory CD4 TCRβ repertoire of healthy adults before and after administration of a hepatitis
95
B vaccine to investigate the impact of preexisting memory CD4 T cells on the immune response
96
to the vaccine. Based on anti-hepatitis B surface (anti-HBs) antibody titers over 365 days,
97
vaccinees were grouped into early, late and non-converters. Our data reveals that individuals
98
with preexisting vaccine-specific CD4 T cell clonotypes in the memory CD4 compartment had
99
earlier emergence of antibodies and mounted a more vigorous CD4 T cell response to the
100
vaccine. Moreover, we identify a set of vaccine-specific TCRβ sequence patterns which can be
101
used to predict which individuals will have an early and more vigorous response to hepatitis B
102
vaccine.
103
104
105
106
107
108
109
110
111
112
113
114
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
7
Methods
115
Human study design and clinical samples. A total of 34 healthy individuals (20-29y: 10, 30-
116
39y: 7, 40-49y: 16, 50+y: 1) without a history of HBV infection or previous hepatitis B
117
vaccination were recruited in this study after obtaining written informed consent. Individuals
118
were vaccinated with a hepatitis B vaccine by intramuscular (m. deltoideus) injection (Engerix-
119
B® containing 20 μg dose of alum-adjuvanted hepatitis B surface antigen, GlaxoSmithKline) on
120
days 0 and 30 (and on day 365). At days 0 (pre-vaccination), 60, 180 and 365, peripheral blood
121
samples were collected on spray-coated lithium heparin tubes, spray-coated K2EDTA
122
(dipotassium ethylenediamine tetra-acetic acid) tubes and serum tubes (Becton Dickinson, NJ,
123
USA). The study was approved by the Ethics Committee of Antwerp University Hospital and
124
University of Antwerp (Antwerp, Belgium).
125
126
Peripheral blood mononuclear cells. Peripheral blood mononuclear cells (PBMC) were
127
isolated by Ficoll-Paque Plus gradient separation (GE Healthcare, Chicago, IL, USA). Cells were
128
stored in 10% dimethyl sulfoxide in fetal bovine serum (Thermo Fisher Scientific, Waltham,
129
MA, USA). After thawing and washing cryopreserved PBMC, cells were cultured in AIM-V
130
medium that contained L-glutamine, streptomycin sulfate at 50 µg/ml, and gentamicin sulfate at
131
10 µg/ml (Thermo Fisher Scientific, Waltham, MA, USA) and supplemented with 5% human
132
serum (One Lambda, Canoga Park, CA, USA).
133
134
Serology and complete blood count. Serum was separated and stored immediately at 80°C
135
until time of analysis. Anti-HBs antibody was titrated in serum from day 0, 60, 180 and 365
136
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
8
using Roche Elecsys® Anti-HBs antibody assay on an Elecsys® 2010 analyzer (Roche, Basel,
137
Switzerland). An anti-HBs titer above 10 IU/ml was considered protective 11.
138
Serum IgG antibodies to Cytomegalovirus (CMV), EpsteinBarr virus viral-capsid antigen
139
(EBV-VCA), and Herpes Simplex virus (HSV)-1 and 2 were determined using commercially
140
available sandwich ELISA kits in accordance with the manufacturer’s instructions.
141
A complete blood count including leukocyte differential was run on a hematology analyzer
142
(ABX MICROS 60, Horiba, Kyoto, Japan).
143
144
Sorting of memory CD4 T cells. Total CD4 T cells were isolated by positive selection using
145
CD4 magnetic microbeads (Miltenyi Biotech, Bergisch Gladbach, Germany). Memory CD4 T
146
cells were sorted after gating on single viable CD3+CD4+CD8CD45RO+ cells. The following
147
fluorochrome-labeled monoclonal antibodies were used for staining: CD3-PerCP (BW264/56)
148
(Miltenyi Biotech), CD4-APC (RPA-T4) and CD45RO-PE (UCHT1) (both from Becton
149
Dickinson, Franklin Lakes, NJ, USA) and CD8-Pacific Orange (3B5) (from Thermo Fisher
150
Scientific, Waltham, MA, USA). Cells were stained at room temperature for 20 min and sorted
151
with FACSAria II (Becton Dickinson). Sytox blue (Thermo Fisher Scientific) was used to
152
exclude non-viable cells.
153
154
Single peptides, matrix peptide pools and epitope mapping. A set of 15-mers peptides with an
155
11-amino acid overlap spanning the 226 amino acids along the small S protein of hepatitis B
156
(HB) surface antigen (HBsAg), also designated as small HBs (SHBs) 12, were synthesized by
157
JPT Peptide Technologies (Berlin, Germany). The set, composed of 54 single peptides (See
158
supplementary table 1), was used in a matrix-based strategy to map epitopes against which the
159
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
9
immune response is directed 13. The matrix layout enables efficient identification of epitopes
160
within the antigen using a minimal number of cells. For this purpose, a matrix of 15 pools, 7
161
rows and 8 columns, referred to as matrix peptide pool, was designed so that each peptide is in
162
exactly one row-pool and one column-pool, thereby allowing for the identification of positive
163
peptides at the intersection of positive pools. Matrix peptide pools that induced a CD4 T cell
164
response (as determined by CD40L/CD154 assay described below) which meets the threshold
165
criteria for a positive response were considered in the deconvolution process. Top six single
166
peptides were considered for peptide-specific T cell expansion and sorting. A master peptide
167
pool is composed of all of the 54 single peptides and was used to identify and sort total vaccine-
168
specific CD4 T cells. Each peptide was used at a final concentration of 2 µg/ml.
169
170
171
Ex vivo T cell stimulation (CD40L/CD154 assay). Thawed PBMC from each vaccinee were
172
cultured in AIM-V medium that contained L-glutamine, streptomycin sulphate at 50 µg/ml, and
173
gentamicin sulphate at 10 µg/ml. (GIBCO, Grand Island, NY) and supplemented with 5% human
174
serum (One Lambda, Canoga Park, CA, USA). Cells were stimulated for 6 hours with 2 μg/ml of
175
each of the 15 matrix peptide pools in the presence of 1 µg/ml anti-CD40 antibody (HB14)
176
(purchased from Miltenyi Biotec, Bergisch Gladbach, Germany) and 1 μg/ml anti-CD28
177
antibody (CD28.2) (purchased from BD Biosciences, Franklin Lakes, NJ, USA).
178
Cells were stained using the following fluorochrome-labelled monoclonal antibodies: CD3-
179
PerCP (BW264/56), CD4-APC (REA623), CD8-VioGreen (REA734) and CD40L-PE (5C8)
180
(purchased from Miltenyi Biotec, Bergisch Gladbach, Germany). Viability dye Sytox blue from
181
Invitrogen (Thermo Fisher Scientific, Waltham, MA, USA) was used to exclude non-viable cells.
182
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
10
Data was acquired on FACSAria II using Diva Software, both from BD Biosciences (Franklin
183
Lakes, NJ, USA), and analyzed on FlowJo software version 10.5.3 (Tree Star, Inc., Ashland, OR,
184
USA). Fluorescence-minus-one controls were performed in pilot studies. Gates for CD40L+CD4
185
T cells were set using cells left unstimulated.
186
187
In vitro T cell expansion and cell sorting. Thawed PBMC were labelled with
188
carboxyfluorescein succinimidyl ester (CFSE) (Invitrogen, Carlsbad, CA, USA) and cultured in
189
AIM-V medium that contained L-glutamine, streptomycin sulphate at 50 µg/ml, and gentamicin
190
sulphate at 10 µg/ml. (GIBCO, Grand Island, NY) and supplemented with 5% human serum
191
(One Lambda, Canoga Park, CA, USA). Cells were stimulated for 7 days with 2 µg/ml of
192
selected single peptides in addition to the master peptides pool. Cells were stained using the
193
following fluorochrome-labelled monoclonal antibodies: CD3-PerCP (BW264/56), CD4-APC
194
(REA623) and CD8-VioGreen (REA734) (purchased from Miltenyi Biotec, Bergisch Gladbach,
195
Germany). Viability dye Sytox blue from Invitrogen (Thermo Fisher Scientific, Waltham, MA,
196
USA) was used to exclude non-viable cells. Single viable CFSElow CD3+ CD8 CD4+ T cells
197
were sorted into 96-well PCR plates containing DNA/RNA Shield (Zymo Research, Irvine, CA,
198
USA) using FACSAria II and Diva Software (BD Biosciences, Franklin Lakes, NJ, USA). For
199
each of the selected single peptides, 500 cells were sorted in two technical replicates. For the
200
master peptide pool, 1000 cells were sorted in two technical replicates. Plates were immediately
201
centrifuged and kept at 20°C before TCR cDNA library preparation and sequencing.
202
203
TCRβ cDNA Library Preparation and Sequencing of memory CD4 T cells. DNA was
204
extracted from sorted memory CD4 T cells using Quick-DNA Microprep kit (Zymo Research,
205
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
11
Irvine, CA, USA). ImmunoSEQ hsTCRB sequencing kit (Adaptive Biotechnologies, Seattle,
206
WA, USA) was used to profile TCRβrepertoire following the manufacturer’s protocol.
207
After quality control using Fragment Analyzer (Agilent, Santa Clara, CA, USA), libraries were
208
pooled with equal volumes. The concentration of the final pool was measured with the Qubit™
209
dsDNA HS Assay kit (Thermo Fisher Scientific, Waltham, MA, USA). The final pool was
210
processed to be sequenced on the Miseq and NextSeq platforms (Illumina, San Diego, CA,
211
USA).
212
213
TCR cDNA Library Preparation and Sequencing of CFSElow CD4 T cells. RNA was
214
extracted from each of the two technical replicates of sorted CFSElow CD4 T cells using Quick-
215
RNA Microprep kit (Zymo Research, Irvine, CA, USA). Without measuring the resulted RNA
216
concentration, an RNA-based library preparation was used. The QIAseq Immune Repertoire
217
RNA Library kit (Qiagen, Venlo, Netherlands) amplifies TCR alpha, beta, gamma and delta
218
chains. After quality control using Fragment Analyzer (Agilent, Santa Clara, CA, USA),
219
concentration was measured with the Qubit™ dsDNA HS Assay kit (Thermo Fisher Scientific,
220
Waltham, MA, USA) and pools were equimolarly pooled and prepared for sequencing on the
221
Nextseq platform (Illumina, San Diego, CA, USA).
222
223
TCRβ Sequence Analysis. TCRβ clonotypes were identified as previously described 14 where a
224
unique TCRβ clonotype is defined as a unique combination of a V gene, CDR3 amino acid
225
sequence, and J gene. All memory CD4 T cell DNA-based TCRβ sequencing reads were
226
annotated using the immunoSEQ analyzer (v2) from Adaptive Biotechnologies. All small bulk
227
RNA-based TCR sequencing reads were annotated using the MiXCR tool (v3.0.7) from the
228
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
12
FASTQ files. As all RNA-based TCR sequencing experiments featured two technical replicates,
229
only those TCR sequences that occurred in both replicates were retained and their counts were
230
summed. Tracking of vaccine-specific TCRβ clonotypes is based on exact TCRβ CDR3 amino
231
acid matches to remove any bias introduced by the different VDJ annotation pipelines. Non-
232
HBsAg TCR annotations were done with the TCRex web tool 15 on the 24th of July, 2019 using
233
version 0.3.0. Inference of similar epitope binding between two TCR sequences is defined
234
according to the Hamming distance (d) calculated on the CDR3 amino acid sequence with a
235
cutoff c, as supported by Meysman et al. 16. All scripts used in this analysis are available via
236
github (https://github.com/pmeysman/HepBTCR).
