Bacterial Protein Secretion Systems - Methods and Protocols (PDFDrive)
Bacterial Protein Secretion Systems - Methods and Protocols (PDFDrive)
Laure Journet
Eric Cascales Editors
Bacterial Protein
Secretion
Systems
Methods and Protocols
Methods in Molecular Biology
Series editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Editors
In their ecological niches, bacteria are in contact with other prokaryotic and eukaryotic
cells. Bacteria therefore evolved mechanisms to communicate and collaborate with these
cells. They also developed belligerent behaviours to eliminate competitors and to infect
eukaryotic host cells. These aggressive actions are mediated by effector toxins with specific
activities which will ultimately cause target cell lysis or re-routing of metabolic or trafficking
pathways in the host.
The proper delivery of bacterial effectors into the milieu or directly into target cells is
assured by dedicated structures called secretion systems. Up to now, nine secretion systems
have been described in bacteria, and additional systems allow the exposition of toxins or
adhesins at the cell surface or at the extremity of a pilus structure. These multi-protein
trans-envelope complexes differ in composition, mechanism of assembly, and mode of
recruitment and transport of toxins. However, studying these macromolecular complexes
requires common techniques, ranging from the bioinformatic identification of machine
components and effectors to methods of defining interactions amongst the different sub-
units and the development of reporters to follow effector translocation in vivo. Finally,
state-of-the-art techniques have made significant progress in the analysis of these large
complexes directly in the bacterial cell or from purified samples.
The purpose of this book on bacterial secretion systems is to provide protocols that cover
the broad arsenal of techniques used to study a secretion system from A to Z: identifying and
localising the different subunits, defining interactions within subunits, monitoring conforma-
tional changes, purifying and imaging large complexes, defining the assembly pathway by
fluorescence microscopy and the role of energy during assembly or secretion, and identifying
secreted effectors as well as using reporters to follow effector transport. Most of these tech-
niques are not restricted to the study of secretion systems but are also of specific interest for
any researcher interested in multi-protein complexes of the bacterial cell envelope.
The book starts with a chapter describing a recently developed software program aimed
at identifying gene clusters encoding secretion systems within bacterial genomes. Then six
chapters describe methods for defining the subcellular localisation of the different subunits
of a multi-protein system: prediction programs, fractionation, cell surface exposition and
isopycnic density gradients to partition inner and outer membranes. Three techniques to
determine the topology of membrane proteins (substituted cysteine accessibility, protease
accessibility and reporter fusions) are then detailed. This is followed by eleven chapters
covering genetic, cytologic and biochemical methods used to study protein–protein and
protein–peptidoglycan interactions. Two chapters are dedicated to methods for unveiling
the role of energy and conformational changes, and then six chapters describe techniques
for purifying and imaging large complexes. Finally, methods to identify effectors as well as
the reporter fusions used to validate effector secretion and translocation are presented.
v
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 525
Contributors
xi
xii Contributors
Abstract
Protein secretion systems are complex molecular machineries that translocate proteins through the outer
membrane, and sometimes through multiple other barriers. They have evolved by co-option of compo-
nents from other envelope-associated cellular machineries, making them sometimes difficult to identify and
discriminate. Here, we describe how to identify protein secretion systems in bacterial genomes using
MacSyFinder. This flexible computational tool uses the knowledge stemming from experimental studies to
identify homologous systems in genome data. It can be used with a set of predefined models—“TXSScan”—
to identify all major secretion systems of diderm bacteria (i.e., with inner and with LPS-containing outer
membranes). For this, it identifies and clusters colocalized components of secretion systems using sequence
similarity searches with hidden Markov model protein profiles. Finally, it checks whether the genetic con-
tent and organization of clusters satisfy the constraints of the model. TXSScan models can be customized
to search for variants of known systems. The models can also be built from scratch to identify novel sys-
tems. In this chapter, we describe a complete pipeline of analysis, including the identification of a reference
set of experimentally studied systems, the identification of components and the construction of their pro-
tein profiles, the definition of the models, their optimization, and, finally, their use as tools to search
genomic data.
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_1, © Springer Science+Business Media LLC 2017
1
2 Sophie S. Abby and Eduardo P.C. Rocha
Fig. 1 Models of protein secretion systems available in TXSScan. The T1SS model is displayed in Fig. 2. Each
box represents a component and its status in the system’s model: “mandatory” (plain), “accessory” (dashed),
or “forbidden” (red cross). Within a system panel: the families of homologous proteins are represented in col-
umns and colored identically. When applicable, the system of origin is indicated next to the box(es). The colo-
calization parameter of the system is indicated (d). When this parameter is specific to a gene (e.g., the relaxase
in T4SS), it is indicated by a subscript (e.g., drel). Additional features specific to a component can be found in
Table 2. Curved double-headed arrows indicate exchangeable components. Figures and legends are freely
reproduced with modification from reference [9] [as specified by the Creative Commons Attribution (CC BY)
license version 4.0]
Table 1
Useful online resources to design models for the detection of secretion systems
the data and software required to use and design models for
MacSyFinder. In the last section, we describe how to define models
and protein profiles to identify protein secretion systems.
2 Materials
2.1 Sequence Data MacSyFinder analyzes protein sequences to identify protein secre-
tion systems. The protein sequences should all be stored in a single
file in Fasta format. This file represents one of several types of
information that must be specified by the “--db-type” option:
●● When the protein sequences are from diverse sources (e.g.,
peptides from a metagenome), the file type is “unordered.” In
this case, the program simply outputs the results of the identi-
fication of the systems’ components using the protein
profiles.
6 Sophie S. Abby and Eduardo P.C. Rocha
●● When the proteins are from a single genome but the relative
order of the corresponding genes in the genome is not known,
the file type is “unordered_replicon.” In this case, the program
can identify components and check whether the quorum of
components is respected. It cannot, however, check the genetic
organization of the model.
●● When proteins are from a single genome and are ordered fol-
lowing the position of genes in the genome, then the file type
is “ordered_replicon.” The “ordered_replicon” mode allows
the use of all available criteria (quorum of components and
genetic organization) to identify instances of the system and as
such is the most powerful mode. It should be used whenever
possible to create, test, and validate a new model and the cor-
responding protein profiles. It will constitute the focus of this
protocol.
●● The “gembase” type is similar to the “ordered_replicon” but
has special identifiers that allow the analysis of multiple
“ordered” genomes in a single step (see MacSyFinder
documentation).
2.3 Software Table 1 displays resources of interest for this protocol. To run
MacSyFinder, one needs to install the NCBI/BLAST tools (in par-
ticular, makeblastdb version 2.8 or higher or formatdb), HMMER,
and MacSyFinder [15, 19, 20]. The latter requires a Python inter-
preter (version 2.7) that must be installed beforehand. See
MacSyFinder’s online documentation for more details [15].
To build novel HMM protein profiles, one also needs a pro-
gram to make multiple sequence alignments (e.g., MAFFT or
Muscle [21, 22]), an alignment editor (e.g., Seaview or Jalview
[23, 24]), and a program to cluster proteins by sequence similarity
(e.g., Silix or MCL [25, 26]). The program MacSyView can be
used to visualize the results of MacSyFinder.
In Silico Identification of Secretion Systems 7
3 Methods
T1SS
Decision rules ABC (essential)
MFP (essential) components list
Model design quorum rules
OMF (essential, multiple systems)
d ≤ 5 genes, OMF in trans co-localization rules
3.1 Compilation Gathering the current biological knowledge of the system, espe-
of Available cially of its experimentally studied instances, may not be simple.
Information The genes and proteins of experimentally studied systems are often
on the System named differently; sequence similarity searches may thus be neces-
sary to establish which components in a system are homologous to
those of other systems. Occasionally, gene fusions and fissions fur-
ther complicate the identification of families of homologs. Once
the relationships of homology between known instances of the sys-
tem are well established, its key components can be inventoried
and qualified (as mandatory, accessory, and forbidden). One can
then characterize the system in terms of its genetic organization
and quorum (i.e., how many components are required for a sys-
tem, see Subsection 3.1.4). The set of experimentally validated pro-
tein secretion systems should be split into two independent data
sets (see Note 1). The reference data set is used to characterize the
protein secretion systems, build the models, and construct the pro-
tein profiles (if required; see Notes 2 and 3). The validation data
set is used to validate the final model (see Subsection 3.5). There
are alternative procedures for systems with few experimentally vali-
dated systems (see Note 4).
3.1.2 Extraction of HMM Each component (called a “gene” in the model) must be associ-
Protein Profiles ated with at least one HMM protein profile. Several public data-
from Databanks banks of protein profiles can be queried by keyword (e.g., the name
of the gene) or by sequence for a match to the component (see
Table 1). InterProScan is a good starting point for this task since it
integrates the information on protein profiles from many data-
banks, including PANTHER, PFAM, SUPERFAMILY, and
TIGRFAM [27].
At the end of this procedure, one often has many profiles for
some components and none for others. One can pick the best
matching profile for each component (see Note 1). However,
sometimes there is no profile that is clearly better than the others.
This occurs when profiles match only certain subfamilies of com-
ponents, for example, because they are very divergent in sequence.
In these cases, it is possible to specify multiple profiles (see Note 5).
Table 2
Keywords to specify models
Keyword
(Command-line override) Required? (Default) Level(s) Description
system Yes Upper (first) Keyword needed to start definition of
system model
inter_gene_max_space Yes system, gene “colocalization” parameter. Defines
(--inter-gene-max-space) maximum number of genes between
two consecutive components of
system for a cluster of components to
be created
min_genes_required No (number of mandatory genes or min_ system Minimum number of mandatory and
(--min-genes-required) mandatory_genes_required if declared) accessory components
min_mandatory_genes_required No (number of mandatory genes, or min_ system Minimum number of mandatory
(--min-mandatory-genes-required) mandatory_genes_required if declared) components
multi_loci No (false, i.e., “0”) Whether system can be encoded on
(--multi-loci) several main loci
gene Yes Second Component of system
multi_system No (false, i.e., “0”) gene Whether gene can participate in several
instances of a system
name Yes gene Name of gene to be used in report files,
and base name of corresponding
HMM protein profile
(continued)
In Silico Identification of Secretion Systems
9
Table 2
10
(continued)
Keyword
(Command-line override) Required? (Default) Level(s) Description
loner No (false, i.e., “0”) gene Whether gene can be found encoded
outside of system’s main loci, i.e., an
exception to colocalization rule
defined at system level
exchangeable No gene Whether gene can be replaced by
another gene in list of components
(quorum); the gene can then be
exchanged with one of its
“homologs” or “analogs”
analogs No gene List of “analogs” to gene
Sophie S. Abby and Eduardo P.C. Rocha
3.1.4 Defining The quorum of the model is the minimum number of components
the Quorum required to validate an instance of the system. It is defined by two
of Components parameters: the minimum number of mandatory components
(“min_mandatory_genes_required”) and the minimum number of
mandatory and accessory components (“min_genes_required”).
The values of both parameters are set by default to the number of
mandatory components, and in this case the accessory components
do not actually count in the quorum. To make them count, one
must specify higher values for the second than for the first crite-
rion. It is particularly important to use prior knowledge to assess
the relevance of a locus lacking a mandatory gene.
The quorum can be optimized using the information from the
secretion systems identified in bacterial genomes (see Note 7).
Changes in the values of the quorum affect the sensitivity and spec-
ificity of the method. Low values authorize the validation of sys-
tems with fewer components that are less similar to the reference
data set, thereby increasing the number of detected systems. This
might come at the cost of misidentifications or the validation of
nonfunctional systems. Running the models with higher values of
the quorum results in the identification of instances that are more
12 Sophie S. Abby and Eduardo P.C. Rocha
similar to the reference data set and thus more likely to be true. On
the other hand, this results in the identification of fewer instances
and may exclude those that are too distinct from those in the refer-
ence data set.
3.2 Model After gathering the available information on the protein secretion
Formulation system, one must write down the model in the relatively simple
XML format predefined for MacSyFinder. A list of the keywords of
this hierarchical XML grammar is presented in Table 2 and in
MacSyFinder’s online documentation.
3.2.1 Defining the Model We illustrate the formulation of a simple model with the example
in an XML Text File: of the type I secretion system. This system has three mandatory
Example 1—T1SS components (Fig. 2): an ABC transporter (“abc”), a membrane
fusion protein (“mfp”), and an outer membrane porin (“omf”).
The latter can be colocalized with the two other components (less
than six genes apart, “inter_gene_max_space” set to 5), or encoded
apart in the genome (loner attribute set to True: “1”). In addition,
omf can be involved in several occurrences of the system (“multi-
system” set to True (“1”)). The three mandatory components are
required to form a full system. The quorum is therefore not speci-
fied; it is left at its default value. The file with the model should be
named after the system (“T1SS.xml”). Its content is displayed in
Fig. 2.
3.2.2 Defining the Model Larger systems tend to require more complex models. This is well
in an XML Text File: exemplified by the model to identify T9SS (file “T9SS.xml” [9]).
Example 2—T9SS This model, in line with the existing literature [28–31], states
that T9SS consists of several mandatory and accessory components
encoded in multiple loci (multi_loci = “1”). The quorum allows
one mandatory and several accessory components to be missing.
Some components form clusters, whereas others are defined as lon-
ers (Fig. 1). The SprA component was matched by several profiles
of the PFAM and TIGRFAM databanks; we used the “exchange-
able” and “homologs” attributes to include them all (see Note 5).
3.3 Running HMM protein profiles should be in individual files named after the
MacSyFinder corresponding component and placed in the same directory. The
path to this directory must be provided with the option “-p”. All
3.3.1 Organizing
profiles must have the same file name extension (“.hmm” by
the Data
default, though it can be changed with the option “--profile-
suffix”). For the T1SS model mentioned earlier, the program
requires three HMM files in a “profiles” directory: “abc.hmm,”
“mfp.hmm,” and “omf.hmm”.
The path to the directory within the current working directory
containing the XML model file must also be given in the input
(option “-d,” named “definitions,” for example).
In Silico Identification of Secretion Systems 13
3.4 Output Files, The results of the detection are printed to files in simple text for-
and Visualization mat that can be read by any word-processing application. They are
of Results stored in the MacSyFinder’s output directory (specified using the
with MacSyView “-o” option, or named automatically). They include configuration
and log files, along with the results of MacSyFinder and
3.4.1 Finding Relevant HMMER. The description of the output files is detailed in
Information MacSyFinder’s documentation: http:// macsyfinder.readthedocs.
in MacSyFinder’s Output org/en/latest/outputs.html.
Files Some output files are particularly useful for improving the
design of a new model. The standard output (“macsyfinder.out”)
shows how the tool processed the instances of the different com-
ponents to validate the systems. Together with the files storing raw
or filtered HMMER hits (in the “hmmer_results” directory), it
gives an overview of the detection process, from the detection of
components to the validation of the clusters. For automated down-
stream analyses of MacSyFinder’s results, the files “macsyfinder.
report” and “macsyfinder.summary” are of particular interest. The
former contains the list of components identified in each instance
with information on the quality of the HMMER matches. The lat-
ter describes the content of each instance of a secretion system and
can be loaded as a dictionary object using a Python script. It can be
particularly useful in the optimization steps (see Notes 6 and 7).
3.4.2 Visualization MacSyView allows the user to visualize the results of MacSyFinder
of Results with MacSyView in a web browser. It reads the “results.macsyfinder.json” output
file. Clicking on a system opens a window with three panels show-
ing the number of components per category, the genomic context,
and the statistics of the detection of each component (Fig. 3).
14 Sophie S. Abby and Eduardo P.C. Rocha
Fig. 3 Screenshots of MacSyView application. (a) The application reads the “results.macsyfinder.json” file from
MacSyFinder’s results directory. (b and c) The selection from a list of detected systems leads to a page with a
description of the instance in relation to the model. (d) The graphical representation of the instance in its
genomic context can be downloaded. (e) A table details the identified components (e.g., name, length) along
with their HMMER scores and other statistics
3.5 Optimization The initial models should be very general to assess the diversity of
and Validation the instances of the system. They can then be optimized iteratively
depending on the results obtained in the analysis of the reference
3.5.1 Optimization
data set (and external data). The output files discussed in Subsection
3.4.2 can be used to identify the parts of the model that must be
improved. The specific case of model optimization for poorly stud-
ied systems is described in Notes 6 and 7.
3.5.2 Validation The final model must be tested on an independent validation data
set (see Note 1). This procedure allows the quantification of the
sensitivity of the model, i.e., to know how well it identifies a secre-
tion system and its different components. It is more difficult to
assess its specificity, i.e., its ability to separate true from false
instances, since usually there is no reliable information on invalid
protein secretion systems.
At the end of the validation process, it might seem tempting
(or necessary) to correct the initial model to identify a larger frac-
tion of the instances of the validation data set. It must be borne in
mind that this violates the hypothesis of independence between the
model and the validation data set. Hence, if the model is changed
In Silico Identification of Secretion Systems 15
4 Notes
Fig. 4 Optimization of the quorum and colocalization parameters for T6SSi. (a) Distribution of number of differ-
ent components of T6SSi colocalized (d ≤ 20). (b) Distribution of maximum distance between consecutive
components in T6SSi detected with d ≤ 20. The minimum number of components required for a T6SSi was set
to 11 in the T6SSi model, which corresponds to the start of the second peak in the distribution represented in
panel (a). T6SSi detected as full systems are colored in blue. Figures and legends are freely reproduced with
modification from reference [9] (as specified by the Creative Commons Attribution (CC BY) license version 4.0)
References
1. Kanonenberg K, Schwarz CK, Schmitt L molecular architecture and mechanism. Nat
(2013) Type I secretion systems—a story of Rev Microbiol 10(5):336–351
appendices. Res Microbiol 164(6):596–604 4. Galan JE, Lara-Tejero M, Marlovits TC,
2. Campos M, Cisneros DA, Nivaskumar M, Wagner S (2014) Bacterial type III secretion
Francetic O (2013) The type II secretion sys- systems: specialized nanomachines for protein
tem—a dynamic fiber assembly nanomachine. delivery into target cells. Annu Rev Microbiol
Res Microbiol 164(6):545–555 68:415–438
3. Korotkov KV, Sandkvist M, Hol WG (2012) 5. Alvarez-Martinez CE, Christie PJ (2009)
The type II secretion system: biogenesis, Biological diversity of prokaryotic type IV
20 Sophie S. Abby and Eduardo P.C. Rocha
secretion systems. Microbiol Mol Biol Rev type IV secretion system. Infect Immun
73:775–808 78(5):1809–1823
6. van Ulsen P, Rahman S, Jong WS, Daleke- 19. Camacho C, Coulouris G, Avagyan V, Ma N,
Schermerhorn MH, Luirink J (2014) Type V Papadopoulos J, Bealer K, Madden TL (2009)
secretion: from biogenesis to biotechnology. BLAST+: architecture and applications. BMC
Biochim Biophys Acta 1843(8):1592–1611 Bioinformatics 10:421
7. Zoued A, Brunet YR, Durand E, Aschtgen 20. Eddy SR (2011) Accelerated profile HMM
MS, Logger L, Douzi B, Journet L, Cambillau searches. PLoS Comput Biol 7(10):e1002195
C, Cascales E (2014) Architecture and assem- 21. Katoh K, Toh H (2010) Parallelization of the
bly of the Type VI secretion system. Biochim MAFFT multiple sequence alignment pro-
Biophys Acta 1843(8):1664–1673 gram. Bioinformatics 26(15):1899–1900
8. McBride MJ, Nakane D (2015) Flavobacterium 22. Edgar RC (2004) MUSCLE: multiple sequence
gliding motility and the type IX secretion sys- alignment with high accuracy and high
tem. Curr Opin Microbiol 28:72–77 throughput. Nucleic Acids Res 32:1792–1797
9. Abby SS, Cury J, Guglielmini J, Néron B, 23. Gouy M, Guindon S, Gascuel O (2010)
Touchon M, Rocha EPC (2016) Identification SeaView version 4: a multiplatform graphi-
of protein secretion systems in bacterial cal user interface for sequence alignment and
genomes. Sci Rep 6:23080 phylogenetic tree building. Mol Biol Evol
10. Ginocchio CC, Olmsted SB, Wells CL, Galan 27(2):221–224
JE (1994) Contact with epithelial cells induces 24. Waterhouse AM, Procter JB, Martin DM, Clamp
the formation of surface appendages on M, Barton GJ (2009) Jalview Version 2—a mul-
Salmonella typhimurium. Cell 76(4):717–724 tiple sequence alignment editor and analysis
11. Peabody CR, Chung YJ, Yen MR, Vidal- workbench. Bioinformatics 25(9):1189–1191
Ingigliardi D, Pugsley AP, Saier MH Jr (2003) 25. Miele V, Penel S, Duret L (2011) Ultra-fast
Type II protein secretion and its relationship sequence clustering from similarity networks
to bacterial type IV pili and archaeal flagella. with SiLiX. BMC Bioinformatics 12:116
Microbiology 149(Pt 11):3051–3072 26. Enright AJ, Van Dongen S, Ouzounis CA
12. Abby SS, Rocha EP (2012) The non-flagellar (2002) An efficient algorithm for large-scale
type III secretion system evolved from the bac- detection of protein families. Nucleic Acids Res
terial flagellum and diversified into host-cell 30(7):1575–1584
adapted systems. PLoS Genet 8(9):e1002983 27. Mitchell A, Chang HY, Daugherty L, Fraser M,
13. Holland IB, Schmitt L, Young J (2005) Type Hunter S, Lopez R, McAnulla C, McMenamin
1 protein secretion in bacteria, the ABC- C, Nuka G, Pesseat S, Sangrador- Vegas A,
transporter dependent pathway. Mol Membr Scheremetjew M, Rato C, Yong SY, Bateman
Biol 22(1–2):29–39 A, Punta M, Attwood TK, Sigrist CJ, Redaschi
14. Paulsen IT, Park JH, Choi PS, Saier MH Jr N, Rivoire C, Xenarios I, Kahn D, Guyot D,
(1997) A family of gram-negative bacterial Bork P, Letunic I, Gough J, Oates M, Haft
outer membrane factors that function in the D, Huang H, Natale DA, Wu CH, Orengo C,
export of proteins, carbohydrates, drugs and Sillitoe I, Mi H, Thomas PD, Finn RD (2015)
heavy metals from gram-negative bacteria. The InterPro protein families database: the
FEMS Microbiol Lett 156(1):1–8 classification resource after 15 years. Nucleic
15. Abby SS, Neron B, Menager H, Touchon M, Acids Res 43(Database issue):D213–D221
Rocha EP (2014) MacSyFinder: a program to 28. Shrivastava A, Johnston JJ, van Baaren JM,
mine genomes for molecular systems with an McBride MJ (2013) Flavobacterium johnso-
application to CRISPR-Cas systems. PLoS One niae GldK, GldL, GldM, and SprA are required
9(10):e110726 for secretion of the cell surface gliding motil-
16. Christie PJ (2004) Type IV secretion: the ity adhesins SprB and RemA. J Bacteriol
Agrobacterium VirB/D4 and related con- 195(14):3201–3212
jugation systems. Biochim Biophys Acta 29. McBride MJ, Zhu Y (2013) Gliding motility
1694(1–3):219–234 and Por secretion system genes are widespread
17. Franco IS, Shuman HA, Charpentier X (2009) among members of the phylum Bacteroidetes.
The perplexing functions and surprising origins J Bacteriol 195(2):270–278
of Legionella pneumophila type IV secretion 30. Zhu Y, McBride MJ (2014) Deletion of the
effectors. Cell Microbiol 11(10):1435–1443 Cytophaga hutchinsonii type IX secretion
18. Gillespie JJ, Brayton KA, Williams KP, Diaz system gene sprP results in defects in glid-
MA, Brown WC, Azad AF, Sobral BW (2010) ing motility and cellulose utilization. Appl
Phylogenomics reveals a diverse Rickettsiales Microbiol Biotechnol 98(2):763–775
In Silico Identification of Secretion Systems 21
31. Kharade SS, McBride MJ (2015) Flavo 34. Finn RD, Tate J, Mistry J, Coggill PC, Sammut
bacterium johnsoniae PorV is required for SJ, Hotz HR, Ceric G, Forslund K, Eddy SR,
secretion of a subset of proteins targeted Sonnhammer EL, Bateman A (2008) The
to the type IX secretion system. J Bacteriol Pfam protein families database. Nucleic Acids
197(1):147–158 Res 36(Database issue):D281–D288
32. Altschul SF, Gish W, Miller W, Myers EW, 35. Haft DH, Selengut JD, Richter RA, Harkins
Lipman DJ (1990) Basic local alignment search D, Basu MK, Beck E (2013) TIGRFAMs and
tool. J Mol Biol 215:403–410 Genome Properties in 2013. Nucleic Acids Res
33. Boyer F, Fichant G, Berthod J, Vandenbrouck 41(Database issue):D387–D395
Y, Attree I (2009) Dissecting the bacterial 36. Quevillon E, Silventoinen V, Pillai S, Harte
type VI secretion system by a genome wide N, Mulder N, Apweiler R, Lopez R (2005)
in silico analysis: what can be learned from InterProScan: protein domains identi-
available microbial genomic resources? BMC fier. Nucleic Acids Res 33(Web Server
Genomics 10:104 issue):W116–W120
Chapter 2
Abstract
Many computational methods are available for predicting protein sorting in bacteria. When comparing
them, it is important to know that they can be grouped into three fundamentally different approaches:
signal-based, global-property-based and homology-based prediction. In this chapter, the strengths and
drawbacks of each of these approaches is described through many examples of methods that predict secre-
tion, integration into membranes, or subcellular locations in general. The aim of this chapter is to provide
a user-level introduction to the field with a minimum of computational theory.
Key words Protein sorting, Subcellular location, Secretion, Transmembrane proteins, Prediction,
Machine learning
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_2, © Springer Science+Business Media LLC 2017
23
24 Henrik Nielsen
never be better than the data used to train them. Nevertheless, the
authors had a point regarding the experimental sources of error
that can easily render a high-throughput experiment less reliable
than a well-trained computational method.
It can be difficult, however, to decide what to believe when the
authors of every computational method tend to describe their per-
formance as being superior to all others. There are different ways
of defining the problem, different ways of measuring the perfor-
mance, and different prerequisites used for prediction. The aim of
this chapter is not to provide a definitive answer to which method
is best for which problem—such a checklist would quickly become
outdated—but instead to give the reader a toolbox for critically
evaluating bioinformatics algorithms. This will involve a number of
examples of computational methods selected for their relevance
for bacteria, with the main focus being on Gram-negative bacteria.
A similar chapter with focus exclusively on Gram-positive bacteria
has been published elsewhere [5]. In general, prediction methods
will be mentioned only if they either provide publicly available web
servers or have strong historical relevance.
(continued)
25
Table 1
26
(continued)
(continued)
27
Table 1
28
(continued)
3 Algorithms for Prediction
Fig. 1 Sequence logo of Tat (twin-arginine translocation) signal peptides from both Gram-positive and Gram-
negative bacteria, aligned to the PROSITE profile PS51318/TAT. The height of each stack of letters corresponds
to the information (conservation) at that position, while the height of each individual letter is proportional to the
fraction of that amino acid at that position. Note that individual sequences may be shorter or longer than 40
amino acids; in the logo, they have been stretched or shortened to fit the model. Picture from PROSITE [28]
made with WebLogo [146]
32 Henrik Nielsen
Gram-negative data
1
0.98
0.96
0.94
0.92 SignalP 3.0 default
sensitivity
0.9 SignalP 4.0 default
0.88
0.86
0.84 SignalP 3.0 no TM
SignalP 3.0 all data
0.82 SignalP 4.0 no TM
SignalP 4.0 all data
0.8
0 0.05 0.1 0.15 0.2
FP rate
Fig. 3 Sequence logo of signal peptides from Gram-negative bacteria, aligned after their cleavage site (between
positions −1 and 0). The visible features are the cleavage site specifying residues in −3 and −1 (strong prefer-
ence for alanine), the hydrophobic region that approximately stretches from −16 to −7, and a preference for
the positively charged lysine in the N-terminal region. Note that no stretching or shortening of the sequences
has been performed; they have simply been aligned by the cleavage site; this is why no completely conserved
methionine is seen on the left-hand side. Picture made with WebLogo [146]
but it is still lower than that of SignalP 3.0, as can be seen from the
ROC curves in Fig. 2.
It should be stressed that the presence of an SP does not neces-
sarily mean that the protein is secreted. First, it may be periplasmic
or integrated into the outer membrane; second, there may be
downstream TMHs keeping the protein integrated in the cytoplas-
mic membrane. It has been reported that cleavable SPs are rarely
found in bacterial cytoplasmic membrane proteins [59], but a
quick search in UniProt [11] revealed that they are not that rare
after all, so a prediction of SPs should always be combined with a
search for TMHs (see Subheading 7) before drawing conclusions
about the SCL.
SignalP and the other SP predictors mentioned so far only pre-
dict classical SPs, translocated by the Sec system and cleaved by
type I signal peptidases. For lipoproteins cleaved by lipoprotein
signal peptidase, there are other prediction methods. LipoP [60]
(see Table 1) is an HMM-based method (although an ANN was
also trained during the development of the method). Even though
LipoP has been trained on sequences from Gram-negative bacteria
only, both the original paper and a later study [61] reported that it
demonstrated good performance on sequences from Gram-positive
bacteria also. Other methods include the ANN-based SPEPlip
[62] (see Table 1), which has separate options for Gram-negative
and Gram-positive bacteria, and the HMM-based PRED-LIPO
[63] (see Table 1), which is specific to Gram-positive bacteria. In
addition to these methods, there is also a profile in PROSITE [28],
dedicated to lipoproteins from both Gram-negative and Gram-
positive bacteria, called PROKAR_LIPOPROTEIN (see Table 1).
A sequence logo of lipoprotein SPs aligned to this model is shown
in Fig. 4. It should be noted that LipoP, SPEPlip, and PRED-
LIPO are all able to predict classical SPs as well, differentiating
between the two types of SP. Additionally, LipoP differentiates
between SPs and N-terminal TMHs.
Fig. 4 Sequence logo of lipoprotein signal peptides from both Gram-positive and Gram-negative bacteria,
aligned to the PROSITE profile PS51257/PROKAR_LIPOPROTEIN. Lipid attachment occurs at the completely
conserved cysteine in position 35. Note that individual sequences may be shorter or longer than 35 amino
acids; in the logo, they have been stretched or shortened to fit the model. Picture from PROSITE [28] made with
WebLogo [146]
Protein Sorting Prediction 39
proteins since they both assume that the submitted protein (only
one at a time is accepted) is a TMBB protein.
Methods that are best at one task are not necessarily the same
ones that are best at another task. In the paper on BOCTOPUS
version 2 [125], the authors compare their performance to a num-
ber of other methods and find that BOCTOPUS 2 is better at
predicting the correct topology than a number of other recent
methods (PRED-TMBB, ProfTMB, and BetAware; transFold and
TMBpro are not included in the benchmark), but HHomp and
BetAware yield better discrimination. Note that TMBB prediction
is still more difficult than TMH prediction; BOCTOPUS 2 reports
69% correct TMBB topologies, in contrast with the 83% correct
TMH topologies reported by TOPCONS.
8 Multicategory Predictors
9 Discussion
output formats as there are servers, and you get a rather frustrating
situation. Even the definitions of SCLs may vary from server to
server; for example, a peripheral membrane protein may be
defined as belonging to the membrane or to the compartment it
protrudes into.
This situation is clearly not ideal for the user, who might prefer
a “one-stop shop” to go to for all sequence-based prediction needs,
an equivalent of UniProt or InterPro. But this kind of confusion is
probably inevitable in a field that is evolving so fast. Scientific com-
petition is basically beneficial, and competing groups should cer-
tainly not be discouraged from publishing their predictors
independently. That being said, prediction servers ought to follow
certain standards concerning usability, definitions, and formats.
Personally, I must admit to having added to the complexity
through my involvement in the servers SignalP, LipoP, and TatP
(see Subheading 5). In hindsight, we should have published LipoP
and TatP not as separate servers but as functionalities within the
SignalP server. Hopefully, the next version of SignalP will be able
to predict all these types of SP in one user interface.
The multicategory prediction methods report quite impressive
performances, and, as described in Subheading 1, the error rates
for prediction may now be lower than the error rates for high-
throughput experiments. However, it is important to keep in mind
that these performances are achieved by analyzing the annotations
of homologs found by sequence similarity searches. I see three
problems with this. First, predictions for novel organisms and
metagenomics samples with few known homologs will necessarily
be harder than for the organisms the training and test sets were
built from, so coverage and precision for such organisms will be
considerably lower than the reported performances. Second, the
annotations used for prediction are themselves error prone and not
necessarily derived from experiments. Especially when relying on
keywords and GO terms, there is a real danger of circular reason-
ing, where annotations based on predictions are used as a basis for
new predictions, which then may enter the databases as annota-
tions. Third, homology-based predictions do not reflect real bio-
logical knowledge about the protein sorting process in the way a
successful signal-based predictor does.
As machine learning algorithms continue to evolve, new classes
of algorithms should also be expected to be applied to predictions
of SCL. In fields such as image processing and speech recognition,
novel types of ANNs—deep and recurrent neural networks—have
been used extensively in recent years [141, 142], and they are
beginning to be employed in bioinformatics as well, for example,
to predict protein secondary structures [143] or alternative splic-
ing [144]. The previously mentioned TMBpro (see Subheading 7)
is an example of a method based on recurrent ANNs. The advan-
tage of recurrent ANNs is that they are naturally designed to handle
52 Henrik Nielsen
References
1. Kyte J, Doolittle RF (1982) A simple method location of proteins. Nucleic Acids Res
for displaying the hydropathic character of a 26:2230–2236
protein. J Mol Biol 157:105–132 9. Hua S, Sun Z (2001) Support vector machine
2. von Heijne G (1983) Patterns of amino acids approach for protein subcellular localization
near signal-sequence cleavage sites. Eur prediction. Bioinformatics 17:721–728
J Biochem 133:17–21 10. Altschul SF, Madden TL, Schaffer AA et al
3. Gardy JL, Laird MR, Chen F et al (2005) (1997) Gapped BLAST and PSI-BLAST: a
PSORTb v.2.0: expanded prediction of bacte- new generation of protein database search
rial protein subcellular localization and programs. Nucleic Acids Res 25:3389
insights gained from comparative proteome 11. The UniProt Consortium (2015) UniProt: a
analysis. Bioinformatics 21:617–623 hub for protein information. Nucleic Acids
4. Rey S, Gardy J, Brinkman F (2005) Assessing Res 43:D204–D212
the precision of high-throughput computa- 12. Nair R, Rost B (2002a) Sequence conserved
tional and laboratory approaches for the for subcellular localization. Protein Sci
genome-wide identification of protein subcel- 11:2836–2847
lular localization in bacteria. BMC Genomics 13. Yu C-S, Chen Y-C, Lu C-H, Hwang J-K
6:162 (2006) Prediction of protein subcellular local-
5. Nielsen H (2016) Predicting subcellular ization. Proteins 64:643–651
localization of proteins by bioinformatic algo- 14. Nair R, Rost B (2002b) Inferring sub-cellular
rithms. In: Bagnoli F, Rappuoli R (eds) localization through automated lexical analy-
Protein export in gram-positive bacteria. sis. Bioinformatics 18(Suppl 1):S78–S86
Current topics in microbiology and immunol-
ogy. Springer, Berlin, Heidelberg 15. Lu Z, Szafron D, Greiner R et al (2004)
Predicting subcellular localization of proteins
6. Nakashima H, Nishikawa K (1994) using machine-learned classifiers. Bioinformatics
Discrimination of intracellular and extracellu- 20:547–556
lar proteins using amino acid composition and
residue-pair frequencies. J Mol Biol 16. Shatkay H, Höglund A, Brady S et al (2007)
238:54–61 SherLoc: high-accuracy prediction of protein
subcellular localization by integrating text
7. Andrade MA, O’Donoghue SI, Rost B (1998) and protein sequence data. Bioinformatics
Adaptation of protein surfaces to subcellular 23:1410–1417
location. J Mol Biol 276:517–525
17. Briesemeister S, Blum T, Brady S et al (2009)
8. Reinhardt A, Hubbard T (1998) Using neu- SherLoc2: a high-accuracy hybrid method for
ral networks for prediction of the subcellular
Protein Sorting Prediction 53
predicting subcellular localization of proteins. 31. Mitchell A, Chang H-Y, Daugherty L et al
J Proteome Res 8:5363–5366 (2015) The InterPro protein families data-
18. Chou K-C, Shen H-B (2010) Cell-PLoc 2.0: base: the classification resource after 15 years.
an improved package of web-servers for pre- Nucleic Acids Res 43:D213–D221
dicting subcellular localization of proteins in 32. Rish I (2001) An empirical study of the naive
various organisms. Nat Sci 2:1090–1103 Bayes classifier. In: IJCAI 2001 workshop
19. Chou K-C, Shen H-B (2006) Large-scale pre- Empir Methods Artif Intell. IBM, New York,
dictions of gram-negative bacterial protein pp 41–46
subcellular locations. J Proteome Res 33. Szafron D, Lu P, Greiner R et al (2004)
5:3420–3428 Proteome analyst: custom predictions with
20. Shen H-B, Chou K-C (2007) Gpos-PLoc: an explanations in a web-based tool for high-
ensemble classifier for predicting subcellular throughput proteome annotations. Nucleic
localization of gram-positive bacterial pro- Acids Res 32:W365–W371
teins. Protein Eng Des Sel 20:39–46 34. Briesemeister S, Rahnenführer J, Kohlbacher
21. Shen H-B, Chou K-C (2010) Gneg-mPLoc: a O (2010) Going from where to why—inter-
top-down strategy to enhance the quality of pretable prediction of protein subcellular
predicting subcellular localization of gram- localization. Bioinformatics 26:1232–1238
negative bacterial proteins. J Theor Biol 35. Hertz JA, Krogh AS, Palmer RG (1991)
264:326–333 Introduction to the theory of neural compu-
22. Shen H-B, Chou K-C (2009) Gpos-mPLoc: a tation. Westview Press, Redwood City, CA
top-down approach to improve the quality 36. Noble WS (2006) What is a support vector
of predicting subcellular localization of machine? Nat Biotechnol 24:1565–1567
gram- positive bacterial proteins. Protein 37. Hobohm U, Scharf M, Schneider R, Sander C
Pept Lett 16:1478–1484 (1992) Selection of representative protein
23. Xiao X, Wu Z-C, Chou K-C (2011) A multi- data sets. Protein Sci 1:409–417
label classifier for predicting the subcellular 38. Höglund A, Dönnes P, Blum T et al (2006)
localization of gram-negative bacterial pro- MultiLoc: prediction of protein subcellular
teins with both single and multiple sites. PLoS localization using N-terminal targeting
One 6:e20592 sequences, sequence motifs and amino acid
24. Wu Z-C, Xiao X, Chou K-C (2012) iLoc- composition. Bioinformatics 22:1158–1165
Gpos: a multi-layer classifier for predicting the 39. Sander C, Schneider R (1991) Database of
subcellular localization of singleplex and mul- homology-derived protein structures and
tiplex gram-positive bacterial proteins. Protein the structural meaning of sequence align-
Pept Lett 19:4–14 ment. Proteins 9:56–68
25. Stormo GD, Schneider TD, Gold L, 40. Nielsen H, Engelbrecht J, von Heijne G,
Ehrenfeucht A (1982) Use of the “percep- Brunak S (1996) Defining a similarity thresh-
tron” algorithm to distinguish translational old for a functional protein sequence pattern:
initiation sites in E. coli. Nucleic Acids Res the signal peptide cleavage site. Proteins
10:2997–3011 24:165–177
26. Schneider TD, Stephens RM (1990) Sequence 41. Nielsen H, Wernersson R (2006) An over-
logos: a new way to display consensus abundance of phase 0 introns immediately
sequences. Nucleic Acids Res 18:6097–6100 after the start codon in eukaryotic genes.
27. Krogh A, Brown M, Mian IS et al (1994) BMC Genomics 7:256
Hidden Markov models in computational 42. Gardy JL, Spencer C, Wang K et al (2003)
biology: applications to protein modeling. PSORT-B: improving protein subcellular
J Mol Biol 235:1501–1531 localization prediction for gram-negative bac-
28. Sigrist CJA, de Castro E, Cerutti L et al (2013) teria. Nucleic Acids Res 31:3613–3617
New and continuing developments at 43. Baldi P, Brunak S, Chauvin Y et al (2000)
PROSITE. Nucleic Acids Res 41:D344–D347 Assessing the accuracy of prediction algo-
29. Finn RD, Bateman A, Clements J et al (2014) rithms for classification: an overview.
Pfam: the protein families database. Nucleic Bioinformatics 16:412–424
Acids Res 42:D222–D230 44. Gorodkin J (2004) Comparing two
30. Haft DH, Selengut JD, Richter RA et al K-category assignments by a K-category cor-
(2013) TIGRFAMs and genome properties in relation coefficient. Comput Biol Chem
2013. Nucleic Acids Res 41:D387–D395 28:367–374
54 Henrik Nielsen
45. von Heijne G (1986) A new method for pre- peptides in gram-negative bacteria. Protein
dicting signal sequence cleavage sites. Nucleic Sci 12:1652–1662
Acids Res 14:4683–4690 61. Rahman O, Cummings SP, Harrington DJ,
46. McGeoch DJ (1985) On the predictive rec- Sutcliffe IC (2008) Methods for the bioinfor-
ognition of signal peptide sequences. Virus matic identification of bacterial lipoproteins
Res 3:271–286 encoded in the genomes of gram-positive
47. von Heijne G, Abrahmsén L (1989) Species- bacteria. World J Microbiol Biotechnol
specific variation in signal peptide design: 24:2377–2382
implications for protein secretion in foreign 62. Fariselli P, Finocchiaro G, Casadio R (2003)
hosts. FEBS Lett 244:439–446 SPEPlip: the detection of signal peptide and
48. Nielsen H, Brunak S, Engelbrecht J, von lipoprotein cleavage sites. Bioinformatics
Heijne G (1997) Identification of prokaryotic 19:2498–2499
and eukaryotic signal peptides and prediction 63. Bagos PG, Tsirigos KD, Liakopoulos TD,
of their cleavage sites. Protein Eng 10:1–6 Hamodrakas SJ (2008) Prediction of lipopro-
49. Nielsen H, Krogh A (1998) Prediction of sig- tein signal peptides in gram-positive bacteria
nal peptides and signal anchors by a hidden with a hidden Markov model. J Proteome Res
Markov model. Proc Int Conf Intell Syst Mol 7:5082–5093
Biol 6:122–130 64. Cristóbal S, de Gier J-W, Nielsen H, von
50. Bendtsen JD, Nielsen H, von Heijne G, Heijne G (1999) Competition between Sec-
Brunak S (2004) Improved prediction of sig- and TAT-dependent protein translocation in
nal peptides: SignalP 3.0. J Mol Biol Escherichia coli. EMBO J 18:2982–2990
340:783–795 65. Rose RW, Brüser T, Kissinger JC, Pohlschröder
51. Petersen TN, Brunak S, von Heijne G, M (2002) Adaptation of protein secretion to
Nielsen H (2011) SignalP 4.0: discriminating extremely high-salt conditions by extensive
signal peptides from transmembrane regions. use of the twin-arginine translocation pathway.
Nat Methods 8:785–786 Mol Microbiol 45:943–950
52. Menne KML, Hermjakob H, Apweiler R 66. Bendtsen JD, Nielsen H, Widdick D et al
(2000) A comparison of signal sequence pre- (2005a) Prediction of twin-arginine signal
diction methods using a test set of signal pep- peptides. BMC Bioinformatics 6:167
tides. Bioinformatics 16:741–742 67. Bagos PG, Nikolaou EP, Liakopoulos TD,
53. Klee E, Ellis L (2005) Evaluating eukaryotic Tsirigos KD (2010) Combined prediction of
secreted protein prediction. BMC Tat and Sec signal peptides with hidden
Bioinformatics 6:1–7 Markov models. Bioinformatics 26:
54. Choo K, Tan T, Ranganathan S (2009) A 2811–2817
comprehensive assessment of N-terminal sig- 68. Binnewies TT, Bendtsen JD, Hallin PF et al
nal peptides prediction methods. BMC (2005) Genome update: protein secretion
Bioinformatics 10:S2 systems in 225 bacterial genomes.
55. Zhang X, Li Y, Li Y (2009) Evaluating signal Microbiology 151:1013–1016
peptide prediction methods for gram-positive 69. Desvaux M, Hébraud M, Talon R, Henderson
bacteria. Biologia (Bratisl) 64:655–659 IR (2009) Secretion and subcellular localiza-
56. Hiller K, Grote A, Scheer M et al (2004) tions of bacterial proteins: a semantic aware-
PrediSi: prediction of signal peptides and ness issue. Trends Microbiol 17:139–145
their cleavage positions. Nucleic Acids Res 70. Bendtsen JD, Kiemer L, Fausbøll A, Brunak S
32:W375–W379 (2005b) Non-classical protein secretion in
57. Gomi M, Sonoyama M, Mitaku S (2004) bacteria. BMC Microbiol 5:58
High performance system for signal peptide 71. Yu L, Guo Y, Li Y et al (2010a) SecretP: iden-
prediction: SOSUIsignal. Chem-Bio Inform tifying bacterial secreted proteins by fusing
J 4:142–147 new features into Chou’s pseudo-amino acid
58. Frank K, Sippl MJ (2008) High-performance composition. J Theor Biol 267:1–6
signal peptide prediction based on sequence 72. Yu L, Luo J, Guo Y et al (2013) In silico iden-
alignment techniques. Bioinformatics 24: tification of gram-negative bacterial secreted
2172–2176 proteins from primary sequence. Comput
59. Broome-Smith JK, Gnaneshan S, Hunt LA Biol Med 43:1177–1181
et al (1994) Cleavable signal peptides are 73. Lloubes R, Bernadac A, Houot L, Pommier S
rarely found in bacterial cytoplasmic mem- (2013) Non classical secretion systems. Res
brane proteins. Mol Membr Biol 11:3–8 Microbiol 164:655–663
60. Juncker AS, Willenbrock H, von Heijne G 74. Luo J, Li W, Liu Z et al (2015) A sequence-
et al (2003) Prediction of lipoprotein signal based two-level method for the prediction of
Protein Sorting Prediction 55
type I secreted RTX proteins. Analyst 140: 88. Dong X, Lu X, Zhang Z (2015) BEAN 2.0:
3048–3056 an integrated web resource for the identifica-
75. Burstein D, Zusman T, Degtyar E et al (2009) tion and functional analysis of type III
Genome-scale identification of Legionella secreted effectors. Database 2015:bav064
pneumophila effectors using a machine learn- 89. Goldberg T, Rost B, Bromberg Y (2016)
ing approach. PLoS Pathog 5:e1000508 Computational prediction shines light on
76. Chen C, Banga S, Mertens K et al (2010) type III secretion origins. Sci Rep 6:34516
Large-scale identification and translocation of 90. Klein P, Kanehisa M, DeLisi C (1985) The
type IV secretion substrates by Coxiella bur- detection and classification of membrane-
netii. Proc Natl Acad Sci U S A spanning proteins. Biochim Biophys Acta
107:21755–21760 815:468–476
77. Lifshitz Z, Burstein D, Peeri M et al (2013) 91. von Heijne G (1992) Membrane protein
Computational modeling and experimental structure prediction: hydrophobicity analysis
validation of the Legionella and Coxiella and the positive-inside rule. J Mol Biol 225:
virulence- related type-IVB secretion signal. 487–494
Proc Natl Acad Sci U S A 110:E707–E715 92. von Heijne G, Gavel Y (1988) Topogenic
78. Zou L, Nan C, Hu F (2013) Accurate predic- signals in integral membrane proteins. Eur
tion of bacterial type IV secreted effectors J Biochem 174:671–678
using amino acid composition and PSSM pro- 93. Paul C, Rosenbusch JP (1985) Folding pat-
files. Bioinformatics 29:3135–3142 terns of porin and bacteriorhodopsin. EMBO
79. Wang Y, Wei X, Bao H, Liu S-L (2014) J 4:1593–1597
Prediction of bacterial type IV secreted effec- 94. Vogel H, Jähnig F (1986) Models for the
tors by C-terminal features. BMC Genomics structure of outer-membrane proteins of
15:50 Escherichia coli derived from raman spec-
80. McDermott JE, Corrigan A, Peterson E et al troscopy and prediction methods. J Mol
(2011) Computational prediction of type III Biol 190:191–199
and IV secreted effectors in gram-negative 95. Krogh A, Larsson B, von Heijne G,
bacteria. Infect Immun 79:23–32 Sonnhammer EL (2001) Predicting trans-
81. Anderson DM, Schneewind O (1997) A membrane protein topology with a hidden
mRNA signal for the type III secretion of Yop Markov model: application to complete
proteins by Yersinia enterocolitica. Science genomes. J Mol Biol 305:567–580
278:1140–1143 96. Tusnády GE, Simon I (2001) The HMMTOP
82. Samudrala R, Heffron F, McDermott JE transmembrane topology prediction server.
(2009) Accurate prediction of secreted sub- Bioinformatics 17:849–850
strates and identification of a conserved puta- 97. Möller S, Croning MDR, Apweiler R (2001)
tive secretion signal for type III secretion Evaluation of methods for the prediction of
systems. PLoS Pathog 5:e1000375 membrane spanning regions. Bioinformatics
83. Arnold R, Brandmaier S, Kleine F et al (2009) 17:646–653
Sequence-based prediction of type III 98. Elofsson A, von Heijne G (2007) Membrane
secreted proteins. PLoS Pathog 5:e1000376 protein structure: prediction versus reality.
84. Löwer M, Schneider G (2009) Prediction of Annu Rev Biochem 76:125–140
type III secretion signals in genomes of gram- 99. Punta M, Forrest LR, Bigelow H et al (2007)
negative bacteria. PLoS One 4:e5917 Membrane protein prediction methods.
85. Wang Y, Zhang Q, Sun M, Guo D (2011) Methods 41:460–474
High-accuracy prediction of bacterial type III 100. Tusnády GE, Simon I (2010) Topology
secreted effectors based on position-specific prediction of helical transmembrane pro-
amino acid composition profiles. teins: how far have we reached? Curr
Bioinformatics 27:777–784 Protein Pept Sci 11:550–561
86. Wang Y, Sun M, Bao H, White AP (2013) 101. Käll L, Krogh A, Sonnhammer EL (2004) A
T3_MM: a Markov model effectively classi- combined transmembrane topology and signal
fies bacterial type III secretion signals. PLoS peptide prediction method. J Mol Biol 338:
One 8:e58173 1027–1036
87. Dong X, Zhang Y-J, Zhang Z (2013) Using 102. Reynolds SM, Käll L, Riffle ME et al (2008)
weakly conserved motifs hidden in secretion Transmembrane topology and signal peptide
signals to identify type-III effectors from bac- prediction using dynamic Bayesian networks.
terial pathogen genomes. PLoS One 8:e56632 PLoS Comput Biol 4:e1000213
56 Henrik Nielsen
103. Jones DT (2007) Improving the accuracy of 117. Bagos P, Liakopoulos T, Spyropoulos I,
transmembrane protein topology prediction Hamodrakas S (2004a) A hidden Markov
using evolutionary information. model method, capable of predicting and
Bioinformatics 23:538–544 discriminating beta-barrel outer membrane
104. Nugent T, Jones DT (2009) Transmembrane proteins. BMC Bioinformatics 5:29
protein topology prediction using support 118. Bagos PG, Liakopoulos TD, Spyropoulos IC,
vector machines. BMC Bioinformatics 10:159 Hamodrakas SJ (2004b) PRED-TMBB: a
105. Viklund H, Bernsel A, Skwark M, Elofsson A web server for predicting the topology of
(2008) SPOCTOPUS: a combined predictor β-barrel outer membrane proteins. Nucleic
of signal peptides and membrane protein Acids Res 32:W400–W404
topology. Bioinformatics 24:2928–2929 119. Bigelow HR, Petrey DS, Liu J et al (2004)
106. Viklund H, Elofsson A (2008) OCTOPUS: Predicting transmembrane beta-barrels in
improving topology prediction by two-track proteomes. Nucleic Acids Res 32:2566–2577
ANN-based preference scores and an 120. Bigelow H, Rost B (2006) PROFtmb: a web
extended topological grammar. Bioinformatics server for predicting bacterial transmembrane
24:1662–1668 beta barrel proteins. Nucleic Acids Res
107. Viklund H, Elofsson A (2004) Best α-helical 34:W186–W188
transmembrane protein topology predictions 121. Bagos P, Liakopoulos T, Hamodrakas S
are achieved using hidden Markov models (2005) Evaluation of methods for predicting
and evolutionary information. Protein Sci the topology of beta-barrel outer membrane
13:1908–1917 proteins and a consensus prediction method.
108. Käll L, Krogh A, Sonnhammer EL (2005) An BMC Bioinformatics 6:7
HMM posterior decoder for sequence feature 122. Jacoboni I, Martelli PL, Fariselli P et al (2001)
prediction that includes homology information. Prediction of the transmembrane regions of
Bioinformatics 21:i251–i257 β-barrel membrane proteins with a neural
109. Bernsel A, Viklund H, Falk J et al (2008) network-based predictor. Protein Sci
Prediction of membrane-protein topology 10:779–787
from first principles. Proc Natl Acad Sci 105: 123. Natt NK, Kaur H, Raghava GPS (2004)
7177–7181 Prediction of transmembrane regions of
110. Hessa T, Meindl-Beinker NM, Bernsel A et al β-barrel proteins using ANN- and SVM-based
(2007) Molecular code for transmembrane- methods. Proteins 56:11–18
helix recognition by the Sec61 translocon. 124. Hayat S, Elofsson A (2012) BOCTOPUS:
Nature 450:1026–1030 improved topology prediction of transmem-
111. Taylor PD, Attwood TK, Flower DR (2003) brane β barrel proteins. Bioinformatics
BPROMPT: a consensus server for membrane 28:516–522
protein prediction. Nucleic Acids Res 125. Hayat S, Peters C, Shu N et al (2016) Inclusion
31:3698–3700 of dyad-repeat pattern improves topology pre-
112. Bernsel A, Viklund H, Hennerdal A, Elofsson diction of transmembrane β-barrel proteins.
A (2009) TOPCONS: consensus prediction Bioinformatics 32:1571–1573
of membrane protein topology. Nucleic 126. Berven FS, Flikka K, Jensen HB, Eidhammer
Acids Res 37:W465–W468 I (2004) BOMP: a program to predict inte-
113. Tsirigos KD, Peters C, Shu N et al (2015) gral β-barrel outer membrane proteins
The TOPCONS web server for consensus encoded within genomes of gram-negative
prediction of membrane protein topology and bacteria. Nucleic Acids Res 32:W394–W399
signal peptides. Nucleic Acids Res 127. Remmert M, Linke D, Lupas AN, Söding
43:W401–W407 J (2009) HHomp—prediction and classifica-
114. Hennerdal A, Elofsson A (2011) Rapid mem- tion of outer membrane proteins. Nucleic
brane protein topology prediction. Acids Res 37:W446–W451
Bioinformatics 27:1322–1323 128. Savojardo C, Fariselli P, Casadio R (2011)
115. Diederichs K, Freigang J, Umhau S et al
Improving the detection of transmembrane
(1998) Prediction by a neural network of β-barrel chains with N-to-1 extreme learning
outer membrane β-strand protein topology. machines. Bioinformatics 27:3123–3128
Protein Sci 7:2413–2420 129. Savojardo C, Fariselli P, Casadio R (2013)
116. Martelli PL, Fariselli P, Krogh A, Casadio R BETAWARE: a machine-learning tool to
(2002) A sequence-profile-based HMM for detect and predict transmembrane beta-barrel
predicting and discriminating β barrel mem- proteins in prokaryotes. Bioinformatics
brane proteins. Bioinformatics 18:S46–S53 29:504–505
Protein Sorting Prediction 57
130. Waldispühl J, Berger B, Clote P, Steyaert J-M 140. Imai K, Asakawa N, Tsuji T et al (2008)
(2006a) transFold: a web server for predicting SOSUI-GramN: high performance prediction
the structure and residue contacts of trans- for sub-cellular localization of proteins in
membrane beta-barrels. Nucleic Acids Res gram-negative bacteria. Bioinformation
34:W189–W193 2:417–421
131. Waldispühl J, Berger B, Clote P, Steyaert J-M 141. Krizhevsky A, Sutskever I, Hinton GE (2012)
(2006b) Predicting transmembrane β-barrels ImageNet classification with deep convolu-
and interstrand residue interactions from tional neural networks. In: Pereira F, Burges
sequence. Proteins 65:61–74 CJC, Bottou L, Weinberger KQ (eds)
132. Randall A, Cheng J, Sweredoski M, Baldi P Advances in neural information processing
(2008) TMBpro: secondary structure, systems, vol. 25, Curran Associates, Inc., Red
β-contact and tertiary structure prediction of Hook, NY, pp 1097–1105
transmembrane β-barrel proteins. 142. Dahl GE, Yu D, Deng L, Acero A (2012)
Bioinformatics 24:513–520 Context-dependent pre-trained deep neural
133. Nakai K, Kanehisa M (1991) Expert system networks for large-vocabulary speech recog-
for predicting protein localization sites in nition. IEEE Trans Audio Speech Lang
gram-negative bacteria. Proteins 11:95–110 Process 20:30–42
134. Yu NY, Wagner JR, Laird MR et al (2010b) 143. Magnan CN, Baldi P (2014) SSpro/ACCpro
PSORTb 3.0: improved protein subcellular 5: almost perfect prediction of protein sec-
localization prediction with refined localization ondary structure and relative solvent accessi-
subcategories and predictive capabilities for all bility using profiles, machine learning and
prokaryotes. Bioinformatics 26:1608–1615 structural similarity. Bioinformatics
135. Magnus M, Pawlowski M, Bujnicki JM
30:2592–2597
(2012) MetaLocGramN: a meta-predictor of 144. Xiong HY, Alipanahi B, Lee LJ et al (2015)
protein subcellular localization for gram- The human splicing code reveals new insights
negative bacteria. Biochim Biophys Acta into the genetic determinants of disease.
1824:1425–1433 Science 347:1254806
136. Ashburner M, Ball CA, Blake JA et al (2000) 145. Sønderby SK, Sønderby CK, Nielsen H,
Gene ontology: tool for the unification of Winther O (2015) Convolutional LSTM
biology. Nat Genet 25:25–29 networks for subcellular localization of pro-
137. Bhasin M, Garg A, Raghava GPS (2005)
teins. In: Dediu A-H, Hernández-Quiroz F,
PSLpred: prediction of subcellular localiza- Martín-Vide C, Rosenblueth DA (eds)
tion of bacterial proteins. Bioinformatics Algorithms for computational biology,
21:2522–2524 Lecture notes in computer science, vol
9199. Springer International Publishing,
138. Goldberg T, Hecht M, Hamp T et al (2014) New York, pp 68–80
LocTree3 prediction of localization. Nucleic
Acids Res 42:W350–W355 146.
Crooks GE, Hon G, Chandonia J-M,
Brenner SE (2004) WebLogo: a sequence
139. Goldberg T, Hamp T, Rost B (2012)
logo generator. Genome Res 14:1188–1190
LocTree2 predicts localization for all domains
of life. Bioinformatics 28:i458–i465
Chapter 3
Cell Fractionation
Melissa Petiti, Laetitia Houot, and Denis Duché
Abstract
Protein function is generally dependent on its subcellular localisation. In Gram-negative bacteria such as
Escherichia coli, a protein can be targeted to five different compartments: the cytoplasm, the inner mem-
brane, the periplasm, the outer membrane and the extracellular medium. Different approaches can be used
to determine the protein localisation within a cell such as in silico identification of protein signal sequences
and motifs, electron microscopy and immunogold labelling, optical fluorescence microscopy, and bio-
chemical technics. In this chapter, we describe a simple and efficient method to isolate the different com-
partments of Escherichia coli by a fractionation method and to determine the presence of the protein of
interest. For inner membrane proteins we propose a method to discriminate between integral and periph-
eral membrane proteins.
Key words Spheroplast, Peptidoglycan, Osmotic shock, Freeze and thaw, Protein solubilisation,
Membrane, Subcellular localisation
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_3, © Springer Science+Business Media LLC 2017
59
60 Melissa Petiti et al.
2 Materials
3 Methods
3.1 Cell In this section, we detail step by step spheroplast preparation from
Fractionation/ Escherichia coli cells (see Note 1) using a method based on lysozyme/
Spheroplast Formation EDTA treatment [3] and a mild osmotic shock [4–6] (see Note 2).
1. Grow a 3 mL starter culture overnight in a lysogeny broth
medium at 37 °C with required antibiotics.
2. Inoculate a 20 mL culture at OD600 = 0.05 and incubate at 37
°C until the optical density of the culture is around 0.8. If
necessary, induce protein production under the required con-
ditions (see Note 3).
3. Take 1 mL of culture and centrifuge for 5 min at 5000 × g to
pellet the cells. Discard the supernatant. Resuspend the pellet
in an appropriate volume of sodium dodecyl sulphate poly-
acrylamide gel electrophoresis (SDS-PAGE) loading buffer.
This fraction will be referred as the total cell fraction (T).
Cell Fractionation 61
The next steps will be performed at 4 °C, and all buffers must
be cooled in advance on ice before use.
4. Centrifuge the remaining culture for 5 min at 5000 × g at 4 °C
(see Note 4). Discard the supernatant.
5. Gently resuspend the cell pellet in 200 μL of TES buffer
(described in materials section, see Note 5). Do not vortex
and do not pipette, only resuspend the cell pellet by inverting
the tube.
6. Add 8 μL of a freshly prepared solution of lysozyme (10 mg/mL
in TES buffer) and mix gently by shaking the tube.
7. Add 720 μL TES buffer diluted twice in water (v/v) and incu-
bate for 30 min on ice. Gently mix the suspension to perform
the osmotic shock by gently inverting and rolling the tube (see
Note 6).
8. Centrifuge at 5000 × g for 5 min at 4 °C. Keep the pellet as the
spheroplast fraction, IM + cytoplasm (+OM), and the superna-
tant as the periplasmic fraction, P.
9. Resuspend the spheroplast fraction in 1 mL of TES buffer
diluted twice in water (v/v) containing 2 mM PMSF, 2 mM
MgCl2 and 10 μg/mL DNase 1 (see Notes 7 and 8).
10. Lyse the spheroplasts by performing four cycles of freezing and
thawing, from −273 °C (liquid azote) to 37 °C (see Note 9).
11. Remove unbroken cells and cell debris by centrifugation at
2000 × g for 5 min. Keep the supernatant as cytoplasmic and
membrane fraction.
12. Centrifuge the supernatant at 120,000 × g, 4 °C, for 45 min.
Use a Beckman Coulter Optima TLX ultracentrifuge and a
TLA55 fixed-angle rotor for small-volume ultracentrifugation
or something similar. The pellet is kept as the membrane frac-
tion and the supernatant is conserved as the cytoplasmic
fraction.
