Download Identification and characterization of novel interaction

Document related concepts

Promoter (genetics) wikipedia , lookup

Epitranscriptome wikipedia , lookup

Metalloprotein wikipedia , lookup

Vectors in gene therapy wikipedia , lookup

G protein–coupled receptor wikipedia , lookup

Point mutation wikipedia , lookup

Biochemical cascade wikipedia , lookup

Eukaryotic transcription wikipedia , lookup

Paracrine signalling wikipedia , lookup

Magnesium transporter wikipedia , lookup

Gene regulatory network wikipedia , lookup

Lac operon wikipedia , lookup

SR protein wikipedia , lookup

Biochemistry wikipedia , lookup

Endogenous retrovirus wikipedia , lookup

Protein wikipedia , lookup

RNA polymerase II holoenzyme wikipedia , lookup

Artificial gene synthesis wikipedia , lookup

Signal transduction wikipedia , lookup

Expression vector wikipedia , lookup

Phosphorylation wikipedia , lookup

Nuclear magnetic resonance spectroscopy of proteins wikipedia , lookup

Bimolecular fluorescence complementation wikipedia , lookup

Gene expression wikipedia , lookup

QPNC-PAGE wikipedia , lookup

Western blot wikipedia , lookup

Transcriptional regulation wikipedia , lookup

Silencer (genetics) wikipedia , lookup

Proteolysis wikipedia , lookup

Interactome wikipedia , lookup

Protein–protein interaction wikipedia , lookup

Two-hybrid screening wikipedia , lookup

Transcript
Identification and characterization of novel
interaction partners of CcpA in Bacillus subtilis
Identifizierung und Charakterisierung neuartiger
Interaktionspartner von CcpA in Bacillus subtilis
Der Naturwissenschaftlichen Fakultät
der Friedrich-Alexander-Universität Erlangen-Nürnberg
zur
Erlangung des Doktorgrades Dr. rer. nat
vorgelegt von
Andrea Wünsche
aus Kecskemet
Als Dissertation genehmigt von den Naturwissenschaftlichen Fakultäten der Universität Erlangen-Nürnberg
Tag der mündlichen Prüfung:
06.07.2012
Vorsitzender der Promotionskommission:
Prof. Dr. Rainer Fink
Erstberichterstatter:
Prof. Dr. Andreas Burkovski
Zweitberichterstatter:
Prof. Dr. Jörg Stülke
TABLE OF CONTENTS
I
Abbreviation index .............................................................................................................................. IV
1
Zusammenfassung ........................................................................................................................ 1
2
Summary ....................................................................................................................................... 2
3
Introduction .................................................................................................................................. 3
3.1
4
Carbon catabolite regulation in bacteria .......................................................................................... 3
3.1.1
CCR in E. coli.................................................................................................................................. 5
3.1.2
CCR in B. subtilis ............................................................................................................................ 7
3.2
Differential regulation by CcpA in B. subtilis ................................................................................ 10
3.3
Scope of the thesis ............................................................................................................................. 11
Materials and methods ............................................................................................................... 12
4.1
Materials ........................................................................................................................................... 12
4.1.1
Chemicals ...................................................................................................................................... 12
4.1.2
Auxiliary Material ......................................................................................................................... 14
4.1.3
Instruments .................................................................................................................................... 14
4.1.4
Commercially available systems („kits“) ...................................................................................... 15
4.1.5
Proteins and Enzymes .................................................................................................................... 16
4.1.6
Oligonucleotides, plasmids and bacterial strains ........................................................................... 16
4.2
Media, buffers and solution ............................................................................................................. 22
4.2.1
Media ............................................................................................................................................. 22
4.2.1.1
Stock solutions for minimal media .................................................................................. 22
4.2.1.2
Additives ......................................................................................................................... 23
4.2.2
Buffers ........................................................................................................................................... 24
4.2.2.1
General buffers ................................................................................................................ 24
4.2.2.2
Buffers and solutions for agarose gel electrophoresis ..................................................... 24
4.2.2.3
Buffers and solutions for Laemmli polyacrylamide gel electrophoresis ......................... 24
4.2.2.4
Buffers and solutions for Schaegger polyacrylamide gel electrophoresis ....................... 25
4.2.2.5
Buffers for staining of PAA gels with coomassie brilliant blue ...................................... 25
4.2.2.6
Buffers for protein purification of SPINE samples ......................................................... 25
4.2.2.7
Buffers for protein purification ....................................................................................... 25
4.2.2.8
Buffers for silver staining ................................................................................................ 26
4.2.2.9
Buffers for immunoblotting............................................................................................. 26
4.3
Methods ............................................................................................................................................. 26
4.3.1
General methods ............................................................................................................................ 26
4.3.2
Growth of bacteria ......................................................................................................................... 27
4.3.2.1
Preparation and transformation of chemically competent E. coli .................................... 27
4.3.2.2
Preparation and transformation of naturally competent B. subtilis.................................. 27
4.3.3
Nucleic acid purification and modification.................................................................................... 28
4.3.3.1
Preparation of chromosomal DNA from B. subtilis ........................................................ 28
4.3.3.2
Polymerase chain reaction (PCR) .................................................................................... 28
4.3.3.3
Colony PCR .................................................................................................................... 28
II
TABLE OF CONTENTS
4.3.3.4
Restriction........................................................................................................................ 29
4.3.3.5
Dephosphorylation........................................................................................................... 29
4.3.3.6
Ligation............................................................................................................................ 29
4.3.3.7
DNA gel electrophoresis ................................................................................................. 29
4.3.3.8
DNA Sequencing ............................................................................................................. 29
4.3.4
4.3.4.1
SDS polyacrylamide gel electrophoresis (PAGE) ........................................................... 30
4.3.4.2
Silver staining of PAA gels ............................................................................................. 30
4.3.4.3
Immunoblot analysis........................................................................................................ 31
4.3.4.4
Growth of bacteria for protein overexpression ................................................................ 31
4.3.4.5
Sonication ........................................................................................................................ 31
4.3.4.6
Purification of CcpA(His)6 from B. subtilis ..................................................................... 32
4.3.4.7
Purification of CodY(His)6 from B. subtilis .................................................................... 32
4.3.4.8
Purification of RpoA(His)6 from B. subtilis .................................................................... 32
4.3.4.9
Purification of αNTD(His)6 from B. subtilis .................................................................... 32
4.3.4.10
Purification of αCTD(His)6 from B. subtilis .................................................................... 33
4.3.4.11
Purification of HPr seryl phosphorylated form from B. subtilis ...................................... 33
4.3.4.12
Purification of Strep-tagged CcpA .................................................................................. 34
4.3.4.13
Purification of Avi-tagged CcpA ..................................................................................... 34
4.3.4.14
Determination of protein concentration ........................................................................... 35
4.3.4.15
Storage and dialysis of proteins ....................................................................................... 35
4.3.4.16
Surface plasmon resonance measurements (SPR) ........................................................... 35
4.3.5
Proteomic analysis ......................................................................................................................... 36
4.3.5.1
Growth conditions and preparation of protein extracts .................................................... 36
4.3.5.2
Strep protein interaction experiment (SPINE) ................................................................. 36
4.3.5.3
MS analysis of SPINE eluates ......................................................................................... 36
4.3.6
Enzyme assays ............................................................................................................................... 37
4.3.6.1
Bacterial two hybrid assay ............................................................................................... 37
4.3.6.2
Liquid β-galactosidase assay in E. coli ............................................................................ 39
4.3.6.3
Liquid β-galactosidase assay in B. subtilis ...................................................................... 39
4.3.7
5
Protein purification and characterization ....................................................................................... 30
Software and computer analyses .................................................................................................... 40
Results .......................................................................................................................................... 41
5.1
Strategy for the isolation and the identification of CcpA interacting proteins involved in
transcriptional activation of ackA by in vivo crosslinking ........................................................................... 41
5.2
Isolation of CcpA complexes involved in transcriptional activation of ackA by SPINE............. 43
5.2.1
Construction of WH1012 and optimization of in vivo crosslinking ............................................... 43
5.2.2
Isolation of StrepCcpA-complexes by SPINE ............................................................................... 45
5.3
Identification of StrepCcpA interaction partners from SPINE by mass spectrometry .............. 46
5.4
Interaction partners of StrepCcpA in vivo ..................................................................................... 48
5.4.1
Analysis of the interaction of StrepCcpA with HPr in vivo ........................................................... 48
TABLE OF CONTENTS
III
5.4.2
Analysis of the interaction of StrepCcpA with Crh in vivo ........................................................... 50
5.4.3
Interaction of StrepCcpA with the α subunit of the RNA polymerase (RpoA) in vivo ................. 51
5.4.4
Interaction of StrepCcpA with CodY in vivo ................................................................................. 52
5.5
BACTH analysis of direct interactions between CcpA and putative interaction partners ........ 54
5.5.1
Interaction analysis of CcpA with RNA polymerase subunits α, β and β′ ..................................... 54
5.5.2
Interaction analysis of CcpA with mutants of the α subunit of RNA polymerase......................... 56
5.5.3
Interaction analysis of CcpA with CodY by BACTH ................................................................... 58
5.5.4
Interaction analysis of CodY with α subunit of RNA polymerase by BACTH............................. 59
5.5.5
Interaction analysis of CcpA with tricarboxylic acid (TCA) cycle enzymes ................................. 59
5.6
Characterization of the complex formation of CcpA, CodY and the α subunit of the RNA
polymerase by in vitro measurements ........................................................................................................... 61
5.6.1
Construction of overexpression vectors for purification of the α subunit of the RNA polymerase
and CodY ..................................................................................................................................................... 61
5.6.2
5.7
SPR analyses of interactions of CcpA, RpoA and CodY............................................................... 64
5.7.1
SPR analyses of interactions between CcpA and RpoA ................................................................ 64
5.7.2
SPR analyses of interactions between CcpA and CodY ................................................................ 65
5.7.3
SPR analyses of interactions between CodY and RpoA ................................................................ 67
5.8
6
Purification of CcpA, RpoA, αNTD, αCTD, HPrSer46P and CodY ............................................. 63
SPR analyses of immobilized CcpA with HPrSer46P and αNTD ................................................ 68
5.8.1
SPR analysis of Strep-tagged CcpA .............................................................................................. 68
5.8.2
Immobilization of CcpA via Avi-tag on an SPR chip ................................................................... 70
5.8.2.1
Enzymatic biotinylation of proteins in vivo based on BirA ............................................. 70
5.8.2.2
Construction and purification of in vivo biotinylated Avi-tagged CcpA ......................... 71
5.8.2.3
SPR measurements of Avi-tagged CcpA with HPrSer46P and αNTD............................ 71
Discussion .................................................................................................................................... 73
6.1
SPINE analysis of StrepCcpA ......................................................................................................... 73
6.2
Binary protein-protein interactions analyzed by BACTH ............................................................ 75
6.3
Interactions of CcpA, CodY and RpoA involved in activation of ackA ....................................... 77
7
References ................................................................................................................................... 81
8
Publications ................................................................................................................................. 93
9
Appendix ..................................................................................................................................... 94
IV
ABBEVIATION INDEX
Abbreviation index
αCTD
αNTD
ADP
Amp
AMP
APS
ATP
B.
bp
BSA
CAA
cAMP
CcpA
CCR
Cm
Crh
DMSO
DNA
DNase
dNTP
DTT
E.
EDTA
Erm
et al.
FBP
Fig.
g
Glc-6-P
HABA
HPr
HPrSer46P
IPTG
Km
kDa
LB
NTA
OD600
ONPG
p. a.
PAA
PAGE
PCR
PTS
PVDF
RNAP
RpoA
RpoB
RpoC
rpm
RT
RU
SDS
Spec
SPR
C-terminal domain of the α subunit of the RNA polymerase
C-terminal domain of the α subunit of the RNA polymerase
Adenosindiphosphate
Ampicillin
Adenosinmomophosphate
Ammoniumperoxodisulfate
Adenosintriphosphate
Bacillus
base pair
bovine serum albumin
casamino acids
cyclic Adenosinmomophosphate
Catabolite control protein A
carbon catabolite regulation
Chloramphenicol
catabolite HPr-like protein
dimethylsulfoxide
desoxyribonucleic acid
desoxyribonuclease
desoxyribonucleotide
Dithiothreitol
Escherichia
Ethylendiaminotetraacetate
Erythromycin
”et alii“ (and others)
fructose 1,6-bisphosphate
figure
gravity (9.81 m/s2)
glucose-6-phosphate
Hydroxy-azophenyl-benzoic-acid
histidine containg protein
histidine containg protein phosphorylated at serine 46 residue
Isopropyl-ß-D-thiogalactopyranoside
Kanamycin
kilo dalton
Luria-Bertani (broth)
Nitrilotetraacidic acid
optical density at 600 nm
o-Nitrophenyl-β-D-galactopyranoside
“pro analysi”
polyacrylamide
polyacrylamid-gelelektrophoresis
polymerase chain reaction
phosphoenolpyruvate:sugar phosphotransferase system
Polyvinylidendifluoride
RNA polymerase
α subunit of the RNA polymerase
β subunit of the RNA polymerase
β′ subunit of the RNA polymerase
revolutions per minute
room temperature
response units
sodiumdodecylsulfate
Spectinomycin
Surface plasmon resonance
ABBREVIATION INDEX
Str
Tab.
TAE
TCA
Tc
TE
TEMED
Tris
wt
x-Gal
% (v/v)
% (w/v)
V
Streptomycin
Table
Tris-acetate-EDTA-buffer
tricarboxylic acid cylce
Tetracycline
Tris-EDTA
N,N,N’,N’-Tetramethyl-ethylendiamine
2-Amino-Hydroxymethylpropan-1,3-diol
wild type
5-Bromo-4-Chloro-3-indolyl-ß-D-galactopyranoside
Volume / volume
Weight / volume
Units
bp
°C
Da
g
h
l
m
M
min
mol
sec
U
V
W
Unit prefixes
base pair
degree centigrade
dalton
gram
hour
liter
meter
molar
minute
mol
second
unit
volt
watt
k
m
µ
n
p
Amino acid nomenclature (IUPAC-IUB, 1969)
A
C
D
E
F
G
H
I
K
L
alanine
cysteine
aspartatic acid
glutamic acid
phenylalanine
glycine
histidine
isoleucine
lysine
leucine
M
N
P
Q
R
S
T
V
W
Y
methionine
asparagine
proline
glutamine
arginine
serine
threonine
valine
tryptophan
tyrosine
kilo (103)
milli (10-3)
micro (10-6)
nano (10-9)
pico (10-12)
ZUSAMMENFASSUNG
1
1 Zusammenfassung
In Gram-positiven Bakterien mit niedrigem GC-Gehalt, wie Bacillus subtilis, wird KohlenstoffKatabolitenregulation überwiegend vom Katabolit Kontroll Protein A (CcpA), einem globalen
Transkriptionsregulator, vermittelt. CcpA reprimiert oder aktiviert die Expression von hunderten von
Genen, die hauptsächlich im Kohlenstoff- und Stickstoffmetabolismus involviert sind. Die
Phosphoproteine HPrSer46P und CrhP sowie die niedermolekularen Effektoren Fruktose-1,6bisphosphat (FBP) und Glukose-6-Phosphat (Glc-6-P) stimulieren die DNA-Bindefähigkeit von CcpA
in Gegenwart einer bevorzugten Kohlenstoffquelle, wie beispielsweise Glukose. Die Bindung dieser
Phosphoproteine an CcpA führt zu Strukturänderungen und ermöglicht die Bindung des CcpAPhosphoprotein-Komplexes
an
die
sogenannten
catabolite
responsive
elements
(cre).
In
vorangegangenen Arbeiten lieferten CcpA-Mutationsanalysen Hinweise für differenzielle Aktivitäten
einiger CcpA-Punktmutanten, da die Regulation von einigen aber nicht von allen Genen aufgehoben
wird und daher auf eine Beteiligung zusätzlicher Interaktionen von CcpA schließen lassen. Zur
Identifizierung solcher bislang unbekannter CcpA-Komplexe, die sowohl bei der Repression als auch
bei der Aktivierung von CcpA regulierten Genen beteiligt sind, wurde deshalb ein in vivo crosslinking
durchgeführt. Um das Verhältnis von CcpA reprimierten zu aktivierten Genen anzugleichen, wurde
die Anzahl einer Sequenz, die ackA Promotorregion, als repräsentatives durch CcpA aktiviertes Gen,
erhöht. Durch massenspektrometrische Analysen konnte HPr als CcpA Interaktionspartner bestätigt
werden. Unter den bisher unbekannten möglichen CcpA Interaktionspartnern befanden sich alle
Untereinheiten der RNA Polymerase sowie der globale Regulator CodY. In detaillierten
Untersuchungen mittels bakteriellem Two-Hybrid-System wurde nur eine direkte Interaktion zwischen
CcpA und der α-Untereinheit der RNA Polymerase (RpoA) detektiert. Die Bindung zwischen CcpA
und RpoA konnte ebenfalls mittels in vivo crosslinking Experimenten und Immunoblotanalysen
nachgewiesen werden. CcpA-RpoA Komplexe waren unabhängig vom Vorhandensein von Glukose
im Kultivierungsmedium detektierbar. Im Gegensatz dazu war die Interaktion zwischen CcpA und
CodY hauptsächlich bei Zugabe von Glukose nachweisbar und Komplexe zwischen CcpA und HPr
konnten ausschließlich durch Zugabe von Glukose zum Medium detektiert werden. In qualitativen und
quantitativen Oberflächenplasmonenresonanzanalysen wurde die Bindung von CcpA sowohl an die
N-, als auch an die C-terminale Domäne von RpoA (αNTD bzw. αCTD) bestätigt. Interaktionen jeder
einzelnen RpoA Domäne (αCTD und αNTD) mit CodY konnten ebenfalls nachgewiesen werden. Die
ermittelten KD-Werte dieser Bindungen liegen zwischen 5 und 8 µM, die Interaktion zwischen CcpA
und CodY wiederum ist mit einem KD-Wert von ca. 60 µM vergleichsweise schwach. Anhand der in
dieser Arbeit ermittelten Ergebnisse wurde ein Model für einen Transkriptionsinitiationskomplex am
ackA-Promotor postuliert.
2
SUMMARY
2 Summary
In Gram-positive bacteria with a low GC-content, e. g. Bacillus subtilis, carbon catabolite
regulation (CCR) is mainly mediated by the carbon catabolite control protein A (CcpA). CcpA is
a global transcriptional regulator that represses or activates hundreds of genes involved in carbon
and nitrogen metabolism. In response to the availability of preferred carbon sources like glucose
the phosphoproteins, HPrSer46P and CrhP, and the low-molecular weight effectors fructose-1,6bisphosphate (FBP) and glucose-6-phosphate (Glc-6-P) stimulate CcpA function. The
phosphoprotein-CcpA interaction triggers structural changes and converts CcpA into its DNA
binding structure which allows binding of catabolite responsive elements (cre). Distinct point
mutations in CcpA abolish regulation of some but not all target genes suggesting additional
interactions of CcpA. Therefore, in vivo crosslinking and mass spectrometry were applied to
identify CcpA complexes which are active in both repression and activation. This experiment was
accomplished with multiple copies of a promoter region of a gene which is activated by CcpA to
compensate for the large excess of promoters which are repressed by CcpA. For this purpose a
B. subtilis strain with multiple copies of the ackA promoter was subjected to in vivo crosslinking
and proteins interacting with CcpA were analyzed by mass spectrometry. Among these with CcpA
interacting proteins identified by mass spectrometry HPr, all RNA polymerase subunits as well as
the global regulator CodY were observed. In bacterial two-hybrid assays combining each RNA
polymerase subunit with CcpA only a binding between CcpA and the α subunit (RpoA) was
detected. In vivo crosslinking combined with immunoblot analyses revealed CcpA-RpoA
complexes in cultures with or without glucose whereas CcpA-HPr and CcpA-CodY complexes
occurred only or predominantly in cultures with glucose. Surface plasmon resonance (SPR)
analyses confirmed binding of CcpA to the N- (αNTD) and C-terminal domains (αCTD) of RpoA
as well as to CodY. Furthermore, binary interactions of CodY with the αNTD and the αCTD were
detected by SPR. The KD values of complexes of CcpA or CodY with the αNTD or the αCTD are
between 5 and 8 µM. CcpA and CodY form a loose complex with a KD of 60 µM. These data
were combined to propose a model for a transcription initiation complex at the ackA promoter.
INTRODUCTION
3
3 Introduction
3.1 Carbon catabolite regulation in bacteria
Bacteria have evolved elaborate mechanisms that facilitate survival in different habitats and
adaption to changing environmental conditions by utilization of many different compounds as a
source of carbon and energy (Görke & Stülke, 2008; Rojo, 2010; Stülke & Hillen, 2000).
Consequently, the expression of metabolic pathways is optimized for efficiency and ecological
fitness in response to the availability of water, osmolarity, oxygen and nutrient (Rojo, 2010;
Stülke & Hillen, 2000). Therefore, bacteria possess different regulatory mechanisms that ensure
survival under changing conditions. One of these evolved regulatory mechanisms is the carbon
catabolite regulation (CCR) which helps bacteria to selectively use the carbon sources that are
most easily accessible when they are exposed to a more or less complicated mixture of available
nutrients (Deutscher, 2008; Görke & Stülke, 2008; Gunnewijk et al., 2001; Stülke & Hillen,
2000). This is important for competition in natural environments, as selection of the preferred
carbon source supports a high growth rate and thus success in competition with other
microorganisms (Brückner & Titgemeyer, 2002; Stülke & Hillen, 2000). In the presence of
glucose the expression of genes required for the utilization of secondary carbon sources is
repressed and the activities of pre-existing enzymes are reduced to prevent waste of resources
(Görke & Stülke, 2008; Stülke & Hillen, 2000).
CCR has been most intensively studied in the Gram-negative model organism Escherichia coli
(Magasanik, 1961; Saier, 1989) as well as in the AT-rich Gram-positive model organism Bacillus
subtilis (Fujita, 2009; Lorca et al., 2005). Although, in both species the physiological outcome is
very similar, CCR is mediated by completely different regulatory mechanisms. In contrast to
E. coli, in which CCR is mediated by prevention of transcriptional activation of catabolic genes in
the presence of glucose, in B. subtilis, CCR is mediated by the catabolite control protein A
(CcpA) a regulator of the LacI / GalR family (Chambliss, 1993; Henkin et al., 1991; Hueck et al.,
1995; Weickert & Adhya, 1992). In both species the regulation of signals leading to CCR is
mediated by the phosphoenolpyruvate:carbohydrate phosphotransferase system (PTS) which also
transports preferred carbon sources, like glucose, into the cell (Cases et al., 2007; Deutscher et al.,
2006; Görke & Stülke, 2008; Postma et al., 1993; Saier & Reizer, 1994; Titgemeyer & Hillen,
2002). The PTS contains two components which are constitutively transcribed: Enzyme I (EI) and
the histidine-containing protein (HPr) and are common to all PTS carbohydrate uptake systems.
Enzyme II (EII) is the sugar-specific component and is a multidomain membrane bound permease
4
INTRODUCTION
which phosphorylates and translocates the sugar through the cytoplasma membrane. Bacteria
usually contain several different EIIs whereby each EII complex consists of three to four domains:
two hydrophilic domains (domains A and B) and one or two hydrophobic integral membrane
domains (domains C and D if present). At least 15 different EII complexes for such carbohydrates
were identified in both model organisms E. coli and B. subtilis (Deutscher et al., 2006; Reizer et
al., 1999).
For transport of carbohydrates EI autophosphorylates from phosphoenolpyruvate (PEP) and
transfers the phosphoryl group to the residue His15 of HPr. Then the EIIAGlc domain is
phosphorylated by HPr and further passes the phosphoryl group to the EIIBGlc domain. The
concomitant phosphorylation and translocation of glucose by EIICGlc result in intracellular
glucose-6-phosphate (Glc-6-P), where it can directly enter the glycolytic pathway (Fig. 3.1)
(Postma et al., 1993).
glucose
His15~ P
PEP
EI
CC
HPr
EIIA
EI~ P
HPr
~
pyruvate
B
P
glucose-6~ P
CM
Figure 3.1: The bacterial phosphoenolpyruvate: glucose phosphotransferase system (PTS).
The phosphoryl group is transferred from phosphoenolpyruvate (PEP) via Enzyme I (EI) to His15 of HPr.
Then the Enzyme IIA domain (EIIAGlc) is phosphorylated by HPr and further passes the phosphoryl group
to the Enzyme IIB domain (EIIBGlc), enables translocation as well as phosphorylation of PTS carbohydrates
like glucose. The phosphoryl groups are highlighted in blue. CM: cytoplasmic membrane (adapted from
Stülke & Hillen, 2000).
In both E. coli and B. subtilis, the expression and activities of enzymes required for the uptake and
catabolism of PTS substrates is mainly positively controlled by transcriptional regulators, which
possess two copies of a structural motif, termed PTS-regulatory domain (PRDs) (Stülke et al.,
1998). The activity of these regulators is controlled by EII- and HPr-dependent phosphorylation of
both PRD copies which are differently involved in the control of activity (Kotrba et al., 2001;
Stülke et al., 1998). The PDRs comprise antiterminators such as LicT (Deutscher et al., 1997;
Schnetz et al., 1996) SacT (Débarbouillé et al., 1990), SacY (Crutz et al., 1990; Steinmetz et al.,
1989) and GlcT (Schmalisch et al., 2003; Stülke et al., 1997) and transcriptional activators such
as LevR (Débarbouillé et al., 1991a; Débarbouillé et al., 1991b) and LicR (Tobisch et al., 1997;
Tobisch et al., 1999a) from B. subtilis (Kotrba et al., 2001). Phosphorylation of PRDs by the
INTRODUCTION
5
sugar specific EIIB inactivates the antiterminators, such as GlcT and SacY, in the absence of their
specific substrate and prevents induction (Bachem & Stülke, 1998; Schmalisch et al., 2003).
However, when the specific substrate is available, EII phosphorylates the substrate while the PRD
remains unphosphorylated. The unphosphorylated PRD of the antiterminator GlcT prohibits the
formation of the terminator structure by binding to sequences in the mRNA leader region, the so
called RAT (ribonucleic-antiterminator) (Langbein et al., 1999). PRD containing activators are
similar, but contain a DNA binding site instead of an RNA binding site (Fujita, 2009; Stülke et
al., 1998; Stülke & Hillen, 2000).
HPrHisP also contributes to CCR by phosphorylation of PRDs. In the presence of the specific
sugar and in the absence of glucose, HPrHisP phosphorylates the antiterminator LicT or the
activator LevR and activates them. In contrast, in the presence of glucose the concentration of
HPrH15P is low and prevents the induction (Débarbouillé et al., 1991b; Martin-Verstraete et al.,
1998; Stülke et al., 1998; Tortosa et al., 2001). This regulatory mechanism is called inducer
prevention since in the presence of glucose, the activation of the PRD containing regulators by
their inducers is prevented. In addition, HPrHis15P is necessary for glycerol utilization. In the
absence of a PTS sugar HPrHis15P phosphorylates the glycerol kinase GlpK and concurrently
stimulates the kinase activity of GlpK which generates glycerol-3-P, the inducer of the glpFK
operon. However, in the presence of PTS sugars GlpK activity is reduced and thus the utilization
of glycerol is abolished (Darbon et al., 1999; Darbon et al., 2002; Stülke & Hillen, 1999).
3.1.1
CCR in E. coli
In E. coli CCR is mainly determined by the phosphorylation state of the glucose-specific
phosphotransferase enzyme EIIAGlc. When cells grow in the presence of a PTS sugar like glucose
the phosphorylgroup from EIIAGlc is transferred to the EIIB domain of the permease EIIBCGlc
which catalyzes transport and simultaneous phosphorylation of the sugar and thus EIIAGlc is
mainly present in the unphosphorylated form (Postma et al., 1993). Unphosphorylated EIIAGlc
interacts with transporters of non-preferred sugars such as the lactose permease (LacY) or
glycerol kinase (GlpK) and inhibits their activity, excluding the inducer of the respective catabolic
operon from the cell (Görke & Stülke, 2008; Saier & Roseman, 1976). This mechanism is termed
inducer exclusion because intracellular formation of the inducer for the respective catabolic