237
238
Predictive HBs-response model. From the single peptide data generated in the matrix peptide
239
pool experiments, we aimed to create a predictive model to enumerate the HBs response from
240
full TCRβ repertoire data. This approach allows for predictions that are epitope-specific rather
241
than simply vaccine-specific. This model was applied in a leave-one-out cross validation so that
242
vaccine-specific TCRβ sequences from a vaccinee are not used to make predictions for the same
243
vaccinee. While the predictive model is derived from epitope-specific data, it cannot be
244
guaranteed that some of the expanded CD4 T cells detected in the in vitro assay are not due to
245
bystander activation. Vaccine-specific TCRβ sequences of vaccinees who did not respond to the
246
vaccine at day 60 (late-converters and non-converters) are expected to be more enriched in cells
247
triggered to expand due to bystander activation. Indeed, running the set of vaccine-specific
248
TCRβ sequences through the TCRex webtool 15 reveals that there is strong enrichment for the
249
CMV NLVPMVATV epitope (P value = 1.16e-71) and the Mart-1 variant ELAGIGILTV
250
epitope (P value = 2.16e-60), which supports the notion that some of these TCRβ sequences
251
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
13
might not be specific to HBsAg. This set of vaccine-specific TCRβ sequences can thus be used
252
to make predictions about possible TCRβ sequences due to bystander activation of CD4 T cells,
253
i.e. common TCRβ sequences that might be present as false positives. The final output of the
254
model is thus a ratio Rhbs for any repertoire repi describing a set of TCRβ sequences trepi:
255
󰇛󰇜   


  
256
with tpep as the set of TCRβ sequences occurring in both biological replicates for a single sample
257
and a single peptide (pep) from the HBsAg matrix peptide pool experiment, and tbystander as the
258
set of TCRβ sequences occurring in both biological replicates of the master peptide pool in any
259
of the non-responding samples. Thus the ratio signifies the number of TCR clonotypes predicted
260
to be reactive against one of the HBsAg peptides, normalized by a count of putative false
261
positive predictions from bystander T-cells.
262
263
Ex vivo T cell phenotyping of vaccine-specific T cells. Thawed PBMC from each vaccinee
264
were cultured in AIM-V medium that contained L-glutamine, streptomycin sulphate at 50 µg/ml,
265
and gentamicin sulphate at 10 µg/ml. (GIBCO, Grand Island, NY) and supplemented with 5%
266
human serum (One Lambda, Canoga Park, CA, USA). Cells were stimulated for 6 hours with 2
267
μg/ml of a master peptide pool representing the full length of the small surface envelope protein
268
of hepatitis B, in the presence of 1 µg/ml anti-CD40 antibody (HB14) (purchased from Miltenyi
269
Biotec, Bergisch Gladbach, Germany) and 1 μg/ml anti-CD28 antibody (CD28.2) (purchased
270
from BD Biosciences, Franklin Lakes, NJ, USA). Cells were stained using the following
271
fluorochrome-labelled monoclonal antibodies:
272
CD3-BV510 (SK7), CD4-PerCP/Cy5.5 (RPA-T4), CD8-APC/Cy7 (SK1), CD45RA-AF488
273
(HI100), CD25-BV421 (M-A251), CD127-BV785 (A019D5) and CD137-PE (4-1BB)
274
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
14
(purchased from BioLegend, San Diego, CA, USA), CXCR5 (CD185)-PE-eFluor 610
275
(MU5UBEE) (from eBioscience, Thermo Fisher Scientific, Waltham, MA, USA) and CD40L-
276
APC (5C8) (purchased from Miltenyi Biotec, Bergisch Gladbach, Germany). Fixable viability
277
dye Zombie NIR™ from BioLegend (San Diego, CA, USA) was used to exclude non-viable
278
cells. Data was acquired on FACSAria II using Diva Software, both from BD Biosciences
279
(Franklin Lakes, NJ, USA), and analyzed on FlowJo software version 10.5.3 (Tree Star, Inc.,
280
Ashland, OR, USA) using gating strategy shown in Fig. S1a. Fluorescence-minus-one controls
281
were performed in pilot studies. Gates for CD40L+ and 4-1BB+ CD4 T cells (Fig. S1b) were set
282
using cells left unstimulated (negative control contained DMSO at the same concentration used
283
to solve peptide pools). In order to account for background expression of CD40L and 4-1BB on
284
CD4 T cells, responses in cells left unstimulated were subtracted from the responses to peptides,
285
and when peptides-specific CD40L+ or 4-1BB+ CD4 T cells were not significantly higher than
286
those detected for cells left unstimulated (using one-sided Fisher's exact test), values were
287
mutated to zero.
288
289
Statistical analysis and data visualization. The two-sided Fisher’s exact test was used to
290
evaluate the significance of relationship between early/late-converters and CMV, EBV or HSV
291
seropositivity. For the visualization of marker expression, TCRβ counts and cell frequencies
292
between time points or groups of vaccinees, ggplot2 (V3.3.2) and ggpubr (V0.2.5) packages in R
293
were used. The Wilcoxon signed-rank test was used to compare two or more groups, with
294
unpaired and paired analysis as necessary. The nonparametric Spearman's rank-order correlation
295
was used to test for correlation. We used the following convention for symbols indicating
296
statistical significance; ns P > 0.05, * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001.
297
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
15
Statistical analysis. The two-sided Fisher’s exact test was used to evaluate the significance of
298
relationship between early/late-converters and CMV, EBV or HSV seropositivity. For the
299
visualization of marker expression, TCRβ counts and cell frequencies between time points or
300
groups of vaccinees, ggpubr (V0.2.5) package in R was used. The Wilcoxon signed-rank test was
301
used to compare two or more groups, with unpaired and paired analysis as necessary. The
302
nonparametric Spearman's rank-order correlation was used to test for correlation. We used the
303
following convention for symbols indicating statistical significance; ns P > 0.05, * P ≤ 0.05, ** P
304
≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001.
305
306
Data availability
307
The sequencing data that support the findings of this study have been deposited on Zenodo
308
(https://doi.org/10.5281/zenodo.3989144). Flow Cytometry Standard (FCS) data files with
309
associated FlowJo workspaces are deposited at flowrepository.org 17 under the following
310
experiment names: epitope mapping: https://flowrepository.org/id/FR-FCM-Z2TN; in vitro T
311
cell expansion: https://flowrepository.org/id/FR-FCM-Z2TM; ex vivo CD4 T cell assay:
312
https://flowrepository.org/id/FR-FCM-Z2TL.
313
314
315
316
317
318
319
320
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
16
Results:
321
Vaccinee cohort can be classified into three groups
322
Out of 34 vaccinees, 21 vaccinees seroconverted (an anti-HBs titer above 10 IU/ml was
323
considered protective 11) at day 60 and were classified as early-converters; 9 vaccinees
324
seroconverted at day 180 or day 365 and were classified as late-converters; remaining 4
325
vaccinees had an anti-HBs antibody titer lower than 10 IU/ml at all time points following
326
vaccination and were classified as non-converters (Fig. 1 and Fig. S2a).
327
Members of Herpesviridae family might alter immune responses to vaccines 18. We found no
328
significant differences in CMV, EBV or HSV seropositivity between the three groups in our
329
cohort (Fig. S2b). Early-converters were slightly younger than late-converters and non-
330
converters were notably younger than both early and late converters (Fig. S2c).
331
332
333
334
335
336
337
338
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
17
339
Figure 1. Hepatitis B vaccination (Engerix-B®) study design.
340
a Hepatitis B (Engerix-B®) vaccination and experimental design. (Top) Timeline of vaccination
341
and blood collection. (Bottom) Memory CD4 T cells were magnetically enriched and FACS-
342
sorted from two time points (day 0 and day 60) for TCRβ repertoire sequencing. Matrix peptide
343
pools were used to map CD4 T cell epitopes of the vaccine from PBMCs collected at day 60 and
344
to select single peptides. After 7 days of in vitro expansion, single peptide-specific and master
345
peptide pool-specific CFSElow CD4 T cells from PBMCs collected at day 60 were FACS-sorted
346
in two technical replicates for TCRβ repertoire sequencing. PBMCs collected at days 0, 60, 180,
347
and 365 were stimulated with the master peptide pool (HBsAg) and assessed for converse
348
expression of 4-1BB and CD40L by flow cytometry.
349
b Vaccinee cohort can be classified into three groups as determined by anti-Hepatitis B surface
350
(anti-HBs) titer over four times points.
351
Early-converters seroconverted at day 60, late-converters seroconverted at day 180 or day 365
352
and nonconverters did not have an anti-HBs titer higher than 10 IU/ml at any of the time points.
353
354
355
356
a b
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
18
Memory CD4 T cell repertoire decreases in clonality following vaccination in early-
357
converters
358
A genomic DNA-based TCRβ sequence dataset of memory CD4 T cells isolated from peripheral
359
blood was generated from a cohort of 34 healthy vaccinees right before vaccination (day 0) and
360
60 days after administration of the first dose of hepatitis B vaccine (30 days after administration
361
of the second vaccine dose).
362
Between 7.32 × 104 and 4.41 × 105 productive TCRβ sequence reads were obtained for each
363
vaccinee at each time point (Fig. S3a). Between 30,000 and 90,000 unique TCRβ sequences
364
were sequenced for each vaccinee at each time point (Fig. S3b). As expected, considering the
365
extremely diverse memory CD4 T cell repertoire 19, only an average of 25% of the TCRβ
366
sequences can be detected at both time points for each vaccinee (Fig. S3c).
367
368
The diversity of the memory CD4 T cell repertoire of each vaccinee at the two time points was
369
explored. Even though the number of unique clones in the memory CD4 TCRβ repertoire
370
remained stable in between the two time points, we detected a significant increase in the TCRβ
371
repertoire Shannon’s entropy for early-converters (Fig. 2a) (P value = 0.042), but not late-
372
converters, suggesting that the memory CD4 T cell repertoires of early-converters have become
373
less clonal, despite the number of distinct TCRβ sequences not changing significantly.
374
375
Unique vaccine-specific TCRβ sequences are trackable within memory CD4 T cell
376
repertoire and increase following vaccination
377
Peripheral blood mononuclear cells from day 60 were labeled with carboxyfluorescein
378
succinimidyl ester (CFSE) and stimulated with a pool of peptides spanning hepatitis B (HB)
379
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
19
surface antigen (HBsAg). At day 7 of in vitro expansion, we sorted CFSElow CD4 T cells 20 and
380
extracted mRNA for quantitative assessment of vaccine-specific TCRβ clonotypes by sequencing
381
(see Methods for details), allowing for the tracking of vaccine-specific TCRβ within memory
382
CD4 T cell repertoire over the two times points, based on CDR3β amino acid sequence mapping.