13. The membrane fraction is suspended in 1 mL TES buffer diluted
twice in water (v/v) or in the desired buffer (see Note 10).
Separation of IM and OM is described in Chapter 6 of this
book.
14. At this step, fractions (OD600 = 0.2–0.4) could be tested for
the presence of the protein of interest by SDS-PAGE and west-
ern blot analysis with required antibodies. As a control, the
same fractions can be tested for the presence of specific IM,
OM, cytoplasm, or periplasm markers.
4 Notes
12. This step can be done overnight in a cold room under rotary
agitation.
13. Be careful; pellets are not always visible after centrifugation.
Carefully place the tubes in the centrifuge to determine the
location of the future pellet.
14. Never let the pellet air-dry completely because this will dra-
matically impede resuspension.
Acknowledgments
References
1. Kaback HR (1972) Transport across isolated 7. Kaback HR (1971) Bacterial membranes.
bacterial cytoplasmic membranes. Biochim Methods Enzymol 22:99–120
Biophys Acta 265:367–416 8. Witholt B, Heerikhuizen HV, De Leij L (1976)
2. Kellenberger E, Ryther A (1958) Cell wall and How does lysozyme penetrate through the
cytoplasmic membrane of Escherichia coli. bacterial outer membrane. Biochim Biophys
J Biophys Biochem Cytol 25:323–326 Acta 443:534–544
3. Neu HC, Heppel LA (1964) The release of 9. Mowbray J, Moses V (1976) The tentative
Ribonuclease into the medium when Escherichia identification in Escherichia coli of a multien-
coli cells are converted to spheroplasts. J Biol zyme complex with glycolytic activity. Eur
Chem 239:3893–3900 J Biochem 66:25–36
4. French C, Keshavarz-Moore E, Ward JM 10. Schook W, Puszkin P, Bloom W, Ores C, Kochwa
(1996) Development of a simple method for S (1979) Mechanochemical properties of brain
the recovery of recombinant proteins from the clathrin: interactions with actin and alpha-actinin
Escherichia coli periplasm. Enzym Microb and polymerization into basketlike structures or
Technol 19:332–338 filaments. Proc Natl Acad Sci U S A 76:116–120
5. Skerra A, Plückthun A (1991) Secretion and in 11. Schnaitman CA (1971) Solubilization of the
vivo folding of the Fab fragment of the antibody cytoplasmic membrane of Escherichia coli by
McPC603 in Escherichia coli: influence of triton X-100. J Bacteriol 108:545–552
disulphides and cis-prolines. Protein Eng 12. Fujiki Y, Fowler S, Shio H, Hubbard AL,
4:971–979 Lazarow PB (1982) Polypeptide and phospho-
6. Nossal NG, Heppel LA (1966) The release of lipid composition of the membrane of rat liver
enzymes by osmotic shock from Escherichia peroxisomes: comparison with endoplasmic
coli in exponential phase. J Biol Chem reticulum and mitochondrial membranes.
241:3055–3062 J Cell Biol 93:103–110
Chapter 4
Abstract
In recent years it has become evident that lipoproteins play crucial roles in the assembly of bacterial
envelope-embedded nanomachineries and in the processes of protein export/secretion. In this chapter we
describe a method to determine their precise localisation, for example inner versus outer membrane, in
Gram-negative bacteria using human opportunistic pathogen Pseudomonas aeruginosa as a model. A fusion
protein between a given putative lipoprotein and the red fluorescent protein mCherry must be created and
expressed in a strain expressing cytoplasmic green fluorescent protein (GFP). Then the peripheral localisa-
tion of the fusion protein in the cell can be examined by treating cells with lysozyme to create spheroplasts
and monitoring fluorescence under a confocal microscope. Mutants in the signal peptide can be engi-
neered to study the association with the membrane and efficiency of transport. This protocol can be
adapted to monitor lipoprotein localisation in other Gram-negative bacteria.
Key words Lipoprotein, Localisation, Cell envelope, Spheroplasts, Bacterial secretion, Fluorescence
microscopy
1 Introduction
†
To the memory of Didier Grunwald, who passed away recently.
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_4, © Springer Science+Business Media LLC 2017
65
66 Maria Guillermina Casabona et al.
2 Materials
Fig. 1 Outer membrane localisation of T6SS signalling lipoprotein TagQ assessed by confocal microscopy. (a)
P. aeruginosa PAO1 expressing cytoplasmic GFP and TagQ-mCherry were imaged directly from exponentially
grown cultures (upper panel) or after treatment with lysozyme (lower panel). Note the red labelling in the
periphery of bacteria and in partially detached OM in spheroplast preparations. Inserts represent a plot
Lipoprotein Localisation 69
2.3 Imaging 1.
Confocal microscope: TCS-SP2 (Leica Microsystems,
Mannheim, Germany) using a DMRE (Leica) upright micro-
scope. For confocal acquisitions, fluorescence is collected
through a spectral detection mode using mechanical filtering.
The different channels are acquired sequentially to avoid fluo-
rescence leakage.
2. Software for image processing like FiJi free software [29].
3 Methods
Fig. 1 (continued) profile of the fluorescence intensity on selected objects obtained by FiJi free software. (b) P.
aeruginosa PAO1 expressing cytoplasmic GFP and the TagQ-mCherry protein lacking cysteine residue within
lipobox motif. The growth conditions, spheroplast preparation, imaging and image treatment were done as for
the wild-type TagQ, represented in a. Note the periphery localisation of TagQΔC-mCherry in bacteria and
absence of labelling in spheroplasts, indicating the loss of the protein during lysozyme treatment. (c) Schema
showing putative localisation of bacterial lipoproteins assessed by analysing mCherry protein fusions by fluo-
rescent microscopy before and after lysozyme treatment. Three options are represented: IM, OM and periplas-
mic localisation
70 Maria Guillermina Casabona et al.
500 and 535 nm for GFP and from 575 to 650 nm for mCherry
(see Note 6). Images (512 × 512 pixels) are obtained with vari-
ous electronic zooms (×8, ×32). The optical sections are
acquired at a focal position corresponding to a median position
in the analysed objects.
5. To determine the relative position for green and red fluores-
cence, display a plot profile of the fluorescence intensity along
the line in the images. Those histograms can be obtained using
FiJi free software (see Fig. 1).
4 Notes
Acknowledgements
We thank Dr. K. M. Sall for initiating studies on TagQ and TssJ1
fusion proteins and Dr. S. Elsen for help in plasmid generation.
MGC was supported by a PhD grant from the French Cystic
Fibrosis Association Vaincre la Mucovisidose. The microscopy
facility is supported by the Biosciences and Biotechnology Institute
of Grenoble (BIG), CEA-Grenoble and the grant to Laboratoire
of Excellence, LabEx GRAL (ANR-10-LABX-49-01).
References
1. Farris C, Sanowar S, Bader MW, Pfuetzner tein RcsF, a new substrate for the periplasmic
R, Miller SI (2010) Antimicrobial peptides protein-disulfide isomerase DsbC. J Biol Chem
activate the Rcs regulon through the outer 286(19):16734–16742
membrane lipoprotein RcsF. J Bacteriol 3. Aliprantis AO, Yang RB, Mark MR, Suggett
192(19):4894–4903 S, Devaux B, Radolf JD et al (1999) Cell
2. Leverrier P, Declercq JP, Denoncin K, activation and apoptosis by bacterial lipopro-
Vertommen D, Hiniker A, Cho SH et al (2011) teins through toll-like receptor-2. Science
Crystal structure of the outer membrane pro- 285(5428):736–739
Lipoprotein Localisation 73
4. Pugsley AP (1993) The complete general for correct assembly of the Pseudomonas
secretory pathway in gram-negative bacteria. aeruginosa type III secretion apparatus in the
Microbiol Rev 57(1):50–108 bacterial membrane and full virulence in vivo.
5. Zuckert WR (2014) Secretion of bacterial lipo- Infect Immun 83(5):1789–1798
proteins: through the cytoplasmic membrane, 17. Guilvout I, Chami M, Engel A, Pugsley AP,
the periplasm and beyond. Biochim Biophys Bayan N (2006) Bacterial outer membrane
Acta 1843(8):1509–1516 secretin PulD assembles and inserts into the
6. Konovalova A, Silhavy TJ (2015) Outer mem- inner membrane in the absence of its pilotin.
brane lipoprotein biogenesis: Lol is not the EMBO J 25(22):5241–5249
end. Philos Trans R Soc Lond Ser B Biol Sci 18. Viarre V, Cascales E, Ball G, Michel GP,
370(1679) Filloux A, Voulhoux R (2009) HxcQ lipose-
7. Babu MM, Priya ML, Selvan AT, Madera M, cretin is self-piloted to the outer membrane
Gough J, Aravind L et al (2006) A database by its N-terminal lipid anchor. J Biol Chem
of bacterial lipoproteins (DOLOP) with func- 284(49):33815–33823
tional assignments to predicted lipoproteins. 19. Aschtgen MS, Bernard CS, De Bentzmann S,
J Bacteriol 188(8):2761–2773 Lloubes R, Cascales E (2008) SciN is an outer
8. Madan Babu M, Sankaran K (2002) membrane lipoprotein required for type VI
DOLOP--database of bacterial lipoproteins. secretion in enteroaggregative Escherichia coli.
Bioinformatics 18(4):641–643 J Bacteriol 190(22):7523–7531
9. Remans K, Vercammen K, Bodilis J, Cornelis 20. Casabona MG, Silverman JM, Sall KM, Boyer
P (2010) Genome-wide analysis and literature- F, Coute Y, Poirel J et al (2013) An ABC trans-
based survey of lipoproteins in Pseudomonas porter and an outer membrane lipoprotein par-
aeruginosa. Microbiology 156(Pt 9):2597–2607 ticipate in posttranslational activation of type
10. Casabona MG, Vandenbrouck Y, Attree I, VI secretion in Pseudomonas Aeruginosa.
Coute Y (2013) Proteomic characterization of Environ Microbiol 15(2):471–486
Pseudomonas aeruginosa PAO1 inner mem- 21. Durand E, Nguyen VS, Zoued A, Logger
brane. Proteomics 13(16):2419–2423 L, Pehau-Arnaudet G, Aschtgen MS et al
11. Fernandez D, Dang TA, Spudich GM, Zhou (2015) Biogenesis and structure of a type VI
XR, Berger BR, Christie PJ (1996) The secretion membrane core complex. Nature
Agrobacterium tumefaciens virB7 gene prod- 523(7562):555–560
uct, a proposed component of the T-complex 22. Felisberto-Rodrigues C, Durand E, Aschtgen
transport apparatus, is a membrane-associated MS, Blangy S, Ortiz-Lombardia M, Douzi B
lipoprotein exposed at the periplasmic surface. et al (2011) Towards a structural comprehen-
J Bacteriol 178(11):3156–3167 sion of bacterial type VI secretion systems:
12. Fernandez D, Spudich GM, Zhou XR, Christie characterization of the TssJ-TssM complex
PJ (1996) The Agrobacterium tumefaciens of an Escherichia coli pathovar. PLoS Pathog
VirB7 lipoprotein is required for stabiliza- 7(11):e1002386
tion of VirB proteins during assembly of the 23. Rao VA, Shepherd SM, English G, Coulthurst
T-complex transport apparatus. J Bacteriol SJ, Hunter WN (2011) The structure of
178(11):3168–3176 Serratia marcescens Lip, a membrane-bound
13. Christie PJ, Cascales E (2005) Structural component of the type VI secretion system.
and dynamic properties of bacterial type IV Acta Crystallogr Sect D 67(Pt 12):1065–1072
secretion systems (review). Mol Membr Biol 24. Basler M, Ho BT, Mekalanos JJ (2013) Tit-
22(1–2):51–61 for-
tat: type VI secretion system counterat-
14. Collin S, Guilvout I, Nickerson NN, Pugsley tack during bacterial cell-cell interactions. Cell
AP (2011) Sorting of an integral outer mem- 152(4):884–894
brane protein via the lipoprotein-specific Lol 25. Alcock F, Baker MA, Greene NP, Palmer T,
pathway and a dedicated lipoprotein pilotin. Wallace MI, Berks BC (2013) Live cell imag-
Mol Microbiol 80(3):655–665 ing shows reversible assembly of the TatA
15. Izore T, Perdu C, Job V, Attree I, Faudry component of the twin-arginine protein
E, Dessen A (2011) Structural character- transport system. Proc Natl Acad Sci U S A
ization and membrane localization of ExsB 110(38):E3650–E3659
from the type III secretion system (T3SS) 26. Guillon L, El Mecherki M, Altenburger S,
of Pseudomonas aeruginosa. J Mol Biol Graumann PL, Schalk IJ (2012) High cel-
413(1):236–246 lular organization of pyoverdine biosynthe-
16. Perdu C, Huber P, Bouillot S, Blocker A, sis in Pseudomonas aeruginosa: clustering of
Elsen S, Attree I et al (2015) ExsB is required PvdA at the old cell pole. Environ Microbiol
14(8):1982–1994
74 Maria Guillermina Casabona et al.
27. Imperi F, Visca P (2013) Subcellular localiza- 31. Thibault J, Faudry E, Ebel C, Attree I, Elsen
tion of the pyoverdine biogenesis machinery S (2009) Anti-activator ExsD forms a 1:1
of Pseudomonas aeruginosa: a membrane- complex with ExsA to inhibit transcription
associated “siderosome”. FEBS Lett of type III secretion operons. J Biol Chem
587(21):3387–3391 284(23):15762–15770
28. Lewenza S, Mhlanga MM, Pugsley AP 32. Hoang TT, Kutchma AJ, Becher A, Schweizer
(2008) Novel inner membrane retention sig- HP (2000) Integration-proficient plasmids for
nals in Pseudomonas aeruginosa lipoproteins. Pseudomonas aeruginosa: site-specific integra-
J Bacteriol 190(18):6119–6125 tion and use for engineering of reporter and
29. Schindelin J, Arganda-Carreras I, Frise E expression strains. Plasmid 43(1):59–72
et al (2012) Fiji: an open-source platform 33. Newman JR, Fuqua C (1999) Broad-host-
for biological-
image analysis. Nat Methods range expression vectors that carry the
9(7):676–682 L-arabinose-inducible Escherichia coli araBAD
30. De Bentzmann S, Giraud C, Bernard CS, promoter and the araC regulator. Gene
Calderon V, Ewald F, Plesiat P et al (2012) 227(2):197–203
Unique biofilm signature, drug susceptibility 34. Chuanchuen R, Narasaki CT, Schweizer HP
and decreased virulence in Drosophila through (2002) Benchtop and microcentrifuge prepa-
the Pseudomonas aeruginosa two-component ration of Pseudomonas aeruginosa competent
system PprAB. PLoS Pathog 8(11):e1003052 cells. Biotechniques 33(4):760. 2–3
Chapter 5
Abstract
Bacterial lipoproteins are characterized by fatty acids that are covalently attached to their amino terminus
via posttranslational modification in the cytoplasmic membrane. Three enzymatic steps are involved in the
synthesis of mature triacylated lipoprotein: prolipoprotein converts into diacylglyceryl-prolipoprotein that
in turn converts into apolipoprotein, which is finally converted into mature triacylated lipoprotein. Here
we describe the detection of one of these intermediate forms of lipoprotein, diacylglyceryl-prolipoprotein,
using 3H-palmitate labeling and inhibition by globomycin and detection by fluorography.
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_5, © Springer Science+Business Media LLC 2017
75
76 Nienke Buddelmeijer
2 Materials
2.1 3H-Palmitate 1. Luria Broth Miller (LB) medium: 5 g yeast extract, 10 g peptone,
Labeling of E. coli 10 g NaCl per liter in ultrapure water. Sterilize by autoclaving
Cultures for 20 min at 120 °C.
2. E. coli strains (see Note 1).
3. 5 mCi/mL of 9,10-3H(N)-palmitate in ethanol. Specific activity:
30–60 Ci(1,11–2,22 TBq)/mmol.
4. Incubator.
2.2 Inhibition of Lsp 1. Luria Broth Miller (LB) medium: 5 g yeast extract, 10 g peptone,
by Globomycin 10 g NaCl per liter in ultrapure water. Sterilize by autoclaving
for 20 min at 120 °C.
2. E. coli strains (see Note 1).
3. Globomycin: use final concentration of 160 μg/mL for E. coli
K12 strains and 5 μg/mL for B strains (see Note 2).
4. Incubator.
2.4 Tris-Tricine Gel 1. Mini-gel caster system and SDS-polyacrylamide gel electro-
Electrophoresis phoresis (PAGE) migration apparatus.
2. Cathode buffer (top, 10×): 1 M Tris, 1 M Tricine, 1% SDS pH
8.25. Do not adjust pH.
3. Anode buffer (bottom, 10×): 1 M Tris–HCl, pH 8.9.
4. Gel buffer (3×): 3 M Tris, 1 M HCl, 0.3% SDS.
5. Acrylamide (49.5%T, 3%C or 6%C). Store at 4 °C.
6. Ammonium persulfate (APS): 10% solution in water. Store at
−20 °C.
7. N, N, N, N′-tetramethyl-ethylenediamine (TEMED). Store
at 4 °C.
8. SDS-PAGE running buffer: 25 mM Tris, 250 mM glycine,
0.1% SDS. Do not adjust pH.
9. SDS loading buffer (3×): 150 mM Tris–HCl, pH 6.8, 6% SDS,
0.3% bromophenol blue, 30% glycerol.
10. Water bath at 100 °C.
11. i-Butanol.
12. Vacuum gel-drying system.
13. Amplify solution.
14. X-ray film.
3 Methods
3.2 Inhibition of Lsp Add globomycin to the cell culture after 1 h of growth in the pres-
by Globomycin ence of 3H-palmitate and let cultures grow for an additional 1 h
(see Note 5).
3.4 Tris-Tricine Gel Tris-Tricine gels are especially useful for separating small proteins
Electrophoresis and peptides (less than 30 kDa) [8].
1. Prepare a mini-gel format separating gel (16%) by mixing
5 mL gel buffer, 6 mL acrylamide solution, and 4 mL water
(total volume 15 mL). Add 5 μL TEMED and 50 μL APS and
cast gel in an 8.6 × 6.8 × 0.75 cm gel holder. Allow space for
stacking gel and overlay with water or i-butanol.
2. Prepare stacking gel (4%) by mixing 3.3 mL gel buffer, 1 mL
acrylamide solution, and 5.7 mL water (total volume 10 mL).
Add 7.5 μL TEMED and 75 μL APS and cast gel and insert
comb immediately.
3.
Load samples on gel along with protein standard.
Electrophorese at 30 V until the samples have entered the
stacking gel and continue at 200 V till the dye front reaches
the bottom of the gel (see Note 9).
4. After migration of samples, pry gel plates open and briefly
wash gel in water.
5. Transfer to amplify solution and let soak for 10 min under
agitation.
6. Dry gel in gel dryer under vacuum for 60 min at 80 °C
(see Note 10).
7. Transfer the gel into a cassette and put an X-ray film over it.
Expose the film for 10 days at −80 °C. Let the cassette warm
up to room temperature before developing the X-ray film
(Fig. 1).
Lipoprotein Labeling 79
4 Notes
References
1. Kovacs-Simon A, Titball RW, Michell SL 6. Hantke K, Braun V (1973) Covalent binding
(2010) Lipoproteins of bacterial pathogens. of lipid to protein. Diglyceride and amide-
Infect Immun 79:548–561 linked fatty acid at the N-terminal end of the
2. Lai J-S, Philbrick WM, Wu HC (1980) Acyl murein-lipoprotein of the Escherichia coli outer
moieties in phospholipids are the precursors membrane. Eur J Biochem 34:284–296
for the fatty acids in murein lipoprotein in 7. Kumamoto CA, Gannon PM (1988) Effects of
Escherichia coli. J Biol Chem 255: Escherichia coli secB mutations on pre-maltose
5384–5387 binding protein conformation and export
3. Buddelmeijer N (2015) The molecular mecha- kinetics. J Biol Chem 263:11554–11558
nism of bacterial lipoprotein modification--how, 8. Schagger H (2006) Tricine-SDS-PAGE. Nat
when and why? FEMS Microbiol Rev Protoc 1:16–22
39:246–261 9. Kiho T et al (2004) Structure-activity relation-
4. Inukai M, Takeuchi K, Shimizu K, Arai M ships of globomycin analogues as antibiotics.
(1978) Mechanism of action of globomycin. Bioorg Med Chem 12:337–361
J Antibiot 31:1203–1205 10. Hussain M, Ichihara S, Mizushima S (1980)
5. Cronan JE Jr, Rock CO (1996) Biosynthesis of Accumulation of glyceride-containing precursor
membrane lipids. In: Neidhardt FC (ed) of the outer membrane lipoprotein in the cyto-
Escherichia coli and Salmonella: molecular and plasmic membrane of Escherichia coli treated with
cellular biology. ASM, Washington, DC globomycin. J Biol Chem 255:3707–3712
Chapter 6
Abstract
In many bacteria, membrane proteins account for around one-third of the proteome and can represent
much more than half of the mass of a membrane. Classic techniques in cell biology can be applied to char-
acterise bacterial membranes and their membrane protein constituents. Here we describe a protocol for
the purification of outer and inner membranes from Escherichia coli. The procedure can be applied with
minor modifications to other bacterial species, including those carrying capsular polysaccharide attached
to the outer membrane.
Key words Sucrose density gradient, Membrane biogenesis, Beta-barrel proteins, Cytoplasmic mem-
brane, Lipoproteins
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_6, © Springer Science+Business Media LLC 2017
81
82 Rhys A. Dunstan et al.
2 Materials
2.2 Sucrose Density 1. 5 mM EDTA, pH 7.5 (see earlier steps). Store at room
Fractionation temperature.
2. Ultra-pure sucrose.
3. Sucrose EDTA fractions: 35–60% (w/w) sucrose, 5 mM
EDTA, pH 7.5 solutions. For example, to make 50% (w/w),
add 15 g sucrose to 15 mL 5 mM EDTA. Pass through a 0.45
μM filter and store at 4 °C.
4. Displacing sucrose solution: 70% (w/w) sucrose, 5 mM EDTA,
pH 7.5 (add 140 g sucrose to 60 mL EDTA) (see Note 3).
5. Beckman Coulter SW 40 Ti tubes (disposable plastic tubes)
and rotor.
6. Density Gradient Fractionation System (Teledyne Isco, Lincoln,
NE, USA)
3 Methods
3.2 Sucrose Density 1. Immediately before use carefully prepare a six-step sucrose gra-
Fractionation dient from 60–35% w/w (1.9 mL of 60%, 55%, 50%, 45%,
40%, 35%) in SW40 tubes. A sharp interphase between layers
should be clearly visible (see Note 6).
2. Layer 400 μL of total membranes on top of a 60–35% w/w
gradient.
3. Spin in an ultracentrifuge using SW40 rotor for 17 h. at 34,000
rpm (205,000 × g), 4 °C (see Note 7).
4. Isolate 1 mL fractions (use ISCO fractionator with 70%
sucrose, 5 mM EDTA, pH 7.5 as displacing fluid, or carefully
pipette 1 mL fractions off top of gradient). Store each fraction
at −80 °C until use (see Note 8).
Membrane Isolation on Sucrose Gradients 85
3.3 Isolation 1. To isolate membranes from specific fractions, add TES buffer
of Membranes (to a final volume of ~8 mL) to each fraction of interest or to
after Density pooled fractions and pellet by ultracentrifugation, 1.5 h, 4 °C,
Fractionation 38,000 rpm (70 Ti .1 rotor).
2. Resuspend each fraction in ~100 μL 25% (w/w) sucrose, 5
mM EDTA, pH 7.5, with a dounce, and store membranes at
−80 °C (see Note 9).
4 Notes
9. The volume of TES used will vary with the amount of mem-
branes isolated or desired membrane concentration for later
experiments.
References
1. Dalbey RE, Wang P, Kuhn A (2011) Assembly D, Lithgow T (2012) Discovery of an arche-
of bacterial inner membrane proteins. Annu typal protein transport system in bacte-
Rev Biochem 80:161–187 rial outer membranes. Nat Struct Mol Biol
2. Okuda S, Tokuda H (2011) Lipoprotein 19(5):506–510
sorting in bacteria. Annu Rev Microbiol 11. Marchand CH, Salmeron C, Bou Raad R,
65:239–259 Meniche X, Chami M, Masi M, Blanot D,
3. Kamio Y, Nikaido H (1976) Outer mem- Daffe M, Tropis M, Huc E, Le Marechal P,
brane of salmonella typhimurium: acces- Decottignies P, Bayan N (2012) Biochemical
sibility of phospholipid head groups to disclosure of the mycolate outer membrane
phospholipase c and cyanogen bromide of Corynebacterium glutamicum. J Bacteriol
activated dextran in the external medium. 194(3):587–597
Biochemistry 15(12):2561–2570 12. Masson L, Holbein BE (1983) Physiology of
4. Smit J, Kamio Y, Nikaido H (1975) Outer sialic acid capsular polysaccharide synthesis in
membrane of salmonella typhimurium: serogroup B Neisseria meningitidis. J Bacteriol
chemical analysis and freeze-fracture studies 154(2):728–736
with lipopolysaccharide mutants. J Bacteriol 13. Siegmund-Schultze N, Kroll HP, Martin
124(2):942–958 HH, Nixdorff K (1991) Composition of the
5. Osborn MJ, Gander JE, Parisi E, Carson outer membrane of Proteus mirabilis in rela-
J (1972) Mechanism of assembly of the tion to serum sensitivity in progressive stages
outer membrane of salmonella typhimurium. of cell form defectiveness. J Gen Microbiol
Isolation and characterization of cytoplas- 137(12):2753–2759
mic and outer membrane. J Biol Chem 14. Jagannadham MV, Abou-Eladab EF, Kulkarni
247(12):3962–3972 HM (2011) Identification of outer mem-
6. Schnaitman CA (1970) Protein composition brane proteins from an Antarctic bacte-
of the cell wall and cytoplasmic membrane of rium Pseudomonas syringae Lz4W. Mol Cell
Escherichia coli. J Bacteriol 104(2):890–901 Proteomics 10(6):M110 004549
7. Yamato I, Anraku Y, Hirosawa K (1975) 15. Bishop RE, Gibbons HS, Guina T, Trent MS,
Cytoplasmic membrane vesicles of Escherichia Miller SI, Raetz CR (2000) Transfer of pal-
coli. A simple method for preparing the cyto- mitate from phospholipids to lipid a in outer
plasmic and outer membranes. J Biochem membranes of gram-negative bacteria. EMBO
77(4):705–718 J 19(19):5071–5080
8. Hobb RI, Fields JA, Burns CM, Thompson 16. Charlson ES, Werner JN, Misra R (2006)
SA (2009) Evaluation of procedures for outer Differential effects of yfgL mutation on
membrane isolation from Campylobacter Escherichia coli outer membrane pro-
jejuni. Microbiology 155(Pt 3):979–988 teins and lipopolysaccharide. J Bacteriol
9. Anwari K, Webb CT, Poggio S, Perry AJ, 188(20):7186–7194
Belousoff M, Celik N, Ramm G, Lovering 17. Steeghs L, de Cock H, Evers E, Zomer B,
A, Sockett RE, Smit J, Jacobs-Wagner C, Tommassen J, van der Ley P (2001) Outer
Lithgow T (2012) The evolution of new membrane composition of a lipopolysaccharide-
lipoprotein subunits of the bacterial outer deficient Neisseria meningitidis mutant.
membrane BAM complex. Mol Microbiol EMBO J 20(24):6937–6945
84(5):832–844 18. Dunstan RA, Hay ID, Wilksch JJ, Schittenhelm
10. Selkrig J, Mosbahi K, Webb CT, Belousoff RB, Purcell AW, Clark J, Costin A, Ramm G,
MJ, Perry AJ, Wells TJ, Morris F, Leyton Strugnell RA, Lithgow T (2015) Assembly of
DL, Totsika M, Phan MD, Celik N, Kelly M, the secretion pores GspD, Wza and CsgG into
Oates C, Hartland EL, Robins-Browne RM, bacterial outer membranes does not require
Ramarathinam SH, Purcell AW, Schembri the Omp85 proteins BamA or TamA. Mol
MA, Strugnell RA, Henderson IR, Walker Microbiol 97(4):616–629
Chapter 7
Abstract
Surface-exposed proteins of Gram-negative bacteria are represented by integral outer membrane beta-
barrel proteins and lipoproteins. No computational methods exist for predicting surface-exposed lipopro-
teins, and therefore lipoprotein topology must be experimentally tested. This chapter describes three
distinct but complementary methods for the detection of surface-exposed proteins: cell surface protein
labeling, accessibility to extracellular protease and antibodies.
Key words Biotinylation, PEGylation, Surface proteolysis, Whole-cell dot blot, Protein topology
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_7, © Springer Science+Business Media LLC 2017
87
88 Anna Konovalova
Table 1
Protein labeling reagents for selective surface labeling
Functional group to
be labeled Name Polar Molecular weight, Da
Primary amine NHS-LC-LC-Biotin − 567.70
NHS-PEG(n)-Biotin + Available in 1–10
kDa range
Sulfhydryl Mal-PEG(n)-Biotin + Available in 1–10
kDa range
Cell Surface Exposure 89
2 Materials
2.1 Cell Surface 1. NHS reagents (see Table 1 for product information). NHS
Labeling Based reagents are moisture sensitive. Store desiccated at 4 °C, and
on Modification equilibrate to room temperature (RT) before opening. Prepare
of Primary Amines 25 mM stock solution according to product instructions
immediately before use.
2. Labeling buffer containing no primary amines, such as phos-
phate buffered saline (PBS): 10 mM Na2HPO4, 1.8 mM
KH2PO4, 2.7 mM KCl, 137 mM NaCl, pH 8.0.
3. Quenching solution: 1 M glycine or 1 M Tris–HCl, pH 8.0.
2.2 Cell Surface 1. Maleimide reagents (see Table 1 for product information).
Labeling Based Maleimide reagents are moisture sensitive. Store desiccated at
on Modification 4 °C, and equilibrate to RT before opening. Prepare 25 mM
of Sulfhydryls stock solution according to product instructions immediately
before use.
2. Labeling buffer containing no sulfhydryls, such as Tris-buffered
saline (TBS): 50 mM Tris–HCl, 150 mM NaCl, pH 7.0 or
PBS: 10 mM Na2HPO4, 1.8 mM KH2PO4, 2.7 mM KCl, 137
mM NaCl, pH 7.0.
3. Tris(2-carboxyethyl)phosphine (TCEP) solution: 500 mM
(optional).
3 Methods
3.1 Cell Surface This protocol is valid for any NHS-based reagent.
Labeling Based
on Modification 1. Collect exponentially growing cells by centrifugation (see Note 1).
of Primary Amines 2. Wash cells three times with ice-cold PBS to remove amine-
containing culture medium.
3. Resuspend cells to 1010 cells/mL.
4. Add NHS reagent to a final concentration of 2.5 mM.
5. Incubate at RT for 30 min.
6. Add one-tenth of the volume of quenching solution.
7. Collect the cells by centrifugation.
8. Wash cells twice with PBS supplemented with 100 mM glycine
or directly in 100 mM Tris–HCl to quench and remove excess
reagent.
9. Analyze by immunoblotting or follow with protein purification
if needed (see Note 2).
3.2 Cell Surface This protocol is valid for any maleimide-based reagent.
Labeling Based
on Modification 1.
Collect exponentially growing cells by centrifugation
of Sulfhydryls (see Note 1).
2. Wash cells three times with ice-cold TBS or PBS.
3. Resuspend cells to 1010 cells/mL.
4. (Optional) If protein contains oxidized (disulfide bonded) cys-
teines, treat cells with 5 mM TCEP in TBS or PBS, pH 7.0, for
30 min at RT. Wash cells twice with TBS or PBS to remove
excess TCEP (see Note 3).
Cell Surface Exposure 93
4 Notes
References
1. Silhavy TJ, Kahne D, Walker S (2010) The 6. Freeman TC Jr, Wimley WC (2010) A highly
bacterial cell envelope. Cold Spring Harb accurate statistical approach for the prediction
Perspect Biol 2(5):a000414 of transmembrane beta-barrels. Bioinformatics
2. Okuda S, Tokuda H (2011) Lipoprotein sort- 26(16):1965–1974
ing in bacteria. Annu Rev Microbiol 7. Singh NK, Goodman A, Walter P, Helms V,
65:239–259 Hayat S (2011) TMBHMM: a frequency pro-
3. Zuckert WR (2014) Secretion of bacterial lipo- file based HMM for predicting the topology
proteins: through the cytoplasmic membrane, of transmembrane beta barrel proteins and
the periplasm and beyond. Biochim Biophys the exposure status of transmembrane resi-
Acta 1843(8):1509–1516 dues. Biochim Biophys Acta
4. Konovalova A, Silhavy TJ (2015) Outer mem- 1814(5):664–670
brane lipoprotein biogenesis: lol is not the end. 8. Hayat S, Elofsson A (2012) BOCTOPUS:
Philos Trans R Soc Lond Ser B Biol Sci improved topology prediction of transmem-
370(1679) brane beta barrel proteins. Bioinformatics
5. Wilson MM, Bernstein HD (2015) Surface- 28(4):516–522
exposed lipoproteins: an emerging secretion 9. Hermanson GT (2013) Bioconjugate tech-
phenomenon in gram-negative bacteria. Trends niques, 3rd edn. Academic press, London,
Microbiol pp 1–1146
Cell Surface Exposure 95
10. Nikaido H (2003) Molecular basis of bacterial 18. Hilz H, Wiegers U, Adamietz P (1975)
outer membrane permeability revisited. Stimulation of proteinase K action by denatur-
Microbiol Mol Biol Rev 67(4):593–656 ing agents: application to the isolation of
11. Konovalova A, Perlman DH, Cowles CE, nucleic acids and the degradation of 'masked'
Silhavy TJ (2014) Transmembrane domain of proteins. Eur J Biochem 56(1):103–108
surface-exposed outer membrane lipoprotein 19. Pinne M, Haake D (2011) Immuno-
RcsF is threaded through the lumen of fluorescence assay of leptospiral surface-exposed
beta-barrel proteins. Proc Natl Acad Sci U S A proteins. J Vis Exp 53
111(41):E4350–E4358 20. Blom K, Lundin BS, Bolin I, Svennerholm A
12. Cowles CE, Li Y, Semmelhack MF, Cristea IM, (2001) Flow cytometric analysis of the localiza-
Silhavy TJ (2011) The free and bound forms of tion of helicobacter pylori antigens during dif-
Lpp occupy distinct subcellular locations in ferent growth phases. FEMS Immunol Med
Escherichia coli. Mol Microbiol Microbiol 30(3):173–179
79(5):1168–1181 21. Matsunaga J, Werneid K, Zuerner RL, Frank
13. Rosenbusch JP (1990) Structural and func- A, Haake DA (2006) LipL46 is a novel surface-
tional properties of porin channels in E. coli exposed lipoprotein expressed during lepto-
outer membranes. Experientia 46(2): spiral dissemination in the mammalian host.
167–173 Microbiology 152(Pt 12):3777–3786
14. Wilson MM, Anderson DE, Bernstein HD 22. Moeck GS, Bazzaz BS, Gras MF, Ravi TS,
(2015) Analysis of the outer membrane pro- Ratcliffe MJ, Coulton JW (1994) Genetic
teome and secretome of Bacteroides fragilis insertion and exposure of a reporter epitope in
reveals a multiplicity of secretion mechanisms. the ferrichrome-iron receptor of Escherichia
PLoS One 10(2):e0117732 coli K-12. J Bacteriol 176(14):4250–4259
15. Pugsley AP, Kornacker MG, Ryter A (1990) 23. Newton SM, Klebba PE, Michel V, Hofnung
Analysis of the subcellular location of pullula- M, Charbit A (1996) Topology of the mem-
nase produced by Escherichia coli carrying the brane protein LamB by epitope tagging and a
pulA gene from Klebsiella pneumoniae strain comparison with the X-ray model. J Bacteriol
UNF5023. Mol Microbiol 4(1):59–72 178(12):3447–3456
16. Pinne M, Haake DA (2009) A comprehensive 24. Schnell U, Dijk F, Sjollema KA, Giepmans BN
approach to identification of surface-exposed, (2012) Immunolabeling artifacts and the need
outer membrane-spanning proteins of for live-cell imaging. Nat Methods 9(2):
Leptospira interrogans. PLoS One 4(6):e6071 152–158
17. Porter WH, Preston JL (1975) Retention of 25. Dinh T, Bernhardt TG (2011) Using
trypsin and chymotrypsin proteolytic activity in s
uperfolder green fluorescent protein for
sodium dodecyl sulfate solutions. Anal periplasmic protein localization studies.
Biochem 66(1):69–77 J Bacteriol 193(18):4984–4987
Chapter 8
Abstract
Inner membrane proteins are inserted into the membrane via α-helices. These helices do not only consti-
tute membrane anchors but may mediate specific interactions with membrane protein partners or partici-
pate in energetic processes. The number, location, and orientation of these helices is referred to as topology.
Bitopic membrane proteins that consist of a single membrane-embedded domain connecting two soluble
domains are distinguished from polytopic ones that consist of multiple membrane-spanning helices con-
nected by extramembrane domains. Defining inner membrane protein topology could be achieved by
different methods. Here we describe a protease accessibility assay that makes it possible to define topology
based on digestion profiles.
Key words Membrane protein, Inner membrane, Insertion, Topology, Transmembrane segment,
Bitopic, Polytopic, Proteolysis, Protease, Proteinase K, Carboxypeptidase Y
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_8, © Springer Science+Business Media LLC 2017
97
98 Maxence S. Vincent and Eric Cascales
Fig. 1 Nomenclature of inner membrane proteins with selected topologies. Shown are the topologies of a
bitopic protein with an N-terminal TMH (a), a bitopic protein with a C-terminal TMH (b) and polytopic proteins
with different numbers of TMH (c). Representative examples of inner membrane (IM) proteins with these
topologies associated with bacterial secretion systems are listed. For polytopic proteins, the number of trans-
membrane segments are indicated in brackets
2 Material
2.1 Cell Growth 1. Lysogeny broth (LB) or recommended medium to grow the
and Spheroplast strain of interest.
Preparation 2. TNS buffer: 20 mM Tris–HCl, pH 8.0, 100 mM NaCl, 30%
sucrose: Dissolve 0.243 g Tris(hydroxymethyl) aminometh-
ane, 0.684 g NaCl, and 30 g sucrose in sterile distilled water
(final volume of 100 mL). Adjust pH to 8.0 with 1 M HCl.
3. TN buffer: 20 mM Tris–HCl, pH 8.0, 100 mM NaCl: Dissolve
0.243 g Tris(hydroxymethyl) aminomethane and 0.684 g
NaCl in 100 mL sterile distilled water. Adjust pH to 8.0 with
1 M HCl.
4. 0.5 M ethylenediaminetetraacetic acid (EDTA), pH 8.0:
Dissolve 1.86 g EDTA (disodium salt) in 10 mL sterile dis-
tilled water. Adjust pH to 8.0 with 10 M NaOH.
5. Lysozyme stock solution (100×), 10 mg/mL lysozyme:
Dissolve 10 mg goose egg lysozyme in 1 mL sterile distilled
water. Store at −20 °C.
6. Incubator.
7. Spectrophotometer to measure bacterial density.
8. Labtop centrifuge.
2.2 Protease 1. Triton X-100 stock solution, 10% Triton X-100: Mix 1 mL 100%
Accessibility Assay Triton X-100 with 9 mL sterile distilled water (see Note 1). Store
at room temperature.
2. Carboxypeptidase Y stock solution (100×), 10 mg/mL car-
boxypeptidase Y: Dissolve 10 mg purified carboxypeptidase Y
in 1 mL sterile distilled water. Store at −20 °C.
3. Proteinase K stock solution (100×), 10 mg/mL proteinase K:
Dissolve 10 mg purified proteinase K in 1 mL sterile distilled
water. Store at −20 °C.
4. Cocktail of protease inhibitors (Complete, F. Hoffmann-La
Roche AG, Basel, Switzerland, or equivalent).
5. Phenylmethylsulfonyl fluoride (PMSF) stock solution (100×),
100 mM PMSF: Dissolve 17.4 mg PMSF in 1 mL absolute
ethanol (see Note 2). Store at −20 °C.
6. 50% trichloroacetic acid (TCA) solution: Dissolve 50 g TCA
in 30 mL distilled water. Complete to 100 mL with distilled
water (see Note 3).
7. Acetone.
8. Vortex.
100 Maxence S. Vincent and Eric Cascales
3 Method
3.2 Protease 1. Divide cell suspension into five samples, numbered 1–5.
Accessibility Sample 1 will remain untreated.
(See Notes 8 and 9) 2. Add 1% (final concentration) of Triton X-100 in samples 3 and
5 to lyse spheroplasts (see Note 9). Mix by vortexing and incu-
bation 10 min on ice.
3. Add carboxypeptidase Y (100 μg/mL final concentration from
10 mg/mL stock solution) in tubes 2 and 3. Incubate for
30 min on ice.
4. Add proteinase K (100 μg/mL final concentration from 10
mg/mL stock solution) in tubes 4 and 5. Incubate for 30 min
on ice.
5. Quench the proteolysis reaction by adding PMSF and inhibi-
tor cocktail in tubes 1–5. Incubate for 5 min on ice.
Protease Accessibility 101
6. Add 0.5 volume 50% TCA in tubes 1–5. Mix by vortexing and
incubate for 20 min on ice.
7. Collect precipitated material by centrifugation at 20,000 × g
for 20 min at 4 °C.
8. Discard supernatant and add 500 μL acetone. Vortex.
9. Collect precipitated proteins by centrifugation at 20,000 × g
for 20 min at 4 °C.
10. Discard supernatant and keep tubes open until pellet is dry
(see Note 10).
Fig. 2 Schematic representation of expected results. The expected immunoblot results for inner membrane
proteins with the topology shown below are schematically represented. Samples 1–5 are shown (1, untreated
sample; 2, carboxypeptidase Y; 3, carboxypeptidase Y on Triton X-100-lysed spheroplasts; 4, proteinase K; 5,
proteinase K on Triton X-100-lysed spheroplasts). The representation of the protein degradation products cor-
responding to the immunodetection bands are shown on the right of each “blot”
102 Maxence S. Vincent and Eric Cascales
4 Notes
Acknowledgements
References
Abstract
A described simple and advanced protocol for the substituted-cysteine accessibility method as applied to
transmembrane (TM) orientation (SCAM™) permits a topology analysis of proteins in their native state
and can be universally adapted to any membrane system to either systematically map an uniform topology
or identify and quantify the degree of mixed topology. In this approach, noncritical individual amino acids
that are thought to reside in the putative extracellular or intracellular loops of a membrane protein are
replaced one at a time by cysteine residue, and the orientation with respect to the membrane is evaluated
using a pair of membrane-impermeable nondetectable and detectable thiol-reactive labeling reagents.
1 Introduction
1.1 Membrane It has been estimated that the vast majority of membrane proteins
Protein Topology adopt α-helical bundle (Fig. 1a) structure, which contains trans-
and Topogenesis membrane domains (TMDs) that span the membrane in a zigzag
but not in a “one-by-one” fashion (Fig. 1b). A fundamental aspect
and primary structural element of the structure of integral mem-
brane proteins is membrane protein topology. Membrane protein
topology refers to the two-dimensional structural information of a
membrane protein and describes the way a polypeptide chain is
arranged in the membrane, i.e., the number of TMDs and their
orientation in the membrane that indicates the sidedness of extra-
membrane domains (EMDs) [1–4]. Although the topology of
membrane proteins provides low-resolution structural information,
it can be a starting point for different biochemical experiments or
modeling of three-dimensional structures. Membrane protein
topology and assembly are governed by structural principles and
topological rules and directed by topogenic signals and sequences
in the nascent polypeptide chain that are recognized and decoded
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_9, © Springer Science+Business Media LLC 2017
105
106 Mikhail Bogdanov
1.3 Justifying The reasons for the broad application of SCAM™ are both concep-
SCAM™ Legacy tual and practical. The strategic use of SCAM™ for mapping mem-
and Advantages brane topology has made it possible to circumvent the many
limitations of alternative methods used to map a topology of inte-
gral membrane proteins:
1. Since only single amino acid changes are made, cysteine chem-
istry has the highest resolution in that water accessibility of
individual cysteine residues can be determined.
2. Cysteine has no or little preference for a particular secondary
structure. Owing to only minmal changes in the primary
sequence, structural perturbation of introduced cysteine muta-
tions is essentially absent or much milder than in other meth-
ods commonly used to determine topology.
3. Detection of engineered cysteine modifications is simple, and
analysis is done by chemical modification using a broad range
of commercially available reagents that differ in charge, size,
mass, and hydrophilicity.
4. These reagents can be used to detect protein sulfhydryl groups
with sensitivities in the femtomole range.
5. Chemical modification can be done using intact cells.
Determination of Membrane Protein Topology 109
1.4 Application In most cases SCAM™ provides topological information after the
of SCAM™ orientation of proteins within membranes is established [5, 19].
The application of this approach allowed either detailed mapping
or significant refinement of the topology of a variety of integral
membrane proteins, including a more accurate mapping of the
ends of TMDs of protein topology that was been established by
other methods [12–14]. However SCAM™ is not only an alterna-
tive approach to low-resolution determination of membrane pro-
tein structure; it also constitutes an attractive independent approach
to dynamic studies of membrane proteins. The dynamic aspects of
protein structure as a function of the physiological state of a cell is
best probed in whole cells or membranes. SCAM™ has been used
to monitor dynamic conformational and topological changes
accompanied by substrate binding and release during enzyme
turnover and function [25, 26]. The labeling of single-cysteine
replacements of a major component (SecG) of the SecYEG trans-
locon with 4-acetamido-4′-maleimidylstilbene-2,2′-disulfonic acid
(AMS) either at rest or during adenosine triphosphate (ATP)-
dependent preprotein translocation clearly demonstrated that a
cytoplasmic region of SecG undergoes topology inversion [27].
Although the labeling patterns derived from SCAM™ assays
usually reflect a steady-state topology of a membrane protein,
semiquantitative analysis of the surface accessibility of individual
cysteines introduced into extramembrane loops can be carried out
at various stages of protein assembly. In this case, SCAM™ can be
used to provide topological information during membrane inser-
tion, folding, and assembly of proteins. Cysteine accessibility dur-
ing bacteriorhodopsin translation was monitored by pulse-chase
radiolabeling and modification by AMS to determine the order
and timing of insertion of TM segments into the membrane of
Halobacterium salinarum [28]. In this in vivo assay, the rate of
insertion of TMDs into the H. salinarum cytoplasmic membrane
was monitored by rapid modification of unique cysteines in extra-
cellular EMDs of the protein with AMS, resulting in a shift in
mobility of the protein in sodium dodecyl sulfate (SDS)-
polyacrylamide gel electrophoresis (PAGE). SCAM™ has also been
utilized to establish a packing geometry of a pilin VirB2 subunit
and its ATP-dependent in and out of membrane dynamics and
reorganization within the T-pilus and T4SS secretion channel [29].
110 Mikhail Bogdanov
1.5 Overview This approach is based on the introduction of cysteine residues one
and General Rationale at a time in the putative EMDs of otherwise cysteine-less mem-
of Topology Mapping brane proteins of interest followed by chemical modification with
Using SCAM™ a membrane-impermeable thiol-specific probes either before or
after compromising cell membrane integrity to determine cysteine
1.5.1 SCAM™ membrane sidedness. Accessibility in whole cells establishes extra-
cellular location, while accessibility only after cell disruption estab-
lishes intracellular location (see Notes 1 and 2). The accessibility of
EMDs flanking a TMD then establishes the orientation of the
TMD with respect to the plane of the membrane bilayer.
1.5.3 Mutation Strategy, The pre-requisite for the method is the generation of the cysteine-
Host and Vector Selections, free template of a protein of interest. The expression of a func-
Construction of Plasmids tional “cysteine-less” template has permitted the use of scanning
Expressing Single-Cysteine cysteine mutagenesis and thiol modification techniques for map-
Derivatives ping membrane topology. All amino acid substitutions should be
verified by DNA sequencing and functional analysis and expres-
sion level by Western blotting of each derivative should be carried
out if possible. Ideally target gene expression should be under
control of an inducible promoter such as OPtac or repressor (TetR)
regulating the promoter (PLtetO-1) to minimize a drastic overex-
pression or continuous expression of potentially disruptive gene
products (see Note 3).
A prerequisite for each cysteine replacement is retention of
function that provides assurance of retention of near native struc-
ture. The native cysteine residues are usually changed into alanine
or serine residues which are small, commonly found in membrane
proteins and appear to be tolerated at most positions thus render-
ing an active protein. A residue that does not tolerate substitution
by Cys makes a crucial contribution to maintaining the structure of
the site, and/or to the folding and function of target protein.
Replacement of charged residues is generally not advised because
these have a high probability of being topogenic signals or may be
involved in long-range charge-charge intramolecular or intermo-
lecular (lipid-protein) interactions. If the protein contains stretches
of residues of intermediate hydrophobicity that cannot unambigu-
ously be identified as membrane spanning TMD, substitutions
should be made approximately every ten residues. The cysteine-
less protein serves also as a negative labeling control to assure that
residues such as lysine and histidine are not labeled by the reagents
(see Note 1). Ideally the protein under study should be devoid of
all native cysteine residues because these residues may also react
Determination of Membrane Protein Topology 113
1.5.4 Cell Growth Cells are first grown overnight at 37 °C in Luria–Bertani (LB)
and Regulated Expression medium supplemented with appropriate antibiotic and then sub-
of Single-Cysteine cultured in the morning to an OD600 of around 0.05 in the LB
Derivatives medium supplemented with appropriate antibiotic to maintain
plasmid-encoding single-cysteine replacements in cysteineless pro-
tein. Carrying plasmids encoding target protein under appropriate
promoters control are induced usually by growth in the presence
of inducer for at least several generations (OD600 reading ~0.5–
0.6) to reach a logarithmic growth phase of cells.
1.5.5 General Protocol Following the expression of monocysteine mutants, cells are sub-
for SCAM™ jected to SCAM™ analysis by in vivo labeling with different detect-
able and nondetectable thiol-reactive reagents [5, 9].
Maleimide-based thiol reagents, which are available in a wide vari-
ety of forms, are particularly suited for SCAM™ [5]. Maleimide
reacts with the ionized form of a thiol group (thiolate anion)
(Fig. 2a), and this reaction requires a water molecule as a proton
acceptor. In most cases the unreactive cysteine residues are located
within the membrane hydrophobic core or in a sterically hindered
environment [5]. The reaction rate of different thiols is controlled
primarily by their surface exposure and proximal environment.
Most experiments utilize biotinylated, radioactive, fluorescent [5],
114 Mikhail Bogdanov
Fig. 3 General strategy for SCAM™ using impermeable MPB and impermeant transparent AMS to probe the
sidedness of EMDs. A target membrane protein (only one TMD hairpin is shown) containing a single cysteine
exposed either to the extracellular (blue, periplasmic) or intracellular (red, cytoplasmic) side of the membrane
is expressed in host cells. Half of the cells are reacted with a detectable thiol reagent MPB to specifically label
the externally exposed cysteine (first row from top) and the other half are reacted with a nondetectable thiol
reagent AMS (third and fourth rows) to protect external cysteines in subsequent labeling steps. Both halves of
the cells are either kept intact (−) or disintegrated (+) by sonication to expose and label previously inaccesible
cytoplasmic cysteine (second and fourth rows). Labeling by MPB that can be blocked completely by pretreatment
with AMS is independent indicative of a periplasm-facing residue (third row). Labeling by MPB that cannot be
blocked by such AMS treatment is independent indicative of a residue that is facing the cytoplasmn (fourth
row). A target protein was immunoprecipitated and resolved by SDS-PAGE, and biotinylated protein was
detected using avidin-HRP and chemiluminescence (right panel)
disrupted cells for the same protein. This approach simplifies the
interpretation of data obtained with a series of protein derivatives
that may express at different levels since conclusions about topol-
ogy are based on the relative reactivity of cysteines in the same
sample before and after cell disruption. Since sample pairs are ana-
lyzed on the same western blot, no signal intensity normalization
is required. The level of expression of any given derivative will
affect the absolute intensity of labeling but not the ratio of the
labeling between the sample pairs.
Although central to the method is the use of detectable thiol-
specific reagents to differentiate intracellular from extracellular
EMDs, confirmation of labeling of external water-exposed cyste-
ines by MPB can be achieved by first blocking putative external
cysteines in intact cells with a thiol-specific reagent that is transpar-
ent in the detection phase of the procedure. Such a preblocking
step also allows selective labeling of luminal (exposed to
Determination of Membrane Protein Topology 117
2 Materials
3 Methods
3.5 Data Analysis The criteria used for determining the location of an introduced
and Interpretation cysteine are as follows. Labeling of a cysteine residue with a
membrane-impermeable sulfhydryl reagent before disruption of
Determination of Membrane Protein Topology 123
4 Notes
during labeling will favor the reaction [5, 9, 10, 30] (Fig. 2a).
However, maleimides are known to react also with primary
amines at pH values above 7.5. An effective control to rule out
nonthiol residue modification is to use a cysteineless template
of the target protein. Unfavorable orientation of a thiol group
owing to a local secondary structure may restrict or prevent
access by large thiol reagents. Increasing the reaction buffer
pH would not only favor alkylation of an extramembrane cys-
teine but also disrupt the local restrictive secondary structure
while truly membrane-embedded cysteines would not be
expected to react. EMDs that are sterically hindered or exhibit
an elevated pKa can be derivatized by increasing the pH up to
10.5 without compromising membrane integrity [10, 30].
However, appropriate controls, such as making known cyto-
plasmically exposed domains inaccessible and removing any
cysteineless target protein label, should be used.
2. Conclusions based on the full reactivity of diagnostic residues
should also be drawn with caution since cysteine residues fac-
ing a hydrophilic pore or near a substrate-binding site may be
within a TM segment but chemically reactive because of water
channels or pockets.
3. The araB system suffers from cell-to-cell heterogeneity when
expression levels vary greatly within the cell population,
thereby making physiological interpretations problematic
because of the induction of promoter yielded mixed popula-
tions of uninduced and fully induced cells. Contrary PLtetO-1
is tightly repressible by the Tet repressor in the absence of
inducer and can be induced with the nontoxic inducer aTc,
which has an increased affinity for repressor TetR and thus can
provide tight and homogeneous cell-to-cell expression.
Overexpression of membrane proteins under the control of
araB or T7 promoter is not recommended or should be carried
out with cation. If the araB system is still utilized a short induc-
tion time (1 h) at a low concetration (0.05%) of arabinose is
recommended. The pET expression systems is widely used
because of its ability to produce large quantities of a desired
protein when activated. However, the use of a pET vector in
SCAM™ is not recommended for the expression of target
membrane proteins because the overloading of cells with a
highly overexpressed target protein can “jam” a translocon and
trigger the accumulation of membrane protein into newly
made intracytoplasmic membranes (Lu, Zheng, and Bogdanov,
unpublished).
4. SCAM™ is based on the controlled membrane permeability of
sulfhydryl reagents. Membranes, either in their native state or
due to experimental manipulation, can be slightly permeable
to labeling reagents. Therefore, optimal labeling conditions
must be established for each reagent and host. The results of
Determination of Membrane Protein Topology 125
References
27. Nagamori S, Nishiyama K, Tokuda H (2002) 35. Dobson L, Remenyi I, Tusnady GE (2015)
Membrane topology inversion of SecG CCTOP: a consensus constrained TOPology
detected by labeling with a membrane- prediction web server. Nucleic Acids Res
impermeable sulfhydryl reagent that causes a 43:W408–W412
close association of SecG with SecA. J Biochem 36. Bayer EA, Zalis MG, Wilchek M (1985)
132:629–634 3-(N-Maleimido-propionyl)biocytin: a versa-
28. Dale H, Angevine CM, Krebs MP (2000) tile thiol-specific biotinylating reagent. Anal
Ordered membrane insertion of an archaeal Biochem 149:529–536
opsin in vivo. Proc Natl Acad Sci U S A 37. Berezuk AM, Goodyear M, Khursigara CM
97:7847–7852 (2014) Site-directed fluorescence labeling
29. Kerr JE, Christie PJ (2010) Evidence for reveals a revised N-terminal membrane topol-
VirB4-mediated dislocation of membrane- ogy and functional periplasmic residues in the
integrated VirB2 pilin during biogenesis of the Escherichia coli cell division protein FtsK. J Biol
agrobacterium VirB/VirD4 type IV secretion Chem 289:23287–23301
system. J Bacteriol 192:4923–4934 38. Moss K, Helm A, Lu Y, Bragin A, Skach WR
30. Xie J, Bogdanov M, Heacock P, Dowhan W (1998) Coupled translocation events generate
(2006) Phosphatidylethanolamine and mono- topological heterogeneity at the endoplasmic
glucosyldiacylglycerol are interchangeable in reticulum membrane. Mol Biol Cell
supporting topogenesis and function of the 9:2681–2697
polytopic membrane protein lactose permease. 39. Woodall NB, Yin Y, Bowie JU (2015) Dual-
J Biol Chem 281:19172–19178 topology insertion of a dual-topology mem-
31. Zhang W, Bogdanov M, Pi J, Pittard AJ, brane protein. Nat Commun 6:8099
Dowhan W (2003) Reversible topological 40. Gafvelin G, von Heijne G (1994) Topological
organization within a polytopic membrane “frustration” in multispanning E. coli inner
protein is governed by a change in membrane membrane proteins. Cell 77:401–412
phospholipid composition. J Biol Chem 41. Nasie I, Steiner-Mordoch S, Schuldiner S
278:50128–50135 (2013) Topology determination of untagged
32. Wang X, Bogdanov M, Dowhan W (2002) membrane proteins. Methods Mol Biol
Topology of polytopic membrane protein sub- 1033:121–130
domains is dictated by membrane phospholipid 42. Gelis-Jeanvoine S, Lory S, Oberto J,
composition. EMBO J 21:5673–5681 Buddelmeijer N (2015) Residues located on
33. Bernsel A, Viklund H, Falk J, Lindahl E, von membrane-embedded flexible loops are essen-
Heijne G, Elofsson A (2008) Prediction of tial for the second step of the apolipoprotein
membrane-protein topology from first princi- N-acyltransferase reaction. Mol Microbiol
ples. Proc Natl Acad Sci U S A 105: 95:692–705
7177–7181 43. Liu Y, Basu A, Li X, Fliegel L (2015)
34. Zhao G, London E (2006) An amino acid Topological analysis of the Na+/H+ exchanger.
“transmembrane tendency” scale that Biochim Biophys Acta 1848:2385–2393
approaches the theoretical limit to accuracy for 44. Abramson J, Smirnova I, Kasho V, Verner G,
prediction of transmembrane helices: Kaback HR, Iwata S (2003) Structure and
relationship to biological hydrophobicity.
mechanism of the lactose permease of
Protein Sci 15:1987–2001 Escherichia coli. Science 301:610–615
Chapter 10
Abstract
Experimental determination of membrane protein topology can be achieved using various techniques. Here
we present the pho-lac dual reporter system, a simple, convenient, and reliable tool to analyze the topology
of membrane proteins in vivo. The system is based on the use of two topological markers with complemen-
tary properties, the Escherichia coli β-galactosidase LacZ, which is active in the cytoplasm, and the E. coli
alkaline phosphatase PhoA, which is active in the bacterial periplasm. Specifically, in this pho-lac gene system,
the reporter molecule is a chimera composed of the mature PhoA that is in frame with the β-galactosidase
α-peptide, LacZα. Hence, when targeted to the periplasm, the PhoA-LacZα dual reporter displays high
alkaline phosphatase activity but no β-galactosidase activity. Conversely, when located in the cytoplasm,
PhoA-LacZα has no phosphatase activity but exhibits high β-galactosidase activity in E. coli cells expressing
the ω fragment of LacZ, LacZω (via the α-complementation phenomenon). The dual nature of the PhoA-
LacZα reporter allows a simple way to normalize both enzymatic activities to obtain readily interpretable
information about the subcellular location of the fusion site between the membrane protein under study
and the reporter. In addition, the PhoA-LacZα reporter permits utilization of dual-indicator agar plates to
easily discriminate between colonies bearing cytoplasmic fusions, periplasmic fusions, or out- of-frame
fusions. In total, the phoA-lacZα fusion reporter approach is a straightforward and rather inexpensive
method of characterizing the topology of membrane proteins in vivo.
Key words Membrane proteins, Membrane topology, Dual reporter system, Phosphatase,
β-galactosidase
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_10, © Springer Science+Business Media LLC 2017
129
130 Gouzel Karimova and Daniel Ladant
2 Materials
3 Methods
3.2 C-Terminal 1. Amplify by PCR the DNA fragments carrying the predeter-
Fusion Approach mined 3′-truncated target protein gene with specific PCR
primers harboring appropriate restriction sites for subcloning
in pKTop plasmid. These PCR primers should be designed in
such a way that the amplified DNA fragment is fused in frame
to the downstream phoA-lacZα reporter gene (i.e., to generate
the expected C-terminal translation fusion).
2. Digest the PCR-amplified DNA fragments with appropriate
restriction enzymes.
3. Ligate the digested PCR-amplified DNA fragments into the
phoA-lacZα reporter vector linearized with the same restric-
tion enzymes.
4. Transform the ligation mixture into E. coli competent cells
(see Notes 3, 4, and 12).
pho-lac Reporter Fusions 135
(1)
(2)
(3)
(4)
(B) (C)
100
C
D8
Arbitary units(%)
Q72
75
M21 50
E57
25
K32
N48
E39 0
Fusion point: D8 M21 K32 E39 N48 L57 Q72
NAR
(PhoA/LacZ): 0.17 1.3 3.0 2.5 2.0 0.5 0.2
Location: cyt tm1 per per tm2 tm2 cyt
Fig. 2 YmgF topology analysis. (a) In silico predicted topological models of YmgF. Predictions were made using
four different methods: (1) PSIPRED, (2) Topcons, (3) Phobius, and (4) TopPred. The red lines indicate the cytosolic
domains, light blue TMSs (TMS1 and TMS2), and the deep blue line the periplasmic part. The small black arrow-
heads at the top of the sequences indicate the positions of the different sites selected to construct fusions with
the Pho-LacZα reporter. (b) Experimental determination of YmgF membrane topology. E. coli DH5α cells express-
ing different YmgF/Pho-LacZα fusions (position of insertion indicated in label) were plated on an indicator medium
containing the two chromogenic substrates Red-Gal (for β-galactosidase activity) and X-Pho (for phosphatase
activity). Blue coloration of the colonies (high phosphatase activity) indicates a membrane or periplasmic location
of the fusion point. Red coloration of the colonies (high β-galactosidase activity) indicates cytosolic location of
fusion point. Control cells (i.e., E. coli DH5α/pKTop) are indicated by the C label. (c) Quantitative enzymatic assays
of various YmgF/PhoA-LacZα fusions. The bar chart (blue bar, phosphatase activity; red bar, β-galactosidase
activity) represents the relative PhoA and LacZ enzymatic activities, measured on liquid cultures of DH5α express-
ing the indicated YmgF/Pho-LacZα fusions (position of insertion indicated on the abscissa). The normalized
activities (NAR) are plotted below as is the deduced subcellular localization of the residues at the position of the
Pho-LacZα insertion (cyt: cytoplasmic; per: periplasmic; tm: TMS)
3.3 Nested Deletion This method makes it possible to obtain a library of randomly
Approach generated 3′-truncations of the target protein gene to be fused to
the phoA-lacZα reporter cassette [19, 25–27, 35].