system and accordingly, the uptake of non-preferred sugars is prevented. Furthermore, non-PTS
substrates like Glc-6-P or lactose increase the ratio of pyruvate / PEP, which also leads to
increasing amounts of unphosphorylated EIIAGlc and thus exerts CCR (Bettenbrock et al., 2007;
Hogema et al., 1998). In contrast, when glucose is being exhausted the ratio of PEP / pyruvate is
high and the amount of phosphorylated EIIAGlc increases while the amount of non-phosphorylated
6
INTRODUCTION
EIIAGlc decreases. Phosphorylated EIIAGlc activates the membrane bound enzyme adenylate
cyclase Cya, which leads to cyclic AMP (cAMP) synthesis. The signal molecule cAMP forms a
complex with the transcription activator CAP (catabolite activating protein, or also called cyclic
AMP (cAMP) receptor protein (CRP)). High cAMP concentrations trigger the formation of
cAMP-CAP complexes, which bind and activate the promoters of catabolic genes and operons.
Non-preferred sugars can then enter the cell and induce the expression of their catabolic pathways
(Fig. 3.2) (Busby & Ebright, 1999; Tagami & Aiba, 1998).
GlpK
LacY
P
~
PEP
glucose
His15
EIIAGlc
HPr
EI
EIICGlc
EIIBGlc
pyruvate
EIIAGlc
HPr
EI
~
~
P
P
Glc-6~ P
AMP
CAP
Cya
cAMP
CAP-cAMP
CM
Activation of catabolic genes and operons
Figure 3.2: CCR in E. coli
CCR in E. coli is mainly mediated by the EIIAGlc domain of the glucose-specific enzyme EIIGlc. In the
presence of glucose EIIAGlc is dephosphorylated and binds to metabolic enzymes and transporters of
secondary carbon sources, such as glycerol kinase (GlpK), lactose permease (LacY) thereby preventing
uptake and catabolism of these carbohydrates. When glucose is being exhausted phosphorylated EIIAGlc is
accumulating and is being able to activate the adenylate cyclase (Cya). Active Cya generates cyclic AMP
(cAMP) which binds CAP. The CAP-cAMP complex binds to promoters of catabolic genes and activates
them. This mechanism is termed inducer exclusion because it prevents the intracellular formation of the
inducer of the respective catabolic system. Glc-6-P: glucose-6-phosphate, CM: cytoplasmic membrane
(adapted from Görke & Stülke, 2008).
INTRODUCTION
3.1.2
7
CCR in B. subtilis
In B. subtilis CCR is mainly exerted by direct transcriptional regulation of the transcription of
genes that are relevant for the transport and metabolism of carbon sources (Deutscher, 2008;
Deutscher et al., 2006; Fujita, 2009; Görke & Stülke, 2008). Whereas the phosphorylation state of
EIIAGlc is crucial for CCR in E. coli, HPr phosphorylation is essential to CCR-related signal
transduction in B. subtilis. HPr contains two phosphorylation sites: residue histidine 15 and
residue serine 46. The catalytic residue His15 phosphorylated by EI at a high level of PEP is
involved in sugar transport and regulation, while Ser46 phosphorylated HPr by HPrK/P is
involved in CCR (Galinier et al., 1998; Reizer et al., 1998). HPrSer46P is the main effector for
the control protein A (CcpA) which is the master regulator in CCR in Gram-positive bacteria with
low GC-content (Görke & Stülke, 2008; Henkin et al., 1991; Warner & Lolkema, 2003; Weickert
& Adhya, 1992). CcpA and HPrSer46P form a ternary complex which induces the binding of this
complex to specific palindromic operator sites, the catabolite responsive elements (cre) and thus
leading to transcriptional regulation (Deutscher et al., 1995; Jones et al., 1997). During glycolytic
degradation of PTS sugars like glucose the concentration of glycolytic intermediates as FBP or
Glc-6-P increase and FBP stimulates the ATP-dependent kinase activity of HPrK/P to
phosphorylate HPr at the serine 46 residue (Galinier et al., 1998; Jault et al., 2000; Poncet et al.,
2004; Reizer et al., 1998). Consequently, in the presence of glucose HPrSer46P increases and
serves as coeffector for CcpA. The binding of HPrSer46P to CcpA causes a rotation of the CcpA
subdomains and the formation of the hinge helix which allows DNA binding (Schumacher et al.,
2004; Stülke & Hillen, 2000). In addition, the low-molecular-weight effectors FBP and Glc-6-P
enhance the interaction between CcpA and HPrSer46P (Deutscher et al., 1995; Seidel et al.,
2005). Under conditions of nutrient limitation the levels of FBP and Glc-6-P decrease while the
level of inorganic phosphate in the cell increases, HPrK/P dephosphorylates HPrSer46P and CCR
is abolished (Mijakovic et al., 2002) (Fig. 3.3).
DNA binding activity of CcpA is not only enhanced by HPrSer46P but also by the HPr-like
protein Crh, a second coeffector for CcpA in B. subtilis (Galinier et al., 1997). Both proteins HPr
and Crh exhibit 45 % sequence similarity and they are also structurally similar (Favier et al.,
2002), however, Crh cannot participate in glucose transport due to the lack of the His15 residue.
Crh has, therefore, an exclusively regulatory function that depends on the HPrK/P mediated
phosphorylation of Ser46 (Galinier et al., 1997). Although, CrhSer46P might interact with CcpA
it substitutes only to some extent for an HPrSer46Ala mutant (Galinier et al., 1999; Galinier et al.,
1997; Zeng et al., 2000). This might be explained by the following observations: (1) CcpA has a
much lower affinity for CrhSerP than for HPrSer46P (Schumacher et al., 2006; Seidel et al.,
8
INTRODUCTION
2005). (2) The expression of Crh is much weaker than that of HPr (Görke et al., 2004). (3) FBP
does not increase the affinity of CrhP to CcpA (Seidel et al., 2005). The function of Crh in
glucose-dependent CCR remains unclear, however, recently an additional function of Crh was
identified in the initiation of a glycolytic bypass as a specific but CcpA-independent function of
Crh seryl phosphorylation by HPrK/P (Landmann et al., 2011; Landmann et al., 2012). In this
process seryl phosphorylation of Crh abolishes the inhibitory interaction of Crh with the
methylglyoxal synthase MgsA and active MgsA prevents then the accumulation of
phosphorylated glycolytic intermediates in the cell. In contrast, HPr does not bind or inhibit MgsA
and, therefore, this inhibition is specific and obviously the main function of Crh under carbon
overflow conditions. However, since CrhSer46P binds CcpA in vivo and CrhP substitutes
HPrSer46P in vivo its function remains partly unclear because a deletion of crh does not influence
CCR.
glucose
P
~
EI
PEP
His15
Glc
EIIC
C
HPr
HPr
EIIAGlc
EIIBGlc
~
pyruvate
EI
HPr
HPr
P
~
Crh
P
PPi
Glc-6~ P
ATP
+
HPrK/P
Pi
CcpA
FBP
ADP
glycolysis
HPr
HPr
Ser46-P
CM
P
P
Crh
cre
Ser46-P
activation or repression
of catabolic genes
Figure 3.3: Carbon catabolite repression (CCR) in B. subtilis
In B. subtilis HPr or Crh can be phosphorylated at Ser46 by the HPr kinase / phosphorylase (HPrK/P). This
phosphorylation occurs when the intracellular concentrations of fructose-1,6-bisphosphate (FBP) and ATP
are high. Both HPrSer46P and CrhP bind to CcpA, respectively. The CcpA-HPrSer46P or CcpA-CrhP
complex bind to cre sites on the DNA, and thereby regulate the transcription of catabolic genes dependent
on the location of the cre with reference to the promoter region. HPrK/P dephosphorylates HPrSerP under
conditions of high inorganic phosphate (Pi) and low ATP. CM: cytoplasmic membrane (adapted from
Görke & Stülke, 2008).
INTRODUCTION
9
In B. subtilis CcpA regulates hundreds of genes involved in carbon metabolism, amino acid
anabolism, overflow metabolism, nitrogen and phosphate metabolism and has substantial effects
on growth, sporulation and competence (Fujita, 2009; Miwa et al., 2000; Moreno et al., 2001;
Tobisch et al., 1999b; Yoshida et al., 2001). In transcriptome studies the vast majority of genes
and operons is repressed by CcpA while only about 50 genes are activated by CcpA (Moreno et
al., 2001). Among the repressed genes are those encoding enzymes required for the utilization of
secondary carbon sources and other metabolic pathways. Genes coding for enzymes of the
tricarboxylic acid (TCA) cycle and the oxidative respiration are also strongly repressed by CcpA
due to the high rate of ATP synthesis at high glucose concentrations (Blencke et al., 2003). In
contrast, the expression of glycolytic genes which catalyze the conversion of triose phosphate to
phosphoenolpyruvate (gapA and the pgk operon) are induced by glucose (Blencke et al., 2003;
Lorca et al., 2005; Lulko et al., 2007; Moreno et al., 2001; Tobisch et al., 1999b). Furthermore,
CcpA also directly or indirectly activates genes which are involved in overflow metabolism such
as ackA, pta and alsS for excretion of acetate or acetoin to prevent accumulation of pyruvate or
acetyl-CoA in the cell (Grundy et al., 1993b; Presecan-Siedel et al., 1999; Renna et al., 1993;
Shivers et al., 2006; Turinsky et al., 1998). Thus, CcpA plays a crucial role in the coordinated
regulation of catabolism and anabolism to ensure optimal cell growth under varying
environmental conditions.
CcpA-dependent CCR is rather complex as CcpA does not only interact with various different
coeffectors and mediators but also binds a number of diverse cre sites in target promoters and
genes (Fujita, 2009; Miwa et al., 2000; Schumacher et al., 2011; Weickert & Chambliss, 1990).
Genes which are activated by CcpA e. g. ackA, pta and the ilv-leu operon, contain single or
multiple copies of cre sites upstream of the promoter (Grundy et al., 1993a; Moreno et al., 2001;
Presecan-Siedel et al., 1999; Shin et al., 1999; Shivers et al., 2006; Tojo et al., 2005; Turinsky et
al., 1998). The mechanism of CcpA-dependent activation is unknown, however, it is assumed that
CcpA might recruit the RNA polymerase by interaction with the α subunit of RNA polymerase or
facilitates open complex formation, which is required for transcription initiation similar to
transcriptional activation in E. coli (Browning & Busby, 2004). In contrast, in repressed genes the
cre sites are located in different regions of the gene. Transcription initiation is prevented when cre
sites overlap with the promoter region such as in amyE and bgl (Kim et al., 2005; Krüger &
Hecker, 1995; Nicholson et al., 1987). By binding to cre sites further downstream of the promoter
such as in the sigL gene and gnt, xyn and xyl operon, RNA polymerase activity is inhibited by
roadblock (Choi & Saier, 2005; Galinier et al., 1999; Kraus et al., 1994; Miwa et al., 1997).
10
INTRODUCTION
3.2 Differential regulation by CcpA in B. subtilis
Analyses of CCR of different promoter-lacZ fusions by point mutations in CcpA in B. subtilis and
B. megaterium revealed that there are not only ccpA mutants which are defective in CCR but there
are also several ccpA mutants which exhibit special regulatory behavior: one set of CcpA variants
exhibit glucose-independent regulation. Mutations of different regions of the CcpA core protein
lead to repression or activation of transcription in the absence of glucose. These residues are
involved in the allosteric switch mechanism or are located in the effector binding cleft and
therefore a site directed mutation probably simulate a low molecular weight effector and keep
CcpA in its DNA binding conformation (Diel, 2005; Küster-Schöck et al., 1999; Seidel, 2005;
Sprehe, 2007; Sprehe et al., 2007). Another set of CcpA mutants display the severe growth defect
on minimal media similar to a ∆ccpA mutant whereas they are able to repress xynP or xylA
expression as by wild type or vice versa (Fuchsbauer, 2003; Seidel, 2005). Moreover, there are
also CcpA variants exerting differential regulation, as some distinct CcpA point mutants regulate
certain promoters similar to the wild type, but fail to regulate other promoters (Sprehe et al., 2007;
Turinsky et al., 2000). In summary, all these data strongly suggest that different residues are
involved in several different mechanisms of transcriptional regulation exerted by CcpA.
Among the CcpA activated genes ackA is of special interest, because several CcpA variants with
point mutations in the hinge helix, the coeffector binding region and the C-terminal subdomain
were active in CCR of ackA, but deficient in regulation of other genes in in vivo and in vitro
studies (Diel, 2005; Horstmann, 2006; Horstmann et al., 2007; Seidel, 2005; Sprehe, 2007; Sprehe
et al., 2007; Turinsky et al., 2000). Furthermore, ackA is activated to different levels depending
on the growth phase (Grundy et al., 1993a). From these results and the versatility of known
interaction partners of CcpA it is assumed that there are very likely further unknown proteinprotein interactions affecting regulation of ackA by CcpA. Interestingly, the the pleiotropic, global
regulator, CodY additionally activates the transcription of ackA (Shivers et al., 2006). This
coregulation of ackA by CcpA and CodY is of special interest because CCR by CcpA is
stimulated by the presence of high FBP concentrations (Schumacher et al., 2007; Seidel et al.,
2005) and CodY binding to the ackA promoter is mainly stimulated by branched chain amino
acids (BCAAs) (Sonenshein, 2005; Sonenshein, 2007). CodY is a transcriptional regulator in
B. subtilis that activates or represses many metabolic genes which involved in nitrogen or carbon
metabolism and in the adaptation to nutritional conditions which limit growth (Fisher et al., 1996;
Molle et al., 2003; Slack et al., 1995; Sonenshein, 2005; Sonenshein, 2007).
INTRODUCTION
11
3.3 Scope of the thesis
The versatility of known CcpA interaction partners, the differential regulation mediated by CcpA
and, in addition, the regulation of some genes to different levels depending on the growth phase,
suggest further unknown protein-protein interactions. Therefore, the overall aim of this thesis was
to identify yet unknown interaction partners of CcpA in B. subtilis by in vivo crosslinking and to
characterize these interactions with appropriate methods.
12
MATERIALS AND METHODS
4 Materials and methods
4.1 Materials
The chemicals used in this work were purchased from Merck (Darmstadt), Roth (Karlsruhe), and
Sigma (Munich) in p. a. quality. All chemicals are summarized in Table 4.1. The following tables list
auxiliary material (Table 4.2), instruments (Table 4.3), commercially available kits (Table 4.4) and
proteins and enzymes (Table 4.5).
4.1.1
Chemicals
Table 4.1: Chemials
Chemical
Source
Acetic acid
Acrylamide
Adenosintriphosphat (ATP)
Agar
Agarose
Ammoniumperoxodisulfate (APS)
Ammoniumsulfate
Ampicillin
Bicine
Boric acid
β-Mercaptoethanol
5-Bromo-4-chlor-3-indolyl-β-D-galactopyranosid (X-Gal)
Bromophenolblue
Casamino acids
Calciumchloride
Chloramphenicol
Coomassie Brilliant Blue
D-desthiobiotin
D(+)-glucose
D(+)-glucose-6-phosphate
Dimethylformamide (DMF)
Dimethylsulfoxid (DMSO)
Dithiotreitol (DTT)
Dipotassiumhydrogenphosphate
Disodiumhydrogenphosphate
dNTPs
D(+)-xylose
EDTA, disodium salt
Erythromycine
Ethanol
Ferric Ammonium Citrate
Formamide
Formic acid
D-fructose
D-fructose-1,6-bisphosphate
Merck, Darmstadt
Roth, Karlsruhe
Roche, Mannheim
Oxoid, Heidelberg
Peqlab, Erlangen
Sigma, Munich
Merck, Darmstadt
Sigma, Munich
Sigma, Munich
Merck, Darmstadt
Merck, Darmstadt
Roth, Karlsruhe
Roth, Karlsruhe
Sigma, Munich
Roth, Karlsruhe
Roche, Mannheim
Gibco / BRL, Karlsruhe
IBA, Göttingen
Merck, Darmstadt
Sigma, München
Sigma, München
Sigma, München
Roth, Karlsruhe
Merck, Darmstadt
Merck, Darmstadt
Roche, Mannheim
Merck, Darmstadt
Merck, Darmstadt
Sigma, München
Roth, Karlsruhe
Sigma, München
Fluka AG, Switzerland
Roth, Karlsruhe
Sigma, München
Fluka AG, Switzerland
MATERIALS AND METHODS
13
Chemical
Source
Glutamic acid
Glycerol
HABA (Hydroxy-azophenyl-benzoic-acid)
Hydrochloric acid 37 %
4-(2-Hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES)
Imidazol
Isoleucine
Isopropyl thio-β-D-galactoside (IPTG)
Kanamycine
Leucine
Maleic acid disodium salt
Magnesiumchloride
Magnesiumsulfate
Manganesesulfate
Methanol
Methionine
Nickelsulfate
o-Nitrophenyl-β-D-galactopyranosid (ONPG)
Paraformaldehyde
Polyethylenglycol 6000
Potassiumdihydrogenphosphate
Potassiumhydroxide
Phenylmethylsulfonylfluorid
Sodiumacetate
Sodiumchloride
Sodiumdihydrogenphosphat
Sodium dodecyl sulfate (SDS)
Sodiumformiate
Sodiumhydroxide
Sodiumsuccinate
Sodiumthiosulfate
SoftLink™ Soft Release Avidin Resin
Spectinomycin
Strep-Tactin Sepharose
Sucrose
Tetracycline
N,N,N’,N’-Tetramethylethylendiamin (TEMED)
Tris-(hydroxymethyl)-aminomethan (Tris-Base)
TritonX-100
Tryptone
Tryptophan
Tween20
Valine
Xylencyanol
X-Gal
Yeast extract
Roth, Karlsruhe
Roth, Karlsruhe
Sigma, München
Roth, Karlsruhe
Roth, Karlsruhe
Merck, Darmstadt
Sigma, München
PEQ Lab, Erlangen
Sigma, München
Sigma, München
Fluka AG, Switzerland
Roth, Karlsruhe
Fluka AG, Switzerland
Merck, Darmstadt
Roth, Karlsruhe
Sigma, München
Roth, Karlsruhe
Sigma, München
Roth, Karlsruhe
Roth, Karlsruhe
Merck, Darmstadt
Merck, Darmstadt
Sigma, München
Merck, Darmstadt
Merck, Darmstadt
Merck, Darmstadt
Roth, Karlsruhe
Merck, Darmstadt
Roth, Karlsruhe
Serva, Heidelberg
Merck, Darmstadt
Promega, Mannheim
Sigma, München
IBA, Göttingen
Merck, Darmstadt
Sigma, München
Merck, Darmstadt
Roth, Karlsruhe
Serva, Heidelberg
Oxoid, Heidelberg
Merck, Darmstadt
Sigma, München
Merck, Darmstadt
Roth, Karlsruhe
Peqlab, Erlangen
Oxoid, Heidelberg
14
4.1.2
MATERIALS AND METHODS
Auxiliary Material
Table 4.2: Auxiliary Material
Auxiliary material
Source
BIAmaintenance Kit for BiacoreX
Centriprep Centrifugal Filter (3 kDa, 10 kDa, 30 kDa)
Chromatograpy material DEAE-Sephacel
Electric dispenser
Eppendorf reaction tubes (1,5 ml; 2 ml)
Gene Amp reaction tubes
Glass bottles
Glass pipettes
Greiner tubes (15 ml; 50 ml)
Half micro vessels
HBS-EP buffer
Hiprep SepharoseQ column
HisTrap columns 1 ml and 5 ml
Microliterpipets
PCR reaction tubes
Petri dishes
Pipettes tips
Poly-Prep Chromatography Columns
Polyvinylidenedifluoride (PVDF) membrane (0.45 µm pore size)
Sensorchip CM5
Sensorchip SA
Size exclusion chromatography column Superdex G75
Size exclusion chromatography column Superdex G200
Sterile filter Minisart (0.2 and 0.45 µm pore size)
StrepTrap column 1 ml
Whatman paper
96 well cell culture plate (sterile, F-bottom, with lid)
GE Healthcare, Munich
Millipore, Eschborn
GE Healthcare, Munich
Eppendorf, Hamburg
Greiner, Nürtingen
Perkin Elmer, Weiterstadt
Schott, Mainz
Brand, Wertheim
Greiner, Nürtingen
Greiner, Nürtringen
GE Healthcare, Munich
GE Healthcare, Munich
GE Healthcare, Munich
Gilson, Düsseldorf
Peqlab, Erlangen
Greiner, Nürtingen
Greiner, Nürtingen
Bio-Rad, Munich
Roth, Karlsruhe
GE Healthcare, Munich
GE Healthcare, Munich
GE Healthcare, Munich
GE Healthcare, Munich
Schleicher & Schüll, Dassel
GE Healthcare, Munich
Millipore, Eschborn
Greiner Bio-One, Kremsmünster
4.1.3
Instruments
Table 4.3: Instruments
Instrument
Source
ABI PRISM 310 Genetic Analyzer
ÄKTA FPLC
ÄKTA Prime
ÄKTA Prime Plus
Analytic Balance Satorius
Autoclave Deutsch and Neumann
Autoclave Systec 5075 EL
Biocad Vision Workstation
BIAcore X
Biofuge centrifuge fresco, pico and primo R
Bio-Print Video-Geldocu-system Mod.215
ChemiDoc XRS+ System
Centrifuge J21B
Applied Biosystems, Weiterstadt
GE Healthcare, Munich
GE Healthcare, Munich
GE Healthcare, Munich
Satorius, Göttingen
Deutsch and Neumann
Systec GmbH, Wettenberg
Applied Biosystems, Weiterstadt
GE Healthcare, Munich
Heraeus Christ, Osterode
Peqlab, Erlangen
Bio-Rad, Munich
Beckmann, Munich
MATERIALS AND METHODS
15
Instrument
Source
Centrifuge Avanti J25
Electrical Dispenser
French Pressure cell press
Heatblock Dri Block DB3
Horizontal shaker
Macro-pipette
Magnetic Heat stirrer
Microliterpipetes Pipetman P20, P200, P1000
Microprocessor pH-meter 766 Calimatic
Milli-Q© PF Plus
Mini-PROTEAN® Tetra Cell
Mini Trans-Blot Electrophoretic Transfer Cell
Mini-V 8·10 Blot Module
Mini Western Blotting System EBX-700
NanoDrop ND1000 Spektrophotometer
Power supply TN 300-120
Power supply
Rotary shaker
Rotary shaker Mod G25
Rotary shaker TM4430
Spectralphotometer Ultrospec 3000
Sonic Sonifier Branson B12
Speed Vac centrifuge
TECAN SpectraFluor Plus
TECAN Infinite F200 Pro
Thermo Cycler primo 96 advanced
Ultra centrifuge
Ultrasonic Sonoplus HD2070 + UW2070
UV Transilluminator 366 nm
Vortex VF2
Water bath TW 12
Water deionization system
Water rotary shaker Innova 3100
Beckmann, Munich
Eppendorf, Hamburg
Thermo Fisher, Schwäbisch-Gmünd
Techne, England
New Brunswick, Neu Isenburg
RCT, Heidelberg
JKA Werk, Staufen
Gilson, Düsseldorf;
Knick, Berlin
Millipore, Eschborn
Bio-Rad, Munich
Bio-Rad, Munich
Life Techn., Gibco / BRL, Karlsruhe
C.B.S. Scientific Company, San Diego
Peqlab, Erlangen
Heinzinger, Rosenheim
Bio-Rad, Munich
GFL, Burgwedel
New Brunswick, Neu Isenburg
New Brunswick, Neu Isenburg
Amersham Biosciences, Freiburg
Braun, Melsungen
Bachofer, Reutlingen
TECAN, Crailsheim
TECAN, Crailsheim
Peqlab, Erlangen
Beckman, Munich
GE Healthcare, Munich
Vetter, Wiesloch
IKA Labortechnik, Staufen
Julabo, Seelbach
Millipore, Schwalbach
New Brunswick, Neu Isenburg
4.1.4
Commercially available systems („kits“)
Table 4.4: Commercially available systems („kits“)
System
Source
Bio-Rad Protein Assay
DNA Leiter-Mix
ECL+ Plus
Nucleobond AX 100
Nucleospin Plasmid
Nucleospin Extract 2 in 1
Pierce ECL Western Blotting Substrate
peqGOLD Protein Standard II
QIAamp® DNA Mini Kit
Ready-To-Go™ PCR beads
Termination-mix
Bio-Rad, Munich
PeqLab, Erlangen
GE Healthcare, Munich
Macherey-Nagel, Düren
Macherey-Nagel, Düren
Macherey-Nagel, Düren
Thermo Scientific, Schwerte
PeqLab, Erlangen
Qiagen, Hilden
GE Healthcare, Munich
Perkin Elmer, Weiterstadt
16
4.1.5
MATERIALS AND METHODS
Proteins and Enzymes
Table 4.5: Proteins and Enzymes
Proteins and Enzymes
Source
Bovine serum albumin (BSA)
DNaseI
Lysozyme
Proteinase K
Phusion DNA Polymerase
Restrictionendonucleases
Sigma, Munich
Sigma, Munich
Merck, Darmstadt
Stratagene, Heidelberg
Finnzymes, Finland
Biolabs, Schwalbach
Roche, Mannheim
Gibco / BRL, Karlsruhe
GE Healthcare, Munich
Sigma, Munich
Roche, Mannheim
Sigma, Munich
Biolabs, Schwalbach
Roche, Mannheim
(Küster et al., 1996)
(Landmann et al., 2011)
Jörg Stülke, department of microbiology,
Göttingen
Sigma, Munich
Neoclone, USA
IBA, Göttingen
Ribonuclease (RNase A)
Shrimp Alkaline Phosphatase
Streptavidine
Taq-DNA-polymerase
T4-DNA-ligase
Anti-CcpA antibody
Anti-Crh antibody
Anti-HPr antibody
Anti-Flag antibody, monoclonal
Anti-RpoA antibody, monoclonal
StrepMAB-Immo anitbody
4.1.6
Oligonucleotides, plasmids and bacterial strains
The following tables summarize oligonucleotides (Table 3.6), plasmids (Table 3.7), and bacterial strains
(Table 3.8) used in this work. All oligonucleotides were purchased from MWG Biotech.
Table 4.6: Oligonucleotides
Oligonucleotide
Sequence (5′′ – 3′′)
CcpA_B2H_BamHI-fwd
ATAGGCGGATCCCATGAGCAATATTAC Cloning of pWH2127, pWH2128
GATCTACGATGTAGCG
pWH2129 and pWH2130
CcpA_B2H_KpnI-rev
ATAAGAGGTACCAATGACTTGGTTGAC Cloning of pWH2127, pWH2128
TTTCTAAGCTCTATACG
pWH2129 and pWH2130
RpoA_B2H-BamHI-fwd
ATACGAGGATCCAATGATCGAGATTGA Cloning of pWH2139, pWH2140
AAAACCAAAAATCGAAAC
pWH2141 and pWH2142
RpoA_B2H_KpnI-rev
ATATGAGGTACCTGATCGTCTTTGCGA Cloning of pWH2139, pWH2140
AGTCCGAGTC
pWH2141 and pWH2142
RpoB_B2H_BamHI-fwd
ATAGGCGGATCCCTTGACAGGTCAACT Cloning of pWH2143, pWH2144
AGTTCAGTATGGAC
pWH2145 and pWH2146
RpoB_B2H_KpnI-rev
ATAAGAGGTACCTATTCTTTTGTTACTA Cloning of pWH2143, pWH2144
CATCGCGTTCAACGTC
pWH2145 and pWH2146
B2H_RpoAdCTD_Accrev
ATAAGTGGTACCTTTTGATCTTCTTCTT Cloning of pWH2155, pWH2156
TTTCAACCAGATTTCAG
pWH2157 and pWH2158
Application
B2H_RpoAdNTD_Bam-f AGAATAGGATCCGGTTGGTTTAACTGA Cloning of pWH2159, pWH2160
CGAAGCTCAACATG
pWH2161 and pWH2162
MATERIALS AND METHODS
17
Oligonucleotide
Sequence (5′′ – 3′′)
RpoC_B2H_SalI-fwd
AGAATAGTCGACCTTGCTAGATGTGAA Cloning of pWH2147, pWH2148
CAATTTTGAGTATATG
RpoC_B2H_KpnI-rev
AGAATAGGTACCAATTCAACCGGGACC Cloning of pWH2147, pWH2148
ATATCG
RpoC1_B2H_PstI-f
AGAATACTGCAGCCTTGCTAGATGTGA Cloning of pWH2149, pWH2150
ACAATTTTGAGTATATG
RpoC2_B2H_PstI-f
AGAATACTGCAGCTTGCTAGATGTGAA Cloning of pWH2149, pWH2150
CAATTTTGAGTATATG
CodY_B2H_BamHI-fwd
ATAGGCGGATCCAATGGCTTTATTACA Cloning of pWH2135, pWH2136,
AAAAACAAG
pWH2137, pWH2138
CodY_B2H_KpnI-rev
ATAAGAGGTACCGTATGAGATTTTAGA Cloning of pWH2135, pWH2136,
TTTTCTAATTCAATTAG
pWH2137, pWH2138
HPr_B2H_BamHI-fwd
ATAGGCGGATCCAATGGCACAAAAAA Cloning of pWH2131, pWH2132,
CATTTAAAGTAACTG
pWH2133, pWH2134
HPr_B2H_KpnI-rev
ATAAGAGGTACCGACTCGCCGAGTCCT Cloning of pWH2131, pWH2132,
TCGC
pWH2133, pWH2134
HPr-S46D-fwd
CAGTTAACCTTAAAGACATTATGGGTG HPr mutagenisis
TTATGTC
citB-B2H-Bam-f
ATAAGAGGATCCAATGGCAAACGAGC Cloning of pWH2376, pWH2377,
AAAAAACTG
pWH2378, pWH2379
citB-B2H-Acc-r
ATAAGAGGTACCTCGGACTGCTTCATT Cloning of pWH2376, pWH2377,
TTTTCACGAAGC
pWH2378, pWH2379
citB-Seq-fwd
CGCAACAATTGCGAATATG
citG-B2H-Bam-f
ATAAGAGGATCCGATGGAATACAGAA Cloning of pWH2380, pWH2381,
TTGAACGAGACACC
pWH2382, pWH2383
citG-B2H-Acc-r
ATAAGAGGTACCGCCGCCTTTGGTTTT Cloning of pWH2380, pWH2381,
ACCATGTCTTC
pWH2382, pWH2383
citZ-B2H-Bam-f
ATAAGAGGATCCGATGACAGCGACAC
GCGGTCTTG
citZ-B2H-Acc-r
ATAAGAGGTACCGCGGCTCTTTCTTCA Cloning of pWH2384, pWH2385,
ATCGGAACGAATTTTTG
pWH2386, pWH2387
icd-B2H-Bam-f
GTGAGAGGATCCGGTGGCACAAGGTG
AAAAAATTACAG
icd-B2H-Acc-r
GTGAGAGGTACCGCGTCCATGTTTTTG Cloning of pWH2388, pWH2389,
ATCAGTTCTTCTC
pWH2390, pWH2391
mdh-B2H-Bam-f
GTGAGAGGATCCGATGGGAAATACTC
GTAAAAAAGTTTCTG
mdh-B2H-Acc-r
GTGAGAGGTACCGCGGATAATACTTTC Cloning of pWH2392, pWH2393,
ATGACATTTTTGACAGATTC
pWH2394, pWH2395
pUT18-Seq-fwd
GCGGATAACAATTTCACACAG
Sequencing of pUT18 vectors
pUT18-Seq-rev
GCGATTTTCCACAACAAGTC
Sequencing of pUT18 vectors
pUT18C-Seq-fwd
GCCGGATGTACTGGAAAC
Sequencing of pUT18C vectors
pUT18C-Seq-rev
TACTGAGAGTGCACCATATTAC
Sequencing of pUT18C vectors
pKNT25-Seq-fwd
TGAGCGGATAACAATTTCACAC
Sequencing of pKNT25 vectors
pKNT25-Seq-rev
TGCGTAACCAGCCTGATG
Sequencing of pKNT25 vectors
pKT25-Seq-fwd
CGATTCGGTGACCGATTAC
Sequencing of pKT25 vectors
Application
Sequencing of citB gene
Cloning of pWH2384, pWH2385,
pWH2386, pWH2387
Cloning of pWH2388, pWH2389,
pWH2390, pWH2391
Cloning of pWH2392, pWH2393,
pWH2394, pWH2395
18
MATERIALS AND METHODS
Oligonucleotide
Sequence (5′′ – 3′′)
Application
pKT25-Seq-rev
GGTAACGCCAGGGTTTTC
Sequencing of pKT25 vectors
Linker 2rep3aa fwd-Bam
AGAATAGGATCCTTCAGGAGGCGGTG
GTTCCGGTG
Cloning of pWH2128-2rep3aa,
pWH2130-2rep3aa,
pWH2132HPrS46D-2rep3aa,
pWH2134HPrS46D-2rep3aa
Linker 2rep3aa rev-Bam
AGAATTGGATCCAGTCTAGAACCGCCA Cloning of pWH2128-2rep3aa,
GAACCGCC
pWH2130-2rep3aa,
pWH2132HPrS46D-2rep3aa,
pWH2134HPrS46D-2rep3aa
Linker 2rep3aa fwd-Kpn
AGAATTGGTACCTTCAGGAGGCGGTGG Cloning of pWH2127-2rep3aa,
TTCCGGTG
pWH2129-2rep3aa,
pWH2131HPrS46D-2rep3aa,
pWH2133HPrS46D-2rep3aa
Linker 2rep3aa rev-Kpn
AGAATTGGTACCAGTCTAGAACCGCCA Cloning of pWH2127-2rep3aa,
GAACCGCC
pWH2129-2rep3aa,
pWH2131HPrS46D-2rep3aa,
pWH2133HPrS46D-2rep3aa
Linker 6rep3aa revAcc65I
AGAATAGGTACCATTCTAGAACCACCA Cloning of pWH2127-6rep3aa,
CTTCCGC
pWH2129-6rep3aa,
pWH2131HPrS46D-6rep3aa,
pWH2133HPrS46D-6rep3aa
Linker 6rep3aa revBamHI
AGAATAGGATCCATTCTAGAACCACCA Cloning of pWH2128-6rep3aa,
CTTCCGC
pWH2130-6rep3aa,
pWH2132HPrS46D-6rep3aa,
pWH2134HPrS46D-6rep3aa
CodY-Flag-Eco-fwd
AGAATAGAATTCCGTCAATTCCCTGAA Cloning of pWH2371
GAATATAC
CodY-Flag-Bam-rev
AGAATAGGATCCATGAGATTTTAGATT Cloning of pWH2371
TTCTAATTCAATTAGG
RpoA-NTD-NheI-fwd
AGAATACTCGAGTTAAGTTAAACCAAC Cloning of pWH2176
GAATATATTAAGGTGTTC
RpoA-XhoI-rev
AGAATACTCGAGTCAATCGTCTTTGCG Cloning of pWH2176
AAGTCC
CodY-NdeI-fwd
AGAATACATATGATGGCTTTATTACAA Cloning of pWH2177
AAAACAAGAATTATTAAC
CodY-XhoI-rev
AGAATACTCGAGTTAATGAGATTTTAG Cloning of pWH2177
ATTTTCTAATTCAATTAGG
pASK-IBA17+_fwd
AGTTATTTTACCACTCCCTATCAG
Sequencing of pASK-IBA17(+)
vectors
pASK-IBA17+_rev
GACGCAGTAGCGGTAAAC
Sequencing of pASK-IBA17(+)
vectors
Strep-CcpA-EcoRI-fwd
ATAAGAGAATTCGAGCAATATTACGAT Cloning of pWH2375
CTACGATGTAGCGAGAG
Strep-CcpA-BamHI-rev
ATAAGAGGATCCTTATGACTTGGTTGA Cloning of pWH2375
CTTTCTAAGCTCTATACG
pAN4-Seq-fwd
GAGCTGTTGACAATTAATCATC
Sequencing of pAN4 or pAC4 vectors
pAN4-Seq-rev
TTATCAGACCGCTTCTGC
Sequencing of pAN4 or pAC4 vectors
AviCcpA-Xho-fwd
AGAATACTCGAGGATGAGCAATATTAC Cloning of pWH2369
GATCTACGATGTAGC
MATERIALS AND METHODS
19
Oligonucleotide
Sequence (5′′ – 3′′)
AviCcpA-Acc-rev
AGAATTAGGTACCTTATGACTTGGTTG Cloning of pWH2369
ACTTTCTAAGCTCTATAC
CcpA-Avi-NcoI-fwd
AGAATACCATGGCGAGCAATATTACGA Cloning of pWH2370
TCTACGATGTAGCGAGAG
CcpA-Avi-XmaI-rev
AGAATTCCCGGGTTGACTTGGTTGACT Cloning of pWH2370
TTCTAAGCTCTATACG
Application
Table 4.7: Plasmids
Plasmid
Characteristics
Reference
pHT304
AmpR, ErmR, ori ColE1, ori1030
(Arantes & Lereclus, 1991)
R
pET3c
Amp , oriBR
Novagen
pWH653
pET3c, ccpAhis6 (B. subtilis)
(Seidel, 2005)
pWH466
pET3c, ptsH (B. subtilis)
(Seidel et al., 2005)
R
R
pWH1520
Amp , Tc , XylR, xylA’, ori pBR, ori BC16
(Rygus & Hillen, 1991)
pWH933
pWH1520 derivative, ackA promoter region
(Bartholomae, 2008)
R
R
pGB382
Amp , Erm , ori ColE1, ori 1030, pdegQ36, (Herzberg et al., 2007)
C-terminal Strep-tag
pWH940
pGP382, ccpA
p4813
pUT18
pUT18C
pKNT25
pKT25
pUT18C-zip
(Bartholomae, 2008)
R
(Reizer et al., 1998)
R
(Karimova et al., 1998)
R
(Karimova et al., 1998)
Amp , hprK
Amp , ori ColE1
Amp , ori ColE1
R
Km , ori p15A
R
Km , ori p15A
R
Amp , ori ColE1
R
(Karimova et al., 1998)
(Karimova et al., 1998)
(Karimova et al., 1998)
pKT25-zip
Km , ori p15A
(Karimova et al., 1998)
pWH2127
pUT18, ccpA
This work
pWH2128
pUT18C, ccpA
This work
pWH2129
pKNT25, ccpA
This work
pWH2130
pKT25, ccpA
This work
pWH2139
pUT18, rpoA
This work
pWH2140
pUT18C, rpoA
This work
pWH2141
pKNT25, rpoA
This work
pWH2142
pKT25, rpoA
This work
pWH2143
pUT18, rpoB
This work
pWH2144
pUT18C, rpoB
This work
pWH2145
pKNT25, rpoB
This work
pWH2146
pKT25, rpoB
This work
pWH2147
pUT18, rpoC
This work
pWH2148
pUT18C, rpoC
This work
pWH2149
pKNT25, rpoC
This work
pWH2150
pKT25, rpoC
This work
20
MATERIALS AND METHODS
Plasmid
Characteristics
Reference
pWH2155
pUT18, rpoA mutant (αNTD (aa 1-244)
This work
pWH2156
pUT18C, rpoA mutant (αNTD (aa 1-244)
This work
pWH2157
pKNT25, rpoA mutant (αNTD (aa 1-244)
This work
pWH2158
pKT25, rpoA mutant (αNTD (aa 1-244)
This work
pWH2159
pUT18, rpoA mutant (αCTD (aa 224-314)
This work
pWH2160
pUT18C, rpoA mutant (αCTD (aa 224-314)
This work
pWH2161
pKNT25, rpoA mutant (αCTD (aa 224-314)
This work
pWH2162
pKT25, rpoA mutant (αCTD (aa 224-314)
This work
pWH2131
pUT18, ptsH
This work
pWH2132
pUT18C, ptsH
This work
pWH2133
pKNT25, ptsH
This work
pWH2134
pKT25, ptsH
This work
pWH2131-HPrS46D
pUT18, ptsH mutant (S46D)
This work
pWH2132-HPrS46D
pUT18C, ptsH mutant (S46D)
This work
pWH2133-HPrS46D
pKNT25, ptsH mutant (S46D)
This work
pWH2134-HPrS46D
pKT25, ptsH mutant (S46D)
This work
pWH2135
pUT18, codY
This work
pWH2136
pUT18C, codY
This work
pWH2137
pKNT25, codY
This work
pWH2138
pKT25, codY
This work
pWH2163
pUT18, ptsI
This work
pWH2164
pUT18C, ptsI
This work
pWH2165
pKNT25, ptsI
This work
pWH2166
pKT25, ptsI
This work
pWH2376
pUT18, citB
This work
pWH2377
pUT18C, citB
This work
pWH2378
pKNT25, citB
This work
pWH2379
pKT25, citB
This work
pWH2380
pUT18, citG
This work
pWH2381
pUT18C, citG
This work
pWH2382
pKNT25, citG
This work
pWH2383
pKT25, citG
This work
pWH2384
pUT18, citZ
This work
pWH2385
pUT18C, citZ
This work
pWH2386
pKNT25, citZ
This work
pWH2387
pKT25, citZ
This work
pWH2388
pUT18, icd
This work
pWH2389
pUT18C, icd
This work
pWH2390
pKNT25, icd
This work
pWH2391
pKT25, icd
This work
pWH2392
pUT18, mdh
This work
pWH2393
pUT18C, mdh
This work
MATERIALS AND METHODS
21
Plasmid
Characteristics
Reference
pWH2394
pKNT25, mdh
This work
pWH2395
pKT25, mdh
This work
R
R
pWH1520
Amp , Tc , xylR, xylA’, ori pBR, ori BC16
(Rygus & Hillen, 1991)
pWH933
pWH1520, ackA promoter
Bartholomae 2008
R
pGP382
AmpR, Erm , C-Strep-tag, ori pBR, ori
BC16
(Herzberg et al., 2007)
pWH940
pGP382, ccpA
Bartholomae 2008
R
pWH338
ccpA-deletion plasmid, Km
(Sprehe et al., 2007)
pET28b
Overexpression vector for fusion of proteins
with N-terminal His-tag, KmR,
Novagen
pWH2175
pET28, rpoA
This work
pWH2176
pET28b, rpoA mutant (αNTD (aa 1-226))
This work
pWH2177
pET28b, codY
This work
pGP1331
Integrative vector for C-terminal fusion of
3 × Flag-tag, AmpR, SpecR
(Lehnik-Habrink et al., 2010)
pWH3171
pGP1331, codY’
This work
pAN4
Overexpression vector for fusion of proteins
with N-terminal Avi-tagTM, AmpR
GeneCopoeiaTM, Avidity, LLC,
Denver, Colorado
pWH2369
pAN4, ccpA
This work
pAC4
Overexpression vector for fusion of proteins
with C-terminal Avi-tagTM, AmpR
GeneCopoeiaTM, Avidity, LLC,
Denver, Colorado
pWH2370
pAC, ccpA
This work
pASK-IBA17(+)
Overexpression vector for fusion of proteins
with N-terminal Strep-tag, AmpR
IBA, Göttingen
pWH2375
pASK-IBA17(+),ccpA
This work
pET28a-αCTD
Overexpression vector for αCTD with