383
We detected a significant increase in the frequency of unique vaccine-specific TCRβ sequences
384
at day 60 post-vaccination compared to pre-vaccination (mean increase = 96.5%, 95% CI = 56.7
385
- 170%) (Fig. 2b). Moreover, this increase was larger for early-converters (mean = 132.1%, 95%
386
CI = 76.4 - 238.2%) than late-converters (mean = 22.1%, 95% CI = 5.9 50.1%). For non-
387
converters the mean was 81.6% (95% CI: [42.7% - 110.6%]). A Wilcoxon test shows that the
388
difference between the increase for the early converters and late converters had a P value of
389
0.04909.
390
As vaccine-specific TCRβ sequences were already detected in the memory CD4 T cell repertoire
391
prior to vaccination, we sought to determine whether the vaccination results in an expansion of
392
those sequences. However, using the abundance of vaccine-specific TCRβ sequences within the
393
memory CD4 T cell repertoire, the data does not support a vaccine-induced expansion of
394
preexisting vaccine-specific TCRβ sequences (Fig. S3d). Thus, although we see a rise in the
395
number of vaccine-specific TCRβ clonotypes from day 0 to day 60, this cannot be attributed to
396
an expansion of preexisting TCRβ clonotypes but rather the recruitment of new TCRβ
397
clonotypes (presumably from the naïve T cell compartment), as visualized for one vaccinee in
398
Fig. 2c.
399
It makes sense to not only look at the difference in vaccine-specific TCRβ sequences between
400
time points, but also explore whether there are differences in the proportion of HBsAg-specific
401
clones in the memory repertoire between early-converters, late-converters and non-converters
402
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
20
after vaccination. In this case, as we aim for a between-vaccinees comparison (in contrast to the
403
within-vaccinees timepoint comparison), we normalize by the number of HBsAg-specific TCRβ
404
found for each vaccinee. Thus, the values are different from those reported before. From this
405
analysis, it can be concluded that there is a difference in HBsAg-specific TCRβ at day 60
406
between the three groups (Fig. 2d) (ANOVA P value = 0.00238). A Wilcoxon test between
407
early-converters and other vaccinees shows a significant P value of 0.000473, thereby implying
408
that early-converters have a higher relative frequency of vaccine-specific TCRβ sequences
409
present in their memory repertoire at day 60 compared to the rest.
410
It is notable that some of the vaccine-specific T cell clonotypes can be detected in the memory
411
repertoire of vaccinees prior to vaccination (Fig. 2e). However, the relative frequency was not
412
statistically significant different between the categories (ANOVA P value = 0.238).
413
414
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
21
415
Figure 2. CD4 T cell memory TCRβ repertoire and vaccine-specific TCRβ clonotypes.
416
a Repertoire diversity and entropy between the two time points
417
b Frequency of unique vaccine-specific TCRβ sequences out of total sequenced TCRβ sequences
418
between two time points for all vaccinees colored by group
419
c Sequenced CD4+ TCR memory repertoire of vaccinee H35 at day 60. Each TCR clonotype is
420
represented by a node. TCRs are connected by an edge if their Hamming distance is one. Only
421
clusters with at least three TCRs are shown. TCR clonotypes in red are the vaccine-specific
422
TCRβ sequences that were not present prior to vaccination.
423
d Frequency of vaccine-specific TCRβ sequences within memory CD4 T cell repertoire
424
normalized by amount of HBsAg-specific TCRβ found for each vaccinee at time point 60 and (e)
425
at time point 0.
426
427
a
d
b
e
c
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
22
HBsAg single peptide-specific TCRβ identification allows predictive modelling of early
428
converters prior to vaccination
429
To quantify the T-cell response at the level of individual peptides that make up the HBsAg, a
430
matrix peptide pool covering 54 overlapping peptides of the HBsAg was used to extract peptide-
431
specific T-cells using a CD40L/CD154 activation-induced marker (AIM) assay (see Methods for
432
details, Fig. S4). The top 6 peptides for each individual were selected for TCR sequencing after a
433
CFSE assay (Supplementary Table 2). In this manner, TCRβ sequences were identified for T-
434
cells reactive against 44 individual HBsAg peptides. These were not uniformly distributed across
435
the HBsAg amino acid sequence, with the most prominent epitopes covering the regions 1-15,
436
129-144, 149-164, 161-176, 181-200, 213-228. For each of those regions, more than 10
437
individuals had a strong T-cell response and more than 150 unique TCRβ sequences could be
438
identified (Fig. 3a).
439
These peptide-specific TCRβ sequences can be utilized in a peptide-TCR interaction classifier to
440
identify other TCRβ that are likely to react against the same HBsAg epitopes, as it has been
441
shown that similar TCRβ sequences tend to target the same epitopes 16,21. These classifications
442
were integrated into a model which outputs a ratio Rhbs for any TCRβ repertoire representing the
443
amount of HBsAg peptide-specific clonotypes. The ratio Rhbs is based on the frequency of
444
putative peptide-specific TCRβ divided by a normalization term for putative false positive
445
predictions due to bystander activations in the training data set. This model applied to the
446
memory repertoire at day 60 shows that early-converters tend to have a higher frequency of
447
putative HBsAg peptide-specific TCRβ, while late-converters tend to have relatively more false
448
positive hits (Fig. 3b). Thus, the defined ratio Rhbs shows significant difference between the
449
early-converters and the late-converters at day 60 (one-sided Wilcoxon-test P value= 0.0313,
450
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
23
Fig. 3c). Furthermore, calculating the Rhbs on the memory repertoires prior to vaccination (day 0)
451
shows a similar difference (one-sided Wilcoxon-test P value= 0.0010, Fig. 3d). In this manner,
452
Rhbs has predictive potential and can be used as a classifier to distinguish early from late-
453
converters prior to vaccination (Fig. 3e), with an AUC of 0.825 (95% CI: 0.657 0.994) in a
454
leave-one-out cross validation setting.
455
While Rhbs is able to differentiate between early-and late-converters, it seems to be worse at
456
distinguishing non-converters. This is mainly due to a single non-converter vaccinee (H21) with
457
a high Rhbs, signifying a high number of putative HBsAg peptide-specific TCRβ in their memory
458
repertoire.
459
460
Predictive capacity of TCRβ repertoire holds true for humoral and cellular immune
461
response
462
Response groups used thus far were established based on anti-HBs titers at different time points.
463
However, response groups can be defined differently if one considers the results from the
464
CD40L assay. For the most part, only those with a high Ab titer at day 60, had a positive value
465
for the CD40L assay (Fig S3e). The only exception was H21, who had a low measurable Ab titer
466
but had a measurable CD40L value. Thus, this vaccinee seems to have had a T-cell response
467
against the vaccine, without developing an antibody response. It is interesting to note that this
468
vaccinee is classified as a non-responder in the data set but it is predicted as a possible strong
469
early-converter by the Rhbs predictive model in the leave-one-out cross validation. Redoing the
470
analysis with Rhbs ratio to predict the CD40L assay at day 60, shows that the Rhbs ratio is a good
471
classifier in a leave-one-out cross validation (Fig. 3f). We tested the predictive capacity of Rhbs
472
ratio at different time points and cell types, and found good AUC values for both 4-
473
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
24
1BBCD40L+ (TCON) and 4-1BB+CD40L (TREG, see further) at day 60, but to a lesser extent at
474
days 180 and 365, thereby indicating that Rhbs predictive model is currently limited to predicting
475
mid-term responses following vaccination (Fig S3f and g).
476
477
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
25
478
Figure 3
479
0
100
200
300
050 100 150 200
HBsAg amino acid position
Unique ep itopespecific TCR .. sequences
MENITSGFLGPLLVLQAGFFLLTRILTIPQSLDSWWTSLNFLGGSPVCLGQNSQSPTSNHSPTSCPPICPGYRWMCLRRFIIFLFILLLCLIFLLVLLDYQGMLPVCPLIPGSTTTNTGPCKTCT TPAQGNSMFPSCCCTKPTDGNCTCIPIPSSWAFAKYLWEWASVRFSWLSLLVPFVQWFVGLSPTVWLSAIWMMWYWGPSLYSIVSPFIPLL
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
26
a. Overview of found HBsAg epitope-specific TCRβ sequences. Each bar corresponds to unique
480
TCRβ sequences found against a single 15mer HBsAg peptide, with 11 amino acid overlap to
481
each subsequent peptide. Bars in blue denote those epitopes for which 10 or more volunteers had
482
a strong T-cell reaction. Motif logos on top of bars denote a sampling of the most common
483
TCRβ amino acid sequence motifs for those epitopes.
484
b. Scatter plot with the percentage predicted epitope-specific and bystander TCRβ sequences.
485
Predictions done as a leave-one-out cross-validation. Each dot represents a vaccinee with the
486
color denoting the responder status (blue: early-converter, yellow: late-converter, red: non-
487
converter)
488
c. HBsAg-predictive ratio R(hbs) when calculated on the memory repertoires at day 60
489
d. HBsAg-predictive ratio R(hbs) when calculated on the memory repertoires at day 0
490
e. ROC curve of using epitope-specific / bystander ratio to differentiate between early-converters
491
and late-converters in a leave-one-out cross validation at day 0. Diagonal line denotes a random
492
classifier. Reported is the area under the curve (AUC) and its 95% confidence interval
493
f. ROC curve of using epitope-specific / bystander ratio to differentiate between samples with a
494
positive CD4L value (> 0) in a leave-one-out cross validation. Diagonal line denotes a random
495
classifier. Reported is the area under the curve (AUC) and its 95% confidence interval. Note that
496
the classifier is worse as several samples (EC, LC and NC) had NA values for the CD154 assay,
497
which were all set to zero.
498
499
500
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
27
Vaccine-specific conventional and regulatory memory CD4 T cells induced in early-
501
converters
502
After showing evidence for the existence of vaccine-specific TCRβ sequences pre-vaccination
503
and that individuals with a higher number of HBsAg peptide-specific clonotypes had earlier
504
seroconversion, we attempted to link this observation to differences in vaccine-specific CD4 T
505
cells responses using CD4 T cell assays. As TREG cells might suppress vaccine-induced immune
506
responses 22, we used activation markers CD40L (CD154) and 4-1BB (CD137) to help delineate
507
the conventional (TCON) and regulatory (TREG) phenotypes of activated CD4 T cells 23,24. In this
508
scheme, after 6 hours of antigen stimulation, CD40L+4-1BB can be used as a signature for
509
antigen-specific CD4 TCON cells, as opposed to CD40L4-1BB+ signature for antigen-specific
510
CD4 TREG cells.
511
Additionally, we added CD25 and CD127 to better identify TREG cells 25,26 and CXCR5 to further
512
distinguish circulatory T follicular helper cells (cTFH) and circulatory T follicular regulatory cells
513
(cTFR) 27,28.
514
Using the converse expression of CD40L and 4-1BB, CD40L+4-1BB and CD40L4-1BB+ CD4
515
T cells had a TCON and TREG phenotype, respectively, as shown by the expression of CD25 and
516
CD127 (Fig. S5a and b), and validate their use for the distinction of activated TCON and TREG
517
cells.