1. Clone the gene encoding the target membrane protein
upstream of the phoA-lacZα reporter gene (e.g., between the
PstI and XbaI sites of pKTop).
136 Gouzel Karimova and Daniel Ladant
3.4 Sandwich Fusion This method allows one to insert the PhoA-LacZα reporter into
Approach various loops (cytosolic or periplasmic) of the otherwise intact
protein [19, 25–27].
1. Design PCR primers to allow for an insertion of the phoA-
lacZα cassette in frame with both the upper and downstream
portions of the target gene.
2. Clone the PCR-amplified phoA-lacZα cassette at selected
restriction sites within the target gene.
3. The selected restriction sites in the target gene may already
preexist within the native sequence or, alternatively, they must
be introduced by site-directed mutagenesis. Caution should
be exercised during construction of recombinant plasmids that
express a hybrid protein between the target membrane protein
(or its fragment) and the PhoA-LacZα reporter (see Notes 12
and 14).
3.5 Analyzing Clones Screening for topological locations can be done directly on agar
on Dual Substrate plates that contain the specific chromogenic substrates for both
Plates PhoA- and LacZ-enzymatic activities (see Notes 15 and 16).
1. Select the E. coli cells (e.g., DH5α or other suitable strains; see
Notes 3, 4, and 12) transformed with the constructed plas-
mids on LB agar plates containing the appropriate antibiotic
(e.g., kanamycin for pKTop) and glucose (0.1–0.2%) and
incubate for 20–24 h at 30 °C to reduce the expression of the
recombinant phoA-lacZα fusions (see Note 12).
2. Streak a single clone of each phoA-lacZα fusion under study
on a fresh dual-indicator plate, containing kanamycin
pho-lac Reporter Fusions 137
3.6 Growth Many different protocols for PhoA and LacZ activity assays have
of Bacterial Culture been described in the literature [22, 23, 31, 37]. We use a simpli-
for Enzymatic Assays fied version of these protocols, in which the E. coli cells are per-
meabilized with chloroform and SDS. The protocols can be easily
adapted for use in a 96-well microtiter plate format.
1. Pick a single colony from a fresh LB/kanamycin/glucose plate
that has been inoculated with DH5α (pKTop-x) cells. Transfer
the colony into 5 mL LB broth with kanamycin (50 μg/mL)
and glucose (0.1%). Grow overnight at 37 °C with shaking
(150–200 rpm).
2. The next day, dilute overnight culture 1:100 in LB broth fresh
medium with kanamycin and let it grow at 37 °C with shaking
(150–200 rpm) for 2.5–3 h (to reach mid-exponential phase
of growth).
3. Add 1 mM IPTG to each culture, to induce expression of
hybrid X/PhoA-LacZα protein, and incubate for an additional
hour with aeration at 37 °C (see Note 17).
4 Notes
Acknowledgment
References
1. von Heijne G (2006) Membrane-protein 12. Silhavy TJ, Shuman HA, Beckwith J et al
topology. Nat Rev Mol Cell Biol 7:909–918 (1977) Use of gene fusions to study outer
2. Islam ST, Lam JS (2013) Topological mapping membrane protein localization in Escherichia
methods for α-helical bacterial membrane pro- coli. Proc Natl Acad Sci U S A 74:5411–5415
teins - an update and a guide. Microbiologyopen 13. Bibi E, Beja O (1994) Membrane topology of
2:350–364 multidrug resistance protein expressed in
3. Dobson L, Remenyi I, Tusnady GE (2015a) The Escherichia coli. N-terminal domain. J Biol
human transmembrane proteome. Biol Direct Chem 269:19910–19915
10:1–18 14. Boyd D, Manoil C, Beckwith J (1987)
4. Chen CP, Rost B (2002) State-of-the-art in Determinants of membrane protein topology.
membrane protein prediction. Appl Bioinforma Proc Natl Acad Sci U S A 84:8525–8529
1:21–35 15. Boyd D, Manoil C, Froshauer S et al (1990)
5. Tusnady GE, Simon I (2010) Topology pre- Use of gene fusions to study membrane-protein
diction of helical transmembrane proteins: how topology. In: Gierash LM, King J (eds) Protein
far have we reached? Curr Protein Pept Sci folding: deciphering the second half of the
11:550–561 genetic code. AAAS Books, Washington
6. Dobson L, Remenyi I, Tusnady GE (2015b) 16. Manoil C (1990a) Analysis of protein localiza-
CCTOP: a consensus constrained TOPology tion by use of gene fusions with complemen-
prediction web server. Nucleic Acids Res tary properties. J Bacteriol 172:1035–1042
43:W408–W412 17. San Millan JL, Boyd D, Dalbey R et al (1989)
7. Peters C, Konstantinos D, Shu N et al (2016) Use of phoA fusions to study the topology of
Improved topology prediction using the termi- the Escherichia coli inner membrane protein
nal hydrophobic helices rule. Bioinformatics leader peptidase. J Bacteriol 171:5536–5541
32:1158–1162 18. Silhavy TJ, Beckwith JR (1985) Uses of lac
8. Manoil C, Beckwith J (1986) A genetic fusions for the study of biological problems.
approach to analyzing membrane protein Microbiol Rev 49:398–418
topology. Science 233:1403–1408 19. Alexeyev MF, Winkler HH (1999) Membrane
9. Manoil C, Mekalanos JJ, Beckwith J (1990) topology of the Rickettsia prowazekii ATP/
Alkaline-phosphatase fusions - sensors of sub- ADP translocase revealed by novel dual pho-lac
cellular location. J Bacteriol 172:515–518 reporters. J Mol Biol 285:1503–1513
10. van Geest M, Lolkema JS (2000) Membrane 20. Langley KE, Villarejo MR, Fowler AV et al
topology and insertion of membrane proteins: (1975) Molecular basis of beta-galactosidase
search for topogenic signals. Microbiol Mol alpha-complementation. Proc Natl Acad Sci U
Biol Rev 64:13–33 S A 72:1254–1257
11. Lee C, Inouye H, Brickman ER et al (1989) 21. Ullmann A, Jacob F, Monod J (1967)
Genetic studies on the inability of beta- Characterization by in vitro complementation of
galactosidase to be translocated across the a peptide corresponding to an operator-proximal
Escherichia coli cytoplasmic membrane. segment of the beta-galactosidase structural
J Bacteriol 171:4609–4616 gene of Escherichia coli. J Mol Biol 24:339–343
142 Gouzel Karimova and Daniel Ladant
Abstract
Bacteria have developed a number of trans-envelope systems to transport molecules or assemble organelles
across bacterial envelopes. However, bacterial envelopes contain a rigid netlike peptidoglycan structure
that protects cells from osmotic lysis. Trans-envelope systems thus must interact with the peptidoglycan
barrier to generate gaps or anchor structures to the peptidoglycan scaffold. Here we describe methods to
use in vivo cross-linking and in vitro co-sedimentation to study protein–peptidoglycan interactions in
Gram-negative bacteria. In particular, we address important considerations to ensure the specificity of the
interactions in question.
Key words Trans-envelope systems, Peptidoglycan, Cross-linking, Co-sedimentation, Muramic acid assay
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_11, © Springer Science+Business Media LLC 2017
143
144 Gang Li and S. Peter Howard
arm. DTSSP can covalently link two primary amines (from ExeA
and peptidoglycan in this case) if they are in close proximity. After
cross-linking, peptidoglycan sacculi are isolated by a modified
small-volume version of the sodium dodecyl sulfate (SDS)-boiling
method to remove noncovalently associated proteins [5]. Purified
peptidoglycan samples are then treated with β-mercaptoethanol to
release cross-linked proteins to be analyzed by SDS-PAGE (poly-
acrylamide gel electrophoresis) and immunoblot.
We also describe a co-sedimentation (pulldown) assay that uses
purified peptidoglycan sacculi to investigate the binding of pro-
teins of interest to peptidoglycan [6]. This is a direct and straight-
forward method to detect protein–petidoglycan interactions
in vitro. We provide a protocol to prepare highly pure peptidogly-
can sacculi using SDS-boiling and enzymic treatments [5]. A colo-
rimetric method is used to quantitate peptidoglycan by measuring
lactic acids released from the muramic acid residues of peptidogly-
can upon acidic and alkaline hydrolysis [7]. We pay particular
attention to the hydrophobic nature of purified peptidoglycan sac-
culi (presumably originating from lipid-linked disaccharide precur-
sors that remain at the ends of glycan strands) [8]. Special
experimental procedures are followed to ease peptidoglycan han-
dling and overcome nonspecific interactions in co-sedimentation
experiments.
2 Materials
2.4 Co-sedimenta- 1. Tween 20 stock solution (10×): 0.5% Tween 20 in water. Store
tion Assay at 4 °C.
2. Binding buffer (10×): 400 mM sodium phosphate, pH 6.5.
3. Bovine albumin stock solution (100×): 1 mg/mL bovine albu-
min in water. Store at −20 °C.
4. Refrigerated microcentrifuge.
3 Methods
3.1 In Vivo 1. Grow bacterial strains under conditions in which the proteins
Cross-Linking of interest are produced and functional. For each cross-linking
experiment, 5–10 mL culture is generally sufficient. A. hydroph-
ila strains cultured in buffered Luria-Bertani (LB) medium are
used in this protocol [4]. In addition to the wild- type A.
hydrophila strain, cells expressing ExeA variants that contain
substitution mutations in the putative peptidoglycan binding
domain are included as controls (see Note 1).
2. Pellet cells by centrifugation at 6000 × g for 5 min. Wash twice
with PBS and resuspend cells in PBS. Adjust cell suspension to
an OD600 of 2.0. Transfer 1 mL cells to microcentrifuge tubes
for cross-linking. To avoid cold shock that may affect cell enve-
lope structure and physiology, perform the preceding steps at
room temperature.
3. Add fresh 10 mM DTSSP solution to a final concentration of
0.5 mM (or a range of 0.1–1 mM in initial experiments). Mix
immediately by inverting the tube several times. Incubate the
146 Gang Li and S. Peter Howard
3.3 Muramic Acid Warning: This colorimetric method uses strong acid and alkaline
Assay of solutions. Wear suitable personal protective equipment and follow
Peptidoglycan laboratory safety guidelines. Alternative methods using high-
performance liquid chromatography or quantitative aminosugar
analysis are described elsewhere [5, 9].
4 Notes
References
1. Höltje JV (1998) Growth of the stress-bearing 6. Li G, Howard SP (2010) ExeA binds to pepti-
and shape-maintaining murein sacculus of doglycan and forms a multimer for assembly of
Escherichia coli. Microbiol Mol Biol Rev the type II secretion apparatus of Aeromonas
62:181–203 hydrophila. Mol Microbiol 76:772–781
2. Dijkstra AJ, Keck W (1996) Peptidoglycan as a 7. Hoijer MA, Melief MJ, van Helden-Meeuwsen
barrier to transenvelope transport. J Bacteriol CG, Eulderink F, Hazenberg MP (1995)
178:5555–5562 Detection of muramic acid in a carbohydrate
3. Scheurwater EM, Burrows LL (2011) fraction of human spleen. Infect Immun
Maintaining network security: how 63:1652–1657
macromolecular structures cross the peptido- 8. Typas A, Banzhaf M, Gross CA, Vollmer W
glycan layer. FEMS Microbiol Lett 318:1–9 (2011) From the regulation of peptidoglycan
4. Howard SP, Gebhart C, Langen GR, Li G, synthesis to bacterial growth and morphology.
Strozen TG (2006) Interactions between pep- Nat Rev Microbiol 10:123–136
tidoglycan and the ExeAB complex during 9. Clarke AJ (1993) Compositional analysis of
assembly of the type II secretin of Aeromonas peptidoglycan by high-performance anion-
hydrophila. Mol Microbiol 59:1062–1072 exchange chromatography. Anal Biochem
5. Glauner B (1988) Separation and quantifica- 212:344–350
tion of muropeptides with high-performance
10. Sulfuric Acid (2015) The Columbia
liquid chromatography. Anal Biochem Encyclopedia, 6th edn. http:// www.encyclo-
172:451–464 pedia.com. Accessed 26 Jan 2016
Chapter 12
Abstract
Most gene clusters encoding multiprotein complexes of the bacterial cell envelope, such as conjugation
and secretion systems, Type IV pili, and flagella, bear a gene encoding an enzyme with peptidoglycan
hydrolase activity. These enzymes are usually glycoside hydrolases that cleave the glycan chains of the pep-
tidoglycan. Their activities are spatially controlled to avoid cell lysis and to create localized rearrangement
of the cell wall. This is assured by interaction with the structural subunits of the apparatus. Here we
describe protocols to test the peptidoglycan hydrolase activity of these proteins in vitro and in solution.
Key words Cell wall, Localized degradation, Peptidoglycan, Lytic transglycosylase, Remazol blue
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_12, © Springer Science+Business Media LLC 2017
151
152 Yoann G. Santin and Eric Cascales
which are necessary for the biogenesis of Type III (T3SS), Type
IV (T4SS), and Type VI (T6SS) secretion systems, respectively [6,
8–18]. However, these enzymes could be deleterious for bacterial
cells, and therefore their activity needs to be restricted to the site
of assembly in order to avoid breaches in the cell wall. Studies
have provided evidence that LTGs are recruited via specific inter-
actions to subunits of the machine [18–21] and, in a few cases,
that these interactions stimulate LTG activity [18, 21, 22].
Methods have been developed and used to test whether puta-
tive LTGs have peptidoglycan hydrolase activities. An indirect
approach is to clone the gene encoding the putative LTG to a signal
sequence in order to address the protein to the periplasm of E. coli
and follow the cell growth after induction as overproduction of the
LTG causes cell lysis [23, 24]. More direct protocols have been
developed using purified LTGs, including zymogram [25, 26].
However, this technique, which consists of subjecting purified
LTGs to sodium dodecyl sulfate-polyacrylamide gel electrophoresis
(SDS-PAGE) in a gel supplemented with purified peptidoglycan,
has limits, such as the refolding of the protein after migration.
Additional approaches, performed in solution, do not require the
denaturation and refolding steps. These turbidometric assays,
detailed in what follows, are methods for following the activity of
purified LTGs on peptidoglycan or peptidoglycan labeled with
remazol brilliant blue (RBB) dye [27, 28]. The peptidoglycan assay
relies on the decrease of absorbance of the peptidoglycan solution
[27], whereas the RBB assay relies on the release of the dye cap-
tured in the peptidoglycan net [28] in the presence of the LTG. In
addition, more precise approaches, such as the analysis of peptido-
glycan degradation products released after incubation of the pepti-
doglycan with the purified protein by reverse-phase high-performance
liquid chromatography coupled to mass spectrometry [29, 30],
make it possible to define the site of cleavage of the enzyme.
2 Material
2.4 RBB-Labeled 1. PBS buffer: Dissolve 1.44 g Na2HPO4, 0.24 g KH2PO4, 0.2 g
Peptidoglycan KCl, and 8 g NaCl in 1 L sterile distilled water. Adjust pH to
Degradation Assay 7.4 with 1 M HCl.
2. Purified protein to be tested.
3. Ethanol 96° or absolute.
4. Lysozyme stock solution: 10 mg/mL egg-white lysozyme in
sterile distilled water.
154 Yoann G. Santin and Eric Cascales
5. Incubate at 37 °C.
6.
Ultracentrifuge (Beckman with TLA100.3 rotor, or
equivalent).
7. Spectrophotometer.
3 Methods
3.1 Peptidoglycan The peptidoglycan purification protocol is adapted from [31, 32].
Purification 1. Grow cells in 400 mL of appropriate medium until culture
reaches an A600 of about 1–1.2.
2. Harvest cells by centrifugation at 10,000 × g for 20 min at
4 °C. Resuspend cells in 20 mL 20 mM Tris–HCl, pH 8.0,
100 mM NaCl. Break cells by three passages at French press or
using an Emulsiflex apparatus.
3. Pellet cell envelopes by centrifugation at 400,000 × g
(90,000 rpm in a Beckman TLA-100.4 rotor) for 45 min at
4 °C. Resuspend cells in 10 mL 0.5 M NaCl.
4. Add 10 mL 8% SDS and incubate for 1 h at 96 °C.
5. Leave solution at room temperature overnight.
6. Pellet peptidoglycan by ultracentrifugation at 400,000 × g at
25 °C for 45 min (see Note 1).
7. Resuspend peptidoglycan fraction in 10 mL 0.5 M NaCl and
add 10 mL 8% SDS. Incubate for 30 min at 96 °C.
8. Pellet peptidoglycan by ultracentrifugation at 400,000 × g at
25 °C for 30 min and resuspend peptidoglycan in 10 mL water.
9. Repeat step 8 twice.
10. Resuspend peptidoglycan in 10 mL 20 mM Tris–HCl, pH 7.2,
50 mM NaCl supplemented with 200 μg/mL α-amylase and
200 μg/mL pronase. Incubate overnight at 37 °C.
11. Add 10 mL 8% SDS and incubate for 1 h at 96 °C.
12. Pellet peptidoglycan by ultracentrifugation at 400,000 × g at
25 °C for 30 min and resuspend peptidoglycan in 10 mL water.
13. Repeat step 12 twice.
14. Resuspend peptidoglycan pellet in 1 mL water. Store at 4 °C.
4 Notes
Acknowledgements
References
1. Demchick P, Koch AL (1996) The perme- 12. Kohler PL, Hamilton HL, Cloud-Hansen
ability of the wall fabric of Escherichia coli and K, Dillard JP (2007) AtlA functions as a
Bacillus subtilis. J Bacteriol 178:768–773 peptidoglycan lytic transglycosylase in the
2. Scheurwater E, Reid CW, Clarke AJ (2008) Neisseria gonorrhoeae type IV secretion system.
Lytic transglycosylases: bacterial space-making J Bacteriol 189:5421–5428
autolysins. Int J Biochem Cell Biol 40:586–591 13. Zhong Q, Shao S, Mu R, Wang H,
3. Scheurwater EM, Burrows LL (2011) Huang S, Han J, Huang H, Tian S (2011)
Maintaining network security: how macromo- Characterization of peptidoglycan hydrolase in
lecular structures cross the peptidoglycan layer. Cag pathogenicity island of Helicobacter pylori.
FEMS Microbiol Lett 318:1–9 Mol Biol Rep 38:503–509
4. Koraimann G (2003) Lytic transglycosyl- 14. García-Gómez E, Espinosa N, de la Mora J,
ases in macromolecular transport systems Dreyfus G, González-Pedrajo B (2011) The
of gram-negative bacteria. Cell Mol Life Sci muramidase EtgA from enteropathogenic
60:2371–2388 Escherichia coli is required for efficient type III
5. Höltje JV (1996) Lytic transglycosylases. EXS secretion. Microbiology 157:1145–1160
75:425–429 15. Arends K, Celik EK, Probst I, Goessweiner-
6. Zahrl D, Wagner M, Bischof K, Bayer M, Mohr N, Fercher C, Grumet L, Soellue C,
Zavecz B, Beranek A, Ruckenstuhl C, Zarfel Abajy MY, Sakinc T, Broszat M, Schiwon K,
GE, Koraimann G (2005) Peptidoglycan deg- Koraimann G, Keller W, Grohmann E (2013)
radation by specialized lytic transglycosylases TraG encoded by the pIP501 type IV secre-
associated with type III and type IV secretion tion system is a two-domain peptidoglycan-
systems. Microbiology 151:3455–3467 degrading enzyme essential for conjugative
transfer. J Bacteriol 195:4436–4444
7. van Heijenoort J (2011) Peptidoglycan hydro-
lases of Escherichia coli. Microbiol Mol Biol 1 6. Laverde Gomez JA, Bhatty M, Christie PJ
Rev 75:636–663 (2014) PrgK, a multidomain peptidoglycan
hydrolase, is essential for conjugative transfer
8. de la Mora J, Ballado T, González-Pedrajo B, of the pheromone-responsive plasmid pCF10.
Camarena L, Dreyfus G (2007) The flagel- J Bacteriol 196:527–539
lar muramidase from the photosynthetic bac-
terium Rhodobacter sphaeroides. J Bacteriol 1 7. Weber BS, Hennon SW, Wright MS, Scott NE,
189:7998–8004 de Berardinis V, Foster LJ, Ayala JA, Adams MD,
Feldman MF (2016) Genetic dissection of the
9. de la Mora J, Osorio-Valeriano M, González- Type VI secretion system in Acinetobacter and
Pedrajo B, Ballado T, Camarena L, Dreyfus G identification of a novel peptidoglycan hydro-
(2012) The C terminus of the flagellar muram- lase, TagX, required for its biogenesis. MBio
idase SltF modulates the interaction with
FlgJ in Rhodobacter sphaeroides. J Bacteriol 1 8. Santin YG, Cascales E (2016) Domestication
194:4513–4520 of a housekeeping transglycosylase for assem-
bly of a type VI secretion system. EMBO Rep
10. Nambu T, Minamino T, Macnab RM, 18(1):138–149
Kutsukake K (1999) Peptidoglycan-
hydrolyzing activity of the FlgJ protein, essen- 1 9. Höppner C, Carle A, Sivanesan D, Hoeppner
tial for flagellar rod formation in Salmonella S, Baron C (2005) The putative lytic trans-
typhimurium. J Bacteriol 181:1555–1561 glycosylase VirB1 from Brucella suis interacts
with the type IV secretion system core compo-
11. Mushegian AR, Fullner KJ, Koonin EV,
nents VirB8, VirB9 and VirB11. Microbiology
Nester EW (1996) A family of lysozyme-like 151:3469–3482
virulence factors in bacterial pathogens of
plants and animals. Proc Natl Acad Sci U S A 2 0. Creasey EA, Delahay RM, Daniell SJ, Frankel
93:7321–7326 G (2003) Yeast two-hybrid system survey
158 Yoann G. Santin and Eric Cascales
Abstract
The bacterial two-hybrid (BACTH, for “Bacterial Adenylate Cyclase-Based Two-Hybrid”) system is a
simple and fast genetic approach to detecting and characterizing protein–protein interactions in vivo. This
system is based on the interaction-mediated reconstitution of a cyclic adenosine monophosphate (cAMP)
signaling cascade in Escherichia coli. As BACTH uses a diffusible cAMP messenger molecule, the physical
association between the two interacting chimeric proteins can be spatially separated from the transcription
activation readout, and therefore it is possible to analyze protein–protein interactions that occur either in
the cytosol or at the inner membrane level as well as those that involve DNA-binding proteins. Moreover,
proteins of bacterial origin can be studied in an environment similar (or identical) to their native one. The
BACTH system may thus permit a simultaneous functional analysis of proteins of interest—provided the
hybrid proteins retain their activity and their association state. This chapter describes the principle of the
BACTH genetic system and the general procedures to study protein–protein interactions in vivo in E. coli.
Key words Two-hybrid system, Protein interaction assay, Membrane protein, cAMP signaling,
Chimeric proteins
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_13, © Springer Science+Business Media LLC 2017
159
160 Gouzel Karimova et al.
1.1 Principle The BACTH bacterial two-hybrid system is a simple and fast
of Bacterial Adenylate approach to detecting and characterizing protein–protein interac-
Cyclase-Based tions in vivo. It offers all the advantages of working with E. coli and
Two-Hybrid System is readily accessible to many researchers having basic knowledge in
standard microbiological and molecular biology techniques (e.g.,
plasmid preparation, bacterial transformation, polymerase chain
reaction (PCR)).
The BACTH system is based on the interaction-mediated
reconstitution of adenylate cyclase enzyme activity in an E. coli cya
mutant, defective in its endogenous adenylate cyclase [3, 10]. It
exploits the fact that the catalytic domain of adenylate cyclase
(CyaA) from B. pertussis [11] consists of two complementary frag-
ments, T25 and T18, that are not active when physically separated
(Fig. 1a). When these two fragments are fused to interacting poly-
peptides, X and Y, heterodimerization of the hybrid proteins results
in functional complementation between the T25 and T18 frag-
ments and, therefore, in cAMP synthesis (Fig. 1b). Cyclic AMP
produced by the reconstituted chimeric enzyme binds to the catab-
olite activator protein (CAP). The cAMP/CAP complex is a pleio-
tropic regulator of gene transcription in E. coli [12]. It turns on the
expression of several resident genes, including genes of the lac and
mal operons involved in lactose and maltose catabolism (Fig. 1c).
Consequently, bacteria become able to utilize lactose or maltose as
the unique carbon source and can be easily distinguished on indi-
cator or selective media [3, 10].
Fig. 1 Principle of BACTH system. (a) When the two fragments of B. pertussis
adenylate cyclase, T25 and T18, are coexpressed as separate polypeptides, they
cannot assemble and there is no enzyme activity. (b) When the T25 and T18 frag-
ments are coexpressed as fusions with polypeptides X and Y that can interact,
the association of the T25-X and T18-Y hybrid proteins reconstitutes the adenyl-
ate cyclase activity. (c) Cyclic AMP synthesized by the reconstituted enzyme
binds to the catabolite activator protein (CAP), and the cAMP/CAP complex can
associate with specific promoter DNA and activates transcription of catabolite
operons (such as lac operon or mal regulon)
2 Materials
2.4 BACTH Reporter 1. E. coli reporter strain carrying a deletion of the cya gene (see
Strains, Plasmids, Note 3).
and Antibodies 2. Set of compatible vectors allowing genetic fusions of proteins
of interest at either the N- or the C-terminus of the T25 frag-
ment (pKT25 and pKNT25) or of the T18 fragment (pUT18
and pUT18C) (see Note 4).
164 Gouzel Karimova et al.
3 Methods
3.2 Construction This section assumes that the reader has background knowledge in
of BACTH Plasmids basic molecular biology techniques. Additional protocols for
Encoding Hybrid molecular cloning, PCR, DNA analysis, and transformation can be
Proteins found in many textbooks (e.g., [18]) or on the Internet.
3.2.1 Standard Cloning 1. Design specific primers to amplify the genes encoding the pro-
of Genes Encoding Proteins teins of interest. The primers should include restriction sites
of Interest into BACTH (e.g., BamHI on 5′ primer and KpnI on 3′ primer) for allow-
Vectors ing oriented cloning of the amplified genes into the BACTH
vectors. Be careful to correctly position these restriction sites
so that the genes of interest will be in frame with the T25 and
T18 open reading frames.
2. PCR amplify the genes encoding the proteins of interest using
a standard protocol [18].
3. Purify the PCR-amplified DNA fragments using a standard
PCR purification kit (available from various companies) and
Bacterial Two-Hybrid Interaction Assays 165
3.2.2 Gateway™ Cloning The Gateway® cloning technology (Life Technologies, Thermo
of Genes Encoding Proteins Fisher Scientific) is used to transfer genes of interest to the BACTH-
of Interest to BACTHGW Gateway destination vectors, pST25-DEST, pSNT25-DEST, and
Vectors pUT18C-DEST [17]. For detailed descriptions of the Gateway®
cloning techniques, the reader may refer to the manufacturer’s
guidelines.
1. PCR-amplify the genes of interest (from genomic DNA or
from other appropriate sources) using appropriate primers that
also contain specific attB sites (see Note 6) and purify the PCR
products as described earlier.
2. Mix the purified PCR products with the pDONR™221 plas-
mid [19], add the BP Clonase™ II enzyme and incubate 2 h at
room temperature to allow the BP recombination reaction.
Add 2 μg proteinase K to terminate recombination reaction
and transform mixture into E. coli XL1 competent cells. Select
transformants on LB plates supplemented with 50 μg/mL
kanamycin [17].
3. Purify plasmid DNA from 3 to 4 independent clones from each
cloning as described earlier, and check the recombinant plas-
mids by restriction analysis and DNA sequencing.
In the resulting plasmids (pDONR™-gene X) the genes of
interest are flanked by attL recombination sites and can be eas-
ily transferred into Gateway® destination vectors by a so-called
LR reaction [19].
4. Mix the entry pDONR™-gene X plasmid with the appropriate
BACTHGW destination vectors, pST25-DEST, pSNT25-
DEST, or pUT18C-DEST. Add the LR Clonase™ enzyme mix
166 Gouzel Karimova et al.
3.4 BACTH Screening The BACTH system can be used to screen libraries to isolate part-
of Interacting ners of a protein of interest (e.g., protein X, classically designated
Partners: Selection as “bait”) as follows:
Procedure 1. Construct a library of genomic DNA (or cDNA) fragments in
on Minimal Medium one of the BACTH vectors, e.g., pKT25, using standard pro-
cedures [18]. Obviously the quality (i.e., complexity) of the
Bacterial Two-Hybrid Interaction Assays 167
4 Notes
Fig. 3 Schematic representation of BACTH plasmids. (a) Standard BACTH plasmids. Yellow and green rect-
angles represent the open reading frames of T25 and T18 fragments, respectively, under the control of the
lac promoter (small arrow). Pink and orange arrows indicate the antibiotic selectable markers and the direc-
tion of transcription. The red and blue boxes indicate the plasmid origin of replication. The hatched boxes
represent the multicloning sequences (MCS) that allow insertion of foreign genes: some unique restriction
sites are displayed above the nucleotide sequence, and the encoded polypeptide sequences are shown
below. (b) Gateway™-compatible, BACTHGW plasmids
Bacterial Two-Hybrid Interaction Assays 173
Acknowledgments
References
1. Fields S, Song O (1989) A novel genetic sys- transcription activation. Curr Opin Struct Biol
tem to detect protein-protein interactions. 14:10–20
Nature 340:245–246 13. Karimova G, Ullmann A, Ladant D (2001)
2. Stynen B, Tournu H, Tavernier J, Van Dijck P Protein-protein interaction between Bacillus
(2012) Diversity in genetic in vivo methods for stearothermophilus tyrosyl-tRNA synthetase
protein-protein interaction studies: from the subdomains revealed by a bacterial two-hybrid
yeast two-hybrid system to the mammalian system. J Mol Microbiol Biotechnol 3(1):
split-luciferase system. Microbiol Mol Biol Rev 73–82
76:331–382 14. Fransen M, Brees C, Ghys K, Amery L,
3. Karimova G, Pidoux J, Ullmann A, Ladant D Mannaerts GP, Ladant D, Van Veldhoven PP
(1998) A bacterial two-hybrid system based on (2002) Analysis of mammalian peroxin interac-
a reconstituted signal transduction pathway. tions using a non-transcription-based bacterial
Proc Natl Acad Sci U S A 95:5752–5756 two-hybrid assay. Mol Cell Proteomics 1:
4. Karimova G, Dautin N, Ladant D (2005) 243–252
Interaction network among Escherichia coli 15. Dautin N, Karimova G, Ladant D (2003)
membrane proteins involved in cell division as Human immunodeficiency virus (HIV) type 1
revealed by bacterial two-hybrid analysis. transframe protein can restore activity to a
J Bacteriol 187:2233–2243 dimerization-deficient HIV protease variant.
5. Jack RL, Buchanan G, Dubini A, Hatzixanthis J Virol 77:8216–8226
K, Palmer T, Sargent F (2004) Coordinating 16. Battesti A, Bouveret E (2012) The bacterial
assembly and export of complex bacterial pro- two-hybrid system based on adenylate cyclase
teins. EMBO J 23:3962–3972 reconstitution in Escherichia coli. Methods
6. Paschos A, den Hartigh A, Smith MA, Atluri 58:325–334
VL, Sivanesan D, Tsolis RM, Baron C (2011) 17. Ouellette SP, Gauliard E, Antosova Z, Ladant
An in vivo high-throughput screening approach D (2014) A Gateway((R)) -compatible bacte-
targeting the type IV secretion system compo- rial adenylate cyclase-based two-hybrid system.
nent VirB8 identified inhibitors of Brucella Environ Microbiol Rep 6:259–267
abortus 2308 proliferation. Infect Immun 18. Sambrook J, Russell DW (2006) The con-
79:1033–1043 densed protocols from molecular cloning: a
7. Cisneros DA, Bond PJ, Pugsley AP, Campos laboratory manual. Cold Spring Harbor
M, Francetic O (2012) Minor pseudopilin self- Laboratory Press, Cold Spring Harbor, NY
assembly primes type II secretion pseudopilus 19. Hartley JL, Temple GF, Brasch MA (2000)
elongation. EMBO J 31:1041–1053 DNA cloning using in vitro site-specific recom-
8. Georgiadou M, Castagnini M, Karimova G, bination. Genome Res 10:1788–1795
Ladant D, Pelicic V (2012) Large-scale study 20. Karimova G, Robichon C, Ladant D (2009)
of the interactions between proteins involved Characterization of YmgF, a 72-residue inner
in type IV pilus biology in Neisseria meningiti- membrane protein that associates with the
dis: characterization of a subcomplex involved Escherichia coli cell division machinery.
in pilus assembly. Mol Microbiol 84:857–873 J Bacteriol 191:333–346
9. Zoued A, Durand E, Brunet YR, Spinelli S, 21. Karimova G, Davi M, Ladant D (2012) The
Douzi B, Guzzo M, Flaugnatti N, Legrand P, beta-lactam resistance protein Blr, a small
Journet L, Fronzes R et al (2016) Priming and membrane polypeptide, is a component of the
polymerization of a bacterial contractile tail Escherichia coli cell division machinery.
structure. Nature 531:59–63 J Bacteriol 194:5576–5588
10. Karimova G, Ullmann A, Ladant D (2000) A 22. Griffith KL, Wolf REJ (2002) Measuring beta-
bacterial two-hybrid system that exploits a galactosidase activity in bacteria: cell growth,
cAMP signaling cascade in Escherichia coli. permeabilization, and enzyme assays in 96-well
Methods Enzymol 328:59–73 arrays. Biochem Biophys Res Commun
11. Ladant D, Ullmann A (1999) Bordatella per- 290:397–402
tussis adenylate cyclase: a toxin with multiple 23. Ouellette SP, Rueden KJ, Gauliard E, Persons
talents. Trends Microbiol 7:172–176 L, de Boer PA, Ladant D (2014) Analysis of
12. Lawson CL, Swigon D, Murakami KS, Darst MreB interactors in Chlamydia reveals a RodZ
SA, Berman HM, Ebright RH (2004) homolog but fails to detect an interaction with
Catabolite activator protein: DNA binding and MraY. Front Microbiol 5:279
176 Gouzel Karimova et al.
24. Robichon C, Karimova G, Beckwith J, Ladant D 26. Ouellette SP, Karimova G, Subtil A, Ladant D
(2011) Role of leucine zipper motifs in associa- (2012) Chlamydia co-opts the rod shape-
tion of the Escherichia coli cell division proteins determining proteins MreB and Pbp2 for cell
FtsL and FtsB. J Bacteriol 193:4988–4992 division. Mol Microbiol 85:164–178
25. Battesti A, Bouveret E (2008) Improvement of 27. Dautin N, Karimova G, Ullmann A, Ladant D
bacterial two-hybrid vectors for detection of fusion (2000) Sensitive genetic screen for protease
proteins and transfer to pBAD-tandem affinity activity based on a cyclic AMP signaling cas-
purification, calmodulin binding peptide, or 6-his- cade in Escherichia coli. J Bacteriol 182:
tidine tag vectors. Proteomics 8:4768–4771 7060–7066
Chapter 14
Abstract
The yeast two-hybrid system is a powerful and commonly used genetic tool to investigate interactions
between artificial fusion proteins inside the nucleus of yeast. Here we describe how to use the Matchmaker
GAL4-based yeast two-hybrid system to detect the interaction of the Agrobacterium type VI secretion
system (T6SS) sheath components TssB and TssC41. The bait and prey gene are expressed as a fusion to
the GAL4 DNA-binding domain (DNA-BD) and GAL4 activation domain (AD, prey/library fusion pro-
tein) respectively. When bait and prey fusion proteins interact in yeast nucleus, the DNA-BD and AD are
brought into proximity, thereby activating the transcription of reporter genes. This technology can be
widely used to identify interacting partners, confirm suspected interactions, and define interacting domains.
Key words Protein–protein interaction, Yeast two-hybrid, Gal4 transcriptional activation domain
(AD), Gal4 DNA-binding domain (BD), Saccharomyces cerevisiae AH109, Type VI secretion system,
TssB, TssC
1 Introduction
The yeast two-hybrid system (Y2H) was first developed in 1989 and
revolutionized the process of searching for and identifying interact-
ing proteins [1]. To date, the Y2H system has proven to be a useful
and sensitive method for detecting not only stable interacting pro-
teins but also weak and transient protein interactions [2]. Because
Y2H is performed in vivo, the great advantage of the system is that
the testing proteins are more likely to be in their native conforma-
tions, which may lead to increased sensitivity and accuracy of detec-
tion [1, 3, 4]. Importantly, the Y2H system is complementary to
biochemical methods such as co-immunoprecipitation/pulldown
followed by western blotting or mass spectrometry analysis to
increase accuracy and dynamics for a more complete and reliable
map of interactions [2]. Notably, the Y2H method has been modi-
fied and improved greatly in recent years, including applications in
protein–DNA interactions and yeast three-hybrid, and has proven to
be amenable to interaction studies of membrane proteins, DNA
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_14, © Springer Science+Business Media LLC 2017
177
178 Jer-Sheng Lin and Erh-Min Lai
(A)
AD
Bait Prey
protein Transcriptional activation
protein
DNA-BD
Co-transformation Selecting
AH109 Yeast transformant
Fig. 1 Principle and experimental flowchart of yeast two-hybrid system. (a) Schematic
diagram of principle of Y2H system. Two testing proteins are each fused with two
different Gal4 domains, with the bait protein fused to the Gal4 DNA-binding domain
(DNA-BD, 1-147 a.a.) and the prey protein fused to the Gal4 transcriptional activation
domain (AD, 768-881 a.a.). In yeast strain AH109, transcriptional activation of the
reporters (ADE2, HIS3, and MEL1) only occurs in a cell that bait protein interact with
prey protein to restore functional Gal4 transcription factor binding to the Gal4-
responsive promoter GAL UAS [3]. (b) Experimental flowchart of Y2H system.
Cotransformation was performed using PEG/LiAc-mediated transformation method.
SD-WL medium is representative of synthetic defined (SD) minimal medium lacking
tryptophan (Trp) and leucine (Leu). SD-WLHA medium is representative of synthetic
dextrose minimal medium lacking Trp, Leu, adenine (Ade), and histidine (His)
Yeast Two-Hybrid 179
ADE2 and HIS3) for transcriptional activation. The Y2H system has
been widely used to detect the interactions of a wide range of pro-
teins from yeast, bacteria, animal, and plant systems. Y2H has been
successfully used to study the interaction of proteins involved in bac-
terial protein secretion from type IV [9–12] and type VI secretion
systems (T4SS, T6SS) in Agrobacterium tumefaciens [13, 14].
Here, the Y2H protocol describes the use of Matchmaker Y2H
system to detect the interaction of the Agrobacterium T6SS sheath
components TssB and TssC41 according to the instructions of the user
manual (Clontech, Mountain View, CA), with minor modifications.
TssB and TssC interact to form a cogwheel-like tubular structure,
which is analogous to the outer sheath structure of a contractile phage,
and wraps around the T6SS tail tube to propel the tail tube toward the
target cell interior upon infection [15, 16]. In A. tumefaciens, we
showed the interaction of the T6SS sheath components TssB and
TssC41 by Y2H assay, copurification in E. coli, and co-IP in A. tumefa-
ciens [14]. For the Y2H assay, each bait and prey plasmid pair was
cotransformed into the Saccharomyces cerevisiae strain AH109. The
transformants were selected by their growth on synthetic dextrose
(SD) minimal medium lacking tryptophan (Trp) and leucine (Leu)
(SD-WL medium), which are the nutritional selection markers for
pGBKT7 and pGADT7, respectively. The positive interaction of
expressed fusion proteins was then determined by their growth on SD
lacking Trp, Leu, adenine (Ade), and histidine (His) (SD-WLHA
medium) at 30 °C for at least 3 days (Fig. 1b). The positive interac-
tions wwere observed only for plasmid pairs expressing TssB and
TssC41 but not when each of them coexpressed with vector only, sug-
gesting the specific interactions of TssB and TssC41 (Fig. 2) [14].
BD-TssC41
BD-TssB
vector
+
vector
AD-TssC41 SD-WLHA
AD-TssB
vector
AD-TssC41 SD-WL
AD-TssB
Fig. 2 TssB and TssC41 interact with each other in yeast strain AH109. SD-WL
medium (SD minimal medium lacking Trp and Leu) was used for the selection of
plasmids. SD-WLHA medium (SD minimal medium lacking Trp, Leu, His, and Ade)
was used for the auxotrophic selection of bait and prey protein interactions. The
positive interaction was determined by the growth on SD-WLHA medium at
30 °C for at least 2 days. The positive control (+) showing interactions of SV40
large T-antigen and murine p53 and negative control (vector) are indicated
(reproduced from [14]; no permission is required for reuse of the content pub-
lished from Public Library of Science, PLoS)
180 Jer-Sheng Lin and Erh-Min Lai
2 Materials
All growth media and solutions are prepared using Milli-Q purified
water and analytical or molecular biology grade reagents.
2.1 Yeast Strain 1. The yeast Saccharomyces cerevisiae strain AH109: The com-
and Vectors (the plete genotype of AH109 is provided in what follows.
Following Information
MATa, trp1–901, leu2–3, 112, ura3–52, his3–200, gal4Δ,
Is According to [3]) gal80Δ, LYS2:: GAL1UAS-GAL1TATA-HIS3, GAL2UAS-GAL2TATA-
ADE2, URA3:: MEL1UAS-MEL1TATA-lacZ.
AH109 strain is gal4 − and gal80 −; this prevents interfer-
ence of native regulatory proteins with the regulatory elements
in the two-hybrid system. AH109 features three reporters,
ADE2, HIS3, and MEL1 (or lacZ), under the control of distinct
GAL4 upstream activating sequences (UASs) and TATA boxes.
2. pGBKT7 vector: The pGBKT7 vector contains a multiple
cloning site (MCS) for cloning to express proteins with
N-terminal fusion to amino acids 1–147 of the GAL4 DNA
binding domain (DNA-BD). In yeast, fusion proteins are
expressed at high levels from the constitutive ADH1 promoter
(PADH1). Transcription is terminated by the T7 and ADH1
transcription termination signals (TADH1). The pGBKT7 vector
can replicate autonomously in both E. coli and S. cerevisiae
from the pUC and 2 μ ori, respectively. The vector carries the
kanamycin-resistant gene for selection in E. coli and the TRP1
nutritional marker for selection in yeast. In addition, pGBKT7
also contains the T7 promoter and a c-Myc epitope tag for
in vitro transcription and translation of the c-Myc-tagged
fusion protein without GAL4 DNA-BD.
3. The pGADT7 vector: The pGADT7 vector contains MCSs
for cloning to express protein with N-terminal fusion to
amino acids 768–881 of the GAL4 activation domain (AD).
In yeast, fusion proteins are expressed at high levels from the
constitutive ADH1 promoter (PADH1). Transcription is ter-
minated at the ADH1 transcription termination signal
(TADH1). The fusion protein is targeted to the yeast nucleus
by the SV40 nuclear localization sequences that have been
added to the activation domain sequence. pGADT7 also
contains the T7 promoter and an HA epitope tag for in vitro
transcription and translation of the HA-tagged fusion pro-
tein without GAL4 AD. The pGADT7 vector can replicate
autonomously in both E. coli and S. cerevisiae from the pUC
and 2 μ ori, respectively. The vector carries the ampicillin-
resistant gene for selection in E. coli and the LEU2 nutri-
tional marker for selection in yeast.
Yeast Two-Hybrid 181
2.2 Yeast Cultures 1. Yeast peptone dextrose adenine (YPDA) medium: 20 g Bacto
and Yeast peptone, 10 g yeast extract, 20 g glucose, 40 mg adenine, 15 g
Transformation [17] agar (For plate use only), add water to 1 L, autoclave.
2. Minimal synthetic defined (SD) plate: 1.675 g yeast nitrogen
base without amino acid, 5 g glucose, 3.75 g Agar, add water
to 250 mL, autoclave. Dropout (DO) supplements (such as
-Trp-Leu or -Trp-Leu-Ade-His) can be added to the Minimal
SD Base to make a synthetic, defined medium lacking the spec-
ified nutrients (see Note 1).
3. Carrier DNA: 10 mg/mL salmon sperm DNA (ssDNA)
(UltraPure™ Salmon Sperm DNA Solution, ThermoFisher),
store at −20 °C (see Note 2).
4. 10× LiAc: 1 M Lithium acetate, pH 7.5 (see Note 3), autoclave
and store at room temperature (RT).
5. 40% polyethylene glycol (PEG) solution: 22 g Polyethylene
glycol (Molecular weight is 6000 or 3350 Da), add 31 mL of
water, autoclave and store at room temperature.
6. Plasmid DNA: ~200 ng per plasmid for co-transformation
(see Note 4).
7. Laminar flow.
2.4 Preparation 1. Yeast peptone dextrose adenine (YPDA) medium: See item 1
of Yeast Cultures in Subheading 2.2.
for Protein Extraction 2. 2× minimal synthetic defined (SD) medium: 40 g Bacto pep-
and Western Blot tone, 20 g yeast extract, 20 g glucose, 80 mg adenine, with
2X-Leu/-Trp dropout (DO) supplement (containing every
essential amino acids except for leucine and tryptophan), add
water to 1 L, autoclave.
mix well, and place on ice. Before use, add 1 mM dithiothreitol
(DTT), 2× protease inhibitor cocktails (stock: 50×), 4 mM
PMSF, mix well, then ready for use.
3 Methods
3.1 Gene The constructs used for Y2H analysis are generated based on the
Construction information of the vector map and MCS provided by the user man-
in pGBKT7 ual (Clontech) [3]. Briefly, bait and prey coding sequence (without
and pGADT7 Vectors stop codon) are PCR-amplified with appropriate primers, digested
with appropriate enzymes, and cloned to the same sites of pGBKT7
or pGADT7 [14].
3.2 Preparation 1. Inoculate 3 mL YPDA with a colony of AH109 (see Note 7)
of Yeast Cultures and incubate at 30 °C overnight (>16 h) with shaking
for Yeast (250 rpm) to the stationary phase (see Note 8).
Transformation [17] 2. Subculture by adding 1 mL AH109 overnight culture into
50 mL fresh YPDA medium.
3. Incubate at 30 °C for 4 h with shaking (250 rpm) (see Note 9).
4. Pour cells into 50 mL tubes and pellet cells at 450 × g for
3 min at 4 °C or RT (see Note 10).
5. Discard supernatant and resuspend cell pellet with 10 mL ster-
ile water by vortexing, and repellet cells at 450 × g for 3 min at
4 °C or RT (see Note 11).
6. Resuspend cell pellet in 100 μL 10× LiAc and 900 μL sterile
water (final concentration is 1× LiAc) (see Note 12). Incubate
cell suspension at 30 °C for 1 h with gentle shaking (150 rpm)
(see Note 13).
7. The suspended yeast competent cells are ready to use for trans-
formation (see Note 14).
3.3 PEG/LiAc- 1. Pretreat ssDNA by heating at 100 °C for 10 min and then put
Mediated on ice for 5–10 min before use (see Note 16).
Transformation 2. Add 80 μL heat-treated ssDNA (10 μg/μL) to 1 mL yeast
of Yeast (Small-Scale competent cell (final concentration ~ 0.8 mg/mL) and mix
Transformation of Bait well (see Note 17).
and Prey Plasmids) 3. Aliquot 100 μL of cell mixture into 1.5 mL microtube, and
(see Note 15) add an approximate amount of plasmid DNA (about 3–5 μL)
(see Note 18). Mix well by vortexing and incubate at 30 °C for
30 min (see Note 19).
4.
Freshly prepare LiAc-PEG solution (10× LiAc: 40%
PEG = 1:10, mix 1 mL 10× LiAc with 10 mL 40% PEG) and
add 700 μL LiAc-PEG solution to cell mixture after 30 min
Yeast Two-Hybrid 183
3.6 Preparation 1. Grow culture from a single colony (see Note 29) in 3 mL
of Yeast Cultures YPDA or 2× SD selection medium (see Note 30) overnight at
for Protein Extraction 30 °C.
(see Note 28) 2. Add 100 μL of overnight culture (OD600 should reach 1.5) in
fresh 5 mL 2× SD selection medium. Incubate at 30 °C with
shaking (about 250 rpm) until OD600 reaches 0.4–0.6.
Depending on tested proteins, it may take 4–5 h to reach
desired cell number (see Note 31).
3. Place cells in 15 mL tubes and pellet cells at 1000 × g for 5 min
at 4 °C.
4. Discard supernatant and resuspend cell pellet with 10 mL ster-
ile water by vortexing (see Note 32).
5. Repellet cells at 1000 × g for 5 min at 4 °C.
6. Repeat steps 4 and 5.
7. Discard supernatant. You can continue to extract yeast protein
or immediately freeze cell pellet by placing tube in liquid nitro-
gen, then store cells at −80 °C until western blot analysis.
3.7 Preparation 1. Keep protein samples on ice, add 100 μL freshly prepared yeast
of Yeast Protein protein extraction buffer to tube followed by addition of 50 μL
Extracts and Western acid-washed glass beads (see Note 33).
Blot Analysis 2. Vortex tubes at maximum speed for 30 s, then place tubes on
ice for 30 s (see Note 34). Repeat this step six times.
3. Transfer supernatant above settled glass beads to a new 1.5 mL
microtube using P200 PIPETMAN, and place tubes on ice.
The supernatant is the first cell extract.
184 Jer-Sheng Lin and Erh-Min Lai
4 Notes
13. This is a very critical step as the excessive speed may cause yeast
cell breakage and reduce the transformation efficiency.
14. Yeast competent cells must be freshly prepared to maintain
high transformation efficiency.
15. All steps of PEG/LiAc-mediated transformation of yeast
should be carried out in a laminar flow under aseptic
conditions.
16. We recommend pretreating ssDNA at 100 °C for only
10–15 min. Prolonged heating may cause instability of ssDNA.
17. This step should be carried out on ice to maintain low
temperature.
18. We recommend using 200 ng/plasmid for PEG/LiAc-
mediated cotransformation of yeast.
19. We recommend vortexing mixture for only 1 s before incuba-
tion in incubator at 30 °C for 30 min. LiAc-PEG solution can
be prepared during incubation time.
20. LiAc-PEG solution is very sticky, so it is better to use blunt-
end tip by cutting the end of a regular pipette tip to draw the
solution. This step is very critical and must be done within
2 min. Otherwise, the following resuspending step will be dif-
ficult to perform. Therefore, avoid handling more than ten
samples at the same time.
21. Resuspend cell mixture immediately by vortexing using maxi-
mum speed for 2–3 s.
22. Before heat shock, we recommend gently shaking microtube
several times to mix cell mixture well.
23. Pellet cells directly by centrifugation at 14,500 × g for 1 min.
It is not necessary to incubate cells on ice before
centrifugation.
24. Final cell suspension can be stored overnight at 4 °C before
use.
25. In general, we recommend picking six to eight single colonies
for further analysis. If possible, choose relatively large colonies,
which usually correlate with high protein expression levels.
26. We highly recommend using a flat toothpick (750 flat tooth-
picks, Diamond Brands) to patch single colony on selection
plate. The use of a sharp toothpick will often cause the break-
age of the agar surface. The yeast cells are ready for further
protein–protein interactions test when the yeast cells have
grown nicely after 2–3 days of incubation.
27. Use a flat toothpick to patch cells on selection plate. In gen-
eral, it is recommended to patch three individual colonies for
186 Jer-Sheng Lin and Erh-Min Lai
Acknowledgements
References
1. Fields S, Song O (1989) A novel genetic sys- VirB9 in the membranes of Agrobacterium
tem to detect protein–protein interactions. tumefaciens. J Bacteriol 179:1211–1218
Nature 340:245–246 11. Das A, Anderson LB, Xie YH (1997)
2. Stasi M, De Luca M, Bucci C (2015) Two- Delineation of the interaction domains of
hybrid-based systems: powerful tools for inves- Agrobacterium tumefaciens VirB7 and VirB9
tigation of membrane traffic machineries. by use of the yeast two-hybrid assay. J Bacteriol
J Biotechnol 202:105–117 179:3404–3409
3. Clontech (2007) Matchmaker™ GAL4 two- 12. Das A, Xie YH (2000) The Agrobacterium
hybrid system 3 & libraries user manual. T-DNA transport pore proteins VirB8, VirB9,
http://www.clontech.com/images/pt/ and VirB10 interact with one another.
PT3247-1.PDF J Bacteriol 182:758–763
4. Chien CT, Bartel PL, Sternglanz R, Fields S 13. Ma LS, Lin JS, Lai EM (2009) An IcmF family
(1991) The two-hybrid system: a method to protein, ImpLM, is an integral inner mem-
identify and clone genes for proteins that inter- brane protein interacting with ImpKL, and its
act with a protein of interest. Proc Natl Acad walker a motif is required for type VI secretion
Sci U S A 88:9578–9582 system-mediated Hcp secretion in
5. Causier B, Davies B (2002) Analysing protein– Agrobacterium tumefaciens. J Bacteriol
protein interactions with the yeast two-hybrid 191:4316–4329
system. Plant Mol Biol 50:855–870 14. Lin JS, Ma LS, Lai EM (2013) Systematic dis-
6. Petschnigg J, Groisman B, Kotlyar M, Taipale section of the Agrobacterium type VI secretion
M, Zheng Y et al (2014) The mammalian- system reveals machinery and secreted compo-
membrane two-hybrid assay (MaMTH) for nents for subcomplex formation. PLoS One
probing membrane–protein interactions in 8:e67647
human cells. Nat Methods 11:585–592 15. Bonemann G, Pietrosiuk A, Diemand A,
7. Reece-Hoyes JS, Barutcu AR, McCord RP, Zentgraf H, Mogk A (2009) Remodelling of
Jeong JS, Jiang L et al (2011) Yeast one-hybrid VipA/VipB tubules by ClpV-mediated thread-
assays for gene-centered human gene regulatory ing is crucial for type VI protein secretion.
network mapping. Nat Methods 8:1050–1052 EMBO J 28:315–325
8. Reece-Hoyes JS, Marian Walhout AJ (2012) 16. Lossi NS, Manoli E, Forster A, Dajani R,
Yeast one-hybrid assays: a historical and techni- Pape T et al (2013) The HsiB1C1 (TssB-
cal perspective. Methods 57:441–447 TssC) complex of the Pseudomonas aerugi-
9. Tsai YL, Chiang YR, Narberhaus F, Baron C, nosa type VI secretion system forms a
Lai EM (2010) The small heat-shock protein bacteriophage tail sheathlike structure. J Biol
HspL is a VirB8 chaperone promoting type IV Chem 288:7536–7548
secretion-mediated DNA transfer. J Biol Chem 17. Ito H, Fukuda Y, Murata K, Kimura A
285:19757–19766 (1983) Transformation of intact yeast cells
10. Baron C, Thorstenson YR, Zambryski PC treated with alkali cations. J Bacteriol
(1997) The lipoprotein VirB7 interacts with 153:163–168
Chapter 15
Abstract
Identifying protein–protein interactions between the machine components of bacterial secretion systems
and their cognate substrates is essential. Establishing which component and substrate interactions are
direct or indirect further facilitates (1) advancing the architecture and assembly of the machines and (2)
understanding the substrates’ translocation mechanistics. Currently, though biochemical means exist for
identifying such direct interactions, they primarily remain in vitro and are quite labor intensive. Thus,
adopting genetic approaches to help visualize these interactions in vivo is quick and advantageous. Here I
describe bimolecular fluorescence complementation and cytology-based two-hybrid assays that could easily
be adopted to understand the bacterial secretions systems.
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_15, © Springer Science+Business Media LLC 2017
189
190 Krishnamohan Atmakuri
2 Materials
3 Methods
3.1 Bimolecular 1. Select fluorescent protein, its halves, and appropriate fusion
Fluorescence sites (see Notes 1 and 2).
Complementation 2. Select appropriate controls (see Note 3).
3. Amplify required DNA fragment by PCR (see Note 4).
4. Use standard restriction-ligation procedures to clone PCR
amplicons into appropriate expression vectors (see Note 5).
5. Transform ligation mixture into electro- or chemically competent
bacteria (see Notes 6 and 7).
Cytology Two-Hybrid 193
3.2 Cytology-Based 1. Select fluorescent protein, its halves, and appropriate fusion
Two-Hybrid sites (see Notes 13 and 14).
2. Select appropriate controls (see Note 15).
3. Amplify the required DNA fragment by PCR (see Note 4).
4. Use standard restriction-ligation procedures to clone PCR
amplicons into appropriate expression vectors (see Note 5).
5. Transform ligation mixture into electro- or chemically competent
bacteria (see Notes 6 and 7).
6. Inoculate four to five colonies from freshly transformed bacteria
separately into required growth media and grow under
required conditions to approximately an OD (optical density
at A600 nm) of 0.1.
7. Induce expression of proteins under study with appropriate
inducers for required duration (see Note 8).
8. Wash a few hundred cells with fresh media (to stop induction).
9. Observe cells under fluorescent microscope (see Notes 9–11).
10. Perform image analyses using ImageJ or commercially available
software that comes with the fluorescent microscopes of most
companies (see Note 16).
4 Notes
Acknowledgments
References
1. Ding Z, Atmakuri K, Christie PJ (2003) The 7. Kodama Y, Hu CD (2012) Biomolecular fluo-
outs and ins of bacterial type IV secretion sub- rescence complementation (BiFC): a 5-year
strates. Trends Microbiol 11:527–535 update and future perspectives. BioTechniques
2. Ding Z, Zhao Z, Jakubowski SJ, Atmakuri K, 53:285–298
Margolin W, Christie PJ (2002) A novel 8. Kerppola TK (2008) Bimolecular Fluorescence
cytology-based, two-hybrid screen for bacteria Complementation (BiFC) analysis as a probe of
applied to protein–protein interaction studies protein interactions in living cells. Annu Rev
of a type IV secretion system. J Bacteriol Biophys 37:465–487
184:5572–5582 9. Hu CD, Chinenov Y, Kerppola TK (2002)
3. Taylor KW, Kim JG, Su XB, Aakre CD, Roden Visualization of interactions among bZIP and
JA, Adama CM, Mudgett MB (2012) Tomato Rel proteins in living cells using bimolecular
TFT1 is required for PAPMP-triggered immu- fluorescence complementation. Mol Cell
nity and mutations that prevent T3S effector 9:789–798
XopN from binding to TFT1 attenuate 10. Hu CD, Kerppola TK (2003) Simultaneous
Xanthomonas virulence. PLoS Pathog visualization of multiple protein interactions in
8:e1002768 living cells using multicolor fluorescence com-
4. Atmakuri K, Ding Z, Christie PJ (2003) VirE2, plementation analysis. Nat Biotechnol 21:
a type IV secretion substrate, interacts with the 539–545
VirD4 transfer protein at cell poles of 11. Jares-Erijman EA, Jovin TM (2003) FRET
Agrobacterium tumefaciens. Mol Microbiol imaging. Nat Biotechnol 21:1387–1395
49:1699–1713 12. Kerppola TK (2006) Design and
5. Kerppola TK (2006) Visualization of molecu- Implementation of Bimolecular Fluorescence
lar interactions by fluorescence complementa- Complementation (BiFC) assays for the visual-
tion. Nat Rev Mol Cell Biol 7:449–456 ization of protein interactions in living cells.
6. Zhang XE, Cui Z, Wang D (2016) Sensing of Nat Protoc 1:1278–1286
biomolecular interactions using fluorescence 13. Shaner NC, Steinbach PA, Tsein RY (2005)
complementing systems in living cells. Biosens A guide to choosing fluorescent proteins. Nat
Bioelectron 76:243–250 Methods 2:905–909
Chapter 16
Abstract
In transenvelope multiprotein machines such as bacterial secretion systems, protein–protein interactions not
only occur between soluble domains but might also be mediated by helix–helix contacts in the inner mem-
brane. Here we describe genetic assays commonly used to test interactions between transmembrane α-helices
in their native membrane environment. These assays are based on the reconstitution of dimeric regulators
allowing the control of expression of reporter genes. We provide detailed protocols for the TOXCAT and
GALLEX assays used to monitor homotypic and heterotypic transmembrane helix–helix interactions.
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_16, © Springer Science+Business Media LLC 2017
199
200 Laureen Logger et al.
1.1 Monitoring TMH Methods of testing the homodimerization of TMHs, such as the λ cI
Homotypic repressor and TOXCAT assays, are based on the one-hybrid reporter
Interactions fusion approach.
The cI transcriptional regulator represses the expression of
early promoters of the bacteriophage λ genome. Repression only
occurs when cI dimerizes, a behavior conferred by the C-terminal
domain. The λcI repressor assay is therefore based on the reconsti-
tution of a dimeric λcI repressor by two interacting fragments [8–10].
The construct consists of a fusion between the monomeric
N-terminal DNA-binding domain of λcI (called cI’) with the TMH
(Fig. 1a). TMH-mediated cI’ dimerization induces binding of cI
to its operator sequence, allowing repression of phage λ early
genes, hence conferring protection against superinfection by phage
λ (Fig. 1a). The cI repressor assay has been successfully used to
demonstrate that the T2SS XcpR and T4SS VirB4 and VirB11
proteins oligomerize [11–13].
The TOXCAT assay is based on the characteristics of the
Vibrio cholerae ToxR regulator: a strict dimerization-dependent
transcriptional activator consisting of an N-terminal helix-turn-
helix DNA- binding domain and a C-terminal dimerization
domain. The construct consists of a fusion in which the TMH is
inserted between the monomeric ToxR DNA-binding domain
and the MalE periplasmic protein (Fig. 1b). By supporting growth
on maltose-minimal media, MalE makes it possible to verify that
the TMH is properly inserted. TMH-mediated ToxR dimeriza-
tion induces binding of ToxR on its operator sequence, allowing
transcription of a reporter gene. In the initial ToxR system, the
reporter gene is lacZ [14], while the TOXCAT assay uses the cat
gene [15] (Fig. 1b). Hence, dimerization of the TMH could then
be assessed by measuring the β-galactosidase and chloramphenicol
acetyltransferase (resistance to chloramphenicol) activities [16].
The TOXCAT assay has been successfully used to provide evi-
dence that the TMH of the T4SS VirB10 subunit oligomerizes
[5]. Further improvements of the ToxR and TOXCAT assays have
been published [17–19].