N-terminal His-tag, KmR
(Nakano et al., 2010)
Table 4.8: Bacterial strains
Strain
Genotype
Reference
DH5α
supE44, ∆lacU109 (φ80lacZ∆M15M),
hsdR17, recA1, endA1, gqrA96, thi-1, relA1
(Ausubel et al., 1994)
XL1blue
endA1, gyrA96(NalR), thi-1, recA1, relA1, lac, Stratagene
glnV44, F'[::Tn10, proAB+, lacIq
∆(lacZ)M15] hsdR17(rK-mK+)
BTH101
F–, cya-99, araD139, galE15, galK16,
rpsL1 (StrR), hsdR2, mcrA1, mcrB1
(Karimova et al., 1998)
BL21(DE3)
F–, dcm, ompT, hsdS (rB-mB-), gal λ(DE3),
pLysS, CmR
Novagen
FT1 / pLysS
BL21(DE3), ∆(ptsHIcrr), KmR, [pLysS,
CmR]
F–, ompT, hsdSB(rB- mB-), pRARE2, CmR
(Parche et al., 1999)
E. coli
Rosetta2
Novagen
22
MATERIALS AND METHODS
Strain
Genotype
Reference
168
trpC2
Bacillus Genetic Stock Centre
WH482
trpC2, amyE::(ackA`-lacZ cat)
(Sprehe et al., 2007)
WH483
trpC2, amyE::(ackA`-lacZ cat), ∆ccpA,
aphA3
(Sprehe et al., 2007)
WH649
trpC2, ∆ccpA, aphA3, KmR
(Sprehe et al., 2007)
WH1012
WH649, pWH933, pWH940
Bartholomae 2008
QB5223
trpC2, ptsH1
(Martin-Verstraete et al., 1995)
WH1124
WH649, pWH933, pGP382
This work
WH1125
QB5223, ∆ccpA, aphA3, pWH933,
pWH940, KmR, ErmR, TcR
This work
WH1129
WH1012, codY’-3 × Flag, SpecR
This work
B. subtilis
4.2 Media, buffers and solution
Media, buffers, and solutions were prepared with Millipore water or deionized water and autoclaved
for 20 min at 121°C and 2bar. Heat labile substances were dissolved and filtered through a sterile filter
(0.2 µM).
4.2.1
Media
LB
1 % (w/v) tryptone, 0.5 % (w/v) yeast extract, 1 % (w/v) NaCl
TYH medium
2 % (w/v) tryptone, 1 % (w/v) yeast extract, 0.5 % (w/v) NaCl,
4.2 mM HEPES, 8.3 mM MgSO4, adjust with KOH pH 7.2-7.4
CSK
1 × C-salts, 0.6 % (w/v) sodiumsuccinate, 0.8 % (w/v)
potassiumglutamate, 250 µM L-tryptophan, 1 × CAF, 1 × III’ salts
CSK with glucose
1 × C-salts, 0.6 % (w/v) sodiumsuccinate, 0.8 % (w/v)
potassiumglutamate, 250 µM L-tryptophan,1 × CAF, 1 × III’ salts,
0.5 % (w/v) glucose
MNGE
1 × MN-medium, 1 % (w/v) glucose, 0.2 % (w/v) potassiumglutamate,
50 µg/ml L-tryptophan, 0.5 × CAF, 3 µM Mg2SO4
Expression mix
2.5 % (w/v) yeast extract, 2.5 % (w/v) CAA, 0.25 mg/ml L-tryptophan
SOB
2 % (w/v) tryptone, 0.5 % (w/v) yeast extract, 8.5 mM NaCl, 2 mM KCl
4.2.1.1 Stock solutions for minimal media
10 × C salts
250 mM (NH4)2SO4, 700 mM K2HPO4, 260 mM KH2PO4
100 × III’ salts
50 mM MgSO4, 1 mM MnSO4
100 × CAF
2.2 mg/ml ferric ammonium citrate
10 × MN medium
440 mM KH2PO4, 600 mM K2HPO4, 30 mM Na3citrate, 150 mM (NH4)2SO4
MATERIALS AND METHODS
23
4.2.1.2 Additives
Agar
For preparation of LB plates, media was supplemented with 1.5 % (w/v) agar and all components were
autoclaved together.
Antibiotics
Antibiotics were prepared as 1,000 fold stock solutions and stored at -20°C. After cooling the media to
about 50°C, the antibiotics were added to the concentration indicated below.
Antibiotic
Application
Solvent
Final concentration
Ampicillin
Chloramphenicol
Chloramphenicol
Erythromycin
Kanamycin
Kanamycin
Spectinomycin
Streptomycin
Tetracycline
E. coli
E. coli
B. subtilis
B. subtilis
E. coli
B. subtilis
B. subtilis
E. coli
B. subtilis
ddH2O
70 % EtOH
70 % EtOH
70 % (v/v) EtOH
ddH2O
ddH2O
ddH2O
ddH2O
70 % (v/v) EtOH
100 µg/ml
25 µg/ml
5 µg/ml
2.5 µg/ml
50 µg/ml and 100 µg/ml
15 µg/ml
100 µg/ml
100 µg/ml
12.5 µg/ml
IPTG
IPTG was dissolved in water as 1 M stock solution, sterile filtered and stored at -20°C. The final
concentration was 1 mM.
Sugars
Sugars were prepared as 20 % (w/v) stock solutions in H2O, sterile filtered and added to the medium
as indicated.
Branched chain amino acids
To ensure comparable growth of different B. subtilis deletion strains, CSK minimal medium was
supplemented with 0.25 ‰ (w/v) L-isoleucine, 0.5 ‰ (w/v) L-leucine, 0.4 ‰ (w/v) L-valine, and
0.2 ‰ (w/v) L-methionine.
24
4.2.2
MATERIALS AND METHODS
Buffers
4.2.2.1 General buffers
TE buffer
10 mM Tris/HCl pH 8,0, 0.1 mM EDTA
TB buffer
10 mM Pipes, 15 mM CaCl2, 250 mM KCl, after adjustment of
pH 6.7 with KOH addition of 55 mM MnCl2; sterilization by
filtration
SET buffer
75 mM NaCl, 20 mM Tris/HCl pH 7.5, 25 mM EDTA pH 8.0
5 × Z-buffer
300 mM NaHPO4, 200 mM NaH2PO4, 50 mM KCl, 5 mM MgCl2,
250 mM β-mercaptoethanol
10 × cell disruption buffer
500 mM Tris/HCl pH 7.5; 2 M NaCl
SBT buffer
50 mM Tris/HCl pH 7.5, 200 mM NaCl, 2 mM DTT
10 × R-buffer
200 mM Tris/HCl pH 7.5, 50 mM MgCl2, 10 mM DTT
Crosslinking-solution
4 % (w/v) paraformaldehyde in 1 × PBS, only dissolves at 60°C
Lysozyme mix
2 × TE, 20 mg/ml lysozyme
4.2.2.2 Buffers and solutions for agarose gel electrophoresis
Agarose gel
0.8 – 2.5 % (w/v) agarose in 1 × TAE
50 × TAE buffer
2 M Tris, 50 mM EDTA, 1 M acidic acid; pH 8.3
DNA loading buffer
0.1 % (w/v) bromphenolblue, 0.1 % (w/v) xylencyanol,
50 % (v/v) glycerol in 1 × TAE
4.2.2.3 Buffers and solutions for Laemmli polyacrylamide gel electrophoresis
Native PAA gel
7.5 % (v/v) Rotiphorese Gel 40 % (acrylamide / bisacrylamide:
37.5:1), 85 mM Tris/HCl pH 8.8, 100 mM Tris/boric acid pH 8.8,
400 µl 10 % (w/v) APS, 5 µl TEMED
10 × native running buffer
192 mM glycine, 50 mM Tris, pH 8.5 – 8.8
5 × native loading buffer
50 % (v/v), glycerol, 1.5 % (w/v) bromphenolblue
SDS stacking gel
4 % (v/v) acrylamide (39:1), 85 mM Tris/HCl pH 6.8, 0.1 % (w/v)
SDS, 400 µl 10 % (w/v) APS, 5µl TEMED
SDS separation gel
7.5 – 12.5 % (v/v) Rotiphorese Gel 40 %
(acrylamide / bisacrylamide: 37.5:1), 85 mM Tris/HCl pH 8.8,
100 mM Tris/boric acid pH 8.8, 0.1 % (w/v) SDS, 400 µl
10 % (w/v) APS, 5 µl TEMED
5 × SDS loading buffer
15 % (v/v) β-mercaptoethanol, 5 % (v/v) SDS, 50 % (v/v) glycerol,
1.5 % (w/v) bromphenolblue
10 × SDS running buffer
192 mM glycine, 50 mM Tris, pH 8.5, 1 % (w/v) SDS
MATERIALS AND METHODS
25
4.2.2.4 Buffers and solutions for Schaegger polyacrylamide gel electrophoresis
SDS separating gel (15 %)
5.6 ml Rotiphorese Gel 40 % (acrylamide / bisacrylamide: 37.5:1),
7.5 ml gel buffer, 8.1 g urea, 1.9 ml water, 80 µl 10 % (w/v) APS,
10 µl TEMED
SDS stacking gel (4.5 %)
0.8 ml Rotiphorese Gel 40 % (acrylamide / bisacrylamide: 37.5:1),
1.9 ml gel buffer, 4.75 ml water, 80 µl 10 % (w/v) APS,
8 µl TEMED
Gel buffer
3 M Tris, 1 M HCl, 0.3 % (w/v) SDS
5 × loading buffer
20 % (w/v) SDS, 60 % (w/v) glycerol, 250 mM Tris/HCl pH 6.8,
10 % (v/v) mercaptoethanol, 0.01 % (w/v) serva blue G
10 × cathode buffer
1 M Tris, 1 M tricine, 1 % (w/v) SDS
10 × anode buffer
2 M Tris/HCl pH 8.9
4.2.2.5 Buffers for staining of PAA gels with coomassie brilliant blue
Wash solution
45 % (v/v) methanol, 10 % (v/v) acidic acid in H2O
Staining solution
0.5 % (v/v) Coomassie brilliant blue R250, 10 % (v/v) acidic acid,
45 % (v/v) methanol, in H2O
De-staining solution
10 % (v/v) acidic acid
4.2.2.6 Buffers for protein purification of SPINE samples
Wash buffer
100 mM Tris-HCl pH 8, 150 mM NaCl, 1 mM EDTA
Elution buffer
100 mM Tris-HCl pH 8, 150 mM NaCl, 1 mM EDTA,
2.5 mM Desthiobiotin
Regeneration-buffer
100 mM Tris-HCl pH 8, 150 mM NaCl, 1 mM EDTA,
1 mM HABA
4.2.2.7 Buffers for protein purification
Nickel affinity chromatography
His A
50 mM NaH2PO4, 500 mM NaCl, 100 mM imidazole
His B
50 mM NaH2PO4, 500 mM NaCl, 500 mM imidazole
Regeneration-buffer
100 mM EDTA, 100 mM NiS04, 300 mM NaCl
Strep affinity chromatography
Strep A
100 mM Tris, pH 8.0, 150 mM NaCl, 1 mM EDTA
Strep B
100 mM Tris-HCl pH 8, 150 mM NaCl, 1 mM EDTA,
2.5 mM Desthiobiotin
Regeneration-buffer
100 mM Tris-HCl pH 8, 150 mM NaCl, 1 mM EDTA,
1 mM HABA
26
MATERIALS AND METHODS
Avidin affinity chromatography
Avi A
50 mM Tris/HCl pH 8.0, 150 mM NaCl
Avi B
50 mM Tris/HCl pH 8.0, 150 mM NaCl, 5 mM D-biotin
Regeneration-buffer
10 % (v/v) acetic acid, 100 mM NaPO4
Size Exclusion chromatography
HBSE buffer
10 mM HEPES, pH 7.4, 150 mM NaCl, 3 mM EDTA
Regereration-buffer
0.5 mM NaOH
4.2.2.8 Buffers for silver staining
Fixing solution
50 % (v/v) Ethanol, 12 % (v/v) acetic acid, 0.1 % (v/v)
formaldehyde 37 %, ad Millipore
Sensitizer
0.02 % (w/v) Na2S2O3, ad Millipore
Silver nitrate solution
0.2 % (w/v) AgNO3, 0.1 % (v/v) formaldehyde 37 %, ad Millipore
Developer
6 % (w/v) Na2CO3, 2 % (v/v) sensitivizing solution, 0.05 % (v/v)
formaldehyde 37 %, ad Millipore
4.2.2.9 Buffers for immunoblotting
Transfer buffer
192 mM glycine, 50 mM Tris, 3 % (v/v) isopropanol, pH 8.5 – 9.0
10 × PBS pH 7.4
580 mM Na2HPO4, 170 mM NaH2PO4, 680 mM NaCl
Blocking solution
5 % (w/v) skim milk powder, 0.1 % (v/v) Tween 20 in 1 × PBS
Wash buffer
1 × PBS, 0.1 % (v/v) Tween 20
Wash solution
Blocking solution/wash buffer: 1:1
4.3 Methods
4.3.1
General methods
Table 4.9: General methods
Methods
Reference
Agarose gel electrophoresis
(Sambrook et al., 1989)
Determination of DNA concentrations
(Sambrook et al., 1989)
Determination of protein concentrations
(Bradford, 1976)
DNA sequencing
(Sanger et al., 1977)
Coomassie staining of protein gels
Ethidiumbromide staining of DNA gels
(Sambrook et al., 1989)
Polyacrylamide gel electrophoresis
(Laemmli, 1970; Schägger & von Jagow, 1987)
Commercially available systems were used as described by the manufactures.
MATERIALS AND METHODS
4.3.2
27
Growth of bacteria
Cells of E. coli and B. subtilis were grown in tubes or baffled flasks for 7 – 12 h at 28°C or 37°C by
vigorous shaking using LB as complex medium or CSK or CSK with glucose medium as minimal
media. Inoculation was performed either by supplying freshly spread colonies or 1 / 100 volume of an
over night culture. Growth was monitored by measuring the OD600.
4.3.2.1
Preparation and transformation of chemically competent E. coli
Chemically competent E. coli cells were prepared as described by Inoue et al. (Inoue et al., 1990). For
this, 4 ml LB medium was inoculated with E. coli cells and grown for 8 h at 37°C. 1 ml of this culture
was used to inoculate 250 ml SOB medium for overnight growth at 20°C in a 1 l flask. On the next
day, when the culture was grown to OD600 ~ 0.4 the cells were chilled on ice for 10 min. Then, the
cells were harvested by centrifugation (4,000 × g, 10 min, 4°C). The cell pellet was resuspended in
80 ml ice-cold TB buffer and centrifuged again (4,000 × g, 10 min, 4°C). After resuspending in 6.4 ml
TB buffer, 400 µl DMSO was carefully added and the cells were incubated for 10 min on ice. Aliquots
of 100 µl were transferred to pre-cooled reaction tubes and subsequently frozen in liquid nitrogen and
stored at -80°C.
For transformation, a 100 µl aliquot of chemically competent E. coli cells was thawed on ice and
plasmid DNA was added. Cells were incubated for 30 min on ice. After heat shock of the cells at 42°C
for 60 sec 400 µl LB medium was added and the cells were grown for 1 h at 37°C. After that, 200 µl of
the cell suspension were spread on selective media and the plates were incubated at 37°C.
4.3.2.2
Preparation and transformation of naturally competent B. subtilis
Naturally competent B. subtilis cells were prepared as described by Kunst et al. (Kunst & Rapoport,
1995). For this, an over night culture of the respective B. subtilis strain was inoculated in 10 ml
MNGE + 1 ml CAA to an OD600 = 0.1 and grown at 37°C. At an OD600 of 1.3 – 1.8 about 11 ml
MNGE medium was added and the cells were grown for another hour at 37°C. Then the competent
cells were harvested by centrifugation at 5,000 rpm for 5 min at RT and the supernatant was decanted
into a sterile greiner tube. The cells were resuspended in 2.6 ml of the supernatant and were adjusted
to 10 % (v/v) glycerol. Aliquots of 300 µl were transferred to reaction tubes and subsequently frozen
in liquid nitrogen and stored at -80°C.
For transformation, a 300 µl aliquot of naturally competent B. subtilis cells were thawed in a waterbath
at 37°C and a solution of 1.7 ml 1 × MN, 43 µl 20 % (v/v) glucose, 34 µl 1 M MgSO4 was added.
400 µl of this cellsupension was mixed with 1 – 5 µg DNA and grown under vigorous shaking at
37°C. A sample of cells transformed without any DNA served as a negative control. After 30 min,
100 µl expression mix was added and the cells were shaken another 60 min at 37°C, before they were
spread on suitable selection plates and incubated at 37°C.
28
4.3.3
MATERIALS AND METHODS
Nucleic acid purification and modification
4.3.3.1 Preparation of chromosomal DNA from B. subtilis
Preparation of chromosomal DNA was performed using phenol chloroform extraction. B. subtilis
strains were grown in 20 ml LB medium overnight. On the next day, cultures were centrifuged
(4,000 × g, 10 min, 4°C) and the pellet was resuspended in 5 ml SET buffer with 100 µl (50 µg/ml)
lyzozyme. After incubation for at least 1 h 600 µl 10 % (w/v) SDS and 150 µl proteinase K
(20 mg/ml) were added and this mixture was incubated at 55°C for 2 h and inverted occasionally.
Then, 2 ml 5 M NaCl was added and mixed by carefully inverting until a white precipitate occurred.
Prior to the addition of 5 ml chloroform, the mixture was put on ice to cool down. After shaking for
30 min at RT the solution was centrifuged at 4,000 × g and 4°C for 15 min. The supernatant
containing the DNA was separated from the organic phase by transferring into a fresh reaction tube
and 0.6 fold volume of isopropanol was added. After carefully inverting, the precipitated DNA was
transferred into a sterile tube, washed twice with 70 % (v/v) ethanol, dried in a Speed Vac and
resolved in 500 µl TE buffer.
4.3.3.2 Polymerase chain reaction (PCR)
The selective amplification of specific DNA fragments was performed by polymerase chain reaction
(PCR) (Mullis et al., 1986). Either genomic DNA, plasmid DNA or a cell suspension, which had
previously been incubated at 95°C for 5 min, were used as template. Primers were supplied by MWG
(Ebersberg) and diluted in H2O to a final concentration of 100 pmol/µl. Depending on the primer
sequence the annealing temperature was calculated. For breaking of hydrogen bonds between guanine
and cytosine 4°C were estimated and for adenine and thymine 2°C. For amplification Phusion DNA
polymerase (Finnzymes, Finland) was used as recommended by the suppliers. The PCR reaction mix
contained: 50 – 100 ng DNA, 10 pmol/µl 3′-Primer, 10 pmol 5′-Primer, 0.5 mM dNTP mixture*,
1 × HF buffer and 2 U Phusion polymerase. The PCR was performed with a Thermocycler. First of all,
template DNA was denatured by an initial incubation at 95°C for 5 min. After that, the following steps
were repeated 30 times: double-stranded DNA was denatured for 30 sec at 95°C. Depending on the
sequence of the primers used, annealing was performed for 30 sec at 56°C to 68°C. At a temperature
of 72°C double-stranded DNA was synthesized by the polymerase. This step was carried out for
30 sec per kbp. By final incubation at 72°C for 5 min, it was ensured that the synthesis of fragments
was completed.
*dNTP mixture: 10 mM dATP, 10 mM dTTP, 10 mM dCTP, 10 mM dGTP
4.3.3.3 Colony PCR
Colony PCR was applied for the screening of transformants. Colonies were directly picked from the
selection plates and employed in the PCR reaction with Ready-To-Go-Taq beads (GE Healthcare,
Munich) for B. subtilis and Taq Polymerase (NEB, Frankfurt / Main) for E. coli with suitable primers
according to the manufacturer instructions.
MATERIALS AND METHODS
29
4.3.3.4 Restriction
DNA restriction was carried out in 1 × restriction buffer as recommended by the manufacturer. The
amount of enzyme and the incubation time varied with the amount of DNA and with the reaction
volume.
4.3.3.5 Dephosphorylation
To dephosporylate Vector DNA the restriction enzymes were inactivated by 15 min incubation at
65°C. The DNA was incubated with 1 U shrimp alkaline phosphatase per 50 ng DNA in the presence
of 1 × dephosphorylation buffer. After 15 min incubation at 37°C the enzyme was inactivated for
20 min at 65°C.
4.3.3.6 Ligation
Ligation of DNA fragments was performed using T4-DNA Ligase in the presence of 1 × ligation
buffer and 1 mM ATP. Normally, 20 – 50 ng of restricted Vector DNA and 20 – 100 ng of DNA
fragment were ligated for 20 min at RT.
4.3.3.7 DNA gel electrophoresis
DNA gel electrophoresis was carried out for analytical- and preparative separation of DNA fragments
using 1 % (w/v) agarose gels. DNA probes were mixed with DNA loading buffer and loaded onto the
gel. Fragments were separated at 80 – 120 V and subsequently stained for 15 min in ethidium bromide
solution. After short washing in water the gel was scanned for further documentation using the BioPrint Video-Geldocu-system (Peqlab, Erlangen).
PEQ Gold “Leitermix” (Peqlab, Erlangen), DNA fragments in kbp:
10 / 8 / 6 / 5 / 4 / 3.5 / 3 / 2.5 / 2 / 1.5 / 1.2 / 1.031 / 0.9 / 0.8 / 0.7 / 0.6 / 0.5 / 0.4 / 0.3 / 0.2 / 0.1.
4.3.3.8 DNA Sequencing
Sequencing of DNA was performed by using the chain termination method described by Sanger et al.
(Sanger et al., 1977). As chain terminators di-desoxynucleotides labeled with fluorescent dye were
used (Big Dye Terminator Mix, Perkin Elmer, Weiterstadt). A sequencing sample was composed of
300 – 500 ng DNA, 5 pmol of an oligonucleotide primer and 3 µl Terminator Ready Reaction Mix.
Each sample was filled with H2O to a final volume of 10 µl. Sequencing reactions were carried out as
recommended by the supplier using the following PCR program:
25 ×
96°C
10 sec
56°C
5 sec
60°C
4 min
The resulting DNA fragments were precipitated as following. Firstly, to the 10 µl reaction mix 90 µl
H2Obidest. and 10 µl 3 M Na-acetate (pH 5.0) were added and mixed by vortexing. Secondly, the
30
MATERIALS AND METHODS
solution was mixed with 250 µl 96 % (v/v) ethanol and centrifuged for 15 min at 13,000 rpm. Then the
pellet was washed with freshly prepared 70 % (v/v) ethanol prior to centrifugation for 5 min at
13,000 rpm. Finally, the pellet was dried in a Speed-Vac, solved in 12 µl formamide, investigated with
the ABI PRISM Genetic Analyser 310 (Applied Biosystems, Freiburg) and sequence data were
evaluated with the Seqman software (DNAstar, Madison, USA).
For further sequence analyses, samples containing 30 µl of plasmid DNA (30 – 100 ng/µl) and 10 µl of
an appropriate sequencing primer (10 pmol/µl) were sent to GATC Biotech (Konstanz). Sequence data
were also analyzed using the Seqman software (DNAstar, Madison, USA).
4.3.4
Protein purification and characterization
4.3.4.1 SDS polyacrylamide gel electrophoresis (PAGE)
Gels (5 – 15 % PAA) with a width of 1 mm were prepared as described by (Laemmli, 1970) (for
protein purification) and by (Schägger & von Jagow, 1987) (for silver staining and immunoblotting).
All samples were mixed with the appropriate SDS-loading buffer and electrophoresis was carried out
at 100 – 150 V. After electrophoresis the gels were fixed for 20 min in wash solution, stained for
30 min in staining solution and destained for several hours in destaining solution.
PEQ Gold protein marker II, Peqlab, Erlangen, molecular weight in kDa:
200 / 150 / 120 / 100 / 90 / 70 / 60 / 50 / 40 / 30 / 25 / 20 / 15 / 10.
4.3.4.2 Silver staining of PAA gels
PAA gels were silver stainded with the following procedure which is compatible for mass
spectrometric analysis. After electrophoresis all following steps were performed on a horizontal shaker
with gentle shaking.
At least one hour or over night
Fixing solution
3 × 20 min
50 % (v/v) ethanol
1 min
Sensitizer
3 × 20 sec
Millipore water
15-25 min
Silver nitrate solution
3 × 20 sec
Millipore water
1-30 min (until staining is sufficient)
Developer
2 × 20 sec
Millipore water
10 min
Stop solution
2 × 20 sec
Millipore water
The stained gels were shrink-wrapped and stored at 4 – 8°C.
MATERIALS AND METHODS
31
4.3.4.3 Immunoblot analysis
For immunoblot analyses of CcpA, HPr, RpoA and CodY-Flag, B. subtilis crude extracts and elution
fractions from SPINE were separated by SDS-PAGE. The gel was washed for 30 min in transfer
buffer; the PVDF membrane was activated for 15 min in methanol and subsequently washed another
15 min in transfer buffer. The proteins were transferred to the membrane by electroblotting at 15 V for
45 min. Afterwards the membrane was treated as followed:
2 × 10 min
wash buffer
3 h or over night
blocking solution
1h
incubation with primary antibody (the primary antibodies were diluted
1 : 1 in wash buffer : wash solution as followed: anti-CcpA 1 : 5,000, antiHPr 1 : 5,000, anti Crh 1 : 10,000, anti-RpoA 1 : 1,000 and anti-Flag
1 : 2,000)
3 × 10 min
wash solution
1h
incubation with secondary antibody (anti-rabbit IgG or anti-mouse IgG
1 : 10,000 in wash buffer : wash solution = 1 : 1)
3 × 10 min
wash solution
5 – 10 min
luminescence reaction with ECL substrate (solution A : B = 1 : 1)
(Thermo Scientific, Schwerte)
CcpA and HPr were detected with rabbit polyclonal antisera raised against B. subtilis CcpA and HPr,
respectively. DnaK, RpoA, CodY-Flag were dected with mouse monoclonal antibodies. The
antibodies were visualized with the corresponding secondary antibodies anti-rabbit IgG-HRP and antimouse IgG-HRP, respectively, with the ECL detection system (GE Healthcare, Munich).
4.3.4.4 Growth of bacteria for protein overexpression
For overexpression 2 × 1 l LB broth were inoculated with an over night culture from cells of the
corresponding E. coli overexpression strain to a start OD600 of ~ 0.05 and the cultures were grown at
37°C under agitation. Growth was monitored by measuring the optical density at 600 nm. For
overproduction of recombinant proteins, the bacteria were grown to OD600 = 0.4 – 0.6 and expression
was induced addition of IPTG to a final concentration of 1 mM. After further incubation for 3 – 4 h
the cells were harvested by centrifugation (5,000 × g, 10 min, 4°C).
4.3.4.5 Sonication
Crude protein extracts were obtained by sonification of cells with a Labsonic sonifier. Usually, cells
were resuspended in a suitable volume of cell disruption buffer (for immunoblotting), 10 mM
Tris/HCl pH 7.5 or His A (for protein preparation). Each buffer contained 1 × complete (Roche,
Mannheim). Lysates were obtained by sonication with a tip size-dependent of the applied volume
(small tip for volumes < 5 ml, large tip for volumes > 5 ml). Three to five 30 sec pulses of 45 W were
performed. During and between each pulse the cell containing tube was cooled on ice. Soluble
proteins were obtained after centrifugation at 4°C for 30 min at 20,000 rpm.
32
MATERIALS AND METHODS
4.3.4.6 Purification of CcpA(His)6 from B. subtilis
C-terminally His6-tagged CcpA, from B. subtilis was expressed in E. coli FT1 pWH653 (Seidel et al.,
2005). A pellet resulting from two liters of overexpression culture was resuspended in 40 ml His A
buffer (80 mM Imidazol with 1 × complete). After cell disruption by sonication 10 mg/ml DNaseI and
5 mg/ml RNase A were added, and the lysate was incubated for 20 min on ice, before insoluble cell
components were separated by centrifugation (20,000 rpm, 40 min, 4°C). Purification was achieved by
Ni-affinity chromatography on a 1 ml Ni2+-NTA column (His trap 1 ml, GE Healthcare). After a
washing step with 20 ml His A buffer, bound CcpA(His)6 was eluted by a linear gradient (25 ml,
0 – 100 % His B), followed by a batch elution (10 ml, 100 % His B). CcpA(His)6 eluted at about
30 – 40 % His B buffer. CcpA(His)6 containing fractions were concentrated to a maximum volume of
5 ml and further purified by size exclusion chromatography on Superdex G200 column, using an
ÄKTA prime chromatograph (GE Healthcare, Munich).
4.3.4.7 Purification of CodY(His)6 from B. subtilis
His-tagged CodY from B. subtilis was overexpressed in E. coli BL21(DE3) pLysS / pWH2177. The
pellets of 2 l culture were resuspended in 50 ml His A buffer (120 mM Imidazol) and 1 × complete
(Roche, Mannheim), disrupted by ultrasonication and incubated with 5 µg/ml RNaseA and 10 µg/ml
DNaseI for 20 min on ice before centrifugation for 30 min at 48,000 × g at 4°C. His-tagged CodY, was
purified by Ni-affinity chromatography on a 5 ml Ni2+-NTA column (His trap 5 ml, GE Healthcare).
After a washing step with at least 50 ml His A, bound His-tagged CodY was eluted by a linear
gradient (50 ml, 0 – 100 % His B), followed by a batch elution (30 ml, 100 % His B). CodY eluted at
about 25 – 35 % His B buffer. CodY containing fractions were concentrated to a maximum volume of
5 ml and further purified by size exclusion chromatography on a Superdex G75 column, using an
ÄKTA prime chromatograph (GE Healthcare, Munich).
4.3.4.8 Purification of RpoA(His)6 from B. subtilis
His-tagged RpoA from B. subtilis was overexpressed in E. coli BL21(DE3) pLysS / pWH2175. The
pellets of 2 l culture were resuspended in 50 ml His A buffer (80 mM Imidazol) and 1 × complete
(Roche, Mannheim), disrupted by ultrasonication and incubated with 5 µg/ml RNaseA and 10 µg/ml
DNaseI for 20 min on ice before centrifugation for 30 min at 48,000 × g at 4°C. His-tagged RpoA, was
purified by Ni-affinity chromatography on a 1 ml NTA column (His trap 5 ml, GE Healthcare). After a
washing step with at least 25 ml His A, bound His-tagged RpoA was eluted by a linear gradient
(30 ml, 0 – 100 % His B), followed by a batch elution (10 ml, 100 % His B). RpoA eluted at about
25 – 65 % His B buffer. RpoA containing fractions were concentrated to a maximum volume of 5 ml
and further purified by size exclusion chromatography on a Superdex G200 column, using an ÄKTA
prime chromatograph (GE Healthcare, Munich).
4.3.4.9 Purification of αNTD(His)6 from B. subtilis
His-tagged αNTD from B. subtilis was overexpressed in E. coli BL21(DE3) pLysS / pWH2176. The
pellets of 2 l culture were resuspended in 50 ml His A buffer (120 mM Imidazol) and 1 × complete,
disrupted by ultrasonication and incubated with 5 µg/ml RNaseA and 10 µg/ml DNaseI for 20 min on
MATERIALS AND METHODS
33
ice before centrifugation for 30 min at 48,000 × g at 4°C. His-tagged αNTD, was purified by
Ni-affinity chromatography on a 5 ml Ni2+-NTA column (His trap 5 ml, GE Healthcare). After a
washing step with at least 50 ml His A, bound His-tagged αNTD was eluted by a linear gradient
(50 ml, 0 – 100 % His B), followed by a batch elution (30 ml, 100 % His B). CodY eluted at about
20 – 35 % His B buffer. αNTD containing fractions were concentrated to a maximum volume of 5 ml
and further purified by size exclusion chromatography on a Superdex G75 column, using an ÄKTA
prime chromatograph (GE Healthcare, Munich).
4.3.4.10 Purification of αCTD(His)6 from B. subtilis
N-terminally (His)6-tagged αCTD was overproduced in E. coli Rosetta2 pET28a- αCTD (Nakano et
al., 2010). The pellet of 1 l culture was resuspended in 25 ml His A buffer (120 mM Imidazol) and
1 × complete, disrupted by ultrasonication and incubated with 5 µg/ml RNaseA and 10 µg/ml DNaseI
for 20 min on ice before centrifugation for 60 min at 48,000 × g at 4°C. αCTD(His)6, was purified by
Ni-affinity chromatography on a 1 ml NTA column (His trap 1 ml, GE Healthcare). After a washing
step with at least 50 ml His A, bound His-tagged αNTD was eluted by a linear gradient (30 ml,
0 – 100 % His B), followed by a batch elution (10 ml, 100 % His B). Due to the lack of any tryptophan
and other aromatic residues, hardly any peak in the elution profile for αCTD was observed.
Nevertheless, αCTD actually eluted at about 60-75 % His B buffer and was clearly detected in SDSPAGE analysis. αCTD containing fractions were concentrated to a maximum volume of 5 ml and
further purified by size exclusion chromatography on a Superdex G75 column, using an ÄKTA prime
chromatograph (GE Healthcare, Munich).
4.3.4.11 Purification of HPr seryl phosphorylated form from B. subtilis
C-terminally His6-tagged CcpA, from B. subtilis was expressed in E. coli FT1 pWH466 as described
(Seidel et al., 2005). Cells of 1 l culture were harvested by centrifugation, resuspended in buffer A and
disrupted by sonication. The crude lysate was incubated with 5 µg/ml RNaseA and 10 µg/ml DNaseI
on ice for 20 min and after centrifugation for 30 min at 48,000 × g at 4°C the soluble proteins were
obtained. Subsequently the proteins were prepurified by heat denaturation for 20 min at 70°C as HPr is
heat stable. The precipitate was then separated from the soluble protein by centrifugation for 30 min at
48,000 × g at 4°C. For in vitro serine phosphorylation a kinase extract from E. coli DH5α p4813 was
added to the heat denatured crude lysates. The reaction mixtures were incubated for 20 min at 37°C
and then immediately loaded on a Sepharose Q FF column (HiPrep Q FF, 20 ml, GE Healthcare,
Munich) for anion exchange chromatography on an ÄKTA prime chromatograph. After washing with
buffer A, proteins were eluted using a linear gradient (50 ml, 0 – 50 % buffer B), followed by a batch
elution (30 ml, 100 % buffer B). Suitable protein fractions were then concentrated in a pressure
dialysis device using YM3 membranes and then purified by size exclusion chromatography on a
Superdex G75 column on an ÄKTA prime chromatograph (GE Healthcare, Munich). Pure protein
fractions were concentrated in Centriprep concentrators and stored at 4°C in HBS-EP buffer.
34
MATERIALS AND METHODS
4.3.4.12 Purification of Strep-tagged CcpA
N-terminally Strep-tagged CcpA, from B. subtilis was expressed in E. coli BL21(DE3) pWH2375. For
overexpression 1 l LB medium was inoculated with 5 ml of an overnight culture. The cells were grown
at 37°C to an OD600 of 0.4 – 0.6 and then gene expression was induced by addition of 200 µg ATC
(Anhydrotetracycline) and further incubation 3 – 4 h. Cells of 1 l culture were harvested by
centrifugation at 5,000 × g for 10 min. For purification the pellet was resuspended in 25 ml Strep A
buffer and 1 × complete (Roche, Mannheim), disrupted by ultrasonication and incubated with 5 µg/ml
RNaseA and 10 µg/ml DNaseI for 20 min on ice before centrifugation for 30 min at 48,000 × g at 4°C.
Strep-tagged CcpA was purified by affinity chromatography on a Streptactin column (1 ml, GE
Healthcare, Munich). After a washing step with 30 ml Strep A, bound Strep-tagged CcpA was eluted
by a batch elution (20 ml, 100 % Strep B). Protein containing fractions were concentrated to a
maximum volume of 5 ml and further purified by size exclusion chromatography on a Superdex G200
column using an ÄKTA prime chromatograph (GE Healthcare, Munich). Pure protein fractions were
concentrated and stored at -20°C in HBSE buffer and 50 % glycerol.
4.3.4.13 Purification of Avi-tagged CcpA
N or C-terminally Avi-tagged CcpA, from B. subtilis was expressed in E. coli AVB101 / pBirAcm
pWH2369 or AVB101/pBirAcm pWH2370, respectively. For overexpression 1 l TYH medium
supplemented with 20 ml 20 % (w/v) glucose was inoculated with 5 ml of an overnight culture. The
cells were grown at 37°C to an OD600 of 0.7 and then gene expression was induced by addition of
10 ml of 5 mM biotin solution* (50 µm final) and IPTG to a final concentration of 1.5 mM and further
incubation 3 – 4 h. Cells of 1 l culture were harvested by centrifugation at 5,000 × g for 10 min. For
purification the pellet was resuspended in 25 ml Avi A buffer and 1 × complete (Roche, Mannheim),
disrupted by ultrasonication and incubated with 5 µg/ml RNaseA and 10 µg/ml DNaseI for 20 min on
ice before centrifugation for 30 min at 48,000 × g at 4°C. AviCcpA was purified by affinity
chromatography on a monomeric Avidin column (Soft link release Avidin resin; Promega,
Mannheim). The use of a monomeric avidin allows biotinylated molecules to be eluted using mild,
non-denaturing conditions. Dissociation of the avidin tetramer into its monomeric subunits leads to a
conformational change within each subunit. This is accompanied by an increase in the KD for biotin to
about 10-7 M, however, the high specificity of binding for biotinylated molecules is retained. This
equilibrium constant readily permits elution under mild conditions, typically using competitive elution
with free biotin. Free monomeric avidin subunits readily reassociate to form tetramers. To preserve
these lower affinity binding sites, avidin monomers must be immobilized on a matrix such that the
subunits are spatially separated.
After a washing step with 30 ml Avi A, bound AviCcpA was eluted by a linear gradient (50 ml,
0 – 100 % Avi B), followed by a batch elution (30 ml, 100 % Avi B). Protein containing fractions
were concentrated to a volume of 5 ml and further purified by size exclusion chromatography on a
Superdex G200 column using an ÄKTA prime chromatograph (GE Healthcare, Munich). Pure protein
fractions were concentrated and stored at -20°C in HBSE buffer and 50 % glycerol.
*Biotin solution: 12 mg D-biotin in 10 ml of warm 10 mM bicine buffer (pH 8.3); filtersterilizing with
a 0.2 µm filter.
MATERIALS AND METHODS
35
4.3.4.14 Determination of protein concentration
Protein concentrations were determined spectrophotometrically using Bio-Rad protein assay. For a
