Bacterial Vaccines (Etc.)
Bacterial Vaccines (Etc.)
Fadil Bidmos
Janine Bossé
Paul Langford Editors
Bacterial
Vaccines
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, UK
Edited by
Paul Langford
Department of Infectious Disease
Imperial College London
London, United Kingdom
This Humana imprint is published by the registered company Springer Science+Business Media, LLC, part of Springer
Nature.
The registered company address is: 1 New York Plaza, New York, NY 10004, U.S.A.
Preface
Vaccination is considered the second most effective public health intervention available, only
following the provision and access to clean water for disease prevention. Compared to the
peak number of deaths prior to introduction of bacterial vaccines against diphtheria, tetanus,
pertussis (whooping cough), and disease caused by Haemophilus influenzae type b, it is
estimated that c. 1.3 million lives, predominantly of children, have been saved annually. The
associated reduction in morbidity by prevention of disease through herd immunity, and
reduction in secondary infections that complicate vaccine-preventable diseases, is estimated
to save 386 million life years and 96 million disability-adjusted life years (DALYs) globally.
In addition to reduction of mortality and morbidity, the use of vaccines has substantial social
benefits. The latter include equity of healthcare, strengthening of health and social care
infrastructure, increased social mobility, improved life expectancy, and empowerment of
women. Economically, in lower- and middle-income countries, an investment of $34 billion
in vaccines resulted in savings of $586 billion from the cost associated with direct illness.
Cost-effectiveness or cost/benefit analyses do not typically consider prevention of long-
term morbidity following acute infection, such as hearing loss or amputation of limbs as
sequelae of meningococcal disease, and the calculated figures can be considered
underestimates.
Given their benefits, it seems a paradox that historically pharmaceutical manufacturers
have been wary of investing in vaccines because of concerns about legal liability and a
comparative low return on investment compared to pharmaceuticals. That situation has
recently started to change in the bacterial arena because of the threats posed by long-
standing diseases where there is still a need for improved vaccines (e.g., for tuberculosis).
In addition, with an aging worldwide population, vaccination is increasingly part of a life-
course strategy to meet the needs of the elderly. However, the main driver is the increase in
worldwide antimicrobial resistance (AMR). As Dame Sally Davies, the UK Chief Medical
Officer, stated: “The world is facing an antibiotic apocalypse. Unless action is taken to halt the
practices that have allowed antimicrobial resistance to spread and ways are found to develop
new types of antibiotics, we could return to the days when routine operations, simple wounds or
straightforward infections could pose real threats to life.” In particular, there is currently much
research focus on vaccine development for the so-called bacterial ESKAPE pathogens:
Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter bauman-
nii, Pseudomonas aeruginosa, and Enterobacter spp. Furthermore, new preventions and/or
antibiotics are urgently needed for other World Health Organization-designated critical or
high priority pathogens such as Neisseria gonorrhoeae, where strains have already been
isolated that are resistant to current recommended treatment regimes. The solution to
AMR will also involve improvement in and introduction of novel bacterial vaccines to reduce
antimicrobial use in animals to prevent transfer of resistance genes through natural transfor-
mation, transconjugation, transduction, and vesiduction in zoonotic pathogens that cause
disease in humans.
Thus, a book on bacterial vaccine methods is extremely timely. We have organized the
chapters into three sections: (1) vaccine antigen discovery; (2) vaccine production and
delivery; and (3) immunology of vaccine candidates. The first two chapters (Ong and He;
Leow et al.) describe reverse vaccinology methods—the use of bioinformatic approaches to
v
vi Preface
identify vaccine candidates, which have been successfully applied in development of the
commercially available Neisseria meningitidis serogroup B Bexsero vaccine. With over
200,000 bacterial genomes publicly available, e.g., through NCBI and EMBL, the use of
reverse vaccinology approaches can be a valuable starting point for immunogenic antigen
and/or epitope discovery, and crucially is not hypothesis-driven. Four further chapters
describe different proteomic approaches for vaccine antigen discovery. These include classic
methods to identify potential immunogenic epitopes, such as immunoprecipitation
(Reglinski), enzymatic surface shaving (Luu and Lan), two-dimensional electrophoresis
combined with western blotting (Obradovic and Wilson), as well as pan-proteomic array
technology (Campo and Oberai) which is also a bottom-up approach starting with genome
data, like reverse vaccinology. Nine chapters are devoted to vaccine production and delivery
systems, illustrating the range and ingenuity of platforms available. These include a relatively
simple yet elegant bacterin inactivation mechanism involving low-energy electron irradia-
tion (Fertey et al.), and bioengineering approaches such as those described for Gram-
negative metal ion transporters (Chauduri et al.), Gram-positive extracellular membrane
vesicles (Stentz et al.), and biological conjugation (Terra and Kay). Different methods for
preparation of outer membrane vesicles/generalized modules for membrane antigens are
presented, including those for secretory production of heterologous antigens (Kawamoto
and Kurihara), for reduction of potential toxicity (Hirayama and Nakao), and for ensuring
quality and stability (Micoli et al.). In addition, two chapters describe platforms that can be
used to present peptide subunits (trimethyl chitosan-based polyelectrolyte complexes)
(Zhao et al.) or multi-epitope fusion antigens (MEFA) (Li et al.), respectively. The final
set of chapters provide methods for assessment of antigens at the discovery, delivery, and
post-licensure phases, including monitoring of immune responses to inhaled antigens
(Ashhurst et al.) and in infants following maternal immunization (Rice and Holder). The
production of a universal human complement source (Alexander et al.) is described, which
could be used to ensure reproducibility of the functional opsonophagocytic activity and
serum bactericidal assays detailed by Semchenko et al. and Wagstaffe et al. Furthermore,
informative infection models, including the ex vivo organ co-perfusion model (Hames et al.)
and controlled human infection model (Dale et al.) described, as well as techniques such as
multi-color flow cytometry and high-dimensional data analysis for analyses of vaccine
responses (Cole et al.) will improve evaluation of prospective vaccines and enable prioritiza-
tion of candidates for field testing, thus speeding up the time to market.
We hope that researchers will not only find specific chapters of interest but also take the
time to review others detailed in the book, as overall they give a snapshot of the variety of
methods available and the extremely high innovative nature of the bacterial vaccine field. We
would like to express our thanks to all of the authors for their valuable contributions,
meeting deadlines, and their understanding through the editing process, and to the series
editor, John Walker, for his excellent guidance.
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 449
Contributors
ABU BAKAR ABDUL MAJEED • Faculty of Pharmacy, Universiti Teknologi MARA, Kuala
Selangor, Selangor, Malaysia
FRANCES ALEXANDER • Public Health England, Porton Down, UK
RENZO ALFINI • GSK Vaccines Institute for Global Health, Siena, Italy
LAUREN ALLEN • Public Health England, Porton Down, UK
ANNELIESE S. ASHHURST • School of Chemistry, The University of Sydney, Sydney, NSW,
Australia; Tuberculosis Research Program Centenary Institute, The University of Sydney,
Camperdown, NSW, Australia; School of Medical Sciences, Faculty of Medicine and Health,
The University of Sydney, Camperdown, NSW, Australia
SAHRA BASHIRI • School of Chemistry & Molecular Biosciences, The University of Queensland,
St Lucia, QLD, Australia
JANA BECKMANN • Fraunhofer Institute for Organic Electronics, Electron Beam and Plasma
Technology FEP, Dresden, Germany
WARWICK J. BRITTON • Tuberculosis Research Program Centenary Institute, The University of
Sydney, Camperdown, NSW, Australia; School of Medical Sciences, Faculty of Medicine and
Health, The University of Sydney, Camperdown, NSW, Australia; Department of Clinical
Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
EMILY BRUNT • Public Health England, Porton Down, UK
JOSEPH J. CAMPO • Antigen Discovery, Inc., Irvine, CA, USA
SIMON R. CARDING • Quadram Institute, Norwich, UK
DAVID CARRENO • Department of Genetics and Genome Biology, University of Leicester,
Leicester, UK
BREEZE CAVELL • Public Health England, Porton Down, UK
SOMSHUKLA CHAUDHURI • Department of Microbiology, Immunology, and Infectious
Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
HANNAH M. CHEESEMAN • Department of Infectious Disease, Imperial College London,
London, UK
CANDY CHUAH • Faculty of Medicine and Health Sciences, Universiti Tunku Abdul
Rahman, Kajang, Selangor, Malaysia
WEN Y. CHUNG • Department of Hepatobiliary and Pancreatic Surgery, University
Hospitals of Leicester, Leicester, UK
MEGAN E. COLE • The Pirbright Institute, Surrey, UK
ADAM P. DALE • Clinical and Experimental Sciences, University of Southampton,
Southampton, UK
ASHLEY R. DENNISON • Department of Hepatobiliary and Pancreatic Surgery, University
Hospitals of Leicester, Leicester, UK
NIKOLAS F. EWASECHKO • Department of Microbiology, Immunology, and Infectious Diseases,
Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
JAMIE E. FEGAN • Department of Molecular Genetics, University of Toronto, Toronto, ON,
Canada
JASMIN FERTEY • Fraunhofer-Institute for Cell Therapy and Immunology IZI, Leipzig,
Germany
ix
x Contributors
Abstract
Reverse vaccinology (RV) is the state-of-the-art vaccine development strategy that starts with predicting
vaccine antigens by bioinformatics analysis of the whole genome of a pathogen of interest. Vaxign is the first
web-based RV vaccine prediction method based on calculating and filtering different criteria of proteins.
Vaxign-ML is a new Vaxign machine learning (ML) method that predicts vaccine antigens based on extreme
gradient boosting with the advance of new technologies and cumulation of protective antigen data. Using a
benchmark dataset, Vaxign-ML showed superior performance in comparison to existing open-source RV
tools. Vaxign-ML is also implemented within the web-based Vaxign platform to support easy and intuitive
access. Vaxign-ML is also available as a command-based software package for more advanced and custom-
izable vaccine antigen prediction. Both Vaxign and Vaxign-ML have been applied to predict SARS-CoV-
2 (cause of COVID-19) and Brucella vaccine antigens to demonstrate the integrative approach to analyze
and select vaccine candidates using the Vaxign platform.
Key words Vaccine, Antigen, Reverse vaccinology, Machine learning, Vaxign, Vaxign-ML, Vaxitop
1 Introduction
1
2 Edison Ong and Yongqun He
Table 1
Vaxign system components
Vaxign system
component Included analysis
Vaxign Subcellular localization
Transmembrane helix
Adhesin probability
Similarity to host (human, mouse, and pig)
Vaxign-ML Extreme gradient boosting machine learning model to predict vaccine
candidate
Vaxitop Vaccine epitope prediction and analysis system based on the principle of reverse
vaccinology
Postprediction analysis IEDB epitope search
IEDB population coverage
EggNOG function
EggNOG orthologs
Pangenome analysis Orthologs’ phylogeny
Multiple sequence alignment
2 Vaxign-ML
2.1 Vaxign-ML The most convenient and straightforward way to use Vaxign-ML
Stand-Alone Web Tool for vaccine candidate prediction is to access the Vaxign-ML stand-
alone web tool http://www.violinet.org/vaxign2/vaxign-ml. A
typical Vaxign-ML web interface is provided in Fig. 1. The standard
procedure for running Vaxign-ML is as follows:
1. Input the protein sequence(s). The following formats are sup-
ported: (a) FASTA format, (b) UniProtKB Protein ID,
(c) NCBI Protein ID, (d) NCBI Protein RefSeq, (e) NCBI
Gene ID, and (f) FASTA File Download Link. Users can also
upload the protein sequence(s) in FASTA format instead of
typing into the input text field.
4 Edison Ong and Yongqun He
Table 2
Protegen protective antigen statistics
Table 2
(continued)
Table 2
(continued)
2.3 SARS-CoV-2 The Vaxign-ML was used to predict COVID-19 vaccine candidates
Example from the SARS-CoV-2 proteome [14]. As expected, the top candi-
date indicated by Vaxign-ML was the SARS-CoV-2 spike protein
(Table 3), which has been the primary target of many vaccines
currently in clinical trials. However, Vaxign-ML also predicted
several nonstructural proteins as a potential vaccine candidate.
Among which, the nsp3 protein was extensively studied in the
Vaxign-ML COVID-19 vaccine prediction study. A separate study
reported that the PL-PRO, as part of the nsp3, was associated with
innate immunity and virulence factor [15]. Overall, Vaxign and
Vaxign-ML offer a valuable resource to select vaccine candidates
and prioritize them for experimental verification.
8 Edison Ong and Yongqun He
Table 3
Predicted SARS-CoV-2 vaccine candidates using Vaxign-ML
The Vaxign-ML has been fully integrated into the Vaxign system.
The overall Vaxign system framework includes the primary Vaxign
analyses, Vaxign-ML vaccine candidate prediction, Vaxitop epitope
prediction, postprediction analysis, and pangenome analysis
(Fig. 2).
Vaccine Design by Reverse Vaccinology and Machine Learning 9
3.2 Vaxign Analyses In addition to the prediction analyses described above, Vaxign also
After Vaccine includes additional analyses, including epitope predictions using
Candidate Prediction Vaxitop epitope prediction, EggNOG Functions and Orthologs,
and pangenome orthologs’ phylogeny. The following is the proce-
dure of how such analyses are performed.
1. Following step 5 in Subheading 3.1 or step 8 in Subheading
3.3, users can perform additional analyses to aid their selection
of vaccine candidates.
2. There are two tabs in the upper panel of the overall result
summary: “Filter Results” and “Analysis.”
3. For the “Filter Results” tab, users can apply different filters to
narrow down the candidate list. These filters include subcellu-
lar localization, number of transmembrane helices, adhesin
probability, and similarity to human/mouse/pig proteins.
Besides, users can also include or exclude candidates with
orthologs in specific pathogen strain(s). (For details of obtain-
ing the ortholog analysis, see Subheading 3.3)
4. For the “Analysis” tab, users can browse the predicted epitopes
and the corresponding population coverage of the restricted
epitopes if the Vaxitop option is selected in Subheading 3.3
step 5 (see Note 2). Users can also view the complete orthologs
table of the pangenome analysis for specific pathogen strain(s).
(For details of obtaining the pangenomic analysis, see Subhead-
ing 3.3)
5. Users can also click the individual input protein, and a web
page will be opened with detailed information of the protein,
including basic information, Vaxitop prediction (if not
selected, a new Vaxitop analysis will be performed automati-
cally), IEDB epitope (see Note 3), IEDB population coverage
(see Note 2), EggNOG Functions and Orthologs (see Note 4),
and pangenome orthologs’ phylogeny and multiple sequence
alignment. (For details of obtaining the pangenomic analysis,
see Subheading 3.3)
3. Enter into the project summary page, and click “Start Vaxign
Dynamic Analysis” button.
4. Users can submit protein sequence(s) for prediction, and the
following formats are supported: (a) FASTA format,
(b) UniProtKB Protein ID, (c) UniProt Proteome ID,
(d) NCBI Protein ID, (e) NCBI Protein RefSeq, (f) NCBI
Gene ID, (g) NCBI BioProject ID, (h) NCBI Nucleotide ID,
and (i) FASTA File Download Link. In addition, users can also
upload the protein sequence(s) in FASTA format instead of
typing into the input text field.
5. Enter the genome group or pathogen name (e.g., Mycobacte-
rium tuberculosis), and genome or strain name (e.g., Mycobac-
terium tuberculosis H37Rv).
6. Select the pathogen organism type, the current Vaxign-ML
supports “Gram positive bacterium”, “Gram negative bacte-
rium”, “Virus”, and “Parasite”.
7. Select whether the basic Vaxign analyses includes (a) subcellular
localization, (b) transmembrane helix, (c) adhesin probability,
and (d) similarity to host (human, mouse, and pig) proteins.
8. Users can choose whether Vaxign-ML is to be included in the
pipeline.
9. Besides the basic Vaxign and Vaxign-ML analyses, the system
also supports epitope prediction via the in-house Vaxitop
program.
10. Click the submit button. Users will be directed to the project
summary page with the Vaxign analysis status displayed.
11. Repeat steps 3–10 for the pathogen strains to be included in
the pangenome analysis.
12. Once the protein sequences of all the pathogen strains are
submitted and the corresponding Vaxign analyses are finished,
users can click the “Run Vaxign Ortholog Analysis” for the
pangenome analysis.
13. Once the analysis is completed, users can click on the “result”
button for each submitted query. Then follow the same proce-
dures (see Subheading 3.4) to include or exclude candidates
with orthologs in specific pathogen strain(s).
14. To facilitate easier Vaxign usage, a set of precomputed queries
were created, which can be accessed from the home page
(or http://www.violinet.org/vaxign2/precompute). The pre-
computed Vaxign results from either the protein level or
genome level. Users are prompted to set up preferred query
criteria; the output data are then provided. The query of pre-
computed Vaxign results is fast.
12 Edison Ong and Yongqun He
Fig. 3 B. abortus 2308 Vaxign and Vaxign-ML analyses. Both Vaxign and Vaxign-
ML were applied to analyze the genome of B. abortus strain 2308. Five different
filtering criteria were used for the filtering analysis
Vaccine Design by Reverse Vaccinology and Machine Learning 13
B.abortus 2308 proteins (total: 3023) Vaxign-ML Predicted results (total: 482)
Unknown Unknown
Cytoplasmic
29% 15%
45%
Cytoplasmic
54%
Periplasmic
Periplasmic 12%
3%
Outer Membrane
Outer Membrane 5%
1%
Extracellular Extracellular
0.4% 3%
Cytoplasmic Cytoplasmic
Membrane Membrane
22% 11%
Fig. 4 Comparison of subcellular locations of all proteins of B. abortus strain 2308 and the 482 vaccine
candidates predicted by Vaxign-ML
4 Notes
Table 4
Predicted B. abortus vaccine candidates using Vaxign and Vaxign-ML
Vaxign-ML
Gene name Protein name score Localization
BAB_RS31580 Flagellar hook protein FlgE 94.8 EC
BR_RS15470 Flagellin 98.6 EC
BAB_RS25510 Hypothetical protein 95.7 EC
BR_RS08885 AprI/Inh family metalloprotease inhibitor, omp19 93.6 OM
flgK Flagellar hook-associated protein FlgK 91.7 OM
BAB_RS19300 LPS-assembly protein LptD 97.5 OM
BOV_RS07445 OmpW family protein 94.2 OM
BAB_RS26700 Outer membrane beta-barrel protein 91.9 OM
BR_RS06570 Outer membrane protein assembly factor BamD 92.5 OM
BAB1_0907 Peptidoglycan-binding LysM:Peptidase M23/M37 96.8 OM
BAB_RS22155 Porin 93.9 OM
BOV_RS00570 Porin family protein 91.1 OM
BR_RS00555 Porin family protein 92.3 OM
BAB_RS27875 Porin family protein 95.2 OM
BR_RS03235 Porin family protein, OMP_b-brl domain-containing 96.4 OM
protein
BAB_RS23735 Porin family protein, OMP_b-brl domain-containing 94 OM
protein
BAB_RS19090 Porin Omp2a 94.9 OM
BAB_RS19100 Porin Omp2b 96.9 OM
ybgF Tol-pal system protein YbgF 96.8 OM
BAB_RS22470 TonB-dependent receptor, Heme transporter BhuA 97.9 OM
BAB_RS31825 TonB-dependent receptor, Heme transporter BhuA 99.3 OM
BAB_RS31975 YadA-like family protein 98.1 OM
Note: The genome of B. abortus strain 2308 was used for this analysis
Acknowledgments
References
1. Pizza M, Scarlato V, Masignani V, Giuliani tumour antigens and subunit vaccines. BMC
MM, Aricò B, Comanducci M, Jennings GT, Bioinformatics 8:4. https://doi.org/10.
Baldi L, Bartolini E, Capecchi B, Galeotti CL, 1186/1471-2105-8-4
Luzzi E, Manetti R, Marchetti E, Mora M, 9. Bowman BN, McAdam PR, Vivona S, Zhang
Nuti S, Ratti G, Santini L, Savino S, JX, Luong T, Belew RK, Sahota H, Guiney D,
Scarselli M, Storni E, Zuo P, Broeker M, Valafar F, Fierer J, Woelk CH (2011) Improv-
Hundt E, Knapp B, Blair E, Mason T, ing reverse vaccinology with a machine
Tettelin H, Hood DW, Jeffries AC, Saunders learning approach. Vaccine 29:8156–8164.
NJ, Granoff DM, Venter JC, Moxon ER, https://doi.org/10.1016/j.vaccine.2011.07.
Grandi G, Rappuoli R (2000) Identification 142
of vaccine candidates against serogroup B 10. Heinson AI, Gunawardana Y, Moesker B, Den-
meningococcus by whole-genome sequencing. man Hume CC, Vataga E, Hall Y, Stylianou E,
Science 287:1816–1820. https://doi.org/10. McShane H, Williams A, Niranjan M, Woelk
1126/science.287.5459.1816 CH (2017) Enhancing the biological relevance
2. Vernikos G, Medini D (2014) Bexsero H of machine learning classifiers for reverse vacci-
chronicle. Pathog Glob Health 108:305–311. nology. Int J Mol Sci 18:312. https://doi.org/
https://doi.org/10.1179/2047773214Y. 10.3390/ijms18020312
0000000162 11. Dalsass M, Brozzi A, Medini D, Rappuoli R
3. Folaranmi T, Rubin L, Martin SW, Patel M, (2019) Comparison of open-source reverse
MacNeil JR (2015) Use of serogroup B menin- vaccinology programs for bacterial vaccine anti-
gococcal vaccines in persons aged 10 years at gen discovery. Front Immunol 10:1–12.
increased risk for serogroup B meningococcal https://doi.org/10.3389/fimmu.2019.
disease: recommendations of the advisory com- 00113
mittee on immunization practices, 2015. 12. Ong E, Wang H, Wong MU, Seetharaman M,
MMWR Morb Mortal Wkly Rep 64:608–612 Valdez N, He Y (2020) Vaxign-ML: supervised
4. Ong E, Wong MU, He Y (2017) Identification machine learning reverse vaccinology model
of new features from known bacterial protec- for improved prediction of bacterial protective
tive vaccine antigens enhances rational vaccine antigens. Bioinformatics 36:3185–3191.
design. Front Immunol 8:1–11. https://doi. https://doi.org/10.1093/bioinformatics/
org/10.3389/fimmu.2017.01382 btaa119
5. Vivona S, Bernante F, Filippini F (2006) 13. Yang B, Sayers S, Xiang Z, He Y (2011) Prote-
NERVE: new enhanced reverse vaccinology gen: a web-based protective antigen database
environment. BMC Biotechnol 6:35. https:// and analysis system. Nucleic Acids Res
doi.org/10.1186/1472-6750-6-35 39:1073–1078. https://doi.org/10.1093/
6. He Y, Xiang Z, Mobley HLT (2010) Vaxign: nar/gkq944
the first web-based vaccine design program for 14. Ong E, Wong MU, Huffman A, He Y (2020)
reverse vaccinology and applications for vaccine COVID-19 coronavirus vaccine design using
development. J Biomed Biotechnol reverse vaccinology and machine learning.
2010:1–15. https://doi.org/10.1155/2010/ Front Immunol 11:1581
297505 15. Shin D, Mukherjee R, Grewe D, Bojkova D,
7. Rizwan M, Naz A, Ahmad J, Naz K, Obaid A, Baek K, Bhattacharya A, Schulz L, Widera M,
Parveen T, Ahsan M, Ali A (2017) VacSol: a Mehdipour AR, Tascher G, Geurink PP,
high throughput in silico pipeline to predict Wilhelm A, van der Heden van Noort GJ,
potential therapeutic targets in prokaryotic Ovaa H, Müller S, Knobeloch K-P,
pathogens using subtractive reverse vaccinol- Rajalingam K, Schulman BA, Cinatl J,
ogy. BMC Bioinformatics 18:1–7. https:// Hummer G, Ciesek S, Dikic I (2020) Papain-
doi.org/10.1186/s12859-017-1540-0 like protease regulates SARS-CoV-2 viral
8. Doytchinova IA, Flower DR (2007) VaxiJen: a spread and innate immunity. Nature 587
server for prediction of protective antigens,
16 Edison Ong and Yongqun He
Abstract
Reverse vaccinology (RV) was first introduced by Rappuoli for the development of an effective vaccine
against serogroup B Neisseria meningitidis (MenB). With the advances in next generation sequencing
technologies, the amount of genomic data has risen exponentially. Since then, the RV approach has widely
been used to discover potential vaccine protein targets by screening whole genome sequences of pathogens
using a combination of sophisticated computational algorithms and bioinformatic tools. In contrast to
conventional vaccine development strategies, RV offers a novel method to facilitate rapid vaccine design and
reduces reliance on the traditional, relatively tedious, and labor-intensive approach based on Pasteur”s
principles of isolating, inactivating, and injecting the causative agent of an infectious disease. Advances in
biocomputational techniques have remarkably increased the significance for the rapid identification of the
proteins that are secreted or expressed on the surface of pathogens. Immunogenic proteins which are able
to induce the immune response in the hosts can be predicted based on the immune epitopes present within
the protein sequence. To date, RV has successfully been applied to develop vaccines against a variety of
infectious pathogens. In this chapter, we apply a pipeline of bioinformatic programs for identification of
Shigella flexneri potential vaccine candidates as an illustration immunoinformatic tools available for RV.
Key words Antigens, Bacteria, Epitope, Immunoinformatics, Outer membrane protein (OMP),
Shigella flexneri, Reverse vaccinology (RV), Vaccine
1 Introduction
17
18 Chiuan Yee Leow et al.
BLASTP (UniProt)
Fig. 1 Flowchart of the use of reverse vaccinology approach for the prediction of vaccine candidates against
Shigella flexneri
20 Chiuan Yee Leow et al.
2 Materials
3 Methods
3.1 Identification 1. Point the web browser to the Vaxign website at http://www.
of Vaccine Targets violinet.org/vaxign/. The Vaccine Design section of the Vax-
Using Vaxign ign page appears (see Note 2).
3.1.1 Select a Genome 2. In the “Vaxign Query” section (Fig. 2a), first go to “Select a
Genome Group.” Select <Shigella (5) > in the drop-down
menu. The number (5) means that Vaxign contains five gen-
omes corresponding to Shigella species (see Note 3).
3. Next, go to “Select a Genome.” Select <Shigella flexneri 2a
strain 2457T genome> in the drop-down menu (see Note 4).
4. Keep the default settings for “Keywords” and “Sort by”
options. Proceed to “Filter Options.”
Fig. 2 Vaxign query submission for genome-based vaccine candidate prediction. (a) Vaxign query web
interface. (b) An example of potential vaccine candidates predicted using Vaxign pipeline
6. In the result page, check one or more box at the left-most side
of the table to select protein(s) for further display.
7. Click link(s) in the “Protein Accession” for detailed informa-
tion, which will be displayed on a new page. The desired
FASTA protein sequences can be saved to appropriate files, or
copied and pasted, as required into subsequent analysis plat-
forms, such as BLASTP, below (see Note 14).
8. If biological function of the protein is of interest, click the
“Run Vaxign COG analysis for all records” link above the
Table. A new page will be displayed. If functional orthologues
of the protein are of interest, click “Show Ortholog Table” link
above the Table. A new page will be displayed.
9. Click the link above the table for “Export all records to MS
Excel file” (for all proteins are selected) or “Export selected to
MS Excel file” (if only certain proteins are selected).
10. Save the MS Excel file on a working directory.
3.2 Identification 1. Point the internet browser to the NCBI BLASTP website at
of Conserved Identity https://blast.ncbi.nlm.nih.gov/Blast.cgi?PAGE¼Proteins
with Other Shigella 2. In the “Enter Query Sequence” box (Fig. 3a),
Strains (a) Copy and paste a either a protein accession number or the
protein sequence (either raw sequence or FASTA-
formatted sequences of proteins identified in Subheading
3.1.2, step 7) into the form field.
(b) Alternatively, the user can upload a protein sequence file
saved in FASTA-format.
(c) Entering a “Job Title” is optional.
(d) Ignore checking “Align two or more sequences” (see Note
15).
(e) Proceed to next section.
3. In the “Choose Search Set” section,
(a) Select the <non-redundant protein sequences (nr) > data-
base in the drop-down menu.
(b) Enter organism(s) to search against. In this case, <Shigella
sonnei (taxid:624)>; <Shigella dysenteriae (taxid:622)>;
and < Shigella boydii (taxid:621) > were selected (see
Note 16).
(c) Do not check the “Exclude” boxes.
(d) Proceed to next section.
4. For “Program Selection,” use the default blastp (protein–pro-
tein BLAST) program.
5. Click the “BLAST” button at the bottom of the page. The
results will appear as in Fig. 3b (see Note 17).
Reverse Vaccinology for S. flexneri Antigen Discovery 23
Fig. 3 An example of BLASTP protein sequence similarity analysis among Shigella spp. proteomes. (a) Protein
sequence submission interface. Outcome of BLASTP analysis. (b) The result showing NP_838365 of S. flexneri
is orthologous in S. dysenteriae
24 Chiuan Yee Leow et al.
3.3 Identification 1. Point the internet browser to the BLAST function within the
of Human Homologs UniProt website at https://www.uniprot.org/blast [27].
2. Copy and paste a protein sequence (raw data or FASTA-
formatted sequences of proteins identified in Subheading
3.1.2, step 7) into the form field (Fig. 4a).
Fig. 4 An example of protein sequence similarity analysis against human whole proteome using BLAST tool in
UniProt. (a) Protein sequence submission interface. (b) Outcome of BLAST analysis showing NP_838365 has
no similar protein sequence found in human proteome
Reverse Vaccinology for S. flexneri Antigen Discovery 25
3.4 Antigenicity 1. Point the internet browser to the VaxiJen v2.0 website at
Analysis http://www.ddg-pharmfac.net/vaxijen/VaxiJen/VaxiJen.
html
2. At “Enter a PROTEIN sequence here,” copy and paste a
protein sequence (plain format) in the form field. Alternatively,
a file containing multiple protein sequences saved in FASTA-
format can be submitted using the “Choose file” button
(Fig. 5a).
3. At “Select a TARGET ORGANISM,” select <Bacteria> in the
scrollable selection menu.
4. Keep default setting for the “THRESHOLD” (cutoff at 0.4)
(see Note 19).
5. Check the “Sequence Output” box. Check the other provided
boxes if user would like to learn more results for the submitted
protein sequence (see Note 20).
6. Click the “Submit” button at the bottom of the page. The
results appear as in Fig. 5b.
7. Five outer membrane proteins predicted to be potential vaccine
candidates against S. flexneri were selected for further analysis,
and the relevant results for these are shown in Table 1. Each
protein was subsequently analyzed to identify potential B- and
T-cell epitopes.
3.5 Prediction 1. Point the internet browser to the predictions page of the
of Linear B-Cell BCPREDS website at http://ailab-projects1.ist.psu.
Epitopes edu:8080/bcpred/predict.html
2. Copy and paste a protein sequence (plain format) in the form
field (Fig. 6a).
3. At “Methods,” select the “Fixed length epitope prediction”
option <BCPred> with a window (epitope length) of
20 amino acids, as set by the default parameters (see Note 21).
4. At “Specificity,” select <75% > in the drop-down menu (see
Note 22).
26 Chiuan Yee Leow et al.
Fig. 5 VaxiJen v2.0 query submission for protein antigenicity prediction. (a) VaxiJen v2.0 query web interface.
(b) An example of result page. The protein (NP_838365) was predicted to be antigenic. Threshold for this
model is 0.4; overall antigen prediction is 0.6201 (predicted to be probable ANTIGEN)
3.6 Prediction of HLA 1. Point the internet browser to the HLApred website at http://
Class I and Class II crdd.osdd.net/raghava/hlapred/ (see Note 23).
T-Cell Epitope 2. Copy and paste a protein sequence (plain format) into the
“Paste Your Protein Sequence” form field, or use the “Upload
Sequence File” option to choose a saved sequence file (Fig. 7).
Reverse Vaccinology for S. flexneri Antigen Discovery 27
Table 1
List of potential vaccine candidates against S. flexneri predicted using RV approach
Fig. 6 BCPreds server for B-cell linear epitope prediction. (a) Cover page of BCPreds. (b) An example of result
page for the prediction of B-cell epitope performed using BCPreds. Table showing seven 20-mers B-cell
epitopes were predicted for NP_838365. Peptide (20 mers) with score >0.75 were predicted to be potential
B-cell linear epitope
Reverse Vaccinology for S. flexneri Antigen Discovery 29
Fig. 7 A web interface of HLAPred. A server for prediction and identification of HLA class I and class II T-cell
epitope using quantitative matrix-based prediction method
4 Notes
Fig. 8 An example of result page (HTML-II view) for HLA class I T-cell epitope predicted using HLAPred.
Predicted binders (9-mer) are displayed as blue colored region, with P1 anchor or the starting residue of each
predicted binding frame as red colored
Fig. 9 An example of result page (HTML-II view) for HLA class II T-cell epitope predicted using HLAPred.
Predicted binders (9-mer) are displayed as blue colored region, with P1 anchor or the starting residue of each
predicted binding frame as red colored
Reverse Vaccinology for S. flexneri Antigen Discovery 31
Acknowledgments
References
of Mycobacterium spp. for peptide-based sub- 25. Singh H, Raghava GP (2003) ProPred1: pre-
unit vaccine design. BioMed Res Int diction of promiscuous MHC class-I binding
2017:4826030. https://doi.org/10.1155/ sites. Bioinformatics 19(8):1009–1014.
2017/4826030 https://doi.org/10.1093/bioinformatics/
16. He Y, Rappuoli R, De Groot AS, Chen RT btg108
(2010) Emerging vaccine informatics. J 26. Singh H, Raghava GP (2001) ProPred: predic-
Biomed Biotechnol 2010:218590. https:// tion of HLA-DR binding sites. Bioinformatics
doi.org/10.1155/2010/218590 17(12):1236–1237. https://doi.org/10.
17. Moise L, Cousens L, Fueyo J, De Groot AS 1093/bioinformatics/17.12.1236
(2011) Harnessing the power of genomics and 27. UniProt C (2019) UniProt: a worldwide hub
immunoinformatics to produce improved vac- of protein knowledge. Nucleic Acids Res 47
cines. Expert Opin Drug Discov 6(1):9–15. (D1):D506–D515. https://doi.org/10.
https://doi.org/10.1517/17460441.2011. 1093/nar/gky1049
534454 28. Yu NY, Wagner JR, Laird MR et al (2010)
18. Kazi A, Chuah C, Majeed ABA et al (2018) PSORTb 3.0: improved protein subcellular
Current progress of immunoinformatics localization prediction with refined localization
approach harnessed for cellular- and antibody- subcategories and predictive capabilities for all
dependent vaccine design. Pathog Glob Health prokaryotes. Bioinformatics 26
112(3):123–131. https://doi.org/10.1080/ (13):1608–1615. https://doi.org/10.1093/
20477724.2018.1446773 bioinformatics/btq249
19. Kazi A, Hisyam Ismail CMK, Anthony AA et al 29. Tusnady GE, Simon I (2001) The HMMTOP
(2020) Designing and evaluation of an transmembrane topology prediction server.
antibody-targeted chimeric recombinant vac- Bioinformatics 17(9):849–850. https://doi.
cine encoding Shigella flexneri outer mem- org/10.1093/bioinformatics/17.9.849
brane antigens. Infect Genet Evol 80:104176. 30. Sachdeva G, Kumar K, Jain P, Ramachandran S
https://doi.org/10.1016/j.meegid.2020. (2005) SPAAN: a software program for predic-
104176 tion of adhesins and adhesin-like proteins using
20. Mount DW (2007) Using the Basic Local neural networks. Bioinformatics 21
Alignment Search Tool (BLAST). CSH Proto- (4):483–491. https://doi.org/10.1093/bioin
cols 2007:pdb top17. https://doi.org/10. formatics/bti028
1101/pdb.top17 31. Raynes JM, Young PG, Proft T et al (2018)
21. Altschul SF, Gish W, Miller W, Myers EW, Lip- Protein adhesins as vaccine antigens for
man DJ (1990) Basic local alignment search Group A Streptococcus. Pathog Dis 76(2).
tool. J Mol Biol 215(3):403–410. https:// https://doi.org/10.1093/femspd/fty016
doi.org/10.1016/S0022-2836(05)80360-2 32. Nagy G, Emody L, Pal T (2008) Strategies for
22. Doytchinova IA, Flower DR (2007) VaxiJen: a the development of vaccines conferring broad-
server for prediction of protective antigens, spectrum protection. Int J Med Microbiol 298
tumour antigens and subunit vaccines. BMC (5–6):379–395. https://doi.org/10.1016/j.
Bioinformatics 8:4. https://doi.org/10. ijmm.2008.01.012
1186/1471-2105-8-4 33. De Groot AS, Ardito M, McClaine EM et al
23. El-Manzalawy Y, Dobbs D, Honavar V (2008) (2009) Immunoinformatic comparison of
Predicting linear B-cell epitopes using string T-cell epitopes contained in novel swine-origin
kernels. J Mol Recognit 21(4):243–255. influenza A (H1N1) virus with epitopes in
https://doi.org/10.1002/jmr.893 2008-2009 conventional influenza vaccine.
24. El-Manzalawy Y, Dobbs D, Honavar V (2008) Vaccine 27(42):5740–5747. https://doi.org/
Predicting flexible length linear B-cell epitopes. 10.1016/j.vaccine.2009.07.040
Comput Syst Bioinformatics Conf 7:121–132
Chapter 3
Abstract
Immunoprecipitation is an affinity purification technique that exploits the highly specific interactions
formed between antibodies and their cognate antigens to purify molecules of interest from complex
biological solutions. The generation of an effective humoral response provides protection against a wide
range of gram-positive pathogens, and thus immunoprecipitation using antibodies purified from immune
humans or animals provides a simple but effective means of isolating prospective vaccine antigens from
fractionated bacterial cells for downstream identification. The commercial availability of antibody prepara-
tions from donated human plasma, containing antibodies against many common gram-positive pathogens,
allows the protocol to be performed in the absence of bespoke vaccination experiments. Thus, immuno-
precipitation has the potential to reduce the number of animals used in vaccine studies by allowing an initial
screen for promising antigens to be conducted in vitro.
1 Introduction
37
38 Mark Reglinski
2 Materials
2.1 Preparation of 1. 30% w/v raffinose (see Note 2): Dissolve 30 g of raffinose in
Gram-Positive Cell 80 ml of deionized water using a magnetic hotplate and a stir
Wall Extracts bar. Add deionized water to 100 ml and autoclave. Store at
room temperature (RT).
2. 1 M Tris–HCl (pH 8): Dissolve 12.1 g of tris base (NH2C
(CH2OH)3) in 80 ml of deionized water and adjust to pH 8
with concentrated HCl. Add deionized water to 100 ml and
store at RT.
3. 10 kU/ml mutanolysin: Add 1 ml of molecular grade water to
a vial containing 10,000 U of mutanolysin. Vortex vigorously
to ensure that the powder has completely dissolved and store at
20 C in 100 μl aliquots.
4. 100 mg/ml lysozyme: Dissolve 100 mg of lysozyme in 1 ml of
molecular grade water. Vortex vigorously to ensure that the
powder has completely dissolved and store at 20 C in 100 μl
aliquots.
5. Protease Inhibitor Cocktail Set III (Millipore).
6. Cell wall extraction buffer: 30% raffinose, 10 mM Tris–HCl,
100 U/ml mutanolysin, 1 mg/ml lysozyme, 1% protease
inhibitor cocktail. Combine 960 μl of 30% raffinose with
10 μl of Tris–HCl, mutanolysin, lysozyme, and protease inhib-
itor cocktail solutions to give 1 ml of cell wall extraction buffer
(see Note 3).
7. 20 ml polypropylene syringes with Luer slip.
8. 18-gauge, 1-in. beveled needles.
9. Slide-A-Lyzer Dialysis Cassettes, 20 K MWCO, 12 ml
(Thermo-Fisher).
10. Vivaspin 20 centrifugal concentrators, MWCO 10 kDa
(Sartorius).
Immunoprecipitation of Vaccine Antigens 39
2.2 Coupling of 1. Coupling buffer (see Note 4): 0.1 M sodium bicarbonate,
Proteins to Cyanogen 0.5 M sodium chloride (pH 8.3). Dissolve 8.4 g of sodium
Bromide Activated bicarbonate (NaHCO3) and 29 g of sodium chloride (NaCl) in
Sepharose 800 ml of deionized water. Adjust to pH 8.3 with 5 M sodium
hydroxide (NaOH, see Note 5) and add deionized water to 1 l.
Store at RT.
2. 1 mM hydrochloric acid (HCl, see Note 6): Dilute 83 μl of
concentrated HCl in 1 L of deionized water. Store at 4 C.
3. 0.2 M glycine: Dissolve 7.5 g of glycine (NH2CH2COOH) in
400 ml of deionized water. Add deionized water to 500 ml and
store at RT.
4. Wash buffer: 0.1 M acetate, 0.5 M sodium chloride (pH 4).
Dissolve 4.2 g of anhydrous sodium acetate (C2H3O2Na) and
14.5 g of NaCl in 400 ml of deionized water. Add 2.2 ml of
glacial acetic acid and adjust to pH 4 using concentrated HCl
(see Note 5). Add deionized water to 500 ml and store at RT.
5. Cyanogen bromide-activated Sepharose (CBr-Sepharose), lyo-
philized powder (Sigma-Aldrich).
6. Econo-Column® Chromatography Column, 1.5 5 cm
(Bio-Rad).
7. Horizontal tube rotator capable of supporting 15 ml centrifuge
tubes.
8. Sodium azide: 1% in aqueous solution, purchased premade
from commercial vendors.
3 Methods
3.1 Preparation of 1. Culture the target bacterium to the desired growth phase
Gram-Positive Cell under appropriate conditions in 500 ml aliquots (see Note 8).
Wall Extracts 2. Centrifuge the culture at 4000 g for 10 min and resuspend
the cell pellet in 10 ml of 30% raffinose.
3. Centrifuge the cell suspension at 4000 g for 10 min and
resuspend the cell pellet in 10 ml of cell wall extraction buffer.
4. Incubate at 37 C for 3 h with occasional agitation.
5. Pellet the protoplasts at 14,000 g for 10 min and aspirate the
supernatant (cell wall extract).
6. Pass the cell wall extract through a 0.22 μm syringe filter and
transfer to a rehydrated 20 kDa MWCO cassette using a 20 ml
syringe and 18-gauge needle (see Note 9).
7. Dialyze the sample against 2 l of coupling buffer for at least 4 h
at 4 C.
8. Discard the spent dialysate and replace with 2 l of fresh cou-
pling buffer. Dialyze the sample overnight at 4 C.
9. Transfer the cell wall extract to a centrifugal filter unit and
concentrate to approximately 1 ml through multiple rounds
of centrifugation at 3000 g in a refrigerated centrifuge (see
Note 10).
10. Measure protein concentration of cell wall extract using a
colorimetric protein assay (see Note 11) and store at 20 C.
3.2 Coupling the Cell 1. Gradually add 250 mg of CBr-Sepharose to 50 ml of ice cold
Wall Proteins to CBr- 1 mM HCl and incubate at RT for 30 min with occasional
Sepharose agitation (see Note 12).
2. Transfer the resin suspension to a 1.5 5 cm chromatography
column and allow HCl to flow through the column.
3. Wash the resin bed with 5 ml of deionized water then with 5 ml
of coupling buffer, and immediately resuspend the resin in
2–4 ml of coupling buffer containing approximately 1 mg of
cell wall extract (see Note 13).
4. Incubate the resin slurry overnight at 4 C using a horizontal
tube rotator to maintain the liquid–solid suspension (see
Note 12).
5. Allow the resin to settle and collect the flow through for
downstream analysis (see Note 14).
6. Block the unreacted groups with 5 ml of 0.2 M glycine for 2 h
at RT using a horizontal tube rotator to maintain the liquid–
solid suspension.
Immunoprecipitation of Vaccine Antigens 41
3.3 Affinity 1. Wash the affinity purification resin extensively with loading
Purification of buffer to remove the sodium azide.
Pathogen-Reactive 2. Apply 5 ml of pooled human immunoglobulin (25 mg of IgG)
Antibodies in loading buffer and incubate at RT for 2 h using a horizontal
tube rotator to maintain the liquid–solid suspension.
3. Allow resin to settle and collect flow-through for downstream
PRAP purifications (see Note 15).
4. Wash the resin with 20 ml of loading buffer and elute the
bound antibody (PRAP) directly into 8 ml of neutralization
buffer using 8 ml of 1 M acetic acid (see Note 16).
5. Wash the column with 10 ml of loading buffer and strip the
remaining antibody using 10 ml of 0.5 M NaOH (see
Note 17).
6. Wash the column with loading buffer until the pH of the flow-
through is neutral.
7. Resuspend the resin in 2 ml of loading buffer supplemented
with 100 μl of 1% sodium azide solution (final concentration
0.05%). Store at 4 C.
8. Transfer the eluted PRAP to a rehydrated 20 kDa MWCO
cassette using a 20 ml syringe and 18-gauge needle.
9. Dialyze the sample against 2 l of coupling buffer for at least 4 h
at 4 C.
10. Discard the spent dialysate and replace with 2 l of fresh cou-
pling buffer. Dialyze the sample overnight at 4 C.
11. Transfer the PRAP to a centrifugal filter unit and concentrate
to 1 mg/ml through multiple rounds of centrifugation at
3000 g in a refrigerated centrifuge (see Note 10).
12. Measure protein concentration of the PRAP and store at 4 C.
42 Mark Reglinski
4 Notes
References
1. Wofsy L, Burr B (1969) The use of affinity 7. Mehta PD (1972) Immunological techniques.
chromatography for the specific purification Indian J Ophthalmol 20(2):49–54
of antibodies and antigens. J Immunol 103 8. Marquis RE (1965) Osmotic stability of bacte-
(2):380–382 rial protoplasts related to molecular size of sta-
2. DeCaprio J, Kohl TO (2019) Tandem Immu- bilizing solutes. Biochem Biophys Res
noaffinity purification using anti-FLAG and Commun 20(5):580–585. https://doi.org/
anti-HA antibodies. Cold Spring Harb Protoc 10.1016/0006-291x(65)90438-9
2019(2). https://doi.org/10.1101/pdb. 9. Reglinski M, Sriskandan S (2019) Treatment
prot098657 potential of pathogen-reactive antibodies
3. Del Villano BC, Defendi V (1973) Characteri- sequentially purified from pooled human
zation of the SV40 T antigen. Virology 51 immunoglobulin. BMC Res Notes 12(1):228.
(1):34–46. https://doi.org/10.1016/0042- https://doi.org/10.1186/s13104-019-4262-
6822(73)90363-2 8
4. Alspaugh MA, Talal N, Tan EM (1976) Differ- 10. Wilson R, Cohen JM, Reglinski M, Jose RJ,
entiation and characterization of autoantibo- Chan WY, Marshall H, de Vogel C,
dies and their antigens in sjögren’s syndrome. Gordon S, Goldblatt D, Petersen FC,
Arthritis Rheum 19(2):216–222. https://doi. Baxendale H, Brown JS (2017) Naturally
org/10.1002/art.1780190214 acquired human immunity to pneumococcus
5. Ohman JL, Lowell FC, Bloch KJ (1974) Aller- is dependent on antibody to protein antigens.
gens of mammalian origin. J Immunol 113 PLoS Pathog 13(1):e1006137. https://doi.
(6):1668 org/10.1371/journal.ppat.1006137
6. Seite JF, Shoenfeld Y, Youinou P, Hillion S 11. Reglinski M, Gierula M, Lynskey NN, Edwards
(2008) What is the contents of the magic RJ, Sriskandan S (2015) Identification of the
draft IVIg? Autoimmun Rev 7(6):435–439. Streptococcus pyogenes surface antigens recog-
https://doi.org/10.1016/j.autrev.2008.04. nised by pooled human immunoglobulin. Sci
012 Rep 5:15825. https://doi.org/10.1038/
srep15825
Chapter 4
Abstract
The bacterial cell surface (surfaceome) is the first site encountered by immune cells and is thus an important
site for immune recognition. As such, the characterization of bacterial surface proteins can lead to the
discovery of novel antigens for potential vaccine development. In this chapter, we describe a rapid 5-min
surface shaving proteomics protocol where live bacterial cells are incubated with trypsin and surface
peptides are “shaved” off. The shaved peptides are subsequently identified with liquid chromatography–-
tandem mass spectrometry (LC-MS/MS). Several checkpoints, including colony forming unit (CFU)
counts, flow cytometry, and a false positive unshaved control, are introduced to ensure cell viability/
membrane integrity are maintained and that proteins identified are true surface proteins. The protein
topology of shaved peptides can be bioinformatically confirmed for surface location. Surface shaving
facilitates identification of surface proteins expressed under different conditions, by different strains as
well as highly abundant essential and immunogenic bacterial surface antigens for potential vaccine
development.
Key words Surface shaving, Surfaceome, Surface proteins, Antigens, Vaccines, Proteomics, Cell
surface, Mass spectrometry, LC-MS/MS, Epitopes
1 Introduction
47
48 Laurence Don Wai Luu and Ruiting Lan
Fig. 1 Overview of the rapid surface shaving protocol for proteomic identification of surface antigens from live
cells. Live whole cells are incubated with trypsin for 5 min to shave off surface peptides. An unshaved control
is incubated under identical conditions without trypsin to identify peptides released during incubation which
could lead to false positive prediction of surface exposure. Cell viability/membrane integrity is assessed using
flow cytometry with propidium iodide (PI) and CFU counts. Shaved peptides are desalted and concentrated
with C18 StageTips and analysed with LC-MS/MS.
2 Materials
2.3 Flow Cytometry 1. FACS buffer (1% BSA and 0.1% sodium azide in 1 PBS): Add
0.5 g BSA and 0.05 g sodium azide to 50 mL of 1 PBS. Filter-
sterilise with a 0.22-μm filter and store at 4 C (see Note 5).
2. 0.5 mg/mL propidium iodide (PI) (see Note 6).
2.4 C18 Clean Up 1. Pierce 200 μL C18 StageTips from Thermo Fisher to desalt
and Liquid and concentrate peptides for LC-MS/MS.
Chromatography– 2. Centrifuge adapter from GL Sciences to hold the StageTips in
Tandem Mass place in 1.5 mL microcentrifuge tubes (Fig. 2).
Spectrometry 3. 0.2% heptafluorobutyric acid (HFBA), HPLC grade.
(LC-MS/MS)
4. 80% acetonitrile, 5% formic acid: 800 μL of acetonitrile (HPLC
grade), 50 μL of formic acid (LC-MS grade), and 150 μL of
ultrapure water.
3. 0.1% formic acid.
Rapid Bacterial Surface Shaving to Identify Surface Antigens 51
Fig. 2 Set up of 200 μL C18 StageTips in the centrifuge adapter and 1.5 mL microcentrifuge tube
3 Methods
3.2 Rapid Surface 1. Centrifuge the 12 h culture at 3300 g for 15 min at 4 C and
Shaving with Trypsin discard the supernatant (see Note 10).
2. Gently wash cells with 1 mL of ice-cold 1 PBS and centrifuge
at 3300 g for 15 min at 4 C. Discard supernatant and repeat
wash step for a total of 3 washes (see Note 11).
3. Gently resuspend cells in 2 mL of 10 mM HEPES (see
Note 12).
4. Split the sample into two by aliquoting 1 mL of the sample into
a TPP TubeSpin bioreactor tube labeled “shaved” and another
labeled as “unshaved control” (see Notes 13 and 14).
5. Add 1 μL of 1 μg/μL trypsin to the “shaved” sample (see
Note 15).
6. Incubate both “shaved” and “unshaved control” samples for
5 min at 37 C with gentle agitation (see Notes 16 and 17).
7. Transfer both samples into two new, appropriately labeled,
1.5 mL microcentrifuge tubes.
8. Optional: aliquot 10 μL of each sample for colony forming unit
(CFU) counts (see Note 18).