Protein-Protein Interaction in the Membrane 201
Fig. 1 Schematic representation of the assays for monitoring TMH homotypic interactions. (a) cI repressor
assay. The TMH of interest (orange) is fused to the cI DNA-binding domain (green). Reconstitution of the cI
dimer results in the repression of early phage λ genes (blue). Repression of phage λ genes confers protection
against phage λ infection. (b) ToxR and TOXCAT assays. The TMH of interest (orange) is fused between the
Vibrio cholerae ToxR DNA-binding domain (red) and the MalE protein (blue). Reconstitution of the ToxR dimer
results in the expression of the reporter genes (blue, lacZ for the ToxR assay, cat for the TOXCAT assay)
1.2 Monitoring TMH Methods of testing the heterodimerization of TMHs, such as the
Heterotypic GALLEX and BACTH assays, are based on the two-hybrid
Interactions reporter fusion approach.
The GALLEX assay is based on the reconstitution of a dimeric
LexA transcriptional repressor by two interacting TMHs. The con-
struct consists of a fusion in which each TMH is inserted between
the monomeric LexA N-terminal DNA-binding domain and the
MalE periplasmic protein. The elegant improvement is that one of
the two TMHs is fused to the wild-type LexA N-terminal
domain (LexAWT), whereas the second TMH is fused to a LexA
N-terminal domain variant bearing a mutation in the DNA-binding
motif (LexA408), allowing recognition of a different operator
sequence (op408). Formation of helix heterodimers induces binding
of LexA/LexA408 on a dual operator sequence (opWT/op408), allow-
ing repression of a reporter gene [20–22] (Fig. 2a).
The BACTH assay is based on the reconstitution of the adenyl-
ate cyclase activity conferred by the T18 and T25 domains of the
Bordetella pertussis Cya protein [23–25] (Fig. 2b). Widely
202 Laureen Logger et al.
Fig. 2 Schematic representation of assays for monitoring TMH heterotypic interactions. (a) GALLEX assay. The
first TMH of interest (orange) is fused between the wild-type LexA DNA-binding domain (WT LexA) and MalE,
whereas the second TMH (blue) is fused between the LexA408 variant (LexA 408) and MalE. Reconstitution of
the LexAWT/LexA408 dimer results in the repression of the reporter gene (blue). (b) BACTH assays. The first TMH
of interest (orange) is fused to the T18 domain of the B. pertussis adenylate cyclase, whereas the second TMH
(blue) is fused to the T25 of adenylate cyclase. Reconstitution of the T18/T25 adenylate cyclase results in the
production of cyclic adenosine monophosphate (cAMP). Binding of cAMP to the catabolite activator protein
(CAP) induces the expression of the reporter gene (blue)
2 Material
3 Methods
3.1 Monitoring TMH 1. Clone the DNA fragment corresponding to the TMH to be stud-
Homotypic ied into the pcckan vector to yield a plasmid producing the ToxR’-
Interactions: TMH-MalE fusion protein. Before testing the homodimerization
The TOXCAT Assay of the TMH, verify that your fusion protein is properly produced
(steps 3–8) and inserted in the inner membrane (steps 9 and
10). The dimerization of the TMH is then assessed by the disk
diffusion assay (steps 11–16) and quantified by measuring the
chloramphenicol acetyltransferase activity (steps 17–26).
Protein-Protein Interaction in the Membrane 205
3.2 Monitoring TMH 1. Clone the DNA fragment corresponding to the first TMH to
Heterotypic be studied (TMH1) into the pBLM100 vector to yield a
Interactions: pBR322 derivative plasmid producing the LexAWT’-TMH1-
The GALLEX Assay MalE fusion protein. Clone the DNA fragment corresponding
to the second TMH to be studied (TMH2) into the pALM148
vector to yield a pACYC184 derivative plasmid producing the
LexA408’-TMH2-MalE fusion protein. Before testing the het-
erodimerization of the TMH, verify that your fusion protein is
properly produced (steps 3–8) and inserted in the inner mem-
brane (steps 9 and 10). The dimerization of the TMH is then
assessed on LB-X-Gal plates (steps 11–14) and quantitated by
measuring the β-galactosidase activity (steps 15–22).
2. Transform the empty pBLM100 and pALM148 vectors as well
as the pBLM100-TMH1 and pALM148-TMH2 constructs
into NT326 or MM39 E. coli competent cells. Select on LB
plates supplemented with ampicillin (pBLM100 derivatives) or
tetracyclin (pALM148 derivatives).
3. Pick a single colony of each transformation and grow cells in
3 mL LB medium supplemented with IPTG and ampicillin or
tetracyclin until an OD600 of 0.8 is reached.
4. Harvest 2 mL of cells by centrifugation at 4000 × g for 5 min.
5. Discard supernatants and resuspend cell pellets into 20 μL of
SDS-PAGE loading buffer.
6. Boil samples for 10 min at 96 °C.
7. Separate proteins by SDS-PAGE and transfer onto nitrocellu-
lose membrane using your favorite protocol.
8. Use western blotting to immunodetect your fusion protein
using commercial anti-MalE (anti-MBP) antibodies.
9. Streak 20 μL of the bacterial culture obtained at step 3 in
Subheading 3.2 onto M9-maltose medium.
10. After incubation for 48 h at 37 °C, verify that your strain grew
on M9-maltose medium.
11. Cotransform pBLM100 and pBLM100-TMH1 vectors in
combination with pALM148 and pALM148-TMH2 vectors
into SU202 E. coli competent cells (see Note 7). Select on LB
plates supplemented with ampicillin and tetracyclin.
Protein-Protein Interaction in the Membrane 207
4 Notes
Acknowledgements
References
1. Lallemand M, Login FH, Guschinskaya N, 2. Ma LS, Lin JS, Lai EM (2009) An IcmF family
Pineau C, Effantin G, Robert X, Shevchik VE protein, ImpLM, is an integral inner membrane
(2013) Dynamic interplay between the peri- protein interacting with ImpKL, and its walker
plasmic and transmembrane domains of GspL a motif is required for type VI secretion system-
and GspM in the type II secretion system. mediated Hcp secretion in Agrobacterium
PLoS One 8:e79562 tumefaciens. J Bacteriol 191:4316–4329
Protein-Protein Interaction in the Membrane 209
3. Aschtgen MS, Gavioli M, Dessen A, Lloubès 15. Russ WP, Engelman DM (1999) TOXCAT: a
R, Cascales E (2010) The SciZ protein anchors measure of transmembrane helix association in
the enteroaggregative Escherichia coli Type VI a biological membrane. Proc Natl Acad Sci U S
secretion system to the cell wall. Mol Microbiol A 96:863–868
75:886–899 16. Joce C, Wiener A, Yin H (2011) Transmembrane
4. Durand E, Zoued A, Spinelli S, Watson PJ, domain oligomerization propensity determined
Aschtgen MS, Journet L, Cambillau C, Cascales by ToxR assay. J Vis Exp 51
E (2012) Structural characterization and oligo- 17. Lindner E, Langosch D (2006) A ToxR-based
merization of the TssL protein, a component dominant-negative system to investigate het-
shared by bacterial type VI and type IVb secre- erotypic transmembrane domain interactions.
tion systems. J Biol Chem 287:14157–14168 Proteins 65:803–807
5. Garza I, Christie PJ (2013) A putative trans- 18. Lindner E, Unterreitmeier S, Ridder AN,
membrane leucine zipper of agrobacterium Langosch D (2007) An extended ToxR
VirB10 is essential for T-pilus biogenesis but not POSSYCCAT system for positive and negative
type IV secretion. J Bacteriol 195:3022–3034 selection of self-interacting transmembrane
6. Schneider D, Finger C, Prodöhl A, Volkmer domains. J Microbiol Methods 69:298–305
T (2007) From interactions of single trans- 19. Lis M, Blumenthal K (2006) A modified,
membrane helices to folding of alpha-helical dual reporter TOXCAT system for monitor-
membrane proteins: analyzing transmembrane ing homodimerization of transmembrane
helix-helix interactions in bacteria. Curr segments of proteins. Biochem Biophys Res
Protein Pept Sci 8:45–61 Commun 339:321–324
7. Fink A, Sal-Man N, Gerber D, Shai Y (2012) 20. Schneider D, Engelman DM (2003) GALLEX,
Transmembrane domains interactions within a measurement of heterologous association of
the membrane milieu: principles, advances transmembrane helices in a biological mem-
and challenges. Biochim Biophys Acta brane. J Biol Chem 278:3105–3111
1818:974–983 21. Cymer F, Sanders CR, Schneider D (2013)
8. Hu JC (1995) Repressor fusions as a tool to Analyzing oligomerization of individual trans-
study protein-protein interactions. Structure membrane helices and of entire membrane
3:431–433 proteins in E. coli: a hitchhiker’s guide to
9. Leeds JA, Beckwith J (1998) Lambda repressor GALLEX. Methods Mol Biol 932:259–276
N-terminal DNA-binding domain as an assay 22. Tome L, Steindorf D, Schneider D (2013)
for protein transmembrane segment interac- Genetic systems for monitoring interactions
tions in vivo. J Mol Biol 280:799–810 of transmembrane domains in bacterial mem-
10. Leeds JA, Beckwith J (2000) A gene fusion branes. Methods Mol Biol 1063:57–91
method for assaying interactions of protein 23. Karimova G, Pidoux J, Ullmann A, Ladant D
transmembrane segments in vivo. Methods (1998) A bacterial two-hybrid system based on
Enzymol 2327:165–175 a reconstituted signal transduction pathway.
11. Turner LR, Olson JW, Lory S (1997) The Proc Natl Acad Sci U S A 95:5752–5756
XcpR protein of Pseudomonas aeruginosa 24. Ladant D, Karimova G (2000) Genetic systems
dimerizes via its N-terminus. Mol Microbiol for analyzing protein-protein interactions in
26:877–887 bacteria. Res Microbiol 151:711–720
12. Dang TA, Zhou XR, Graf B, Christie PJ (1999) 25. Battesti A, Bouveret E (2012) The bacterial
Dimerization of the agrobacterium tumefaciens two-hybrid system based on adenylate cyclase
VirB4 ATPase and the effect of ATP-binding reconstitution in Escherichia coli. Methods
cassette mutations on the assembly and func- 58:325–334
tion of the T-DNA transporter. Mol Microbiol 26. Karimova G, Dautin N, Ladant D (2005)
32:1239–1253 Interaction network among Escherichia coli
13. Rashkova S, Zhou XR, Chen J, Christie PJ membrane proteins involved in cell division
(2000) Self-assembly of the Agrobacterium as revealed by bacterial two-hybrid analysis.
tumefaciens VirB11 traffic ATPase. J Bacteriol J Bacteriol 187:2233–2243
182:4137–4145 27. Sivanesan D, Hancock MA, Villamil Giraldo
14. Langosch D, Brosig B, Kolmar H, Fritz HJ AM, Baron C (2010) Quantitative analy-
(1996) Dimerisation of the glycophorin A sis of VirB8-VirB9-VirB10 interactions pro-
transmembrane segment in membranes probed vides a dynamic model of type IV secretion
with the ToxR transcription activator. J Mol system core complex assembly. Biochemistry
Biol 263:525–530 49:4483–4493
210 Laureen Logger et al.
28. Cisneros DA, Bond PJ, Pugsley AP, Campos platform for tail tube/sheath polymerization.
M, Francetic O (2012) Minor pseudopilin self- PLoS Genet 11:e1005545
assembly primes type II secretion pseudopilus 34. Zoued A, Durand E, Brunet YR, Spinelli S, Douzi
elongation. EMBO J 31:1041–1053 B, Guzzo M, Flaugnatti N, Legrand P, Journet L,
29. Georgiadou M, Castagnini M, Karimova G, Fronzes R, Mignot T, Cambillau C, Cascales E
Ladant D, Pelicic V (2012) Large-scale study (2016) Priming and polymerization of a bacterial
of the interactions between proteins involved contractile tail structure. Nature 531:59–63
in type IV pilus biology in Neisseria meningiti- 35. Llosa M, Zunzunegui S, de la Cruz F (2003)
dis: characterization of a subcomplex involved Conjugative coupling proteins interact with cog-
in pilus assembly. Mol Microbiol 84:857–873 nate and heterologous VirB10-like proteins while
30. Zoued A, Durand E, Bebeacua C, Brunet YR, exhibiting specificity for cognate relaxosomes.
Douzi B, Cambillau C, Cascales E, Journet L Proc Natl Acad Sci U S A 100:10465–10470
(2013) TssK is a trimeric cytoplasmic protein 36. Segura RL, Aguila-Arcos S, Ugarte-Uribe B,
interacting with components of both phage- Vecino AJ, de la Cruz F, Goñi FM, Alkorta I
like and membrane anchoring complexes of the (2013) The transmembrane domain of the
type VI secretion system. J Biol Chem T4SS coupling protein TrwB and its role in
288:27031–27041 protein-protein interactions. Biochim Biophys
31. Pais SV, Milho C, Almeida F, Mota LJ (2013) Acta 1828:2015–2025
Identification of novel type III secretion 37. Sawma P, Roth L, Blanchard C, Bagnard D,
chaperone- substrate complexes of Chlamydia Crémel G, Bouveret E, Duneau JP, Sturgis JN,
trachomatis. PLoS One 8:e56292 Hubert P (2014) Evidence for new homotypic
32. Pineau C, Guschinskaya N, Robert X, Gouet P, and heterotypic interactions between trans-
Ballut L, Shevchik VE (2014) Substrate recog- membrane helices of proteins involved in
nition by the bacterial type II secretion system: receptor tyrosine kinase and neuropilin signal-
more than a simple interaction. Mol Microbiol ing. J Mol Biol 426:4099–4111
94:126–140 38. Dimitrova M, Younès-Cauet G, Oertel-
33. Brunet YR, Zoued A, Boyer F, Douzi B, Buchheit P, Porte D, Schnarr M, Granger-
Cascales E (2015) The type VI secretion Schnarr M (1998) A new LexA-based genetic
TssEFGK-VgrG phage-like baseplate is system for monitoring and analyzing protein
recruited to the TssJLM membrane complex heterodimerization in Escherichia coli. Mol
via multiple contacts and serves as assembly Gen Genet 257:205–212
Chapter 17
Abstract
Proteins often do not function as single substances but rather as team players in a dynamic network.
Growing evidence shows that protein–protein interactions are crucial in many biological processes in living
cells. Genetic (such as yeast two-hybrid, Y2H) and biochemical (such as co-immunoprecipitation, co-IP)
methods are the methods commonly used at the beginning of a study to identify the interacting proteins.
Immunoprecipitation (IP), a method using a target protein-specific antibody in conjunction with Protein
A/G affinity beads, is a powerful tool to identify molecules that interact with specific proteins. Therefore,
co-IP is considered to be one of the standard methods of identifying or confirming the occurrence of
protein–protein interaction events in vivo. Co-IP experiments can identify proteins via direct or indirect
interactions or in a protein complex. Here, we use Agrobacterium type VI secretion system (T6SS) sheath
components TssB-TssC41 interaction as an example to describe the principle, procedure, and experimental
problems of co-IP.
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_17, © Springer Science+Business Media LLC 2017
211
212 Jer-Sheng Lin and Erh-Min Lai
Agarose
Bead
Protein
A/G
Agarose
Bead
Protein
A/G
Add antibody Add Protein
A/G beads
Protein sample
(Usually a cell lysate)
Discard
Centrifugation
Supernatant
Agarose
Bead
Protein
A/G
Wash
Non-bound sample
components
Sample buffer
Elute
Agarose
Bead
Protein
A/G
Fig. 1 Schematic diagram of principle of co-IP. Antigen-containing protein sample (usually a cell lysate), spe-
cific antibody, and affinity beads (usually protein A/G, which can specifically bind to conserved region of anti-
body) are added sequentially for binding reaction. The affinity beads with bound proteins are collected by
centrifugation. The supernatant containing unbound proteins is discarded and further washed away during
washing steps. Antibody and antigen are eluted with a buffer that dissociates proteins from affinity beads.
Purified protein complexes can be further used for immunoblot or other biochemical analysis
Co-Immunoprecipitation 213
TssB 15
Fig. 2 Co-IP analysis of TssB and TssC41 in A. tumefaciens. Total protein extracts
isolated from A. tumefaciens wild-type strain C58 treated with DTBP cross-linker
were solubilized by buffer containing 1% SDS, then diluted in Triton X-100-
containing solution for IP. Coprecipitated proteins were identified by western
blotting. Co-IP was also performed with antibody against RNA polymerase alpha
subunit (RpoA) as a negative control. The proteins analyzed and sizes of molecu-
lar weight standards are indicated on the left and right, respectively, and by
arrows when necessary (I input, IP immunoprecipitation) (reproduced from Ref.
[10]; no permission is required for reuse of content published from Public Library
of Science, PLOS
2 Materials
2.3 Protein Sample 1. Protein A-Sepharose™ CL4B (GE Healthcare Life Sciences)
Preclearing or equivalent (see Note 6).
2. 2 mL microtube.
3. Rotating wheel.
2.5 Purification 1. NP1 buffer supplemented with 1% Triton X-100 (see Note 7).
and Isolation 2. NP1 buffer supplemented with 0.1% Triton X-100.
of Protein Complexes
3. Elution buffer: 0.1 M glycine–HCl, pH 2.5.
4. 2× SDS sample buffer: 100 mM Tris–HCl, pH 6.8, 4% SDS,
20% glycerol, 5% 2-mercaptoethanol, 2 mM EDTA, 0.1 mg/
mL bromophenol blue (see Note 8).
3 Methods
3.3 Protein Sample The preclearing step will reduce the background noise caused by the
Preclearing adhesion of some protein components to the protein A-Sepharose.
and Coupling
of Antibody to Protein 1. For each 2 mL of the detergent-solubilized solution, add a 60
A/G Beads μL bed volume of protein A-Sepharose and incubate for 60 min
at room temperature with rotation (see Note 21).
2. Remove protein A-Sepharose with nonspecifically bound proteins
by centrifugation for 5 min at 5000 × g at 4 °C (see Note 22).
216 Jer-Sheng Lin and Erh-Min Lai
4 Notes
Acknowledgements
References
1. Kessler SW (1975) Rapid isolation of anti- tools.thermofisher.com/content/sfs/bro-
gens from cells with a staphylococcal protein chures/TR0064-Immunoprecipitation-guide.
A-antibody adsorbent: parameters of the inter- pdf.
action of antibody-antigen complexes with 4. Lee C (2007) Coimmunoprecipitation assay.
protein A. J Immunol 115:1617–1624 Methods Mol Biol 362:401–406
2. Kessler SW (1976) Cell membrane anti- 5. Aebersold R, Mann M (2003) Mass
gen isolation with the staphylococcal pro- spectrometry- based proteomics. Nature
tein A-antibody adsorbent. J Immunol 422:198–207
117:1482–1490 6. Gevaert K, Vandekerckhove J (2000) Protein
3. ThermoFisher (2009) Immunoprecipitation identification methods in proteomics.
(IP) technical guide and protocols. https:// Electrophoresis 21:1145–1154
Co-Immunoprecipitation 219
7. Atmakuri K, Cascales E, Christie PJ (2004) 10. Lin JS, Ma LS, Lai EM (2013) Systematic dissec-
Energetic components VirD4, VirB11 and tion of the Agrobacterium type VI secretion sys-
VirB4 mediate early DNA transfer reactions tem reveals machinery and secreted components
required for bacterial type IV secretion. Mol for subcomplex formation. PLoS One 8:e67647
Microbiol 54:1199–1211 11. Ma LS, Narberhaus F, Lai EM (2012) IcmF
8. Atmakuri K, Cascales E, Burton OT, Banta family protein TssM exhibits ATPase activity
LM, Christie PJ (2007) Agrobacterium ParA/ and energizes type VI secretion. J Biol Chem
MinD-like VirC1 spatially coordinates early 287:15610–15621
conjugative DNA transfer reactions. EMBO 12. Elion EA (2007) Detection of protein-protein
J 26:2540–2551 interactions by coprecipitation. Curr Protoc
9. Anderson LB, Hertzel AV, Das A (1996) Immunol Chapter 8:Unit 8.7
Agrobacterium tumefaciens VirB7 and VirB9 13. Cascales E, Christie PJ (2004) Definition of a
form a disulfide-linked protein complex. Proc bacterial type IV secretion pathway for a DNA
Natl Acad Sci U S A 93:8889–8894 substrate. Science 304:1170–1173
Chapter 18
Abstract
The discovery of protein–protein interaction networks can lead to the unveiling of protein complex(es)
forming cellular machinerie(s) or reveal component proteins of a specific cellular pathway. Deciphering
protein–protein interaction networks therefore contributes to a deeper understanding of how cells func-
tion. Here we describe the protocol to perform tandem affinity purification (TAP) in bacteria, which
enables the identification of the partners of a bait protein under native conditions. This method consists in
two sequential steps of affinity purification using two different tags. For that purpose, the bait protein is
translationally fused to the TAP tag, which consists of a calmodulin binding peptide (CBP) and two immu-
noglobulin G (IgG) binding domains of Staphylococcus aureus protein A (ProtA) that are separated by the
tobacco etch virus (TEV) protease cleavage site. After the first round of purification based on the binding
of ProtA to IgG coated beads, TEV protease cleavage releases CBP-tagged bait-protein along with its
partners for a second round of purification on calmodulin affinity resin and leaves behind protein contami-
nants bound to IgG. Creating the TAP-tag translational fusion at the chromosomal locus allows detection
of protein interactions occurring in physiological conditions.
Key words Protein–protein interaction, Protein complex, Affinity purification, Tandem affinity puri-
fication (TAP), Calmodulin binding peptide (CBP), ProtA, Tobacco etch virus (TEV), Escherichia
coli, Salmonella
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_18, © Springer Science+Business Media LLC 2017
221
222 Julie P.M. Viala and Emmanuelle Bouveret
specific needs. For example, the extensive washes and the duration
of the procedure only allow for the recovery of relatively stable com-
plexes. For the detection of more transient or unstable interactions,
a cross-linking procedure can be applied before purification [13].
224 Julie P.M. Viala and Emmanuelle Bouveret
Fig. 2 Creation of a C-terminal TAP-tag translational fusion at chromosomal locus. Scheme of TAP-tag trans-
lational fusion at chromosomal locus (a) and corresponding nucleotidic and protein TAP-tag sequences (b).
CBP sequence is in clear purple; uppercase at the beginning of the nucleotidic CBP sequence corresponds to
the primer sequence mentioned in Subheading 3.1, TEVprotease cleavage site is in yellow, and ProtA sequence
is in dark purple, with stop codon in red
2 Materials
2.2 Protein 1. Phosphate buffered saline (PBS): 8 g NaCl, 0.2 g KCl, 0.2 g
Cytoplasmic Extract KH2PO4, 2.9 g Na2HPO4, make up to 1 L with distilled water.
Autoclave and store at room temperature.
2. 10% Nonidet P-40 (NP-40 or Igepal): Mix 10 mL NP-40 in
90 mL distilled water, pass through a 0.2 μm filter, and store at
room temperature (see Note 3).
3. ProtA binding buffer: 10 mM Tris–HCl, pH 8, 150 mM NaCl,
0.1% NP-40. Approximately 50 mL will be required per sample
TAP in Bacteria 227
3 Methods
3.1 Verification 1. Prepare a cytoplasmic or a crude protein extract (see Note 8).
of Expression of TAP- 2. Load 10 μg of protein extract (or proteins corresponding to a
Tag Translational bacterial sample of 0.3 OD600 unit) on a SDS-PAGE and pro-
Fusion by Western ceed to transfer and western blot to verify production of hybrid
Blot protein (see Note 9).
3. Perform a one-step western blot using PAP antibody (see
Note 10) and using an appropriate substrate to detect horse-
radish peroxidase activity (see Note 11).
3.3 Tandem Affinity From here, carry out all procedures with gloves to avoid contami-
Purification nation of your sample(s) with keratine.
1. Put 200 μL IgG Sepharose beads in a disposable chromatog-
raphy column and wash by gravity with 5 mL ProtA binding
buffer.
2. Binding of ProtA tag to IgG Sepharose beads. After washing the
beads, close the bottom of the chromatography column, and,
using a pipette, transfer 9 mL of the cytoplasmic protein extract.
Close the top of the column and put on a wheel for 2 h at 4 °C.
3. Remove first the top plug of the column and then the bottom
one. Leave the unbound material flow by gravity and discard.
4. Wash three times the IgG beads with 10 mL of ProtA binding
buffer.
5. TEVprotease cleavage. Close bottom of column, fill it with 1 mL
TEV cleavage buffer and 100 units of AcTEV™ protease. Close
top of column and put on wheel at room temperature for 1 h.
6. Remove top and bottom plugs and recover elution fraction by
gravity. Add additional 200 μL TEV cleavage buffer in column
to recover as much material as possible from sides of column.
7. Add 3 mL calmodulin binding buffer and 3 μL 1 M CaCl2
(see Note 13) to elution fraction.
8. Binding by the CBP tag part on calmodulin affinity resin. Put
200 μL calmodulin affinity resin in a new disposable chroma-
tography column, and wash it with 5 mL calmodulin binding
buffer. Then close bottom of column.
9. Add the 4.2 mL of the elution fraction (obtained at steps 6
and 7). Close top of column and put on wheel for 1 h at 4 °C.
10. Remove first the top plug of the column and then the bottom
one. Leave unbound material to flow by gravity and discard.
11. Wash three times calmodulin affinity resin with 10 mL calmod-
ulin binding buffer.
12. Elution. Elute with five times 200 μL of calmodulin elution
buffer.
13. Pool fractions 2, 3, and 4 and proceed to TCA precipitation of
elution fraction 1, pooled fractions 2, 3, and 4, and fraction 5.
230 Julie P.M. Viala and Emmanuelle Bouveret
3.4 TCA Precipitation 1. To each of the eluted protein samples add 1/100th of 16 mg/
mL sodium deoxycholate. Vortex and leave on ice 30 min.
2. Add TCA to 10% final. Vortex and leave on ice 30 min.
3. Centrifuge 15 min, 15,000 × g at 4 °C.
4. Wash twice with TCA washing buffer.
5. Leave pellets to dry on bench and resuspend in 20 μL protein
loading buffer 1×.
3.5 Analysis 1. Load totality of samples on SDS-PAGE (see Note 14) and
by SDS-PAGE stain with coomassie blue.
and Mass 2. Unstain and then rinse with distilled water.
Spectrometry
3. Cut bands to identify partner proteins by mass spectrometry
(see Note 15).
4 Notes
6. Add DTT to the volume of buffer you will need when starting
the experiment. DTT is necessary for TEV activity.
7. Add β-mercaptoethanol to the volume of buffer you will need
when starting the experiment.
8. Also plan to prepare a protein extract of an untagged strain as
the negative control of the experiment.
9. Translational TAP tag fusion adds 20 kDa to the mass of the
protein of interest; 3 kDa corresponds to the CBP tag and
15 kDa to the ProtA tag.
10. Immunoglobulins will bind the ProtA fragment of the TAP tag.
11. In our experience, the detection of a tagged protein in crude
extracts using this PAP antibody is mandatory for a successful
TAP purification.
12. A French press or cell disruptor might be more gentle for pre-
serving protein complexes.
13. The addition of extra CaCl2 is required to quench the EDTA
that was previously necessary for TEV protease activity.
14. Usually 12% SDS-PAGE allows visualization of low- and high-
molecular-weight proteins.
15. Use one blade per band.
References
12. Babu M, Butl G, Pogoutse O, Li J, Greenblatt JF plex that participates in mRNA degradation.
et al (2009) Sequential peptide affinity purifica- EMBO J 19:1661–1671
tion system for the systematic isolation and iden- 16. So EC, Schroeder GN, Carson D, Mattheis C,
tification of protein complexes from Escherichia Mousnier A et al (2016) The Rab-binding pro-
coli. Methods Mol Biol 564:373–400 files of bacterial virulence factors during infec-
13. Stingl K, Schauer K, Ecobichon C, Labigne A, tion. J Biol Chem 291:5832–5843
Lenormand P et al (2008) In vivo interactome 17. Sana TG, Baumann C, Merdes A, Soscia C, Rattei
of Helicobacter pylori urease revealed by tan- T et al (2015) Internalization of Pseudomonas
dem affinity purification. Mol Cell Proteomics aeruginosa strain PAO1 into epithelial cells is
7:2429–2441 promoted by interaction of a T6SS effector with
14. Puig O, Caspary F, Rigaut G, Rutz B, the microtubule network. MBio 6:e00712
Bouveret E et al (2001) The tandem affinity 18. Battesti A, Bouveret E (2008) Improvement of
purification (TAP) method: a general proce- bacterial two-hybrid vectors for detection of
dure of protein complex purification. Methods fusion proteins and transfer to pBAD-tandem
24:218–229 affinity purification, calmodulin binding pep-
15. Bouveret E, Rigaut G, Shevchenko A, Wilm tide, or 6-histidine tag vectors. Proteomics
M, Seraphin B (2000) A Sm-like protein com- 8:4768–4771
Chapter 19
Abstract
To efficiently transport proteins into and across cellular membranes, specialized transport machineries
engage in recognition events with different domains of their client proteins, forming sequential intermedi-
ate complexes. The short-lived nature of these interactions poses a big challenge in the identification of the
key factors involved in transport reactions and their mechanism of action. Site-directed photocrosslinking
is a powerful method for the detection and accurate mapping of interacting protein domains. This chapter
describes a protocol that combines site-directed photocrosslinking to metabolic labeling of proteins and
lipids as a method to characterize, with temporal and spatial resolution, the interactions of a secretory
protein as it transverses the bacterial envelope.
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_19, © Springer Science+Business Media LLC 2017
233
234 Raffaele Ieva
2 Materials
2.2 Expression of a 1. 10× M9 salt (67.8 g/L Na2HPO4, 30 g/L KH2PO4, 5 g/L
EspP Variant NaCl, 10 g/L NH4Cl).
Containing Bpa and 2. M9 complete medium containing 1× M9 salt, 1 mM MgSO4,
Pulse-Chase Metabolic 0.1 mM CaCl2, 0.2% w/v glycerol, 40 μg/mL l-amino acids
Labeling of Cells Using (except methionine and cysteine).
35
S-Labeled Amino 3. One disposable 125 mL Erlenmeyer flask.
Acids
4.
A high-specific activity mixture of 35
S-cysteine and
35
S-methionine (1075–1175 Ci/mmol).
5. A stock solution of non-radiolabeled methionine and cysteine
(100 mM methionine, 100 mM cysteine).
6. 100 mM isopropyl-β-d-thio-galactoside (IPTG) stock solution.
7. Bpa (Bachem).
8. Water bath shaker.
2.3 Metabolic
1. One disposable 125 mL Erlenmeyer flask.
Labeling
with 32P-Labeled 2. Modified G56 medium (45 mM MES, pH 7.0, 10 mM KCl,
10 mM MgCl2, 15 mM (NH4)2SO4, 5 μg/L thiamine, 0.2%
Inorganic Phosphate
glycerol, 40 μg/mL l-amino acids).
and Expression
of an EspP Variant 3. A stock solution of 0.5 M KH2PO4.
Containing Bpa 4. Radioactive KH232PO4 (900–1100 mCi/mmol).
236 Raffaele Ieva
2.5 Immunoprecipita 1. Antisera specific for EspP, BamA, and BamB or other proteins
tion and Sodium of interest.
Dodecyl Sulfate 2. Protein solubilization buffer (15% glycerol, 200 mM Tris base,
(SDS)-Polyacrylamide 15 mM EDTA, 4% SDS, 2 mM phenylmethylsulfonyl fluoride
Gel Electrophoresis (PMSF)).
(PAGE) Analysis of 3. Radioimmunoprecipitation assay (RIPA) buffer (50 mM Tris–
Photocrosslinked HCl, pH 8, 150 mM NaCl, 1% Igepal CA-630, 0.5% sodium
Proteins deoxycholate, 0.1% SDS).
4. Staphylococcus aureus protein A-Sepharose beads.
5. SDS-polyacrylamide gels.
6. 4× SDS-PAGE sample buffer (8% SDS, 40% glycerol, 240 mM
Tris–HCl, pH 6.8, 1.6% β-mercaptoethanol, 0.04% bromo-
phenol blue).
7. Gel drying system.
8. Storage phosphor screen.
3 Method
3.1 Strategy Design 1. Clone a construct encoding the selected protein of interest
and Plasmid under the control of an inducible promoter in an expression
Construction plasmid that can be propagated in the desired bacterial model
organism. In this example, RI23, an RB11-derived plasmid,
harbors a construct encoding EspP(586TEV) (see Note 1)
under the control of the IPTG-inducible lac promoter. The
selected model organism is the laboratory E. coli strain AD202,
an MC4100-derived strain that lacks the gene encoding the
outer membrane protease OmpT [13].
2. Use a PCR-based site-specific mutagenesis approach to incor-
porate an amber codon at a specific position of the protein of
interest. In this example, the mutagenesis approach is con-
ducted on pRI23 to replace with an amber codon the EspP
Trp 1149 codon, generating pRI23-1149Bpa (see Note 2).
3. Digest the parental DNA used as template in the PCR with the
restriction enzyme DpnI.
4. Transform ultracompetent E. coli cells using a small aliquot of
the amplified DNA product.
In vivo Site-Directed Photocrosslinking 237
3.2 Preparation of Two methodologies of metabolic labeling are used (Fig. 1). In one
Cell Cultures for EspP culture 35S labeling is conducted following a “pulse-chase” proce-
Expression and dure, in which all newly synthesized proteins become radiolabeled by
Metabolic Labeling exposing cells for a short time period to 35S-methionine and
35
S-cysteine (pulse phase). Subsequently, the addition of an excess of
non-radiolabeled (“cold”) methionine and cysteine prevents further
incorporation of radiolabeled amino acids into proteins, thereby end-
ing the pulse phase. The fate of radiolabeled proteins can be chased
over time (chase phase). By combining 35S-labeling with site-specific
Fig. 1 Flow diagram of the illustrated experimental procedure. Two cultures are conducted in parallel. One
culture is subjected to pulse-chase labeling using 35S-methionine and 35S-cysteine in order to label all newly
synthesized proteins (left). The other culture is subjected to labeling with 32P-inorganic phosphate in order to
label cellular phospholipids (right)
238 Raffaele Ieva
–– P +UV.
32
Fig. 2 Transient and stable interactions detected at a specific position of the EspP β domain. AD202 cells
transformed with plasmids RI23-1149Bpa and pDULE-pBpa are subjected either to pulse-chase labeling with
radioactive amino acids (lanes 1–18) or to labeling with radioactive phosphate (lanes 19, 20). Each sample is
divided into two equal aliquots, one of which is subjected to UV irradiation (lanes 10–18 and 20). All samples
are subjected to immunoprecipitation using the indicated antisera. The crystal structure of EspP (PDB: 2QOM)
is shown to highlight the position of W1149
4 Notes
migrates 2–4 kDa higher than the EspP β domain (Fig. 2, lanes
10–12, product labeled “3”). This crosslinking product is gen-
erated with high efficiency, and its amount is proportional to the
amount of mature EspP β domain that forms over time (lanes
10–12). Thus crosslinking product 3 results from a stable inter-
action of the mature EspP β domain with a low- molecular-
weight factor. In a parallel experiment, cellular phospholipids are
labeled with 32P. Upon UV irradiation, EspP-specific antibodies
precipitate a 32P-labeled product that runs with the same appar-
ent molecular weight of crosslinking product 3 (Fig. 2, lane 20).
Furthermore, this crosslinking product can be detected using
antibodies specific for E. coli LPS [10, 11]. Thus, product 3
reveals an interaction of EspP with lipids of the outer leaflet of
the outer membrane.
15. This protocol detects two types of interactions for EspP amino
acid position 1149: (i) transient interactions with BamA and
BamB at an early stage of EspP biogenesis, prior to passenger
domain cleavage; (ii) an interaction of the mature EspP β
domain with the LPS of the outer membrane, following secre-
tion and cleavage of the passenger domain, that remains stable
over time. Together with analyses of the interactions occurring
at other amino acids of EspP, the site-directed photocrosslink-
ing approach in metabolically labeled cells has helped to
describe consecutive intermediate steps of the autotransporter
assembly reaction in the bacterial outer membrane [9–11]. In
brief, distinct autotransporter segments interact with periplas-
mic chaperons such as Skp and SurA at an early stage of its
transport through the cellular envelope. At a later time, tran-
sient interactions with one of three subunits of the BAM com-
plex (BamA, BamB, and BamD) are mapped at amino acid
positions that are located approximately 120° from each other
at the periplasmic side of the folded EspP β-barrel, suggesting
the formation of an intermediate where the assembling EspP β
domain is at the center of the BAM complex. On the circum-
ference of the folded barrel, amino acid 1149 is positioned
between the BamA and the BamB interacting sites. At a subse-
quent stage, a stable interaction of position 1149 with the
outer membrane LPS indicates the release of the EspP β
domain by the BAM complex into the lipid bilayer.
Acknowledgements
References
1. Holland IB (2010) The extraordinary diver- during its translocation across the bacterial
sity of bacterial protein secretion mechanisms. outer membrane. Proc Natl Acad Sci U S A
Methods Mol Biol 619:1–20 106:19120–19125
2. Ellman J, Mendel D, Anthony-Cahill S et al 10. Ieva R, Tian P, Peterson JH et al (2011)
(1991) Biosynthetic method for introducing Sequential and spatially restricted interac-
unnatural amino acids site-specifically into pro- tions of assembly factors with an autotrans-
teins. Methods Enzymol 202:301–336 porter beta domain. Proc Natl Acad Sci U S A
3. Chin JW, Martin AB, King DS et al (2002) 108:E383–E391
Addition of a photocrosslinking amino acid to 11. Pavlova O, Peterson JH, Ieva R et al (2013)
the genetic code of Escherichia coli. Proc Natl Mechanistic link between β barrel assembly and
Acad Sci U S A 99:11020–11024 the initiation of autotransporter secretion. Proc
4. Dormán G, Prestwich GD (1994) Natl Acad Sci U S A 110:E938–E947
Benzophenone photophores in biochemistry. 12. Ganong BR, Leonard JM, Raetz CR (1980)
Biochemistry 33:5661–5673 Phosphatidic acid accumulation in the mem-
5. Dautin N, Bernstein HD (2007) Protein secre- branes of Escherichia coli mutants defective
tion in gram-negative bacteria via the auto- in CDP-diglyceride synthetase. J Biol Chem
transporter pathway. Annu Rev Microbiol 255:1623–1629
61:89–112 13. Akiyama Y, Ito K (1990) SecY protein, a
6. Leyton DL, Rossiter AE, Henderson IR membrane- embedded secretion factor of E
(2012) From self sufficiency to dependence: coli, is cleaved by the ompT protease in vitro.
mechanisms and factors important for auto- Biochem Biophys Res Commun 167:711–715
transporter biogenesis. Nat Rev Microbiol 14. Barnard TJ, Dautin N, Lukacik P et al (2007)
10:213–225 Autotransporter structure reveals intra-barrel
7. Hagan CL, Silhavy TJ, Kahne D (2011) cleavage followed by conformational changes.
β-Barrel membrane protein assembly by the Nat Struct Mol Biol 14:1214–1220
Bam complex. Annu Rev Biochem 80:189–210 15. Young TS, Ahmad I, Yin JA et al (2010) An
8. Noinaj N, Rollauer SE, Buchanan SK (2015) enhanced system for unnatural amino acid
The β-barrel membrane protein insertase mutagenesis in E coli. J Mol Biol 395:361–374
machinery from Gram-negative bacteria. Curr 16. Dautin N, Barnard TJ, Anderson DE et al
Opin Struct Biol 31:35–42 (2007) Cleavage of a bacterial autotransporter
9. Ieva R, Bernstein HD (2009) Interaction of an by an evolutionarily convergent autocatalytic
autotransporter passenger domain with BamA mechanism. EMBO J 26:1942–1952
Chapter 20
Abstract
Determining protein partners is an essential step toward understanding protein function and identifying
relevant biological pathways. Many methods exist for investigating protein–protein interactions. The pull-
down assay is an in vitro technique used to detect physical interactions between two or more proteins and
an invaluable tool for confirming a predicted protein–protein interaction or identifying novel interacting
partners. This method typically involves the use of affinity purification with various wash and elution steps.
In this chapter, we describe how an interaction between two purified bacterial proteins or between bacte-
rial and eukaryotic proteins can be detected by pull-down experiments.
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_20, © Springer Science+Business Media LLC 2017
247
248 Arthur Louche et al.
2 Materials
2.2 Pull- 1. 1 M Tris–HCl, pH 7.5 stock solution. Weigh 121.1 g Tris base
Down Assays and transfer to a 1 L graduated cylinder. Add water to 800 mL,
mix, adjust pH with HCl, and make up to 1 L with water.
Store at room temperature (see Note 1).
2. 5 M NaCl stock solution. Weigh 292.2 g NaCl and transfer to
a 1 L graduated cylinder. Add water to 800 mL, stir, and adjust
volume to 1 L with water (see Note 1).
3. Equilibrium buffer (see Note 2): 20 mM Tris–HCl, pH 7.5,
250 mM NaCl. Mix 1 mL 1 M Tris–HCl, pH 7.5 stock solu-
tion with 2.5 mL 5 M NaCl stock solution in a 50 mL centri-
fuge tube, and add water to a volume of 50 mL. Keep at 4 °C
(see Note 3).
4. Elution buffer (see Note 2): 20 mM Tris–HCl, pH 7.5, 250
mM NaCl, 500 mM imidazole. Weigh 1.7 g imidazole in 50
mL solution of equilibrium buffer. Keep at 4 °C (see Note 3).
5. Purified His-tagged protein (bait).
6. Ni-NTA agarose beads: 6% beaded agarose (cross-linked), pre-
charged with Ni2+ (Protino® Ni-NTA Agarose, Macherey
Nagel, or equivalent). Store at 4 °C (see Note 4).
7. 0.8 mL empty columns for gravity flow (Pierce™ Centrifuge
Columns, Thermo Fisher Scientific, or equivalent).
8. Refrigerated microcentrifuge.
2.3 Sodium Dodecyl 1. Resolving gel: 1.5 M Tris–HCl, pH 8.8. Weigh 90.8 g, transfer
Sulfate (SDS) to 500 mL graduated cylinder, and add 300 mL water. Adjust
Polyacrylamide Gel pH with HCl and fill with water to 500 mL. Store at room
Components temperature.
2. Stacking gel buffer: 0.5 M Tris–HCl, pH 6.8. Weigh 30.275 g,
transfer to 500 mL graduated cylinder, and add 300 mL water.
250 Arthur Louche et al.
3 Methods
3.1 Preparation 1. Seed eukaryotic cells at 5.105 in 10 cm cell culture dish
of Cell Lysate (see Note 7) and incubate overnight at 37 °C in CO2.
2. Transfect cells with plasmid containing gene of interest fused
to a specific tag with appropriate transfection reagent for time
necessary for optimal expression of protein (16–24 h is usually
a good range).
3. Cool cells by placing plates on ice, wash cells with 1× PBS. Add
2 mL cold PBS and harvest cells using cell scraper.
4. Centrifuge 5 min at 80 × g at 4 °C.
5. Resuspend cells with 200 μL RIPA buffer supplemented with
antiprotease cocktail.
6. Incubate on ice 20 min and mix gently every 5 min with a
P200 micropipette.
7. Stock prepared cells at −80 °C (see Note 8).
8. Right before pull-down experiment, thaw prepared cell extract.
Centrifuge at 17,000 × g at 4 °C for 20 min. Use the superna-
tant as prey by following step 9 in Subheading 3.2 (see
Note 9).
3.2 Pull-Down Assay 1. Transfer 120 μL Ni-NTA agarose beads to gravity flow column
Using Cell Lysate (see Note 12).
as Prey (See Notes 10 2. Centrifuge column for 1 min at 1000 × g at 4 °C. Discard
and 11) flow-through.
Pull-Down Assays 251
3.3 Pull-Down Assay 1. Incubate 50 μg His-tagged bait protein with 50 μg purified prey
Using Purified Protein protein in total volume of 400 μL equilibrium buffer (see Note
as Prey (See Note 11) 23) 2 h 30 min at 4 °C under agitation (see Notes 17 and 24).
2. Add 80 μL Ni-NTA agarose beads to gravity flow column and
follow steps 1–4 of Subheading 3.2.
252 Arthur Louche et al.
4 Notes
References
1. Costa TRD, Felisberto-Rodrigues C, Meir A, Cascales E (2014) Architecture and assembly of
Prevost MS, Redzej A, Trokter M, Waksman the Type VI secretion system. Biochim Biophys
G (2015) Secretion systems in Gram-negative Acta 1843:1664–1673
bacteria: structural and mechanistic insights. 5. Boucrot E, Henry T, Borg J-P, Gorvel J-P,
Nat Rev Microbiol 13:343–359 Méresse S (2005) The intracellular fate of
2. McBride MJ, Nakane D (2015) Flavobacterium Salmonella depends on the recruitment of
gliding motility and the type IX secretion sys- kinesin. Science 308:1174–1178
tem. Curr Opin Microbiol 28:72–77 6. Laemmli UK (1970) Cleavage of structural
3. Rodríguez-Negrete E, Bejarano ER, Castillo proteins during the assembly of the head of
AG (2014) Using the yeast two-hybrid sys- bacteriophage T4. Nature 227:680–685
tem to identify protein-protein interactions. 7. Di Guan C, Li P, Riggs PD, Inouye H (1988) Vectors
Methods Mol Biol 1072:241–258 that facilitate the expression and purification
4. Zoued A, Brunet YR, Durand E, Aschtgen M-S, of foreign peptides in Escherichia coli by fusion
Logger L, Douzi B, Journet L, Cambillau C, to maltose-binding protein. Gene 67:21–30
Chapter 21
Abstract
Surface plasmon resonance (SPR) is one of the most commonly used techniques to study protein–protein
interactions. The main advantage of SPR is it gives on the ability to measure the binding affinities and
association/dissociation kinetics of complexes in real time, in a label-free environment, and using relatively
small quantities of materials. The method is based on the immobilization of one of the binding partners,
called the ligand, on a dedicated sensor surface. Immobilization is followed by the injection of the other
partner, called the analyte, over the surface containing the ligand. The binding is monitored by subsequent
changes in the refractive index of the medium close to the sensor surface upon injection of the analyte.
During the last 10 years, SPR has been intensively used in the study of secretion systems because of its
ability to detect highly dynamic complexes that are difficult to investigate using other techniques.
This chapter will guide users in the setup of SPR experiments in order to identify protein complexes and
to assess their binding affinity or kinetics. It will include detailed protocols for (i) the immobilization of
proteins with the amine coupling capture method, (ii) analyte-binding analysis, (iii) affinity/kinetic mea-
surements, and (iv) data analysis.
Key words Surface Plasmon Resonance, Protein–protein interaction, Analyte, Ligand, Affinity,
Kinetics, BIAcore T200
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_21, © Springer Science+Business Media LLC 2017
257
258 Badreddine Douzi
such dynamic systems owing to its ability to detect both weak and
strong interactions ranging from the millimolar to the nanomolar
range [13, 14]. SPR can be used as a primary tool to screen inter-
acting partners or as a validation tool for interactions previously
identified by other methods (e.g., bacterial two-hybrid, co-
immunoprecipitation, chemical crosslinking). The determination
of the affinity or kinetics of an interaction, as can be done by SPR,
is fundamental to understanding the nature of binding at the cel-
lular level.
In SPR terminology, the immobilized biomolecule is called the
ligand and the binding partner present in solution is called the
analyte. SPR uses an optical method based on the detection of very
tiny modifications of the refractive index of the medium in close
proximity to a metal surface. The binding of the analyte to the
ligand induces a change in the mass concentration at the metal
surface and, consequently, in the refractive index, which is con-
verted to resonance or response units (RUs). The metal surface
typically consists of a thin gold layer and contains flow cells with
very small volume (less than 100 nL). In a typical SPR experiment,
the first step consists in the immobilization of the ligand in one
flow cell by covalent or noncovalent capture methods. In the sec-
ond step, the analyte is injected into the flow cell containing the
immobilized ligand. The presence of a reference flow cell, which
can either be empty or contain a protein irrelevant for the studied
complex, is important to monitor the true binding of the analyte
to the ligand. The binding of the analyte on the ligand induces a
change on the mass concentration at the surface of the flow cell.
The signal induced by the binding of the analyte to the metal sur-
face in the reference flow cell is subtracted from the signal obtained
from the binding of the analyte to the ligand. Consequently, upon
binding, the curve that we observe, called a sensogram, is divided
into three distinct parts (Fig. 1). The first phase is the association
phase, during which the analyte molecules bind to the binding sites
of the ligand, leading to the increase in RUs. The curve obtained
can be used to calculate the rate of association (kon). The next phase
is a steady state, during which the association/dissociation events
are equal. The level of RUs obtained in this equilibrium phase is
called the response at equilibrium, Req, and can be used to calcu-
late the binding affinity (KD). Upon stopping the injection of the
analyte over the flow cell, the dissociation phase takes place: the
ligand dissociates from the surface as the running buffer flows over
the chip. The dissociation of the analyte from the ligand binding
site induces a decrease in RUs. The corresponding curve can be
used to measure the rate of dissociation (koff). The full removal of
the analyte from the ligand binding sites takes place during the
regeneration step, which is determinant for keeping the surface
intact and ready for new injection cycles.
Surface Plasmon Resonance 259
Fig. 1 Different phases of an SPR sensogram. Initial baseline is obtained after the immobilization of the ligand.
Following injection of analyte over the ligand, the increase in the RUs corresponds to the association phase.
When the number of association and dissociation events is equal, the equilibrium level is maintained. The
dissociation phase starts when stopping the analyte injection: the decrease in the RUs corresponds to the
flowing out of the analyte from the ligand surface. The regeneration step aims at the full removal of the analyte
and at reaching the baseline level
2 Materials
3 Methods
2. Check the purity and the stability of the proteins at least 1 day
before doing the experiment. This can be done by sodium
dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis
(PAGE) and coomassie blue staining (see Note 9).
3. Thaw proteins on ice for 30 min.
4. Centrifuge proteins at 16,000 × g for 20 min at 4 °C.
5. Collect supernatant and keep it on ice on a new 1.5 mL
Eppendorf tube.
6. Measure concentration using NanoDrop or alternative spec-
troscopic method (Bradford or bicinchoninic acid assays). Try
to use the same method during the SPR experiment.
3.2 Material 1. Turn on BIAcore T200 system and prime system with filtered
and Buffer Preparation ultrapure water (see Note 10).
2. Take sensor chip CM5 from 4 °C and keep it at room tempera-
ture (RT) for at least 1 h before experiment.
3. Prepare 1 L running buffer by diluting stock solution ten times.
4. Filter running buffer using 0.22 μM membrane filter.
5. Prime system using running buffer at least three times to be
sure that all system tubing is properly flushed with running
buffer.
6. Insert CM5 sensor chip on sensor chip port and close sensor
port.
7. Prime system three times.
8. When priming is finished, the system will be automatically
shifted to a continuous standby flow.
9. Start a manual run by setting flow path 2 and flow rate of 30
μL min−1.
10. Prepare two vials, one containing 400 μL running buffer and
the other with 400 μL 50 mM NaOH regeneration solution.
11. Eject rack tray and place vials in rack.
12. The rack will be inserted after 1 min.
13. Inject running buffer three times for 1 min.
14. Inject regeneration solution for 30 s after each running buffer
injection until baseline becomes stable.
3.3 pH Scouting (See 1. Open Biacore T200 control software and go to File/Open/
Note 11) New wizard template/Immobilization scouting wizard/New.
2. The next steps consist in setting the different parameters:
–– Specify different solutions used for your experiments and
corresponding pH.
–– Set protein injection and dissociation times; injection time
can be fixed at 2 min, dissociation time at 1 min.
262 Badreddine Douzi
3.4.2 Manual Method 1. In Biacore T200 control software, open manual run.
2. Set flow rate to 10 μL min−1.
3. Select flow path 2 (for flow cell 2).
Surface Plasmon Resonance 263
4. Select start.
5. Dilute your protein in 100 μL 10 mM sodium acetate with
suitable pH and at desired concentration (10–50 μg mL−1).
6. Mix 120 μL EDC (0.4 M in water) and 120 μL NHS (0.1 M
in water).
7. Prepare 120 μL ethanolamine (1 M ethanolamine–HCl at
pH 8.5).
8. Inject EDC/NHS mixture between 6 and 10 min over surface.
9. After activation step, set flow rate to 5 μL min−1.
10. Inject ligand (see Note 17).
11. Inject ethanolamine for 5 min.
12. To calculate the binding capacity, subtract RUs obtained after
activation from RUs obtained after ethanolamine deactivation.
13. Wait until signal is stabilized to perform analyte-binding analysis
(see Note 18).
3.6 Analyte Binding Despite the availability of wizard methods in the Biacore T200
Analysis (See Note 20) control software (wizard template), it is recommended to perform
a manual run (see Note 21).
1. In Biacore T200 control software, open manual run.
2. Set flow rate to 30 μL min−1.
3. Select flow path 2-1 (analyte will be injected over flow cells 1
and 2).
4. Select start.
5. Thaw analyte on ice for 30 min.
6. Measure analyte concentration (see Subheading 3.1).
7. Make dilution of analyte on running buffer. If you estimate
that the affinity is in the micromolar range, prepare 50 μM of
analyte.
264 Badreddine Douzi
3.8 Affinity 1. Go to T200 Biacore software control dialog interface and open
Measurements (See File/Open/New wizard template/Kinetics-affinity.
Notes 25–27) 2. Specify flow path (2-1) if ligand is immobilized.
3. Specify chip type (CM5).
4. Select regeneration, if needed.
5. Select three startup cycles with buffer to stabilize baseline sig-
nal before starting experiment.
6. Specify contact time, flow rate, and dissociation time (see Note 28).
7. Specify contact time and flow rate of regeneration solution.
8. Specify stabilization period (100 s).
9. Fill sample identity and specify serial twofold dilutions of ana-
lyte used for experiment.
10. Prepare analyte (see Subheading 3.1).
11. Prepare ten twofold dilutions of analyte starting from 10 × KD
as specified in wizard template (see Note 29). Dilutions must
be prepared on vials (see Note 30).
12. Place vials in corresponding positions.
13. Start experiment.
Surface Plasmon Resonance 265
4 Notes
3500
30000 pH 4
pH 4.5
25000
Response Units
20000
pH 5
(RUs)
15000
10000
5000
pH 5.5
0
-5000
-50 0 50 100 150 200 250 300 350 400
Time (sec)
Fig. 3 pH scouting of TssE protein from Type VI secretion system over CM5 sensor surface. The pH of each
solution tested is indicated at the top of the corresponding sensogram
270 Badreddine Douzi
34. Mass transport is one of the more widely known limitations for
the determination of the association rate constant. Mass trans-
port takes place when the rate of analyte binding is higher than
the rate of diffusion of the analyte. By contrast, a limitation on
the determination of the dissociation rate constant comes from
the rebinding of the analyte to the ligand owing to the ineffi-
cient effusion of the free analyte from the ligand surface. These
issues can be avoided by the immobilization of a low amount
of ligand (50–150 RUs) and performing the binding analysis
at a high flow rate (30–100 μL min−1).
35. To perform the experiment, the analyte concentrations must
be varied from 0.1 × KD to 10 × KD.
36. The best way to analyze the kinetic data is to use the Biacore
T200 evaluation software. Kinetics data are interpreted in
terms of an interaction model, and the kinetic constants
obtained from the SPR data analysis are apparent constants,
which are valid in the context of the binding model adopted. A
simple model used to determine the rates of an interaction is
the Langmuir model, in which it is assumed that molecule A
binds B with a 1:1 stoichiometry and the binding events are
independent and equivalent.
Acknowledgments
References
1. Barison N, Lambers J, Hurwitz R, Kolbe M role in the assembly of a quaternary complex
(2012) Interaction of MxiG with the cytosolic within the T2SS pseudopilus. J Biol Chem
complex of the type III secretion system 284:34580–34589
controls
Shigella virulence. FASEB 5. Douzi B, Spinelli S, Blangy S, Roussel A,
J 26:1717–1726 Durand E, Brunet YR, Cascales E, Cambillau C
2. Benabdelhak H, Kiontke S, Horn C, Ernst R, (2014) Crystal structure and self-interaction of
Blight MA, Holland IB, Schmitt L (2003) A the type VI secretion tail-tube protein from
specific interaction between the NBD of the enteroaggregative Escherichia coli. PLoS One
ABC-transporter HlyB and a C-terminal frag- 9:e86918
ment of its transport substrate haemolysin A. J 6. Felisberto-Rodrigues C, Durand E, Aschtgen
Mol Biol 327:1169–1179 MS, Blangy S, Ortiz-Lombardia M, Douzi B,
3. Douzi B, Ball G, Cambillau C, Tegoni M, Cambillau C, Cascales E (2011) Towards a
Voulhoux R (2011) Deciphering the Xcp structural comprehension of bacterial type VI
Pseudomonas aeruginosa type II secretion secretion systems: characterization of the TssJ-
machinery through multiple interactions with TssM complex of an Escherichia coli pathovar.
substrates. J Biol Chem 286:40792–40801 PLoS Pathog 7:e1002386
4. Douzi B, Durand E, Bernard C, Alphonse S, 7. Girard V, Cote JP, Charbonneau ME, Campos
Cambillau C, Filloux A, Tegoni M, Voulhoux R M, Berthiaume F, Hancock MA, Siddiqui N,
(2009) The XcpV/GspI pseudopilin has a central Mourez M (2010) Conformation change in a
Surface Plasmon Resonance 275
Abstract
Changes in conformation can alter a protein’s vulnerability to proteolysis. Thus, in vivo differential
proteinase sensitivity provides a means for identifying conformational changes that mark discrete states in
the activity cycle of a protein. The ability to detect a specific conformational state allows for experiments
to address specific protein–protein interactions and other physiological components that potentially
contribute to the function of the protein. This chapter presents the application of this technique to the
TonB-dependent energy transduction system of Gram-negative bacteria, a strategy that has refined our
understanding of how the TonB protein is coupled to the ion electrochemical gradient of the cytoplasmic
membrane.
Key words Protein conformation, Ion electrochemical gradient, Proteinase sensitivity, TonB,
Spheroplast, Energy transduction
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_22, © Springer Science+Business Media LLC 2017
277
278 Ray A. Larsen
Fig. 1 Identification of an ion electrochemical responsive TonB conformation by differential proteinase K sus-
ceptibility. Western blots of whole-cell (WC), spheroplast (S), and CCCP-treated spheroplast (S-CCCP) samples,
treated with (+) or without (−) proteinase K (“Prot K”), are shown. The sample set on the left was generated
from the wild-type strain W3110 and is wild type for TonB, ExbB, and ExbD. The sample set in the middle was
generated from the W3110 derivative and is wild type for TonB but does not express ExbB and ExbD. The
sample set on the right was generated from a W3110 derivative bearing a tonB allele that encodes an inactive
TonB with a deletion of the valine residue from position 17 and is wild type for ExbB and ExbD. Samples were
developed on 11% SDS-polyacrylamide gels, transferred to polyvinylidene fluoride membranes and probed
with the monoclonal antibody 4H4 (specific for TonB residues 79–84). The positions and apparent molecular
masses of intact TonB and the proteinase K-resistant fragment are indicated on the left of the panel. Reproduced
in part from Larsen et al. [5] with permission from John Wiley & Sons
2 Materials
2.1 Spheroplast 1. Luria–Bertani (LB) agar medium ([16]) (see Note 3).
Production 2. Supplemented M9 medium: Prepare a 10× stock solution con-
taining 60 g Na2HPO4, 30 g KH2PO4, 5 g NaCl, and 10 g
NH4Cl per liter. Sterilize the 10× solution by autoclaving and
store at room temperature. To make the culture medium, add
10 mL of the 10× solution to 85.6 mL sterile distilled water,
then supplement this mixture with 2 mL of a filter-sterilized
20% (w/v) carbohydrate source (see Note 4), 1 mL 20% (w/v)
vitamin-free casamino acids (autoclaved), 1 mL filter-sterilized
0.4% (w/v) tryptophan, 200 μL filter-sterilized 0.2% (w/v)
thiamin (see Note 5), 100 μL 1 M MgSO4, 100 μL 0.5 M
CaCl2, and 50 μL 0.1 (w/v) FeCl3·6H2O (see Notes 6 and 7).
3. Isopropyl β-d-1-thiogalactopyranoside (IPTG): 1 M (see
Note 8).
4. Buffer 1: 200 mM Tris–acetate, pH 8.2, 500 mM sucrose,
0.5 mM ethylenediaminetetraacetic acid (EDTA). Keep solution
at 4 °C.
5. Buffer 2: 200 mM Tris–acetate, pH 8.2. Keep solution at
4 °C.
6. Buffer 3: 200 mM Tris–acetate, pH 8.2, 250 mM sucrose,
20 mM MgSO4 (see Note 9). Keep solution at 4 °C.
7. Lysozyme: Stock solution of 2 mg lysozyme in 1 mL sterile
water (see Note 10). Keep solution at 4 °C.
8. Incubator.
9. Spectrophotometer (see Note 11).
10. Microcentrifuge and microfuge tubes.
11. Micropipette or a pulled Pasteur pipet and a rubber bulb.
Assessing Protein Conformational Changes 281
3 Methods
3.1 Production 1. Select an isolated colony from an LB agar stock plate (see Note 3)
of Spheroplasts and inoculate 5 mL of supplemented M9 medium in a stan-
dard culture tube. Incubate at 37 °C overnight with shaking
(~200 rpm).
2. Inoculate a fresh culture tube containing 5 mL of supple-
mented M9 medium with 25 μL of the overnight culture and
incubate as described earlier, with periodic spectrophotomet-
ric monitoring of growth (see Notes 11 and 14).
3. At or near an A550 of 0.2, add 5 μL 1 M IPTG (1 mM final
concentration) to each culture to induce β-galactosidase expres-
sion for subsequent measurement of spheroplast integrity.
4. At an A550 of 0.4, harvest cells as six 500 μL samples into pre-
chilled 1.5 mL Eppendorf microfuge tubes, and centrifuge at
~20,000 × g for 5 min at 4 °C (see Note 15).
5. Remove supernatant by manual aspiration using either a micro-
pipette with a disposable tip or a pulled Pasteur pipet and a
rubber bulb. Discard the supernatant and place tubes on ice.
6. Four of the six tubes are used to generate spheroplasts. To each
of these tubes, add 250 μL chilled (4 °C) buffer 1. Suspend
pellets by gently pipetting up and down with a micropipette.
When fully suspended, add 20 μL of the 2 mg/mL lysozyme
solution. Mix by gently flicking each tube, then immediately
add 250 μL chilled (4 °C) buffer 2. Mix gently as above, then
incubate on ice for 5 min (see Note 16). Proceed to step 7
while incubating these tubes, then proceed to step 8.