standard a solution of BSA was used, which had been adjusted to a concentration of 200 µg/ml.
4.3.4.15 Storage and dialysis of proteins
CcpA(His)6, RpoA(His)6, αNTD(His)6, αCTD(His)6 and CodY(His)6 preparations were adjusted to
50 % (w/v) glycerol, 1 × HBSE and stored at -20°C. For SPR analyses all proteins were freshly
purified except αCTD(His)6 which was dialyzed 3 – 4 times against the 1,000 fold volume of HBSEbuffer under slow stirring at 4°C. Slide-A-Lyzer Dialysis units were used for dialysis of small volumes
(up to 100 µl).
4.3.4.16 Surface plasmon resonance measurements (SPR)
SPR measurements with CcpA, CodY, αCTD or αNTD each from B. subtilis, were performed on a
BIAcoreX instrument at 25°C (BIAcoreX, Uppsala, Sweden). For the analysis of protein–protein
interactions αNTD, αCTD or CodY, respectively, were immobilized by amine coupling on the
carboxylated dextran matrix of a CM5 sensorchip (Biacore AB) in flowcell 2. After activation of the
carboxylated dextran matrix of a CM5 chip by injecting 35 µl of a mixture containing 50 mM NHS
(N-hydroxysuccinimide) and 200 mM EDC (1-ethyl-3-(3-dimethylaminoproply)carbodiimide) the
proteins were injected at about 0.5 µM to 1 µM concentrations in 10 mM sodiumacetate, pH 4.5 and
about 3,000 – 4,500 RU of the protein of interest were coupled. After coupling residual activated
carboxyl groups were deactivated by injection of 1 M ethanolamine hydrochloride⁄NaOH, pH 8.5.
Flowcell 1 served as a reference and was only activated and deactivated with ethanolamine without
coupling of a ligand. HBS-EP buffer (Biacore, GE Healthcare) was used as a running buffer during
immobilization and at 25 µl/min during interaction analyses. The analyzed proteins were titrated in
50 µl to 100 µl volumes. The titrations for the kinetic measurements have been carried out at least
twice for each protein complex, CcpA–αNTD, CcpA–αCTD, CodY–αNTD and CodY–αCTD. The
equilibrium constant of CcpA–CodY was determined by Langmuir fits of plots from the steady state
response vs. the analyte concentrations from at least two different titrations.
For analyzing the protein-protein interaction the protein solutions were freshly prepared as stock
solutions in HBS-EP and adjusted to pH 7.4. To avoid bulk shift effects the proteins were supplied in
increasing concentrations both in the analyte mixture and in the running buffer.
36
4.3.5
MATERIALS AND METHODS
Proteomic analysis
4.3.5.1 Growth conditions and preparation of protein extracts
The B. subtilis strains were grown in 10 ml LB medium supplemented either with or without
1 % (w/v) glucose in 100 ml Erlenmeyer flasks at 180 rpm and 37°C. Growth was monitored by
measurement of the OD600. Cells were harvested during exponential growth phase at an OD600 of about
0.4 – 0.5 by centrifugation (10,000 rpm, 4°C, 10 min) and washed twice with ice-cold cell disruption
buffer. Bacteria were resuspended in cell disruption buffer with 1 × complete (Roche, Mannheim) and
disrupted by ultrasonication. After cell disruption, the cell lysate was cleared by centrifugation at
20,000 rpm and 4°C for 30 min. The protein concentration of the supernatant was determined by
Bio-Rad assay and the remaining soluble protein was stored at -20°C.
4.3.5.2 Strep protein interaction experiment (SPINE)
B. subtilis WH1012, WH1124, WH1125 and WH1129, encoding StrepCcpA was used for in vivo
crosslinking experiments (SPINE). After growing on selective LB plates (Km, Erm, Tc, (Spec)) 4 ml
of LB with appropriate selective markers were inoculated with one single colony of each strain over
day at 37°C until the OD600 reached ~ 0.5. Afterwards an over night culture with the appropriate
minimal medium (CSK with or without 0.5 % (w/v) glucose) was inoculated to an OD600 ~ 0.05 and
grown at 37°C. The next day 2 l flasks containing each 1 l of CSK medium with or without
0.5 % glucose were inoculated with the corresponding over night culture to a start OD600 ~ 0.02 and
grown at 37°C. When the cells entered the log phase at OD600 ~ 0.35 each culture was divided into two
parts. The first part was immediately harvested (5,000 × g, 10 min, 4°C) and served as
non-crosslinking control. For in vivo crosslinking the second part of the cells were treated with 90 ml
4 % (w/v) formaldehyde-solution for 20 min and then harvested (5,000 × g, 10 min, 4°C). Next, the
cells of both pellets were each resuspended in 10 ml washing buffer. The cells were disrupted at least
5 times by French Press using the 35 ml piston (1,000 psi). After removing the cell debris by
ultracentrifugation (45,000 rpm, 1 h, 4°C) the soluble proteins were isolated by Strep affinity
chromatography via gravimetric 500 µl Strep-Tactin® columns (IBA, Göttingen, Germany). The
bound protein complexes were washed 4 times with 1.5 ml of disruption buffer before the
Strep-tagged protein complexes were eluted with 250 µl and 3 times 500 µl elution buffer containing
2.5 mM D-desthiobiotin. After elution, the crosslinked complexes were resolved by heating the
samples for 30 min at 100°C prior to electrophoresis. The released interaction partners were visible as
additional bands next to the Strep-tagged protein.
4.3.5.3 MS analysis of SPINE eluates
To discriminate between signals of specifically crosslinked interaction partners of CcpA and coeluted
impurities, for the preparation of SPINE eluates subjected to MS analysis, cultures were grown in
CSK medium either in the presence of 14N or 15N (medium supplemented with 15N Ammonium
sulphate, L-leucine, L-isoleucine, L-methionine and L-valine, but normal 14N tryptophan) (Dreisbach
et al., 2008). SPINE was carried out with 1:1 mixtures of crosslinked 14N- and non-crosslinked 15Nlabeled cultures and vice versa. Following reduction by 2.5 mM DTT (1 h at 60°C) two technical
replicates (each 2 µg) per eluate were subjected to proteolytic digest with trypsin in a ratio of 1:25.
MATERIALS AND METHODS
37
Desalting of peptide extracts and subsequent LC-ESI-MS / MS analysis on a LTQ-FTICR mass
spectrometer (Thermo Scientific, Bremen, Germany) equipped with a nano-UPLC (Waters,
Manchester, U.K.) was performed as previously described (Hammer et al., 2011). Data sets were
searched using the Sequest algorithm rel. 2.7 (Sorcerer built 4.04, Sage-N Research Inc., Milpitas, CA,
U.S.A.) against a forward-reverse Swiss-Prot database limited to Bacillus subtilis, 4244 entries, (Barbe
et al., 2009). Tandem MS data sets were searched twice to match spectra to peptides generated from
14
N as well as 15N labeled peptides. Parent mass tolerance (MS) was set to 10 ppm and fragment mass
tolerance 1 Da. Methionine oxidation was considered as optional modification as well as 14N
tryptophane in the 15N search. Protein identification was based on at least two significant peptides
annotated in Scaffold rel. 3.3 (Proteome Software, Portland, OR) at a Peptide Prophet probability of
95 % corresponding to false positive rate <0.5 %. Quantification was planned on base of peptide signal
intensities. However, most of the proteins were only identified after crosslinking but were not
observed at all in non-crosslinked samples although the bait CcpA was obtained in comparable
amounts. Data following this pattern cannot be handled well by software packages (Census, TPP)
commonly used for quantification of labeled samples. Therefore, quantification was performed based
on the number of spectral counts obtained per protein (Old et al., 2005). Proteins were considered as
putative interaction partners of CcpA if enrichment of more 1.5 fold was observed in both crosslinked
samples analyzed in duplicate (total four data sets) and quantitative value (spectral count) was 6 in at
least one sample.
4.3.6
Enzyme assays
4.3.6.1 Bacterial two hybrid assay
The bacterial Two-Hybrid-System (BACTH) (Karimova et al., 1998) in E. coli is a genetic test system
which allows the identifcation of directly interacting proteins by the functional reconstitution of the
adenylate cyclase activity of the Bordetella pertussis adenylate cyclase, Cya. This enzyme is
composed of two complementary fragments, T25 and T18, that are both necessary to form an active
enzyme. In this assay these two cya fragments, T18 and T25, are physically separated by expression of
each fragment on vectors in an E. coli ∆cya strain, and therefore they cannot reconstitute a functional
enzyme. However, when T25 and T18 domains are fused to peptides or proteins that are able to
interact, the complex formation of these fusion proteins results in a functional complementation
between the adenylate cyclase fragments and consequently to the synthesis of cAMP. The signal
molecule cAMP binds to CAP, the catabolite activating protein, and the CAP-cAMP complex triggers
the transcriptional activation of catalytic operons such as lac. Then, bacteria are able to utilize lactose
or maltose as carbon source and can bei distinguished on indicator or selective media. Here, the
expression of the reporter gene lacZ can be analyzed in a β-galactosidase activity assay (Figure 4.1).
38
MATERIALS AND METHODS
A
B
T25
T18
C
X
T18
Y
T25
T25
ATP
T18
cAMP
cAMP
ATP
cAMP
D
cAMP
CAP
CAP
+
CAP
CAP
on
cAMP-CAP
promoter
reporter gene
(lac, mal)
Figure 4.1: Principle of the E. coli two-hybrid system (BACTH) which is based on functional
reconstiution of CyaA fragments.
(A) Schematic of the basic principle of functional in vivo complementation between the two fragments of
the catalytic domain of Bordetella pertussis adenylate cyclase. The two boxes represent the T25 and T18
fragments of the Cya protein. (B) The two fragments T25 and T18, physically separated when coexpressed
as independent polypeptides, are unable to interact and AMP synthesis does not occur. (C) If two
interacting proteins, X (blue) and Y (red), are fused to the T18 and the T25 fragments, the interaction
causes a close proximity of both Cya domains and results in functional complementation. This leads to the
conversion of ATP to cAMP. (D) Schematic of the readout of the complementation. The complementation
of T18 and T25 leads to cAMP synthesis. cAMP binds to CAP and the cAMP-CAP complex can recognize
specific promoters and activate the transcription of reporter genes, such as the lacZ gene.
The vectors, which either contain the T18 or the T25 domain of the adenylate cyclase, can be used to
fuse the interesting proteins to the N- or the C-terminus of the T18 or the T25 domain (e. g.
T25-proteinX or proteinX-T25). The name of the vectors, the name of the resulting fusion protein and
a schematic of the fusion protein is listed in Table 4.10. Proteins fused to these domains are hereinafter
named as described in Table 4.10.
Table 4.10: Vectors, their function and the designation of the protein fusions
Vector
Name of the fusion protein
pUT18C
T18-protein X
pUT18
protein X-T18
pKNT25
T25-protein X
pKT25
protein X-T25
Fusion protein
X
T18
T18
X
T25
X
X
T25
If the tested proteins interact in the reporter strain, the T18 and the T25 fragments can recognize each
other and then they can catalyze the conversion of ATP to cAMP. As positive control for proteinprotein interactions the fusions of the adenylate cyclase fragments to the yeast GCN4 leucine zipper
(Karimova et al., 1998), are used.
MATERIALS AND METHODS
39
4.3.6.2 Liquid β-galactosidase assay in E. coli
After cotransformation of strain BTH101 with the two complementary Cya fragments T18 and T25
constructs (plasmids expressubg T18 and T25 fusion protein, respectively) and growth of the
transformants on the seletive Media plates (LB, Amp, Km, X-Gal and ITPG) three test tubes with 4 ml
LB-media (Amp, Km, ITPG) were each inoculated with a single colony per construct combination.
The over night cultures were grown at 30°C. For each of the three cultures per construct combination,
a fresh test tube was inoculated in fresh media (4 ml LB-medium with Amp, Km, ITPG) to an
OD600 of ~ 0.1. The interaction of pUT18C-zip (Cya fragment T18 fused to GCN4-leucine-zipper) and
pKT25-zip (Cya fragment T25 fused to GCN4-leucine-zipper) was set to 100 % as reference control,
which typically varied from 950 to 1700 Miller units in the individual experiments. As negative
control for interaction the strain was cotransformed with unfused Cya domains expressing plasmids
pUT18C and pKT25. The cells were incubated at 30°C under vigorous agitation to an OD600 reached
0.4 – 0.6 and subsequently 1 ml of the cell culture was frozen in liquid nitrogen, to stop growth. The
aliquots were stored at -80°C.
For determination of β-galactosidase activities 100 µl of the cell suspension was filled in glass tubes
containing 725 µl LB 200 µl 5 × Z-buffer. 800 µl LB with 200 µl 5 × Z-buffer served as reference.
After the addition of 25 µl 0.1 % SDS and 50 µl chloroform to each reaction sample, the glass tubes
were vortexed and incubated at 28°C for 20 min for cell lysis. To start the reaction 200 µl ONPG
solution (4 mg/ml in 1 × Z buffer) were added, subsequently the glass tubes were thoroughly vortexed.
The enzyme β-galactosidase catalyzes the hydrolysis of ONPG to galactose and o-nitrophenol which
can be detected photometrically at a wavelength of 420 nm. As soon as reaction mixtures turned to a
clear yellow color in comparison to the control sample with no cell suspension, the reaction was
stopped by the addition of 500 µl 1 M Na2CO3 and vortexing. The reaction time was recorded and then
300 µl of each sample were pipetted into a 96-well-plate. The absorption of these solution was
measured at 420 nm and 550 nm, respectively, using the TECAN SpectraFluor Plus. The
β-galactosidase activities [Miller-Units] were calculated as follows:
1.75 A420: Absorption of o-nitrophenol 5 mM MgSO4
A550: light scattering of cell debris 250 mM β-mercaptoethanol
A600: cell density at 600 nm
t: time interval between start and end of the reaction in minutes
V: volume of cell suspension used
1.75: correction factor for B. subtilis cells scattering light at 420 nm
4.3.6.3 Liquid β-galactosidase assay in B. subtilis
An over night culture grown without glucose was inoculated in fresh media without and with
1 % (w/v) glucose to an OD600 = 0.1. The cells were incubated at 37°C under vigorous agitation until
the desired OD600 was reached subsequently 1 ml of cell culture was frozen in liquid nitrogen and
stored at -80°C. For determination of β-galactosidase activities 100 µl of the cell suspension was filled
in glass tubes containing 900 µl Z-buffer. 1 × Z-buffer served as reference. After the addition of 10 µl
40
MATERIALS AND METHODS
lysozyme solution (10 mg/ml in Z-buffer), glass tubes were vortexed and incubated at 28°C for
20 min. After addition of 20 µl Triton X-100 and 10 min incubation at 28°C the reaction was started
by addition of 200 µl ONPG (4 mg/ml in 1 × Z-buffer) followed by and thoroughly vortexing. As soon
as reaction mixtures turned yellow, the reaction was stopped by the addition of 500 µl 1 M Na2CO3.
The reaction time was recorded and then 300 µl of each sample were pipetted into a 96-well-plate. The
absorption of these solution was measured at 420 nm and 550 nm, respectively, using the TECAN
SpectraFluor Plus. The β-galactosidase activities [Miller-Units] were calculated as described in
4.3.6.2.
4.3.7
Software and computer analyses
Table 4.11: Software used in this work
Program
Applicance
Adobe® Photoshop® CS4
Digital image processing
Vector NTI Suite 8.0 Informax, Inc. Bethesda,
USA
Processing of DNA sequence data and administration of
vector database
Clone Manager 5
Construction of plasmids, Processing of DNA sequence data
and administration of vector database
DNASTAR SeqManTM II
Sequence alignment editor (abi-files)
Microsoft® Office Standard Edition 2003
Generation of text documents, diagrams and tables
SigmaPlot 9.0
Calculation of binding constants
Pymol 0.99rc1
Model building tools for molecular graphics
EndNote 10.0
Generating reference index
Table 4.12: Internet services used in this work
URL
Provider
Appliance
http://genolist.pasteur.fr/Colibri
Institute Pasteur, Paris, France
Genome database of E. coli
http://genolist.pasteur.fr/SubtiList
Institute Pasteur, Paris, France
Genome database of B. subtilis
http://www.subtiwiki.unigoettingen.de/wiki/index.php
University of Göttingen,
Germany
Genome database of B. subtilis
http://www.ch.embnet.org/software/
ClustalW.html
EMBnet; Swiss Institute of
Bioinformatics, Switzerland
DNA and protein database
http://www.expasy.ch/tools/pi_tool.html
Swiss Institute of Bioinformatics,
Switzerland
Proteomic database,
calculation of isoelectric
points and molecular weights
http://www.ncbi.nlm.nih.gov
National Center for
Biotechnology Information at the
National Institutes of Health,
Bethesda, USA
DNA and protein database
searches, BLAST server;
literature research
RESULTS
41
5 Results
5.1 Strategy for the isolation and the identification of CcpA interacting
proteins involved in transcriptional activation of ackA by in vivo
crosslinking
CcpA plays an important role as a transcriptional repressor, however, it also activates the
transcription of genes. One of these, ackA, is involved in overflow metabolism. In the presence of
glucose ackA is activated to different levels depending on the growth phase (Grundy et al., 1993a)
and it is cooperatively coactivated by the pleiotropic global regulator CodY (Shivers et al., 2006).
Interestingly, distinct CcpA variants with point mutations in the hinge helix, the coeffector
binding region and the C-terminal subdomain abolish the regulation of some but not all target
genes of CcpA in vivo and in vitro studies (Diel, 2005; Horstmann et al., 2007; Seidel et al., 2005;
Sprehe et al., 2007; Turinsky et al., 2000). From these results and the versatility of known
interaction partners of CcpA it is assumed that there are very likely further unknown interaction
partners of CcpA.
For identification of these unknown interaction partners in vivo crosslinking experiments were
performed. Here, the Strep Protein Interaction Experiment (SPINE) was applied using
Strep-tagged CcpA. SPINE allows the isolation of complexes formed in vivo between a target
protein and its interaction partners. It is based on reversible crosslinking of proteins with a
Strep-tagged target protein which is mediated by addition of formaldehyde. Because
formaldehyde easily passes membranes and since it connects only biomolecules in close
proximity to each other (~ 0.2 nm), this small molecule is commonly used for crosslinking protein
complexes in vivo (Das et al., 2004; Dedon et al., 1991). The crosslinked protein-complexes are
subsequently purified with the Strep-tagged target protein by affinity purification via specific
binding to streptactin. To resolve the crosslinks the Strep-tagged protein complexes eluted are
boiled and separated via SDS-PAGE. The gel is then analyzed for additional bands either visually
by silver staining, or experimentally by immunoblot analysis and mass spectrometry. A scheme of
the SPINE protocol is depicted in Figure 5.1.
42
RESULTS
20 min of incubation
H
H
+ formaldehyde
H
H
C=O
C=O
+
distance 2Å
growth of the strain
with Strep-tagged
protein
cell harvesting
cell disruption
ultracentrifugation
resolving
of complex
by heating
affinity
purification
tagged protein
interaction protein
SDS-PAGE
crosslinked
complex
Figure 5.1: Procedure of protein crosslinking, complex isolation and detection by SPINE
Growing cells of B. subtilis are treated with formaldehyde for 20 min and then harvested. Next the cells are
disrupted and crosslinked Strep-tagged protein complexes are isolated from the soluble fraction of the
crude lysate by Strep affinity chromatography. After elution, the crosslinked complexes are resolved by
heating the samples prior to electrophoresis. The released interaction partners are visible as additional
bands next to the Strep-tagged protein. The coeluted proteins are identified by immunoblotting or mass
spectrometry.
RESULTS
43
5.2 Isolation of CcpA complexes involved in transcriptional activation of
ackA by SPINE
5.2.1
Construction of WH1012 and optimization of in vivo crosslinking
The SPINE approach is based on the isolation of crosslinked complexes of Strep tagged target
proteins and was applied to identify CcpA complexes which were active in repression and
acitivation. Since the majority of the genes are only repressed by CcpA, this experiment was
accomplished with multiple copies of the ackA promoter which is activated by CcpA. For this
purpose, an ackA promoter fragment was cloned in the high copy vector pWH1520 and the
resulting plasmid was named pWH933. In addition, ccpA was fused to Strep-tag coding DNA in
the vector pWH940 to express StrepCcpA for affinity purification. The CCR activity of this
StrepCcpA was similar to wildtype CcpA (Bartholomae, 2008). To express only Strep-tagged
CcpA in the cells, a ccpA deletion mutant, B. subtilis WH649, was transformed with pWH933 and
pWH940 and the resulting in strain WH1012 (Bartholomae, 2008), which was used in the
following experiments.
First, the expression of episomally encoded StrepCcpA was analyzed by immunoblotting and
compared to chromosomally encoded CcpA. Therefore, cells of the following strains were
investigated: B. subtilis 168 trp-, WH1012 and the control strain WH1124. This strain is a ∆ccpA
mutant derived from WH1012 and carries the empty strep vector pGP382 instead of pWH940
(StrepCcpA). The crude protein lysates obtained in the mid exponential phase were analyzed with
an anti-CcpA antibody and an anti-DnaK antibody (Fig. 5.2).
1
2
3
4
5
DnaK
CcpA
Figure 5.2: Steady state level of CcpA in B. subtilis 168, WH1012 and WH1124
CcpA amounts (lower row) in crude protein lysates of cells isolated in the mid exponential growth phase
from the strains B. subtilis 168 trp- (lane 3), WH1012 (lane 4) and WH1124 (∆ccpA) (lane 5). Purified Histagged CcpA served as control (lane 1); lane 2 was empty. As loading control, DnaK was detected with a
monoclonal anti-DnaK antibody (upper row).
With respect to the loading control DnaK, Strep-tagged CcpA is expressed at a higher level than
chromosomally encoded CcpA. No signal was detected in the control strain WH1124 (∆ccpA), as
expected.
44
RESULTS
Next, an ackA’::’lacZ fusion was analyzed to determine the optimal time point and glucose
concentration for isolating the maximum amount of StrepCcpA complexes. Therefore, the strain
WH482 was grown in the presence of BCAAs with glucose concentrations increasing from 0 % to
2 %. The growth curves and the corresponding determined β-galactosidase activities were
measured in triplicates and the results are shown in Figure 5.3.
-35 -10
ackA-cre1
ackA-cre2
ackA‘::‘lacZ
10,00
10.0
3000
2800
2600
2200
2000
1,00
1.00
OD600
1800
0 % glucose
0.1 % glucose
0.5 % glucose
0,10
0.10
1600
1400
1 % glucose
1200
2 % glucose
1000
800
β-galactosidase activity [MU]
2400
600
400
200
0
0,01
0.01
0
1
2
3
4
5
6
7
8
9
10
11
12
Time [h]
Figure 5.3: Growth curves and β-galactosidase activity measurement of WH482 (ackA’::’lacZ) grown
with different glucose concentrations
The diagram shows a scatter plot (rhombi) of the OD600 values of the cultures and a bar chart of Miller
Units against time. OD600 and β-galactosidase activity were determined every hour from cells grown in
CSK medium with the following glucose concentrations: 0 % glucose (black), 0.1 % glucose (red), 0.5 %
glucose (green), 1 % glucose (yellow) and 2 % glucose (blue). A scheme of the genetic elements of the
ackA promoter is shown in the insert. All experiments were carried out in triplicates and the results are
given as averages of these three replicates.
The β-galactosidase activities increased for all glucose concentrations during the exponential
phase. In the stationary phase the β-galactosidase activities decreased to a minimum of 50 MU.
The highest β-galactosidase activity of about 1400 MU was measured in the mid exponential
growth phase in medium supplemented with 0.5 % glucose. This corresponds to a seven-fold
activation compared to the strain grown in medium lacking glucose. Therefore, in this work in
vivo crosslinking experiments were performed with cultures grown in CSK and BCAAs with
0.5 % glucose in the mid-exponential growth phase.
RESULTS
5.2.2
45
Isolation of StrepCcpA-complexes by SPINE
Putative CcpA interaction partners were detected in silver-stained gels after SDS-PAGE. Elution
fractions from Strep affinity chromatography of the strains WH1012 (StrepCcpA) and WH1124
(∆ccpA) grown with and without glucose were analyzed. Typical protein patterns are shown in
Figure 5.4.
WH1012
(StrepCcpA)
kDa
M
+
+
+
-
+
WH1124
(∆ccpA)
-
+
+
+
-
+
glucose
-
formaldehyde
200
100
70
50
40
30
CcpA-Strep
20
1
2
3
4
5
6
7
8
9
10
Figure 5.4: SDS-PAGE from eluates of SPINE with Strep-tagged CcpA
A silver-stained gel with SPINE eluates from the B. subtilis strains WH1012 and WH1124 is shown in this
figure. This gel contains a molecular weight standard (M) in lane 1, samples from the StrepCcpA
containing strain WH1012 in lanes 2-5, samples from the ∆ccpA mutant WH1124 in lanes 6-9 and purified
StrepCcpA in lane 10. The presence or absence of glucose in the cultures is indicated by glucose + or
glucose − above the respective lanes. Samples harvested before addition of formaldehyde are labeled with
formaldehyde − and samples harvested after formaldehyde treatment with formaldehyde +.
As shown in Figure 5.4, several proteins coeluted with StrepCcpA without crosslinking,
indicating strong complex formation of these proteins with StrepCcpA. The addition of
formaldehyde increased the number of copurified proteins since weak binding of interacting
molecules or interactions with a high on-off rate were fixed to each other. Interestingly, the
protein patterns, the number and the individual amount of the proteins crosslinked to StrepCcpA
were similar in cells grown in the presence or in the absence of glucose. These coeluted proteins
after SPINE visible in silver stained SDS-PAGEs were putative complex partners of CcpA and
were therefore identified by mass spectrometry as described in chapter 1.3. Furthermore, this
experiment shows that the Streptactin matrix does not bind and elute crude lysate proteins which
would falsify SPINE with StrepCcpA.
46
RESULTS
5.3 Identification of StrepCcpA interaction partners from SPINE by mass
spectrometry
The proteins coeluted with StrepCcpA in SPINE (chapter 5.2.2) were identified by mass
spectrometry in collaboration with Dr. Elke Hammer and Prof. Dr. Uwe Völker, Ernst-MoritzArndt University of Greifswald. This analysis revealed more than 600 proteins (MS, data not
shown). However, formaldehyde primarily targets lysine amino groups and side chains of
adenine, guanine and cytosine which are in close vicinity to each other (~ 0.2 nm) (Das et al.,
2004). Therefore, it was assumed that several DNA or RNA binding proteins identified by mass
spectrometry were crosslinked indirectly by DNA or RNA bridges.
In order to discriminate between signals of specifically crosslinked interaction partners of CcpA
and coeluted impurities, the SPINE procedure was modified. For this purpose, proteins were
labeled in vivo either with
14
N or 15N, as described by Dreisbach et al. (Dreisbach et al., 2008).
The basis of this metabolic labeling is the incorporation of different N-isotopes into all amino
acids and then into all proteins. Identical but
14
N or
15
N labeled peptides form diffently treated
cultures can be discriminated by MS due to their different masses. The ratios between the signal
intensities of the 14N or 15N labeled proteins provide the relative abundance of the peptides in the
different original cell cultures. Consequently, the ratio of the signal intensity of a crosslinked 14N
labeled peptide compared to the non-crosslinked 15N labeled peptide is high when a protein forms
a complex with StrepCcpA only after crosslinking. Very stable complexes might result in a 1:1
ratio at high signal intensities.
Therefore, in this work two cultures of the strain WH1012 were grown in glucose containing
minimal medium with either (14NH4)2SO4 and
14
N BCAAs or (15NH4)2SO4 and
15
labeled medium. In the mid-exponential growth phase only the cells grown in the
14
N BCAAs
N medium
were treated with formaldehyde before the cells of both cultures were harvested. Prior to cell
disruption and StrepCcpA affinitiy chromatography the cells of the crosslinked 14N- and the noncrosslinked
15
N-labeled culture were mixed 1:1. Finally, this eluate was analyzed by mass
spectrometry (Figure 5.5).
RESULTS
14N
47
medium
+ Formaldehyde
cell harvesting
cell disruption
+
purification
15N
medium
cell harvesting
+
mass
spectrometry
Figure 5.5: Modification of SPINE by 14N or 15N in vivo labeling prior to mass spectrometric analysis
Cells of WH1012 were grown to the mid exponential growth phase and then only the 14N labeled culture
was treated with formaldehyde. After sedimentation and resuspension in cell disruption buffer the cells of
both cultures were mixed 1:1. This mixture was lysed and the StrepCcpA complexes were purified. Finally,
the eluate was analyzed by mass spectrometry. The formaldehyde-dependent crosslinking was also
performed vice versa with the 15N in vivo labeled cells.
This experiment was also performed with formaldehyde treated
14
15
N labeled cells and untreated
N labeled cells. A protein identified in MS was only considered to be an interaction partner of
CcpA if it was identified in both experiments with at least two different peptides and at least 6
spectra in one MS sample. Furthermore, enrichment by crosslinking should be higher than 1.5
fold by comparing the number of spectral counts in crosslinked samples with that in samples
without crosslinking (Appendix Table A1, MS analysis of the samples was performed in
collaboration with Dr. Elke Hammer and Prof. Uwe Völker (Ernst-Moritz-Arndt-Universität
Greifswald)). Proteins with an enrichment ratio lower than 1.5 were considered to be
non-specifically coeluating from the streptactin matrix regardless of their count of identified
peptides or recorded spectra. The application of these selection criteria reduced the number of
potential interaction partners to 222 proteins (see Appendix Table 1) pointing to a broad
interactome of CcpA. A subpopulation of 143 proteins was only detected after crosslinking,
whereas 79 proteins were additionally detected in lower abundance in non-crosslinked samples. A
selection of 44 identified proteins with known function is listed in Table 5.1. These proteins can
be classified roughly into 4 different groups. The first group is obviously functionally related to
CcpA because it contains proteins involved in transcription and regulation. The proteins involved
in sugar transport, glycolysis and the TCA cycle build the second group. These proteins are either
related to coeffectors of CcpA or their expression is regulated by CcpA. Less obvious are
interactions with the metabolic enzymes found in the third group or the proteins involved in
sporulation, competence and cell division in the fourth group. Among the putative interaction
partners in the first group HPr, the RNA polymerase subunits and CodY are of particular interest,
because they are involved in the activation of ackA transcription.
48
RESULTS
Table 5.1: Overview of CcpA interaction partners identified in mass spectrometry
Functional category / Cellular process
Identified proteins
Transcription
RpoA, RpoB, RpoC, GreA, NusA
Regulators
CodY, CheV, TenA
Sugar transport
HPr, PtsI
Glycolysis
FbaA, Eno, PfkA, Pgi, Pyk
TCA
Mdh, CitB, CitG, CitZ, Icd, SucD
Biotin and fatty acid biosynthesis
FabD, FabG, FabI
Amino acid metabolism
GlnA, GltA, GltB, AroF, ArgG
Purine and pyrimidine synthesis
GuaA, GuaB, PurA, PurB, PurH
Sporulation
SpoVG, Spo0M
Competence
SrfAA, SrfAB, SrfAC
Cell division
FtsZ, FtsH, DivIVA, MinD, GidA
5.4 Interaction partners of StrepCcpA in vivo
Some of the putative interaction partners detected by MS actually might form complexes with
CcpA since formaldehyde crosslinks molecules in close vicinity. However, the spectral counts of
some proteins, like HPr and CodY, were lower than expected (Appendix Table A1).
Consequently, in order to examine whether the selection criteria applied to the MS analysis were
appropriate the elution fractions from SPINE subjected to MS were also applied to SPINEimmunoblot analyses to identify already established and as well as novel interaction partners of
CcpA.
5.4.1
Analysis of the interaction of StrepCcpA with HPr in vivo