9. Pellet cells in both samples by centrifugation at maximum
speed (20,238 g) for 1 min at 4 C (see Note 19).
10. For both the “shaved” and “unshaved control” samples, trans-
fer the respective supernatants into new, appropriately labeled,
1.5 mL microcentrifuge tubes. The cell pellets from each sam-
ple are kept on ice until processed for flow cytometry (see
Subheading 3.3).
11. Add 1 μL of 1 μg/μL trypsin to the supernatant of the
“unshaved control” sample and incubate for 5 min at 37 C.
Keep the “shaved” sample supernatant on ice to prevent fur-
ther trypsin digestion while the “unshaved control” is under-
going trypsin digestion.
12. Add 1% of formic acid (10 μL) to both samples to inactivate
trypsin digestion (see Note 20).
3.3 Determining Cell 1. Prepare a positive control for cell death by resuspending 1 loop-
Viability After Rapid ful of Bvg+ B. pertussis grown on BG agar in 1 mL of 1x PBS
Surface Shaving Using and heat at 70 C for 30 min. Centrifuge at maximum speed
Propidium Iodide (PI). (20,238 g) for 1 min at 4 C. Discard supernatant and
resuspend cells in 900 μL of FACS buffer (see Note 21).
Rapid Bacterial Surface Shaving to Identify Surface Antigens 53
3.4 C18 Peptide 1. Prepare the “shaved” and “unshaved control” supernatant
Clean-Up samples (from Subheading 3.2, step 12) for C18 clean-up by
and LC-MS/MS mixing 50 μL of sample with 20 μL of 0.2% HFBA in appropri-
ately labeled 1.5 mL microcentrifuge tubes (see Note 23).
2. For each sample, place a C18 StageTip into a centrifuge adapter
and insert into an appropriately labeled 1.5 mL microcentri-
fuge tube (Fig. 2).
3. Initialize the C18 StageTips by adding 20 μL of 80% acetoni-
trile, 5% formic acid. Centrifuge at 200 g for 2 min (see
Note 24).
4. Reequilibrate the C18 StageTips with 20 μL of 0.2% HFBA
and centrifuge at 200 g for 2 min. Discard flowthrough.
5. Load each 70 μL sample (from Subheading 3.4, step 1) into a
prepared C18 StageTip and centrifuge at 200 g for 2 min (see
Notes 25 and 26).
6. Wash the samples on the C18 StageTips with 20 μL of 0.2%
HFBA and centrifuge at 200 g for 2 min.
7. Transfer the C18 StageTips with bound sample, along with
their centrifuge adaptors, to new appropriately labeled 1.5 mL
microcentrifuge tubes and elute samples with 20 μL of 80%
acetonitrile, 5% formic acid and centrifuge at 200 g for 2 min.
8. Vacuum-dry samples (see Note 27).
9. Resuspend dried peptides in 10 μL of 0.1% formic acid and
perform LC-MS/MS (see Note 28).
3.5 Bioinformatics 1. Load the resulting LC-MS/MS raw datafiles into Mascot
Analysis Daemon (v.2.5.1) for protein identification with the following
and Identification search parameters: instrument type ¼ ESI-type; peptide toler-
of Surface Proteins ance ¼ 4 ppm; MS/MS tolerance ¼ 0.4 Da; variable modifica-
tions ¼ Carbamidomethyl (C) and Oxidation (M); enzyme
54 Laurence Don Wai Luu and Ruiting Lan
Fig. 3 (a) Example screenshot of the Samples tab in Scaffold illustrating the number of proteins and peptides
identified in the unshaved control (1191_C in the purple box outline) and shaved samples (1191_T in the red
box outline). The settings for protein threshold, peptide threshold, and minimum number of peptides per
protein are shown in the green box outline. (b) The highlighted peptide (MLDTTVALMSAK) from the cytoplasmic
protein glyceraldehyde-3-phosphate dehydrogenase was identified in the unshaved control sample and is
removed from the shaved samples by unticking the “valid” box (black box outline) in the Proteins tab
Rapid Bacterial Surface Shaving to Identify Surface Antigens 55
4 Notes
Fig. 4 Topology of peptides identified by rapid surface shaving (in green) were mapped to their proteins using
Protter v1.0. (a-e) display 5 well-characterized B. pertussis autotransporter proteins that are either current
antigens found in the pertussis acellular vaccine, or proposed to be good candidates as future vaccine
antigens (A. Pertactin (Prn); B. Tracheal colonization factor A (TcfA); C. Bordetella resistance to killing A (BrkA);
D. Virulence associated gene 8 (Vag8); and E. Subtilisin (SphB1). For A-E, N-signal peptides are shown in red,
autotransporter passenger domains, which are known to be surface exposed, are colored purple while channel
domains, which are embedded into the membrane, are shown in grey. In (f), the Bordetella Bvg-intermediate
phase protein (BipA) is displayed. BipA is an outer membrane ligand binding protein and another proposed
potential vaccine antigen. The shaved peptides (green) from BipA are mapped to the predicted surface
exposed area from Phobius. The bacterial outer membrane is depicted as an orange bar and the numbers
1 and 2 are the predicted BipA transmembrane domains that anchor the protein to the outer membrane.
(Reprinted from Vaccine, 38 (3), Luu LDW, Octavia S, Aitken C, Zhong L, Raftery MJ, Sintchenko V and Lan R,
Surfaceome analysis of Australian epidemic Bordetella pertussis reveals potential vaccine antigens, 539–548,
2020, with permission from Elsevier)
105
104
Foward Scatter A
103
102
–102
Fig. 5 Flow cytometry dot plots of propidium iodide stained cells indicating intact
membrane integrity and minimal cell lysis from cells subjected to rapid surface
shaving protocol (purple dots). Unshaved control cells (incubated in the absence
of trypsin) are illustrated as pink dots, while the heated treated cells (positive
control for cell death) are shown as red dots. (Reprinted from Vaccine, 38 (3),
Luu LDW, Octavia S, Aitken C, Zhong L, Raftery MJ, Sintchenko V and Lan R,
Surfaceome analysis of Australian epidemic Bordetella pertussis reveals
potential vaccine antigens, 539–548, 2020, with permission from Elsevier)
24. For each centrifugation step, check that all liquid has flowed
through the StageTip. If there is remaining liquid, increase
centrifugal force by 100 g and centrifuge again until all liquid
has passed.
25. Centrifuge samples at maximum speed for 1 min prior to
loading onto the StageTips to remove precipitates which may
block liquid from passing through.
26. Keep the flowthrough from the load and wash steps in case
peptides have not bound and store it at 80 C. If after
LC-MS/MS of the C18-eluted samples (see Subheading 3.4,
step 9), no/minimal peptides are detected especially in the
“shaved” samples, this may indicate that the peptides did not
bind to the C18 StageTips. If this is the case, the flowthrough
can be reloaded into a new C18 StageTip that has been initi-
alized and reequilibrated (see Subheading 3.4, steps 3–5) and
cleaned up again.
60 Laurence Don Wai Luu and Ruiting Lan
References
1. Molloy MP, Herbert BR, Slade MB, application to complete genomes. J Mol Biol
Rabilloud T, Nouwens AS, Williams KL, Goo- 305(3):567–580
ley AA (2000) Proteomic analysis of the 11. K€all L, Krogh A, Sonnhammer EL (2004) A
Escherichia coli outer membrane. Eur J Bio- combined transmembrane topology and signal
chem 267(10):2871–2881 peptide prediction method. J Mol Biol 338
2. Rodrı́guez-Ortega MJ, Norais N, Bensi G, (5):1027–1036
Liberatori S, Capo S, Mora M, Scarselli M, 12. Omasits U, Ahrens CH, Müller S, Wollscheid B
Doro F, Ferrari G, Garaguso I (2006) Charac- (2013) Protter: interactive protein feature visu-
terization and identification of vaccine candi- alization and integration with experimental
date proteins through analysis of the group A proteomic data. Bioinformatics 30
Streptococcus surface proteome. Nat Biotechnol (6):884–886
24(2):191–197 13. Aramaki T, Blanc-Mathieu R, Endo H,
3. Walters MS, Mobley HL (2009) Identification Ohkubo K, Kanehisa M, Goto S, Ogata H
of uropathogenic Escherichia coli surface pro- (2020) KofamKOALA: KEGG ortholog
teins by shotgun proteomics. J Microbiol assignment based on profile HMM and adap-
Methods 78(2):131–135 tive score threshold. Bioinformatics 36
4. Olaya-Abril A, Jiménez-Munguı́a I, Gómez- (7):2251–2252
Gascón L, Rodrı́guez-Ortega MJ (2014) Sur- 14. Huerta-Cepas J, Forslund K, Coelho LP,
fomics: shaving live organisms for a fast pro- Szklarczyk D, Jensen LJ, Von Mering C, Bork
teomic identification of surface proteins. J P (2017) Fast genome-wide functional annota-
Proteome 97:164–176 tion through orthology assignment by
5. Solis N, Larsen MR, Cordwell SJ (2010) eggNOG-mapper. Mol Biol Evol 34
Improved accuracy of cell surface shaving pro- (8):2115–2122
teomics in Staphylococcus aureus using a false- 15. Sherman BT, Lempicki RA (2009) Systematic
positive control. Proteomics 10 and integrative analysis of large gene lists using
(10):2037–2049 DAVID bioinformatics resources. Nat Protoc 4
6. Luu LDW, Octavia S, Aitken C, Zhong L, Raf- (1):44
tery MJ, Sintchenko V, Lan R (2019) Surfa- 16. Vita R, Mahajan S, Overton JA, Dhanda SK,
ceome analysis of Australian epidemic Martini S, Cantrell JR, Wheeler DK, Sette A,
Bordetella pertussis reveals potential vaccine Peters B (2019) The immune epitope database
antigens. Vaccine 38(3):539–548 (IEDB): 2018 update. Nucleic Acids Res 47
7. Bark SKN, Ahmad R, Dantzler K, Lukens AK, (D1):D339–D343
De Niz M, Szucs MJ, Jin X, Cotton J, 17. Luo H, Lin Y, Gao F, Zhang C-T, Zhang R
Hoffmann D, Bric-Furlong E (2018) Quanti- (2014) DEG 10, an update of the database of
tative proteomic profiling reveals novel plasmo- essential genes that includes both protein-
dium falciparum surface antigens and possible coding genes and noncoding genomic ele-
vaccine candidates. Mol Cell Proteomics 17 ments. Nucleic Acids Res 42(D1):D574–D580
(1):43–60 18. Thalen M, van den IJssel J, Jiskoot W,
8. Nancy YY, Wagner JR, Laird MR, Melli G, Zomer B, Roholl P, de Gooijer C, Beuvery C,
Rey S, Lo R, Dao P, Sahinalp SC, Ester M, Tramper J (1999) Rational medium design for
Foster LJ (2010) PSORTb 3.0: improved pro- Bordetella pertussis: basic metabolism. J Bio-
tein subcellular localization prediction with technol 75(2):147–159
refined localization subcategories and predic- 19. Thalen M, van der Ark A, van den Ijssel J, van
tive capabilities for all prokaryotes. Bioinfor- Straaten I, Jansen D, Beuvery C, Martens D,
matics 26(13):1608–1615 Tramper J (2008) Improving the cellular per-
9. Armenteros JJA, Tsirigos KD, Sønderby CK, tussis vaccine: increased potency and consis-
Petersen TN, Winther O, Brunak S, von tency. Vaccine 26(5):653–663. https://doi.
Heijne G, Nielsen H (2019) SignalP 5.0 org/10.1016/j.vaccine.2007.11.047
improves signal peptide predictions using 20. Luu LDW, Octavia S, Zhong L, Raftery M,
deep neural networks. Nat Biotechnol 37 Sintchenko V, Lan R (2017) Characterisation
(4):420–423 of the Bordetella pertussis secretome under dif-
10. Krogh A, Larsson B, Von Heijne G, Sonnham- ferent media. J Proteome 158:43–51
mer EL (2001) Predicting transmembrane pro- 21. Luu LDW, Octavia S, Zhong L, Raftery MJ,
tein topology with a hidden Markov model: Sintchenko V, Lan R (2018) Proteomic
62 Laurence Don Wai Luu and Ruiting Lan
adaptation of Australian epidemic Bordetella 24. Paoletti AC, Parmely TJ, Tomomori-Sato C,
pertussis. Proteomics 18(8):1700237 Sato S, Zhu D, Conaway RC, Conaway JW,
22. Herigstad B, Hamilton M, Heersink J (2001) Florens L, Washburn MP (2006) Quantitative
How to optimize the drop plate method for proteomic analysis of distinct mammalian
enumerating bacteria. J Microbiol Methods 44 mediator complexes using normalized spectral
(2):121–129 abundance factors. Proc Natl Acad Sci U S A
23. Hoofnagle AN, Whiteaker JR, Carr SA, 103(50):18928–18933
Kuhn E, Liu T, Massoni SA, Thomas SN, 25. Keasey S, Pugh C, Tikhonov A, Chen G,
Townsend RR, Zimmerman LJ, Boja E Schweitzer B, Nalca A, Ulrich RG (2010) Pro-
(2016) Recommendations for the generation, teomic basis of the antibody response to mon-
quantification, storage, and handling of pep- keypox virus infection examined in
tides used for mass spectrometry–based assays. cynomolgus macaques and a comparison to
Clin Chem 62(1):48–69 human smallpox vaccination. PLoS One 5
(12):e15547
Chapter 5
Abstract
Antigen selection is a critical step in subunit vaccine design, especially if the goal is to identify antigens that
can be bound by neutralizing antibodies to prevent invasion of cells by intracellular bacteria. Here, we
describe a method involving two dimensional gel electrophoresis (2-DE) coupled with western blotting
(WB) and mass spectrometry (MS) to identify bacterial proteins that: (1) interact with the host target cell
proteins, and (2) are targeted by antibodies from sera from infected animals. Subsequent steps would be
performed to validate that the bacteria are targeted by neutralizing antibodies to prevent invasion of the
eukaryotic cells.
1 Introduction
63
64 Milan Obradovic and Heather L. Wilson
2 Materials
All reagents are analytical grade unless otherwise specified and solu-
tions were made using deionized water. All reagents, solutions and gels
were used at room temperature, unless specified otherwise. All steps
are performed in 1.5 mL Eppendorf tubes unless specified otherwise.
2.4 10% SDS 1. Sodium dodecyl sulfate (SDS) equilibration buffer 2 210 mL:
PAGE Gel 6 M urea, 75 mM Tris–HCl pH 8.8, 29.3% glycerol, 2% SDS,
0.002% bromophenol blue (can be stored at 20 C in 20 mL
aliquots).
2. DTT, 100 mg per 210 mL equilibration buffer (see Note 4).
3. 2.5% iodoacetamide, 250 mg per 210 mL equilibration buffer
(see Note 4).
4. Resolving gel buffer (total 25.32 mL for two gels; see Note 5):
25.32 mL double-distilled water (ddH2O), 21.36 mL 30%
acrylamide mix, 16.04 mL 1.5 M Tris pH 8.8, 0.64 mL 10%
SDS, 0.64 mL 10% ammonium persulphate (APS), and
25.6 μL tetramethylethylenediamine (TEMED).
5. Silver stain kit.
3 Methods
13. For analytical gels, add 250 μg IPEC + Cy-5 labeled L. intra-
cellularis proteins to rehydration buffer up to the 250 μL
volume. For MS preparatory gels, add 600 μg of L. intracellu-
laris proteins in rehydration buffer up to the 250 μL volume.
3.2 Rehydration 1. Pipet 250 μL of protein sample in rehydration buffer into one
of Two 13 Cm pH 4–7 lane of the Ettan IPGphor rehydration strip apparatus. Slowly
IPG Strips (See Note 9) discharge the sample into the rack lane about the same length
of the strip. Avoid creating bubbles (see Note 10).
2. Remove strip from a 20 C freezer.
3. Carefully peel off the protective backing from the strip.
4. Gently place the strip (gel side down) onto the sample, being
careful not to create bubbles and ensuring the entire strip is in
contact with the sample.
5. Cover strip with 3–4 mL of Dry Strip Cover fluid.
6. Place the lid on a rack and leave on a level bench for at least 10 h
or overnight, in the dark.
3.3 IEF of Rehydrated 1. Place strip in a strip holder, gel side up—positive end of gel at
Strips Using IPGphor the pointed end of the holder and negative end all the way to
the flat side.
2. Place 1–1.5 cm long paper strips (presoaked with water and
blotted almost completely dry) on each end of the strip ensur-
ing some overlap with the gel.
3. Place electrodes on the ends of a filter paper not in contact with
the gel.
4. Completely cover with Dry Strip Cover fluid (around 4.5 mL).
5. Ensure that pointed ends of holders (with the positive end of
the strip) are in the positive area of the apparatus.
6. Run focusing: 150 V step and hold for 3 h.
300 V step and hold for 1200 Volt hours (Vh).
1000 V gradient for 3900 Vh.
8000 V gradient for 13,500 Vh.
8000 V step and hold for 25,000 Vh.
7. After focusing, strips are placed in a tube, gel side up, and
stored at 80 C until ready for the next step (see Note 11).
3.4 10% SDS-PAGE 1. Run the second dimension on a medium size 10% SDS-PAGE
Gel, Using Bio-Rad gel, using Bio-Rad PROTEAN II xi cell apparatus connected to
Protean II Xi Cell a circulating water bath to maintain temperature at around
Apparatus 10 C.
2. Thaw the required number of strips at room temperature
(RT) for 15–30 min.
68 Milan Obradovic and Heather L. Wilson
3.7 Excising Gel 1. Excise gel with silver-stained dots which correspond to IR-800
Plugs for Mass labeled proteins detected by WB analysis using a sterile biopsy
Spectrometry punch (3 mm diameter) in a biological cabinet to avoid con-
tamination of gel samples with environmental proteins
(Fig. 1b).
2. Collect gel plugs and store them in Eppendorf tube with
100 μL ultrapure water at 20 C. Send gel plug samples for
MS analysis (see Notes 17 and 18).
70 Milan Obradovic and Heather L. Wilson
4 Notes
14. Shorter time could be used but with higher voltage such as
25 V for 30 min. The time and voltage should be established
based on the size of the gel and semidry apparatus protocol.
15. From our experience, hyperimmune rabbit serum in 5% milk in
TBST could be used again up to 4 times if stored at 20 C.
16. There are multiple image software and projectors available for
processing of immunoblot image and correlation of protein
spots to the preparative gel. This is useful if larger and complex
proteome is analyzed. Although we did not have this equip-
ment, we performed selection and Cy5 staining of proteins (see
Subheading 3.1), thus reducing the number of proteins on the
membrane and increasing their detection. We were able to
compare the printed image of the immunoblot to the silver
stained gel and select gel plugs for MS with great accuracy.
17. Gel plug samples (annotated as 1.4, 2.3, 3.1, 3.2, and
4 (Fig. 1b)) were sent to Plateforme Protéomique Centre de
Recherche du CHU de Québec CHUL, Québec, Canada for
MS analysis.
Acknowledgments
References
1. Rabilloud T, Strub JM et al (2001) A compari- immunogenic proteins. Electrophoresis 20
son between Sypro ruby and ruthenium II tris (4–5):1001–1010
(bathophenanthroline disulfonate) as fluores- 6. Haas G, Karaali G et al (2002) Immunopro-
cent stains for protein detection in gels. Prote- teomics of Helicobacter pylori infection and
omics 1(5):699–704 relation to gastric disease. Proteomics 2
2. Lahner E, Bernardini G et al (2011) Immuno- (3):313–324
proteomics of Helicobacter pylori infection in 7. Hagan EC, Mobley HLT (2007) Uropatho-
patients with atrophic body gastritis, a predis- genic Escherichia coli outer membrane antigens
posing condition for gastric cancer. Int J Med expressed during urinary tract infection. Infect
Microbiol 301(2):125–132 Immun 75(8):3941–3949
3. Havlasova J, Hernychova L et al (2005) Pro- 8. Enany S, Yoshida Y et al (2013) Two dimen-
teomic analysis of anti-Francisella tularensis sional electrophoresis of the exo-proteome
LVS antibody response in murine model of produced from community acquired methicil-
tularemia. Proteomics 5(8):2090–2103 lin resistant Staphylococcus aureus belonging to
4. Kellner R, Lichtenfels R et al (2002) Targeting clonal complex 80. Microbiol Res 168
of tumor associated antigens in renal cell carci- (8):504–511
noma using proteome-based analysis and their 9. Salzano AM, Novi G et al (2013) Mono-
clinical significance. Proteomics 2 dimensional blue native-PAGE and
(12):1743–1751 bi-dimensional blue native/urea-PAGE
5. Pitarch A, Pardo M et al (1999) or/SDS-PAGE combined with nLC-ESI-LIT-
Two-dimensional gel electrophoresis as analyt- MS/MS unveil membrane protein heteromeric
ical tool for identifying Candida albicans
Identification of Lawsonia intracellularis Neutralizing Antibody Targets. . . 73
and homomeric complexes in Streptococcus 11. Obradovic MR, Wilson HL (2020) Immune
thermophilus. J Proteome 94:240–261 response and protection against Lawsonia
10. Obradovic M, Pasternak JA et al (2019) Immu- intracellularis infections in pigs. Vet Immunol
noproteomic analysis of Lawsonia intracellu- Immunopathol 219:109959
laris identifies candidate neutralizing antibody 12. Lawson GH, McOrist S et al (1993) Intracel-
targets for use in subunit vaccine development. lular bacteria of porcine proliferative enteropa-
Vet Microbiol 235:270–279 thy: cultivation and maintenance in vitro. J Clin
Microbiol 31(5):1136–1142
Chapter 6
Abstract
High-density protein microarray is an established technology for characterizing host antibody profiles
against entire pathogen proteomes. As one of the highest throughput technologies for antigen discovery,
proteome microarrays are a translational research tool for identification of vaccine candidates and biomar-
kers of susceptibility or protection from microbial challenge. The application has been expanded in recent
years due to increased availability of bacterial genomic sequences for a broader range of species and strain
diversity. Panproteome microarrays now allow for fine characterization of antibody specificity and cross-
reactivity that may be relevant to vaccine design and biomarker discovery, as well as a fuller understanding of
factors underlying themes of bacterial evolution and host–pathogen interactions. In this chapter, we present
a workflow for design of panproteome microarrays and demonstrate statistical analysis of panproteomic
human antibody responses to bacterial vaccination and challenge. Focus is particularly drawn to the
bioinformatics and statistical tools and providing nontrivial, real examples that may help foster hypotheses
and rational design of panproteomic studies.
Key words Protein microarray, Panproteome, Antigenic diversity, Antibody profiling, Differential
analysis
1 Introduction
75
76 Joseph J. Campo and Amit Oberai
Fig. 1 Proteome microarray development and antibody profiling workflow (reproduced from https://
antigendiscovery.com/adi-proteome-microarray-technology/ with permission from Antigen Discovery, Inc.)
Fig. 2 Selection of S. pneumoniae genes for inclusion in panproteome microarray. Among 616 sequenced
pediatric clinical isolates, genes present in at least 20% of the bacterial population were included in an array
containing 4296 proteins
2 Materials
3 Methods
3.1 Select Pathogen Identify an organism or set of organisms from which to perform
genome-wide homology analysis prior to designing the contents of
a panproteome microarray. Base the selection of organisms on the
research questions, but balance factors such as limitation of array
space (i.e., number of proteins), and quality and source of genomic
information. See Note 7 for considerations in selecting organisms.
3.2 Identify The goal is to capture species or strain diversity by identifying the
Conserved and Diverse panproteome, or the highly diverse proteins, for example those
Proteins under immune pressure. The steps involved in capturing the pan-
proteome are as follows, using an example of 3 selected species:
1. Download whole proteome sequences for the group of species
or strains of the pathogens of interest that will constitute the
panproteome microarray. The fasta format files “cds_from_ge-
nomic.fna” and “translated_cds.faa” (nucleotide and protein
files, respectively) are downloaded from NCBI Genbank or
Refseq, or they may be obtained from private curated whole
genome sequencing data (see Note 8).
2. Run BLASTp for all vs. all sequence homology comparison for
the selected species or strains. The stand-alone BLAST is run in
the windows command prompt (cmd) application. The first
step in running BLASTp is to format each of the proteome
fasta files for BLAST using the “makeblastdb” command. For
comparing 3 species A, B, and C, with proteome sequence files
A_translated_cds.faa, B_translated_cds.faa, and C_transla-
ted_cds.faa, the makeblast db command would be as follows:
3.3 Classify Proteins Classify proteins by intersections between groups, here using the
in the Core 3 species example homology analysis in Subheading 3.2.
and Panproteome
1. The proteins that intersect in groups A-B-C, A-B-C and A-B-C
are the protein sequences that are common to all species. This
group of proteins forms the core proteome. Define vectors
group_abc.vec as the vector containing proteins sequence
IDs from proteins in group A-B-C, group_bac.vec as the
vector containing protein sequence IDs from proteins in group
A-B-C, and group_cab.vec as vector containing the protein
sequence IDs from proteins in group A-B-C, as follows:
Table 1
Brucella species homology analysis for classification of the core proteome and panproteome
Panproteome Analysis of Human Antibody Responses 85
Fig. 3 Defining variants for the four core variable Streptococcus pneumoniae antigens. Each histogram shows
the distribution of pairwise similarities between representatives of (a) PspA, (b) PspC, (c) ZmpA, and (d) ZmpB.
The vertical red lines on each plot represent the thresholds that were used as a cut-off to define distinct
variants: 0.525 for PspA; 0.675 for PspC; 0.575 for ZmpA, and 0.475 for ZmpB (reproduced from [4] with
permission from PNAS)
Table 2
T. pallidum subsp. pallidum (T.p.p.) Nichols and SS14 strain homology analysis for classification of
the core proteome and panproteome
Table 3
Clostridium species homology analysis for classification of the core proteome (yellow highlighted for
3 or more species) and panproteome
Fig. 4 Distribution of percent identity between Clostridium species. The histograms show for pairwise BLASTp
results for (a) C. difficile vs. C. perfringens and (b) C. scindens vs. P. bifermentans showing that most genes
are poorly conserved between species
3.5 Statistical Typical methods for univariate differential reactivity analysis have
Analysis been reported extensively for proteome microarrays (see https://
of the Panproteome antigendiscovery.com/publications/), which also apply for pan-
Antibody Response proteome analyses. Examples are given from our studies of panpro-
to Vaccines teome antibody responses to a S. pneumoniae whole cell vaccine,
and Challenge showing responses to both the core proteome and diverse core
loci [5].
– To investigate homology between whole cell vaccine strains and
proteins present on the panproteome microarray, follow the
methods described in Subheading 3.2, steps 1–6 to perform
pairwise BLASTp using amino acid sequences of each protein on
the array as queries against the vaccine strain genome as the
subject. This provides a distribution of homology (% sequence
identity) or, conversely, divergence (100 – (% sequence iden-
tity)) from the vaccine strain (Fig. 5).
– To investigate panproteomic immunogenicity profiles, differen-
tial reactivity analysis can be performed using standard para-
metric and nonparametric methods such as t-tests, Wilcoxon’s
rank-sum tests, empirical Bayes models or multivariable linear
models, as appropriate for the study design. Example code for
performing empirical Bayes moderated t-tests on array data and
example output in a study of immunogenicity of a S. pneumoniae
whole cell vaccine (Fig. 6):
> library(limma)
> # sample.classes is a vector of classes for group comparison
> # int.data.df is a data frame containing the matrix of data
with rows as proteins and columns as samples
> trt <- as.factor(sample.classes)
> design <- model.matrix(~trt) # Create a design matrix to fit
limma model
> model <- lmFit(int.data.df, design = design) # Fit ANOVA
model and build contrasts
> result <- eBayes(model)
> ebayes <- topTable(result, coef = 2, number = nrow(int.data.
df),
90 Joseph J. Campo and Amit Oberai
Fig. 5 Distribution of percent identity between S. pneumoniae panproteome microarray proteins and the
S. pneumoniae whole cell vaccine strain RM200 showing high level sequence identity for most proteins
constituting the core proteome and greater diversity for a subset of proteins on the microarray (reproduced
from [5] under a CC-BY license)
Fig. 6 Volcano plot showing the statistical and biological significance of changes in IgG binding following
S. pneumoniae whole cell vaccination, showing increase in antibodies targeting diverse proteins in the
panproteome. Points corresponding to variants of diverse core loci PspA, PspC, ZmpA, or ZmpB are colored
in purple, blue, green and orange, respectively (reproduced from [5] under a CC-BY license)
genelist = row.names(int.data.df),
sort.by = "none", p.value = 1.1)
Fig. 7 Changes in IgG binding to S. pneumoniae diverse core loci following vaccination with a whole cell
vaccine. The variants with higher similarity to the RM200 vaccine strain tended to have greater immunoge-
nicity for ZmpA, showing a strain-transcendent immune response (reproduced from [5] under a CC-BY license)
4 Notes
References
1. Driguez P, Doolan DL, Molina DM et al 6. Yu NY, Wagner JR, Laird MR et al (2010)
(2015) Protein microarrays for parasite antigen PSORTb 3.0: improved protein subcellular
discovery. Methods Mol Biol 1201:221–233. localization prediction with refined localization
https://doi.org/10.1007/978-1-4939-1438- subcategories and predictive capabilities for all
8_13 prokaryotes. Bioinformatics 26:1608–1615.
2. Köser CU, Ellington MJ, Cartwright EJP et al https://doi.org/10.1093/bioinformatics/
(2012) Routine use of microbial whole btq249
genome sequencing in diagnostic and public 7. Krogh A, Larsson B, von Heijne G, Sonnham-
health microbiology. PLoS Pathog 8: mer EL (2001) Predicting transmembrane pro-
e1002824. https://doi.org/10.1371/journal. tein topology with a hidden Markov model:
ppat.1002824 application to complete genomes. J Mol Biol
3. Croucher NJ, Finkelstein JA, Pelton SI et al 305:567–580. https://doi.org/10.1006/
(2013) Population genomics of post-vaccine jmbi.2000.4315
changes in pneumococcal epidemiology. Nat 8. Marchler-Bauer A, Derbyshire MK, Gonzales
Genet 45:656–663. https://doi.org/10. NR et al (2015) CDD: NCBI’s conserved
1038/ng.2625 domain database. Nucleic Acids Res 43:
4. Croucher NJ, Campo JJ, Le TQ et al (2017) D222–D226. https://doi.org/10.1093/nar/
Diverse evolutionary patterns of pneumococcal gku1221
antigens identified by pangenome-wide immu- 9. Smyth GK (2004) Linear models and empirical
nological screening. Proc Natl Acad Sci 114: bayes methods for assessing differential expres-
E357–E366. https://doi.org/10.1073/pnas. sion in microarray experiments. Stat Appl
1613937114 Genet Mol Biol 3:1–69
5. Campo JJ, Le TQ, Pablo JV et al (2018) 10. van der Maaten L, Hinton G (2008) Visualiz-
Panproteome-wide analysis of antibody ing high-dimensional data using t-SNE. J
responses to whole cell pneumococcal vaccina- Mach Learn Res 9:2579–2605
tion. eLife 7:e37015. https://doi.org/10. 11. Benjamini Y, Hochberg Y (1995) Controlling
7554/eLife.37015 the false discovery rate: a practical and powerful
96 Joseph J. Campo and Amit Oberai
approach to multiple testing. J Royal Stat Soc 13. Arora N, Schuenemann VJ, J€ager G et al
Ser B 57:289–300 (2016) Origin of modern syphilis and emer-
12. Pětrošová H, Pospı́šilová P, Strouhal M et al gence of a pandemic Treponema pallidum clus-
(2013) Resequencing of Treponema pallidum ter. Nat Microbiol 2:16245. https://doi.org/
ssp. pallidum strains Nichols and SS14: correc- 10.1038/nmicrobiol.2016.245
tion of sequencing errors resulted in increased 14. Camponovo F, Campo JJ, Le TQ et al (2020)
separation of syphilis Treponeme subclusters. Proteome-wide analysis of a malaria vaccine
PLoS One 8:e74319. https://doi.org/10. study reveals personalized humoral immune
1371/journal.pone.0074319 profiles in Tanzanian adults. Elife 9:e53080.
https://doi.org/10.7554/eLife.53080
Chapter 7
Abstract
Vaccines consisting of whole inactivated bacteria (bacterins) are generated by incubation of the pathogen
with chemicals. This is a time-consuming procedure which may lead to less immunogenic material, as
critical antigenic structures can be altered by chemical modification. A promising alternative approach is
low-energy electron irradiation (LEEI). Like other types of ionizing radiation, it mainly acts by destroying
nucleic acids but causes less damage to structural components like proteins. As the electrons have a limited
penetration depth, LEEI is currently used for sterilization of surfaces. The inactivation of pathogens in
liquids requires irradiation of the culture in a thin film to ensure complete penetration. Here, we describe
two approaches for the irradiation of bacterial suspensions in a research scale. After confirmation of
inactivation, the material can be directly used for vaccination, without any purification steps.
Key words Bacterial inactivation, Low-energy electron irradiation, LEEI, Bacterial vaccine, Bacterin,
Electron beam
1 Introduction
97
98 Jasmin Fertey et al.
https://www.ebeamtechnologies.com/en/products). However,
they are currently not suited for irradiating liquids and the tech-
nique is mainly used for surface sterilization. The major challenge
for LEEI in inactivating liquid solutions is the limited penetration
depth of the electrons. To overcome this limitation, the liquid has
to be irradiated in a thin film, and manufacturing of larger volumes
in a multiliter scale with this technique is challenging. With the
currently available commercial devices specified for surface sterili-
zation, this can be achieved by using Petri dishes, followed by
covering the liquid with a piece of polypropylene foil [13, 14]. How-
ever, amounts that are required for larger study groups or larger
animals are difficult to produce with this small-scale approach.
Further development of irradiation technologies for larger volumes
is therefore required.
We have recently described the development of the first
research-scale prototype for LEEI of liquids in higher throughput
[8], and this development will form the basis for commercially
available devices. We have also shown that LEEI can be used to
generate efficient vaccines against viruses, parasites, and gram-neg-
ative bacteria [13, 15, 16]. In this chapter, we demonstrate both
small-scale (see Subheading 3.2) and large-scale (see Subheading
3.3) methods to generate LEEI-inactivated bacterial vaccines and
to evaluate their antigenic components (see Subheading 3.4) before
administration into animals. As an example, we chose the gram-
negative bacterium Rodentibacter pneumotropicus to describe the
procedures for small-scale and larger-scale irradiation experiments.
R. pneumotropicus belongs to the family Pasteurellaceae and is
related to other human and veterinary pathogens such as Haemo-
philus influenzae, Actinobacillus pleuropneumoniae, and Pasteurella
multocida. It is an opportunistic pathogen commonly found in the
microbiome of the respiratory tract of mice and rats. Primary or
secondary infections can result in severe diseases, and make
R. pneumotropicus one of the most prevalent infectious agents in
laboratory rodents [17].
2 Materials
2.1 Buffers, 1. Brain Heart Infusion broth: 37.0 g per 1 L distilled water.
Reagents and Media Autoclave and store at 4 C (see Note 1).
2. Phosphate buffered saline (PBS), 10 stock: 1.37 M NaCl,
270 mM KCl, 100 mM Na2PO4, 20 mM KH2PO4 pH 7.4 (see
Note 2).
100 Jasmin Fertey et al.
2.3 Specialized 1. Electron irradiation device (Fig. 1a) with module for auto-
Equipment mated bag transport (Fig. 2), for bag irradiation experiments.
Bacterial Inactivation by Low-Energy Electron Irradiation 101
Fig. 1 (a) Photograph of the custom-built LEEI device. The picture shows the closed device with the electron
beam source and the leaden irradiation chamber in the middle. Feed lines with flexible hoses for the in- and
outlet of liquids (e.g., for cooling inside the irradiation chamber) are guided through the metal box followed by
baffles to shield the environment from occurring Bremsstrahlung. The movable module holder can be rolled
out of the irradiation chamber and carries the modules (shown in Fig. 3) for liquid handling. The irradiation
chamber was constructed as a research-scale prototype and different experimental modules designed for
liquid handling can be inserted completely [8]. This increases the overall footprint of the electron beam device.
(b) Photograph of the commercially available EB-Lab 200 (https://www.ebeamtechnologies.com/en/eblab).
The picture shows the opened chamber, where the sample holder for Petri plates is placed, and the cover is
closed before irradiation. The sample is transported to the left for irradiation where the electron beam source
is located
3 Methods
3.1 Preparing the 1. Inoculate bacteria in 50 mL growth medium and incubate over
Bacterial Solution for night at 37 C, 180 rpm (see Note 10).
Irradiation 2. Harvest bacteria by centrifugation (3452 g, 4 C, 10 min)
(see Note 11).
102 Jasmin Fertey et al.
Bag with
pathogens
in solution
Conveyor Belts
Fig. 2 Schematic drawing of the module. The disposable bag containing the
pathogen solution is placed on the left side between the two conveyor belts
(marked by the red line). The roller transports the bag through the irradiation
zone (marked by radiation warning sign). The bag is stretched by the braking
roller to limit the liquid height and to enable the low-energy electrons to
penetrate the bag completely. The bag with the irradiated solution (marked in
green) is transported to the right and can be taken out of the module after the
irradiation is completed
Fig. 3 (a) Sample preparation for LEEI in a small-scale experimental setup. 230 μL of bacteria are pipetted to
the center of a Petri dish and covered with a piece of round, sterilized oriented polypropylene (OPP)-foil
forming the thin liquid film. (b) Photograph of sample holder with prepared Petri dishes, placed in the center of
the sample holder. The picture shows the sample holder already inserted into the irradiation device
3.2 Irradiation in a 1. Take the sample holder out of the irradiation device and put
Small-Scale into a biological safety cabinet (BSC). Discard lids from the
Experimental Setup Petri dishes, as they are not needed anymore. A maximum of six
dishes can be placed on the sample holder and processed at the
same time.
2. For each sample, pipet 230 μL of washed bacteria to the center
of a Petri dish and cover the drop with a piece of sterilized
OPP-foil (Fig. 3a). Be careful to avoid bubbles during pipet-
ting and spreading the liquid, since they might affect the height
of the liquid film (see Note 16).
3. Put the Petri dishes in the center of the sample holder and
insert the sample holder in the device (Fig. 3b) (see Note 17).
4. Close the cover of the irradiation chamber, set the desired
parameters and start the irradiation process (see Note 18).
5. Take the sample holder out of the irradiation device and put it
back into the BSC.
6. Recover the liquid from Petri dishes by carefully tapping the
edge of the dish on the surface of the BSC and collecting the
liquid that runs down to the edge using a pipet. Transfer the
sample to a sterile 1.5 mL tube.
7. For inactivation testing, inoculate 10% of the recovered sample
into fresh growth medium and incubate at 37 C overnight.
Include a nonirradiated sample as positive control for growth
(see Note 19).
8. Save 10–20% of the recovered sample for checking the antige-
nicity (see Subheading 3.4). Ensure to save the same amount of
the nonirradiated sample, since this will be the reference for the
antigenicity testing.
9. Store the residual sample at 80 C until use.
104 Jasmin Fertey et al.
12 cm
15 cm
10 cm
39 cm 65 cm
11 cm
3.3 Irradiation 1. Prepare irradiation bags by placing two layers of PET/PE film
in Bags carefully over one another. Seal the long (65 cm) sides with the
80–100 cm impulse sealer, taking care that the two layers are
properly aligned on top of each other. Then seal the bottom of
the bag, creating the seam about 10–11 cm from the bottom
edge (Fig. 4), using the 30 cm impulse sealer (see Note 7).
Make sure that all three sides are completely sealed (Figs. 4 and
5) and that the sealing seams are intact to ensure that no
infectious content spills out (see Note 20). Leave the top of
the bag open for filling (Fig. 5).
2. In a biological safety cabinet (BSC), fill 20 mL of washed
bacteria in PBS or other buffer into a prepared irradiation bag
using a 10 mL or 20 mL pipet and a pipetting aid (Fig. 5). Use
one bag per irradiation dose to be tested and include an extra
Bacterial Inactivation by Low-Energy Electron Irradiation 105
sealed seam
(both sides)
sealed seam
(top)
removal of
excessive air
sealed seam
(bottom)
Fig. 5 sample preparation for LEEI in disposable bags. 20 mL of washed bacteria are transferred to PET/PE-
bags (see Fig. 1 for dimensions). Air is removed from the bag and the top is sealed using an impulse sealing
machine
3.4 Determining the 1. In a BSC, resuspend pelleted bacteria in the initially noted
Antigenicity of the volume of buffer (PBS or coating buffer).
Irradiated Sample by 2. Always include the nonirradiated sample as positive control for
ELISA the ELISA experiment. Make sure that the nonirradiated sam-
ple is diluted in the same way as the irradiated sample. Include
the buffer (e.g., PBS) used for resuspending the bacteria before
irradiation in the experiment as background negative control
(see Note 25).
3. Dilute samples in coating buffer and transfer 100 μL of each
dilution per well in duplicates or triplicates to the ELISA plate.
Usually, 1–5 μL of sample (prepared from washed bacteria with
an OD600 of 1–2) in 100 μL coating buffer per well are suffi-
cient for obtaining optimal signals (see Note 26). Incubate the
plate on a shaker over night at 4 C.
4. On the next day, wash the plate 3 times with 200 μL per well
PBS-T (see Note 27).
Bacterial Inactivation by Low-Energy Electron Irradiation 107
4 Notes
time, only the amount needed for each assay run should be
prepared.
5. We recommend using polyethylene terephthalate/polyethyl-
ene (PET/PE) material for the bags and oriented polypropyl-
ene (OPP) for the round plastic-foils for irradiation [18]. In
general, PET/PE and round pieces of OPP foil are available at
specialized packaging companies that provide packaging solu-
tions for medical or food products (e.g., sicht-pack HAGNER
GmbH or tbs-pack GmbH, both in Germany). We recommend
ordering PET/PE film rolls with the intended bag width (i.e.,
12 cm), and enough length to generate the desired number of
bags, each 65 cm long (Fig. 4).
6. For the small-scale irradiation experiments, it is crucial to avoid
shadowing effects during irradiation, otherwise there is the
chance that the sample is not evenly irradiated. Therefore, the
OPP foil must have a smaller diameter than the Petri plates. It is
recommended to sterilize the round OPP-foils before use by
putting them into 70% ethanol for 15 min, then allow to dry on
paper towels in a BSC. This treatment also removes any nonor-
ganic contaminations.
7. It is possible to use the 80–100 cm impulse sealer for all sides.
However, we prefer to have a smaller (30 cm) impulse sealer in
the BSC to directly seal the bags after filling. This makes the
disinfection (by spraying or wiping off the bag) much easier
because no infectious content spills out.
8. We tried different Petri dishes and identified Corning® Pri-
maria™ to be the best choice. The very hydrophilic surface
treatment ensures that the liquid is evenly distributed under the
OPP-foil, providing a homogenous liquid layer. In untreated
Petri dishes the liquid stays more or less as a drop on the
surface, making it difficult to achieve a liquid layer with an
even height. We therefore recommend using specially treated
Petri dishes (comparable to Corning® Primaria™).
9. In most cases, Nunc PolySorp® surface coated ELISA plates
worked well for coating the irradiated samples; however, there
have been some exceptions where other surfaces performed
better. If weak ELISA-signals are observed due to insufficient
coating, checking other plates (such as Nunc MaxiSorp® or
MultiSorp®) might improve the results.
10. We have also performed experiments with other bacteria, for
example, Escherichia coli and Bacillus cereus. We usually store
the bacteria at 80 C as cryostocks that can be used directly
for inoculation. We observe optimal growth if the bacteria are
precultured in a lower volume, such as 5 mL, at 37 C,
180 rpm on the day before the inoculation for the experiment
and use 0.5 mL of this culture for inoculation of the final
culture.
Bacterial Inactivation by Low-Energy Electron Irradiation 109
16. Ensure that the liquid sample is in the center of the dish.
Otherwise, shadowing effects from the side of the Petri dish
might lead to incomplete inactivation.
17. Regardless of the number of Petri dishes that are used for each
irradiation run, always ensure that they are in the center of the
sample holder (Fig. 4b). If they are placed at the side, shadow-
ing effects may occur, and the sample is not completely
irradiated.
18. The small-scale setup is optimal for determining the required
inactivation dose and for measuring the antigenicity of the
inactivated sample. It can be used as a small-scale test before
the actual experiment. In this case, several doses should be
tested, and CFU/mL should be determined afterward in addi-
tion to liquid culture for checking the inactivation kinetics. In
this way, a killing curve and the minimal dose required for
inactivation can efficiently be established.
19. The nonirradiated control consists of an extra Petri dish with
sample that is treated in the same way (covering with foil) like
the samples, except without the irradiation process. The sam-
ples can stay in the BSC, while the other samples are irradiated.
20. Make sure that the sealer is not too hot and the seams are
tightly sealed.
21. Removing air is crucial for processing of the bag and efficient
inactivation. There should be as little air as possible in the
closed bag. During irradiation, degassing of the buffer solution
might be observed, in this case air bubbles will be visible in the
irradiated bag. It is crucial that the thin liquid film that is
generated by stretching the bag between two rollers (Fig. 2)
is less than 150 μm thick to ensure complete penetration of the
electrons to the bottom part of the bag. It is recommended
that extra sealed seams (i.e., more than one) are added on the
top of the bag to ensure that infectious content is not released
when the bag is squeezed.
22. Depending on the velocity with which the bag is transported,
the irradiation process takes approx. 2–5 min per bag. Since the
applied dose is a product of applied energy and velocity of the
sample, these parameters have to be determined empirically
and controlled using commercially available, adjusted dosime-
ter films (e.g., Risø B3, Risø High Dose Reference Laboratory,
Denmark). For commercially available irradiation devices, as
used for the small-scale experiments, the dose application is
precalibrated and usually no dosimetry is needed.
23. Cooling the irradiated solution might help to slow down resid-
ual hydrolysis reactions in the liquid after irradiation and
improve antigenicity due to less formation of free radicals.
Bacterial Inactivation by Low-Energy Electron Irradiation 111
References
1. Hoelzer K, Bielke L, Blake DP et al (2018) 8. Fertey J, Thoma M, Beckmann J et al (2020)
Vaccines as alternatives to antibiotics for food Automated application of low energy electron
producing animals. Part 1: challenges and irradiation enables inactivation of pathogen-
needs. Vet Res 49:1–10. https://doi.org/10. and cell-containing liquids in biomedical
1186/s13567-018-0560-8 research and production facilities. Sci Rep
2. Babb R, Chen A, Hirst TR et al (2016) Intra- 10:12786. https://doi.org/10.1038/
nasal vaccination with γ-irradiated Streptococcus s41598-020-69347-7
pneumoniae whole-cell vaccine provides 9. Espinosa AC, Jesudhasan P, Arredondo R et al
serotype-independent protection mediated by (2012) Quantifying the reduction in potential
B-cells and innate IL-17 responses. Clin Sci health risks by determining the sensitivity of
130:697–710. https://doi.org/10.1042/ poliovirus type 1 chat strain and rotavirus
CS20150699 SA-11 to electron beam irradiation of iceberg
3. Bordin AI, Pillai SD, Brake C et al (2014) lettuce and spinach. Appl Environ Microbiol
Immunogenicity of an electron beam inacti- 78:988–993. https://doi.org/10.1128/
vated Rhodococcus equi vaccine in neonatal AEM.06927-11
foals. PLoS One 9:e105367. https://doi.org/ 10. Skowron K, Paluszak Z, Olszewska H et al
10.1371/journal.pone.0105367 (2014) Effectiveness of high energy electron
4. Jwa MY, Jeong S, Ko EB et al (2018) Gamma- beam against spore forming bacteria and
irradiation of Streptococcus pneumoniae for the viruses in slurry. Radiat Phys Chem
use as an immunogenic whole cell vaccine. J 101:36–40. https://doi.org/10.1016/j.
Microbiol 56:579–585. https://doi.org/10. radphyschem.2014.04.001
1007/s12275-018-8347-1 11. IAEA Safety standards (2010) Radiation safety
5. Alizadeh E, Orlando TM, Sanche L (2015) of gamma, electron and x-ray irradiation facil-
Biomolecular damage induced by ionizing ities. IAEA Safety standards series. https://
radiation: the direct and indirect effects of www-pub.iaea.org/MTCD/publications/
low-energy electrons on DNA. Annu Rev PDF/Pub1454_web.pdf
Phys Chem 66:379–398. https://doi.org/10. 12. Silindir M, Özer AY (2009) Sterilization meth-
1146/annurev-physchem-040513-103605 ods and the comparison of E-beam sterilization
6. Brown F (1995) Formaldehyde as an inacti- with gamma radiation sterilization. FABAD J
vant. Vaccine 13:231. https://doi.org/10. Pharm Sci 34:43–53
1016/0264-410x(95)93142-v 13. Fertey J, Bayer L, Grunwald T et al (2016)
7. Delrue I, Verzele D, Madder A et al (2012) Pathogens inactivated by low-energy-electron
Inactivated virus vaccines from chemistry to irradiation maintain antigenic properties and
prophylaxis: merits, risks and challenges. induce protective immune responses. Viruses
Expert Rev Vaccines 11:695–719. https:// 8:319. https://doi.org/10.3390/v8110319
doi.org/10.1586/erv.12.38 14. Bayer L, Fertey J, Ulbert S et al (2018) Immu-
nization with an adjuvanted low-energy
Bacterial Inactivation by Low-Energy Electron Irradiation 113
Abstract
Metal ion transporters in the outer membrane of gram-negative bacteria that are responsible for acquiring
iron and zinc are attractive vaccine targets due to their essential function. The core function is mediated by
an integral outer membrane TonB-dependent transporter (TBDT) that mediates the transport of the metal
ion across the outer membrane. Some TBDTs also have a surface lipoprotein (SLP) that assists in the
efficient capture of the metal ion-containing host protein from which the metal ion is extracted. The
challenges in producing the integral outer membrane protein for a commercial subunit vaccine prompted us
to develop a hybrid antigen strategy in which surface loops of the TBDT are displayed on the lipoprotein,
which can readily be produced as a soluble protein. The focus of this chapter will be on the methods for
production of hybrid antigens and evaluating the immune response they elicit.
Key words Hybrid antigens, TonB-dependent transporter, Surface lipoprotein, Outer membrane
proteins, ELISA, Transferrin binding proteins
1 Introduction
115
116 Somshukla Chaudhuri et al.
Fig. 1 TonB-dependent transporters (TBDTs) such as transferrin binding protein A (TbpA) and surface
lipoproteins (SLP) such as TbpB both have loops (red) that are anchored by antiparallel beta-strands
(green). Loops on the soluble SLP can be replaced with loops from TBDTs to generate novel hybrid antigens
2 Materials
2.3 Stock Solutions 1. 1 M Tris pH 8.0: Weigh out 242.28 g of Tris and transfer to a
of Components large beaker. Add 1400 mL of distilled water, dissolve solids
of Protein Purification with agitation, then transfer solution to a graduated cylinder
Buffers and make up volume to ~1950 mL to leave room for the
120 Somshukla Chaudhuri et al.
2.6 Materials 1. DNA and protein sequences of the SLP scaffold and the TBDT.
for Hybrid Antigen 2. Access to programs/software for protein modeling, protein
Design visualization, and gene cloning and visualization (see Note 4).
2.8 ELISA Reagents 1. PBST: Add 100 mL of 10 PBS to a 1-L graduated cylinder.
Make up volume to 1 L using distilled water. Add 0.5 mL of
Tween-20 (final concentration: 0.05%) using a wide-bore 1 mL
pipette tip. Pipette up and down numerous times to ensure that
all the viscous detergent is mixed into the PBS. Shake the bottle
to mix well. Store at room temperature. This can be prepared
several days in advance.