282 Ray A. Larsen
3.2 Proteinase Preparations are divided into pairs for analysis. The whole-cell con-
Accessibility trol pair was produced separately at the same time the four tubes of
spheroplasts were produced. The spheroplast tubes are now divided
into two groups: The first is used to evaluate baseline proteinase
susceptibility, and the second pair of tubes is used to examine the
effect of collapsing the ion electrochemical gradient of the cyto-
plasmic (i.e., spheroplast) membrane on the conformation of
TonB. Take care to maintain all tubes at 4 °C and to handle tubes
gently through the following steps.
1. Add the protonophore CCCP to two spheroplast tubes as
3.4 μL of a 15 mM solution of CCCP in DMSO, for a final
concentration of ~50 μM (see Note 19). These two tubes
become the second pair. The two remaining spheroplast
tubes (pair 1) and the two whole-cell tubes receive 3.4 μL of
DMSO alone.
2. Add 6.3 μL of the 2.0 mg/mL solution of proteinase K to one
tube of the whole-cell pair and one tube of each spheroplast
pair (for a final concentration of 25 μg/mL). Add 6.3 μL
water to the other tube in each pair. Incubate all tubes for
15 min at 4 °C, with each tube gently flicked every several
minutes to maintain cells and spheroplasts in suspension (see
Note 20).
3. Add 5 μL 100 mM PMSF to each tube (for a final concentra-
tion of 1 mM), mix gently, and incubate for 2 min at 4 °C to
inactivate proteinase K.
4. Optional: Remove two 100 μL aliquots from each tube for the
determination of β-galactosidase activity. Store on ice while
step 5 is performed, then proceed to step 6.
5. Add 500 μL (300 μL if step 4 performed) of cold (4 °C) 10%
(w/v) TCA to both whole-cell tubes and to each tube in sphero-
plast pairs 1 and 2. Mix by inverting several times, then incubate
the samples on ice for 15 min to precipitate total protein
(see Note 21). Proceed to step 6 while incubating these tubes.
Assessing Protein Conformational Changes 283
4 Notes
References
1. Oldfield CJ, Dunker AK (2014) Intrinsically C-terminal domain of TonB and interaction
disordered proteins and intrinsically disordered studies with TonB box peptides. J Mol Biol
protein regions. Annu Rev Biochem 83: 345:1185–1197
553–584 4. Postle K, Larsen RA (2007) TonB dependent
2. Johnson DE, Xue B, Sickmeier MD, Meng J, energy transduction between outer and cyto-
Cortese MS, Oldfield CJ, Gall TL, Dunker AK, plasmic membranes. Biometals 20:453–465
Uversky VN (2012) High-throughput charac- 5. Larsen RA, Thomas MG, Postle K (1999)
terization of intrinsic disorder in proteins from Protonmotive force, ExbB and ligand-bound
the protein structure initiative. J Struct Biol FepA drive conformational changes in
180:201–215 TonB. Mol Microbiol 31:1809–1824
3. Peacock RS, Weijie AM, Howard SP, Price FD, 6. Larsen RA, Deckert G, Kastead K, Devanathan
Vogel HJ (2005) The solution structure of the S, Keller KL, Postle K (2007) His20 provides
Assessing Protein Conformational Changes 287
the sole functionally significant side chain in 12. Randall LL, Hardy SJS (1986) Correlation of
the essential TonB transmembrane domain. competence for export with lack of tertiary
J Bacteriol 189:2825–2833 structure of the mature species: a study in vivo
7. Ollis AA, Postle K (2012) Identification of of maltose-binding protein in E. coli. Cell
functionally important TonB-ExbD periplas- 46:921–928
mic domain interactions in vivo. J Bacteriol 13. Cascales E, Gavioli M, Sturgis JN, Lloubés R
194:3078–3087 (2000) Proton motive force drives the interac-
8. Ollis AA, Kumar A, Postle K (2012) The ExbD tion of the inner membrane TolA and outer
periplasmic domain contains distinct functional membrane pal proteins in Escherichia coli. Mol
regions for two stages in TonB energization. Microbiol 38:904–915
J Bacteriol 194:3069–3077 14. Germon P, Ray MC, Vianney A, Lazzaroni JC
9. Gresock MG, Kastead KA, Postle K (2015) (2001) Energy-dependent conformational
From homodimer to heterodimer and back: change in the TolA protein of Escherichia coli
elucidating the TonB energy transduction involves its N-terminal domain, TolQ, and
cycle. J Bacteriol 197:3433–3445 TolR. J Bacteriol 183:4110–41104
10. Fischer E, Günter K, Braun V (1989) 15. Hill CW, Harnish BW (1981) Inversions
Involvement of ExbB and TonB in transport between ribosomal RNA genes of Escherichia
across the outer membrane of Escherichia coli: coli. Proc Natl Acad Sci U S A 78:7069–7072
phenotypic complementation of exbB mutants 16. Miller JH (1972) Experiments in molecular
by overexpressed tonB and physical stabiliza- genetics. Cold Spring Harbor Press, Cold
tion of TonB by ExbB. J Bacteriol 171: Spring Harbor
5127–5134 17. Larsen RA, Myers PS, Skare JT, Seachord CL,
11. Larsen RA, Thomas MG, Wood GE, Postle K Darveau RP, Postle K (1996) Identification of
(1994) Partial suppression of an Escherichia coli TonB homologs in the family Enterobacteriaceae
TonB transmembrane domain mutation and evidence for conservation of TonB-
(∆V17) by a missense mutation in ExbB. Mol dependent energy transduction complexes.
Microbiol 13:627–640 J Bacteriol 178:1363–1373
Chapter 23
Abstract
Bacterial secretion systems are among the largest protein complexes in prokaryotes and display remarkably
complex architectures. Their assembly often follows clearly defined pathways. Deciphering these pathways
not only reveals how bacteria accomplish building these large functional complexes but can provide crucial
information on the interactions and subcomplexes within secretion systems, their distribution within bac-
teria, and even functional insights. The emergence of fluorescent proteins has provided a new powerful
tool for biological imaging, and the use of fluorescently labeled components presents an interesting
method to accurately define the biogenesis of macromolecular complexes. Here we describe the use of this
method to decipher the assembly pathway of bacterial secretion systems.
Key words Fluorescence microscopy, Biogenesis, Secretion systems, Fluorescently labeled proteins,
Macromolecular complexes, Epistasis experiments, Subcellular localization
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_23, © Springer Science+Business Media LLC 2017
289
290 Abdelrahim Zoued and Andreas Diepold
2 Materials
3 Methods
3.1 Preparation 1. Streak bacteria from −80 °C stock onto LB agar plates con-
of Bacteria and Setup taining the required additives and antibiotics and incubate at
of Microscopy the required temperature (e.g., 37 °C) until single colonies are
Equipment visible (usually 12–36 h).
2. Inoculate overnight cultures of bacteria with single colonies
from agar plates and grow in shaking incubator at required
temperature and agitation.
3. On the next day, determine the optical density at 600 nm
wavelength (OD600) of the overnight culture and inoculate a
main culture to OD600 that is suitable for the expression of the
analyzed secretion system (a culture volume of 5 mL is suffi-
cient, and an OD600 of 0.1 is a good starting point for many
systems).
4. Incubate bacteria in a shaking incubator until they reach the
early stationary phase (usually 1–2 h).
5. Induce secretion system according to standard conditions
(e.g., temperature shift, addition of inducer).
6. In the meantime, prepare a 1.5% solution of low-fluorescence
agarose in microscopy buffer (see Note 8). While less than 100
μL of agarose solution is required per strain, a higher volume
292 Abdelrahim Zoued and Andreas Diepold
3.2 Microscopy 1. Place a drop of immersion oil onto the center of the cover slip,
and flip the slide so that the cover slip faces the objective.
Carefully insert the slide into the microscope and bring the
lens into contact with the immersion oil.
Protein Dynamics by Fluorescence Microscopy 293
Fig. 2 Schematic representation of bacteria loaded onto cover slip and covered
with agar pad
3.3 Image 1. Phase contrast and fluorescence images can be adjusted and
Processing merged using ImageJ or equivalent software (see Note 16).
2. Slight movements of the whole field during the time of the
experiment can be corrected by registering individual frames
using the StackReg plug-in in ImageJ [19].
3. Blurring of the image can be reduced by deconvolution, a
mathematical postimaging process that removes or reassigns
the fraction of detected photons caused by out-of-focus struc-
tures. This is especially useful when three-dimensional infor-
mation from z stacks is available. Care should be taken not to
mistake deconvolution artifacts for clustering, and negative
controls are mandatory when applying deconvolution.
4. Detection and quantification of bacteria and fluorescent foci
can be performed in ImageJ or using the Oufti software pack-
age (formerly known as MicrobeTracker [21]).
4 Notes
References
1. Tseng T-T, Tyler B, Setubal J (2009) Protein bacteria: structural and mechanistic insights.
secretion systems in bacterial-host associations, Nat Rev Microbiol 13:343–359
and their description in the Gene Ontology.
3.
Kimbrough TG, Miller SI (2000)
BMC Microbiol 9:S2 Contribution of Salmonella typhimurium type
2. Costa TRD, Felisberto-Rodrigues C, Meir A, III secretion components to needle complex
Prevost MS, Redzej A, Trokter M, Waksman G formation. Proc Natl Acad Sci U S A
(2015) Secretion systems in Gram-negative 97:11008–11013
298 Abdelrahim Zoued and Andreas Diepold
4. Sukhan A, Kubori T, Wilson J, Galán JE structural components and involves the for-
(2001) Genetic analysis of assembly of the mation of an YscV oligomer. Mol Microbiol
Salmonella enterica serovar Typhimurium 82:502–514
type III secretion-associated needle complex. 17. Aguilar J, Zupan J, Cameron TA, Zambryski
J Bacteriol 183:1159–1167 PC (2010) Agrobacterium type IV secretion
5. Kimbrough TG, Miller SI (2002) Assembly system and its substrates form helical arrays
of the type III secretion needle complex of around the circumference of virulence-induced
Salmonella typhimurium. Microbes Infect cells. Proc Natl Acad Sci U S A 107:3758–3763
4:75–82 18. Durand E, Nguyen VS, Zoued A, Logger L,
6. Ogino T, Ohno R, Sekiya K, Kuwae A, Péhau-Arnaudet G, Aschtgen M-S, Spinelli
Matsuzawa T, Nonaka T, Fukuda H, Imajoh- S, Desmyter A, Bardiaux B, Dujeancourt A,
Ohmi S, Abe A (2006) Assembly of the type Roussel A, Cambillau C, Cascales E, Fronzes
III secretion apparatus of enteropathogenic R (2015) Biogenesis and structure of a type
Escherichia coli. J Bacteriol 188:2801–2811 VI secretion membrane core complex. Nature
7. Fronzes R, Schäfer E, Wang L, Saibil H, 523:555–560
Orlova E, Waksman G (2009) Structure of a 19. Schneider CA, Rasband WS, Eliceiri KW
type IV secretion system core complex. Science (2012) NIH Image to ImageJ: 25 years of
323:266–268 image analysis. Nat Methods 9:671–675
8. Schraidt O, Lefebre MD, Brunner MJ, Schmied 20. Köhler A (1893) Ein neues Beleuchtungsverfahren
WH, Schmidt A, Radics J, Mechtler K, Galán für mikrophotographische Zwecke. Z Wiss
JE, Marlovits TC (2010) Topology and organi- Mikrosk 10:433–440
zation of the Salmonella typhimurium type III 21. Paintdakhi A, Parry B, Campos M, Irnov I,
secretion needle complex components. PLoS Elf J, Surovtsev I, Jacobs-Wagner C (2015)
Pathog 6:e1000824 Oufti: an integrated software package for high-
9. Reichow SL, Korotkov KV, Hol WGJ, Gonen accuracy, high-throughput quantitative micros-
T (2010) Structure of the cholera toxin secre- copy analysis. Mol Microbiol 99:767–777
tion channel in its closed state. Nat Struct Mol 22. Shaner NC, Steinbach PA, Tsien RY (2005)
Biol 17:1226–1232 A guide to choosing fluorescent proteins. Nat
10. Chandran Darbari V, Waksman G (2015) Methods 2:905–909
Structural biology of bacterial type IV secre- 23. Adams S, Campbell R, Gross L, Martin B,
tion systems. Annu Rev Biochem 84:603–629 Walkup G, Yao Y, Llopis J, Tsien RY (2002)
11. Rose P, Fröbel J, Graumann PL, Müller M New biarsenical ligands and tetracysteine
(2013) Substrate-dependent assembly of the motifs for protein labeling in vitro and in vivo:
Tat translocase as observed in live Escherichia synthesis and biological applications. J Am
coli cells. PLoS One 8:e69488 Chem Soc 124:6063–6076
12. Alcock F, Baker MAB, Greene NP, Palmer T, 24. Andresen M, Schmitz-Salue R, Jakobs S
Wallace MI, Berks BC (2013) Live cell imaging (2004) Short tetracysteine tags to beta-tubulin
shows reversible assembly of the TatA compo- demonstrate the significance of small labels for
nent of the twin-arginine protein transport sys- live cell imaging. Mol Biol Cell 15:5616–5622
tem. Proc Natl Acad Sci U S A 110:3650–3659 25. Enninga J, Mounier J, Sansonetti P, Tran Van
13. Lybarger S, Johnson TL, Gray M, Sikora A, Nhieu G, Van Nhieu GT (2005) Secretion of
Sandkvist M (2009) Docking and assembly of type III effectors into host cells in real time.
the type II secretion complex of Vibrio chol- Nat Methods 2:959–965
erae. J Bacteriol 191:3149–3161 26. Diepold A, Kudryashev M, Delalez NJ, Berry
14. Johnson TL, Sikora AE, Zielke RA, Sandkvist RM, Armitage JP (2015) Composition, forma-
M (2013) Fluorescence microscopy and pro- tion, and regulation of the cytosolic C-ring, a
teomics to investigate subcellular localization, dynamic component of the type III secretion
assembly, and function of the type II secretion injectisome. PLoS Biol 13:e1002039
system. Methods Mol Biol 966:157–172 27. Poulter NS, Pitkeathly WTE, Smith PJ,
15. Diepold A, Amstutz M, Abel S, Sorg I, Jenal U, Rappoport JZ (2015) In: Verveer PJ (ed)
Cornelis GR (2010) Deciphering the assembly Advanced fluorescence microscopy. Springer,
of the Yersinia type III secretion injectisome. New York
EMBO J 29:1928–1940 28. MacDonald L, Baldini G, Storrie B (2015)
16. Diepold A, Wiesand U, Cornelis GR (2011) Does super-resolution fluorescence micros-
The assembly of the export apparatus copy obsolete previous microscopic approaches
(YscR,S,T,U,V) of the Yersinia type III secre- to protein co-localization? Methods Mol Biol
tion apparatus occurs independently of other 1270:255–275
Chapter 24
Abstract
Membrane proteins can assemble and form complexes in the cell envelope. In Gram-negative bacteria, a
number of multiprotein complexes, including secretion systems, efflux pumps, molecular motors, and pilus
assembly machines, comprise proteins from the inner and outer membranes. Besides the structures of
isolated soluble domains, only a few atomic structures of these assembled molecular machines have been
elucidated. To better understand the function and to solve the structure of protein complexes, it is thus
necessary to design dedicated production and purification processes. Here we present cloning procedures
to overproduce membrane proteins into Escherichia coli cells and describe the cloning and purification
strategy for the Type VI secretion TssJLM membrane complex.
Key words Membrane protein complexes, Escherichia coli, T7 overexpression, Protein purification
1 Introduction
1.1 Cloning Vectors Several inducible promoters have been described and are available
to overexpress a gene of interest. These promoters are usually
cloned into vectors that also contain the gene encoding the cognate
regulatory protein and a transcriptional terminator to prevent non-
productive transcription from the downstream gene [1–7]. The tac
and trc promoters that contain the −35 and −10 sequences from
the trp and lacUV5 promoters, respectively, have been optimized
for high expression levels [1]. The most famous regulated E. coli
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_24, © Springer Science+Business Media LLC 2017
299
300 Eric Durand and Roland Lloubes
2 Materials
3 Methods
3.1 Cloning TssJLM To overproduce the T6SS TssJLM membrane complex, the tssJ,
Membrane Complex tssL, and tssM genes were assembled in an artificial operon with
individual optimized ribosome-binding sites (RBS). In addition,
based on previous data regarding permissive positions, each sub-
unit was tagged with a specific tag (see Fig. 1).
The pRSF–TssJStrep-TssLFLAG-His6TssM plasmid was constructed
by both RSl (see Note 4) and RSl-free (see Note 2, [23]) cloning
methods (Fig. 1).
1. PCR amplify the tssJ gene (encoding the TssJ lipoprotein)
using high-fidelity Taq DNA polymerase and primers contain-
ing the NdeI (5′ primer) and the XhoI RS (3′ primer) extensions.
Membrane Protein Complex Purification 303
Fig. 1 Cloning of synthetic tssJLM operon encoding the T6SS membrane core complex. (a) The tssL, tssM, and
tssJ genes were PCR amplified from the entero-aggregative sci-1 T6SS operon. DNA sequences were added
encoding an optimized RBS at the 5′ end of each gene and sequences encoding a C-terminal Strep-II
tag (STREP) for TssJ and N-terminal 6×His and Flag tags for TssM and TssL, respectively. (b) The tssJLM
artificial operon was cloned in the pRSF-Duet vector. (c) Schemes representing the protein constructs, the
subdomain boundaries, and some indicated characteristics (TM transmembrane segments, SP signal peptide,
and CYS acylated cysteine)
4 Notes
Acknowledgements
References
high-level expression by vectors containing the Escherichia coli as revealed by LILBID mass
arabinose PBAD promoter. J Bacteriol 177: spectrometry. Biochemistry 50:8950–8956
4121–4130 17. Sverzhinsky A, Fabre L, Cottreau AL, Biot-
6. Haldimann A, Daniels LL, Wanner BL (1998) Pelletier DM, Khalil S, Bostina M, Rouiller I,
Use of new methods for construction of tightly Coulton JW (2014) Coordinated rearrange-
regulated arabinose and rhamnose promoter ments between cytoplasmic and periplasmic
fusions in studies of the Escherichia coli phos- domains of the membrane protein complex
phate regulon. J Bacteriol 180:1277–1286 ExbB-ExbD of Escherichia coli. Structure
7. Balzer S, Kucharova V, Megerle J, Lale R, 22:791–797
Brautaset T, Valla S (2013) A comparative 18. Yonekura K, Maki-Yonekura S, Homma M
analysis of the properties of regulated promoter (2011) Structure of the flagellar motor protein
systems commonly used for recombinant gene complex PomAB: implications for the torque-
expression in Escherichia coli. Microb Cell Fact generating conformation. J Bacteriol 193:
12:26 3863–3870
8. Studier FW (1991) Use of bacteriophage T7 19. Kim JS, Jeong H, Song S, Kim HY, Lee K,
lysozyme to improve an inducible T7 expres- Hyun J, Ha NC (2015) Structure of the tripar-
sion system. J Mol Biol 219:37–44 tite multidrug efflux pump AcrAB-TolC sug-
9. Schlegel S, Löfblom J, Lee C, Hjelm A, Klepsch gests an alternative assembly mode. Mol Cells
M, Strous M, Drew D, Slotboom DJ, de Gier 38:180–186
JW (2012) Optimizing membrane protein 20. Low HH, Gubellini F, Rivera-Calzada A,
overexpression in the Escherichia coli strain Braun N, Connery S, Dujeancourt A, Lu F,
Lemo21(DE3). J Mol Biol 423:648–659 Redzej A, Fronzes R, Orlova EV, Waksman G
10. Dubendorff JW, Studier FW (1991) (2014) Structure of a type IV secretion system.
Controlling basal expression in an inducible T7 Nature 508:550–553
expression system by blocking the target T7 21. Durand E, Nguyen VS, Zoued A, Logger L,
promoter with lac repressor. J Mol Biol Péhau-Arnaudet G, Aschtgen MS, Spinelli S,
219:45–59 Desmyter A, Bardiaux B, Dujeancourt A,
11. Schwarz D, Junge F, Durst F, Frölich N, Roussel A, Cambillau C, Cascales E, Fronzes R
Schneider B, Reckel S, Sobhanifar S, Dötsch V, (2015) Biogenesis and structure of a type VI
Bernhard F (2007) Preparative scale expression secretion membrane core complex. Nature
of membrane proteins in Escherichia coli-based 523:555–560
continuous exchange cell-free systems. Nat 22. Bakelar J, Buchanan SK, Noinaj N (2016) The
Protoc 2:2945–2957 structure of the β-barrel assembly machinery
12. Guihard G, Boulanger P, Bénédetti H, Lloubes complex. Science 351:180–186
R, Besnard M, Letellier L (1994) Colicin A 23. Unger T, Jacobovitch Y, Dantes A, Bernheim
and the Tol proteins involved in its transloca- R, Peleg Y (2010) Applications of the
tion are preferentially located in the contact Restriction Free (RF) cloning procedure for
sites between the inner and outer membranes molecular manipulations and protein expres-
of Escherichia coli cells. J Biol Chem sion. J Struct Biol 172:34–44
269:5874–5880 24. Tabor S, Richardson CC (1985) A bacterio-
13. Cascales E, Lloubes R, Sturgis JN (2001) The phage T7 RNA polymerase/promoter system
TolQ-TolR proteins energize TolA and share for controlled exclusive expression of specific
homologies with the flagellar motor proteins genes. Proc Natl Acad Sci U S A 82:1074–1078
MotA-MotB. Mol Microbiol 42:795–807 25. Studier FW, Moffatt BA (1986) Use of bacte-
14. Celia H, Noinaj N, Zakharov SD, Bordignon E, riophage T7 RNA polymerase to direct selec-
Botos I, Santamaria M, Barnard TJ, Cramer tive high-level expression of cloned genes.
WA, Lloubes R, Buchanan SK (2016) Structural J Mol Biol 189:113–130
insight into the role of the Ton complex in 26. Narayanan A, Ridilla M, Yernool DA (2011)
energy transduction. Nature 538:60–65 Restrained expression, a method to overpro-
15. Pramanik A, Zhang F, Schwarz H, Schreiber F, duce toxic membrane proteins by exploiting
Braun V (2010) ExbB protein in the cytoplas- operator-repressor interactions. Protein Sci
mic membrane of Escherichia coli forms a stable 20:51–61
oligomer. Biochemistry 49:8721–8728 27. Stuchlík S, Turna J (1998) Overexpression of
16. Pramanik A, Hauf W, Hoffmann J, Cernescu M, the FNR protein of Escherichia coli with T7
Brutschy B, Braun V (2011) Oligomeric struc- expression system. Folia Microbiol (Praha)
ture of ExbB and ExbB-ExbD isolated from 43:601–604
Membrane Protein Complex Purification 309
28. Doherty AJ, Connolly BA, Worrall AF (1993) and E1 interact with a component of their
Overproduction of the toxic protein, bovine translocation system. EMBO J 10:1989–1995
pancreatic DNaseI, in Escherichia coli using a 32. Derouiche R, Bénédetti H, Lazzaroni JC,
tightly controlled T7-promoter-based vector. Lazdunski C, Lloubes R (1995) Protein com-
Gene 136:337–340 plex within Escherichia coli inner membrane.
29. van den Ent F, Löwe J (2006) RF cloning: a TolA N-terminal domain interacts with TolQ
restriction-free method for inserting target and TolR proteins. J Biol Chem 270:
genes into plasmids. J Biochem Biophys 11078–11084
Methods 67:67–74 33. Konovalova A, Silhavy TJ (2015) Outer mem-
30. Jeong J, Yim H, Ryu J, Lee H, Lee J, Seen D, brane lipoprotein biogenesis: Lol is not the
Kang S (2012) One-step sequence- and end. Philos Trans R Soc Lond B Biol Sci
ligation-independent cloning as a rapid and 370:1679
versatile cloning method for functional genom- 34. Ringquist S, Shinedling S, Barrick D, Green L,
ics studies. Appl Environ Microbiol 78: Binkley J, Stormo GD, Gold L (1992)
5440–5443 Translation initiation in Escherichia coli:
31. Bénédetti H, Lazdunski C, Lloubes R (1991) sequences within the ribosome-binding site.
Protein import into Escherichia coli: colicins A Mol Microbiol 6:1219–1229
Chapter 25
Abstract
Pili are widespread among bacteria. Type IVa pili (T4aP) are associated with a variety of bacterial functions,
including adhesion, motility, natural transformation, biofilm formation, and force-dependent signaling.
In pathogenic bacteria, T4aP play a crucial role during infection and have been the subject of hundreds of
studies. Methods for the isolation and purification of T4aP were first described in the 1970s. Purified pili
have been used for studies of filament protein content, morphology, immunogenicity, post-translational
modifications, and X-ray crystallography. We detail a tried-and-true method of isolating large amounts of
native T4aP from bacterial surfaces. The method requires supplies and equipment that are available in
most microbiology labs.
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_25, © Springer Science+Business Media LLC 2017
311
312 Alba Katiria Gonzalez Rivera and Katrina T. Forest
2 Materials
2.2 Collection of Pili 1. High-speed floor model centrifuge and appropriate rotor
(e.g., Beckman JA25.50).
2. Oak Ridge centrifuge tubes with O-ring screw caps (maxi-
mum relative centrifugal force (rcf) > 17,500 × g). These are
needed to handle Biosafety Level-2 (BSL-2) organisms.
3. 250-mL beaker.
4. Magnetic stir bar and magnetic stir plate.
5. Parafilm.
6. 25 and 5 mL serological pipets.
Isolation of T4P 313
3 Methods
3.2 Collection of Pili 1. Transfer samples to centrifuge tubes with O-ring screw caps.
Fill only half of the centrifugation tube to allow good vortexing
action.
2. Shear pili from cells by vortexing the cell suspension three
times in 1 min bursts at maximum strength. Cool on ice for
2 min between vortexing steps. Collect sample for sodium
dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis
(PAGE) analysis. In choosing a volume to load into gel, keep
in mind this solution will have a high protein concentration.
Isolation of T4P 315
Shear pili
by vortexing
Remove cells
by centrifugation
P1 S1
Collect pili
by centrifugation
S2
P2
Resuspend pili
in PDB
Optional
Centrifugation S3
P3
Dialyze against PBB
Collect pili
by centrifugation
S4 P4
Fig. 2 Workflow for T4aP isolation. Bacteria are suspended in PDB and pili are
sheared by vortexing (S0). Cell debris and bacteria are removed by centrifugation
(P1). The remaining solubilized pili (S1) are aggregated by dialyzing against PBB
and collected by centrifugation (P2). For purification purposes, pili in P2 are
resuspended in PDB (S3) and aggregated by another dialysis against PBB (P4).
Cycles of resuspension and aggregation can be repeated to improve pilus purity,
although at a loss of overall yield
3.4 Assessment 1. To monitor pilus purity, run pilus preparation samples in 16%
of Results Tricine-SDS-PAGE gel (Fig. 3a, Table 1) and visualize by
Coomassie Brilliant Blue or silver staining. The latter also
permits estimation of LPS contamination. Yields of ~0.2–
0.5 mg of pili per plate at the S3 step can be achieved;
although the overall yield of pilin falls with each cycle, the
purity increases (Fig. 3a, Table 1).
Isolation of T4P 317
Table 1
Purification yields at each step
P3 S3 P4 S4
Total protein (mg) 1 0.9 16a 30.5 10 11.7 6.4 12.6
Purity (%) 84 92 71 88 81 98 70 82
Pilin yield/loss (mg) 1 0.5 11 26.8 8 11.5 4.5 14.5
Approximate purity was estimated by image analysis of the Coomassie-stained gel in Fig. 3a using ImageJ [23]. Protein
concentration was estimated using OD280 and a calculated extinction coefficient for pilin of 14,100/M/cm (as calcu-
lated using ExPasy ProtParam [24]). We present data for two representative examples of yield from the protocol
described here. The left-hand columns are based on a purification by a relatively inexperienced researcher using 44
plates with volumes exactly as described in protocol (and shown in Fig. 3a). The right-hand columns are based on a
purification by a more practiced investigator from 32 plates with correspondingly reduced volumes
a
Note in this case the S3 concentration was estimated from the dialysate after S3 dialysis against PBB rather than in the
initial S3. Thus, this value is an underestimate of the true yield at the S3 stage
Fig. 3 Pili isolated from P. aeruginosa strain K ΔpilT. (a) Proteins were separated
in 16% Tricine-SDS-PAGE gel (molecular mass markers are in kDa). (b) Whole
cells and (c) purified pili (plus contaminating flagella) analyzed by negative stain-
ing and transmission electron microscopy (scale bars: 100 nm)
4 Notes
Acknowledgements
References
1. Hansen JK, Forest KT (2006) Type IV pilin bacterial pili in disease, purification and proper-
structures: insights on shared architecture, ties of Gonococcal pilus vaccine for Gonorrhea.
fiber assembly, receptor binding and type II In: Brooks GF (ed) Immunobiology of
secretion. J Mol Microbiol Biotechnol 11: Neisseria gonorrhoeae: proceedings of a confer-
192–207 ence held in San Francisco, CA. American
2. MacRae TH, Dobson WJ, McCurdy HD Society for Microbiology, Washington, DC
(1977) Fimbriation in gliding bacteria. Can 11. Li J, Lim MS, Li S, Brock M, Pique ME,
J Microbiol 23:1096–1108 Woods VL Jr, Craig L (2008) Vibrio cholerae
3. Henrichsen J, Blom J (1975) Examination of toxin-coregulated pilus structure analyzed by
fimbriation of some gram-negative rods with hydrogen/deuterium exchange mass spec-
and without twitching and gliding motility. trometry. Structure 16:137–148
Acta Pathol Microbiol Scand B 83:161–170 12. Forest KT, Dunham SA, Koomey M, Tainer JA
4. Swanson J (1973) Studies on gonococcus (1999) Crystallographic structure reveals phos-
infection. IV. Pili: their role in attachment of phorylated pilin from Neisseria: phosphoserine
gonococci to tissue culture cells. J Exp Med sites modify type IV pilus surface chemistry and
137:571–589 fibre morphology. Mol Microbiol 31:743–752
5. Saarimaa C, Peltola M, Raulio M, Neu TR, 13. Parge HE, Bernstein SL, Deal CD, McRee DE,
Salkinoja-Salonen MS, Neubauer P (2006) Christensen D, Capozza MA, Kays BW, Fieser
Characterization of adhesion threads of TM, Draper D, So M (1990) Biochemical
Deinococcus geothermalis as type IV pili. purification and crystallographic characteriza-
J Bacteriol 188:7016–7021 tion of the fiber-forming protein pilin from
6. Imam S, Chen Z, Roos DS, Pohlschroder M Neisseria gonorrhoeae. J Biol Chem 265:
(2011) Identification of surprisingly diverse 2278–2285
type IV pili, across a broad range of gram- 14. Parge HE, Forest KT, Hickey MJ, Christensen
positive bacteria. PLoS One 6:e28919 DA, Getzoff ED, Tainer JA (1995) Structure
7. Melville S, Craig L (2013) Type IV pili in of the fibre-forming protein pilin at 2.6 Å reso-
Gram-positive bacteria. Microbiol Mol Biol lution. Nature 378:32–38
Rev 77:323–341 15. Craig L, Taylor RK, Pique ME, Adair BD,
8. Berry JL, Pelicic V (2015) Exceptionally wide- Arvai AS, Singh M, Lloyd SJ, Shin DS, Getzoff
spread nanomachines composed of type IV pil- ED, Yeager M, Forest KT, Tainer JA (2003)
ins: the prokaryotic Swiss Army knives. FEMS Type IV pilin structure and assembly: X-ray
Microbiol Rev 39:134–154 and EM analyses of Vibrio cholerae toxin-
coregulated pilus and Pseudomonas aeruginosa
9. Pelicic V (2008) Type IV pili: e pluribus unum? PAK pilin. Mol Cell 11:1139–1150
Mol Microbiol 68:827–837
16. Schagger H (2006) Tricine-SDS-PAGE. Nat
10. Brinton CC, Bryan J, Dillon J-A, Guerina N, Protoc 1:16–22
Jen Jacobson L, Labik A, Lee S, McMichael J,
Polen S, Rogers K, ACC T, SCM T (1978) 17. Bradley DE (1974) The adsorption of
Uses of pili in Gonorrhea control: role of Pseudomonas aeruginosa pilus-dependent bac-
320 Alba Katiria Gonzalez Rivera and Katrina T. Forest
Abstract
Bacterial protein secretion systems serve to translocate substrate proteins across up to three biological
membranes, a task accomplished by hydrophobic, membrane-spanning macromolecular complexes. The
overexpression, purification, and biochemical characterization of these complexes is often difficult, imped-
ing progress in understanding the structure and function of these systems. Blue native (BN) polyacryl-
amide gel electrophoresis (PAGE) allows for the investigation of these transmembrane complexes right
from their originating membranes, without the need for long preparative steps, and is amenable to the
parallel characterization of a number of samples under near-native conditions. Here we present protocols
for sample preparation, one-dimensional BN PAGE and two-dimensional BN/sodium dodecyl sulfate
(SDS)-PAGE, as well as for downstream analysis by staining, immunoblotting, and mass spectrometry on
the example of the type III secretion system encoded on Salmonella pathogenicity island 1.
Key words Bacterial secretion systems, Membrane proteins, Blue native polyacrylamide gel electro-
phoresis, Two-dimensional polyacrylamide gel electrophoresis, Sucrose gradients, Bacterial cell frac-
tionation, Type III secretion systems, Salmonella typhimurium, Escherichia coli
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_26, © Springer Science+Business Media LLC 2017
321
322 Susann Zilkenat et al.
2 Materials
Table 1
Sucrose solutions for two gradients
Percentage of Buffer
sucrose (%) Buffer 2× M 57% sucrose 1× M
(w/w) Sucrose (g) (mL) solution (mL) (mL)
57 22.8 17.2
32 12.8 10.0
Percentage of Buffer
sucrose (%) Buffer 2× M 55% sucrose 1× M
(w/w) Sucrose (g) (mL) solution (mL) (mL)
55 19.25 13.5
50 4.55 0.45
45 4.1 0.91
40 3.6 1.36
35 3.2 1.8
30 2.7 2.3
2.2 Blue Native PAGE 1. Precast native polyacrylamide gradient gels, e.g., Novex®
NativePAGE™ Bis–Tris or SERVAGel™ N gel systems.
2.2.1 One-Dimensional
BN PAGE Using Precast 2. 10× BN anode buffer: 500 mM Bis-Tris-HCl, pH 7.0. Store at
Mini Gels 4 °C.
3. 10× BN cathode buffer A: 500 mM Tricine, 150 mM Bis–Tris,
0.2% (w/v) Serva Blue G. Do not adjust pH and store at 4 °C.
4. 10× BN cathode buffer B: 500 mM Tricine, 150 mM Bis–Tris.
Do not adjust pH and store at 4 °C.
5. NativeMark™ Unstained Protein Standard (Thermo Fisher).
2.3 Protein 1. Fixing solution: 50% (v/v) ethanol, 3% (w/v) phosphoric acid
Detection, in ddH2O.
and Analysis 2. Serva Blue G (see Note 2).
2.3.1 Colloidal 3. ddH2O.
Coomassie Staining 4. Neuhoff’s solution: 16% (w/v) ammonium sulfate, 25% (v/v)
methanol, 5% (v/v) phosphoric acid, fill up to 100% with ddH2O.
5. Storage solution: 5% (v/v) glacial acetic acid in ddH2O.
2.3.2 Silver Staining Because silver staining is extremely sensitive to trace impurities in
water, it is strongly recommended to use high-quality ddH2O in all
recipes and protocol steps.
1. Fixing solution: 45% (v/v) methanol and 5% (v/v) glacial ace-
tic acid in ddH2O.
2. Sensitizing solution: 0.02% (w/v) sodium thiosulfate in
ddH2O. Always prepare fresh.
3.
Silver nitrate solution: 0.1% (w/v) silver nitrate in
ddH2O. Always prepare fresh.
4. Developing solution: 2% (w/v) sodium carbonate and 0.04%
(v/v) formalin in ddH2O. Prepare this solution the day of use
and add formalin to the developer at most 1 h before use.
5. Stop solution: 1% (v/v) glacial acetic acid in ddH2O.
6. Plastic containers: Polyethylene trays are recommended (see
Note 11).
7. Rocking table.
8. Special waste container for silver nitrate waste.
2.3.3 Immunoblotting 1. 10× SDS-PAGE running buffer: 0.25 M Tris base, 1.92 M gly-
Using Dual-Color Detection cine, 1% (w/v) SDS.
2. 10× transfer buffer: 0.25 M Tris base, 1.92 M glycine, 0.05%
SDS.
3. Polyvinylidene fluoride (PVDF) membrane.
4. Wet blot unit.
326 Susann Zilkenat et al.
5. 100% methanol.
6. 10× Tris-buffered saline (TBS): 200 mM Tris-HCl, 1.5 M
NaCl, pH 8.0.
7. TBS-T: TBS/0.05% Tween 20.
8. 0.1% Ponceau S, 5% acetic acid in ddH2O.
9. Primary antibodies.
10. Secondary DyLight antibodies (Thermo Fisher).
11. LiCor Odyssey infrared scanner.
12. Image Studio software (LiCor) or Quantity One software
(Bio-Rad).
3 Methods
3.1 Sample This protocol serves to extract membrane protein complexes from
Preparation small amounts of whole bacterial cells. The extracted proteins are
best run on BN mini gels and analyzed by immunoblotting as
3.1.1 Extraction
described previously [13].
of Membrane Proteins
from Whole Bacterial Cells 1. All steps are to be performed at 4 °C or on ice unless otherwise
stated.
2. Harvest 0.4–0.6 optical density units (ODU, see Note 12) of
bacterial cells by centrifugation for 2 min at 5000 × g and 4 °C.
3. Aspirate supernatant.
4. Optional: Snap freeze cell pellet in liquid nitrogen and store
cell pellet at −80 °C (see Note 13). Thaw pellet on ice to
continue.
5. Resuspend bacterial cell pellet in 10 μL freshly prepared buffer
K, supplemented with protease inhibitor cocktail, 10 μg/mL
lysozyme, 10 μg/mL DNase, and 1 mM MgSO4 (see Note 14).
6. Incubate for 30 min on ice to allow for cell wall digestion.
Blue Native PAGE 327
3.1.2 Extraction This protocol describes the preparation of small amounts of crude
of Membrane Proteins bacterial membranes and the extraction of membrane protein com-
from Crude Bacterial plexes thereof (see Note 18). The extracted proteins are best run on
Membrane Preparations BN mini gels and analyzed by immunoblotting as described previ-
ously [16]; however, we have also had good experience with the
analysis of these preparations by two-dimensional BN/SDS-PAGE.
1. All steps are to be performed at 4 °C or on ice unless otherwise
stated.
2. Harvest 2–10 ODU of bacterial cells by centrifugation for
2 min at 5000 × g and 4 °C (see Notes 19 and 20).
3. Aspirate supernatant.
4. Optional: Snap freeze cell pellet in liquid nitrogen and store
cell pellet at −80 °C (see Note 13). Thaw pellet on ice to
continue.
5. Resuspend bacterial cell pellet in 750 μL freshly prepared buf-
fer K supplemented with protease inhibitor cocktail, 10 μg/
mL lysozyme,10 μg/mL DNase (see Note 14).
6. Incubate for 30 min on ice to allow for cell wall digestion.
7. Meanwhile, prepare 2 mL screw-cap tubes with 500 μL glass
beads. Label tubes on lid and on sides. Place tubes on ice for
cooling (see Notes 21 and 22).
8. Add 0.8 μL 1 M MgSO4 to each sample (final concentration).
9. Transfer cell suspension to a bead-containing tube and close
properly.
10. Break up cells by bead milling (2 min continuous mode using
SpeedMill Plus, 2 × 20 s at 4 m/s using FastPrep-24). For
cooling of samples, cool down the SpeedMill Plus sample
holder to −20 °C or place the samples on ice for 5 min after
first 20 s cycle when using the FastPrep-24.
328 Susann Zilkenat et al.
3.1.3 Preparation This protocol describes the large-scale preparation of crude mem-
of Crude Membranes branes that are used for further membrane fractionation using
for Sucrose Density sucrose density gradient centrifugation described in Subheadings
Gradient Centrifugation 3.1.4 and 3.1.5 (see Note 25).
1. All steps are to be performed at 4 °C or on ice unless otherwise
stated.
2. Harvest 500–2000 ODU of bacterial cells by centrifugation
for 15 min at 6000 × g and 4 °C, e.g., in Beckman JLA-8.1000
(see Note 26).
3. Pour off supernatant.
4. Resuspend bacterial pellet in 35 mL cold PBS.
5. Transfer suspension to 50 mL Falcon tube.
6. Pellet bacteria by centrifugation for 10 min at 6000 × g and
4 °C.
7. Resuspend bacterial pellet in 10–15 mL buffer K supplemented
with protease inhibitor cocktail, 1 mM EDTA, 10 μg/mL
DNase, and 10 μg/mL lysozyme (all final concentrations).
8. Pass the cell suspension twice through a French press at
124 MPa (high position, 1000–1100 units).
9. Add MgSO4 to a final concentration of 1 mM to activate the
DNase. Mix.
10. Pellet cell debris by centrifugation for 20 min at 24,000 × g
and 4 °C (see Notes 27 and 28).
11. Transfer supernatant to suitable ultracentrifugation tubes or
bottles, e.g., for Beckman Type 45 Ti. Fill up bottles with buf-
fer K.
12. Pellet crude membranes by centrifugation for 45 min at
234,000 × g and 4 °C (45,000 rpm in Beckman Type 45 Ti).
13. Discard supernatant. Wipe off residual supernatant with lint-
free tissue.
14. Resuspend crude membranes in 500 μL buffer 1× M. Use a cut
1 mL pipette tip to detach membrane from tube wall. Transfer
crude suspension to 1 mL dounce homogenizer. Homogenize
with loose piston 15 times.
15. Store crude membrane suspension on ice until further use and
proceed to alternatives (see Subheading 3.1.4 or 3.1.5).
3.1.4 Membrane This protocol describes the preparation of separated inner and
Fractionation by Sucrose outer membranes of Gram-negative bacteria using sucrose density
Density Gradient gradient centrifugation and extraction of membrane protein com-
Centrifugation Using plexes thereof. Sucrose gradient formation and fractionation are
a Biocomp Gradient Station described in two ways: either supported by a Biocomp gradient
station (this section, see Note 29) or manually without the need
for specialized equipment (see Subheading 3.1.5).
330 Susann Zilkenat et al.
Fig. 1 Preparation of sucrose gradients (a–e) using Biocomp Instruments Gradient Station or (f–g) by hand. For
details see Subheadings 3.1.4 and 3.1.5
14. Carefully place tubes on ice once the run is finished. Proceed
to fractionation using Biocomp gradient station fractionator
following step 15.
15. Label 1.5 mL tubes for collecting gradient fractions (13 per
sample). Place tubes on ice.
16. Visually check if membranes are clearly separated and mem-
brane bands are approximately at the same height. Make note
if this is not the case (see Note 34). Take a photo of your
gradient.
17. Program fractionation protocol: Speed 0.3 mm/s, distance 6.6
mm, number of fractions 12, rinse #0 (no automatic washing
steps). Check manual for programming instructions.
332 Susann Zilkenat et al.
3.1.5 Membrane 1. Carefully pour gradient layers on top of each other, starting
Fractionation by Sucrose with 55% sucrose solution. Use 5 mL pipettes and Peleus ball
Density Gradient or a cut 1 mL pipette tip (Fig. 1f) (see Note 37).
Centrifugation Using 2. Carefully place gradients on ice until further use. Proceed to
a Manual Sucrose Step step 3 for ultracentrifugation of sucrose gradients.
Gradient
3. Carefully layer membrane suspension from step 15 in
Subheading 3.1.3 on top of continuous 30–55% step gradient
(see Note 31, Fig. 1e, g).
4. Tare two opposing tubes with buffer 1× M to <0.01 g.
5. Centrifuge gradients for 14 h at 41,000 rpm and 4 °C in a
Beckman SW41 Ti swing-out rotor (287,000 × g) (see Note
32). Set to slow acceleration and slow brake (see Note 33).
6. Carefully place tubes on ice once run is finished. Proceed to
fractionation of sucrose gradient by hand following step 7.
7. Label 1.5 mL tubes for collecting gradient fractions (13 per
sample). Place tubes on ice.
8. Visually check if membranes are clearly separated and mem-
brane bands are approximately at same height. Make note if
this is not the case. Take a photo of your gradient.
Blue Native PAGE 333
3.1.6 Preparing 1. Dilute sucrose at least 1:3 with buffer 1× M. Mix well.
Membrane Fractions 2. Pellet membranes by centrifugation for 45 min at 230,000 × g
from Sucrose Gradient and 4 °C (e.g., in Beckman Type 70.1 Ti).
Fractionations
for Downstream
3. Aspirate supernatant.
Experiments 4. Resuspend each membrane pellet in 200 μL buffer L.
5. Measure protein concentration by BCA assay according to
manufacturer’s instructions (see Note 38).
334 Susann Zilkenat et al.
3.2 Blue Native PAGE One-dimensional BN PAGE using precast mini gels accommo-
dates all previously described sample preparations but is best suited
3.2.1 One-Dimensional
for analytical amounts of membrane protein complexes and down-
BN PAGE Using Precast
stream immunoblotting. If preparative amounts of membrane
Mini Gels
complexes are needed, or the investigation of entire complexomes
is sought, larger gel format may be better suited. Preparation and
run of larger gel formats are described in Subheading 3.2.2. 3–12%
gradient gels are suited for the analysis of protein complexes
between 100 kDa and 5 MDa, 4–16% gradient gels for the analysis
of protein complexes between 30 kDa and 1 MDa.
Table 2
Solutions for casting of BN gradient gel
3% 12% 4% 16% 3%
ddH2O 6.0 mL 1.05 mL 5.6 mL – 5.1 mL
3× BN gel buffer 3.5 mL 3.5 mL 3.5 mL 3.5 mL 3.0 mL
Acrylamide (30% 1.05 mL 4.2 mL 1.4 mL 5.6 mL 0.9 mL
T, 3% C)
Glycerol (80%) – 1.75 mL – 1.4 mL –
TEMED 5.25 μL 5.25 μL 5.25 μL 5.25 μL 4 μL
APS (10%) 52.5 μL 52.5 μL 52.5 μL 52.5 μL 40 μL
2. Fix the plastic film on the lower edge of one of the glass plates
using double-sided Scotch tape. Make sure that the hydro-
philic side of the plastic film is facing up.
3. Place a few drops of ddH2O between the glass plate and plastic
film. Wipe tightly along film with tissue to remove excess water
and trapped air.
4. Slightly grease 1.0 mm spacers on one side with gel seal. Place
spacers with greased side on plastic film.
5. Finish assembly of gel casting assembly according to manufac-
turer’s instructions.
6. Prepare solutions for casting of BN gradient gel according to
recipes in Table 2. Do not add APS at this point (see Note 47).
7. Assemble gradient maker including magnet stirrer and peristal-
tic pump. Squeeze end of outlet tubing between glass plates of
gel casting assembly.
8. Add APS to gel solution of lower percentage, mix well, and
pour solution into distal chamber of gradient maker. Add APS
to gel solution of higher percentage, mix well, and pour solu-
tion into proximal chamber of gradient maker (see Note 48).
9. Switch on peristaltic pump. Open outlet valve and then
chamber-connecting valve. Pour gradient gel (see Note 49).
10. When all the gel solution has entered the gel, add APS to stack-
ing gel solution, mix, and pour solution into proximal cham-
ber of gradient maker (see Note 50). Continue pouring gel.
11. Carefully place comb (10 or 15 wells) into stacking gel (see
Note 51). Take care not to destroy gradient.
12. Allow gel to polymerize for at least 4 h at room temperature.
Gels can be stored after polymerization up to 1 week (4 °C and
moist).
338 Susann Zilkenat et al.
13. Remove comb from gel, rinse gel and wells with ddH2O, and
assemble gel running equipment in cold room (see Note 52).
Fill anode buffer tank with 2 L cold BN anode buffer.
14. Pour cold BN cathode buffer A into wells of gel. Do not fill up
entire cathode buffer tank because the blue stain will make
loading of sample very difficult.
15. Load suitable protein standard, e.g., 10 μL NativeMark from
Thermo Fisher, into one well of gel (see Note 43).
16. Load solubilized and coomassie-treated samples from step 12
from Subheading 3.1.1, step 26 from Subheading 3.1.2, step
12 from Subheading 3.1.6, or step 16 from Subheading 3.1.7
into desired wells of gel.
17. Carefully fill up cathode buffer tank with cold BN cathode
buffer A. Take care not to wash out samples from wells.
18. Run electrophoresis at 4 °C as follows: 130 V and 30 mA for 1
h, 150 V and 30 mA for 14 h, or until thick blue front runs out
of gel (see Note 43).
19. Proceed to second dimension—SDS-PAGE (see Note 53).
Pour as many polyacrylamide gels (resolving and stacking gel)
as needed to analyze each lane of BN PAGE. Use a polyacryl-
amide concentration for resolving gel that allows resolution of
all proteins expected to compose complex of interest (e.g.,
12% polyacrylamide).
20. After pouring stacking gel, place a preparative comb (one long
well matching length and width of one lane of first-dimension
BN gel) in gel and allow polymerization at room temperature
(see Note 54).
21. Disassemble first-dimension BN PAGE assembly (obtained
from step 15). Remove glass plate facing gel (not plastic film)
and spacers.
22. Using razor blades, scrape off partition walls of wells and thickly
stained coomassie front if not everything has run out of gel.
23. Lift up plastic-backed gel and remove double-sided Scotch tape.
24. Using scissors, cut out lanes of first-dimension run.
25. Place lane strips into SDS equilibration buffer to denature pro-
tein complexes and reduce disulfide bonds. Incubate for
20 min (see Note 55).
26. Remove comb of second-dimension gel and overlay top of gel
with hot low-melting agarose solution.
27. Place lane strips of first-dimension gel onto second-dimension
gel (see Notes 56 and 57).
28. Drop 5 μL of protein size ladder onto a small piece of filter
paper (3 × 3 mm). Place marker-soaked filter paper on top of
second-dimension gel next to BN gel lane strip.
Blue Native PAGE 339
3.3 Protein Detection The described protocol according to Neuhoff et al. [24] allows for
and Analysis the highly sensitive detection of proteins by coomassie G.
3.3.1 Colloidal
1. After electrophoresis, place gel in a clean plastic container and
Coomassie Staining
rinse briefly in ddH2O. Reduce gel manipulation to minimum
and always wear powder-free gloves.
2. Decant ddH2O. Add enough fixing solution to completely
immerse gel and incubate o/n at room temperature with gen-
tle agitation (see Note 58).
3. Decant fixing solution and wash gel three times in ddH2O,
30 min for each wash.
4. Decant ddH2O. Equilibrate gel in Neuhoff’s solution for 1 h.
The exact volume of Neuhoff’s solution must be known in
order to add the right amount of Serva Blue G in the next step.
5. Add Serva Blue G powder to a final concentration of 0.1%
(w/v) (e.g., 0.1 g/100 mL Neuhoff’s solution) and stain for
up to 48 h (see Note 59).
6. Transfer gel into a clean container and wash several times with
ddH2O. Gently remove coomassie particles from gel surface.
7. Decant ddH2O and store gel in 5% acetic acid solution at 4 °C
until further usage (see Note 60).
3.3.2 Silver Staining (MS The described protocol according to Shevchenko et al. [25] allows
Compatible) for a highly sensitive detection of proteins.
1. After electrophoresis, place gel in tray and rinse briefly with
ddH2O. Always manipulate gel wearing powder-free gloves
that have been rinsed with deionized water to avoid fingerprint
contamination and pressure marks.
2. Decant ddH2O. Immerse gel in fixing solution for 20–30 min;
gently agitate (see Note 61).
3. To remove acid, rinse gel with ddH2O for at least 20 min (see
Note 62).
4. Soak gel in sensitizing solution for 1–2 min (see Note 63).
5. Decant sensitizing solution. Rinse gel twice with ddH2O,
1 min for each wash.
6. Pour out ddH2O and impregnate gel in chilled silver nitrate
solution for 20–40 min, preferably at 4 °C (see Note 64).
340 Susann Zilkenat et al.
7. Discard solution and rinse gel twice in water, 1 min for each
wash (see Note 65).
8. Develop by adding a sufficient amount of developing solution
to completely cover gel. Agitate until intensity of staining is as
desired (see Note 66).
9. Once the adequate degree of staining is achieved, decant devel-
oping solution. Add stop solution and incubate for 30 min.
Agitate slowly.
10. Store silver-stained gel in stop solution at 4 °C until scanning
or preparation of bands for MS.
3.3.3 Immunoblotting BN PAGE facilitates the analysis of protein complexes that often
Using Dual-Color Detection contain more than one kind of protein component. Quantitative
analysis of the composition of protein complexes by BN PAGE is
very powerful when a simultaneous detection of two different pro-
teins within the same complex band is achieved by dual-color
immunoblotting. The protocol described here uses infrared-
fluorescent secondary antibodies and a LiCor Odyssey scanner.
Although the protocol is described for immunoblotting of BN
gels, it can be easily adapted for two-dimensional BN/SDS-PAGE
or regular SDS-PAGE. Figure 4 shows an example of a dual-color
immunoblotting of a BN PAGE-separated T3SS needle complex.
1. Equilibrate BN gels in SDS-PAGE running buffer for
15–20 min (see Note 67).
2. Assemble wet blot sandwich according to manufacturer’s
instructions. Use only PVDF membranes because coomassie G
binds irreversibly to nitrocellulose.
3. Transfer proteins according to the instructions of the wet blot
equipment manufacturer.
4. When the transfer is finished, take out the PVDF membrane,
place it in a dark container (see Note 68), and rinse it three or
four times in 100% MetOH to remove all bound coomassie G
(see Note 69).
5. Decant MetOH and rinse PVDF membrane with TBS until
wetted.
6. Stain PVDF membrane with Ponceau S solution to visualize
unstained native protein standard.
7. When sufficient staining is achieved, rinse PVDF membrane
with ddH2O until bands become visible and mark bands with
a pencil.
8. Destain PVDF membrane completely using ddH2O.
9. Block unoccupied binding sites of PVDF membrane by incubat-
ing in 5% skim milk/TBS or 3% BSA/TBS for 1 h (see Note 70).
10. Incubate PVDF membrane with primary antibody diluted to
desired concentration TBST (1 × TBS plus 0.05% Tween-20)
for 1 h (see Note 71).
Blue Native PAGE 341
11. Wash the PVDF membrane three times 15 min with TBST.
12. Incubate PVDF membrane with secondary infrared-fluorescent
antibody diluted to desired concentration in TBST for 1 h (see
Note 72).
13. Wash PVDF membrane three times for 15 min with TBST.
14. Scan PVDF membrane in LiCor Odyssey scanner according to
manufacturer’s instructions.
15. Analyze protein bands using western blot analysis tool of
Image Studio software (LiCor). A typical result of dual-color
immunoblotting of a type III secretion system is shown in
Fig. 4.
3.3.4 Preparation of BN MS is well suited to detect the composition of protein complexes
PAGE-Separated separated by BN PAGE; however, it can be difficult to distinguish
Complexes for Analysis a true complex component from an unrelated comigrating protein.
by Mass Spectrometry Distinction can be achieved using suitable controls and bioinfor-
matic analysis, as described by Fischer et al. [15], but also by the
mass spectrometrical profiling of complete lanes of BN gels [26],
as described in what follows. Figure 5 shows the MS-based analysis
of lane profiles of immunoprecipitated T3SS needle complexes.
342 Susann Zilkenat et al.
Fig. 5 Example of MS–based analysis of a lane profile of BN PAGE-separated T3SS needle complex. BN lane
of immunoprecipitated S. typhimurium SPI-1 type III secretion needle complex stained with colloidal coo-
massie, divided into 20 gel slices. Protein intensity profiles based on quantification by MS is shown for indi-
cated proteins. InvA, SpaS, SpaP, and SpaQ are components of the export apparatus of the type III secretion
needle complex. InvG, PrgH, and PrgK are components of the needle complex base. PrgI and PrgJ are secreted
substrates of the type III secretion system forming the filament structures of the needle complex. All these
components form the stable complex that was co-immunoprecipitated. The true components belonging to this
complex can be deduced from the similar protein intensity profiles in slices 2 and 3. It is also apparent that a
large fraction of non-needle-complex-associated SpaS and InvA are present in the sample (slices 15–20).
InvC, SpaO, and OrgA are cytosolic components of the type III secretion system that are not stably associated
with the needle complex and just coprecipitate during purification. This is reflected by the sharp peaking of the
intensity of these proteins in slice 2 but not slice 3. The profile of the unrelated outer membrane protein OmpA
is shown as a control
Blue Native PAGE 343
4 Notes
Fig. 7 BN PAGE analysis of T3SS needle complex using a set of different deter-
gents. Crude membranes of S. typhimurium were extracted with indicated deter-
gents and subsequently analyzed by BN PAGE and immunoblotting. The
FLAG-tagged T3SS export apparatus component SpaS was probed with a pri-
mary anti-FLAG M2 mouse monoclonal antibody and a secondary DyLight 800
nm goat anti-mouse antibody. The band of SpaS in the type III secretion needle
complex is indicated
ing the membrane extraction (at least 1:2), and spin down the
crude membrane pellet again to repeat the gradient.
34. If the membrane bands are not at an equal height in the differ-
ent samples, the most common problem is the pipetting of the
gradient. Either the half-full mark for the wrong cap type
(long/short) was used or the sucrose solutions were not prop-
erly dissolved before layering the gradient. Accidental stirring
of the solutions while underlaying can also lead to aberrations
in gradient formation.
35. For the following samples: Move down position of piston until
position value is exactly 0.00. If you move too far, make note of
the position and reset to 0 to continue; however, fractions will
not be identical to the first sample. Do NOT press “UP” at this
point. Pressing “UP” will move the piston all the way to the top
again, mixing at least the upper part of your gradient.
36. Fractionation of a continuous gradient does not require rins-
ing of sample tubing after each fraction. If a discrete fraction is
targeted, rinsing and blowing off tubing before start of frac-
tionation is recommended.
37. The phase boundaries should be visible from an angle. If you
cannot see any boundaries at all, you have mixed the layers and
should make new gradients.
38. The BCA assay is largely tolerant to lipids in the sample and
therefore preferred over Bradford or Lowry assays. For better
comparability, always use the same standard protein to make a
standard curve, e.g., BSA.
39. The extraction power of a given detergent toward a protein of
interest can be tested by extracting crude membranes with a
protein concentration equivalent of 1–10 mg/mL BSA in
1 mg increments with 1% of a given detergent. The highest
concentration that is still correlated to a linear increase in
extracted protein (testable, for example, by immunoblotting)
is the optimal concentration to use for extraction.
40. Immunoprecipitation can be performed using suitable anti-
bodies or any other epitope tags that tolerate the detergents
used. In an optimal case, the 3× FLAG epitope-tagged fusion
protein is encoded in its natural context, e.g., on the chromo-
some or on its virulence plasmid. However, also expression of
plasmid-based artificial operons encoding all or some complex
partners of interest (of which one is 3× FLAG-tagged) is
possible.
41. During immunoprecipitation with beads, choose a tube
according to your sample volume. It should be one-third to
two-thirds full and rotated over its own head to guarantee a
good distribution of beads in the sample over the whole incu-
bation time.
348 Susann Zilkenat et al.
Acknowledgments
References
1. Schägger H, von Jagow G (1991) Blue native coli twin-arginine translocation apparatus
electrophoresis for isolation of membrane pro- incorporates a distinct form of TatABC com-
tein complexes in enzymatically active form. plex, spectrum of modular TatA complexes and
Anal Biochem 199:223–231 minor TatAB complex. J Mol Biol 346:
2. Bessonneau P, Besson V, Collinson I, Duong F 295–305
(2002) The SecYEG preprotein translocation 4.