The role of HPrSer46P as main coeffector for CcpA was intensely studied and is well
characterized (Deutscher et al., 1994; Fujita, 2009). Although there is a large body of evidence for
complex formation of CcpA with HPrSer46P in vitro (Deutscher et al., 1995; Horstmann et al.,
2007; Schumacher et al., 2004; Seidel et al., 2005), the physical interaction between both proteins
has not been demonstrated in vivo, yet. HPr fulfilled the criteria applied to the MS analysis and
was identified as an interaction partner of CcpA. However, the spectral count of 6 spectra was
lower than expected (Appendix Table 1), which is likely a reflection of the small size of the
protein (88 amino acids), which yields a few tryptic peptides that are theoretically accessible by
MS. Therefore, and in order to test whether the experimental SPINE setup used in this work is
RESULTS
49
suitable to detect known interaction partners of CcpA, the presence of the HPr was analyzed. For
this purpose, as an additional control approach elution fractions from SPINE applied to MS were
also analyzed for HPr by immunoblotting with an anti-HPr antibody (Fig. 5.6).
WH1012
(CcpA-Strep)
WH1124
(∆ccpA)
HPr
+
-
1
2
+
+
3
+
-
+
-
-
glucose
+
-
+
formaldehyde
4
5
6
7
Figure 5.6: In vivo evidence for CcpA-HPr by immunoblotting
An immunoblot analysis for HPr in SPINE eluates from the B. subtilis strains WH1012 (StrepCcpA) and
WH1124 (∆ccpA) is depicted in this figure. The presence or the absence of glucose in the cultures is
indicated above the respective lanes by “glucose +” or “glucose −”. Samples harvested before addition of
formaldehyde are labeled with “formaldehyde −” and after formaldehyde treatment with “formaldehyde +”.
This blot contained purified HPr as positive control in lane 1, eluates from the ∆ccpA mutant WH124 in
lanes 2-3 and samples from the StrepCcpA containing strain WH1012 in lanes 4-7.
Analysis of SPINE eluates from cultures with glucose show that HPr was only detected in the
presence of StrepCcpA and after formaldehyde crosslinking. Without formaldehyde HPr was not
detected. The analogous experiment from cultures without glucose also did not show a HPr signal.
This is in good agreement with studies that show that PTS sugars like glucose trigger CCR by
CcpA via complex formation with HPrSer46P (Görke & Stülke, 2008).
Although HPr was only coeluted in crosslinked SPINE eluates of WH1012 when glucose was
available, the influence of the Ser46 phosphorylation status of HPr remained unclear in this
experimental approach. In order to address this question, the strain WH1125 was constructed
which basically has the same genotype as WH1012, but a substitution of ptsH for the ptsH1 allele
which encodes for a HPrSer46Ala mutant. The results from SPINE with the strains WH1012
(StrepCcpA, HPr(WT)), WH1124 (∆ccpA, HPr(WT)) and WH1125 (StrepCcpA, HPrSer46Ala)
are shown in Figure 5.7.
50
RESULTS
StrepCcpA
HPr
1
HPr(WT)
2
∆ccpA
HPrS46A
3
crude lysate
HPr(WT)
4
∆ccpA
StrepCcpA
HPr(WT)
5
HPrS46A
6
HPr(WT)
7
eluted fractions
Figure 5.7: In vivo evidence for CcpA-HPrSer46P interaction by immunoblotting
In this figure an immunoblot analysis is shown for HPr in SPINE eluates from the B. subtilis strains
WH1012 (StrepCcpA), WH1124 (∆ccpA) and WH1125 (StrepCcpA, HPrSer46Ala). The blot contains
crude lysates and crosslinked elution fractions of StrepCcpA-protein complexes of the strains WH1012
(StrepCcpA, HPr (WT)) in lane 2 and 5, WH1125 (StrepCcpA, HPrSer46Ala) in lane 3 and 6 and WH1124
(∆ccpA) in lane 4 and 7. Purified HPr in lane 1 serves as a control. All samples were derived from cultures
grown in CSK supplemented with 0.5 % glucose.
Figure 5.7 illustrates the expected presence of HPr in all crude lysates before complex
purification. In the fractions containing purified StrepCcpA complexes a signal for HPr was only
detected in the strain WH1012, carrying the wildtype HPr, but not in the HPrSer46Ala strain
WH1125. This result confirmed the important role of residue serine 46 of HPr for the interaction
with CcpA.
5.4.2
Analysis of the interaction of StrepCcpA with Crh in vivo
Besides HPrSer46P as main coeffector, CrhSer46P also leads to the stimulation of CcpA binding
to cre (Galinier et al., 1997). However, like HPr the physical interaction between CcpA and Crh
in vivo remained to be elucidated. In order to test, if Crh interacts with CcpA or Crh is able to
substitute for the HPrSer46Ala mutant SPINE eluates of WH1012 (StrepCcpA, HPr(WT)) and
WH1125 (StrepCcpA, HPrSer46Ala) were investigated for the presence of Crh (Fig. 5.8).
RESULTS
51
1
2
3
crude lysate
4
5
flowthrough
6
7
8
9
CrhP
eluted fractions
Figure 5.8: Analysis of the CcpA-Crh interaction in vivo by immunoblotting
In this figure an immunoblot analysis is shown for Crh in SPINE eluates from the B. subtilis strains
WH1012 (StrepCcpA), WH1124 (∆ccpA) and WH1125 (StrepCcpA, HPrSer46Ala). The blot contains
crude lysates, flowthrough and crosslinked elution fractions of StrepCcpA-protein complexes of the strains
WH1124 (∆ccpA) (lanes 1, 4 and 7), WH1012 (StrepCcpA, HPr (WT)) (lanes 2, 5 and 8) and WH1125
(StrepCcpA, HPrSer46Ala) (lanes 3, 6 and 9). Purified Crh in lane 10 served as a control. All samples were
derived from cultures grown in CSK supplemented with 0.5 % glucose.
Crh was detected in all crude lysates before complex purification and in the flowthrough of all
strains. Interestingly, Crh was not detected in the fractions containing purified StrepCcpA
complexes of all investigated strains.
5.4.3
Interaction of StrepCcpA with the α subunit of the RNA polymerase (RpoA)
in vivo
Although most genes and operons are repressed by CcpA, in transcriptomic studies about 50
genes were activated by CcpA (Moreno et al., 2001) Interestingly, CcpA variants with point
mutations in the coeffector binding region were only active in CCR of ackA (Diel, 2005;
Horstmann et al., 2007; Seidel et al., 2005; Sprehe et al., 2007; Turinsky et al., 2000). In addition
to the previous studies, in this work the RNA polymerase subunits α, β and β′ were identified
mass spectrometry (chapter 1.3). Based on these data an interaction between CcpA and the RNA
polymerase was assumed.
In order to verify the results of MS analysis, a SPINE-immunoblot analysis was also performed
for the α subunit of RNA polymerase. For this purpose, SPINE eluates of the strains WH1012
(StrepCcpA) and WH1124 (∆ccpA) were analyzed for the presence of RpoA by immunoblotting
with a monoclonal anti-RpoA antibody (Fig. 5.9).
52
RESULTS
WH1124
(∆ccpA)
WH1012
(StrepCcpA)
+
-
-
-
+
+
-
-
2
3
4
5
+
-
+
1
eluted fractions
crude lysate
+
glucose
+
+
-
+
formaldehyde
6
7
8
eluted fractions
Figure 5.9: In vivo evidence for CcpA-RpoA interaction by immunoblotting
In this figure an immunoblot analysis is shown for RpoA in SPINE eluates from the B. subtilis strains
WH1012 (StrepCcpA) and WH1124 (∆ccpA). The blot contains crude lysates and crosslinked elution
fractions of StrepCcpA-protein crude lysates (lanes 3) and in the crosslinked elution fractions of
StrepCcpA-complexes of the strains WH1124 (∆ccpA) (lanes 1 and 2) and of the strain WH1012
(StrepCcpA) in (lanes 4, and 5 to 8). The presence or absence of glucose in the cultures is indicated by
“glucose +” or “glucose −” above the respective lanes. Samples harvested before addition of formaldehyde
are labeled with “formaldehyde −” and samples harvested after formaldehyde treatment with
“formaldehyde +”.
In both strains, WH1012 (StrepCcpA) and WH1124 (∆ccpA), RpoA was detected in the crude
lysates before SPINE. After treatment with formaldehyde, RpoA was detected in the SPINE
eluates from the StrepCcpA expressing strain cultured in the presence and in the absence of
glucose, indicating that the presence of glucose is not a prerequisite for the interaction of CcpA
with the α subunit of the RNA polymerase.
5.4.4
Interaction of StrepCcpA with CodY in vivo
Interestingly, full transcriptional activation of ackA requires another global regulator CodY
(Shivers et al., 2006; Turinsky et al., 1998; Turinsky et al., 2000). CodY binds upstream of CcpA
at the ackA promoter and is able to activate ackA transcription alone, similar to CcpA, but
cooperative activation by both regulators is much stronger (Shivers et al., 2006). Although, these
studies focused on the physiological activation of different mutations either CcpA and the
promoter region of ackA and the identification of binding sites of both CcpA and CodY, the
evidence for a physical interaction between both regulators remained to be experimentally
verified.
The complex formation of CcpA and CodY was investigated by SPINE-immunoblot analysis.
Because an antibody raised against CodY was not available, CodY was indirectly detected via
Flag-tag by construction of a CodY-3 × Flag-tag fusion protein. For this purpose, the integrative
plasmid pGP1331 (Stülke, Göttingen) was used, because it allows the fusion of a 3 × Flag-tag to
RESULTS
53
the C-terminus of a protein, which is expressed from its natural locus on the chromosome. For this
approach, first × 600 bp from the 3′ end of codY were amplified by PCR and cloned into
pGP1331, resulting in pWH2371 (Fig. 5.10). Then, WH1012 was transformed with pWH2371
resulting in strain WH1129, which expressed CodY with a C-terminal 3 × Flag-tag from its native
promoter. Finally, WH1129 was analyzed for integration of the Flag-tag by sequencing.
SpecR
Figure 5.10: Plasmid pWH2371
used for construction of a
'codY
CodY-3 × Flag-tag fusion protein
The plasmid pWH2371 is a vector,
which carries sequences coding for
BamHI
the C-terminal sequence of CodY
3 × Flag-tag
with a C-terminal 3 × Flag-tag. This
DYKDHDGDYKDHDIDYKDDDDK vector is a derivative of pGP1331,
which allows the fusion of a
3 × Flag-tag to the C-terminus of a
protein of interest at its natural locus.
EcoRI
pWH2371
~6000 kb
AmpR
rep(pMB1)
The complex formation of CcpA and CodY in vivo was investigated by SPINE-immunoblot
analysis. For this purpose, cells of the stains WH1124 (∆ccpA), WH1012 (StrepCcpA) and
WH1129 (WH1012, CodY-Flag) were cultured with and without 0.5 % glucose and SPINE was
performed. The 3 × Flag-tag was detected with a monoclonal anti-Flag antibody (Fig. 5.11).
WH1124
(∆ccpA)
WH1129
(StrepCcpA,
CodY-3 × Flag)
WH1012
(StrepCcpA)
CL
+
-
+
+
+
-
+
+
+
-
+
+
1
2
3
4
5
6
7
-
8
+
glucose
formaldehyde
9
Figure 5.11: In vivo evidence forCcpA-CodY interaction by immunoblotting
In this figure an immunoblot analysis is shown for CodY-3 × Flag in SPINE eluates from the B. subtilis
strains WH1012 (StrepCcpA), WH1124 (∆ccpA) and WH1129 (StrepCcpA, CodY-3 × Flag). The blot
contains elution fractions of StrepCcpA of WH1124 (∆ccpA) in lanes 2-3, WH1012 (StrepCcpA) in lanes
4-5, and WH1129 in lanes 6-9. As control crude lysate (CL) of WH1129 in lane 1 was used. The presence
or absence of glucose in the cultures is indicated above the figure by “glucose +” or “glucose −”. Samples
which were harvested before addition of formaldehyde are labeled with “formaldehyde −” and after
formaldehyde treatment with “formaldehyde +”.
54
RESULTS
CodY-3 × Flag was present in the crude lysate of WH1129 (StrepCcpA, CodY-3 × Flag) before
SPINE, as expected. The Flag-tagged CodY was only detected in the SPINE eluates from
formaldehyde treated cultures expressing StrepCcpA and CodY-3 × Flag. A very strong
CodY-3 × Flag signal was detected in eluates from cultures grown with glucose. In contrast,
cultures without glucose yielded only a faint band for CodY-3 × Flag in the immunoblot. Elution
fractions of the control strains WH1124 (∆ccpA) and WH1012 (StrepCcpA, CodY) showed
expectedly no signal for the Flag-tag.
5.5 BACTH analysis of direct interactions between CcpA and putative
interaction partners
In SPINE, the subunits α (RpoA), β (RpoB) and β′ (RpoC) of the RNA polymerase core enzyme
were coeluted in a complex with CcpA. However, the SPINE analysis does not contain any
information if CcpA interacts with all or only with two or just one of the RNAP subunits because
formaldehyde also crosslinks the subunits in the RNA polymerase heterooligomer. Therefore, an
alternative screen was performed to analyze binary protein interactions with CcpA. For this
purpose, a bacterial two-hybrid assay (BACTH) in E. coli (Karimova et al., 1998) was performed
to analyze the binary interactions of the RNA polymerase subunits with CcpA in vivo and to
specify which RNA polymerase component interacts with CcpA. The BACTH assay is based on
reconstitution of adenylate cyclase activity when the T18 and T25 domains of Bordetella pertussis
Cya are connected via interactions between proteins fused to them (chapter 4.3.6.1 and 4.3.6.2).
Moreover, several further proteins, which were identified in StrepCcpA complexes (Table 5.1),
were also analyzed for the existance of interactions with CcpA.
5.5.1
Interaction analysis of CcpA with RNA polymerase subunits α, β and β′′
For the interaction analysis between CcpA and the RNA polymerase in this approach both Cya
domains, T18 and T25, were separately fused to full-length copies of CcpA and the target proteins
RpoA, RpoB and RpoC, respectively. In order to increase the chance of proper binding of T18 to
T25 in a protein complex, the number of possible orientations of T18 and T25 is increased by
fusing each domain either to the N-terminus or the C-terminus of each interaction partner. Then
every possible combination of an N- or C-terminal T18 fusion protein with an N- or C-terminal
T25 fusion protein was analyzed for restored Cya activity. Fusions of the GCN4 domain of the
leucine zipper to each Cya domain served as positive control. Since none of the combinations of
unfused Cya domains with any of the analyzed proteins fused to the complementary Cya domain
RESULTS
55
resulted in Cya activity, it was concluded that the assay only detects highly specific interactions.
Pairs of T18- and T25-protein fusions were coexpressed in the indicator strain BTH101 (∆cya),
and screened for possible interactions by a qualitative plate assay for β-galactosidase activity. The
T25-zip
T25
RpoC-T25
T25-RpoC
RpoB-T25
T25-RpoB
RpoA-T25
T25-RpoA
CcpA-T25
T25-CcpA
results of the BACTH assay are depicted in Figure 5.12.
CcpA-T18
T18-CcpA
RpoA-T18
T18-RpoA
RpoB-T18
T18-RpoB
RpoC-T18
T18-RpoC
T18
T18C-zip
Figure 5.12: BACTH analysis of the direct interactions between CcpA and the RNA polymerase
subunits α (RpoA), β (RpoB) and β′ (RpoC)
The figure shows a BACTH assay of E. coli BTH101 strains expressing different combinations of N- or Cterminal fusions of the T18 or the T25 domains of the B. pertussis adenylate cyclase with the interactants
CcpA, RpoA, RpoB and RpoC. Each protein was fused to either domain of Cya. Fusions to T25 are listed
above and T18 fusions are listed on the left side in the figure. N-terminal fusions to the T18 Cya domain
are indicated as “proteinX-T18” and C-terminal fusions “T18-proteinX” and an equally denomination for
fusion to the T25 Cya domain. T18 and T25 indicate the combination of the unfused Cya domains and are
the negative control. T18C-zip and T25-zip indicate the fusions of Cya domains with the leucine zipper
serving as a positive control. Blue-colored colonies indicate interaction of the proteins fused to the
adenylate cyclase domains.
Self-pairing of CcpA or the α subunit of the RNA polymerase, respectively, resulted in strong
signals confirming dimerization of both proteins but not for the self pairing of RpoB and RpoC.
The combination of CcpA and the α subunit of the RNA polymerase also showed detectable Cya
activity and thus complex formation. The absence of enzymatic activity in BACTH analyses of
Cya-CcpA fusions with different combinations of the Cya fusions to the β (RpoB) or the
β′ subunit (RpoC) did not restore Cya activity and, consequently, did not show a direct interaction
of CcpA with RpoB or RpoC.
56
5.5.2
RESULTS
Interaction analysis of CcpA with mutants of the α subunit of RNA polymerase
The α subunit of the RNA polymerase of B. subtilis is a 314 amino acids protein, containing an Nterminal subdomain (αNTD) (amino acids 1-231) and a C-terminal domain (αCTD) (amino acids
245-314) which are separated by a 13 aa linker (232-244) (Fig. 5.13).
α
NTD
1 – 231
αNTD
NTD
1 – 231
αCTD
linker
CTD
232 – 244
245 – 314
linker
232 – 244
linker
CTD
232 – 244
245 – 314
Figure 5.13: RpoA subdomains used in this work
The entire α subunit of the RNA polymerase contains the N-terminal domain (aa 1 – 231), the linker
(aa 232 – 244) and the C-terminal domain (aa 245 – 314). Below the two deletion mutants used in this
approach are illustrated. The αNTD lacks the C-terminal domain. In contrast in the αCTD the N-terminal
domain is deleted.
In order to discriminate whether the N- or the C-terminal domain of RpoA is interacting with
CcpA, the αNTD and the αCTD were either fused to the N- or the C-terminal Cya domain and
combined with the complementary CcpA Cya fusions. The results of the BACTH assay are
depicted in Fig. 5.14A. Interestingly, pairing of CcpA with disrupted α subunit fusions showed
different signals. CcpA pairing with fusions of the αNTD resulted in significant signals. In
contrast, the combination of CcpA with the αCTD did not restore Cya activity. The self-pairing of
the αNTD or of the αCTD revealed detectable signals. The pairing of the GCN4 domains of the
leucine zipper as a positive control in BACTH showed a strong self-interaction, as anticipated.
Unfused Cya domains showed no activity in combination with any of the analyzed proteins
(Fig. 5.14A).
.
T25-zip
T25
αCTD-T25
T25-αNTD
T25-αNTD
T25-αCTD
RpoA-T25
RpoA-T25
αNTD-T25
T25-RpoA
T25-RpoA
A
CcpA-T25
57
T25-CcpA
RESULTS
CcpA-T18
T18-CcpA
RpoA-T18
T18-RpoA
αNTD-T18
T18-αNTD
αCTD-T18
T18-αCTD
T18
T18C-zip
αCTD-T25
T25
T25-zip
4.7*
n.a.
T18-CcpA 120.9‡ 94.3* 40.7* 67.8* 57.0* 55.4* 13.5* 3.6*
4.0*
n.a.
αNTD-T25
4.5*
CcpA-T25
CcpA-T18 90.4# 104.0# 12.1* 38.4* 37.2* 77.0* 7.2*
T25-CcpA
T25-αCTD
B
RpoA-T18
7.2*
8.8*
53.7* 52.7* 66.4* 110.4# 20.1*
6.9*
4.7*
n.a.
T18-RpoA
5.0*
5.4*
52.7* 39.8# 42.4* 78.7*
4.5*
93.3*
4.0*
n.a.
αNTD-T18
24.4
14.8* 83.2‡ 58.0* 64.7* 48.5*
9.9*
5.0*
3.7*
n.a.
T18-αNTD
3.3*
22.7* 50.9* 52.9* 59.2* 34.6* 19.2*
6.5*
2.7*
n.a.
αCTD-T18
2.6*
2.7* 103.4‡ 1.9*
2.4*
2.5*
n.a.
T18-αCTD
2.1*
1.8*
3.4*
37.3* 49.1*
2.6*
2.7*
73.7#
3.0*
n.a.
T18
2.5*
2.4*
2.4*
2.5*
6.7*
3.1*
2.9*
2.6*
3.4*
n.a.
T18C-zip
n.a.
n.a.
n.a.
n.a.
n.a.
n.a.
n.a.
n.a.
n.a. 100.0#
35.2# 11.5* 36.8#
Figure 5.14: BACTH analysis of the direct interaction of CcpA and RpoA and the different domains
of the α subunit of RNA polymerase
The figure shows a BACTH assay of E. coli BTH101 strains expressing different combinations of N- or Cterminal fusions of the T18 or the T25 domains of the B. pertussis adenylate cyclase with the interactants
CcpA, RpoA and the N- and the C-terminal domain of RpoA (indicated as αNTD and αCTD). T18 and T25
indicate the combination of the unfused Cya domains and are the negative control. The combination of
T18C-zip and T25-zip served as a positive control (A) Blue-colored colonies indicate interaction of the
proteins fused to the adenylate cyclase domains. (B) Quantitative β-galactosidase activities of the
corresponding strains. Bold numbers depict β-galactosidase activities ≥ 50 % of the positive control. All
experiments were carried out in triplicates and the results are given as averages of these three
replicates.Standard deviations based on T18-zip and T25-zip self interaction on 100 %: *: ≤ 5 %, #: ≤ 10 %,
‡
: ≤ 20 %; n.a.: not analyzed.
58
RESULTS
The efficiency of the complementation between the hybrid proteins of all transformants was
quantified by determining the β-galactosidase activities, which are correlated with the cellular
cAMP level (Karimova et al., 1998). The transformants that coexpressed fusion proteins of CcpA
and full-length RpoA or CcpA and αNTD produced β-galactosidase activities between 5 % and
77 % of the positive control. Combinations of the unfused Cya domains with any of the analyzed
proteins showed β-galactosidase activities < 7 %. In conclusion the quantitative β-galactosidase
activities correspond to the qualitative plate assay and verfied the interaction between CcpA and
the α subunit of the RNA polymerase (Fig. 5.14B).
5.5.3
Interaction analysis of CcpA with CodY by BACTH
As the analysis of SPINE eluates by MS and immunoblotting identified CodY in CcpAStrep
complexes, the direct interaction of both regulators CcpA and CodY investigated by BACTH. For
this purpose, analogous to the experiments of CcpA and the RpoA, CodY was fused to the N- and
C-terminus of each Cya domain, respectively. All possible combinations of N- and C-terminal
fusion proteins of CcpA and CodY were analyzed for restored Cya activity. The results of the
CcpA-T18
T18-CcpA
T18-CodY
CodY-T18
T18
T18-zip
T25-zip
T25
T25-CodY
CodY-T25
T25-CcpA
CcpA-T25
BACTH assay are depicted in Figure 5.15.
Figure 5.15: BACTH analysis of the
direct interaction of CcpA and CodY
The figure shows a BACTH assay of
E. coli BTH101 strains expressing different
combinations of N- or C-terminal fusions
of the T18 or the T25 domains with the
interactants CcpA and CodY. T18 and T25
are the negative control and the
combination of T18C-zip and T25-zip
served as a positive control. Blue-colored
colonies indicate interaction of the proteins
fused to the adenylate cyclase domains.
Besides the leucine-zipper self-pairing, only the combinations for self-pairing of CcpA and
CodY restored Cya activity in the BACTH assay. The combination of CcpA and CodY did
not show detectable adenlyate cyclase activities. Interestingly, the strain BTH101 (∆cya) was
not able to grow after cotransformation of T18-CodY and CodY-T25, a combination for
CodY self-pairing. Unfused Cya domains showed as expected no Cya activity in combination
with CcpA or CodY.
RESULTS
5.5.4
59
Interaction analysis of CodY with α subunit of RNA polymerase by BACTH
CodY, represses transcription of genes but can also activates some genes (Molle et al., 2003;
Sonenshein, 2007). As described before (chapter introduction and results) the full transcriptional
activation of ackA (Shivers et al., 2006), requires CodY. Thus, a direct interaction of CodY with
the α subunit of the RNA polymerase seemed possible. Therefore, this putative direct interaction
CodY-T18
T18-CodY
RpoA-T18
T18-RpoA
T18
T18C-zip
T25-zip
T25
RpoA-T25
T25-RpoA
T25-CodY
CodY-T25
of CodY and RpoA was investigated by BACTH (Fig. 5.16).
Figure 5.16: BACTH analysis of the
direct interaction of CodY and RpoA
The figure shows a BACTH assay of E. coli
BTH101 strains expressing different
combinations of N- or C-terminal fusions of
the T18 or the T25 domains with the
interactants CodY and RpoA. T18 and T25
are the negative control and the
combination of T18C-zip and T25-zip
served as a positive control. Blue-colored
colonies indicate interaction of the proteins
fused to the adenylate cyclase domains.
In the BACTH analysis only the self-pairing of CodY, RpoA and the leucine zipper showed Cya
activity. The combinations of CodY and RpoA were not able to restore Cya activity. CodY or
RpoA showed no activity in combinations with unfused Cya domains, as expected.
5.5.5
Interaction analysis of CcpA with tricarboxylic acid (TCA) cycle enzymes
In B. subtilis, the activities of the TCA cycle enzymes major citrate synthase (CitZ), aconitase
(CitB) and isocitrate dehydrogenase (Icd) are reduced when the cells were grown in the presence
of glucose and glutamate (Blencke et al., 2003; Hanson & Cox, 1967; Jourlin-Castelli et al., 2000;
Kim et al., 2002; Sonenshein, 2007). CcpA is involved in glucose-dependent repression of almost
all TCA cycle genes (Tobisch et al., 1999a). While CcpA represses transcription of citZ directly,
citB expression is obviously released from repression in a ∆ccpA mutant by indirect effects
(Blencke et al., 2003; Kim et al., 2002). Recently, interactions among TCA enzymes were
detected which suggest a TCA cycle metabolon (Meyer et al., 2011). Interestingly, in SPINE
eluates of CcpAStrep several TCA cylce enzymes were identified as abundant proteins. These
proteins were CitB (Aconitase), CitG (Fumarat hydratase), CitZ (major citrate synthase) Icd
(Isocitrate dehydrogenase) and Mdh (Malate dehydrogenase). Therefore, CcpA and CitB, CitG,
CitZ, Icd and Mdh were combined in BACTH analyses (Fig. 5.17).
CitG-T25
T25-CitG
CitZ-T25
T25-CitZ
T25-Icd
Mdh-T25
T25-Mdh
T25
T25-zip
T25-CitB
Icd-T25
T25
CitB-T25
T25-CcpA
CcpA-T25
A
T25-CcpA
RESULTS
CcpA-T25
60
CcpA-T18
T18-CcpA
CitB-T18
T18-CitB
CitG-T18
T18-CitG
CitZ-T18
T18-CitZ
T18
B
T25-zip
T18-zip
CcpA-T18
T18-CcpA
Icd-T18
T18-Icd
Mdh-T18
T18-Mdh
T18
T18-zip
Figure 5.17: BACTH analysis of the direct interactions between CcpA and and the TCA enzymes
CitB, CitG, CitZ, Icd and Mdh
The figure shows BACTH analyses of E. coli BTH101 strains expressing different combinations of N- or
C-terminal fusions of the T18 or the T25 domains with the interactants CcpA, CitB, CitG and CitZ (A) and
CcpA, Icd and Mdh (B). Blue-colored colonies indicate interaction of the proteins fused to the adenylate
cyclase domains.
RESULTS
61
The BACTH assays show that all combinations for self-pairing of CcpA and CitZ, respectively,
resulted in strong signals confirming multimerization of these proteins (Fig. 5.17A). The Cya
acitiviy for self-pairing of CitB, CitG, Icd and Mdh was detectable only in some combinations
(Fig. 5.17A and 5.17B). Several combinations of CcpA with CitG, CitZ, Icd and Mdh showed
detectable Cya activities and therefore complex formation (Fig. 5.18A and 5.18B). Combinations
of CcpA with CitB showed no Cya activity and suggest that there is no direct interaction between
these proteins (Fig. 5.17B). In conclusion, qualitative plate assays revealed positive interactions
between CcpA and CitG, CitZ, Icd and Mdh, but not between CcpA and CitB by BACTH.
5.6 Characterization of the complex formation of CcpA, CodY and the
α subunit of the RNA polymerase by in vitro measurements
The motivation for the projects described in the following chapters was to validate and
characterize interactions which are involved in complex formation of CcpA, CodY and the
α subunit of the RNA polymerase by suitable in vitro measurements with purified proteins. The
data revealed from these analyses in combination with the results obtained from SPINE and
BACTH was used to explain the interactions of CcpA which are required for transcriptional
activation.
5.6.1
Construction of overexpression vectors for purification of the α subunit of the
RNA polymerase and CodY
The α subunit of the RNA polymerase of B. subtilis is a protein which contains 314 amino acids
and which is subdivided into the the N-terminal domain (αNTD) which contains the amino acids
1 – 231 and the C-terminal domain (αCTD) which contains the amino acids 245 – 314. Both
domains are connected by a linker with a length of 14 amino acids (232 – 245) (Fig. 5.18).
62
RESULTS
αNTD
MIEIEKPKIE TVEISDDAKF GKFVVEPLER GYGTTLGNSL RRILLSSLPG
50
αNTD
AAVTSIQIDG VLHEFSTIEG VVEDVTTIIL HIKKLALKIY SDEEKTLEID
100
αNTD
VQGEGTVTAA DITHDSDVEI LNPDLHIATL GENASFRVRL TAQRGRGYTP
150
αNTD
ADANKRDDQP IGVIPIDSIY TPVSRVSYQV ENTRVGQVAN YDKLTLDVWT
αNTD
200
linker
DGSTGPKEAI ALGSKILTEH LNIFVGLTDE AQHAEIMVEK EEDQKEKVLE
250
αCTD
MTIEELDLSV RSYNCLKRAG INTVQELANK TEEDMMKVRN LGRKSLEEVK
300
αCTD
AKLEELGLGL RKDD
314
Figure 5.18: Protein sequence of RpoA of B. subtilis
The figure shows the protein sequence of the α subunit of the RNA polymerase of B. subtilis, which
contains 314 amino acids. This protein is subdivided into the the N-terminal domain (αNTD) which
contains the amino acids 1 – 231 (dark grey box) and the C-terminal domain (αCTD) which contains the
amino acids 245 – 314 (light grey box). Both domains are connected by a linker with a length of 14 amino
acids (232 – 245) (black line).
For interaction analyses His-tagged CcpA, RpoA, αNTD, αCTD and CodY from B. subtilis
should be overexpressed and purified. For this purpose, the promoterless rpoA and codY genes
were amplified from chromosomal DNA from B. subtilis 168. The overexpression of αNTD was
generated by amplification of a DNA fragment encoding for the αNTD (amino acids 1 – 244) of
rpoA by PCR. All PCR products were digested with XhoI and NheI and XhoI and NdeI,
respectively, and cloned into the overexpression vector pET28b, linearized with the same
enzymes. The resulting plasmids pWH2175, pWH2176 and pWH2177, allow IPTG induced
overexpression of N-terminally His-tagged RpoA, αNTD and CodY, respectively, from B. subtilis
in E. coli BL21(DE3), which expresses T7 polymerase. For overexpression of CcpA from
B. subtilis, the strain FT1 / pWH653 was used which expresses an IPTG inducible C-terminally
His-tagged CcpA protein (Seidel, 2005). N-terminally His6–tagged αCTD, which expresses the
C-terminal domain of the RNA polymerase (245 – 314) was purified from the strain
Rosetta 2 / pET28a-αCTD (Nakano et al., 2010). The overexpression of either protein is described
in the Chapters 4.3.4.5 to 4.3.4.10.
RESULTS
5.6.2
63
Purification of CcpA, RpoA, αNTD, αCTD, HPrSer46P and CodY
N-terminally His-tagged RpoA, αNTD, αCTD and CodY, were purified by Ni2+-affinity
chromatography and the pure protein fractions were then subsequently applied to a size exclusion
chromatography to remove the retaining imidazole and to equilibrate the protein in HBSE buffer
as described in the chapters 4.3.4.6 to 4.3.4.11. His-tagged CcpA was purified as described in
(Seidel, 2005). Figure 5.19A and B show representative Coomassie-stained 12.5 % SDS-PAGEs
with elution fractions of His-tagged αNTD after Ni2+-affinity chromatography (Fig. 5.19A) and
elution fractions after size exclusion chromatography (Fig. 5.19B). Figure 5.19C shows
Coomassie stained SDS-PAGEs with the purified proteins RpoA, CcpA, CodY, αNTD, αCTD and
HPrSer46P used in the following chapters.
A
M 1 2 3 4 5 6 7 8 9 10 11 12 13 14
B
M 15 16 17 18 19 20 21 22 23 24 25 26 27 28
αNTD
C
1
2
3
M
4
M
5
6
200
200
120
70
85
70
50
40
50
RpoA
CcpA
30
25
40
30
αNTD
CodY
25
HPrSer46P
15
10
αCTD
Figure 5.19: Protein purification of αNTD-his6 from B. subtilis from E. coli BL21(DE3) by affinity
purification and size exclusion chromatography
(A) Analytical gel electrophoresis (SDS-PAGE, 12.5 %) of samples of His6-tagged αNTD after Ni-affinity
chromatography. 10 µl of each fraction was analyzed. The soluble fractions of the pre-induction sample
(lanes 1), the sample before purification (lanes 2) and washing (lanes 3 – 4) and elution fractions (lanes
5 – 14) were analyzed. M: molecular weight standard. Fractions 6 – 14 were unified and the imidazole was
removed by size exclusion chromatography (B) with superdex G75. 10 µl of each elution fractions were
tested (lanes 15 – 28). M: molecular weight standard. (C) These gels illustrate the purity of the protein
preparations used for in vitro interaction analyses. The figure shows coomassie stained 12.5 % and 15 %
SDS PAA gels, respectively, loaded with 5 µg of RpoA (lane 1), 10 µg of CcpA (lane 2), 8 µg of CodY
(lane 3), 8 µg of αNTD (lane 4) and 2 µg of αCTD (lane 5) and 7 µg HPrSer46P (lane 6), respectively.
M: molecular weight standard. Only the pure protein species were detectable in each lane.
64
RESULTS
5.7 SPR analyses of interactions of CcpA, RpoA and CodY
Complex formation of CcpA with RpoA and CodYwere analyzed by surface plasmon resonance
(SPR) analyses. SPR is a sensitive method to observe biomolecular interactions in vitro. This
surface analysis method is based on the detection of changes in the refractive index of the adjacent
medium next to a metal such as silver and gold (Liedberg et al., 1995). A slight change at the
interface leads to a change in the SPR signal and allows precise measurements of surface
molecular interactions in real-time.
SPR was demonstrated to be suitable for analyses of B. subtilis CcpA-HPrSer46P complex
formation and for studies of the effect of the low molecular weight effectors FBP and Glc-6-P on
these interactions and binding to cre-sequences (Horstmann et al., 2007; Seidel et al., 2005).
Therefore, this sensitive method seemed to be suitable for interaction analyses of CcpA, RpoA
and CodY and is described in the following sections.
5.7.1
SPR analyses of interactions between CcpA and RpoA
The interaction between CcpA and αNTD from B. subtilis was analyzed by SPR measurements to
validate and quantify the interaction of CcpA with RpoA in vitro. First, the binding of CcpA to
αNTD was investigated. Therefore, about 4500 RU αNTD was covalently coupled to a CM5 chip