2. Blocking solution: Add 2.5 g of skim milk powder to 25 mL of
PBST in a 50-mL conical tube and vortex to mix. Make up
volume to 50 mL with more PBST. This can be prepared a day
in advance and stored at 4 C. Bring to room temperature prior
to use. Alternatively, 5% w/v bovine serum albumin (BSA) can
also be used as a blocking reagent: Add 50 g of BSA to 1 L of
PBS and dissolve with stirring. Once dissolved, filter-sterilize
and store at 4 C.
3. Diluent solution: Add 1.25 g of skim milk powder to 25 mL of
PBST in a 50-mL conical tube and vortex to mix. Make up
volume to 50 mL with more PBST. This can be prepared a day
in advance and stored at 4 C. Bring to room temperature prior
to use.
4. Transferrin solution: Using a stock solution of 0.5 mg/mL
transferrin conjugated to horseradish peroxidase (Tf-HRP),
prepare a 1:1000 dilution for a working solution in 2.5% skim
milk solution in PBST just prior to use. Vortex to mix well.
Design and Production of Hybrid Antigens 123
3 Methods
3.1 Designing 1. Acquire published structures of the SLP scaffold and the
and Preparing Hybrid TBDT. If structures are not available, generate structural mod-
Antigen Genes els of the scaffold and the TBDT in silico. Programs such as
I-TASSER, RaptorX, or Phyre2 can all be used.
2. Visualize the models using PyMOL.
3. Identify the anchoring residue sites of loops on the SLP scaf-
fold and the TBDT (see Note 6). Locate the antiparallel beta-
strands in each model and label the residues in alternating color
leading up to the “loop,” as shown in Fig. 2 for the example
with TBDT (A. baumannii ZnuD) and SLP scaffold (deriva-
tive of TbpB from N. meningitidis). The splicing site of the
SLP scaffold is determined by selecting anchoring residues that
are next to each other (parallel to each other) and have side
chains that face the same direction on the scaffold (“out” and
124 Somshukla Chaudhuri et al.
Fig. 2 Example of hybrid antigen design. (a) Computer generated model of the TBDT, ZnuD, from Acinetobacter
baumannii with a loop highlighted (red) where the residues of the anchoring beta-barrel are colored differently
depending on whether their side chains are oriented toward the inside of the barrel (orange) or the outside/
away from the barrel (cyan). (b) The scaffold, a derivative of the SLP TbpB C-lobe from Neisseria meningitidis
strain M982 (PDB 5KKX) [1], is labeled similarly to a, with alternating residues colored. (c) The loop from the
ZnuD is determined by selecting parallel anchoring residues (side chains facing the same direction) on the
beta-barrel. If the anchoring residues of the loop are facing in (black arrow in c), then the anchoring residues
of the scaffold face out (black arrow in d) to maintain the antiparallel beta-strand formation
3.2 Expressing 1. Thaw out the plasmid encoding the desired hybrid protein
Hybrid Protein designed in Subheading 3.1, as well as the competent cells
Antigens in E. coli derived from the strain of E. coli to be used for protein expres-
Using a T7 Expression sion (see Note 10). The plasmid concentration should be
Vector ~100 ng/μL, and competent cells should be stored in 50- or
100-μL aliquots. After removing the competent cells from the
80 C freezer, let the cells sit on ice for 20 min prior to
transformation.
2. Add 1–2 μL of plasmid to the competent cells and let the cells
sit on ice for 30 min. Next, transfer to a prechilled 14-mL
round-bottom culture tube.
3. Prepare a 42 C water bath or heat block, then “heat-shock”
the cells at this temperature for 30–60 s, followed by a 2-min
incubation on ice. Promptly add 700 μL of LB medium, then
incubate the cells with shaking at 37 C for 1 h.
4. Add 6 mL of LB containing the appropriate antibiotic (i.e., add
6 μL of 100 mg/mL ampicillin if you are using ampicillin
resistance to select for the presence of your plasmid) to the
culture and incubate with shaking at 37 C for another 4 h.
5. Approximately 1–1.5 h before the end of the 4-h incubation
period, prepare the autoinduction medium by aseptically add-
ing 75 mL NPS, 30 mL 5052, and 1.5 mL MgSO4 to 1.5 L of
ZY media (see Note 11). Four 4-L flasks containing 1.5 L of
media in each are used in a typical protein production run, but
the culture volume can be adjusted depending on the expected
yield of the protein being produced. Add the appropriate anti-
biotic at half the normal concentration (i.e., use 50 μg/mL
instead of 100 μg/mL if using ampicillin). Prewarm the media
in 37 C shaking incubator at a shaking speed of ~50 rpm (see
Note 12).
6. At the end of the 4-h incubation mentioned in step 4, inocu-
late 1.5 L of autoinduction media with 1.5 mL of the starter
culture. Repeat as needed, depending on the number of flasks
being used.
7. Incubate the flasks at 37 C with vigorous shaking (175 rpm)
for 18 h, then adjust the temperature to 20 C and incubate for
another 24 h (see Note 13).
8. Harvest cells by centrifugation at 5000 g for 25 min at 4 C.
9. Prepare Resuspension Buffer (25 mL per L of culture) and add
1 protease inhibitor tablet, 6 mL of 10 mg/mL lysozyme, and
200 μL of 5 mg/mL DNase I (increase the amounts of these
reagents as necessary if the culture volume is greater than 6 L).
Mix in an appropriate-sized beaker, keeping in mind that the
cell pellets will subsequently be transferred to this beaker.
Design and Production of Hybrid Antigens 127
Fig. 4 Representative SDS-PAGE results for purification of a soluble hybrid antigen using the TbpB derived
from Haemophilus influenzae as an example. (a) SDS-PAGE results for 10-μL aliquots of fractions from a
Ni-NTA (IMAC—immobilized metal affinity chromatography) column. FT1 indicates a sample from the pooled
flow through buffer containing proteins that did not bind to the column. Fractions A3-D9 represent the specific
wells of the 96-well collection block that collected the fractions eluted with the imidazole gradient. Fractions
A3–D9 were pooled based on the presence of the 130-kDa MBP-TbpB fusion protein for subsequent TEV
cleavage. (b) SDS-PAGE results for a 10-μL aliquot of the MBP-TbpB fusion protein following cleavage with
TEV protease. The gel confirms that TEV cleavage was complete as there is no MBP-TbpB band present with
TbpB, MBP and TEV protease as the main protein bands. Applying this sample to a Ni-NTA column removes the
MbpB and TEV, leaving TbpB as the main protein (similar to the purified LbpB example shown in Fig. 5)
Fig. 5 Representative SDS-PAGE results following the separation of MBP and TEV
protease from the recombinant protein of interest using a Ni-NTA column. A
modified Neisseria gonorrhoeae lactoferrin binding protein B (LbpB) is provided
as an example. Lane 1: flow through containing the LbpB; lane 2: pooled eluted
fractions (MBP and TEV protease are both present here); lane 3: concentrated
sample of flow through (lane 1). Each lane was loaded with a 10-μL aliquot
3. Lyse the cells by sonicating for two 5 min periods on ice using
conditions that do not result in overheating the samples. These
generally involve short pulses with cooling periods of 2–4 times
longer. We use a QSonica sonicator (pulse 03, amplitude 65%).
4. Centrifuge the lysate for 35 min at 20,440 g at 4 C.
5. Collect the supernatant and filter through a 0.2-μm filter.
6. Add 3 mL of Ni-NTA resin for every 50 mL of filtered lysate
(see Note 23), then mix gently in 50-mL conical tubes using a
rotary shaker overnight (~16 h) at room temperature.
7. Prepare four buffers with decreasing concentrations of urea
(3 M, 1.5 M, 0.75 M, 0.375 M) by starting with Denaturing
Wash Buffer and subsequently performing serial dilutions,
using Refolding Wash Buffer as a diluent.
8. Centrifuge the lysate–resin mixture for 10 min at 3220 g at
room temperature, then decant the supernatant. Next, using
18 mL (or 6 volumes of Ni-NTA resin) of the initial buffer in
the aforementioned serial dilution (50 mM NaH2PO4,
300 mM NaCl, 3 M urea, 5 mM imidazole, pH 8.0), resuspend
the Ni-NTA resin and incubate the resulting mixture on a
rotary shaker for 30 min at room temperature.
9. Repeat step 8 for each of the subsequent buffers in the serial
dilution series prepared in step 7.
10. Resuspend the Ni-NTA resin in 18 mL Refolding Wash Buffer,
then transfer the mixture to a gravity column.
11. Elute any proteins bound to the Ni-NTA using 18 mL of
Refolding Elution Buffer (see Note 24). The eluted volume
can be collected in 1-mL fractions.
12. Verify the presence of the desired hybrid antigen using SDS-
PAGE, then perform steps 8–12 described under Section.
3.5 Cloning of Tbdt 1. Identify mature sequence of TBDT using the online software
in Fusion SignalP (see Note 25).
with Streptavidin 2. Design primers to amplify the selected gene or synthesize the
Binding Protein (SBP) sequence.
3. Clone the TBDT gene into plasmid pE5771 or an alternate
expression plasmid containing SBP (see Note 26).
4. Transform the newly constructed plasmid into E. coli TOP10
cells for long-term storage, as described in Subheading 3.1,
steps 9–15.
3.7 Coating 1. Pour the SBP-TBDT–PBST mixture into a reagent boat, and,
Streptavidin ELISA using a multichannel pipette, pipette out 100 μL into each well
Plates with Detergent- of a streptavidin-coated 96-well plate (see Note 31).
Extracted SBP-TBDT 2. Incubate at room temperature for 1 h, then wash with 250 μL/
well of PBST three times (see Notes 32 and 33).
3. Add 250 μL of the prepared 5% blocking solution to each well
after the wash step, incubate at room temperature again for 1 h,
then wash with 250 μL/well of PBST three times.
3.8 Assessing 1. During the blocking step (Subheading 3.7, step 3), add 1 μL
Conformation of TbpA Tf-HRP to 1 mL of diluent solution (i.e., final dilution of
by Its Ability to Bind 1/1000).
to Transferrin 2. Add 100 μL of the Tf-HRP solution to each of four wells (i.e.,
two test and two control wells), incubate for 1 h, then wash
with 250 μL of PBST three times.
3. Add 50 μL of TMB to the four wells and develop in the dark
(a cupboard/drawer is adequate) for 20 min, then add 25 μL of
4 N HCl to stop the reaction. Measure the optical density at
450 nm (see Note 34 and Fig. 7).
3.9 Assessing 1. During the blocking step (Subheading 3.7, step 3), thaw
antibody Titers serum samples on ice for at least 20 min and vortex to mix well.
in Serum Samples 2. Add 100 μL of diluent solution to each well. Add additional
from Immunized diluent solution in the first well of each row (A to H). The
Animals amount of additional diluent solution will depend on the
desired starting dilution for the serum used in the assay, such
that the final volume of diluent with added serum is 200 μL.
4 Notes
12. This step is not always necessary; however, we find that pre-
warming may enhance bacterial growth and protein yields
compared to inoculation at room temperature.
13. The yield of protein after growth at 37 C appears to predict
whether an additional period of growth at low temperature will
lead to an increased yield. The expression plasmids with lower
yields after growth at 37 C (perhaps unstable or prone to
degradation) do not achieve enhanced yields with an additional
incubation period at low temperature.
14. For efficient resuspension of the cell pellets, scoop out each cell
pellet using a spatula and transfer it to the beaker containing
the Resuspension Buffer mixture. Then, transfer ~20 mL of the
Resuspension Buffer mixture into each centrifugation bottle
using a serological pipettor to resuspend any remaining bacte-
ria. Finally, transfer each of the resulting cell suspensions to the
beaker containing the cell pellets and the remaining Resuspen-
sion Buffer mixture.
15. Perform four discrete passes through the homogenizer and
avoid recirculating the sample as it results in uneven lysis—
that is, collect the lysate in a beaker after each independent pass
through the homogenizer instead of continuously directing
the sample back through the homogenizer.
16. Use appropriate size filtration devices for filtering the superna-
tant. When using syringe filters, limit the volume to 25–50 mL
of lysate, depending on viscosity. If the sample cannot be
filtered easily using a 0.2-μm syringe filter, use a 0.45-μm filter
first before passing the lysate through the 0.2-μm filter. If the
lysate still cannot be easily filtered, an additional 200 μL of
DNase I (5 mg/mL) can be added and mixed in with the lysate
for 30 min using a magnetic stir bar prior to filtration.
17. If the protein antigen of interest is readily detected in the
supernatant, this suggests that the recombinant protein is suf-
ficiently soluble to allow for the purification of the protein
using the protocol outlined in Subheading 3.3. If the SDS-
PAGE gel indicates the absence of the protein in the superna-
tant, or “soluble fraction,” this suggests that constitutive
expression of the protein in the cytoplasm of E. coli has resulted
in aggregation and misfolding of the protein. Thus, due to
the apparent insolubility resulting from improper folding of
the protein of interest, the protein is likely present alongside
the cell debris in the pellet following centrifugation (Subhead-
ing 3.2, step 12). In this circumstance we recommend restart-
ing the protein production run (beginning at the start of
Subheading 3.2) and proceeding directly to Subheading 3.4
immediately following Subheading 3.2, step 8.
Design and Production of Hybrid Antigens 137
References
1. Fegan JE, Calmettes C, Islam EA, Ahn SK, factors in Actinobacillus pleuropneumoniae
Chaudhuri S, Yu RH, Gray-Owen SD, Moraes serotype 7 infection. FEMS Microbiol Lett
TF, Schryvers AB (2019) Utility of hybrid 209(2):283–287
transferrin binding protein antigens for protec- 9. Anderson JE, Hobbs MM, Biswas GD, Spar-
tion against pathogenic Neisseria species. Front ling PF (2003) Opposing selective forces for
Immunol 10:247. https://doi.org/10.3389/ expression of the gonococcal lactoferrin recep-
fimmu.2019.00247 tor. Mol Microbiol 48(5):1325–1337. https://
2. Qamsari MM, Rasooli I, Chaudhuri S, Astaneh doi.org/10.1046/j.1365-2958.2003.
SDA, Schryvers AB (2020) Hybrid antigens 03496.x
expressing surface loops of ZnuD from Acine- 10. Hooda Y, Lai CCL, Moraes TF (2017) Identi-
tobacter baumannii is capable of inducing pro- fication of a largefamily of Slam-dependent sur-
tection against infection. Front Immunol face lipoproteins in gram-negative bacteria.
11:1–13. https://doi.org/10.3389/fimmu. Front Cell Infect Microbiol 7:207. https://
2020.00158 doi.org/10.3389/fcimb.2017.00207
3. Noinaj N, Guillier M, Barnard TJ, Buchanan 11. Frandoloso R, Martinez-Martinez S,
SK (2010) TonB-dependent transporters: reg- Calmettes C, Fegan J, Costa E, Curran D,
ulation, structure, and function. Annu Rev Yu R, Gutierrez-Martin CB, Rodriguez-Ferri
Microbiol 64:43–60. https://doi.org/10. EF, Moraes TF, Schryvers AB (2015) Non-
1146/annurev.micro.112408.134247 binding site-directed mutants of transferrin
4. Postle K, Kadner RJ (2003) Touch and go: binding protein B enhances their immunoge-
tying TonB to transport. Mol Microbiol 49 nicity and protective capabilities. Infect Immun
(4):869–882 83(3):1030–1038
5. Hood MI, Skaar EP (2012) Nutritional immu- 12. Calmettes C, Alcantara J, Yu R-H, Schryvers
nity: transition metals at the pathogen-host AB, Moraes TF (2012) The structural basis of
interface. Nat Rev Microbiol 10(8):525–537. transferrin sequestration by transferrin binding
https://doi.org/10.1038/nrmicro2836 protein B. Nat Struct Mol Biol 19
6. Morgenthau A, Pogoutse A, Adamiak P, Moraes (3):358–360. https://doi.org/10.1038/
TF, Schryvers AB (2013) Bacterial receptors for nsmb.2251
host transferrin and lactoferrin: molecular 13. Noinaj N, Buchanan SK, Cornelissen CN
mechanisms and role in host-microbe interac- (2012) The transferrin-iron import system
tions. Future Microbiol 8(12):1575–1585 from pathogenic Neisseria species. Mol Micro-
7. Cornelissen CN, Kelley M, Hobbs MM, biol 86(2):246–257. https://doi.org/10.
Anderson JE, Cannon JG, Cohen MS, Sparling 1111/mmi.12002
PF (1998) The transferrin receptor expressed 14. Fegan JE, Yu R-H, Islam EA, Schryvers A
by gonococcal strain FA1090 is required for (2021) Development of a non-biased, high-
the experimental infection of human male throughput ELISA for the rapid evaluation of
volunteers. Mol Microbiol 27(3):611–616 immunogenicity and cross-reactivity. J Immu-
8. Baltes N, Hennig-Pauka I, Gerlach GF (2002) nol Methods 493:113027
Both transferrin binding proteins are virulence
Chapter 9
Abstract
A variety of delivery vehicles have been explored as adjuvanting/delivery platforms for peptide-based
subunit vaccines. Polysaccharide-based systems have been found to be especially attractive due to their
immune stimulating properties, biodegradability, biocompatibility, and low toxicity. Among them, chitosan
and its derivatives are the most common cationic nanocarriers used for the delivery of antigens. Trimethyl
chitosan (TMC) is a partially quaternized, water-soluble, and mucoadhesive derivative of chitosan. This
chapter describes the preparation of a TMC-based polyelectrolyte complex as a delivery system for peptide
subunit vaccines.
Key words Trimethyl chitosan, Alginate, Polyelectrolyte complexes, Peptide subunit vaccine,
Nanoparticles
1 Introduction
141
142 Lili Zhao et al.
Fig. 1 Schematic illustration of the preparation of the LCP-1-loaded polyelectrolyte complex: LCP-1/alginate/
TMC. (a) Primary complex (LCP-1/alginate), (b) Ternary PEC (LCP-1/alginate/TMC)
2 Materials
2.2 Formulation and 1. Stock solution of alginate sodium in Milli-Q water (2 mg/mL).
Characterization of 2. Stock solution of trimethyl chitosan (TMC) in Milli-Q water
PECs (LCP-1/Alginate/ (2 mg/mL).
TMC)
3. Stock solution of LCP-1 in Milli-Q water (1 mg/mL).
4. Phosphotungstic acid (0.5% w/v).
2.3 Equipment 1. Magnetic stirrer with hot plate, magnetic stir bar, and ice bath.
2. Thermometer.
3. Condenser.
4. Freeze dryer.
5. Nuclear magnetic resonance (NMR) spectrometer (300 MHz
or higher).
6. Probe-type sonicator.
7. Malvern Zetasizer Nano-ZS (Malvern Instruments Ltd., UK or
equivalent) to measure dynamic light scattering (DLS).
8. Transmission electron microscope.
9. Dialysis bag (3500 Da).
10. Balance, microbalance.
11. Laboratory glassware and apparatus.
3 Methods
3.1 Synthesis of TMC 1. To 250 mL round bottom flask add low molecular weight
chitosan (1 g) and sodium iodide (2.4 g), add 40 mL of NMP.
2. Stir the mixture until fully dissolved.
3. Add 6 mL of 15% aqueous NaOH solution and stir the mixture
for 20 min. The temperature should be kept at 60 C (see
Note 1).
4. Add 6 mL of the methylation agent, methyl iodide, to the
reaction mixture. Attach a reflux condenser to the flask and
stir the mixture for 1 h at 60 C.
5. Slowly pour the reaction mixture into 50 mL of diethyl ether–
ethanol solution to precipitate the product (see Note 2).
144 Lili Zhao et al.
3.2 Preparation of 1. Dilute the alginate stock solution (2 mg/mL) to 0.1, 0.2, 0.4,
PECs (LCP-1/Alginate/ 0.6, 0.8, and 1.0 mg/mL, with Milli-Q water (see Note 6).
TMC) 2. Transfer 100 μL of LCP-1 solution (100 μg) into a 2 mL
3.2.1 Prepare the centrifuge tube; dilute 10 times with Milli-Q water.
Primary Complexes (LCP-1/ 3. Add 50 μL of the diluted alginate solution (0.1 mg/mL, con-
Alginate) taining 5 μg alginate) into the centrifuge tube and mix
thoroughly.
4. Sonicate the mixture for 4 min (2 2 min) using a probe-type
sonicator at 120 W (duration 2 min, duty cycle 50%) in an
ice bath.
5. Transfer the mixture to a scintillation vial and stir it continu-
ously for 1 h at room temperature to stabilize the complexes.
6. Measure the particle size, polydispersity index (PDI), and zeta
potential using dynamic light scattering (DLS).
146 Lili Zhao et al.
Fig. 3 Optimization of the ratio of LCP-1 to alginate for the preparation of primary complexes (LCP-1/alginate):
the optimum ratio is indicated by white shading. (a) The bars show size (nm), while the line represents
polydispersity index (PDI). (b) The bars show zeta potential (mV). LCP-1/alginate ¼ 10:4, size 177 2 nm, PDI
0.205 0.01, zeta potential 37.1 0.7 mV
3.2.2 Preparation of 1. Dilute TMC stock solution (2 mg/mL) to 0.6, 1.0, 1.2, and
Ternary PECs (LCP-1/ 1.6 mg/mL with Milli-Q water.
Alginate/TMC) 2. Add 50 μL of TMC solution (0.6 mg/mL, containing 30 μg
TMC) dropwise to freshly prepared LCP-1/alginate complex
suspension (containing 100 μg of LCP-1, 40 μg of alginate).
3. Stir the solution for 1 h at room temperature to produce the
final PEC nanoparticles.
4. Measure the particle size, PDI, and zeta potential using DLS.
5. Repeat steps 2–4 five times, except at step 2, add 50 μL of
TMC solution at concentrations of 0.6, 1.0, 1.2, 1.6, and
2.0 mg/mL (instead of 50 μL of the diluted TMC solution)
to produce ternary PEC nanoparticles at ratios of 10:4:3,
10:4:5, 10:4:6, 10:4:8, and 10:4:10, respectively (see Note 8).
6. Measure the particle size, PDI, and zeta potential of the pri-
mary complexes and final ternary PEC nanoparticles using
DLS. The expected particle size and zeta potential for different
concentration are shown in Fig. 4 (see Note 9).
7. Examine the morphology of the PEC nanoparticles using
transmission electron microscopy. Drop PEC nanoparticles
(dilution might be required) onto a carbon-coated copper
grid and leave it for 1 min to allow the nanoparticles to deposit
Chitosan-Based Nanovaccine 147
Fig. 4 Optimization of PEC nanoparticle (LCP-1/alginate/TMC) preparation using DLS monitoring: the optimum
mixing ratio is indicated by white shading. (a) The bars show size (nm), while the line represents PDI. (b) The
bars show zeta potential (mV). LCP-1/alginate/TMC ¼ 10:4:8, size 237 4 nm, PDI 0.199 0.01, zeta
potential 29.3 1.5 mV
4 Notes
References
1. Nevagi RJ, Toth I, Skwarczynski M (2018) 5. Fujita Y, Taguchi H (2011) Current status of
Peptide-based vaccines. In: Koutsopoulos S multiple antigen-presenting peptide vaccine
(ed) Peptide applications in biomedicine, bio- systems: application of organic and inorganic
technology and bioengineering. Woodhead nanoparticles. Chem Cent J 5(1):48
Publishing, Oxford 6. Skwarczynski M, Toth I (2014) Recent
2. Skwarczynski M, Toth I (2016) Peptide-based advances in peptide-based subunit nanovac-
synthetic vaccines. Chem Sci 7(2):842–854 cines. Nanomedicine 9:2657–2669
3. Bartlett S, Skwarczynski M, Toth I (2020) 7. Reddy ST, Van Der Vlies AJ, Simeoni E et al
Lipids as activators of innate immunity in pep- (2007) Exploiting lymphatic transport and
tide vaccine delivery. Curr Med Chem 27 complement activation in nanoparticle vac-
(17):2887–2901 cines. Nat Biotechnol 25(10):1159–1164
4. Nevagi RJ, Skwarczynski M, Toth I (2019) 8. Henriksen-Lacey M, Christensen D, Bramwell
Polymers for subunit vaccine delivery. Eur VW et al (2011) Comparison of the depot
Polym J 114:397–410 effect and immunogenicity of liposomes based
Chitosan-Based Nanovaccine 149
Abstract
Vaccines are regarded as the most cost-effective countermeasure against infectious diseases. One challenge
often affecting vaccine development is antigenic diversity or pathogen heterogeneity. Different strains
produce immunologically heterogeneous virulence factors, therefore an effective vaccine needs to induce
broad-spectrum host immunity to provide cross-protection. Recent advances in genomics and proteomics,
particularly computational biology and structural biology, establishes structural vaccinology and highlights
the feasibility of developing effective and precision vaccines. Here, we introduce the epitope- and structure-
based vaccinology platform multiepitope-fusion-antigen (MEFA), and provide instructions to generate
polyvalent MEFA immunogens for vaccine development. Conceptually, MEFA combines epitope vaccinol-
ogy and structural vaccinology to enable a protein immunogen to present heterogeneous antigenic domains
(epitopes) and to induce broadly protective immunity against different virulence factors, strains or diseases.
Methodologically, the MEFA platform first identifies a safe, structurally stable and strongly immunogenic
backbone protein and immunodominant (ideally neutralizing or protective) epitopes from heterogeneous
strains or virulence factors of interest. Then, assisted with protein modeling and molecule dynamic
simulation, MEFA integrates heterogeneous epitopes into a backbone protein via epitope substitution for
a polyvalent MEFA protein and mimics epitope native antigenicity. Finally, the MEFA protein is examined
for broad immunogenicity in animal immunization, and assessed for potential application for multivalent
vaccine development in preclinical studies.
Key words MEFA (multiepitope fusion antigen), Vaccinology platform, Multivalent vaccine, Epi-
topes, Structural vaccinology
1 Introduction
151
152 Siqi Li et al.
Fig. 1 Illustration of constructing a polyvalent MEFA immunogen by using the MEFA vaccinology platform.
Surface-exposed epitopes of a backbone immunogen are substituted with protective or immunodominant
epitopes from heterogeneous virulence factors or strains of interest to construct an MEFA immunogen
2 Materials
2.1 Computation MEFA backbone protein selection, epitope identification, and pro-
and Programs tein modeling are carried out on standard desktop or laptop com-
puters through open-access servers. Atomic molecular dynamics
simulation requires a local server and remote access to license-
based CHARMM server or open-source GROMACS-5.0.7 MD
server.
Programs or servers that are applied for in silico analyses are as
follows.
1. IEDB (http://www.iedb.org/) or BepiPred (http://www.cbs.
dtu.dk/services/BepiPred/).
2. ExPASy (https://www.expasy.org/).
3. PyMol (https://www.pymol.org/).
4. Phyre2 (http://www.sbg.bio.ic.ac.uk/~phyre2/html/page.
cgi?id¼index).
5. GROMACS (http://www.gromacs.org/).
6. Or CHARMM (https://www.charmm.org/), and MMTSB
toolset (http://blue11.bch.msu.edu/mmtsb/Main_Page).
2.2 A Backbone The MEFA platform requires a backbone protein and epitopes of
Protein heterogeneous strains or virulence factors of interest to construct
and Heterogeneous an MEFA immunogen. A backbone protein should be safe and
Epitopes of Interest structurally stable, preferably a mutant key virulence factor which
becomes avirulent after epitope substitution and induces protective
154 Siqi Li et al.
2.4 Protein 1. SDS-PAGE, for MEFA protein Coomassie blue stain or West-
Biochemical Property ern blot with specific antibodies.
and Immunogenicity 2. Coomassie blue stain buffer (0.125% Coomassie blue R350,
Characterization 50% methanol, 10% acetic acid), and destain buffer (20% meth-
anol, 10% acetic acid, in ddH2O).
3. Protein immunogenicity studies: MEFA protein-induced T cell
and/or B cell immune responses are examined in animal immu-
nization studies. Antigen specific adaptive immunity and
in vitro protective activities, which vary and are specific to the
pathogen of interest, are measured to assess MEFA immuno-
gen broad immunity (B cell epitopes are used for MEFA immu-
nogen construction in this protocol).
Animal immunization: 8-week-old mice, 25-gauge needle and
1-mL syringe, alum or incomplete Freund’s adjuvant; mouse fecal
and serum samples collected for measuring antigen-specific anti-
body responses.
Antibody titration: 96-well microtiter plates, ELISA coating
antigens specific to the virulence factors targeted by MEFA protein,
HRP-conjugated goat anti-mouse IgG and IgA secondary antibo-
dies, 3,30 ,5,50 -tetramethybenzidine (TMB) Microwell Peroxidase
Substrate, a 96-well plate reader.
Antibody in vitro protection: assays to evaluate in vitro protec-
tion of MEFA-induced antibodies vary and are specific to patho-
gens or virulence factors of interest.
Preclinical challenge studies: if animal challenge models are
available, immunizing animals with an MEFA protein to assess
epitope-specific immunogenicity, then challenging the immunized
animals with pathogen strains to measure cross protection and to
evaluate potential application of an MEFA immunogen for multi-
valent vaccine development.
3 Methods
3.1 Backbone Online programs that predict epitopes, locate epitope positions,
Protein Selection image protein secondary structure, and examine protein stability
are applied to select a suitable backbone protein. B cell epitopes on
a backbone protein, including epitope amino acid sequence, posi-
tion, and antigenic score, are predicted and analyzed by using
156 Siqi Li et al.
Fig. 2 Illustration of in silico identifying B cell immunodominant epitopes with IEDB program. Left: a diagram to
show in silico predicted epitopes, with antigenic scores and locations, from a backbone protein. Right: a table
to indicate epitope position, sequence and length
3. Input protein amino acid sequence and name your optional job
description.
4. Select “Intensive” as the modeling and analysis mode.
5. Press “Phyre Search.”
6. Receive email notices of computation updates, results in a “xxx.
pdb file format” (xxx is the name of your optional job descrip-
tion; the confidence in the intensive model should be greater
than 90%), and a link to view your protein model and 3D
structure image.
7. Download your “xxx.pdb” file from Phyre2 site, view protein
structure, and further analyze “xxx.pdb” data for 3D structure
with PyMol/ EdiPyMol.
8. Download PyMol (https://www.pymol.org/).
9. Open your Phyre2 file “xxx.pdb” through PyMol program by
using “File”!“Open”!“Downloads” by selecting your “xxx.
pdb file.
10. Select the “Display” tab on the top to view protein amino acid
sequence.
11. Select the “S” tab to show “surface” at the right for display
type of the secondary structure in a 3D model, and the “C” for
a uniform color for the backbone protein model (Fig. 3a).
12. Mark epitope amino acid sequences in different colors to image
the location and surface exposure of each epitope (Fig. 3b) (see
Note 5).
Fig. 3 Illustration of a backbone protein 3D model (a) and position of a backbone epitope (b), by PyMol
program
158 Siqi Li et al.
3.2 Identification With a desirable backbone immunogen selected, epitopes from the
of Epitopes from heterogeneous strains or virulence factors of interest must be iden-
Target Heterogeneous tified. Protective epitopes are ideal for MEFA immunogen con-
Strains or Virulence struction. Having epitope native antigenicity retained, an MEFA
Factors immunogen carrying multiple protective epitopes of various strains
or virulence factors induces broadly protective immune responses.
As pointed out in Subheading 3.1, protective epitopes from well
characterized pathogens are deposited at IEDB (www.iedb.org)
and can be used directly for MEFA immunogen construction. For
strains or virulence factors without protective epitopes being iden-
tified, immunodominant epitopes in silico identified can be used
first; eventually protective epitopes selected by screening immuno-
dominant ones in empirical studies are included in an MEFA
immunogen.
3.2.1 To Predict Epitopes Immunodominant linear epitopes from each virulence factor or
of Heterogeneous Strains strain are predicted with IEDB, the same as backbone protein
or Virulence Factors epitope prediction described in Subheading 3.1.1, with the only
of Interest difference is the input of different amino acid sequences.
3.3.1 GROMACS 1. Download and install SSH client from link https://www.
to Characterize an MEFA netsarang.com/en/all-downloads/ (Xshell for Windows OS);
Protein log in from your desktop as an SSH client to connect remote
server (Linux OS) and execute commands (note: more infor-
mation at https://netsarang.atlassian.net/wiki/spaces/
ENSUP/pages/31555780/Getting+Started).
2. Download and install GROMACS (http://www.gromacs.org/
) and FFTW (version 3.3.8) (http://www.fftw.org/download.
html).
3. Input “MEFA protein (or backbone or each virulence factor).
pdb” file into Xshell generated from Phyre2 under your server
account.
4. Generate an MEFA protein topology form the input pdb.file,
with command gmx pdb2gmx -f MEFA protein.pdb -o
mefa_processed.gro -water spce, with the selection of GRO-
MOS96 53a6 force field.
5. Select periodic boundary conditions (PBC) at “cubic” shape
and “1.0 nm” parameters with gmx editconf -f mefa_pro-
cessed.gro -o mefa_newbox.gro -c -d 1.0 -bt cubic.
160 Siqi Li et al.
6. Enter solvent water into the simulation box with gmx solvate
-cp mefa_newbox.gro -cs spc216.gro -o mefa_solv.gro -p
topol.top.
7. Conduct system neutralization with command gmx grompp -f
ions.mdp -c mefa_solv.gro -p topol.top -o ions.tpr, add
counter ions through “genion” with gmx genion -s ions.tpr
-o mefa_solv_ions.gro -p topol.top -pname NA -nname CL
-np 9.
8. Minimize energy with commands gmx grompp -f minim.
mdp -c mefa_solv_ions.gro -p topol.top -o em.tpr and
gmx mdrun -v -deffnm em.
9. Equilibrate NVT with gmx grompp -f nvt.mdp -c em.gro -p
topol.top -o nvt.tpr and gmx mdrun -deffnm nvt.
10. Equilibrate NPT with command gmx grompp -f npt.mdp -c
nvt.gro -t nvt.cpt -p topol.top -o npt.tpr and gmx mdrun
-deffnm npt.
11. Carry out molecule dynamics (MD) simulations, with gmx
grompp -f md.mdp -c npt.gro -t npt.cpt -p topol.top -o
md_0_1.tpr and gmx mdrun -deffnm md_0_1.
12. Calculate Cα root-mean-square deviation (RMSD) and root-
mean-square fluctuations (RMSF) (Fig. 4), with commands
gmx trjconv -s md_0_1.tpr -f md_0_1.xtc -o
md_0_1_noPBC.xtc -pbc mol -ur compact, gmx rms -s
md_0_1.tpr -f md_0_1.trr -o rmsd.xvg, and gmx rmsf -s
md_0_1.tpr -f md_0_1.xtc -o rmsf.xvg, respectively.
Fig. 4 GROMOS molecule dynamic simulation to show root-mean-square deviation—RMSD (a) and root-
mean-square fluctuation—RMSF (b) of a backbone protein and an MEFA immunogen protein. RMSD and
RMSF are used to assess structural and antigenic similarity between a backbone protein and an MEFA protein,
or between an epitope on the MEFA and the epitope on the native virulence factor protein. Boxed is a backbone
protein epitope and a substituting epitope on an MEFA protein
MEFA for Multivalent Vaccines 161
Fig. 5 GROMOS molecule dynamic simulation to show solvent average surface area (SASA) of a backbone
protein (or an MEFA immunogen protein, or the epitope of interest on an MEFA protein or native virulence
factor protein). SASA is used to assess structural and antigenic similarity between a backbone protein and an
MEFA protein, or between an epitope on the MEFA and the epitope on the native virulence factor protein
3.4 MEFA Protein After protein structure stability and epitope antigenicity have been
Expression, Extraction, assessed and verified in silico, we synthesize an MEFA immunogen
and Immunogenicity gene and clone the gene for protein expression and extraction. The
Characterization MEFA gene can be inserted into the pET28a expression vector
directly at synthesis, or with two restriction enzyme sites flanked
at the MEFA gene for cloning into the pET28a vector, for expres-
sion of 6xHis-tagged or tag-less recombinant MEFA protein in
E. coli strain BL21 (DE3), by following standard molecular biology
protocols [22]. MEFA protein expression and extraction are ver-
ified by SDS PAGE with Coomassie blue staining and western
blotting with specific antibodies; MEFA protein broad immunoge-
nicity is confirmed in mouse immunization.
3.4.1 MEFA Protein Recombinant 6xHis-tag MEFA protein is purified with Ni-NTA
Expression and Extraction agarose in a column (www.qiagen.com), whereas tag-less MEFA
protein expressed in inclusion body is extracted with B-PER™
Bacterial Protein Extraction Reagent (https://www.thermofisher.
com).
1. Pick a colony from overnight growth on a LB/kan agar plate,
culture in 5–10 ml 2 YT medium supplemented with kana-
mycin (30 μg/mL) at 37 C overnight on a shaker (200 rpm).
2. Transfer 2–3 mL bacterial growth to 200 mL 2 YT medium
supplemented with kanamycin (30 μg/mL) in a 500 mL flask,
culture at 37 C on a shaker (200 rpm) till the OD600 reaches
0.5–0.7.
MEFA for Multivalent Vaccines 163
3.4.2 MEFA Protein Eight-week-old female BALB/c mice, 8–10 per group, are com-
Immunization monly used for immunization with MEFA protein to examine
and MEFA-Specific broad immunogenicity (see Note 9).
Antibody Titration
1. Mix and emulsify 50 μg MEFA protein (in 50 μL PBS) with an
equal amount of incomplete Freund’s adjuvant (IFA); 50 μL
PBS mixed with IFA as a negative control.
2. Inject each mouse with MEFA protein (or PBS for the control
group) intraperitoneally (IP) [or intramuscularly (IM), subcu-
taneously (SC), or intradermally (ID)].
3. Two booster injections (of the same dose and route of the
primary) are followed at the interval of 2 weeks.
4. Collect mouse serum and fecal samples 2 weeks after the final
booster (see Note 10).
5. Store serum and fecal supernatant at 80 C until the use for
antibody titration and antibody neutralization studies.
6. To titrate antigen-specific antibody responses, coat 96-well
microtiter 2HB ELISA plates with epitope specific antigen
(50–100 ng of each virulence factor protein in 100 μL
0.05 M carbonate-bicarbonate buffer, pH 9.6; per well) over-
night at 4 C.
7. Discard coating reagent, wash wells three times with PBST
(PBS with 0.05% Tween 20), 150 μL PBST per well.
MEFA for Multivalent Vaccines 165
4 Notes
Acknowledgments
References
1. Collaborators GD, Collaborators GH (2017) https://doi.org/10.1016/j.vaccine.2004.11.
Global, regional, and national disability- 010
adjusted life-years (DALYs) for 333 diseases 12. Huang JC, Duan QD, Zhang WP (2018) Sig-
and injuries and healthy life expectancy nificance of enterotoxigenic Escherichia coli
(HALE) for 195 countries and territories, (ETEC) heat-labile toxin (LT) enzymatic sub-
1990-2016: a systematic analysis for the global unit epitopes in LT enterotoxicity and immu-
burden of disease study 2016. Lancet 390 nogenicity. Appl Environ Microbiol 84(15):
(10100):1260–1344 e00849-00818. https://doi.org/10.1128/
2. Global health estimates 2016: Deaths by cause, AEM.00849-18
age, sex, by country and by region, 2000–2016 13. Lu T, Moxley RA, Zhang W (2019) Mapping
(2018). https://www.who.int/healthinfo/ the neutralizing epitopes of enterotoxigenic
global_burden_disease/estimates/en/index1. Escherichia coli (ETEC) K88 (F4) fimbrial
html. Accessed 21 Jan 2021 adhesin and major subunit FaeG. Appl Environ
3. Nabel GJ (2013) Designing tomorrow’s vac- Microbiol 85(11):e00329–e00319. https://
cines. New Engl J Med 368(6):551–560. doi.org/10.1128/AEM.00329-19
https://doi.org/10.1056/NEJMra1204186 14. Lu T, Seo H, Moxley RA, Zhang W (2019)
4. Mascola JR, Ahmed R (2015) Vaccines: vac- Mapping the neutralizing epitopes of F18 fim-
cines for cancer and infectious diseases. Curr brial adhesin subunit FedF of enterotoxigenic
Opin Immunol 35:V–Vii. https://doi.org/10. Escherichia coli (ETEC). Vet Microbiol
1016/j.coi.2015.07.009 230:171–177. https://doi.org/10.1016/j.
5. Ruan X, Sack DA, Zhang W (2015) Genetic vetmic.2019.02.015
fusions of a CFA/I/II/IV MEFA (multiepi- 15. Lu T, Moxley RA, Zhang W (2020) Applica-
tope fusion antigen) and a toxoid fusion of tion of a novel epitope and structure
heat-stable toxin (STa) and heat-labile toxin vaccinology-assisted fimbria-toxin multiepi-
(LT) of enterotoxigenic Escherichia coli tope fusion antigen of enterotoxigenic Escher-
(ETEC) retain broad anti-CFA and antitoxin ichia coli for multivalent vaccine development
antigenicity. PLoS One 10(3):e0121623 against porcine post-weaning diarrhea. Appl
6. Duan Q, Lee KH, Nandre RM, Garcia C, Environ Microbiol 86(24):e00274-20.
Chen J, Zhang W (2017) MEFA (multiepitope https://doi.org/10.1128/AEM.00274-20
fusion antigen)-novel technology for structural 16. Vita R, Mahajan S, Overton JA, Dhanda SK,
vaccinology, proof from computational and Martini S, Cantrell JR et al (2019) The
empirical immunogenicity characterization of immune epitope database (IEDB): 2018
an enterotoxigenic Escherichia coli (ETEC) update. Nucleic Acids Res 47(D1):
Adhesin MEFA. J Vaccines Vaccin 8(4):367. D339–D343. https://doi.org/10.1093/nar/
https://doi.org/10.4172/2157-7560. gky1006
1000367 17. Artimo P, Jonnalagedda M, Arnold K,
7. Seo H, Duan Q, Zhang W (2020) Vaccines Baratin D, Csardi G, de Castro E et al (2012)
against gastroenteritis, current progress and ExPASy: SIB bioinformatics resource portal.
challenges. Gut Microbes 11(6):1486–1517. Nucleic Acids Res 40(W1):W597–W603.
https://doi.org/10.1080/19490976.2020. https://doi.org/10.1093/nar/gks400
1770666 18. Kelley LA, Mezulis S, Yates CM, Wass MN,
8. Lycke N, Holmgren J (1986) Strong adjuvant Sternberg MJE (2015) The Phyre2 web portal
properties of cholera toxin on gut mucosal for protein modeling, prediction and analysis.
immune responses to orally presented antigens. Nat Protoc 10(6):845–858. https://doi.org/
Immunology 59(2):301–308 10.1038/nprot.2015.053
9. Clements JD, Hartzog NM, Lyon FL (1988) 19. Chivian D, Baker D (2006) Homology model-
Adjuvant activity of Escherichia coli heat-labile ing using parametric alignment ensemble gen-
enterotoxin and effect on the induction of oral eration with consensus and energy-based
tolerance in mice to unrelated protein antigens. model selection. Nucleic Acids Res 34(17):
Vaccine 6(3):269–277 e112. https://doi.org/10.1093/nar/gkl480
10. Lycke N (2005) Targeted vaccine adjuvants 20. Raman S, Vernon R, Thompson J, Tyka M,
based on modified cholera toxin. Curr Mol Sadreyev R, Pei JM et al (2009) Structure pre-
Med 5(6):591–597 diction for CASP8 with all-atom refinement
11. Freytag LC, Clements JD (2005) Mucosal using Rosetta. Proteins Struct Funct Genet
adjuvants. Vaccine 23(15):1804–1813.
MEFA for Multivalent Vaccines 169
Abstract
Bacterial extracellular vesicles (BEVs) possess features that make them well suited for the delivery of
therapeutics and vaccines. This chapter describes methods for engineering the commensal human intestinal
bacterium Bacteroides thetaiotaomicron (Bt) to produce BEVs carrying vaccine antigens and accompanying
methods for isolating and purifying BEVs for mucosal vaccination regimens.
1 Introduction
171
172 Régis Stentz et al.
2 Materials
2.1 Synthetic Gene 1. Protein sequence (e.g., stalk of the hemagglutinin antigen of
Design influenza A virus strain H5N1 [8, 17]).
2. Secretion sequence signal (e.g., N-terminal sequence of Bt
outer membrane protein A [OmpA] (BT_3852)).
3. Optional additional sequences (e.g., His-tag or FLAG-tag
sequences).
4. Escherichia coli/Bacteroides shuttle expression vector sequence
(e.g., pGH90 [18]) (see Note 3).
Production, Isolation, and Characterization of Bioengineered Bacterial. . . 173
Fig. 1 Overview of the production and extraction of bioengineered bacterial extracellular vesicles (BEVs)
derived from B. thetaiotaomicron for the development of novel vaccines. Immunization and sample collection
for preclinical development and evaluation are described in Notes 1 and 2
2.2 Cloning The required materials for the cloning of the conjugative plasmid
of Synthetic Gene harboring the gene of interest are as follows:
2.2.2 Transformation 1. Crushed ice for thawing the MAX Efficiency® DH10B compe-
of Competent Cells tent cells.
2. S.O.C. medium: commercially available.
3. Luria-Bertani (LB) agar plates: prepare according to the man-
ufacturer’s instructions.
4. Shaking incubator.
5. LB broth for the dilution of the bacterial cell suspension:
prepare according to the manufacturer’s instructions.
6. Agar plates containing ampicillin (working concentration 100
μg/mL).
7. 1.5 mL centrifuge tubes.
8. Water bath.
2.3 Conjugative 1. E. coli donor strain containing shuttle vector carrying gene of
Transfer of Shuttle interest.
Vector into Bt 2. E. coli helper strain, e.g., J53/R751 [19].
3. 5 mL LB bottle with ampicillin 100 μg/mL (or another suit-
able antibiotic).
4. 5 mL LB bottle with trimethoprim 200 μg/mL (or another
suitable antibiotic)
5. Brain heart infusion (BHI) agar supplemented with hemin and
antibiotics (gentamicin 200 μg/mL and/or erythromycin 5
μg/mL, or another suitable antibiotic): Dissolve 18.5 g of
BHI powder and 7.5 g of agar in 0.5 liter of deionized water
and add 0.75 μM of hemin (BHIH). Autoclave the medium
and leave for a minimum of 24 h in an anaerobic cabinet to fully
deoxygenate.
6. Filter disc 0.45 μm pore size, 25 mm.
7. 50 mL conical centrifuge tubes.
8. Tweezers.
9. Sterile 25 mL wide neck universal glass bottle.
2.4 Assessing 1. BHIH agar plates containing gentamicin 200 μg/mL and
Protein Expression erythromycin 5 μg/mL.
and Secretion into BEV 2. Sterile inoculation loop(s).
2.4.1 Culture of Bt 3. 20 mL BHIH containing erythromycin 5 μg/mL in Universal
Transconjugants bottles.
4. Anaerobic cabinet.
5. 50 mL conical centrifuge tubes.
6. Refrigerated centrifuge.
7. 20 C freezer.
2.7 EVs Purification In this part of the chapter, the use of size-exclusion chromatogra-
phy is described for the removal of remaining proteins. Two
options are proposed: 2.7.1 and 2.7.2 for an increased resolution.
3 Methods
3.1 Synthetic Gene The gene can be synthesized de novo using commercial gene syn-
Design thesis services. The N-terminus of the protein of interest is fused in
frame to the signal peptide of the product of BT_3852 (OmpA of
Bt); MKKILMLLAFAGVASVASA. The chimeric protein sequence
is tested in silico for cleavage of the OmpA signal sequence using
http://www.cbs.dtu.dk/services/SignalP/. If unsuccessful,
change or add amino acids as appropriate to the N-terminus of
the gene of interest, downstream from the signal peptide sequence.
To facilitate immunodetection and/or purification of the protein in
downstream applications, a fusion tag can be added to the gene. It
is important that the coding sequence of the desired protein incor-
porates codon usage optimization for expression in Bt, which is
usually provided as part of gene synthesis services. The desired
target sequence is then integrated into an acceptor vector.
3.2 Cloning of Gene 1. Digest the plasmid containing the synthetic gene with restric-
of Interest tion enzymes to excise the gene from the vector carrying the
synthetic gene (e.g., pEX-K168).
3.2.1 Generation
of Recombinant DNA 2. Digest the E. coli/Bacteroides shuttle vector pGH90 (see Sub-
heading 2.1) (see Note 6).
3. Ligate the gene into the digested pGH090 expression vector to
allow translational fusion (see Note 7).
4. Place the vial with the bacteria suspension on ice for 30 min.
5. Induce a heat-shock at 42 C for 30 s.
6. Incubate the bacteria on ice for an additional 5 min.
7. Add 950 μL S.O.C. medium.
8. Incubate at 37 C with agitation (250 rpm).
9. Seed 100 μL of the bacterial suspension onto agar plates con-
taining ampicillin.
10. Incubate the plates for 16–18 h at 37 C.
3.2.3 Screening 1. Pick individual colonies from agar plates and add each colony
of Cloned to a tube containing 5 mL LB medium and ampicillin.
Recombinant DNA 2. Incubate the liquid cultures at 37 C and 250 rpm for 16–18 h.
3. Isolate plasmid DNA from each culture using a small-scale
isolation kit (e.g., QIAprep Spin Miniprep Kit) according to
the manufacturer’s instructions.
4. Digest the plasmid DNA using the appropriate restriction
enzymes (see Note 8).
5. Resolve digested DNA on a TBE gel.
6. Confirm the identity of the plasmids containing the insert of
the expected size by DNA sequencing using appropriate
primers.
3.3 Conjugative 1. Prepare BHIH agar plates either with or without gentamycin
Transfer of Shuttle and erythromycin.
Vector into Bt 2. Grow cultures of the E. coli donor strain with ampicillin (con-
3.3.1 Triparental Mating taining the plasmid with the correct inserted sequence, see
Procedure Subheading 3.2.3) and the E. coli helper strain with trimetho-
prim in 10 mL of LB, at 37 C, under agitation for 16–18 h. In
parallel, grow culture of the Bt recipient strain in BHIH in an
anaerobic cabinet at 37 C for 16–18 h.
3. Inoculate 10 mL of LB with 100 μL of the E. coli donor strain
and the helper strain (no antibiotics added) cultures and incu-
bate for 2 h at 37 C with agitation (e.g., 200 rpm). In parallel,
inoculate in 30 mL BHIH contained in a 50 mL tube with 800
μL of the Bt culture and incubate for 2 h at 37 C in an
anaerobic cabinet.
4. Add donor and helper cultures to the Bt recipient in the 50 mL
tube, mix by vortexing briefly and centrifuge at 2000 g for
15 min at 20 C.
5. Remove the supernatant and resuspend cells in 100 μL of
BHIH. Transfer cell suspension to the surface of a sterile 0.45
μm filter placed on a BHIH agar plate. Incubate the plate
aerobically for 16–18 h at 37 C.
180 Régis Stentz et al.
3.4 Assessing 1. Pick 4 individual colonies and restreak each one on separate
Protein Expression BHIH agar plates containing gentamicin and erythromycin.
and Secretion into 2. Incubate the plates anaerobically at 37 C for 48 h.
BEVs
3. Inoculate bottles containing 20 mL of BHIH with each of the
3.4.1 Culture of Bt 4 isolated clones.
Transconjugants 4. Incubate the bottles anaerobically at 37 C for 48 h.
5. Centrifuge the 20 mL of culture in 50 mL tubes, at 6000 g
for 15 min, at 4 C.
6. Collect the supernatant.
7. Wash cell pellets once in PBS before storing at 20 C prior to
analysis.
3.4.2 Cell Total Protein 1. Resuspend thawed cell pellets in 250 μL of 0.2 M Tris–HCl
Extraction (pH 7.2).
2. Disrupt the cells via sonication using eight 10-s pulses (ampli-
tude, 6 μm), with 30-s pauses on ice between each pulse.
3. Cell extracts are obtained after centrifugation at 14,000 g for
30 min at 4 C and harvesting the supernatant.
4. Measure total protein concentration of each supernatant/sam-
ple using the Bio-Rad protein assay according to the manufac-
turer’s instructions using BSA to generate a standard curve.