Wiedemann N, Kozjak V, Chacinska A,
channel is a conformationally dynamic and Schönfisch B, Rospert S, Ryan MT, Pfanner N,
dimeric structure. EMBO J 21:995–1003 Meisinger C (2003) Machinery for protein
3. Oates J, Barrett CML, Barnett JP, Byrne KG, sorting and assembly in the mitochondrial
Bolhuis A, Robinson C (2005) The Escherichia outer membrane. Nature 424:565–571
Blue Native PAGE 351
5. Kikuchi S, Hirohashi T, Nakai M (2006) workflow for affinity purification mass spec-
Characterization of the preprotein translocon trometry data. J Proteome Res 13:2239–2249
at the outer envelope membrane of chloro- 16. Monjarás Feria JV, Lefebre MD, Stierhof YD,
plasts by blue native PAGE. Plant Cell Physiol Galán JE, Wagner S (2015) Role of autocleavage
47:363–371 in the function of a type III secretion specificity
6. Wagner S, Baars L, Ytterberg AJ, Klussmeier A, switch protein in Salmonella enterica serovar
Wagner CS, Nord O, Nygren PA, van Wijk KJ, Typhimurium. MBio 6:e01459–e01415
de Gier JW (2007) Consequences of mem- 17. Zilkenat S, Franz-Wachtel M, Stierhof YD,
brane protein overexpression in Escherichia Galán JE, Macek B, Wagner S (2016)
coli. Mol Cell Proteomics 6:1527–1550 Determination of the stoichiometry of the
7. Baars L, Wagner S, Wickström D, Klepsch M, complete bacterial type III secretion needle
Ytterberg AJ, van Wijk KJ, de Gier JW (2008) complex using a combined quantitative pro-
Effects of SecE depletion on the inner and teomic approach. Mol Cell Proteomics
outer membrane proteomes of Escherichia coli. 18. Houben ENG, Bestebroer J, Ummels R,
J Bacteriol 190:3505–3525 Wilson L, Piersma SR, Jiménez CR, Ottenhoff
8. Wagner S, Klepsch MM, Schlegel S, Appel A, THM, Luirink J, Bitter W (2012) Composition
Draheim R, Tarry M, Högbom M, van Wijk KJ, of the type VII secretion system membrane
Slotboom DJ, Persson JO, de Gier JW (2008) complex. Mol Microbiol 86:472–484
Tuning Escherichia coli for membrane protein 19. Wittig I, Braun HP, Schägger H (2006) Blue
overexpression. Proc Natl Acad Sci U S A native PAGE. Nat Protoc 1:418–428
105:14371–14376 20. Schraidt O, Marlovits TC (2011) Three-
9. Wickström D, Wagner S, Baars L, Ytterberg dimensional model of Salmonella’s needle
AJ, Klepsch M, van Wijk KJ, Luirink J, de Gier complex at subnanometer resolution. Science
JW (2011) Consequences of depletion of the 331:1192–1195
signal recognition particle in Escherichia coli. 21. Loquet A, Sgourakis NG, Gupta R, Giller K,
J Biol Chem 286:4598–4609 Riedel D, Goosmann C, Griesinger C, Kolbe
10. Wickström D, Wagner S, Simonsson P, Pop O, M, Baker D, Becker S, Lange A (2012) Atomic
Baars L, Ytterberg AJ, van Wijk KJ, Luirink J, model of the type III secretion system needle.
de Gier JWL (2011) Characterization of the Nature 486:276–279
consequences of YidC depletion on the inner 22. Klepsch M, Schlegel S, Wickström D, Friso G,
membrane proteome of Escherichia coli using van Wijk KJ, Persson JO, de Gier JW, Wagner
2D blue native/SDS-PAGE. J Mol Biol S (2008) Immobilization of the first dimension
409:124–135 in 2D blue native/SDS-PAGE allows the rela-
11. Krall L, Wiedemann U, Unsin G, Weiss S, tive quantification of membrane proteomes.
Domke N, Baron C (2002) Detergent extrac- Methods 46:48–53
tion identifies different VirB protein subassem- 23. Schlegel S, Klepsch M, Wickström D, Wagner
blies of the type IV secretion machinery in the S, de Gier JW (2010) Comparative analysis of
membranes of Agrobacterium tumefaciens. cytoplasmic membrane proteomes of
Proc Natl Acad Sci U S A 99:11405–11410 Escherichia coli using 2D blue native/SDS-
12. Kuroda T, Kubori T, Thanh Bui X, Hyakutake PAGE. Methods Mol Biol 619:257–269
A, Uchida Y, Imada K, Nagai H (2015) 24. Neuhoff V, Arold N, Taube D, Ehrhardt W
Molecular and structural analysis of Legionella (1988) Improved staining of proteins in poly-
DotI gives insights into an inner membrane acrylamide gels including isoelectric focusing
complex essential for type IV secretion. Sci Rep gels with clear background at nanogram sensi-
5:10912 tivity using Coomassie Brilliant Blue G-250
13. Wagner S, Königsmaier L, Lara-Tejero M, and R-250. Electrophoresis 9:255–262
Lefebre M, Marlovits TC, Galán JE (2010) 25. Shevchenko A, Wilm M, Vorm O, Mann M
Organization and coordinated assembly of the (1996) Mass spectrometric sequencing of pro-
type III secretion export apparatus. Proc Natl teins silver-stained polyacrylamide gels. Anal
Acad Sci U S A 107:17745–17750 Chem 68:850–858
14. Lara-Tejero M, Kato J, Wagner S, Liu X, Galán 26. Sessler N, Krug K, Nordheim A, Mordmüller
JE (2011) A sorting platform determines the B, Macek B (2012) Analysis of the Plasmodium
order of protein secretion in bacterial type III falciparum proteasome using blue native
systems. Science 331:1188–1191 PAGE and label-free quantitative mass spec-
15. Fischer M, Zilkenat S, Gerlach RG, Wagner S, trometry. Amino Acids 43:1119–1129
Renard BY (2014) Pre- and post-processing
Chapter 27
Abstract
The unique property of electron cryotomography (ECT) is its capability to resolve the structure of mac-
romolecular machines in their cellular context. The integration of ECT data with high-resolution struc-
tures of purified subcomplexes and live-cell fluorescence light microscopy can generate pseudo-atomic
models that lead to a mechanistic understanding across size and time scales. Recent advances in electron
detection, sample thinning, data acquisition, and data processing have significantly enhanced the applica-
bility and performance of ECT. Here we describe a detailed workflow for an ECT experiment, including
cell culture, vitrification, data acquisition, data reconstruction, tomogram analysis, and subtomogram aver-
aging. This protocol provides an entry point to the technique for students and researchers and indicates
the many possible variations arising from specific target properties and the available instrumentation.
Key words Tilt series, Plunge freezing, Electron cryomicroscopy, Reconstruction, Cryogen,
Segmentation
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_27, © Springer Science+Business Media LLC 2017
353
354 Gregor L. Weiss et al.
2 Materials
2.1 Plunge Freezing 1. Sample support: EM grids with a holey carbon support such as
Quantifoils (Quantifoil Micro Tools, Germany) or C-Flats
(Protochips, USA). Here we use Quantifoil R2/2 copper grids
(200 mesh). For grid specifications see Note 1.
2. Cleaning and hydrophilization of grids: Glow discharge system
such as Emitech K100X (Quorum Technologies, UK) and a
glass slide as grid support.
3. Preparation of gold fiducial markers: Bovine serum albumin
(BSA), double-distilled (dd) water, 10 nm gold nanoparticles,
and a tabletop centrifuge.
4. Vitrification of cells on grids: Plunge-freezing device such as
EM GP (Leica Microsystems, Germany), Cryoplunge 3
(Gatan, USA), or a custom-built model. Here we use a Vitrobot
MK II (FEI, USA).
5. Blotting of excess liquid: Whatman filter paper (diameter: 47 mm,
grade #1) and a device for punching holes into the filter paper.
6. Cryogen for plunge freezing: Ethane/propane (37%/63%
vol/vol; ±2%) gas mixture.
7. Cooling agent: Liquid nitrogen (LN2) in a 4 L cryo-dewar.
8. Grid storage: Cryo-grid storage boxes and transfer release tool
(TGS Technologies) or custom-made systems. Long-term LN2
storage dewars and 50 mL Falcon tubes attached to strings.
9. Cryo-transfer dewars (350 mL) for grid box transfer.
3 Methods
3.1 Cultivation Culture the bacteria. Optimize the growth conditions (e.g.,
of Bacteria medium, temperature, gas atmosphere, shaking/static incubation,
liquid/solid medium) to obtain a high expression level for the
secretion system of interest (see Note 3) while at the same time
minimizing the cell diameter, resulting in higher-quality data (see
Note 4). Ideally, cells are grown in a few milliliters of liquid
medium. For a protocol on how to proceed with cells grown on
solid medium, see Note 5. For a description of how to grow
Electron Cryo-Tomography 357
5. Mix the bacterial culture with gold fiducial markers (4:1 vol/
vol). Prepare this shortly before plunge freezing to avoid alter-
ations of the sample.
6. Pick up a glow-discharged grid with dry Vitrobot tweezers.
Mount the tweezers on the Vitrobot and lift the grid up into
the blotting chamber by pressing the foot pedal. Apply 3–4 μL
of the cell/gold suspension onto the carbon side of the grid
and continue by pressing the foot pedal. The grid is then auto-
matically blotted from both sides, removing excess liquid.
After the drain time, the grid is plunged into the liquid eth-
ane/propane mixture. Due to the high thermal conductivity of
the cryogen, freezing happens very rapidly without the forma-
tion of ice crystals, resulting in vitreous ice [42, 45]. See Note 9
for ways to increase the amount of cells on the grid.
7. From this point, the grids must be kept at liquid nitrogen tem-
perature to prevent thawing, ice contamination, and devitrifi-
cation. Carefully remove the grid from the cryogen and place
it into a labeled and precooled grid storage box.
8. In rare cases, the expression level or assembly of a secretion
system might be affected by the short incubation time on the
grid or blotting of excess liquid. To verify the assembly state of
a fluorescently tagged secretion system after plunge freezing,
the frozen grid can be analyzed in a cryogenic fluorescence
light microscope [25].
9. For storage, grid boxes are dropped into 50 mL Falcon tubes.
The tubes are closed with lids attached to strings and kept in
long-term liquid nitrogen storage dewars.
3.3 Loading Grids 1. Cool down the loading station chamber and cryopump. Place
into Electron the grid box and Polara cartridges into the chamber. Load a
Microscope C-clip ring into a C-clip tool and cool the tip using LN2. Using
cooled forceps, transfer a grid into a cartridge and clamp it
with the C-clip ring. Repeat for up to six grids/cartridges total.
2. Detach the cooled multispecimen holder (MSH) from the
Polara and attach it to the loading station. Vent the holder and
insert the cartridge rod into the chamber. Load the cartridges
into the parking positions of the MSH with the threads facing
inward. Retract the rod and pump the vacuum in the MSH
using the rough pump and cryopump. Start the turbomolecu-
lar pump of the Polara. Detach the MSH from the loading
station and attach it to the Polara. Click “Pump airlock” in the
“Setup” tab of the microscope software and open valves A and
B when prompted by the software.
3. Push the MSH to the intended cartridge position. The first
position refers to the lowest parking position in the cryo load-
ing station. Use the cooled insertion rod to pick up a cartridge
from the MSH. Retract the insertion rod and the MSH.
Electron Cryo-Tomography 359
3.4 Basic Microscope Microscope alignments change over time and must be adjusted
Alignments/ before data collection. Here we describe the procedure for a cooled
Preparations FEI Polara instrument equipped with a Gatan imaging filter and
K2 Summit direct electron detector.
1. Choose a magnification for data collection. The pixel size on
the specimen level should be at least two times smaller than the
intended resolution. Keep in mind that it might be necessary
to bin the data during processing, resulting in a larger final
pixel size. The following microscope preparations should be
made at the chosen magnification.
2. To center the condenser (C2) aperture, move the stage in X/Y
to an area of broken sample support (hole) and save this posi-
tion in the “Stage” tab of the microscope software. Drop down
the fluorescent screen, condense, and spread the beam repeat-
edly. Adjust the condenser aperture screws so that the beam
condenses and spreads centered without lateral movement.
3. To correct for an astigmatic beam, activate the condenser stig-
mators in the “Direct alignments” tab and adjust them using
the X/Y multifunction knobs.
4. Pivot points are aligned to ensure that the beam stays centered
on the camera when the focus is being changed. In “Direct
alignments,” select “Beam tilt ppx.” Condense the beam and
minimize movements by adjusting the X/Y multifunction
knobs. Repeat for “Beam tilt ppy,” click “done,” and spread
the beam. Lift the viewing screen.
5. To adjust the position of the grid so that the target does not
move laterally during tilting (eucentric height), move the stage
in X/Y to center a feature, e.g., an ice particle on carbon. Start
the “Wobbler” in the “Stage” tab and adjust the height of the
grid by pressing the “+/−“ Z-axis buttons to minimize move-
ments of the centered feature (use the K2 detector to view with
an exposure time of 0.1 s at data collection magnification).
6. Focus the feature using the focus knob.
7. Press the “Eucentric focus” button on the microscope control
panel
8. To center the beam along the optical axis of the objective lens,
select “Tomo rotation center” in “Direct alignments.”
Minimize movements of the image (viewed on K2 detector in
linear mode) using the X/Y multifunction buttons. Using the
focus step size knob, the amplitude of the objective lens cur-
rent oscillation can be adjusted, which results in different
intensities of image movement.
360 Gregor L. Weiss et al.
3.5 Automated The next step is the acquisition of tilt series (2D projection images
Sequential Tilt Series from different angles). The sequential recording of tilt series for a
Acquisition number of targets is performed automatically using a suitable soft-
ware package (see Subheading 2.2, step 2). Here we describe the
use of UCSF Tomography [32]. The program operates by record-
ing a low-magnification atlas of a grid region for the localization of
targets, intermediate-magnification images for the inspection of
potential targets, and final data collection on a selection of targets.
For this purpose, the program specifies five different modes that
correspond to different settings of the microscope and the camera:
Atlas, Search, Track, Focus, and Collect. Table 1 lists the main
function for each mode and a set of parameters that can serve as a
good starting point for data collection.
Electron Cryo-Tomography 361
Table 1
UCSF tomography modes and example parameters
Fig. 1 Steps in ECT data acquisition and analysis. Examples are shown for (a) an atlas covering several
squares, (b) a search image of a bacterial cell lying across the carbon support (c) an individual tilt image of
arrays of metamorphosis-associated contractile structures, (d) a slice through a cryotomogram that was
reconstructed from the tilt series shown in c, (e) two views of a segmentation of the tomogram shown in d,
(f) a slice through an average generated from subvolumes of tomograms similar to d, (g) an isosurface of the
average shown in f. c–e were generated with data from [16]. Bars, 1000 nm in a and b, 100 nm in c–e,
10 nm in f and g
Insert the base name for tilt series into the “File” field and
locate the target file in the “Target” field. Press “More” to set
additional imaging parameters. Check “Dose Fract” to enable
dose fractionation and enter total exposure time and subframe
exposure time (see Note 12). Select “UShort” and “Cryo” for
“MRC Data Type.” Check “Align” for “ZLP Alignment” (to
align zero-loss peak before each tilt series) and “Close at End”
(to close column valves after data acquisition).
12. To set the electron dose, move to a hole in the carbon support
and switch to Collect mode. Acquire an image with DM and
read out the dose for the image (e−/pixel). By adjusting the
beam intensity, spot size, and exposure time, set the dose for
individual tilt images. See Note 12 for considerations on total
dose, dose rate, spot size, exposure time, and dose-fractionation
parameters.
364 Gregor L. Weiss et al.
3.6 Data Processing 1. Motion correction. Imperfections in the microscope stage and
the incident electron beam result in sample movement during
exposure [23]. To correct for this motion, the projection
images for each tilt image are read out as subframes (dose frac-
tionation) and saved as image stacks. The subframes of a given
tilt image are then computationally aligned to each other, aver-
aged, and saved. Move the uncorrected tilt series and the cor-
responding subframes to an empty folder. Rename each of the
numbered subframe stacks from “(…).mrc001” to “(…)_001.
mrc,” “(…).mrc002” to “(…)_002.mrc,” and so on. Open a
terminal window within that folder and enter the command
“alignframes –stack <uncorrected tilt series.mrc> <sub-frames-
basename_*> <output file.mrc>.” The measured drift for each
frame set can be monitored in the terminal window.
2. The next step is the reconstruction of the tilt series into a 3D
image (tomogram). Move the motion-corrected tilt series to a
new directory. Open a terminal window, change to the preced-
ing directory, and start the IMOD program eTomo. The docu-
mentation for programs of the IMOD package can be accessed
via the Help menu. Locate the tilt series (stack) under the
“Build Tomogram” tab. See Note 14 for automated data
reconstruction.
3. Specify tilt series parameters. Enter the diameter of the gold
fiducials (10 nm), enter tilt series axis type (single axis), and
choose “cryosample.adoc” as the system template. Click “Scan
header” to read pixel size and image rotation from the tilt
series file. Click “View raw image stack” to inspect the selected
tilt series (see Subheading 3.7, step 1 on using 3dmod for data
visualization). Specify tilt images to be excluded from recon-
struction (e.g., images in which the grid bar blocks the beam
at high tilts) to the “Exclude views” field. Click “Create Com
Scripts.”
Electron Cryo-Tomography 365
4. The procedure now follows the eTomo panels from top (“Pre-
processing”) to bottom (“Clean Up”). Note that intermediate
files are saved and it is possible to go back and rerun certain
steps with changed parameters. Also note that moving the
mouse over an input field in IMOD will show more informa-
tion on the required input parameters.
5. “Pre-processing.” To remove pixels with aberrantly high inten-
sity. Set 12 for “Peak criterion” and 9 for “Difference crite-
rion.” Click “Create Fixed Stack” and inspect the processed tilt
series by clicking “View Fixed Stack.” Click “Use Fixed Stack”
and proceed by clicking “Done.”
6. “Coarse Alignment.” To align successive tilt images, click
“Calculate Cross-Correlation.” Bin the coarsely aligned image
stack by 2 and check “Reduce size with antialiasing filter”
(note that this binning will not affect the size of the final recon-
struction). Click “Generate Coarse Aligned Stack” and inspect
the aligned stack by clicking “View Aligned Stack in 3dmod.”
Use “Midas” to fix any misaligned image pairs. Proceed with
“Done.”
7. “Fiducial Model Gen.” The selection of gold markers and the
tracking of the selected markers throughout the tilt series can
be performed automatically using the program RAPTOR, or
manually. Here, we manually select the fiducial markers after
selecting “Make seed and track” and “Make seed model manu-
ally.” Open the aligned tilt series by clicking “Seed Fiducial
Model.” Select 10–20 gold fiducial markers distributed over
the entire 0° tilt image, and save the model before closing
3dmod. Select the “Track Beads” tab and track the fiducials
over the whole tilt series with “Track Seed Model.” Open the
fiducial model by clicking “Fix Fiducial Model” and modify
the model to minimize the number of gaps and to fix mis-
tracked fiducials. Save the model, click “Done,” and proceed.
8. “Fine Alignment.” Calculate the fine alignment with “Compute
Alignment.” Inspect by clicking “View/Edit Fiducial Model.”
Inspect fiducials by clicking “Go to Next Big Residual” and if
appropriate fix the model by clicking “Move Point by Residual.”
Repeat until there are no more residuals. Save model and
repeat “Compute alignment.” Proceed with “Done.”
9. “Tomogram Positioning.” Enter “1500” as the preliminary
tomogram Z-height in the field “Positioning tomogram thick-
ness” and click “Create Whole Tomogram.” Click “Create
Boundary Model” and flip the tomogram 90° using “Edit >
Image > Flip/Rotate.” Indicate the boundaries of the cell on a
central slice (“View axis position”) by drawing two horizontal
lines. Repeat this for one higher and one lower slice (“View
axis position”). Save the model by typing “s,” close 3dmod,
and press “Compute Z Shift & Pitch Angles.” Click “Create
Final Alignment” and proceed with “Done.”
366 Gregor L. Weiss et al.
10. “Final Aligned Stack.” Enter the binning factor in the “Aligned
image stack binning” field. We usually use a factor of 2, result-
ing in data with 1920 pixels in X and 1854 pixels in Y. Check
“Use linear interpolation” and “Reduce size with antialiasing
filter.” Click “Create Full Aligned Stack.” Other, optional pro-
cessing steps in this tab allow for the erasing of gold beads,
CTF correction, and 2D filtering.
11. “Tomogram Generation.” Choose between two different
tomogram reconstruction algorithms: weighted backprojec-
tion in Fourier space [46] or the Simultaneous Iterative
Reconstruction Technique (SIRT) [47]. Click “Generate
Tomogram” and proceed with “Done.”
12. “Post-processing.” Open the tomogram with “3dmod Full
Volume.” Select a slice with structures of interest. Use the con-
trast sliders to determine the maximum and minimum values
for “black” and “white,” needed to visualize the structure of
interest. Enter the numbers into the “Scale to match contrast”
fields. For “Reorientation,” choose either “Rotate around X”
or “Swap Y and Z dimensions.” Note that this option will
affect the handedness of the data. The correct option must be
determined as described previously [48]. Click “Trim Volume”
and “Done.”
13. “Clean Up.” Select all listed intermediate files and click “Delete
Selected” to save disk space. Finish by clicking “Done.” Note
that the “Com Scripts Interface” for a specific reconstruction
can be reopened using the “.edf” file.
3.7 Data 1. View the tomogram. Open the “.rec” file in 3dmod of the
Visualization IMOD package using the command “3dmod <input file.rec>”
(see Fig. 1d for an example). 3dmod offers different viewing
tools, including “ZAP,” “XYZ,” and “Slicer.” “Slicer” is par-
ticularly useful to detect macromolecular complexes (such as
secretion systems), based on the possibilities of rotating the
tomogram around all three axes (use sliders “X-, Y-,
Z-rotation”), pan up and down slice by slice (use slider “View
axis position”), and average multiple slices to enhance contrast
(enter value in “Img”). To measure distances, select “model”
mode in the 3dmod information window, click the left mouse
button on point A, move the mouse to point B, and type “q.”
The A-B distance (in 3D) will be reported in the log of the
3dmod information window.
2. Generate 3D models by segmentation and visualize them.
Segmentation (visualization of selected pixels) can be per-
formed automatically, e.g., by applying a density threshold
(use 3dmod’s “Isosurface” in the “Image” drop-down menu)
or by algorithms that trace specific features such as membranes
or filaments (e.g., Shape, at bio3d.colorado.edu, or other
automated segmentation approaches [49–51]). The poor con-
Electron Cryo-Tomography 367
dow. Using the “Slicer,” rotate the tomogram to the best pos-
sible view of a particular secretion system. Click with the
middle mouse button on the feature of interest to define the
center of a subvolume. Click a second point to indicate the
orientation of the subvolume (e.g., perpendicular to the cell
envelope or along a tube). The program draws a line between
both model points (green line and circles in Fig. 2.). Type “n”
(for new Contour) and repeat for all subvolumes in this tomo-
gram. Record the number of points within the object and save
the model as a “.mod” file. Repeat the procedure for addi-
tional tomograms and save the corresponding model files.
Note that all tomograms should have the same pixel size.
2. Compute an initial motive list. This list contains information
about the orientation of the modeled subvolumes. Run the
IMOD command “stalkInit <input file.mod>” in the folder
where the model is located.
3. Open PEET via the eTomo graphical user interface and select
a base name and a folder. Parameters from previous projects
can be imported.
4. Load the tomograms and corresponding models (“head.mod”
file, generated by the stalkInit command) into the respective
fields of the “Volume Table.”
5. As “Reference” for the first round of alignment, choose a ran-
dom subvolume.
Table 2
PEET subtomogram averaging iteration table
4 Notes
1. The choice of the hole size and the distance between holes in
the carbon film depends on the sample and the data collection
magnification. For Quantifoils, both parameters are specified
in the grid type, e.g., R2/1 designates a grid with 2 μm hole
diameter and 1 μm hole distance. Larger hole diameters and
smaller hole distances provide more area for imaging without a
carbon background; however, they result in higher fragility
(resulting in more breakage during transfers and more beam-
induced sample movement during data collection). The mesh
number specifies the number of squares on the grid. A higher
mesh number results in smaller area per square, which in turn
provides higher stability.
2. Alternative 300 kV instruments are the Titan Krios (FEI),
Titan Halo (FEI), and JEOL3200 (JEOL, Japan). A 200 kV
instrument, such as a Tecnai F20 (FEI) with cryosample holder
(Gatan) or Talos Arctica (FEI), can be used for thin specimens
or for screening freezing conditions. The imaging filter is criti-
cal for improving the signal-to-noise ratio by removing inelas-
tically scattered electrons, in particular for thicker samples.
Electron Cryo-Tomography 371
Acknowledgments
References
16. Shikuma NJ, Pilhofer M, Weiss GL, Hadfield 28. Hagen WJH, Wan W, Briggs JAG (2016)
MG, Jensen GJ, Newman DK (2014) Marine Implementation of a cryo-electron tomogra-
tubeworm metamorphosis induced by arrays of phy tilt-scheme optimized for high resolution
bacterial phage tail-like structures. Science subtomogram averaging. J Struct Biol
343:529–533 29. Bharat TAM, Russo CJ, Löwe J, Passmore LA,
17. Hu B, Morado DR, Margolin W, Rohda JR, Scheres SHW (2015) Advances in single-
Arizmendi O, Picking WL, Picking WD, Liu particle electron cryomicroscopy structure
J (2015) Visualization of the type III secretion determination applied to sub-tomogram aver-
sorting platform of Shigella flexneri. Proc Natl aging. Structure 23:1743–1753
Acad Sci U S A 112:1047–1052 30. Suloway C, Pulokas J, Fellmann D, Cheng A,
18. Kudryashev M, Diepold A, Amstutz M, Guerra F, Quispe J, Stagg S, Potter CS,
Armitage JP, Stahlberg H, Cornelis GR (2015) Carragher B (2005) Automated molecular
Yersinia enterocolitica type III secretion injec- microscopy: the new Leginon system. J Struct
tisomes form regularly spaced clusters, which Biol 151:41–60
incorporate new machines upon activation. 31. Mastronarde DN (2005) Automated electron
Mol Microbiol 95:875–884 microscope tomography using robust predic-
19. Nans A, Kudryashev M, Saibil HR, Hayward tion of specimen movements. J Struct Biol
RD (2015) Structure of a bacterial type III 152:36–51
secretion system in contact with a host mem- 32. Zheng SQ, Keszthelyi B, Branlund E, Lyle JM,
brane in situ. Nat Commun 6:10114 Braunfeld MB, Sedat JW, Agard DA (2007)
20. Chang YW, Rettberg LA, Treuner-Lange A, UCSF tomography: an integrated software
Iwasa J, Segaard-Anderson L, Jensen GJ suite for real-time electron microscopic tomo-
(2016) Architecture of the type IVa pilus graphic data collection, alignment, and recon-
machine. Science 351:1165–1172 struction. J Struct Biol 157:138–147
21. Kühlbrandt W (2014) The resolution revolu- 33. Li X, Mooney P, Zheng S, Booth CR, Braunfeld
tion. Science 343:1443–1444 MB, Gubbens S, Agard DA, Cheng Y (2013)
22. McMullan G, Faruqi AR, Clare D, Henderson Electron counting and beam-induced motion
R (2014) Comparison of optimal performance correction enable near-atomic-resolution
at 300 keV of three direct electron detectors single-
particle cryo-EM. Nat Methods
for use in low dose electron microscopy. 10:584–590
Ultramicroscopy 147:156–163 34. Mastronarde DN (2008) Correction for non-
23. Campbell MG, Cheng A, Brilot AF, Moeller A, perpendicularity of beam and tilt axis in tomo-
Lyumkis D, Veesler D, Pan J, Harrison SC, graphic reconstructions with the IMOD
Potter CS, Carragher B, Grigorieff N (2012) package. J Microsc 230:212–217
Movies of ice-embedded particles enhance 35. Kremer JR, Mastronarde DN, McIntosh JR
resolution in electron cryo-microscopy.
(1996) Computer visualization of three-
Structure 20:1823–1828 dimensional image data using IMOD. J Struct
24. Danev R, Buijsse B, Khoshouei M, Plitzko JM, Biol 116:71–76
Baumeister W (2014) Volta potential phase 36. Kunz M, Frangakis AS (2014) Super-sampling
plate for in-focus phase contrast transmission SART with ordered subsets. J Struct Biol
electron microscopy. Proc Natl Acad Sci U S A 188:107–115
111:15635–15640 37. Chen Y, Förster F (2014) Iterative reconstruc-
25. Briegel A, Chen S, Koster AJ, Plitzko JM, tion of cryo-electron tomograms using non-
Schwartz CL, Jensen GJ (2010) Correlated uniform fast Fourier transforms. J Struct Biol
light and electron cryo-microscopy. Methods 185:309–316
Enzymol 481:317–341 38. Agulleiro JI, Fernandez JJ (2015) Tomo3D
26. Chang YW, Chen S, Tocheva EI, Treuner-Lange 2.0—exploitation of advanced vector eXten-
A, Löbach S, Søgaard-Anderson L, Jensen GJ sions (AVX) for 3D reconstruction. J Struct
(2014) Correlated cryogenic photoactivated Biol 189:147–152
localization microscopy and cryo- electron 39. Schindelin J, Arganda-Carreras I, Frise E,
tomography. Nat Methods 11:737–739 Kaynig V, Longair M, Pietzsch T, Preibisch S,
27. Rigort A, Bäuerlein FJB, Villa E, Eibauer M, Rueden C, Saalfeld S, Schmid B, Tinevez JY,
Laugks T, Baumeister W, Plitzko JM (2012a) White DJ, Hartenstein V, Eliceiri K, Tomancak
Focused ion beam micromachining of eukary- P, Cardona A (2012) Fiji: an open-source plat-
otic cells for cryoelectron tomography. Proc form for biological-image analysis. Nat
Natl Acad Sci U S A 109:4449–4454 Methods 9:676–682
Electron Cryo-Tomography 375
Abstract
Structural studies of biocomplexes using single-particle cryo-electron microscopy (cryo-EM) is now a
well-established technique in structural biology and has become competitive with X-ray crystallography.
The latest advances in EM enable us to determine structures of protein complexes at 3–5 Å resolution for
an extremely broad range of sizes from ~200 kDa up to hundreds of megadaltons (Bartesaghi et al.,
Science 348(6239):1147–1151, 2051; Bai et al., Nature 525(7568):212–217, 2015; Vinothkumar et al.,
Nature 515(7525):80–84, 2014; Grigorieff and Harrison, Curr Opin Struct Biol 21(2):265–273, 2011).
The majority of biocomplexes comprise a number of different components and are not amenable to crys-
tallisation. Secretion systems are typical examples of such multi-protein complexes, and structural studies
of them are extremely challenging. The only feasible approach to revealing their spatial organisation and
functional modification is cryo-EM. The development of systems for digital registration of images and
algorithms for the fast and efficient processing of recorded images and subsequent analysis facilitated the
determination of structures at near-atomic resolution. In this review we will describe sample preparation
for cryo-EM, how data are collected by new detectors, and the logistics of image analysis through the basic
steps required for reconstructions of both small and large biological complexes and their refinement to
nearly atomic resolution. The processing workflow is illustrated using examples of EM analysis of a Type
IV Secretion System.
Key words Cryo-electron microscopy, Sample preparation, Single particle analysis, Image processing,
Type IV secretion system
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_28, © Springer Science+Business Media LLC 2017
377
378 Tiago R.D. Costa et al.
Fig. 2 EM maps of different T4SS structures. (a) Cryo-EM structure of core outer-membrane complex at 15 Å
resolution. (b) Cryo-EM structure of the core outer-membrane complex at 12.4 Å resolution. (c) Structure of
almost complete the T4SS complex (in negative stain) at overall resolution of ~20 Å
Fig. 3 Cryo-EM sample preparation. (a) 3 mm copper mesh grid covered with a film of holey carbon. (b) Magnified
image of square patch showing microscopic holes in carbon. (c) Enlarged image of a single hole containing a
layer of vitrified ice with protein molecules. (d) Cross section of a hole with particles embedded in ice
Fig. 4 Sample vitrification. Left panel: grid with sample applied being held in twee-
zers; right panel : after blotting excess of a sample, grid is plunged into container
filled with liquid ethane. The top level of the container should be immersed in the
nitrogen atmosphere that has a temperature slightly above the temperature of
liquid nitrogen. The grid is then transferred into the grid holder. The transfer must
be done without taking the grid out of the nitrogen atmosphere
3.1 CCD Cameras EM, like all modern photographic systems, is related to the acqui-
sition of images and now uses digital cameras. All digital cameras
convert analogue optical signals into digital format so the steps in
developing and scanning film have become redundant. The first
digital cameras used charge-coupled device (CCD) sensors, which
had been invented in 1969 by W.S. Boyle and G.E. Smith at Bell
Telephone Laboratories (Nobel Prize, 2009) [27]. The concept
behind this device was based on the transformation of an analogue
signal, such as photon energy, into an electrical charge using spe-
cially designed photo sensors. The magnitude of the charge regis-
tered by the sensor is proportional to the energy of photons
absorbed by the sensor. CCD chips consist of an array of photosen-
sitive elements. In the readout mechanism, charges are successively
transferred to a reading register, amplified, and converted into a
digital signal. The number of reading registers determines the
speed of the CCD image recording. Since there are only a few of
them, the readout of these cameras is not very high.
However, in electron microscopes, the process of recording
electrons is more complicated than with photosensors, which are
not able to register electrons. Therefore, the sensors were modi-
fied in such a way that a mono- or polycrystalline scintillator that
converts electron energy into photons was placed on top of the
photosensor, and only then were photons converted into an elec-
trical signal (Fig. 5a) [28, 29]. Unfortunately, a CCD camera’s
sensitivity decreases with increased voltage of an electron micro-
scope, so thicker scintillator layers are needed to improve the elec-
tron detection efficiency. Thick layers of scintillators affect the
image quality, which is degraded because the higher-energy elec-
trons are scattered through several adjacent sensors, reducing the
image resolution. Nonetheless, these cameras provided experience
and understanding for the development of automated data collec-
tion in EM.
3.2 Direct Electron In the last decade, new digital detectors have been designed
Detectors enabling the detection of electrons without the intermediate step
of transforming electrons into photons and then into an electrical
signal (Fig. 5b). Direct detector devices (DDDs) use an array of
radiation-hardened active pixel sensors (a pixel circuit) which are
integrated into a silicon complementary metal-oxide semiconduc-
tor (CMOS) chip [30, 31]. In this case the electron energy is trans-
formed directly into an electrical signal. Another advance in this
384 Tiago R.D. Costa et al.
Fig. 5 Digital cameras. (a) In CCD-based cameras, the electrons hit a scintillator,
generating light, which is partially captured by fibre optics, and directed onto the
cooled CCD chip. (b) In direct detectors (two right panels ), active pixel sensors,
mostly based on CMOS technology, are capable of capturing and directly detect-
ing incident electrons
The ratio depends on the spatial frequency (sizes of the details)
of the image. A perfect detector would not distort the input signal,
so in an ideal system the output should be the same as the input.
Therefore, the DQE of an ideal system would be equal to one for
all frequencies. In reality, cameras distort the fine details in images,
and this is reflected by a significant decline in the DQE at high
frequencies [34, 35].
Direct detectors make it possible to register electrons within a
high range of energy and are now used in 300 keV microscopes.
Cryo Electron Microscopy 385
3.3 Micrograph Cryo-EM images using DDD cameras can record from 7 sub-
Subframe Alignment frames on Falcon (FEI) to 40–50 subframes using direct electron
(DE) or K2 (Gatan) cameras. Thus, the data recorded with DDD
cameras represent sets of image frames (movies) that can be
motion-corrected. Such high rates of image recording can reveal
distortions of images induced by drift of the grid (sample) within
the EM. Typically, frame alignment starts from frame N−1 that is
aligned to the last frame. Then these two images are summed, and
frame N−2 will be aligned to this sum. Then frames N−1 and N−2
will be summed, and frame N−3 is aligned to this new sum. The
process is repeated in the same way towards the first frame. There
are variations in algorithms when summation is done not for two
frames for the following alignment but for four or five frames or all
frames. Alignment is refined iteratively when on the next round of
alignment the total sum obtained during the previous round is
used as a reference. The entire procedure improves, firstly, SNR of
the reference and then the quality of the alignment. Nowadays a
number of software packages can be used for the frame alignment
[36, 38–41]. The image shown in Fig. 6a represents the sum of the
original frames without any correction. The trajectory of the image
shift during these several exposures indicates that the movement of
initial shifts of the sample is large initially but then slows down
(Fig. 6b). A power spectrum from the sum of the frames without
motion correction demonstrates that the Thon rings are not very
sharp: they fade fast owing to the small shifts in different directions
shown in Fig. 6b (red dots). When the movie frames are aligned
(the motion correction), the Thon rings become symmetrical,
going up to 3 Å (Fig. 6c). This indicates the presence of high-
resolution details in the images. Summation of the motion-
corrected subframes generates the final sharper image (Fig. 6d).
Fig. 6 Cryo-EM images with motion correction. (a) Representative cryo-EM image of vitrified T4SS particles. (b)
Trace of motion in X and Y directions of frames. (c) Left : power spectrum from the sum of raw movie frames
without motion correction. Right : power spectrum from the sum of movie frames after motion correction. (d) Sum
of movie frames that were shifted according to determined shifts shown in (b). Protein is black in these images
Fig. 7 CTF with envelope function. Dotted blue line : amplitude of all frequencies
in perfect microscope; green line: effect of envelope function on CTF (red) result-
ing in suppression of high spatial frequencies
4.2 Defocus To correct an image for CTF effects and obtain an image that cor-
Determination responds to the projection, it is necessary to determine its defocus
and Correction of CTF and to check it for astigmatism and drift. The nominal value of
defocus set on the microscope does not usually represent the actual
defocus obtained in the final digital image or micrograph. This
occurs because, although the acceleration voltage and spherical
aberration remain constant, the common deviations in sample
thickness and the position of the supporting film will cause local
variations in defocus. As a result, the CTF related to the defocus
should be determined for each image frame. Finding the exact level
of defocus and astigmatism in cryo-EM images is of absolutely cru-
cial importance when working on the production of a high-
resolution structure.
CTF determination is performed by calculating the sum of
power spectra (or amplitudes) of small patches (256 × 256 or
slightly larger) from the sum of all subframes. This spectrum is cor-
related with a number of CTFs theoretically calculated in a range
of possible defocus values. A maximum correlation between the
observed and a theoretical CTF would indicate the actual defocus
of the values and will define frequencies where the phases must be
flipped (Fig. 8). Different options for (semi-) automated defocus
determination are available in a number software packages, such as
EMAN2, CTFIND, and IMAGIC5 [55–57].
Astigmatic images have power spectra which are not rotation-
ally symmetric, and this can complicate and reduce the accuracy of
CTF determination. Generally cryo-EM images which have greater
than 5% astigmatism are not used for further processing except in
special cases where strong astigmatism can be used to recover
information in areas where the CTF crosses zeroes. The level of
astigmatism can be calculated as follows:
(
Astigmatism = Defocus max - Defocus min / Defocus avg )
An EM projection image is only considered a faithful represen-
tation of the observed object of interest if it has been corrected for
the CTF modulation effects of the microscope. This can only be
done after CTF determination. Phase correction is carried out in
reciprocal space by multiplying the alternating rings of the CTF by
Cryo Electron Microscopy 391
Fig. 9 CTF correction. (a) CTF oscillates changing contrast from negative to posi-
tive depending on frequencies. Information is lost only where CTF crosses zero
line. (b) Negative lobes of uncorrected CTF are flipped over to positive (correction
of CTF by phase flipping). The missing information can be recovered by collecting
images at different defocus levels which fill these zero regions with information
4.3 Particle Selection The structural analysis process in EM begins with selecting images
of individual particles from micrographs. This involves recording
their unique locations (x,y) within the image field and saving these
coordinates in a data file that will be used in the next steps of pro-
cessing. This can be done interactively using packages like Xmipp
[59], EMAN2 [55], Ximdisp [60], RELION (semi-automated
selection of cryo-EM particles in RELION-2 [61], and others.
The simplest way to do this is to select a single particle image by
clicking on the image with a mouse. The coordinates of these
points will be stored and then used to extract individual particles
within a square box of designated dimension, for example, 500 ×
500 pixels. The cut-out area must be large enough to retain all the
image data around the object with as little background as possible.
Particles can also be selected automatically with particle identifica-
tion/selection programs, for example, Autopicker [62], BShow
[63], and FindEM [64]. These programs use the assessment of
local correlation to measure the degree of similarity between refer-
ence images and then a small area of the raw micrograph. Areas
which show maximum correlation to the references are boxed out.
392 Tiago R.D. Costa et al.
The poor contrast in ice images and the presence of artefacts which
could resemble target particles can generally increase the inaccu-
racy of automatic selection.
The images of selected particles must not overlap with other
particles and particles in images should not be distorted. In Fig. 10
we show an example of a micrograph of a vitrified sample of the
T4SS core OMC. Images were recorded on a 4096 × 4096 Gatan
CCD camera with a low electron dose on a Tecnai F20 FEG micro-
scope operating at a voltage of 200 kV, a magnification of 68,100,
and a defocus range of 1250–3500 nm.
rinorm
,j = (( r
i, j ) )
- ravg / s old s new ,
σold and σnew are the standard deviations of the original and target
images respectively, and ρi,j is the density of a pixel in the image
array coordinates.
Cryo Electron Microscopy 393
4.5 Alignment One of the most important steps in image processing is the reduc-
of Particle Images tion of noise and enhancement of the signal. Different factors,
such as insufficient coherence of the electron beam, quality of
amorphous ice (due to uneven distribution of salts and some other
effects in buffers), supporting films, and the noise of registering
cameras, contribute to decreasing the SNR in particle images.
Since these types of noise are not related to a signal from the sam-
ple, the averaging of particle images improves the SNR. However,
to retrieve reliable information using averaging, images must rep-
resent the same particles in the same orientations [44]. Therefore,
images of particles in the same orientations should be identified,
and before averaging, the images should be aligned rotationally
and translationally with respect to each other. Alignment compares
all images to a reference image and shifts them so that they are in
the same position as the reference. Usually, normalised particle
images should be centred or aligned to the reference that repre-
sents a typical view of the complex.
One of the possible options for starting analysis is to align all
images of a data set to the rotationally averaged total sum of all
images (with no alignment). The centred images are then sub-
jected to multivariate statistical analysis (MSA) (see following dis-
cussion) to obtain a number of averages of images which are
aligned only translationally and grouped according to common
features. The best characteristic views (averages of groups of images
with the lowest variations among them) are used for multi-reference
alignment. The most reliable classes are centred and used as new
references for the next round of alignment (Fig. 11). The proce-
dure can be repeated several times alternating with MSA [65]. In
other cases one can use alignment in Fourier space [66] or so-
called reference-free alignment implemented in EMAN2 and
SPIDER [55, 67].
394 Tiago R.D. Costa et al.
4.6 Statistical To improve the SNR, aligned images which have high similarity
Analysis between each other should be grouped together, and for this pur-
and Classification pose, statistical analysis and classification are used. Different
approaches have been developed to reduce a large number of vari-
ables to a limited number of important parameters [68].
4.6.1 Principal Principal component analysis (PCA) reduces the number of variables
Component Analysis to find the most significant variations in the measurements [44, 65].
The essence of the procedure is a transformation of a set of observa-
tions of possibly correlated variables (in our case images) into a set of
values of uncorrelated variables called principal components. In the
complete representation, a number of the principal components are
equal to the number of original variables. However, since images
contain a high level of noise, the number of the meaningful compo-
nents becomes much smaller. The principal components are described
by the eigenvectors of the data matrix. PCA is the simplest of the
true eigenvector-based multivariate analyses [65, 68].
4.7.1 Random Conical The random conical tilt (RCT) technique is used to obtain an ini-
Tilt tial model of a new complex about which little is known. The
method is based on a typical property of samples having a prefer-
ential orientation on a grid when negatively stained. The RCT
Cryo Electron Microscopy 397
Fig. 13 Sinograms and sinocorrelation functions for three projections. A sample object is a model of the T4SS
core outer-membrane complex, which has 14-fold symmetry. Images corresponding to projections are num-
bered 1–3. S1, S2, and S3 (sinograms) are sets of 1D projections of corresponding projections 1–3. CSC12 and
CSC32 correspond to cross-sinogram correlation (CSC) functions between projections 1 and 2 and projections
3 and 2, respectively. Each point of the CSC function represents the correlation coefficient of a pair of lines
from the two sinograms. There are 14 common lines between each pair of images since the object has 14-fold
symmetry. The highest correlations (rainbow circles ) point to the position of common lines. Some of them are
shown by dashed lines between projections 2 and 1, and the solid red line shows one common line between
projections 2 and 3 (the other are not shown). Each CSC function has all peaks doubled because projections
from 180° to 360° mirror those from 0° to 180°. The angular distance between the common lines (red solid
and dashed ) shows the angle α13 between projections 1 and 3
400 Tiago R.D. Costa et al.
R = D /N
4.9 Structure All single-particle EM packages use nearly the same procedure for
Refinement the refinement of structures after the first 3D model is obtained
(Fig. 1). It is done by a realignment of single-particle images with
new references obtained from the new model. It is often combined
with the determination of angles: projection matching with a
smaller angular increment or a local refinement of the angles [88].
Reprojections of the new model can be used as a new anchor set in
angular reconstitution to refine the orientation of the classes. The
anchor set is a set of projections calculated from the first 3D model
which are used to determine orientations for new classes or
realigned images. The angular increment between projections used
as the anchor set is usually chosen so that 100–150 projections
(which is much less compared to projection matching) will be cal-
culated and used for the refinement of angles. New Euler angles
are found and assigned to the new class averages. Sorting classes
402 Tiago R.D. Costa et al.
Fig. 15 Reconstruction using Fourier transform (FT) of classes (images). (a) T4SS
core outer-membrane complex (14-fold symmetry) is observed in different ori-
entations on the supporting film. (b) Projections of these particles that are equiv-
alent to EM images in the direction of the electron beam. (c) FT from projections
shown in b. s : FT of side view; e : FT of end view. The corresponding images are
shown in b. (d) A pair of 2D transforms that share at least one common line in
reciprocal space. Common lines between the side view (s) projection and the end
view (e) projection are indicated by green lines ; purple line: common line
between side views (shown in s1 and s7). The angles between pairs of common
lines determine the relative Euler angle orientations. (e) An inverse Fourier trans-
formation of the combined 2D transforms generates an improved real-space
structure seen as electron density
4.10 Evaluation When the map of a new structure is obtained, the molecular mass
of Quality Structure and its oligomeric state should be estimated. The map should be
inspected at the 1σ threshold of the density level, which typically
corresponds to the molecular weight of the complex in the study.
If the complex consists of several interacting proteins, the map
Cryo Electron Microscopy 403
Fig. 16 Examples of Fourier shell correlation (FSC) and resolution assessments. (a) FSC curvature of two inde-
pendent 3D structures that have been well aligned and masked with a loose soft mask shown as a wide halo
around the particle images. A resolution assessed at the 0.5 level is 15.6 Å. (b) Here the second structure is
Cryo Electron Microscopy 405
Fig. 16 (continued) rotated by 10° around the rotational axis. While the overall shape still coincides well
between both structures small details are not in the register. It is reflected in the FSC curve which declines
much faster and the resolution at the 0.5 threshold indicates only 27 Å, corresponding to a size of major
domains in the structure. (c) The structures are not aligned. FSC falls down even earlier at lower frequencies
indicating only consistency in overall sizes. (d) FSC between two 3D structures when the same tight mask was
applied. The increase at high frequencies indicates the correlation between masks imposed on the structures,
and here the resolution is overestimated
406 Tiago R.D. Costa et al.
4.10.3 Local Estimation The third approach whose popularity has risen in recent years anal-
of Resolution yses the 3D density maps obtained from a whole data set by com-
puting the cross-correlation between neighbouring voxels in the
Fourier domain, the Fourier Neighbor Correlation (FNC). A 3D
mask is applied on a 3D structure in such a manner that values
outside the mask are altered for zeroes and pixels remain unchanged
only in part of the cube. Any density inside the structure mask
counts as signal plus noise, whereas any density outside this mask
counts as noise. This operation can be represented in Fourier space
as a convolution of the Fourier transform of the mask with the
Fourier transform of the noise. The convolution provides an assess-
ment of correlations between the Fourier terms. The method has
been implemented in a computer program called RMEASURE
[100]. It is used for 3D reconstructions only.
The ResMap algorithm [101] is based on initialising a
local sinusoid model at r = 2d, where d is the voxel spacing in ang-
stroms (Å). Likelihood ratio tests are conducted at all voxels in the
volume. At a fixed wavelength equal to d, the standard likelihood
ratio test can detect whether a local sinusoid is a meaningful part of
the model approximation. The test requires an estimate of the
noise variance, which can be evaluated from the region surround-
ing the structure. The smallest r at which the likelihood ratio test
passes at a given p-value defines the resolution. The p-value is the
measure of whether the outcome of the attempt is due to an actual
effect or a mere random chance. Voxels that pass the test are
assigned a resolution r, while those that fail the test will be exam-
ined at a larger r. The algorithm produces a local resolution map
with a number assigned to every voxel in the density map.
Fig. 17 Fitting of atomic model (PDB: 3JQO) to cryo-EM map (EMD-2232) of core
outer-membrane complex. (a) Side view. (b) Top view. (c) Central cross section
6 Conclusions
Acknowledgments
The authors thank Dr. H. White for reading the manuscript and
useful discussions that led to improvements in the manuscript.
This work was funded by MRC Grant MR/K012401/1 to
E.V.O. The authors apologise for not covering all methods fully
owing to space constraints.
References
1. Steitz TA (2008) A structural understanding 9. Cheng Y, Grigorieff N, Penczek PA, Walz T
of the dynamic ribosome machine. Nat Rev (2015) A primer to single-particle cryo-
Mol Cell Biol 9(3):242–253 electron microscopy. Cell 161(3):438–449
2. Bartesaghi A, Merk A, Banerjee S, Matthies 10. Costa TR, Felisberto-Rodrigues C, Meir A,
D, Wu X, Milne JL, Subramaniam S (2015) Prevost MS, Redzej A, Trokter M, Waksman
2.2 A resolution cryo-EM structure of beta- G (2015) Secretion systems in Gram-negative
galactosidase in complex with a cell-permeant bacteria: structural and mechanistic insights.
inhibitor. Science 348(6239):1147–1151 Nat Rev Microbiol 13(6):343–359
3. Bai XC, Yan C, Yang G, Lu P, Ma D, Sun L, 11. Ilangovan A, Connery S, Waksman G (2015)
Zhou R, Scheres SH, Shi Y (2015) An atomic Structural biology of the Gram-negative bac-
structure of human gamma-secretase. Nature terial conjugation systems. Trends Microbiol
525(7568):212–217 23(5):301–310
4. Vinothkumar KR, Zhu J, Hirst J (2014) 12. Fronzes R, Schafer E, Wang L, Saibil HR,
Architecture of mammalian respiratory com- Orlova EV, Waksman G (2009) Structure of a
plex I. Nature 515(7525):80–84 type IV secretion system core complex.
5. Grigorieff N, Harrison SC (2011) Near- Science 323(5911):266–268
atomic resolution reconstructions of icosahe- 13. Rivera-Calzada A, Fronzes R, Savva CG,
dral viruses from electron cryo-microscopy. Chandran V, Lian PW, Laeremans T, Pardon
Curr Opin Struct Biol 21(2):265–273 E, Steyaert J, Remaut H, Waksman G, Orlova
6. Grant T, Grigorieff N (2015) Automatic esti- EV (2013) Structure of a bacterial type IV
mation and correction of anisotropic magnifi- secretion core complex at subnanometre reso-
cation distortion in electron microscopes. lution. EMBO J 32(8):1195–1204
J Struct Biol 192(2):204–208 14. Low HH, Gubellini F, Rivera-Calzada A,
7. Scheres SH (2010) Classification of structural Braun N, Connery S, Dujeancourt A, Lu F,
heterogeneity by maximum-likelihood meth- Redzej A, Fronzes R, Orlova EV, Waksman G
ods. Methods Enzymol 482:295–320 (2014) Structure of a type IV secretion system.
8. Orlova EV, Saibil HR (2010) Methods for Nature 508(7497):550–553
three-dimensional reconstruction of hetero- 15. Spence JCH (2003) High resolution micros-
geneous assemblies. Methods Enzymol copy, 3rd edn. Oxford University Press,
482:321–341 New York
410 Tiago R.D. Costa et al.
16. Dubochet J, Adrian M, Chang JJ, Homo JC, 31. Veesler D, Campbell MG, Cheng A, Fu CY,
Lepault J, McDowall AW, Schultz P (1988) Murez Z, Johnson JE, Potter CS, Carragher
Cryo-electron microscopy of vitrified speci- B (2013) Maximizing the potential of elec-
mens. Q Rev Biophys 21(2):129–228 tron cryomicroscopy data collected using
17. Jaffe JS, Glaeser RM (1987) Difference direct detectors. J Struct Biol
Fourier analysis of “surface features” of bacte- 184(2):193–202
riorhodopsin using glucose-embedded and 32. Cunningham IA (1999) Practical digital
frozen-hydrated purple membrane. imaging and PACS. Advanced Medical
Ultramicroscopy 23(1):17–28 Publishing for American Association of
18. Dubochet J, Lepault J, Freeman R, Berriman Physicists in Medicine, USA
A, Homo JC (1982) Electron microscopy of 33. McMullan G, Chen S, Henderson R, Faruqi
frozen water and aqueous solutions. J Microsc AR (2009) Detective quantum efficiency of
124(3):219–237 electron area detectors in electron micros-
19. Lepault J, Dubochet J (1986) Electron copy. Ultramicroscopy 109(9):1126–1143
microscopy of frozen hydrated specimens: 34. McMullan G, Faruqi AR, Clare D, Henderson
preparation and characteristics. Methods R (2014) Comparison of optimal perfor-
Enzymol 127:719–730 mance at 300keV of three direct electron
20. Cabra V, Samso M (2015) Do’s and don’ts of detectors for use in low dose electron micros-
cryo-electron microscopy: a primer on sample copy. Ultramicroscopy 147:156–163
preparation and high quality data collection 35. Ruskin RS, Yu Z, Grigorieff N (2013)
for macromolecular 3D reconstruction. J Vis Quantitative characterization of electron
Exp 95:52311 detectors for transmission electron micros-
21. Grassucci RA, Taylor DJ, Frank J (2007) copy. J Struct Biol 184(3):385–393
Preparation of macromolecular complexes for 36. Bammes BE, Rochat RH, Jakana J, Chen
cryo-electron microscopy. Nat Protoc DH, Chiu W (2012) Direct electron detec-
2(12):3239–3246 tion yields cryo-EM reconstructions at reso-
22. Adrian M, Dubochet J, Lepault J, McDowall lutions beyond 3/4 Nyquist frequency.
AW (1984) Cryo-electron microscopy of J Struct Biol 177(3):589–601
viruses. Nature 308(5954):32–36 37. Milazzo AC, Moldovan G, Lanman J, Jin L,
23. Baker LA, Rubinstein JL (2010) Radiation Bouwer JC, Klienfelder S, Peltier ST, Ellisman
damage in electron cryomicroscopy. Methods MH, Kirkland AI, Xuong NH (2010)
Enzymol 481:371–388 Characterization of a direct detection device
24. Tivol WF, Briegel A, Jensen GJ (2008) An imaging camera for transmission electron
improved cryogen for plunge freezing. Microsc microscopy. Ultramicroscopy 110(7):744–747
Microanal 14(5):375–379 38. Campbell MG, Cheng A, Brilot AF, Moeller
25. Vos MR, Bomans PH, Frederik PM, A, Lyumkis D, Veesler D, Pan J, Harrison SC,
Sommerdijk NA (2008) The development of Potter CS, Carragher B, Grigorieff N (2012)
a glove-box/Vitrobot combination: air-water Movies of ice-embedded particles enhance
interface events visualized by cryo- TEM. resolution in electron cryo-microscopy.
Ultramicroscopy 108(11):1478–1483 Structure 20(11):1823–1828
26. Jensen GJ (2010) Cryo-EM. Part A: sample 39. Li X, Mooney P, Zheng S, Booth CR, Braunfeld
preparation and data collection. Preface. MB, Gubbens S, Agard DA, Cheng Y (2013)
Methods Enzymol 481:xv–xvi Electron counting and beam-induced motion
27. Boyle WS, Smith GE (1970) Charge coupled correction enable near- atomic-
resolution sin-
semiconductor devices. J Bell Syst Tech gle-particle cryo-EM. Nat Methods
49(4):587–593 10(6):584–590
28. McMullan G, Cattermole DM, Chen S, 40. Abrishami V, Vargas J, Li X, Cheng Y,
Henderson R, Llopart X, Summerfield C, Marabini R, Sorzano CO, Carazo JM (2015)
Tlustos L, Faruqi AR (2007) Electron imag- Alignment of direct detection device micro-
ing with Medipix2 hybrid pixel detector. graphs using a robust optical flow approach.
Ultramicroscopy 107(4–5):401–413 J Struct Biol 189(3):163–176
29. Faruqi AR, Henderson R (2007) Electronic 41. Afanasyev P, Ravelli RB, Matadeen R, De
detectors for electron microscopy. Curr Opin Carlo S, van Duinen G, Alewijnse B, Peters PJ,
Struct Biol 17(5):549–555 Abrahams JP, Portugal RV, Schatz M, van
30. Ramachandra R, Bouwer JC, Mackey MR, Heel M (2015) A posteriori correction of cam-
Bushong E, Peltier ST, Xuong NH, Ellisman era characteristics from large image data sets.
MH (2014) Improving signal to noise in Sci Rep 5:10317
labeled biological specimens using energy- 42. Glaeser RM (1971) Limitations to significant
filtered TEM of sections with a drift correc- information in biological electron microscopy
tion strategy and a direct detection device. as a result of radiation damage. J Ultrastruct
Microsc Microanal 20(3):706–714 Res 36(3):466–482
Cryo Electron Microscopy 411
43. Chiu W, Jeng TW (1982) Electron radiation 59. de la Rosa-Trevin JM, Oton J, Marabini R,
sensitivity of protein crystals. Ultramicroscopy Zaldivar A, Vargas J, Carazo JM, Sorzano CO
10(1–2):63–69 (2013) Xmipp 3.0: an improved software
44. Frank J (2006) Three dimensional electron suite for image processing in electron micros-
microscopy of macromolecular assemblies: copy. J Struct Biol 184(2):321–328
visualization of biological molecules in their 60. Smith JM (1999) Ximdisp–a visualization
native state, 2nd edn. Oxford University tool to aid structure determination from
Press, New York electron microscope images. J Struct Biol
45. Egerton RF, Li P, Malac M (2004) Radiation 125(2–3):223–228
damage in the TEM and SEM. Micron 61. Scheres SH (2015) Semi-automated selection
35(6):399–409 of cryo-EM particles in RELION-1.3.
46. Burmeister WP (2000) Structural changes in J Struct Biol 189(2):114–122
a cryo-cooled protein crystal owing to radia- 62. Langlois R, Pallesen J, Ash JT, Nam Ho D,
tion damage. Acta Crystallogr D Biol Rubinstein JL, Frank J (2014) Automated par-
Crystallogr 56(Pt 3):328–341 ticle picking for low-contrast macromolecules
47. Taylor KA, Glaeser RM (1976) Electron in cryo-electron microscopy. J Struct Biol
microscopy of frozen hydrated biological 186(1):1–7
specimens. J Ultrastruct Res 55(3):448–456 63. Heymann JB, Belnap DM (2007) Bsoft:
48. Chiu W (1986) Electron microscopy of fro- image processing and molecular modeling for
zen, hydrated biological specimens. Annu electron microscopy. J Struct Biol
Rev Biophys Biophys Chem 15:237–257 157(1):3–18
49. Knapek E, Dubochet J (1980) Beam damage 64. Roseman AM (2004) FindEM--a fast, effi-
to organic material is considerably reduced in cient program for automatic selection of par-
cryo-electron microscopy. J Mol Biol 141(2): ticles from electron micrographs. J Struct Biol
147–161 145(1–2):91–99
50. Bartesaghi A, Matthies D, Banerjee S, Merk 65. Van Heel M, Portugal RV, Schatz M (2009)
A, Subramaniam S (2014) Structure of beta- Multivariate statistical analysis in single parti-
galactosidase at 3.2-a resolution obtained by cle (Cryo) electron microscopy. In: Verkley A,
cryo-electron microscopy. Proc Natl Acad Sci Orlova E (eds) An electronic textbook: elec-
U S A 111(32):11709–11714 tron microscopy in life science. 3D-EM
51. Carlson DB, Evans JE (2012) Low-dose Network of Excellence, London
imaging techniques for transmission electron 66. Grigorieff N (2007) FREALIGN: high-
microscopy. The transmission electron micro- resolution refinement of single particle struc-
scope. InTech, China tures. J Struct Biol 157(1):117–125
52. Erickson HP, Klug A (1971) Measurement and 67. Frank J, Radermacher M, Penczek P, Zhu J,
compensation of defocusing and aberrations by Li Y, Ladjadj M, Leith A (1996) SPIDER and
Fourier processing of electron micrographs. WEB: processing and visualization of images
Philos Trans R Soc B 261(837):105–118 in 3D electron microscopy and related fields.
53. Wade RH (1992) A brief look at imaging and J Struct Biol 116(1):190–199
contrast transfer. Ultramicroscopy 46:145–156 68. Bartholomew DJ, Steele F, Galbraith J,
54. Thon F (1966) Zur Defokussierungsabh ä Moustaki I (2008) Analysis of multivariate
ngigkeit des Phasenkontrastes bei der social science data. Statistics in the social and
elektronenmikroskopischen Abbildung. behavioral sciences series, 2nd edn. Taylor &
Naturforschg 21a:476–478 Francis, USA
55. Tang G, Peng L, Baldwin PR, Mann DS, 69. Myung IJ (2003) Tutorial on maximum likeli-
Jiang W, Rees I, Ludtke SJ (2007) EMAN2: hood estimation. J Math Psyc 47(1):90–100
an extensible image processing suite for elec- 70. Sigworth FJ (1998) A maximum-likelihood
tron microscopy. J Struct Biol 157(1):38–46 approach to single-particle image refinement.
56. Rohou A, Grigorieff N (2015) CTFFIND4: J Struct Biol 122(3):328–339
fast and accurate defocus estimation from 71. Scheres SH (2012) A Bayesian view on cryo-
electron micrographs. J Struct Biol 192(2): EM structure determination. J Mol Biol
216–221 415(2):406–418
57. van Heel M, Harauz G, Orlova EV, Schmidt 72. Macqueen J (1967) Some methods for classi-
R, Schatz M (1996) A new generation of the fication and analysis of multivariate observa-
IMAGIC image processing system. J Struct tions. Proc Fifth Berkeley Symp Math Stat
Biol 116(1):17–24 Prob 1:281–297
58. Wiener N (1964) Extrapolation, interpola- 73. Ward JHJ (1963) Hierarchical grouping to
tion, and smoothing of stationary time series. optimize an objective function. J Am Stat
Wiley, New York Assoc 58(301):236–244
412 Tiago R.D. Costa et al.
74. Guan W, Lockwood A, Inkson BJ, Mobus G 89. Glaeser RM, Downing KH, DeRosier DJ,
(2011) A piezoelectric goniometer inside a Chiu W, Frank J (2007) Electron crystallog-
transmission electron microscope goniome- raphy of biological macromolecules. Oxford
ter. Microsc Microanal 17(5):827–833 University Press, New York
75. Van Heel M (1987) Angular reconstitution: a 90. van Heel M, Schatz M (2005) Fourier shell
posteriori assignment of projection directions correlation threshold criteria. J Struct Biol
for 3D reconstruction. Ultramicroscopy 151(3):250–262
21(2):111–123 91. Rosenthal PB, Henderson R (2003) Optimal
76. van Heel M, Orlova EV, Harauz G, Stark H, determination of particle orientation, abso-
Dube P, Zemlin F, Schatz M (1997) Angular lute hand, and contrast loss in single-particle
reconstitution in three-dimentional electron electron cryomicroscopy. J Mol Biol 333(4):
microscopy: historical and theoretical aspects. 721–745
Scanning Microsc 11:195–210 92. Scheres SH, Chen S (2012) Prevention of over-
77. Crowther RA (1971) Procedures for three- fitting in cryo-EM structure determination.
dimensional reconstruction of spherical Nat Methods 9(9):853–854
viruses by Fourier synthesis from electron 93. Chen S, McMullan G, Faruqi AR, Murshudov
micrographs. Philos Trans R Soc Lond Ser B GN, Short JM, Scheres SH, Henderson R
Biol Sci 261(837):221–230 (2013) High-resolution noise substitution to
78. Fuller SD (1987) The T=4 envelope of measure overfitting and validate resolution in
Sindbis virus is organized by interactions with 3D structure determination by single particle
a complementary T=3 capsid. Cell 48(6): electron cryomicroscopy. Ultramicroscopy
923–934 135:24–35
79. Herman GT (1980) Image reconstruction 94. Unser M, Trus BL, Steven AC (1987) A new
from projections: the fundamentals of com- resolution criterion based on spectral signal-
puterized tomography. Academic, New York to-noise ratios. Ultramicroscopy 23(1):39–51
80. Penczek PA (2010) Fundamentals of three- 95. Unser M, Trus BL, Frank J, Steven AC
dimensional reconstruction from projections, (1989) The spectral signal-to-noise ratio res-
vol 482. Methods in enzymology: Cryo-EM, olution criterion: computational efficiency
part B, 3-D reconstruction. Academic, Elsevier, and statistical precision. Ultramicroscopy
San Diego, CA 30(3):429–433
81. Orlova EV, Saibil HR (2011) Structural anal- 96. Unser M, Sorzano CO, Thevenaz P, Jonic S,
ysis of macromolecular assemblies by electron El-Bez C, De Carlo S, Conway JF, Trus BL
microscopy. Chem Rev 111(12):7710–7748 (2005) Spectral signal-to-noise ratio and res-
82. Harauz G, van Heel M (1986a) Exact filters olution assessment of 3D reconstructions.
for general geometry three-dimensional J Struct Biol 149(3):243–255
reconstruction. Optik 73:146–156 97. Penczek PA (2002) Three-dimensional spec-
83. Orlov SS (1976) Theory of three dimensional tral signal-to-noise ratio for a class of recon-
reconstruction—conditions of a complete set of struction algorithms. J Struct Biol 138(1–2):
projections. Sov Phys Crystallogr 20:312–314 34–46
84. Radermacher M (1988) Three-dimensional 98. Kessel M, Radermacher M, Frank J (1985)
reconstruction of single particles from ran- The structure of the stalk surface layer of a
dom and nonrandom tilt series. J Electron brine pond microorganism: correlation aver-
Microsc Tech 9(4):359–394 aging applied to a double layered lattice struc-
85. De Rosier DJ, Klug A (1968) Reconstruction ture. J Microsc 139(Pt 1):63–74
of three dimensional structures from electron 99. van Heel M, Hollenberg J (1980) The
micrographs. Nature 217(5124):130–134 stretching of distorted images of two-
86. Crowther RA, DeRosier DJ, Klug A (1970) dimensional crystals. Electron microscopy at
The reconstruction of a three-dimensional molecular dimensions. Springer, Berlin
structure from projections and its application 100. Sousa D, Grigorieff N (2007) Ab initio reso-
to electron microscopy. Proc R Soc A 317 lution measurement for single particle struc-
(1530) tures. J Struct Biol 157(1):201–210
87. DeRosier DJ, Moore PB (1970) 101. Kucukelbir A, Sigworth FJ, Tagare HD
Reconstruction of three-dimensional images (2014) Quantifying the local resolution of
from electron micrographs of structures with cryo-EM density maps. Nat Methods
helical symmetry. J Mol Biol 52(2):355–369 11(1):63–65
88. Penczek PA (2008) Single particle recon- 102. Zhang R, Alushin GM, Brown A, Nogales E
struction. In: Shmueli U (ed) International (2015) Mechanistic origin of microtubule
tables for crystallography. Springer, New York, dynamic instability and its modulation by EB
pp 375–388 proteins. Cell 162(4):849–859
Cryo Electron Microscopy 413
103. Clare DK, Orlova EV (2010) 4.6A cryo-EM refinement guided by cryo-EM density.
reconstruction of tobacco mosaic virus from Structure 16(2):295–307
images recorded at 300 keV on a 4k x 4k 110. Lopez-Blanco JR, Chacon P (2013) iMOD-
CCD camera. J Struct Biol 171(3): FIT: efficient and robust flexible fitting based
303–308 on vibrational analysis in internal coordinates.