518
We detected a significant increase in the frequency of CD40L+4-1BB and CD40L4-1BB+
519
memory CD4 T cells at day 60 in our cohort (Fig. 4a) that correlated positively with the increase
520
in antibody titer between day 0 and day 365 (Fig. 4b and Fig. S6). Upon a closer look, the
521
induction of both signatures of vaccine-specific memory CD4 T cells was only true for early-
522
converters (Fig. 4c, see Fig. S7a for non-converters and Fig. S7b for vaccine-specific CD4 T
523
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
28
cells) while late-converters did not show a detectable memory CD4 T cell response. Although a
524
subset of both early and late-converters had detectable memory CD4 T cell responses prior to
525
vaccination, we detected no significant difference in the frequency of CD40L+4-1BB and
526
CD40L4-1BB+ memory CD4 T cells between the two groups at day 0 (Fig. 4d).
527
Collectively, flow cytometry data reveal that the expression of CD40L and 4-1BB in our ex vivo
528
assay is consistent with our serological data and reflects the lack of seroconversion at day 60 in
529
late-converters. However, it does not support the existence of more vaccine-specific memory
530
CD4 T cells in early-converters prior to vaccination.
531
532
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
29
533
Figure 4 Hepatitis B vaccine induces a vaccine-specific CD40L+4-1BB− and CD40L−4-1BB+
534
memory CD4 T cell response in early-converter vaccinees.
535
PBMCs from vaccinees were stimulated with 2 μg/ml of the master peptide pool (HBsAg) and
536
assessed for converse expression of 4-1BB and CD40L by flow cytometry on days 0, 60, 180,
537
and 365. Shown is number of of vaccine-specific memory CD4 T cells out of 1x106 memory
538
CD4 T cells after subtraction of responses in negative control.
539
a Aggregate analysis from vaccinees (including early, late and non-converters) showing a peak
540
of vaccine-specific CD40L+4-1BB and CD40L4-1BB+ memory CD4 T cell at day 60 (day 60
541
after 1st dose of the vaccine and day 30 after 2nd dose), declining thereafter. Shown are
542
proportions of vaccine-specific memory CD4 T cells out of total memory CD4 t cells.
543
b Correlation between the difference in antibody titer between day 365 and day 0 and vaccine-
544
specific CD40L+4-1BB and CD40L4-1BB+ memory CD4 T cell at day 60
545
c Aggregate analysis from early and late-converter vaccinees showing a significant induction of
546
vaccine-specific CD40L+4-1BB and CD40L4-1BB+ memory CD4 T cell in early-converters
547
and lack thereof in late-converters.
548
d Aggregate analysis from early and late-converter vaccinees showing no significant differences
549
in vaccine-specific CD40L+4-1BB and CD40L4-1BB+ memory CD4 T cell at day 0.
550
Wilcoxon signed-rank with unpaired and paired analysis as necessary; statistical significance
551
was indicated with ns P > 0.05, * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001
552
rs, Spearman correlation coefficient, −1 ≤ rs ≤ 1; rs and p value by Spearman’s correlation test
553
a b
cd
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
30
An expanded subset of 4-1BB+CD45RA TREG cells is a prominent feature of late-
554
converters
555
In order to detect any distinct signatures of early and late-converters, we analyzed pre-
556
vaccination flow cytometry data to examine major CD4 T cell subsets; TH, TREG, cTFH and cTFR
557
cells. Using manual gating in which regulatory T cells (TREG) were defined as viable
558
CD3+CD4+CD8CD25+CD127CXCR5 and were further divided into CD45RA+ and CD45RA
559
TREG cells, we identified a significantly higher frequency of 4-1BB+ CD45RA TREG cells in late-
560
converters compared to early-converters (Fig. 5a and Fig. S8).
561
TREG cells showed higher 4-1BB expression compared to TH, cTFH and cTFR cells (Fig. 5b) and
562
within TREG subset, CD45RA TREG cells showed significantly higher expression of 4-1BB,
563
accompanied with a higher expression of CD25, compared to CD45RA+ TREG cells (Fig. 5c). In
564
this scheme, CD45RA TREG can be divided into 4-1BB+CD25high and 4-1BB-CD25int subsets. It
565
is worth noting here that no differences were detected in the frequency of CD45RA or
566
CD45RA+ TREG cells within CD4 T cell compartment between the two groups (Fig. 5d), and that
567
the composition of TREG compartment that is distinct between the two groups (Fig. 5e).
568
In summary, an expanded subset of 4-1BB+CD45RA TREG cells pre-vaccination is a prominent
569
feature of a delayed and modest immune response to hepatitis B vaccine in our cohort.
570
571
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
31
572
Figure 5 An expanded 4-1BB+CD45RA TREG cells within TREG compartment is is a
573
prominent feature in late-converters prior to vaccination.
574
PBMCs from vaccinees at day 0 (prior to vaccination) were phenotyped for expression of
575
markers of TREG.
576
a Aggregate analysis of 4-1BB+CD45RA TREG within CD45RA TREG CD4 T cells in early and
577
late and non-converter vaccinees before vaccination
578
b Aggregate analysis of the median fluorescence intensity of 4-1BB in TH, cTFH, TREG and cTFR
579
cells before vaccination.
580
c Aggregate analysis of the median fluorescence intensity of 4-1BB (left panel) and CD25 (right
581
panel) in CD45RA TREG and CD45RA+ TREG cells before vaccination.
582
d Frequency of TREG, CD45RA TREG and CD45RA+ TREG cells within total CD4 T cells in early,
583
late and non-converter vaccinees before vaccination.
584
e Composition of TREG compartment as determined by expression of 4-1BB and CD45RA in
585
early, late and non-converter vaccinees before vaccination.
586
Wilcoxon signed-rank with unpaired and paired analysis as necessary; statistical significance
587
was indicated with ns P > 0.05, * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001
588
d
a b c
e
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
32
589
Discussion
590
In this study, we used high-throughput TCRβ repertoire profiling and ex vivo T cell assays to
591
characterize memory CD4 T cell repertoires before and after immunization with hepatitis B
592
vaccine, an adjuvanted subunit vaccine, and tracked vaccine-specific TCRβ clonotypes over two
593
time points. As antigen-naïve adults were found to have an unexpected abundance of memory-
594
phenotype CD4 T cells specific to viral antigens 7,29, we sought to investigate the influence that
595
preexisting memory CD4 T cells can have on vaccine-induced immunity.
596
Commercially available HBV vaccines produces a robust and long-lasting anti-HBs response,
597
and protection is provided by induction of an anti-HBs (antibody against HBV surface antigen)
598
titer higher than 10 mIU/mL after a complete immunization schedule of 3 doses 30. However, 5-
599
10% of healthy adult vaccinees fail to produce protective titers of anti-HBs and can be classified
600
as non-responders 30. In our cohort, 13 vaccinees did not seroconvert by day 60 (30 days
601
following administration of the second vaccine dose), as determined by antibody titer. Out of this
602
group, 9 vaccinees seroconverted by day 180 or day 365, referred to here as late-converters, and
603
4 vaccinees did not seroconvert, referred to here as non-converters.
604
A hallmark of adaptive immunity is a potential for memory immune responses to increase in
605
both magnitude and quality upon repeated exposure to the antigen 31. Our systems immunology
606
data supports the theory that preexisting memory CD4 T cell TCRβ sequences specific to
607
HBsAg, the antigenic component of the current hepatitis B vaccine, predict which individuals
608
will mount an early and more vigorous immune response to the vaccine as evidenced by a higher
609
fold change in anti-HBs antibody titer and a more significant induction of antigen-specific CD4
610
T cells. It is postulated that preexisting memory CD4 T cell clonotypes are generated due to the
611
highly degenerate nature of T cell recognition of antigen/MHC and are cross-reactive to
612
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
33
environmental antigens 8. For example, preexisting memory CD4 T cells are well-established in
613
unexposed HIV-seronegative individuals, although at a significantly lower magnitude than HIV-
614
exposed seronegative individuals 32,33, and were likely primed by exposure to environmental
615
triggers or the human microbiome.
616
617
We and others have shown before that the TCRβ repertoire of CD4 T cells encodes the antigen
618
exposure history of each individual and that antigen-specific TCRβ sequences could serve to
619
automatically annotate the infection or exposure history 14,34,35. In this study, we show that
620
similar principles can be used to study vaccine responsiveness. Specifically, the recruitment of
621
novel vaccine-specific T-cell clonotypes into memory compartment following vaccination can be
622
tracked by examining the CD4 memory TCRβ repertoire over time. While we observed no
623
increase in the frequency of the vaccine-specific memory T-cells, as the time point may have
624
missed the peak of the clonal expansion of effector CD4 T cells as was reported before 3638, a
625
significant rise in the number of unique vaccine-specific T-cell clonotypes was detected.
626
Nevertheless, this observation is consistent with earlier T cell immune repertoire sequencing
627
studies that showed that antigen-specific TCRβ sequences do not always overlap with those
628
TCRβ sequences that increase in frequency after infection or vaccination 39. More interestingly,
629
individuals with the earlier and more robust response against the vaccine, had a telltale antigen-
630
specific signature in their memory TCRβ repertoire prior to vaccination, despite the lack of
631
HBsAg antibodies or prior vaccination history.
632
633
Detection of this vaccine-specific signature was possible due to the development of a novel
634
predictive model that used epitope-specific TCRβ sequences from one set of individuals to make
635
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
34
predictions about another. A correction factor was needed to account for the occurrence of
636
bystander activated T-cells within the original epitope-specific TCRβ sequences. Indeed, in those
637
vaccinees without a positive antibody titer at day 60, any putative vaccine-specific T-cells may
638
be considered as bystander vaccination. This was supported by predictions using the TCRex tool
639
15, which matched these TCR sequences to common viral or other epitopes. It is of note that
640
these TCRβ sequences are matched with CD8 T cell epitopes, while they originate from isolated
641
CD4 T cells. This is likely due to the great similarity between the TCRβ sequences of CD4 and
642
CD8 T cells as noted in prior research 16.
643
644
However, our in vitro antigen-specificity data, using an assay that enables discrimination of
645
TCON and TREG cells using the converse expression of the activation markers CD40L and 4-1BB
646
23,40, failed to show a significant difference in preexisting antigen-specific CD4 T cells between
647
early and late-converters prior to vaccine administration. It is plausible that the signal is below
648
the detection limit of the assay and that more sensitive assays that require pre-enrichment of
649
CD40L+ and 4-1BB+ T cells (using magnetic beads) 41 or cultured ELISpot assay 42 are needed to
650
capture preexisting vaccine-specific memory CD4 T cells directly from human peripheral blood.
651
Another plausible explanation is that our activation proteins, CD40L and 4-1BB, might be
652
unsuitable to detect preexisting memory CD4 T cells but this is unlikely as both proteins have
653
been used successfully in similar studies 43,44. A different explanation may be that the diversity of
654
preexisting antigen-specific CD4 T clonotypes as determined by TCRβ sequencing is not
655
reflected in the quantitative measurements of the fractional cell counts.