3.4.3 BEV Total Protein 1. Filter the 20 mL supernatants through 0.22 μm pore-size PES
Extraction membranes to remove debris and cells.
2. Concentrate the supernatants by ultrafiltration (100 kDa
molecular weight cutoff, Vivaspin 20) to a final volume of
250 μL.
3. Discard the filtrate.
4. Rinse the retentate with 20 mL of 0.2 M Tris–HCl, pH 7.2 and
concentrated to 250 μL.
5. Collect the retentate and disrupt the vesicles via sonication
using eight 10-s pulses (amplitude, 6 μm), with 30-s pauses
on ice between each pulse.
6. Measure the total protein content and concentration using the
Bio-Rad protein assay according to the manufacturer’s instruc-
tions using BSA to generate a standard curve.
Production, Isolation, and Characterization of Bioengineered Bacterial. . . 181
3.4.4 Protein Western 1. Add BEV and cell extracts obtained in Subheading 3.4.3 to
Blotting/Antigen loading buffer containing 0.4 M freshly prepared dithiothreitol
Immunodetection (DTT).
2. Load 7 μg of the total protein onto a 12% precast gel and
separate by electrophoresis at 180 V for 40 min.
3. Transfer the proteins from the gel onto a polyvinylidene
difluoride (PVDF) membrane using the XCell II™ Blot Mod-
ule or equivalent (according to the manufacturer’s instruc-
tions) at 25 V for 2 h in a solution containing Tris-Glycine
transfer buffer and methanol 20% (v/v).
4. Incubate the membrane with blocking buffer by gently shaking
for 30 min at 20 C using an orbital shaker.
5. Discard the blocking solution and incubate the membrane for
16–18 h at 4 C in blocking buffer containing primary anti-
body (Usually 1:1000 to 1:10000).
6. After washing 3 times with TBST, membranes are incubated
with HRP-conjugated secondary antibody in blocking buffer
for 1 h at 20 C.
7. After 3 washes with TBST, Enhanced Chemiluminescent sub-
strate (ECL) is added to detect bound antibody.
3.4.6 BDM+ Bacterial Initially BHIH was used as a standard medium for generating Bt
Culture BEVs [8, 16]. However, considering the need to reduce and even
exclude animal-derived products from medical/therapeutic formu-
lations to be used in humans, we have modified a chemically defined
Bacteroides growth media (BDM) [20] (see Subheading 2.5.2) for
Bt BEV production.
1. Inoculate 10 mL of BHIH with a frozen stock for ~16 h.
2. Inoculate 10 mL of BDM+ with 100 μL BHIH culture from
step 1 for ~8 h.
3. Inoculate 500 mL of BDM+ with 0.5 mL of the preinoculum
for 17 h (starting OD600 ~ 0.005) with gentle stirring.
3.5 Isolation of BEVs During the filtration process, the membrane will retain any mole-
cules >100 kDa including BEVs (retentate) and concentrate them
in the reservoir, whereas molecules <100 kDa will be removed in
the flow-through (filtrate) directly to the waste. BEVs isolation
should be performed at ambient (20–22 C) temperature.
Three procedures are used in a step wise manner for isolating
BEVs, which are illustrated in Fig. 2.
Fig. 2 BEV isolation module system. Option 1. Sample concentration. Option 2. Module rinsing/decontamina-
tion. Option 3. Collection of concentrated samples
12. Place 0.5–1 mL of PBS in the reservoir and pump through the
system at speed setting 1–2 to flush out remaining BEVs.
Collect in the original 15 mL tube or microcentrifuge tube.
Final volume collected depends on concentration of BEVs
needed.
13. Switch off the pump.
14. Centrifuge at 15,000 g for 20 min at 4 C to remove any
precipitate.
15. Filter-sterilize the supernatant using a 0.22 μm syringe filter,
collecting the filtrate in sterile 1.5 mL lo-bind microcentrifuge
tubes or 15 mL tube.
3.6 BEVs Purification Contaminants (e.g., proteins) of BEV preparations can be removed
by size exclusion chromatography (SEC). We recommend using the
method described in Subheading 3.7, step 1 for routine prepara-
tions and in Subheading 3.7, step 2 if increased resolution is
needed, for instance, to size fractionate BEVs.
3.6.1 Routine Purification 1. Bring the BEVs preparation, column buffer (PBS pH 7.4) and
qEVoriginal/35 nm SEC column(s) to ~20 C. Do not remove
the column caps until operational temperature is reached.
2. Secure the column in a vertical position using a stand.
3. Carefully remove the column top-cap.
4. Attach a column reservoir (if available) and add 1.5 column
volume of buffer (PBS 1, 15 mL).
5. Remove the bottom column cap and allow the buffer to run
under gravity to waste.
6. If any buffer other than PBS is used, flush with at least 3 column
volumes of the buffer (>30 mL).
7. The column will stop flowing when the buffer has entered the
loading frit.
8. Load 0.5–1 mL of isolated BEVs onto the loading frit.
Production, Isolation, and Characterization of Bioengineered Bacterial. . . 185
3.7 BEVs Size Size and concentration of isolated BEVs suspension is determined
and Concentration by nanoparticle tracking analysis (NTA) using a suitable NTA
Analysis instrument. The protocol described below is for the ZetaView
PMX-220 TWIN instrument from Particle Metrix GmbH.
1. Prepare instrument set up according to the manufacturer’s
instructions.
2. Inject 10–20 mL particle-free deionized water using a 10 mL
syringe to perform cell quality check; avoid injecting air
bubbles.
3. Inject 5–10 mL of 1:25,000 100 nm polystyrene standard bead
suspension to perform focus auto-alignment.
4. Inject 10–20 mL particle-free deionized water using a 10 mL
syringe to rinse instrument; avoid injecting air bubbles.
5. Dilute aliquots of BEVs suspension in 1:1000 to 1:20,000 in
particle-free deionized water.
6. Inject 1 mL sample with a syringe; avoid injecting air bubbles.
186 Régis Stentz et al.
4 Notes
Acknowledgments
References
14. Smith CJ, Rocha ER, Paster BJ (2006) The 19. Shoemaker NB, Getty C, Gardner JF et al
medically important Bacteroides spp. in health (1986) Tn4351 transposes in Bacteroides spp.
and disease. In: Dworkin M, Falkow S, and mediates the integration of plasmid R751
Rosenberg E, Schleifer KH, Stackebrandt E into the Bacteroides chromosome. J Bacteriol
(eds) The prokaryotes. Springer, New York, 165:929–936. https://doi.org/10.1128/jb.
NY. https://doi.org/10.1007/0-387-30747- 165.3.929-936.1986
8_14 20. Bryant WA, Stentz R, Le Gall G et al (2017) In
15. Human Microbiome Project Consortium silico analysis of the small molecule content of
(2012) Structure, function and diversity of outer membrane vesicles produced by Bacter-
the healthy human microbiome. Nature oides thetaiotaomicron indicates an extensive
486:207–214. https://doi.org/10.1038/ metabolic link between microbe and host.
nature11234 Front Microbiol 8:2440. https://doi.org/10.
16. Carvalho AL, Miquel-Clopés A, Wegmann U 3389/fmicb.2017.02440
et al (2019) Use of bioengineered human com- 21. Horn N, Carvalho AL, Overweg K et al (2016)
mensal gut bacteria-derived microvesicles for A novel tightly regulated gene expression sys-
mucosal plague vaccine delivery and immunisa- tem for the human intestinal symbiont Bacter-
tion. Clin Exp Immunol 196:287–304. oides thetaiotaomicron. Front Microbiol
https://doi.org/10.1111/cei.13301 7:1080. https://doi.org/10.3389/fmicb.
17. Valkenburg SA, Mallajosyula VV, Li OT et al 2016.01080
(2016) Stalking influenza by vaccination with 22. Durant L, Stentz R, Noble A et al (2020)
pre-fusion headless HA mini-stem. Sci Rep Bacteroides thetaiotaomicron-derived outer
6:22666. https://doi.org/10.1038/ membrane vesicles promote regulatory den-
srep22666 dritic cell responses in health but not in inflam-
18. Wegmann U, Horn N, Carding SR (2013) matory bowel disease. Microbiome 8:88.
Defining the Bacteroides ribosomal binding https://doi.org/10.1186/s40168-020-
site. Appl Environ Microbiol 79:1980–1989. 00868-z
https://doi.org/10.1128/AEM.03086-12
Chapter 12
Abstract
Extracellular membrane vesicles (EMVs) produced by Gram-negative bacteria are useful as a vaccine
platform. During growth in broth at 18 C, Shewanella vesiculosa HM13 produces a large number of
EMVs that contain a 49-kDa major cargo protein, named P49. Enhanced green fluorescent protein fused to
the C-terminus of P49 is delivered to EMVs, suggesting that P49 is useful as a carrier to target foreign
proteins to EMVs for production of artificial EMVs with desired functions. This method is potentially
useful for the preparation of designed vaccines and is described in detail in this chapter.
Key words Extracellular membrane vesicles, Protein fusion, Recombinant protein production, Pro-
tein secretion, Protein transport
1 Introduction
191
192 Jun Kawamoto and Tatsuo Kurihara
Fig. 1 EMVs and their cargo proteins from S. vesiculosa HM13. (a) Field emission-scanning electron
microscopy image of S. vesiculosa HM13. Blebs on the cell surface and vesicles secreted into the extracellular
milieu were observed. White triangles indicate blebbing of the outer membrane to produce EMVs. Bar
indicates 100 nm. (b) Transmission electron microscopy (TEM) observation of EMVs. TEM images demonstrate
that S. vesiculosa HM13 produces EMVs with a uniform diameter of approximately 50 nm. The inset
represents a magnified image of the boxed area. Bar indicates 500 and 50 nm, respectively. (c) SDS-PAGE
gel image of PVF and purified EMVs. The EMVs of S. vesiculosa HM13 carry a single major cargo protein, P49.
(Parts of this figure have been reproduced/modified from [8], with permission from Frontiers Media SA)
2 Materials
3 Methods
Table 1
Primers used in PCR reactions
a
Complementary sequences used to fuse amplicons for construction of pKP49eGFP are indicated, where appropriate, by
matched text color
Kanr
Kanr
oriT
pKNOCK-Km pKP49eGFP
oriT R6K
Plasmid eGFP -ori
construction 3 -region of P49-
R6K-ori coding gene w/o
stop codon
Conjugative transformation
& homologous recombination
S. vesiculosa HM13-Rifr
genome stop
P49-eGFP
genome Kanr
stop stop
P49-gene 3 - pKNOCK-specific primer
region primer (pK-check-rev)
(P49C-fwd)
Fig. 2 Schematic illustration of the construction of the eGFP-fused P49 expression strain of S. vesiculosa
HM13. A 523 bp sequence corresponding to the 30 end of the P49 gene was fused to the 717 bp eGFP gene,
and cloned into the pir-dependent suicide plasmid, pKNOCK. Integration of the plasmid into the genome by
homologous recombination was confirmed by PCR with a set of primers annealing to the P49 gene and the
pKNOCK-specific region
15. For further confirmation, the plasmids were extracted from the
transformants and analyzed by sequencing using primers
pK-check-fwd/pK-check-rev.
16. Store glycerol stocks of confirmed culture at 80 C until use
as plasmid-donor cells.
3.1.3 Verification 1. Prepare a standard Taq polymerase PCR reaction mixture con-
of Plasmid Integration into taining a P49C-fwd and pK-check-rev (Fig. 2), allowing
the S. vesiculosa 10–20 μL per colony to be tested.
HM13-Rifr Chromosome 2. Aliquot 10–20 μL PCR reaction mixture, and mix in single
colonies to be tested, in respective PCR tubes. Amplify
30 cycles using manufacturer’s recommendations for times
and temperatures for each step.
3. Analyze 10 μL of each PCR product by 1% agarose gel
electrophoresis.
4. Check the size of amplified gene fragments, which should be
1,304 bp for correct clones.
5. Isolate single colonies for which targeted plasmid integration
was detected and preserve confirmed S. vesiculosa P49-eGFP
cultures using cryopreservation beads at 80 C until use.
198 Jun Kawamoto and Tatsuo Kurihara
3.3 EMV Preparation 1. Transfer the 5 mL culture to 1.5 mL tubes and pellet the
bacterial cells by centrifugation at 6,800 g and 4 C for
15 min.
2. Carefully transfer the supernatants to new 1.5 mL tubes.
3. Collect the cell pellets into a single 1.5 mL tube and wash the
combined pellet three times with 1 mL of fresh LB medium,
discarding the supernatant after each wash (see Note 13).
4. Keep cells on ice and use them for soluble and insoluble protein
preparation described in Subheading 3.4.1, or store at 80 C
until use.
5. Centrifuge the supernatants from step 2 at 13,000 g and
4 C for 15 min to remove possible contaminants such as
broken cell debris in EMV-containing fraction.
6. Filter the supernatants through a 0.45-μm PES syringe filter to
remove residual cellular materials.
7. Transfer the filtrates into a 16 76 mm polycarbonate ultra-
centrifuge tube, prepare a weight-matched balance tube, and
ultracentrifuge at 100,000 g and 4 C for 2 h (see Note 14).
8. Transfer the supernatant (see Note 15) to new 1.5 mL tubes
and store as a post-vesicle fraction (PVF) at 80 C until use.
9. Add 1 mL DPBSS to the ultracentrifuge tubes and suspend the
pellet (see Note 16).
10. Collect the EMV suspension into 1.5 mL tubes (see Note 17)
for Western blotting, or store at 80 C until use.
Low Temperature Production of Engineered Vesicles 199
3.4 Localization 1. Suspend the cells (prepared in Subheading 3.3, step 4) in 10
Analysis of P49-Fusion volume of DPBSS (typically 300–500 μL for cells from a 5 mL
Protein culture sample).
3.4.1 Preparation
2. Sonicate cells on ice for 3 min (0.5 s ON, 1 s OFF, at 20%
of Cellular Protein Samples amplitude).
3. Transfer the suspension to a new 1.5 mL tube and pellet
undisrupted cells at 20,500 g and 4 C for 15 min.
4. Transfer the supernatant into a 16 76 mm polycarbonate
ultracentrifuge tube, add 5 mL DPBSS, and prepare a weight-
matched balance tube (see Note 14).
5. Ultracentrifuge at 100,000 g and 4 C for 2 h.
6. Collect the supernatant as soluble protein fraction and store at
80 C for Western blot analysis.
7. Add 50 μL DPBSS into the ultracentrifuge tube to resuspend
the pellet by pipetting or gently vortexing.
8. Transfer the suspension to a new 1.5 mL tube.
9. Store the suspension as insoluble protein fraction at 80 C for
Western blot analysis.
3.4.2 Sample 1. Add 1/10 volume of 100% TCA to each of the four samples
Preparation for SDS-PAGE (i.e., soluble and insoluble cell protein fractions, PVF,
and Western Blotting and EMVs).
2. Vortex and incubate on ice for 30 min.
3. Centrifuge at 20,500 g and RT for 30 min.
4. Discard supernatants.
5. Add 20 μL prechilled acetone to each tube and wash by
pipetting.
6. Centrifuge at 20,500 g and RT for 30 min.
7. Discard supernatants and repeat the wash step twice.
8. Air-dry the pellets for 5 min.
9. Add 40 μL of 1 SDS-sample buffer to each tube and suspend
samples by pipetting.
10. Boil the suspensions at 100 C for 5 min and cool at RT.
3.4.3 SDS-PAGE 1. For each sample prepared in Subheading 3.4.2, mix 2.4 μL
and Western Blotting with 27.6 μL of 1 SDS-sample buffer and load 10 μL
(corresponding to 100 μL culture) to respective lanes of a
precast 5–20% gradient polyacrylamide gel.
2. Run the gel at 120 V in 1 SDS-running buffer until the dye
front reaches the bottom of the gel.
3. For semidry Western-blot transfer of the proteins (see Note
18), cut the PVDF membrane to the size of the separation
200 Jun Kawamoto and Tatsuo Kurihara
P49-eGFP
Cell
l.
l. F Vs
so EM
So In PV
75 P49-fused
eGFP
30
P49-free
eGFP
(Mass/kDa)
4 Notes
Acknowledgments
References
1. Schwechheimer C, Kuehn MJ (2015) Outer- 3. Nagakubo T, Nomura N, Toyofuku M (2020)
membrane vesicles from Gram-negative bacte- Cracking open bacterial membrane vesicles.
ria: biogenesis and functions. Nat Rev Micro- Front Microbiol 10:3026. https://doi.org/
biol 13:605–619. https://doi.org/10.1038/ 10.3389/fmicb.2019.03026
nrmicro3525 4. Anand D, Chaudhuri A (2016) Bacterial outer
2. Toyofuku M, Tashiro Y, Hasegawa Y et al membrane vesicles: new insights and applica-
(2015) Bacterial membrane vesicles, an over- tions. Mol Membr Biol 33:125–137. https://
looked environmental colloid: biology, envi- doi.org/10.1080/09687688.2017.1400602
ronmental perspectives and applications. Adv 5. McCarthy PC, Sharyan A, Sheikhi Moghad-
Colloid Interf Sci 226:65–77. https://doi. dam L (2018) Meningococcal vaccines: current
org/10.1016/j.cis.2015.08.013 status and emerging strategies. Vaccines (Basel)
Low Temperature Production of Engineered Vesicles 205
Abstract
Bacteria are known to release nanometer scale proteoliposomes termed bacterial membrane vesicles (MVs),
and it is considered that native and bioengineered MVs would be applicable for development of acellular
vaccines and novel drug delivery systems in medical settings. However, important considerations for
manufacturing purposes include the varied productivity of MV among bacterial species and strains, as
well as endotoxicity levels due to the lipopolysaccharide component. The method for MV induction using
glycine described here is simple and provides a solution to these problems. Glycine weakens bacterial
peptidoglycans and significantly increases bacterial MV formation, while the relative endotoxin activity of
glycine-induced MVs is extremely reduced as compared to that of noninduced MVs. Nevertheless, glycine-
induced MVs elicit strong immune responses at levels nearly equivalent to those of noninduced MVs. Taken
together, the present method for induction by glycine is convenient for research studies of bacterial MVs
and has potential for use in medical applications including vaccine development.
1 Introduction
207
208 Satoru Hirayama and Ryoma Nakao
Fig. 2 Noninduced and glycine-induced MVs of E. coli. Noninduced MVs (a) and glycine-induced MVs (b) of a
flagellar-deficient derivative of E. coli Nissle 1917 were purified by ultracentrifugation, then observed by
transmission electron microscopy. The mean of diameter of glycine-induced MVs was apparently greater than
that of noninduced MVs. No flagella were observed as the flagella master regulator gene flhD was deleted in
both preparations [5], while fimbrial structures (black arrow heads) associated with MVs were noted in both.
Scale bars represent 100 nm
2 Materials
2.2 Quantification 1. Bovine serum albumin (BSA) solution: Dissolve BSA in dis-
of MV Components tilled water to obtain 10 mg/mL stock solution. Dispense
aliquots into microtubes and store at 20 C.
2.2.1 Quantification
of the Amount of Total 2. 96-well microtiter plates (see Note 7).
Protein in MVs
Low Endotoxin Glycine-Induced Membrane Vesicles 211
2.3 Evaluation In 1. Mouse macrophage-like cell line J774.1 (see Note 10).
Vitro: 2. Supplemented RPMI 1640 medium (S-RPMI): RPMI 1640
Cytokine-Inducing supplemented with 10% heat-inactivated fetal calf serum,
Activity of MVs 100 units/mL penicillin G, and 0.1 mg/mL streptomycin
2.3.1 Addition of MVs
sulfate.
to Macrophage-like Cells 3. 90-mm nontreated culture dishes (see Note 10).
and Sample Preparation 4. CO2 incubator.
5. Microscope.
6. Centrifuge.
7. 50-mL polypropylene centrifuge tubes.
8. Trypan blue solution: 0.4% (w/v) trypan blue dissolved in
distilled water.
9. Cell counting system.
212 Satoru Hirayama and Ryoma Nakao
2.3.2 Quantitative 1. RNA purification kit suitable for use with cultured cells (e.g.,
Real-Time PCR RNeasy Mini Kit, QIAGEN, Hilden, Germany).
2. Centrifuge.
3. 15-mL or 50-mL polypropylene centrifuge tubes.
4. Real-time PCR cDNA synthesis kit (e.g., ReverTra Ace qPCR
RT Master Mix with gDNA Remover, TOYOBO, Tokyo,
Japan).
5. 96-well PCR plates with plate seals.
6. Thermal cycler.
7. Forward and reverse primers and TaqMan probes for detection
of the expression of the target cytokines and β-actin internal
control (Table 1).
8. Enzyme and buffer for real-time PCR (e.g., Premix Ex Taq
[Probe qPCR], TaKaRa Bio, Shiga, Japan).
9. TE buffer: 10 mmol/L Tris-HCl (pH 8.0), 1 mmol/L EDTA
(pH 8.0), autoclaved.
10. DNA standard for absolute quantification: Prepare template
DNA of interest (e.g., plasmid cloned target sequences or
synthesized gene). Adjust to 0.5 1010 copies/μL with TE
buffer.
11. Real-time PCR instrument.
Table 1
Primers and probes for quantitative real-time PCR
3 Methods
3.1 Induction 1. For seed culture, pick a single colony of E. coli from an over-
of E. coli MV night LB agar plate culture and inoculate a small amount (e.g.,
Production by Addition 2 mL) of LB broth. Incubate at 37 C for 8 h with shaking at
of Glycine ca. 150 rpm.
2. Inoculate 1/200 volume of E. coli culture into 80 mL of LB
broth without glycine (noninduction control) (see Note 15),
and LB broth with glycine added to a final concentration of
~2.0% (see Note 16). Incubate at 37 C for 16 h with shaking at
ca. 150 rpm (see Note 17).
3. Centrifuge bacterial cultures at 7,200 g for 30 min at 4 C
and collect the culture supernatants.
4. Filter culture supernatants using 0.45-μm PVDF filters to
completely remove remaining bacterial cells (see Note 4).
5. Ultracentrifuge culture supernatants at 103,800 g for 2 h at
4 C to obtain MV pellets.
6. Suspend MV pellets in small amount (e.g., 1.5 mL) of PBS or
20 mM Tris-HCl (pH 8.0) (see Notes 6, 18 and 19).
3.2 Quantification The amounts of total protein and total lipid in MVs can be quanti-
of MV Components fied by the Bradford and FM4-64-based methods, respectively. We
routinely perform the Bradford method for quality control of MVs,
and sometimes use the FM4-64-based method as an alternative
quality control technique. The endotoxin activity of MVs can be
also measured by the Limulus assay and normalized by the amount
of protein or lipid contained in MVs.
3.2.1 Quantification 1. Make a two-fold serially diluted solution (e.g., 0.1–1000 μg/
of the Amount of Total mL) of standard BSA with distilled water.
Protein in MVs 2. Dilute MV samples two-fold serially with distilled water (see
Note 20).
3. Pipette 10 μL each of standard BSA and MV samples into
appropriate wells of a 96-well microtiter plate (see Notes 7
and 21).
4. Add appropriate volume (e.g., 200 μL/well) of dye reagent for
Bradford protein assay (see Note 22).
5. Incubate the plate for 5–15 min at room temperature.
6. Measure absorbance at 595 nm.
7. Estimate the amount of protein contained in the MV samples
from the calibration curve created by the BSA standards.
216 Satoru Hirayama and Ryoma Nakao
3.3 Evaluation In This section presents two methods for assessing MV immunoactiv-
Vitro: ity using a mouse macrophage-like cell line; quantification of intra-
Cytokine-Inducing cellular cytokine mRNA expression levels and determination of the
Activity of MVs amount of cytokines into the medium. Different cytokines (and/or
their specific mRNA expression profiles) may be evaluated, affect-
ing the choice of reagents used for the real-time PCR and ELISA
methods described. We normally assay for IL-6, IL-12 and TNF-α,
and β-actin for internal control.
3.3.2 Quantitative 1. Prepare total RNA from collected cells prepared in Subheading
Real-Time PCR 3.3.1 using an RNA purification kit appropriate for culture
cells, according to the manufacturer’s instructions.
2. Synthesize cDNA from total RNA using the cDNA synthesis
kit for real-time PCR, according to the manufacturer’s
instructions.
3. Prepare a master mixture for quantitative real-time PCR con-
taining 0.04 μM each of forward and reverse primers and
probe, specific for the cytokine-encoding mRNA to be quanti-
fied, and the appropriate concentration of enzyme and buffer
(20 μL/PCR reaction).
4. Dispense 18 μL/well of the master mixture into a 96-well real-
time PCR plate.
5. Dilute the appropriate standard plasmid tenfold serially (e.g.,
100–109 copies per 2 μL) with sterile water or TE buffer.
6. Add 2 μL each of the serially diluted standard plasmid and the
cDNA samples to appropriate wells (Fig. 3) containing the
PCR mixture dispensed in step 4.
7. Seal the plate and perform real-time PCR reaction (1 cycle of
95 C for 30 s, 40 cycles consisting of 95 C for 5 s and 60 C
for 34 s). Set to measure the fluorescence of the sample at
60 C.
8. Create a calibration curve using Ct (threshold cycle) values of
standards and plasmid copy numbers. Use the calibration curve
to determine the copy number of the target gene contained in
the cDNA sample (see Note 25).
3.3.3 Cytokine ELISA 1. To coat an ELISA plate with a capture antibody specific for the
cytokine to be quantified, dilute the antibody with PBS and
dispense 100 μL/well into a 96-well microtiter plate.
2. Seal the plate with plastic wrap and incubate overnight at room
temperature.
3. Remove contents and wash the plate with 300 μL/well PBST
four times.
4. Dispense 300 μL/well blocking buffer into the plate.
5. Seal the plate with plastic wrap and incubate for 1 h at room
temperature.
6. Remove contents and wash the plate with 300 μL/well PBST
four times.
7. Twofold serially dilute the recombinant cytokine protein stan-
dard (e.g., ranging from 28 to 21 ng/mL) with PBST. Also,
dilute culture supernatants prepared in Subheading 3.3.1 with
PBST (see Note 26).
Low Endotoxin Glycine-Induced Membrane Vesicles 219
Fig. 3 Example 96-well plate layout for real-time PCR detection of cytokine expression. Dilutions of standards
are tested in duplicate, whereas each of the 21 cDNA samples from stimulated J774.1 cells are tested in
triplicate. NC ¼ negative control
Fig. 4 Example 96-well plate layout for detection of cytokine concentration by ELISA. Dilutions of recombinant
cytokine standards are tested in duplicate, whereas each of the 21 culture supernatant samples from
stimulated J774.1 cells may be tested either in triplicate for a single dilution, or in three different concentra-
tions (serial twofold dilutions). NC ¼ negative control
13. Remove contents and wash the plate with 300 μL/well PBST
four times.
14. Add 100 μL/well of enzyme-conjugated avidin, appropriately
diluted with diluent, as recommended by the manufacturer.
15. Seal the plate with plastic wrap and incubate at room tempera-
ture for 30 min.
16. Remove contents and wash the plate with 300 μL/well PBST
four times.
17. Add 100 μL/well of substrate solution into the plate.
18. Sequentially (e.g., 10 min, 30 min, 60 min, and 120 min)
measure the absorbance of 405 nm (for AP) or 650 nm (for
HRP) (see Note 27).
3.4 Evaluation In Intranasally immunize mice with OVA as an antigen and bacterial
Vivo: Mucosal MVs as an adjuvant to evaluate the adjuvant activity of MVs. The
Adjuvanticity of MVs immunization schedule is shown in Fig. 5.
3.4.2 Sample Collection 1. To collect saliva samples, inject 200 μL of the parasympathetic
from Mice stimulant solution into the abdominal cavity. When saliva secre-
tion is induced, collect saliva from the mouth with a pipette.
Store at 80 C (see Note 31).
2. For serum samples, collect whole blood from anesthetized
mice by cardiac puncture through the diaphragm with 1-mL
syringe and 26G needle (see Notes 32 and 33). Centrifuge
blood samples at 300 g for 10 min to precipitate blood
cells, collect the serum in new microtubes. Store at 20 C.
3. For nasal wash samples, remove the head of the deceased
mouse after exsanguination and dissect out the lower jaw.
Insert a syringe needle (21G nonbeveled) into the nasal cavity
from the posterior opening, and flush with 1 mL of PBS con-
taining 0.1% BSA. Collect the outflow from the nostrils in 1.5-
mL microtube. Repeat the flushing step three times, pool the
replicates and centrifuge at 300 g for 10 min to remove cell
debris. Store at 20 C.
3.4.3 ELISA The samples collected in Subheading 3.4.2 may be assayed for
for Evaluation of Mucosal OVA-specific IgG, IgA, IgM, and IgE antibodies, as desired, by
Adjuvanticity of MVs using the appropriate enzyme-linked detection antibody. Each type
of sample (i.e., saliva, serum, nasal wash) is tested separately, but a
single ELISA plate can be used to assay for two types of
OVA-specific antibodies, for example, IgG and IgA (Fig. 6).
1. To coat ELISA plates, dispense 100 μL/well of 0.1 μg/μL
OVA in ELISA coating buffer (i.e., 10-μg OVA per well).
2. Seal the plate with plastic wrap and incubate overnight at 4 C.
3. Remove contents and wash the plate with 300 μL/well PBST
three times.
4. Dispense 150 μL/well of 1% skim milk in PBST to block.
222 Satoru Hirayama and Ryoma Nakao
Fig. 6 Example 96-well plate layout for samples assayed for OVA-specific IgG and IgA antibodies. Sets of
samples (saliva, serum, or nasal wash) are tested in duplicate (each at a single dilution) for OVA-specific IgG
(top half of the plate) and IgA (bottom half of the plate) by using the appropriate enzyme-linked detection
antibody
4 Notes
Acknowledgments
References
1. Schwechheimer C, Kuehn MJ (2015) Outer- 8. Rembacken B, Snelling A, Hawkey P,
membrane vesicles from gram-negative bacte- Chalmers D, Axon A (1999) Non-pathogenic
ria: biogenesis and functions. Nat Rev Micro- Escherichia coli versus mesalazine for the treat-
biol 13(10):605–619 ment of ulcerative colitis: a randomised trial.
2. McBroom AJ, Johnson AP, Vemulapalli S, Lancet 354(9179):635–639
Kuehn MJ (2006) Outer membrane vesicle 9. Kruis W, Frič P, Pokrotnieks J, Lukáš M,
production by Escherichia coli is independent Fixa B, Kaščák M, Kamm M, Weismueller J,
of membrane instability. J Bacteriol 188 Beglinger C, Stolte M (2004) Maintaining
(15):5385–5392 remission of ulcerative colitis with the probiotic
3. Turnbull L, Toyofuku M, Hynen AL, Escherichia coli Nissle 1917 is as effective as
Kurosawa M, Pessi G, Petty NK, Osvath SR, with standard mesalazine. Gut 53
Cárcamo-Oyarce G, Gloag ES, Shimoni R (11):1617–1623
(2016) Explosive cell lysis as a mechanism for 10. Behnsen J, Deriu E, Sassone-Corsi M, Raffa-
the biogenesis of bacterial membrane vesicles tellu M (2013) Probiotics: properties, exam-
and biofilms. Nat Commun 7(1):1–13 ples, and specific applications. Cold Spring
4. Toyofuku M (2019) Bacterial communication Harb Perspect Med 3(3):a010074
through membrane vesicles. Biosci Biotechnol 11. Ichinohe T, Watanabe I, Ito S et al (2005)
Biochem 83(9):1599–1605 Synthetic double-stranded RNA poly(I:C)
5. Hirayama S, Nakao R (2020) Glycine signifi- combined with mucosal vaccine protects
cantly enhances bacterial membrane vesicle against influenza virus infection. J Virol 79
production: a powerful approach for isolation (5):2910–2919
of LPS-reduced membrane vesicles of probiotic 12. Hirayama S, Nakao R (2021) Intranasal vaccine
Escherichia coli. Microb Biotechnol 13 study using Porphyromonas gingivalis mem-
(4):1162–1178 brane vesicles: isolation method and applica-
6. Vollmer W, Blanot D, De Pedro MA (2008) tion to a mouse model. Methods Mol Biol
Peptidoglycan structure and architecture. 2210:157–166. https://doi.org/10.1007/
FEMS Microbiol Rev 32(2):149–167 978-1-0716-0939-2_15
7. Hammes W, Schleifer K, Kandler O (1973) 13. Smith PK, Krohn RI, Hermanson GT, Mallia
Mode of action of glycine on the biosynthesis AK, Gartner FH, Provenzano M, Fujimoto
of peptidoglycan. J Bacteriol 116 EK, Goeke NM, Olson BJ, Klenk D (1985)
(2):1029–1053 Measurement of protein using bicinchoninic
acid. Anal Biochem 150(1):76–85
Chapter 14
Abstract
Outer membrane vesicles (OMV) represent a promising platform for the development of vaccines against
bacterial pathogens. More recently, bacteria have been genetically modified to increase OMV yield and
modulate the design of resulting particles, also named generalized modules for membrane antigens
(GMMA). OMV/GMMA resemble the bacterial surface of the pathogen, where key antigens to elicit a
protective immune response are and contain pathogen-associated molecular patterns (e.g., lipopolysacchar-
ides, lipoproteins) conferring self-adjuvanticity. On the other hand, OMV/GMMA are quite complex
molecules and a comprehensive panel of analytical methods is needed to ensure quality, consistency of
manufacture and to follow their stability over time. Here, we describe several procedures that can be used
for OMV/GMMA characterization as particles and for analysis of key antigens displayed on their surface.
Key words Outer membrane vesicles, OMV, GMMA, Vaccines, Analytical methods
1 Introduction
227
228 Francesca Micoli et al.
lumen
LPS OM
PP PG
IM
Cytoplasm
Abbreviations
LPS: Lipopolysaccharides
OAg: O-antigen
OM: Outer membrane
PG: Peptidoglycan
PP: Periplasm
IM: Inner membrane
Table 1
Methods for OMV/GMMA characterization
2 Materials
2.3 Size Exclusion 1. Tosoh TSK gel 6000 PW column (30 cm 7.5 mm; cat.
High-Performance 805765) connected in series with a Tosoh TSK gel 4000 PW
Liquid column (30 cm 7.5 mm; cat. 805763) and with Tosoh TSK
Chromatography gel PWH guard column (7.5 cm 7.5 mm; cat. 806732).
with Multiangle Light 2. HPLC system equipped with UV, fluorimeter, and multiangle
Scattering static light scattering (MALS) detectors (e.g., DAWN
(HPLC-SEC/MALS) HELEOS II, Wyatt).
3. Filter PBS eluent through 0.22 μm filter before use.
2.6 Micro BCA 1. Micro BCA Protein Assay Kit (Thermo Fisher, cat. 23235).
2.7 Ultracentri- 1. Ultracentrifuge equipment with appropriate rotor (see Note 1).
fugation
Table 2
Preparation of acid solution “TFA-HCl Mixture”
TFA HCl
Final volume mL mL mL
20 2.6 17.4
25 3.3 21.7
30 3.9 26.1
35 4.6 30.4
40 5.2 34.8
45 5.9 39.1
50 6.5 43.5
55 7.2 47.8
60 7.8 52.2
65 8.5 56.5
2.15 Competitive- 1. 10 Sample Dilution buffer (SDB) (10 PBS containing 1.0%
ELISA (cELISA) BSA and 0.5% Tween 20): Add 10 PBS into a graduated
cylinder up to 1.8 L. Add gently 10.0 mL of Tween 20 into
the cylinder followed by 20.0 g BSA. Stir for 1 h and if needed
continue stirring until the BSA is completely dissolved. When
the BSA is completely dissolved, add 10 PBS up to 2 L and
stir again the solution for few min. Check the pH of the
solution (which should be in the range 6.5 0.5) using a pH
meter or a pH paper. Filter-sterilize the solution into two 1 L
bottles using two bottle-top 0.22 μm filter units.
2. SDB working solution: In a graduated cylinder add water up to
the volume needed to have a 1:10 dilution, according to the
total volume to be prepared. Add 10 SDB (e.g., one 50 mL
aliquot 10 SDB in 450 mL water). Stir for few min with a
magnetic stirrer. Filter-sterilize the solution into 1 L bottles
using a bottle-top 0.22 μm filter unit.
OMV/GMMA Analytics 237
2.16 OAg Extraction 1. 10 Acetate buffer (final 100 mM pH 3.9 after ten-fold dilu-
tion): Weigh 1099 mg of AcONa and transfer it in a 100 mL
graduated cylinder. Add, in sequence, 50 mL of water and
4950 μL of acetic acid. Add water up to 100 mL, insert a
magnetic stirring bar and homogenize the solution.
2.17 Size Exclusion 1. Tosoh TSK gel G3000 PWXL column (30 cm 7.8 mm; cat.
High-Performance 808021) with a Tosoh TSK gel PWXL guard column
Liquid (4.0 cm 6.0 mm; cat. 808033).
Chromatography, 2. Eluent 0.1 M NaCl, 0.1 M NaH2PO4, 5% ACN, pH 7.2:
Derivatization Weigh 5.85 g of NaCl, 3.22 g of NaH2PO4 and 10.37 g of
with Semicarbazide Na2HPO4 and transfer the powders inside a 1 L glass
(HPLC-SEC SCA) graduated cylinder. Add water up to 850 mL in the 1 L
238 Francesca Micoli et al.
2.19 Hestrin To prepare solutions from powders, use magnetic stir bars and a
Colorimetric Method magnetic stirrer to accelerate the dissolution.
1. 1 M Acetic acid: Dilute in a 100 mL glass graduate cylinder
5.7 mL of acetic acid up to 100 mL with water.
2. 1 mM AcONa solution, pH 4.5: Weigh 8.2 mg of AcONa
anhydrous and dissolve it in a 100 mL glass graduated cylinder
with 80 mL of water. The pH is adjusted to 4.5 with acetic acid
1 M solution. Calculate the total water volume of the solution
needed to get the correct concentration using the following
equation:
sodium acetate weight ðmgÞ
Total mL of water ¼
0:082
Add water to the cylinder in order to reach the calculated
volume.
3. 30 mg/mL acetylcholine: Resuspend the contents of a 150 mg
vial of acetylcholine chloride using 5 mL of AcONa solution
1 mM, pH 4.5. Prepare just prior use.
OMV/GMMA Analytics 239
3 Methods
3.1 Dot Dot blot/western blot are used for verifying OMV/GMMA iden-
Blot/Western Blot tity. Specific primary antibodies are used according to the key
antigen(s) present on the samples. In the Materials section we
have reported as an example the use of a primary antibody (and
corresponding secondary antibody) specific for S. Typhimurium
OAg, key component of S. Typhimurium GMMA [14].
1. Prepare a positive control (+ CTRL) by diluting the
OMV/GMMA sample to use as positive control to 8 μg/mL
(protein concentration) with PBS. Use PBS as negative control
( CTRL).
2. Dilute each OMV/GMMA sample to be analyzed to 8 μg/mL
(protein concentration) with PBS.
3. Cut the 0.2 μm polyvinylidene difluoride (PVDF) membrane
(other membrane materials could be more suitable, depending
on the product type) in order to have a resulting area large
enough to cover the Dot-blot apparatus area.
4. Wet the PVDF membrane in a plastic case with a suitable
volume of methanol for about 1 min, manually shaking the
case in order to make the membrane entirely wet.
5. Transfer the membrane in another case filled with PBS and
leave the membrane with PBS for 3 min, shaking manually
the case in order to facilitate the entire wetting of the
240 Francesca Micoli et al.
3.2 Dynamic Light DLS is one of the methods that can be used for OMV/GMMA
Scattering particle size determination [9]. This technique allows determina-
tion of a mean hydrodynamic diameter (Z-average diameter) and a
OMV/GMMA Analytics 241
10
0
0.1 1 10 100 1000 10000
Size (d.nm)
Fig. 3 HPLC-SEC analyses (fluorescence emission profiles) of a purified S. Enteritidis GMMA sample (a), of a
protein standards mixture (b) and of the GMMA sample spiked with such standard (c), showing ability of this
method to separate GMMA particles from soluble proteins of different size
Fig. 4 HPLC-SEC analysis (UV detection at 260 and 280 nm), purified S. Enteritidis GMMA
with fit degree 1 (see Note 12). On the 90 detector (LS11)
chromatogram, select as peak for the average radius calculation
the time range corresponding to 20% height in OMV/GMMA
peak leading and tailing (see Note 13). Check the fitting of the
244 Francesca Micoli et al.
3.4 Nanoparticle 1. Set the instrument as follows: minimum track length ¼ auto-
Tracking Analysis matic settings, minimal expected particle size and blur
(NTA) setting ¼ applied, viscosity settings for water with automatic
correction for the temperature, syringe pump speed 20.
2. Dilute the sample just before the analysis to 10–100 ng/mL of
protein (the concentration needs to be adjusted depending on
the sample) using low-bind material (pipette tips and tubes)
and acquire five replicate videos of 30 s at 25 frames per second,
generating five replicate histograms to be averaged.
3. Analyze particles movement with camera level ¼ 16, slider
shutter ¼ 1300 and slider gain ¼ 512. Test and adjust detec-
tion threshold value for the sample appearance. Figure 6 shows
a typical NTA graph for a GMMA sample.
Concentration
1000
900
800
700
600
500
Inte 400
nsit 300
SEn BPR 60~28.nano
y (a 200
.u.) )
100 n(nm
Size SEn BPR~15-40-42
0
Fig. 6 NTA graph of S. Enteritidis GMMA (as an example), in which particle size, particle concentration, and
relative intensity are plotted together
191 kDa
97 kDa
64 kDa
51 kDa
39 kDa
28 kDa
19 kDa
14 kDa
3.6 Micro BCA Micro BCA analysis is used to quantify OMV/GMMA total protein
content.
1. Set the spectrophotometer wavelength at 562 nm.
2. Set a thermostatic bath to 60 C.
3. To prepare 20 μg/mL BSA solution, open a BSA Standard
ampoule from the Micro BCA kit, mix gently. Transfer 100
μL of BSA 2 mg/mL in a Falcon tube and weigh. Calculate the
amount of water to add using the following formula:
Water ðmgÞ ¼ 100 2 mg=mL BSA weight ðmgÞ 2 mg=mL BSA weight ðmgÞ
OMV/GMMA Analytics 247
Table 3
Dilutions for preparing calibration curve for Micro BCA analysis
Table 4
Dilutions for preparing calibration curve for Lowry analysis
3.9 Amino Acid Amino acid analysis is another method to quantify total protein
Analysis content of OMV/GMMA samples. In contrast to the Micro BCA
and Lowry described above, Amino Acid Analysis quantifies actual
content of each amino acid present in the sample, instead of assum-
ing similar behavior of OMV/GMMA sample as a BSA standard in
the colorimetric assays.
MS analysis can also be used to quantify specific proteins in
OMV/GMMA that are relevant for vaccine efficacy. Such methods
need to be set up and optimized specifically for the protein of
interest. Examples can be found in [10, 18]. Before amino acid
content determination, the time required to hydrolyze the sample
has to be defined with a hydrolysis kinetic study (i.e., hydrolysis for
250 Francesca Micoli et al.
Table 5
Dilutions for preparing curve for amino acidic analysis
Amino acid 250 nmol/mL amino acid standard Water 2500 nmol/mL amino acid standard
nmol/mL solution (μL) (μL) solution (μL)
0 0 500 0
15 30 470 0
25 50 450 0
62.5 125 375 0
125 250 250 0
187.5 375 125 0
250 0 1800 200
Fig. 8 Gradient program for amino acid analysis by HPLC-RP. A -AccQTag Ultra
Eluent A; B -AccQTag Ultra Eluent B/10; C -Water (Filtered 0.22 μm); D -AccQTag
Ultra Eluent B
Lys - 6.908
A
Val - 7.229
Tyr - 6.991
Leu - 7.919
Phe - 8.027
lle - 7.845
Met - 7.115
Cys - 6.836
Drv - 6.680
Pro - 5.778
Ala - 5.141
Thr - 4.735
Glu - 4.339
NH3 - 1.764
Arg - 3.222
Gly - 3.399
Ser - 3.076
His - 2.141
3.798
Brv 6.675
B
Lys - 6.907
NH3 - 1.763
Asp - 3.801
Leu - 7.928
Ala - 5.144
Val - 7.234
Phe - 8.035
Glu - 4.339
Gly - 3.403
Thr - 4.739
Tyr - 6.991
lle - 7.853
Pro - 5.774
Met - 7.119
Ser - 3.077
3.559
Arg - 3.217
His - 2.136
7.615
7.759
7.412
6.155
5.403
5.050
2.531
1.50 2.00 2.50 3.00 3.50 4.00 4.50 5.00 5.50 6.00 6.50 7.00 7.50 8.00
Minutes
Fig. 9 Chromatograms (260 nm profiles) of standard amino acids mixture (a) and of a purified S. Typhimurium
GMMA sample after hydrolysis and derivatization for amino acid analysis (b)
3.10 Total Sugar LPS is one of the main constituents of OMV (naturally released)/
Quantification GMMA membranes (Fig. 1). LPS molecules are constituted of
three main portions: Lipid A is linked to the 3-deoxy-D-manno-
octulosonic acid (KDO) terminus of the core region, which in turn
is attached to the OAg chain (Fig. 10). Capsular polysaccharides
can also be present on OMV/GMMA surface depending on the
OMV/GMMA Analytics 253
Fig. 10 Schematic representation of LPS molecules, constituted by lipid A, core region and repeating units of
the OAg chain
3.11 HPAEC-PAD This method allows quantification of neutral sugars, that are usually
(Neutral Sugars) [14] found as constituents of LPS core regions or OAg chains (such as in
S. Typhimurium, S. Enteritidis, Salmonella Paratyphi A, Shigella
flexneri). Concentrations of Rha, Gal, Glc, Man can be estimated
through this analysis.
1. In 2 mL screw cap vials, prepare in duplicate the dilutions of
calibration curve standard solutions, starting from the 11.25
μg/mL neutral sugar standard mix, as indicated in Table 6.
2. For triplicate analysis, prepare three 2 mL screw cap vials con-
taining 450 μL of the diluted sample.
3. To each vial containing standards or samples, add 150 μL 8 M
TFA (final concentration 2 M), close and vortex each vial for
few seconds.
4. Place all vials in a rigid cardboard rack and incubate in a pre-
heated oven at 100 C for 4 h.
5. After this time, allow all the vials to cool at 2–8 C for 30 min.
6. Remove the caps and dry the standards and samples overnight
in centrifugal evaporator at RT in order to remove the TFA.
254 Francesca Micoli et al.
Table 6
Dilutions for preparing calibration curve for HPAEC-PAD neutral sugars analysis
3.12 HPAEC-PAD This method allows quantification of amino uronic acids, present
(Amino Uronic Acid) for example in Shigella sonnei OAg and capsular polysaccharide
[13] displayed on OMV/GMMA surface.
1. In 2 mL screw cap vials, prepare in duplicate the dilutions of
calibration curve standard solutions, starting from the 2.56
OMV/GMMA Analytics 255
Fig. 11 HPAEC-PAD profile of standard neutral sugar mixture (a) and of a purified S. Typhimurium GMMA
sample (b)
Table 7
Dilutions for preparing calibration curve for HPAEC-PAD amino uronic acid analysis
Fig. 12 HPAEC-PAD profile of S. sonnei standard OAg (a) and of a S. sonnei GMMA sample (b) after acid
hydrolysis resulting in the formation of 2-amino-2-deoxy-α-L-altropyranuronic acid quantified in the analysis
Table 8
Dilutions for preparing calibration curve for Dische determination
3.14 HPLC-MS The lipid A present in the sample is quantified through the
3-hydroxymyristic or 3-hydroxylauric acids present as primary
esters in its structure (in Fig. 13 the lipid A structures of S. sonnei
OMV/GMMA Analytics 259
Fig. 13 Lipid A structures of S. sonnei (a) and N. meningitidis (b) reported as examples with the 3-hydroxy fatty
acids (in red) quantified by HPLC-MS after the hydrolysis of ester bonds
Table 9
Summary of the method time events
ESI Sheath gas pressure ¼ 25, ESI Aux gas pressure ¼ 10,
Capillary temperature ¼ 300 C, Tube lens offset ¼ 70,
Skimmer Offset ¼ 10, q cell collision gas Argon ¼ 1 mTorr,
Collision energy ¼ 16 V, Data type ¼ centroid.
3. Set up the MS scan event (time segment 0–10 min) with the
following parameters: Q1 with 215 (for 3OH-La or 243 for
3OH-My) 0.02 M/z as precursor, Q3 with 59 0.02 M/z as
product using scan time 0.5 s (see Note 36).
4. Program the time events of the HPLC instrumental method as
reported in Table 9.
5. Set up the instrument for the analysis connecting MS Divert
valve, SPE cartridge, RP-column and tubes as shown in Fig. 14
closing valve position 4 with a screw cap fitting. Position the
ESI cone as shown in Fig. 15 (B level).
6. To calculate the dead volume (needed only for first time setup;
see Note 37) assemble the HPLC-QqQ equipment as reported
in Fig. 14 connecting the ESI-MS directly to position 5 without
the C8 column. Program the HPLC pump, divert valve and
QqQ spectrometer as reported in Table 10. Program the MS
spectrometer to measure 3OH-La or 3OH-My according to
the specific sample to be analyzed. Inject 1 μL of a standard
dilution (no subjected to hydrolysis treatment) and collect the
chromatogram. In Fig. 16 examples of chromatograms
acquired by this way are reported.
Consider the time needed for starting the leading of the
first peak (i.e., 2.5 min in the Fig. 16) and calculate the eluent B
volume pumped at that time with its flow rate (in the example:
2.5–1 ¼ 1.5 min; 1.5 min * 0.25 mL/min ¼ 375 μL). This is
the dead volume of eluent B to be pumped after SPE wash step
with eluent A before commute the divert valve. In terms of
OMV/GMMA Analytics 261
Fig. 14 SPE, RP-column and tubes connections in the two different valve
positions
Lasting ð min Þ of dead volume compensation step at flow rate of 0:5 mL= min ¼
ðfirst peak leading time ð min Þ 1Þ 0:25
¼
0:5
262 Francesca Micoli et al.
Table 10
Instrument setting for dead volume calculation
Table 11
Dilutions for preparing calibration curve for HPLC-MS lipid A quantification analysis
the area of the first injection is lower than the other replicates),
one injection for each calibration standard in order of increas-
ing concentration, one injection for each sample replicate, one
injection for each calibration standard in order of increasing
concentration. At the end of sample and standard list runs,
after the last chromatographic analysis is completed, store the
column with 10 column volumes (1.6 mL) of eluent E. After
several analyses, Column Cleaning may be needed (see Note
39). In Fig. 17 are reported chromatograms related to
3OH-La (RT 4.71 min) and 3OH-My (RT 5.57 min).
Fig. 17 Examples of chromatograms related to 3OH-La (RT 4.71 min) (a) and 3OH-My (RT 5.57 min) (b)
quantification by HPLC-MS.
OMV/GMMA Analytics 265
3.15 MALDI-TOF MS 1. Dilute the OMV/GMMA sample with PBS to a final concen-
tration close to 1 mg/mL of protein.
2. In a Wheaton 2 mL screw cap vial, add 200 μL of sample and 50
μL of acetic acid 5% solution.
3. Close the container and keep it at 100 C in a preheated oven
for 2 h (see Note 40).
4. Cool the vial, vortex it, and transfer the whole content in a
2 mL Eppendorf tube.
5. Centrifuge at 14,000 g, 10 C, for 15 min and discard the
supernatant.
6. Add 1.5 mL of water to the pellet and resuspend it.
7. Centrifuge at 14,000 g, 10 C, for 15 min, discard the
supernatant.
8. Add 1.5 mL of water to the pellet and resuspend it.
9. Centrifuge at 14,000 g, 10 C, for 15 min, discard the
supernatant.
10. Dry the pellet overnight in a centrifugal evaporator.
11. Just before deposition on MALDI plate, dissolve the whole
dried pellet in 100 μL of 4:1 chloroform–MeOH by vortexing.