104. Brown A, Long F, Nicholls RA, Toots J,
J Struct Biol 184(2):261–270
Emsley P, Murshudov G (2015) Tools for 111. Adams PD, Afonine PV, Bunkoczi G, Chen
macromolecular model building and refine- VB, Davis IW, Echols N, Headd JJ, Hung
ment into electron cryo-microscopy recon- LW, Kapral GJ, Grosse-Kunstleve RW, McCoy
structions. Acta Crystallogr D Biol Crystallogr AJ, Moriarty NW, Oeffner R, Read RJ,
71(Pt 1):136–153 Richardson DC, Richardson JS, Terwilliger
105. Kelley LA, Mezulis S, Yates CM, Wass MN, TC, Zwart PH (2010) PHENIX: a compre-
Sternberg MJ (2015) The Phyre2 web portal hensive python-based system for macromo-
for protein modeling, prediction and analysis. lecular structure solution. Acta Crystallogr D
Nat Protoc 10(6):845–858 Biol Crystallogr 66(Pt 2):213–221
106. Yang J, Yan R, Roy A, Xu D, Poisson J, Zhang 112. Zhu J, Cheng L, Fang Q, Zhou ZH, Honig B
Y (2015) The I-TASSER suite: protein struc- (2010) Building and refining protein models
ture and function prediction. Nat Methods within cryo-electron microscopy density maps
12(1):7–8 based on homology modeling and multiscale
107. Pettersen EF, Goddard TD, Huang CC,
structure refinement. J Mol Biol
Couch GS, Greenblatt DM, Meng EC, Ferrin 397(3):835–851
TE (2004) UCSF chimera—a visualization 113. Lindert S, Alexander N, Wotzel N, Karakas
system for exploratory research and analysis. M, Stewart PL, Meiler J (2012) EM-fold: de
J Comput Chem 25(13):1605–1612 novo atomic-detail protein structure determi-
108. Emsley P, Cowtan K (2004) Coot: model- nation from medium-resolution density maps.
building tools for molecular graphics. Acta Structure 20(3):464–478
Crystallogr D Biol Crystallogr 60(Pt 12 Pt 114.
Ramachandran GN, Ramakrishnan C,
1):2126–2132 Sasisekharan V (1963) Stereochemistry of
109. Topf M, Lasker K, Webb B, Wolfson H, Chiu polypeptide chain configurations. J Mol Biol
W, Sali A (2008) Protein structure fitting and 7:95–99
Chapter 29
Abstract
The assembly of filamentous appendages at the surface of bacteria is essential in many infection mecha-
nisms. The extent of mechanical, dynamical, and functional properties of such appendages is very diverse,
ranging from a structural scaffold of the pathogen–host cell interaction to cell motility, surface adhesion,
or the export of virulence effectors. In particular, the architectures of several bacterial secretion systems
have revealed the presence of filamentous architectures, known as pili, fimbriae, andneedles. At the macro-
scopic level, filamentous bacterial appendages appear as thin extracellular filaments of several nanometers
in diameter and up to several microns in length. The structural characterization of these appendages at
atomic-scale resolution represents an extremely challenging task because of their inherent noncrystallinity
and very poor solubility. Here, we describe protocols based on recent advances in solid-state NMR spectros-
copy to investigate the secondary structure, subunit–subunit protein interactions, symmetry parameters,
and atomic architecture of bacterial filaments.
Key words Solid-state nuclear magnetic resonance, Structure determination, Pilus, Needle, Protein
assembly, Protein complex, Helical symmetry
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_29, © Springer Science+Business Media LLC 2017
415
416 Birgit Habenstein and Antoine Loquet
Fig. 1 Overview of structural investigations of type III secretion needle and type 1
pilus by SSNMR-based approaches. (a) Schematic representation of type III injec-
tisome based on cryo-EM maps of Shigella flexneri needle (accession number
EMD-5352 [2]) and Salmonella typhimurium needle complex (accession number
EMD-1875 [70]). (b) SSNMR-based atomic model of S. typhimurium needle
filaments [1], side perspective and top view, PDB accession number 2LPZ. (c) Top
view of chaperone–usher pilus structure obtained by cryo-EM (accession number
EMD-3222 [6]). (d) SSNMR-based atomic resolution model (PDB accession
number 2N7H [5])
418 Birgit Habenstein and Antoine Loquet
Fig. 2 In vitro bacterial filament reconstitution for SSNMR analysis. (a) Expression of labeled protein subunits in
minimal medium containing labeled 13C, 15N sources. One colony of the transformed E. coli bacteria must be
selected from an agar plate to inoculate a preculture, which is further used for inoculating the main culture. (b) The
cells are destroyed and the protein subunit is harvested and purified on an adequate column system. (c) The pure
protein subunits are concentrated (or diluted) to a reasonable assembly concentration in the assembly buffer, which
should be meticulously tested and optimized at the optimal pH and salt concentration. The assembly conditions are
usually optimal under slow shaking. The subunits then adopt their native conformation by assembling (represented
here). (d) Filament formation should be visualized by transmission electron microscopy, as illustrated here for a
protein filament (subunit size of 14 kDa)
Solid-State NMR 419
2 Materials
2.1 In Vitro Protein SSNMR structural investigations require the use of uniformly
Expression, (or selectively) 13C/15N samples. In this protocol, the sample pro-
Purification, and Self- duction is based on the in vitro polymerization of protein subunits
Assembly into filaments. Here, the purpose is to give an overall description
of Isotopically of the requirements and their particularities throughout the
13
C-/15N-Labeled SSNMR sample preparation process using Escherichia coli as a
Protein Subunits standard expression system (see Notes 1–3).
1. Expression vector encoding the protein construct with, for
example, a (His)7-tag. Expression vector, typically pET21,
containing the DNA fragment encoding the protein sequence
of interest; a (His)7-tag is often used for protein purification.
2. E.coli expression strain. A commonly used protein expression
strain for NMR studies is E. coli BL21 (DE3).
3. Isopropyl-thio-β-d-galactopyranoside (IPTG), stock solution
(1 M).
4. A culture medium. For producing unlabeled protein standard
Lysogeny Broth LB is used. For isotope-labeled proteins and
expression tests, M9 minimal medium is used, following the
composition (see Table 1).
5. In case an isotopically labeled sample is produced, different
labeled 13C sources will be needed (see Subheading 3.1.2).
To obtain a uniformly 13C/15N ([U-13C/15N])-labeled sam-
ple, the minimal medium is supplemented with uniformly
13
C-labeled glucose (glc) and 15N-labeled NH4Cl to corre-
spondingly labeled 13C and 15N atoms. As part of the struc-
tural studies it is necessary to selectively label the proteins by
supplementing the M9 medium instead of uniformly
13
C-labeled glc with selectively 13C-labeled glc or glycerol (gly)
[27–30]. The selectively 13C-labeled precursors are [1-13C]-glc
and [2-13C]-glc or [1,3-13C]-glyc and [2-13C]-glyc. The two
distinct selective labeling schemes, two for glc and two for glyc
labeling, are complementary in their resulting amino acid
labeling patterns, which arise from the metabolic pathways
[27–29].
6. Incubator/shaker.
7. Purification elements: usually Äkta (GE Healthcare) or Bio-rad
chromatography system, purification chromatography column(s),
desalting column, centrifuges, centrifugal filter units.
8. pH meter.
9. Purification buffers.
420 Birgit Habenstein and Antoine Loquet
Table 1
M9 Minimal medium composition
Component M9 medium
NaCl 0.5 g/L
KH2PO4 3 g/L
Na2HPO4 6.7 g/L
MgSO4 1 mM
ZnCl2 10 μM
FeCl3 1 μM
CaCl2 100 μM
Table 2
Websites of computational programs and databases
3 Methods
3.1 Isotope-Labeled 1. Transform E.coli strain (e.g., BL21 DE3) with vector, includ-
In Vitro Protein ing protein-coding DNA, and plate it on LB agar plates con-
Expression, taining appropriate antibiotic.
Purification, 2. Inoculate a preculture of ≤10 mL LB medium with one iso-
and Self-Assembly lated culture picked from the agar plate (see Note 5). Incubate
3.1.1 Expression
at 37 °C while shaking (rotation values around 200 rpm) until
of Unlabeled Subunit
exponential growth phase is reached (0.6 < OD600 < 1).
Proteins 3. Inoculate main culture (LB) with preculture. To optimize pro-
tein expression, purification, and assembly conditions, as well as
to obtain a preliminary NMR signature (see Subheading 3.3), it
is appropriate to use unlabeled LB medium. With this low-cost
filament production, the conditions can be optimized and the
produced quantity estimated. Based on a 1D 13C spectrum
recorded at natural abundance (unlabeled material), a prelimi-
nary analysis of the structural complexity in the filament can be
obtained.
4. Induce protein expression with 0.75–1 mM IPTG at an opti-
cal density measured at a wavelength of 600 nm (OD600) of
0.8–1. Test the optimal expression time by sodium dodecyl
sulfate (SDS) gels, usually between 3 and 6 h at 37 °C, but
sometimes protein expression can be optimal at low tempera-
tures overnight. Harvest cells by centrifugation on a table cen-
trifuge (6000 rpm, 30 min, 4 °C) and store at −80 °C until
purification.
3.1.2 Expression 1. Transform E. coli strain (e.g., BL21 DE3) with vector, includ-
of Isotopically Labeled ing protein-coding DNA, and plate it on LB agar plates con-
Subunit Proteins taining appropriate antibiotic.
2. A preculture of ≤10 mL LB medium should be inoculated
with one isolated culture picked from the agar plate (see Note 5),
incubated at 37 °C while shaking (rotation values around
200 rpm) until the exponential growth phase is reached (0.6 <
OD600 < 1). Centrifuge down the preculture and remove
supernatant.
3. Inoculate main culture—M9 medium with unlabeled (step 4)
or labeled carbon and nitrogen sources (step 5)—with the
harvested cells (preculture of ≤10 mL LB per liter main
culture).
4. First perform a protein production in M9 medium supple-
mented with unlabeled glucose and NH4Cl to quantify the
production yield in M9 medium (see Note 6).
5. For the subsequent production of uniformly 13C/15N-labeled
([U-13C/15N]-labeled) protein in M9 medium (see Fig. 3a),
Solid-State NMR 423
3.1.3 Purification The purification protocol should be set up using unlabeled pro-
and Polymerization teins to avoid unnecessary expenses.
of Subunit Proteins
1. Thaw cells on ice and resuspend in appropriate lysis buffer.
2. Lyse E. coli cells by appropriate lysis method (e.g., sonication,
mechanical disruption or chemical lysis).
3. Set up a purification protocol. If the protein is produced in
inclusion bodies, the purification generally needs to be carried
out under denaturing conditions (e.g., 6–8 M urea or guani-
dine). In this case, a refolding step needs to be scheduled,
which might be concurrent with the desalting step. If the pro-
tein stays folded in the cytoplasm, the desalting step might not
be required and a simple concentration after purification might
be sufficient.
4. Concentrate the soluble protein fraction to a final concentra-
tion favorable to correct assembly. Typical values for protein
concentration range between 0.1 and 1 mM.
5. Keep protein solution under smooth shaking conditions for at
least 1 week. The solution becomes turbid upon filament for-
mation. After 1 week, filament formation should be checked
by EM. If filaments have formed, it is recommended to keep
assembled fibrils at 4 °C and add an antibacterial agent such as
sodium azide (0.02% (w/v)) to avoid sample contamination.
Fig. 3 Strategies for isotopic labeling of protein subunits. Homogeneous labeling schemes, based on (a) uni-
formly 13C-labeled subunits (white ), (b) selectively [1-13C]-glucose-labeled subunits (green), and (c) selectively
[2-13C]-glucose-labeled subunits (pink ). [1-13C]- and [2-13C]-glucose labeling schemes are homogeneous in
the sense that all protein subunits are labeled with the same scheme, although the isotopically labeled carbon
positions are selective. (d) Labeling patterns for the amino acid valine for each of the labeling schemes.
Isotopically labeled atomic positions are underlined and colored according to the cases a–c
3.2 Solid-State 1. Centrifuge aggregated filament sample and remove and keep the
NMR Setup supernatant at 4 °C until SSNMR experiments are successful.
Wash sample one to five times with ultrapure water if a high con-
3.2.1 Rotor Packing
centration of salts was used in the assembly buffer (see Note 8).
Centrifugation is needed between the washing steps, and
Solid-State NMR 425
3.2.2 Magic-Angle Several preparatory and optimization steps are required to carry out
and Pulse Calibration MAS SSNMR spectroscopy on biological samples. SSNMR experi-
ments are designed using a so-called NMR pulse sequence, which
contains a single or a series of radiofrequency pulses and one or
more acquisition times to record the NMR signal. The procedures
and parameters described in what follows are typical in the opera-
tion of a Bruker spectrometer in MAS configuration. A probehead
for 3.2 mm MAS rotors of an 800 MHz spectrometer is shown
in Fig. 4b.
1. Adjust angle of rotor relative to magnetic field (called the magic
angle) at a value of ~54.7°. A sample of KBr in powder form is
used to record a 79Br detected 1D spectrum under a spinning
frequency of 5 kHz; a single scan is enough to observe the sig-
nal on most spectrometers. The spinning frequency gives rise to
so-called spinning sidebands at the distance of the spinning
frequency (in this case 5 kHz) from the 79Br signal. Adjust the
angle by optimizing the linewidth of the spinning sidebands,
i.e., the sideband signal to the maximum intensity. The benefi-
cial effect of MAS is illustrated in Fig. 4c.
2. Calibrate the hard NMR pulses on standard reference samples,
usually Adamantane for calibrating the 1H pulse and a 13C/15N-
labeled amino acid (e.g., 13C/15N-labeled histidine hydrochlo-
ride monohydrate in polycrystalline powder form) for 13C and
15
N pulses. 90° pulses are optimized by adjusting the pulse
length or the pulse power. For example, to obtain the 90°
pulse values, the 180° pulse can be found by optimizing the
signal decrease until it disappears.
426 Birgit Habenstein and Antoine Loquet
Fig. 4 Solid-state experimental setup. (a) SSNMR MAS rotors with 7, 4, 3.2, and
2.5 mm diameter. (b) Bruker triple resonance 1H/13C/15N MAS 3.2 mm probehead.
(c) Carbon-detected 1H-13C CP spectrum of a protein filament recorded under MAS
condition with 1H decoupling during acquisition (SPINAL64) [54] (black ), under
MAS condition without 1H decoupling (red ), and without MAS (blue )
3.2.3 Sample Once the NMR setup on reference samples has been done, the
Temperature and Chemical sample of interest is introduced into the spectrometer to adjust the
Shift Calibration sample temperature and calibrate the chemical shift.
1. To obtain a precise estimation of the sample temperature dur-
ing an SSNMR experiment, it is recommended to use an inter-
nal calibration to avoid inaccurate measurements of the probe
thermocouple. On hydrated samples such as the filament sam-
ples described in this chapter, the sample temperature can be
calibrated using the difference between the DSS 1H resonance
(observed at 0 ppm) and the supernatant water resonance
Solid-State NMR 427
3.3.1 CP-Based
1.
Introduce the unlabeled filament sample into the
One-Dimensional spectrometer.
Experiment 2. Set the spinning frequency to 11 kHz and simultaneously cool
down the probehead to maintain a sample temperature in the
probehead of around 5–10 °C (see Subheading 3.2.3).
3. Setup a 1D 1H-13C CP experiment following the hard pulse
optimization on a reference sample (see Subheading 3.2.2). For
most natural abundance filament samples, the 13C signal is vis-
ible only after several hours of acquisition, making the optimi-
zation of pulse lengths, decoupling, and CP transfers extremely
time consuming and practically almost impossible. For this
experiment, the values should therefore be taken from the
values obtained in the reference sample optimization. Set the
number of scans to 20k (20,480), the CP contact time to 1 ms,
and the decoupling strength to 90 kHz. Different causes might
be at play if the spectrum shows no signal (see Note 9).
3.3.2 Interpretation The carbon-detected 1D 1H-13C CP spectrum is very informative
of a CP-Based One- since the linewidths are directly correlated to the degree of molec-
Dimensional Experiment: ular order and the resonance peak distribution indicates the sec-
Structural Elements ondary structure content. Figure 5 illustrates the information
and Homogeneity content of a 1D 1H-13C CP versus a 1D INEPT-based experiment
recorded on a bacterial filament.
428 Birgit Habenstein and Antoine Loquet
Fig. 5 SSNMR characterization of rigid and mobile protein segments in a protein filamentous assembly.
(a) Schematic representation of a bacterial filament, including rigid and mobile protein segments. (b) Cross
polarization (CP)-based experiments reveal residues contributing to the rigid core of the assembly. INEPT-
based experiments probe the presence of mobile segments, illustrated on a protein filament with a subunit
size of 14 kDa. (c) The chemical shift value of Cα, Cβ, and carbonyl C′ atoms is sensitive to the secondary
structure, and the chemical shift distribution is indicative of the secondary structure composition; illustrated
on a 1D CP spectrum of two protein filaments (both subunit sizes of 9 kDa) containing β-strand (blue) and
α-helical (red ) conformation
Solid-State NMR 429
3.3.3 INEPT-Based Although a CP-based experiment can reveal rigid residues, SSNMR
One-Dimensional also has the ability to probe residues with higher mobility. INEPT-
Experiment: Mobility based experiments are used to perform a through-bond transfer
and probe protein segments with increased molecular mobility
(from picoseconds to nanoseconds). Such INEPT-based experi-
ments are useful for detecting the presence of mobile protein seg-
ments in comparison with the rigid core probed by CP-based
experiments; both approaches are complementary in the delinea-
tion of different protein segments or domains with different mobil-
ity (see Fig. 5). The following procedure is used to carry out the
INEPT-based approach.
1. Optimize a 1H-13C INEPT transfer using GARP [56] proton
decoupling with a radio frequency of 5 kHz. Set up a 1D spec-
trum with 4000 scans and a recycle delay of 1 s.
2. Analysis: The interpretation of a 1D 13C-detected INEPT
spectrum (e.g., Fig. 5b) depends mostly on the amount of vis-
ible signals. If a low amount of signals is present, the signal
assignment to the amino acid carbons is possible (residue-type
assignment). The shifts in an INEPT experiment are usually
very close to chemical shifts typical for random coil conforma-
tion and can be assigned using the published 13C random coil
chemical shifts (e.g., [36]). Always carefully check the reso-
nances of your buffer components that might remain after
washing. If signals do not correspond to protein signals and
cannot be identified, it is recommended to record a solution
NMR spectrum of the buffer only to clearly assign buffer
signals.
3. Analysis: Based on the 1H/13C/15N chemical shift database
(e.g., the useful paper by Jardetzky et al. [36]), residue spin
systems can mostly be assigned unambiguously. As described
earlier, the residues observed in INEPT-based experiments
often exhibit random coil chemical shifts. Therefore, a com-
parison of the chemical shifts assigned in the INEPT spectrum
to the random coil standard values allows for estimating the
degree of mobility of the residue present in this dynamic
regime. The amino acids observed in the INEPT spectrum
compared to the primary sequence of a protein might make it
possible to identify the mobile protein segment, sometimes
with high confidence if a single copy of one residue type is
present in the primary sequence.
4. For all other described analyses of the mobile elements a
[U-13C]- or [U-13C/15N]-labeled sample is necessary.
5. If multiple residues of the same residue type are present
showing slightly different chemical shifts or difficulties in
disambiguating signals, 2D 1H-13C INEPT and 1H-(13C)-13C
or (1H-)13C-13C INEPT-TOBSY [57, 58] can be recorded.
Solid-State NMR 431
Fig. 6 Fingerprint SSNMR experiments for structural investigation of filamentous appendages. (a) 2D 13C-13C
PDSD spectrum recorded with a short mixing time to observe intraresidue correlations. (b) 2D 15N-13Cα spec-
trum to probe intraresidue backbone correlations. These experiments are illustrated on the S. typhimuriumtype
III secretion needle [1]
432 Birgit Habenstein and Antoine Loquet
Fig. 7 Comparison of theoretical 2D NCA spectra for each amino acid, for three different labeling schemes:
[U-13C]-glc (a), [1-13C]-glc (b), and [2-13C]-glc (c). (d) Spectral comparison of [U-13C] and [2-13C]-glc leads to
the identification of leucine correlations. (e) Spectral comparison of [U-13C] and [1-13C]-glc leads to the identi-
fication of several amino acid correlations
Fig. 8 Strategy for SSNMR resonance assignment. Intraresidue (a–d) and interresidue (e–h) correlations are
established through the connectivity between the carbon–carbon and the nitrogen–carbon atoms. (a) C-C PDSD
experiment for all intraresidue 13C-13C correlations (short mixing time). (b) N-C specific-CP for intraresidual 15N-13Cα
correlations. (c) N-(Cα)-Cβ (15N-13Cα specific-CP, DREAM) for intraresidual 15N-13Cα-13Cβ correlations. (d) N-(Cα)-C′
(15N-13Cα specific-CP, MIRROR) for intraresidual 15N-13Cα-13C’ correlations. (e) C-C PDSD experiment for predomi-
nantly intraresidue and sequential 13C-13C correlations (intermediate mixing time). (f) N-C′ (15N-13C′ specific-CP)
for sequential 15N-13C′ correlations. (g) Cα-(N)-C′ (13Cα-15 N specific-CP, 15N-13C’ specific-CP) for sequential
13
Cα-13C′ correlations. (h) N-(C′)-Cα (15N-13C’ specific-CP, MIRROR) for sequential 15N-13C’-13Cα correlations
Solid-State NMR 433
3.4.3 Sequential The assignment procedure described in what follows requires uni-
Assignment formly or uniformly and selectively 13C/15N-labeled samples for
steps 1 and 2 and a [U-13C/15N]-labeled sample for steps 3 and 4.
1. Set up a 2D 13C-13C PDSD experiment using a mixing time of
200 ms. The intermediate mixing time allows for the interresidue
Solid-State NMR 435
3.4.4 Determination 1. The assigned 13Cα, 13Cβ resonances make it possible to iden-
of Secondary Structure tify the secondary chemical shift [64] ΔδCα-ΔδCβ, indicative
and Topology of Protein of the secondary structure.
Subunit 2. Calculate 13Cα(assigned)-13Cα(random coil) and 13Cβ(assigned)-
13
Cβ(random coil). Chemical shift values of amino acids in
random coil conformation can be obtained from [36].
3. Plot ΔδCα-ΔδCβ, negative values for >3 residues in a row are
indicative of β-strands and positive for α-helical conformation.
It appears that amino acids such as glycine and proline show
unusual chemical shift values, especially when they occur as
secondary structure breakers. Caution must be taken when
defining the limits of secondary structure elements from the
secondary chemical shifts.
4. Use TALOS+ [39] or PREDITOR [38] to predict the protein
dihedral angles from the assigned chemical shifts. The phi/psi
dihedral angle restraints will be used later as structural input
during the modeling process.
3.5 SSNMR Several types of structural restraints are used in the structural mod-
Structural Restraints eling of macromolecular protein filaments. The following list
(Subheadings 3.5.1–3.5.3) follows chronological order in a basic
SSNMR structure determination process of a macromolecular
assembly.
3.5.1 Dihedral Angle The dihedral angles can be derived from the SSNMR chemical shifts
Restraints based on the TALOS+ routine (step 2 in Subheading 3.4.4) and are
introduced as structural restraints in the protocol of the selected
modeling software to perform the structure determination.
3.5.2 Collection Following the nomenclature used in solution NMR, SSNMR dis-
of Distance Restraints tance restraints are classified in different categories depending
on the number of residues separating the residues involved in the
Solid-State NMR 437
Fig. 9 Strategies for heterogeneous isotopically labeled filament samples. Heterogeneous labeling schemes
based on (a) an equimolar mixture of uniformly 13C and uniformly 15N labeled subunits (blue and white, respec-
tively), (b) an equimolar mixture of [1-13C]-glucose- and [2-13C]-glucose-labeled subunits (green and magenta,
respectively), and (c) a diluted mixture (1/4) of 13C-labeled subunits in an unlabeled subunit background (white
and yellow, respectively)
Solid-State NMR 439
3.6 SSNMR Structure Modeling the 3D structure aims at the generation of a PDB file
Calculation containing all atom coordinates for each protein subunit within the
filament assembly (Fig. 10). However, depending on the quality of
the SSNMR spectra, the amount of SSNMR structural restraints or
the access to complementary structural data from other techniques,
the modeling process can lead to 3D models with different degrees
of precision, ranging from pseudo-atomic resolution structures to
cartoonlike 3D models based on the molecular topology.
1. Prepare the SSNMR distance restraint lists (unambiguous and
ambiguous lists) encoding for intramolecular (i.e., intrasubunit)
distances and TALOS-based dihedral restraints.
Fig. 10 (a, b) T3SS needle filament and its basic building block, consisting of the smallest asymmetric unit contain-
ing all subunit–subunit interfaces. (c) Based on the detection of intermolecular SSNMR restraints, (d) distance
restraints at subunit–subunit interfaces can be derived. (e) SSNMR atomic structure of T3SS building block
440 Birgit Habenstein and Antoine Loquet
Fig. 11 Integrative structure determination process based on SSNMR. The different tasks are numbered and
asterisks indicate the data that can be integrated. Green arrows and underlining highlight intermediate results,
if the necessary data are available. Step 1 assignment process; 2 assignment process of long-range contacts
in SSNMR data; 3 secondary structure determination; 4 structure modeling with integration of different struc-
tural data obtained from biophysical techniques. # The monomer structure can be obtained from x-ray crystal-
lography or solution NMR
3.7.1 SSNMR Analysis This subheading corresponds to steps 1 and 3 illustrated in Fig. 11.
Guided by Solution NMR
1. If the solution NMR 13C chemical shifts of a monomer or a
Chemical Shifts
truncated version of the monomer are available, predict a 2D
13
C-13C spectrum and superimpose it to the experimental
SSNMR 13C-13C spectrum recorded on the assembly, using,
for example, the CCPNMR analysis prediction tool. If a
monomer crystal structure is available, predict the 13C chemi-
cal shifts (seeprediction programs listed in Subheading 2.3) to
further predict a 2D SSNMR 13C-13C.
2. Plot the predicted 2D 13C-13C on the experimental SSNMR
spectrum and compare the two spectra. If an experimental
peak and a predicted peak correspond in an unoverlapped
region of the spectrum, you can use with caution the pre-
dicted peak as assignment to start a sequential assignment
stretch. Be aware that this assignment is only predicted and
very liable to be incorrect. Nevertheless, it can facilitate/
accelerate the sequential assignment process (described in
Subheading 3.4.3).
3. If the solution NMR chemical shifts of a monomeric subunit
are available, plot the chemical shift differences between
solution NMR and SSNMR data obtained on the protein
sequence. Where differences larger than the error bar are
observed, structural differences between the monomeric state
and the monomer in the assembly might occur or an interaction
442 Birgit Habenstein and Antoine Loquet
4 Notes
Acknowledgments
The authors thank their past and present colleagues, in particular Prof.
Adam Lange at the Leibniz-Institut für Molekulare Pharmakologie
for his guidance during the author postdoctoral periods and his main
intellectual contribution for the T3SS needle and type I pilus projects.
This work was further supported by the Fondation pour la Recherche
Médicale (FRM-AJE20140630090 to A.L.), the ANR (13-PDOC-
0017-01 to B.H. and ANR-14-CE09-0020-01 to A.L.), the FP7
program (FP7-PEOPLE-2013-CIG to A.L.), the IdEx Bordeaux
University (Chaire d’Installation to B.H.)and the European
Research Council (ERC) under the European Union’s Horizon
2020 research and innovation program (ERC Starting Grant to
A.L., agreement 105945). Erick Dufourc is acknowledged for his
continuous support.
References
1. Loquet A, Sgourakis NG, Gupta R, Giller K, insights into its assembly and signaling mecha-
Riedel D, Goosmann C et al (2012) Atomic nisms. Proc Natl Acad Sci U S A 109:
model of the type III secretion system needle. 4461–4466
Nature 486:276–279 3. Blocker AJ, Deane JE, Veenendaal AK, Roversi P,
2. Fujii T, Cheung M, Blanco A, Kato T, Blocker Hodgkinson JL, Johnson S et al (2008) What’s
AJ, Namba K (2012) Structure of a type III the point of the type III secretion system needle?
secretion needle at 7-A resolution provides Proc Natl Acad Sci U S A 105:6507–6513
446 Birgit Habenstein and Antoine Loquet
4. Sauer FG, Futterer K, Pinkner JS, Dodson KW, 18. Tang M, Comellas G, Rienstra CM (2013)
Hultgren SJ, Waksman G (1999) Structural Advanced solid-state NMR approaches for
basis of chaperone function and pilus biogene- structure determination of membrane proteins
sis. Science 285:1058–1061 and amyloid fibrils. Acc Chem Res 46:
5. Habenstein B, Loquet A, Hwang S, Giller K, 2080–2088
Vasa SK, Becker S et al (2015) Hybrid struc- 19. Weingarth M, Baldus M (2013) Solid-state
ture of the type 1 pilus of Uropathogenic NMR-based approaches for supramolecular
Escherichia coli. Angew Chem Int Ed Engl structure elucidation. Acc Chem Res 46:
54:11691–11695 2037–2046
6. Hospenthal MK, Redzej A, Dodson K, Ukleja 20. Loquet A, Habenstein B, Lange A (2013)
M, Frenz B, Rodrigues C et al (2016) Structure Structural investigations of molecular machines
of a chaperone-usher pilus reveals the molecu- by solid-state NMR. Acc Chem Res 46:
lar basis of rod uncoiling. Cell 164:269–278 2070–2079
7. Geibel S, Waksman G (2014) The molecular 21. Yan S, Suiter CL, Hou G, Zhang H, Polenova T
dissection of the chaperone-usher pathway. (2013) Probing structure and dynamics of pro-
Biochim Biophys Acta 1843:1559–1567 tein assemblies by magic angle spinning NMR
8. Chandran Darbari V, Waksman G (2015) spectroscopy. Acc Chem Res 46:2047–2058
Structural biology of bacterial type IV secre-
22. Tycko R, Wickner RB (2013) Molecular
tion systems. Annu Rev Biochem 84:603–629 structures of amyloid and prion fibrils: con-
9. Melville S, Craig L (2013) Type IV pili in sensus versus controversy. Acc Chem Res
Gram-positive bacteria. Microbiol Mol Biol 46:1487–1496
Rev 77:323–341 23. Hong M, Zhang Y, Hu F (2012) Membrane
10. Stones DH, Krachler AM (2015) Fatal attrac- protein structure and dynamics from NMR
tion: how bacterial adhesins affect host signal- spectroscopy. Annu Rev Phys Chem 63:1–24
ing and what we can learn from them. Int 24. Egelman EH (2015) Three-dimensional
J Mol Sci 16:2626–2640 reconstruction of helical polymers. Arch
11. Cheung M, Shen DK, Makino F, Kato T, Biochem Biophys 581:54–58
Roehrich AD, Martinez-Argudo I et al (2015) 25. Egelman EH (2010) Reducing irreducible
Three-dimensional electron microscopy recon- complexity: divergence of quaternary structure
struction and cysteine-mediated crosslinking and function in macromolecular assemblies.
provide a model of the type III secretion sys- Curr Opin Cell Biol 22:68–74
tem needle tip complex. Mol Microbiol 26. Demers JP, Habenstein B, Loquet A, Kumar
95:31–50 Vasa S, Giller K, Becker S et al (2014) High-
12. Rathinavelan T, Lara-Tejero M, Lefebre M, resolution structure of the Shigella type-III
Chatterjee S, McShan AC, Guo DC et al secretion needle by solid-state NMR and cryo-
(2014) NMR model of PrgI-SipD interaction electron microscopy. Nat Commun 5:4976
and its implications in the needle-tip assembly 27. Hong M (1999) Determination of multiple
of the Salmonella type III secretion system. ***φ***-torsion angles in proteins by
J Mol Biol 426:2958–2969 selective and extensive (13)C labeling and two-
13. Jones CH, Pinkner JS, Roth R, Heuser J, dimensional solid-state NMR. J Magn Reson
Nicholes AV, Abraham SN et al (1995) FimH 139:389–401
adhesin of type 1 pili is assembled into a fibril- 28. Castellani F, van Rossum B, Diehl A, Schubert
lar tip structure in the Enterobacteriaceae. Proc M, Rehbein K, Oschkinat H (2002) Structure
Natl Acad Sci U S A 92:2081–2085 of a protein determined by solid-state
14. Campos M, Nilges M, Cisneros DA, Francetic O magic-angle-spinning NMR spectroscopy.
(2010) Detailed structural and assembly model Nature 420:98–102
of the type II secretion pilus from sparse data. 29. Lundstrom P, Teilum K, Carstensen T,
Proc Natl Acad Sci U S A 107:13081–13086 Bezsonova I, Wiesner S, Hansen DF et al (2007)
15. Habenstein B, Loquet A. (2015). Solid-state Fractional 13C enrichment of isolated carbons
NMR: an emerging technique in structural using [1-13C]- or [2-13C]-glucose facilitates
biology of self-assemblies. Biophys Chem. the accurate measurement of dynamics at back-
16. Meier BH, Bockmann A (2015) The structure bone Calpha and side-chain methyl positions in
of fibrils from ‘misfolded’ proteins. Curr Opin proteins. J Biomol NMR 38:199–212
Struct Biol 30:43–49 30. Loquet A, Giller K, Becker S, Lange A (2010)
17. Miao Y, Cross TA (2013) Solid state NMR and Supramolecular interactions probed by 13C-13C
protein-protein interactions in membranes. solid-state NMR spectroscopy. J Am Chem Soc
Curr Opin Struct Biol 23:919–928 132:15164–15166
Solid-State NMR 447
by adiabatic passage Hartmann-Hahn cross- 64. Luca S, Filippov DV, van Boom JH, Oschkinat
polarization. J Magn Reson Ser A 118: H, de Groot HJ, Baldus M (2001) Secondary
140–144 chemical shifts in immobilized peptides and
58. Andronesi OC, Becker S, Seidel K, Heise H, proteins: a qualitative basis for structure refine-
Young HS, Baldus M (2005) Determination of ment under magic angle spinning. J Biomol
membrane protein structure and dynamics by NMR 20:325–331
magic-angle-spinning solid-state NMR spec- 65. Lewandowski JR, De Paepe G, Griffin RG
troscopy. J Am Chem Soc 127:12965–12974 (2007) Proton assisted insensitive nuclei cross
59. Ader C, Frey S, Maas W, Schmidt HB, Gorlich polarization. J Am Chem Soc 129:728–729
D, Baldus M (2010) Amyloid-like interactions 66. Bardiaux B, Malliavin T, Nilges M (2012)
within nucleoporin FG hydrogels. Proc Natl ARIA for solution and solid-state
Acad Sci U S A 107:6281–6285 NMR. Methods Mol Biol 831:453–483
60. Szeverenyi NM, Sullivan MJ, Maciel GE 67. Guerry P, Herrmann T (2012) Comprehensive
(1982) Observation of spin exchange by two- automation for NMR structure determina-
dimensional Fourier-transform C-13 cross tion of proteins. Methods Mol Biol 831:
polarization-magic-angle spinning. J Magn 429–451
Reson 47:462–475 68. Morag O, Sgourakis NG, Baker D, Goldbourt
61. Verel R, Ernst M, Meier BH (2001) Adiabatic A (2015) The NMR-Rosetta capsid model of
dipolar recoupling in solid-state NMR: the M13 bacteriophage reveals a quadrupled
DREAM scheme. J Magn Reson 150:81–99 hydrophobic packing epitope. Proc Natl Acad
62. Westfeld T, Verel R, Ernst M, Bockmann A, Sci U S A 112:971–976
Meier BH (2012) Properties of the DREAM 69. Rieping W, Habeck M, Nilges M (2005)
scheme and its optimization for application to Inferential structure determination. Science
proteins. J Biomol NMR 53:103–112 309:303–306
63. Scholz I, Hodgkinson P, Meier BH, Ernst M 70. Schraidt O, Marlovits TC (2011) Three-
(2009) Understanding two-pulse phase- dimensional model of Salmonella’s needle
modulated decoupling in solid-state NMR. J complex at subnanometer resolution. Science
Chem Phys 130:114510 331:1192–1195
Chapter 30
Abstract
Protein transport across the cytoplasmic membrane is coupled to energy derived from adenosine triphos-
phate hydrolysis or the protein motive force (pmf). A sophisticated, multi-component type III secretion
system exports substrate proteins of both the bacterial flagellum and virulence-associated injectisome sys-
tem of many Gram-negative pathogens. The type-III secretion system is primarily a pmf-driven protein
exporter. Here, I describe methods to investigate the export of substrate proteins into the culture super-
natant under conditions that manipulate the pmf.
Key words Type III secretion system, Bacterial flagellum, Protein export, Proton motive force, ∆pH
gradient, ∆Ψ gradient, Ionophore, Carbonyl cyanide m-chlorophenylhydrazone (CCCP), Valinomycin
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_30, © Springer Science+Business Media LLC 2017
449
450 Marc Erhardt
(f-T3SS: FlhA FlhB FliO FliP FliQ FliR; v-T3SS: SctV SctU SctR
SctS SctT) form the export gate and are involved in the primary
substrate recognition, substrate unfolding, energy transduction
and protein transport across the cytoplasmic membrane. An associ-
ated ATPase complex (f-T3SS: FliH FliI FliJ; injectisome: SctL
SctN SctO) has a facilitating role in substrate recognition, sub-
strate unfolding and energy transduction, but is not strictly required
for protein export [9].
A scaffold structure formed by an integral membrane ring
(MS-ring; flagellum: FliF; injectisome: SctD SctJ) is essential for
the assembly of a functional core export apparatus [10]. In addi-
tion, accessory proteins form a cytoplasmic ring that facilitates sub-
strate recognition and binding of ATPase complex components
(flagellum: FliG FliM FliN; injectisome: SctQ) [11–13].
The contribution of the pmf and ATP hydrolysis to the protein
export process via T3SS has been examined using T3SS-dependent
substrate protein secretion of mutant strains and after treatment
with compounds that modulate the pmf. Here, I describe a meth-
odology to analyse the protein export of an f-T3SS-specific sub-
strate into culture supernatant in the presence or absence of
compounds interfering with the pmf.
2 Materials
3 Methods
3.2 Protein 1. Remove 0.5 mL of bacterial culture and record OD600 for nor-
Fractionation malization purposes (see step 4 in Subheading 3.2.1 and step
7 in Subheading 3.2.2).
2. Centrifuge 2 mL of bacterial culture at 10,000 × g for 5 min in
a tabletop centrifuge and transfer 1.8 mL of the supernatant to
a new centrifugation tube. Discard remaining supernatant and
store pellet on ice until further treatment (label ‘cellular
fraction’).
3. Centrifuge supernatant at 10,000 × g for 5 min in a tabletop
centrifuge and transfer 1.6 mL of the supernatant to a new cen-
trifugation tube (label ‘supernatant fraction’) (see Note 11).
4. Two alternative methods can be used for collecting proteins
from the culture supernatant: filtration on nitrocellulose filters
and TCA precipitation.
Energy Requirements of Type-III Protein Secretion 453
3.2.1 Protein Extraction 1. Filter supernatant from step 3 in Subheading 3.2 through a
by Filtration over prewetted nitrocellulose filter with a pore size of 0.45 μm for
Nitrocellulose Filter protein binding.
(See Note 12) 2. Elute proteins by addition of 40 μL 2× SDS sample buffer and
heat treatment for 30 min at 65 °C.
3. Resuspend bacterial pellet from step 2, Subheading 3.2, in 50
μL 2× SDS sample buffer and heat for 10 min at 95 °C.
4. Adjust cellular and supernatant fractions to 20 OD600 equiva-
lents per microlitre by addition of 2× SDS sample buffer and
keep on ice or store at −20 °C until further use.
3.3 Immunoblotting 1. Load 200 OD600 equivalents of cellular and supernatant frac-
tions onto 4–20% precast gels and perform protein separation
by SDS-PAGE in standard Tris-glycine buffer.
2. Following separation, electrotransfer proteins to a 0.2 μm pore
size Hybond-P PVDF transfer membrane (see Note 6) or a
0.45 μm pore size nitrocellulose membrane using a Trans-Blot
transfer apparatus.
3. Perform immunodetection using appropriate concentrations
of primary and secondary antibodies. In case of TH3730 or
TH10874, secreted and cellular FlgM protein is detected
using serum-purified anti-FlgM rabbit polyclonal antibodies
(dilution 1:10,000, [14]) and horseradish-peroxidase-conju-
gated anti-rabbit polyclonal antibodies (dilution 1:10,000,
BioRad).
454 Marc Erhardt
3.4 Assays to Inhibit 1. Grow bacterial cultures as described in Subheading 3.1, steps
Proton Motive Force 1–3.
3.4.1 Disruption 2. Pellet bacterial culture by 5 min centrifugation at 10,000 × g
of Proton Motive Force and resuspend in 3 mL LB medium containing 0–20 μM
Using Carbonyl Cyanide carbonyl cyanide m-chlorophenylhydrazone (see Notes 14
m-Chlorophenylhydrazone and 15).
(See Note 13) (See Fig. 1a) 3. Wash bacterial cells by centrifugation at 10,000 × g for 5 min.
Resuspend pellet in 3 mL LB medium containing 0–20 μM
carbonyl cyanide m-chlorophenylhydrazone and inducer as in
step 3, Subheading 3.1.
4. Resume incubation at 37 °C for 30 min in a water bath shaker
at 200 rpm and continue with step 6, Subheading 3.1.
Fig. 1 (a) Effect of inhibition of pmf on FlgM export. FlgM secretion was inhibited by
addition of 10 μM uncoupling agent carbonyl cyanide m-chlorophenyl hydrazone
(CCCP) and completely abolished by treatment with 20 μM CCCP in strain TH10874
(arabinose-inducible flgM ). Cytoplasmic FlgM levels remained constant. (b) Effect
of inhibition of ∆Ψ component of pmf on FlgM export. FlgM secretion was inhibited
by addition of valinomycin in the presence of K+. Cells were pretreated with 120
mM Tris–HCl to permeabilize the outer membrane to valinomycin where indicated.
(c) Effect of inhibition of ∆pH on FlgM export. Secretion of FlgM for cultures grown
in pH 5 was inhibited by addition of 34 mM potassium acetate. Adapted with per-
mission from Macmillan Publishers Ltd: Nature [6], copyright 2008
Energy Requirements of Type-III Protein Secretion 455
3.4.2 Disruption of ΔΨ 1. Grow bacterial cultures as described in Subheading 3.1, steps
Component of Proton 1–3.
Motive Force by K+/ 2. Pellet bacterial culture by 5 min centrifugation at 10,000 × g
Valinomycin (See Note 16) and resuspend in 3 mL LB medium containing 120 mM Tris–
(See Fig. 1b) HCl, pH 7.3. Incubate for 2 min (see Note 17).
3. Pellet bacterial culture by 5 min centrifugation at 10,000 × g
and resuspend in 3 mL LB medium containing 120 mM Tris–
HCl, pH 7.3, and 0–40 μM valinomycin in the presence or
absence of 150 mM KCl (see Note 15).
4. Pellet bacterial culture by 5 min centrifugation at 10,000 × g,
discard supernatant and resuspend in 3 mL LB medium con-
taining 120 mM Tris–HCl, pH 7.3, and 0–40 μM valinomycin
in the presence or absence of 150 mM KCl.
5. Resume incubation at 37 °C for 30 min in a water bath shaker
at 200 rpm and continue with step 6, Subheading 3.1.
3.4.3 Disruption of ΔpH 1. Grow bacterial cultures as described in Subheading 3.1, steps
Component of Proton 1–3.
Motive Force by Potassium 2. Pellet bacterial culture by 5 min centrifugation at 10,000 × g
Acetate (See Note 18) (See and resuspend in 3 mL LB medium.
Fig. 1c)
3. Wash bacterial cells by centrifugation at 10,000 × g for 5 min
and discard supernatant.
4. Resuspend pellet in 3 mL LB medium at pH 7 or pH 5 in the
presence or absence of 34 mM potassium acetate, respectively.
Add inducer of flagellar genes transcription as required for
strains TH3730 and TH10874 (see Note 15).
5. Resume incubation at 37 °C for 30 min in a water bath shaker
at 200 rpm and continue with step 6, Subheading 3.1.
4 Notes
Acknowledgments
References
1. Wickner W, Schekman R (2005) Protein trans- tein secretion in Salmonella enterica. PLoS
location across biological membranes. Science Genet 10:e1004800
310:1452–1456 10. Morimoto YV, Ito M, Hiraoka KD, Che YS,
2. Erhardt M, Namba K, Hughes KT (2010) Bai F, Kami-Ike N, Namba K, Minamino T
Bacterial nanomachines: the flagellum and type (2014) Assembly and stoichiometry of FliF and
III injectisome. Cold Spring Harb Perspect FlhA in Salmonella flagellar basal body. Mol
Biol 2:a000299 Microbiol 91:1214–1226
3. Minamino T (2014) Protein export through 11. McMurry JL, Murphy JW, Gonzalez-Pedrajo
the bacterial flagellar type III export pathway. B (2006) The FliN-FliH interaction medi-
Biochim Biophys Acta 1843:1642–1648 ates localization of flagellar export ATPase
4. Diepold A, Wagner S (2014) Assembly of the FliI to the C ring complex. Biochemistry
bacterial type III secretion machinery. FEMS 45:11790–11798
Microbiol Rev 38:802–822 12. Erhardt M, Hughes KT (2010) C-ring require-
5. Wilharm G, Lehmann V, Krauss K, Lehnert ment in flagellar type III secretion is bypassed
B, Richter S, Ruckdeschel K, Heesemann by FlhDC upregulation. Mol Microbiol
J, Trulzsch K (2004) Yersinia enterocolitica 75:376–393
type III secretion depends on the proton 13. Diepold A, Kudryashev M, Delalez NJ, Berry
motive force but not on the flagellar motor RM, Armitage JP (2015) Composition, forma-
components MotA and MotB. Infect Immun tion, and regulation of the cytosolic c-ring, a
72:4004–4009 dynamic component of the type III secretion
6. Paul K, Erhardt M, Hirano T, Blair DF, injectisome. PLoS Biol 13:e1002039
Hughes KT (2008) Energy source of flagellar 14. Hughes KT, Gillen KL, Semon MJ, Karlinsey
type III secretion. Nature 451:489–492 JE (1993) Sensing structural intermediates in
7. Minamino T, Namba K (2008) Distinct roles bacterial flagellar assembly by export of a nega-
of the FliI ATPase and proton motive force tive regulator. Science 262:1277–1280
in bacterial flagellar protein export. Nature 15. Minamino T, Morimoto YV, Hara N, Namba
451:485–488 K (2011) An energy transduction mechanism
8. Lee PC, Zmina SE, Stopford CM, Toska J, used in bacterial flagellar type III protein
Rietsch A (2014) Control of type III secretion export. Nat Commun 2:475
activity and substrate specificity by the cyto- 16. Minamino T, Imae Y, Oosawa F, Kobayashi Y,
plasmic regulator PcrG. Proc Natl Acad Sci U Oosawa K (2003) Effect of intracellular pH on
S A 111:E2027–E2036 rotational speed of bacterial flagellar motors.
9. Erhardt M, Mertens ME, Fabiani FD, Hughes J Bacteriol 185:1190–1194
KT (2014) ATPase-independent type-III pro-
Chapter 31
Abstract
Bacterial secretion systems allow the transport of proteins, called effectors, as well as external machine
components in the extracellular medium or directly into target cells. Comparison of the secretome, i.e. the
proteins released in the culture medium, of wild-type and mutant cells provides information on the secre-
tion profile. In addition, mass spectrometry analyses of the culture supernatant of bacteria grown in liquid
culture under secreting conditions allows the identification of secretion system substrates. Upon identifica-
tion of the substrates, the secretion profile serves as a tool to test the functionality of secretion systems.
Here we present a classical method used to concentrate the culture supernatant, based on trichloroacetic
acid precipitation.
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_31, © Springer Science+Business Media LLC 2017
459
460 Nicolas Flaugnatti and Laure Journet
2 Materials
3 Methods
4 Notes
1. For a safe and easy preparation, avoid weighting out the TCA
crystalline powder as it becomes easily syrupy upon contact
with humidity. The TCA solution must be kept in a dark glass
bottle. It is very corrosive and should be handled with care
with suitable protection. Do not use plastic containers.
2. DOC may be used as a carrier to assist protein precipitation.
If using DOC, add the DOC stock solution at the final con-
centration of 0.16 mg/mL to the cell-free fraction obtained in
step 6, vortex and leave on ice for 30 min; then proceed to
TCA precipitation as described in step 7. DOC should be
washed out with further acetone washing steps (repeat steps
10 and 11 three times). However, this could be a problem
with further mass spectrometry analysis.
3. Tabletop centrifuge at maximum speed may be sufficient;
however, we generally use a higher speed. TCA-resistant tubes,
such as Eppendorf tubes (check with your manufacturer for
tube compatibility), should be used.
4. In principle, any 0.22 μm filter may be used. However, we had
experience with a secreted protein that was retained on polyvi-
nylidene fluoride filters, so we moved to Polyether sulfone
(PES) filters. Be aware that the material of the filter may be of
importance.
5. A “non-secreting strain” should be used as a control, such as a
mutant in a core component, the ATPase energizing the assem-
bly of the secretion machinery or the substrate transport.
6. You must find conditions where secretion can be detected
in vitro. Because effectors can be secreted at low levels, high-
sensitivity mass spectrometry methods may be required [10].
If the secretion system is not produced in laboratory conditions,
native endogenous promoter(s) may be swapped for an induc-
ible promoter (e.g. Ptac, Plac, PBAD) to artificially induce the
expression of the secretion system [17].
TCA Precipitation 463
Acknowledgments
References
1. Costa TR, Felisberto-Rodrigues C, Meir A, 4. Coulthurst SJ, Lilley KS, Hedley PE, Liu H,
Prevost MS, Redzej A, Trokter M, Waksman G Toth IK, Salmond GP (2008) DsbA plays a
(2015) Secretion systems in Gram-negative critical and multifaceted role in the production
bacteria: structural and mechanistic insights. of secreted virulence factors by the phytopatho-
Nat Rev Microbiol 13:343–359 gen Erwinia carotovora subsp. atroseptica.
2. Cornelis GR, Biot T, Lambert de Rouvroit C, J Biol Chem 283:23739–23753
Michiels T, Mulder B, Sluiters C, Sory MP, 5. Kazemi-Pour N, Condemine G, Hugouvieux-
Van Bouchaute M, Vanooteghem JC (1989) Cotte-Pattat N (2004) The secretome of the
The Yersinia yop regulon. Mol Microbiol 3: plant pathogenic bacterium Erwinia chrysan-
1455–1459 themi. Proteomics 4:3177–3186
3. Beuzon CR, Banks G, Deiwick J, Hensel M, 6. Sikora AE, Zielke RA, Lawrence DA, Andrews
Holden DW (1999) pH-dependent secretion PC, Sandkvist M (2011) Proteomic analysis of
of SseB, a product of the SPI-2 type III secre- the Vibrio cholerae type II secretome reveals
tion system of Salmonella typhimurium. Mol new proteins, including three related serine
Microbiol 33:806–816 proteases. J Biol Chem 286:16555–16566
464 Nicolas Flaugnatti and Laure Journet
7. Burtnick MN, Brett PJ, DeShazer D (2014) 12. Veith PD, Chen YY, Gorasia DG, Chen D,
Proteomic analysis of the Burkholderia pseudo- Glew MD, O’Brien-Simpson NM, Cecil JD,
mallei type II secretome reveals hydrolytic Holden JA, Reynolds EC (2014)
enzymes, novel proteins, and the deubiquitin- Porphyromonas gingivalis outer membrane
ase TssM. Infect Immun 82:3214–3226 vesicles exclusively contain outer membrane
8. Hood RD, Singh P, Hsu F, Güvener T, Carl and periplasmic proteins and carry a cargo
MA, Trinidad RR, Silverman JM, Ohlson BB, enriched with virulence factors. J Proteome
Hicks KG, Plemel RL, Li M, Schwarz S, Wang Res 13:2420–2432
WY, Merz AJ, Goodlett DR, Mougous JD 13. Hwang BJ, Chu G (1996) Trichloroacetic acid
(2010) A type VI secretion system of precipitation by ultracentrifugation to concen-
Pseudomonas aeruginosa targets a toxin to trate dilute protein in viscous solution.
bacteria. Cell Host Microbe 7:25–37 BioTechniques 20:982–984
9. Russell AB, Singh P, Brittnacher M, Bui NK, 14. Ozols J (1990) Amino acid analysis. Methods
Hood RD, Carl MA, Agnello DM, Schwarz S, Enzymol 182:587–601
Goodlett DR, Vollmer W, Mougous JD (2012) 15. Wessel D, Flügge UI (1984) A method for the
A widespread bacterial type VI secretion effec- quantitative recovery of protein in dilute solu-
tor superfamily identified using a heuristic tion in the presence of detergents and lipids.
approach. Cell Host Microbe 11:538–549 Anal Biochem 138:141–143
10. Fritsch MJ, Trunk K, Diniz JA, Guo M, Trost 16. Caldwell RB, Lattemann CT (2004) Simple
M, Coulthurst SJ (2013) Proteomic identifica- and reliable method to precipitate proteins
tion of novel secreted antibacterial toxins of the from bacterial culture supernatant. Appl
Serratia marcescens type VI secretion system. Environ Microbiol 70:610–612
Mol Cell Proteomics 12:2735–2749
17. Gueguen E, Cascales E (2013) Promoter
11. Deng W, de Hoog CL, Yu HB, Li Y, Croxen swapping unveils the role of the Citrobacter
MA, Thomas NA, Puente JL, Foster LJ, Finlay rodentium CTS1 type VI secretion system in
BB (2010) A comprehensive proteomic analysis interbacterial competition. Appl Environ
of the type III secretome of Citrobacter roden- Microbiol 79:32–38
tium. J Biol Chem 285:6790–6800
Chapter 32
Abstract
The bacterial type VI secretion system (T6SS) is a secretory apparatus encoded by many Gram-negative bac-
teria. The T6SS facilitates the secretion and injection of toxic effector proteins into host cells, providing a
competitive advantage to bacteria encoding this machinery. The activity of the T6SS can be monitored by
probing for the conserved tubule component Hcp, which is secreted to the supernatants by the T6SS. Detection
of Hcp in culture supernatants is indicative of an active T6SS, but this secretion system is often tightly regu-
lated or inactive under laboratory conditions and different bacterial strains display differing Hcp secretion
phenotypes. Herein, we describe an enzyme-linked immunosorbent assay (ELISA) and colony blot methods
to facilitate large-scale screening of isolates for Hcp secretion and, thus, T6SS activity.
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_32, © Springer Science+Business Media LLC 2017
465
466 Brent S. Weber et al.
2 Materials
3 Methods
3.1 Hcp ELISA 1. The day prior to beginning the experiment, inoculate LB agar
plate(s) with the bacterial strain(s) of interest and incubate
overnight at 37 °C. This should be done in a manner that
ensures well-defined single colonies are present the next day.
2. Prepare 96-well plates for bacterial growth: Aseptically add
200 μL LB broth to the wells of a 96-well plate. Using sterile
toothpicks, inoculate individual colonies from plates grown
overnight into each well. If available, inoculate a positive and
negative control strain into the last two wells (e.g., strain that
secretes Hcp and an hcp mutant). Place 96-well plate in a
humidified chamber (see Note 2) at 37 °C with shaking (200
rpm) overnight (see Note 3).
468 Brent S. Weber et al.
3. The next day, retrieve 96-well plate and measure the OD600 in
a plate reader for a measure of bacterial cell growth. Then,
centrifuge the plate at 5000 × g for 10 min to pellet the bacte-
rial cells.
4. Meanwhile, transfer 25 μL blocking buffer to the wells of a
96-well high-binding ELISA plate.
5. Using a multichannel pipette, carefully transfer 75 μL of the
supernatants to the high-binding ELISA plate. Avoid pipetting
any cellular material (see Note 4). Be sure to keep the original
plate with the cell pellets as this will serve as the master stock
for any positive wells later on. The plate can be kept at 4 °C, or
alternatively add glycerol to the pellets and store at −80 °C for
the long term.
6. Incubate ELISA plate at room temperature for 1.5 h.
7. After incubation, remove solution from plate and wash three
times with PBS. Then completely fill wells with blocking buffer
(5% skim milk in PBS). Place on a rocking platform at room
temperature for 1 h.
8. Remove blocking solution, wash once with PBS.
9. Add 6 μL anti-Hcp antibody to 12 mL 2.5% skim milk in PBST
(1:2000 dilution) (see Note 5). Pipet 100 μL of this solution
into each well of the ELISA plate and place on room-
temperature rocking platform for 1 h.
10. Remove solution and wash thoroughly three to five times with
PBST.
11. Add 2.4 μL goat anti-rabbit HRP conjugate to 12 mL 2.5%
skim milk in PBST (1:5000 dilution) and pipet 100 μL of this
solution into each well of the ELISA plate. Incubate for 1 h at
room temperature on rocking platform.
12. Remove solution and wash thoroughly three to five times with
PBST.
13. Add 100 μL TMB substrate to wells. Gently shake the plate or
place on rocking platform. Watch plate for appearance of blue
color, which may be very rapid or take several minutes(see
Note 6). At desired time, measure the A650nm of the plate (see
Note 7). This can be used in conjunction with the A600nm taken
earlier to compare wells relative to growth. Take a photograph
of the plate if desired (see Fig. 1).
3.2 Colony Blot All liquids should be discarded as per biohazard protocols. All steps
for Hcp SECRETION are performed at room temperature unless otherwise stated.
1. Inoculate LB agar plate(s) with bacteria of interest in a way
that allows defined single colonies to grow. For large-scale
screening of colonies, LB agar plates are inoculated by spread
Screens for Protein Secretion 469
Fig. 1 (a) Example of using Hcp ELISA to screen multiple isolates of a given species at once. Here, 45 different
isolates of various Acinetobacter species were screened for Hcp secretion in duplicate. The results highlight
the variability among different species for T6SS activity within a given genus. (b) Screening a single strain, A.
baumannii ATCC 17978, for Hcp secretion. Single colonies of the strain were inoculated and used to perform
the Hcp ELISA. For this strain, a plasmid encodes the repressors for T6SS but can be lost upon culture without
selection [12]
Fig. 2 Use of colony blots to isolate active T6SS mutants from a T6SS-inactive strain. (a) Example of an Hcp
colony blot used to screen transposon mutants of A. baumannii strain 1225, which has an inactive T6SS and
does not secrete Hcp under normal laboratory conditions. Arrows indicate strong signals from single colonies
probed with the anti-Hcp antibody. (b) Hcp colony blot used on patch plated A. baumannii transposon mutants.
Colonies I and II show strong signals for anti-Hcp. (c) Western blot for Hcp expression in whole cells (WC) and
secretion in supernatant (SUP) of colonies I and II from figure (b). RNAP antibody used as lysis and loading
control. Wild-type A. baumannii 1225 expresses but does not secrete Hcp and is used as a negative control
4 Notes
References
Abstract
An accurate and complete roster of the Type III effector (T3E) proteins translocated by the P. syringae
Type III secretion system (T3SS) into host cells is critical to understanding the pathogen’s interactions
with plants. The adenylate cyclase (Cya) reporter offers a highly sensitive and robust assay for monitoring
the translocation of T3Es. T3Es are fused to the calmodulin-dependent adenylate-cyclase domain of
CyaA. The T3E targets Cya for translocation through the T3SS into the host cell at which point it is acti-
vated by calmodulin and converts adenosine triphosphate into cyclic adenosine monophosphate (cAMP).
The T3SS translocation-dependent increase in cAMP concentration in plant cells is then measured with an
enzyme-linked immunosorbent assay kit. The Cya reporter can be used to determine whether a candidate
protein is translocated by T3SS or to measure relative levels of T3SS translocation in a semiquantitative
manner.