656
657
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
35
TREG cells represent about 5 10% of human CD4 T cell compartment and are identified by the
658
constitutive surface expression of CD25, also known as IL-2 receptor α subunit (IL-2Ra), and the
659
nuclear expression of forkhead family transcription factor 3 (Foxp3), a lineage specification
660
factor of TREG cells 45. Regulatory memory T cells play a role in the mitigation of tissue damage
661
induced by effector memory T cells during protective immune responses, resulting in a selective
662
advantage against pathogen-induced immunopathology 4649. Several studies have identified CD4
663
TREG cells with specificity to pathogen-derived peptides in murine models and showed evidence
664
for an induced expansion of TREG cells followed by an emergence and a long-term persistence of
665
TREG cells with a memory phenotype and potent immunosuppressive properties 47,50. Blom et al.
666
reported a significant and transient activation of TREG cells (identified by upregulation of CD38
667
and Ki67) in humans 10 days after administration of live attenuated yellow fever virus 17D
668
vaccine 37. The induction of vaccine-specific TREG cells in our cohort is unexpected and the role
669
it might play in vaccine-induced immunity warrants further investigation.
670
671
The association of an expanded 4-1BB+ CD45RA TREG subset with a delayed immune response
672
to hepatitis B vaccine was not described before. Miyara et al. showed that blood contains two
673
distinct subsets of stable and suppressive TREG cells: resting TREG, identified as
674
FOXP3lowCD45RA+ CD4 T cells, and activated TREG, identified as FOXP3highCD45RA CD4 T
675
cells. They further noted that activated TREG cells constitute a minority subset within cord blood
676
TREG cells and increase gradually with age 51. As activated TREG cells were shown to have an
677
increased expression of proteins indicative of activation, including ICOS and HLA-DR 5155, it
678
might be the case that an upregulation of 4-1BB is one more feature of this population or a subset
679
thereof. Moreover, TREG cells in mice were shown to modulate TFH formation and GC B cell
680
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
36
responses and to diminish antibody production in a CTLA-4 mediated suppression 56.
681
Interestingly, CD45RA TREG cells were shown to be more rich in preformed CTLA-4 stored in
682
intracellular vesicles compared to CD45RA+ TREG cells 51.
683
4-1BB was shown to be constitutively expressed by TREG cells 57 and that 4-1BB+ TREG cells are
684
functionally superior to 4-1BB TREG cells in both contact-dependent and contact-independent
685
immunosuppression 58. 4-1BB+ TREG cells are the major producers of the alternatively-spliced
686
and soluble isoform of 4-1BB among T cells 58. 4-1BB was shown before to be preferentially
687
expressed on TREG cells compared with other non-regulatory CD4 T cell subsets 57 and that 4-
688
1BB-costimualtion induces the expansion of TREG cells both in vitro and in vivo 59. Moreover,
689
agonistic anti-4-1BB mAbs have been shown to abrogate T cell-dependent antibody responses in
690
vivo 60 and to ameliorate experimental autoimmune encephalomyelitis by skewing the balance
691
against TH17 differentiation in favor of TREG differentiation 61. It is plausible that the expansion
692
4-1BB+CD45RA TREG cells in late-converters is involved in the suppression of GC vaccine-
693
specific TFH cells and the ensuing antibody response in our cohort, but this remains speculative
694
and further research is warranted.
695
696
It is enticing to speculate that the preexisting memory CD4 T cells result from the complex
697
interplay between cellular immunity and the human microbiome. A role for the microbiota in
698
modulating immunity to viral infection was suggested in 1960’s 62, and since then we gained
699
better understanding of the impact of the various components of the microbiota including
700
bacteria, fungi, protozoa, archaea and viruses on the murine and human immune systems 63.
701
Viral clearance of hepatitis B virus infection depends on the age of exposure and neonates and
702
young children are less likely to spontaneously clear the virus 64. Han-Hsuan et al. have shown
703
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
37
evidence in mice that this age-dependency is mediated by gut microbiota that prepare the liver
704
immunity system to clear HBV, possibly via a TLR4 signaling pathway 65. In this study, young
705
mice that have not reached an equilibrium in the gut microbiota, exhibited prolonged HBsAg
706
persistence, impaired anti-HBs antibody production, and limited Hepatitis B core antigen
707
(HBcAg)-specific IFNγ+ splenocytes. More recently, Tingxin et al. provided evidence for a
708
critical role of the commensal microbiota in supporting the differentiation of GC B cells, through
709
follicular T helper (TFH) cells, to promote the anti-HBV humoral immunity 66.
710
711
Our study bears some intrinsic limitations. A major drawback is the restricted number of days at
712
which TCRβ repertoire was profiled, as vaccine-specific perturbations within the repertoire may
713
occur at different time points for early, late and non-converters. Additionally, more in-depth
714
characterization and functional studies on 4-1BB+CD45RA TREG cells could have helped shed
715
more light on the role they play in vaccine-induced immunity. Future studies in larger cohorts
716
and with a more comprehensive TCRβ repertoire profiling and CD4 T cells immunophenotyping
717
are required to validate our findings.
718
719
In conclusion, our analysis of the memory CD4 T cell repertoire has uncovered a role for
720
preexisting memory CD4 T cells in naïve individuals in mounting an earlier and more vigorous
721
immune response to hepatitis B vaccine and argue for the utility of pre-vaccination TCRβ
722
repertoire in the prediction of vaccine-induced immunity. Moreover, we identify a subset of 4-
723
1BB+ memory TREG cells that is expanded in individuals with delayed immune response to the
724
vaccine, which might further explain the heterogeneity of response to hepatitis B vaccine.
725
726
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
38
727
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
39
AUTHOR CONTRIBUTIONS
728
Conception: PM, BO
729
Design: GE, PM, EB, AS, GM, PVD, PB, KL, VVT, BO
730
Experiments: GE, PM, EB, NDN, HJ, AS
731
Data-analysis: GE, PM, NDN, BO
732
Supervision: HDR, EL, PT, GM, PVD, PB, KL, VVT, BO
733
First draft: GE, PM, BO
734
Contributed to the paper: all authors
735
736
COMPETING FINANCIAL INTERESTS
737
Parts of the contents of this manuscript form the topic of patent EPO 19159931.5.
738
VVT is an employee of Johnson & Johnson since 1/11/2019 and remains currently employed at
739
the University of Antwerp.
740
741
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
40
References
742
1. Rudolph, M. G., Stanfield, R. L. & Wilson, I. A. HOW TCRS BIND MHCS, PEPTIDES, AND CORECEPTORS. Annu. Rev.
743
Immunol. 24, 419466 (2006).
744
2. Esensten, J. H., Helou, Y. A., Chopra, G., Weiss, A. & Bluestone, J. A. CD28 Costimulation: From Mechanism to Therapy. Immunity
745
44, 973988 (2016).
746
3. Curtsinger, J. M. & Mescher, M. F. Inflammatory cytokines as a third signal for T cell activation. Curr. Opin. Immunol. 22, 33340
747
(2010).
748
4. Turner, S. J., La Gruta, N. L., Kedzierska, K., Thomas, P. G. & Doherty, P. C. Functional implications of T cell receptor diversity.
749
Current Opinion in Immunology 21, 286290 (2009).
750
5. Krangel, M. S. Mechanics of T cell receptor gene rearrangement. Current Opinion in Immunology 21, 133139 (2009).
751
6. Farber, D. L., Yudanin, N. A. & Restifo, N. P. Human memory T cells: Generation, compartmentalization and homeostasis. Nature
752
Reviews Immunology 14, 2435 (2014).
753
7. Su, L. F., Kidd, B. A., Han, A., Kotzin, J. J. & Davis, M. M. Virus-Specific CD4+ Memory-Phenotype T Cells Are Abundant in
754
Unexposed Adults. Immunity 38, 373383 (2013).
755
8. Sewell, A. K. Why must T cells be cross-reactive? Nature Reviews Immunology 12, 669677 (2012).
756
9. Wilson, D. B. et al. Specificity and degeneracy of T cells. Mol. Immunol. 40, 10471055 (2004).
757
10. Reese, T. A. et al. Sequential Infection with Common Pathogens Promotes Human-like Immune Gene Expression and Altered Vaccine
758
Response. Cell Host Microbe 19, 713719 (2016).
759
11. Keating, G. M. & Noble, S. Recombinant Hepatitis B Vaccine (Engerix-B). Drugs 63, 10211051 (2003).
760
12. Shouval, D. Hepatitis B vaccines. J. Hepatol. 39, 7076 (2003).
761
13. Precopio, M. L. et al. Optimizing peptide matrices for identifying T-cell antigens. Cytom. Part A 73, 10711078 (2008).
762
14. de Neuter, N. et al. Memory CD4+T cell receptor repertoire data mining as a tool for identifying cytomegalovirus serostatus. Genes
763
Immun. (2018). doi:10.1038/s41435-018-0035-y
764
15. Gielis, S. et al. Detection of Enriched T Cell Epitope Specificity in Full T Cell Receptor Sequence Repertoires. Front. Immunol. 10,
765
(2019).
766
16. Meysman, P. et al. On the viability of unsupervised T-cell receptor sequence clustering for epitope preference. Bioinformatics 35,
767
14611468 (2018).
768
17. Spidlen, J., Breuer, K., Rosenberg, C., Kotecha, N. & Brinkman, R. R. FlowRepository: A resource of annotated flow cytometry
769
datasets associated with peer-reviewed publications. Cytom. Part A 81 A, 727731 (2012).
770
18. Furman, D. et al. Cytomegalovirus infection enhances the immune response to influenza. Sci. Transl. Med. 7, 281ra43 (2015).
771
19. Klarenbeek, P. L. et al. Human T-cell memory consists mainly of unexpanded clones. Immunol. Lett. 133, 4248 (2010).
772
20. Becattini, S. et al. Functional heterogeneity of human memory CD4+ T cell clones primed by pathogens or vaccines. Science (80-. ).
773
347, 400406 (2015).
774
21. De Neuter, N. et al. On the feasibility of mining CD8+ T cell receptor patterns underlying immunogenic peptide recognition.
775
Immunogenetics 70, 159168 (2018).
776
22. Brezar, V. et al. T-regulatory cells and vaccination “pay attention and do not neglect them”: Lessons from HIV and cancer vaccine
777
trials. Vaccines 4, (2016).
778
23. Schoenbrunn, A. et al. A converse 4-1BB and CD40 ligand expression pattern delineates activated regulatory T cells (Treg) and
779
conventional T cells enabling direct isolation of alloantigen-reactive natural Foxp3+ Treg. J. Immunol. 189, 598594 (2012).
780
24. Elias, G., Ogunjimi, B. & Van Tendeloo, V. Activation-induced surface proteins in the identification of antigen-responsive CD4 T
781
cells. Immunology Letters 219, 17 (2020).
782
25. Liu, W. et al. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J. Exp. Med.
783
203, 17011711 (2006).
784
26. Seddiki, N. et al. Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J.
785
Exp. Med. 203, 16931700 (2006).
786
27. Bentebibel, S.-E., Schmitt, N., Banchereau, J. & Ueno, H. Human tonsil B-cell lymphoma 6 (BCL6)-expressing CD4+ T-cell subset
787
specialized for B-cell help outside germinal centers. Proc. Natl. Acad. Sci. 108, E488E497 (2011).
788
28. Fonseca, V. R. et al. Human blood Tfr cells are indicators of ongoing humoral activity not fully licensed with suppressive function. Sci.
789
Immunol. 2, (2017).