12. For each standard and sample to be assayed, pipet 2 μL into a
0.5 mL tube, add 2 μL of Super-DHB solution, and mix them
carefully pipetting.
13. Withdraw 2 μL of the solution and load it on the target plate.
14. Let the spot dry at RT, possibly with gentle air flux (i.e., plate
left under chemical hood near the front border).
15. To acquire MS Spectra, set up the spectrometer in order to
work in negative, reflectron mode and acquire MS in the range
1000–3000 Da.
16. Calibrate the spectrometer by shooting with laser at the pep-
tide standard spot coordinate.
17. For each sample, record the mass of the sample by shooting
with the laser at the sample spot coordinate (see Note 41).
Shoot with a laser intensity suitable for obtaining resolution of
isotopic cluster peaks (usually less than 50% of its maximum),
in different positions within the sample spot, summing the
result spectrum until clear peaks appear over the baseline
noise (Fig. 18) (see Note 42).
266 Francesca Micoli et al.
A 1585.592 B 1585.499
1000 1250
Penta- Penta-acylated
800 acylated 1000
Intensity a.u.
Intensity a.u.
600 Penta-
Penta- 750
acylated+P
acylated+P
1505.448
400 500
1665.542
200 250
1279.175
0 0
1000 1200 1400 1600 1800 1200 1400 1600 1800
m/z m/z
Fig. 18 MALDI-MS spectra of pentaacylated lipid A of S. Enteritidis GMMA isolated by mild hydrolysis in
acetate buffer pH 4.5 with 3% N-Octyl-β-D-glucopyranoside preserving the lipid A structure (a), compared to
acetic acid hydrolysis (b)
3.16 cELISA [15] The cELISA working principle is based on the competition
between the coating antigen and the specific antigen to quantify
in OMV/GMMA samples for the antigen-specific primary anti-
body. The more antigen is present on OMV/GMMA, the less
primary antibody can bind to the coating antigen, and the less
signal can be detected by ELISA. The antigen of interest on
OMV/GMMA is quantified by comparing the ELISA signal
obtained with a standard curve. The standard curve is built by
spiking the primary antibody with known amounts of
OMV/GMMA displaying the antigen of interest.
MS analysis can also be used for quantifying specific protein
antigens, as well as to determine entire OMV/GMMA protein
composition [10].
1. Prepare 10 sequential dilutions (2- or 3-fold), named as Std01-
Std10, of a freshly prepared OMV/GMMA sample solution,
starting from a concentration previously set, based on the
specific OMV/GMMA sample and antibodies used. To prepare
350 μL of each standard (sufficient for 50 μL each, plated in
duplicate on each of three replicate plates), begin by weighing
the appropriate volume of undiluted standard (Std01) in a tube
and add the appropriate volume of dilution buffer. To prepare
Std02 to Std10, weigh in the corresponding tubes the appro-
priate volume of previous standard and add the appropriate
volume of dilution buffer.
2. Transfer 350 μL of each standard dilution point into successive
wells in a row of a 1 mL deep-well plate. Use the diluted
standards within 1 day.
3. Add 350 μL dilution buffer to each of two blank wells in the
same row as the standards.
4. For positive controls, prepare three test dilutions of the sample
used to prepare the standards, each a total of 0.8 mL in SDB in
2 mL tubes.
OMV/GMMA Analytics 267
3.17 OAg Extraction OMV/GMMA treatment at low pH and high temperature results
in cleavage of the labile linkage between KDO, at the proximal end
of the core oligosaccharide, and lipid A, releasing the OAg-core
chains or core alone in the supernatant (Fig. 10) and causing
OMV/GMMA proteins and lipids to precipitate. The extracted
sugars reflect the composition of LPS molecules on
OMV/GMMA. They can be constituted by OAg chains attached
to the core region (from LPS) and/or core region only (from
lipooligosaccharides). OAg-core (simply indicated as OAg from
here after) and core chains can be easily isolated (e.g., by size
exclusion chromatography) and characterized in depth. Hydrolysis
can be performed with acetate buffer at pH 3.9, or in 1% acetic acid,
based on the OAg stability. The advantage to use 1% acetic acid is
that the supernatant can be directly dried for further analysis with-
out any desalting step (that can be eventually performed by the use
a Cytiva PD10).
1. For polysaccharide extraction with acetate buffer, in a screw cap
vial, add 0.9 mL of OMV/GMMA sample and 100 μL of 10
acetate buffer.
OMV/GMMA Analytics 269
3.18 HPLC-SEC SCA OAg/core samples are analyzed by HPLC-SEC. A refractive index
detector can be used to estimate apparent molecular size using
dextrans (in the range 5–150 kDa) to run a calibration curve and
GPC software. The samples are analyzed after derivatization with
Semicarbazide to quantify the KDO present at the reducing end of
OAg/core chains. This reaction is performed as a slight modifica-
tion of the Semicarbazide assay for α-ketoacid determination [23]
and allows calculation of molar concentration of the chains and
quantification of molar ratio of populations at different size when
present.
1. Using water, prepare in tubes, the dilutions of KDO ammo-
nium salt 40 μg/mL standard solution in duplicate
(as indicated in Table 12). Vortex all standard dilution tubes
for few seconds.
2. Mix the OMV/GMMA sample in order to homogenize the
content.
Table 12
Dilutions for preparing calibration curve for HPLC-SCA analysis
Fig. 19 HPLC-SEC chromatogram of OAg/core extracted from S. Typhimurium GMMA (Fig. 10): detection at
252 nm after derivatization with semicarbazide (a); detection in differential refractive index (dRI) of the
underivatized sample (b)
272 Francesca Micoli et al.
0.15 [rel]
a NAc of GalNAc
OAc on
Rhaˡˡ4Ac when Rha are
NAc of not Ac
OAc on GalNAc
Rhaˡˡ3Ac
0.10
0.05
0.00
5 4 3 2 [ppm]
[rel]
b NAc of GalNAc
when Rha are
not Ac
12
10
8
6
Free
acetate
4
2
0
5 4 3 2 [ppm]
Fig. 20 1H NMR spectra of S. flexneri 6 capsular polysaccharide isolated from GMMA pre (a) and post (b) de-O-
acetylation in NaOD
3.20 Hestrin The O-acetyl ester content can also be measured by the Hestrin
Colorimetric Method colorimetric method. This method is based on the reaction of the
ester groups with hydroxylamine in a basic media to form hydro-
xamic acid which, at low pH, generates a complex with Fe3+ with a
maximum absorbance at 540 nm. The Hestrin method suffers from
interference by common salts like phosphates; for this reason, OAg
extracted samples often need to be desalted for it to work properly.
O-acetylation is expressed as molar ratio (%) between acetic esters
and OAg repeating units (see Note 46).
OMV/GMMA Analytics 273
Table 13
Dilutions for preparing calibration curve for O-acetyl determination by Hestrin colorimetric method
4 Notes
23. Before taking up the standard solution for transfer, rinse the tip
with it by pipetting up and down multiple times.
24. Before taking up the reagent for transfer, rinse the tip with it by
pipetting up and down multiple times.
25. Do not add derivatizing agent to several vials and vortex them
all at the end but proceed to capping and vortexing immedi-
ately for each vial.
26. If a binary UPLC system is used for the analysis, refer to the
Waters Kit instruction material for the appropriate eluent
scheme and gradient program.
27. Being the AMQ the product of derivatizing reagent reaction
with water, if a degraded (i.e. hydrolyzed for humidity) reagent
is used this test doesn’t work.
28. After the last chromatographic analysis is completed, store the
column in 18 mM NaOH solution.
29. In this case, a standard OAg sample is used for building the
calibration curve, as the monomer (2-amino-2-deoxy-α-L-
altropyranuronic acid) resulting from sample hydrolysis and
quantified by the chromatographic analysis is not commercially
available.
30. After the last chromatographic analysis is completed, store the
column in 100 mM NaOH solution.
31. It is advisable, due to the short incubation time, to proceed in
parallel with less than 15 standard/sample tubes. If the number
of samples is higher, proceed with a cluster of 10 tubes for each
5 min incubation.
32. Both ABS have to be lower than 1.2 in order to be considered
acceptable.
33. Do not leave concentrated sulfuric acid solution in polystyrene
cuvettes but appropriately discard the solution just after usage
(cuvettes are slowly corroded).
34. LAL is not deemed appropriate, as samples contain such high
endotoxin units that they typically require a dilution factor of
million-billion.
35. M/z accuracy check needs to be done at first implementation of
the procedure on a new instrument or after a calibration of the
MS instrument. This is required, as accuracy issues in M/z,
especially for 59 Da fragment, were experienced. The check is
performed by direct infusion of 3-OH-lauric acid/3-OH-myr-
istic acid standard diluted in 70% ACN 0.05% FA to about
0.5–1 nmol/mL. Perform direct infusion of the standards
and acquire/visualize in real time with the following para-
meters: First set Q1 in full scan mode and note the 215 M/z
closest peak and/or the 243 M/z closest peak: each of these
OMV/GMMA Analytics 277
Table 14
Column cleaning procedure
Eluent C Eluent D
Min % %
0 75 25
2 75 25
32 0 100
35 75 25
278 Francesca Micoli et al.
41. The highest intensity spectra are usually recovered far from big
crystal formations.
42. Using the MALDI instrument in reflectron mode, it is likely to
find as highest peak exact m/z + 1 or exact m/z + 2 for the
isotopic abundance of the elements present in the structure.
During hydrolysis or matrix desorption, lipid A can lose one
phosphate (80 m/z) or other substituents so more than one
peak can be observed for one product. Note that +22 m/z/
+38 m/z peaks, respectively, represent +Na and + K adducts.
43. Y ¼ ((b/(a Log10(X)))ˆ(d) 1)/c, where a, b, c, and
d represent values of the curve parameters (i.e., respectively:
a ¼ minimum asymptote, b ¼ Hill’s slope, c ¼ inflection point,
and d ¼ maximum asymptote), Y the amount determined, and
X the absorbance reading.
44. Perform system equilibration with elution buffer before start-
ing the analysis. This equilibration step lasts 70 min if the
column system has to be equilibrated with a different eluent;
60 min or more are needed also to warm up the PDA lamps
before the analysis.
45. The first 1H NMR spectrum is recorded to ensure the absence
of impurities or other signals at the same chemical shift of the
acetate anion released after de-O-acetylation of the sample that
would interfere with the quantification of the O-acetyl content.
46. Considering the multiple steps to prepare the sample for the
O-acetylation measurement (OAg extraction, desalting), prior
to calculating the O-acetyl ester/repeating unit ratio, it is
recommended to measure the OAg quantity on the same trea-
ted sample prepared for ester measurements and use this value
for the further calculation.
Acknowledgments
References
1. Micoli F, MacLennan CA (2020) Outer mem- 74(1):81–94. https://doi.org/10.1128/
brane vesicle vaccines. Semin Immunol mmbr.00031-09
50:101433. https://doi.org/10.1016/j. 3. Gerke C, Colucci AM, Giannelli C et al (2015)
smim.2020.101433 Production of a Shigella sonnei vaccine based
2. Ellis TN, Kuehn MJ (2010) Virulence and on generalized modules for membrane anti-
immunomodulatory roles of bacterial outer gens (GMMA), 1790GAHB. PLoS One 10
membrane vesicles. Microbiol Mol Biol Rev
OMV/GMMA Analytics 279
Abstract
The production of conjugate vaccines within an E. coli (Escherichia coli) host provides an inexhaustible
supply without the need for culture of pathogenic organisms. The machinery for expression of glycan and
acceptor protein, as well as the coupling enzyme, are all housed within the E. coli chassis, meaning that there
are no additional steps required for individual purification and chemical conjugation of components. In
addition, there are far fewer purification steps necessary to obtain a purified glycoconjugate for use in
vaccine testing. Here we describe production and purification of a HIS-tagged Campylobacter jejuni AcrA
protein conjugated to Streptococcus pneumoniae serotype 4 capsule.
1 Introduction
281
282 Emily J. Kay and Vanessa S. Terra
Fig. 1 Glycoengineering approach to the production and purification of glycoconjugate vaccines. An E. coli cell
is transformed with the components needed to generate the glycoconjugate protein, shown here encoded on
plasmids for (1) glycan expression; (2) target protein with leader sequence and glycosylation sequons; (3) the
enzyme responsible for coupling the glycan to the protein. Any of the components could be inserted into the
chromosome, as in the protocol described in this chapter where PglB (the coupling enzyme is inserted onto the
chromosome. The resulting glycoconjugate must then undergo several rounds of purification to remove
contaminants and unconjugated glycan. (This figure was created with BioRender.com)
2 Materials
2.1 Cell Growth 1. E. coli W311B containing plasmids pWA2 and pB4 (see
Note 1).
2. Growth medium SSOB: 2% tryptone, 0.5% yeast extract,
171 mM NaCl, 2.5 mM KCl, 10 mM MgCl2, 10 mM
MgSO4. Weigh 40 g tryptone, 10 g yeast extract, 20 g NaCl,
372 mg KCl, 1.904 g MgCl2 and 4.928 g MgSO4·7H2O and
284 Emily J. Kay and Vanessa S. Terra
Fig. 2 (a) AKTA trace of AcrA-SP4 glycoconjugate eluted from the His-Trap
column with imidazole. The green line denotes increasing concentration of
imidazole, the x axis denotes the volume of liquid passed through the column,
and the Y axis denotes the UV absorbance at 280 nm, as an indication of eluted
protein concentration. Fractions are labeled by their position in the AKTA
collection tray, which accommodates 15 tubes per row. The red lines indicate
the 1 mL fractions collected between A1 and B14. (b) Selected eluate fractions
separated on SDS-PAGE gel followed by Western blot with anti-His tag fluores-
cent antibody (red) and anti-SP4 capsule fluorescent antibody (green), as
visualized by a LiCor Odyssey digital imager. M molecular weight marker
PageRuler Plus
Fig. 3 (a) AKTA trace of AcrA-SP4 glycoconjugate eluted from the Resource S column with NaCl. The green line
denotes increasing concentration of NaCl, the x axis denotes the volume of liquid passed through the column,
and the Y axis denotes the UV absorbance at 280 nm, as an indication of eluted protein concentration.
Fractions are labeled by their position in the AKTA collection tray, which accommodates 15 tubes per row. The
red lines indicate the 1 mL fractions collected between A1 and B6. Selected eluate fractions were separated
on SDS-PAGE gel followed by either (b) Western blot with fluorescent anti-His tag antibody (red) and
fluorescent anti-SP4 capsule antibody (green), or (c) Coomassie staining, both visualized by a LiCor Odyssey
digital imager. S1–S4 denote high salt fractions collected after the salt concentration was increased to 1 M
NaCl. M molecular weight marker PageRuler Plus
2.9 Coomassie 1. Staining tray big enough to hold SDS-PAGE gel and around
Staining 30 mL liquid.
Production of Vaccines Using Biological Conjugation 289
3 Methods
3.2 Cell Lysis 1. Thaw pellet by submerging storage tube in cold water for
15 min.
2. Resuspend cell pellet (3–4 g wet weight) in 100 mL lysis buffer
+ 1 mg/mL lysozyme (1.2 mL 100 mg/mL) + 10 μL benzo-
nase (2.5 KU) (see Note 10).
3. Lyse with a Stansted High Pressure cell homogenizer or similar
high-pressure cell homogenizer. Prerinse the homogenizer
with 70% ethanol, distilled water and lysis buffer before use.
Sample should be passed through the high-pressure cell
homogenizer as many times as required for the resuspended
pellet suspension to significantly clear.
4. After lysis, centrifuge sample at 3000 g for 15 min at 4 C to
pellet large debris.
5. Supernatant should be decanted into fresh centrifuge tubes and
insoluble cell debris removed by centrifugation at 7800 g for
1 h at 4 C.
290 Emily J. Kay and Vanessa S. Terra
3.5 Western Blot 1. Cut nitrocellulose membrane to the size of the gel and soak in
MilliQ water for 10 min, then transfer to Towbin buffer (see
Note 17).
2. Cut six pieces of filter paper to the size of the gel and soak them
in Towbin buffer.
3. Assemble the transfer stack in the semi-dry transfer unit as
follows, using tweezers to handle filter paper and membrane:
on the anode, place three Towbin-soaked filter papers followed
by the nitrocellulose membrane. Submerge the rinsed gel in
Towbin buffer, then carefully place it on the membrane being
careful not to move it once it has touched the membrane. At
this point air bubbles should be removed by gently rolling a
clean object over the top. The remaining three Towbin-soaked
filter papers should be placed on top of the stack and air
bubbles removed. The cathode is then placed on top and the
electrodes connected to the power pack.
4. The transfer is run at 1 mA/cm2 for 1 h before removing the
membrane to a suitable clean washing tray.
5. Wash the membrane once with PBS to remove residual Towbin
buffer and then incubate with PBS-M for 1 h at room
temperature.
6. Pour off the PBS-M and wash the membrane three times with
around 30 mL PBS-T. Gentle agitation should be applied
during incubation of the wash steps by using a rocker or orbital
shaker at room temperature.
7. Incubate the membrane with a mix of the two primary anti-
bodies diluted in 10 mL PBS-M (1:10000 mouse anti-His
antibody and 1:1000 rabbit anti-SP4 antibody) for 1 h at
room temperature for simultaneous detection of protein
(anti-His) and glycan (anti-SP4) (see Note 18).
292 Emily J. Kay and Vanessa S. Terra
3.6 Coomassie Stain 1. Place the rinsed gel in a staining tray with around 30 mL of
Coomassie stain. Place on a rocker or orbital shaker for for
2–4 h, until the gel is a uniform blue colour.
2. Pour off the Coomassie stain and de-stain using de-staining
solution for 4–24 h, changing the de-stain solution periodically
until background is clear and protein bands are prominent.
3. Image the gel using any image detection equipment or camera
against a white background.
3.7 Desalting 1. Prepare an endotoxin removal column the day before use.
and Endotoxin Twist off bottom closure, loosen cap and place in 50 mL
Removal centrifuge tube. Spin at 500 g for 1 min. Discard solution.
Plug the bottom with the plug provided with the column and
add 8 mL 0.2 N NaOH, replace cap and invert several times.
Incubate overnight at room temperature.
2. Pool selected HisTrap eluate fractions from Subheading 3.3
that contain target protein, as determined by Western blot and
Coomassie-stained gel in Subheadings 3.4–3.6 (Fig. 2) (see
Note 20).
3. Prewash a 10 kDa MWCO centrifuge filter column (e.g.,
Vivaspin) twice by adding the maximum fill volume of MilliQ
water at centrifuging at 4000 g for 1 min, then load pooled
sample onto the column and concentrate until sample volume
is no more than 2.5 mL (see Note 21).
4. Use PD-10 desalting column gravity protocol. Remove storage
buffer by pouring it off the column. Drip through 25 mL
PD-10 equilibration buffer, then apply a maximum of 2.5 mL
sample (see Note 22).
5. Discard flow through and then elute sample with 3.5 mL
equilibration buffer, collecting the eluate in 1 mL fractions.
6. Clean the PD-10 column with 25 mL equilibration buffer,
then store in 20% ethanol (see Note 23).
Production of Vaccines Using Biological Conjugation 293
7. Pool the first 2.375 mL of eluate from PD-10 column and add
125 μL 2 M NaCl to increase the salt concentration to 0.1 M.
8. To prepare an endotoxin removal column, remove bottom
plug and cap and centrifuge column at 500 g 1 min, then
wash with 8 mL each of 2 M NaCl, followed by water, and
3 rounds of endotoxin removal buffer, with centrifugation at
500 g 1 min for each wash.
9. Plug the endotoxin removal column and add 2.5 mL sample
(salt adjusted PD-10 eluate from step 6), replace cap and invert
several times. Incubate the column at 4 C with end-over-end
mixing, on a tube rotator fitted with a “Ferris Wheel” disk,
overnight.
10. Remove bottom plug and cap and centrifuge the column at
500 g 1 min to elute sample off the endotoxin removal
column into a fresh collection tube (see Note 24).
11. Remove salt before anion exchange by using PD-10 desalting
columns as in steps 4–6.
3.8 Cation Exchange 1. Prepare the Resource S column (see Note 25) by running
Chromatography through 5 CV each of MilliQ water, start buffer, elution buffer,
and start buffer before loading sample.
2. The desalted sample in PD-10 equilibration buffer must be
diluted in start buffer before loading onto the column (e.g.,
3 mL sample diluted up to 35 mL with start buffer).
3. After loading, the column is attached to the AKTA and washed
with 10–20 mL start buffer until the UV baseline is steady (see
Note 26).
4. The sample is eluted from the column using a salt gradient up
to 0.5 M NaCl (feed line A ¼ start buffer, feed line B ¼ elution
buffer. Change from 100% A to 50% B over 20 mL, with 1 mL
fractions collected).
5. The eluted fractions can be analyzed at this stage by SDS-
PAGE with Coomassie staining and by Western blot as in
Subheadings 3.4–3.6 (Fig. 3).
6. Clean the Resource S column on the AKTA using a high salt
wash of 10 mL 1 M NaCl (this can be achieved by using only
feed line B, that is, increase from 50% B to 100% B for 10 min)
(see Note 27).
7. The AKTA is cleaned with 30 mL 0.5 M NaOH, distilled water,
and 20% EtOH.
3.9 Buffer Exchange 1. Pool the desired fractions from Resource S column (see Note
and Storage 28), concentrate to around 2.5 mL and buffer exchange using
Vivaspin 10 kDa MWCO centrifuge filter column, as described
in Subheading 3.7, step 3 (again, prewash filter twice with
MilliQ water before loading sample).
294 Emily J. Kay and Vanessa S. Terra
4 Notes
15. As the protein AcrA has two glycosylation sites there should be
two bands visible above the unglycosylated protein, denoting
short repeat units attached at one or both glycosylation sites. In
addition, glycan bands should be visible above the protein,
showing polymerized glycan attached to the protein. Control
samples of protein only and glycan only can be included in
Western blots for comparison. Any glycan visible at the bottom
of the gel will be glycan that is not attached to the target
protein.
16. It is recommended to store columns stripped and only recharge
with Ni2+ before use. A full cleaning of columns is done using
5–10 CV water, 5–10 CV stripping buffer, 60 CV 1 M NaOH
(contact time at least 1 h), 5–10 CV wash II buffer and 5–10
CV 20% ethanol (protocol can be found in the manufacturer’s
manual). Stripping Buffer: 20 mM NaH2PO4, 0.5 M NaCl,
and 50 mM EDTA at pH 7.4. To 200 mL of distilled water add
0.69 g of NaH2PO4, 7.305 g of NaCl, and 3.653 g of EDTA.
Adjust to pH 7.4 with NaOH and make up to volume to
250 mL with distilled water. The EDTA will not dissolve
until the pH is adjusted.
17. We recommend use of Hybond™-C Extra nitrocellulose mem-
brane (Amersham Biosciences, UK) for better results.
18. A 10 mL volume of primary antibody solution in PBS-M is
generally enough to completely cover a membrane in a tray
11 14 cm. If antibody is scarce then a heat-sealable bag,
the size of the membrane, may be used to reduce the volume of
the primary antibody solution required to completely cover the
membrane. The aim is to cover the membrane with solution at
all times without air bubbles crossing the surface. In the exper-
iment described in this chapter, both primary antibodies are
added for simultaneous detection, which requires a detection
system that can detect two different secondary signals at once
(e.g., fluorescence). If this is not available, the primary anti-
bodies should be added separately.
19. There are different detection systems that can be used for
Western blotting. For example, horseradish peroxidase,
HRP-conjugated secondary antibodies may be used, followed
by an enhanced chemiluminescence (ECL) detection kit. The
membrane can then be exposed to X-ray film in the dark and
then developed. The disadvantage is that only one primary
antibody can be used at a time, so either two gels for each
sample could be run, or the membrane stripped and reprobed.
20. Different fractions may be pooled according to downstream
application or desired ratio of glycosylated to unglycosylated
protein. As the sample will be further purified by ion exchange
chromatography, it is preferable to pool as many fractions as
Production of Vaccines Using Biological Conjugation 297
26. The maximum pressure for the Resource S column is 1.5 MPa,
therefore flow rates should be adjusted so as not to exceed this
pressure.
27. The Resource S column is cleaned with 5–10 CV each of 2 M
NaCl, 1 M NaOH, 2 M NaCl, distilled water, start buffer,
distilled water, and 20% ethanol–0.2 M sodium acetate. If the
column is blocked, as indicated by no flow through the column
at the maximum pressure tolerance for the column, then inject
with 1 mg/mL pepsin in a solution of 0.5 M NaCl and 0.1 M
acetic acid, and leave overnight before flushing through with
5–10 CV of distilled water and cleaning as above. Once the
column is in 20% ethanol–0.2 M sodium acetate it may be
stored at 4 C until next use.
28. As with the initial His-purified sample, different fractions may
be pooled according to downstream application or desired
ratio of glycosylated to unglycosylated protein. In this example,
fractions A9–A15 (Fig. 3) were pooled. This was to capture as
much protein as possible that is glycosylated while minimizing
the proportion of contaminating protein (i.e., protein bands
that appear on the Coomassie stained gel that are absent from
the Western blot).
29. Vivaspin columns can be used for buffer exchange by diluting
the concentrated sample up to the original volume with PBS
and concentrating again. Repeating the process three times. If
preferred, alternative buffer exchange methods may be used
(e.g., dialysis cassettes).
30. Sugar content can be quantified using, for example, hot phe-
nol–sulfuric acid method [28] or high-performance anion-
exchange chromatography with pulsed amperometric detec-
tion (HPAEC–PAD) on a Dionex system [29].
31. Lyophilizing glycoconjugates suspended in PBS will result in
salts being retained in the sample, therefore water should be
used for reconstitution. Buffer exchange into 50 mM ammo-
nium bicarbonate buffer may be preferable, because it sublimes
into ammonia, water, and carbon dioxide upon lyophilization,
often without causing measurable harm to the protein. A
50 mM ammonium bicarbonate buffer, pH 7.8, is made by
dissolving 4 g NH4HCO3 in 800 mL distilled water, then
making the final volume up to 1 L. If ammonium bicarbonate
buffer is used then the sample should be reconstituted in PBS.
If a lyophilizer is not available, then the purified glycoconjugate
may be stored at 80 C, but freeze–thaw cycles should be
avoided.
Production of Vaccines Using Biological Conjugation 299
References
1. Parkhill J, Wren BW, Mungall K et al (2000) 10. Pan C, Sun P, Liu B et al (2016) Biosynthesis of
The genome sequence of the food-borne path- conjugate vaccines using an O-linked glycosyl-
ogen Campylobacter jejuni reveals hypervari- ation system. mBio 7(2):e00443–e00416.
able sequences. Nature 403(6770):665–668. https://doi.org/10.1128/mBio.00443-16
https://doi.org/10.1038/35001088 11. Harding CM, Nasr MA, Scott NE et al (2019)
2. Wacker M, Linton D, Hitchen PG et al (2002) A platform for glycoengineering a polyvalent
N-linked glycosylation in Campylobacter jejuni pneumococcal bioconjugate vaccine using
and its functional transfer into E. coli. Science E. coli as a host. Nat Commun 10(1):891.
298(5599):1790–1793. https://doi.org/10. https://doi.org/10.1038/s41467-019-
1126/science.298.5599.1790 08869-9
3. Fisher AC, Haitjema CH, Guarino C et al 12. Naegeli A, Neupert C, Fan Y-Y et al (2014)
(2011) Production of secretory and extracellu- Molecular analysis of an alternative
lar N-linked glycoproteins in Escherichia coli. N-glycosylation machinery by functional trans-
Appl Environ Microbiol 77(3):871–881. fer from Actinobacillus pleuropneumoniae to
https://doi.org/10.1128/aem.01901-10 Escherichia coli. J Biol Chem 289
4. Yates LE, Natarajan A, Li M et al (2019) (4):2170–2179. https://doi.org/10.1074/
Glyco-recoded Escherichia coli: jbc.M113.524462
Recombineering-based genome editing of 13. Kowarik M, Young NM, Numao S et al (2006)
native polysaccharide biosynthesis gene clus- Definition of the bacterial N-glycosylation site
ters. Metab Eng 53:59–68. https://doi.org/ consensus sequence. EMBO J 25
10.1016/j.ymben.2019.02.002 (9):1957–1966. https://doi.org/10.1038/sj.
5. Strutton B, Jaffé SRP, Pandhal J, Wright PC emboj.7601087
(2018) Producing a glycosylating Escherichia 14. Georgiou G, Segatori L (2005) Preparative
coli cell factory: the placement of the bacterial expression of secreted proteins in bacteria: sta-
oligosaccharyl transferase pglB onto the tus report and future prospects. Curr Opin
genome. Biochem Biophys Res Commun 495 Biotechnol 16(5):538–545. https://doi.org/
(1):686–692. https://doi.org/10.1016/j. 10.1016/j.copbio.2005.07.008
bbrc.2017.11.023 15. Cuccui J, Wren B (2015) Hijacking bacterial
6. K€ampf MM, Braun M, Sirena D et al (2015) In glycosylation for the production of glycocon-
vivo production of a novel glycoconjugate vac- jugates, from vaccines to humanised glycopro-
cine against Shigella flexneri 2a in recombinant teins. J Pharm Pharmacol 67(3):338–350.
Escherichia coli: identification of stimulating https://doi.org/10.1111/jphp.12321
factors for in vivo glycosylation. Microb Cell 16. Herbert JA, Kay EJ, Faustini SE et al (2018)
Factories 14(1):12. https://doi.org/10. Production and efficacy of a low-cost recombi-
1186/s12934-015-0195-7 nant pneumococcal protein polysaccharide
7. Caillava AJ, Ortiz GE, Melli LJ et al (2019) conjugate vaccine. Vaccine 36
Improving bioreactor production of a recom- (26):3809–3819. https://doi.org/10.1016/j.
binant glycoprotein in Escherichia coli: effect of vaccine.2018.05.036
specific growth rate on protein glycosylation 17. Feldman MF, Wacker M, Hernandez M et al
and specific productivity. Biotechnol Bioeng (2005) Engineering N-linked protein glycosyl-
116(6):1427–1438. https://doi.org/10. ation with diverse O antigen lipopolysaccharide
1002/bit.26953 structures in Escherichia coli. Proc Natl Acad
8. Ihssen J, Haas J, Kowarik M et al (2015) Sci U S A 102(8):3016–3021. https://doi.
Increased efficiency of Campylobacter jejuni org/10.1073/pnas.0500044102
N-oligosaccharyltransferase PglB by structure- 18. Sutcliffe JG (1978) Nucleotide sequence of the
guided engineering. Open Biol 5 ampicillin resistance gene of Escherichia coli
(4):140227–140227. https://doi.org/10. plasmid pBR322. Proc Natl Acad Sci U S A
1098/rsob.140227 75(8):3737–3741. https://doi.org/10.1073/
9. Faridmoayer A, Fentabil MA, Haurat MF et al pnas.75.8.3737
(2008) Extreme substrate promiscuity of the 19. Kay EJ, Yates LE, Terra VS et al (2016)
Neisseria oligosaccharyl transferase involved in Recombinant expression of Streptococcus pneu-
protein O-glycosylation. J Biol Chem 283 moniae capsular polysaccharides in Escherichia
(50):34596–34604. https://doi.org/10. coli. Open Biol 6(4):150243. https://doi.org/
1074/jbc.M807113200 10.1098/rsob.150243
300 Emily J. Kay and Vanessa S. Terra
20. Kovach ME, Elzer PH, Hill DS et al (1995) an inhibitory peptide. Sci Rep 8(1):16297.
Four new derivatives of the broad-host-range https://doi.org/10.1038/s41598-018-
cloning vector pBBR1MCS, carrying different 34534-0
antibiotic-resistance cassettes. Gene 166 26. Garcia-Quintanilla F, Iwashkiw JA, Price NL
(1):175–176. https://doi.org/10.1016/ et al (2014) Production of a recombinant vac-
0378-1119(95)00584-1 cine candidate against Burkholderia pseudomal-
21. Bachmann BJ (1996) Derivations and geno- lei exploiting the bacterial N-glycosylation
types of some mutant derivatives of Escherichia machinery. Front Microbiol 5:381. https://
coli K-12. In: Neidhardt FC (ed) Escherichia doi.org/10.3389/fmicb.2014.00381
coli and Salmonella: cellular and molecular 27. Zimmerman T, Petit Frère C, Satzger M et al
biology, 2nd Ed edn. ASM Press, (2006) Simultaneous metal chelate affinity
Washington, DC, pp 2460–2488 purification and endotoxin clearance of recom-
22. Thomas CM (2014) Plasmid incompatibility. binant antibody fragments. J Immunol Meth-
In: Bell E (ed) Molecular life sciences: an ency- ods 314(1):67–73. https://doi.org/10.1016/
clopedic reference. Springer New York, j.jim.2006.05.012
New York, NY, pp 1–3. https://doi.org/10. 28. Masuko T, Minami A, Iwasaki N et al (2005)
1007/978-1-4614-6436-5_565-2 Carbohydrate analysis by a phenol–sulfuric acid
23. Carattoli A (2009) Resistance plasmid families method in microplate format. Anal Biochem
in Enterobacteriaceae. Antimicrob Agents Che- 339(1):69–72. https://doi.org/10.1016/j.
mother 53(6):2227–2238. https://doi.org/ ab.2004.12.001
10.1128/AAC.01707-08 29. Talaga P, Vialle S, Moreau M (2002) Develop-
24. Lizak C, Gerber S, Numao S et al (2011) X-ray ment of a high-performance anion-exchange
structure of a bacterial oligosaccharyltransfer- chromatography with pulsed-amperometric
ase. Nature 474(7351):350–355. https://doi. detection based quantification assay for pneu-
org/10.1038/nature10151 mococcal polysaccharides and conjugates. Vac-
25. Napiórkowska M, Boilevin J, Darbre T et al cine 20(19):2474–2484. https://doi.org/10.
(2018) Structure of bacterial oligosaccharyl- 1016/S0264-410X(02)00183-4
transferase PglB bound to a reactive LLO and
Chapter 16
Abstract
Lipopeptides or lipoproteins show potential as safe and effective subunit vaccines for protection against
bacterial pathogens. Provided suitable adjuvants are selected, such as the TLR2-stimulating molecules
Pam2Cys and Pam3Cys, these may be formulated as inhalational vaccines to optimize localized pulmonary
immune responses. Here, we present methods to assess antigen-specific memory lymphocyte responses to
novel vaccines, with a focus on immune responses in the lung tissue and bronchoalveolar space. We describe
detection of T-cell responses via leukocyte restimulation, followed by intracellular cytokine staining and
flow cytometry, enzyme-linked immunosorbent spot assay (ELISpot), and sustained leukocyte restimula-
tion for detection of antigen-specific memory responses. We also detail assessment of antibody responses to
vaccine antigens, via enzyme-linked immunosorbent assay (ELISA)-based detection. These methods are
suitable for testing a wide range of pulmonary vaccines.
Key words Tuberculosis, Inhalational vaccination, Lungs, Bronchoalveolar lavage, Lipoprotein, ELI-
Spot, ELISA, Intracellular cytokine staining, Flow cytometry, Leukocyte restimulation
1 Introduction
301
302 Anneliese S. Ashhurst et al.
2 Materials
Prepare all solutions using ultrapure water and store at room tem-
perature (~22 C), unless indicated otherwise. Standard cell culture
consumables and plasticware, including 70 μm filters, should be
sterile. Follow all institutional biosafety and chemical hazard reg-
ulations, including when disposing of waste.
2.1 Leukocyte All reagents should be sterile. Saline buffer may be autoclaved,
Preparation otherwise filter buffers or stock solutions through a nonpyrogenic
0.22 μm membrane filter.
1. 1 phosphate buffered saline (PBS), pH 7.4.
2. Fluorescence activated cell sorting (FACS) buffer (keep at
4 C): PBS, 2% v/v fetal bovine serum (FBS) or fetal calf
serum (FCS), 2 mM ethylenediaminetetraacetic acid (EDTA).
3. Complete cell culture media (keep at 4 C): Roswell Park
Memorial Institute (RPMI) 1640 Medium (containing L-glu-
tamine), 10% FBS or FCS, 0.05 mM 2-Mercaptoethanol,
100 U/ml penicillin–streptomycin.
4. PBS/Heparin (keep at 4 C): PBS, 20 U/ml Heparin.
Immunological Assessment of Inhalational Lipoprotein Vaccines 305
Table 1
Comparison of key methods for assessing T-cell cytokine responses to peptide or protein vaccine
antigens
Major reportable
Method outcome(s) Key advantages Disadvantages
Leukocyte Proportion of antigen- Flexibility—Simultaneous Cannot provide
restimulation, ICS responsive cytokine single-cell level quantitation of the
and flow positive T-cells, detection of multiple cytokines released
cytometry including single-cell different phenotypes Requires access to a
level analysis of High sensitivity—Large multiparameter flow
multifunctionality and numbers of cells can be cytometer and analysis
phenotype analyzed to allow software
detection of rare Depending on
antigen-responsive experiment design,
populations may be costly
ELISpot Enumeration of antigen- High sensitivity Limited to detection of
specific cells capable of Cost-effective one cytokine only
releasing the detected High-throughput (unless variant
cytokine methods such as dual-
color [40] or
FluoroSpot [41] are
utilized)
Cannot determine the
phenotype of cells
releasing the detected
cytokine
Sustained leukocyte Quantitative detection of High-sensitivity, Takes longer
restimulation— cytokine released particularly useful for Cannot determine the
Recall of memory Can measure multiple detection of memory phenotype of cells
antigen-specific cytokines if multiple T-cell responses (not releasing the detected
T-cell responses ELISAs are performed recently activated) cytokine
on cell supernatants, or Cost-effective Assessment of cellular
if multiplex methods High-throughput proliferation by
such as cytokine bead Cell supernatants can be radiological or
array are used stored frozen and fluorescent methods
Can also examine antigen- cytokines quantitated may be costly
specific cell proliferation when convenient
in the same assay, either Offers several ways to
simply by enumerating determine cellular
cells, or by radioisotope proliferation, which
or fluorescent methods can be selected based
on the resources
available
3 Methods
Carry out all procedures at room temperature (RT; ~22 C), unless
indicated otherwise.
3.1 Isolation of BAL Leukocytes should be collected and processed using aseptic tech-
and Lung Leukocytes niques, with sterile reagents and equipment. Keep tissue or cells at
from Vaccinated Mice 4 C or on wet ice as much as is practical, to slow down metabolic
processes that alter cell phenotype and decrease the rate of cell
death. Keep FACS buffer and complete cell culture media at 4 C.
3.1.1 Bronchoalveolar this is likely to cause injury to the trachea, chest cavity, and
Lavage Collection and Cell blood vessels, resulting in difficulty performing the lavage.
Isolation 2. Immediately after confirmation of euthanasia, spray 70% etha-
nol on the neck to sterilize and make an incision in the skin.
Incise away the muscles to expose the trachea. Use two pairs of
curved forceps to gently tease away the membrane covering the
trachea.
3. Slide one pair of curved forceps under the trachea, to provide
stability during intubation.
4. Very carefully, with the bevel facing upward, punch a hole in
one side of the trachea with a 21 GA needle.
5. Insert a ~1.8 in. 20 GA flexible cannula, attached to a 1 ml
syringe containing 1 ml sterile PBS, into the hole created by the
needle to 0.5–1.5 cm depth. Do not exceed this, as it may
damage the lungs. With a second pair of fine forceps, pinch
the trachea to seal the cannula firmly in place. Remove the pair
of forceps under the trachea.
6. Perform lavage: slowly inject the 1 ml PBS into the lungs—you
should see the lungs inflate and the chest expand as the lungs
fill with PBS. There should be no leakage of PBS from the
trachea or lungs. If this occurs, it is likely the seal with the
forceps on the trachea is not sufficiently firm, or, there has been
a rupture in the trachea.
7. Pull back on the syringe to collect as much fluid from the lungs
as possible, usually 700–900 μl. Dispense into an Eppendorf or
Falcon tube. In a healthy naı̈ve mouse, there should be no
obvious evidence of blood in the bronchoalveolar lavage fluid
(BALF); however, this may occur in mice with inflammation or
infection in the lungs.
8. Centrifuge the BAL for 5 min (300–500 g, 4 C). Collect the
supernatant—this is the BALF and can be used in assays to
quantitate the presence of cytokines/chemokines or antibodies
in the airways by ELISA. If not used immediately, this should
be stored at 80 C to 30 C. It may be beneficial to add
protease inhibitors to prevent degradation of protein analytes.
9. Resuspend the cells in ~200 μl FACS buffer or complete cell
culture media by gently pipetting up and down. Count viable
cells by trypan blue exclusion. The number of cells will be
variable, but in a healthy mouse expect ~1 105 or less, and
up to 1 106 following pulmonary immunization or if inflam-
mation or infection is present.
3.1.2 Lung Leukocytes 1. Euthanize the mouse as per the requirements of your institu-
tional animal ethics committee, avoiding cervical dislocation.
308 Anneliese S. Ashhurst et al.
3.2.1 Restimulation 1. Add 2–4 106 leukocytes (see Note 1) from each sample into a
of Antigen-Specific T-cells well of a 96-well round- or V-bottom plate. Use one well for
in Ex vivo Culture each antigen you wish to restimulate the sample with.
2. Centrifuge the plate to pellet cells (3–5 min, 300–500 g) and
discard the supernatant (see Note 2). Resuspend the cells, by
gently pipetting up and down (see Note 3), in 200 μl per well
complete culture media containing the peptide or protein vac-
cine antigen of interest (5–10 μg/ml is generally suitable). Also
prepare negative and positive control samples (see Note 4).
3. Incubate the plate at 37 C, 5% CO2 to allow the antigen to be
processed for presentation on MHCI/II to T-cells. This incu-
bation time may require optimization. As a guide: for peptide
restimulation 1–2 h; for protein restimulation, 3–4 h.
4. Add Brefeldin A to each well (10 μg/ml; this will block trans-
port of proteins from the Golgi) and pipette gently to resus-
pend the cells. Incubate (humidified) at 37 C, 5% CO2 for
4–6 h to allow intracellular accumulation of the cytokines (see
Note 5).
3.2.2 Immunostaining The number and type of cell surface or intracellular markers
Cells for Detection selected for immunostaining will vary greatly depending on the
of Intracellular Cytokine experimental design. In addition, the monoclonal antibody panels
will vary between laboratories, accounting for the configuration of
the cytometer available for data acquisition, as well as more practical
considerations such as antibody stocks already within the laboratory
and the budget available. We recommend the following papers to
assist in the critical steps of selecting antibody combinations for use
in flow cytometry, optimization of antibody dilutions and tips for
data acquisition on the cytometer [34, 35]. The antibody clones
and fluorophores utilized to generate the sample data in Fig. 1 are
included in Note 6, but this is by no means restrictive.
From this step onward, keep the cells on wet ice or at 4 C to
prevent any further metabolic changes or increasing cell death. As
vaccine experiments often involve testing many samples, to improve
efficiency and consistency, we strongly recommend performing cell
staining in a 96-well plate, rather than staining cells in individual
FACS tubes.
Immunological Assessment of Inhalational Lipoprotein Vaccines 311
Fig. 1 Gating strategy for analysis of intracellular stained flow cytometry data, to quantitate the proportion of
antigen-specific cytokine producing T-cells and determine polyfunctional T-lymphocyte responses to immu-
nization. Debris and dead cells are excluded, CD4+ or CD8+ T-cells selected, cytokine positive cells gated,
followed by Boolean gating to enumerate the frequency of polyfunctional populations. A representative
immunostained lung sample from a Pam2Cys-ESAT61–20-TB10.43–11 intranasally immunized mouse is
shown after ex vivo stimulation in the presence of Brefeldin A (10μg/ml) with (a) ESAT61–20 peptide (10μg/
ml) recall, (b) TB10.43–11 peptide (10μg/ml) recall and (c) no peptide recall, used as a negative control to set
gates for cytokine producing cells
312 Anneliese S. Ashhurst et al.
3.3 IFNγ ELISpot First described by Czerkinsky in 1983, ELISpot provides a quanti-
tative measure of cells in the sample capable of releasing antibody or
a particular cytokine, specifically in response to a recall antigen [36–
38]. It is widely considered a sensitive and reproducible technique
and is utilized in both research and clinical diagnostic settings
[39]. Similar to plate-based sandwich ELISA, ELISpot utilizes
complementary antibody pairs that recognize different epitopes of
the desired analyte, in this case IFNγ, and combines this with
membrane-based Western blotting techniques. The first of the
antibody pairs is coated onto a polyvinylidene difluoride (PVDF)-
backed microtiter plate, then leukocytes are cultured in the plate
with the vaccine peptide or protein antigen of interest. During the
incubation, if the sample contains antigen-specific cells capable of
releasing IFNγ, this is released and captured in place on the mem-
brane by the coating antibody. Leukocytes are then washed out of
the plate and IFNγ is detected with a biotinylated or directly
enzyme-conjugated complementary antibody. If needed, avidin or
streptavidin conjugated enzyme is bound to the antibody, then
substrate solution is added to form colored spots at the sites of
cytokine secretion on the membrane. These can then be counted to
quantitate the number of spot-forming units per well, and extra-
polated to provide the number of responding cells per million
leukocytes.
While the protocol presented here provides the materials and
method for detection of murine IFNγ, this may be adjusted to
detect other cytokines or antibodies of interest using complemen-
tary monoclonal antibody pairs at optimized concentrations. Mod-
ification of the cell culture conditions and incubation period may
also be required.
Subheadings 3.3.1 and 3.3.2 should be performed under asep-
tic conditions. The subsequent steps need not be sterile.
314 Anneliese S. Ashhurst et al.
3.3.1 Preparation 1. Wet the membrane at the base of the wells of the ELISpot plate
and Coating with 40 μl of 35% ethanol (v/v prepared in milliQ sterile water)
of the ELISpot Plate for less than 1 min (see Note 11). Wash the plate three times by
pipette (see Note 12) with 200 μl sterile PBS per well, adding
the first wash straight on top of the ethanol. Ensure the mem-
branes do not dry out for the remainder of the protocol. Leave
final PBS wash on until ready to commence the next step.
2. Coat each well of the ELISpot plate with the primary antibody,
purified anti-mouse IFNγ antibody (clone AN18) by pipetting
100 μl antibody diluted in sterile PBS into each well. Incubate
overnight at 4 C. Alternatively incubate at room temperature
for a minimum of 2 h.
3. Pipette out the primary antibody solution and wash with 200 μl
sterile PBS three times as before.
4. To ensure proteins do not bind nonspecifically to the mem-
brane, add 200 μl complete cell culture media into each well as
a blocking agent. Incubate at 37 C for 2 h, or at room
temperature (22 C) if for longer.
3.3.3 Staining 1. Decant cells and wash plate six times with PBST (0.1% Tween
and Developing 20), 200 μl per well, with a pipette. Leave final wash on while
the ELISpot Plate you make up the secondary antibody solution.
2. Add 100 μl per well biotinylated anti-mouse IFNγ antibody
(clone XMG.12, see Note 15) prepared at an appropriate dilu-
tion in 0.5% BSA (w/v)/PBS and incubate at room tempera-
ture for greater than 2 h, or alternatively leave overnight at
4 C.
3. Decant secondary antibody solution and wash plate six times
with PBST as before. Leave final wash on while you make up
secondary antibody label solution.
Immunological Assessment of Inhalational Lipoprotein Vaccines 315
3.5 Quantitation The majority of clinically utilized vaccines aim to induce humoral
of Vaccine-Induced immunity against the pathogen of interest, with a level of vaccine-
Antigen-Specific specific antibodies determined that correlates with protective
Antibodies by ELISA immunity. Antibodies generated against the vaccine antigen can
be readily detected in serum samples by straightforward ELISA
methods. As mice may be bled routinely throughout vaccination
experiments, serum may be collected sequentially from individual
animals to monitor changes to the isotype and titer of antibodies
over time. Take care to detect isotypes specific to the experimental
mouse strain, for instance, C57BL/6 mice produce IgG2c instead
of the IgG2a produced by BALB/c. However, vaccine-induced
antibody may also be found in many peripheral tissues and samples
taken at euthanasia, such as BALF as described in Subheading
3.1.1, nasal washes or peritoneal washes, may also be assessed for
the presence of vaccine-induced antibodies. This may be particu-
larly valuable to detect mucosal IgA if vaccination is delivered by
inhalation [19].
Here we present a standard indirect ELISA-based method to
quantitatively determine vaccine antigen-specific antibody titer in
serum or BALF.
1. Coat high-binding 96-well ELISA plates with 100 μl per well
of the vaccine peptide or protein antigen, at 1 μg/ml diluted in
coating buffer. Seal plate and incubate for 2 h at 37 C or
overnight at 4 C.
2. Decant coating solution, then thoroughly wash the plate by
filling wells completely with wash buffer then decanting, 4–6
times. This may be performed using a squirt bottle or an
automated plate washer, to forcefully wash the wells. Tap
plate firmly on absorbent paper toweling to blot.
3. Add 200 μl blocking buffer per well, seal plate and incubate at
37 C for 1 h.
4. While the ELISA plate is blocking, prepare serial dilutions of
samples in blocking buffer, in a separate nonbinding 96-well
plate. For serum, we recommend a starting dilution of 1:100,
performing an 8 to 12-point curve and 2–5 fold dilution series.
For BALF samples, start at 1:20 dilution. All samples should be
run in technical duplicate (see Note 19).