Key words Pseudomonas syringae, Type III secretion system, Type III translocation, Translocation
reporter, Adenylate cyclase, Calmodulin, cAMP, ELISA
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_33, © Springer Science+Business Media LLC 2017
473
474 Suma Chakravarthy et al.
2 Materials
3 Methods
3.2 Plant Inoculation 1. Bring plants from the greenhouse or chamber 1 or 2 days prior
(See Notes 10 and 11) to plant inoculation and keep them on the laboratory bench.
We have used Nicotiana benthamiana, tobacco, and Arabidopsis
for inoculation of the Cya reporter strains. N. benthamiana
plants should be 4 to 6 weeks old, tobacco 5 to 8 weeks old,
and Arabidopsis 4 to 5 weeks old.
2. Start fresh plates for each DC3000 strain, pCPP5371::EOI,
pCPP5388 (positive control), and T3SS- pCPP5388 (negative
control) on KB agar with 10 μg/mL gentamicin and grow at
30 °C for 1–2 days.
3. From the primary plates use a wooden dowel to scoop up an
entire isolated colony, resuspend in 150–200 μL liquid KB,
and mix well by vortexing. Spot the entire volume onto a fresh
plate of KB agar with 10 μg/mL gentamicin. Swirl the plate
around to spread the bacteria on the entire surface of the plate.
Refresh all the strains similarly. Leave the plates open in the
laminar flow hood for about 5–10 min to dry and then incu-
bate at 30 °C overnight (up to 18 h).
480 Suma Chakravarthy et al.
3.3 Direct Direct cAMP ELISA kit is used to quantitatively measure cAMP in
cAMP Assay the leaf samples. See the manufacturer’s instructions for additional
details on conducting the assay and data analysis.
1. Bring all cAMP ELISA buffers to room temperature before use
(see Note 13).
2. Vortex the frozen tubes rapidly so the copper BBs grind the
tissue into a fine powder. Proceed to perform the assay. Ground
tissue can also be frozen at −80 °C until use (see Note 14).
3. Add 300 μL 0.1 M HCl to the frozen tissue. Vortex vigorously
to mix. Tissue will turn brown in a few minutes. This is
normal.
4. Centrifuge at ≥12,000 relative centrifugal force (RCF) for
10 min. Resuspend the pellet by vortexing and physical agita-
tion and recentrifuge at ≥12,000 RCF for 10 min to create a
tight pellet. Transfer the supernatant to a fresh microcentri-
fuge tube, taking care to avoid any leaf debris (see Note 15).
5. Extracted supernatants of each sample need to be diluted so
that cAMP concentrations are within the range of the cAMP
standard curve. This must be empirically determined for each
EOI vector/host plant/bacterial concentration combination.
We have used 10-fold to 300-fold dilutions in different experi-
ments. More concentrated samples may be required to measure
lower levels of translocation. The 50-fold dilution described in
what follows has been used successfully in Arabidopsis.
6. Using a multichannel pipette and a 96-well plate, set up the
following dilution series:
Mix 50 μL extract supernatant with 200 μL 0.1 M HCl (1:5).
Mix 30 μL 1:5 diluted sample with 270 μL 0.1 M HCl (1:10)
(1:50 final).
7. Prepare cAMP standards. Diluted standards should be used
within 60 min of preparation. Glass or polypropylene, but not
polystyrene, tubes may be used with the standards.
8. Label five 12 × 75 mm Pyrex culture tubes #S1– #S5.
9. Pipet 900 μL of the 0.1 M HCl into tube #S1.
10. Pipet 750 μL of the 0.1 M HCl into tubes #S2– #S5.
11. Add 100 μL of the 2000 pmol/mL standard stock into tube
#S1. Vortex vigorously.
12. Add 250 μL of tube #S1 to tube #S2 and vortex vigorously.
482 Suma Chakravarthy et al.
3.4 Bradford Assay All standards and samples should be run in duplicate. Sixteen wells
will be used for the standards. BSA concentration in the prediluted
standard kit: 125, 250, 500, 750, 1000, 1500, and 2000 μg/mL.
Use water for the 0 μg/mL “standard.”
These steps can be performed with a multichannel pipette.
1. In a 96-well plate add 99 μL water to each standard well and
80 μL water to each sample well.
2. Add 1 μL of the standards to the appropriate wells (1:100
dilution).
3. Add 20 μL of the 1:5 diluted samples to the appropriate wells
(1:25 final dilution).
4. Add 100 μL Bradford dye to each well, pipetting up and down
to mix.
5. Wells with higher protein concentrations should turn bluer.
6. Use a bubble breaker to remove any bubbles in the wells.
7. Incubate 10 min at room temperature.
8. Measure absorbance at 595 nm.
3.5 Data Analysis The principle behind the data analysis is as follows. (1) Generate
standard curves of pmol cAMP/mL and protein μg/mL concen-
tration. (2) Use the standard curves to calculate the pmol cAMP/
mL and protein μg/mL of your samples. (3) Divide the sample
cAMP value by the sample protein value to get pmol cAMP/μg
protein. This analysis can be conducted using Microsoft Excel.
Use the mean value of technical duplicate wells for all
calculations.
cAMP concentration calculation from 405 nm absorbance
values.
1. Subtract the blank well value from the NSB, B0, standard, and
sample values.
2. Subtract the blank-adjusted NSB value from the blank-adjusted
B0, standard, and sample values. These are the Net OD values.
3. Use the Net OD values to calculate the percentage of bound
cAMP for the standards. (S1/B0)*100, (S2/B0)*100….
4. Plot the percentage of bound (y) for the standards (linear scale)
vs. the pmol/mL values (x) of the standards (log scale). The
cAMP standard concentrations for S1–S5 are as follows:
484 Suma Chakravarthy et al.
4 Notes
Acknowledgments
The authors would like to thank Dr. Lisa Schechter, Dr. Hai Li
Wei, Dr. Sebastien Cunnac, Dr. Alan Collmer, and Dr. Sheng Yang
He for significant contributions to the development and refine-
ment of the procedure described in this chapter. This work was
supported by National Science Foundation grant IOS-1025642
and the Gordon and Betty Moore Foundation (GBMF3037).
References
1. Cunnac S, Chakravarthy S, Kvitko BH, Russell 9. den Dulk-Ras A, Vergunst AC, Hooykaas PJ
AB, Martin GB, Collmer A (2011) Genetic dis- (2014) Cre reporter assay for translocation
assembly and combinatorial reassembly identify (CRAfT): a tool for the study of protein translo-
a minimal functional repertoire of type III cation into host cells. Methods Mol Biol 1197:
effectors in Pseudomonas syringae. Proc Natl 103–121
Acad Sci U S A 108:2975–2980 10. Schechter LM, Valenta JC, Schneider DJ,
2. Fouts DE, Badel JL, Ramos AR, Rapp RA, Collmer A, Sakk E (2012) Functional and
Collmer A (2003) A pseudomonas syringae pv. computational analysis of amino acid patterns
tomato DC3000 Hrp (Type III secretion) dele- predictive type III secretion system substrates
tion mutant expressing the Hrp system of bean in Pseudomonas syringae. PLoS One 7:e36038
pathogen P. syringae pv. syringae 61 retains nor- 11. Crabill E, Joe A, Block A, van Rooyen JM,
mal host specificity for tomato. Mol Plant- Alfano JR (2010) Plant immunity directly or
Microbe Interact 16:43–52 indirectly restricts the injection of type III
3. Alfano JR, Collmer A (2004) Type III secre- effectors by the Pseudomonas syringae type III
tion system effector proteins: double agents in secretion system. Plant Physiol 154:233–244
bacterial disease and plant defense. Annu Rev 12. Wei HL, Chakravarthy S, Worley JN, Collmer
Phytopathol 42:385–414 A (2013) Consequences of flagellin export
4. Wei CF, Kvitko BH, Shimizu R, Crabill E, through the type III secretion system of
Alfano JR, Lin NC, Martin GB, Huang HC, Pseudomonas syringae reveal a major difference
Collmer A (2007) A Pseudomonas syringae pv. in the innate immune systems of mammals and
tomato DC3000 mutant lacking the type III the model plant Nicotiana benthamiana. Cell
effector HopQ1-1 is able to cause disease in Microbiol 15:601–618
the model plant Nicotiana benthamiana. Plant 13. Oh HS, Kvitko BH, Morello JE, Collmer A
J 51:32–46 (2007) Pseudomonas syringae lytic transglycosyl-
5. Schechter LM, Vencato M, Jordan KL, ases coregulated with the type III secretion system
Schneider SE, Schneider DJ, Collmer A (2006) contribute to the translocation of effector pro-
Multiple approaches to a complete inventory teins into plant cells. J Bacteriol 189:8277–8289
of Pseudomonas syringae pv. tomato DC3000 14. Badel JL, Shimizu R, Oh HS, Collmer A
type III secretion system effector proteins. Mol (2006) A Pseudomonas syringae pv. Tomato
Plant-Microbe Interact 19:1180–1192 avrE1/hopM1 mutant is severely reduced in
6. Schechter LM, Roberts KA, Jamir Y, Alfano growth and lesion formation in tomato. Mol
JR, Collmer A (2004) Pseudomonas syringae Plant-Microbe Interact 19:99–111
type III secretion system targeting signals and 15. Lam HN, Chakravarthy S, Wei HL, BuiNguyen
novel effectors studied with a Cya translocation H, Stodghill PV, Collmer A, Swingle BM,
reporter. J Bacteriol 186:543–555 Cartinhour SW (2014) Global analysis of the
7. Garcia JT, Ferracci F, Jackson MW, Joseph SS, HrpL regulon in the plant pathogen
Pattis I, Plano LR, Fischer W, Plano GV (2006) Pseudomonas syringae pv. tomato DC3000
Measurement of effector protein injection by reveals new regulon members with diverse
type III and type IV secretion systems by using functions. PLoS One 9:e106115
a 13-residue phosphorylatable glycogen syn- 16. Kvitko BH, Ramos AR, Morello JE, Oh HS,
thase kinase tag. Infect Immun 74:5645–5657 Collmer A (2007) Identification of harpins in
8. Sory MP, Cornelis GR (1994) Translocation of Pseudomonas syringae pv. tomato DC3000,
a hybrid YopE-adenylate cyclase from Yersinia which are functionally similar to HrpK1 in pro-
enterocolitica into HeLa cells. Mol Microbiol moting translocation of type III secretion sys-
14:583–594 tem effectors. J Bacteriol 189:8059–8072
Chapter 34
Abstract
Among the bacterial secretion systems, the Type III, IV, and VI secretion systems enable bacteria to secrete
proteins directly into a target cell. This specific form of secretion, referred to as translocation, is essential
for a number of pathogens to alter or kill targeted cells. The translocated proteins, called effector proteins,
can directly interfere with the normal processes of the targeted cells, preventing elimination of pathogens
and promoting their multiplication. The function of effector proteins varies greatly depending on the
considered pathogen and the targeted cell. In addition, there is often no magic bullet, and the number of
effector proteins can range from a handful to hundreds, with, for instance, a substrate of over 300 effector
proteins of the Icm/Dot Type IV secretion system in the human pathogen Legionella pneumophila.
Identifying, detecting, and monitoring the translocation of each of the effector proteins represents an
active field of research and is key to understanding the bacterial molecular weaponry. Translational fusion
of an effector with a reporter protein of known activity remains the best method to monitor effector trans-
location. The development of a fluorescent substrate for the TEM-1 beta-lactamase has turned this
antibiotic-resistant protein into a highly versatile reporter system for investigating protein transfer events
associated with microbial infection of host cells. Here we describe a simple protocol to assay the transloca-
tion of an effector protein by the Icm/Dot system of the human pathogen Legionella pneumophila.
Key words Effector protein, Type IV secretion system, β-lactamase fusion, CCF4, Fluorescence,
Legionella pneumophila
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_34, © Springer Science+Business Media LLC 2017
489
490 Julie Allombert et al.
Extracytosolic compartment
(media, vesicular compartement)
Bacterial cell
CCF2/AM
No fluorescence 'blaM-X TEM-X
520 nm 447 nm
FRET
409 nm 409 nm
Green Blue
fluorescence fluorescence
Intracellular compartment
2 Materials
2.2 Legionella 1. ACES-buffered Yeast Extract (AYE) medium: For 1 L dissolve 12
pneumophila Media g yeast extract and 10 g N-(2-Acentamido)-2-aminoethanesulfonic
and Bacterial Growth acid (ACES), adjust pH to 6.9 with 1 M KOH. Add 10 mL cys-
teine 40 g/L and 10 mL iron pyrophosphate 30 g/L. Fill volume
to 1 L with distilled water and filter sterilize.
2. Charcoal yeast extract (CYE) plates: For 1 L dissolve 10 g yeast
extract and 10 g ACES, adjust pH to 6.9 with 1 M KOH, add
15 g agar and 2 g activated charcoal, and autoclave. Add 10
mL filter sterilized cysteine 40 g/L and 10 mL filter-sterilized
ferric nitrate 25 g/L. When appropriate add 5 μg/mL
chloramphenicol
and 1 mM isopropyl β-d-1-
thiogalactopyranoside (IPTG) (see Note 2).
3. Disposable 13 mL polypropylene snap-cap tubes, sterile.
4. 1.5 mL microcentrifuge tubes, sterile.
5. 30 °C incubator.
6. Orbital shaker, 30 °C.
7. Spectrophotometer and cuvettes.
TEM-1 Beta-Lactamase Reporter System 493
2.3 Cell Culture 1. RPMI medium supplemented with 10% fetal bovine serum
and Differentiation (FBS) and glutamine (i.e., RPMI 1640 GlutaMAXTM, Gibco).
When appropriate add 5 μg/mL chloramphenicol and 1 mM
IPTG.
2. Phorbol 12-myristate 13-acetate (PMA): 0.1 M.
3. Culture flask, 25 cm2, sterile.
4. Disposable 15 mL polypropylene snap-cap tubes, sterile.
5. 96-well black polystyrene microplates with clear bottom, sterile.
6. CO2 incubator, 37 °C.
7. Malassez counting chamber.
494 Julie Allombert et al.
3 Methods
3.1 Growth The infecting strain should be grown under conditions that have
of Infecting Legionella been previously determined to result in a successful infection. These
pneumophila Strains conditions may vary depending on the strain and species used but
should include chloramphenicol to maintain the plasmid and IPTG
to induce expression of the tested effector fusion proteins.
1. Streak L. pneumophila strains carrying pXDC61-derived plas-
mids from a frozen stock to CYE plates supplemented with
chloramphenicol and then incubated at 30 °C for 5 days.
2. With a sterile loop, scrape off a few colonies and transfer to a
1.5 mL microcentrifuge tube containing 1 mL sterile ultrapure
water. Resuspend bacteria by repeated pipetting.
3. Measure the optical density at 600 nm (OD600) of a tenfold
diluted bacterial suspension.
4. In a sterile 13 mL tube, inoculate 2 mL AYE supplemented
with chloramphenicol and IPTG with appropriate volume of
previous bacterial suspension to reach a starting OD600 of 0.3.
Incubate at 30 °C in orbital shaker for 3 days (see Note 4).
5. Validate the beta-lactamase fusions production by western blot
(see Note 5).
TEM-1 Beta-Lactamase Reporter System 495
3.2 Maintenance U937 cells are monocytes that grow as a suspension and should be
and Differentiation maintained at a cell density of between 1.105 and 2.106 viable
of U937 Target Cells cells/mL.
1. Seed a 25 cm2 culture flask with U937 cells from a frozen stock
or from a previous culture flask in 10 mL RPMI medium sup-
plemented with glutamine and FBS. The culture flask is incu-
bated at 37 °C in a CO2 incubator for 5 days.
2. Determine the cellular concentration of the U937 cell culture
with a Malassez counting chamber.
3. Transfer 1.107 cells to a sterile 15 mL conical tube and centri-
fuge 5 min at 880 g.
4. Discard the supernatant and gently resuspend the pellet of cells
in 10 mL RPMI supplemented with glutamine and FBS (pre-
heated to 37°C) by slow repeated pipetting. This gives a cell
suspension of 1.106 cells/mL. Add 0.5 μL PMA.
5. Distribute 100 μL of cell suspension per well of a 96-well
microplate (105 cells/well). Leave three wells without U937
cells (medium alone). They will be used for the blank fluores-
cence measurement.
6. Incubate at 37 °C in CO2 incubator for 3 days. Following this
incubation, the previously spherical and nonadherent cells
should now be differentiated into macrophage-like cells that
adhere to the bottom of the well and display a spread-out
morphology.
3.4 Visualization Following fluorescence quantifications, the infected cells may also be
of Effector observed under a microscope to assess the percentage of translocation-
Translocation Using positive (blue) and translocation-negative cells (green).
Fluorescence 1. Follow the protocol of Subheading 3.3 until step 8.
Microscope
2. Place the plate on an inverted microscope equipped with a 40×
or 60× objective.
3. Observe cells with the beta-lactamase filter set (Ex: 405 ± 10;
dichroic mirror: 425; Em: 435 long-pass). Using this filter set,
both green and blue cells can be visualized simultaneously.
4. Alternatively, blue cells can be visualized with a (DAPI) filter
set (340–380 nm excitation and 425 nm long-pass emission).
Green cells can be visualized with the filter set commonly used
for the visualization of GFP. Overexposure of the cells with this
filter set may bleach the fluorescein moiety of CCF2
TEM-1 Beta-Lactamase Reporter System 497
Fig. 3 Typical results of a translocation assay. (a) Raw data, blank-subtracted fluorescence signals (relative
fluorescence unit, RFU), and fluorescence ratio (emission 460 nm/530 nm) obtained for secretion by wild-type
(WT) or ∆dotA L. pneumophila strain Lens of the known Dot/Icm effector LepA and the nonsecreted cytoplas-
mic protein FabI. Secretion assays were done in triplicate for each strain. Fluorescence measurements were
made with a Tecan M200 Infinite microplate reader equipped with a monochromator and a fluorescence top
reading module. The measurement program includes fluorescence readings with an excitation wavelength of
405 nm, emission wavelengths of 460 and 530 nm, and a gain set at 135. (b) Graphical representation of mean
emission ratio for LepA and FabI and corresponding standard deviations. Depending on the gain set for each
of the two fluorescence measurements, this ratio can change significantly. Data acquired under different gain
or in different plate reader can be normalized by setting the 460/530 ratio of the uninfected cells to 1. (c)
Fluorescence images of a typical translocation assay. Upon excitation at 405 nm, two images were captured
at 460 and 530 nm and merged
4 Notes
References
1. Costa TRD et al (2015) Secretion systems 8. Lodoen MB, Gerke C, Boothroyd JC (2010) A
in Gram-negative bacteria: structural and highly sensitive FRET-based approach reveals
mechanistic insights. Nat Rev Microbiol secretion of the actin-binding protein toxo-
13(6):343–359 filin during Toxoplasma gondii infection. Cell
2. Sory MP, Cornelis GR (1994) Translocation of Microbiol 12(1):55–66
a hybrid YopE-adenylate cyclase from Yersinia 9. Mills E, Baruch K, Charpentier X, Kobi S,
enterocolitica into HeLa cells. Mol Microbiol Rosenshine I (2008) Real-time analysis of
14(3):583–594 effector translocation by the type III secretion
3. Day JB, Ferracci F, Plano GV (2003) system of enteropathogenic Escherichia coli.
Translocation of YopE and YopN into eukary- Cell Host Microbe 3(2):104–113
otic cells by Yersinia pestis yopN, tyeA, sycN, 10. Charpentier X et al (2009) Chemical genetics
yscB and lcrG deletion mutants measured reveals bacterial and host cell functions critical
using a phosphorylatable peptide tag and for type IV effector translocation by Legionella
phosphospecific antibodies. Mol Microbiol pneumophila. PLoS Pathog 5(7):e1000501
47(3):807–823 11. Harmon DE, Davis AJ, Castillo C, Mecsas
4. Lee VT, Anderson DM, Schneewind O (1998) J (2010) Identification and characterization of
Targeting of Yersinia Yop proteins into the small-molecule inhibitors of Yop translocation
cytosol of HeLa cells: one-step translocation in Yersinia pseudotuberculosis. Antimicrob
of YopE across bacterial and eukaryotic mem- Agents Chemother 54(8):3241–3254
branes is dependent on SycE chaperone. Mol 12. Marketon MM, DePaolo RW, DeBord KL,
Microbiol 28(3):593–601 Jabri B, Schneewind O (2005) Plague bacteria
5. Charpentier X, Oswald E (2004) Identification target immune cells during infection. Science
of the secretion and translocation domain of 309(5741):1739–1741
the enteropathogenic and enterohemorrhagic 13. Geddes K, Cruz F, Heffron F (2007) Analysis
Escherichia coli effector Cif, using TEM-1 of cells targeted by Salmonella Type III secre-
beta-lactamase as a new fluorescence-based tion in vivo. PLoS Pathog 3(12):e196
reporter. J Bacteriol 186(16):5486–5495 14. de Felipe KS et al (2008) Legionella eukaryotic-
6. Zlokarnik G et al (1998) Quantitation of tran- like type IV substrates interfere with organelle
scription and clonal selection of single living trafficking. PLoS Pathog 4(8):e1000117
cells with beta-lactamase as reporter. Science 15. Steinberg TH, Newman AS, Swanson JA,
279(5347):84–88 Silverstein SC (1987) Macrophages possess pro-
7. Pechous RD, Goldman WE (2015) benecid-inhibitable organic anion transporters
Illuminating targets of bacterial secretion. that remove fluorescent dyes from the cytoplas-
PLoS Pathog 11(8):e1004981 mic matrix. J Cell Biol 105(6 Pt 1):2695–2702
Chapter 35
Abstract
The identification of effector proteins delivered into mammalian host cells by bacterial pathogens possessing
syringelike nanomachines is an important step toward understanding the mechanisms underlying the viru-
lence of these pathogens. In this chapter, we describe a method based on mammalian tissue culture infec-
tion models where incubation with a nonionic detergent (Triton X-100) enables solubilization of host cell
membranes but not of bacterial membranes. This allows the isolation of a Triton-soluble fraction lacking
bacteria but enriched in proteins present in the host cell cytoplasm and plasma membrane. Using appropriate
controls, this fraction can be probed by immunoblotting for the presence of bacterial effector proteins
delivered into host cells.
Key words Bacterial protein secretion system, Type III secretion, Effector, Translocation, Detergent
solubilization, SDS-PAGE, Immunoblotting
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_35, © Springer Science+Business Media LLC 2017
501
502 Irina S. Franco et al.
that does not affect the integrity of bacterial membranes. The nonionic
detergents Triton X-100 [4] and digitonin [5] have been widely
used for this purpose, based on the inability of Triton X-100 to
solubilize outer membranes of Gram-negative bacteria (although it
can solubilize inner membranes) [6–8], and on the specificity of
digitonin for cholesterol-rich membranes [9]. Subsequent high-
speed centrifugation allows the separation of detergent-soluble
(supernatant) and insoluble components (pellet) of the lysate,
where the supernatant comprises cytoplasmic and plasma mem-
brane components (including delivered effector proteins) and the
pellet retains unbroken bacteria and nuclei that remained intact.
Analysis of these fractions by immunoblotting makes it possible to
confirm the presence of an effector protein of interest in the super-
natant fraction, which is taken as evidence of effector translocation.
A critical control involves probing for a bacterial protein that is not
delivered into host cells. This ensures that during experimental
manipulation there was no contamination of the supernatant frac-
tion with cytosolic bacterial proteins.
Differential solubilization can be applied to monitor effector
translocation by different Gram-negative bacteria and types of host
cells. Furthermore, if effector-specific antibodies are available, dif-
ferential solubilization can be used to monitor the translocation of
endogenously expressed and nonmodified effector proteins. This is
in contrast to other effector translocation assays that require the
modification of the gene encoding the effector to produce a protein
with an epitope tag or fused to a reporter protein, usually expressed
from a plasmid and often from an exogenous promoter. The dif-
ferential solubilization procedure using Triton X-100 is illustrated
by the two protocols detailed in what follows, consisting in the
monitoring of type III secretion (T3SS)-mediated translocation (1)
of the Yersinia enterocolitica effector YopE, expressed from its own
promoter using a nonmodified wild-type strain to infect RAW
264.7 murine macrophage-like cells (see Fig. 1), and (2) of the
Salmonella enterica serovar Typhimurium (S. Typhimurium)
effector SteA with a C-terminal double hemagglutinin epitope tag
(SteA-2HA), expressed from its own promoter but encoded in an
exogenous low-copy plasmid, during infection of HeLa cells
(see Fig. 2). While YopE is translocated by extracellular Yersinia
into host cells [2], SteA can also be translocated into the host cell
cytoplasm from intracellular Salmonella residing within a mem-
brane-bound vacuole [10], which further illustrates the versatility
of the procedure.
Assessing Effector Translocation by Differential Solubilization 503
2 Materials (See Note 1)
2.1 Cell Culture, 1. Cell lines: HeLa (clone HtTA-1) and RAW 264.7 cells
Infection, (European Collection of Authenticated Cell Cultures,
and Preparation of Cell ECACC).
Extracts 2. Bacterial strains and plasmids: Y. enterocolitica E40 (pYV40)
(wild-type) and Y. enterocolitica E40 (pMSL41) (yscNΔ169–177;
deficient in YscN ATPase that is essential for the activity of the
Yersinia T3SS) [11], S. Typhimurium steA mutant (an isogenic
derivative of S. Typhimurium strain NCTC 12023 [identical to
ATCC 14208s]) [12], carrying low-copy pWSK129-derived
plasmids (six to eight copies per cell) [13] expressing C-terminal
2 × HA epitope-tagged wild-type SteA (SteAWT-2HA) or mutant
SteA with lysine residue 36 replaced by alanine (SteAK36A-2HA)
under the control of the steA promoter [10, 14].
3. Dulbecco’s Modified Eagle’s Medium (DMEM) supplemented
with 10% (v/v) heat-inactivated fetal bovine serum (FBS)
(DMEM + FBS), stored at 4 °C. Commercial 500 mL bottles
of heat-inactivated FBS are stored at −20 °C. The FBS is
thawed by incubation at 4 °C for 48 h, followed by prepara-
tion of aliquots in 50 mL tubes stored at −20 °C. To prepare
DMEM + FBS, the aliquots are thawed in a 37 °C water bath
and added to a commercial 500 mL bottle of DMEM. Do not
add antibiotics to cell culture medium.
4. Earle’s Buffered Salt Solution pH 7.4 (EBSS). Store at room
temperature.
5. TrypLE™ Express (Thermo Fisher Scientific). Store at room
temperature.
6. Phosphate-buffered saline (PBS 1×): 137 mM NaCl, 2.7 mM
KCl, 10 mM Na2HPO4, 1.8 mM KH2PO4, pH 7.4. Store at
room temperature. Prepared by diluting a stock of commercial
PBS 10× in double-distilled water (ddH2O), followed by ster-
ilization by autoclaving.
7. Nalidixic acid 3.5 mg/mL: Dissolve appropriate amount in
0.1 M NaOH and filter (0.22 μm) sterilize. Store at −20 °C.
Keep working aliquots at 4 °C.
8. Kanamycin 50 mg/mL: Dissolve appropriate amount in
ddH2O and filter (0.22 μm) sterilize. Store at −20 °C. Keep
working aliquots at 4 °C.
9. Lysogeny broth (LB) medium: Dissolve appropriate amount
of LB powder in ddH2O and sterilize by autoclaving. Store at
room temperature. Freshly supplemented with kanamycin
(to 50 μg/mL) to grow S. Typhimurium strains.
10. LB agar: Dissolve appropriate amount of LB powder in
ddH2O, add agar to 1.6% (w/v), and sterilize by autoclaving
Assessing Effector Translocation by Differential Solubilization 505
2.3 Immunoblotting 1. Transfer buffer: 0.025 M Tris, 192 mM glycine, and 20%
(v/v) methanol. Store at room temperature.
2. PBS 10×: 1.37 M NaCl, 0.027 M KCl, 0.1 M Na2HPO4,
0.02 M KH2PO4. Weigh adequate amounts of each of the
reagents, dissolve in ddH2O, adjust to final volume, and steril-
ize by autoclaving. Store at room temperature.
3. Washing solution (PBST): PBS 1× containing 0.2% (v/v)
Tween 20. Store at room temperature.
4. Blocking solution: PBST containing 4% (w/v) skim milk
powder: Dissolve the appropriate amount of skim milk pow-
der in PBST (see Note 6). Prepare fresh and store at 4 °C for
up to 2 days.
5. Stripping buffer: 25 mM glycine, pH 2, 1% (w/v) SDS:
Dissolve an adequate amount of glycine in ddH2O, adjust pH
to 2 with HCl, add 20% (w/v) SDS to a final concentration of
1% (w/v), and adjust to desired volume using ddH2O. Store
at room temperature.
6. Nitrocellulose membranes, 0.2 μm pore size (see Note 7).
7. Ponceau solution: 0.1% (w/v) Ponceau solution in 0.5% (v/v)
acetic acid: Dissolve Ponceau S in H2O and glacial acetic acid.
8. Whatman paper.
9. Autoradiography films.
10. Primary antibodies (all stored at −20 °C): Mouse monoclonal
anti-DnaK (clone 8E2/2; Millipore; used at 1:5000); rat
monoclonal anti-HA (clone 3F10; Roche; used at 1:1000),
mouse monoclonal anti-TEM-1 (QED Bioscience; used at
1:500); mouse monoclonal anti-α-tubulin (clone B-5-1-2;
Sigma-Aldrich; used at 1:1000); rabbit polyclonal anti-SycO
([15]; used at 1:500); rabbit polyclonal anti-YopE ([16];
used at 1:1000).
Assessing Effector Translocation by Differential Solubilization 507
3 Methods
3.1 Infection of RAW 1. RAW 264.7 cells are maintained in DMEM + FBS (with no
264.7 Cells by Y. antibiotics) at 37 °C in a humidified atmosphere with 5% (v/v)
enterocolitica CO2. The cells are used for up to 15–20 passages. The cells are
and Preparation routinely tested for mycoplasma contamination, using
of Triton-Soluble Venor®GeM Advance (Minerva Biolabs GmbH).
and Triton-Insoluble 2. The day before the infection prepare RAW 264.7 cells and
Fractions grow Y. enterocolitica strains: (1) seed RAW 264.7 cells at a
density of 1 × 106 cells per well in six-well tissue culture plates;
and (2) grow Y. enterocolitica in 5 mL BHI, overnight at 26 °C,
with continuous shaking (130 rpm).
3. Dilute the bacterial cultures grown overnight to an optical
density at 600 nm (OD600) of 0.2 in fresh BHI and resume
growth at 26 °C with continuous shaking (130 rpm) for 2 h
(see Note 8).
4. To induce expression of the Yersinia T3SS genes, quickly shift
the bacterial cultures to a shaking water bath (130 rpm) at
37 °C and incubate for an additional 30 min (see Note 9).
5. Centrifuge 1.5 mL of the bacterial culture (17,000 × g, 1 min;
see Note 10) and resuspend the bacterial pellet in 1 mL
DMEM + FBS and measure the OD600.
6. Calculate the volume of the bacterial suspension that needs to
be added to the RAW 264.7 cells to have a multiplicity of
infection (MOI) of 50, i.e., 5 × 107 bacteria per well
(see Note 11).
7. Add the calculated volume to the seeded RAW 264.7 cells.
Carefully swirl the plates to obtain an even infection.
8. Incubate the infected cells for 3 h at 37 °C in a humidified
atmosphere of 5% (v/v) CO2.
9. After 3 h of infection, replace the medium of the infected cells
by DMEM + FBS (previously warmed at 37 °C) containing
50 μg/mL gentamicin to kill extracellular bacteria (50 μg/mL)
and incubate for an additional 2 h at 37 °C in a humidified
atmosphere of 5% (v/v) CO2.
508 Irina S. Franco et al.
10. From this point, all manipulation should be done on ice and
using ice-cold solutions.
11. Wash infected cells twice with ice-cold 1× PBS.
12. Add 250 μL 1× PBS containing 0.1% (v/v) Triton X-100 and
a protease inhibitor cocktail (see Notes 12 and 13).
13. Incubate cells for 10 min on ice.
14. To remove cells from the wells, pipet up and down several
times (about 15–20 times) and transfer cells to a 1.5 mL tube.
15. Centrifuge samples at 17,000 × g for 15 min (see Note 10) at
4 °C. Remove the top 200 μL of supernatant and repeat this
centrifugation step (see Note 14). Recover the top 100 μL of
this second centrifugation step and add 25 μL 5× SDS-PAGE
loading buffer (this is the Triton-soluble fraction).
16. Remove all supernatant from the pellet of the first centrifuga-
tion and resuspend it in 200 μL 1× SDS-PAGE loading buffer
(this is the Triton-insoluble fraction).
17. Incubate samples for 10 min at 95–100 °C.
18. Use immediately 30 μL of the Triton-soluble fraction and
20 μL of the Triton-insoluble fraction for immunoblotting
(see subsequent discussion) or keep samples at −20 °C or
−80 °C until use.
3.2 Infection 1. HeLa cells are maintained in DMEM + FBS (with no antibiotics)
of HeLa Cells at 37 °C in a humidified atmosphere with 5% (v/v) CO2. The
by S. Typhimurium cells are used for up to 15–20 passages. The cells are routinely
and Preparation tested for mycoplasma contamination using Venor®GeM
of Triton-Soluble Advance (Minerva Biolabs GmbH).
and Triton-Insoluble 2. The day before the infection prepare HeLa cells and grow
Fractions S. Typhimurium strains: (1) seed HeLa cells at a density of
2.5 × 105 cells per well in six-well tissue culture plates; and (2)
grow S. Typhimurium in 5 mL LB overnight at 37 °C, with
continuous shaking (130 rpm) (see Note 15).
3. Dilute 1:33 the bacterial cultures grown overnight in 5 mL
fresh LB medium and grow the bacterial culture for 3 h 30 min
at 37 °C with continuous shaking (130 rpm) (see Note 16).
4. 5–10 min before the bacterial incubation has ended (step 3),
wash once the seeded HeLa cells with previously warmed
EBSS and incubate for 15–20 min at 37 °C in a humidified
atmosphere of 5% (v/v) CO2.
5. Measure OD600 of the bacterial culture.
6. Dilute the bacterial culture in 5 mL EBSS (previously warmed at
37 °C) to have a MOI of 100 (i.e., to 1.25 × 106 bacteria/mL)
when adding 2 mL of this suspension to the seeded HeLa cells
(see Note 17).
Assessing Effector Translocation by Differential Solubilization 509
4 Notes
Acknowledgments
References
1. Costa TR, Felisberto-Rodrigues C, Meir A, 5. Lee VT, Anderson DM, Schneewind O (1998)
Prevost MS, Redzej A, Trokter M, Waksman G Targeting of Yersinia Yop proteins into the
(2015) Secretion systems in Gram-negative cytosol of HeLa cells: one-step translocation of
bacteria: structural and mechanistic insights. YopE across bacterial and eukaryotic mem-
Nat Rev Microbiol 13:343–359 branes is dependent on SycE chaperone. Mol
2. Sory MP, Cornelis GR (1994) Translocation of Microbiol 28:593–601
a hybrid YopE-adenylate cyclase from Yersinia 6. Schnaitman CA (1971) Effect of ethylenedi-
enterocolitica into HeLa cells. Mol Microbiol aminetetraacetic acid, Triton X-100, and lyso-
14:583–594 zyme on the morphology and chemical
3. Charpentier X, Oswald E (2004) Identification composition of isolate cell walls of Escherichia
of the secretion and translocation domain of coli. J Bacteriol 108:553–563
the enteropathogenic and enterohemorrhagic 7. Schnaitman CA (1971) Solubilization of the
Escherichia coli effector Cif, using TEM-1 beta- cytoplasmic membrane of Escherichia coli by
lactamase as a new fluorescence-based reporter. Triton X-100. J Bacteriol 108:545–552
J Bacteriol 186:5486–5495 8. Birdsell DC, Cota-Robles EH (1968) Lysis of
4. Collazo CM, Galan JE (1997) The invasion- spheroplasts of Escherichia coli by a non-ionic
associated type III system of Salmonella detergent. Biochem Biophys Res Commun
typhimurium directs the translocation of Sip 31:438–446
proteins into the host cell. Mol Microbiol 9. Esparis-Ogando A, Zurzolo C, Rodriguez-
24:747–756 Boulan E (1994) Permeabilization of MDCK
Assessing Effector Translocation by Differential Solubilization 515
cells with cholesterol binding agents: dependence subcellular targeting within host cells. Cell
on substratum and confluency. Am J Phys Microbiol 18:949–969
267:C166–C176 15. Letzelter M, Sorg I, Mota LJ, Meyer S, Stalder
10. Domingues L, Holden DW, Mota LJ (2014) The J, Feldman M, Kuhn M, Callebaut I, Cornelis
Salmonella effector SteA contributes to the control GR (2006) The discovery of SycO highlights a
of membrane dynamics of Salmonella-containing new function for type III secretion effector
vacuoles. Infect Immun 82:2923–2934 chaperones. EMBO J 25:3223–3233
11. Sory MP, Boland A, Lambermont I, Cornelis 16. Diepold A, Amstutz M, Abel S, Sorg I, Jenal
GR (1995) Identification of the YopE and U, Cornelis GR (2010) Deciphering the
YopH domains required for secretion and assembly of the Yersinia type III secretion
internalization into the cytosol of macro- injectisome. EMBO J 29:1928–1940
phages, using the cyaA gene fusion approach. 17. VanRheenen SM, Luo ZQ, O’Connor T,
Proc Natl Acad Sci U S A 92:11998–12002 Isberg RR (2006) Members of a Legionella
12. Figueira R, Watson KG, Holden DW, Helaine pneumophila family of proteins with ExoU
S (2013) Identification of Salmonella pathoge- (phospholipase A) active sites are translocated
nicity island-2 type III secretion system effec- to target cells. Infect Immun 74:3597–3606
tors involved in intramacrophage replication of 18. Denecker G, Totemeyer S, Mota LJ,
S. enterica serovar Typhimurium: implications Troisfontaines P, Lambermont I, Youta C,
for rational vaccine design. MBio 4:e00065 Stainier I, Ackermann M, Cornelis GR (2002)
13. Wang RF, Kushner SR (1991) Construction of Effect of low- and high-virulence Yersinia
versatile low-copy-number vectors for cloning, enterocolitica strains on the inflammatory
sequencing and gene expression in Escherichia response of human umbilical vein endothelial
coli. Gene 100:195–199 cells. Infect Immun 70:3510–3520
14. Domingues L, Ismail A, Charro N, Rodriguez- 19. Drecktrah D, Knodler LA, Ireland R, Steele-
Escudero I, Holden DW, Molina M, Cid VJ, Mortimer O (2006) The mechanism of
Mota LJ (2016) The Salmonella effector SteA Salmonella entry determines the vacuolar envi-
binds phosphatidylinositol 4-phosphate for ronment and intracellular gene expression.
Traffic 7:39–51
Chapter 36
Abstract
Anti-bacterial activity assays are an important tool in the assessment of the ability of one bacterium to kill
or inhibit the growth of another, for example during the study of the Type VI secretion system (T6SS) and
the anti-bacterial toxins it secretes. The method we describe here can detect the ability of a bacterial strain
to kill or inhibit other bacterial cells in a contact-dependent manner when co-cultured on an agar surface.
It is particularly useful since it enumerates the recovery of viable target cells and thus enables quantification
of the anti-bacterial activity. We provide a detailed description of how to measure the T6SS-dependent
anti-bacterial activity of a bacterium such as Serratia marcescens against a competitor prokaryotic organ-
ism, Escherichia coli, and also describe possible variations in the method to allow adaptation to other
attacker and target organisms.
Key words Gram-negative bacteria, Protein secretion system, Type VI secretion system, Co-culture
assay, Anti-bacterial activity, Bacterial competitive fitness, Toxin/immunity pair
1 Introduction
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9_36, © Springer Science+Business Media LLC 2017
517
518 Juliana Alcoforado Diniz et al.
Fig. 1 Schematic overview of anti-bacterial activity assay. Following overnight growth on a solid agar surface,
the target strain (Ta) and different attacker strains (wild type, WT; T6SS inactive mutant, ΔT6; a mutant lacking
a toxin of interest, ΔTX; and a no-attacker, medium-only control, LB) are mixed in a ratio of 5 attackers to 1
target and spotted onto a solid agar surface. After a defined incubation period at the required temperature, the
co-culture spots (LB + target, green; wild type + target, blue; ΔT6SS + target, red; and Δtoxin + target, grey)
are scraped off, the cells resuspended and serial dilutions prepared. For an initial trial, these dilutions from
neat (N) to 10−6 (−6) are spotted on an agar plate supplemented with antibiotic selective for growth of the
target only. After incubation, estimation of target recovery from the trial plate is used to determine the dilution
which will provide a few tens of single colonies per plate in the actual experiment. For the actual experiment,
an appropriate volume of the correct dilution of the co-culture is spread on a selective plate with a glass
spreader and the colonies are counted following overnight incubation; replicate experiments provide fully
quantitative data. See text for full details
2 Materials
3 Methods
7. Spot 25 μL of each mixture onto an LB agar plate (all the spots
from one replicate, i.e. one co-culture spot for each attacker,
go on the same plate). When the co-culture is being performed
at 37 °C (see Note 3), pre-warm this plate.
8. Wait 5 min to allow the spots to dry and immediately put plates
in the incubator at 37 °C for 4 h (see Note 4).
9. Following the incubation period, scrape off each spot using a
sterile disposable loop and resuspend all the cells on the loop
in 1 mL sterile LB; mix thoroughly on a vortex for approxi-
mately 30 s or until the pellet is completely resuspended.
10. Prepare serial tenfold dilutions of each resuspension from neat
to 10−6 using sterile LB (e.g. 90 μL LB plus 10 μL preceding
dilution). Make sure to change pipette tips and to vortex 5 s
between each dilution step. Then put diluted samples onto anti-
biotic-containing selective medium to enumerate the surviving
target cells, in one of two formats described in steps 11 and 12.
11. Trial: When testing a particular attacker/target combination
for the first time, it is advisable to perform a trial experiment to
determine the correct dilution to spread on a selective plate to
yield a few tens of single colonies. For this, perform a full serial
tenfold dilution of the resuspended cells, from neat to 10−6,
and spot 5 μL of each dilution onto an LB + streptomycin
plate, as shown in Fig. 1. Incubate at 37 °C (or the target
organism’s preferred growth temperature) overnight or until
single colonies have grown.
12. Proper experiment: Prepare a suitable dilution of the resus-
pended cells for each attacker/target pair (based on the trial)
and spread 50 μL or 100 μL onto LB + streptomycin plates.
We recommend using a bent glass rod, sterilised by dipping in
ethanol and removal using a flame, to spread the cell suspen-
sion evenly over the agar surface. Incubate at 37 °C (or the
target organism’s preferred growth temperature) overnight or
until single colonies have grown. It is important to obtain
well-spaced single colonies; if this does not happen, adjust the
volume or dilution spread on the plate.
13. Count the colonies on the enumeration plates, using a tally or
pen-style colony counter if preferred. Calculate the number of
viable target cells recovered, expressed as cfu per co-culture
spot. See also Notes 5 and 6.
Example (Fig. 2):
Target with LB only: 30 colonies in 50 μL of 10−6 dilu-
tion → 30 × (1000/50) × 106 = 6 × 108 cells/spot.
Target with wild-type S. marcescens: 11 colonies in 100 μL of
10−2 dilution → 11 × (1000/100) × 102 = 1.1 × 104 cells/spot.
Target with T6SS mutant: 45 colonies in 100 μL of 10−6 dilu-
tion → 45 × (1000/100) × 106 = 4.5 × 108 cells/spot.
522 Juliana Alcoforado Diniz et al.
4 Notes
Acknowledgements
References
1. Alcoforado Diniz J, Liu YC, Coulthurst SJ type VI secretion system spike. Nature 500:
(2015) Molecular weaponry: diverse effectors 350–353
delivered by the Type VI secretion system. Cell 6. Russell AB, Hood RD, Bui NK et al (2011)
Microbiol 17:1742–1751 Type VI secretion delivers bacteriolytic effec-
2. Cianfanelli FR, Monlezun L, Coulthurst SJ tors to target cells. Nature 475:343–347
(2016) Aim, load, fire: the Type VI secretion 7. Schwarz S, West TE, Boyer F et al (2010)
system, a bacterial nanoweapon. Trends Micro Burkholderia Type VI secretion systems have
biol 24:51–62 distinct roles in eukaryotic and bacterial cell
3. Durand E, Cambillau C, Cascales E et al interactions. PLoS Pathog 6:e1001068
(2014) VgrG, Tae, Tle, and beyond: the versa- 8. Gueguen E, Cascales E (2013) Promoter
tile arsenal of Type VI secretion effectors. swapping unveils the role of the Citrobacter
Trends Microbiol 22:498–507 rodentium CTS1 type VI secretion system in
4. Russell AB, Peterson SB, Mougous JD (2014) interbacterial competition. Appl Environ Micro
Type VI secretion system effectors: poisons with biol 79:32–38
a purpose. Nat Rev Microbiol 12:137–148 9. Hachani A, Lossi NS, Filloux A (2013) A visual
5. Shneider MM, Buth SA, Ho BT et al (2013) assay to monitor T6SS-mediated bacterial
PAAR-repeat proteins sharpen and diversify the competition. J Vis Exp 20:50103
524 Juliana Alcoforado Diniz et al.
10. Alcoforado Diniz J, Coulthurst SJ (2015) 14. Hood RD, Singh P, Hsu F et al (2010) A type
Intraspecies competition in Serratia marcescens VI secretion system of Pseudomonas aeruginosa
is mediated by Type VI-secreted Rhs effectors targets a toxin to bacteria. Cell Host Microbe
and a conserved effector-associated accessory 7:25–37
protein. J Bacteriol 197:2350–2360 15. Ma LS, Hachani A, Lin JS et al (2014)
11. English G, Trunk K, Rao VA et al (2012) New Agrobacterium tumefaciens deploys a super-
secreted toxins and immunity proteins encoded family of type VI secretion DNase effectors
within the Type VI secretion system gene clus- as weapons for interbacterial competition in
ter of Serratia marcescens. Mol Microbiol 86: planta. Cell Host Microbe 16:94–104
921–936 16. Macintyre DL, Miyata ST, Kitaoka M et al
12. Fritsch MJ, Trunk K, Diniz JA et al (2013) (2010) The Vibrio cholerae type VI secretion
Proteomic identification of novel secreted anti- system displays antimicrobial properties. Proc
bacterial toxins of the Serratia marcescens Type Natl Acad Sci U S A 107:19520–19524
VI secretion system. Mol Cell Proteomics 17. Casadaban MJ, Cohen SN (1979) Lactose
12:2735–2749 genes fused to exogenous promoters in
13. Murdoch SL, Trunk K, English G et al (2011) one step using a Mu-lac bacteriophage:
The opportunistic pathogen Serratia marces- in vivo probe for transcriptional control
cens utilizes Type VI secretion to target bacte- sequences. Proc Natl Acad Sci U S A 76:
rial competitors. J Bacteriol 193:6057–6069 4530–4533
Index
Laure Journet and Eric Cascales (eds.), Bacterial Protein Secretion Systems: Methods and Protocols, Methods in Molecular
Biology, vol. 1615, DOI 10.1007/978-1-4939-7033-9, © Springer Science+Business Media LLC 2017
525
Bacterial Protein Secretion Systems: Methods and Protocols
526 Index
Electron microscopy (EM) Immunoprecipitation (IP)������������������������115, 117, 120, 125,
cryo-electron microscopy������������������������������������ 381, 409 126, 162, 169, 211, 213, 233, 236, 238, 240–242, 324,
electron cryo-tomography�����������������������������������353–373 330, 334–335, 347
EM grid������������������������������������������������������������������� 354, 355, Infection����������������������������������� 179, 201, 305, 415, 492, 494,
357, 381 495, 498, 501–505, 507–510, 512
Enzyme-linked immunosorbent assay (ELISA)����������������90, Insertion����������������32, 66, 106, 107, 109–111, 126, 134–136,
466–469, 471, 474, 476, 481, 482 151, 171, 172, 200, 207, 235, 242, 305, 306, 358, 359
Escherichia coli (E. coli)���������������������������������59, 60, 62, 76, Interactome����������������������������������������������������������������������221
77, 79, 81, 82, 89, 102, 106, 110, 119, 125, 130, 131, Ion electrochemical gradient���������������������������� 277, 279, 282
133–137, 139, 140, 146, 152, 156, 160, 163, 165,
167, 169–171, 174, 179, 180, 190–192, 202, 203, L
205, 206, 222, 234–237, 244, 253, 260, 269, 280, Lambda Red based recombination�����������������������������������222
283, 296, 300, 301, 303–305, 334, 346, 378, 418, Legionella pneumophila�����������������������492, 494–495, 497, 498
419, 422, 423, 443, 467, 475, 478, 519, 520 Lipobox������������������������������������������������������ 66, 68–69, 71, 75
Lipoprotein labeling�����������������������������������������������������������77
F
Lipoproteins�����������������������������������38, 65–72, 75–78, 87, 89,
Fluorescence microscopy�����������������67–69, 72, 292, 293, 297 91, 235, 279, 302, 306
Fluorescent reporter����������������������������������������� 191, 192, 519 Lysozyme������������������������������� 60–63, 67–71, 82, 84, 99, 100,
Fractionation�������������������� 59–64, 83–85, 323–324, 329–333, 102, 153, 156, 214, 215, 217, 280, 281, 285, 300, 302,
346, 347, 363, 364, 372, 384, 387, 450–453, 490 304, 322, 326, 329, 344, 371
Fractionator���������������������������������������������������������� 84, 85, 332 Lytic transglycosylases (LTGs)����������������� 151, 152, 155, 156
Freeze and thaw����������������������������������������������������������������327
M
G
MacConkey/maltose medium�����������163, 166, 169, 173, 208
β-galactosidase����������������������������������130, 131, 133, 135, 137, Machine learning���������������������������������������29, 31–34, 40, 41,
139, 140, 162, 163, 167–170, 190, 200, 206–208, 44–46, 48, 50, 51, 110
281–283, 286 MacSyFinder�������������������������������������������4, 5, 7, 9–10, 18, 19
GAL4 DNA binding domain (DNA-BD)�������������� 178, 180 Maleimide����������������������������������������88, 91, 92, 94, 113–115,
Gal4 transcriptional activation domain����������������������������178 120–121, 124–126
GALLEX������������������������������������������������� 200–204, 206–208 Mass spectrometry����������������������������152, 177, 212, 221, 222,
Genetic assay������������������������������������������������������������ 159, 162 230, 322, 335, 340–344, 349, 350, 460–462, 466
Globomycin������������������������������������������������������������ 76, 77, 79 MCherry. See Fluorescent reporters
Membrane fraction�������������������������������������60, 61, 63, 81, 82,
H 125, 138, 323–324, 329–334
HeLa cells������������������������������������������������502, 503, 508–510, Membrane preparations��������������������� 82, 322–323, 327–329
512, 513 Membrane protein complexes��������������������������������������44, 51, 62,
Helix–helix interactions�������������������������������������������� 200, 202 81–86, 97–102, 105–126, 129–141, 199, 235, 301, 321,
Hidden Markov model (HMM)����������������������� 4–12, 16, 31, 322, 324, 326–329, 334–335
32, 36, 38, 39, 44–46, 111 Muramic acid assay�������������������������������������������������� 145, 147
Host cell����������������������������������� 116, 125, 226, 247, 248, 473,
N
474, 489, 490, 492, 498, 501–503, 512
Hybrid protein������������������������������������������131, 136, 159–164, Naïve Bayes classifier����������������������������������������������������32, 48
166–171, 226, 228 n-dodecyl- β -D-maltopyranoside (DDM)���������������������302,
307, 322, 327, 328, 334, 341, 345
I n-dodecyl-β-D-maltoside. See n-dodecyl- β
Identification -D-maltopyranoside (DDM)
of components���������������������������������������������� 2, 3, 5, 7, 17 Negative staining (NS)������������������������������������� 307, 317, 379
of effectors������������������������������������������������������������������463 N-hydroxysuccinimide (NHS)-based reagent������������ 88, 259
of the secretion systems������������������������������������ 1–19, 459 Nicotiana benthamiana������������������������������ 476, 479, 480, 485
Image processing������������������������������������������51, 69, 291, 294, Ni-NTA agarose beads����������������������������� 248–251, 253, 254
378, 393, 398, 405
O
Immunoblotting. See Western blot
Immunodetection���������������������������������69, 90, 100–102, 130, One-dimensional BN PAGE�������������������������������������������322
162, 203, 204, 252, 349, 453 One-hybrid�����������������������������������������������������������������������200
Bacterial Protein Secretion Systems: Methods and Protocols
527
Index
Orientation����������������������������� 94, 97, 98, 105, 107–112, 115, Pseudomonas aeruginosa���������������������������������� 66–69, 71, 267,
119, 123, 124, 129, 267, 306, 367, 368, 395–398, 401 311–313, 317, 318, 519
Ortho-nitrophenyl-β-D-galactoside (ONPG)����������������131, Pull-down������������������������������������������������� 169, 177, 248–254
133, 137, 139, 163, 168, 204, 207 Pulse-chase��������������������������������109, 234, 235, 237–241, 243
Osmotic shock��������������������������������������������������������������60–63 Purified peptidoglycan���������������������� 144–147, 149, 152–156
Outer membrane protein (OMP)���������������������47, 82, 85, 87,
97, 235, 342 R
Receiver operating characteristic (ROC) curve������������35, 36
P
Reconstruction������������������������� 296, 354, 356, 364–366, 372,
Palmitate labeling���������������������������������������������������������76, 77 373, 379, 387, 388, 394, 397–403, 405, 408, 416
p-benzoyl-l-phenylalanine (Bpa)������������� 234–236, 239, 242 Red-Gal (6-chloro-3-indolyl-β-D-galactoside)��������������131,
Peptidoglycan�����������������������������������59, 62, 79, 84, 143–149, 133, 139
151–156, 278, 285 Regular expression��������������������������������������������������������������39
Peptidoglycan-binding domain����������������������������������������145 Remazol blue���������������������������������������������������� 152, 153, 155
Performance measures��������������������������������������������������������35 Reporter gene�����������������������������������130, 132, 134, 135, 170,
Peripheral proteins��������������������������������������������������������63, 87 178, 200–202, 208, 485
Pho-lac dual reporter system (or pho-lac reporter Restriction site/ligation free cloning methods���������� 136, 300
fusions)������������������������������������������� 98, 132, 135, 140
Pilus��������������97, 109, 131, 143, 312–318, 415–418, 443, 445 S
Plant inoculation���������������������������������������������� 476, 479–481 Saccharomyces cerevisiae���������������������������������������������� 179, 180
Plunge freezing����������������������������������355, 357–358, 371, 381 Salmonella enterica serovar Typhimurium���������� 450, 502, 503
p-nitrophenyl phosphate (pNPP)������������� 131, 133, 138, 139 SDS-boiling������������������������������������������������������������� 144, 146
Polytopic������������������������ 97, 98, 102, 108–112, 119, 131, 139 Secretomes������������������������������������������������������������������������460
Positive-inside rule�������������������������������������������������������������44 Sec system��������������������������������������������������������������������������38
Prediction���������������������������� 52, 111, 112, 129, 134, 135, 441 Segmentation��������������������������������������������������� 363, 366, 367
Prey��������������������������� 178, 179, 182–183, 190, 248, 250–255 Sequence logo��������������������������������������������������� 31, 32, 37, 38
Pronase����������������������������������������������������� 144, 147, 153, 154 Sequence similarity search���������������������������2, 5, 8, 15, 16, 51
ProtA�������������������������������������������������222, 224–227, 229, 231 Serratia marcescens��������������������������������������������� 519, 521, 523
Protease���������������� 84, 88–90, 94, 98–102, 181, 186, 214, 215, Serva Blue G�������������������������������������322, 324, 325, 339, 344
217, 222, 224–227, 229–231, 236, 249, 250, 279, 300, Shearing�������������������������������������������������������������������311–319
302, 304, 322, 326, 327, 329, 344, 505, 508, 509 Signal peptide (or Signal sequence)����������������� 23, 31, 36–39,
Protease accessibility (or Proteinase accessibility)���������98–102, 45, 66, 75, 76, 91, 152, 300
281–283 Silver staining����������������������������222, 322, 325, 339–340, 344
Protease inhibitor��������������������������� 63, 90, 99, 102, 181, 182, Single-particle analysis�����������������������������������������������������403
214, 215, 217, 230, 249, 302, 304, 322, 326, 327, 329, Site-directed mutagenesis������������ 71, 119, 136, 191, 192, 235
344, 505, 508 Site-directed photocrosslinking�������������������������������� 234, 235
Protein A/G-agarose affinity resin��������������������������� 120, 122 Sodium carbonate������������������������������������������������� 60, 62, 325
Protein A/G Sepharose����������������������������������������������������117 Sodium dodecyl sulphate polyacrylamide gel electrophoresis
Proteinase K��������������89–91, 93, 99–102, 165, 166, 278, 279, (SDS-PAGE)���������������������������������61, 62, 77, 79, 82,
281–283, 285, 286, 478 85, 100–102, 109, 115, 116, 120, 122, 146, 148, 152,
Protein complex�������������������91, 190, 199, 212–214, 216, 221, 203–206, 211, 214, 216, 222, 226, 228, 230, 231, 236,
222, 231, 248, 299–301, 321, 322, 326–329, 334–336, 241, 243, 248, 250, 252, 261, 266, 313, 314, 316–318,
338, 340, 341, 344, 345, 377–409 322, 324–325, 328, 330, 334, 336–340, 348, 451,
Protein G-Sepharose����������������������������������������������������76, 78 453, 460, 461, 463, 503, 506, 508–511, 513
Protein-lipid interaction���������������������������������������������������199 Solid-state NMR������������������������������������������������������415–445
Protein overproduction�����������������������������������������������������299 Solubilisation�������������������������������������������� 60–63, 76, 78, 126
Protein-peptidoglycan interaction������������������������������������149 Sorting signals���������������������������������������23, 24, 29, 30, 52, 69
Protein-protein interaction�������������������������������� 81, 159–174, Spheroplast�����������������������������������������59–63, 67–72, 99–102,
177–186, 199, 211–218, 221–231, 247–255, 277, 279 278–283, 285, 286
Protein sorting��������������������������������������������������������������23–52 Structure determination���������������������������������� 106, 353, 417,
Proteolysis��������������� 63, 89–93, 100–102, 277, 278, 280, 286 429, 436, 440, 441, 443
Proton motive force Subcellular location (SCL) (or Subcellular
∆pH���������������������������������������������������������������������������454 localization)���������������������������������23, 29, 48, 131, 135
∆Ψ������������������������������������������������������������������������������� 454 Substituted cysteine accessibility method
Protonophore���������������������������������������������������� 279, 282, 286 (SCAM)��������������������������������������������������������� 98, 126
Bacterial Protein Secretion Systems: Methods and Protocols
528 Index
Sucrose gradient (or Sucrose density Trypsin����������������������������������������������������������� 63, 89, 90, 102
gradient)����������� 69, 82, 84, 85, 323–324, 329–334, 344 Twin-arginine protein translocation�����������������������������������39
Superfolder green fluorescentprotein. See Fluorescent Two-dimensional BN/SDS PAGE������������������ 322, 336, 340
reporters Two-hybrid system������������������������������������������� 177, 178, 180
Supernatant������������������������������������������������������� 60–63, 70, 78, Type III secretion (T3SS)������������������������2–4, 6, 8, 15, 17, 41,
84, 100, 101, 122, 148, 156, 182–184, 205, 206, 212, 42, 50, 66, 98, 143, 151, 179, 200, 213, 226, 289, 295,
215, 216, 218, 240, 250, 253, 261, 281–283, 286, 304, 301, 307, 321, 322, 336, 340–342, 345, 377–380, 386,
316, 327–329, 333–335, 357, 422, 424, 426, 444, 450, 392, 393, 399, 402, 409, 415–418, 431, 439, 440, 443,
452, 453, 455, 456, 463, 465, 468, 470, 481, 486, 495, 449–456, 459, 473, 474, 476, 492, 499, 502, 503
502, 508, 509, 512 Type IV secretion (T4SS)������������������������������� 6, 98, 109, 143
Support vector machine������������������� 24, 31, 33, 40, 44, 47, 49 Type VI secretion system (T6SS)���������������������� 6, 17–19, 40,
Surface exposed lipoproteins����������������������������������������87, 89 66, 68–69, 98, 152, 179, 199, 213, 226, 269, 270, 289,
Surface plasmon resonance (SPR)����������������������������257–274 301–303, 460, 465–471, 501, 517–522
T U
Tandem Affinity Purification (TAP)�������� 222–226, 228–231 Ultracentrifugation���������������������������������61, 81, 85, 146, 148,
TEM-1 beta-lactamase reporter��������������� 490, 492, 493, 497 154–156, 304, 328–330, 332, 346
Tilt series�����������������������������������354, 356, 360–365, 371, 372 Urea�������������������������������������������������������������� 60, 63, 125, 423
Tobacco Etch Virus (TEV)�������222, 224–227, 229, 231, 300
Topology W
lipoprotein topology�����������������������������������������������������91 Western blot��������������������������������� 61, 62, 112, 116, 117, 120,
membrane/Inner membrane protein 122, 125, 162, 168, 169, 181, 183–184, 186, 205, 206,
topology�������������������������������������������������� 97, 101, 102 212–214, 216, 218, 226, 228, 248, 253, 279, 341, 451,
transmembrane β-barrel/OMPs topology��������������44, 47 460, 462, 466, 470, 494, 498
T7 overexpression�������������������������������������������������������������300 Whole-cell dot blot������������������������������������������������������92, 93
TOXCAT�����������������������������������������������������������������200–206
Toxin/immunity pairs����������������������������������������������� 518, 519 X
Transglycosylase. See Lytic transglycosylase
X-Gal�����������������������������������������������163, 164, 166, 167, 169,
Transient protein interaction (or transient
173, 204, 206–208
interaction)��������������������������������������������������� 177, 244
X-Pho (5-bromo-4-chloro-3-indolyl-phosphate or
Transmembranedomain. See Transmembrane segment
BCIP)�������������131, 133, 135, 137, 138, 140, 163, 204
Transmembranehelix. See Transmembrane segment
Transmembraneprotein. See Membrane protein Y
Trans-membrane segment���������������������������� 36, 98, 129, 303
Tris-Tricine gel electrophoresis Yeast transformation������������������������������������������������� 181, 182
(Tricine-SDS-PAGE)�������������� 77–79, 313, 316–318 Yeast two-hybrid (Y2H)���������������������������159, 162, 178, 179,
Triton X-10060, 62, 76, 79, 99–102, 121, 126, 213–217, 307, 182, 186, 213, 247
345, 502, 505, 508, 509, 513 Yersinia enterocolitica������������������������� 502–505, 507–508, 512