and titrated with CcpA concentrations increasing from 0.5 µM to 30 µM. The shape and the
increase in signal height of the sensorgrams shown in Fig. 5.20A indicate highly specific binding
of CcpA to the immobilized αNTD of RpoA. The SPR analysis of CcpA and the αCTD of RpoA
was carried out analogously and showed similar sensorgrams (Fig. 5.20B). The titration could not
be performed with higher concentrations than 30 µM due to insufficient solubility of CcpA at
concentrations above 40 µM. The kinetic constants of complex association and dissociation and
the equilibrium constants were determined for both interactions: ka = 9.4 ± 5.6 × 102 Ms-1,
kd = 2.8 ± 0.8 × 10-3 s-1 and KD = 5.0 ± 3.5 × 10-6 M for the binding of CcpA to the αNTD and
ka = 2.2 ± 0.8 × 102 Ms-1, kd = 1.5 ± 0.2 × 10-3 s-1 and KD = 7.9 ± 3.6 × 10-6 M for the binding of
CcpA to the αCTD. The fits correspond to a Langmuir model for a 1:1 interaction
(Fig. 5.20A and B).
RESULTS
65
A
350
30 µM CcpA
20 µM CcpA
15 µM CcpA
10 µM CcpA
5 µM CcpA
2 µM CcpA
1 µM CcpA
0.5 µM CcpA
Resp. Diff. [RU]
300
250
200
150
100
50
0
0
50
100
Time [s]
150
200
250
B
120
Resp. Diff. [RU]
100
30 µM CcpA
20 µM CcpA
10 µM CcpA
5 µM CcpA
2 µM CcpA
1 µM CcpA
80
60
40
20
0
0
50
100
150
200
Time [s]
250
300
350
400
Figure 5.20: Quantitative analyses of CcpA and RpoA interactions by SPR
The sensorgrams show titrations of (A) immobilized αNTD with CcpA, (B) immobilized αCTD with
CcpA. (C) immobilized CodY with CcpA, (D) immobilized αNTD with CodY and (E) immobilized αCTD
with CcpA. Concentrations of the analyte are depicted on the right side next to each sensorgram.
5.7.2
SPR analyses of interactions between CcpA and CodY
CodY was detected as an interaction partner of CcpA in SPINE-MS and it is required together
with CcpA for full activation of ackA (Shivers et al., 2006). For this purpose, CodY was purified
to homogeneity (Fig. 5.19C) and used in SPR analyses to study the interaction of CcpA with
CodY. CcpA binding to CodY was studied by titration of CodY immobilized on a CM5 chip with
CcpA concentrations increasing from 1 µM to 30 µM. The titration showed specific binding with
rapid association and dissociation phases and increasing signal intensity. An equilibrium analysis
for a 1:1 Langmuir model yielded a KD of 5.9 ± 0.1 × 10-5 M (Fig. 5.21A).
66
RESULTS
A
120
Resp. Diff. [RU]
100
30 µM CcpA
20 µM CcpA
10 µM CcpA
5 µM CcpA
2 µM CcpA
1 µM CcpA
80
60
40
20
0
0
150 200
Time [s]
250
300
350
30 µM αCTD
20 µM αCTD
10 µM αCTD
5 µM αCTD
2 µM αCTD
1 µM αCTD
0.5 µM αCTD
0 µM αCTD
0 µM CcpA
C
60
100
20 µM αNTD
10 µM αNTD
5 µM αNTD
2 µM αNTD
1 µM αNTD
0.5 µM αNTD
0 µM αNTD
0 µM CcpA
50
Resp. Diff. [RU]
100
40
30
20
10
0
80
Resp. Diff. [RU]
B
50
60
40
20
0
-20
-10
-40
-20
-60
0
50
100
150 200
Time [s]
250
300
350
0
50
100
150 200
Time [s]
250
300 350
Figure 5.21: Analyses of CcpA and CodY interactions by SPR
The sensorgrams show titrations of immobilized immobilized CodY with CcpA (A) and titrations of
immobilized CodY with 10 µM CcpA with increasing concentrations of either αNTD (B) or αCTD (C).
Concentrations of each analyte are depicted on the right side next to the corresponding sensorgram.
Furthermore, the influence of RpoA for the interaction between CcpA and CodY was investigated
by cotitrations of immobilized CodY with CcpA and αNTD and CcpA and αCTD, respectively.
The binding CcpA to immobilized CodY was hardly altered by addition increasing concentrations
of αNTD as well as αCTD (Fig. 5.21B and C).
RESULTS
5.7.3
67
SPR analyses of interactions between CodY and RpoA
CodY was detected as an interaction partner of CcpA in SPINE-MS and it is required together
with CcpA for full activation of ackA (Shivers et al., 2006). For this purpose, CodY was used in
SPR analysis to test whether CodY may form complexes with RpoA. Then, either αCTD or αNTD
of RpoA were coupled to a CM5 chip and 1 µM to 40 µM solutions of CodY were injected. The
sensorgrams in Fig. 5.22D and 5.22E show specific binding of CodY to both, αCTD and αNTD as
well. Both interactions were quantified by a 1:1 Langmuir analysis of association and dissociation
kinetics and revealed the following rate and equilibrium constants: ka = 5.2 ± 0.4 × 102 Ms-1,
kd = 1.7 ± 0.1 × 10-3 s-1 and KD = 5.9 ± 4.5 × 10-6 M for the CodY-αCTD complex and of
ka = 1.1 ± 0.8 × 103 Ms-1, kd = 3.9 ± 0.8 × 10-3 s-1 and KD = 8.1 ± 7.5 × 10-6 M for the CodY-αNTD
complex (Fig. 5.22D and E).
A
70
Resp. Diff. [RU]
60
50
20 µM CodY
15 µM CodY
5 µM CodY
3 µM CodY
1 µM CodY
0.5 µM CodY
40
30
20
10
0
0
50
100
150
200
Time [s]
250
300
350
400
B
120
Resp. Diff. [RU]
100
40 µM CodY
20 µM CodY
15 µM CodY
10 µM CodY
5 µM CodY
2 µM CodY
1 µM CodY
80
60
40
20
0
0
50
100
150
200
250
300
350
400
Time [s]
Figure 5.22: Quantitative analysis of CodY and RNAP interactions by SPR
The sensorgrams show titrations of (A) immobilized αNTD with CodY and (B) immobilized αCTD with
CcpA. Concentrations of the analyte are depicted on the right side next to each sensorgram.
68
RESULTS
5.8 SPR analyses of immobilized CcpA with HPrSer46P and αNTD
The interactions of CcpA with αNTD, αCTD and CodY, respectively, were observed in SPR
measurements only when CcpA was added with increasing concentrations. When CcpA was
coupled to the chip via amine coupling no response was obtained upon addition of αNTD, αCTD
and CodY suggesting that CcpA the respective interaction surface of CcpA was not accessible.
However, the titrations described in chapter 5.7 were not performed with higher concentrations of
CcpA due to insufficient solubility of CcpA at concentrations above 40 µM. Therefore,
immobilization of CcpA via tag on an SPR chip could be an alternative strategy.
5.8.1
SPR analysis of Strep-tagged CcpA
In a first approach Strep-tagged CcpA was coupled on a StrepMAB-Immo antibody coated CM5
chip according to the method described by Schmidt et al. (Schmidt & Skerra, 2007) (Fig. 5.23A).
First, the ccpA gene was cloned into the pASK-IBA vector, which expresses CcpA with an Nterminal Strep-tag and was named pWH2375. Then, StrepCcpA was purified from E. coli
BL21(DE3) / pWH2375 to homogeneity. After coating of a CM5 chip with a StrepMAB-Immo
antibody (Fig. 5.23B) purified StrepCcpA was captured. However, StrepCcpA dissociated rapidly
from the surface (Fig. 5.23C) and, therefore, this method was not suitable for analysis of
interactions between CcpA and interaction partners.
RESULTS
69
B
30000
A
CM5 chip
Coupling of
StrepMAB-Immo Ab
Y
Y
Y
Y
Resp. Diff. [RU]
25000
Y
Y
Saturation
with
ethanolamine
Injection of
StrepMABImmo Ab
5000
Time [s]
4000
3500
Addition of analyte
(interaction partner)
3000
Resp. Diff. [RU]
Y
Y
Activation
with
NHS + EDC
10000
0 200 400 600 800 100012001400 1600 1800 2000
C
Y
15000
0
Immobilization of
ligand (Streptagged protein)
Y
20000
500 nM StrepCcpA
2500
2000
1500
1000
500
0nM StrepCcpA
0
0
100 200 300 400 500 600 700 800 900 1000
Time [s]
Figure 5.23: Strategy for immobilization of Strep-tagged CcpA on an SPR chip and coupling of
StrepCcpA on an StrepMAB-Immo coated SPR chip
This figures shows the strategy for immobilization of a Strep-tagged protein on an SPR chip (A). A CM5
chip is coated with a StrepMAB-Immo antibody via amine coupling, before the Strep-tagged protein
(ligand) is immobilized on the chip. Then binding of the analyte to the ligand is analyzed. (B) The
sensorgram shows the sensorgram of the amide coupled StrepMAB-Immo Ab in FC2 of a CM5 chip. (C)
This sensorgram shows the immobilization of 500 nM StrepCcpA on the StrepMAB-Immo coated CM5
chip.
70
5.8.2
RESULTS
Immobilization of CcpA via Avi-tag on an SPR chip
5.8.2.1 Enzymatic biotinylation of proteins in vivo based on BirA
Biotin is able to bind with a high affinity and specificity to avidin or streptavidin (KD = 10-15 M),
which is the strongest non-covalent binding known (Green, 1990). Due to this high affinity and
specificity of the biotin-avidin interaction, biotinylation of biomolecules has been exploited for a
large number of biotechnological applications, like immobilization, purification and detection of
proteins (Cognet et al., 2005; Parrott & Barry, 2000).
Chemical biotinylation of proteins is not site specific and might lead to inactivation of some
biological molecules (Bayer & Wilchek, 1990; Jokiranta & Meri, 1993). In contrast, enzymatic
biotinylation of the Avi-tag mimics the physiological biotinylation of proteins using a biotin
accepting peptide (Cull & Schatz, 2000). The Avi-tag is a 15 amino acid biotin accepting peptide
(Schatz, 1993), which contains an artificial biotinylation site for the biotin protein ligase BirA of
E. coli (Beckett et al., 1999) (Fig. 5.24A). BirA catalyzes the covalent attachment of biotin to the
ε amino group of the lysine residue (Eisenberg et al., 1982) within the Avi-tag (Schatz, 1993) in
an ATP-dependent reaction (Barker & Campbell, 1981; Chapman-Smith & Cronan, 1999a;
Chapman-Smith & Cronan, 1999b) (Fig. 5.24B). When the Avi-tag is fused to a protein, it is
possible to biotinylate the fusion protein in vivo (Cronan, 1990; Cronan & Reed, 2000).
A
B
Avitag Sequence: GLNDIFEAQKIEWHE
Biotin + ATP
biotinyl-5′-AMP + apo-protein
biotinyl-5′-AMP + PPi
(1)
biotinylated holo-protein + AMP
(2)
Figure 5.24: Avi-tag sequence and the BirA-dependent biotinylation reaction
This figure shows the Avi-tag sequence which can be site specifically biotinylated at the lysine residue
(highlighted in red) (A) and the BirA-dependent covalent attachment of the biotin prosthetic group to a
specific lysine of the biotin carboxyl carrier domain of biotin-dependent carboxylases in a two-step reaction
(B).
RESULTS
71
5.8.2.2 Construction and purification of in vivo biotinylated Avi-tagged CcpA
For expression of Avi-tagged CcpA, the ccpA gene was cloned into pAC4 and E. coli AVB101
was transformed with the resulting plasmid pWH2370. The protein expression E. coli strain
AVB101 simultaneously expresses the Avi-tagged fusion protein and BirA (Fig. 5.25). BirA
recognizes the Avi-tag sequence and monobiotinylates specifically the central lysine of the Avitag in vivo in the presence of biotin (Beckett et al., 1999; Schatz, 1993).
Ptrc
CcpA
lacIQ
Ptac
BirA
Avitag
pBirAcm
pWH2370
~ 7.9kb
5.2kb
pMB1
CmR
AmpR
p15A
Figure 5.25: Scheme of the in vivo
biotinylation strain AVB101
The E. coli overexpression strain
AVB101 is shown schematically,
which expresses the IPTG inducible
plasmids pWH2370, which expresses
C-terminally Avi-tagged (blue arrow)
CcpA (red box) and pBirAcm
encoding the biotin protein ligase
BirA (green arrow).
Expression of BirA and Avi-tagged CcpA was simultaneuously induced by addition of IPTG and
D-biotin was added as a substrate for BirA at the same time. Biotinylated Avi-tagged CcpA was
then purified by affinity chromatography using a monomeric avidin-matrix which allows the
elution of biotinylated molecules by competitive elution with free biotin. The pure protein
fractions were then subsequently applied to a size exclusion chromatography to remove the
retaining biotin and to equilibrate the protein in HBSE buffer as described in chapter 4.3.4.13.
5.8.2.3 SPR measurements of Avi-tagged CcpA with HPrSer46P and αNTD
For qualitative SPR analysis, about 3,000 RU neutravidine were bound to a CM5 chip via amine
coupling. Neutravidine is a streptavidin analog, which avoids non-specific accumulated of
charged proteins like HPrSer46P in the chip matrix. After that, about 1,000 RU biotinylated
C-terminally Avi-tagged CcpA was captured stepwise (Fig. 5.26A). Injections with increasing
concentrations (from 1 to 50 µM) of HPrSer46P led to increasing responses (Fig. 5.26B) and
confirmed the results of previous analyses using amide coupling (Horstmann et al., 2007; Seidel
et al., 2005). Furthermore, the shape and the increase in signal height of the sensorgrams of
injections with increasing concentrations of αNTD (from 1 µM to 20 µM) indicate highly specific
binding of αNTD to immobilized CcpA (Fig. 5.26C). These titrations confirmed the results
obtained by titrations of amine coupled αNTD with CcpA (chapter 5.7.1).
72
RESULTS
A
B
1200
120
Resp. Diff. [RU]
Resp. Diff. [RU]
1000
800
600
400
200
100
200
300 400
Time [s]
Titration of immobilized
100 biotinylated C-terminally
Avi-tagged CcpA
500
600
80
60
40
20
0
700
50 µM αNTD
20 µM αNTD
10 µM αNTD
5 µM αNTD
1 µM αNTD
0 µM αNTD
50
0
D
Resp. Diff. [RU]
0
Resp. Diff. [RU]
100
0
0
C
50 µM HPrS46P
20 µM HPrS46P
10 µM HPrS46P
5 µM HPrS46P
1 µM HPrS46P
0 µM HPrS46P
140
50
100
Time [s]
Titration of immobilized
150 biotinylated N-terminally
Avi-tagged CcpA
100
150
200
100 µM αNTD
10 µM αNTD
0 µM αNTD
50
0
-50
-100
0
50
100 150 200
Time [s]
250
300 350
0
50
100
Time [s]
150
200
Figure 5.26: SPR analysis of AviTagged-CcpA with HPrSer46P and αNTD
The sensorgrams shows the immobilization of 250 nM C-terminally Avi-tagged CcpA to a neutravidine
coated CM5 chip (A). The sensorgram (B) shows titrations of immobilized C-terminally Avi-tagged CcpA
with HPrSer46P. The sensorgrams (C) and (D) show titrations of immobilized C-terminally Avi-tagged
CcpA with αNTD (C) and of immobilized N-terminally Avi-tagged CcpA with αNTD (D). Concentrations
of the analytes are depicted on the right side next to each sensorgram.
While titrations of N-terminally Avi-tagged CcpA with HPrSer46P yielded almost equal results
for CcpA interaction (data not shown), injections with increasing concentrations of αNTD (from
1 µM to 100 µM) did not yield any response (Fig. 5.26D).
DISCUSSION
73
6 Discussion
6.1 SPINE analysis of StrepCcpA
The global transcription regulator CcpA is a multifaceted protein. It represses or activates a large
number of genes and is controlled by a diverse set of phosphoproteins and small molecules
(Deutscher, 2008; Deutscher et al., 2006; Fujita, 2009; Moreno et al., 2001; Sonenshein, 2007).
This versatility generates different regulatory outputs and quite a few of those are not understood
completely at the molecular level. In particular, differential regulation of genes and the molecular
mechanism for activation of genes by CcpA have not been studied in detail yet and both aspects
suggest the presence of further unknown interaction partners for CcpA. The first approach by
which this was addressed in this work was a SPINE analysis in order to identify interaction
partners of CcpA in vivo. The addition of formaldehyde to growing cultures caused efficient
crosslinking of biomolecules and the approach was specific for Strep-tagged complexes since in
the ∆ccpA mutant strain WH1124, lacking a Strep-tag, hardly any protein was copurified. Strepaffinity purified CcpA complexes from crosslinked lysates contained a high number of different
proteins whereas only a few proteins were copurified in the same experiment with
non-crosslinked cultures. Interestingly, in the SDS-PAGE analysis of these SPINE eluates the
protein patterns and the band intensities were similar for samples from cultures grown with or
without glucose. These crosslinked elution fractions were analyzed by MS in collaboration with
Dr. Elke Hammer and Prof. Dr. Uwe Völker (Ernst-Moritz-Arndt-Universität Greifswald). As
formaldehyde connects biomolecules unspecifically, SPINE was performed with mixtures of
formaldehyde treated
15
N and untreated
14
N labeled cells according to Dreisbach et al. as
described in chapter 5.2 to distinguish specifically crosslinked proteins from non-specifically
crosslinked ones (Dreisbach et al., 2008). The MS analysis of the crosslinked elution fractions
from cells grown with glucose revealed 619 proteins. The application of the selection criteria
described in chapter 5.3 reduced the number of potential interaction partners to 222 proteins
identified by MS. From these potential interactions partners 143 were only detected in crosslinked
elution fractions, such as HPr and CodY, indicating either a weak binding to CcpA or interaction
dynamics with fast off rates of the interactants. 38 proteins were not only detected in all
crosslinked elution fractions but also in all non-crosslinked ones pointing to a stronger binding or
a binding with a slow off rate. The remaining 41 proteins were detected in all crosslinked elution
fractions and in some but not all non-crosslinked elution fractions as well.
74
DISCUSSION
The identified interaction partners can be subdivided into four functional categories: (1)
Functionally related interaction partners were the subunits of the RNAP (RpoA, RpoB, RpoC),
transcriptional regulators like CodY, TenA or CheV. (2) Enzymes involved in sugar transport
(including HPr), glycolysis and tricarboxylic acid cycle. (3) Proteins involved in biotin and fatty
acid biosynthesis, amino acid metabolism, purine and pyrimidine synthesis. (4) Proteins involved
in sporulation, competence development and, unexpectedly, proteins involved in cell division and
were also detected. Since formaldehyde crosslinks proteins with DNA, too, it is also possible that
the non-specific DNA binding proteins and some of the other putative interaction partners
detected by MS might result from indirect crosslinking with protein-DNA complexes located in
close vicinity to DNA bound CcpA. Furthermore, SPINE-MS revealed also 33 proteins with yet
unknown function. Considering the important role of CcpA in B. subtilis physiology it is not
surprising that a large number of putative CcpA interaction partners were detected by MS after
SPINE. However, SPINE-MS identified also ribosomal proteins, proteases, chaperones and stress
proteins that are likely crosslinked with CcpA due to contacts during translation, maintenance and
degradation of CcpA. Since formaldehyde crosslinks proteins with DNA, too, it is also possible
that the non-specific DNA binding proteins and some of the other putative interaction partners
detected by MS might result from indirect crosslinking with protein-DNA complexes located in
close vicinity to DNA bound CcpA. Therefore, many of the proteins listed in Table A1
(Appendix) (data from SPINE eluates of WH1012 with and without glucose derive from MS
analysis performed in collaboration with Dr. Elke Hammer and Prof. Dr. Uwe Völker, ErnstMoritz-Arndt-Universität Greifswald) originate from contacts which do not influence the
regulatory activity of CcpA, however, not all proteins are functionally relevant. Consequently, for
detailed analyses the interaction partners of CcpA listed in the Table A1 (Appendix) should be
carefully preselected based on physiological but also on experimental data indicating an
involvement in CcpA-dependent regulation.
Interestingly, the MS analysis of elution fractions from SPINE did not reveal the known
interaction partners like HPr or HPrSer46P as the most abundant proteins, even though the
HPrSer46P-CcpA interaction is well established and has been characterized in many other studies
(Horstmann et al., 2007; Reizer et al., 1996; Schumacher et al., 2004; Seidel et al., 2005).
However, immunoblotting clearly proved the presence of HPr in the SPINE eluates and thus
indicated that HPr was likely missing at least partially in the MS analysis. This is very likely due
to the low molecular weight (88 amino acids) of HPr and the corresponding small number of
specific peptides might explain the remarkable low number of identified peptides in MS for HPr
or HPrSer46P. Immunoblotting identified HPr in SPINE elutions from cells grown in medium
supplemented with glucose but not in a ptsH1 mutant. Complex formation in the presence, but not
DISCUSSION
75
in the absence of glucose, as well as the lack of StrepCcpA-HPrSer46Ala complexes in SPINE
eluates is consistent with previous in vivo and in vitro studies (Fujita, 2009; Stülke & Hillen,
2000).
Although, HPrSer46P is the main coeffector, CrhSer46P also leads to the stimulation of CcpA
binding to cre (Galinier et al., 1997). However, Crh was not detected in SPINE eluates, neither in
the wildtype strain nor in the ptsH1 mutant strain. This agrees with studies which showed that a
ptsH1 mutant encoding for HPrS46A exhibits strongly reduced CCR at many genes as CrhSer46P
can only partially substitute for HPrSer46P (Galinier et al., 1999; Galinier et al., 1997; Zeng et
al., 2000). Thus there are obviously less CcpA-CrhP complexes. There are only few specific
peptides for the small Crh protein (85 amino acids) available and thus the determination of Crh is
even more difficult than detection of HPr. Thus, the number of Crh specific peptides might be
below the selection criteria and, consequently, Crh was not detected in MS analysis. This is in
good agreement with the results of the SPINE immunoblot analysis in which a signal for Crh was
not observed, too. Furthermore, the initiation of a glycolytic bypass was identified as an
exclusively Crh specific but CcpA-independent function of Crh (Landmann et al., 2011;
Landmann et al., 2012). The methylglyoxal synthase MgsA is inhibited by interaction with Crh,
however, CrhSerP abolishes this inhibitory interaction and thereby prevents accumulation of
phosphorylated glycolytic intermediates in the cell. However, since CrhSer46P binds CcpA in
vitro and CrhSer46P substitutes HPrSer46P in vivo at least partially in an HPrSer46Ala mutant
(Galinier et al., 1997; Singh et al., 2008), it is not clear how or under which conditions CrhSer46P
is important for CcpA-dependent CCR.
6.2 Binary protein-protein interactions analyzed by BACTH
In SPINE in vivo crosslinking is mediated by formaldehyde. Because formaldehyde reacts with
amino and imino groups of proteins and nucleic acids (Das et al., 2004), indirect binding of
proteins via DNA and RNA, can be misinterpreted for direct binding (Spencer et al., 2003).
Therefore, it is very likely that some of the putative interaction partners detected by MS might
form complexes with proteins located in close vicinity to DNA bound by CcpA and consequently
a BACTH system was applied to verify binary protein-protein interactions.
In contrast to enzymes involved in glycolysis, CcpA represses the synthesis of almost all the TCA
cycle enzymes in the presence of glucose (Tobisch et al., 1999a). These genes are derepressed by
growth of the cells in medium containing glutamate or ribose (Sonenshein, 2007). While CcpA
represses transcription of citZ directly, the expression of citB is obviously indirectly repressed by
CcpA (Blencke et al., 2003; Kim et al., 2002). Interestingly, in SPINE eluates several TCA cylce
76
DISCUSSION
enzymes were identified with a large number of specific peptides. BACTH analyses indicated that
CcpA interacts weakly with CitZ, Icd and Mdh. Combinations of CcpA with CitB and CitG
showed no interaction with CcpA in BACTH assays. In the presence of glucose CcpA activates
the transcription of genes of the overflow metabolism and those for utilization of carbon sources
of the TCA cycle are repressed. Recently, Meyer et al. detected protein-protein interactions
among TCA enzymes and suggested a TCA cycle metabolon (Meyer et al., 2011). In presence of
glucose CcpA represses citZ transcription (Sonenshein, 2007) and additionally CcpA might
interact with Mdh, Icd and CitZ, the core TCA metabolon. However, there is no evidence for a
specific role of CcpA-TCA metabolon interactions and the function of CcpA interactions with
TCA cycle enzymes remains unclear.
Although CodY was identified in SPINE eluates by MS and immunoblotting in cells grown with
and without glucose, the BACTH analyses did not show any interaction of CodY with CcpA and
RpoA and of CcpA and αCTD, respectively. However, the interaction of CcpA and HPr and
HPrS46D, a mutant which binds CcpA, was also not detected by the BACTH assay (Liebl, 2009).
This might be due to steric hindrance or wrong orientation of the Cya domains in the fusion
proteins, preventing Cya dimerization and thus Cya activity. In one approach, linkers with
different lengths, 35 and 15 amino acids (Ün, 2009), respectively, were inserted between each Cya
fusion of either CcpA or HPrS46D, however, Cya activity was not restored (data not shown). It is
well known that only a fraction of the actual protein-protein interactions is detected by each
individual method for investigation of binary protein-protein interactions (Li et al., 2004; Rual et
al., 2005; Yu et al., 2008). False negatives in two-hybrid screens may occur out of several
reasons: (1) Misfolding of fusion proteins abolish the interactions (Strauch & Georgiou, 2007).
(2) The Cya domains cannot reconstitute for a functional enzyme in a complex with the fusion
proteins as their orientation in the fusion protein is insufficient for proper dimerization of the
fused Cya domains. Different protein sizes of the interaction partners or the physical distance of
the Cya domains can prevent an actual positive interaction. (3) The interaction requires
posttranslational modification which induce conformational changes on the interaction surfaces of
the proteins (Ito et al., 2000).
The orientation of the small 8 kDa αCTD in a complex with the 38 kDa CcpA monomer is likely
insufficient for proper dimerization of the fused Cya domains and might explain the negative
result in the BACTH assay. This might also explain the missing interaction of CcpA and HPr or
HPrS46D and also the negative results for interactions of CodY and CcpA or RpoA in the
BACTH assays.
DISCUSSION
77
6.3 Interactions of CcpA, CodY and RpoA involved in activation of ackA
Transcriptional activation in bacteria is mostly mediated by interactions of transcriptional
activators with different subdomains of the RNA polymerase and influences the recruitment of
RNA polymerase in the transcription initiation. These interactions increase RNA polymerase
binding to the promoter or facilitate the open complex formation (Browning & Busby, 2004;
Busby & Ebright, 1994). In E. coli several activators directly interact with at least one of each
subunit of the RNA polymerase (α, β, β′ and ω) (Hochschild & Dove, 1998). Only few activators
contact the β or the β′ subunits of the RNA polymerase (Kulkarni & Summers, 1999; Lee &
Hoover, 1995; Miller et al., 1997; Szalewska-Palasz et al., 1998). Most transcriptional activators
primarily interact with α or σ subunits (Busby & Ebright, 1994; Hochschild & Dove, 1998) such
as the transcriptional regulators CAP, FNR, IHF, FIS, NtrC and MarA in E. coli (Browning &
Busby, 2004). Interactions of activators with the αCTD stabilize the binding of the RNA
polymerase to the promoter (Gourse et al., 2000). In contrast the main function of the αNTD is the
dimerization of the RNA polymerase and activators binding to the αNTD mainly increase the rate
of isomerization (Ishihama, 1993; Niu et al., 1996) and is the target of only few activators (Niu et
al., 1996).
Among the interaction partners of CcpA, isolated by SPINE and identified by MS, the
transcriptional regulator CodY and all RNA polymerase subunits, RpoA, RpoB and RpoC were
observed. The SPINE-immunoblot analysis with an anti-RpoA antibody confirmed the presence
of RpoA in the elution fractions from in vivo crosslinking of cells grown with or without glucose.
In contrast to the data derived from SPINE, in the BACTH assays only the interaction of CcpA
with RpoA was detected, but not with RpoB and RpoC. This is likely due to the fact that
crosslinking in SPINE connects all proteins in heterooligomeric complexes, such as the RNA
polymerase subunits (α2ββ′), whereas the two-hybrid assay only detects binary interaction
partners. In more detail, the BACTH assay revealed interactions between CcpA and RpoA or the
αNTD and self-interactions of CcpA, RpoA and the αNTD due to their dimerization. The SPR
analysis confirmed binding of CcpA to the αNTD and detected additionally binding of CcpA to
the CTD of RpoA as well as interactions of CodY with both subunits of RpoA. These different
observations are not surprising because in SPR the analyte can bind in every possible orientation,
as long as the interaction surface of the ligand is exposed to the solvent whereas the BACTH
depends on a proper orientation of the interacting proteins. The interactions of CcpA and CodY
with the αNTD are unusual as transcriptional activators mainly interact with the different
activating regions of αCTD or with the σ subunit of RNA polymerase (Browning & Busby, 2004).
Interestingly, the only protein which is known so far to interact with the αNTD is the functional
78
DISCUSSION
homologue of CcpA in E. coli, the catabolite activating protein CAP (Browning & Busby, 2004;
Niu et al., 1996). In E. coli, CAP-αNTD contacts are involved in regulation of galactosidase
expression at the galP1 and melR promoters (Niu et al., 1996). Interestingly, in B. subtilis both
regulators, CcpA and CodY, interact with both the αCTD and the αNTD of the RNA polymerase.
The KDs of 5 and 8 µM of the CcpA complexes with the RpoA subunits are similar to that of CAP
with the αCTD and the αNTD in E. coli (Niu et al., 1996). The intracellular concentrations of
CcpA and RpoA are in the low micromolar range for CcpA (Miwa et al., 1994) 15 – 30 µM for
the RNAP holoenzyme (Doherty et al., 2010; Lopez de Saro et al., 1999). This combination of the
KDs and the intracellular concentrations of CcpA and RpoA suggests that a subpopulation of
CcpA forms complexes with the α subunit already before DNA binding, as it was demonstrated
for CAP, SoxS and MarA (Heyduk et al., 1993; Martin et al., 2002; Shah & Wolf, 2004). This is
also in good agreement with the result of SPINE because CcpA-RpoA complexes were detected in
eluates from cells grown with and without glucose. This result suggests that preformed complexes
of CcpA with RpoA or with the RNA polymerase holoenzyme are directed to a cre site for gene
activation after glucose-dependent binding of HPrSer46P. However, it is not clear if a
CcpA-RpoA complex can also bind to cre sites of repressed genes or if it plays any role in
repression. The SPR analyses in this study show also that CodY binds the αCTD as well as the
αNTD of RpoA at similar KDs like CcpA. This means that CodY is able to bind to RNA
polymerase independently from CcpA. This is in good agreement with studies on ackA expression
because, like CcpA, CodY alone is also able to activate ackA (Shivers et al., 2006). The latter
publication also demonstrated that cooperative activation of ackA by CcpA and CodY exceeds the
sum of the individual activation. This synergism might be due to the rather weak complex
formation between CodY and CcpA. The high KD for CcpA-CodY maintains individual control of
gene expression by either regulator because it prevents permanent binding to each other in the
cytoplasm. On the other hand, binding of both regulators to consecutive DNA binding sites in
close vicinity, e.g. in the ackA regulatory region, leads to an increased local concentration which
likely allows complex formation even for the high KD of 60 µM. This assumption is strongly
supported by the results from SPINE with cultures grown with and without glucose. CcpA binds
DNA in the presence of glucose. Consequently, CodY-CcpA complex formation occurred
strongly in cells grown with glucose and only marginally in the absence of glucose. Furthermore,
neither the αCTD nor the αNTD inhibited the binding of CcpA to CodY in SPR indicating that the
αCTD and the αNTD do not interact with CcpA at overlapping binding sites.
However, amine coupling of ligands has a commonly known great disadvantage, as by this
random immobilization method, the ligand is bound to the chip surface in various orientations.
The presence of lysine groups in the vicinity of the binding site on the protein surface might not
DISCUSSION
79
be suitable for coupling of this protein. The binding site might be blocked or hampered, thus
influencing the binding constants.
The biotin / streptavidin interaction is a high affinity, non-covalent binding which is also suitable
for immobilization of ligands on the sensor chip surface. Biotinylated molecues are coupled sitespecifically on the chip surface, due to the very strong interaction between streptavidin and a
biotinylated molecule (KD 10-15 M). In contrast to unspecific in vitro biotinylation systems, the
Avi-tag is site-specifically biotinylated by BirA. The biotinylated proteins can be immobilized
directly on streptavidin or neutravidine coated sensor chips. The major advantage of this
immobilization method over the amine coupling is that all immobilized proteins have the same
orientation due to site-directed coupling via the biotinylated Avi-tag. Therefore, CcpA was fused