Immunological Assessment of Inhalational Lipoprotein Vaccines 317
4 Notes
Acknowledgments
References
1. Krammer F (2020) SARS-CoV-2 vaccines in receptor 2 ligand. J Med Chem 55
development. Nature 586(7830):516–527 (15):6968–6974
2. WHO (2019) Global Tuberculosis Report. 8. McDonald DM, Byrne SN, Payne RJ (2015)
World Health Organization, Geneva Synthetic self-adjuvanting glycopeptide cancer
3. Andersen P, Doherty TM (2005) The success vaccines. Front Chem 3:60
and failure of BCG—implications for a novel 9. Bettahi I, Dasgupta G, Renaudet O, Chentoufi
tuberculosis vaccine. Nat Rev Microbiol 3 AA, Zhang X, Carpenter D, Yoon S, Dumy P,
(8):656–662 BenMohamed L (2008) Antitumor activity of a
4. Sable SB, Posey JE, Scriba TJ (2019) Tubercu- self-adjuvanting glyco-lipopeptide vaccine
losis vaccine development: Progress in clinical bearing B cell, CD4+ and CD8+ T cell epi-
evaluation. Clin Microbiol Rev 33(1): topes. Cancer Immunol Immunother 58
e00100–e00119 (2):187
5. Zhu X, Ramos TV, Gras-Masse H, Kaplan BE, 10. McDonald DM, Wilkinson BL, Corcilius L,
BenMohamed L (2004) Lipopeptide epitopes Thaysen-Andersen M, Byrne SN, Payne RJ
extended by an Nϵ-palmitoyl-lysine moiety (2014) Synthesis and immunological evalua-
increase uptake and maturation of dendritic tion of self-adjuvanting MUC1-macrophage
cells through a toll-like receptor-2 pathway activating lipopeptide 2 conjugate vaccine can-
and trigger a Th1-dependent protective immu- didates. Chem Commun 50
nity. Eur J Immunol 34(11):3102–3114 (71):10273–10276
6. McDonald DM, Hanna CC, Ashhurst AS, 11. Belnoue E, Mayol JF, Carboni S, Di Berardino
Corcilius L, Byrne SN, Payne RJ (2018) Syn- BW, Dupuychaffray E, Nelde A, Stevanovic S,
thesis of a self-adjuvanting MUC1 vaccine via Santiago-Raber ML, Walker PR, Derouazi M
diselenide-selenoester ligation-deselenization. (2019) Targeting self and neo-epitopes with a
ACS Chem Biol 13(12):3279–3285 modular self-adjuvanting cancer vaccine. JCI
7. Abdel-Aal A-BM, El-Naggar D, Zaman M, Insight 5(11):e127305
Batzloff M, Toth I (2012) Design of Fully 12. Moyle PM, Toth I (2008) Self-adjuvanting
Synthetic, self-Adjuvanting vaccine incorporat- lipopeptide vaccines. Curr Med Chem 15
ing the tumor-associated carbohydrate Tn anti- (5):506–516
gen and Lipoamino acid-based toll-like
322 Anneliese S. Ashhurst et al.
13. Anderson RJ, C-w T, Daniels NJ, Compton BJ, 22. Satti I, Meyer J, Harris SA, Thomas Z-RM,
Hayman CM, Johnston KA, Knight DA, Griffiths K, Antrobus RD, Rowland R,
Gasser O, Poyntz HC, Ferguson PM, Larsen Ramon RL, Smith M, Sheehan S,
DS, Ronchese F, Painter GF, Hermans IF Bettinson H, McShane H (2014) Safety and
(2014) A self-adjuvanting vaccine induces immunogenicity of a candidate tuberculosis
cytotoxic T lymphocytes that suppress allergy. vaccine MVA85A delivered by aerosol in
Nat Chem Biol 10:943 BCG-vaccinated healthy adults: a phase 1, dou-
14. Azmi F, Ahmad Fuaad AA, Giddam AK, Batzl- ble-blind, randomised controlled trial. Lancet
off MR, Good MF, Skwarczynski M, Toth I Infect Dis 14(10):939–946
(2014) Self-adjuvanting vaccine against group 23. WHO (2012) Measles aerosol vaccine project -
a streptococcus: application of fibrillized pep- report to SAGE. WHO, Geneva, Switzerland
tide and immunostimulatory lipid as adjuvant. 24. Counoupas C, Ferrell KC, Ashhurst A, Bhatta-
Bioorg Med Chem 22(22):6401–6408 charyya ND, Nagalingam G, Stewart EL, Feng
15. Batzloff MR, Hartas J, Zeng W, Jackson DC, CG, Petrovsky N, Britton WJ, Triccas JA
Good MF (2006) Intranasal vaccination with a (2020) Mucosal delivery of a multistage sub-
Lipopeptide containing a Conformationally unit vaccine promotes development of lung-
constrained conserved minimal peptide, a uni- resident memory T cells and affords interleu-
versal T cell epitope, and a self-Adjuvanting kin-17-dependent protection against pulmo-
lipid protects mice from group a streptococcus nary tuberculosis. NPJ Vaccines 5(1):105
challenge and reduces throat colonization. J 25. O’Garra A, Redford PS, McNab FW, Bloom
Infect Dis 194(3):325–330 CI, Wilkinson RJ, Berry MPR (2013) The
16. Rai PK, Chodisetti SB, Maurya SK, Nadeem S, immune response in tuberculosis. Annu Rev
Zeng W, Janmeja AK, Jackson DC, Agrewala Immunol 31(1):475–527
JN (2018) A lipidated bi-epitope vaccine com- 26. Counoupas C, Triccas JA, Britton WJ (2019)
prising of MHC-I and MHC-II binder pep- Deciphering protective immunity against
tides elicits protective CD4 T cell and CD8 T tuberculosis: implications for vaccine develop-
cell immunity against Mycobacterium tuberculo- ment. Expert Rev Vaccines 18(4):354–364
sis. J Transl Med 16(1):279 27. Flórido M, Pillay R, Gillis CM, Xia Y, Turner
17. Xu Z, Rivera-Hernandez T, Chatterjee O, SJ, Triccas JA, Stambas J, Britton WJ (2015)
Walker MJ, Moyle PM (2020) Semisynthetic, Epitope-specific CD4+, but not CD8+, T-cell
self-adjuvanting vaccine development: effi- responses induced by recombinant influenza a
cient, site-specific sortase A-mediated conjuga- viruses protect against Mycobacterium tubercu-
tion of toll-like receptor 2 ligand FSL-1 to losis infection. Eur J Immunol 45(3):780–793
recombinant protein antigens under native 28. Barletta KE, Cagnina RE, Wallace KL, Ramos
conditions and application to a model group SI, Mehrad B, Linden J (2012) Leukocyte
A streptococcal vaccine. J Control Release compartments in the mouse lung: distinguish-
317:96–108 ing between marginated, interstitial, and alveo-
18. Ashhurst A, McDonald D, Hanna C, lar cells in response to injury. J Immunol
Stanojevic V, Britton WJ, Payne RJ (2019) Methods 375(1–2):100–110
Mucosal vaccination with a self-adjuvanted 29. Sakai S, Kauffman KD, Schenkel JM, McBerry
lipopeptide is immunogenic and protective CC, Mayer-Barber KD, Masopust D, Barber
against Mycobacterium tuberculosis. J Med DL (2014) Cutting edge: control of Mycobac-
Chem 62(17):8080–8089 terium tuberculosis infection by a subset of lung
19. Hanna CC, Ashhurst AS, Quan D, Maxwell parenchyma–homing CD4 T cells. J Immunol
JWC, Britton WJ, Payne RJ (2021) Synthetic 192(7):2965–2969
protein conjugate vaccines provide protection 30. Flórido M, Muflihah H, Lin LCW, Xia Y,
against Mycobacterium tuberculosis in mice. Sierro F, Palendira M, Feng CG, Bertolino P,
Proc Natl Acad Sci U S A 118(4): Stambas J, Triccas JA, Britton WJ (2018) Pul-
e2013730118 monary immunization with a recombinant
20. Tyne AS, Chan JGY, Shanahan ER, influenza a virus vaccine induces lung-resident
Atmosukarto I, Chan H-K, Britton WJ, West CD4+ memory T cells that are associated with
NP (2013) TLR2-targeted secreted proteins protection against tuberculosis. Mucosal
from Mycobacterium tuberculosis are protective Immunol 11:1743–1752
as powdered pulmonary vaccines. Vaccine 31 31. Ashhurst AS, Flórido M, Lin LCW, Quan D,
(40):4322–4329 Armitage E, Stifter SA, Stambas J, Britton WJ
21. Sabin AB (1983) Immunization against mea- (2019) CXCR6-deficiency improves the con-
sles by aerosol. Rev Infect Dis 5(3):514–523 trol of pulmonary mycobacterium tuberculosis
Immunological Assessment of Inhalational Lipoprotein Vaccines 323
Abstract
The Bordetella pertussis bacterium is the causative agent of whooping cough (pertussis disease). Following
recent outbreaks of pertussis, disproportionately affecting young infants, several countries have introduced
maternal pertussis vaccination strategies, aimed at boosting transplacental transfer of protective antibodies
during pregnancy. Given historical associations between high maternal antibody and blunted infant
responses to vaccination, subsequent research studies have investigated the impact of maternal pertussis
vaccine on infant humoral responses. However, far less is known about the potential impact of the vaccine
on innate immunity. Here, we describe methods to detect in vitro cellular responses to B. pertussis in
mothers and their infants using a B. pertussis stimulation assay and multiplex cytokine assays to address this
research question.
Key words Maternal vaccination, Pregnancy, Bacterial infection, Innate immunity, Cytokines,
Pediatrics
1 Introduction
325
326 Thomas Rice and Beth Holder
2 Materials
2.1 Patient Samples Samples are required from mother-infant pairs, with equal numbers
of Tdap-vaccinated and unvaccinated pregnancies. The gestational
age at time of vaccination of the mother, and the brand of vaccine
administered should be recorded.
1. Maternal sample(s): at minimum, one sample is required from
the mother; usually at term. For practical reasons, this sample is
often taken near the time of birth, when mothers attend a
healthcare setting. The timing of the sample (before/after
birth) and the mode of delivery should be considered, as
labor is an inflammatory process. If samples are taken after
birth, it is preferable to use samples from caesarean sections.
Likewise, if patients are delivering vaginally, it is preferable to
collect blood prior to commencement of labor (see Note 1).
Ideally, gestation-matched controls from unvaccinated preg-
nancies should also be obtained.
2. Infant samples: at minimum, cord blood taken at birth can
serve as a proxy for neonatal blood (see Note 2). If the aim is
to investigate impact on infant vaccine responses, infant blood
is additionally required at 7 weeks of age (1 week prior to
primary pertussis immunization at 8 weeks) and at 5 months
of age (1 month after completion of the primary pertussis
immunization at 4 months; see Note 3).
2.4 In Vitro Whole 1. 96-well sterile round-bottom tissue culture plate with lid.
Blood 2. Tissue culture hood.
Stimulation Assay
3. Humidified cell culture incubator set to 5% CO2 and 37 C.
4. S-RPMI: Commercially available Roswell Park Memorial Insti-
tute (RPMI 1640) medium supplemented with 10% decom-
plemented fetal calf serum and 1% penicillin–streptomycin.
5. Heat-killed B. pertussis bacteria.
6. 5 mg commercially available bacterial Lipopolysaccharide
(LPS).
7. 3.5 mL bijou.
8. Wet ice.
2.5 Detection 1. Meso Scale Discovery Proinflammatory Panel 1 Human kit (see
of Cytokines by Notes 5 and 6).
Multiplex 2. MESO QuickPlex SQ 120 plate reader and Discovery Work-
and Singleplex Assays bench 4.0 software (free to download).
3. Commercial IL-8 ELISA.
4. High-binding 96-well ELISA plates.
5. Non–high-binding 96-well plate.
6. Reagent reservoirs.
7. Multichannel pipette—ranges 10 μL–200 μL.
8. Adhesive plate sealers.
9. Phosphate buffered saline (PBS): 137 mM NaCl, 10 mM phos-
phate, 2.7 mM KCl, in H2O. Adjust to a final pH of 7.4.
10. ELISA wash buffer: 1 PBS, 0.05% Tween 20.
11. Wet ice.
328 Thomas Rice and Beth Holder
12. Vortexer.
13. 1.5 mL microcentrifuge tubes.
14. Plate shaker.
15. Rocking platform or tube rotator (optional).
16. 2 N H2SO4.
17. Absorbance microplate reader capable of reading 450 nm and
accompanying analysis software.
2.6 Contribution This assay is an adaption of the in vitro whole blood B. pertussis
of the Plasma stimulation assay. As such, reagents and equipment as described in
Compartment in Cord Subheadings 2.2–2.4 are required, in addition to those listed
Blood Cytokine below.
Responses 1. Serum or plasma blood collection tubes.
to B. pertussis
2. Pools of cord blood serum or plasma from unvaccinated and
Tdap-vaccinated pregnancies—40 μL required per assay (see
Note 7).
3. Centrifuge.
4. Microcentrifuge tubes.
3 Methods
3.2 Bulk Preparation Plates are prepared in bulk and frozen to reduce variability and to
of B. pertussis simplify the logistics of working with fresh blood samples. Treat-
Stimulation Plates ments are prepared at 2 the final required concentration, as they
will be diluted by the addition of an equal volume of prepared
blood sample in the stimulation assay (see Subheading 3.4, step 3).
1. To prepare ten plates (each with one set of treatments), thaw
one aliquot of frozen heat-killed B. pertussis stock and four
aliquots of frozen LPS stock on ice (see Note 10).
2. Label ten 96-well round-bottom tissue culture plates with
“B. pertussis stimulation” and the date of preparation.
3. Working in a tissue culture hood, prepare 3.5 mL of bacteria at
1 105 CFU/mL. From a stock of 1 108 CFU/mL (see
Note 11), transfer 35 μL of bacteria to 3465 μL of S-RPMI.
Gently vortex to mix.
4. Pour carefully into a reagent reservoir and, using a multichan-
nel pipette, add 100 μL per well in triplicate (in three adjacent
wells in a row) in each of the ten 96-well plates (see Note 12).
5. For the LPS positive control, pool four aliquots of LPS stock in
a sterile 3.5 mL bijou. Vortex gently to mix (see Note 13).
6. Pour into a reagent reservoir and, using a multichannel pipette,
add 100 μL per well in triplicate, adjacent to the wells contain-
ing the heat-killed B. pertussis, in each of the ten 96-well plates.
7. For the no-treatment (i.e., negative control) wells, add 100 μL
per well of S-RPMI to the plates in triplicate, adjacent to the
wells containing the LPS positive control aliquots.
8. Seal plates with the accompanying lid and freeze at 80 C for
future use (see Note 14).
3.3 Processing 1. Peripheral venous blood samples (minimum 1 mL) from mums
of Maternal, Cord, and babies are collected into blood collection tubes (see Note
and Infant 15).
Whole Blood 2. Immediately invert the blood sample tubes end to end at least
twice to ensure blood is mixed with the anticoagulant.
3. Keep tubes at room temperature and process within 2 h (see
Subheading 3.4, step 2).
3.5 Meso Scale MSD plates can only be used once. Therefore, it is important to
Discovery (MSD) accumulate samples before analyzing cytokine levels in supernatants
Multiplex using this assay.
Cytokine Assay 1. Prepare a plate plan for your experiment (see Note 17).
2. Thaw eight sets of supernatant samples (from Subheading 3.4)
on ice and allow all assay reagents to reach room temperature
before being used (see Note 18).
3. Reconstitute the cytokine calibrator provided in the kit by
adding 1 mL of Diluent 2 to lyophilized MSD Calibrator
Blend and vortexing (see Note 19). Allow the reconstituted
calibrator to sit for a minimum of 5 min before using.
4. In this time, add 300 μL of Diluent 2 to each of eight 1.5 mL
microfuge tubes.
5. Prepare a dilution series of the calibrator by transferring 100 μL
to the first of the tubes containing Diluent 2, prepared above,
to create a fourfold dilution. Mix well by vortexing.
6. Transfer 100 μL to the subsequent tube and repeat this process
to serially dilute 6 more times, leaving the eighth tube with
Diluent 2 alone. This final tube will serve as the calibration
blank.
7. Using a multichannel pipette, aliquot 90 μL Diluent 2 per well
into ten columns of wells in a 96-well plate (not supplied in the
MSD kit; use any non–high-binding plate), according to the
MSD plate layout (see Note 17). This plate will be used for the
dilution of supernatant samples only.
8. Dilute thawed supernatant samples (from step 2 above) 1:10
by transferring 10 μL of each sample, using a multichannel
pipette, into appropriate wells (according to prepared plate
layout) containing 90 μL Diluent 2 and mix by pipetting up
and down at least five times. Change tips between sets of
samples. All samples are tested at a single dilution of 1:10.
9. Add 50 μL of standard and samples to the MSD plate according
to the plate layout (see Note 20).
10. Cover plates with an adhesive plate sealer and incubate for 2 h
at room temperature on a plate shaker at 300 rpm.
Detecting Responses to Maternal Tdap Vaccination 331
3.6 IL-8 ELISA 1. Prepare a plate plan for your experiment (see Note 23).
2. Prepare 1 IL-8 capture antibody in 1 coating buffer (see
Note 24), by adding 40 μL antibody to 10 mL coating buffer
per 96-well plate (see Note 25).
3. Coat ELISA plate with 100 μL per well of IL-8 capture anti-
body (see Note 26).
4. Seal the plate with an adhesive plate sealer and incubate over-
night at 4 C.
5. Aspirate and wash wells three times with 200 μL ELISA wash
buffer to remove unbound antibody (see Note 27). Allow wells
to soak in wash buffer for at least 30 s. Afterward, blot on tissue
paper to remove and excess wash buffer from wells.
6. To prevent nonspecific binding of antigens and antibodies,
block wells with 200 μL 1 diluent (see Note 28).
7. Seal and incubate the plate at room temperature for a minimum
of 1 h.
8. Whilst the plate is being blocked, thaw supernatants on ice (see
Note 29).
9. Using standard nonsterile 96-well plates (not high-binding),
dilute assay supernatants to an intermediate dilution of 1:50 in
PBS according to the plate plan designed in step 1 (see Note
30). For example, in one plate, dilute 10 μL of sample in 90 μL
of PBS for a 1:10 dilution. In a separate plate, take 20 μL of the
diluted sample from plate one, and add to 80 μL PBS (making
an overall intermediate dilution of 1:50; see Note 31). Ensure
332 Thomas Rice and Beth Holder
during dilution steps that sample and PBS are well mixed by
pipetting up and down at least five times. When used in step 15
below, the final dilution for samples will be 1:500 (see Note
32).
10. Reconstitute the IL-8 standard provided in the kit using the
amount of deionized water stated on the vial, and place on a
rocking platform or tube rotator for at least 15 min to solubi-
lize (see Note 33).
11. Whilst the standard is being reconstituted, wash the blocked
ELISA plate twice as described in step 5, and blot-dry. It is
important not to leave the wells dry for long periods of time
between steps.
12. To columns 1 and 2 of the plate, apart from wells A1 and B1
which will contain the reconstituted standard, add 100 μL of
1 diluent. In addition, 100 μL of 1 diluent should be added
to the two designated blank wells G12 and H12.
13. To all wells that will receive samples, add 90 μL of 1 diluent
(see Note 34).
14. Add 200 μL of the reconstituted IL-8 standard to wells A1 and
B1. Using a multichannel pipette, take 100 μL from A1 and B1
into A2 and B2, pipetting up and down to mix with the diluent
present in the well. Change tips, and repeat the process a
further six times, discarding 100 μL from A8 and B8. This
will create a serially diluted eight-point standard curve.
15. Add 10 μL of prediluted 1:50 sample from step 9 above to
designated sample wells containing 90 μL of 1 diluent,
making a final sample dilution of 1:500.
16. Once all samples and standards are added, seal the plate with an
adhesive plate sealer and incubate at room temperature for 2 h.
17. Prepare biotin-conjugated IL-8 detection antibody by adding
40 μL antibody to 10 mL 1 diluent per 96-well plate, or the
same volumes as for capture antibody if running partial plates
(see Note 25).
18. Wash and aspirate plates four times with wash buffer as
described in step 5.
19. Add 100 μL diluted IL-8 detection antibody per well, seal plate
and incubate at room temperature for 1 h (see Note 35).
20. Prepare avidin-conjugated horseradish peroxidase (HRP)
enzyme by adding 40 μL HRP conjugate to 10 mL 1 diluent
for a 96-well plate, or same volumes as for capture antibody if
running partial plates (see Note 25).
21. Wash and aspirate plates four times with wash buffer as
described in step 5.
Detecting Responses to Maternal Tdap Vaccination 333
22. Add 100 μL prepared HRP per well, seal plate and incubate at
room temperature for 30 min.
23. Wash and aspirate plates six times with wash buffer as described
in step 5.
24. Add 100 μL of 3,30 ,5,50 -tetramethylbenzidine (TMB) per well
(see Note 36).
25. Monitor the colour change of sample and standard wells from
clear to blue to ensure samples and standards do not become
saturated. When the standard curve is colored blue, with a
visible difference between the middle standards (see Note
37), stop the reaction by the addition of 100 μL 2 N H2SO4
(see Note 38).
26. Immediately read the plates at 450 nm using an absorbance
microplate reader. If wavelength subtraction is available, sub-
tract the values of readings at 570 nm from those at 450 nm
and analyze data (see Note 39).
3.7 Contribution 1. Collect blood in blood collection tubes. The assay requires
of the Plasma 400 μL of cord whole blood.
Compartment in Cord 2. Dilute cord blood 1:5 in S-RPMI. After dilution, remove
Blood Cytokine 700 μL of this into a microcentrifuge tube for use in control
Responses wells.
to B. pertussis 3. Centrifuge the rest of the diluted blood at 500 g for 5 min.
4. Remove the top clear plasma layer, without disturbing the cells
beneath and record the volume taken off. Replace this volume
with S-RPMI (see Note 40).
5. Prepare 96-well cell culture stimulation plate (see Fig. 1 for
layout). To eight wells (1A-1D and 2A-2D), add 80 μL S-
RPMI. To eight wells (3A-3D and 4A-4D), add B. pertussis
prepared in 80 μL S-RPMI. To 2 wells (5A and 6A), add
100 μL of 2 μg/mL LPS.
1 2 3 4 5 6
No treatment A S-RPMI
-Plasma B
B. pertussis
-Tdap serum/plasma C
+Tdap serum/plasma D LPS
Fig. 1 Plate layout showing the stimulation conditions for the plasma removal
assay. Cord blood is stimulated with S-RPMI, B. pertussis and LPS. Stimulations
are done with plasma removed from cord blood and replaced with S-RPMI
(plasma), with plasma removed and replaced with cord plasma/serum from
unvaccinated pregnancies (Tdap serum/plasma) or with plasma removed and
replaced with cord plasma/serum from Tdap-vaccinated pregnancies (+Tdap
serum/plasma)
334 Thomas Rice and Beth Holder
4 Notes
16. Be careful not to disturb the cell pellet sitting at the bottom of
the well. Approximately 160 μL of supernatant can be removed
using a multichannel pipette without disturbing the cell pellet.
This can be done in two 80 μL draws into one plate, mixing by
pipetting up and down, and then removing 80 μL into the
second plate.
17. MSD plates are precoated with capture antibodies for each
target, located on independent spots within each of the
96 wells on the plate. MSD plates are expensive and can only
be used once; therefore, it is good to make maximal use of the
available wells. For the experiment described in this chapter,
each blood sample requires nine wells in order to test the
effects of three treatments, each tested in triplicate. A sug-
gested MSD plate plan that can accommodate eight sets of
samples is shown in Fig. 2. As columns 1 and 2 will contain
duplicates of standards, the eight sets of samples must be
arranged in in consecutive rows (A to H) of nine columns,
which would leave one column empty. In order not to waste
these wells, other samples, unrelated to the experiment
described in this chapter may be tested in this column. A
different layout will be required for the samples from Subhead-
ing 3.7.
18. Upon the first thaw of MSD assay diluents, aliquot into smaller
volumes for future use to avoid repetitive freeze–thaw cycles.
8 mL aliquots of Diluent 2 and 3 mL aliquots of Diluent 3 can
be made. Aliquots can be kept at 20 C.
19. Each kit contains a vial of lyophilized calibrator per MSD plate.
The reconstituted calibrator is stable for 1 day at 2–8 C.
1 2 3 4 5 6 7 8 9 10 11 12
A Standard
B Blank
C
D B. pertussis
E LPS
F
S-RPMI
G
H Empty well
Fig. 2 MSD plate layout. Shown is the suggested layout for testing of eight sets of blood samples, each
stimulated with the three treatments (S-RPMI, B. pertussis, and LPS) run in triplicate. The assay standards and
blanks are applied to wells in columns 1 and 2, and the eight sets of samples are applied, one per row, in wells
of columns 3 to 11, as shown
Detecting Responses to Maternal Tdap Vaccination 337
20. It is important that the full surface of the well is covered with
sample. Reverse pipetting can help to prevent bubbles in the
wells. To reverse pipette, set the pipette to the desired volume.
When pressing the pipette plunger down to take up a solution,
go past the first stop. Immerse the tip in the liquid, and slowly
release the plunger to full extension. The takes up more vol-
ume of solution than set on the pipette. When dispensing, press
the plunger down to the first stop. A small volume of liquid will
remain in the tip, which helps to prevent bubbles.
21. For the detection antibody cocktail, use 60 μL per antibody, or
60 9 ¼ 540 μL. For 3 mL total volume, add 2.46 mL Diluent
3. NB: the IL-8 antibody is not included in the cocktail, as
sample levels are regularly out of range in the MSD assay, so are
tested separately by ELISA.
22. Read Buffer is provided at 4 concentration, dilute to 2 in
deionized water. For a 96-well plate, add 7.5 mL of Read
Buffer to 7.5 mL of deionized water.
23. A suggested ELISA plate plan that can accommodate eight sets
of samples, tested at a single dilution, is shown in Fig. 3.
Columns 1 and 2 will contain duplicates of standards, and the
eight sets of samples are tested in consecutive rows (A to H) of
columns 3 to 11. The final column contains two wells used for
blanks and six empty wells. A different layout will be required
for the samples from Subheading 3.7.
24. Coating buffer is provided as a 10 stock, dilute to 1 in
deionized water. For one full plate, dilute 1 mL coating buffer
in 9 mL deionized water.
1 2 3 4 5 6 7 8 9 10 11 12
A Standard
B Blank
C
D B. pertussis
E LPS
F
S-RPMI
G
H Empty well
Fig. 3 IL-8 ELISA plate layout. Shown is the suggested layout for testing of eight sets of blood samples, each
stimulated with the three treatments (S-RPMI, B. pertussis, and LPS) run in triplicate. The assay standards are
applied to wells in columns 1 and 2, and the eight sets of samples are applied, one per row, in wells of
columns 3 to 11, as shown. Two blank wells are included in column 12
338 Thomas Rice and Beth Holder
Table 1
Volumes of reagents for partial ELISA plates
25. Partial ELISA plates can be run using the reagent volumes
shown in Table 1.
26. Pipette into wells by placing the tips of the pipettes on the rim
of one side of the well. This will help prevent contamination
between wells in later steps.
27. Wells can be washed and aspirated using an automatic plate
washer, or the contents can be tipped out into a laboratory sink
and washed with a multichannel pipette as described earlier in
the protocol (see Subheading 3.5, step 12).
28. Diluent is provided as a 5 stock to be diluted to 1 in
deionized water. Add 10 mL diluent to 40 mL deionized
water. The excess 1 diluent can be saved for later steps in
the ELISA experiment.
29. The IL-8 ELISA and MSD assays can be performed separately,
as stimulation supernatants are frozen in two separate plates, to
minimize freeze-thawing.
30. Standards are prepared separately, so there is no need to add
PBS to columns 1 and 2 for the sample predilution steps.
31. A 1:50 dilution can be done in 96-well plates in two steps (1:10
followed by 1:5) as described, with a final (1:10) dilution being
done in the ELISA plate, giving an overall sample dilution of
1:500. However, these volumes can be changed, as long as the
desired final 1:500 dilution of sample is achieved. For accuracy,
it is advised to avoid pipetting volumes less than 5 μL.
32. A dilution of 1:500 was determined to be optimal for sample
testing at a single dilution.
33. If no rocking platform or tube rotators are available, mixing
can be done periodically over the 15 min by hand. Invert the
bottle gently, avoiding foaming.
34. This volume may change depending on the order in which
samples are diluted to make a 1:500 final dilution, as long as
the final volume in the ELISA plate is 100 μL (see Note 31).
35. After the sample incubation, all subsequent reagents should be
added at a consistent speed and order across the plate to make
the incubation time in each well as consistent as possible.
36. TMB is a chromogenic substrate that reacts with HRP, result-
ing in a colour change when the assay target is present in each
Detecting Responses to Maternal Tdap Vaccination 339
sample. When pipetting this into the wells, lean the tips on the
opposite side of the well to previous steps to avoid cross-
contamination.
37. It is important to ensure the reaction is not stopped too early,
before wells with low-level concentrations have developed, or
too late, when the colour change becomes saturated leading to
inaccurate standard curve and/or plateauing of samples. It can
be helpful to use standard rows C and D as a guide; as long as
there is still a difference in colour between these wells, the
reaction can continue. If samples wells turn brightly colored
immediately and before the standard curve can develop, stop
the reaction as the sample concentrations will be too high and
out of range. If needed, repeat experiment with a new dilution.
38. Sulfuric acid is a highly corrosive chemical that is potentially
explosive in concentrated form. It can cause severe skin burns,
can irritate the nose and throat and cause difficulties breathing
if inhaled. Laboratory personal protective equipment should
be worn when using, including gloves and a lab coat.
39. The accompanying analysis software to the microplate reader
will produce a concentration of IL-8 in each sample by extra-
polating from the standard curve values, taking into account
the sample dilution factor.
40. Be careful not to disturb the blood layer below the plasma. It
can help to remove the final volume of plasma with a smaller
volume pipette, such as P200 micropipette.
References
1. Sawyer M, Liang JL, Messonnier N, Clark TA 5. Palmeira P, Quinello C, Silveira-Lessa AL et al
(2012) Updated recommendations for use of (2012) IgG placental transfer in healthy and
tetanus toxoid, reduced diphtheria toxoid, and pathological pregnancies. Clin Dev Immunol
acellular pertussis vaccine (Tdap) in pregnant 2012:985646
women--advisory committee on immunization 6. Byrne P, McGuirk P, Todryk S, Mills KHG
practices (ACIP). Morb Mortal Wkly Rep 62 (2004) Depletion of NK cells results in disse-
(7):131–135 minating lethal infection with Bordetella per-
2. Wiley KE, Massey PD, Cooper SC et al (2013) tussis associated with a reduction of antigen-
Pregnant women’s intention to take up a post- specific Th1 and enhancement of Th2, but
partum pertussis vaccine, and their willingness not Tr1 cells. Eur J Immunol 34
to take up the vaccine while pregnant: a cross (9):2579–2588
sectional survey. Vaccine 31(37):3972–3978 7. Higgins SC, Lavelle EC, McCann C et al
3. Donegan K, King B, Bryan P (2014) Safety of (2003) Toll-like receptor 4-mediated innate
pertussis vaccination in pregnant women in IL-10 activates antigen-specific regulatory T
UK: observational study. BMJ 349(9526): cells and confers resistance to Bordetella pertus-
g4219 sis by inhibiting inflammatory pathology. J
4. Gall SA, Myers J, Pichichero M (2011) Mater- Immunol 171(6):3119–3127
nal immunization with tetanus–diphtheria–- 8. Mahon BP, Sheahan BJ, Griffin F et al (1997)
pertussis vaccine: effect on maternal and Atypical disease after Bordetella pertussis respi-
neonatal serum antibody levels. Am J Obstet ratory infection of mice with targeted disrup-
Gynecol 204(4):334. e1 tions of interferon-γ receptor or
340 Thomas Rice and Beth Holder
immunoglobulin μ chain genes. J Exp Med 186 the first week of human life follow a robust
(11):1843–1851 developmental trajectory. Nat Commun 10
9. Kroes MM, Mariman R, Hijdra D et al (2019) (1):1–14
Activation of human NK cells by Bordetella 11. Thalen M, Van Den Ijssel J, Jiskoot W et al
pertussis requires inflammasome activation in (1999) Rational medium design for Bordetella
macrophages. Front Immunol 10:2030 pertussis: basic metabolism. J Biotechnol 75
10. Lee AH, Shannon CP, Amenyogbe N et al (2–3):147–159
(2019) Dynamic molecular changes during
Chapter 18
Abstract
Complement is a key component of functional immunological assays used to evaluate vaccine-mediated
immunity to a range of bacterial and viral pathogens. However, standardization of these assays is compli-
cated due to the availability of a human complement source that lacks existing antibodies acquired either
through vaccination or natural circulation of the pathogen of interest. We have developed a method for
depleting both IgG and IgM in 200 mL batches from pooled hirudin-derived human plasma by sequential
affinity chromatography using a Protein G Sepharose column followed by POROS™ CaptureSelect™ IgM
Affinity resin. The production of large IgG- and IgM-depleted batches of human plasma that retains total
hemolytic and alternative pathway activities allows for improved assay standardization and comparison of
immune responses in large clinical trials.
Key words Complement system, IgG depletion, IgM depletion, Human plasma, FPLC
1 Introduction
341
342 Frances Alexander et al.
2 Materials
2.1 Plasma 1. Freshly drawn blood from volunteer donors (see Note 1), using
and Blood Plasma butterfly cannulas and 50 mL syringe.
Preparation 2. Recombinant hirudin mature variant from Pichia pastoris
freeze dried by request, >1 104 antithrombin units
(ATU)/mg (Creative BioMart). Reconstitute hirudin to
20 mg/mL using sterile water for irrigation (see Note 2).
Add sterile hirudin to 50 mL polypropylene tubes to give
2 mg per 50 mL whole blood.
3. Benchtop centrifuge: speeds up to 3000 g.
2.4 Chromatography 1. IgG depletion resin: 360 mL Protein G Sepharose . Linear flow
Columns rate ¼ 150–250 cm/h.
2. IgG depletion column: 400 50 mm h internal diameter.
3. IgM depletion Resin: 70 mL POROS™ CaptureSelect™ IgM
Affinity resin. Linear flow rate ¼ 150 cm/h.
Large-Scale Depletion of IgG and IgM from Human Plasma 345
2.5 Chromatography 1. AKTA FPLC, fraction collector placed within a Class II micro-
Systems biological safety cabinet (MSC II), at 22 C with UV monitor
of 280 nm, conductivity cell and pH monitor.
2. AKTA Purifier, in cold room (or 4 C) with UV monitor of
280, 254, 215 nm, conductivity cell, pH, and temperature
monitor.
2.6 Concentration 1. Sterilized dialysis tubing: Snakeskin 3.5 K molecular weight cut
of Plasma, off (MWCO) tubing, autoclaved in HPLC water.
Complement, and C1q 2. Polyethylene glycol 20,000 Da.
3 Method
All steps should be carried out using aseptic technique and where
possible within an MSC II to maintain sterility, the exterior of all
equipment should be sprayed with 70% ethanol (see Note 5). To
prevent activation of complement during the processing care must
be taken to avoid certain plastics (see Note 6).
3.1 Plasma 1. Collect venous human blood from multiple donors using but-
Preparation terfly cannulas and 50 mL syringes and immediately decant into
polypropylene centrifuge tubes containing hirudin (final con-
centration 0.04 mg/mL) on ice. Mix the blood gently with the
hirudin by inversion of the tube, without frothing.
2. Centrifuge at 3000 g for 10 min to separate red blood cells.
All particulate matter is removed, and the native plasma is snap
frozen in small batches, see Notes 7 and 8.
3. Defrost the small batches under running water and pool before
snap freezing in 45 mL aliquots.
4. On day of depletion, defrost the plasma in approximately
200 mL batches and centrifuge again at 3000 g for 10 min
to remove any particulates or clots, see Note 9. Take 5 1 mL
Large-Scale Depletion of IgG and IgM from Human Plasma 347
3.2 Column Setup 1. Place the columns in a clamp within a laminar flow cabinet or
and Sanitization MSC II and dismantle and check the O rings and adaptors (top
and base) for integrity.
3.2.1 Column
Preparation 2. Wash the columns with sterile HPLC grade water (also sanitize
with 0.1 M sodium hydroxide if previously used) and immerse
all wet areas in sterile 70% ethanol for 12 h minimum. Immerse
the top adaptor separately (not within the column).
3. Discard the 70% ethanol and replace with a sterile 20% ethanol
wash before packing the columns with the appropriate resin, see
Note 10.
4. Place the top adaptor on a sterile surface (foil or paper) and
insert and seal when the column has been packed.
3.2.2 Sanitization 1. Assemble the chromatography systems with blank lines in place
of Chromatography of columns, with dual end connector switches.
Systems with Sodium 2. Set the pressure limit for system to 0.5 Pa.
Hydroxide (See Notes 11
3. Purge the system with water or 20% ethanol followed by water
and 12)
to neutralize the pH and remove all traces of salt from the
system. Monitor the pH readings as the valves and lines change;
a plateaued and neutral trace is required and may take 3–6
system volumes (sv) to achieve. A program can be used to
prime pumps and then purge all lines, valves, ports and fraction
collector in sequence.
4. Prime the system with 0.1 M sodium hydroxide and run a
program to sanitize all pumps, valves, ports, lines fraction
collector, and dead space for 1 h contact time.
5. Using a program (see Note 13), prime and flush with HPLC
grade water to remove sodium hydroxide from all pumps,
valves, lines, and dead space settings until the absorbance,
conductivity, and pH traces indicate a neutral pH or the trace
has stabilized. Ensure the sample line is included.
6. The cold room setting may require use of 20% ethanol to clear
Bernoulli effects and enhance clearance of edge effect of the
sodium hydroxide from the tubing and valves—this should be
run through as previously stated. Gather the collection lines in
a single sterile collection vessel and allow these lines to void
20% ethanol.
7. The system is now sterile and ready to be equilibrated. The
columns may now be connected into separate column ports if
they are already in 20% ethanol.
348 Frances Alexander et al.
3.2.3 Sanitization 1. Ensure columns are primed with 20% ethanol prior to transfer
of Columns at 4 C to 4 C.
with 70% Ethanol (See 2. Set pressure limit for 4 C system to 0.35 Pa.
Note 14)
3. Insert the columns into separate column ports in the system,
with 20% ethanol running at a low flow rate to avoid air in lines.
Reverse flow may be applied to the columns. Spray connection
points externally with 70% ethanol when connecting columns
to maintain sterility.
4. Run columns separately for 3–6 column volumes (cv) in 20%
ethanol or until conductivity has plateaued.
5. Prime the pump and flush bypass lines with 70% ethanol.
6. For the Protein G column: run 70% ethanol through the
column for 1 h contact time, and then flush with 20% ethanol.
Close the column top and bottom manual switch valves.
7. For the POROS™ CaptureSelect™ IgM column: equilibrate
the column for 12 h contact time then flush with 20% ethanol.
Close the column top and bottom manual switch valves.
8. Ensure both columns are equilibrated in 20% ethanol. This will
take a minimum of 6 cv.
9. When both columns have been sanitized and equilibrated back
to 20% ethanol, they are ready for use or storage.
3.3 IgG and IgM 1. Ensure the system and columns have been sanitized and are
Depletion of Plasma ready for use.
3.3.1 Equilibration 2. Set the pressure limits for the system and column to 0.35 Pa for
of the Chromatography the Protein G column and 0.5 Pa for POROS™ CaptureSe-
Systems and Columns lect™ column.
at 4 C 3. Open the manual switches and run 20% ethanol slowly through
both column ports.
4. Each column should be connected separately to the chroma-
tography system, see Note 15. Flush columns and all parts of
the system individually with water, including the sample pump.
Continue to flush each column with water until both the UV
and conductivity traces have stabilized.
5. Repeat step 4 using PBS for a minimum of 6 cv and ensure
pH trace has reached 7.4.
3.3.2 Loading 1. Ensure columns and system have been sanitized and equili-
the Plasma onto brated in PBS at 4 C and plasma is prepared for use (as de-
the Columns scribed in Subheading 3.2).
2. Set the pressure limits to 0.35 Pa for the system and columns.
3. Connect the columns together with the Protein G (for IgG
depletion) in front of the POROS™ CaptureSelect™ (for IgM
depletion) (see Fig. 1). Allow the pressure and monitor traces to
Large-Scale Depletion of IgG and IgM from Human Plasma 349
Fig. 1 Schematic diagram showing the setup of the chromatography system and columns for IgG and IgM
depletion at 4 C. The system is equilibrated and primed with PBS using pump A or B. Plasma is loaded onto
the columns via pump C and passes first over the Protein G Sepharose column, then the POROS™
CaptureSelect™ IgM Affinity column. UV, conductivity, and pH monitors are used to assess the composition
of column breakthrough, which is then directed to waste or collected accordingly
adjust and stabilize by flushing the columns with PBS for 3–
6 cv (use cv of both columns combined) and autozero the UV
detector once this is achieved. Use manual switch valves and
spray externally with 70% ethanol to ensure sterility when con-
necting the columns.
4. Aseptically place the collection lines into separate sterile collec-
tion vessels.
5. Set pump C running with the sample feed directed to the
system bypass waste and use pump A or B on a slower flow
rate, to pump PBS over the columns to waste. Once pumps are
running, apply the plasma to the columns using pump
C. Gradually raise the flow rate of pump C to approximately
5 mL/min.
350 Frances Alexander et al.
6. When all plasma has been loaded onto the system, transfer
pump C into PBS, taking care not to allow air into the lines
and clear plasma from the sample line and pump. Once cleared,
pump C can be switched off and pump A or B can be used to
direct PBS over the columns.
7. Monitor the UV 280 nm trace and collect the breakthrough
from 20 mAU until the traces indicate the breakthrough has
cleared and the monitors have plateaued again. Collect the
breakthrough until the 280 nm trace has dropped to
20 mAU. This indicates that large proteins have passed over
the columns and been collected.
8. Once the breakthrough is collected, flush the sample pump
with PBS to void, stop all pumps and disconnect columns
from the system and each other and seal with the manual
switches. Transfer columns to the second chromatography sys-
tem at 22 C and allow to acclimatize—this may take up to 3 h.
Sanitize the 4 C system as described in Subheading 3.2 (this
does not have to be carried out immediately after removal of
columns).
9. Keeping the breakthrough at 4 C, pool fractions before
removing QC samples (see Subheading 3.5) and concentrating
the depleted plasma (see Subheading 3.4.3).
3.4 C1q Elution, 1. Set up the fraction collector within an MSC II (to maintain
Concentration sterility) and sanitize both the system and fraction collector.
of Components Aseptically load 15 mL tubes (minus lids) into the fraction
and Reconstitution collector carousel and only insert this once the system has
been sanitized (avoid sodium hydroxide splashing into tubes).
3.4.1 Elution of C1q
at 22 C with Salt Gradient
2. Ensure columns are acclimatized to 22 C and equilibrated
(See Note 16)
in PBS.
3. With the system acclimatized to 22 C, set the pressure limits to
0.35 Pa for the system and columns (see Note 17), and auto-
zero the UV. Autozero with PBS flowing through the
bypass line.
4. Insert the Protein G column between column selection valves
(see Fig. 2) and start pump A (PBS) at a flow rate of 1 mL/min.
5. Apply a linear salt gradient of 0–1.5 M NaCl, at a flow rate of
1.6 mL/min for 1100 min (total 1760 mL). Collect 12 mL
fractions in an MSC II to maintain sterility.
6. Identify fractions containing C1q using the UV monitor trace
and SDS PAGE (see Note 18). Pool all fractions with C1q and
concentrate (see Subheading 3.4.3).
Large-Scale Depletion of IgG and IgM from Human Plasma 351
Fig. 2 Schematic diagram showing the setup of the chromatography system and columns for the elution of
C1q at room temperature. Pump A is primed with PBS, and pump B is primed with PBS supplemented with
1.5 M NaCl. C1q is then eluted from the Protein G Sepharose using a salt gradient and collected in 12 mL
fractions. The UV monitor trace can be used in identifying which fractions contain eluted C1q
3.4.2 Elution of IgG 1. Ensure the salt gradient has been completed for the Protein G
and IgM for Reuse column and C1q has been collected (see Subheading 3.4.1).
of Column (See Note 19) Ensure the POROS™ CaptureSelect™ column is acclimatized
to room temperature.
2. Connect columns to the system in parallel (see Fig. 3).
3. Equilibrate the Protein G column separately back into PBS.
4. Apply the elution buffer, using pump C, to the Protein G
column for a maximum of 1 h or until a peak has eluted.
5. Apply PBS to the column until it is equilibrated before flushing
with water and 20% ethanol.
6. Collect the peaks manually and assess the protein profile by
SDS PAGE and western blotting, see Subheading 3.5.1, to
ensure antibodies have been eluted.
7. Repeat steps 3–6 for the Poros™ CaptureSelect™ column to
elute the IgM separately.
352 Frances Alexander et al.
Fig. 3 Schematic diagram showing the setup of the chromatography system and columns for the IgG and IgM
elution at room temperature. Pump C is primed with elution buffer and antibodies are eluted from the
respective columns into multiple collection lines. The UV monitor trace can be used in identifying which
fractions contain eluted antibody
3.4.3 Concentration 1. Set up sterile dialysis tubing, forceps, scissors, and lint-free
of Plasma and C1q paper in an MSC II.
and Recovery from Dialysis 2. Work aseptically in an MSC II but do not spray the outside of
Tubing tubing containing depleted plasma or C1q with 70% ethanol.
3. Fill the dialysis tubing with the depleted plasma breakthrough
or C1q fractions, void any residual air and knot the ends,
leaving 5 cm lengths of tubing post knots. Dry down the
outside of the tubing. Weigh the tubing once filled, for use as
a guide to the rate of concentration.
Large-Scale Depletion of IgG and IgM from Human Plasma 353
3.4.4 Reconstitution 1. Pre-chill the labeled internally threaded cryovials and racks.
of Complement with C1q 2. Defrost the required depleted plasma and the paired C1q.
and Aliquoting for Storage
3. Centrifuge the depleted plasma at 3000 g for 10 min to
remove all particulates or clots.
4. Within an MSC II, pool all depleted plasma and C1q into a
sterile glass Duran bottle on ice, mix gently and the aliquot into
50 mL falcon tubes (for ease of aliquoting) and store at 4 C for
immediate aliquoting.
5. Aseptically pipette into 0.5 mL or 1 mL aliquots as required.
Snap-freeze the vials (see Note 4) and store at 80 C.
3.5 Batch Quality 1. Samples are loaded onto 4–12% SDS-PAGE under reduced
Testing conditions; native plasma and reconstituted complement are
loaded at a 1:20 dilution, while C1q and all other samples are
3.5.1 SDS-PAGE
loaded neat.
and Western Blotting
2. One gel should be stained for protein and the subsequent gels
should be transferred to polyvinylidene difluoride (PVDF)
membrane by western blotting and probed with anti-IgG and
anti-IgM primary antibodies to show complete removal of the
immunoglobulins from the final depleted plasma.
3.5.2 Bioburden Check 1. Apply 20 μL of each sample to each of the three types of agar
plate.
2. Incubate at 37 C for 5 days at 5% CO2.
3. Observe contaminant growth.
4. Complete sterility is required for use of the depleted plasma in
functional assays therefore discard any batch where any micro-
bial contamination is seen.
354 Frances Alexander et al.
Table 1
Serum bactericidal activity reciprocal titers against Neisseria meningitidis strain NZ98/254 and
B. pertussis strain B1917 using three different batches of IgG- and IgM-depleted plasma. Reciprocal
bactericidal titers are accepted within 1 doubling dilution of the mean assigned titer calculated
through repeated testing; NT, not tested
4 Notes
1. The volunteer blood and plasma have not been screened for
infectious agents and should always be handled wearing gloves,
safety glasses, and lab coat by staff who have been vaccinated
against Hepatitis B.
2. Add sterile water to the hirudin vial and leave for 30 min before
aliquoting the resuspended hirudin to required polypropylene
tubes. The tubes may be frozen if not used immediately. Safety
note: This is a concentrated and irreversible anticoagulant; care
must be taken when handling and the use of sharps should be
avoided.
3. All buffers must be filtered, prepared from the highest quality
grade components possible, and using HPLC grade water.
When preparing the buffers, a sterile lid is aseptically placed
on the filtered buffer bottle and covered with a sterile foil cover.
All buffers and solutions are prepared and filtered ahead of the
purification, the only one prepared fresh on the day is glycine
buffer for the antibody elution. Buffers are left to equilibrate to
required temperature for 3 h prior to use.
4. Snap freezing is performed using an ethanol bath containing
dry ice (solid carbon dioxide pellets). The solution may be as
cold as 70 C so care must be taken to not freeze fingers or
splash ethanol over the body, face, or clothes. The cryotubes
used should be selected to avoid both activation of the comple-
ment and plasma coagulation; the “O” ring and lid composi-
tion should be considered, and in our hands we found
internally threaded cryovials gave the better results. Tubes are
filled to 80% maximum volume, tightly sealed, and immersed
to the level of the liquid to ensure even and immediate super-
cooling of the contents. After reconstitution of the 200 mL
complement, the solution is subdivided into 4 or 5 prechilled
polypropylene tubes or glass vessels and remains chilled until
dispensed. The complement is dispensed as 0.2–1 mL aliquots
into pre-chilled and pre-labeled cryovials ready for use. The
tubes are tightly sealed and placed in racks sitting in the ethanol
bath sufficient to cover the contents but not the lid or the seal.
The use of cryogenic storage boxes with fixed hollow pronged
dividers allows rapid permeation of the freezing mixture
356 Frances Alexander et al.
15. Columns should only be added to the systems when the system
contains a sterile and compatible buffer/solution. The flow
rate should be set to a slow flow rate (e.g., 0.5–1.0 mL/
min). The upper blanking line switch should be opened, and
the blanking line uncoupled downstream of the valve so the
liquid flows through; these are held in one hand and sprayed
with 70% ethanol. The column top adaptor line is uncoupled
downside of the column top switch while the base switch
remains closed and the top line sprayed immediately with
70% ethanol. The top adaptor line is inserted into the down-
stream of the flowing blanking line. The base adaptor switch is
uncoupled to prevent back pressure. This process is repeated
with the base switches. The column switches should be period-
ically rotated into the system to ensure they become regularly
sanitized with sodium hydroxide.
16. C1q is in an equilibrium within the plasma and bound to
circulating antibody [34, 35]. When the base plasma prepara-
tion is collected in the column breakthrough, a proportion of
the C1q remains attached to the antibody which is now bound
to the column. Antibody binds to the column by the Fc region,
and so to avoid possible leaching of the antibody by the sudden
addition of high salt to the column, a slow and gentle salt
gradient is applied, thereby resulting in the C1q ionically dis-
sociating from the antibody, and consequently leaving the
antibody attached to the Protein G matrix. In our hands,
transferring the Protein G column from 4 C and equilibrating
to 22 C gives clear separation of the C1q from other proteins
along the gradient. The C1q peak is very minor (~40 mAU)
and may be easily missed (M Pangburn personal communica-
tion and Ref. 31) without analysis by SDS PAGE. Pure C1q is
observed in the second peak of the elution profile
corresponding to ~40% pump B elution. Alternatively, an iso-
cratic elution may be made to clear all proteins by stepping
directly to 40% high salt buffer concentration, and then 100%
to clear all. Elution of the C1q may be scaled up or down using
the ratio of the gradient over time for different column sizes.
17. Use of newer AKTA systems (or other chromatography sys-
tems) allows the selection of the column delta pressure and so
finely tunes the pressure exerted across the column without
being tripped by the internal pressures of the pump. This is
noticeable when eluting the C1q from the Protein G column at
low flow rates when the piston pump reverse flow creates a
pressure in excess of that set for the pressure limit.
18. Sampling of the fractions is made from every sixth tube and the
fractions collected for all six either side of the last C1q sample
identified by SDS PAGE. The C1q remains in the MSC II until
the SDS PAGE gel profile has been confirmed (using neat
360 Frances Alexander et al.
References
bactericidal antibody activity. In: Vaccine. Else- 13. Ji X, Olinger GG, Aris S, Chen Y, Gewurz H,
vier BV, Amsterdam, pp 2222–2227 Spear GT (2005) Mannose-binding lectin
5. Terajima M, Cruz J, Co MDT, Lee J-H, binds to Ebola and Marburg envelope glyco-
Kaur K, Wrammert J, Wilson PC, Ennis FA proteins, resulting in blocking of virus interac-
(2011) Complement-dependent lysis of influ- tion with DC-SIGN and complement-
enza a virus-infected cells by broadly cross- mediated virus neutralization. J Gen Virol
reactive human monoclonal antibodies. J 86:2535–2542. https://doi.org/10.1099/vir.
Virol 85:13463–13467. https://doi.org/10. 0.81199-0
1128/JVI.05193-11 14. Maslanka SE, Gheesling LL, Libutti DE,
6. Townsend K, Ladhani SN, Findlow H, Borrow Donaldson KBJ, Harakeh HS, Dykes JK,
R (2014) Evaluation and validation of a serum Arhin FF, Devi SJN, Frasch CE, Huang JC,
bactericidal antibody assay for Haemophilus Kriz-Kuzemenska P, Lemmon RD,
influenzae type b and the threshold of protec- Lorange M, Peeters CCAM, Quataert S, Tai
tion. Vaccine 32:5650–5656. https://doi.org/ JY, Carlone GM, Mills EL, Ashton FE,
10.1016/j.vaccine.2014.08.010 Diepevaen J, Bielecki J, Bybel M, Cloutier M,
7. Aase A, Herstad TK, Merino S, Brandsdal KT, Poolman JT (1997) Standardization and a
Berdal BP, Aleksandersen EM, Aaberge IS multilaboratory comparison of Neisseria
(2007) Opsonophagocytic activity and other meningitidis serogroup a and C serum bacteri-
serological indications of Bordetella pertussis cidal assays. Clin Diagn Lab Immunol
infection in military recruits in Norway. Clin 4:156–167. https://doi.org/10.1128/cdli.4.