790
29. Su, L. F. & Davis, M. M. Antiviral memory phenotype T cells in unexposed adults. Immunol. Rev. 255, 95109 (2013).
791
30. Meireles, L. C., Marinho, R. T. & Van Damme, P. Three decades of hepatitis B control with vaccination. World J. Hepatol. 7, 212732
792
(2015).
793
31. Sallusto, F., Lanzavecchia, A., Araki, K. & Ahmed, R. From vaccines to memory and back. Immunity 33, 451463 (2010).
794
32. Ritchie, A. J. et al. Differences in HIV-Specific T Cell Responses between HIV-Exposed and -Unexposed HIV-Seronegative
795
Individuals. J. Virol. 85, 35073516 (2011).
796
33. Campion, S. L. et al. Proteome-wide analysis of HIV-specific naive and memory CD4+ T cells in unexposed blood donors. J. Exp.
797
Med. 211, 12731280 (2014).
798
34. Emerson, R. O. et al. Immunosequencing identifies signatures of cytomegalovirus exposure history and HLA-mediated effects on the T
799
cell repertoire. Nat. Genet. 49, 659665 (2017).
800
35. DeWitt, W. S. et al. Human T cell receptor occurrence patterns encode immune history, genetic background, and receptor specificity.
801
Elife 7, (2018).
802
36. Kohler, S. et al. The early cellular signatures of protective immunity induced by live viral vaccination. Eur. J. Immunol. 42, 23632373
803
(2012).
804
37. Blom, K. et al. Temporal Dynamics of the Primary Human T Cell Response to Yellow Fever Virus 17D As It Matures from an
805
Effector- to a Memory-Type Response. J. Immunol. 190, 21502158 (2013).
806
38. Pogorelyy, M. V. et al. Precise tracking of vaccine-responding T cell clones reveals convergent and personalized response in identical
807
twins. Proc. Natl. Acad. Sci. U. S. A. 115, 1270412709 (2018).
808
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
41
39. DeWitt, W. S. et al. Dynamics of the Cytotoxic T Cell Response to a Model of Acute Viral Infection. J. Virol. 89, 45174526 (2015).
809
40. Frentsch, M. et al. Direct access to CD4+ T cells specific for defined antigens according to CD154 expression. Nat. Med. 11, 1118
810
1124 (2005).
811
41. Bacher, P. et al. Antigen-reactive T cell enrichment for direct, high-resolution analysis of the human naive and memory Th cell
812
repertoire. J. Immunol. 190, 396776 (2013).
813
42. Reece, W. H. H. et al. A CD4+ T-cell immune response to a conserved epitope in the circumsporozoite protein correlates with
814
protection from natural Plasmodium falciparum infection and disease. Nat. Med. 10, 406410 (2004).
815
43. Bacher, P. et al. Identification of Immunogenic Antigens from Aspergillus fumigatus by Direct Multiparameter Characterization of
816
Specific Conventional and Regulatory CD4+ T Cells. J. Immunol. 193, 33323343 (2014).
817
44. Bacher, P. et al. Antigen-specific expansion of human regulatory T cells as a major tolerance mechanism against mucosal fungi.
818
Mucosal Immunol. 7, 916928 (2014).
819
45. Rudensky, A. Y. Regulatory T cells and Foxp3. Immunol. Rev. 241, 260268 (2011).
820
46. Lovelace, P. & Maecker, H. T. Multiparameter Intracellular Cytokine Staining. in Methods in molecular biology (Clifton, N.J.) 1678,
821
151166 (2018).
822
47. Lin, P. H. et al. Vaccine-induced antigen-specific regulatory T cells attenuate the antiviral immunity against acute influenza virus
823
infection. Mucosal Immunol. 11, 12391253 (2018).
824
48. Garner-Spitzer, E. et al. Tick-Borne Encephalitis (TBE) and Hepatitis B Nonresponders Feature Different Immunologic Mechanisms in
825
Response to TBE and Influenza Vaccination with Involvement of Regulatory T and B Cells and IL-10. J. Immunol. 191, 24262436
826
(2013).
827
49. Lanteri, M. C. et al. Tregs control the development of symptomatic West Nile virus infection in humans and mice. J. Clin. Invest. 119,
828
32663277 (2009).
829
50. Sanchez, A. M., Zhu, J., Huang, X. & Yang, Y. The Development and Function of Memory Regulatory T Cells after Acute Viral
830
Infections. J. Immunol. 189, 28052814 (2012).
831
51. Miyara, M. et al. Functional Delineation and Differentiation Dynamics of Human CD4+ T Cells Expressing the FoxP3 Transcription
832
Factor. Immunity 30, 899911 (2009).
833
52. Booth, N. J. et al. Different Proliferative Potential and Migratory Characteristics of Human CD4 + Regulatory T Cells That Express
834
either CD45RA or CD45RO. J. Immunol. 184, 43174326 (2010).
835
53. Ito, T. et al. Two Functional Subsets of FOXP3+ Regulatory T Cells in Human Thymus and Periphery. Immunity 28, 870880 (2008).
836
54. Mason, G. M. et al. Phenotypic Complexity of the Human Regulatory T Cell Compartment Revealed by Mass Cytometry. J. Immunol.
837
195, 20302037 (2015).
838
55. Mohr, A., Malhotra, R., Mayer, G., Gorochov, G. & Miyara, M. Human FOXP3+ T regulatory cell heterogeneity. Clinical and
839
Translational Immunology 7, (2018).
840
56. Wing, J. B., Ise, W., Kurosaki, T. & Sakaguchi, S. Regulatory T cells control antigen-specific expansion of Tfh cell number and
841
humoral immune responses via the coreceptor CTLA-4. Immunity 41, 10131025 (2014).
842
57. McHugh, R. S. et al. CD4(+)CD25(+) immunoregulatory T cells: gene expression analysis reveals a functional role for the
843
glucocorticoid-induced TNF receptor. Immunity 16, 31123 (2002).
844
58. Kachapati, K. et al. The B10 Idd9.3 locus mediates accumulation of functionally superior CD137(+) regulatory T cells in the nonobese
845
diabetic type 1 diabetes model. J. Immunol. 189, 500115 (2012).
846
59. Zheng, G., Wang, B. & Chen, A. The 4-1BB costimulation augments the proliferation of CD4+CD25+ regulatory T cells. J. Immunol.
847
173, 242834 (2004).
848
60. Mittler, R. S., Bailey, T. S., Klussman, K., Trailsmith, M. D. & Hoffmann, M. K. Anti-4-1BB monoclonal antibodies abrogate T cell-
849
dependent humoral immune responses in vivo through the induction of helper T cell anergy. J. Exp. Med. 190, 153540 (1999).
850
61. Kim, Y. H. et al. 4-1BB Triggering Ameliorates Experimental Autoimmune Encephalomyelitis by Modulating the Balance between
851
Th17 and Regulatory T Cells. J. Immunol. 187, 11201128 (2011).
852
62. Robinson, C. M. & Pfeiffer, J. K. Viruses and the Microbiota. Annu. Rev. Virol. 1, 5569 (2014).
853
63. Winkler, E. S. & Thackray, L. B. A long-distance relationship: the commensal gut microbiota and systemic viruses. Current Opinion in
854
Virology 37, 4451 (2019).
855
64. Yuen, M.-F. et al. Hepatitis B virus infection. Nat. Rev. Dis. Prim. 4, 18035 (2018).
856
65. Chou, H. H. et al. Age-related immune clearance of hepatitis B virus infection requires the establishment of gut microbiota. Proc. Natl.
857
Acad. Sci. U. S. A. 112, 21752180 (2015).
858
66. Wu, T. et al. CD4+ T cells play a critical role in microbiota-maintained Anti-HBV immunity in a mouse model. Front. Immunol. 10,
859
(2019).
860
861
862
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
42
Supplemental tables
863
Supplementary Table 1. List of 54 single peptides, each 15 AA long with an 11-amino acid
864
overlap spanning the 226 amino acids along the small S protein of hepatitis B (HB) surface
865
antigen (HBsAg)
866
867
Peptide
No.
15 AA with 11 AA overlap
No. of AA at which the peptide start
1
MENITSGFLGPLLVL
1
2
TSGFLGPLLVLQAGF
5
3
LGPLLVLQAGFFLLT
9
4
LVLQAGFFLLTRILT
13
5
AGFFLLTRILTIPQS
17
6
LLTRILTIPQSLDSW
21
7
ILTIPQSLDSWWTSL
25
8
PQSLDSWWTSLNFLG
29
9
DSWWTSLNFLGGSPV
33
10
TSLNFLGGSPVCLGQ
37
11
FLGGSPVCLGQNSQS
41
12
SPVCLGQNSQSPTSN
45
13
LGQNSQSPTSNHSPT
49
14
SQSPTSNHSPTSCPP
53
15
TSNHSPTSCPPICPG
57
16
SPTSCPPICPGYRWM
61
17
CPPICPGYRWMCLRR
65
18
CPGYRWMCLRRFIIF
69
19
RWMCLRRFIIFLFIL
73
20
LRRFIIFLFILLLCL
77
21
IIFLFILLLCLIFLL
81
22
FILLLCLIFLLVLLD
85
23
LCLIFLLVLLDYQGM
89
24
FLLVLLDYQGMLPVC
93
25
LLDYQGMLPVCPLIP
97
26
QGMLPVCPLIPGSTT
101
27
PVCPLIPGSTTTNTG
105
28
LIPGSTTTNTGPCKT
109
29
STTTNTGPCKTCTTP
113
30
NTGPCKTCTTPAQGN
117
31
CKTCTTPAQGNSMFP
121
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
43
32
TTPAQGNSMFPSCCC
125
33
QGNSMFPSCCCTKPT
129
34
MFPSCCCTKPTDGNC
133
35
CCCTKPTDGNCTCIP
137
36
KPTDGNCTCIPIPSS
141
37
GNCTCIPIPSSWAFA
145
38
CIPIPSSWAFAKYLW
149
39
PSSWAFAKYLWEWAS
153
40
AFAKYLWEWASVRFS
157
41
YLWEWASVRFSWLSL
161
42
WASVRFSWLSLLVPF
165
43
RFSWLSLLVPFVQWF
169
44
LSLLVPFVQWFVGLS
173
45
VPFVQWFVGLSPTVW
177
46
QWFVGLSPTVWLSAI
181
47
GLSPTVWLSAIWMMW
185
48
TVWLSAIWMMWYWGP
189
49
SAIWMMWYWGPSLYS
193
50
MMWYWGPSLYSIVSP
197
51
WGPSLYSIVSPFIPL
201
52
LYSIVSPFIPLLPIF
205
53
VSPFIPLLPIFFCLW
209
54
IPLLPIFFCLWVYI
213
868
869
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
44
Supplementary Table 2. Overview of the single peptides tested for each vaccinee in the
870
CFSE assay
871
Vaccinee
Gender
Age
Status
peptides_number
single peptides
2
F
43.6
Late-converter
6
51, 35, 50, 34, 54, 38
3
F
48.5
Late-converter
6
53, 5, 37, 49, 1, 33
4
M
47.1
Late-converter
6
21, 20, 5, 53, 4, 52
6
F
44.3
Late-converter
6
23, 39, 7, 21, 37, 5
7
M
38.3
Early-converter
2
17, 33
8
M
43.4
Early-converter
3
47, 41, 42
10
F
33.2
Early-converter
5
49, 1, 33, 41, 9
11
F
39
Early-converter
6
53, 50, 5, 2, 8, 21
13
F
45.9
Early-converter
3
24, 8, 16
14
M
36.3
Late-converter
2
45, 47
17
F
26.5
Early-converter
6
10, 42, 9, 41, 14, 46
18
M
43.3
Early-converter
4
6, 38, 5, 37
19
M
41.3
Early-converter
3
40, 16, 8
20
F
48.7
Early-converter
2
6, 1
21
F
27.2
Non-converter
6
2, 1, 3, 42, 41, 43
22
F
22.3
Early-converter
4
10, 13, 12, 16
23
F
43.9
Late-converter
6
51, 54, 48, 43, 47, 46
24
F
47.7
Late-converter
6
22, 24, 23, 20, 18, 46
25
F
22.6
Non-converter
6
29, 30, 28, 21, 22, 20
26
F
41.2
Late-converter
6
23, 47, 17, 41, 7, 49
28
F
41.7
Early-converter
6
54, 38, 52, 36, 40, 39
29
M
39.5
Early-converter
6
7, 47, 6, 1, 23, 46
30
F
45.1
Late-converter
6
22, 46, 54, 19, 20, 43
31
M
32.4
Early-converter
6
54, 38, 49, 33, 53, 37
32
F
31.4
Early-converter
6
7, 4, 1, 6, 31, 28
33
F
41
Early-converter
6
1, 41, 7, 5, 47, 45
34
M
22.5
Early-converter
5
7, 4, 5, 2, 6
35
F
50.2
Early-converter
6
46, 45, 47, 22, 21, 23
36
F
23.2
Early-converter
6
54, 47, 46, 39, 48, 38
38
F
23.2
Early-converter
6
37, 34, 33, 35, 5, 2
39
M
45.8
Early-converter
6
51, 50, 53, 49, 35, 34
40
F
21.3
Early-converter
5
33, 38, 37, 39, 40
41
M
21.6
Non-converter
6
23, 22, 7, 6, 17, 1
42
M
22.7
Non-converter
4
1, 33, 25, 41
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
45
872
Figure S1. Gating strategy of ex vivo T cell phenotyping of vaccine-specific T cells.