to the Avi-tag because other interaction analyses with RpoA and CodY could be maintained on
one chip surface. Biotinylated C- and N-terminally Avi-tagged CcpA were able to bind
HPrSer46P suggesting that the HPrSer46P binding sites of CcpA are exposed in both N- and Cterminally coupled Avi-tagged CcpA. In contrast, only αNTD bound to C-terminally immobilized
but not to N-terminally coupled CcpA, indicating that the N-terminally immobilized protein is
blocked for αNTD binding. Thus, αNTD binding to CcpA might also overlap with the N-terminal
subdomain of CcpA.
Although, binding of HPrSer46P and of αNTD was detected qualitatively using biotinlyated CcpA
coupled to a neutravidine chip, this experimental approach was not used for analysis of CcpA
interactions because of the high “bulk effects” at higher concentrations of αNTD. The bulk effect
describes a signal offset due to a refractive index mismatch by contribution of the bulk solute
concentration to the SPR response that occurs in addition to the response which is clearly
observed as “peak” in the sensorgrams at the beginning and the end of an injection. Because this
bulk effect might impair the kinetic analyses which was not the case to the amine coupling of the
proteins on a CM5 chip, these measurements were not used for kinetic analyses.
In summary, the data is in good agreement with published in vivo analyses because only a
∆ccpA∆codY double mutant abolishes activation of ackA, the ∆ccpA and ∆codY single mutants
activate ackA intermediately whereas ackA activation in wildtype cells clearly exceeds ackA
expression of the single deletion mutants (Shivers et al., 2006). Moreover, in vitro analyses
showed CodY and CcpA high affinity binding to DNA motifs located -83 bp and -56.5 bp
upstream of the promoter, respectively (Grundy et al., 1993a; Shivers et al., 2006; Turinsky et al.,
1998). Taking all this into account the regulatory protein-DNA complex shown in Figure 6.1 for
cooperative initiation of transcription of ackA can be proposed. This assumption is supported as
follows: 1. CcpA and CodY interact both with the αNTD and the αCTD. 2. CcpA binds to the cre
close to the αNTD but CodY binds a distant DNA binding site. 3. In vitro, CcpA binds to CodY in
80
DISCUSSION
the absence of HPrSer46P. 4. αNTD and αCTD bind to CodY at a different binding site than
CcpA. 5. HPrSer46P points towards CodY in an HPrSer46P-CcpA-cre complex (Schumacher et
al., 2004) and presents an obstacle preventing CodY from directly contacting the αNTD. The most
likely orientation for CodY to form a complex with CcpA and the αNTD at the same time is,
therefore, to induce a bend in the DNA and to contact its interaction partners.
CodY
αCTD
αNTD
β´
β
HPrSer46P
σ
CcpA
-35
-10
Figure 6.1: Putative model for the transcriptional activation of ackA
Schematic representation of an arrangement of CcpA, CodY and the RNAP in a initiation complex which
involves all interactions detected in this study: blue CcpA homodimer; red: HPrSer46P; orange: CodY
homodimer; green homodimer of the α subtunit of the RNA polymerase, grey: β, β′ subunits of the
RNA polymerase; black: σ factor; grey boxes: DNA binding domains of CcpA (cre) and CodY.
In summary, CodY and RpoA as novel interaction partners of CcpA were identified and
characterized and a model for a transcription initiation complex was proposed. Further studies
may focus on the identification of residues, which are involved in the interaction.
Moreover, after careful preselection of the interaction partners of CcpA listed in Table A1
(Appendix) additional direct interaction partners of CcpA might be identified and analyzed in
detail.
REFERENCES
81
7 References
Arantes, O. & Lereclus, D. (1991). Construction of cloning vectors for Bacillus thuringiensis. Gene
108, 115-119.
Ausubel, F. M., Brent, R., Kingston, R. E., Moore, D. D., Seidman, J. G., Smith, J. A. & Struhl,
K. (1994). Current Protocols in Molecular Biology: New York: Greene Publishing Assoziates.
Bachem, S. & Stülke, J. (1998). Regulation of the Bacillus subtilis GlcT antiterminator protein by
components of the phosphotransferase system. J Bacteriol 180, 5319-5326.
Barbe, V., Cruveiller, S., Kunst, F., Lenoble, P., Meurice, G., Sekowska, A., Vallenet, D., Wang,
T., Moszer, I., Medigue, C. & Danchin, A. (2009). From a consortium sequence to a unified
sequence: the Bacillus subtilis 168 reference genome a decade later. Microbiology 155, 1758-1775.
Barker, D. F. & Campbell, A. M. (1981). The birA gene of Escherichia coli encodes a biotin
holoenzyme synthetase. J Mol Biol 146, 451-467.
Bartholomae, M. (2008). Identifizierung und Signaltransduktion der Interaktionspartner der
Kohlenstoffkatabolitenregulation in Bacillus subtilis. Lehrstuhl für Mikrobiologie, Diplomarbeit,
Friedrich-Alexander-Universität Erlangen-Nürnberg.
Bayer, E. A. & Wilchek, M. (1990). Protein biotinylation. Methods Enzymol 184, 138-160.
Beckett, D., Kovaleva, E. & Schatz, P. J. (1999). A minimal peptide substrate in biotin holoenzyme
synthetase-catalyzed biotinylation. Protein Sci 8, 921-929.
Bettenbrock, K., Sauter, T., Jahreis, K., Kremling, A., Lengeler, J. W. & Gilles, E. D. (2007).
Correlation between growth rates, EIIACrr phosphorylation, and intracellular cyclic AMP levels in
Escherichia coli K-12. J Bacteriol 189, 6891-6900.
Blencke, H. M., Homuth, G., Ludwig, H., Mäder, U., Hecker, M. & Stülke, J. (2003).
Transcriptional profiling of gene expression in response to glucose in Bacillus subtilis: regulation of
the central metabolic pathways. Metab Eng 5, 133-149.
Bradford, M. (1976). A rapid and sensitive method for quantitation of microgram quantities of
protein utilizing the principle of protein-dye binding. Anal. Biochem. 72, 248-254.
Browning, D. F. & Busby, S. J. (2004). The regulation of bacterial transcription initiation. Nat Rev
Microbiol 2, 57-65.
Brückner, R. & Titgemeyer, F. (2002). Carbon catabolite repression in bacteria: choice of the carbon
source and autoregulatory limitation of sugar utilization. FEMS Microbiol Lett 209, 141-148.
Busby, S. & Ebright, R. H. (1994). Promoter structure, promoter recognition, and transcription
activation in prokaryotes. Cell 79, 743-746.
Busby, S. & Ebright, R. H. (1999). Transcription activation by catabolite activator protein (CAP). J
Mol Biol 293, 199-213.
82
REFERENCES
Cases, I., Velazquez, F. & de Lorenzo, V. (2007). The ancestral role of the phosphoenolpyruvatecarbohydrate phosphotransferase system (PTS) as exposed by comparative genomics. Res Microbiol
158, 666-670.
Chambliss, G. H. (1993). Carbon source-mediated catabolite repression. In Bacillus subtilis and other
Gram-positive bacteria, (ed. A. L. Sonenshein J. A. Hoch & R. Losick), pp. 213-218. Washington
D.C.: American Society for Microbiology.
Chapman-Smith, A. & Cronan, J. E., Jr. (1999a). The enzymatic biotinylation of proteins: a posttranslational modification of exceptional specificity. Trends Biochem Sci 24, 359-363.
Chapman-Smith, A. & Cronan, J. E., Jr. (1999b). Molecular biology of biotin attachment to
proteins. J Nutr 129, 477S-484S.
Choi, S. K. & Saier, M. H., Jr. (2005). Regulation of sigL expression by the catabolite control
protein CcpA involves a roadblock mechanism in Bacillus subtilis: potential connection between
carbon and nitrogen metabolism. J Bacteriol 187, 6856-6861.
Cognet, I., Guilhot, F., Gabriac, M., Chevalier, S., Chouikh, Y., Herman-Bert, A., Guay-Giroux,
A., Corneau, S., Magistrelli, G., Elson, G. C., Gascan, H. & Gauchat, J. F. (2005). Cardiotrophinlike cytokine labelling using BirA biotin ligase: a sensitive tool to study receptor expression by
immune and non-immune cells. J Immunol Methods 301, 53-65.
Cronan, J. E., Jr. (1990). Biotination of proteins in vivo. A post-translational modification to label,
purify, and study proteins. J Biol Chem 265, 10327-10333.
Cronan, J. E., Jr. & Reed, K. E. (2000). Biotinylation of proteins in vivo: a useful posttranslational
modification for protein analysis. Methods Enzymol 326, 440-458.
Crutz, A. M., Steinmetz, M., Aymerich, S., Richter, R. & Le Coq, D. (1990). Induction of
levansucrase in Bacillus subtilis: an antitermination mechanism negatively controlled by the
phosphotransferase system. J Bacteriol 172, 1043-1050.
Cull, M. G. & Schatz, P. J. (2000). Biotinylation of proteins in vivo and in vitro using small peptide
tags. Methods Enzymol 326, 430-440.
Darbon, E., Ito, K., Huang, H. S., Yoshimoto, T., Poncet, S. & Deutscher, J. (1999). Glycerol
transport and phosphoenolpyruvate-dependent enzyme I- and HPr- catalysed phosphorylation of
glycerol kinase in Thermus flavus. Microbiology 145, 3205-3212.
Darbon, E., Servant, P., Poncet, S. & Deutscher, J. (2002). Antitermination by GlpP, catabolite
repression via CcpA and inducer exclusion triggered by P-GlpK dephosphorylation control Bacillus
subtilis glpFK expression. Mol Microbiol 43, 1039-1052.
Das, P. M., Ramachandran, K., vanWert, J. & Singal, R. (2004). Chromatin immunoprecipitation
assay. Biotechniques 37, 961-969.
Débarbouillé, M., Arnaud, M., Fouet, A., Klier, A. & Rapoport, G. (1990). The sacT gene
regulating the sacPA operon in Bacillus subtilis shares strong homology with transcriptional
antiterminators. J. Bacteriol. 172, 3966-3973.
Débarbouillé, M., Martin-Verstraete, I., Klier, A. & Rapoport, G. (1991a). The levanase regulator
LevR of Bacillus subtilis has domains homologous to both σ54- and PTS-dependent regulators. Proc
Natl Acad Sci USA 88, 2212-2216.
REFERENCES
83
Débarbouillé, M., Martin-Verstraete, I., Klier, A. & Rapoport, G. (1991b). The transcriptional
regulator LevR of Bacillus subtilis has domains homologous to both σ54- and phosphotransferase
system-dependent regulators. Proc Natl Acad Sci USA 88, 2212-2216.
Dedon, P. C., Soults, J. A., Allis, C. D. & Gorovsky, M. A. (1991). A simplified formaldehyde
fixation and immunoprecipitation technique for studying protein-DNA interactions. Anal Biochem
197, 83-90.
Deutscher, J. (2008). The mechanisms of carbon catabolite repression in bacteria. Curr Opin
Microbiol 11, 87-93.
Deutscher, J., Fischer, C., Charrier, V., Galinier, A., Lindner, C., Darbon, E. & Dossonnet, V.
(1997). Regulation of carbon metabolism in gram-positive bacteria by protein phosphorylation. Folia
Microbiol 42, 171-178.
Deutscher, J., Francke, C. & Postma, P. W. (2006). How phosphotransferase system-related protein
phosphorylation regulates carbohydrate metabolism in bacteria. Microbiol Mol Biol Rev 70, 939-1031.
Deutscher, J., Küster, E., Bergstedt, U., Charrier, V. & Hillen, W. (1995). Protein kinasedependent HPr/CcpA interaction links glycolytic activity to carbon catabolite repression in grampositive bacteria. Mol Microbiol 15, 1049-1053.
Deutscher, J., Reizer, J., Fischer, C., Galinier, A., Saier Jr., M. H. & Steinmetz, M. (1994). Loss
of protein kinase-catalyzed phosphorylation of HPr, a phosphocarrier protein of the
phosphotransferase system, by mutation of the ptsH gene confers catabolite repression resistance to
several catabolic genes of Bacillus subtilis. J. Bacteriol. 176, 3336-3344.
Diel,
M.
(2005).
Fluoreszenzspektroskopische
Analyse
der
Interaktionen
von
Einzeltryptophanmutanten des Katabolit-Kontrollproteins CcpA aus Bacillus subtilis mit Cofaktoren
und cre. Lehrstuhl für Mikrobiologie, Doktorarbeit, Friedrich-Alexander Universität ErlangenNürnberg.
Doherty, G. P., Fogg, M. J., Wilkinson, A. J. & Lewis, P. J. (2010). Small subunits of RNA
polymerase: localization, levels and implications for core enzyme composition. Microbiology 156,
3532-3543.
Dreisbach, A., Otto, A., Becher, D., Hammer, E., Teumer, A., Gouw, J. W., Hecker, M. &
Völker, U. (2008). Monitoring of changes in the membrane proteome during stationary phase
adaptation of Bacillus subtilis using in vivo labeling techniques. Proteomics 8, 2062-2076.
Eisenberg, M. A., Prakash, O. & Hsiung, S. C. (1982). Purification and properties of the biotin
repressor. A bifunctional protein. J Biol Chem 257, 15167-15173.
Favier, A., Brutscher, B., Blackledge, M., Galinier, A., Deutscher, J., Penin, F. & Marion, D.
(2002). Solution structure and dynamics of Crh, the Bacillus subtilis catabolite repression HPr. J Mol
Biol 317, 131-144.
Fisher, S. H., Rohrer, K. & Ferson, A. E. (1996). Role of CodY in regulation of the Bacillus subtilis
hut operon. J Bacteriol 178, 3779-3784.
Fuchsbauer, N. (2003). Zur molekularen Funktion des Regulatorproteins CcpA aus Bacillus subtilis.
Lehrstuhl für Mikrobiologie, Doktorarbeit, Friedrich-Alexander Universität Erlangen-Nürnberg.
84
REFERENCES
Fujita, Y. (2009). Carbon catabolite control of the metabolic network in Bacillus subtilis. Biosci
Biotechnol Biochem 73, 245-259.
Galinier, A., Deutscher, J. & Martin-Verstraete, I. (1999). Phosphorylation of either Crh or HPr
mediates binding of CcpA to the Bacillus subtilis xyn cre and catabolite repression of the xyn operon. J
Mol Biol 286, 307-314.
Galinier, A., Haiech, J., Kilhofer, M.-C., Jaquinod, M., Stülke, J., Deutscher, J. & MartinVerstraete, I. (1997). The Bacillus subtilis crh gene encodes a HPr-like protein involved in carbon
catabolite repression. Proc Natl Acad Sci USA 94, 8439-8444.
Galinier, A., Kravanja, M., Engelmann, R., Hengstenberg, W., Kilhoffer, M. C., Deutscher, J. &
Haiech, J. (1998). New protein kinase and protein phosphatase families mediate signal transduction in
bacterial catabolite repression. Proc Natl Acad Sci USA 95, 1823-1828.
Görke, B., Fraysse, L. & Galinier, A. (2004). Drastic differences in Crh and HPr synthesis levels
reflect their different impacts on catabolite repression in Bacillus subtilis. J Bacteriol 186, 2992-2995.
Görke, B. & Stülke, J. (2008). Carbon catabolite repression in bacteria: many ways to make the most
out of nutrients. Nat Rev Microbiol 6, 613-624.
Gourse, R. L., Ross, W. & Gaal, T. (2000). UPs and downs in bacterial transcription initiation: the
role of the α subunit of RNA polymerase in promoter recognition. Mol Microbiol 37, 687-695.
Green, N. M. (1990). Avidin and streptavidin. Methods Enzymol 184, 51-67.
Grundy, F. J., Waters, D. A., Allen, S. H. & Henkin, T. M. (1993a). Regulation of the Bacillus
subtilis acetate kinase gene by CcpA. J Bacteriol 175, 7348-7355.
Grundy, F. J., Waters, D. A., Takova, T. Y. & Henkin, T. M. (1993b). Identification of genes
involved in utilization of acetate and acetoin in Bacillus subtilis. Mol Microbiol 10, 259-271.
Gunnewijk, M. G., van den Bogaard, P. T., Veenhoff, L. M., Heuberger, E. H., de Vos, W. M.,
Kleerebezem, M., Kuipers, O. P. & Poolman, B. (2001). Hierarchical control versus autoregulation
of carbohydrate utilization in bacteria. J Mol Microbiol Biotechnol 3, 401-413.
Hammer, E., Goritzka, M., Ameling, S., Darm, K., Steil, L., Klingel, K., Trimpert, C., Herda, L.
R., Dörr, M., Kroemer, H. K., Kandolf, R., Staudt, A., Felix, S. B. & Völker, U. (2011).
Characterization of the human myocardial proteome in inflammatory dilated cardiomyopathy by labelfree quantitative shotgun proteomics of heart biopsies. J Proteome Res 10, 2161-2171.
Hanson, R. S. & Cox, D. P. (1967). Effect of different nutritional conditions on the synthesis of
tricarboxylic acid cycle enzymes. J Bacteriol 93, 1777-1787.
Henkin, T., Grundy, F., Nicholson, W. & Chambliss, G. (1991). Catabolite repression of α-amylase
gene expression in Bacillus subtilis involves a trans-acting gene product homologous to the
Escherichia coli lacI and galR repressors. Mol Microbiol 5, 575-584.
Herzberg, C., Weidinger, L. A., Dörrbecker, B., Hübner, S., Stülke, J. & Commichau, F. M.
(2007). SPINE: a method for the rapid detection and analysis of protein-protein interactions in vivo.
Proteomics 7, 4032-4035.
Heyduk, T., Lee, J. C., Ebright, Y. W., Blatter, E. E., Zhou, Y. & Ebright, R. H. (1993). CAP
interacts with RNA polymerase in solution in the absence of promoter DNA. Nature 364, 548-549.
REFERENCES
85
Hochschild, A. & Dove, S. L. (1998). Protein-protein contacts that activate and repress prokaryotic
transcription. Cell 92, 597-600.
Hogema, B. M., Arents, J. C., Bader, R., Eijkemans, K., Yoshida, H., Takahashi, H., Aiba, H. &
Postma, P. W. (1998). Inducer exclusion in Escherichia coli by non-PTS substrates: the role of the
PEP to pyruvate ratio in determining the phosphorylation state of enzyme IIAGlc. Mol Microbiol 30,
487-498.
Horstmann, N. (2006). Influence of various coeffectors on differential carbon catabolite regulation
exerted by CcpA. Lehrstuhl für Mikrobiologie, Doktorarbeit, Friedrich-Alexander Universität
Erlangen-Nürnberg.
Horstmann, N., Seidel, G., Aung-Hilbrich, L. M. & Hillen, W. (2007). Residues His-15 and Arg-17
of HPr participate differently in catabolite signal processing via CcpA. J Biol Chem 282, 1175-1182.
Hueck, C. J., Kraus, A., Schmiedel, D. & Hillen, W. (1995). Cloning, expression and functional
analyses of the catabolite control protein CcpA from Bacillus megaterium. Mol. Microbiol. 16, 855864.
Inoue, H., Nojima, H. & Okayama, H. (1990). High efficiency transformation of Escherichia coli
with plasmids. Gene 96, 23-28.
Ishihama, A. (1993). Protein-protein communication within the transcription apparatus. J Bacteriol
175, 2483-2489.
Ito, T., Tashiro, K., Muta, S., Ozawa, R., Chiba, T., Nishizawa, M., Yamamoto, K., Kuhara, S. &
Sakaki, Y. (2000). Toward a protein-protein interaction map of the budding yeast: A comprehensive
system to examine two-hybrid interactions in all possible combinations between the yeast proteins.
Proc Natl Acad Sci USA 97, 1143-1147.
Jault, J. M., Fieulaine, S., Nessler, S., Gonzalo, P., Di Pietro, A., Deutscher, J. & Galinier, A.
(2000). The HPr kinase from Bacillus subtilis is a homo-oligomeric enzyme which exhibits strong
positive cooperativity for nucleotide and fructose 1,6-bisphosphate binding. J Biol Chem 275, 17731780.
Jokiranta, T. S. & Meri, S. (1993). Biotinylation of monoclonal antibodies prevents their ability to
activate the classical pathway of complement. J Immunol 151, 2124-2131.
Jones, B. E., Dossonnet, V., Küster, E., Hillen, W., Deutscher, J. & Klevit, R. E. (1997). Binding
of the catabolite repressor protein CcpA to its DNA target is regulated by phosphorylation of its
corepressor HPr. J Biol Chem 272, 26530-26535.
Jourlin-Castelli, C., Mani, N., Nakano, M. M. & Sonenshein, A. L. (2000). CcpC, a novel regulator
of the LysR family required for glucose repression of the citB gene in Bacillus subtilis. J Mol Biol 295,
865-878.
Karimova, G., Pidoux, J., Ullmann, A. & Ladant, D. (1998). A bacterial two-hybrid system based
on a reconstituted signal transduction pathway. Proc Natl Acad Sci USA 95, 5752-5756.
Kim, H. J., Roux, A. & Sonenshein, A. L. (2002). Direct and indirect roles of CcpA in regulation of
Bacillus subtilis Krebs cycle genes. Mol Microbiol 45, 179-190.
Kim, J. H., Yang, Y. K. & Chambliss, G. H. (2005). Evidence that Bacillus catabolite control
protein CcpA interacts with RNA polymerase to inhibit transcription. Mol Microbiol 56, 155-162.
86
REFERENCES
Kotrba, P., Inui, M. & Yukawa, H. (2001). Bacterial phosphotransferase system (PTS) in
carbohydrate uptake and control of carbon metabolism. J Biosci Bioeng 92, 502-517.
Kraus, A., Hueck, C., Gärtner, D. & Hillen, W. (1994). Catabolite repression of the Bacillus subtilis
xyl operon involves a cis element functional in the context of an unrelated sequence, and glucose
exerts an additional xylR-dependent repression. J Bacteriol 176, 1738-1745.
Krüger, S. & Hecker, M. (1995). Regulation of the putative bglPH operon for aryl-beta-glucoside
utilization in Bacillus subtilis. J Bacteriol 177, 5590-5597.
Kulkarni, R. D. & Summers, A. O. (1999). MerR cross-links to the α, β, and σ70 Subunits of RNA
polymerase in the preinitiation complex at the merTPCAD promoter. Biochemistry 38, 3362-3368.
Kunst, F. & Rapoport, G. (1995). Salt stress is an environmental signal affecting degradative
enzyme synthesis in Bacillus subtilis. J Bacteriol 177, 2403-2407.
Küster-Schöck, E., Wagner, A., Völker, U. & Hillen, W. (1999). Mutations in catabolite control
protein CcpA showing glucose-independent regulation in Bacillus megaterium. J Bacteriol 181, 76347638.
Küster, E., Luesink, E. J., de Vos, W. M. & Hillen, W. (1996). Immunological Crossreactivity to
Catabolite control Protein CcpA from Bacillus megaterium is found in many Gram-positive Bacteria.
FEMS Microbiol. Lett. 139, 109-115.
Laemmli, U. K. (1970). Cleavage of structural proteins during the assembly of the head of
bacteriophage T4. Nature 227, 680-685.
Landmann, J. J., Busse, R. A., Latz, J. H., Singh, K. D., Stülke, J. & Görke, B. (2011). Crh, the
paralogue of the phosphocarrier protein HPr, controls the methylglyoxal bypass of glycolysis in
Bacillus subtilis. Mol Microbiol 82, 770-787.
Landmann, J. J., Werner, S., Hillen, W., Stülke, J. & Görke, B. (2012). Carbon source control of
the phosphorylation state of the Bacillus subtilis carbon-flux regulator Crh in vivo. FEMS Microbiol
Lett 327, 47-53.
Langbein, I., Bachem, S. & Stülke, J. (1999). Specific interaction of the RNA-binding domain of the
Bacillus subtilis transcriptional antiterminatior GlcT with its RNA target, RAT. Journal of Molecular
Biology 293, 795-805.
Lee, J. H. & Hoover, T. R. (1995). Protein crosslinking studies suggest that Rhizobium meliloti C4dicarboxylic acid transport protein D, a σ54-dependent transcriptional activator, interacts with σ54 and
the β subunit of RNA polymerase. Proc Natl Acad Sci USA 92, 9702-9706.
Lehnik-Habrink, M., Pförtner, H., Rempeters, L., Pietack, N., Herzberg, C. & Stülke, J. (2010).
The RNA degradosome in Bacillus subtilis: identification of CshA as the major RNA helicase in the
multiprotein complex. Mol Microbiol 77, 958-971.
Li, S., Armstrong, C. M., Bertin, N., Ge, H., Milstein, S., Boxem, M., Vidalain, P. O., Han, J. D.,
Chesneau, A., Hao, T., Goldberg, D. S., Li, N., Martinez, M., Rual, J. F., Lamesch, P., Xu, L.,
Tewari, M., Wong, S. L., Zhang, L. V., Berriz, G. F., Jacotot, L., Vaglio, P., Reboul, J.,
Hirozane-Kishikawa, T., Li, Q., Gabel, H. W., Elewa, A., Baumgartner, B., Rose, D. J., Yu, H.,
Bosak, S., Sequerra, R., Fraser, A., Mango, S. E., Saxton, W. M., Strome, S., Van Den Heuvel, S.,
Piano, F., Vandenhaute, J., Sardet, C., Gerstein, M., Doucette-Stamm, L., Gunsalus, K. C.,
REFERENCES
87
Harper, J. W., Cusick, M. E., Roth, F. P., Hill, D. E. & Vidal, M. (2004). A map of the interactome
network of the metazoan C. elegans. Science 303, 540-543.
Liebl, A. (2009). Untersuchung der Interaktion von CcpA mit HPr und HPrS46D mittels BACTH.
Lehrstuhl für Mikrobiologie, Bachelorarbeit, Friedrich-Alexander Universität Erlangen-Nürnberg.
Liedberg, B., Nylander, C. & Lundstrom, I. (1995). Biosensing with surface plasmon resonance how it all started. Biosens Bioelectron 10, i-ix.
Lopez de Saro, F. J., Yoshikawa, N. & Helmann, J. D. (1999). Expression, abundance, and RNA
polymerase binding properties of the δ factor of Bacillus subtilis. J Biol Chem 274, 15953-15958.
Lorca, G. L., Chung, Y. J., Barabote, R. D., Weyler, W., Schilling, C. H. & Saier, M. H., Jr.
(2005). Catabolite repression and activation in Bacillus subtilis: dependency on CcpA, HPr, and HprK.
J Bacteriol 187, 7826-7839.
Lulko, A. T., Buist, G., Kok, J. & Kuipers, O. P. (2007). Transcriptome analysis of temporal
regulation of carbon metabolism by CcpA in Bacillus subtilis reveals additional target genes. J Mol
Microbiol Biotechnol 12, 82-95.
Magasanik, B. (1961). Catabolite repression. Cold Spring Harb Symp Quant Biol 26, 249-256.
Martin-Verstraete, I., Charrier, V., Stülke, J., Galinier, A., Erni, B., Rapoport, G. & Deutscher,
J. (1998). Antagonistic effects of dual PTS-catalysed phosphorylation on the Bacillus subtilis
transcriptional activator LevR. Mol Microbiol 28, 293-303.
Martin-Verstraete, I., Stülke, J., Klier, A. & Rapoport, G. (1995). Two different mechanisms
mediate catabolite repression of the Bacillus subtilis levanase operon. J Bacteriol 177, 6919-6927.
Martin, R. G., Gillette, W. K., Martin, N. I. & Rosner, J. L. (2002). Complex formation between
activator and RNA polymerase as the basis for transcriptional activation by MarA and SoxS in
Escherichia coli. Mol Microbiol 43, 355-370.
Meyer, F. M., Gerwig, J., Hammer, E., Herzberg, C., Commichau, F. M., Völker, U. & Stülke, J.
(2011). Physical interactions between tricarboxylic acid cycle enzymes in Bacillus subtilis: evidence
for a metabolon. Metab Eng 13, 18-27.
Mijakovic, I., Poncet, S., Galinier, A., Monedero, V., Fieulaine, S., Janin, J., Nessler, S.,
Marquez, J. A., Scheffzek, K., Hasenbein, S., Hengstenberg, W. & Deutscher, J. (2002).
Pyrophosphate-producing protein dephosphorylation by HPr kinase/phosphorylase: a relic of early
life? Proc Natl Acad Sci USA 99, 13442-13447.
Miller, A., Wood, D., Ebright, R. H. & Rothman-Denes, L. B. (1997). RNA polymerase β' subunit:
a target of DNA binding-independent activation. Science 275, 1655-1657.
Miwa, Y., Nagura, K., Eguchi, S., Fukuda, H., Deutscher, J. & Fujita, Y. (1997). Catabolite
repression of the Bacillus subtilis gnt operon exerted by two catabolite-responsive elements. Mol
Microbiol 23, 1203-1213.
Miwa, Y., Nakata, A., Ogiwara, A., Yamamoto, M. & Fujita, Y. (2000). Evaluation and
characterization of catabolite-responsive elements (cre) of Bacillus subtilis. Nucleic Acids Res 28,
1206-1210.
88
REFERENCES
Miwa, Y., Saikawa, M. & Fujita, Y. (1994). Possible function and some properties of the CcpA
protein of Bacillus subtilis. Microbiology 140, 2567-2575.
Molle, V., Nakaura, Y., Shivers, R. P., Yamaguchi, H., Losick, R., Fujita, Y. & Sonenshein, A. L.
(2003). Additional targets of the Bacillus subtilis global regulator CodY identified by chromatin
immunoprecipitation and genome-wide transcript analysis. J Bacteriol 185, 1911-1922.
Moreno, M., Schneider, B., Maile, R., Weyler, W. & Saier, M., Jr. (2001). Catabolite repression
mediated by the CcpA protein in Bacillus subtilis: novel modes of regulation revealed by wholegenome analyses. Mol Microbiol 39, 1366-1381.
Mullis, K., Faloona, F., Scharf, S., Saiki, R., Horn, G. & Erlich, H. (1986). Specific enzymatic
amplification of DNA in vitro: the polymerase chain reaction. Cold Spring Harb Symp Quant Biol 51,
263-273.
Nakano, M. M., Lin, A., Zuber, C. S., Newberry, K. J., Brennan, R. G. & Zuber, P. (2010).
Promoter recognition by a complex of Spx and the C-terminal domain of the RNA polymerase α
subunit. PLoS One 5, e8664.
Nicholson, W. L., Park, Y.-K., Henkin, T. M., Won, M., Weickert, M. J., Gaskell, J. A. &
Chambliss, G. H. (1987). Catabolite repression-resistant mutations of the Bacillus subtilis α-amylase
promoter affect transcription levels and are in an operator-like sequence. J. Mol. Biol. 198, 609-618.
Niu, W., Kim, Y., Tau, G., Heyduk, T. & Ebright, R. H. (1996). Transcription activation at class II
CAP-dependent promoters: two interactions between CAP and RNA polymerase. Cell 87, 1123-1134.
Old, W. M., Meyer-Arendt, K., Aveline-Wolf, L., Pierce, K. G., Mendoza, A., Sevinsky, J. R.,
Resing, K. A. & Ahn, N. G. (2005). Comparison of label-free methods for quantifying human
proteins by shotgun proteomics. Mol Cell Proteomics 4, 1487-1502.
Parche, S., Schmid, R. & Titgemeyer, F. (1999). The phosphotransferase system (PTS) of
Streptomyces coelicolor identification and biochemical analysis of a histidine phosphocarrier protein
HPr encoded by the gene ptsH. Eur J Biochem 265, 308-317.
Parrott, M. B. & Barry, M. A. (2000). Metabolic biotinylation of recombinant proteins in
mammalian cells and in mice. Mol Ther 1, 96-104.
Poncet, S., Mijakovic, I., Nessler, S., Gueguen-Chaignon, V., Chaptal, V., Galinier, A., Boel, G.,
Maze, A. & Deutscher, J. (2004). HPr kinase/phosphorylase, a Walker motif A-containing
bifunctional sensor enzyme controlling catabolite repression in Gram-positive bacteria. Biochim
Biophys Acta 1697, 123-135.
Postma, P. W., Lengeler, J. W. & Jacobson, G. R. (1993). Phosphoenolpyruvate:carbohydrate
phosphotransferase systems of bacteria. Microbiol. Rev. 57, 543-504.
Presecan-Siedel, E., Galinier, A., Longin, R., Deutscher, J., Danchin, A., Glaser, P. & MartinVerstraete, I. (1999). Catabolite regulation of the pta gene as part of carbon flow pathways in
Bacillus subtilis. J. Bacteriol. 181, 6889-6897.
Reizer, J., Bachem, S., Reizer, A., Arnaud, M., Saier, M. H., Jr. & Stülke, J. (1999). Novel
phosphotransferase system genes revealed by genome analysis - the complete complement of PTS
proteins encoded within the genome of Bacillus subtilis. Microbiology 145, 3419-3429.
REFERENCES
89
Reizer, J., Bergstedt, U., Galinier, A., Küster, E., Saier, M. H., Jr., Hillen, W., Steinmetz, M. &
Deutscher, J. (1996). Catabolite repression resistance of gnt operon expression in Bacillus subtilis
conferred by mutation of His-15, the site of phosphoenolpyruvate-dependent phosphorylation of the
phosphocarrier protein HPr. J Bacteriol 178, 5480-5486.
Reizer, J., Hoischen, C., Titgemeyer, F., Rivolta, C., Rabus, R., Stülke, J., Karamata, D., Saier,
M. H., Jr. & Hillen, W. (1998). A novel protein kinase that controls carbon catabolite repression in
bacteria. Mol Microbiol 27, 1157-1169.
Renna, M. C., Najimudin, N., Winik, L. R. & Zahler, S. A. (1993). Regulation of the Bacillus
subtilis alsS, alsD, and alsR genes involved in post-exponential-phase production of acetoin. J
Bacteriol 175, 3863-3875.
Rojo, F. (2010). Carbon catabolite repression in Pseudomonas: optimizing metabolic versatility and
interactions with the environment. FEMS Microbiol Rev 34, 658-684.
Rual, J. F., Venkatesan, K., Hao, T., Hirozane-Kishikawa, T., Dricot, A., Li, N., Berriz, G. F.,
Gibbons, F. D., Dreze, M., Ayivi-Guedehoussou, N., Klitgord, N., Simon, C., Boxem, M.,
Milstein, S., Rosenberg, J., Goldberg, D. S., Zhang, L. V., Wong, S. L., Franklin, G., Li, S.,
Albala, J. S., Lim, J., Fraughton, C., Llamosas, E., Cevik, S., Bex, C., Lamesch, P., Sikorski, R.
S., Vandenhaute, J., Zoghbi, H. Y., Smolyar, A., Bosak, S., Sequerra, R., Doucette-Stamm, L.,
Cusick, M. E., Hill, D. E., Roth, F. P. & Vidal, M. (2005). Towards a proteome-scale map of the
human protein-protein interaction network. Nature 437, 1173-1178.
Rygus, T. & Hillen, W. (1991). Inducible high-level expression of heterologous genes in Bacillus
megaterium using the regulatory elements of the xylose-utilization operon. Appl. Microbiol.
Biotechnol. 35, 594-599.
Saier, M. H., Jr. (1989). Protein phosphorylation and allosteric control of inducer exclusion and
catabolite repression by the bacterial phosphoenolpyruvate:sugar phosphotransferase system.
Microbiol. Rev. 53, 109-120.
Saier, M. H., Jr. & Reizer, J. (1994). The bacterial phosphotransferase system: new frontiers 30
years later. Mol Microbiol 13, 755-764.
Saier, M. H., Jr. & Roseman, S. (1976). Sugar transport. The crr mutation: its effect on repression of
enzyme synthesis. J Biol Chem 251, 6598-6605.
Sambrook, J., Fritsch, E. F. & Maniatis, T. (1989). Molecular cloning: a laboratory manual. Cold
Spring Harbor, N. Y.: Cold Spring Harbor Laboratory Press.
Sanger, F., Nicklen, S. & Coulson, A. R. (1977). DNA sequencing with chain terminating inhibitors.
Proc Natl Acad Sci USA 74, 5463-5467.
Schägger, H. & von Jagow, G. (1987). Tricine-sodium dodecyl sulfate-polyacrylamide gel
electrophoresis for the separation of proteins in the range from 1 to 100 kDa. Anal Biochem 166, 368379.
Schatz, P. J. (1993). Use of peptide libraries to map the substrate specificity of a peptide-modifying
enzyme: a 13 residue consensus peptide specifies biotinylation in Escherichia coli. Biotechnology (N
Y) 11, 1138-1143.
90
REFERENCES
Schmalisch, M. H., Bachem, S. & Stülke, J. (2003). Control of the Bacillus subtilis antiterminator
protein GlcT by phosphorylation. Elucidation of the phosphorylation chain leading to inactivation of
GlcT. J Biol Chem 278, 51108-51115.
Schmidt, T. G. & Skerra, A. (2007). The Strep-tag system for one-step purification and high-affinity
detection or capturing of proteins. Nat Protoc 2, 1528-1535.
Schnetz, K., Stülke, J., Gertz, S., Krüger, S., Krieg, M., Hecker, M. & Rak, B. (1996). LicT, a
Bacillus subtilis transcriptional antiterminator protein of the BglG family. Journal of Bacteriology
178, 1971-1979.
Schumacher, M. A., Allen, G. S., Diel, M., Seidel, G., Hillen, W. & Brennan, R. G. (2004).
Structural basis for allosteric control of the transcription regulator CcpA by the phosphoprotein HPrSer46-P. Cell 118, 731-741.
Schumacher, M. A., Seidel, G., Hillen, W. & Brennan, R. G. (2006). Phosphoprotein Crh-Ser46-P
displays altered binding to CcpA to effect carbon catabolite regulation. J Biol Chem 281, 6793-6800.