Vaccine Immunol 14:855–862. https://doi. 2.156-167.1997
org/10.1128/CVI.00081-07 15. Yu J, Tostanosk LH, Peter L, Mercad NB,
8. Guttormsen H, Liu Y, Paoletti L (2008) Func- McMahan K, Mahrokhia SH, Nkolol JP,
tional activity of antisera to group B strepto- Liu J, Li Z, Chandrashekar A, Martine DR,
coccal conjugate vaccines measured with an Loos C, Atyeo C, Fischinger S, Burk JS, Slei
opsonophagocytosis assay and HL-60 effector MD, Chen Y, Zuiani A, Lelis FJN, Travers M,
cells. Hum Vaccin Vaccin 4:370–374 Habibi S, Pessaint L, Van Ry A, Blade K,
Brown R, Cook A, Finneyfrock B, Dodson A,
9. Herbert J, Thomas S, Brookes C, Turner C, Teow E, Velasco J, Zahn R, Wegmann F,
Turner P, Nosten F, LeDoare K, Hudson M, Bondzi EA, Dagotto G, Gebr MS, He X,
Heath PT, Gorringe A, Taylor S (2015) Jacob-Dolan C, Kirilova M, Kordana N,
Antibody-mediated complement C3b/iC3b Lin Z, Maxfiel LF, Nampanya F,
binding to group B streptococcus in paired Nityanandam R, Ventur JD, Wan H, Cai Y,
mother and baby serum samples in a refugee Chen B, Schmid AG, Weseman DR, Bari RS,
population on the Thailand-Myanmar border. Alter G, Andersen H, Lewi MG, Barou DH
Clin Vaccine Immunol 22:319–326. https:// (2020) DNA vaccine protection against
doi.org/10.1128/CVI.00803-14 SARS-CoV-2 in rhesus macaques. Science
10. Thomas SR, Leung S, Knox K, Wilkinson 369:806–811. https://doi.org/10.1126/sci
TMA, Staples KJ, Lestrate P, Wauters D, ence.abc6284
Gorringe A, Taylor SC (2018) Development 16. Granoff DM, Welsch JA, Ram S (2009) Bind-
of flow cytometric opsonophagocytosis and ing of complement factor H (fH) to Neisseria
antibody-mediated complement deposition meningitidis is specific for human fH and inhi-
assays for non-typeable Haemophilus influen- bits complement activation by rat and rabbit
zae. BMC Microbiol 18:167. https://doi. sera. Infect Immun 77:764–769
org/10.1186/s12866-018-1314-5
17. Gill CJ, Ram S, Welsch JA, De Tora L, Ane-
11. Lesne E, Cavell BE, Freire-Martin I, mona A (2011) Correlation between serum
Persaud R, Alexander F, Taylor S, bactericidal activity against Neisseria meningi-
Matheson M, van Els CACM, Gorringe A tidis serogroups a, C, W-135 and Y measured
(2020) Acellular pertussis vaccines induce using human versus rabbit serum as the com-
anti-pertactin bactericidal antibodies which plement source. Vaccine 30:29–34. https://
drives the emergence of Pertactin-negative doi.org/10.1016/j.vaccine.2011.10.068
strains. Front Microbiol 11:2108. https://doi.
org/10.3389/fmicb.2020.02108 18. Ray TD, Lewis LA, Gulati S, Rice PA, Ram S
(2011) Novel blocking human IgG directed
12. Mehlhop E, Whitby K, Oliphant T, Marri A, against the pentapeptide repeat motifs of Neis-
Engle M, Diamond MS (2005) Complement seria meningitidis lip/H.8 and Laz lipopro-
activation is required for induction of a protec- teins. J Immunol 186:4881–4894. https://
tive antibody response against West Nile virus doi.org/10.4049/jimmunol.1003623
infection. J Virol 79:7466–7477. https://doi.
org/10.1128/JVI.79.12.7466-7477.2005
362 Frances Alexander et al.
19. Giuntini S, Reason DC, Granoff DM (2012) of classical complement activation. Mol Immu-
Combined roles of human IgG subclass, alter- nol 47:2154–2160
native complement pathway activation, and 28. Brookes C, Freire-Martin I, Cavell B,
epitope density in the bactericidal activity of Alexander F, Taylor S, Persaud R, Fry N,
antibodies to meningococcal factor H binding Preston A, Diavatopoulos D, Gorringe A
protein. Infect Immun 80:187–194. https:// (2018) Bordetella pertussis isolates vary in
doi.org/10.1128/IAI.05956-11 their interactions with human complement
20. Beernink PT, Shaughnessy J, Braga EM, Liu Q, components. Emerg Microbes Infect 7:81.
Rice PA, Ram S, Granoff DM (2011) A menin- https://doi.org/10.1038/s41426-018-
gococcal factor H binding protein mutant that 0084-3
eliminates factor H binding enhances protec- 29. Staples KJ, Taylor S, Thomas S, Leung S,
tive antibody responses to vaccination. J Cox K, Pascal TG, Ostridge K, Welch L, Tuck
Immunol 186:3606–3614. https://doi.org/ AC, Clarke SC, Gorringe A, Wilkinson TMA
10.4049/jimmunol.1003470 (2016) Relationships between mucosal antibo-
21. Mutti M, Ramoni K, Nagy G, Nagy E, Szijártó dies, non-Typeable Haemophilus influenzae
V (2018) A new tool for complement research: (NTHi) infection and airway inflammation in
in vitro reconstituted human classical comple- COPD. PLoS One 11:e0167250. https://doi.
ment pathway. Front Immunol 9:2770. org/10.1371/journal.pone.0167250
https://doi.org/10.3389/fimmu.2018. 30. Le Doare K, Faal A, Jaiteh M, Sarfo F, Taylor S,
02770 Warburton F, Humphries H, Birt J, Jarju S,
22. Brookes C, Kuisma E, Alexander F, Allen L, Darboe S, Clarke E, Antonio M, Foster-
Tipton T, Ram S, Gorringe A, Taylor S (2013) Nyarko E, Heath PT, Gorringe A, Kampmann
Development of a large scale human comple- B (2017) Association between functional anti-
ment source for use in bacterial immunoassays. body against Group B Streptococcus and
J Immunol Methods 391:39–49. https://doi. maternal and infant colonization in a Gambian
org/10.1016/j.jim.2013.02.007 cohort. Vaccine 35:2970. https://doi.org/10.
23. Berends ETM, Kuipers A, Ravesloot MM, 1016/J.VACCINE.2017.04.013
Urbanus RT, Rooijakkers SHM (2014) Bacte- 31. Kolb WP, Kolb LM, Podack ER (1979) C1q:
ria under stress by complement and coagula- isolation from human serum in high yield by
tion. FEMS Microbiol Rev 38:1146–1171. affinity chromatography and development of a
https://doi.org/10.1111/1574-6976.12080 highly sensitive hemolytic assay. J Immunol
24. Amara U, Rittirsch D, Flierl M, Bruckner U, 122:2103–21011
Klos A, Gebhard F, Lambris JD, Huber-Lang 32. Björck L, Kronvall G (1984) Purification and
M (2008) Interaction between the coagulation some properties of streptococcal protein G, a
and complement system. Adv Exp Med Biol novel IgG-binding reagent. J Immunol
632:71–79 133:969–974
25. Mollnes TE, Brekke O-L, Fung M, Fure H, 33. Tao MH, Canfield SM, Morrison SL (1991)
Christiansen D, Bergseth G, Videm V, Lappe- The differential ability of human IgG1 and
gård KT, Köhl J, Lambris JD (2002) Essential IgG4 to activate complement is determined
role of the C5a receptor in E coli-induced oxi- by the COOH-terminal sequence of the CH2
dative burst and phagocytosis revealed by a domain. J Exp Med 173:1025–1028. https://
novel lepirudin-based human whole blood doi.org/10.1084/jem.173.4.1025
model of inflammation. Blood 34. Wang G, de Jong RN, van den Bremer ETJ,
100:1869–1877 Beurskens FJ, Labrijn AF, Ugurlar D, Gros P,
26. Vidarsson G, van der Pol W-L, van den Elsen Schuurman J, Parren PWHI, Heck AJR (2016)
JMH, Vilé H, Jansen M, Duijs J, Morton HC, Molecular basis of assembly and activation of
Boel E, Daha MR, Corthésy B, van de Winkel complement component C1 in complex with
JGJ (2001) Activity of human IgG and IgA immunoglobulin G1 and antigen. Mol Cell
subclasses in immune defense against Neisseria 63:135–145. https://doi.org/10.1016/j.
meningitidis serogroup B. J Immunol molcel.2016.05.016
166:6250–6256. https://doi.org/10.4049/ 35. Feinstein A, Richardson N, Taussig MI (1986)
jimmunol.166.10.6250 Immunoglobulin flexibility in complement
27. Kojouharova M, Reid K, Gadjeva M (2010) activation. Immunol Today 7:169–174
New insights into the molecular mechanisms
Chapter 19
Abstract
There is no vaccine available to prevent Neisseria gonorrhoeae infection, however there is currently a high
level of interest in developing gonococcal vaccines due to the increasing number of cases and continuing
emergence of antimicrobial resistance worldwide. A key aspect of vaccine development is the investigation
of the functional immune response raised to the vaccine targets under investigation. Here, we describe two
assays used to assess the functional immune response raised against gonococcal vaccine targets: the serum
bactericidal assay (SBA) and the opsonophagocytic assay (OPA).
1 Introduction
363
364 Evgeny A. Semchenko et al.
Fig. 1 Schematic overview of the serum bactericidal assay (SBA) and the opsonophagocytic assay (OPA). For
both SBA and OPA, serial dilutions of antibody are prepared, then Neisseria gonorrhoeae (Ng; ~103 colony
forming units (CFU)) are added and incubated at 37 C/5% CO2 for 15 min. Then complement is added (and
polymorphonuclear leukocyte (PMNs) are added for OPA only) and incubated for 1 h. Samples (5 μL neat, and
dilutions) are then plated and incubated for 16 h. Finally, CFU counts and the SBA or OPA titer are determined
2 Materials
3 Methods
3.1 Preparation 1. Prepare and autoclave the following: PBS, 1.7% NaCl (w/v),
of Assay Media 1 M CaCl2, 1 M MgCl2, milli-Q H2O.
2. Prepare 10% (w/v) saponin in GCB and sterilize by passing
through a 0.22 μm filter.
3. Supplement RPMI with 0.15 mM CaCl2, 0.5 mM MgCl2 and
0.5% (w/v) HSA. Sterilize media through a 0.22 μm filter and
store in the fridge (see Note 1).
4. Bring media to room temperature (RT) before using in the
assays.
3.3 Heat Inactivation 1. If using immune serum (mouse, rabbit, human etc.) as a source
of Immune Sera of antibodies for the assay, inactivate heat labile complement
proteins by incubating serum on a block heater (set to 56 C)
for 1 h, then briefly centrifuge (see Note 5). This step is not
necessary if using purified antibodies.
2. Test bactericidal activity of heat-inactivated serum against
N. gonorrhoeae using the same conditions as in the assay (i.e.,
heat-inactivated serum alone should not kill N. gonorrhoeae
after 1 h at 37 C).
3.4 Preparation 1. Collect blood from healthy volunteers (e.g., using Vacuette® Z
of Serum or equivalent Serum Clot Activator Tubes with Gel Separator)
for the Complement and clot as per the manufacturer’s instructions (e.g., invert
Source several times and incubate at room temperature for 10 min
before centrifuging at 2000 g for 10 min, room tempera-
ture). Transfer serum into tubes on ice before combining,
aliquoting and storing (see Note 6).
Gonococcal Vaccine Evaluation 367
3.5 Preparation 1. Collect blood from healthy volunteers (e.g., using Vacuette®
of Polymorphonuclear- K3EDTA blood collection tubes or equivalent). Invert tubes
Cells several times to ensure adequate mixing with the anticoagulant.
2. Dispense 6 mL of PolymorphPrep™ or equivalent into a
15 mL tube and carefully overlay with an equal volume of
freshly collected blood (see Note 10).
3. Centrifuge the tube in a swinging-bucket rotor centrifuge at
500 g for 30 min (RT, no brakes). After centrifugation
carefully transfer the tube to the work bench.
4. Using a serological pipette, carefully collect the PMN buffy
coat and transfer it into a 50 mL tube (see Note 11).
5. Gently add HBSS to the PMN buffy coat, bringing the final
volume to 50 mL. Invert the tube several times to mix the
contents.
6. Centrifuge the tube at 200 g for 10 min.
7. Carefully remove the supernatant and resuspend the PMN
pellet in 10 mL of sterile milli-Q H2O.
368 Evgeny A. Semchenko et al.
3.6 SBA Assay 1. In a round-bottom 96-well plate prepare two-fold serial dilu-
tions of antibody (triplicate wells) (see Note 13).
2. To all appropriate test wells, add bacterial inoculum
(~103 CFU). Place the lid on the plate and incubate at
37 C/CO2 for 15 min.
3. While incubating the assay plate, plate out the bacterial inocu-
lum to determine the bacterial CFU (see Note 14). In a sepa-
rate 96-well plate, prepare serial dilutions of the bacterial
inoculum (e.g., add 5 μL of inoculum to 45 μL of GCB in
the first set of triplicate wells). Mix the contents by repeated
pipetting and transfer 5 μL into subsequent wells containing
45 μL of GCB (1/10 dilution). After performing several ten-
fold dilutions, plate 5 μL spots from all wells onto a GC agar
plates. Allow the spots on the agar plate to dry and then
incubate in 37 C/CO2 incubator overnight.
4. Remove the assay plate from the incubator and add required
volume of serum as a complement source (see Note 9). With-
out delay, make up the final volume to 100 μL with assay media
and return the plate to the 37 C/CO2 incubator. Incubate the
plate for 1 h.
5. While the assay plate is incubating, prepare a 96-well dilution
plate by adding 45 μL of GC broth into appropriate wells for
serial dilutions and plating of bacteria. Also, prepare two GC
agar plates for each set of triplicate wells (i.e., neat/undiluted
and 1/10 dilution).
6. Remove the assay plate from the incubator and mix the con-
tents of wells by gently, repeated pipetting. Transfer 5 μL from
all the wells in the assay plate into prepared 96-well dilution
plate. Plate out 5 μL spots on prepared GC agar plates. Allow
the spots on the agar plates to dry and then incubate in 37 C/
CO2 incubator overnight.
7. Next day, count CFU and determine the SBA titer, defined as
the lowest concentration/dilution of antibody that caused
50% killing relative to no treatment control.
Gonococcal Vaccine Evaluation 369
4 Notes
15. The OPA assay plate set up is identical to SBA, however this
assay may require additional control wells (i.e., PMN and
serum only, PMN only). The presence of complete comple-
ment in the OPA assay means that some killing may also be due
to bacterial lysis via SBA. As such it is recommended that SBA
and OPA assays are performed simultaneously if direct compar-
ison is required.
References
1. Gottlieb SL, Ndowa F, Hook EW 3rd, Deal C, blocking antibodies. Infect Immun 85:
Bachmann L, Abu-Raddad L et al (2020) Gon- e00898–e00816. https://doi.org/10.1128/
ococcal vaccines: public health value and pre- IAI.00898-16
ferred product characteristics; report of a 10. Semchenko EA, Day CJ, Seib KL (2020) The
WHO global stakeholder consultation, January Neisseria gonorrhoeae vaccine candidate NHBA
2019. Vaccine 38:4362–4373. https://doi. elicits antibodies that are bactericidal, opsono-
org/10.1016/j.vaccine.2020.02.073 phagocytic and that reduce gonococcal adher-
2. WHO (2012) Global incidence and prevalence ence to epithelial cells. Vaccines (Basel) 8:219.
of selected curable sexually transmitted infec- https://doi.org/10.3390/vaccines8020219
tions - 2008. World Health Organization, 11. Jen FE, Semchenko EA, Day CJ, Seib KL, Jen-
Geneva nings MP (2019) The Neisseria gonorrhoeae
3. Braxton J, Davis D, Emerson B, Flagg E, methionine sulfoxide reductase (MsrA/B) is a
Grey J, La Zetta et al (2018) Sexually transmit- surface exposed, immunogenic, vaccine candi-
ted disease surveillance 2017. Centers for Dis- date. Front Immunol 10:137. https://doi.
ease Control and Prevention, Atlanta. https:// org/10.3389/fimmu.2019.00137
www.cdc.gov/std/stats17/2017-STD-Surveil 12. Gulati S, Beurskens FJ, de Kreuk BJ, Roza M,
lance-Report_CDC-clearance-9.10.18.pdf. Zheng B, DeOliveira RB et al (2019) Comple-
Accessed 18 Jan 2021 ment alone drives efficacy of a chimeric antigo-
4. Kirby Institute (2018) HIV, viral hepatitis and nococcal monoclonal antibody. PLoS Biol 17:
sexually transmissible infections in Australia: e3000323. https://doi.org/10.1371/journal.
annual surveillance report 2018. The Kirby pbio.3000323
Institute, UNSW Sydney 13. McQuillen DP, Gulati S, Rice PA (1994)
5. Unemo M (2020) Optimizations to keep Complement-mediated bacterial killing assays.
gonorrhoea treatable and reduce antimicrobial Methods Enzymol 236:137–147. https://doi.
resistance selection. Nat Rev Urol org/10.1016/0076-6879(94)36013-8
17:609–610. https://doi.org/10.1038/ 14. Findlow J, Balmer P, Borrow R (2019) A
s41585-020-00378-5 review of complement sources used in serum
6. Edwards JL, Jennings MP, Apicella MA, Seib bactericidal assays for evaluating immune
KL (2016) Is gonococcal disease preventable? responses to meningococcal ACWY conjugate
The importance of understanding immunity vaccines. Hum Vaccin Immunother
and pathogenesis in vaccine development. Crit 15:2491–2500. https://doi.org/10.1080/
Rev Microbiol 42:928–941. https://doi.org/ 21645515.2019.1593082
10.3109/1040841X.2015.1105782 15. Borrow R, Aaberge IS, Santos GF, Eudey TL,
7. Rice PA, Shafer WM, Ram S, Jerse AE (2017) Oster P, Glennie A et al (2005) Interlaboratory
Neisseria gonorrhoeae: drug resistance, mouse standardization of the measurement of serum
models, and vaccine development. Annu Rev bactericidal activity by using human comple-
Microbiol 8(71):665–686. https://doi.org/ ment against meningococcal serogroup b,
10.1146/annurev-micro-090816-093530 strain 44/76-SL, before and after vaccination
8. Plotkin SA (2010) Correlates of protection with the Norwegian MenBvac outer mem-
induced by vaccination. Clin Vaccine Immunol brane vesicle vaccine. Clin Diagn Lab Immunol
17:1055–1065. https://doi.org/10.1128/ 12:970–976. https://doi.org/10.1128/
CVI.00131-10 CDLI.12.8.970-976.2005
9. Semchenko EA, Day CJ, Seib KL (2017) MetQ 16. Lucidarme J, Louth J, Townsend-Payne K,
of Neisseria gonorrhoeae is a surface-expressed Borrow R (2019) Meningococcal
antigen that elicits bactericidal and functional serogroup A, B, C, W, X, and Y serum
372 Evgeny A. Semchenko et al.
bactericidal antibody assays. Methods Mol Biol 18. Jódar L, Butler J, Carlone G, Dagan R,
1969:169–179. https://doi.org/10.1007/ Goldblatt D, K€ayhty H et al (2003) Serological
978-1-4939-9202-7_12 criteria for evaluation and licensure of new
17. Dwyer M, Gadjeva M (2014) Opsonophago- pneumococcal conjugate vaccine formulations
cytic assay. Methods Mol Biol 1100:373–379. for use in infants. Vaccine 21:3265–3272.
https://doi.org/10.1007/978-1-62703-724- https://doi.org/10.1016/s0264-410x(03)
2_32 00230-5
Chapter 20
Abstract
The opsonophagocytic killing assay (OPKA) is designed to measure the functionality of strain-specific
antibodies and, therefore, assess protective immunity or the immunogenicity of Group A Streptococcus
(GAS) (type A Streptococcus pyogenes) vaccines. Opsonization of GAS for phagocytosis is an important
mechanism by which antibodies protect against disease in vivo. The Opsonophagocytic Index or Opsonic
Index (OI) is the estimated dilution of antisera that kills 50% of the target bacteria. Here, we describe the
protocol of the standardized GAS OPKA developed by Jones et al., 2018.
Key words Antibody, Phagocytosis, Opsonophagocytic killing assay, S. pyogenes, HL-60 cells, Baby
rabbit complement, In vitro, Functional assay
1 Introduction
373
374 Helen Wagstaffe et al.
2 Materials
3 Methods
3.1 HL-60 Master HL-60 cells are promyelocytic leukemia cells which are differen-
Stock Propagation tiated to a neutrophil-like cell with 0.8% dimethylformamide
from Master Cell (DMF) for use in the assay. All cell culture is to be undertaken in
Bank Stock a culture hood under sterile conditions. Warm culture medium to
37 C before use.
1. Add 10 ml culture medium to a 15 ml centrifuge tube. Thaw
the master stock of HL-60 cells rapidly by swirling in a 37 C
water bath. Transfer the cells into the culture medium.
2. Centrifuge the tube at 350 g for 5 min at room temperature
(RT). Remove the supernatant.
3. Resuspend the cells in culture medium to a final concentration
of 2 105 per ml and transfer to a culture flask. Incubate at
37 C, 5% CO2.
4. When the cell density reaches 5 105 cells per ml, add further
fresh culture medium to readjust the concentration to 2 105
per ml (see Note 2). Use multiple flasks when the volume
reaches the capacity of the flask. To avoid risk of contamination,
the medium must not reach the cap when the flask is
horizontal.
5. When the concentration reaches 5 105 cells per ml in
10 flasks, freeze the cells. Transfer the contents of the flasks
into 50 ml centrifuge tubes and spin at 350 g for 5 min.
6. Extract and discard the supernatant, being careful not to dis-
turb the pellet.
7. Add 2.5 ml freezing medium to each 50 ml centrifuge tube and
resuspend each pellet.
8. Combine the contents of all tubes in a single culture flask and
aliquot 1 ml into cryovials (each vial should contain
1 107 cells).
376 Helen Wagstaffe et al.
3.4 Bacterial Master All bacterial culture to be undertaken in a bacterial culture hood
Stock Preparation under sterile conditions.
and Maintenance Frozen vials of GAS strains are stored at 80 C. To maximize
the integrity of the bacterial master stocks, the vials should remain
Group A Streptococcus OPK Assay 377
frozen at all times by keeping on dry ice. Vials can be stored for
several years.
3.4.1 From Clinical Swabs can be stored at 4 C for up to 4 months depending on the
Swabs strain or for longer time if stored at 80 C.
1. Streak the swab along the edge of a horse blood agar plate.
2. Rotate the plate by 90 , streak using a sterile loop and repeat
once more using the same loop (optional, use a new loop, pass
through the final streak and streak on clean zones toward the
center of the plate to ensure isolated colonies).
3. Incubate overnight at 37 C, 5% CO2 until colonies appear.
4. To create a master stock, harvest enough colonies on a loop and
mix well in 1 ml of storage buffer before storing at 80 C.
3.4.2 From Plated Plated bacteria can be stored at 4 C for up to 4 weeks, depending
Bacteria on the storage condition. To create a master stock, harvest enough
colonies on a loop and mix well in 1 ml of storage buffer before
storing at 80 C.
3.6 Human Serum Human serum must be handled in accordance with local guidelines
Sample Preparation and stored at 80 C. Prior to testing, the serum must be thawed at
RT and heat-inactivated by incubating at 56 C for a minimum of
30 min. Allow the samples to cool to RT before use in an assay.
Samples can be stored at 4 C for up to 1 month during the testing
process to avoid repeated freeze–thaw cycles (if sample requires
retesting).
3.7 Preparing Caution must be exercised when handling BRC as its components
Working Aliquots are extremely heat-sensitive. When receiving BRC stock from the
of Baby Rabbit supplier, ensure that the contents are entirely frozen and transfer to
Complement (BRC) 80 C storage immediately. There is the potential for high degrees
of variation in performance between BRC lots; therefore, prospec-
tive lots need screening before use in an assay.
1. Defrost each bottle of BRC in cold water with constant agita-
tion, for example, inside an ice box containing a mixture of ice
and cold water, placed on top of an orbital shaker.
2. Label tubes with the lot number and aliquot date. Place tubes
on ice to cool. Place the bottle of BRC on ice as soon as it has
thawed.
3. Quickly aliquot the BRC placing the aliquots in ice until fin-
ished (this can be done in a culture hood to ensure sterility).
4. Store all aliquots at 80 C until required.
3.8 Preparation This procedure describes the process for one 200 ml flask; volumes
of Differentiated HL-60 can be altered accordingly. The concentration of cells required for
Cells (for Use use in the assay is 1 107 per ml. Do not use cells >1.5 106 per
in the Assay) ml at initial count. See Note 4 for more detail on HL-60 cell
acceptance criteria.
1. Resuspend differentiated cells by shaking gently to ensure
equal distribution throughout the flask. Count cells and
check viability by trypan blue exclusion. Cell viability must be
80%.
2. Decant cells into four 50 ml centrifuge tubes under sterile
conditions. Spin tubes for 5 min at 350 g at RT.
3. Remove the supernatant and resuspend each 50 ml of cells in
50 ml HBSS (without Ca/Mg). Centrifuge for 5 min at
350 g at RT.
4. Remove the supernatant and resuspend each 50 ml tube in
50 ml HBSS (+Ca/Mg).
Group A Streptococcus OPK Assay 379
3.9 Preparation 1. Remove one vial of bacteria from the 80 C freezer and
of Bacteria (for Use defrost.
in the Assay) 2. Centrifuge the tube for 2 min at 13,000 g at RT.
3. Remove the supernatant using a pipette, being careful not to
dislodge the pellet of cells.
4. Add 1 ml of OPS buffer to each tube and vortex. Centrifuge
the tube at 13,000 g for 2 min at RT.
5. Carefully remove the supernatant and resuspend the pellet in
0.5 ml OPS buffer.
Fig. 1 Optimal dilution 1 experiment procedure. Bacteria is serially diluted in a plate 1 then transferred to plate
2 when BRC and HL-60 cells are added. After incubation, the mixture is spotted onto THY agar and incubated
overnight
3.10.2 Optimal Dilution 1. Make up each dilution of bacteria working stock determined in
Experiment 2 OD1 required for the OD2 experiment.
2. Assay a known positive quality control (QC) serum, Control A
(heat inactivated BRC, no serum) and Control B (active BRC,
no serum) as detailed in Subheading 3.11 (each dilution of
bacteria will be added to four columns of one plate) (Fig. 2).
3. Select the dilution that yields ~100 CFU in Control B and the
OI of the known positive QC serum within the predetermined
accepted range.
3.11 OPKA Procedure The following procedure details the quantities required for four
96-well plates. Unknown test sera can be used neat or prediluted in
OPS buffer, for example, at 1:4 or 1:30 dilution.
1. Prepare 400 ml overlay agar. Store in a 50 C water bath until
required. Ensure a minimum of 16 THY agar plates are avail-
able for use.
2. Remove the BRC from the 80 C freezer and defrost on ice.
3. Label each round-bottomed 96-well microtiter plate with plate
ID (A, B, C or D) and sample layout. The procedure described
382 Helen Wagstaffe et al.
Fig. 2 Optimal dilution 2 experiment plate layout. Each test dilution of bacteria is assayed with a dilution series
of known positive serum, Control A and Control B
3.12 Data Analysis 1. The OI is the estimated dilution of serum that kills 50% of the
target bacteria.
384 Helen Wagstaffe et al.
3.12.1 Limits The following applies when samples are run neat.
of Detection
1. The upper limit of detection (ULD) is 8748. This is calculated
by taking the fold dilution of serum [3] to the power of
7 (number of fold dilutions), and multiplying by 4 (total vol-
ume in well (80 μl) divided by volume of serum in well (20 μl)).
2. However, Opsotiter automatically multiplies all 8748 results by
2 for statistical purposes. The “OI final” on the results sheet is
therefore displayed as 17,496.
3. Samples that have an opsonic index higher than the upper limit
of detection may be repeated at a predilution of 1 in 30.
4. The lower limit of detection (LLD) is 4, as this is the initial
dilution of serum once all components have been added to the
plate. However Opsotiter automatically divides all results of
4 by 2 for statistical purposes. The “OI final” on the results
sheet is therefore displayed as 2.
5. The lower limit of quantification (LLOQ) was determined for
seven different clinically relevant GAS strains (see Note 7).
Samples below LLOQ can be reported as half LLOQ for
statistical purposes.
3.12.2 Tentative Plate 1. The level of NSK is determined as the killing induced by BRC
Acceptance Criteria and HL-60 cells alone, in the absence of serum. This is deter-
mined as the difference between Control A and Control B: 1—
(average Control B CFU/average Control A CFU) 100.
This should be kept at 35%; see Note 5.
2. The CFU obtained in Control B should be kept between
50 and 150 to ensure accurate and reproducible counting on
the automated colony counter.
3. A QC sample should be included on all plates run, the OI result
should remain between a predetermined acceptability range.
3.12.3 Tentative Sample 1. Sample replicates must cross the 50% killing limit within one
Acceptance Criteria 3-fold dilution. If they do not cross the 50% killing limit, then
the sample will be retested.
Group A Streptococcus OPK Assay 385
4 Notes
References
1. Barth DD, Moloi A, Mayosi BM, Engel ME Organization Research and Development tech-
(2020) Prevalence of group a streptococcal nology roadmap and preferred product charac-
infection in Africa to inform GAS vaccines for teristics. Clin Infect Dis 69(5):877–883
rheumatic heart disease: a systematic review and 4. Lancefield RC (1957) Differentiation of group a
meta-analysis. Int J Cardiol 307:200–208 streptococci with a common R antigen into
2. Sims Sanyahumbi A, Colquhoun S, Wyber R, three serological types, with special reference to
Carapetis JR (2016) Global disease burden of the bactericidal test. J Exp Med 106
group a streptococcus. In: Streptococcus pyogenes: (4):525–544
basic biology to clinical manifestations. Univer- 5. Jones S, Moreland NJ, Zancolli M, Raynes J,
sity of Oklahoma Health Sciences Center, Okla- Loh JMS, Smeesters PR et al (2018) Develop-
homa City (OK) ment of an opsonophagocytic killing assay for
3. Vekemans J, Gouvea-Reis F, Kim JH, Excler JL, group a streptococcus. Vaccine 36
Smeesters PR, O’Brien KL et al (2019) The path (26):3756–3763
to group a streptococcus vaccines: World Health
Chapter 21
Abstract
Neisseria lactamica is a nonpathogenic commensal of the human upper respiratory tract that has been
associated with protection against N. meningitidis colonization and disease. We have previously utilized the
N. lactamica controlled human infection model to investigate the protective effect of N. lactamica
colonization on N. meningitidis colonization, the nature of cross-reactive immune responses mounted
toward N. meningitidis following N. lactamica colonization, and the microevolution of N. lactamica over a
5-month colonization period. More recently, we have assessed the possibility of utilizing genetically
modified strains of N. lactamica to enable use of the commensal as a vehicle for prolonged exposure of
the nasopharynx of humans to antigens of interest, expressed in carried organisms. A controlled infection
with N. lactamica expressing the meningococcal antigen NadA has been executed and the results demon-
strate that this strategy is effective at generating immune responses to the target antigen. Throughout this
chapter, we outline in a step-by-step manner the methodologies utilized when performing controlled
human infection with N. lactamica including procedures relating to: (1) the dilution of N. lactamica
stock vials to derive intranasal inocula, (2) the delivery of intranasal inocula to human volunteers, (3) the
determination of N. lactamica colonization status following intranasal inoculation using oropharyngeal
swabbing and nasal wash sampling, (4) the microbiological procedures utilized to identify N. lactamica
colonization among study volunteers, and (5) the identification of N. lactamica colonies as strain
Y92-1009 using polymerase chain reaction.
Key words Neisseria lactamica, Neisseria meningitidis, Controlled human infection model, CHIME,
Human challenge
1 Introduction
387
388 Adam P. Dale et al.
2 Materials
2.3 Taking 1. Sterile throat swab for microbiological culture with an appro-
and Processing priate transport medium (e.g., TS/5-17 Probact Amies Clear
an Oropharyngeal medium in a tube with a polystyrene viscose tip swab).
Throat Swab 2. GC-X-gal plate.
to Determine Volunteer 3. Dedicated room within a clinical environment, for example, a
N. lactamica CRF, complete with an examination couch.
Colonization Status
4. Class II MSC housed within an Advisory Committee on Dan-
gerous Pathogens (ACDP) containment level 2 (CL2)
laboratory.
5. Appropriate laboratory waste container.
6. Incubator set at 37 C, 5% CO2.
2.4 Taking 1. Sterile saline (0.9%) for irrigation or injection (20 ml) at room
and Processing temperature.
a Nasal Wash Sample 2. 2 10 ml sterile syringes.
to Determine Volunteer
3. Sterile plastic Petri dish.
N. lactamica
Colonization Status
4. 2 GC-X-gal plates.
5. Sterile PBS.
6. Sterile 50 ml centrifuge tube.
7. Set of calibrated Gilson-style pipettes and sterile tips.
8. Sterile microbiology plate spreaders.
9. Disposable gloves and aprons.
10. Tissues.
11. Transport box with ice.
12. Dedicated room within a clinical environment, for example, a
CRF, complete with an examination couch.
13. Class II MSC housed within an ACDP CL2 laboratory.
14. Centrifuge.
15. Appropriate laboratory waste container.
16. Incubator set at 37 C, 5% CO2.
Table 1
Nucleotide sequences of primers for Y92-1009-specific, multiplex PCR. (Forward—FOR; Reverse—
REV)
Nucleotide sequence
Name (50 to 30 ) Product size (bp)
SeqA_FOR gtgctgaatttatagacgggc 388
SeqA_REV aagctaggtctacttggtttag
SeqB_FOR caattgtttaccgccctgcc 273
SeqB_REV gtttcctgccaatcaatccc
SeqC_FOR agggaccgacatctttcatac 594
SeqC_REV ttgcaggctctttccaaac
lacZ_FOR cgggcaaacttgcgcgg 745
lacZ_REV gcaaaccgaaacggggcagg
3 Methods
3.3 Taking 3. While sitting upright, ask the volunteer to tilt their head back
and Processing and open their mouth wide.
Oropharyngeal Throat 4. When the head of the swab has made contact with the pharyn-
Swabs to Determine geal wall, behind the uvula, stroke it across the pharyngeal wall
Volunteer N. lactamica five times.
Colonization Status 5. Remove the swab from the oral cavity, place it into the Amies
transport medium, seal the swab and transfer it immediately to
the class II MSC for further processing.
6. Working in the class II MSC, apply the swab contents directly
to the GC-X-gal plate by streaking the swab vertically in a
continuous fashion while rotating the plate and swab tip.
Ensure the swab makes contact with all areas of the agar.
7. Dispose of the swab in the appropriate waste container and
incubate the GC-X-gal plate immediately at 37 C, 5% CO2 for
48 h.
8. Following 48 h incubation, assess the GC-X-gal plate for
growth of putative colonies of N. lactamica in line with the
methodology outlined in Subheading 3.5.
N. lactamica Controlled Human Infection Model 397
3.4 Taking 1. Explain the procedure to the volunteer and obtain informed
and Processing Nasal consent.
Wash Samples 2. Ask the patient to don a disposable apron to protect their
to Determine Volunteer clothing.
N. lactamica 3. Position the patient supine with a pillow placed under their
Colonization Density shoulders to enable extension of the neck backward.
4. Don an apron and a pair of gloves.
5. Draw up 10 ml of sterile 0.9% saline into the syringe and expel
any air.
6. Ask the volunteer to hold the Petri dish on their chest to enable
collection of the nasal wash fluid (leave the lid on the Petri dish
at this point).
7. Ask the volunteer to open their mouth, extend their head
backward, and position their tongue to avoid swallowing the
saline. Remind them to breathe through their mouth and not
to swallow.
8. Once in position and the volunteer has signaled they are ready,
place the tip of the syringe in their nostril and gently instill
10 ml of saline. Ask them to hold the saline in their nasophar-
ynx for 1 min.
9. After 1 min, ask the volunteer to open the lid of the Petri dish
and to lean forward as fast as possible to allow the saline to exit
the nose passively and be caught in the Petri dish.
10. Withdraw the nasal wash specimen from the Petri dish using
the syringe and transfer it to the centrifuge tube.
11. Repeat steps 5–10 using the alternate nostril.
12. Transport the nasal wash specimen on ice immediately to the
microbiology laboratory for processing.
13. Measure and record the total volume of nasal wash fluid
retrieved from the volunteer.
14. Centrifuge the nasal wash fluid at 5000 g for 10 min to pellet
the bacteria.
15. Carefully decant the supernatant and resuspend the pellet in a
total volume of 300 μl PBS before vortexing for 30 s to loosen
the mucous.
16. Inoculate 2 GC-X-gal plates with 250 μl and 25 μl volumes of
the resuspended pellet, respectively, and lawn over the plates
using individual microbiology spreaders.
17. Incubate the GC-X-gal plates immediately at 37 C, 5% CO2,
for 48 h.
18. Following 48 h incubation, assess the GC-X-gal plates for
growth of putative colonies of N. lactamica in line with the
methodology outlined in Subheading 3.5.
398 Adam P. Dale et al.
3.5 Identification 1. Working in a class II MSC, visually inspect the GC-X-gal agar
of N. lactamica plates inoculated with throat swab or nasal wash contents for
Colonies from Throat evidence of putative N. lactamica colonies, that is, blue colo-
Swab and Nasal Wash nies (beta-galactosidase positive) with morphology consistent
Cultures with Neisseria spp.
2. Pick a well-defined, accessible blue colony and subculture it by
streaking it onto GC-X-gal agar with a 10 μl bacteriological
loop. Incubate at 37 C, 5% CO2, for 24 h.
3. Assuming a pure subculture is obtained, produce a stock of the
isolate in an aliquot of bacterial storage medium. Macerate
colonies against the side of the cryogenic storage vial to pro-
duce a suspension, then incubate for 10 min and freeze at
80 C.
4. Check the oxidase status of the organism by spreading a small
amount of a single blue colony onto an oxidase strip using a
10 μl bacteriological loop. A positive oxidase test (strip turns
blue/black where contact with colony is made) would be in
keeping with potential N. lactamica and should prompt a
Gram stain to be performed. If the bacteria are oxidase nega-
tive, then the isolate is not N. lactamica and the prepared stock
(step 3, above) can be discarded.
5. To perform a Gram stain, the following steps should be
followed:
(a) Place a drop of distilled water onto a clean glass micro-
scope slide and emulsify a small amount of a single blue
colony into it using a 10 μl bacteriological loop.
(b) Dry and fix the slide by placing on a heat block at 65 C.
(c) Remove the slide from the class II MSC and flood the slide
with 0.5% crystal violet solution and leave for 30 s.
(d) Wash off the crystal violet solution with tap water and
then flood the slide with Gram’s acetone and wash off
with tap water rapidly.
(e) Flood the slide with carbolfuchsin or safranin solution and
leave for 30 s.
(f) Wash off the carbolfuchsin or safranin with tap water and
dry with clean blotting paper.
(g) View under oil immersion at 100 magnification.
6. A Gram stain that reveals Gram-negative diplococci is consis-
tent with N. lactamica and should prompt speciation using a
biochemical test strip, for example, the API® NH (Biomerieux)
(see Note 4).
N. lactamica Controlled Human Infection Model 399
3.6.2 Performance 1. If using boiled bacterial lysates, either fresh or frozen, remove
of Y92-1009-Specific PCR bacterial debris from each suspension by centrifugation of the
lysates at 17,000 g for 10 min. Note that the Y92-1009-
specific PCR will also amplify target sequences from purified
genomic DNA. Solutions of extracted genomic DNA are most
easy to use at a concentration of 50 μg/ml.
2. Don pre-PCR laboratory coat, safety spectacles and gloves.
3. Decontaminate working area by liberal application of 10%
(v/v) bleach solution.
4. Wait for 10 min for the bleach to inactivate environmental
amplicons and other potential DNA contaminants. Change
gloves.
5. Meanwhile: gather together the appropriate number of ali-
quots of the ‘Y92-1009-specific 1 Master Mix’, such that
there is one aliquot of 1 Master Mix for each sample to be
analysed. Gather additional aliquots of 1 Master Mix suffi-
cient to include one positive and one negative control reaction
for every two rows of wells that will be filled during agarose gel
electrophoresis. Note that each PCR control tube will provide
sufficient material to load 2 wells of control material. One well
containing positive control material and one well containing
negative control material must be loaded in each row of wells,
to allow for: (1) visual comparison of sample amplicons to
those generated from wild type genomic DNA extracted from
N. lactamica strain Y92-1009, and (2) to show that no con-
taminating DNA was present in the 1 Master Mix,
respectively.
N. lactamica Controlled Human Infection Model 401
A B C D E F G
800 bp
600 bp
400 bp
200 bp
6. Spray working area with distilled water and then mop up the
resulting moisture with paper toweling. Carefully dispose of
the paper toweling in the waste bin. Change gloves.
7. For each sample, carefully transfer 1 μl of the lysate or purified,
genomic DNA (50 ng) into a single 24 μl aliquot of “Y92-
1009-specific 1 Master Mix.” Appropriately label each PCR
tube with the laboratory marker pen.
8. Into one of the remaining 1 Master Mix aliquots, add 1 μl of
N. lactamica gDNA derived from wild type strain Y92-1009
(50 ng), and into another add 1 μl of DNase/RNase-free water.
Appropriately label each tube with the laboratory marker pen.
9. Retain all 1.5 ml microcentrifuge tubes containing the boiled
lysates until a given lysate has been definitively identified.
Boiled lysates can be refrozen at 20 C.
10. Place all PCR tubes containing complete reaction mixtures
(including controls) into the Thermal Cycler. Set the Thermal
Cycler to run according to the parameters shown in Note 5.
11. Analyse PCR amplicons on an agarose gel and analyze the
banding pattern present in each lane of the gel, with reference
to Fig. 2.
4 Notes
STAGE A: 1 5 min at 98 C
STAGE B: 35 20 s at 98 C (melting)
15 s at 63 C (annealing)
26 s at 72 C (extension)
STAGE C: 1 5 min at 72 C
(Optional): Infinite at 10 C
References
1. Laver JR, Hughes SE, Read RC (2015) Neis- on Neisseria lactamica outer membrane vesi-
serial molecular adaptations to the nasopharyn- cles. Clin Vaccine Immunol 16:1113–1120
geal niche. Adv Microb Physiol 66:323–355 8. Evans CM, Pratt CB, Matheson M et al (2011)
2. Christensen H, May M, Bowen L et al (2010) Nasopharyngeal colonization by Neisseria lac-
Meningococcal carriage by age: a systematic tamica and induction of protective immunity
review and meta-analysis. Lancet Infect Dis against Neisseria meningitidis. Clin Infect Dis
10:853–861 52:70–77
3. Maiden MCJ, Ibarz-Pavón AB, Urwin R et al 9. Deasy AM, Guccione E, Dale AP et al (2015)
(2008) Impact of meningococcal serogroup C Nasal inoculation of the commensal Neisseria
conjugate vaccines on carriage and herd immu- lactamica inhibits carriage of Neisseria menin-
nity. J Infect Dis 197:737–743 gitidis by young adults: a controlled human
4. Gold R, Goldschneider I, Lepow ML et al infection study. Clin Infect Dis 60:1512–1520
(1978) Carriage of Neisseria meningitidis and 10. Pandey AK, Cleary DW, Laver JR et al (2017)
Neisseria lactamica in infants and children. J Neisseria lactamica Y92-1009 complete
Infect Dis 137:112–121 genome sequence. Stand Genomic Sci 12:41
5. Cartwright KA, Stuart JM, Jones DM et al 11. Pandey AK, Cleary DW, Laver JR et al (2018)
(1987) The stonehouse survey: nasopharyn- Microevolution of Neisseria lactamica during
geal carriage of meningococci and Neisseria nasopharyngeal colonisation induced by con-
lactamica. Epidemiol Infect 99:591–601 trolled human infection. Nat Commun 9:4753
6. Coen PG, Cartwright K, Stuart J (2000) Math- 12. Dale AP, Theodosiou AA, Laver JR et al (2020)
ematical modeling of infection and disease due Controlled human infection with Neisseria lac-
to Neisseria meningitidis and Neisseria lacta- tamica induces B-cell responses that are cross-
mica. Int J Epidemiol 29:180–188 reactive with Neisseria meningitidis. J Immu-
7. Gorringe AR, Taylor S, Brookes C et al (2009) nol 204:231.21
Phase I safety and immunogenicity study of a 13. Gbesemete D, Laver JR, de Graaf H et al
candidate meningococcal disease vaccine based (2019) Protocol for a controlled human
404 Adam P. Dale et al.
infection with genetically modified Neisseria 14. Laver JR, Gbesemete D, Dale AP et al (2021) A
lactamica expressing the meningococcal vac- recombinant commensal bacteria elicits heter-
cine antigen NadA: a potent new technique ologous antigen-specific immune responses
for experimental medicine. BMJ Open 9: during pharyngeal carriage. Sci Transl Med
e026544 13:eabe8573
15. APIWEB™, Biomerieux. https://apiweb.bio
merieux.com/login. Accessed 27 Sep 2020
Chapter 22
Abstract
Classical in vivo infection models are oftentimes associated with speculation due to the many physiological
factors that are unseen or not accounted for when analyzing experimental outputs, especially when solely
utilizing the classic approach of tissue-derived colony-forming unit (CFU) enumeration. To better under-
stand the steps and natural progression of bacterial infection, the pathophysiology of individual organs with
which the bacteria interact in their natural course of infection must be considered. In this case, it is not only
important to isolate organs as much as possible from additional physiological processes, but to also consider
the dynamics of the bacteria at the cellular level within these organs of interest. Here, we describe in detail
two models, ex vivo porcine liver and spleen coperfusion and a murine infection model, and the numerous
associated experimental outputs produced by these models that can be taken and used together to
investigate the pathogen–host interactions within tissues in depth.
Key words Ex vivo perfusion, Murine infection model, Immunohistochemistry, Confocal micros-
copy, Fiji, InForm, Image analysis, Correlates of protection
1 Introduction
405
406 Ryan G. Hames et al.
2 Materials
3 Methods
3.1 Bacterial Strains Bacteria are cultured to the mid-logarithmic phase and aliquots are
and Culture Conditions stored at 80 C with a cryopreservative such as glycerol. The
media selected will depend on the requirements of your bacterial
strain. For example, S. pneumoniae grows well in Brain Heart
Infusion (BHI) broth and BAB culture plates with 3% v/v defibri-
nated horse blood.
3.1.1 Broth 1. Prepare the relevant bacterial growth medium (e.g., BHI;
14.8 g in 400 mL distilled water) and autoclave at 121 C 15 psi
(103 kPa) for 20 min.
2. Ensure color and media pH is within the parameters recom-
mended by the manufacturer.
3. Store media at room temperature (RT).
3.1.2 Blood Agar Base 1. Prepare BAB (16 g in 400 mL of distilled water) and autoclave
(BAB) Culture Plates + 3% at 121 C 15 psi (103 kPa) for 20 min.
v/v Horse Blood 2. Once autoclaved, allow to cool to around 56 C, then add 3%
v/v sterile defibrinated horse blood (12 mL blood in 400 mL
molten agar) and mix (see Note 6).
3. Pour the agar mix into Petri dishes. 400 mL of medium should
make roughly twenty 100 mm 15 mm plates (around 20 mL
per plate).
4. Leave to set in a sterile environment; once set the plates can be
inverted. For long-term storage, store inverted at 4 C.
3.1.3 Bacterial Infection 1. From laboratory stocks, streak to single colonies on blood agar
Stocks plates. Record strain identifier.
2. Confirm the strain, in case it carries an antibiotic resistant
marker, by placing an antibiotic disc onto the plate at the site
of the initial streak with flamed forceps.
3. Incubate the plates inverted at the ideal temperature and time
conditions for the strain selected.
4. Inoculate a sweep of colonies into 10 mL broth in a universal
tube with a tight cap.
5. After 3–3.5 h, check the OD600 nm of the culture. The OD600
nm of the culture should be between 0.4 and 0.6; this
Analyzing Macrophage Infection at the Organ Level 411
3.2 Murine All procedures performed herein were done in the UK in accor-
Infection Model dance with the UK Home Office license P7B01C07A, and were
approved by the University of Leicester Ethics Committee. Proce-
dures documented here are for example purposes only; the actual
procedures utilized in experimental projects should be in line with
the relevant legislation and regulations applicable to the country
where experiments are undertaken.
3.2.1 Infection Dose 1. Thaw an aliquot of bacteria at RT and add the corresponding
volume of broth or PBS, based on the viability counts of the
stocks, to reach the desirable infection dose (e.g.,
1 107 CFU/mL of S. pneumoniae is required for a dose of
1 106 CFU per mouse with an 100 μL inoculation volume).
2. Make a serial dilution of the infection dose in a BAB plate, and
incubate the plate at the optimal time and temperature for the
strain used, to retrospectively determine the viable CFU in the
inoculum (see Note 7).
3. The prepared infection dose should be used within 30 min and
kept at RT.
4. Prepare the individual doses of 100 μL in insulin syringes
equipped with a 29G needle immediately prior to infection.
3.2.2 Intravenous Route 1. Acclimate 6–8-week-old, sex-matched mice in the animal facil-
of Infection ity environment for at least 1 week prior to infection. Animals
should be acclimated under the standard lighting and temper-
ature conditions in individually HEPA-filtered cages with ster-
ile bedding and nesting and free access to food and water
provided ad libitum.
2. Ear tag or tattoo the mice for identification.
412 Ryan G. Hames et al.
3.2.3 Monitor Signs 1. Following IV inoculation, monitor the mice every 6–8 h for
of Disease and Euthanasia determination of signs of disease (see Note 11).
2. At predetermined time points after infection or upon reaching
the severity limit outlined in the project license, euthanize the
mice using an approved humane killing method such as cervical
dislocation. Confirm the mouse is dead by cutting the femoral
artery (see Note 12).
3.2.4 Organ Recovery 1. Before organ collection, prepare and label two sterile tubes for
and Bacterial Enumeration each organ to be collected. Add a small volume of PBS or broth
into each tube.
2. Disinfect the work surfaces using 70% v/v ethanol. On a sterile
surgical field, prepare a cleaned dissection board with all sterile
dissection instruments.
3. Pin the mouse carcass to the dissection board with the abdo-
men facing up. Spray the fur of the chest and abdomen with
disinfectant such as 70% v/v ethanol to reduce contamination
by hair.
4. Using sterilized forceps, grasp the skin above the urethral
opening and cut along the ventral midline from the groin to
the sternum using fine point sharp scissors. Peel the skin back
from the peritoneal wall underneath using forceps.
Analyzing Macrophage Infection at the Organ Level 413
3.3 Ex Vivo Porcine To prevent extensive ischemia resulting from delays in perfusion,
Liver and Spleen the circuit should be set up prior to organ retrieval. The circuit
Coperfusion Model described herein consists of a reservoir which supplies blood to the
portal circulation and one which supplies the systemic circulation.
3.3.1 Set up of the Ex A diagrammatic representation of the liver–spleen perfusion circuit
Vivo Perfusion Circuit can be found in Fig. 1.
1. Remove the perfusion circuit from its packaging under aseptic
conditions.
2. Connect the drainage lines which will originate from the infe-
rior vena cava and splenic vein to the systemic reservoir.
3. Connect the systemic reservoir to the centrifugal pump fol-
lowed by the oxygenator.
4. Following the oxygenator, the line is split into three outlets.
These should be connected via a cannula to the hepatic artery,
splenic artery, and the reservoir which supplies blood to the
hepatic portal vein once the organ is retrieved.