873
a Gating strategy started by a lymphocyte gate, followed by gating on viable CD3+CD8 T cells.
874
Doublets were excluded using
875
doublet discrimination (area against the height of forward scatter pulse) before gating on CD4+ T
876
cells. Next, CD45RA, CXCR5, CD25 and CD127 were used to identify main subsets of CD4 T
877
cells using Boolean gates as specified in the accompanying table.
878
b Shown an example of gating for CD154 (CD40L) and CD137 (4-1BB) for cells left
879
unstimulated (left) and cells stimulated with a master peptide pool (right) for an early-converter
880
vaccinee at day 60.
881
882
SSC-A
FSC-A
CD3-Alexa Fluor 430
CD8-Viability-DUMP
CD4-PerCP-Cy5.5
FSC-H
FSC-A
FSC-H
CD137pos
0.049
CD154pos
0.050
CD154-APC
CD137-PE
CD137pos
0.13
CD154pos
0.21
CD154-APC
CD137-PE
CD25-BV421
CD127-BV785
CD137neg_CD154pos
CD137pos_CD154neg
CD45RA-AF488
CXCR5-PE-Cy7
CD25-BV421
CD127-BV785
CD45RACXCR5 CD25 CD127
Naive T
H
+
low + high
Memory T
H
low + high
Naive T
FH
+ +
low + high
Memory T
FH
+
low + high
CD45RA
+
T
REG
++
low
CD45RA
T
REG
+
low
CD45RA
+
T
FR
+ + +
low
CD45RA
T
FR
+ +
low
a
b
FSC-A
FSC-H
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
46
883
Figure S2.
884
a Anti-Hepatitis B surface (anti-HBs) titer of vaccinees over four times points, facetted by
885
groups of early, late and non-converters. An anti-HBs titer above 10 IU/ml was considered
886
protective. Early-converters seroconverted at day 60, late-converters seroconverted at day 180 or
887
day 365 and nonconverters did not have an anti-HBs titer higher than 10 IU/ml at any of the
888
time points. b CMV, EBV and HSV seropositivity in the three groups of the cohort as
889
determined by serum IgG antibodies to CMV, EBV-VCA, and HSV-1 and 2 using sandwich
890
ELISA. c Age of vaccinees per group.
891
Wilcoxon signed-rank with unpaired and paired analysis as necessary; statistical significance
892
was indicated with ns P > 0.05, * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001
893
894
a
b c
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
47
895
Figure S3
896
c
b
ns
0.000
0.001
0.002
0.003
0.004
0.005
Day0 Day60
HBVspecific Tcell frequency
a
d
ef
CD40L
+
4-1BB
-
memory CD4 T cell
g
CD40L
-
4-1BB
+
memory CD4 T cell
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
48
a Scatter plot of the DNA-based TCRβ reads for each vaccinee at each time point
897
b Scatter plot of number of unique TCRβ amino acid sequences for each vaccinee at each time
898
point, where the shape denotes the response as based on antibody titer.
899
c Overview of unique TCRβ amino acid sequences in the memory CD4 T cell repertoire of each
900
vaccinee. The bottom blue bar denotes those TCR sequences that were found at both time points.
901
The green and red bars denote the number of unique TCR sequences at each time point. The total
902
bar height thus represents the total number of unique memory CD4 T cell clonotypes sequences
903
for a specific vaccinee.
904
d Change in frequency of those HBV-specific T-cells present at both time points. The (ns) mark
905
denotes a non-significant paired Wilcoxon signed-rank test (P value = 0.7577).
906
e Antibody-defined categories (EC, LC, NC)) plotted against the values from the CD40L assay
907
f ROC curve for the Rhbs predictive model from day 0 data in a leave-one-out cross-validation
908
compared to CD40L+4-1BB- (TCON) memory CD4 T cell values for each vaccinee at time
909
points 60 (AUC = 0.84), 180 (AUC = 0.56) and 365 (AUC = 0.57).
910
g ROC curve for the Rhbs predictive model from day 0 data in a leave-one-out cross-validation
911
compared to CD40L-4-1BB+ (TREG) memory CD4 T cell values for each vaccinee at time
912
points 60 (AUC = 0.62), 180 (AUC = 0.56) and 365 (AUC = 0.52).
913
914
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
49
915
Figure S4. Overview of the in vitro expansion experiments for all vaccinees, vaccinees per
916
group and for each vaccinee.
917
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
50
918
Figure S5. CD40L+4-1BB and CD40L4-1BB+ CD4 T cells have a TCON and TREG
919
phenotype, respectively.
920
921
CD137pos
0.049
CD154pos
0.050
CD154-APC
CD137-PE
CD137pos
0.13
CD154pos
0.21
CD154-APC
CD137-PE
CD25-BV421
CD127-BV785
CD137neg_CD154pos
CD137pos_CD154neg
CD45RA-AF488
CXCR5-PE-Cy7
CD25-BV421
CD127-BV785
CD40L
+
4-1BB
CD4 T cells
CD40L
4-1BB
+
CD4 T cells
a b
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
51
922
Figure S6
923
Correlation between the difference in antibody titer between day 365 and day 0 and vaccine-
924
specific CD40L+4-1BB and CD40L4-1BB+ memory CD4 T cell at day 60 colored by vaccinee
925
group and labeled with vaccinee ID.
926
rs, Spearman correlation coefficient, −1 ≤ rs ≤ 1; rs and p value by Spearman’s correlation test
927
928
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
52
929
Figure S7 Hepatitis B vaccine induces a vaccine-specific CD4 T cell response in early-
930
converter vaccinees.
931
PBMCs from vaccinees were stimulated with 2 μg/ml of a pool of peptides of HBsAg and
932
assessed for converse expression of 4-1BB and CD40L by flow cytometry on days 0, 60, 180,
933
and 365.
934
a Aggregate analysis from early, late and non-converter vaccinees showing a significant
935
induction of vaccine-specific CD40L+4-1BB and CD40L4-1BB+ memory CD4 T cell in early-
936
converters and lack thereof in late and non-converters. Shown is number of of vaccine-specific
937
memory CD4 T cells out of 1x106 memory CD4 T cells after subtraction of responses in negative
938
control (see Methods for details).
939
b Aggregate analysis from early, late and non-converter vaccinees showing a significant
940
induction of vaccine-specific CD40L+4-1BB and CD40L4-1BB+ CD4 T cell in early-
941
converters and lack thereof in late and non-converters. Shown is number of of vaccine-specific
942
CD4 T cells out of 1x106 CD4 T cells after subtraction of responses in negative control (see
943
Methods for details).
944
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
53
945
Figure S8
946
Aggregate analysis of 4-1BB+CD45RA TREG within CD45RA TREG CD4 T cells in early, late
947
and non-converter vaccinees at days 0, 60, 180 and 365.
948
Wilcoxon signed-rank with unpaired and paired analysis as necessary; statistical significance
949
was indicated with ns P > 0.05, * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001
950
951
.CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprintthis version posted August 25, 2020. . https://doi.org/10.1101/2020.08.22.262568doi: bioRxiv preprint
... Pre-existing memory B cells also affect the outcome of a quadrivalent influenza vaccine (QIV); when pre-existing B cell memory exhibits a dominant response to a particular subtype (subtype immunodominance), Ab response to QIV was positively correlated with the preexisting memory (93). Also, it was recently reported that the kinetics and magnitude of Ab response to a hepatitis B vaccine were significantly increased in the presence of hepatitis vaccine-specific memory CD4 + T cells (94). Ab response to HCoV can be observed in most adults (95) and the S2 domain of SARS-CoV-2 S exhibits relatively high amino acid sequence homology with those of seasonal HCoVs (up to 42%) (90). ...
Article
Full-text available
The global outbreak of coronavirus disease 2019 (COVID-19) is still threatening human health, economy, and social life worldwide. As a counteraction for this devastating disease, a number of vaccines are being developed with unprecedented speed combined with new technologies. As COVID-19 vaccines are being developed in the absence of a licensed human coronavirus vaccine, there remain further questions regarding the long-term efficacy and safety of the vaccines, as well as immunological mechanisms in depth. This review article discusses the current status of COVID-19 vaccine development, mainly focusing on antigen design, clinical trials in later stages, and immunological considerations for further study.
Article
T cells exercise a multitude of functions such as cytotoxicity, secretion of immunomodulating cytokines or regulation of tolerance, collectively resulting in an effective control of immune-related disease. Through the unique mechanism of V(D)J recombination, T cells express a highly specific receptor complex known as the T-cell receptor (TCR). Single-cell sequencing technologies have paved the road for interrogating the transcriptome and the paired αβ TCR repertoire of a single T cell in tandem. In contrast, conventional bulk methods are restricted to only one layer of information. This combination of transcriptomic- and repertoire information can provide novel insight into the functional character of T cell immunity. Recently, single-cell technologies have gained in popularity due to improvements in throughput, decrease in cost and the ability for multi-modal experiments that integrate different information layers. Consequently, this prompts the need for the development of novel computational tools that integrate transcriptomic profiles and corresponding features of the TCR repertoire. Here we discuss the current progress in the field of single-cell T cell sequencing, with a focus on the multimodality of new approaches that allow the paired profiling of cellular phenotype and clonotype information. In addition, this review provides detailed descriptions of recent computational developments for analyzing single-cell TCR sequencing data in an integrative manner using novel computational approaches. Finally, we present an overview of the available software tools that can be used to perform integrative analysis of gene expression and TCR profiles.