Schumacher, M. A., Seidel, G., Hillen, W. & Brennan, R. G. (2007). Structural Mechanism for the
Fine-tuning of CcpA Function by The Small Molecule Effectors Glucose 6-Phosphate and Fructose
1,6-Bisphosphate. J Mol Biol 368, 1042-1050.
Schumacher, M. A., Sprehe, M., Bartholomae, M., Hillen, W. & Brennan, R. G. (2011).
Structures of carbon catabolite protein A-(HPr-Ser46-P) bound to diverse catabolite response element
sites reveal the basis for high-affinity binding to degenerate DNA operators. Nucleic Acids Res 39,
2931-2942.
Seidel, G. (2005). Interactions and molecular function of the transcriptional regulator CcpA from
Bacillus subtilis and B. megaterium. Lehrstuhl für Mikrobiologie, Doktorarbeit, Friedrich-Alexander
Universität Erlangen-Nürnberg.
Seidel, G., Diel, M., Fuchsbauer, N. & Hillen, W. (2005). Quantitative interdependence of
coeffectors, CcpA and cre in carbon catabolite regulation of Bacillus subtilis. FEBS J 272, 2566-2577.
Shah, I. M. & Wolf, R. E., Jr. (2004). Novel protein-protein interaction between Escherichia coli
SoxS and the DNA binding determinant of the RNA polymerase α subunit: SoxS functions as a cosigma factor and redeploys RNA polymerase from UP-element-containing promoters to SoxSdependent promoters during oxidative stress. J Mol Biol 343, 513-532.
Shin, B. S., Choi, S. K. & Park, S. H. (1999). Regulation of the Bacillus subtilis
phosphotransacetylase gene. J Biochem (Tokyo) 126, 333-339.
Shivers, R. P., Dineen, S. S. & Sonenshein, A. L. (2006). Positive regulation of Bacillus subtilis
ackA by CodY and CcpA: establishing a potential hierarchy in carbon flow. Mol Microbiol 62, 811822.
Singh, K. D., Schmalisch, M. H., Stülke, J. & Görke, B. (2008). Carbon catabolite repression in
Bacillus subtilis: quantitative analysis of repression exerted by different carbon sources. J Bacteriol
190, 7275-7284.
Slack, F. J., Serror, P., Joyce, E. & Sonenshein, A. L. (1995). A gene required for nutritional
repression of the Bacillus subtilis dipeptide permease operon. Mol. Microbiol. 15, 689-702.
REFERENCES
91
Sonenshein, A. L. (2005). CodY, a global regulator of stationary phase and virulence in Grampositive bacteria. Curr Opin Microbiol 8, 203-207.
Sonenshein, A. L. (2007). Control of key metabolic intersections in Bacillus subtilis. Nat Rev
Microbiol 5, 917-927.
Spencer, V. A., Sun, J. M., Li, L. & Davie, J. R. (2003). Chromatin immunoprecipitation: a tool for
studying histone acetylation and transcription factor binding. Methods 31, 67-75.
Sprehe, M. (2007). Mechanismen der Katabolitenregulation in Bacilli. Lehrstuhl für Mikrobiologie,
Doktorarbeit, Friedrich-Alexander Universität Erlangen-Nürnberg.
Sprehe, M., Seidel, G., Diel, M. & Hillen, W. (2007). CcpA mutants with differential activities in
Bacillus subtilis. J Mol Microbiol Biotechnol 12, 96-105.
Steinmetz, M., Le Coq, D. & Aymerich, S. (1989). Induction of saccharolytic enzymes by sucrose in
Bacillus subtilis: evidence for two partially interchangeable regulatory pathways. J Bacteriol 171,
1519-1523.
Strauch, E. M. & Georgiou, G. (2007). A bacterial two-hybrid system based on the twin-arginine
transporter pathway of E. coli. Protein Sci 16, 1001-1008.
Stülke, J., Arnaud, M., Rapoport, G. & Martin-Verstraete, I. (1998). PRD - a protein domain
involved in PTS-dependent induction and carbon catabolite repression of catabolic operons in bacteria.
Mol Microbiol 28, 865-874.
Stülke, J. & Hillen, W. (1999). Carbon catabolite repression in bacteria. Current Opinion in
Microbiology 2, 195-201.
Stülke, J. & Hillen, W. (2000). Regulation of carbon catabolism in Bacillus species. Annu Rev
Microbiol 54, 849-880.
Stülke, J., Martin-Verstraete, I., Zagorec, M., Rose, M., Klier, A. & Rapoport, G. (1997).
Induction of the Bacillus subtilis ptsGHI operon by glucose is controlled by a novel antiterminator,
GlcT. Mol Microbiol 25, 65-78.
Szalewska-Palasz, A., Wegrzyn, A., Blaszczak, A., Taylor, K. & Wegrzyn, G. (1998). DnaAstimulated transcriptional activation of oriλ: Escherichia coli RNA polymerase β subunit as a
transcriptional activator contact site. Proc Natl Acad Sci USA 95, 4241-4246.
Tagami, H. & Aiba, H. (1998). A common role of CRP in transcription activation: CRP acts
transiently to stimulate events leading to open complex formation at a diverse set of promoters.
EMBO J 17, 1759-1767.
Titgemeyer, F. & Hillen, W. (2002). Global control of sugar metabolism: a gram-positive solution.
Antonie Van Leeuwenhoek 82, 59-71.
Tobisch, S., Glaser, P., Krüger, S. & Hecker, M. (1997). Identification and characterization of a
new β-glucoside utilization system in Bacillus subtilis. J. Bacteriol. 179, 496-506.
Tobisch, S., Stülke, J. & Hecker, M. (1999a). Regulation of the lic operon of Bacillus subtilis and
characterization of potential phosphorylation sites of the LicR regulator protein by site-directed
mutagenesis. J Bacteriol 181, 4995-5003.
92
REFERENCES
Tobisch, S., Zühlke, D., Bernhardt, J., Stülke, J. & Hecker, M. (1999b). Role of CcpA in
regulation of the central pathways of carbon catabolism in Bacillus subtilis. J Bacteriol 181, 69967004.
Tojo, S., Satomura, T., Morisaki, K., Deutscher, J., Hirooka, K. & Fujita, Y. (2005). Elaborate
transcription regulation of the Bacillus subtilis ilv-leu operon involved in the biosynthesis of branchedchain amino acids through global regulators of CcpA, CodY and TnrA. Mol Microbiol 56, 1560-1573.
Tortosa, P., Declerck, N., Dutartre, H., Lindner, C., Deutscher, J. & Le Coq, D. (2001). Sites of
positive and negative regulation in the Bacillus subtilis antiterminators LicT and SacY. Mol Microbiol
41, 1381-1393.
Turinsky, A. J., Grundy, F. J., Kim, J. H., Chambliss, G. H. & Henkin, T. M. (1998).
Transcriptional activation of the Bacillus subtilis ackA gene requires sequences upstream of the
promoter. J Bacteriol 180, 5961-5967.
Turinsky, A. J., Moir-Blais, T. R., Grundy, F. J. & Henkin, T. M. (2000). Bacillus subtilis ccpA
gene mutants specifically defective in activation of acetoin biosynthesis. J Bacteriol 182, 5611-5614.
Ün, S. (2009). Charakterisierung von Peptiden für die Bindung essentieller Penicillin-bindender
Proteine und die Variationen der Linkerlänge einzelkettiger TetR Varianten. Lehrstuhl für
Mikrobiologie, Doktorarbeit, Friedrich-Alexander Universität Erlangen-Nürnberg.
Warner, J. B. & Lolkema, J. S. (2003). CcpA-dependent carbon catabolite repression in bacteria.
Microbiol Mol Biol Rev 67, 475-490.
Weickert, M. J. & Adhya, S. (1992). A family of bacterial regulators homologous to Gal and Lac
Repressors. J. Biol. Chem. 267, 15869-15874.
Weickert, M. J. & Chambliss, G. H. (1990). Site-directed mutagenesis of a catabolite repression
operator sequence in Bacillus subtilis. Proc. Natl. Acad. Sci. USA 87, 6238-6242.
Yoshida, K., Kobayashi, K., Miwa, Y., Kang, C. M., Matsunaga, M., Yamaguchi, H., Tojo, S.,
Yamamoto, M., Nishi, R., Ogasawara, N., Nakayama, T. & Fujita, Y. (2001). Combined
transcriptome and proteome analysis as a powerful approach to study genes under glucose repression
in Bacillus subtilis. Nucleic Acids Res 29, 683-692.
Yu, H., Braun, P., Yildirim, M. A., Lemmens, I., Venkatesan, K., Sahalie, J., HirozaneKishikawa, T., Gebreab, F., Li, N., Simonis, N., Hao, T., Rual, J. F., Dricot, A., Vazquez, A.,
Murray, R. R., Simon, C., Tardivo, L., Tam, S., Svrzikapa, N., Fan, C., de Smet, A. S., Motyl, A.,
Hudson, M. E., Park, J., Xin, X., Cusick, M. E., Moore, T., Boone, C., Snyder, M., Roth, F. P.,
Barabasi, A. L., Tavernier, J., Hill, D. E. & Vidal, M. (2008). High-quality binary protein
interaction map of the yeast interactome network. Science 322, 104-110.
Zeng, X., Galinier, A. & Saxild, H. H. (2000). Catabolite repression of dra-nupC-pdp operon
expression in Bacillus subtilis. Microbiology 146, 2901-2908.
PUBLICATIONS
93
8 Publications
Wünsche A., Hammer E., Bartholomae M., Völker U., Burkovski A., Seidel G. & Hillen W.
(2012). CcpA forms complexes with CodY and RpoA in Bacillus subtilis. FEBS J (in Press)
Bertram R., Wünsche A., Sprehe M. & Hillen W. (2006). Regulated expression of HPrK/P
does not affect carbon catabolite repression of the xyn operon and of rocG in Bacillus subtilis.
FEMS Microbiol Lett., 259, 147-152.
APPENDIX
94
9 Appendix
9.1 Proteins identified across all crosslinked and non-crosslinked samples of WH1012 grown with glucose
The following Table A1 shows the data from MS analysis of crosslinked and non-crosslinked eluates of WH1012 grown with glucose. The MS analysis
of the samples was performed in collaboration with Dr. Elke Hammer and Prof. Dr. Uwe Völker (Ernst-Moritz-Arndt-Universität Greifswald).
Table A1: Proteins identified across all cross-linked samples with an enrichment ratio higher than 1.5fold and at least 6 spectra in one sample
N14
Gene
name
Accession
Number
tufA
metE
srfAA
fusA
srfAB
rpoC
rpoB
purB
dhbF
BSU01130
BSU13180
BSU03480
BSU01120
BSU03490
BSU01080
BSU01070
BSU06440
BSU31960
gapA
dnaK
gltA
Bio View: Identified Proteins (625)
elongation factor Tu
methionine synthase
surfactin synthetase / competence
elongation factor G
surfactin synthetase / competence
RNA polymerase beta' subunit
RNA polymerase beta subunit
adenylsuccinate lyase
involved in 2,3-dihydroxybenzoate
biosynthesis
BSU33940 Glyceraldehyde 3-phosphate
dehydrogenase, NAD-dependent,
glycolytic enzyme
BSU25470 class I heat-shock protein (molecular
chaperone)
BSU18450 large subunit of glutamate synthase
MW
crosslink_1
N15
N14
no
crosscrosslink_2
link_1
N15
N15
cR1
crossno
link / no crosscrosscross- link_3
link_2
link
44 kDa
87 kDa
402 kDa
77 kDa
401 kDa
134 kDa
134 kDa
49 kDa
264 kDa
82
63
61
53
51
35
33
30
30
22
7
14
3
10
13
8
n.d.
3
78
64
59
59
52
34
35
26
28
24
4
11
3
19
13
8
n.d.
3
3.5
11.5
4.8
18.7
3.6
2.7
4.3
36 kDa
29
9
21
66 kDa
29
9
169 kDa
27
n.d.
N14
N15
no
crosscrosslink_4
link_3
N14
cR2
crossno
link / no
crosscrosslink_4
link
27
9
19
4
12
16
9
n.d.
9
63
44
29
36
46
24
19
23
17
24
8
18
2
8
11
11
n.d.
6
2.2
5.6
1.9
12.7
4.3
1.9
1.8
9.7
51
52
40
40
39
28
16
23
19
10
2.6
22
10
24
10
2.3
28
9
3.2
23
13
22
16
1.6
25
n.d.
17
n.d.
14
n.d.
2.4
APPENDIX
95
N14
Gene
name
Accession
Number
tsf
gndA
BSU16500 elongation factor Ts
BSU23860 NADP-dependent phosphogluconate
dehydrogenase
BSU06480 phosphoribosylformylglycinamidine
synthase
BSU33900 enolase, glycolytic/ gluconeogenic
enzyme, universally conserved protein
BSU18000 trigger enzyme: aconitase and RNA
binding protein
BSU40100 alkyl hydroperoxide reductase (large
subunit) / NADH dehydrogenase
BSU06520 phosphoribosylaminoimidazole
carboxamide formyltransferase
BSU32670 synthesis of Fe-S-clusters
BSU28290 ketol-acid reductoisomerase (2,3dihydroxy-3-methylbutanoate, 2acetolactate)
BSU03510 surfactin synthetase / competence
BSU35300 preprotein translocase subunit (ATPase)
BSU29130 isocitrate dehydrogenase
BSU00860 ATP-dependent Clp protease, ATPase
subunit
BSU04580 DEAD-box RNA helicase
BSU22880 similar to ribosomal protein S1
BSU28220 ATP-dependent Clp protease ATP-binding
subunit (class III heat-shock protein)
BSU03360 putative metallochaperone
BSU16630 translation initiation factor IF-2
BSU38960 unknown
purL
eno
citB
ahpF
purH
sufB
ilvC
srfAC
secA
icd
clpC
cshA
ypfD
clpX
yciC
infB
yxjG
Bio View: Identified Proteins (625)
MW
crosslink_1
N15
N14
no
crosscrosslink_2
link_1
N15
N15
cR1
crossno
link / no crosscrosscross- link_3
link_2
link
32 kDa
52 kDa
27
26
n.d.
n.d.
30
29
n.d.
n.d.
80 kDa
25
10
23
11
47 kDa
24
n.d.
19
99 kDa
23
n.d.
55 kDa
21
56 kDa
N14
N15
no
crosscrosslink_4
link_3
N14
cR2
crossno
link / no
crosscrosslink_4
link
20
21
n.d.
n.d.
17
17
n.d.
n.d.
22
14
18
11
1.6
n.d.
17
5
12
5
2.9
24
n.d.
20
n.d.
19
n.d.
n.d.
21
n.d.
11
n.d.
8
n.d.
21
n.d.
22
n.d.
14
n.d.
12
n.d.
53 kDa
37 kDa
21
21
n.d.
2
21
23
n.d.
n.d.
18
10
2
n.d.
15
8
n.d.
4
144 kDa
96 kDa
46 kDa
90 kDa
20
19
19
18
n.d.
n.d.
4
n.d.
22
18
22
19
n.d.
n.d.
6
n.d.
14
16
13
10
n.d.
n.d.
6
n.d.
10
13
12
8
n.d.
n.d.
7
n.d.
57 kDa
42 kDa
46 kDa
18
18
18
n.d.
n.d.
5
19
16
17
n.d.
2
8
11
15
13
n.d.
2
8
7
16
12
n.d.
3
8
45 kDa
79 kDa
43 kDa
18
17
16
5
n.d.
n.d.
21
20
19
4
n.d.
n.d.
15
16
17
10
n.d.
n.d.
16
16
15
10
n.d.
n.d.
2.3
22.0
4.1
17.0
2.7
4.3
16.5
4.5
1.9
6.2
1.6
1.6
APPENDIX
96
N14
Gene
name
Accession
Number
aspS
guaA
mdh
pgcA
BSU27550
BSU06360
BSU29120
BSU09310
mtnK
hbs
metK
glnA
BSU13560
BSU22790
BSU30550
BSU17460
glyA
tkt
bdhA
mtnA
accC
BSU36900
BSU17890
BSU06240
BSU13550
BSU24340
sufD
gatA
ileS
guaB
tig
rnjA
rpsC
yceH
pdhD
BSU32700
BSU06680
BSU15430
BSU00090
BSU28230
BSU14530
BSU01220
BSU02940
BSU14610
ykpA
BSU14430
crosslink_1
N15
N14
no
crosscrosslink_2
link_1
N15
N15
cR1
crossno
link / no crosscrosscross- link_3
link_2
link
Bio View: Identified Proteins (625)
MW
aspartyl-tRNA synthetase
GMP synthase (glutamine-hydrolysing)
malate dehydrogenase
alpha-phosphoglucomutase, required for
UDP-glucose synthesis
5-methylthioribose kinase
non-specific DNA-binding protein Hbsu
S-adenosylmethionine synthetase
trigger enzyme: glutamine synthetase and
effector of TnrA and GlnR
serine hydroxymethyltransferase
transketolase
acetoine/ butanediol dehydrogenase
5-methylthioribose-1-phosphate isomerase
acetyl-CoA carboxylase (biotin
carboxylase subunit)
synthesis of Fe-S-clusters
production of glutamyl-tRNA (Gln)
isoleucyl-tRNA synthetase
IMP dehydrogenase
trigger factor (prolyl isomerase)
RNase J1
ribosomal protein S3
similar to toxic anion resistance protein
dihydrolipoamide dehydrogenase E3
subunit of both pyruvate dehydrogenase
and 2-oxoglutarate dehydrogenase
complexes
similar to ABC transporter (ATP-binding
protein)
66 kDa
58 kDa
34 kDa
65 kDa
16
16
16
16
n.d.
n.d.
2
3
17
13
17
16
n.d.
n.d.
n.d.
5
45 kDa
10 kDa
44 kDa
50 kDa
16
16
16
15
4
3
5
n.d.
17
15
14
20
6
5
5
n.d.
45 kDa
72 kDa
37 kDa
39 kDa
49 kDa
15
15
15
15
15
n.d.
n.d.
n.d.
n.d.
2
16
17
15
13
15
n.d.
n.d.
n.d.
n.d.
n.d.
48 kDa
53 kDa
105 kDa
53 kDa
47 kDa
62 kDa
24 kDa
42 kDa
50 kDa
15
15
14
14
14
14
14
14
14
2
3
n.d.
n.d.
n.d.
n.d.
3
2
7
19
15
14
16
12
13
15
15
12
n.d.
3
n.d.
n.d.
n.d.
3
4
2
9
61 kDa
13
n.d.
13
n.d.
16.5
4.0
3.3
3.9
3.0
15.0
17.0
5.0
9.0
4.1
7.3
1.6
N14
N15
no
crosscrosslink_4
link_3
N14
cR2
crossno
link / no
crosscrosslink_4
link
15
11
16
9
n.d.
n.d.
2
n.d.
11
13
13
7
n.d.
n.d.
n.d.
2
17
17
10
10
2
7
6
n.d.
15
13
12
9
4
5
5
n.d.
12
11
11
10
12
n.d.
n.d.
n.d.
n.d.
n.d.
13
9
11
9
14
n.d.
n.d.
n.d.
n.d.
n.d.
8
12
10
10
10
12
14
13
12
3
5
n.d.
n.d.
n.d.
n.d.
3
6
7
13
11
7
7
12
11
10
12
16
2
3
n.d.
n.d.
n.d.
n.d.
3
5
6
9
n.d.
8
n.d.
14.5
8.0
5.3
2.5
2.0
4.2
2.9
4.0
2.3
2.2
APPENDIX
97
N14
Gene
name
Accession
Number
pdhA
BSU14580 pyruvate dehydrogenase (E1 alpha
subunit)
BSU28630 phenylalanyl-tRNA synthetase (beta
subunit)
BSU29470 acetate kinase
BSU06210 unknown
BSU08760 sporulation-control gene
BSU29140 citrate synthase
BSU29220 malic enzyme
BSU14590 pyruvate dehydrogenase (E1 beta subunit)
BSU22620 histidinol-phosphate aminotransferase /
tyrosine and phenylalanine
aminotransferase
BSU27850 quinolinate synthetase
BSU32690 cysteine desulfurase
BSU06180 phage shock protein A homolog
BSU37120 fructose 1.6-bisphosphate aldolase.
glycolytic/ gluconeogenic enzyme
BSU35670 UTP-glucose-1-phosphate
uridylyltransferase. general stress protein
BSU02890 similar to tellurium resistance protein
BSU28470 aspartokinase II (alpha and beta subunits)
BSU06450 phosphoribosylaminoimidazole
succinocarboxamide synthase
BSU36810 ATP synthase (subunit beta)
BSU16940 multifunctional protein involved in
homologous recombination and DNA
repair (LexA-autocleavage)
BSU33910 phosphoglycerate mutase, glycolytic /
gluconeogenic enzyme
pheT
ackA
ydjI
spo0M
citZ
ytsJ
pdhB
hisC
nadA
sufS
pspA
fbaA
gtaB
yceC
lysC
purC
atpD
recA
pgm
Bio View: Identified Proteins (625)
MW
crosslink_1
N15
N14
no
crosscrosslink_2
link_1
N15
N15
cR1
crossno
link / no crosscrosscross- link_3
link_2
link
N14
N15
no
crosscrosslink_4
link_3
N14
cR2
crossno
link / no
crosscrosslink_4
link
42 kDa
13
n.d.
12
n.d.
5
n.d.
7
n.d.
88 kDa
13
n.d.
14
n.d.
11
n.d.
9
n.d.
43 kDa
36 kDa
30 kDa
42 kDa
44 kDa
35 kDa
40 kDa
13
13
13
13
13
13
12
n.d.
2
2
n.d.
n.d.
5
n.d.
13
12
13
15
13
17
12
n.d.
n.d.
3
2
2
7
n.d.
15
9
9
8
8
11
10
n.d.
n.d.
n.d.
3
2
8
n.d.
12
9
8
9
4
11
10
2
n.d.
2
n.d.
2
6
n.d.
41 kDa
45 kDa
25 kDa
30 kDa
12
12
12
12
n.d.
n.d.
n.d.
n.d.
10
15
13
10
n.d.
n.d.
n.d.
n.d.
8
8
8
14
n.d.
n.d.
2
4
6
8
8
11
n.d.
n.d.
n.d.
n.d.
8.0
6.3
33 kDa
12
n.d.
14
n.d.
9
2
10
4
3.2
22 kDa
44 kDa
27 kDa
12
12
12
5
n.d.
2
11
12
13
5
2
n.d.
2.3
12.0
12.5
11
7
12
3
4
5
9
10
10
4
2
4
2.9
2.8
2.4
51 kDa
38 kDa
12
12
3
6
14
15
3
6
4.3
2.3
10
14
5
6
10
9
6
8
1.8
1.6
56 kDa
11
n.d.
12
n.d.
9
n.d.
11
n.d.
12.5
5.2
14.0
13.0
2.5
13.5
8.5
5.7
3.0
1.6
APPENDIX
98
N14
Gene
name
Accession
Number
hisS
ppaC
yxjH
purF
BSU27560
BSU40550
BSU38950
BSU06490
ptsI
BSU13910
prsA
BSU09950
lutB
proS
citG
cysK
BSU34040
BSU16570
BSU33040
BSU00730
cysS
metS
purA
yjoB
BSU00940
BSU00380
BSU40420
BSU12420
ykwC
yyaF
argG
ftsH
pnpA
metI
BSU13960
BSU40920
BSU29450
BSU00690
BSU16690
BSU11870
rpoA
BSU01430
crosslink_1
N15
N14
no
crosscrosslink_2
link_1
N15
N15
cR1
crossno
link / no crosscrosscross- link_3
link_2
link
Bio View: Identified Proteins (625)
MW
histidyl-tRNA synthetase
inorganic pyrophosphatase
unknown
glutamine phosphoribosyldiphosphate
amidotransferase
Enzyme I, general (non sugar-specific)
component of the PTS. Enzyme I transfers
the phosphoryl group from
phosphoenolpyruvate (PEP) to the
phosphoryl carrier protein (PtsH |HPr)
protein secretion (post-translocation
molecular chaperone)
lactate catabolic enzyme
prolyl-tRNA synthetase
fumarase
trigger enzyme: cysteine synthetase A and
control of CymR activity
cysteine-tRNA synthetase
methionyl-tRNA synthetase
adenylosuccinate synthetase
AAA family, ATPase activity , similar to
cell-division protein FtsH
putative beta-hydroxyacid dehydrogenase
GTP-binding protein/ GTPase
argininosuccinate synthase, reversible
ATP-dependent metalloprotease
polynucleotide phosphorylase, RNase
O-succinylhomoserine lyase (L-cysteine,
H2S, methanethiol, elimination)
RNA polymerase alpha subunit
48 kDa
34 kDa
43 kDa
52 kDa
11
11
11
11
n.d.
n.d.
n.d.
n.d.
13
10
14
7
n.d.
n.d.
n.d.
n.d.
63 kDa
11
3
11
3
33 kDa
11
2
12
4
53 kDa
63 kDa
51 kDa
33 kDa
10
10
10
10
n.d.
n.d.
n.d.
n.d.
13
8
8
10
54 kDa
76 kDa
48 kDa
49 kDa
10
10
10
10
n.d.
n.d.
n.d.
n.d.
31 kDa
40 kDa
45 kDa
71 kDa
77 kDa
42 kDa
10
10
10
10
10
10
35 kDa
10
N14
N15
no
crosscrosslink_4
link_3
N14
cR2
crossno
link / no
crosscrosslink_4
link
12
7
11
2
n.d.
n.d.
n.d.
n.d.
11
6
10
4
n.d.
n.d.
n.d.
n.d.
3.7
12
2
10
2
5.5
3.8
10
4
9
2
3.2
n.d.
n.d.
n.d.
n.d.
8
8
6
9
n.d.
n.d.
n.d.
n.d.
5
8
4
8
n.d.
n.d.
n.d.
n.d.
8
9
10
7
n.d.
n.d.
n.d.
n.d.
5
6
10
5
n.d.
n.d.
n.d.
n.d.
4
4
8
4
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
4
9
8
11
11
12
11
n.d.
n.d.
n.d.
n.d.
n.d.
3
3.0
5
5
7
9
5
7
n.d.
n.d.
2
3
2
3
9
4
7
10
7
6
n.d.
n.d.
n.d.
n.d.
n.d.
3
7.0
6.3
6.0
2.2
3
11
3
3.5
11
4
8
5
2.1
APPENDIX
99
N14
Gene
name
Accession
Number
rpsD
pdxS
BSU29660 ribosomal protein S4
BSU00110 pyridoxal-5'-phosphate synthase (synthase
domain)
BSU31750 putative nicotinate
phosphoribosyltransferase
BSU11690 hydroxyethylthiazole phosphate
biosynthesis
BSU22710 chorismate synthase
BSU15910 beta-ketoacyl-acyl carrier protein
reductase
BSU18440 small subunit of glutamate synthase
BSU25070 similar to peptidoglycan acetylation,
1-hydroxy-2-methyl-2-(E)-butenyl-4diphosphate synthase
BSU28260 3-isopropylmalate dehydratase
(large subunit)
BSU30320 Leu-tRNA synthetase
BSU16600 transcription termination factor
BSU00510 phosphoribosylpyrophosphate synthetase,
universally conserved protein
BSU06420 phosphoribosylaminoimidazole
carboxylase (ATP-dependent)
BSU15590 sulfate adenylyltransferase
BSU21870 dihydroxy-acid dehydratase (2,3dihydroxy-3-methylbutanoate, 2,3dihydroxy-3-methylpentanoate)
BSU11720 enoyl-acyl carrier protein reductase
BSU01030 ribosomal protein L1
BSU40090 alkyl hydroperoxide reductase (small
subunit)
pncB
thiG
aroF
fabG
gltB
ispG
leuC
leuS
nusA
prs
purE
sat
ilvD
fabI
rplA
ahpC
Bio View: Identified Proteins (625)
MW
crosslink_1
N15
N14
no
crosscrosslink_2
link_1
N15
N15
cR1
crossno
link / no crosscrosscross- link_3
link_2
link
N14
N15
no
crosscrosslink_4
link_3
N14
cR2
crossno
link / no
crosscrosslink_4
link
23 kDa
32 kDa
10
10
n.d.
5
7
9
2
3
8.5
2.4
5
11
3
5
5
11
2
6
2.0
2.0
56 kDa
10
n.d.
9
2
9.5
6
3
6
4
1.7
27 kDa
10
4
10
6
2.0
7
5
8
4
1.7
43 kDa
26 kDa
9
9
n.d.
n.d.
11
6
n.d.
n.d.
10
8
n.d.
n.d.
10
5
n.d.
n.d.
55 kDa
41 kDa
9
9
n.d.
n.d.
14
10
n.d.
n.d.
5
6
n.d.
n.d.
5
9
n.d.
n.d.
52 kDa
9
n.d.
10
n.d.
8
n.d.
8
n.d.
92 kDa
42 kDa
35 kDa
9
9
9
n.d.
n.d.
n.d.
10
12
10
n.d.
n.d.
n.d.
3
7
6
n.d.
n.d.
n.d.
3
7
6
n.d.
n.d.
n.d.
17 kDa
9
n.d.
10
n.d.
5
n.d.
6
n.d.
43 kDa
60 kDa
9
9
n.d.
n.d.
10
8
n.d.
n.d.
7
4
n.d.
2
4
3
n.d.
n.d.
3.5
28 kDa
25 kDa
21 kDa
9
9
9
4
4
2
10
9
12
4
2
2
10
6
9
3
2
3
11
9
8
2
2
3
4.2
3.8
2.8
2.4
3.0
5.3
APPENDIX
100
N14
Gene
name
Accession
Number
ftsZ
BSU15290 cell-division initiation protein (septum
formation)
BSU07350 similar to benzaldehyde dehydrogenase
BSU32710 ABC transporter (ATP-binding protein),
synthesis of Fe-S clusters
BSU22360 asparagyl-tRNA synthetase
BSU31980 2,3-dihydroxybenzoate-AMP ligase
(enterobactin synthetase component E)
BSU11540 oligoendopeptidase
BSU32110 ferredoxin-NAD(P)+ oxidoreductase
BSU24080 valine dehydrogenase, isoleucine
dehydrogenase, L-leucine dehydrogenase
BSU31350 glucose 6-phosphate isomerase,
glycolytic / gluconeogenic enzyme
BSU14770 similar to GTP-binding elongation factor
BSU23850 glucose 6-phosphate dehydrogenase,
pentose-phosphate pathway
BSU01370 adenylate kinase
BSU30230 lysine-8-amino-7-oxononanoate
aminotransferase
BSU13620 1,2,-dihydroxy-3-keto-5methylthiopentene dioxygenase
BSU13590 2,3-diketo-5-methylthiopentyl-1-phosphate
enolase, Rubisco-like protein
BSU29180 pyruvate kinase, glycolytic enzyme
BSU16100 succinyl-CoA synthetase (alpha subunit)
BSU30540 asparagine synthase (glutaminehydrolysing)
BSU36830 ATP synthase (subunit alpha)
yfmT
sufC
asnS
dhbE
pepF
yumC
bcd
pgi
ylaG
zwf
adk
bioA
mtnD
mtnW
pyk
sucD
asnB
atpA
Bio View: Identified Proteins (625)
MW
crosslink_1
N15
N14
no
crosscrosslink_2
link_1
N15
N15
cR1
crossno
link / no crosscrosscross- link_3
link_2
link
N14
N15
no
crosscrosslink_4
link_3
N14
cR2
crossno
link / no
crosscrosslink_4
link
40 kDa
9
n.d.
10
2
9.5
7
4
5
2
2.0
53 kDa
29 kDa
9
9
2
n.d.
7
11
3
3
3.2
6.7
5
8
3
5
5
8
2
4
2.0
1.8
49 kDa
60 kDa
8
8
n.d.
n.d.
8
8
n.d.
n.d.
8
9
n.d.
n.d.
5
11
n.d.
n.d.
77 kDa
37 kDa
40 kDa
8
8
8
n.d.
n.d.
n.d.
8
12
9
n.d.
n.d.
n.d.
3
8
2
n.d.
n.d.
n.d.
2
7
4
n.d.
n.d.
n.d.
51 kDa
8
n.d.
10
n.d.
5
n.d.
4
n.d.
68 kDa
56 kDa
8
8
n.d.
n.d.
9
9
n.d.
n.d.
5
4
n.d.
n.d.
6
5
n.d.
n.d.
24 kDa
50 kDa
8
8
n.d.
n.d.
7
8
n.d.
n.d.
6
3
n.d.
n.d.
7
5
n.d.
n.d.
21 kDa
8
n.d.
8
n.d.
4
n.d.
5
n.d.
45 kDa
8
n.d.
9
n.d.
5
n.d.
4
n.d.
62 kDa
31 kDa
73 kDa
8
8
8
n.d.
n.d.
n.d.
6
9
9
n.d.
n.d.
n.d.
7
8
5
n.d.
n.d.
3
5
7
7
n.d.
n.d.
3
2.0
55 kDa
8
6
9
4
6
4
8
3
2.0
1.7
APPENDIX
101
N14
Gene
name
Accession
Number
cheV
BSU14010 modulation of CheA activity in response to
attractants
BSU41010 glucose-inhibited division protein, tRNA
uridine 5-carboxymethylaminomethyl
modification enzyme
BSU21770 threonine dehydratase
BSU22370 aspartate transaminase
BSU11240 carbamoyl-phosphate transferase-arginine
subunit B)
BSU25260 glycyl-tRNA synthetase (beta subunit)
BSU28870 translation initiation factor IF-3
BSU37330 arginyl-tRNA synthetase, universally
conserved protein
BSU22600 3-phosphoshikimate 1carboxyvinyltransferase
BSU16750 aspartate-semialdehyde dehydrogenase
BSU31970 isochorismatase
BSU38860 UDP glucose 4-epimerase
BSU28440 succinate dehydrogenase (flavoprotein
subunit)
BSU32250 threonine synthase
BSU31370 similar to NADH-dependent butanol
dehydrogenase
BSU01190 ribosomal protein L2
BSU02910 general stress protein, similar to tellurium
resistance protein
BSU32750 methionine ABC transporter (ATP-binding
protein)
BSU11190 N-acetyl-g-glutamyl-phosphate reductase
gidA
ilvA
aspB
carB
glyS
infC
argS
aroE
asd
dhbB
galE
sdhA
thrC
yugJ
rplB
yceE
metN
argC
Bio View: Identified Proteins (625)
MW
crosslink_1
N15
N14
no
crosscrosslink_2
link_1
N15
N15
cR1
crossno
link / no crosscrosscross- link_3
link_2
link
N14
N15
no
crosscrosslink_4
link_3
N14
cR2
crossno
link / no
crosscrosslink_4
link
35 kDa
7
n.d.
7
n.d.
5
n.d.
4
n.d.
70 kDa
7
n.d.
8
n.d.
7
n.d.
3
n.d.
47 kDa
43 kDa
113 kDa
7
7
7
n.d.
n.d.
n.d.
7
10
6
n.d.
n.d.
n.d.
6
7
8
n.d.
n.d.
n.d.
7
6
6
n.d.
n.d.
n.d.
76 kDa
20 kDa
63 kDa
7
7
7
n.d.
n.d.
n.d.
4
7
6
n.d.
n.d.
n.d.
5
10
2
n.d.
n.d.
n.d.
4
11
2
n.d.
n.d.
n.d.
45 kDa
7
n.d.
8
n.d.
6
n.d.
5
n.d.
38 kDa
35 kDa
37 kDa
65 kDa
7
7
7
7
n.d.
n.d.
n.d.
n.d.
8
8
7
8
n.d.
n.d.
n.d.
n.d.
7
6
3
3
n.d.
n.d.
n.d.
n.d.
6
6
4
6
n.d.
n.d.
n.d.
n.d.
37 kDa
43 kDa
7
7
n.d.
n.d.
8
8
n.d.
n.d.
2
5
n.d.
n.d.
2
5
n.d.
n.d.
30 kDa
21 kDa
7
7
n.d.
n.d.
6
6
n.d.
n.d.
6
4
n.d.
n.d.
6
7
n.d.
2
38 kDa
7
n.d.
8
2
7.5
3
n.d.
2
n.d.
38 kDa
7
n.d.
7
3
4.7
5
n.d.
5
n.d.
5.5
APPENDIX
102
N14
Gene
name
Accession
Number
rplL
gatB
pfkA
salA
lonA
BSU01050
BSU06690
BSU29190
BSU01540
BSU28200
murB
BSU15230
obg
BSU27920
pyrC
ykuQ
BSU15500
BSU14180
cysC
pyrF
argJ
bioB
cysI
dapG
deoD
dltA
fabD
BSU15600
BSU15550
BSU11200
BSU30200
BSU33430
BSU16760
BSU19630
BSU38500
BSU15900
fabL
folE2
BSU08650
BSU03340
glmS
BSU01780
ribA
BSU23260
crosslink_1
N15
N14
no
crosscrosslink_2
link_1
N15
N15
cR1
crossno
link / no crosscrosscross- link_3
link_2
link
Bio View: Identified Proteins (625)
MW
ribosomal protein L12
production of glutamyl-tRNA (Gln)
phosphofructokinase, glycolytic enzyme
negative regulator of scoC expression
class III heat-shock ATP-dependent
protease
UDP-N-acetylenolpyruvoylglucosamine
reductase
GTP-binding protein involved in initiation
of sporulation
dihydroorotase
similar to tetrahydrodipicolinate
succinylase
adenylyl-sulfate kinase
orotidine 5'-phosphate decarboxylase
N-acetylglutamate synthase
biotin synthase
sulfite reductase (NADPH2)
aspartokinase I (alpha and beta subunits)
purine nucleoside phosphorylase
D-alanyl-D-alanine carrier protein ligase
malonyl CoA-acyl carrier protein
transacylase
enoyl-acyl carrier protein reductase
GTP cyclohydrolase IB, replaces FolE
under conditions of zinc starvation
glutamine-fructose-6-phosphate
transaminase
GTP cyclohydrolase II/ 3,4-dihydroxy-2butanone 4-phosphate synthase
13 kDa
53 kDa
34 kDa
39 kDa
87 kDa
7
7
7
7
6
2
n.d.
2
4
n.d.
6
9
6
4
6
2
2
2
n.d.
n.d.
33 kDa
6
n.d.
7
48 kDa
6
n.d.
47 kDa
25 kDa
6
6
23 kDa
26 kDa
43 kDa
37 kDa
65 kDa
43 kDa
25 kDa
56 kDa
34 kDa
3.3
8.0
3.3
2.8
N14
N15
no
crosscrosslink_4
link_3
N14
cR2
crossno
link / no
crosscrosslink_4
link
6
6
4
7
5
2
3
2
3
n.d.
5
6
4
6
3
2
3
2
5
n.d.
n.d.
3
n.d.
4
n.d.
4
n.d.
5
n.d.
2
n.d.
n.d.
n.d.
7
7
n.d.
n.d.
5
7
n.d.
n.d.
5
4
n.d.
n.d.
6
6
6
6
6
6
6
6
6
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
5
7
5
6
4
6
4
7
3
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
4
4
7
4
2
4
2
4
3
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
4
4
5
4
3
2
2
3
5
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
27 kDa
35 kDa
6
6
n.d.
n.d.
6
6
n.d.
n.d.
5
4
n.d.
n.d.
3
4
n.d.
n.d.
65 kDa
6
n.d.
4
n.d.
5
n.d.
4
n.d.
44 kDa
6
n.d.
5
n.d.
2
n.d.
2
n.d.
2.8
2.0
2.0
1.6
APPENDIX
103
N14
Gene
name
Accession
Number
uvrA
yhfJ
ykvY
greA
thrS
trxA
BSU35160
BSU10250
BSU13860
BSU27320
BSU28950
BSU28500
ptsH
BSU13900
yqeY
ytiA
BSU25400
BSU30700
ywjH
minD
folD
BSU37110
BSU27990
BSU24310
codY
BSU16170
tpx
dnaN
BSU29490
BSU00020
yxxG
gltX
BSU39220
BSU00920
rnjB
dhbC
sodA
BSU16780
BSU31990
BSU25020
crosslink_1
N15
N14
no
crosscrosslink_2
link_1
N15
N15
cR1
crossno
link / no crosscrosscross- link_3
link_2
link
N14
N15
no
crosscrosslink_4
link_3
N14
cR2
crossno
link / no
crosscrosslink_4
link
Bio View: Identified Proteins (625)
MW
excinuclease ABC (subunit A)
similar to lipoate-protein ligase
similar to Xaa-Pro dipeptidase
transcription elongation factor
threonyl-tRNA synthetase (major)
antioxidative action by facilitating the
reduction of other proteins by cysteine
thiol-disulfide exchange
HPr, General component of the sugar
phosphotransferase system (PTS).
unknown
general stress protein, binds in the
stationary phase to the ribosome, replaces
RpmE under conditions of zinc limitation
transaldolase
cell-division inhibitor (septum placement)
methylenetetrahydrofolate dehydrogenase
(NADP)
regulation of a large region in response to
branched-chain amino acid limitation
probable thiol peroxidase
DNA polymerase III (beta subunit), beta
clamp
unknown
glutamyl-tRNA synthetase, universally
conserved protein
RNase J2
isochorismate synthase
superoxide dismutase, general stress
protein
106 kDa
38 kDa
40 kDa
17 kDa
74 kDa
11 kDa
6
6
6
6
6
6
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
5
5
6
6
6
7
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
6
3
5
4
7
7
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
2
3
4
3
5
7
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
9 kDa
6
n.d.
4
n.d.
4
n.d.
3
n.d.
17 kDa
10 kDa
6
6
n.d.
n.d.
7
7
n.d.
n.d.
2
4
n.d.
n.d.
3
4
n.d.
n.d.
23 kDa
29 kDa
31 kDa
6
6
6
n.d.
n.d.
n.d.
5
5
5
n.d.
n.d.
n.d.
5
4
4
n.d.
2
2
7
5
3
n.d.
n.d.
2
29 kDa
6
n.d.
4
n.d.
3
n.d.
3
n.d.
18 kDa
42 kDa
6
6
n.d.
2
7
6
2
2
6.5
3.0
5
4
n.d.
2
4
5
2
n.d.
4.5
4.5
16 kDa
56 kDa
6
5
2
n.d.
6
9
n.d.
n.d.
6.0
6
3
2
n.d.
5
4
2
n.d.
2.8
61 kDa
43 kDa
22 kDa
5
5
5
n.d.
n.d.
n.d.
9
8
7
n.d.
n.d.
n.d.
8
3
3
n.d.
n.d.
n.d.
6
3
4
n.d.
n.d.
n.d.
4.5
1.8
APPENDIX
104
N14
Gene
name
Accession
Number
gyrA
murE
BSU00070 DNA-Gyrase (subunit A)
BSU15180 UDP-N-acetylmuramoyl-L-alanyl-Dglutamyl-meso-2,6-diaminopimelate
synthetase
BSU35310 general stress protein, similar to Sigma-54
(SigL) modulating factor of gram-negative
bacteria
BSU33400 glyoxal reductase, general stress protein
BSU29560 similar to acetate-CoA ligase
BSU01770 phosphoglucosamine mutase, required for
cell wall synthesis
BSU34030 lactate catabolic enzyme
BSU18550 similar to xylulokinase
BSU20800 unknown
BSU16520 ribosome recycling factor
BSU00120 pyridoxal-5'-phosphate synthase
(glutaminase domain)
BSU26230 unknown
BSU01420 ribosomal protein S11
BSU01150 ribosomal protein S10
BSU00820 lysyl-tRNA synthetase
BSU15980 signal recognition particle (SRP)
component
BSU22960 trigger enzyme: glutamate dehydrogenase
(cryptic in 168 and derivatives)
BSU21600 similar to delta-endotoxin
BSU35360 flagellin protein
BSU32000 2,3-dihydro-2,3-dihydroxybenzoate
dehydrogenase
BSU11650 thiaminase II
yvyD
yvgN
ytcI
glmM
lutC
yoaC
yopQ
frr
pdxT
yqaP
rpsK
rpsJ
lysS
ffh
gudB
yokG
hag
dhbA
tenA
Bio View: Identified Proteins (625)
MW
crosslink_1
N15
N14
no
crosscrosslink_2
link_1
N15
N15
cR1
crossno
link / no crosscrosscross- link_3
link_2
link
N14
N15
no
crosscrosslink_4
link_3
N14
cR2
crossno
link / no
crosscrosslink_4
link
92 kDa
54 kDa
5
5
n.d.
n.d.
7
7
n.d.
n.d.
3
5
n.d.
n.d.
5
4
n.d.
n.d.
22 kDa
5
n.d.
6
n.d.
6
n.d.
3
n.d.
32 kDa
59 kDa
48 kDa
5
5
5
n.d.
n.d.
n.d.
6
6
6
n.d.
n.d.
n.d.
3
5
3
n.d.
n.d.
n.d.
2
2
2
n.d.
n.d.
n.d.
26 kDa
54 kDa
54 kDa
21 kDa
21 kDa
5
5
5
5
5
n.d.
n.d.
n.d.
n.d.
n.d.
6
6
6
6
6
n.d.
n.d.
n.d.
n.d.
n.d.
6
4
3
4
3
n.d.
n.d.
n.d.
n.d.
n.d.
4
2
3
4
4
n.d.
n.d.
n.d.
n.d.
n.d.
35 kDa
14 kDa
12 kDa
58 kDa
50 kDa
5
5
5
4
4
n.d.
n.d.
n.d.
n.d.
n.d.
5
5
4
7
7
n.d.
n.d.
n.d.
n.d.
n.d.
6
6
6
4
4
n.d.
n.d.
3
n.d.
n.d.
6
5
5
4
3
n.d.
n.d.
n.d.
n.d.
n.d.
47 kDa
4
n.d.
7
n.d.
4
n.d.
2
n.d.
41 kDa
33 kDa
27 kDa
4
4
4
n.d.
n.d.
n.d.
6
6
6
n.d.
n.d.
n.d.
3
4
5
n.d.
n.d.
n.d.
2
5
3
n.d.
n.d.
n.d.
27 kDa
4
n.d.
6
n.d.
4
n.d.
6
n.d.
3.7
APPENDIX
105
N14
Gene
name
Accession
Number
Bio View: Identified Proteins (625)
divIVA BSU15420 cell-division initiation protein (septum
placement)
spoVG BSU00490 negative effector of asymmetric septation
at the onset of sporulation
BSU02900
general stress protein, similar to tellurium
yceD
resistance protein
BSU01440 ribosomal protein L17
rplQ
BSU16160 two-component ATP-dependent protease,
clpY
ATPase subunit
BSU11700 hydroxyethylthiazole phosphate
thiF
biosynthesis
BSU13010 general stress protein
ykgB
BSU01040 ribosomal protein L10
rplJ
MW:
cR:
n. d.:
red highlighted:
Molecular weight
combined ratio
not detected
experimentally validated interaction partners
MW
crosslink_1
N15
N14
no
crosscrosslink_2
link_1
N15
N15
cR1
crossno
link / no crosscrosscross- link_3
link_2
link
N14
N15
no
crosscrosslink_4
link_3
N14
cR2
crossno
link / no
crosscrosslink_4
link
19 kDa
4
n.d.
5
n.d.
6
n.d.
6
n.d.
11 kDa
4
n.d.
5
n.d.
6
n.d.
5
n.d.
21 kDa
4
n.d.
4
n.d.
6
n.d.
3
n.d.
14 kDa
53 kDa
4
4
n.d.
n.d.
4
6
n.d.
2
6
6
n.d.
2
3
5
n.d.
2
36 kDa
3
n.d.
7
n.d.
7
n.d.
6
n.d.
38 kDa
18 kDa
2
2
n.d.
n.d.
5
6
n.d.
n.d.
6
5
n.d.
3
4
2
n.d.
n.d.
5.0
2.8
2.3