414 Ryan G. Hames et al.
Fig. 1 The ex vivo normothermic porcine liver and spleen coperfusion. A schematic of the complete perfusion
circuit. The circuit described here consists of a sterile organ chamber that houses the porcine spleen and liver,
cannulated via the splenic artery, hepatic artery, and portal vein respectively. Autologous porcine blood
supplemented with heparin drains passively into a blood reservoir which is fed by saline infusion bags
supplemented with epoprostenol sodium. Blood subsequently flows through the pump and water bath which
ensures physiological temperature and pressure. Following oxygenation, the blood line splits into three, one
for each vessel: splenic artery, hepatic artery and portal vein (via a portal reservoir). The pressure of each
vessel is relayed to, and can be controlled by, the control console. Once the perfusion is deemed stable, the
circuit can be infected via syringe, and blood samples taken from, the inlet/outlet—commonly a three-way
stopcock. (Figure was generated using BioRender)
3.3.2 Organ Retrieval 1. Allow the local qualified team at the abattoir to euthanize pigs
by exsanguination from the jugular vein following stunning
(UK common practice) and collect around 3 L of autologous
blood in a container supplemented with 25,000 units of
heparin.
2. For spleen retrieval: perform a laparotomy and sternotomy
incision and access the celiac trunk to isolate the main splenic
artery and vein.
3. Cannulate the main splenic artery in situ with a catheter of
appropriate size and secure by tying in place with suture thread.
Carefully ligate and divide other associated vessels.
Analyzing Macrophage Infection at the Organ Level 415
3.3.3 Ex Vivo Perfusion 1. Flush organs with 250 mL saline by gentle squeezing of the
and Bacterial Infection infusion bag to remove excess preservative solution prior to
plugging into the perfusion circuit.
2. Using a measuring cylinder, pour 3 L of autologous pig blood
into the systemic reservoir, and apply a gentle flow rate to the
circuit such that each organ input line is gently releasing blood.
Start the saline infusion lines.
3. Plug the cannulated vessels into the circuit ensuring that no air
bubbles are introduced.
4. Ensure normothermia by setting the heat exchange unit to
37 C, and set the oxygenator to a physiological level of 2 L/min.
5. Once plugged in, set the perfusion pressure to 80 Hg/mm for
the splenic artery and hepatic artery, and 10 Hg/mm for the
portal vein for the length of the experiment. Monitor the flow
rate through each vessel at hourly intervals.
6. Twenty to thirty milliliters sodium bicarbonate can now be
added to the circuit perfusate to minimize the effect of tissue
acidosis, in addition to epoprostenol sodium which should be
added to the saline infusion bag to a final concentration of 2.5
μg/mL to facilitate vasodilation and thus effective oxygenation
of the tissues (see Note 13).
7. Once a stable perfusion flow rate and blood gas parameters
have been achieved, infect the circuit with bacteria via syringe
into the circuit input, commonly a three-way stopcock (see
Note 14).
8. At each predetermined time point, use a scalpel to excise a
1 cm2 tissue biopsy from the organ, which should then be
halved, and take 10 mL of blood via one of the circuit inputs
(see Note 15).
416 Ryan G. Hames et al.
3.4 Sample 1. Place a metal freezing container on dry ice and fill it with
Preparation 2-methylbutane up to 1 cm in depth (see Note 16).
3.4.1 Sample Freezing 2. Fill approximately one third of an embedding mold with OCT.
3. Using forceps, place the sample in the middle of the mold.
Ensure the biopsy is submerged in OCT by adding more if
necessary.
4. Place the mold into the 2-methylbutane, being careful not to
get any liquid onto the sample, and let it freeze.
5. Take the frozen sample out of the 2-methylbutane and store it
at 80 C until use.
3.4.2 Sample Sectioning 1. Switch on and set up your cryostat as per manufacturer’s
instructions, setting the internal temperature to 20 C. Insert
the blade and the glass anti-roll plate (see Note 17).
2. Once the temperature is reached, remove the OCT block from
its mold. Add a small amount of OCT onto the specimen disk.
Once the OCT starts to freeze, mount the sample onto the
specimen disk. Once the OCT fully freezes (~5 min) and the
sample is securely attached to the specimen disk, mount the
specimen disk onto the stage.
3. Set the section thickness (see Note 18).
4. Move the specimen as close as possible to the blade by using the
stage forward button (if applicable).
Analyzing Macrophage Infection at the Organ Level 417
3.4.3 Indirect It is important to keep the antibodies, dyes and section out of light
Immunohistochemistry as much as possible throughout the staining process to reduce the
Staining amount of photobleaching that occurs and thus increasing the
quality and fluorescence intensity of the sample.
1. Select the relevant primary and secondary antibodies to label
individual cell types and bacteria, as well as fluorescent dyes to
label nuclei (commonly DAPI) and cell macromolecules (see
Note 20).
2. Dilute the primary and secondary antibodies and dyes in block-
ing solution, and temporarily store away from light at 4 C (see
Note 21).
3. On the slide, circle around the section with a hydrophobic pen
and let dry. Alternatively, place the slides into a specialized
staining rack.
4. Add the fixation buffer (see Note 22).
5. Wash thoroughly with PBS then permeabilize the sample by
adding 0.1% v/v Triton X-100 for 10 min at RT.
6. Block the sample with blocking solution for 30 min to 1 h.
7. Add the primary antibodies and incubate for 1 h at RT (see
Note 23).
8. Wash thoroughly with PBS.
9. Add the secondary antibodies with any dyes and incubate for
45 min at RT (see Note 24).
10. Wash thoroughly with PBS and add DAPI for 10 min.
11. Wash thoroughly with PBS and one final time with distilled
water.
12. Add a drop of mountant onto the sample followed by a cover-
slip (see Note 25).
13. Wipe away any excess mountant from the periphery of the
coverslip and seal the around the edges of the coverslip with
cover slip adhesive or clear nail polish. Let the adhesive dry.
14. The slide is ready to use or can be stored at either 4 C or
20 C until use (see Note 26).
418 Ryan G. Hames et al.
3.5 Sample 1. Wipe the slides to be scanned with 70% v/v IMS.
Microscopy and Image 2. With a fine permanent marker, draw a complete box on the
Analysis underside of the slides around the sample.
3.5.1 Fluorescence 3. Load the slides into the slide carriers (see Note 27).
Whole-Slide Scanning 4. Load the Vectra Polaris software, and load the carriers into the
slide scanner (see Note 28).
5. Select “Edit Protocol”!“New. . .” to create a new protocol. A
new protocol should be created for each combination of anti-
bodies used on the slides to be scanned. Enter a suitable
protocol name, ensure fluorescence imaging mode is selected,
select “multispectral slide scan” (Opal 4 color) and select
(or “create”) a study in which to save the protocols and result-
ing scan images (see Note 29).
6. Under the slide scan settings, change the pixel resolution to
“0.25 μm (40)” and select “auto update” for the exposure
times when prompted. Select “Select Scan Bands” and check
the desired Opals depending on the fluorophores used in this
particular protocol. The color of signal resulting from each
Opal can also be changed here (see Note 30).
7. Select “Scan Exposures”!“Load Carrier” and select a rack
containing a slide stained with the antibody panel
corresponding to the protocol that is being created. With the
DAPI channel automatically selected, locate the sample by
clicking various locations on the appropriate slide depiction to
the right of the live image, or by clicking on the sides of the live
image itself. Alternatively, use “Take Overview” to scan the
entirety of all slides in the carrier, hence showing exactly
where the samples are located (see Note 31).
8. Move around the sample and use “Autofocus” and “Autofluor-
escence” until there are no more overexposed (red) areas in the
image. Repeat this step for the remaining antibody channels,
then select “Back” (see Note 32).
9. Save the protocol. Repeat steps 5–9 to create a new protocol
for each antibody combination used in the slides to be scanned.
Once all protocols have been created, click “Back” (see
Note 33).
10. Select “Scan Slides.” Select the gray circle next to each slot to
bring up the edit slides window. Change the task for all slides to
be scanned to “Scan Slides,” then enter the appropriate study,
protocol and desired slide ID. Select “Scan.”
11. Once scanning is complete, the scanned images of the sample (.
qptiff file) and slide label photos (.tiff file) will appear in the
study folder which was selected prior to scanning. The scans
can then be opened with Phenochart, or analyzed using Fiji or
inForm.
Analyzing Macrophage Infection at the Organ Level 419
Fig. 2 inForm image analysis window. A screenshot of the inform image analysis window with tools utilized in
this methodology labeled as appropriate. Options on the horizontal toolbar include zoom, draw training regions
(used in the tissue segmentation step), component/composite view (used in the cell segmentation and
phenotyping steps), and edit the view for adjusting channel brightness and contrast. Options on the vertical
toolbar include buttons for the toggling on/off of the tissue segmentation, nuclear segmentation and cytoplasm
segmentation maps along with the cell phenotypes. The image batch containing image stamps is shown at the
bottom of the window
3.5.2 inForm Image Whole slide scan images cannot be directly opened in the inForm
Analysis software, therefore “stamp” sections of the images to be analyzed
must be prepared beforehand in Phenochart. When analyzing the
images in inForm, refer to Fig. 2 for a view of the processing
window with labeled buttons that are used in the methodology
described here.
1. Open the desired image(s) to be analyzed in the Phenochart
software and login using the appropriate institutional
password.
2. Select “Stamp” ! “Select for: inForm Projects”. Select an
appropriate size in fields for the stamp and select areas on the
image to be analyzed in inform (see Note 34).
3. Open the images to be analyzed in inForm through
“File”!“Open”!“Image.” The stamps of each image will be
opened separately in the batch view.
4. Select “Configure. . .” to configure the analysis workflow.
Select “Trainable Tissue Segmentation,” “Adaptive Cell Seg-
mentation,” and “Phenotyping” while skipping the
“Score” step.
420 Ryan G. Hames et al.
3.5.3 Fiji Image Analysis 1. Load the Fiji software and open the image to be analyzed by
selecting “Plugins”!“Bio-Formats”!“Bio-Formats
Importer.”
2. In the “Bio-Formats Import Options,” ensure that the color
mode is set to composite (autoscale checked) and that channels
are split into separate windows.
3. Select the series with highest resolution (Series 1) in the “Bio-
Formats Series Options” screen. Click “OK.”
4. On the nuclear marker (commonly DAPI) channel, use the
“Polygon selections” tool in the toolbar to create a region of
interest (ROI) around the entire sample. Add this ROI to the
ROI manager by pressing Ctrl+T, and rename it as “whole slice
ROI” and save.
5. On a macrophage marker channel, alter the threshold by select-
ing “Image”!“Adjust”!“Threshold.” Ensure that the “Dark
background” box is unchecked, and set the maximum fluores-
cence (bottom slider) to its highest level, and the minimum
fluorescence (upper slider) to an appropriate level as to include
the majority of fluorescent signal while negating background.
Click “Apply” to apply the selected threshold levels (see Note
49).
6. The image should now be binary: black with white signal. If
this is reversed, select “Process”!“Binary”!“Options,” select
“Black background” and reapply the threshold settings.
7. If there should be any artefacts in the image that have been
included in the threshold selection, these can be omitted by
drawing around them with the “Polygon selections” or “Free-
hand selections” tools and pressing the delete key.
8. Omit any signal outside of the sample by selecting the “whole
slice ROI” from the ROI manager, inverting the selection by
selecting “Edit”!“Selection”!“Make Inverse” and pressing
the delete key (see Note 50).
9. Select “Edit”!“Selection”!“Create selection” to create
ROIs around the macrophage marker signal. Ensure that the
binary white signal and not the surroundings are selected and
subsequently add the ROIs to the ROI manager using Ctrl+T.
Rename this ROI as the appropriate macrophage marker ROI
and save.
10. On the bacteria channel, apply a threshold to include all bacte-
ria within the sample, as described in step 5. The image should
have a black background with white signal; alter the Binary
settings as outlined in step 6 to reverse this (see Note 51).
11. On the bacteria channel, select the whole slice ROI. Select
“Analyze”!“Analyze Particles. . .” and check the “Summa-
rize” box. The size can be adjusted depending on the average
Analyzing Macrophage Infection at the Organ Level 423
4 Notes
bottle was too warm when the horse blood was added. If solid
chunks of agar are poured, the agar was too cold when the
horse blood was added.
7. Serial dilutions from neat to 1:100,000 can be achieved by
diluting 20 μL into 180 μL media in a 96-well plate five
times. A multichannel pipette can then be used to plate 5 μL
of all dilutions onto the plate. To calculate CFU/mL, count
the number of colonies in the highest dilution where bacteria
can still be accurately counted, multiply by the dilution factor
(e.g., 1 104 for the fourth dilution of the neat sample) and
divide by 0.005.
8. This causes vasodilation and provides better vein visibility. Do
not overheat the animal by leaving the animal in the chamber
for more than 10 min. This will cause dehydration and make
accessing the vein and administering the dose more difficult.
9. Sedation or anesthesia for intravenous inoculation is unneces-
sary for trained and experienced personnel. Placing food in the
opposite end of the restraint tube can encourage the mouse to
enter the tube themselves.
10. If the needle is correctly inserted, blood should flash into the
syringe when the plunger is gently withdrawn and the inocu-
lum will flow easily during administration. Do not force the
inoculum—this means the dose is being improperly adminis-
tered to tissue surrounding the vein and can cause distress to
the animal. If the proper placement cannot be confirmed,
attempt to place the needle in a more proximal position
where the vein is wider in diameter.
11. The frequency of which the mice should be observed and/or
scored will depend on the guidelines specific to your project.
12. The time points utilized in the study will depend on experi-
mental objectives; however, a frequently used time course for
the study of early stages of pathogenesis is up to 8 h
postinfection.
13. The volume of sodium bicarbonate that should be added is
dependent on the pH yielded by blood gas analysis and as such
the actual volume required can vary. In our perfusion circuit,
we utilize 200 mL infusion bags of saline, which requires the
addition of 0.5 mg epoprostenol sodium.
14. In our experience, a stable flow rate is reached after around
30 min of perfusion. Our perfusion circuits are infected with
around 1 107 CFU of S. pneumoniae or Klebsiella pneumo-
niae, although the infection bacteria and dosage should be
specifically tailored to the experimental aims and objectives.
Ensure blood is taken into the syringe and dispelled multiple
times to ensure all the bacteria enter the circuit.
Analyzing Macrophage Infection at the Organ Level 425
21. The blocking solution used here is 5% v/v goat serum in PBS;
however, other type of sera used for blocking (i.e., bovine
serum albumin) in IHC are also common. The antibodies
and dyes should be diluted to a concentration outlined by the
manufacturer, although 1:200 for primary antibodies and
1:500 for secondary antibodies are common dilutions.
22. 4% v/v formaldehyde is commonly used in immunohistochem-
istry; however, different fixation buffers such as methanol can
be used in certain applications. The base can also vary depend-
ing on application and often yields different staining intensity
and clarity. A common fixation buffer is PBS, as used here.
23. Alternatively, primary antibodies can often be incubated over-
night at 4 C or for 1 h at 37 C. In some cases, manufacturers
provide specific incubation times for antibodies which should
be adhered to.
24. From this step onwards it is extremely important to keep the
sample out of direct light as much as possible as the
fluorochrome-conjugated secondary antibodies are light sensi-
tive and can become photobleached when exposed to bright
light for extended periods.
25. A coverslip with thickness that is compatible with the micro-
scope should be used. When placing the coverslip onto the
slide, lower down one end slowly to prevent the formation of
air bubbles. If any bubbles form underneath the coverslip, a
gentle rolling pressure to one end can expel the bubbles
through the end of the coverslip.
26. Storage at 20 C is recommended for long-term storage, to
preserve the brightness of the fluorescent tags and dyes,
although care should be taken to minimize the number of
freeze–thaw cycles. 4 C is suitable for short-term storage
when planning to use the samples within the same week.
27. Take a photo of the slides in the carriers to refer back to during
the scan exposures step, thus ensuring that the slides identity
and the location of the sample on the slides can easily be found.
28. Document which slide carriers are placed at which position in
the rack. You will refer back to these when imaging protocols
are assigned to individual slides in the scanning step.
29. Oftentimes, similar sample types stained with the same panel of
antibodies will yield similar fluorescence intensities, that is,
mouse spleens stained with the same antibody combination in
parallel will appear similar. In these cases, one protocol can be
used for all slides harboring the same antibody panel. However,
if the resulting scans show over/underexposed signal between
slides, a protocol should be created for, and assigned to, each
individual slide.
Analyzing Macrophage Infection at the Organ Level 427
Acknowledgements
We thank John Isherwood and Rohan Kumar for help with the
perfusion of the porcine organs at explant, the staff of Joseph
Morris Butchers, and Sarah Glenn and the staff of the Leicester
Preclinical Research Facility for support with the mouse experi-
ments. The grant was in part supported by a collaboration agree-
ment with the University of Oxford and grants from the MRC
MR/M003078/1 and BBSRC BB/S507052/1 to MRO. ZJ is
funded by BBSRC BB/S507052/1.
References
humoral immunity against experimental pneu- ex vivo perfused model: results with the por-
mococcal sepsis. Vaccine 29(46):8241–8249. cine liver--kidney circuit over 24 h. Artif
https://doi.org/10.1016/j.vaccine.2011.08. Organs 37(5):457–466. https://doi.org/10.
119 1111/aor.12003
16. Rukke HV, Kalluru RS, Repnik U et al (2014) 21. Daniel CR, Labens R, Argyle D et al (2018)
Protective role of the capsule and impact of Extracorporeal perfusion of isolated organs of
serotype 4 switching on Streptococcus mitis. large animals—bridging the gap between
Infect Immun 82(9):3790–3801. https://doi. in vitro and in vivo studies. ALTEX 35
org/10.1128/IAI.01840-14 (1):77–98. https://doi.org/10.14573/altex.
17. Yue F, Cheng Y, Breschi A et al (2014) A 1611291
comparative encyclopedia of DNA elements in 22. Meurens F, Summerfield A, Nauwynck H et al
the mouse genome. Nature (London) 515 (2012) The pig: a model for human infectious
(7527):355–364. https://doi.org/10.1038/ diseases. Trends Microbiol 20(1):50–57.
nature13992 https://doi.org/10.1016/j.tim.2011.11.002
18. Steiniger BS (2015) Human spleen microanat- 23. Ramos-Vara JA, Miller MA (2013) When tissue
omy: why mice do not suffice. Immunology antigens and antibodies get along: revisiting
145(3):334–346. https://doi.org/10.1111/ the technical aspects of immunohistochemis-
imm.12469 try--the red, brown, and blue technique. Vet
19. Mestas J, Hughes CCW (2004) Of mice and Pathol 51(1):42–87. https://doi.org/10.
not men: differences between mouse and 1177/0300985813505879
human immunology. J Immunol 172 24. Butler AJ, Rees MA, Wight DGD et al (2002)
(5):2731–2738. https://doi.org/10.4049/ Successful extracorporeal porcine liver perfu-
jimmunol.172.5.2731 sion for 72 h. Transplantation 73
20. Chung WY, Gravante G, Al-Leswas D et al (8):1212–1218
(2013) The development of a multiorgan
Chapter 23
Abstract
Vaccines induce a highly complex immune reaction in secondary lymphoid organs to generate immunolog-
ical memory against an antigen or antigens of interest. Measurement of post immunization immune
responses generated by specialized lymphocyte subsets requires time-dependent sampling, usually of the
blood. Several T and B cell subsets are involved in the reaction, including CD4 and CD8 T cells, T follicular
helper cells (Tfh), and germinal center B cells alongside their circulating (c) counterparts; cTfh and
antibody secreting cells. Multicolor flow cytometry of peripheral blood mononuclear cells (PBMC) coupled
with high-dimensional analysis offers an opportunity to study these cells in detail. Here we demonstrate a
method by which such data can be generated and analysed using software that renders multidimensional
data on a two dimensional map to identify rare vaccine-induced T and B cell subsets.
Key words Flow cytometry, High-dimensional data analysis, t-SNE, FlowSOM, T cells, Vaccine,
immunophenotyping, FlowJoTM
1 Introduction
433
434 Megan E. Cole et al.
2 Materials
Table 1
The fluorochrome for each marker has been selected based on the configuration of a Becton
Dickinson (BD) LSRFortessa flow cytometer instrument, following the general principles of multicolor
panel design BV is Brilliant Violet™. BUV dyes are BD Horizon Brilliant™ Ultraviolet. Fluorochrome-
conjugated antibodies were obtained from BD Biosciences and BioLegend
3 Methods
Table 2
Examples of antibody clones that could be used in a multicolor flow cytometry panel. Users should
check with suppliers for the fluorescent antibody clone-conjugates available when designing the
panel
its handling. In this protocol, live cells are fixed with 4% parafor-
maldehyde before acquiring on the flow cytometer.
Sterile working
Standard laboratory aseptic technique should be used through-
out to avoid contamination of live samples. Surfaces, working area,
and equipment are cleaned with 70% ethanol.
Before embarking on the wet-lab procedures, draw up a
96-well plate plan, including control wells and test wells. Use this
as a reference throughout the experiment.
3.2 Preparation This method can be used on PBMC prepared from fresh whole
of PBMC blood or raised from viable cryopreservation.
for Multidimensional
1. Suspend the washed PBMC in FACS wash buffer and count the
Flow Cytometry cells by trypan blue exclusion or using a cell counter and record
Analysis viability. Adjust the concentration to 1 106 cells/100 μl.
2. Prepare the live–dead cell control sample. Program the heat
block to 70 C. Take 0.5 106 PBMC in FACS wash buffer
and kill the cells by heating in a sealed plastic tube at 70 C for
10 min. Combine the killed 0.5 106 PBMCs in FACS wash
buffer with the same quantity of live cells and make up the vol-
ume to 200 μl. Transfer to a 96-well U bottomed plate.
3. Transfer 2 106 cells per well of live PBMC to the labeled
96-well U bottomed plate.
4. Centrifuge at 800 g for 3 min and remove the supernatant.
5. Resuspend the cells in all wells in 100 μl diluted viability dye
buffer, except for the unstained control well, where 100 μl PBS
is used.
6. Incubate for 5 min at RT.
7. Stop the staining by adding 100 μl of FACS wash buffer.
8. Centrifuge at 800 g for 3 min and remove the supernatant.
Multicolor Flow Cytometry and High-Dimensional Data Analysis to Probe. . . 439
3.3 Acquiring Events 1. Open the instrument software (e.g., BD FACSDiva™ Software
on the Flow Cytometer BD Biosciences) and set up the experiment layout on the
instrument.
2. Run the compensation controls and adjust the voltages for each
parameter for clear negative and positive signal separation with
minimal compensation values (see Note 5).
3. Acquire and record at least one million events in the live cell
lymphocyte gate for analysis of rare populations.
3.4 Data Analysis In this section, we describe how to use the data analysis software,
FlowJoTM (BD Life Sciences) for basic data analysis and plug-ins to
visualize high-dimensional data on a two dimensional (2D) map, to
enable the identification of cell clusters that share similar expression
patterns but would be otherwise difficult to extract for analysis
using conventional analysis methods (see Note 6).
3.4.1 Preliminary 1. Check data quality with FlowJo™ by choosing “Inspect” from
Analysis the populations band of the workspace task in the workspace or
by double-clicking on the circle badge to the left of a sample.
2. Cross-check the compensation matrix or, if necessary, recalcu-
late a new compensation matrix with the compensation
controls.
440 Megan E. Cole et al.
3.4.2 Data Down These steps define the number of events for analysis and combine
Sampling them into one file.
and Concatenation
1. Import all exported CD4+ T cell FSC files into one new
workspace.
2. Use the down sample plugin to define the number of events for
analysis, this step will ensure the analysis is not biased due to an
uneven number of events from a few samples or time points.
3. Concatenate the down-sampled populations into one new FCS
file for further analysis, and gate the individual samples based
on their sample ID with the concatenated file.
3.4.3 Running the t-SNE 1. Run the t-SNE plugin on FlowJo™ with the concatenated
data sample to generate x- and y- axis parameters, which project
similar populations from the high-dimensional space into a 2D
plot. Select markers that are important and meaningful to the
research question, to define the x- and y- axis. The running
time for the algorithm depends on the computer RAM capac-
ity, the number of events in the sample, and the number of
markers selected for analysis. The plugin for t-SNE is fully
integrated, and no R or extra package is needed.
2. The 2D plot created shows all events from the concatenated
samples, or one can select the individual sample ID gate to
show the individual sample on the plot. Choose the heatmap
mode to show the expression level of each parameter.
3. To project the conventional populations onto the t-SNE plot,
select the population with t-SNE x-axis and y-axis and copy the
plot onto the experiment layout, and overlay other populations
for comparison.
4. An example of simple data analysis using this method is shown
below (Figs. 1, 2, and 3).
3.4.4 FlowSOM [9] 1. FlowSOM is an algorithm to visualize the cell subsets with Self-
Organizing Maps (SOMs) and Minimal Spanning Trees, which
can reveal how all markers are behaving on all cells and can
detect subsets that might otherwise be missed. The FlowSOM
Plugin needs R packages to run under the command of Flow-
JoTM software (see Note 7 and 8).
2. Run FlowSOM by clicking on the FlowSOM tab and select the
parameters for clusters generation. The outcome of the algo-
rithm run will include distinct populations that are used as
Multicolor Flow Cytometry and High-Dimensional Data Analysis to Probe. . . 441
SSC-W
FSC-W
SSC-A
0 0 0
0 50K 100K 150K 200K 250K 0 50K 100K 150K 200K 250K 0 50K 100K 150K 200K 250K
FSC-A FSC-A SSC-A
CD4
103 103 103
0 0 0
0 50K 100K 150K 200K 250K 0 103 104 –103 0 103 104 105
FSC-A CD3 CD8
Fig. 1 Preliminary gating strategy for targeting populations. For each sample, check the sample quality and
select the populations of interest using known markers. The gating strategy shown here is an example to
enrich CD4+ T cells for further analysis
4 Notes
1. These buffers are optimal for cell viability and must be prepared
fresh on the day. For storage up to 1 month at 2–8 C, 0.25 g
sodium azide (antibacterial agent) can be added.
2. Panel design: Successful flow cytometry experiments require
robust panel design. This process is time consuming but,
done correctly, reaps reward. The first stage is to decide how
many target antigens will be in the panel. This will be depen-
dent on the flow cytometer available so always check this first
before designing the panel. The exact configuration and capa-
bility of the flow cytometer must be understood. Determine
the possible fluorochromes in the panel using a matrix and
include the excitation and emission spectra of these fluoro-
chromes. A spectra analyzer or spectrum viewer can help with
viewing the excitation and emission parameters. Spectral over-
lap can result in data that are difficult to interpret and should be
442 Megan E. Cole et al.
Sample A
Heatmap key
120 120 120
tSNE y
CCR7
tSNE y
tSNE y
90 90 90
expression
range 60 60 60
30 30 30
0 0 0
0 30 60 90 120 0 30 60 90 120 0 30 60 90 120
tSNE x tSNE x tSNE x
120 20 120
TSNE y
90 90
tSNE y
tSNE y
90
60 60 60
30 30 30
0 0 0
0 30 60 90 120 0 30 60 90 120 0 30 60 90 120
tSNE x tSNE x tSNE x
Time point 1 Time point 2 Time point 3
Fig. 2 t-SNE plots of individual samples to show the difference in cluster abundancy. The selected plots show
a heatmap of CCR7 expression on the concatenated CD4+ T cell FCS file. A cluster with relatively low
expression of CCR7 (red arrow) is more abundant in sample A, compared with sample B at three different time
points
Key ID
105
Name
CD45RA-CCR7- 104
CCR7
CD27
CD45RA-CCR7+
103
CD45RA+CCR7-
0
CD45RA+CCR7+ PD-1
-103
120
CXCR3
tSNE y
90
60
CD38
30
0
0 30 60 90 120
tSNE x CD28
Fig. 3 Overlay of known cell subsets onto the t-SNE plot to reveal the complexity of the populations. In this
example, the CD45RACCR7+ population (orange) and the CD45RACCR7 population (green) distribute into
different cluster areas on the t-SNE plot. The abundancy of each cell subset within individual samples is
shown on the top of the histogram overlay
Pop12 700
Pop1
600
Pop0
Pop6 500
Pop3
Pop7 400
Pop4
Pop9 300
Pop8
200
Pop10
Pop13
Pop5
Pop2
Pop11
CD38
CXCR3
PD-1
ICOS
CCR7
CD45RA
CD32
CXCR5
CD27
CD127
CD28
B
ID
120
CD27
90
tSNE y
PD-1
60
ICOS
30
0
CD127
0 30 60 90 120
tSNE x CXCR3
Name CD45RA
Pop 10
CD38
Pop 9
Pop 8 CD32
Pop 4
Pop 3 CXCR5
Pop 2
Pop 1 CCR7
Pop 0
CD28
Fig. 4 Heatmap of metapopulations; diverse populations identified with the FlowSOM algorithm (A) and how
those populations present on the t-SNE plot (B)
Fig. 5 The Minimum Spanning Tree generated by FlowSOM analysis. Each node is represented as a star chart,
where the color-coded height of each segment shows the expression level of that marker. Every chart has a
background colour indicating its meta-population assignment. On the tree, the distance of nodes clustersed
together with each other within a branch shows their similarity to each other. The abundance of each node is
represented by the size of its symbol
CD27
CD127
103 103 103
0 0 0
–103 –103 –103
–103 0 103 104 105 –103 0 103 104 105
–103 0 103 104 105
CD45RA CD38
CD28
Name
105 105 Pop10
Pop9
104 104
CXCR3
ICOS
Pop8
103 103
Pop4
0 0 Pop3
–103 Pop2
–103
–103 0 103 104 105 Pop1
–103 0 103 104 105
PD-1 CXCR5 Pop0
Fig. 6 Identification of FlowSOM populations with selected markers. Like the use of the heatmap, overlay of
FlowSOM populations with selected markers can help to reveal the nature of the populations and possible
gating strategy for future experiments. For example, population2 (orange) is high in CD27, CD38, PD-1, and
ICOS expression
Acknowledgements
The FluAGE study was conducted with a grant from the National
Institute for Health Research (NIHR) Imperial Biomedical
Research Centre (BRC). The views expressed in this publication
are those of the author(s) and not necessarily those of the NHS, the
National Institute for Health Research or the Department of
Health. The authors would like to thank the participants in the
FluAGE study and the staff of the Imperial College Clinical Trials
Centre, and the Department of Infectious Disease, Imperial Col-
lege London." If a conflicts of interest statement is required then
please write "K.P. reports grants from the National Institute for
Health Research and the Medical Research Council UK Research
and Innovation. K.P. is chief investigator for the Imperial College
London COVID-19 vaccine development programme and princi-
pal investigator for the University of Oxford COVID-19 vaccine
development trials. K.P. reports personal fees from Sanofi, and
honoraria received from the British Society for Antimicrobial Ther-
apy and ITV plc.
References
A BLAST+ .....................................................................78, 92
Bordetella pertussis (B. pertussis) ......................... 4, 48, 49,
Adjuvants ........................................... 112, 141, 155, 163, 51–54, 56, 57, 59, 60, 325–330, 332–337, 343,
167, 172, 207, 209, 219, 302, 303, 309 354, 355
Affinity purification ............................................ 39, 41, 43
Bronchoalveolar ............................................................ 307
Alginate................................................................. 142–148 Brucella ..................................................... 3, 4, 12, 13, 18,
Amino acids ................................... 25, 27, 33, 60, 86, 90, 76, 83, 84, 94
138, 155–158, 178, 231, 232, 249–252, 275, 282
Antibodies ............................... 37–39, 41–45, 60, 63–72, C
75–95, 99, 107, 111, 117, 118, 123, 132, 133,
139, 140, 155, 162–166, 175, 181, 186, 194, C1q ............................................................. 343, 345, 346,
200, 201, 204, 212, 214, 218, 219, 221, 222, 349–354, 357–360
225, 230, 239, 240, 266–268, 274, 284, 285, Candidates .............................................. 1–3, 6, 8–11, 13,
288, 291, 292, 295, 296, 304, 305, 307, 309, 14, 17–19, 21, 25, 27, 31–33, 56, 118, 166
310, 312, 314–320, 325, 331, 332, 334, Capsule ................................................237, 282, 284, 285
336–338, 341–343, 351–353, 355, 359, 360, Carriage ................................................................ 387–389
363–371, 373–386, 388, 405, 407, 409, 417, Cell-mediated ................................................................ 303
418, 421, 425–427, 433–435, 437–439, 442, Cell wall extracts ..........................................38, 40, 42–45
443, 445 CHARMM ................................................. 153, 159, 160,
Antigenicity ................................... 6, 19, 25–27, 32, 101, 162, 165, 166
103, 105, 106, 109, 110, 118, 152, 158, 159, Chromatography ............................... 39, 40, 42, 47, 127,
161, 162, 165 128, 134, 135, 137, 177, 230, 232–234, 237,
Antigen presenting cells (APCs) ......................... 302, 435 238, 267, 282, 283, 286, 290, 293, 296, 298,
Antigens...................................................... 2–4, 6, 12, 13, 342, 344, 345, 347–352, 356–359
18–20, 26, 31–33, 37–45, 47–60, 63, 64, 75, 77, Circulating Tfh (cTfh) .................................................. 433
85, 86, 93, 94, 111, 115–142, 151–167, 171, Class I ..............................................18–20, 26, 27, 29–33
172, 175, 178, 181, 186, 207, 219, 224, 227, Class II ........................................... 19, 20, 26, 27, 29–31,
228, 239, 266, 267, 302–305, 309, 310, 33, 54, 197, 309, 345, 365, 390, 391, 393, 394,
312–318, 320, 331, 364, 389, 425, 441 396, 398, 434
Clostridium spp. ................................................. 76, 83, 94
B Clusters ................................................434, 439–443, 445
Clusters of orthologous genes (COGs)............ 22, 83, 92
Baby rabbit complement (BRC) ........................ 374, 375, Colorimetric ...................................... 39, 40, 42, 75, 247,
378–380, 383–385
249, 255, 272, 273
Bacterial extracellular vesicles Complement.......................................... vi, 341–360, 364,
(BEVs).....................................172–175, 177, 178, 366–371, 373, 374
180–186, 188
ComplexHeatmap .....................................................78, 93
Bacterin..............................................................................vi Conjugation ............................................... 180, 281–299,
Bacteroides thetaiotaomicron 302, 303
(B. thetaiotaomicron)................................ 171–173 Conjugative ................................ 173, 174, 179–180, 197
B cell ........................................14, 18–20, 25, 26, 28, 31,
Controlled human infection model experiment
33, 155, 156, 166, 228, 389, 433 (CHIME).................................................. 388, 394
Bicinchoninic acid (BCA) protein Co-perfusion ................................................413–416, 423
assay............................. 39, 64, 66, 224, 229, 231,
Coupling........................................................... 39–44, 283
246–249, 294 Crossflow ultrafiltration ................................................ 171
Bioinformatics ............................ 1, 18, 19, 51, 53–55, 77
449
BACTERIAL VACCINES: METHODS AND PROTOCOLS
450 Index
Cultures ................................................40, 49, 51, 52, 56, G
57, 107–111, 125, 126, 128, 131, 137, 162, 163,
174, 176–177, 179–181, 194–199, 202, 203, Generalized modules for membrane antigens
208, 210, 211, 215, 217–219, 223, 224, 281, (GMMA)................................................... 227–278
289, 295, 304, 305, 307–310, 312, 314–316, Genes ................................................. 3, 9, 11, 14, 18, 56,
318–320, 327–330, 332, 335, 374–376, 378, 64, 75–77, 83, 88, 92, 94, 122–125, 130, 131,
385, 391, 398, 399, 407, 410–411 134, 135, 138, 152, 154, 158, 162, 172–174,
Cytokines ................................... 212, 213, 217–220, 225, 178–179, 194, 196, 197, 202, 208, 209, 212,
303, 305, 307, 309–313, 315, 316, 318–320, 218, 225, 389
325–328, 330–332, 334, 335 Gene synthesis ............................................................... 178
Glycine .......................................39, 40, 43, 65, 175, 193,
D 208–210, 215, 224, 288, 355, 360
Glycoconjugates ......................................... 282–285, 294,
De novo .......................................................................... 178 295, 298, 387, 389
Differential analysis ......................................................... 92 Glycoprotein...................................................................... 8
Dische ......................................................... 229, 234–236, Glycosylation ..................... 282, 283, 292, 294, 296, 297
255, 257, 258 Gram-negative .......................................... 6, 9, 11, 17, 48,
Dynamic light scattering (DLS) ..................................143, 63–72, 87, 99, 112, 115–140, 172, 191, 227,
144, 146, 147, 204, 229, 230, 240, 241 335, 398, 399
Gram-positive ....................................................4, 6, 9, 11,
E
37–45, 48, 87, 95, 373
Electrons .......................................... vi, 97–112, 143, 146, GROMACS ................................................................... 159
147, 192, 204, 209, 225 Group A Streptococcus (GAS) ............................. 373–376,
Endotoxin...........................................137, 207–225, 276, 380, 384, 386
282, 283, 287, 292, 293, 295, 297
Enzyme-linked immunosorbent assays H
(ELISA)..............................................99–101, 105, Hestrin ........................................229, 238, 239, 272, 273
107–109, 111, 117, 118, 122, 123, 131, 132, High-performance liquid chromatography
139, 140, 155, 163, 212–214, 217–222, 304, (HPLC)..................................... 50, 229–232, 238,
305, 307, 312, 316, 317, 321, 326, 327, 242, 250, 251, 260, 262, 270, 274, 344, 345,
331–334, 337, 338 347, 355, 360
Enzyme-linked immunosorbent spot assay High-performance liquid chromatography mass
(ELISpot)................................................. 304, 305, spectrometry (HPLC-MS)...................... 234–236,
312–315, 319, 320 257, 259, 260, 262–265
Epitopes ................................. 2, 8–11, 14, 18–21, 25–33, HL-60 cells.......................................................... 375, 376,
45, 48, 55, 76, 115, 152–163, 165–167, 302, 378–380, 383–385
312, 320 HLA ..........................................19, 20, 26, 27, 29, 30, 33
Escherichia coli (E. coli) ....................................... 108, 125, Human.....................................2, 5, 9–12, 17, 19, 24, 25,
126, 130, 135–138, 154, 162, 167, 173, 174, 27, 32, 33, 38, 39, 41, 43–45, 54, 63, 75–83,
178–180, 194, 195, 197, 202, 208–210, 215, 85–87, 89, 90, 92–95, 99, 117, 151–153, 166,
223, 281–283, 289, 294 327, 334, 341–360, 364–366, 373–403, 406
Extracellular....................................... 12, 13, 94, 95, 116, Human challenge ......................................................75–95
171–189, 191, 192 Humoral .............................. 18, 303, 304, 316, 388, 405
Ex vivo ....................................... 309–313, 405, 406, 408, Hybrids ................................................................. 115–140
409, 413–416, 425
I
F
IgG.............................................41, 44, 90, 91, 123, 133,
Flow cytometry .......................................... 48–50, 52, 53, 155, 165, 194, 200, 214, 221, 222, 225, 230,
57–59, 303, 305, 309–313, 319, 385, 433–446 341–360
FlowJo® ........................................................ 58, 313, 436, IgM ......................................................221, 225, 341–360
439, 440, 446 Immunization................................................. vi, 117, 129,
FlowSOM ...................................................................... 440 134, 152, 155, 158, 162–165, 167, 171, 173,
Fluorescence whole-slide scanning ..................... 418–419 187, 189, 214, 220, 221, 302, 303, 307, 311,
Fusion tag ............................................................. 135, 178 325, 326, 433, 434
BACTERIAL VACCINES: METHODS AND PROTOCOLS
Index 451
Immunodetection ....................................... 175, 178, 181 M
Immunogenicity ................................................18, 86, 91,
153–155, 162–166, 171, 172, 191, 374, 434 Machine learning (ML) .................................... 1–15, 420,
Immunohistochemistry 427, 429, 434
(IHC) ..............................406, 407, 417, 425, 426 Matrix-assisted laser desorption/ionization time-of-flight
Immunoinformatics ..................................................17, 19 mass spectrometry (MALDI-TOF MS) ........... 236
Immunophenotyping.................................................... 433 Mass spectrometry (MS)............................ 22, 47, 50, 53,
Immunoprecipitation...........................................vi, 37–45 58, 60, 63, 64, 67, 69, 70, 72, 229, 235, 242, 249,
Inactivation................................................... 98, 103, 106, 259, 260, 266, 276, 277
107, 109, 110, 366 Maternal........................................................... vi, 325–339
Infants .............................................................. vi, 325–339 Membranes .............................. 12–14, 18–20, 25, 27, 48,
Infection models ................................166, 387–403, 406, 49, 55–60, 65, 69, 70, 72, 86, 92, 95, 99, 100,
408, 411–413 115–140, 155, 164, 172–187, 191–193, 199,
Inhalational vaccination ................................................ 303 200, 207–225, 227, 239, 240, 274, 286–288,
Innate......................................................6, 325, 326, 334, 291, 292, 296, 304, 305, 307, 312, 314, 315,
335, 370, 405, 406 319, 320, 353, 360, 388, 389
Inoculation ........................................ 108, 136, 174, 219, Memory ...................................... 303–305, 309, 315, 316
225, 365, 377, 385, 388–391, 393–396, 402, Meso scale discovery (MSD) ............ 327, 330, 331, 334,
403, 411, 412, 424 336–338
Insoluble proteins ............................................... 117, 118, MHC ......................................................... 18, 31, 32, 309
128, 130, 198, 199 Microarrays ........................................... 75–79, 83, 85, 86,
Intestinal pig epithelial cell 89, 90, 92–95
(IPEC) ............................................. 64, 66, 67, 71 Microscopy ................................146, 147, 192, 204, 209,
Intravenous.......................................................... 309, 406, 407, 409, 413, 416, 418
411, 412, 424 Monoclonal ................................................ 175, 305, 310,
Intravenous immunoglobulin G 312, 318, 343, 360
(IVIG) ..................................................... 38, 43, 44 Multi-epitope fusion antigen (MEFA) .......... vi, 151–167
Ionizing ........................................................................... 98 Multivalent ........................................................... 151–167
Isoelectric focusing (IEF) ........................... 65, 67, 70, 71 Murine .......................................302, 303, 312, 318, 320,
Isolation ..............................................134, 148, 171–187, 406, 408, 411–413, 425
224, 253, 303, 305, 307–309
N
In vitro ....................................................18, 64, 155, 158,
165, 166, 209, 211–213, 217, 218, 220, Nanoparticles......................................141, 143, 146–148,
326–328, 343 178, 185, 204, 230, 242
In vitro transcription and translations (IVTT) .............. 75 Neisseria gonorrhoeae (N. gonorrhoeae) .............. 129, 363,
In vivo ................................................ 6, 18, 64, 118, 135, 364, 366, 367, 370
152, 165, 209, 214, 219, 221, 222, 405 Neisseria lactamica (N. lactamica) .................... 388–396,
398–403
L Neisseria meningitidis
Lavage ................................................................... 303, 307 (N. meningitidis).......................... 4, 18, 117, 123,
Lawsonia intracellularis 124, 281, 354, 355, 364, 387–389, 405
(L. intracellularis) .....................64, 66, 67, 70, 71 NERVE.............................................................................. 2
Leukocytes .................................................. 303–310, 312, Neutralizing.................................... 63–72, 152, 154, 162
314–317, 320 Neutrophils........................................................... 365, 373
Limulus amebocyte lysate (LAL) ....................... 137, 211, Nuclear magnetic resonance
216, 223, 259, 295 (NMR) ..............................................143–145, 238
Lipoproteins ...........................27, 88, 116, 228, 301–321
O
Liquid chromatography-tandem mass spectrometry
(LC-MS/MS) ............................ 47–50, 53, 59, 60 OAg extraction....................................237, 267, 269, 278
Liver ............................................................ 406, 413–416, Opsonophagocytic assay (OPA) ......................... 364, 365,
423, 425 369–371
Low-energy electron irradiation Opsonophagocytic killing assay
(LEEI).....................................................vi, 97–112 (OPKA)...........................373, 374, 380, 382, 383
Lymphocytes ........................................................ 434, 439 Orthologous.................................................................... 23
BACTERIAL VACCINES: METHODS AND PROTOCOLS
452 Index
P PSORTb ...................................31, 51, 55, 78, 86, 92, 94
Pulmonary ..................................302, 303, 307, 309, 320
Pam2Cys ............................................................... 302, 311 Purifications..........................................31, 37, 41, 43, 44,
Pam3Cys ........................................................................ 302 118–121, 127–130, 135–138, 177, 178,
Pangenome ...................................................................... 76 184–185, 189, 212, 218, 224, 283, 286, 287,
Panproteomes.................................. 76–79, 82–90, 92–95 290, 295, 355
Pathogen-reactive antibody pool
(PRAP).................................................... 38, 41, 42 Q
Peptides ............................................1, 27, 28, 37, 48–50,
53–60, 88, 95, 118, 132, 135, 141–148, 171, Quantitative real-time PCR........................ 212, 217, 218
172, 178, 235, 259, 282, 302, 303, 305,
R
309–312, 314–316, 318
Peptidoglycans................................................48, 207, 208 Radiation ................................................................ 98, 102
Periplasmic............................................... 13, 95, 138, 172 Radioimmunoprecipitation assay (RIPA) ................64, 66
PglB .....................................................281, 283, 285, 294 Responses ................................................. 14, 18, 86, 141,
Phagocytosis ......................................................... 302, 383 152–155, 158, 163, 166, 301–321, 325–339,
Plasma .......................................... 38, 326, 328, 332–334, 342, 389, 405, 433, 434
339, 341–360, 370 Reverse vaccinology (RV) ................................. v, vi, 1–14,
Polyclonal ............................................................... 99, 111 17–19, 27, 32
Polyelectrolyte ................................................. vi, 141–148 Rodentibacter pneumotropicus
Polymerase chain reaction (PCR) ...............................124, (R. pneumotropicus) ...........................99, 101, 107
125, 193–197, 202, 212, 217–219, 389,
392–393, 399–401 S
Polymorphonuclear leukocyte...................................... 364
SARS-CoV-2 ........................................................... 3, 6, 8,
Polysaccharides .......................................43, 97, 142, 172,
301, 406
228, 253–255, 267, 269, 270, 272, 294, 388, 389
Scores ...........................................6, 8, 14, 20, 27, 28, 80,
Porcine.................. 5, 117, 406, 408, 409, 413–416, 430
86–89, 92, 94, 155, 156, 158, 159, 165, 419
Predictions ..................................................... 1–3, 6, 8–13,
Secretion ............................................... 57, 172, 174–175,
18–21, 25–29, 31–33, 48, 49, 57, 60, 86, 87, 89,
180–182, 221, 312
92, 94, 134, 156, 166
Self-adjuvanting.................................................... 142, 302
Pregnancies...................................................325–339, 402
Sequons ................................................................ 281–283
Profiling .....................................................................76, 77
Serum bactericidal activity
Propidium iodide (PI) ............................................ 49, 50,
(SBA)..............341–344, 354, 355, 364, 367–371
52, 53, 56, 57, 59
Shaving .................................................................vi, 48–60
Protegenicity ..................................................................... 6
Shewanella vesiculosa (S. vesiculosa) ..................... 191–204
Proteinase K assay ................................................ 178, 186
Shigella flexneri (S. flexneri) .....................................17–34
Protein G ............................................................. 342–344,
Shuttle vector ...............................................174, 178–180
348, 349, 351, 359, 360
SignalP ...............................................................51, 55, 78,
Protein glycan coupling technology
86, 87, 94, 130, 178
(PGCT) .............................................................. 281
Size exclusion chromatography
Proteins............................................. 1–3, 6, 8–14, 18–27,
(SEC) ............................................... 177, 184, 189
31–33, 37, 39–45, 47, 48, 51, 53–57, 60, 63, 64,
Size exclusion high-performance liquid chromatography
66–72, 75–83, 85, 86, 88–90, 92–95, 97, 98, 111,
with multiangle laser light scattering (HPLC-
115–140, 152–167, 171, 172, 174–175, 177,
SEC/MALS) ................................... 241, 242, 244
178, 180–182, 184–186, 191–204, 208–212,
Sodium dodecyl sulfate-polyacrylamide gel
215, 218, 223, 224, 227–229, 231, 239,
electrophoresis (SDS-PAGE)...................... 65, 67,
241–243, 245, 247, 249, 251, 257, 259, 266,
68, 70, 71, 127–130, 136, 155, 162–164, 192,
267, 275, 277, 281–285, 290–292, 294–298,
193, 199–201, 229–231, 242, 245, 246, 274,
302, 303, 305, 307, 309, 310, 312, 314–316,
284, 285, 287, 288, 290, 291, 293, 345, 349,
318, 320, 342–346, 350, 351, 353, 354, 358,
351, 353, 359
359, 366, 389, 434, 444
Soluble protein .............................................127–129, 199
Proteomics.................................................... vi, 18, 47–61,
Spectroscopy.................................................................. 144
75, 152, 202
Spleens ............................................... 309, 406, 408, 409,
Protoplasts .................................................................40, 43
413–416, 423, 425, 426
BACTERIAL VACCINES: METHODS AND PROTOCOLS
Index 453
Staining ............................................ 68, 71, 72, 245, 246, Transports.............................................47, 100, 102, 115,
285, 288–293, 297, 303, 305, 310, 312, 314, 117, 135, 191, 192, 201, 309, 310, 391, 394, 396
315, 317–319, 409, 417, 420, 421, 425, 426, Treponema pallidum (T. pallidum)...........................5, 76,
428, 434, 438, 439, 442, 443, 446 83, 86, 90
Staphylococcus aureus (S. aureus) ...................4, 43, 44, 63 Trimethyl chitosan (TMC) .................................. 142–148
Streptococcus pneumoniae Triparental mating ........................................................ 179
(S. pneumoniae) ..........................4, 18, 43, 76, 77, Tuberculosis (TB) ............................................... 4, 11, 18,
83, 85, 86, 90, 91, 94, 282, 406, 410, 411, 424 152, 302–304
Streptococcus pyogenes (S. pyogenes)..............................4, 48 2-Dimensional gel electrophoresis (2-DE) .............63, 64
Structural vaccinology .................................................. 152
Subunit ..............................vi, 20, 64, 141–148, 171, 302 V
Support vector machine.................................................... 2
Vaccines.................................... v, vi, 1–3, 6, 8–10, 12–14,
Surfaceome ......................................................... 47, 56, 59 17–32, 34, 37–45, 47–60, 64, 75, 76, 78–83,
Swabs ...........................................76, 377, 385, 389, 391, 85–87, 89–95, 97–112, 116, 118, 141–148,
394, 396, 398, 399
151–167, 171–189, 191, 207, 209, 227, 228,
249, 281–298, 301–321, 326, 334, 363–371,
T
373, 374, 387, 388, 405, 433, 434
T7....................................................... 117, 118, 125, 126, Vaxign ...............................................2, 3, 6, 8–14, 18–22,
131, 135, 137, 138 29, 31, 32, 34
T cells ....................................... 14, 18–20, 25–27, 29–31, Vaxign-ML......................................................2, 3, 6, 8–13
33, 155, 166, 302–305, 309–313, 315, 316, Vaxitop .............................................................. 2, 8–11, 32
318–320, 325, 440, 441 Vesicles ........................................... vi, 171–189, 191–204,
t-distributed stochastic neighbor embedding 207–225, 227, 388
(t-SNE) ..........................................................93, 94 Viability.................................................48, 49, 52, 53, 56,
Testing .......................................101, 103, 106, 107, 109, 58, 107, 109, 111, 223, 312, 319, 320, 370, 376,
111, 224, 225, 302, 304, 310, 336–338, 345, 378, 379, 385, 402, 403, 411, 435, 437, 438,
346, 353, 354, 378, 403, 405 441–443
Titration............................. 142, 143, 155, 163, 165, 442 Virulence.............................................6, 54, 56, 152–155,
TMHMM ....................................... 51, 55, 78, 86, 92, 94 158–161, 163, 165, 166
TonB-dependent ..................................... 13, 14, 115, 116
Transconjugants .......................................... 174, 180, 202 W
Transferrin ...................64, 115–118, 122, 132, 138, 139 Western blotting (WB) .......... 63, 64, 69, 175, 181, 186,
Translational fusions ..................................................... 178 198–201, 204, 312, 351, 353
Transporter .....................................................14, 115, 116
Whole blood stimulation .............................................. 327