Article
Full-text available
Naive and memory CD4+ T cells reactive with human immunodeficiency virus type 1 (HIV-1) are detectable in unexposed, unimmunized individuals. The contribution of preexisting CD4+ T cells to a primary immune response was investigated in 20 HIV-1-seronegative volunteers vaccinated with an HIV-1 envelope (Env) plasmid DNA prime and recombinant modified vaccinia virus Ankara (MVA) boost in the HVTN 106 vaccine trial (clinicaltrials.gov NCT02296541). Prevaccination naive or memory CD4+ T cell responses directed against peptide epitopes in Env were identified in 14 individuals. After priming with DNA, 40% (8/20) of the elicited responses matched epitopes detected in the corresponding preimmunization memory repertoires, and clonotypes were shared before and after vaccination in 2 representative volunteers. In contrast, there were no shared epitope specificities between the preimmunization memory compartment and responses detected after boosting with recombinant MVA expressing a heterologous Env. Preexisting memory CD4+ T cells therefore shape the early immune response to vaccination with a previously unencountered HIV-1 antigen.
Article
Full-text available
High-throughput T cell receptor (TCR) sequencing allows the characterization of an individual's TCR repertoire and directly queries their immune state. However, it remains a non-trivial task to couple these sequenced TCRs to their antigenic targets. In this paper, we present a novel strategy to annotate full TCR sequence repertoires with their epitope specificities. The strategy is based on a machine learning algorithm to learn the TCR patterns common to the recognition of a specific epitope. These results are then combined with a statistical analysis to evaluate the occurrence of specific epitope-reactive TCR sequences per epitope in repertoire data. In this manner, we can directly study the capacity of full TCR repertoires to target specific epitopes of the relevant vaccines or pathogens. We demonstrate the usability of this approach on three independent datasets related to vaccine monitoring and infectious disease diagnostics by independently identifying the epitopes that are targeted by the TCR repertoire. The developed method is freely available as a web tool for academic use at tcrex.biodatamining.be.
Article
Full-text available
The ability of the host to clear hepatitis B virus (HBV) is closely correlated to the establishment of commensal microbiota. However, how microbiota affects anti-HBV immunity is still unclear. Using a well-known hydrodynamical HBV transfection mouse model and treatment with antibiotics (Atb), we explored the change in adaptive immunity (CD4⁺ cells, germinal center B cells and anti-HBs Ab). In our setting, normal mice exhibited complete clearance of HBV within 6 weeks post-hydrodynamic injection (HDI) of HBV-containing plasmid, whereas Atb-treated mice lost this capacity, showing high serum level of hepatitis B surface antigen (HBsAg) without hepatitis B surface antibodies (anti-HBs), similar as what happened in Rag1−/− mice or CD4−/− mice, suggesting that microbiota may influence the function of CD4⁺ T cells. Furthermore, the numbers of splenic and hepatic effector CD4⁺ T cells (CD44hiCD62L⁻CD4⁺ T cells) both decreased with impaired function (IFN-γ synthesis), resulting in lower frequency of germinal center B cells and CD4⁺ follicular helper T cells, and impaired anti-HBs production. We further tried to find the bacterial species responsible for maintaining anti-HBV immunity, and found that each antibiotic alone could not significantly influence HBV clearance compared to antibiotic combination, suggesting that global commensal microbial load is critical for promoting HBV clearance. We also confirmed that TLRs (e.g., TLR2, 4, 9) are not major players in immune clearance of HBV using their agonists and knock-out mice. These results suggest that commensal microbiota play an important role in maintaining CD4⁺ T cell immunity against HBV infection.
Article
Full-text available
The T cell receptor (TCR) repertoire encodes immune exposure history through the dynamic formation of immunological memory. Statistical analysis of repertoire sequencing data has the potential to decode disease associations from large cohorts with measured phenotypes. However, the repertoire perturbation induced by a given immunological challenge is conditioned on genetic background via major histocompatibility complex (MHC) polymorphism. We explore associations between MHC alleles, immune exposures, and shared TCRs in a large human cohort. Using a previously published repertoire sequencing dataset augmented with high-resolution MHC genotyping, our analysis reveals rich structure: striking imprints of common pathogens, clusters of co-occurring TCRs that may represent markers of shared immune exposures, and substantial variations in TCR-MHC association strength across MHC loci. Guided by atomic contacts in solved TCR:peptide-MHC structures, we identify sequence covariation between TCR and MHC. These insights and our analysis framework lay the groundwork for further explorations into TCR diversity.
Article
Full-text available
T-cell receptor (TCR) repertoire data contain information about infections that could be used in disease diagnostics and vaccine development, but extracting that information remains a major challenge. Here we developed a statistical framework to detect TCR clone proliferation and contraction from longitudinal repertoire data. We applied this framework to data from three pairs of identical twins immunized with the yellow fever vaccine. We identified 500-1500 responding TCRs in each donor and validated them using three independent assays. While the responding TCRs were mostly private, albeit with higher overlap between twins, they could be well predicted using a classifier based on sequence similarity. Our method can also be applied to samples obtained post-infection, making it suitable for systematic discovery of new infection-specific TCRs in the clinic.
Article
Full-text available
Peptide-based T cell vaccines targeting the conserved epitopes of influenza virus can provide cross-protection against distantly related strains, but they are generally not immunogenic. Foreign antigen-specific regulatory T (Treg) cells are induced under subimmunogenic conditions peripherally, although their development and role in vaccine-mediated antiviral immunity is unclear. Here, we demonstrated primary vaccination with peptides alone significantly induced antigen-specific Foxp3+ Treg cells, which were further expanded by repeated vaccination with unadjuvanted peptides. Certain adjuvants, including CpG, suppressed the induction and expansion of antigen-specific Treg cells by peptide vaccination. Interestingly, secondary influenza virus infection significantly increased the frequency of preexisting antigen-specific Treg cells, although primary infection barely induced them. Importantly, specific depletion of vaccine-induced antigen-specific Treg cells promoted influenza viral clearance, indicating their inhibitory role in vivo. Immunization with CpG-adjuvanted peptides by the subcutaneous prime–intranasal-boost strategy restricted the recruitment and accumulation of antigen-specific Treg cells in lung, and stimulated robust T cell immunity. Finally, subcutaneous prime–intranasal-boost immunization with CpG-adjuvanted peptides or whole-inactivated influenza vaccines protected mice from heterosubtypic influenza virus infection. In conclusion, antigen-specific Treg cells induced by peptide vaccines attenuate the antiviral immunity against influenza virus infection. CpG-adjuvanted peptide vaccines provide heterosubtypic influenza protection probably by inhibiting Treg development and enhancing T cell immunity.
Article
Full-text available
FOXP3-expressing CD4⁺ T regulatory (Treg) cells are instrumental for the maintenance of self-tolerance. They are also involved in the prevention of allergy, allograft rejection, foetal rejection during pregnancy and of exaggerated immune response towards commensal pathogens in mucosal tissues. They can also prevent immune responses against tumors and promote tumor progression. FOXP3-expressing Treg cells are not a homogenous population. The different subsets of Treg cells can have different functions or roles in the maintenance of immune homeostasis and can therefore be differentially targeted in the management of autoimmune diseases or in cancer. We discuss here how Treg cell subsets can be differentiated phenotypically, functionally and developmentally in humans.
Article
Identification of antigen specificity of CD4 T cells is instrumental in understanding adaptive immune responses in health and disease. The high diversity of CD4 T cell repertoire combined with the functional heterogeneity of the compartment poses a challenge to the assessment of CD4 T cell responses. In spite of that, multiple technologies allow direct or indirect interrogation of antigen specificity of CD4 T cells. In the last decade, multiple surface proteins have been established as cytokine-independent surrogates of in vitro CD4 T cell activation, and have found applications in the live identification and isolation of antigen-responsive CD4 T cells. Here we review the current knowledge of the surface proteins that permit identification of viable antigen-responsive CD4 T cells with high specificity, including those capable of identifying specialized CD4 T subsets such as germinal center follicular helper T cells and CD4 regulatory T cells.
Article
Recent advances defining the role of the commensal gut microbiota in the development, education, induction, function, and maintenance of the mammalian immune system inform our understanding of how immune responses govern the outcome of systemic virus infection. While characterization of the impact of the local oral, respiratory, dermal and genitourinary microbiota on host immune responses and systemic virus infection is in its infancy, the gut microbiota interacts with host immunity systemically and at distal non-gastrointestinal tract sites to modulate the pathogenesis of systemic viruses. Gut microbes, microbe-associated molecular patterns, and microbe-derived metabolites engage receptors expressed on the cell surface, in the endosome, or in the cytoplasm to orchestrate optimal innate and adaptive immune responses important for controlling systemic virus infection.
Article
Motivation: The T-cell receptor (TCR) is responsible for recognizing epitopes presented on cell surfaces. Linking TCR sequences to their ability to target specific epitopes is currently an unsolved problem, yet one of great interest. Indeed, it is currently unknown how dissimilar TCR sequences can be before they no longer bind the same epitope. This question is confounded by the fact that there are many ways to define the similarity between two TCR sequences. Here we investigate both issues in the context of TCR sequence unsupervised clustering. Results: We provide an overview of the performance of various distance metrics on two large independent data sets with 412 and 2835 TCR sequences respectively. Our results confirm the presence of structural distinct TCR groups that target identical epitopes. In addition, we put forward several recommendations to perform unsupervised T-cell receptor sequence clustering. Availability and implementation: Source code implemented in Python 3 available at https://github.com/pmeysman/TCRclusteringPaper. Supplementary information: Supplementary data are available at Bioinformatics online.
Article
Hepatitis B virus (HBV) is a hepatotropic virus that can establish a persistent and chronic infection in humans through immune anergy. Currently, 3.5% of the global population is chronically infected with HBV, although the incidence of HBV infections is decreasing owing to vaccination and, to a lesser extent, the use of antiviral therapy to reduce the viral load of chronically infected individuals. The course of chronic HBV infection typically comprises different clinical phases, each of which potentially lasts for decades. Well-defined and verified serum and liver biopsy diagnostic markers enable the assessment of disease severity, viral replication status, patient risk stratification and treatment decisions. Current therapy includes antiviral agents that directly act on viral replication and immunomodulators, such as interferon therapy. Antiviral agents for HBV include reverse transcriptase inhibitors, which are nucleoside or nucleotide analogues that can profoundly suppress HBV replication but require long-term maintenance therapy. Novel compounds are being actively investigated to achieve the goal of HBV surface antigen seroclearance (functional cure), a serological state that is associated with a higher remission rate (thus, no viral rebound) after treatment cessation and a lower rate of cirrhosis and hepatocellular carcinoma. This Primer addresses several aspects of HBV infection, including epidemiology, immune pathophysiology, diagnosis, prevention and management.